Towards the Immunogenic Hyperthermic Action

Towards the Immunogenic Hyperthermic Action: Modulated ElectroHyperthermia

A B S T R A C T

Hyperthermia treatment for solid tumors is a long-used, but poorly accepted method in clinical use. Modulated electro-hyperthermia (mEHT, trade name: oncothermia®) changes the paradigm, introduces a novel, cellularly selective and immunogenic cell-ruination. The mEHT method produces tumor-vaccination, presenting the unharmed genetic information of cancer cells to immune cells [1]. The mEHT method is approved in more than 30 countries. Its phase II/III clinical applications indicate a broad perspective.

Keywords

Hyperthermia, oncology, immunogenic cell death

Introduction

Hyperthermia in oncology faces certain challenges [2]. The poor performances of the hyperthermia acceptance in the medical community developed a general opinion, blaming the physics and the technical solutions for unsuccessful processes. Modulated electro-hyperthermia (mEHT, trade name oncothermia®) represents a new paradigm of hyperthermia treatment for solid tumors, which gives a hint on how to apply physics in this complex phenomenon [3, 4]. The 30-year history of mEHT covers multiple discoveries and many publications [5]. The mEHT technique heats malignant cells selectively. Contrary to the conventional isothermal approach, it follows the natural heterogeneity of the tumor-structure by selective energy-absorption [6]. The selection uses the electromagnetic heterogeneity of the malignant lesion [7]. Furthermore, the cancer lesion drastically modifies the collective harmony in the attacked organ, which could be repaired in the time-fractal modulation of the mEHT technology [8]. The main physiology parameters of malignant cells differ from their healthy counterpart. Conductivity, due to the high metabolic rate, and the variation of the dielectric constant of the microenvironments of the cells, due to the loss of intercellular bonds are higher [9, 10]. The selective frequency dispersion, which focuses on the lipid-protein interactions, chooses the intracellular proteins as targets [11, 12]. The main medical task to treat advanced malignant diseases requests that the systemic treatment is effective on possible micro and macrometastases all over the body. The local technique of mEHT produces immune effects, which does the required systemic task [13].

Method

The mEHT method focuses on the bioelectromagnetic field of malignant cells. It uses sophisticated technical mechanisms to select and “gently” kill cancer cells while presenting genetic information about these cells to the immune cells. This “message” carried by various proteins is able to produce the presentation of antigens, developing the T-cells for targeting and killing cancer cells all over the body.

The technique is a capacitive impedance coupled with electromagnetic heating. However, the coupling is not plane wave as usual for capacitive methods, but strict impedance tuning to the cancerous target [14]. Interconnected conductive and dielectric effects establish the base for the impedance-selection process, realizing a cellular selection [15, 16]. The mEHT technique uses 13.56 MHz radiofrequency [17]. An amplitude modulation technique supports the focusing of the electric field on the cellular membrane with a time-fractal (1/f) pattern [18]. The similarity of the electric field and temperature actions allows the creation of the desired effects [19]. Cellular selection is performed with high preciosity, showing the excitation of the rafts of the cytoplasmic membrane well [20, 21]. The selective heating of the membrane rafts affects the cytoskeleton and its polymerization too [22, 23]. The physical effects of the mEHT method are described with various aspects, discussing the non-thermal energy-exchange possibility in connection with thermal conditions [24-27].

The dose in the selective heterogenic heating differs from the general dose concept of conventional hyperthermia [28, 29]. This dose is based on energy absorption, similarly, to ionizing radiation therapies [30]. Energy absorption depends on the absorbed power and of course, is connected to the temperature that produced it, but it describes the real effects more accurately [31]. It is a generalization of the conventional hyperthermia dose [32]. It is a real advantage that the penetration depth of the selective heating is larger than what the isothermal heating achieves [33]. Importantly, the heat-up period of the treatment, when the energy absorption is quasi-adiabatic, provides the best effect, offering the optimization of the treatment protocol [34].

