search
for
 About Bioline  All Journals  Testimonials  Membership  News


Journal of Cancer Research and Therapeutics
Medknow Publications on behalf of the Association of Radiation Oncologists of India (AROI)
ISSN: 0973-1482 EISSN: 1998-4138
Vol. 2, Num. 2, 2006, pp. 41-46

Journal of Cancer Research and Therapeutics, Vol. 2, No. 2, April-June, 2006, pp. 41-46

Review Article

Hyperthermia today: Electric energy, a new opportunity in cancer treatment

Department of Oncology, San Giuseppe General Hospital, Empoli (Florence), Italy. *Department of Biotechnics, Faculty Engineering, Szent Istvan University, Gödölla, Hungary
Correspondence Address:Medical Oncology Unit, San Giuseppe General Hospital, Viale Paladini 40, 50053 Empoli (Florence), oncologiaempoli@usl11.toscana.it

Code Number: cr06010

Abstract

Hyperthermia is an ancient, but nowadays rapidly developing treatment method in tumor-therapy. Its new paradigm applied in the electro-hyperthermia (oncothermia), which provides energy by means of electric-field and produces non-equilibrium thermal situation in the tissue. The temperature gradients formed in stationer conditions, destroy the membrane of the malignant cells and selectively eliminate the cancer tissue. The characteristic control parameter is the absorbed energy-dose, which is partly used to make the distortions, partly to increase the temperature of the target. This type of technique could be applied for some tumor sites, including brain, soft tissues, liver and abdominal masses, pancreatic cancer, head and neck tumors as well.

Keywords: Hyperthermia, solid tumors, electric energy.

Introduction

Cancer and its treatment have been one of the greatest challenges in the medical science for centuries. Nowadays, enormous economic and human resources are involved in this field, but according to the epidemic data the solution will still be awaited for. Sure, cancer is not the first and probably not the last one among the diseases which despite of the exceptional human efforts have not had any cure for a long time. The use of hyperthermia for cancer therapy has been documented for thousands of years. The first provable application was attributed to Hypocrites, whose approach of course was mainly supported by the Greek philosophy, where the fire (heat) had the highest level of abilities and freedom. The method was later forgotten. It was revived around the end of the 19th century, when the deep penetrating energy transfer was solved by electromagnetic way. As early as in 1912, a controlled Phase II clinical study on 100 patients was published, showing the benefit of the thermo-radiation therapy.[1] Nevertheless, hyperthermia is still in its starting phase, it carries all the problems of the method in early stages: not enough scientific proofs have been collected yet. The situation today is similar to that of radiology at its early stages. When ionizing radiation was first discovered, many hypothesized its usefulness in oncology, yet its exact techniques, dose, contraindications, limits and the conditions of optimal treatment were determined only several decades later. This is a natural process: every beginning shows these development.

Hyperthermia suffers from a lack of standards and a lack of scientific consensus about its effects on malignant and healthy tissues. In order that hyperthermia will gain widespread approval and clinical use, the technique requires extensive further research and standardization. Many believe in oncological thermo-therapy and many regard it as quackery. There is a definite group of physicians who submit that hyperthermia has a strong curative force in oncology; however, another group exists believing the opposite. Sure, both the positive and the negative believers are not helpful to clarify the situation. Fortunately, science is not the question of belief; it is a question of proofs and results, which has to be carefully analyzed. We need interdisciplinary scientific approaches and hypotheses to go ahead with the topic.

There are intensive discussions in scientific communities on the mechanism of oncological hyperthermia[2] and so it′s not a surprise, that nowadays most oncological conferences deal with hyperthermia. There is an increasing number of the relevant published books and periodicals as well as a large number of scientific articles published in high ranked, good impact factor journals.[3] The increasing number of applications and clinical trials at universities, clinics, hospitals and institutes prove the feasibility and applicability of clinical hyperthermia in cancer therapies.

State-of-art

Some widely accepted effects characterize the classical hyperthermia:

  • Higher baseline temperature.[4]
  • Vascular changes,[5],[6],[7] increased heat conduction,[8] selective increase in tumor temperature[9] providing an effective heat trap.[10]
  • Cellular membrane changes,[11],[12],[13] it can change lipid-protein interactions[14] and it can denature proteins.[15]
  • Changes the active membrane transport,[16] the membrane capacity,[17] the membrane potential,[18] the cellular function[19],[20] and causes thermal block of electrically excitable cells.[18],[21]
  • Increases the biochemical reaction rates[22] resulting in hypoxia[23] and anaerobe metabolism producing lactate.[24]
  • Causes ATP depletion.[24]
  • DNA replication is slowing down.[25],[26]
  • Enhances the immune reactions,[25] with increase of natural killer cell activity[27] and distributes tumor-specific antigens on the surface of various tumor cells[28] and assists in their secretion into the extracellular electrolyte.[29]
  • Hyperthermia and especially its electric field induced realization, has significant pain-reduction during treatments.[30]
  • It has synergy with ionizing-radiation[31] and by chemotherapies.[32],[33]
  • Could make previously dangerous operations possible.[34] Postoperative application prevents relapses and metastatic processes.[35] Intraoperative radiofrequency ablation has also been used to improve surgical outcomes.[36]
  • Combination of the hyperthermia with the gene-therapy looks very promising, therapy in advanced breast cancer patients[37] and enhances the local rate of release from liposomes.[38]

Although hyperthermia can have significant benefits, there are several well-known problems to be solved.

