Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 244
1.
Sci Rep ; 14(1): 9305, 2024 04 23.
Article En | MEDLINE | ID: mdl-38653804

Dysregulated nuclear-cytoplasmic trafficking has been shown to play a role in oncogenesis in several types of solid tumors and hematological malignancies. Exportin 1 (XPO1) is responsible for the nuclear export of several proteins and RNA species, mainly tumor suppressors. KPT-330, a small molecule inhibitor of XPO1, is approved for treating relapsed multiple myeloma and diffuse large B-cell lymphoma. Cutaneous T-cell lymphoma (CTCL) is an extranodal non-Hodgkin lymphoma with an adverse prognosis and limited treatment options in advanced stages. The effect of therapeutically targeting XPO1 with KPT-330 in CTCL has not been established. We report that XPO1 expression is upregulated in CTCL cells. KPT-330 reduces cell proliferation, induces G1 cell cycle arrest and apoptosis. RNA-sequencing was used to explore the underlying mechanisms. Genes associated with the cell cycle and the p53 pathway were significantly enriched with KPT-330 treatment. KPT-330 suppressed XPO1 expression, upregulated p53, p21WAF1/Cip1, and p27Kip1 and their nuclear localization, and downregulated anti-apoptotic protein (Survivin). The in vivo efficacy of KPT-330 was investigated using a bioluminescent xenograft mouse model of CTCL. KPT-330 blocked tumor growth and prolonged survival (p < 0.0002) compared to controls. These findings support investigating the use of KPT-330 and next-generation XPO1 inhibitors in CTCL.


Apoptosis , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Exportin 1 Protein , Karyopherins , Lymphoma, T-Cell, Cutaneous , Receptors, Cytoplasmic and Nuclear , Triazoles , Tumor Suppressor Protein p53 , Humans , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Lymphoma, T-Cell, Cutaneous/drug therapy , Lymphoma, T-Cell, Cutaneous/pathology , Lymphoma, T-Cell, Cutaneous/metabolism , Lymphoma, T-Cell, Cutaneous/genetics , Apoptosis/drug effects , Animals , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Karyopherins/metabolism , Karyopherins/antagonists & inhibitors , Mice , Cell Line, Tumor , Triazoles/pharmacology , Cell Proliferation/drug effects , Hydrazines/pharmacology , Hydrazines/therapeutic use , Xenograft Model Antitumor Assays , Signal Transduction/drug effects , Gene Expression Regulation, Neoplastic/drug effects
2.
J Oncol Pharm Pract ; 30(3): 535-546, 2024 Apr.
Article En | MEDLINE | ID: mdl-38454813

OBJECTIVE: Multiple myeloma cells resist standard therapies due to overexpression of the transport protein, exportin 1. Selinexor is a novel drug that targets the Exportin 1 protein in these cells. DATA SOURCE: A comprehensive search was done, and data showing the efficacy and safety of selinexor in relapsed/refractory multiple myeloma was collected using PubMed, Google Scholar, and clincialtrials.gov. DATA SUMMARY: Results from the clinical trials STORM, BOSTON, and STOMP were included. Parts I and II of the STORM trial revealed a progression-free survival (PFS) of 4.7 and 3.7 months, a median duration of response of 6.2 and 4.4 months, and an overall survival of 7.3 and 8.4 months, respectively. BOSTON trial's SVd arm (selinexor, bortezomib, and dexamethasone) had a median follow-up period of 13.2 months and an mPFS of 13.93 months. The Vd arm (bortezomib and dexamethasone) had a median follow-up duration of 16.5 months and an mPFS of 9.46 months. The STOMP trial is still active and has limited data available. The SKd arm (selinexor, carfilzomib, and dexamethasone) reported an overall response rate of 66.7% in patients with triple refractory multiple myeloma, and 82% in patients with high-risk cytogenetics. The SPd arm (selinexor, pomalidomide, and dexamethasone) shows an overall response rate of 54.30% in pomalidomide naïve-nonrefractory, 35.70% in pomalidomide refractory and 60% in those dosed at RP2D. SRd arm (selinexor, lenalidomide, and dexamethasone) shows an overall response rate of 91.7% in lenalidomide naïve and 12.5% in lenalidomide refractory patients. SVd (selinexor, bortezomib, and dexamethasone) arm reported an overall response rate of 63% in all patients while the SDd arm (selinexor, daratumumab, and dexamethasone) showed an overall response rate of 73%. CONCLUSION: To improve the outcome of patients with relapsed/refractory multiple myeloma, it is critical to develop new therapies, assess potential therapeutic synergies, and overcome drug resistance by determining the efficacy of multiple myeloma therapies across multiple disease subgroups.


