Skip to main content
Clinical and Translational Medicine logoLink to Clinical and Translational Medicine
. 2024 May 23;14(5):e1684. doi: 10.1002/ctm2.1684

The nuclear export protein exportin‐1 in solid malignant tumours: From biology to clinical trials

Chuanxi Lai 1,2, Lingna Xu 1,2,, Sheng Dai 1,2,
PMCID: PMC11116501  PMID: 38783482

Abstract

Background

Exportin‐1 (XPO1), a crucial protein regulating nuclear‐cytoplasmic transport, is frequently overexpressed in various cancers, driving tumor progression and drug resistance. This makes XPO1 an attractive therapeutic target. Over the past few decades, the number of available nuclear export‐selective inhibitors has been increasing. Only KPT‐330 (selinexor) has been successfully used for treating haematological malignancies, and KPT‐8602 (eltanexor) has been used for treating haematologic tumours in clinical trials. However, the use of nuclear export‐selective inhibitors for the inhibition of XPO1 expression has yet to be thoroughly investigated in clinical studies and therapeutic outcomes for solid tumours.

Methods

We collected numerous literatures to explain the efficacy of XPO1 Inhibitors in preclinical and clinical studies of a wide range of solid tumours.

Results

In this review, we focus on the nuclear export function of XPO1 and results from clinical trials of its inhibitors in solid malignant tumours. We summarized the mechanism of action and therapeutic potential of XPO1 inhibitors, as well as adverse effects and response biomarkers.

Conclusion

XPO1 inhibition has emerged as a promising therapeutic strategy in the fight against cancer, offering a novel approach to targeting tumorigenic processes and overcoming drug resistance. SINE compounds have demonstrated efficacy in a wide range of solid tumours, and ongoing research is focused on optimizing their use, identifying response biomarkers, and developing effective combination therapies.

Key Points

  • Exportin‐1 (XPO1) plays a critical role in mediating nucleocytoplasmic transport and cell cycle.

  • XPO1 dysfunction promotes tumourigenesis and drug resistance within solid tumours.

  • The therapeutic potential and ongoing researches on XPO1 inhibitors in the treatment of solid tumours.

  • Additional researches are essential to address safety concerns and identify biomarkers for predicting patient response to XPO1 inhibitors.

Keywords: cancer, drug resistance, XPO1, XPO1 inhibitor


Exportin‐1 (XPO1), a key protein for nuclear export (proteins and RNAs) and cell cycle regulation, plays a significant role in the development of solid tumours and the emergence of drug resistance. While XPO1 inhibitors demonstrate promising therapeutic potential in preclinical and clinical studies for solid tumours, treatment safety and biomarkers of response require further exploration.

graphic file with name CTM2-14-e1684-g003.jpg

1. INTRODUCTION

The nuclear envelope separates eukaryotic cells into distinct nuclear and cytoplasmic compartments. 1 The nuclear pore complex (NPC) serves as an essential conduit, facilitating the exchange of substantial quantities of protein and RNA cargoes between the nucleus and the cytoplasm, a critical process for maintaining optimal cellular function. 2 Smaller molecules such as ions or metabolites can rapidly diffuse through the NPC, while larger entities such as proteins and RNA molecules are restricted and move less efficiently unless they assist a member of the karyopherin (Kap) β family of transport proteins. 3 To date, more than 20 human Kaps have been identified. Although the functions of two isoforms of Kaps are unknown, 18 isoforms of Kaps are known to play crucial roles in nuclear transport, including import, export and biportin functions. 4 , 5 Kaps act as carrier molecules that facilitate cargo protein or RNA import into and export out of the nucleus via the NPC by recognising either a nuclear localisation signal (NLS) or a nuclear export signal (NES). 6 Four classes of NLSs and one class of NES have been identified, including (i) the classic basic amino acid NLS sequences recognised by a heterodimer composed of importins α and β; (ii) the proline‐tyrosine NLS that binds KAPβ2 or KAPβ2b; (iii) the isoleucine‐lysine NLS that binds yeast Kap121; (iv) the transportin 3‐binding arginine‐serine repeat NLS found in RS domains; and (v) a hydrophobic leucine‐rich NES recognised by exportin‐1 (XPO1). 5 , 6

XPO1 plays an essential role as an exportin in transporting a diverse array of cargoes, including RNAs, tumour suppressor proteins and cell cycle regulators, such as p53, adenomatous polyposis coli (APC), retinoblastoma (RB), p21, p27, forkhead box protein O (FOXO), signal transducer and activator of transcription 3 (STAT3), inhibitor of nuclear factor kappa B (IκB) and breast cancer susceptibility gene 1 (BRCA1). 7 , 8 , 9 XPO1 is overexpressed in numerous cancer tissues, and its overexpression is accompanied by disease progression, therapeutic resistance and decreased overall survival (OS) or progression‐free survival (PFS). Consequently, the potential of XPO1 as a therapeutic target for anticancer treatments has been widely researched over the past few decades. 7 , 8 , 9

Selective inhibitor of nuclear export (SINE) compounds, which inhibit the expression of XPO1, represent a groundbreaking class of anticancer agents. Moreover, these agents are highly valued for their antitumour effectiveness in patients with various pretreated haematological malignancies and solid tumours. 10 , 11 Selinexor (an inhibitor of XPO1), the most prominent member of the SINE compound group, has been extensively evaluated in phase I and II clinical trials for various cancers. In addition, selinexor has achieved Food and Drug Administration(FDA) approval for the treatment of multiple myeloma (MM) and relapsed or refractory diffuse large B‐cell tumours. 12 , 13 Despite its primary application in treating haematologic malignancies, a plethora of preclinical studies and clinical trials suggest that selinexor also holds significant promise for the treatment of solid tumours. In this review, we provide a comprehensive introduction of the function of XPO1 and the role of XPO1 inhibitors in managing solid malignant tumours.

2. PHYSIOLOGICAL FUNCTIONS OF XPO1

XPO1 was first isolated from Schizosaccharomyces pombe and was originally named chromosomal region maintenance 1 (CRM1); this gene maintains high‐order chromosomal structure. 14 Subsequent studies revealed that CRM1 in S. cerevisiae mediates the nuclear export of proteins and mRNAs, and this gene was therefore renamed XPO1. 15 As a member of the Kapβ family of transport factors, XPO1 has been identified as a primary nuclear export protein that accepts leucine‐rich NESs, which can be found in many proteins as well as RNAs, and then transports them to the cytoplasm via the support of the small guanosine triphosphate (GTPase) Ran. 16 , 17 , 18 , 19 , 20 The concentration of the GTPase Ran determines the direction of nucleoplasmic transport. Ran guanine exchange factor, also called regulator of chromosome condensation 1 (RCC1), GTPase‐activating protein (RanGAP) and RanGTP‐binding protein 1/2 (RanBP1/2), plays important roles in maintaining the differential concentrations of Ran in the nucleus and cytoplasm. In the nucleus, RCC1 converts RanGDP to RanGTP, while in the cytoplasm, RanGAP and RanBP1/2 convert RanGTP to RanGDP. 21 XPO1 binds cargo proteins/RNAs and RanGTP, assembling a stable ternary export complex that is collaboratively exported into the cytoplasm through the NPC. Once the complex is exported into the cytoplasm, it is disassembled via RanGAP, which promotes the dephosphorylation of RanGTP to RanGDP. 22 , 23 Interestingly, XPO1 is then recycled to the nucleus for subsequent transport. 24

2.1. Structure of XPO1

Native full‐length human XPO1 comprises 1071 amino acids and has 20 HEATs. 25 , 26 The canonical HEAT repeat is composed of two antiparallel helices, A and B, with approximately 40−50 residues, while H20, as a C‐terminal region, has three helices, H20', H20A and H20B. 25 With the use of a combination of X‐ray crystallography, homology modelling and electron microscopy, the understanding of XPO1 function has improved. 27 H1‐H3, the CRIME domain (CRM1, importin β, etc.), which shares homology with importin β, is an N‐terminal region that participates in RanGTP binding. 28 Interestingly, NES cargos bind the outer surface of XPO1 in a bidirectional manner and interact with H11‐H12 and H13‐H14, two NES epitopes. 17 , 25 , 29 XPO1 is known to recognise leucine‐rich NES proteins that bear a specific short peptide sequence comprising 10 ordered amino acid residues, defined as Φ1 × 2‐3Φ2 × 2‐3‐Φ3XΦ4. Here, Φ signifies isoleucine, leucine, methionine, phenylalanine or valine, and X represents any other amino acid. 30 Interestingly, the location of XPO1 Cys528 within the leucine‐rich NES binding groove indicates its potential as a target for inhibiting the formation of export complexes. 31 Leptomycin B (LMB) inhibits nuclear export by modifying the Cys528 residue of XPO1. 32 A large‐scale analysis of 322 cancers revealed three hotspots of high‐recurrence mutations in XPO1, namely, E571, D624 and R749. 33 E571K is the most common mutation of XPO1. The Q626 mutation of XPO1 is second only to the E571 mutation in lung cancer, which is associated with poor prognosis. 34 Moreover, the E571K mutation in XPO1 has been identified as a key factor in cancer development because it augments the affinity between XPO1 and negatively charged C‐terminal NESs to promote nuclear export (Figure 1). 35

FIGURE 1.