Discussion

mEHT has a long history from laboratory experiments to human therapies [35]. The excitation of the membrane rafts is accompanied by the point-connected cells, which are common at the end of mitosis (cytokinetic phase) [36, 37]. The heat-map difference between the conventionally heated and mEHT treated cell-suspension showed certain deviations in up, and down-regulated genes in GeneChip measurement [38]. Importantly, the combination of mEHT with chemotherapy in vitro and in vivo as well as with radiation therapy in vitro and in vivo show the broad application variability of the method in preclinical experiments, which was also shown in clinical phase in both chemotherapies and radiation therapies (See details below) [39-44]. The thermal dose with temperature mapping is also shown in vivo, as well as the radiation equivalent of mEHT is estimated [45, 46].

mEHT causes massive apoptosis by exciting the death-toll receptors and transient reaction-potential channels of the membrane [47, 48]. The observed apoptotic cell-distraction is significantly more intensive with mEHT than with water-bath or other conventional capacitive coupling hyperthermia methods [49]. The large number of apoptotic bodies, together with the correct time-sequences of cell membrane exposure of Calreticulin, the release of HMGB1 protein, and membrane expression of HSP70 form a damage-associated molecular pattern. The spatiotemporal series of liberated molecules from the cytoplasm to the extracellular electrolyte (Calreticulin - “eat me signal”, HMGB1- “danger signal”, released HSP70- “info signal”, and extracellular ATP - “find me signal”) triggers immunogenic cell-death (ICD) [50]. ICD initiates the maturation of the dendritic cells (DCs) and forms a tumor-specific antigen presentation, shaping CD4+ and CD8+ T-cells in situ, real-time, without extra-corporal laboratory preparations. The prepared T-cells act throughout the whole body by blood flow transports, causing an abscopal effect on distant micro and macrometastases, increasing the survival time as well [51]. The concomitant application of the (anyway ineffective) unmatured DCs form antigen presentation cells (APCs), and significantly increase the survival [52, 53]. Importantly the re-challenge of the same tumor into the cured animal was unsuccessful; the process has worked like a tumor-vaccination [1].

The mEHT method has a broad application possibility in oncologic clinical use [54]. mEHT is a complementary method and could be applied together with all other conventional treatments, including chemotherapies and radiotherapies [3, 55, 56]. Importantly, the strong selective energy absorption on the membrane rafts helps the cellular killing as well as the drug penetration into the cell, and the moderate isothermal heat effect pumps the blood to the selected tumor helping the drug delivery and also improving the oxygenation which sensitizes the radiotherapy action. Oxygen delivery is strongly supported by the separation of the grouped erythrocytes (destroys the rouleaux formation of them), so oxygen delivery does not need high local isothermal temperature [57]. The moderate isothermal gain of temperature does not improve the metastatic dissemination, which could be a problem in higher heating.

Such a sensitive organ as the brain could be safely treated by a dose-escalation safety trial for advanced, relapsed glioblastoma patients, and an effective curative potential was presented in this group of patients, integrated the therapy in complex manner in the clinical practice [58-60]. A larger group of brain gliomas shows the same positive effect, and another trial has detected the immunogenic behaviour in a complex integrative therapy [61, 62]. This brain treatment can be personalized, and it shows its advantages in both clinical and economic considerations in meta-analyses [63, 64]. The results in rectal cancer, gastric cancer, and others, show the gastrointestinal capability of mEHT, strongly supported by clinical trials on primary and metastatic origin of liver cancer [65-70]. The anyway problematic inoperable pancreatic cancer also shows the feasibility of mEHT in this disease [71, 72]. The worldwide problematic large morbidity of lung cancers needs the complex solutions of the therapy, which could be given by mEHT [73]. The high complexity of the therapy could use different methods complementary to chemotherapy, including mEHT too [74]. Complementary mEHT could be used together with chemotherapy and radiotherapy too, even in the aggressive small-cell lung carcinoma [75-77]. There was an interesting observation, a successful clinical study with intravenous high-dose vitamin C for heavily pretreated, advanced, refractory non-small-cell lung cancer [78].

mEHT proved its feasibility in gynaecological cancers too. Both the local tumor suppression and longer survival time were observed in advanced cervical cancers by mEHT with chemotherapy compared to chemotherapy alone [79]. Chemo-radiotherapy for recurrent cervical cancer was also more effective in case of the complementary application of mEHT [80]. A phase III extended clinical trial on local disease control in HIV-positive and negative cervical cancer shows significant improvement [44]. The survival of cervical cancer patients with or without associated HIV infection and treated with mEHT combined with chemo-radiotherapy shows the strong capability of the method for long-term measures as well [81]. An important addition is that the treatment-related toxicity of the mEHT was suppressed, and at the same time, the treatment improves the quality of life of HIV-positive cervical cancer patients [82]. The detailed analysis of nodal disease on PET/CT scans in patients with HIV-positive and negative locally advanced cervical cancer shows the improvement of the patient status metabolically too [83]. The treatment of ovarian cancer has also shown advantages with the complementary application of mEHT in a clinical trial, and the successful mEHT complementary treatment of advanced ovarian cancer with thermo-chemotherapy and adjuvant immune therapy presented a significant improvement [84, 85]. Others independently showed the feasibility of complex multimodal immunotherapy for patients with ovarian cancer [86]. The complex approach showed its capability well in breast cancer too, which was also well-supported by immunotherapy [87, 88]. The larger group of breast cancer patients also proved the efficacy of the mEHT method in a single-centre experience [89]. The treatment of advanced triple-negative breast cancers by mEHT is also feasible [90].