Hydrazines , Multiple Myeloma , Triazoles , Multiple Myeloma/drug therapy , Humans , Hydrazines/therapeutic use , Triazoles/therapeutic use , Dexamethasone/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm , Progression-Free Survival , Exportin 1 Protein , Karyopherins/antagonists & inhibitors , Bortezomib/therapeutic use , Receptors, Cytoplasmic and Nuclear , Antineoplastic Agents/therapeutic use , Neoplasm Recurrence, Local/drug therapy
3.
Expert Opin Pharmacother ; 25(4): 421-434, 2024 Mar.
Article En | MEDLINE | ID: mdl-38503547

INTRODUCTION: Selinexor, an XPO1 inhibitor, has emerged as a promising therapeutic option in the challenging landscape of relapsed/refractory multiple myeloma (RRMM). AREAS COVERED: This article provides a review of selinexor, with a focus on available clinical studies involving MM patients and its safety profile. Clinical trials, such as STORM and BOSTON, have demonstrated its efficacy, particularly in combination regimens, showcasing notable overall response rates (ORR) and prolonged median progressionfree survival (mPFS). Selinexor's versatility is evident across various combinations, including carfilzomibdexamethasone (XKd), lenalidomidedexamethasone (XRd), and pomalidomidedexamethasone (XPd), with efficacy observed even in tripleclass refractory and highrisk patient populations. However, challenges, including resistance mechanisms and adverse events, necessitate careful management. Realworld evidence also underscores selinexor's effectiveness in RRMM, though dose adjustments and supportive measures remain crucial. Ongoing trials are exploring selinexor in diverse combinations and settings, including pomalidomidenaïve patients and postautologous stem cell transplant (ASCT) maintenance. EXPERT OPINION: The evolving landscape of selinexor's role in the sequencing of treatment for RRMM, its potential in highrisk patients, including those with extramedullary disease, as revealed in the most recent international meetings, and ongoing investigations signal a dynamic era in myeloma therapeutics. Selinexor emerges as a pivotal component in multidrug strategies and innovative combinations.


Hydrazines , Multiple Myeloma , Triazoles , Multiple Myeloma/drug therapy , Humans , Hydrazines/therapeutic use , Hydrazines/adverse effects , Triazoles/therapeutic use , Triazoles/adverse effects , Karyopherins/antagonists & inhibitors , Exportin 1 Protein , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Progression-Free Survival
4.
Nature ; 627(8002): 212-220, 2024 Mar.
Article En | MEDLINE | ID: mdl-38355801

Circular RNAs (circRNAs), which are increasingly being implicated in a variety of functions in normal and cancerous cells1-5, are formed by back-splicing of precursor mRNAs in the nucleus6-10. circRNAs are predominantly localized in the cytoplasm, indicating that they must be exported from the nucleus. Here we identify a pathway that is specific for the nuclear export of circular RNA. This pathway requires Ran-GTP, exportin-2 and IGF2BP1. Enhancing the nuclear Ran-GTP gradient by depletion or chemical inhibition of the major protein exporter CRM1 selectively increases the nuclear export of circRNAs, while reducing the nuclear Ran-GTP gradient selectively blocks circRNA export. Depletion or knockout of exportin-2 specifically inhibits nuclear export of circRNA. Analysis of nuclear circRNA-binding proteins reveals that interaction between IGF2BP1 and circRNA is enhanced by Ran-GTP. The formation of circRNA export complexes in the nucleus is promoted by Ran-GTP through its interactions with exportin-2, circRNA and IGF2BP1. Our findings demonstrate that adaptors such as IGF2BP1 that bind directly to circular RNAs recruit Ran-GTP and exportin-2 to export circRNAs in a mechanism that is analogous to protein export, rather than mRNA export.


Active Transport, Cell Nucleus , Cell Nucleus , RNA Transport , RNA, Circular , Active Transport, Cell Nucleus/physiology , Cell Nucleus/metabolism , Guanosine Triphosphate/metabolism , Karyopherins/antagonists & inhibitors , Karyopherins/deficiency , Karyopherins/genetics , Karyopherins/metabolism , Nuclear Proteins/metabolism , ran GTP-Binding Protein/metabolism , RNA, Circular/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA-Binding Proteins/metabolism , Exportin 1 Protein/metabolism , Protein Transport
5.
Leukemia ; 37(10): 2036-2049, 2023 10.
Article En | MEDLINE | ID: mdl-37528310

The first-in-class inhibitor of exportin-1 (XPO1) selinexor is currently under clinical investigation in combination with the BTK inhibitor ibrutinib for patients with chronic lymphocytic leukaemia (CLL) or non-Hodgkin lymphoma. Selinexor induces apoptosis of tumour cells through nuclear retention of tumour suppressor proteins and has also recently been described to modulate natural killer (NK) cell and T cell cytotoxicity against lymphoma cells. Here, we demonstrate that XPO1 inhibition enhances NK cell effector function against primary CLL cells via downregulation of HLA-E and upregulation of TRAIL death receptors DR4 and DR5. Furthermore, selinexor potentiates NK cell activation against CLL cells in combination with several approved treatments; acalabrutinib, rituximab and obinutuzumab. We further demonstrate that lymph node associated signals (IL-4 + CD40L) inhibit NK cell activation against CLL cells via upregulation of HLA-E, and that inhibition of XPO1 can overcome this protective effect. These findings allow for the design of more efficacious combination strategies to harness NK cell effector functions against CLL.