Standardization
Hyperthermia dosing and treatment standardization is still a significant problem. Everybody agrees that hyperthermia is an overheating of the targeted tissue, but the definitions strictly differ on the heat-dose and the temperature issue.

Hot spots
Inadequate focusing can dangerously overheat the healthy tissue, causing unwanted burn and necrosis.

Heat Shock Protein (HSP) production
Heat can induce HSP production. HSP-assisted adaptation mechanisms decrease the efficacy of hyperthermia and can aid in the development of resistance to heat, chemo and radiation therapies.

Many believe that the single most important factor in hyperthermia is tumor temperature. On the other hand, there are no doubts about the strong heat-dose (energy absorption) dependence, which is shown by the treatment-time relevance in laboratory and clinical results.[39] However, the application of lower temperatures for longer time periods (same dose) treatments also showed surprisingly good efficacy for whole-body hyperthermia treatments.[40] This finding supports the opinion that the delivered heat dose (absorbed energy) or applied field[41] (electromagnetic influence) are the primary determinants of efficacy. More recently, numerous scientific theories concentrate on the vital significance of the thermally induced but basically non-thermal effects.[42] They back up their view by the thermally and non-thermally generated chaperone proteins, which are most of the case heat-shock proteins (HSP).[43]

New paradigm: Electro-hyperthermia

Recently, scientists have begun to realize that hyperthermia induced temperature gradients could have significant biological effects. A new branch of hyperthermia, known as extracellular hyperthermia[44] (or electro-hyperthermia, oncothermia) has been developed around this concept. Although this new technique recognizes the benefits of increased tissue temperature and its biological consequences it also argues that non-equilibrium thermal effects are partially responsible for the observed clinical deviations from the purely temperature-based treatment theory.

Oncothermia is devoted to enhance the efficiency of conventional hyperthermia by additional, non-equilibrium thermal effects with the aim of suppressing the existing disadvantages of the classical thermal treatments. The electric field energy matching (capacitive coupling) has smaller penetration depth relative to the magnetic field, however, the absorbed energy is significantly increased. On the other hand, the penetration depth of the radiative (antenna-array coupled) applications is only one third of that of the capacitive coupling. Moreover the electric field offers important selectivity factors to use. The energy absorption at the applied frequency is proportional to the tissue conductivity and the square root of the dielectric constant of the targeted material. Due to its intensive metabolic activity, the conductivity in malignant tissue is higher than that of normal tissue;[45],[46] as well as the dielectric constant of the extracellular matrix at the applied frequencies is also higher in the malignant tissue than in the healthy one.[45],[47] It has also been observed that the dielectric constants in the malignant tissue are far from homogeneous[48],[49],[50] and this is supported by theoretical considerations.[51] In consequence good selectivity could be achieved at relatively low frequencies. Further focusing effect can be derived from the coherent electric waves,[52],[53],[54] with spontaneous breakdown of the polarization symmetries. Therefore the electric coupling could select between the healthy and tumor-tissues.

The energy absorption for these effects is more significant than the temperature; so we have to characterize the hyperthermia by thermal dose and not by temperature. Thermal dose changes many energetic processes in the tissue and in their physiology. Most of the desired changes (structural and chemical) need energy consumption, which will be missing to rise the temperature. The heat, causing only the temperature rise, is not involved in the actual distortion, that is "lost" to make the job. The non-equilibrium thermodynamics describes how the absorbed heat could excite various (e.g. diffusional, electric, chemical, etc.) processes; which drives the distortion efficacy as well. These phenomena are completely out of the possibility of temperature characterization.

Electro-hyperthermia is based on a capacitively-coupled energy transfer applied at a frequency that is primarily absorbed in the extracellular matrix due to its inability to penetrate the cell membrane.[55] Although these temperature gradients typically relax within a few milliseconds, a constant energy delivery can maintain this gradient for extended periods of time. An externally applied electric field can maintain temperature gradients of 1 K/μm, creating a permanent heat flow of 1500 nW/μm 2, which is well above the natural heat-flow (20 nW/μm 2) across the target cell membranes. This gradient and the resulting heat flow can produce 150 pA/μm 2 currents through the membrane primarily by Na+ influx into the cell, which significantly exceed the typical 12 pA/μm 2 sodium efflux present. This depolarizes and therefore destabilizes the membrane and stimulates Na+ /K+ pump activity. This requires ATP resulting in further heat production at the membrane. The membrane permeability of water is much higher than for ions, therefore it is the main transported component in thermo-dynamic coupling. A thermal flux of 0.001 K/nm can therefore build up pressure reaching 1.32 MPa. Since malignant cells typically have relatively rigid membranes due to increased phospholipids concentrations,[56] an increase in pressure will selectively destroy malignant cells before it affects healthy ones.