The primary leiomyosarcoma of the breast following salvage mEHT and pazopanib, as well as recurrent metastatic sarcoma, indicates the applicability of the method in this type of solid tumors, which was proven in more cases, and in clinical trial too [91-93]. The advantages of the complex approach are shown by an 8-year observational study for the survival time of various advanced solid tumors treated complementarily by mEHT, as well as for various malignant diseases when mEHT was applied independently [94, 95]. Cases that respond to the mEHT monotherapy are also collected, showing success, and sometimes turning the acute, fatal cancer to a chronic, long-term, manageable disease [96].

The abscopal effect using the local mEHT allows the overall action in the system; it treats the micro and macrometastases in distant locations [97]. The abscopal immune actions are shown in some clinical applications for advanced metastatic cancers including brain tumors too [62, 87, 88, 98]. The potentiation of the abscopal outcome was one of the successful parameters in advanced cervical cancer treatment with mEHT and was proven in some cases in combination with radiotherapy too [99, 100]. It shows the effects of tumor-directed immunotherapy [101].

Conclusion

The method mEHT has a broad application possibility in combination with other conventional cancer treatments or even in monotherapy application when the condition of the patient does not allow the complementary therapy. mEHT extends the local hyperthermia treatment to a systemic one by the abscopal effect, making the treatment of advanced cancers with distant metastases available.

The preclinical and clinical results support the preliminary expectations formulated on theoretical and model basis. The observed clinical results allow the development of a guideline for local hyperthermia treatment concentrating on mEHT in oncology, with the authorship of 28 physicians from 12 countries (Austria, Canada, China, Germany, Greece, Hungary, Italy, Switzerland, South-Korea, South-Africa, Spain, and Taiwan) [102].

Download PDF View PDF


Article Info

Article Type
Research Article

Publication history

Received:

Accepted:

Published:


Copyright

© 2023 Andras Szasz. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Hosting by Science Repository.