Histocompatibility Antigens Class I , Hydrazines , Karyopherins , Leukemia, Lymphocytic, Chronic, B-Cell , Receptors, Cytoplasmic and Nuclear , Humans , Karyopherins/antagonists & inhibitors , Karyopherins/metabolism , Killer Cells, Natural/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Hydrazines/pharmacology , Histocompatibility Antigens Class I/metabolism , Exportin 1 Protein , HLA-E Antigens
6.
Med ; 3(11): 730-732, 2022 11 11.
Article En | MEDLINE | ID: mdl-36370692

We urgently need to identify new druggable targets for pediatric cancers. Coutinho et al.1 identified and validated XPO1 aberrant activity in patients with Wilms tumor (WT) and malignant rhabdoid tumor (MRT) using public databases and report an early proof-of-concept bench-to-bedside translation of XPO1 inhibition in pediatric WT and MRT.


Karyopherins , Rhabdoid Tumor , Wilms Tumor , Child , Humans , Genomics , Karyopherins/antagonists & inhibitors , Karyopherins/genetics , Precision Medicine , Receptors, Cytoplasmic and Nuclear/genetics , Exportin 1 Protein
7.
Nat Cancer ; 3(7): 837-851, 2022 07.
Article En | MEDLINE | ID: mdl-35668193

Selinexor is a first-in-class inhibitor of the nuclear exportin XPO1 that was recently approved by the US Food and Drug Administration for the treatment of multiple myeloma and diffuse large B-cell lymphoma. In relapsed/refractory acute myeloid leukemia (AML), selinexor has shown promising activity, suggesting that selinexor-based combination therapies may have clinical potential. Here, motivated by the hypothesis that selinexor's nuclear sequestration of diverse substrates imposes pleiotropic fitness effects on AML cells, we systematically catalog the pro- and anti-fitness consequences of selinexor treatment. We discover that selinexor activates PI3Kγ-dependent AKT signaling in AML by upregulating the purinergic receptor P2RY2. Inhibiting this axis potentiates the anti-leukemic effects of selinexor in AML cell lines, patient-derived primary cultures and multiple mouse models of AML. In a syngeneic, MLL-AF9-driven mouse model of AML, treatment with selinexor and ipatasertib outperforms both standard-of-care chemotherapy and chemotherapy with selinexor. Together, these findings establish drug-induced P2RY2-AKT signaling as an actionable consequence of XPO1 inhibition in AML.


Leukemia, Myeloid, Acute , Proto-Oncogene Proteins c-akt , Animals , Antineoplastic Combined Chemotherapy Protocols , Karyopherins/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Mice , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Purinergic P2Y2/metabolism , United States , Exportin 1 Protein
8.
J Virol ; 96(3): e0127321, 2022 02 09.
Article En | MEDLINE | ID: mdl-34757845

After receptor-mediated endocytosis and endosomal escape, adenoviral capsids can travel via microtubule organizing centers to the nuclear envelope. Upon capsid disassembly, viral genome import into nuclei of interphase cells then occurs through nuclear pore complexes, involving the nucleoporins Nup214 and Nup358. Import also requires the activity of the classic nuclear export receptor CRM1, as it is blocked by the selective inhibitor leptomycin B. We have now used artificially enucleated as well as mitotic cells to analyze the role of an intact nucleus in different steps of the viral life cycle. In enucleated U2OS cells, viral capsids traveled to the microtubule organizing center, whereas their removal from this complex was blocked, suggesting that this step required nuclear factors. In mitotic cells, on the other hand, CRM1 promoted capsid disassembly and genome release, suggesting a role of this protein that does not require intact nuclear envelopes or nuclear pore complexes and is distinct from its function as a nuclear export receptor. Similar to enucleation, inhibition of CRM1 by leptomycin B also leads to an arrest of adenoviral capsids at the microtubule organizing center. In a small-scale screen using leptomycin B-resistant versions of CRM1, we identified a mutant, CRM1 W142A P143A, that is compromised with respect to adenoviral capsid disassembly in both interphase and mitotic cells. Strikingly, this mutant is capable of exporting cargo proteins out of the nucleus of living cells or digitonin-permeabilized cells, pointing to a role of the mutated region that is not directly linked to nuclear export. IMPORTANCE A role of nucleoporins and of soluble transport factors in adenoviral genome import into the nucleus of infected cells in interphase has previously been established. The nuclear export receptor CRM1 promotes genome import, but its precise function is not known. Using enucleated and mitotic cells, we showed that CRM1 does not simply function by exporting a crucial factor out of the nucleus that would then trigger capsid disassembly and genome import. Instead, CRM1 has an export-independent role, a notion that is also supported by a mutant, CRM1 W142A P143A, which is export competent but deficient in viral capsid disassembly, in both interphase and mitotic cells.