A relevant characterization of oncological hyperthermia for quality guidelines has to be started to define the aims: to destroy the malignant cells. This demand contains some more precise requests: act selectively on the malignant cells, block the further proliferation and stop the dissemination of tumor-cells, etc. The demands actually do not contain any temperature request; the temperature could be a tool only for this job. If a bio-system undergoes chemical reactions, the non-temperature terms of the internal energy become important.[57] In despite of the same temperature was reached by conventional and microwave heating, the in-vivo reaction was significantly different.[58]

In despite of its inadequate character, the temperature has gradually become the base of hyperthermia quality assurance and treatment control. The physiologically and physically well studied extracellular ionic environment is used to control the treatment, serves for comparison and gives information for the physician about the treatment success in-situ. The ion-concentration in extracellular electrolyte (ECM) definitely depends on the metabolic rate, on the chemical reactions and on the structural changes. To control the energy induced distortion processes the ion-density and the actual structural changes could be well followed by the simple technique of complex bio-impedance;[59],[60] uses special frequency dispersion of the actual tissue. As early as 1940 both the whole-body electrolyte status[61] and the local changes (ECG)[62] were studied by the method. Nowadays, it is commercially applied, (T-Scan TS2000) and for breast tumor diagnostics received the FDA approval in 1999. Various important parameters had been measured by this method (histological,[63] coagulative necrosis,[64] apoptosis,[65] ischemia.[66],[67] In addition, the temperature of the tissue[68] and the Arrhenius activation energy[69] could be monitored by impedance. It adequately measures the distortion made by irradiation,[70] as well as the drug-effect can also be controlled,[71] moreover, the wound healing is also objectively traceable.[72] It is widely applied for RF ablation/interstitial techniques, without any extra control of the temperature.[73],[74]

Results

OncoThermia results were mainly measured by the survival analysis (Kaplan-Meier distribution) and considered the quality of life by objective and subjective parameters. The results are amazingly good. Some examples are collected below, which are rarely treated by hyperthermia.

Brain
The brain treatment is generally out of scopes of hyperthermia with conventional methods. However, oncothermia is able to treat brain with excellent results.[75] Oncothermia is applied for the advanced brain tumors (anaplastic astrocytoma and glioblastoma multiforme)[76],[77],[78] and the survival analysis shows a great success,.(overall survival median/mean: 27.3/40.6 (n=29) and 14.1/17.4 (n=33) months for astrocytoma and glioblastoma respectively).

Liver
The liver hyperthermia is a complicated issue because of the large blood perfusion and sensitivity due to the chemo-toxicity from previous treatments. Oncothermia results are also exceptionally good for that organ.[79] Overall median survival for patients with liver metastases from colorectal primary (n=80) is also remarkable (median is 24.1 months) by oncothermia treatment.[80]

Pancreas
The pancreas carcinoma is a rapid and aggressive disease, also not much conventional hyperthermia results could find in this location. However, oncothermia has good results in survival.[81],[82],[83] The advanced pancreas carcinoma study[84] (N=129) shows also very good response for the oncothermia treatment (median/mean 8/12.5 (n=85) and 6.5/8.6 (n=34) months for active and control groups respectively).

Lung
The lung is also a complicated issue for hyperthermia because of the permanent cooling-ventilation of the breathing. Our method, the electro-hyperthermia due to the non-equilibrium approach is an excellent treatment for that as well.[85],[86] For example, oncothermia successfully applied for advanced non-small-cell lung cancer[87] (median of overall survivals (n=200) are 36.3, 20.3 and 11.4 months for not advanced, advanced (operable) and advanced (not operable) cases, respectively).

Bone
The bone is the other problematic issue for hyperthermia because of the low density of the bone compared to the adjoining tissues. Excellent bone results could be achieved by oncothermia as a part of a complex therapy.[88]

Conclusion

Hyperthermia is an emerging effective treatment method in oncology. It has became a new modality of cancer treatments, showing significant improvements in tumor response rates and patient morbidity in combination with other treatment methods, such as surgery, chemotherapy, radiation therapy and gene-therapy or applied as a single therapy. Nevertheless, hyperthermia is still in its infancy. It lacks standards and a scientific consensus about its effects on malignant and healthy tissues and the current techniques used to treat patients vary significantly from antenna-array focused electromagnetic energy delivery methods to non-thermal low-power current applications. In order to gain wide-spread approval and clinical use for hyperthermia, the technique requires further extensive research and standardization. Hyperthermia′s update technique, the oncothermia is highly selective and safe, providing all the positive effects of the conventional hyperthermia with additional new advantages. Its working principle is mainly based on the extracellular and highly focused actions, extending the thermal treatment efficiency by non-thermal effects and by non-equilibrium selection and distortion of cellular membranes in tumors. We are convinced that the perspectives of hyperthermia in oncology are very bright and promising. What we have in hand is a practically non toxic effect with huge potential and advantages.