DOI: 10.31487/j.COR.2020.09.07


Author Info

Andras Szasz


Corresponding Author
Andras Szasz
Professor, Chair, Department of Biotechnics, St. Istvan University, Hungary
  1. Andocs G, Szasz O, Szasz A, Iluri N (2015) Tumor vaccination patent, Nr. US20150217099A1, (2016) Nr. EP2780024B1.
  2. Minnaar C (2020) Challenges associated with hyperthermia, in book Challenges and solutions of oncological hyperthermia. 1-31.
  3. Szasz A, Szasz N, Szasz O (2010) Oncothermia-Principles and practices, Springer, Heidelberg.
  4. Wust P, Nadobny J, Zschaeck S, Ghadjar P (2020) Physics of hyperthermia-Is physics really against us?, in book Challenges and solutions of oncological hyperthermia 346-376.
  5. Szasz A, Iluri N, Szasz O (2013) Local hyperthermia in Oncology-To Choose or not to Choose? A chapter in book: Hyperthermia 1-82.
  6. Szasz A (2019) Thermal and nonthermal effects of radiofrequency on living state and applications as an adjuvant with radiation therapy. J Radiation Cancer Res 10: 1-17.
  7. Szasz A (2013) Electromagnetic effects in nanoscale range. Cellular Response to Physical Stress and Therapeutic Applications. Nova Science Publishers.
  8. Szasz O, Andocs G, Meggyeshazi N (2013) Modulation effect in oncothermia. Hindawi Publishing Corporation Conference Papers in Medicine 2013.
  9. Warburg O (1966) Oxygen, The Creator of Differentiation, Biochemical Energetics. Acad Press, New York, (Warburg O: The Prime Cause and Prevention of Cancer, Revised lecture at the meeting of the Nobel-Laureates on June 30, 1966 at Lindau, Lake Constance, Germany).
  10. Szentgyorgyi A (1968) Bioelectronics, A Study on Cellular Regulations, Defense and Cancer. Acad Press New York, London.
  11. Martinsen OG, Grimnes S, Schwan HP (2002) Interface phenomena and dieslectric properties of biological tissue, Encyclopedia of Surface and Colloid Science, CRC Press, Taylor & Francis Group 2643-2652.
  12. B E Pennock, H P Schwan (1969) Further observations on the electrical properties of hemoglobin bound water. J Phys Chem 73: 2600-2610. [Crossref]
  13. Dank M, Meggyeshazi N, Szigeti Gy, Andocs G (2016) Immune effects by selective heating of membrane rafts of cancer-cells. ASCO Annual Meeting.
  14. Ferenczy GL, Szasz A (2020) Technical challenges and proposals in oncological hyperthermia, in book Challenges and solutions of oncological hyperthermia 72-90.
  15. Szigeti GP, Szasz O, Hegyi G (2017) Connections between Warburg’s and Szentgyorgyi’s Approach about the Causes of Cancer. J Neoplasm 1: 1-13.
  16. Szasz A (2013) Challenges and Solutions in Oncological Hyperthermia. Thermal Med 29: 1-23.
  17. Baronzio G, Parmar G, Ballerini M, Szasz A, Baronzio M et al. (2014) A brief overview of hyperthermia in cancer treatment. J Integrative Oncol 3: 1.
  18. Szasz A, Szasz O (2020) Time-fractal modulation of modulated electro-hyperthermia (mEHT), in book Challenges and solutions of oncological hyperthermia 377-415.
  19. Vincze Gy, Szasz A (2018) Similarities of modulation by temperature and by electric field. OJBIPHY 8: 95-103.
  20. Szasz O, Szasz AM, Minnaar C, Szasz A (2017) Heating preciosity - trends in modern oncological hyperthermia. Open J Biophys 7: 116-144.
  21. Papp E, Vancsik T, Kiss E, Szasz O (2017) Energy absorption by the membrane rafts in the modulated electro-hyperthermia (mEHT). Open J Biophys 7: 216-229.
  22. Vincze Gy, Sziget GyP, Szasz A (2016) Reorganization of the cytoskeleton. J Adv Biology 9: 1872-1882.
  23. Vincze Gy, Szasz A (2015) Reorganization of actin filaments and microtubules by outside electric field. J Adv Biology 8: 1514-1518.
  24. Szasz A (2015) Bioelectromagnetic Paradigm of Cancer Treatment Oncothermia. Bioelectromagnetic Subtle Energy Medicine 323-336.
  25. Peter Wust, Pirus Ghadjar, Jacek Nadobny, Marcus Beck, David Kaul et al. (2019) Physical analysis of temperature-dependent effects of amplitude-modulated electromagnetic hyperthermia. Int J Hyperthermia 36: 1246-1254. [Crossref]
  26. Peter Wust, Benedikt Kortüm, Ulf Strauss, Jacek Nadobny, Sebastian Zschaeck et al. (2020) Non-thermal effects of radiofrequency electromagnetic fields. Sci Rep 10: 13488. [Crossref]
