This research program was initiated following observations that a whole body exposure to a mild, fever-like hyperthermia (39.5 degree C for 6-8 hours, i.e., """"""""physiological hyperthermia""""""""), resulted in alternations in lymphocyte structure, inhibition of tumor growth and increased tumor cell apoptosis in murine models. While this response is not in itself, curative, the fact that such a mild treatment would have any anti-tumor activity at all was somewhat unexpected. The major hypothesis being tested to explain these observations is that this fever-like treatment enhances the immune response or facilitates the immunological rejection of tumors. From this hypothesis, the investigators predict that the moderate hyperthermia protocol will synergize with other therapies (vaccines, adoptive T cell therapy or cytokine treatments) that stimulate the anti-tumor immune response. Their preliminary and published data strongly support these assumptions. The major goal of this renewal application continues to be a determination of the molecular and immunological mechanism(s) underlying the anti-tumor effects of physiological hyperthermia so that its usefulness can be fully exploited in the clinic. Since the immune system is composed of multiple systems and cells upon which physiological hyperthermia could exert its effects, the investigators have continued to evaluate molecular endpoints in Aims 1 and 2 that would be important for all aspects of immune response: gene and protein expression, and lymphocyte membrane organization. Moreover, the investigators have chosen to evaluate functional endpoints that represent a convergence of many important components and levels of the immune system, i.e., immune response to several specific tumor antigens (Aim 3). Finally, they have chosen to evaluate whether whole body hyperthermia would be synergistic with other immunotherapies, such as vaccines, T cell or cytokine administration (Aim 4). All of these aims are logically derived from the progress made by the investigators in the first period of funding. Moreover, compelling preliminary data has been obtained strongly supporting the use of fever-like hyperthermia in combination with vaccines or cytokines for the treatment of cancer. With a better understanding of the molecular and cellular mechanisms underlying the anti-tumor effects of physiological heating, the investigator hopes to more rationally define how this approach can be utilized clinically. Since a Phase I trial using fever-like hyperthermia alone (to demonstrate its safety before combining it with other treatments), is just being completed the investigators will be poised to apply the information obtained from the studies described in the present proposal to the rapid development of new Phase I/II trials at this Institute.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
2R01CA071599-04
Application #
6288572
Study Section
Radiation Study Section (RAD)
Program Officer
Stone, Helen B
Project Start
1997-12-24
Project End
2003-11-30
Budget Start
2001-01-09
Budget End
2001-11-30
Support Year
4
Fiscal Year
2001
Total Cost
$265,059
Indirect Cost
Name
Roswell Park Cancer Institute Corp
Department
Type
DUNS #
City
Buffalo
State
NY
Country
United States
Zip Code
14263
Zynda, Evan R; Grimm, Melissa J; Yuan, Min et al. (2015) A role for the thermal environment in defining co-stimulation requirements for CD4(+) T cell activation. Cell Cycle 14:2340-54
Winslow, Timothy B; Eranki, Annu; Ullas, Soumya et al. (2015) A pilot study of the effects of mild systemic heating on human head and neck tumour xenografts: Analysis of tumour perfusion, interstitial fluid pressure, hypoxia and efficacy of radiation therapy. Int J Hyperthermia 31:693-701
Dayanc, Baris E; Bansal, Sanjay; Gure, Ali Osmay et al. (2013) Enhanced sensitivity of colon tumour cells to natural killer cell cytotoxicity after mild thermal stress is regulated through HSF1-mediated expression of MICA. Int J Hyperthermia 29:480-90
Lee, Chen-Ting; Zhong, Lingwen; Mace, Thomas A et al. (2012) Elevation in body temperature to fever range enhances and prolongs subsequent responsiveness of macrophages to endotoxin challenge. PLoS One 7:e30077
Mace, Thomas A; Zhong, Lingwen; Kokolus, Kathleen M et al. (2012) Effector CD8+ T cell IFN-? production and cytotoxicity are enhanced by mild hyperthermia. Int J Hyperthermia 28:9-18
Lee, Chen-Ting; Repasky, Elizabeth A (2012) Opposing roles for heat and heat shock proteins in macrophage functions during inflammation: a function of cell activation state? Front Immunol 3:140
Sen, Arindam; Capitano, Maegan L; Spernyak, Joseph A et al. (2011) Mild elevation of body temperature reduces tumor interstitial fluid pressure and hypoxia and enhances efficacy of radiotherapy in murine tumor models. Cancer Res 71:3872-80
Mace, Thomas A; Zhong, Lingwen; Kilpatrick, Casey et al. (2011) Differentiation of CD8+ T cells into effector cells is enhanced by physiological range hyperthermia. J Leukoc Biol 90:951-62
Beachy, Sarah H; Repasky, Elizabeth A (2011) Toward establishment of temperature thresholds for immunological impact of heat exposure in humans. Int J Hyperthermia 27:344-52
Fisher, Daniel T; Chen, Qing; Skitzki, Joseph J et al. (2011) IL-6 trans-signaling licenses mouse and human tumor microvascular gateways for trafficking of cytotoxic T cells. J Clin Invest 121:3846-59

Showing the most recent 10 out of 40 publications