References

1.Muller C. Therapeutische Erfahrungen an 100 mit kombination von Rontgenstrahlen un Hochfrequenz, resp. Diathermie behandeleten bosartigen Neubildungen. Munchener Medizinische Wochenschrift 1912;28:1546-9.  Back to cited text no. 1    
2.Szasz A, Szasz O, Szasz N. Physical background and technical realizations of hyperthermia, edited by Baronzio GF, Hager ED: Locoregional Radiofrequency-Perfusional and Whole-body Hyperthermia in Cancer Treatment: New Clinical Aspects, Eurokah.com and Springer Science Business Media: 2005.  Back to cited text no. 2    
3.Nielsen OS, Horsman M, Overgaard J:A future of hyperthermia in cancer treatment? (Editorial Comment). Eur J Cancer 2001;37:1587-9.  Back to cited text no. 3    
4.Head JF, Wang Fen, Lipari CA, Elliot RL. The important role of Infrared Imaging in Breast cancer. IEEE Engg Med Biol 2000;19:52-7.  Back to cited text no. 4    
5.Vaupel P, Kallinowski FP. Blood flow, oxygen and nutrient supply, and microenvironment of human tumors: A review. Cancer Res 1989;49:6449-65.  Back to cited text no. 5    
6.Dudar TE, Jain RK. Differential response of normal and tumor microcirculation to hyperthermia. Cancer Res 1984;44:605-12.  Back to cited text no. 6  [PUBMED]  
7.Song CW, Lokshina A, Rhee JG, Patten M, Lewitt SH. Implication of blood-flow in hyperthermic treatment of tumors, IEEE Trans. Biomed. Eng. BME 1984;31:9-16.  Back to cited text no. 7    
8.Song CW, Choi IB, Nah BS, Sahu SK, Osborn JL. Microvasculature and Persfusion in Normal Tissues and Tumors, Thermoradiometry and Thermochemotherapy. (Eds. Seegenschmiedt MH, Fessenden P, Vernon CC). 1995;1:139-56.  Back to cited text no. 8    
9.Vaupel P. Phatophysiological Mechanisms of Hyperthermia in Cancer Therapy, M. Gautherie (ed.), Biological Basis of Oncologic thermotherapy. Springer Verlag: Berlin Heidelberg; 1990. p. 73-134.  Back to cited text no. 9    
10.Takana Y. Thermal Responses of Microcirculation and Modification of Tumor Blood Flow in Treating the Tumors, Thermotherapy for Neoplasia, Inflammation, and Pain (Eds. M. Kosaka, T. Sugahara, K.L. Schmidt, E. Simon). Springer Verlag: Tokyo; 2001. p. 408-19.  Back to cited text no. 10    
11.Heilbrunn LV. The colloid chemistry of protoplasm. Am J Physiol 1924;69:190-9.  Back to cited text no. 11    
12.Yatvin MB, Dennis WH. Membrane lipid composition and sensitivity to killing by hyperthermia, Procaine and Radiation, In : Cancer Therapy by Hyperthermia and Radiation, Editors: Streffer C, vanBeuningen D, Dietzel F, Rottingen E, Robinson JE, Scherer E, Seeber S, Trott KR. Urban & Schwarzenberg: Baltimore, Munich; 1978. p. 157-9.  Back to cited text no. 12    
13.Streffer C. Biological Basis of Thermotherapy (with special reference to Oncology), In:Biological Basis of Oncologic Thermotherapy, Ed: Gautherie M, Springer Verlag: Berlin; 1990. p. 1-72.  Back to cited text no. 13    
14.Bowler K, Duncan CJ, Gladwell RT, Davison TF. Cellular heat injury. Comp Biochem Physiol 1973;45A:441-50.  Back to cited text no. 14    
15.Belehradek J. Physiological aspects of heat and cold. Am Rev Physiol 1957;19:59-82.  Back to cited text no. 15  [PUBMED]  
16.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. Int J Hyperther 1997;13:227-34.  Back to cited text no. 16  [PUBMED]  
17.Wallach DF. Action of Hyperthermia and Ionizing radiation on plasma membranes, In:Cancer Therapy by Hyperthermia and Radiation, Editors: Streffer C, vanBeuningen D, Dietzel F, Rottingen E, Robinson JE, Scherer E, Seeber S, Trott KR. Urban & Schwarzenberg: Baltimore, Munich; 1978. p. 19-28.  Back to cited text no. 17    
18.Weiss TF. Cellular Biophysics, Electrical properties, MIT Press: Cambridge; 1996.  Back to cited text no. 18    