  27. Szasz A (2019) Thermal and nonthermal effects of radiofrequency on living state and applications as an adjuvant with radiation therapy. J Radiation Cancer Res 10: 1-17.
  28. A Szasz, Gy Vincze (2006) Dose concept of oncological hyperthermia: heat-equation considering the cell destruction. J Cancer Res Ther 2: 171-181. [Crossref]
  29. Szasz O, Szasz A (2016) Heating, efficacy and dose of local hyperthermia. Open J Biophys 6: 10-18.
  30. Sun Young Lee, Gyula Peter Szigeti, Attila Marcell Szasz (2019) Oncological hyperthermia: The correct dosing in clinical applications. Int J Oncol 54: 627-643. [Crossref]
  31. Szasz O, Szigeti GyP, Szasz A (2016) Connections between the specific absorption rate and the local temperature. Open J Biophys 6: 53-74.
  32. Vincze Gy, Szasz O, Szasz A (2015) Generalization of the thermal dose of hyperthermia in oncology. Open J Biophys 5: 97-114.
  33. Szasz O, Szigeti GyP, Vancsik T, Szasz A (2018) Hyperthermia dosing and depth of effect. Open J Biophys 8: 31-48.
  34. Patrick Hung Ju Kao, Chia Hung Chen, Yuk Wah Tsang, Chen Si Lin, Hsin Chien Chiang et al. (2020) Relationship between Energy Dosage and Apoptotic Cell Death by Modulated Electro-Hyperthermia. Sci Rep 10: 8936. [Crossref]
  35. G Andocs, O Szasz, A Szasz (2009) Oncothermia treatment of cancer: from the laboratory to clinic. Electromagn Biol Med 28: 148-165. [Crossref]
  36. Szasz O, Andocs G, Kondo T, Rehman MU, Papp E et al. (2015) Heating of membrane raft of cancer-cells, ASCO Annual Meeting. J Clin Oncol 33.
  37. Andocs G, Rehman MU, Zhao QL, Papp E, Kondo T et al. (2015) Nanoheating without Artificial Nanoparticles Part II. Experimental support of the nanoheating concept of the modulated electro-hyperthermia method, using U937 cell suspension model. Biology Med 7: 1-9.
  38. G Andocs, M U Rehman, Q L Zhao, Y Tabuchi, M Kanamori et al. (2016) Comparison of biological effects of modulated electro-hyperthermia and conventional heat treatment in human lymphoma U937 cell. Cell Death Discov 2: 16039. [Crossref]
  39. Jihye Cha, Tae Won Jeon, Chang Geol Lee, Sang Taek Oh, Hee Beom Yang et al. (2015) Electro-hyperthermia inhibits glioma tumorigenicity through the induction of E2F1-mediated apoptosis. Int J Hyperthermia 31: 784-792. [Crossref]
  40. Tamas Vancsik, Gertrud Forika, Andrea Balogh, Eva Kiss, Tibor Krenacs (2019) Modulated electro-hyperthermia induced p53 driven apoptosis and cell cycle arrest additively support doxorubicin chemotherapy of colorectal cancer in vitro. Cancer Med 8: 4292-4303. [Crossref]
  41. Beomseok Son, Jaewan Jeon, Sungmin Lee, Hyunwoo Kim, Hyunkoo Kang et.al. (2019) Radiotherapy in combination with hyperthermia suppresses lung cancer progression via increased NR4A3 and KLF11 expression. Int J Radiat Biol 95: 1696-1707. [Crossref]
  42. Kim J K, Prasad B, Kim S (2017) Temperature mapping and thermal dose calculation in combined radiation therapy and 13.56 MHz radiofrequency hyperthermia for tumor treatment. 1004718.
  43. Fiorentini G, Casadei V (2018) Modulated Electro-Hyperthermia (mEHT) in Integrative Cancer Treatment for Relapased Malignant Gliobastroma and Astrocytoma: a retrospective multicenter controlled study. Oncothermia J 24: 464-481.
  44. Minnaar CA, Kotzen JA, Ayeni OA (2019) The effect of modulated electro-hyperthermia on local disease control in HIV-positive and -negative cervical cancer women in South Africa: Early results from a phase III randomized controlled trial. Plos One 14: e0217894.
  45. Bibin Prasad, Subin Kim, Woong Cho, Suzy Kim, Jung Kyung Kim et al. (2018) Effect of tumor properties on energy absorption, temperature mapping, and thermal dose in 13,56-MHz radiofrequency hyperthermia. J Therm Biol 74: 281-289. [Crossref]
  46. Prasad B, Kim S, Cho W, Kim JK, Kim YA et al. (2019) Quantitative estimation of the equivalent radiation dose escalation using radiofrequency hyperthermia in mouse xenograft models of human lung cancer. Sci Rep, Nature 9: 3942.
  47. N Meggyeshazi, G Andocs, L Balogh, P Balla, G Kiszner et al. (2014) DNA fragmentation and caspase-independent programmed cell death by modulated electrohyperthermia. Strahlenther Onkol 190: 815-822. [Crossref]