FIGURE 1

(A) Schematic structure of exportin‐1 (XPO1). Boxes 1−20 represent the HEAT repeat motifs. The CRIME domain (XPO1, importin β, etc.), which corresponds to HEAT repeats 1−3, shares homology with importin β and helps recognise RanGTP. H11‐12 and H13‐14 have two NES epitopes that can bind NES cargoes. However, Cys528 is modified by leptomycin B (LMB) and targets the central region of the XPO1 NES‐binding cleft. Moreover, a mutation hotspot (E571K) plays a role in oncogenic processes. (B) Schematic diagram of the physiological activities of XPO1. In terms of nuclear‐cytoplasmic transport, XPO1 and RanGDP undergo nuclear‐cytoplasmic transport via Ran guanine exchange factor (RanGEP), where they form XPO1‐RanGTP and further bind to cargo proteins. The substances involved in this translocation are generally proteins and RNAs, and the proteins can include tumour suppressors, cell cycle‐related proteins and oncogenes. When cargo is transported to the cytoplasm, the XPO1‐RanGTP complex and cargo are redissolved into XPO1, RanGDP and cargo (protein or RNA) with the help of GTPase‐activating protein (RanGAP) and RanGTP‐binding protein 1/2 (RanBP1/2). Among these components, XPO1 re‐enters the nucleus. 36 (C) Schematic diagram of XPO1‐mediated RNAs nuclear export.

2.2. Functions of XPO1

XPO1, the predominant protein of the Kap protein family, plays a critical role in shuttling numerous leucine‐rich NES proteins and RNAs from the nucleus, requiring the help of the small GTPases Ran and GTP to form a complex to promote importin‐RanGTP complex transfer to the cytoplasm. 20 In addition to its significant role in nuclear‐cytoplasmic transport, XPO1 also plays an important role in the regulation of mitosis, specifically during the interphase of the cell cycle. 37

2.2.1. Protein export

XPO1 is essential for exporting NES proteins into the cytoplasm. Rev and the protein kinase inhibitor cyclic adenosine monophosphate (cAMP)‐dependent protein kinase were among the first proteins identified as NESs. 29 , 38 To date, the new database ValidNESs (http://prodata.swmed.edu/LRNes) shows that more than 221 proteins with diverse functions rely on XPO1 transport into the cytoplasm. 39 These proteins, including cell cycle regulators (p21, cyclin B1, D1), tumour suppressor proteins (BRCA1/2, p53, APC, Rb, cellular‐abelsongene (c‐ABL)), antiapoptotic proteins (survivin) and autophagy‐related proteins (beclin‐1 and yes‐associated protein 1), are essential for regulating normal cellular physiological activities. 40 , 41 , 42 , 43 , 44 , 45 To date, extensive studies have been conducted on the proteins exported by XPO1. Further investigation of the cargo spectrum of XPO1 revealed additional functions involved in the spatial regulation of vesicle coat assembly, centrosomes, autophagy, peroxisome biogenesis, the cytoskeleton, ribosome maturation, translation and mRNA degradation, which suggests that protein export via XPO1 is a widespread and intricate process. 46 The diverse functions of XPO1 indicate its potential as a therapeutic target in various diseases.

2.2.2. RNA export

XPO1 was initially shown to export mRNA from the nucleus to the cytoplasm through its interaction with the RNA‐binding protein Rev. 38 Subsequent evidence has demonstrated the vital function of XPO1 in the nuclear export of various types of RNA, such as ribosomal RNAs (rRNAs), uridine‐rich small nuclear RNAs (U snRNAs), specific mRNAs, miRNAs and transfer RNA (tRNAs). 18 , 47 , 48 XPO1 is involved in the synthesis and processing of rRNA, in which it exports the pre‐40S and pre‐60S ribosomal subunits to the cytoplasm; this process is followed by splicing and translation. 49 , 50 , 51 , 52 An important aspect of the function of XPO1 is its regulation of heterogeneous nRNA (pre‐mRNA) splicing through its influence on the maturation of snRNAs. 53 U snRNAs are transcribed in the nucleus and form complexes with phosphorylated adaptors for RNA export (PHAX), RanGTP, XPO1 and the cap‐binding complex. The complex is then transferred to the cytoplasm for further assembly and modification, where it eventually forms U snRNPs that re‐enter the nucleus for complete maturation and participation in splicing functions. 38 , 54 , 55 , 56 , 57 , 58 Although genome‐wide screens in Drosophila spp. have suggested that bulk mRNAs are exported through nuclear RNA export factor 1(NXF1)/TAP, which are expressed in all tissue types, subsequent research has demonstrated that XPO1 is involved in exporting a subset of specialised mRNAs in both humans and yeast. 59 , 60 , 61 Additionally, research has revealed interactions between XPO1 and NXF3, a member of the NXF family, through a NES, which mediates the export of several mRNAs. 62 XPO1 interacts with other adaptor proteins, including leucine‐rich pentatricopeptide repeat protein), eukaryotic translation initiator factor 4E (eIF4E) and HuR (with the help of APRIL and pp32), and facilitates the export of specific mRNAs. 63 Moreover, XPO1 also plays a role in the export of several alternative small noncoding RNAs, including microRNAs and tRNAs. 61 Most miRNAs are transcribed and possess cap structures as well as poly(A) tails to form primary miRNAs via RNA polymerase II in the nucleus. 64 Long pri‐miRNAs are further processed into hairpin‐shaped precursor miRNAs (pre‐miRNAs) by a microprocessor that includes the double‐stranded RNase III enzyme DROSHA and its necessary cofactors, double‐stranded RNA‐binding protein DiGeorge syndrome critical region 8. 65 , 66 , 67 Previous studies have shown that XPO5 mediates the nuclear transport of pre‐miRNAs. 68 , 69 Subsequent studies have demonstrated that XPO1 selectively transports a novel class of (m7G)‐capped pre‐miRNAs. 70 , 71

2.2.3. Other functions of XPO1

An increasing body of evidence has demonstrated the critical role of XPO1 not only in mediating nucleocytoplasmic transport, cell division and the cell cycle but also in spindle formation. 72 XPO1, located on kinetochores, has been found to stabilise microtubule movement and support chromosome segregation by forming a complex with RanGAP1 and RanBP2. 73 , 74 , 75 During the cell cycle, XPO1 is present at the centrosome, where it assists in the recruitment of pericentriolar proteins as well as in spindle localisation. 76 Abnormal expression of XPO1 is linked to cellular instability. Elevated XPO1 expression potentially accelerates the cell cycle, while decreased or absent XPO1 expression might disrupt the integrity of mitogenic granules and telomeres. 77 , 78 Furthermore, XPO1 may perform a regulatory function in early embryonic development, highlighting its broad influence on cellular processes. 79

3. ROLES OF XPO1 IN CANCER

XPO1 is a critical nuclear export protein that plays a vital role in sustaining normal cellular physiological functions. 80 Intriguingly, XPO1 expression is markedly increased in a wide array of tumour cells, including osteosarcoma, glioma, lung cancer, ovarian cancer, pancreatic cancer, oesophageal cancer, gastric cancer and colorectal cancer (CRC) cells. The upregulation of XPO1 expression disrupts the balance of nucleoplasmic transport and leads to the mislocalisation of tumour suppressor proteins and oncoproteins (Table 1), which consequently promotes tumour progression and potentially fosters therapeutic resistance. 81 , 82 , 83 , 84 , 85 , 86 , 87 , 88 Despite its recognised significance, the mechanism underlying the overexpression of XPO1 in numerous tumours has not been elucidated. Accumulating evidence suggests a correlation between the aberrant nucleocytoplasmic transport and elevated XPO1 expression in tumours, leading to the occurrence and development of several cancers. 22 The upregulation of XPO1 allows key cellular proteins, such as p27Kip1 and p53, to be transported into the cytoplasm and inactivates inappropriate cell growth or DNA repair. 89 Similarly, oncogenes such as STAT3 and c‐myc activate downstream signalling pathways to stimulate proliferation and inhibit apoptosis upon export. 89 Inhibition of XPO1 promotes the nuclear accumulation of TSPs and oncogenes and promotes the apoptosis of tumour cells. 90 Several studies have shown that inhibiting XPO1 induces tumour cell apoptosis by triggering intranuclear accumulation of p53 or p27. 91 , 92 STAT3 phosphorylation or acetylation activates downstream growth signals, promotes cell proliferation and prevents apoptosis. 93 The CREB‐binding protein (CBP) induces acetylation of STAT3 to regulate transcriptional activity. 94 XPO1 inhibition represses STAT3 activation by affecting CBP activity to reduce the level of STAT3 acetylation, which blocks the interaction between STAT3 and the survivin promoter. 95 C‐myc plays a key role as a regulator in a variety of tumours. 96 mTOR‐dependent 4EBP1 phosphorylation has been shown to be a key factor by which myc maintains tumour survival. 97 eIF4E, a translation regulator downstream of mTOR, plays an important role in myc transcriptional regulation. 98 KPT‐330 inhibits the nuclear export of c‐myc mRNA by inducing the nuclear accumulation of XPO1 and eIF4E complexes. 99 Subsequent studies have shown that inhibition of XPO1 also regulates c‐myc expression through other pathways, such as the upregulation of miR‐145. 100 Therefore, inhibition of XPO1 may constitute a new strategy for tumour therapy (Figure 2).