19.Mikkelsen RB, Verma SP, Wallach DF. Hyperthermia and the membrane potential of erythrocyte membranes as studied by Raman Spectroscopy, In : Cancer Therapy by Hyperthermia and Radiation, Eds: Streffer C, vanBeuningen D, Dietzel F, Rottingen E, Robinson JE, Scherer E, Seeber S, Trott KR. Urban & Schwarzenberg: Baltimore, Munich; 1978. p. 160-2.  Back to cited text no. 19    
20.Hahn GM. The heat-shock response:Effects before, during and after Gene activation, In:Biological Basis of Oncologic Thermotherapy, Ed: Gautherie M. Springer Verlag: Berlin; 1990. p. 135-59.  Back to cited text no. 20    
21.Hodgkin AL, Katz B. The effect of temperature on the electrical activity of the giant axon of the squid. J Physiol 1949;108:37-77.  Back to cited text no. 21    
22.Weiss TF. Cellular Biophysics. Transport, MIT Press: Cambridge; 1996.  Back to cited text no. 22    
23.Dewhirst MW, Ozimek EJ, Gross J, Cetas TC. Will hyperthermia conquer the elusive hypoxic cell? Radiology 1980;137:811-7.  Back to cited text no. 23  [PUBMED]  
24.Vaupel PW, Kelleher DK. Metabolic status and reaction to heat of Normal and tumor tissue, Seegenschmiedt MH, Fessenden P, Vernon CC (Eds.) Thermo-radiotherapy and Thermo-chemiotherapy, Biology, physiology and physics. Springer Verlag: Berlin Heidelberg; 1996. p. 157-76.  Back to cited text no. 24    
25.Keszler G, Csapo Z, Spasokoutskaja T, Sasvary-Szekely M, Virga S, Demeter A, et al . Hyperthermy increase the phosporylation of deoxycytidine in the membrane phospholipid precursors and decrease its incorporation into DNA. Adv Exper Med Biol 2000;486:33-7.  Back to cited text no. 25    
26.Dikomey E, Franzke J. Effect of heat on induction and repair of DNA strand breaks in X-irradiated CHO cells. Int J Radiat Biol 1992;61:221-34.  Back to cited text no. 26    
27.Shen RN, Lu L, Young P, Shidnia H, Hornback NB, Broxmeyer HE. Influence of elevated temperature on natural killer cell activity, lymphokine-activated killer cell activity and lecitin-dependent cytotoxicity of human umbilical cord blood and adult blood cells. Int J Radiat Oncol Biol Phys 1994;29:821-6.  Back to cited text no. 27  [PUBMED]  
28.Srivastava PK, DeLeo AB, Old LJ. Tumor Rejection Antigens of Chemically Induced Tumors of Inbred Mice. Proc Natl Acad Sci USA 1986;83:3404-11.  Back to cited text no. 28  [PUBMED]  [FULLTEXT]
29.Csermely P, Schnaider T, Soti C, Prohaszka Z, Nardai G. The 90 kDa Molecular Chaperone Family: Structure, Function and Clinical Applications, A Comprehensive Review. Pharmacol Therap 1998;79:129-68.  Back to cited text no. 29  [PUBMED]  [FULLTEXT]
30.Gonzalez-Gonzalez D. Thermo-radiotherapy for tumors of the lower gastro-instenstinal tract, M. H. Seegenschmiedt, P. Fessenden, C. C.Vernon (Eds.) Thermo-radiotherapy and Thermo-chemiotherapy, Biology, physiology and physics. Springer Verlag: Berlin Heidelberg; 1996. p. 105-19.  Back to cited text no. 30    
31.Urano M, Douple E. Hyperthermia and Oncology Vol. 2, Biology of thermal potentiation of radiotherapy. VSP Utrecht: The Netherlands; 1989.  Back to cited text no. 31    
32.Urano M, Douple E. Hyperthermia and Oncology Vol.4, Chemopotentiation by Hyperthermia. VSP Utrecht: The Netherlands; 1994.  Back to cited text no. 32    
33.Kawasaki S, Jun-Ichi A, Shibuya K, Kuroda M, Hiraki Y. Recent Aspects of Elucidating the Cellular Basis of Thermochemotherapy, Thermotherapy for Neoplasia, Inflammation, and Pain (Eds. M. Kosaka, T. Sugahara, K.L. Schmidt, E. Simon). Springer Verlag: Tokyo; 2001. p. 424-32.  Back to cited text no. 33    
34.Masunaga SI, Hiraoka M, Akuta K, Nishimura Y, Nagata Y, Jo S, et al . Non-randomized trials of thermoradiotherapy versus radiotherapy for preoperative treatment of invasive urinary bladder cancer. J Jap Soc Ther Radiol Oncol 1990;2:313-20.  Back to cited text no. 34    
35.Kodama K, Doi O, Higashiyama M, Yokouchi H, Tatsuda M. Long -term results of postoperative Intrathoracic Chemo-thermotherapy for lung cancer with pleural dissemination. Cancer 1993;72:100-6.  Back to cited text no. 35    
