Prostate cancer (PCa) is still a leading cause of cancer related-deaths in the US and the treatment of advanced disease is still limited. As prostate cancer can have a widely variable course of progression, identification of factors that preferentially associate with aggressive form of tumors is of the utmost importance to be able to improve treatment for advanced disease. Almost one third of human cancers including PCa are proved a causative link with chronic inflammation and infection. The risk of prostate cancer is strongly associated with persistent inflammation in the prostate. Over the past decade, heme oxygenase-1 (HO-1) and heme degradation product, carbon monoxide (CO) have evolved into accepted protective molecules in inflammatory, proliferative and oxidative-stress related disorders with the greatest support for these observations coming from the knockout mice and HO-1-deficient human. The importance of heme degradation pathway in tumor-associated macrophages to modulate cancer growth however has not been yet studied. Our major hypothesis is that HO-1 acting via CO constrains the development and progression of prostate cancer via modulation of macrophage activities: phagocytosis and cytokine production through Nalp3-caspase-1-IL1? signaling pathway. In this proposal, we shall test the role of HO-1 and heme degradation products as protective molecules in tumor-associated macrophages in prostate cancer.

Public Health Relevance

Inflammation, a seven hallmark of cancer, is a critical component of cancerogenesis and in many situations is present before a malignant transformation occurs. Heme oxygenase-1 (HO-1), a member of the family of immunoregulatory proteins, is the rate-limiting enzyme in the degradation of heme and we posit is critical in tumor-associated inflammation and control of tumorigenesis. We hypothesized that induction of HO-1 and application of carbon monoxide will lead to faster and more efficient reprogramming of innate immune towards anti-cancerous response. We will dissect the role of the heme pathway by using our newly generated HO-1(hmox-1-fl/fl) conditional knockout mice in the transgenic model of PCa and/or infection-associated PCa.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Exploratory/Developmental Grants (R21)
Project #
1R21CA169904-01A1
Application #
8511905
Study Section
Transplantation, Tolerance, and Tumor Immunology (TTT)
Program Officer
Howcroft, Thomas K
Project Start
2013-08-06
Project End
2015-07-31
Budget Start
2013-08-06
Budget End
2014-07-31
Support Year
1
Fiscal Year
2013
Total Cost
$227,070
Indirect Cost
$96,570
Name
Beth Israel Deaconess Medical Center
Department
Type
DUNS #
071723621
City
Boston
State
MA
Country
United States
Zip Code
02215
Wegiel, Barbara; Vuerich, Marta; Daneshmandi, Saeed et al. (2018) Metabolic Switch in the Tumor Microenvironment Determines Immune Responses to Anti-cancer Therapy. Front Oncol 8:284
Seth, Pankaj; Csizmadia, Eva; Hedblom, Andreas et al. (2017) Deletion of Lactate Dehydrogenase-A in Myeloid Cells Triggers Antitumor Immunity. Cancer Res 77:3632-3643
Wang, Yiqiang; Hedblom, Andreas; Koerner, Steffi K et al. (2016) Novel synthetic chalcones induce apoptosis in the A549 non-small cell lung cancer cells harboring a KRAS mutation. Bioorg Med Chem Lett 26:5703-5706
Nemeth, Zsuzsanna; Csizmadia, Eva; Vikstrom, Lisa et al. (2016) Alterations of tumor microenvironment by carbon monoxide impedes lung cancer growth. Oncotarget 7:23919-32
Nemeth, Zsuzsanna; Li, Mailin; Csizmadia, Eva et al. (2015) Heme oxygenase-1 in macrophages controls prostate cancer progression. Oncotarget 6:33675-88
Otterbein, Leo E; Fan, Zhigang; Koulmanda, Maria et al. (2015) Innate immunity for better or worse govern the allograft response. Curr Opin Organ Transplant 20:8-12
Wegiel, Barbara; Larsen, Rasmus; Gallo, David et al. (2014) Macrophages sense and kill bacteria through carbon monoxide-dependent inflammasome activation. J Clin Invest 124:4926-40
Wegiel, B; Hedblom, A; Li, M et al. (2014) Heme oxygenase-1 derived carbon monoxide permits maturation of myeloid cells. Cell Death Dis 5:e1139
Wegiel, Barbara; Gallo, David; Csizmadia, Eva et al. (2013) Carbon monoxide expedites metabolic exhaustion to inhibit tumor growth. Cancer Res 73:7009-21