TABLE 1.

Exportin‐1 (XPO1) mediates the nuclear export of cancer‐related cargoes.

Category Cargo Biological process Related cancer
Tumour suppressor proteins APC Wnt signaling pathway Colorectal cancer (CRC) 261
p21 Cip1 Cell cycle Breast cancer, chronic myeloid leukaemia
p53 Apoptosis, cell cycle, DNA damage, DNA repair Ovarian cancer, breast cancer, lung cancer, gastric cancer, gallbladder cancer, neuroblastoma, CRC 8 , 111 , 201 , 262 , 263 , 264 , 265
p27 Kip1  Cell cycle Cholangiocarcinoma, osteosarcoma 266 , 267
RB  Transcription factor Lung cancer, cervical cancer 90 , 268
PALB2 DNA repair Breast cancer, pancreatic cancer 269 , 270
TET2 Cell cycle Small intestinal neuroendocrine tumours 271
FOXO Apoptosis, transcription regulation Breast, prostate, thyroid cancer, glioblastoma, melanoma 114 , 272
VDUP1  Tumour and metastasis suppressor Gastric cancer, cervical cancer, lung cancer 273
MLH1 Cell cycle, DNA damage, DNA repair CRCs 274
KLF6 Transcription regulation Prostate cancer, gastric cancer 275 , 276
RASSF2  Cell growth Lung cancer 277
TOB  TOB/BTG antiproliferative (APRO) protein family Breast cancer 278
Oncoproteins Cyclin D1 Cell cycle, cell division, DNA damage Prostate cancer, multiple myeloma (MM) 279 , 280
FAM21 Protein transport Pancreatic cancer 281
AKT1 Apoptosis, carbohydrate metabolism Thyroid cancer, breast cancer 133 , 282
SMAD4 Transcription regulation Pancreatic cancer, CRC 283 , 284
Anti‐apoptotic proteins Nucleophosmin DNA repair, cell cycle Acute myeloid, leukemia 285
Survivin Apoptosis, cell cycle Breast cancer, liposarcoma, prostate cancer, ovarian cancer 95 , 286 , 287
Transcription factors RUNX  Transcription factor Breast cancer, gastric cancer, skin squamous cell carcinoma, esophageal cancer, lung cancer, colon cancer, pancreatic cancer, prostate cancer 288 , 289
IκBα  Transcription factor MM, lung cancer, breast cancer, chronic lymphocytic leukemia, T‐cell lymphoma, ovarian cancer 192 , 290 , 291 , 292 , 293 , 294

Abbreviations: APC, adenomatous polyposis coli; FOXO, forkhead box protein O; IκBα, inhibitor of nuclear factor kappa B alpha.

FIGURE 2.

FIGURE 2

The mechanism of XPO1 in normal and malignant cells. In normal cells, XPO1 maintains normal nuclear export, while in tumour cells, increased XPO1 expression promotes nuclear export leading to a large number of mislocated proteins and RNAs in the cytoplasm. Inhibition of XPO1 induced nuclear accumulation of tumour suppressors, oncogenes and RNAs.

4. ROLES OF XPO1 IN DRUG RESISTANCE

Drug resistance is an inevitable problem in cancer treatment. Accumulating evidence suggests that XPO1 may mediate drug resistance in multiple tumour types by promoting the nuclear export of multiple drug targets. Protein mislocalisation via XPO1 is a significant contributor to tumour drug resistance. 101 We classified the following as several reasons for increased nucleoplasmic transport (Figure 3). High XPO1 expression often indicates poor survival and chemical resistance. 102 In breast cancer patients >40 years of age, cytoplasmic BRCA1 expression was inversely associated with metastasis‐free survival. 103 Inhibition of XPO1 sensitised hepatocellular carcinoma cells to sorafenib. 104 NESs on cargo proteins can be exposed through different modifications, including phosphorylation, dephosphorylation, acetylation, SUMOylation and ubiquitination, which induce export. 105 Abnormal activation of signalling pathways plays an important role in NES exposure. Overactivation of protein kinase B (AKT) induces FOXO phosphorylation and promotes XPO1‐mediated FOXO nucleoplasmic transport, which causes FOXO to lose its ability to induce apoptosis and transcriptional regulation. 106 Notably, the XPO1 E571K mutation was shown to have greater affinity for negatively charged C‐terminal NESs than for other NESs, which may lead to the mislocalisation of several proteins. The XPO1 E571K mutation promotes the accumulation of TRAF2 (tumour necrosis factor receptor‐associated factor 2) in the cytoplasm, which is negatively correlated with the survival of CRC patients. 33 , 107

FIGURE 3.

FIGURE 3

Roles of XPO1 in drug resistance. Cargo protein modification, XPO1 mutation or increased XPO1 expression promote nuclear export and induce tumour resistance.

4.1. Platinum‐based chemotherapeutic agents

Moreover, platinum resistance frequently impedes the effectiveness of chemotherapy, and numerous factors contribute to this resistance. Several studies have shown that increased expression of galectin‐3 (Gal‐3) decreases sensitivity to platinum chemotherapy drugs. 108 , 109 Gal‐3, a member of the β‐galactoside‐carbohydrate binding protein family, is associated with carcinogenesis and malignant potential in the majority of tumours. 110 In response to apoptotic stimuli, Gal‐3 translocates from the nucleus to the cytoplasm to prevent apoptosis. It has been reported that phosphorylated Gal‐3 at serine 6 enhances nuclear export and promotes antiapoptotic activity. Another study revealed that XPO1 expression was increased in cisplatin‐resistant ovarian cancer cells. 111 Breast cancer tumour cells exposed to LMB or cisplatin retain Gal‐3 in the nucleus, where it induces apoptosis. 108 KPT‐185 and KPT‐330 resensitise cisplatin‐resistant ovarian cancer cells to cisplatin by inhibiting XPO1 expression. 111 Thus, inhibition of XPO1 maintains the nuclear localisation of Gal‐3, inducing apoptosis in platinum‐resistant cells. 101 , 112 Subsequent studies revealed that FOXO1 and high mobility group box 1 (HMGB1), which participate in DNA repair and extracellular signalling, play important roles in platinum resistance. 113 , 114 One study showed that increased FOXO1 nuclear translocation decreased cisplatin sensitivity in non‐small cell lung cancer (NSCLC). 115 Similarly, cisplatin induces HMGB1 expression in tumour cells, which in turn suppresses cisplatin‐induced apoptosis. 116 The suppression of XPO1 expression by KPT‐330 triggers the nuclear relocation of FOXO1 and HMGB1, thereby overcoming platinum resistance in tumour cells. 113 , 114

4.2. Anthracycline drugs

Despite medical advancements, the mortality rate of head and neck squamous cell carcinoma (HNSCC) has remained consistent for several decades at approximately 45%. 117 This persistent statistic is largely attributable to the development of drug resistance in patients. Several studies have revealed that in more than 80% of patients with HNSCC, the transcription factor E2F7 is mislocalised to the cytoplasm. 118 , 119 E2F7 regulates the production of proteins that confer resistance to a class of chemotherapy drugs known as anthracyclines. One study showed that E2F7 expression was positively correlated with sphingosine kinase 1 (SPHK1) expression. 117 SPHK1 and SPHK2 (two sphingosine kinase isoforms) catalyse the adenosine triphosphate (ATP)‐dependent phosphorylation of sphingosine to form sphingosine 1‐phosphate (S1P), which is responsible for the regulation of a wide range of biological effects, including proliferation and survival. 120 The phorbol 12‐myristate 13‐acetate (PMA, a protein kinase C activator)‐induced translocation of SPHK1 to the plasma membrane significantly increased SPHK1 activity. 121 Several studies have suggested that anthracycline resistance in osteosarcoma may be related to increased expression of SPHK1. 122 , 123 Phenoxodiol blocks the activation of SPHK1, and anthracycline is then able to promote ceramide accumulation in osteosarcoma cells, mediate the activation of the apoptosis signal‐regulating kinase 1 (ASK1)/c‐Jun N‐terminal kinase (JNK) pathway and negatively regulate Akt activity, suggesting that the inhibition of SPHK1 may restore the sensitivity of osteosarcoma to anthracycline. 122 Similarly, E2F7‐induced doxorubicin resistance is mediated by SPHK1‐dependent AKT activation in squamous cell carcinomas. 117 Furthermore, KPT‐330 has been shown to mediate the nuclear retention of E2F7, leading to the repression of SPHK1, which in turn increases anthracycline sensitivity 119 These findings reveal some of the mechanisms underlying anthracycline resistance and provide new treatment strategies that warrant further clinical evaluation in patients with HNSCC and osteosarcoma.