  48. Vincze Gy, Szigeti Gy, Andocs G, Szasz A (2015) Nanoheating without Artificial Nanoparticles. Biology Med 7: 249. [Crossref]
  49. Kai Lin Yang, Cheng Chung Huang, Mau Shin Chi, Hsin Chien Chiang, Yu Shan Wang et.al. (2016) In vitro comparison of conventional hyperthermia and modulated electro-hyperthermia. Oncotarget 7: 84082-84092. [Crossref]
  50. Gabor Andocs, Nora Meggyeshazi, Lajos Balogh, Sandor Spisak, Mate Elod Maros et al. (2014) Upregulation of heat shock proteins and the promotion of damage-associated molecular pattern signals in a colorectal cancer model by modulated electrohyperthermia. Cell Stress Chaperones 20: 37-46. [Crossref]
  51. Wei Qin, Yasunori Akutsu, Gabor Andocs, Akiko Suganami, Xin Hu et al. (2014) Modulated electro-hyperthermia enhances dendritic cell therapy through an abscopal effect in mice. Oncol Rep 32: 2373-2379. [Crossref]
  52. Yuk Wah Tsang, Cheng Chung Huang, Kai Lin Yang, Mau Shin Chi, Hsin Chien Chiang et al. (2015) Improving immunological tumor microenvironment using electro-hyperthermia followed by dendritic cell immunotherapy. BMC Cancer 15: 708. [Crossref]
  53. Tamas Vancsik, Csaba Kovago, Eva Kiss, Edina Papp, Gertrud Forika et al. (2018) Modulated electro-hyperthermia induced loco-regional and systemic tumor destruction in colorectal cancer allografts. J Cancer 9: 41-53. [Crossref]
  54. Attila M Szasz, Carrie Anne Minnaar, Gyongyver Szentmártoni, Gyula P Szigeti, Magdolna Dank et al. (2019) Review of the clinical evidences of modulated electro-hyperthermia (mEHT) method: an update for the practicing oncologist. Front Oncol 9: 1012. [Crossref]
  55. Szasz O (2013) Renewing Oncological Hyperthermia-Oncothermia. Open J Biophys 3: 245-252.
  56. Szasz O (2020) Development in oncological hyperthermia, Chapter 11 in book New insights into physical science. Book Publisher International 1: 1-13.
  57. Saupe H, Szigeti GyP, Andocs G (2016) Why modulated electrohyperthermia (mEHT) destroys the rouleaux formation of erythrocytes? J Advances Biology 9: 1945-1955.
  58. Caecilia Wismeth, Christine Dudel, Christina Pascher, Paul Ramm, Torsten Pietsch et al. (2010) Transcranial electro-hyperthermia combined with alkylating chemotherapy in patients with relapsed high-grade gliomas: phase I clinical results. J Neurooncol 98: 395-405. [Crossref]
  59. Fiorentini G, Sarti D, Casadei V (2020) Modulated electro-hyperthermia for the treatment of relapsed brain gliomas, in book Challenges and solutions of oncological hyperthermia 110-125.
  60. Giammaria Fiorentini, Donatella Sarti, Carlo Milandri, Patrizia Dentico, Andrea Mambrini et al. (2019) Modulated electrohyperthermia in integrative cancer treatment for relapsed malignant glioblastoma and astrocytoma: Retrospective multicenter controlled study. Integr Cancer Ther. 18: 1534735418812691. [Crossref]
  61. Sahinbas H, Groenemeyer DHW, Boecher E, Szasz A (2007) Retrospective clinical study of adjuvant electro-hyperthermia treatment for advanced brain-gliomas. Deutsche Zeitschrift fuer Onkologie 39: 154-160.
  62. Van Gool SW, Makalowski J, Feyen O, Prix L, Schirrmacher V et al. (2018) The induction of immunogenic cell death (ICD) during maintenance chemotherapy and subsequent multimodal immunotherapy for glioblastoma (GBM). Austin Oncol Case Rep 3: 1-8.
  63. Van Gool SW, Makalowski J, Domogalla MP (2020) Personalised medicine in glioblastoma multiforme, in book Challenges and solutions of oncological hyperthermia 126-158.
  64. Sergey V Roussakow (2017) Clinical and economic evaluation of modulated electrohyperthermia concurrent to dose-dense temozolomide 21/28 days regimen in the treatment of recurrent glioblastoma: a retrospective analysis of a two-centre German cohort trial with systematic comparison and effect-to-treatment analysis. BMJ Open 7: e017387. [Crossref]
  65. S H You, S Kim (2019) Feasibility of modulated electro-hyperthermia in preoperative treatment for locally-advanced rectal cancer: Early phase 2 clinical results. Neoplasma 67: 677-683. [Crossref]