36.Scott A, Izzo F, Fleming RY, Ellis LM, Delrio P, Roh MS, et al . Intraoperative radiofrequency ablation of cryoablation for hepatic malignances. Am J Surg 1999;178:592-9.  Back to cited text no. 36    
37.Ohtsuru A, Braiden V, Cao Y, Kosaka M, Yamashita S. Cancer Gene Therapy in Conjunction with Hyperthermia Under the Control of Heat-Inducible Promoter, Thermotherapy for Neoplasia, Inflammation, and Pain (Eds. M. Kosaka, T. Sugahara, K.L. Schmidt, E. Simon). Springer Verlag: Tokyo; 2001. p. 464-70.  Back to cited text no. 37    
38.Gaber MH, Wu NZ, Hong K. Thermosensitive liposomes: Extravasation and relase of contents in tumor microvascular networks. Int J Radiat Oncol Biol Phys 1996;36:1177-87.  Back to cited text no. 38    
39.Field SB. Biological Aspects of Hyperthermia, Physics and Technology of Hyperthermia, Field SB, Franconi C, (Eds.) NATO ASI Series, E: Applied Sciences, No.127. Martinus Nijhoff Publ: Dordrecht/Boston; 1987. p. 19-53.  Back to cited text no. 39    
40.Kraybill W, Olenki T. A phase I study of fever-range whole body hyperthermia (FR-WBH) in patients with advanced solid tumors: Correlation with mouse models, Int. J. Hyperthermia, 2002, Vol. 18, No. 3, 253-266 and Burd R, Dziedzic ST: Tumor Cell Apoptosis, Lymphocyte Recruitment and Tumor Vascular Changes Are Induced by Low Temperature, Long Duration (Fever-Like) Whole Body Hyperthermia. J Cell Physiol 1998;177:137-47.  Back to cited text no. 40    
41.Szasz A, Szasz O, Szasz N. Electrohyperthermia: A new paradigm in cancer thermapy, Wissenschaft & Forschung. Deutsche Zeitschrift Für Onkologie 2001;33:91-9.  Back to cited text no. 41    
42.de Pomarai D, Daniels C, David H, Allan J, Duce I, Mutwakil M, et al . Non-thermal heat-shock response to microwaves. Nature 2000;405:417-8.  Back to cited text no. 42    
43.Bukau B, Horwich AL. The HSP70 and HSP60 chaperone machines. Cell 1998;92:351-66.  Back to cited text no. 43  [PUBMED]  [FULLTEXT]
44.Szasz A, Vincze GY, Szasz O, Szasz N. An energy analysis of extracellular hyperthermia, accepted for publication in Magneto- and electro-biology. 2003.  Back to cited text no. 44    
45.Smith SR, Foster KR, Wolf GL. Dielectric Properties of VX-2 Carcinoma Versus Normal Liver Tissue. IEEE Trans Biomed Eng 1986;33:522-4.  Back to cited text no. 45  [PUBMED]  
46.Dissado LA, Alison JM, Hill RM, McRae DA, Esrick MA. Dynamic Scaling in the Dielectric Response of Excised EMT-6 Tumours Undergoing Hyperthermia. Phys Med Biol 1995;40:1067-84.  Back to cited text no. 46  [PUBMED]  [FULLTEXT]
47.Szent-Gyorgyi A. Electronic Biology and Cancer. Marcel Dekker: New York - Basel; 1976.  Back to cited text no. 47    
48.Cope FW. A review of the application of solid state physics concepts in biologcal systems. J Biol Phys 1974;3:1-41.  Back to cited text no. 48    
49.Damadian R. Tumor Detection by Nuclear Magnetic Resonance. Science 1971;171:1151-3.  Back to cited text no. 49  [PUBMED]  
50.Hazlewood CF, Nichols BL, Chamberlain NF.. Evidence for the existence of a minimum of two phases of ordered water in skeletal muscle. Nature 1969;222:747-50.  Back to cited text no. 50  [PUBMED]  
51.Szent-Gyorgyi A. The Living State and Cancer. Marcel Dekker: New York-Basel; 1976.  Back to cited text no. 51    
52.DelGiudice E, Doglia S, Milani M, Vittiello G. Electromagnetic Field and Spontaneous Symmetry Breaking in Biological Matter. Nucl Physics 1986;B275:185-99.  Back to cited text no. 52    
53.DelGiudice E, Doglia S, Milani M. Self-focusing and Ponderomotive Forces of Coherent Electric Waves: A Mechanism for Cytoskeleton Formation and Dynamics, In : Coherent Excitations in Biological Systems, Eds. Frohlich H, Kramer F. Springer Verlag: Berlin-Heidelberg; 1983. p. 123-7.  Back to cited text no. 53    