Adenoviridae Infections/metabolism , Adenoviridae Infections/virology , Adenoviridae/physiology , Capsid/metabolism , Host-Pathogen Interactions , Karyopherins/metabolism , Nuclear Envelope/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Active Transport, Cell Nucleus , Adenoviridae/drug effects , Cell Line , Genome, Viral , Humans , Karyopherins/antagonists & inhibitors , Karyopherins/chemistry , Karyopherins/genetics , Microtubules/metabolism , Models, Molecular , Mutation , Protein Conformation , Protein Transport , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Structure-Activity Relationship , Virus Replication , Exportin 1 Protein
9.
J Med Chem ; 64(21): 15534-15548, 2021 11 11.
Article En | MEDLINE | ID: mdl-34669417

Chromosome region maintenance 1 (CRM1) is a major nuclear export receptor protein and contributes to cell homeostasis by mediating the transport of cargo from the nucleus to the cytoplasm. CRM1 is a therapeutic target comprised of several tumor types, including osteosarcoma, multiple myeloma, gliomas, and pancreatic cancer. In the past decade, dozens of CRM1 inhibitors have been discovered and developed, including KPT-330, which received FDA approval for multiple myeloma (MM) and diffuse large B-cell lymphoma (DLBCL) in 2019 and 2020, respectively. This review summarizes the biological functions of CRM1, the current understanding of the role CRM1 plays in cancer, the discovery of CRM1 small-molecule inhibitors, preclinical and clinical studies on KPT-330, and other recently developed inhibitors. A new CRM1 inhibition mechanism and structural dynamics are discussed. Through this review, we hope to guide the future design and optimization of CRM1 inhibitors.


Antineoplastic Agents/pharmacology , Drug Discovery , Karyopherins/antagonists & inhibitors , Lymphoma, Large B-Cell, Diffuse/drug therapy , Multiple Myeloma/drug therapy , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Dose-Response Relationship, Drug , Humans , Karyopherins/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Molecular Structure , Multiple Myeloma/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Exportin 1 Protein
10.
Radiat Oncol ; 16(1): 191, 2021 Sep 28.
Article En | MEDLINE | ID: mdl-34583727

Glioblastoma is the most common primary brain malignancy and carries with it a poor prognosis. New agents are urgently needed, however nearly all Phase III trials of GBM patients of the past 25 years have failed to demonstrate improvement in outcomes. In 2019, the National Cancer Institute Clinical Trials and Translational Research Advisory Committee (CTAC) Glioblastoma Working Group (GBM WG) identified 5 broad areas of research thought to be important in the development of new herapeutics for GBM. Among those was optimizing radioresponse for GBM in situ. One such strategy to increase radiation efficacy is the addition of a radiosensitizer to improve the therapeutic ratio by enhancing tumor sensitivity while ideally having minimal to no effect on normal tissue. Historically the majority of trials using radiosensitizers have been unsuccessful, but they provide important guidance in what is required to develop agents more efficiently. Improved target selection is essential for a drug to provide maximal benefit, and once that target is identified it must be validated through pre-clinical studies. Careful selection of appropriate in vitro and in vivo models to demonstrate increased radiosensitivity and suitable bioavailability are then necessary to prove that a drug warrants advancement to clinical investigation. Once investigational agents are validated pre-clinically, patient trials require consistency both in terms of planning study design as well as reporting efficacy and toxicity in order to assess the potential benefit of the drug. Through this paper we hope to outline strategies for developing effective radiosensitizers against GBM using as models the examples of XPO1 inhibitors and HDAC inhibitors developed from our own lab.


Brain Neoplasms/radiotherapy , Glioblastoma/radiotherapy , Radiation-Sensitizing Agents/therapeutic use , Clinical Trials as Topic , Humans , Karyopherins/antagonists & inhibitors , Neoplastic Stem Cells/drug effects , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Valproic Acid/therapeutic use , Exportin 1 Protein
11.
Sci Rep ; 11(1): 19223, 2021 09 28.
Article En | MEDLINE | ID: mdl-34584169

Respiratory syncytial virus (RSV) is the primary cause of serious lower respiratory tract disease in infants, young children, the elderly and immunocompromised individuals. Therapy for RSV infections is limited to high risk infants and there are no safe and efficacious vaccines. Matrix (M) protein is a major RSV structural protein with a key role in virus assembly. Interestingly, M is localised to the nucleus early in infection and its export into the cytoplasm by the nuclear exporter, exportin-1 (XPO1) is essential for RSV assembly. We have shown previously that chemical inhibition of XPO1 function results in reduced RSV replication. In this study, we have investigated the anti-RSV efficacy of Selective Inhibitor of Nuclear Export (SINE) compounds, KPT-335 and KPT-185. Our data shows that therapeutic administration of the SINE compounds results in reduced RSV titre in human respiratory epithelial cell culture. Within 24 h of treatment, RSV replication and XPO1 expression was reduced, M protein was partially retained in the nucleus, and cell cycle progression was delayed. Notably, the effect of SINE compounds was reversible within 24 h after their removal. Our data show that reversible inhibition of XPO1 can disrupt RSV replication by affecting downstream pathways regulated by the nuclear exporter.


