scholarly journals Selinexor—A Drug Review

2021 ◽  
Vol 42 (04) ◽  
pp. 360-363
Author(s):  
Hridya Jayamohanan ◽  
Vaibhav Venniyoor ◽  
Keechilat Pavithran

AbstractSelinexor developed by Karyopharm Therapeutics is the first orally available small-molecule inhibitor of exportin-1 (XPO1). XPO-1 is a protein transporter responsible for the export of macromolecules such as tumor suppressor proteins and oncoprotein mRNAs from the nucleus to the cytoplasm; its inhibition results in blocking of multiple oncogenic pathways. Overexpression of XPO1 is seen in multiple myeloma and various other malignancies and is a poor prognostic marker. Pivotal positive trials have resulted in the approval of selinexor for use in refractory or relapsed diffuse large B cell lymphoma and multiple myeloma. In this review, we briefly cover the drug development, mechanism of action, indications, and toxicities of the drug, and the major pivotal trials.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 606-606
Author(s):  
Michael Milhollen ◽  
Usha Narayanan ◽  
Allison J Berger ◽  
Michael Thomas ◽  
Tary Traore ◽  
...  

Abstract MLN4924 is a first-in-class, small molecule inhibitor of the Nedd8 Activating Enzyme (NAE) in Phase I clinical trials in hematological malignancies. Inhibition of NAE by MLN4924 leads to decreased neddylation and inhibition of cullin-dependent ubiquitin ligase (CDL) activity. CDLs are enzyme complexes which control the ubiquitination and degradation of proteins with important roles in cell cycle progression and cell survival. CDL-mediated degradation of pIkBa regulates NF-kB signaling by freeing cytoplasmic NF-kB transcription factors to translocate to the nucleus promoting cell proliferation and survival. In tumors dependent on the NF-kB pathway for growth and survival, we hypothesized that MLN4924 inhibition of CDL activity would prevent pIkBa degradation and inhibit NF-kB signaling. We utilized models of ABC-like Diffuse Large B-cell Lymphoma (ABC-like DLBCL, OCI-Ly10 and OCI-Ly3 cells) dependent on NF-kB signaling for survival and Germinal Center B-cell like DLBCL (GCB-like DLBCL, OCI-Ly19 and OCI-Ly7 cells) that are not dependent on NF-kB signaling for survival. In vitro, we show that NAE inhibition by MLN4924 in ABC-like DLBCL produces marked stabilization of pIkBa, inhibits p65 nuclear translocation and NF-KB gene transcription demonstrating an inhibition of NF-kB signaling. The inhibition of NF-KB signaling in Ly10 cells results in a G1 phenotype and an acute induction of apoptosis. In contrast, in GCB-like DLBCL we observed an elevation of multiple substrates of the CDLs, an accumulation of cells with increased DNA content (>4N) followed by a DNA damage response and induction of cell death. This mechanism of action in GCB-like DLBCL cells is observed in other tumor cell lines that are not dependent on NF-kB signaling for survival. In vivo administration of MLN4924 to mice bearing xenograft tumors of OCI-Ly10 and OCI-Ly19 resulted in a pharmacodynamic response of NAE pathway inhibition. In both models, a single dose of MLN4924 resulted in time and dose-dependent inhibition of total neddylated cullin levels and stabilization of CDL substrates including the CDL3Keap1 substrate, Nrf-2. Notably, in the OCI-Ly10 model, a single dose of MLN4924 resulted in a marked elevation of pIkBa levels, indicative of NF-kB pathway inhibition, and induction of apoptosis. In both OCI-Ly10 and OCI-Ly19 xenograft models, inhibition of the NAE pathway following repeated daily and intermittent dosing of MLN4924 translated into significant tumor growth inhibition. In the OCI-Ly10 model tumor regressions were observed showing this model to be particularly sensitive to MLN4924 treatment, reflecting the addiction of these tumors to NF-kB signaling. Additionally we demonstrate an inhibition of the NAE pathway and NF-KB signaling in a primary human tumor DLBCL xenograft model (PHTX-22L) resulting in tumor regressions following MLN4924 treatment. In summary, in tumors dependent on NF-kB signaling for growth and survival, MLN4924 inhibition of CDL activity provides a novel mechanism for targeted NF-kB pathway modulation and therapeutic intervention. In addition, these data demonstrate that MLN4924 is a novel agent that has broad activity in pre-clinical models of lymphoma.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4435-4435
Author(s):  
Matko Kalac ◽  
Michael Mangone ◽  
Alison Rinderspacher ◽  
Shi-Xian Deng ◽  
Luigi Scotto ◽  
...  