54.DelGiudice E, Doglia S, Milani M. Self-Focusing of Frohlich Waves and Cytoskeleton Dynamics. Physics Lett 1982;90A:104-6.  Back to cited text no. 54    
55.Kotnik T, Miklavcic D. Theoretical evaluation of the distributed power dissipation in biological cells exposed to electric field. Bioelectromagnetics 2000;21:385-94.  Back to cited text no. 55  [PUBMED]  [FULLTEXT]
56.Galeotti T, Borrello S, Minotti G, Masotti L. Membrane Alterations in Cancer Cells: the role of Oxy Radicals, An. New York Acad. Sci. Vol. 488, Membrane Pathology, Bianchi G, Carafoli E, Scarpa A, editors. 1986. p. 468-80.  Back to cited text no. 56    
57.Katchalsky A, Curran PF. Nonequilibrium Thermodynamics in Biophysics. Harvard University Press: Cambridge, MA, USA; 1967.  Back to cited text no. 57    
58.de Pomarai DC, Daniels H, David J, Allan I, Duce M, Mutwakil D, et al . Non-thermal heat-shock response to microwaves. Nature 2000;405:417-8.  Back to cited text no. 58    
59.Scholz B, Anderson R. On Electrical Impedance Scanning - Principles and Simulations. Electromedica 2000;68:35-44.  Back to cited text no. 59    
60.Sha L, Wand ER, Story B. A rewiev of dielectric properties of normal and malignant breast, Proceedings. IEEE South East Con 2002. p . 457-62.  Back to cited text no. 60    
61.Barnett A. Electrical method for studying water metabolism and transloction in body segments. Proc Soc Exp Biol Med 1940;44:142-7.  Back to cited text no. 61    
62.Nyboer JS, Bango A, Barnett, Halsey RH. Radiocardiograms - the electrical impedance changes of the heart in relation to electrocardiograms and heart sounds. J Clin Invest 1940;19:963-6.  Back to cited text no. 62    
63.McRae DA, Esrick MA. The dielectric parameters of excised EMT-6 tumours and their change during hyperthermia. Phys Med Biol 1992;37:2045-58.  Back to cited text no. 63  [PUBMED]  [FULLTEXT]
64.McRae DA, Esrick MA, Mueller SC. Changes in the non-invasive, in vivo electrical impedance of the xenograpfts during the necrotic cell-response sequence. Int J Radiat Oncol Biol Phys 1999;43:849-57.  Back to cited text no. 64  [PUBMED]  [FULLTEXT]
65.Shchepotin IB, McRae DA, Shabahang M, Buras RR, Evans SR. Hyperthermia and verapamil inhibit the growth of human colon cancer xenografts in vivo through apoptosis, Anticancer Res 1997;17:2213-6.  Back to cited text no. 65  [PUBMED]  
66.Casas O, Bragos R, Riu PJ, Rossel J, Tresanchez M, Warren M, et al . In vivo and in situ ischemic tissue characterization using electrical impedance spectroscopy, In:Electrical Bioimpedance Methods:Applications to Medicine and Biotechnology, Riu P, Rosell J, Bragos R, Casas O. (Eds.). Ann New York Acad Sci 1999;873:51-8.  Back to cited text no. 66    
67.Gheorghiu M, Gersing E, Gheorghiu E. Quantitative analysis of impedance spectra of organs during ischemia, In: Electrical Bioimpedance Methods: Applications to Medicine and Biotechnology, Riu P, Rosell J, Bragos R, Casas O. (Eds.). Ann New York Acad Sci 1999;873:65-71.  Back to cited text no. 67  [PUBMED]  [FULLTEXT]
68.Gersing E. Monitoring temperature induced changes in tissue during hyperthermia by impedance methods, In:Electrical Bioimpedance Methods:Applications to Medicine and Biotechnology, Riu P, Rosell J, Bragos R, Casas O. (Eds.). Ann New York Acad Sci 1999;873:13-20.  Back to cited text no. 68    
69.McRae DA, Esrick MA. Changes in electrical impedance of skeletal muscle measured during hyperthermia. Int J Hyperthermia 1993;9:247-61.  Back to cited text no. 69  [PUBMED]  
70.Osterman KS, Paulsen KD, Hoopes PJ. Application of linear circuit models to impedance spectra in irradiated muscle, In:Electrical Bioimpedance Methods: Applications to Medicine and Biotechnology, Riu P, Rosell J, Bragos R, Casas O. (Eds.). Ann New York Acad Sci 1999;873:21-9.  Back to cited text no. 70  [PUBMED]  [FULLTEXT]