  66. Iyikesici MS, Slocum A, Turkmen E, Akdemir O, Slocum AK  et al. (2016) Complete response of locally advanced (stage III) rectal cancer to metabolically supported chemoradiotherapy with hyperthermia. Int J Cancer Res Mech 2.
  67. M S Iyikesici (2020) Survival outcomes of metabolically supported chemotherapy combined with Ketogenic diet, hyperthermia, and hyperbaric oxygen therapy in advanced gastric cancer. Niger J Clin Pract 23: 734-740. [Crossref]
  68. Garay T, Kiss E, Szentmartoni Gy (2020) Gastrointestinal cancer series treated with modulated electro-hyperthermia (mEHT) – A single centre experience, in book Challenges and solutions of oncological hyperthermia. 159-162.
  69. Gennaro Gadaleta Caldarola, Stefania Infusino, Ida Galise, Girolamo Ranieri, Gianluca Vinciarelli et al. (2014) Sorafenib and locoregional deep electro-hyperthermia in advanced hepatocellular carcinoma. A phase II study. Oncol Lett 8: 1783-1787. [Crossref]
  70. E D Hager, H Dziambor, D Höhmann, D Gallenbeck, M Stephan et al. (1999) Deep hyperthermia with radiofrequencies in patients with liver metastases from colorectal cancer. Anticancer Res 19: 3403-3408. [Crossref]
  71. Mehmet Salih Iyikesici (2020) Long-term survival outcomes of metabolically supported chemotherapy with Gemcitabine-based or FOLFIRINOX regimen combined with Ketogenic diet, hyperthermia, and hyperbaric oxygen therapy in metastatic pancreatic cancer. Complement Med Res 27: 31-39. [Crossref]
  72. Volovat C, Volovat SR, Scripcaru V, Miron L (2014) Second-line chemotherapy with gemcitabine and oxaliplatin in combination with loco-regional hyperthermia (EHY-2000) in patients with refractory metastatic pancreatic cancer - preliminary results of a prospective trial. Romanian Reports Physics 66:166-174.
  73. Andras Szasz (2014) Current status of oncothermia therapy for lung cancer. Korean J Thorac Cardiovasc Surg 47: 77-93. [Crossref]
  74. Mehmet Salih Iyikesici (2019) Feasibility study of metabolically supported chemotherapy with weekly carboplatin/paclitaxel combined with ketogenic diet, hyperthermia and hyperbaric oxygen therapy in metastatic non-small cell lung cancer. Int J Hyperthermia 36: 445-454. [Crossref]
  75. Lee DY, Park SJ, Jung HC, Byun ES, Haam SJ et al. (2015) The Outcome of the Chemotherapy and Oncothermia for Far Advanced Adenocarcinoma of the Lung: Case reports of four patients. Adv Lung Cancer 4: 1-7.
  76. Seung Gu Yeo (2015) Definitive radiotherapy with concurrent oncothermia for stage IIIB non-small-cell lung cancer: A case report. Exp Ther Med 10: 769-772. [Crossref]
  77. Lee DJ, Haam SJ, Kim TH, Lim JY, Kim EJ et al. (2013) Oncothermia with chemotherapy in the patients with Small Cell Lung Cancer. Hindawi Publishing Corporation Conference Papers in Medicine 2013.
  78. Junwen Ou, Xinyu Zhu, Pengfei Chen, Yanping Du, Yimin Lu et al. (2020) A randomized phase II trial of best supportive care with or without hyperthermia and vitamin C for heavily pretreated, advanced, refractory non-small-cell lung cancer. J Adv Res 24: 175-182. [Crossref]
  79. Sun Young Lee, Na Ri Lee, Dong Hyu Cho, Jung Soo Kim et al. (2017) Treatment outcome analysis of chemotherapy combined with modulated electro-hyperthermia compared with chemotherapy alone for recurrent cervical cancer, following irradiation. Oncol Lett 14: 73-78. [Crossref]
  80. Lee SY (2020) Concurrent chemo-hyperthermia for recurrent cervical cancer after previous CCRT, in book Challenges and solutions of oncological hyperthermia. 163-186.
  81. Minnaar C, Baeyens A, Kotzen J, Vangu MD (2018) Survival of cervical cancer patients with or without associated HIV infection and treated with modulated electro-hyperthermia combined with chemo-radiotherapy, 32nd Annual Meeting of the European Hyperthermia Society, OP 13. Strahlenther Onkol 194: 476.
  82. Carrie Anne Minnaar, Jeffrey Allan Kotzen, Thanushree Naidoo, Mariza Tunmer, Vinay Sharma et al. (2020) Analysis of the effects of mEHT on the treatment-related toxicity and quality of life of HIV-positive cervical cancer patients. Int J Hyperthermia 37: 263-272. [Crossref]
  83. Minnaar CA, Baeyens A, Aeni OA (2019) Defining characteristics of nodal disease on PET/CT scans in patients with HIV-positive and -negative locally advanced cervical cancer in South Africa. Tomography 5: 339-345.