Abstract The first two authors contributed equally to this work Identifying pharmacologic strategies to inhibit the activation of NF-κB and its target genes has been a major research pursuit. To date, no direct inhibitors of the NF-κB subunits have been explored in the clinic. Based on the constitutive activation of NF-κB in diffuse large B-cell lymphoma (DLBCL), we used this disease model to develop drugs targeting NF-κB. Using a fluorescence-based high throughput screening (HTC) approach, a unique N-quinoline-benzenesulfonamide (NQBS) scaffold was identified as potential small molecule inhibitor of the NF-κB pathway. A confocal microscopy based HTC assay performed in human umbilical vein endothelial cells (HUVEC) identified hit compounds that contained a unique NQBS core structure. The assay screened for compounds that inhibited nuclear translocation of NF-κB subunits in TNFα-induced HUVEC cells. To date over 100 NQBS analogs have been synthesized with varying potency and cytotoxicity in inhibiting growth of DLBCL lines (OCI-Ly10, RIVA, HBL-1 and OCI-Ly3). Cytotoxicity assays demonstrated that the most potent compounds exhibit IC50s in the 0.5 to 1.5 µM range. These most potent NQBS analogs identified as CU-O42 CU-O47 and CU-O75 were also able to induce apoptosis and caspase activation. Apoptosis was preceded by exclusion of the NF-κB proteins from the nucleus. To analyze the localization of NF-κB proteins within the cell compartments before and after the treatment with CU-O42, CU-O47 and CU-O75, we used confocal microscopy, electromobility shift (EMSA) and ELISA assays. Control cells tested positive for p50/p65 both within the cytoplasm and the nucleus. Following treatment with CU-O42 NF-κB was sequestered within the cytoplasm of the cell which occurred as early as 3h after exposure. In addition, all three analogs reduced the nuclear levels of NF-κB in a concentration-dependent manner when measured by EMSA and ELISA. Furthermore, CU-O47 and CU-O75 were able to inhibit TNFα induced luciferase expression in a HEK293T cell model where luciferase is controlled by an NF-κB promoter. A KINOMEscan platform (examining the activity of over 450 different kinases) showed that no NQBS analog screened (CU-O42 and CU-O75) inhibited any of the kinases in the assay. In addition, a proteasome inhibition assay tested negative for trypsin-like and chromotrypsin-like protease activity (CU-O42, CU-O47 and CU-O75). Stabilization of the inactive trimer of p50, p65 and IκBα was hypothesized as a potential mechanism of action of CU-O42 and CU-O75 through Internal Coordinate Mechanics (ICM) software. This binding hypothesis was further corroborated by cellular thermal shift assays (CETSA) with an increase of the IκBα melting temperatures (2.5-3°C) in whole cell lysates following rapid (30min) exposure to CU-O42 and CU-O75. Using a genome-wide regulatory network perturbation analysis (DeMAND) based on the RNA-Seq data collected from OCI-Ly10 cells treated with CU-O75, we identified IκBα as one of the potential targets of the compounds. Gene set enrichment analysis demonstrated NF-κB target gene downregulation using IC20 of CU-O75 at 24h (p=0.045). In vivo experiments were conducted in two models: (1) xenografts with human DLBCL cell lines of both ABC and GC subtype; and (2) myc cherry luciferase mouse model where mice spontaneously develop aggressive lymphomas. In both models, CU-O42 was able to inhibit tumor growth. Interestingly, in the xenograft model, malignant cell growth was inhibited in both ABC (HBL-1) and GC (OCI-Ly1) cells when compared to controls (p=0.01 and p=0.02). However, overall survival of mice with ABC xenografts treated with CU-042 significantly exceeded the survival of mice with GC xenografts (p<0.01) suggesting a more sustainable response in this subtype of disease, consistent with its dependency on NF-κB. Identification of a unique NQBS scaffold has led to the chemical synthesis of over 100 structural analogs with a potent inhibition on NF-κB nuclear translocation. They display potent activity across a panel of lymphoma cell lines, producing a survival benefit in mice implanted with an ABC-subtype of lymphoma. ICM, CETSA and DeMAND suggest that this is a direct effect mediated on the proteins within the p65/p50/IκBα complex. These findings point to a novel mechanism of action and warrant further research into potential clinical translation of this class of small molecules. Disclosures Califano: Thermo Fischer Scientific: Consultancy; Ipsen pharmaceuticals: Consultancy; Cancer Genetics Inc: Consultancy; Therasis Inc: Employment. O'Connor:Spectrum Pharmaceuticals: Consultancy, Honoraria, Research Funding; Takeda Millennium: Consultancy, Honoraria, Research Funding; Celgene: Consultancy, Research Funding; Bristol-Myers Squibb Company: Consultancy; Novartis: Consultancy, Honoraria; Seattle Genetics: Consultancy; Bayer: Consultancy, Honoraria; Mundipharma: Consultancy, Honoraria, Research Funding; Acetylon Pharmaceuticals, INC: Consultancy.


2020 ◽  
Author(s):  
Issa Ismail Issa ◽  
Rasmus Froberg Brøndum ◽  
Hanne Due ◽  
Linnéa Schmidt ◽  
Martin Bøgsted ◽  
...  

2020 ◽  
Vol 13 (11) ◽  
pp. 396
Author(s):  
Massimo Offidani ◽  
Maria Teresa Petrucci

Multiple myeloma (MM) is the second most common hematological cancer after diffuse large B-cell lymphoma, accounting for about 10% of all blood cancers [...]


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3483-3483
Author(s):  
Changju Qu ◽  
Yadong Liu ◽  
Kranthi Kunkalla ◽  
Nitin K Agarwal ◽  
Estelle Bourbon ◽  
...  

