학술논문

Disruption of IRE1α through its kinase domain attenuates multiple myeloma
Document Type
article
Source
Proceedings of the National Academy of Sciences of the United States of America. 116(33)
Subject
Rare Diseases
Cancer
Hematology
Orphan Drug
Aetiology
2.1 Biological and endogenous factors
Aged
Animals
Bortezomib
Endoplasmic Reticulum Stress
Endoribonucleases
Female
Gene Expression Regulation
Neoplastic
Humans
Lenalidomide
Male
Mice
Middle Aged
Multiple Myeloma
Protein Kinase Inhibitors
Protein Serine-Threonine Kinases
Signal Transduction
Unfolded Protein Response
X-Box Binding Protein 1
Xenograft Model Antitumor Assays
multiple myeloma
endoplasmic reticulum stress
unfolded protein response
inositol-requiring enzyme 1
kinase inhibitors
Language
Abstract
Multiple myeloma (MM) arises from malignant immunoglobulin (Ig)-secreting plasma cells and remains an incurable, often lethal disease despite 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 co-opt the IRE1α-XBP1s pathway; however, the validity of IRE1α as a potential MM therapeutic target is controversial. 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 established frontline antimyeloma agents, bortezomib and lenalidomide. Mechanistically, IRE1α perturbation inhibited expression of key components of the endoplasmic reticulum-associated degradation machinery, as well as secretion of Ig light chains and of cytokines and chemokines known to promote MM growth. Selective IRE1α kinase inhibition reduced viability of CD138+ plasma cells while sparing CD138- cells derived from bone marrows of newly diagnosed or posttreatment-relapsed MM patients, in both US- and European Union-based cohorts. Effective IRE1α inhibition preserved glucose-induced insulin secretion by pancreatic microislets and viability of primary hepatocytes in vitro, as well as normal tissue homeostasis in mice. These results establish a strong rationale for developing kinase-directed inhibitors of IRE1α for MM therapy.