4.3. Tamoxifen

Tamoxifen is one of the most potent endocrine therapeutic agents for treating estrogen receptor (ER)α [+] in breast cancer. However, the effectiveness of endocrine therapies has limitations, as evidenced by instances of tumour recurrence or metastasis in ERα (+) patients. 124 , 125 Tamoxifen resistance involves complex mechanisms, including loss of ER, increase in growth factor receptors (such as human epidermal growth factor receptor 2HER2) and aberrant activation of signalling pathways (such as phosphoinositide 3‐kinase (PI3K)/AKT signalling). 126 Recently, the protein interferon alpha inducible protein 27 (IFI27) was shown to reduce tumour cell sensitivity to tamoxifen by mediating the nuclear export of ERα via XPO1. 124 HER2 overexpression was associated with poor prognosis in patients treated with tamoxifen, mainly due to the activation of other survival signalling pathways (such as the PI3K/AKT signalling pathway). 127 The expression of XPO1 and HER‐2 is positively correlated, and inhibition of XPO1 induces HER‐2 nuclear accumulation. 87 , 128 The G‐protein‐coupled ER (GPER) is strongly associated with tamoxifen resistance in breast cancer. 129 A subsequent study revealed that tumour cells induced GPER nuclear export via an XPO1‐dependent pathway in cancer‐associated fibroblasts by activating the PI3K/AKT signalling pathway. 130 Several studies have shown that inhibiting the activation of the PI3K/AKT signalling pathway increases the sensitivity of breast cancer to tamoxifen. 131 , 132 In addition, tamoxifen treatment induced an increase in XPO1 expression, which may suggest that the subcellular localisation of key proteins is related to the tamoxifen treatment response. 125 One study showed that KPT‐330 enhanced the sensitivity of breast cancer cells to tamoxifen by inhibiting the nuclear export of ERK5. 125 However, there is a risk of tumour recurrence in patients treated with KPT‐330 alone, which warrants further exploration of the combination of KPT‐330 and tamoxifen. Interestingly, the combination of XPO1 and tamoxifen blocked the activation of the AKT signalling pathway, induced autophagy, improved the treatment sensitivity to tamoxifen and reshaped the tumour metabolic pathway, which provided a new approach for the future treatment of breast cancer. 133 , 134

4.4. Gefitinib

Lung cancer remains the leading cause of cancer‐related deaths worldwide. 135 NSCLC represents approximately 85% of all lung cancer cases. Currently, epidermal growth factor (EGF) receptor tyrosine kinase inhibitors (EGFR‐TKIs), such as gefitinib, are the primary treatment for advanced NSCLC patients with EGFR mutations. 136 Despite the initial efficacy of EGFR‐TKIs, approximately 65% of EGFR‐TKI‐sensitive NSCLC patients develop resistance to these medications within 9 to 13 months of treatment. 137 DEAD‐box helicase 17 (DDX17) is a multifunctional ATP‐dependent RNA/DNA helicase that is involved in the initiation and development of a variety of tumours. 138 In NSCLC, the upregulation of DDX17 is associated with increased gefitinib resistance, while the silencing of DDX17 can partially reverse this sensitivity. 139 One study showed that increased DDX17 expression promoted β‐catenin transcriptional activity. 140 As a member of the canonical Wnt pathway, β‐catenin participates in the regulation of cell proliferation through nuclear translocation. 141 Several studies have shown that gefitinib resistance is associated with increased β‐catenin transcriptional activity and that inhibition of β‐catenin partially restores gefitinib sensitivity. 142 , 143 Furthermore, blocking DDX17 nuclear export by inhibiting the binding of XPO1 significantly reduced the nuclear translocation of β‐catenin, which improved the sensitivity of NSCLC to gefitinib. 139 These results provide a new therapeutic strategy for the clinical treatment of gefitinib‐resistant NSCLC.

4.5. Other drugs

Gemcitabine is a basic chemotherapy drug that is often limited by patient tolerance. 144 Gemcitabine resistance is extremely complex and involves alterations in the tumour microenvironment. 145 One study revealed that the AKT, p53 and hypoxia‐inducible factor‐1 signalling pathways are associated with gemcitabine resistance. 145 The key molecules of the above pathways are regulated by the nuclear export of XPO1. Thus, inhibition of nuclear export appears to ameliorate gemcitabine resistance. A previous study showed that KPT‐330 enhanced the antitumour effect of gemcitabine by inducing the nuclear accumulation of p27 and decreasing survivin. 146 Recently, a study revealed that combination treatment with KPT‐330 and gemina‐nab‐paclitaxel improved the survival time of Pdx‐1‐Cre (KPC) model mice. 147 CD44, a cell surface protein, has been reported to be associated with gemcitabine resistance. 148 The combination of KPT‐330 and gemcitabine‐nab‐paclitaxel significantly reduced the number of CD44‐positive cells. The extracellular matrix between tumour cells hinders contact between gemcitabine and tumour cells, leading to the development of gemcitabine resistance. 149 The combination of KPT‐330 and gemcitabine‐nab‐paclitaxel decreased MMP7 and epithelial cellular adhesion molecule, which play important roles in the regulation of the ECM, and increased the therapeutic sensitivity to gemcitabine. 147

Similarly, XPO1 also mediates resistance to other drugs. The application of KPT‐330 to CYH33‐resistant breast cancer cells enhances the localisation of p53 in the nucleus, leading to cell cycle arrest and a reversal of CYH33 resistance. 150 Lenvatinib resistance is a common clinical hurdle in the treatment of thyroid cancer. Research has indicated that an XPO1 inhibitor can suppress the growth of lenvatinib‐resistant 8505C thyroid mesenchymal carcinoma cells both in vivo and in vitro. 151

5. PAST, PRESENT,AND FUTURE OF XPO1 INHIBITORS

5.1. XPO1 inhibitors in preclinical and early clinical development

XPO1 has emerged as a promising potential target in cancer therapy because of its consistent involvement in a wide range of tumour types. Nevertheless, the development of XPO1 inhibitors has encountered substantial challenges. Natural inhibitors of XPO1 (Table 2) and its derivatives have been linked to severe toxic side effects, prompting researchers to explore the potential of small molecule inhibitors. 152 , 153 , 154 , 155 , 156 There are many ongoing studies regarding SINE compounds. Notably, KPT‐330 has achieved FDA approval for the treatment of MM and relapsed or refractory diffuse large B‐cell tumours, marking a significant step forward in the field. 12 , 13

TABLE 2.

Natural products of XPO1 inhibitor.

XPO1 inhibitor drugs Function Restriction Effect in clinical trial
LMB Alkylated amino acid residues of Cys528 at the active site of XPO1 295 , 296 , 297 , 298 , 299 , 300 Severe malaise and anorexia 301 No partial response
Anguinomycins NR NR
Ratjadones High cytotoxicity 302 NR
Piperlongumine NR Effective inhibition of human acute myeloid leukaemia cells 303
Curcumin Low oral bioavailability 304 >120 clinical trials, but no validated clinical trials have been successful 304 , 305
Plumbagin Limited solubility, oral bioavailability and unknown toxicity 306 NR
Oridonin Increased the expression of nucleoporin98 307 Uncertain toxicity 308 NR

Abbreviations: LMB, Leptomycin B; NR, not reported.

5.2. Natural inhibitors and their analogues

LMB is an antifungal agent generated by the bacterium Streptomyces that is an inaugural natural inhibitor capable of inhibiting XPO1 to achieve antitumour effects. 154 , 157 , 158 Regrettably, the corresponding clinical study abruptly ended due to the intense severity of the toxic side effects of LMB, which included severe malaise and anorexia and no partial response (PR). 159 Subsequent discoveries of natural inhibitors akin to LMB, including anguinomycins and ratjadones A through D, presented significant challenges for use in oncological treatment, primarily due to their debilitating toxicity similar to that of LMB. 160 , 161

Several natural XPO1 inhibitors, including piperlongumine, curcumin, plumbagin and oridonin, have been identified in plant extracts. These compounds have demonstrated potential antitumour effects across a variety of tumour cells. 162 , 163 , 164 , 165 , 166 , 167 , 168 , 169 , 170 , 171 , 172 , 173 Similarly, semisynthetic inhibitors exhibit significant antitumour efficacy. However, the advancement of these semisynthetic inhibitors in clinical studies has been impeded due to potential toxicological responses that remain unverified. 174 , 175

5.3. Selective inhibitors

To mitigate the toxic side effects associated with LMB and its analogues during clinical trials, researchers have used the consensus‐induced fit docking methodology. This approach not only decreases the financial burden of drug development but also enhances the diversity of potential therapeutic agents. Utilising computer simulations within dedicated research is particularly effective for creating compounds that are collectively known as SINE compounds (Figure 4). 176

FIGURE 4.