Acrylates/pharmacology , Karyopherins/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Respiratory Syncytial Virus Infections/drug therapy , Triazoles/pharmacology , Viral Matrix Proteins/metabolism , Virus Replication/drug effects , A549 Cells , Acrylates/therapeutic use , Cell Nucleus/metabolism , Drug Evaluation, Preclinical , Humans , Karyopherins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/metabolism , Triazoles/therapeutic use , Exportin 1 Protein
12.
Am J Hum Genet ; 108(9): 1669-1691, 2021 09 02.
Article En | MEDLINE | ID: mdl-34314705

Transportin-2 (TNPO2) mediates multiple pathways including non-classical nucleocytoplasmic shuttling of >60 cargoes, such as developmental and neuronal proteins. We identified 15 individuals carrying de novo coding variants in TNPO2 who presented with global developmental delay (GDD), dysmorphic features, ophthalmologic abnormalities, and neurological features. To assess the nature of these variants, functional studies were performed in Drosophila. We found that fly dTnpo (orthologous to TNPO2) is expressed in a subset of neurons. dTnpo is critical for neuronal maintenance and function as downregulating dTnpo in mature neurons using RNAi disrupts neuronal activity and survival. Altering the activity and expression of dTnpo using mutant alleles or RNAi causes developmental defects, including eye and wing deformities and lethality. These effects are dosage dependent as more severe phenotypes are associated with stronger dTnpo loss. Interestingly, similar phenotypes are observed with dTnpo upregulation and ectopic expression of TNPO2, showing that loss and gain of Transportin activity causes developmental defects. Further, proband-associated variants can cause more or less severe developmental abnormalities compared to wild-type TNPO2 when ectopically expressed. The impact of the variants tested seems to correlate with their position within the protein. Specifically, those that fall within the RAN binding domain cause more severe toxicity and those in the acidic loop are less toxic. Variants within the cargo binding domain show tissue-dependent effects. In summary, dTnpo is an essential gene in flies during development and in neurons. Further, proband-associated de novo variants within TNPO2 disrupt the function of the encoded protein. Hence, TNPO2 variants are causative for neurodevelopmental abnormalities.


Developmental Disabilities/genetics , Drosophila Proteins/genetics , Eye Diseases, Hereditary/genetics , Intellectual Disability/genetics , Karyopherins/genetics , Musculoskeletal Abnormalities/genetics , beta Karyopherins/genetics , ran GTP-Binding Protein/genetics , Alleles , Amino Acid Sequence , Animals , Developmental Disabilities/metabolism , Developmental Disabilities/pathology , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Eye Diseases, Hereditary/metabolism , Eye Diseases, Hereditary/pathology , Female , Gene Dosage , Gene Expression Regulation, Developmental , Genome, Human , Humans , Infant , Infant, Newborn , Intellectual Disability/metabolism , Intellectual Disability/pathology , Karyopherins/antagonists & inhibitors , Karyopherins/metabolism , Male , Musculoskeletal Abnormalities/metabolism , Musculoskeletal Abnormalities/pathology , Mutation , Neurons/metabolism , Neurons/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Whole Genome Sequencing , beta Karyopherins/metabolism , ran GTP-Binding Protein/metabolism
13.
Antiviral Res ; 192: 105115, 2021 08.
Article En | MEDLINE | ID: mdl-34157321

The novel coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the recent global pandemic. The nuclear export protein (XPO1) has a direct role in the export of SARS-CoV proteins including ORF3b, ORF9b, and nucleocapsid. Inhibition of XPO1 induces anti-inflammatory, anti-viral, and antioxidant pathways. Selinexor is an FDA-approved XPO1 inhibitor. Through bioinformatics analysis, we predicted nuclear export sequences in the ACE-2 protein and confirmed by in vitro testing that inhibition of XPO1 with selinexor induces nuclear localization of ACE-2. Administration of selinexor inhibited viral infection prophylactically as well as therapeutically in vitro. In a ferret model of COVID-19, selinexor treatment reduced viral load in the lungs and protected against tissue damage in the nasal turbinates and lungs in vivo. Our studies demonstrated that selinexor downregulated the pro-inflammatory cytokines IL-1ß, IL-6, IL-10, IFN-γ, TNF-α, and GMCSF, commonly associated with the cytokine storm observed in COVID-19 patients. Our findings indicate that nuclear export is critical for SARS-CoV-2 infection and for COVID-19 pathology and suggest that inhibition of XPO1 by selinexor could be a viable anti-viral treatment option.


