Abstract

Interactions between immune and tumor cells in the tumor microenvironment (TME) often impact patient outcome, yet remain poorly understood. In addition, the effects of biophysical features such as hypoxia [low oxygen (O2)] on cells within the TME may lead to tumor evasion. Gamma delta T cells (γδTcs) naturally kill transformed cells and are therefore under development as immunotherapy for various cancers. Clinical trials have proven the safety of γδTc immunotherapy and increased circulating γδTc levels correlate with improved patient outcome. Yet, the function of γδTc tumor infiltrating lymphocytes in human breast cancer remains controversial. Breast tumors can be highly hypoxic, thus therapy must be effective under low O2 conditions. We have found increased infiltration of γδTc in areas of hypoxia in a small cohort of breast tumors; considering their inherent plasticity, it is important to understand how hypoxia influences γδTc function. In vitro, the cell density of expanded primary healthy donor blood-derived human γδTc decreased in response to hypoxia (2% O2) compared to normoxia (20% O2). However, the secretion of macrophage inflammatory protein 1α (MIP1α)/MIP1β, regulated on activation, normal T cell expressed and secreted (RANTES), and CD40L by γδTc were increased after 40 h in hypoxia compared to normoxia concomitant with the stabilization of hypoxia inducible factor 1-alpha protein. Mechanistically, we determined that natural killer group 2, member D (NKG2D) on γδTc and the NKG2D ligand MHC class I polypeptide-related sequence A (MICA)/B on MCF-7 and T47D breast cancer cell lines are important for γδTc cytotoxicity, but that MIP1α, RANTES, and CD40L do not play a direct role in cytotoxicity. Hypoxia appeared to enhance the cytotoxicity of γδTc such that exposure for 48 h increased cytotoxicity of γδTc against breast cancer cells that were maintained in normoxia; conversely, breast cancer lines incubated in hypoxia for 48 h prior to the assay were largely resistant to γδTc cytotoxicity. MICA/B surface expression on both MCF-7 and T47D remained unchanged upon exposure to hypoxia; however, ELISAs revealed increased MICA shedding by MCF-7 under hypoxia, potentially explaining resistance to γδTc cytotoxicity. Despite enhanced γδTc cytotoxicity upon pre-incubation in hypoxia, these cells were unable to overcome hypoxia-induced resistance of MCF-7. Thus, such resistance mechanisms employed by breast cancer targets must be overcome to develop more effective γδTc immunotherapies.

Highlights

  • Low oxygen (O2) levels characterize the microenvironment of many solid tumors, occurring as a consequence of structurally disorganized blood vessels and tumor growth that exceeds the rate of vascularization

  • In order to determine whether gamma delta T cells (γδTcs) are present in areas of hypoxia in breast tumors, we performed immunohistochemistry to detect the hypoxia marker Carbonic anhydrase IX (CAIX) and γδTc using single stains of serial sections from a panel of 17 breast tumors (Table 1)

  • Gamma delta T cells are being developed as immunotherapeutic agents for a variety of cancer indications and clinical trials (Phase I/II) far have shown excellent safety profiles [36]

Read more

Summary

Introduction

Low oxygen (O2) levels (hypoxia) characterize the microenvironment of many solid tumors, occurring as a consequence of structurally disorganized blood vessels and tumor growth that exceeds the rate of vascularization. Cells express genes that are essential for their survival. In tumor cells, this O2-regulated gene expression leads to more aggressive phenotypes, including those that increase the ability of cells to resist therapy, recruit a vasculature and metastasize [2,3,4]. There is a growing body of evidence correlating tumor hypoxia with poor clinical outcome for patients with a variety of cancers [5,6,7]. O2 availability has been shown to regulate immune editing, allowing cancer cells to evade the immune system via a variety of mechanisms [8]. While many studies have focused on myeloid-derived suppressor cells or conventional CD8+ T cells [8], so far none have considered the impact of tumor hypoxia on gamma delta T cells (γδTcs)

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call