FIGURE 4

Structures of XPO1 inhibitors.

The SINE compounds include KPT‐301, KPT‐330, KPT‐335, KPT‐185, KPT‐276, KPT‐251 and KPT‐8602. KPT‐301, a trans isomer of KPT185, is often used as a negative control for SINEs due to its low activity. 177 , 178 Extensive research has indicated that KPT‐185 has robust antitumour effects in vitro. However, the efficacy of the antitumour effects of KPT‐185 in vivo did not meet the anticipated expectations. Subsequent clinical trials have shown encouraging antitumour outcomes. Remarkably, KPT‐330 gained the approval of the FDA for treating MM and relapsed or refractory diffuse large B‐cell tumours. 12 , 13 , 179 , 180 , 181 Collectively, these findings underscore the therapeutic potential of SINE compounds in the management of tumours.

6. THE STUDY OF SINE COMPOUNDS IN VARIOUS SOLID TUMOURS

Following the remarkable success of KPT‐330 in treating haematological malignancies, a crucial question arises: Do KPT‐330 and its analogous derivatives demonstrate significant antitumour efficacy in solid tumours?

6.1. Glioblastoma

KPT‐330 and KPT‐276 effectively inhibited the growth of glioma cells in vitro and enhanced survival rates in an orthotopic patient‐derived xenograft (PDX) model. 182 , 183 KPT‐330 treatment exhibited a potent proapoptotic effect on tumour cells, and its efficacy was notably augmented when KPT‐330 was combined with Bcl‐xL and the Bcl‐2 inhibitor ABT263, compared to that of the individual treatments, thereby leading to superior tumour suppression both in vivo and in vitro. 184 , 185 Intriguingly, while treatment with KPT‐330 or the XPO1 inhibitor S109 alone did not markedly improve survival rates in vivo, it amplified the sensitivity of tumour cells to radiotherapy, hence offering a fresh perspective for future treatment strategies. 182 , 186 Moreover, studies on high‐grade gliomas have shown that nerve growth factor receptors heighten the responsiveness of KPT‐330, a discovery that could mitigate the issue of single‐drug resistance and provide insights for future combination therapies. 187

6.2. Lung cancer

KPT‐330 inhibited the proliferation of 11 NSCLC cell lines in vitro, and combination with either cisplatin or the Bcl‐xL inhibitor A‐1331852 resulted in more pronounced inhibition than a single treatment. 184 , 188 XPO1 treatment in combination with cisplatin/irinotecan or in combination with the poly (adenosine diphosphate (ADP)‐ribose) polymerase (PARP) inhibitor BMN673 has significant efficacy in vivo, ameliorating chemoresistance. 189 , 190 In addition, XPO1 was surprisingly effective in treating kirsten ratsarcoma viral oncogene homolog (KRAS) ‐mutant lung cancer; in combination with sotorasib (a KRAS‐mutant inhibitor), XPO1 not only inhibited tumour cell proliferation in vitro but also effectively inhibited KRAS12C‐mutated lung cancer cells in SCLC xenografts, increasing the chances of survival in mice. 191 , 192 , 193 Although XPO1 has an extremely low mutation rate, the occurrence of a mutation poses a significant survival risk for patients with NSCLC, and further research is needed. 34

6.3. Breast cancer

KPT‐185, KPT‐251, KPT‐276 and KPT‐330 have been shown to significantly inhibit the growth of breast cancer cell lines both in vivo and in vitro. 95 , 194 Research has indicated that KPT‐330 combined with paclitaxel or eribulin has greater antitumour efficacy than carboplatin or doxorubicin alone, both in vitro and in triple‐negative breast cancer (TNBC) PDX models. 195 , 196 In a separate study, KPT‐330, when used in combination with GSK2126458, a PI3K/mTOR inhibitor, was shown to significantly reduce tumour burden, compared to monotherapy in TNBC PDX models. 197 Moreover, the binding of KPT‐330 to the TRAIL‐R2xCD3 BsAb has been shown to stimulate apoptosis in TNBC cells. 198 KPT‐330 combined with olaparib enhances antitumour activity in vivo and in vitro, regardless of the presence of a BRCA1 mutation in TNBC, indicating a potential direction for future treatment strategies. 199 It has also been suggested that the antitumour and antiproliferative effects of KPT‐330 can be amplified by elevating miR‐34a levels, enhancing the effectiveness of treatment for TNBC. 200 The combination of KPT‐330 and tamoxifen has been shown to be effective in treating tumours in tamoxifen‐resistant tumour xenografts and in vitro. These findings suggest that KPT‐330 could restore the sensitivity of tamoxifen‐resistant cells and prevent recurrence in vivo. 125 , 133 , 134 Additionally, for TP53 wild‐type (WT) breast cancer, KPT‐330 may enhance proliferation inhibition when combined with tucidinostat, a type I histone deacetylase inhibitor, in vitro and significantly inhibit tumour growth in cell‐derived xenografts (CDXs). 201

6.4. Pancreatic cancer

KPT‐185 or KPT‐330, when used individually, effectively inhibited the proliferation of pancreatic cancer cells and demonstrated significant efficacy in halting tumour growth in xenograft models. 100 , 146 , 202 KPT‐330 treatment in combination with gemcitabine markedly enhances the antitumour effects of KPT‐330 against pancreatic cancer both in vitro and in vivo, leading to a decrease in tumour volume and a reduction in liver metastasis. 146 In addition, another study revealed that combining KPT‐330 with gemcitabine supplemented with nab‐paclitaxel resulted in synergistic inhibition of pancreatic cancer cell proliferation. This combination was more effective at halting the growth of PDXs and orthotopic tumours than was any monotherapy. 203 Among pancreatic cancers characterised by high rates of KRAS mutation, KPT‐330 has been found to counteract the drug resistance of KRAS G12C‐resistant pancreatic cancer cells. Combining KPT‐330 with KRAS G12C inhibitors not only drastically inhibited the proliferative capacity of these drug‐resistant cell lines in vitro but also exerted significant tumour‐suppressing effects in a CDX model. 191

6.5. CRC

KPT‐330 effectively impeded the proliferation of CRC cell lines in vitro, and its synergistic use with proteasome inhibitors significantly enhanced its antitumour effect both in vitro and in vivo. 9 , 204 Furthermore, a subsequent study indicated that a sequential regimen involving KPT‐330/KPT‐8602 and the ataxia telangiectasia and Rad3‐related (ATR) inhibitor AZD6738 offered superior efficacy, compared to either agent used independently, thereby extending survival time in TP53‐mutant models of CRC. 205 KPT‐330 also amplified tumour radiosensitivity, thereby bolstering tumour suppression in vitro and in vivo. 206 , 207 These investigations could lead to valuable reference points for devising future strategies for combination therapy involving the use of other drugs for tumour management.

6.6. Cervical cancer

The administration of LMB to HeLa cells suppressed cellular resistance in vitro and additionally hindered the recovery of apoptotic cells. 208 In a separate study, the combination of KPT‐330 and A‐1331852 was found to effectively stimulate apoptosis in HeLa cells in a controlled laboratory environment. 184

6.7. Ovarian cancer

Research has indicated that KPT‐330 and KPT‐185 have a profound impact on cell apoptosis in vitro, markedly increasing the percentage of apoptotic cells. These agents have also been demonstrated to significantly inhibit tumour proliferation in human ovarian carcinoma xenografts and orthotopic models. 114 , 196 , 209 , 210 KPT‐330 in combination with cisplatin exhibited an enhanced tumour‐suppressing effect on platinum‐sensitive ovarian cancer cells. These synergistic effects work by boosting platinum sensitivity in these cancer cells. 111 , 114 In addition, KPT‐330 combined with other drugs, such as RG7388 (an MNM2 inhibitor) or olaparib, synergistically impedes ovarian cancer cell proliferation in vitro by promoting apoptosis. The combination of KPT‐330 with topotecan or olaparib significantly decreases tumour nodule size, inhibits tumour cell proliferation and extends survival in vivo. 196 , 209 , 210 To address the issue of tumour cell resistance caused by the long‐term use of SINEs, strategies such as inhibiting the NRG1/EBB3 signalling pathway have shown potential effectiveness. 211