COVID-19 Drug Treatment , Hydrazines/pharmacology , SARS-CoV-2/drug effects , Triazoles/pharmacology , Active Transport, Cell Nucleus/drug effects , Angiotensin-Converting Enzyme 2/metabolism , Animals , Antiviral Agents/pharmacology , COVID-19/virology , Chlorocebus aethiops , Cytokines , Ferrets , Humans , Karyopherins/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Respiratory System/drug effects , Respiratory System/virology , SARS-CoV-2/metabolism , Tumor Suppressor Proteins/metabolism , Vero Cells , Virus Replication , Exportin 1 Protein
14.
J Hematol Oncol ; 14(1): 97, 2021 06 24.
Article En | MEDLINE | ID: mdl-34167562

BACKGROUND: T cell acute lymphoblastic leukemia (T-ALL) is a high-risk subtype that comprises 10-15% of childhood and 20-25% of adult ALL cases. Over 70% of T-ALL patients harbor activating mutations in the NOTCH1 signaling pathway and are predicted to be sensitive to gamma-secretase inhibitors. We have recently demonstrated that selective inhibition of PSEN1-containing gamma-secretase complexes can overcome the dose-limiting toxicity associated with broad gamma-secretase inhibitors. In this study, we developed combination treatment strategies with the PSEN1-selective gamma-secretase inhibitor MRK-560 and other targeted agents (kinase inhibitors ruxolitinib and imatinib; XPO-1 inhibitor KPT-8602/eltanexor) for the treatment of T-ALL. METHODS: We treated T-ALL cell lines in vitro and T-ALL patient-derived xenograft (PDX) models in vivo with MRK-560 alone or in combination with other targeted inhibitors (ruxolitinib, imatinib or KPT-8602/eltanexor). We determined effects on proliferation of the cell lines and leukemia development and survival in the PDX models. RESULTS: All NOTCH1-signaling-dependent T-ALL cell lines were sensitive to MRK-560 and its combination with ruxolitinib or imatinib in JAK1- or ABL1-dependent cell lines synergistically inhibited leukemia proliferation. We also observed strong synergy between MRK-560 and KPT-8602 (eltanexor) in all NOTCH1-dependent T-ALL cell lines. Such synergy was also observed in vivo in a variety of T-ALL PDX models with NOTCH1 or FBXW7 mutations. Combination treatment significantly reduced leukemic infiltration in vivo and resulted in a survival benefit when compared to single treatment groups. We did not observe weight loss or goblet cell hyperplasia in single drug or combination treated mice when compared to control. CONCLUSIONS: These data demonstrate that the antileukemic effect of PSEN1-selective gamma-secretase inhibition can be synergistically enhanced by the addition of other targeted inhibitors. The combination of MRK-560 with KPT-8602 is a highly effective treatment combination, which circumvents the need for the identification of additional mutations and provides a clear survival benefit in vivo. These promising preclinical data warrant further development of combination treatment strategies for T-ALL based on PSEN1-selective gamma-secretase inhibition.


Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Karyopherins/antagonists & inhibitors , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Imatinib Mesylate/therapeutic use , Mice , Molecular Targeted Therapy , Nitriles/therapeutic use , Presenilin-1/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Exportin 1 Protein
15.
J Med Chem ; 64(10): 6596-6607, 2021 05 27.
Article En | MEDLINE | ID: mdl-33974430

Nuclear export factor chromosome region maintenance 1 (CRM1) is an attractive anticancer and antiviral drug target that spurred several research efforts to develop its inhibitor. Noncovalent CRM1 inhibitors are desirable, but none is reported to date. Here, we present the crystal structure of yeast CRM1 in complex with S109, a substructure of CBS9106 (under clinical test). Superimposition with the LFS-829 (another covalent CRM1 inhibitor) complex inspired the design of a noncovalent CRM1 inhibitor. Among nine synthesized compounds, noncovalent CRM1 inhibitor 1 (NCI-1) showed a high affinity to human and yeast CRM1 in the absence or presence of GST-bound Ras-related nuclear protein (RanGTP). Unlike covalent inhibitors, the crystal structure showed that NCI-1 is bound in the "open" nuclear export signal (NES) groove of CRM1, simultaneously occupying two hydrophobic pockets. NCI-1 additionally inhibited the nuclear export and proliferation of cells harboring the human CRM1-C528S mutant. Our work opens up the avenue of noncovalent CRM1 inhibitor development toward a more potent, less toxic, and broad-spectrum anticancer/antiviral therapy.


