RT Journal Article SR Electronic T1 Disruption of IRE1α through its Kinase Domain Attenuates Multiple Myeloma JF bioRxiv FD Cold Spring Harbor Laboratory SP 495242 DO 10.1101/495242 A1 Jonathan M Harnoss A1 Adrien Le Thomas A1 Scot A Marsters A1 David A Lawrence A1 Min Lu A1 Yung-Chia Ariel Chen A1 Jing Qing A1 Klara Totpal A1 David Kan A1 Ehud Segal A1 Heidi Ackerly Wallweber A1 Weiru Wang A1 Kevin Clark A1 Susan Kaufman A1 Maureen Beresini A1 Wendy Sandoval A1 Maria Lorenzo A1 Jiansheng Wu A1 Justin Ly A1 Tom De Bruyn A1 Amy Heidersbach A1 Benjamin Haley A1 Alvin Gogineni A1 Robby Weimer A1 Dong Lee A1 Marie-Gabrielle Braun A1 Joachim Rudolph A1 Michael J VanWyngarden A1 Daniel W Sherbenou A1 Patricia Gomez-Bougie A1 Martine Amiot A1 Diego Acosta-Alvear A1 Peter Walter A1 Avi Ashkenazi YR 2018 UL http://biorxiv.org/content/early/2018/12/14/495242.abstract AB Multiple myeloma (MM) arises from malignant immunoglobulin-secreting plasma cells and remains an incurable, often lethal disease despite recent therapeutic advances. The unfolded-protein response sensor IRE1α supports protein secretion by deploying a kinase-endoribonuclease module to activate the transcription factor XBP1s. MM cells may coopt the IRE1α-XBP1s pathway; however, the validity of IRE1α as a potential MM therapeutic target is controversial. Here we show that genetic disruption of IRE1α or XBP1s, or pharmacologic IRE1α kinase inhibition, attenuated subcutaneous or orthometastatic growth of MM tumors in mice, and augmented efficacy of two well-established frontline antimyeloma agents, bortezomib or lenalidomide. Mechanistically, IRE1α perturbation inhibited expression of key components of the ER-associated degradation machinery, as well as cytokines and chemokines known to promote MM growth. Selective IRE1α kinase inhibition reduced viability of CD138+ plasma cells while sparing CD138− cells from bone marrow of newly diagnosed MM patients or patients whose disease relapsed after 1 - 4 lines of treatment in both US- and EU-based cohorts. IRE1α inhibition preserved survival and glucose-induced insulin secretion by pancreatic microislets. Together, these results establish a strong therapeutic rationale for targeting IRE1α with kinase-based small-molecule inhibitors in MM.Significance statement Multiple myeloma (MM) is a lethal malignancy of plasma cells. MM cells have an expanded endoplasmic reticulum (ER) that is constantly under stress due to immunoglobulin hyperproduction. The ER-resident sensor IRE1α mitigates ER stress by expanding the ER’s protein-folding capacity while supporting proteasomal degradation of misfolded ER proteins. IRE1α elaborates these functions by deploying its cytoplasmic kinase-RNase module to activate the transcription factor XBP1s. The validity of IRE1α as a potential therapeutic target in MM has been questioned. Using genetic and pharmacologic disruption in vitro and in vivo, we demonstrate that the IRE1α-XBP1s pathway plays a critical role in MM growth. We further show that IRE1α’s kinase domain is an effective and safe potential small-molecule target for MM therapy.