  84. Heon Jong Yoo , Myong Cheol Lim, Sang Soo Seo, Sokbom Kang, Jungnam Joo et al. (2019) Phase I/II clinical trial of modulated electro-hyperthermia treatment in patients with relapsed, refractory or progressive heavily treated ovarian cancer. Jpn J Clin Oncol 49: 832-838. [Crossref]
  85. R Kleef, S Kekic, N Ludwig (2012) Successful treatment of advanced ovarian cancer with thermochemotherapy and adjuvant immune therapy. Case Rep Oncol 5: 212-215. [Crossref]
  86. Van Gool S, Makalowski J, Marko M (2019) Multimodal immunotherapy for patients with ovarian cancer. Oncothermia J 27: 138-152.
  87. Mehmet Salih İyikesici, Abdul Kadir Slocum, Ayshe Slocum, Ferhan Bulent Berkarda, Miriam Kalamian et al. (2017) Efficacy of metabolically supported chemotherapy combined with ketogenic diet, hyperthermia, and hyperbaric oxygen therapy for stage IV triple-negative breast cancer. Cureus 9: e1445. [Crossref]
  88. Volker Schirrmacher, Wilfried Stücker, Maria Lulei, Akos Sigmund Bihari, Tobias Sprenger et al. (2015) Long-term survival of a breast cancer patient with extensive liver metastases upon immune and virotherapy: a case report. Immunotherapy 7: 855-860. [Crossref]
  89. Szasz AM, Szentmartoni Gy, Garay T, Reka Mohacsi, Mariann Kvansika (2020) Breast cancer series treated with modulated electro-hyperthermia (mEHT)- A single centre experience, in book Challenges and solutions of oncological hyperthermia 105-109.
  90. Garay T, Borbényi E, Szasz AM (2020) Treatment of locally advanced triple-negative breast cancer with oncothermia, in book Challenges and solutions of oncological hyperthermia 187-191.
  91. Sun Young Lee, Na Ri Lee (2018) Positive response of a primary leiomyosarcoma of the breast following salvage hyperthermia and pazopanib. Korean J Intern Med 33: 442-445. [Crossref]
  92. Jeung TS, Ma SY, Choi J, Yu J, Lee SY et al. (2015) Results of oncothermia combined with operation, chemotherapy and radiation therapy for primary, recurrent and metastatic sarcoma. Case Reports Clin Med 4: 157-168.
  93. Volovat C, Volovat SR, Scripcaru V, Lupascu C (2014) The results of combination of ifosfamid and locoregional hyperthermia (EHY 2000) in patients with advanced abdominal soft-tissue sarcoma after relapse of first line chemotherapy. Romanian Rep Physics 66: 175-181.
  94. Parmar G, Rurak E, Elderfield M (2020) 8-year observational study on naturopathic treatment with modulated electro-hyperthermia (mEHT): A single-centre experience, in book Challenges and solutions of oncological hyperthermia 227-266.
  95. Lee Y (2013) Oncothermia Application for Various Malignant Diseases. Hindawi Publishing Corporation Conference Papers in Medicine 2013.
  96. Jeung TS, Ma SY, Yu J, et al. (2013) Cases that respond to oncothermia monotherapy. Conf Papers Medicine 2013.
  97. Szasz O (2020) Local treatment with systemic effect: Abscopal outcome, in book Challenges and solutions of oncological hyperthermia 192-205.
  98. Volker Schirrmacher, Akos Sigmund Bihari, Wilfried Stücker, Tobias Sprenge (2014) Long-term remission of prostate cancer with extensive bone metastases upon immuno- and virotherapy: A case report. Oncology Lett 8: 2403-2406. [Crossref]
  99. Carrie Anne Minnaar, Jeffrey Allan Kotzen, Olusegun Akinwale Ayeni, Mboyo Di Tamba Vangu, Ans Baeyen (2020) Potentiation of the abscopal effect by modulated electro-hyperthermia in locally advanced cervical cancer patients. Front Oncol 10: 1-8. [Crossref]
  100. Mau Shin Chi, Minesh P Mehta, Kai Lin Yang, Hung Chih Lai, Ying Chu Lin et al. (2020) Putative abscopal effect in three patients treated by combined radiotherapy and modulated electrohyperthermia. Front Oncol 9: 254. [Crossref]
  101. Chi K H (2020) Tumour-directed immunotherapy: Clinical results of radiotherapy with modulated electro-hyperthermia, in book Challenges and solutions of oncological hyperthermia 206-226.
  102. Szasz AM, Arkosy p, Arrojo EE (2020) Guidelines for local hyperthermia treatment in oncology, in book Challenges and solutions of oncological hyperthermia A 32-71.