Drug Design , Fungal Proteins/antagonists & inhibitors , Karyopherins/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Small Molecule Libraries/chemistry , Active Transport, Cell Nucleus/drug effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Fungal Proteins/metabolism , Humans , Karyopherins/metabolism , Molecular Dynamics Simulation , Pyridines/chemistry , Pyridines/metabolism , Pyridines/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Saccharomyces cerevisiae/metabolism , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Exportin 1 Protein
16.
Traffic ; 22(7): 221-229, 2021 07.
Article En | MEDLINE | ID: mdl-34021516

Dysregulation of the nuclear export machinery mediated by chromosomal maintenance 1 (CRM1, also known as exportin-1), is closely associated with various human disorders, such as breast cancer. Previously, we identified sulforaphene and its synthetic analogues as covalent inhibitors of CRM1. Herein, we describe the discovery and biological evaluation of another sulforaphene synthetic analogue, LFS-31, as a potential CRM1 inhibitor. In addition, we investigated the reversible binding mechanism of LFS-31 with CRM1 through molecular simulations coupled with bio-layer interferometry (BLI) and found relatively high binding affinity (KD = 43.1 ± 35.3 nM) between the LFS-31 and CRM1 groups. We found that LFS-31 exhibited a stronger growth suppression of triple-negative breast cancer (TNBC) cells than non-TNBC cells, and had minimal effect on normal breast cells. Pharmacological treatment of TNBC cells with LFS-31 at nanomolar concentrations led to the nuclear retention of IkBα resulting in strong suppression of NF-κB transcriptional activity and attenuated cell growth and proliferation, which collectively contributed to the antitumor responses. To the best of our knowledge, this is the first study to demonstrate the use of a sulforaphene analogue as a potent CRM1 inhibitor that targets the NF-κB signaling pathway for the targeted therapy of TNBC.


Karyopherins/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Triple Negative Breast Neoplasms , Active Transport, Cell Nucleus , Cell Line, Tumor , Cell Proliferation , Humans , NF-kappa B/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/drug therapy , Exportin 1 Protein
17.
J Nat Prod ; 84(5): 1478-1488, 2021 05 28.
Article En | MEDLINE | ID: mdl-33890470

The protein chromosome region maintenance 1 (CRM1) is an important nuclear export factor and drug target in diseases such as cancer and viral infections. Several plant-derived CRM1 inhibitors including plumbagin and oridonin possess potent antitumor activities. However, their modes of CRM1 inhibition remain unclear. Here, a multimutant CRM1 was engineered to enable crystallization of these two small molecules in its NES groove. Plumbagin and oridonin share the same three conjugation sites in CRM1. In solution, these two inhibitors targeted more CRM1 sites and inhibited its activity through promoting its aggregation, in addition to directly targeting the NES groove. While the plumbagin-bound NES groove resembles the NES-bound groove state, the oridonin complex reveals for the first time a more open NES groove. The observed greater NES groove dynamics may improve cargo loading through a "capture-and-tighten" mechanism. This work thus provides new insights on the mechanism of CRM1 inhibition by two natural products and a structural basis for further development of these or other CRM1 inhibitors.


Diterpenes, Kaurane/pharmacology , Karyopherins/antagonists & inhibitors , Naphthoquinones/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Amino Acid Sequence , Molecular Structure , Mutant Proteins/antagonists & inhibitors , Protein Engineering , Protein Structure, Tertiary , Exportin 1 Protein
18.
Int J Nanomedicine ; 16: 2833-2847, 2021.
Article En | MEDLINE | ID: mdl-33883894

INTRODUCTION: Peptides can be rationally designed as non-covalent inhibitors for molecularly targeted therapy. However, it remains challenging to efficiently deliver the peptides into the targeted cells, which often severely affects their therapeutic efficiency. METHODS: Herein, we created a novel non-covalent peptide inhibitor against nuclear export factor CRM1 by a structure-guided drug design method and targetedly delivered the peptide into cancer cells by a nanoparticle-mediated gene expression system for use as a cancer therapy. RESULTS: The nuclear export signal (NES)-optimized CRM1 peptide inhibitor colocalized with CRM1 to the nuclear envelope and inhibited nuclear export in cancer cell lines in vitro. The crystal structures of the inhibitors complexed with CRM1 were solved. In contrast to the covalent inhibitors, the peptides were similarly effective against cells harboring the CRM1 C528S mutation. Moreover, a plasmid encoding the peptides was delivered by a iRGD-modified nanoparticle to efficiently target and transfect the cancer cells in vivo after intravenous administration. The peptides could be selectively expressed in the tumor, resulting in the efficient inhibition of subcutaneous melanoma xenografts without obvious systemic toxicity. DISCUSSION: This work provides an effective strategy to design peptide-based molecularly targeted therapeutics, which could lead to the development of future targeted therapy.


