Renal cell carcinoma (RCC) encompasses 90% of kidney cancers and 3% of adult cancers. In the U.S. alone, more than 30,000 new cases of RCC are diagnosed and nearly 13,000 patients die from RCC annually. RCC is categorized into three distinct histologic subtypes of which clear cell (ccRCC) is the most common and lethal form, accounting for nearly 90% of RCC-related deaths. The majority of patients now diagnosed with ccRCC will present with organ-confined tumors seemingly amenable to surgical extirpation. Yet, almost half of these patients will experience metastatic relapse after surgery. Treatment options for metastatic ccRCC are limited both in terms of scope and efficacy. Thus, key advances to improve ccRCC patient care will necessarily include: 1) better methods to identify patients at high risk for metastatic relapse following surgery;2) a better understanding of molecular mechanisms that drive ccRCC metastatic progression;and 3) rational, mechanism- targeted therapies to treat metastatic disease in ccRCC patients. Such improvements could emanate from the introduction of newer, more effective drugs or optimization of combination therapies. In this proposal, emphasis will be placed on the latter topic to gain a better understanding of key issues related to the immunobiology and immunotherapeutic treatment of advanced ccRCC. We have recently reported that ccRCC tumors can aberrantly express several relatively novel immune cell coregulatory ligands, namely B7-H1, B7-H3 and B7-H4. We have further shown that enhanced tumor expression of these B7-H ligands can predict aggressive ccRCC behavior including enhanced risk for cancer progression and cancer-related death. Collectively, these observations raise the distinct possibility that human tumors might employ B7-H ligands to disarm host antitumoral immunity in order to promote malignant progression. Thus, tumor-associated B7-H ligands will likely prove useful to refine prognostic algorithms to pinpoint high-risk patients who are most prone to ccRCC progression and death. However, whether tumor- associated B7-H ligands actually function as clinical inhibitors of cell-mediated antitumoral immunity to promote malignant progression remains to be determined. This proposal is singularly devoted to studies that, we believe, will directly improve clinical ccRCC patient care. Specifically, our studies will rigorously test the prognostic and immunotherapeutic potential of B7-H ligands aberrantly expressed by ccRCC. Specifically, we will address a most vital question: """"""""Do B7-H ligands within clinical tumors truly function to impair antitumoral immunity in cancer patients?"""""""" Such findings will help to establish the merits (and limitations) of in vivo B7-H ligand blockade as a clinical immunotherapeutic approach to treat human forms of malignancy. Unifying Hypothesis for Specific Aims 1- 4: The overall hypothesis for this proposal is that the aggressive behavior of ccRCC tumors stems, at least in part, from the ability of tumors to elaborate an array of homologous but spatially distributed B7-H ligands that act in concert to undermine cell-mediated immunity in cancer patients. We further postulate that the most aggressive of ccRCC tumors concurrently express increased levels of other proteins that promote tumor cell proliferation and survival and, may also serve as antigenic moieties to lure immune cells into a perilous intratumoral environment. Thus, each aim of our proposal has been crafted to be free-standing, each addressing key clinical and scientific issues pertaining to B7-H ligands and moving from a macroscopic to microscopic understanding of B7-H ligands as immune- regulators and immunotherapeutic targets for clinical ccRCC.
Specific Aim 1. Test if putative immunosuppressive, tumor-associated B7-H ligands (B7-H1, B7-H3, B7-H4, and PD-1) can collaborate with each other, as well as with other molecular markers that assist in fostering tumor cell proliferation and survival (IMP3, CAIX, Ki-67, and survivin) to improve outcome prediction for ccRCC patients.
Specific Aim 2. Test whether ccRCC metastases are enriched for prognostic B7-H related molecules (B7-H1, B7-H3, B7-H4, and PD-1) as well as IMP3, Ki-67, and survivin when compared against patient-matched primary tumors from which metastases arise.
Specific Aim 3. Test if soluble B7-H (sB7-H) ligands can be detected in the sera of ccRCC patients to improve strategies to monitor and, perhaps, immunotherapeutically treat ccRCC patients.
Specific Aim 4. Test if tumor-associated B7-H ligands inhibit T and NK cell-mediated immune responses within clinical ccRCC specimens using in situ IHC analysis in combination with in vitro assays of immune cell function.

