Hypoxic Adaptation and Arsenic Trioxide Treatment in Small Cell Lung Carcinoma

University dissertation from Molecular Medicine

Abstract: Small cell lung carcinoma (SCLC) is a very aggressive solid tumor and is often widely metastasized by the time of diagnosis. Despite good response to the initial chemotherapy, SCLC cells often develop multidrug resistance to conventionally used chemotherapeutic drugs, which cause almost all SCLC tumors to relapse. The 5-year survival rate for patients with limited disease is around 20% while it is only a few percent for patients with a disseminating disease. Arsenic trioxide (As2O3) is one of the oldest medicines used for treatment of different diseases and it is today used as first-line treatment for patients with relapsed or refractory acute promyelocytic leukemia. Here, we demonstrate that As2O3 is cytotoxic to SCLC cells and xenotransplanted SCLC tumors at clinically relevant concentrations and the effect is also sustained at hypoxic conditions. Areas of low oxygen tensions, hypoxia, are a common characteristic in solid tumors and are associated with aggressive tumor behavior, treatment resistance and poor outcome in several tumor forms. In response to hypoxia, tumor cells induce a transcriptional shift which is mainly regulated by the transcription factors hypoxia-inducible factor (HIF)-1 and HIF-2. HIF proteins consist of two subunits, an oxygen-regulated α-subunit and a constitutively expressed β-subunit. Previous reports have shown that the transcription factors are differentially regulated over time; HIF-1 primarily mediates the acute hypoxic response, whereas HIF-2 dominates during more chronic phases of hypoxia. We found that SCLC tumor specimens and cells lack expression of HIF-2α protein while HIF-1α is expressed at both acute and prolonged hypoxia. In addition, SCLC cells have a high adaptive capacity to hypoxia including a high proliferation rate and low cell death, even though we demonstrated a modest induction of well-known hypoxia-driven genes. We further show that knockdown of HIF1A using siRNA or shRNA, is not significantly affecting the cell viability of cultured SCLC cells at moderate and severe hypoxia or tumor take and tumor growth in SCLC xenografts. We found that SCLC cells are dependent on glutamine metabolism for cell viability and proliferation, in a HIF-independent fashion. The SCLC cells used here are MYC and MYCL amplified and MYC overexpression is known to stimulate glutaminolysis and lipogenesis. In HIF1A repressed cells that overexpress MYC, genes involved in these pathways are further up-regulated at hypoxic conditions. Taken together, our data indicate that the adaptive capacity to hypoxia is partially HIF-independent in MYC amplified SCLC cells.

  CLICK HERE TO DOWNLOAD THE WHOLE DISSERTATION. (in PDF format)