71.Santini MT, Cametti C, Zimatore G, Malorni W, Benassi M, Gentile FP, et al . A dielectric relaxation study on the effects of the antitumor drugs Lomidamineand Rhein on the membrane electrical properties of Erlich ascites tumour cells. Anticancer Res 1995;15:29-36.  Back to cited text no. 71    
72.Keese CR, Wegener J, Walker SR, Giaever I. Electrical wound-healing assay for cells in vitro, PNAS. Proc Nat Acad Sci USA 2004;101:1554-9.  Back to cited text no. 72  [PUBMED]  [FULLTEXT]
73.Avitall B, Mughal K, Hare J, Helms R, Krum D. The effects of electrode-tissue contact on radiofrequency lesion generation. Pacing Clin Electrophysiol 1997;20:2899-910.  Back to cited text no. 73  [PUBMED]  
74.Schmidt D, Trubenbach J, Konig CW, Brieger J, Duda S, Claussen O, et al . Radiofrequency ablation ex vivo: comparison of the efficacy impedance control mode versus manual control mode by using internally cooled clustered electrode. Rofo Fortschr Geb Rontgenstr Neuen Bildgeb Verfahr 2003;175:967-72.  Back to cited text no. 74    
75.Hager ED, Dziambor H, App EM, Popa C, Popa O, Hertlein M. The treatment of patients with high-grade malignant gliomas with RF-hyperthermia. Proc ASCO 2003;22:118.  Back to cited text no. 75    
76.Szasz A, Szasz O, Szasz N. Electro-hyperthermia:principles and practice. 14th ICACT Conference: Paris; 2003.  Back to cited text no. 76    
77.Sahinbas H. Deep RF hyperthermia treatment of advanced gliomas. Oncology Conference: Basel; 2004.  Back to cited text no. 77    
78.Sahinbas H, Deep RF. Hyperthermia: an effective treatment of advanced gliomas. ESHO Conference: Graz; 2005. p. 8-11.  Back to cited text no. 78    
79.Hager ED, Dziambor H, Hohmann D, Gallenbeck D, Stephan M, Popa C, et al . Deep hyperthermia with radiofrequencies in patients with liver metastases from colorectal cancer. Anticancer Res 1999;19:3403-8.  Back to cited text no. 79    
80.Varkonyi A. Klinikai tapasztalatok elυrehaladott tüdυ daganatok hipertermiaαs kezelιsιben. Szent Istvan Egyetemi Napok: Godollo; 2003. p. 26-7.  Back to cited text no. 80    
81.Hager ED, Süβe B, Popa C, Schrittwieser G, Heise A, Kleef R. Complex therapy of the not in sano respectable carcinoma of the pancreas - a pilot study. J Can Res Clin Oncol 1994;120:R47.  Back to cited text no. 81    
82.Hager ED, Dziambor H, Hoehmann D. Survival and quality of life patients with advanced pancreatic cancer. Proc ASCO 2002;21:136b.  Back to cited text no. 82    
83.Dani A. Clinical experience of electro-hyperthermia for advanced lung tumors. ESHO Conference: Munich; 2003.  Back to cited text no. 83    
84.Dani A, Varkonyi A, Nyiro I, Osvath M. Clinical experience of electro-hyperthermia for advanced pancreatic tumors. ESHO Conference: Munich; 2003.  Back to cited text no. 84    
85.Hager ED, Krautgartner I, Popa C, H φhmann D, Dziambor H. Deep Hyperthermia with short waves of patients with advanced stage lung cancer. Hyperthermia in clinical practice. XXII Meeting of the International Clinical Hyperthermia Society: 1999.  Back to cited text no. 85    
86.Dani A. Clinical experience of electro-hyperthermia for advanced NSC lung cancer. Sypmosium Hyperthermie: K φln; 2003.  Back to cited text no. 86    
87.Dani A. Electro-hyperthermia for advanced pancreas tumors. Degro: Erfurt; 2004. p. 10-3.  Back to cited text no. 87    
88.Bogovic J, Douwes F, Muravjov G, Istomin J. Posttreatment histology and microcirculation status of osteogenic sarcoma after a neoadjuvant chemo and radiotherapy in combination with local electromagnetic hyperthermia. Onkologie 2001;24:55-8.  Back to cited text no. 88    

Copyright 2006 - Journal of Cancer Research and Therapeutics

Home Faq Resources Email Bioline
© Bioline International, 1989 - 2024, Site last up-dated on 01-Sep-2022.
Site created and maintained by the Reference Center on Environmental Information, CRIA, Brazil
System hosted by the Google Cloud Platform, GCP, Brazil