Oncotarget

Research Papers:

PRIMA-1 targets the vulnerability of multiple myeloma of deregulated protein homeostasis through the perturbation of ER stress via p73 demethylation

Phaik Ju Teoh, Chonglei Bi, Chirackal Sintosebastian, Liang Seah Tay, Rafael Fonseca and Wee Joo Chng _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2016; 7:61806-61819. https://doi.org/10.18632/oncotarget.11241

Metrics: PDF 2220 views  |   HTML 2393 views  |   ?  


Abstract

Phaik Ju Teoh1,4, Chonglei Bi1, Chirackal Sintosebastian2, Liang Seah Tay3, Rafael Fonseca2, Wee Joo Chng1,2,4

1Cancer Science Institute Singapore, National University of Singapore, Singapore

2Department of Hematology-Oncology, Mayo Clinic, Scottsdale, Arizona, USA

3Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore

4Department of Haematology-Oncology, National University Cancer Institute, Singapore

Correspondence to:

Wee Joo Chng, email: [email protected]

Keywords: multiple myeloma, PRIMA-1, ER stress, p73, bortezomib

Received: May 05, 2016    Accepted: July 28, 2016    Published: August 12, 2016

ABSTRACT

Despite therapeutic advancement, multiple myeloma (MM) remains incurable with drug resistance being one of the main challenges in the clinic. Myeloma cells possess high protein secretory load, leading to increased intracellular endoplasmic reticulum (ER) stress. Hence, they are vulnerable to further perturbation to its protein homeostasis. In studying the therapeutic mechanism of PRIMA-1 (mutant-p53-reactivating-agent), we uncovered its novel p53-independent-mechanism that can be exploited for myeloma. Despite its inability in restoring the wild type-p53 protein conformation and transcriptional function in the mutant-p53-human-myeloma-cells, PRIMA-1 was efficacious against myeloma cells with differential p53 genotypes. Strikingly, cells without p53 expression demonstrated highest drug sensitivity. Genome-wide gene-expression analysis revealed the involvement of ER stress/UPR-pathway in inducing PRIMA-1-toxicity. UPR markers, HSP70, CHOP and GADD34, were significantly up-regulated, concomitantly with the induction of apoptosis. Furthermore, there was a global attenuation of protein synthesis, correlated with phospho-eIF2a up-regulation. Mechanistically, we identified that PRIMA-1 could cause the demethylation of TP73, through DNMT1 depletion, to subsequently enhance UPR. Of clinical significance, we observed that PRIMA-1 had additive therapeutic effects with another UPR-inducing-agent, bortezomib. Importantly, it can partially re-sensitize bortezomib-resistant cells to bortezomib. Given that MM is already stressed at the baseline in the ER, our results implicated that PRIMA-1 is a potential therapeutic option in MM by targeting its Achilles heel.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 11241