6.8. Prostate cancer

Various SINEs, such as KPT‐127, KPT‐185, KPT‐205, KPT‐225, KPT‐251 and KPT‐330, have been found to inhibit the growth of prostate cancer cells by inducing apoptosis and arresting the cell cycle in vitro. 40 , 177 , 212 However, the inhibitory effect of KPT‐301 on prostate cancer cells was not obvious in vitro. 177 Another study showed that KPT‐330 or KPT‐251 in a systemic metastasis model significantly reduced the likelihood of bone metastasis from prostate cancer and improved survival time in mice. 212 Interestingly, KPT‐330 or KPT‐251 not only enhanced the effectiveness of the chemotherapeutic drug docetaxel (DTX) in DTX‐sensitive prostate cancer cells but also resensitised DTX‐resistant prostate cancer cells. When these agents are combined with DTX, tumour cell proliferation is inhibited, and apoptosis is induced, effectively extending the survival time of mice. 213 To improve the effectiveness of treatment for desmoplastic‐resistant prostate cancer, KPT‐330 was combined with anti‐androgen receptor (AR) drugs, such as enzalutamide, which have been shown to synergistically enhance antitumour effects in vitro and vivo. Similarly, KPT‐8602, which is used in combination with these anti‐AR agents, has been shown to synergistically inhibit tumour growth and prolong the survival time of mice in vivo. 214 Furthermore, the pairing of KPT‐8602 with PARP inhibitors demonstrated increased effectiveness in inhibiting the proliferation of castration‐resistant prostate cancer cells by inducing apoptosis. 215

6.9. Sarcoma

Extensive research has illuminated the notable tumour‐suppressing potential of SINEs in different types of sarcomas, including gastrointestinal mesenchymal tumours (GISTs), liposarcomas, smooth muscle sarcomas, rhabdomyosarcomas, undifferentiated sarcomas, alveolar soft part sarcomas (ASPSs), Kaposi's sarcomas, Ewing sarcomas (EWSs) and osteosarcomas. 216 , 217 , 218 , 219 , 220

KPT‐330 treatment had potent, dose‐dependent inhibitory effects on GIST cells. It effectively impeded cell proliferation by inducing G1‐phase arrest and triggering apoptosis. Furthermore, in three distinct GIST xenograft models, KPT‐330 successfully inhibited tumour growth and cell proliferation. 216

In liposarcoma, KPT‐330 reduced tumour cell proliferation in vitro by initiating cell cycle arrest, promoting apoptosis and decreasing tumour size and volume in multiple xenograft models. 216 , 220 , 221 Another study demonstrated that the antitumour impact of KPT‐330 treatment, when applied to three PDXs and two dedifferentiated liposarcoma tumour cell lines, was considerably greater than that of doxorubicin. This was evidenced by its superior tumour response and ability to induce apoptosis. 220 , 222 KPT‐330 treatment in conjunction with carfilzomib disrupted ribosome biogenesis networks or inhibited the pro‐survival factor prolin‐rich Akt substrate of 40 kD (PRAS40), thus decreasing tumour cell activity. Interestingly, the combined antitumour efficacy of KPT‐330 and carfilzomib appeared to be dose‐dependent. A low dose of carfilzomib combined with KPT‐330 has been shown to exhibit a superimposed effect, whereas a higher dose has demonstrated a synergistic effect. 220 , 223

In ASPS, although in vitro results suggested that two specific cell lines, ASPS‐KY and ASPS‐1, exhibited resistance to KPT‐330, compelling in vivo evidence demonstrated that KPT‐330 significantly hampered tumour growth and induced cell death in certain ASPS xenograft models. 216 With respect to EWS, in vitro experiments demonstrated that KPT‐330 substantially inhibited most EWS cell lines. However, the A672 and CHP100 cell lines showed a lack of therapeutic sensitivity. 220 Investigations further revealed that combining KPT‐330 with either carfilzomib or crizotinib (a c‐mesenchymal‐epithelial transition factor (c‐MET) inhibitor) markedly enhanced apoptosis in vitro, demonstrating superior efficacy, compared to monotherapy. 219 The insulin‐like growth factor 1 (IGF1) receptor (IGF‐1R) has emerged as a promising therapeutic target for EWS. Interestingly, the knockdown of XPO1 expression resulted in increased expression of IGFBP3, an IGF1‐binding protein that regulates IGF signalling. 219 Accordingly, the combination of KPT‐330 and linsitinib (an IGF‐1R inhibitor) effectively restrains tumour cell proliferation and has synergistic efficacy, compared to monotherapy. 219

In osteosarcoma, KPT‐335 effectively inhibited cell proliferation, regardless of whether it was used independently or in combination with the proteasome inhibitor bortezomib (BORT). Additionally, KPT‐330 helps increase the sensitivity of tumour cells to radiation therapy, thereby augmenting the overall therapeutic impact of the treatment. 218 , 220 , 224

6.10. Melanoma

Different SINEs, including KPT‐185, KPT‐276, KPT‐330 and KPT‐335, have been demonstrated to significantly inhibit the proliferation of melanoma cells and promote apoptosis in vitro. 225 , 226 Empirical evidence from in vivo studies has shown that treatment with KPT‐330 or KPT‐276 alone reduces tumour growth. 225 Additionally, KPT‐330 can be utilised in combination with immunosuppressive agents, including programmed cell death protein 1 (PD‐1) inhibitors, programmed cell death protein 1 ligand 1 (PD‐L1) inhibitors and cytotoxic T‐lymphocyte‐associated protein 4 (CTLA4) inhibitors, for more effective melanoma treatment. 227 However, the daily administration of KPT‐330 potentially leads to a diminished antitumour effect of combined treatment with KPT‐330 and PD‐L1 inhibitors. 228 The strategic scheduling of treatment dosages may play a crucial role in enhancing melanoma immunotherapy efficacy. In addition, the concept of combination therapy holds promise for clinical treatment guidelines (Table 3).

TABLE 3.

Summary of preclinical study of selective inhibitor of nuclear export (SINE) compounds efficacy in solid cancers in vitro and in vivo.

SINE Cancer Antitumour effect in vivo/in vitro Effect of combination therapy in vivo/in vitro
KPT‐127 Pancreatic cancer  In vitro 272 NR
Prostate cancer In vitro 309 NR
KPT‐185 Lung cancer In vitro 310 , 311 Cisplatin in vitro 190 ; irinotecan in vitro 190
Triple‐negative breast cancer In vitro 95 , 312 NR
Gastric cancer In vitro 178 , 313 NR
Pancreatic cancer Both 272 , 314 NR
Ovarian cancer In vitro 315 NR
Kidney cancer In vitro 316 NR
Prostate cancer In vitro 177 NR
Melanoma In vitro 317 NR
KPT‐205 Pancreatic cancer  In vitro 272 NR
Prostate cancer In vitro 309 NR
KPT‐225 Prostate cancer In vitro 309 NR
KPT‐227 Pancreatic cancer  In vitro 272 NR
KPT‐251 Glioblastoma In vitro 182 NR
Triple‐negative breast cancer In vitro 95 NR
Pancreatic cancer Both 314 NR
Kidney cancer Both 316 NR
Prostate cancer Both 177 , 309 Docetaxel (DTX) in vitro and vivo 318
Diffuse malignant peritoneal mesothelioma Both 319 Survivin inhibitor in vitro 319
Melanoma Both 317 PLX‐4032, a BRAF inhibitor, in vitro
KPT‐276 Glioblastoma Both 182 NR
Triple‐negative breast cancer In vitro 95 NR
Non‐small cell lung cancer In vivo 311 NR
Gastric cancer In vivo 313 NR
Diffuse malignant peritoneal mesothelioma Both 319 Survivin inhibitor in vitro 319
Melanoma Both 317 PLX‐4720, a BRAF inhibitor in vitro and vivo 317
KPT‐301 Prostate cancer In vitro 177 NR
KPT‐330 Glioblastoma Both 182 Bcl‐xL/Bcl‐2 inhibitor in vitro and vivo 320 ; Irradiation in vitro and in vivo 186 ; olaparib, a poly(ADP‐ribose) polymerase (PARP) inhibitor in vitro and vivo 321
Lung cancer Both Enzalutamide in vivo 322 ; cisplatin in vitro and vivo 190 , 322 ; irinotecan in vitro and vivo 190 ; KRAS G12C inhibitor in vitro and vivo 191 ; PARP1 inhibition in vitro and vivo 189 ; Bcl‐xL inhibitor in vitro and vivo 184
Breast cancer Both 95 , 312 Tamoxifen in vitro and vivo 134 ; olaparib, a PARP inhibitor in vitro and vivo 199 ; LOM612, a small molecule activator of FOXO nuclear‐cytoplasmic shuttling in vitro and in vivo 323 ; a PI3K/mTOR inhibitor in vitro and in vivo 197 ; tucidinostat in vitro and vivo 201 ; Y219, a novel proteasome inhibitor in vitro and vivo 324
Gastric cancer Both 178 , 313 Nab‐paclitaxel in vitro and vivo 178
Pancreatic cancer Both Gemcitabine in vitro and vivo 146 ; gemcitabine plus nab‐paclitaxel in vitro and vivo 147 , 233
CRC Both Bortezomib (BORT) in vitro and vivo 204 ; irradiation in vitro and in vivo 207 ; ATR inhibitors in vitro 205
Cervical cancer In vitro A Bcl‐xL inhibitor in vitro 184
Ovarian cancer Both 315 Decitabine in vitro 325 ; an inhibitor of MDM2‐p53 interaction in vitro 209 ; cisplatin in vitro and vivo 315 , 326 ; olaparib in vitro and vivo 327
Prostate cancer Both 309 Abiraterone in vitro 214 ; enzalutamide in vitro and vivo 214 , 322 ; DTX in vitro and vivo 318 ; cisplatin in vitro and vivo 322
Sarcoma Both BORT in vitro and vivo 328
Gastrointestinal stromal tumours Both 216 Imatinib in vitro 216
Liposarcoma Both 221 Carfilzomib in vitro 329
Dedifferentiated liposarcoma Both 216 NR
Sarcoma of soft part of alveolar Both 216 NR
Ewing sarcoma Both Crizotinib, a c‐MET inhibitor, in vitro 219 ; linsitinib in vitro and vivo 219
Osteosarcoma In vitro Irradiation in vitro 330
Leiomyosarcoma Both 216 NR
Uterine leiomyosarcoma Both Doxorubicin/gemcitabine in vitro and vivo 331
Rhabdomyosarcoma In vitro 216 NR
Undifferentiated sarcomas Both 216 Palbociclib in vitro and in vivo 332
Diffuse malignant peritoneal mesothelioma Both 319 Survivin inhibitor in vitro 319
Melanoma Both 317 PLX‐4720, a BRAF inhibitor in vivo 317 ; anti‐CTLA4/anti‐PD‐1/anti‐PD‐L1 antibodies in vivo 333
KPT‐335 Esophageal cancer Both 334 NR
Neuroblastoma Both 264 NR
KPT‐8602 Gastric cancer In vitro 178 NR
Hepatocellular carcinoma Both Sorafenib in vivo 335
CRC Both ATR inhibitors in vitro and vivo 205
Prostate cancer Both PARP inhibitors in vitro 336 ; abiraterone in vivo 214 ; enzalutamide in vitro 214