Intracellular Space/metabolism , Karyopherins/antagonists & inhibitors , Melanoma, Experimental/drug therapy , Nanoparticles/chemistry , Peptides/pharmacology , Peptides/therapeutic use , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , A549 Cells , Active Transport, Cell Nucleus/drug effects , Amino Acid Sequence , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Gene Transfer Techniques , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Karyopherins/chemistry , Karyopherins/metabolism , Melanoma, Experimental/pathology , Mutant Proteins/metabolism , Mutation/genetics , Nanoparticles/ultrastructure , Nuclear Export Signals , Peptides/chemistry , Protein Binding/drug effects , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Viral Nonstructural Proteins/chemistry , Exportin 1 Protein
19.
Chembiochem ; 22(12): 2116-2123, 2021 06 15.
Article En | MEDLINE | ID: mdl-33887086

Selinexor, a covalent XPO1 inhibitor, is approved in the USA in combination with dexamethasone for penta-refractory multiple myeloma. Additional XPO1 covalent inhibitors are currently in clinical trials for multiple diseases including hematologic malignancies, solid tumor malignancies, glioblastoma multiforme (GBM), and amyotrophic lateral sclerosis (ALS). It is important to measure the target engagement and selectivity of covalent inhibitors to understand the degree of engagement needed for efficacy, while avoiding both mechanism-based and off-target toxicity. Herein, we report clickable probes based on the XPO1 inhibitors selinexor and eltanexor for the labeling of XPO1 in live cells to assess target engagement and selectivity. We used mass spectrometry-based chemoproteomic workflows to profile the proteome-wide selectivity of selinexor and eltanexor and show that they are highly selective for XPO1. Thermal profiling analysis of selinexor further offers an orthogonal approach to measure XPO1 engagement in live cells. We believe these probes and assays will serve as useful tools to further interrogate the biology of XPO1 and its inhibition in cellular and in vivo systems.


Amyotrophic Lateral Sclerosis/drug therapy , Antineoplastic Agents/pharmacology , Glioblastoma/drug therapy , Hydrazines/pharmacology , Karyopherins/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Triazoles/pharmacology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Hydrazines/chemistry , Karyopherins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Triazoles/chemistry , Exportin 1 Protein
20.
Invest New Drugs ; 39(5): 1357-1365, 2021 10.
Article En | MEDLINE | ID: mdl-33909232

Background Selinexor, a first-in-class, oral selective inhibitor of nuclear export (SINE) compound inhibits Exportin-1(XPO1), had demonstrated synergistic activity with many chemotherapies and conferred in vivo antitumor efficacy in hematologic as well as solid tumors. Methods This open-label, single-center, multi-arm phase 1b study used a standard 3 + 3 design and a "basket type" expansion. Selinexor with intravenous topotecan was given in one of the 13 parallel arms. Patients with advanced or metastatic relapsed/refractory solid tumors following prior systemic therapy, or in whom the addition of selinexor to standard chemotherapy deemed appropriate, were eligible. Results Fourteen patients with the median age of 61 years (range, 22-68years) were treated, and the most common cancer types were gynecological cancers; ovarian (n = 5), endometrial (n = 2), and 1 each with fallopian tube and vaginal cancers. Of the 14 patients treated, 12 (86 %) had at least one treatment-related adverse event (TRAE). The most common TRAEs were anemia (71 %), thrombocytopenia (57 %), hyponatremia (57 %), vomiting (57 %), fatigue (50 %), nausea (50 %), and neutropenia (36 %). Two patients had dose limiting toxicities. One patient dosed at selinexor 80 mg had grade 3 nausea and vomiting and one patient dosed at selinexor 60 mg experienced grade 4 neutropenia and thrombocytopenia. Of the 13 efficacy evaluable patients, one (8 %) with endometrial cancer achieved unconfirmed partial response (uPR) and the time-to-treatment failure (TTF) was 48 weeks, whereas 6 of the 13 (46 %) patients had stable disease (SD) contributing to the clinical benefit rate of 46 %. The median TTF for all patients was 9 weeks (range, 2-48weeks). Conclusions Once weekly selinexor in combination with topotecan was viable and showed some preliminary tumor efficacy. The recommend phase 2 dose of selinexor was 60 mg once weekly in combination with IV topotecan.Trial registration: NCT02419495. Registered 14 April 2015, https://clinicaltrials.gov/ct2/show/NCT02419495.


Hydrazines/therapeutic use , Karyopherins/antagonists & inhibitors , Neoplasms/drug therapy , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Triazoles/therapeutic use , Active Transport, Cell Nucleus/drug effects , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dose-Response Relationship, Drug , Female , Humans , Hydrazines/administration & dosage , Hydrazines/adverse effects , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/pathology , Topotecan/therapeutic use , Triazoles/administration & dosage , Triazoles/adverse effects , Exportin 1 Protein
...