Public Health Relevance

Clear cell renal cell carcinoma (ccRCC) is the most lethal form of kidney malignancy, killing nearly 13,000 patients annually. Current treatment options for metastatic ccRCC are limited as evident in 2 year survival rates of 10-20%. Advancing ccRCC patient care centers around several areas including better methods for identifying patients at high risk for metastatic relapse following surgery, more thorough understanding of molecular mechanisms that drive ccRCC metastatic progression, and developing rational mechanism-targeted therapies to treat metastatic ccRCC disease. We have determined that ccRCC tumors can aberrantly express several relatively novel immune cell coregulatory ligands, namely B7-H1, B7-H3, and B7-H4, the expression of which predict aggressive ccRCC behavior and increased cancer-related death. These observations raise the distinct possibility that human tumors, especially ccRCC, might employ B7-H ligands to blunt patients'anti-tumor immune responses, thereby promoting malignant progression. Tumor- associated B7-H ligands may likely prove useful to refine prognostic algorithms to pinpoint high-risk patients who are most prone to ccRCC progression and death. However, whether tumor-associated B7-H ligands actually function as clinical inhibitors of cell-mediated antitumoral immunity to promote malignant progression remains to be determined, as does the rationale for B7-H blockade as immunotherapeutic means to improve treatment of advanced ccRCC (or any other malignancy for that matter). This proposal is singularly devoted to studies that, we believe, will directly improve clinical ccRCC patient care. Specifically, our studies will rigorously test the prognostic and immunotherapeutic potential of B7-H ligands aberrantly expressed by ccRCC. Specifically, we will address a most vital question: Do B7-H ligands within clinical tumors truly function to impair antitumoral immunity in cancer patients? Such findings will help to establish the merits (and limitations) of in vivo B7-H ligand blockade as a clinical immunotherapeutic approach to treat human forms of malignancy.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA134345-06
Application #
8589371
Study Section
Special Emphasis Panel (ZRG1-ONC-S (02))
Program Officer
Mckee, Tawnya C
Project Start
2008-12-01
Project End
2015-06-30
Budget Start
2014-07-01
Budget End
2015-06-30
Support Year
6
Fiscal Year
2014
Total Cost
$268,069
Indirect Cost
$98,083
Name
Mayo Clinic, Rochester
Department
Type
DUNS #
006471700
City
Rochester
State
MN
Country
United States
Zip Code
55905
Eckel-Passow, Jeanette E; Serie, Daniel J; Cheville, John C et al. (2017) BAP1 and PBRM1 in metastatic clear cell renal cell carcinoma: tumor heterogeneity and concordance with paired primary tumor. BMC Urol 17:19
Dronca, Roxana S; Dong, Haidong (2015) Immunomodulatory antibody therapy of cancer: the closer, the better. Clin Cancer Res 21:944-6
Park, Sean S; Dong, Haidong; Liu, Xin et al. (2015) PD-1 Restrains Radiotherapy-Induced Abscopal Effect. Cancer Immunol Res 3:610-9
Mansfield, Aaron Scott; Roden, Anja C; Peikert, Tobias et al. (2014) B7-H1 expression in malignant pleural mesothelioma is associated with sarcomatoid histology and poor prognosis. J Thorac Oncol 9:1036-40
Gibbons, Rachel M; Liu, Xin; Harrington, Susan M et al. (2014) B7-H1 signaling is integrated during CD8(+) T cell priming and restrains effector differentiation. Cancer Immunol Immunother 63:859-67
Liu, Xin; Gibbons, Rachel M; Harrington, Susan M et al. (2013) Endogenous tumor-reactive CD8(+) T cells are differentiated effector cells expressing high levels of CD11a and PD-1 but are unable to control tumor growth. Oncoimmunology 2:e23972
Frigola, Xavier; Inman, Brant A; Krco, Christopher J et al. (2012) Soluble B7-H1: differences in production between dendritic cells and T cells. Immunol Lett 142:78-82
Gibbons, Rachel M; Liu, Xin; Pulko, Vesna et al. (2012) B7-H1 limits the entry of effector CD8(+) T cells to the memory pool by upregulating Bim. Oncoimmunology 1:1061-1073
Pulko, Vesna; Harris, Kimberley J; Liu, Xin et al. (2011) B7-h1 expressed by activated CD8 T cells is essential for their survival. J Immunol 187:5606-14
Frigola, Xavier; Inman, Brant A; Lohse, Christine M et al. (2011) Identification of a soluble form of B7-H1 that retains immunosuppressive activity and is associated with aggressive renal cell carcinoma. Clin Cancer Res 17:1915-23

Showing the most recent 10 out of 26 publications