Abstract Abstract 3483 Activated Hedgehog (HH) signaling was identified in our laboratory to contribute to cell survival, proliferation and chemotolerance of diffuse large B-cell lymphoma (DLBCL) (Leukemia 2010;24:1025 & Oncogene 2011, in press). The HH receptor complex is integrated by two main proteins, smoothened (SMO) and patched-1 (PTCH1). SMO is a seven-transmembrane G protein-coupled receptor that transduces HH signal to the cytoplasm and has glioma-associated oncogene homologue (GLI) proteins as major signaling transcription effectors. PTCH1 is a 12 transmembrane protein that inhibits SMO in the absence of HH ligands. Here, we investigated potential cross talk between SMO with the activation status of PI3K/AKT and NF-kB, two relevant oncogenic pathways in DLBCL. Using a small interfering (si)RNA approach and DLBCL cell lines of germinal center (GC) and activated B-cell (ABC) type we found that the expression levels of SMO modulate the activation status of AKT and canonical NF-KB pathways in DLBCL cells of GC type and mainly AKT in those of ABC type. In DLBCL cells of GC cell type, silencing SMO resulted in decrease of the phosphorylation levels of ser473p-AKT and ser536p-P65 and silencing PTCH1 resulted in increase of the phosphorylation levels of both proteins. The same silencing experimental approach in DLBCL cells of ABC type resulted in similar modulation in the activation status of the AKT but not, or to less extent, in the activation of NF-kB pathway. The modulation of the activation status of the NF-KB pathway was also confirmed using protein nuclear extracts and DNA binding ELISA assays. In cells of both DLBCL subtypes, silencing of the SMO transcriptional effector GLI1 showed no changes in the activation status of both pathways. The modulation in the activation status of AKT and NF-KB was also detected using SMO inhibitors, cyclopamine-KAAD and HhAntag (Genentech Inc) or activators, purmorphamine and recombinant HH protein. Combinatorial treatments with increasing concentrations of SMO inhibitors (cyclopamine and HhAntag [1.6, 3.2 and 4.8 μM]) with minimal lethal doses of Ly294002 (PI3K inhibitor) or BAY-11 (NF-KB inhibitor) were also performed. Using cell viability, and apoptosis (Annexin-V) assays, we found that combined treatments of PI3K or NF-KB inhibitors with a SMO inhibitor resulted in an additive/synergistic decrease of cell viability and increase of apoptosis in comparison to the treatments with SMO inhibitors alone. Taken together, our data shed novel light on the contribution of SMO on the activation of PI3K/AKT and NF-kB pathways in DLBCL. Our data also provide a rationale to use SMO inhibitors in combination with inhibitors of other oncogenic pathways such as PI3K/AKT and/or NF-KB for the treatment of patients with DLBCL. Disclosures: No relevant conflicts of interest to declare.


2016 ◽  
Vol 34 (23) ◽  
pp. 2698-2704 ◽  
Author(s):  
Alexander M. Lesokhin ◽  
Stephen M. Ansell ◽  
Philippe Armand ◽  
Emma C. Scott ◽  
Ahmad Halwani ◽  
...  

Purpose Cancer cells can exploit the programmed death-1 (PD-1) immune checkpoint pathway to avoid immune surveillance by modulating T-lymphocyte activity. In part, this may occur through overexpression of PD-1 and PD-1 pathway ligands (PD-L1 and PD-L2) in the tumor microenvironment. PD-1 blockade has produced significant antitumor activity in solid tumors, and similar evidence has emerged in hematologic malignancies. Methods In this phase I, open-label, dose-escalation, cohort-expansion study, patients with relapsed or refractory B-cell lymphoma, T-cell lymphoma, and multiple myeloma received the anti–PD-1 monoclonal antibody nivolumab at doses of 1 or 3 mg/kg every 2 weeks. This study aimed to evaluate the safety and efficacy of nivolumab and to assess PD-L1/PD-L2 locus integrity and protein expression. Results Eighty-one patients were treated (follicular lymphoma, n = 10; diffuse large B-cell lymphoma, n = 11; other B-cell lymphomas, n = 10; mycosis fungoides, n = 13; peripheral T-cell lymphoma, n = 5; other T-cell lymphomas, n = 5; multiple myeloma, n = 27). Patients had received a median of three (range, one to 12) prior systemic treatments. Drug-related adverse events occurred in 51 (63%) patients, and most were grade 1 or 2. Objective response rates were 40%, 36%, 15%, and 40% among patients with follicular lymphoma, diffuse large B-cell lymphoma, mycosis fungoides, and peripheral T-cell lymphoma, respectively. Median time of follow-up observation was 66.6 weeks (range, 1.6 to 132.0+ weeks). Durations of response in individual patients ranged from 6.0 to 81.6+ weeks. Conclusion Nivolumab was well tolerated and exhibited antitumor activity in extensively pretreated patients with relapsed or refractory B- and T-cell lymphomas. Additional studies of nivolumab in these diseases are ongoing.


Sign in / Sign up

Export Citation Format

Share Document