Abbreviations: FOXO, forkhead box protein O; NR, not reported.

7. CLINICAL TRIALS OF XPO1 INHIBITORS

Given the noteworthy therapeutic effectiveness of SINEs in preclinical trials for solid tumours, a growing number of comprehensive clinical studies have investigated the impacts of selinexor, either as a standalone treatment or in combination with other agents. Table 4 provides an overview of completed studies that have contributed to our understanding of the efficacy of selinexor in treating solid tumours. To date, most clinical trials of the application of selinexor in solid tumours are still in the recruitment phase. This suggests that the promising potential of KPT‐330 for treating solid tumours warrants further exploration to establish a safe, effective and rational treatment plan. To date, KPT‐330 has achieved incremental success in clinical trials across several types of solid tumours, underscoring its potential role in advancing cancer therapy.

TABLE 4.

Summary of clinical trial results on selinexor's efficacy in solid cancers.

NCT Trial Completed or not Population and tumour type Selinexor and other treatment Outcomes Haematopoietic‐related adverse events
NCT01896505 Phase Ib trial 337 Completed 54 patients with advanced refractory bone or soft tissue sarcoma 30 mg/m2, 50 mg/m2 or flat dose of 60 mg 33% (17/52) had SD for ≥4 months Grade 3 or 4 toxicities were thrombocytopenia (7.7%), anaemia (9.7%), lymphopenia (9.7%) and leukopenia (9.7%)
NCT02215161 Phase II trial 338 No 14 patients in abiraterone‐ and/or enzalutamide‐refractory metastatic castration‐resistant prostate cancer 65 mg/m2 BIW and subsequently 60 mg/m2 BIW 14.2% (2/14) had PR and 28.6% (4/14) had SD Grade 3 or 4 toxicities were anaemia (21%), neutrophil count decreased (7%), hypophosphatemia (7%)
NCT02419495 Phase Ib trial 339 No 13 patients with advanced solid tumours Selinexor 60 mg BIW or 60–80 mg QW + carboplatin with paclitaxel 7.7% (1/13) had PR and 38.5% (5/13) had SD Grade 3 or 4 toxicities were neutropenia (85%), leukopenia (85%), thrombocytopenia (85%), anaemia (69%)
NCT03042819 Phase Ib trial 340 Completed 25 patients with advanced soft tissue sarcomas Selinexor 60 mg OR 80 mg QW; doxorubicin 75 mg/m2 Q3W 20.8% (5/24) had PR, and 62.5% (15/24) had SD Grade 3 or 4 toxicities were neutropenia (56%), febrile neutropenia (28%) and anaemia (24%)
NCT02178436 Phase Ib trial 341 Completed 9 patients with pancreatic ductal adenocarcinoma Selinexor QW; GEM 1000 mg/m2 QW and nab‐paclitaxel 125 mg/m2 QW 40% (2/5) had PR and 40% (2/5) had SD Grade 3 or 4 toxicities were anaemia, thrombocytopenia, leukopenia and lymphopenia
NCT01607905 Phase I trial 342 Completed 189 patients with advanced solid tumours Selinexor dose escalation: 3–85 mg/m2  0.7% (1/152) had CR, 3.9% (6/152) had PR and 17.8% (27/152) had SD for ≥4 months Grade 3 or 4 toxicities were thrombocytopenia (16%) and hyponatremia (13%)
NCT02269293 Phase I trial 343 Completed 23 patients with advanced ovarian or endometrial cancers (ECs)

1 of 4 alternating regimens:

1. Selinexor 30 mg/m2 BIW, carboplatin AUC 5 IV, paclitaxel 175 mg/m2 IV Q3W;

2. Selinexor 30 mg/m2 BIW, carboplatin AUC 5 IV, paclitaxel 80 mg/m2 IV QW;

3. Selinexor 60 mg/m2 QW, carboplatin AUC 5 IV, paclitaxel 80 mg/m2 IV QW;

4. Selinexor 60 mg/m2 QW, carboplatin AUC 5 IV, paclitaxel 175 mg/m2 IV Q3W

4.3% (1/23) had CR, 52.2% (12/23) had PR and 13% had SD Grade 3 or 4 toxicities were leukopenia (70%), neutropenia (70%), lymphopenia (61%) and anaemia (57%)
NCT02419495 Phase Ib trial 344 No 14 patients with advanced or metastatic solid tumours Selinexor 60 mg BIW or 60–80 mg QW + topotecan 0.5 to 1.5 mg/m2 daily for 5 days 46.2% (6/13) had SD Grade 3 or 4 were anaemia (21 %), neutropenia (21 %), thrombocytopenia (21 %) and leukopenia (14 %)
NCT02025985 Phase II trial 345 Completed 114 patients with recurrent gynaecological malignancies 35 or 50 mg/m2 BIW or 50 mg/m2 QW 11.2% (11/98) had PR and 23.5% (23/98) had SD Grade 3 or 4 were thrombocytopenia (17%) and anaemia (10%)
NCT02419495 Phase Ib trial 346 No 35 patients with metastatic solid tumours Selinexor 60 or 80 mg BIW + paclitaxel 80 mg QW 12.9% (4/31) had PR and 32.3% (10/31) had SD Grade 3 or 4 were neutropenia (46%) and anaemia (31%)
NCT02606461 Phase II‐III trial 347 Completed 285 patients with dedifferentiated liposarcoma Selinexor 60 mg BIW or placebo BIW 2.66% (5/188) had PR and 59.0% (111/188) had SD Grade 3 or 4 were anaemia (18.7%), thrombocytopenia (10.2%) and neutropenia (9.1%)
NCT03555422 Phase III trial 348 No 263 patients with advanced or recurrent EC Selinexor 80 mg QW or placebo QW NR Grade 3 or 4 were neutropenia (8.8%), thrombocytopenia (7.0%), anaemia (2.3%) and leukopenia (1.2%)
NCT02419495 Phase Ib trial 349 No 31 patients with advanced solid tumours and triple‐negative breast cancer

Two stages:

1. Selinexor 60 mg BIW, eribulin 1 mg/m2 QW;

2. Selinexor 80 mg QW, eribulin 1 mg/m2 QW;

9.7% (3/31) had PR and 58.1% (18/31) had SD Grade 3 or 4 were neutropenia (29%), leukopenia (26%) and anaemia (16%)
NCT02384850  Phase I trial 350 No 10 patients with metastatic CRC Selinexor 20 or 40 mg + 5‐fluorouracil, leucovorin and oxaliplatin (mFOLFOX6) NR No obvious grade 3 or 4 were observed
NCT02137356 Phase I trial 351 No 11 patients with locally advanced rectal cancer Selinexor 20 mg/m2 BIW + chemoradiation, or 35 mg/m2 BIW+ chemoradiation, or 35 mg/m2 BIW + 2 weeks after chemoradiation NR No obvious grade 3 or 4 were observed
NCT02402764 Phase II trial 352 Completed 10 patients with metastatic triple‐negative breast cancer 60 mg BIW 30% (3/10) had SD Grade 3 or 4 was platelet count decreased (10%)
NCT01986348 Phase II trial 353 No 76 patients with recurrent glioblastoma Arm B: 50 mg/m2 BIW Arm B: 8.3% (2/24) had PR, 25% (6/24) had SD Grade 3 or 4 were thrombocytopenia (4.4%), leukopenia (5.9%), neutropenia (11.8%) and lymphopenia (5.9%)
Arm C: 60 mg flat dose BIW Arm C: 7.7% ( 1/13) had PR, 30.8%  (4/13) had SD
Arm D: 80 mg flat dose QW Arm D: 3.3% (1/30) had CR, 6.7% (2/30) had PR, 23.3% (7/30) had SD
NR Phase I trial 354 22 patients with advanced CRC 4 schedules WT: 12.5% (1/8) had disease control rate (DCR) of >3 months NR
1: 40 mg/m2 BIW until 28 days RAS/AKT‐mut: 40% (4/10) had DCR of >3 months
2: 50 mg/m2 QW continuous until 28 days
3: 40 mg/m2 BIW for 2 weeks, until 21 days
4: 20 mg/m2 TIW until 28 days
NCT02120222 Phase I trial 355 Completed 7 patients with unresectable melanoma 50 mg/m2 BIW 14.3% (1/7) had a minor response, 57.2% (4/7) experienced SD Grade 3 or 4 were neutropenia (14.3%), thrombocytopenia (42.9%), anaemia (14.3%)
NCT04256707 Phase I/II trial 356 No 29 patients with CRC Selinexor 80 mg QW and pembrolizumab 200 mg IV every 3 weeks 38.9% (7/18) had SD NR
NCT02078349 Phase I trial Completed 120 patients with advanced or metastatic solid tumour malignancies 3 schedules NR NR
1:50 mg/m2 QW continuous until 28 days
2:40 mg/m2 BIW continuous until 21 days
3:20 mg/m2 TIW continuous until 28 days
NCT02250885 Phase II trial Completed 14 patients with poorly differentiated lung and gastrointestinal and pancreatic neuroendocrine tumours 50 mg/m2 BIW on weeks 1, 2 and 3 of each 4‐week cycle NR NR
NCT03095612 Phase I/II trial Completed 40 patients with NSCLC Selinexor QW or BIW and DTX Q3W (75 mg/m2 IV) NR NR

Abbreviations: BIW, twice a week; CR, completed response; NR, not reported; NSCLC, non‐small cell lung cancer; PR, partial response; Q3W, thrice a week; QW, once a week; SD, stable disease; TIW, thrice a week.

Source: All data are from the US National Institutes of Health (NIH) clinical trial database (clinicaltrials.gov).

7.1. Glioblastoma

In a phase II clinical study (NCT01986348) of the use of oral selinexor for the treatment of glioblastoma, 76 patients were enrolled. Although varying degrees of treatment‐related adverse events were reported with the use of oral selinexor, these adverse effects were mitigated by adjusting the drug dosage. 229 For the nonsurgical treatment cohort (n = 68), 28% (19/68) of the tumours exhibited a significant reduction in tumour size. Additionally, this group also reported an objective response rate (ORR) of 10% (3/30), and the highest 6‐month PFS rate of 17.7% was noted in the oral 80 mg/once weekly treatment subgroup. This subgroup was unique because it achieved a complete response (3.3%, 1/30) and two instances of PR (6.7%, 2/30). In contrast, in the surgical treatment cohort (n = 8), immunohistochemistry indicated that selinexor treatment effectively reduced cell proliferation (Ki67: pretreatment vs. posttreatment 29% ± 3% vs. 13% ± 0.8%, p = .012) and promoted apoptosis (cleaved caspase‐3: pretreatment 2% ± 0.7% vs. 28% ± 3.0%, p = .003). Remarkably, one patient continued oral selinexor treatment for up to 42 months. The promising efficacy and safety outcomes observed in this trial suggest that further exploration of the potential of selinexor for treating glioblastoma is warranted.

7.2. Breast cancer

In a phase II clinical study (NCT02402764) of selinexor administration for the treatment of metastatic TNBC (n = 10), patients were orally administered 60 mg of selinexor twice a week, during which the drug was well tolerated. 230 However, the combined efficacy—considering completed response (CR), PR and stable disease (SD)—was rather limited at only 30% (three out of 10 patients). In addition, both in vivo and in vitro studies have shown that combined treatment has a more substantial therapeutic effect on tumours than monotherapy. 195 , 196 , 197 , 198 Despite these findings, the study was unfortunately terminated due to interim results failing to meet expectations. Nevertheless, the results underscore the need for additional clinical trials involving selinexor in combination with other drugs for breast cancer treatment. In another study (NCT02419495), selinexor in combination with paclitaxel was administered to two breast cancer patients. The results showed SD in 50% of the patients (one out of 2 patients) and PD in the other 50% (one out of 2 patients). 231

7.3. Pancreatic cancer

According to a study regarding the use of selinexor for the treatment of advanced solid tumours in Asia, patients with solid malignancies with PI3K/AKT/rat sarcoma (RAS) pathway mutations had a median PFS of 40 days, whereas the PFS was 32 days for the other six pancreatic cancer patients. 232 Furthermore, the study succeeded in establishing the optimal dose of selinexor for the forthcoming phase 2 study, setting the stage for subsequent efficacy validation. To further evaluate the substantial clinical efficacy of selinexor in combination with gemcitabine and nab‐paclitaxel, a total of nine patients were enrolled in this study (NCT02178436). 233 Over the course of the research period, 22.2% (2/9) of the patients treated with this combination achieved a PR, while another 22.2% (2/9) achieved SD. Remarkably, one patient even achieved a 16‐month PFS following treatment, culminating in an OS time of 22 months. These findings underscore the promising potential of selinexor for the treatment of pancreatic cancer.

7.4. CRC

In the first clinical study (NCT01607905) regarding the use of selinexor for the treatment of advanced solid tumours (n = 189), histochemical analyses performed approximately 4 weeks before and after selinexor treatment revealed that selinexor reduced the number of tumour cells by promoting apoptosis and reducing tumour cell proliferation. 10 Furthermore, one patient showed PD in imaging studies. In a subset of the study, biopsies were taken from 43 patients with CRC, revealing that 29 patients had KRAS mutations. Among the patients with KRAS mutations, 44% exhibited SD, whereas only 12% of the patients in the KRAS WT group exhibited SD. A subsequent phase I trial examining the impact of selinexor on patients with advanced CRC harbouring either WT or RAS/AKT pathway mutations aimed at evaluating treatment efficacy. 234 Interestingly, PFS was longer in patients with RAS/AKT pathway mutations than in those with WT RAS/AKT pathway expression (78 days vs. 50 days). Moreover, the disease control rate (DCR) was greater in patients with RAS/AKT mutations (four out of 10 or 40%) than in WT patients (one out of eight or 12.5%). Finally, in a recent study assessing selinexor tolerance in Asian patients with advanced solid tumours (n = 14), the median PFS was reported to be 42 days. 232

To enhance our understanding of the safety and tolerability of selinexor in combination with calcium 5‐fluorouracil, folinic acid and oxaliplatin (mFOLFOX6) for treating metastatic CRC, a phase I clinical trial (NCT02384850) was conducted with 10 patients. 235 Regrettably, the trial encountered a setback when dose‐limiting toxicity necessitated the withdrawal of four patients. Even after the selinexor dose was reduced, another four patients independently chose to withdraw from the study following the first treatment cycle. Moreover, the treatment's effectiveness was insufficient to achieve the anticipated clinical outcomes. For future investigations, adjusting the drug combination and treatment dosage regimen may be beneficial. Previous animal research has indicated that treatment with selinexor augments sensitivity to radiotherapy in mouse xenograft models. 206 , 207 A new phase I clinical trial (NCT02137356) enrolled 11 patients with rectal cancer to study the efficacy and safety of different doses of selinexor (20, 35, 50 mg/m2) in combination with neoadjuvant radiotherapy. 236 However, two patients from the highest dose group opted out of the trial due to poor tolerance of subsequent treatments. Of the nine remaining patients, two exhibited a PR, achieving a near complete or complete pathologic response. For the optimal delineation of an appropriate selinexor dose in conjunction with neoadjuvant radiotherapy, a phase II clinical trial is advised. Furthermore, a phase I open‐label study (NCT04256707) investigated the efficacy of selinexor combined with pembrolizumab for patients (= 29) resistant to chemotherapy in CRC. 237 Of the 18 evaluated patients, 38.9% (7/18) had SD, and 83.3% (6/7) of those with SD had RAS mutations. This study underscores the effectiveness of selinexor in combination with pembrolizumab in combating chemotherapy‐resistant CRC. Given the satisfactory safety profile of this combination, further exploration of CRC treatment modalities could be conducted, particularly focused on RAS‐mutant CRC.

7.5. Prostate cancer

In an initial clinical investigation (NCT01607905) regarding the use of selinexor for the treatment of advanced solid tumours (n = 189), histochemical analysis was performed approximately 4 weeks prior to and after selinexor treatment. Treatment with selinexor noticeably reduced the tumour cell count by inducing apoptosis and curtailing tumour cell proliferation. 10 Concurrently, PD was observed by imaging in one patient with prostate cancer. Regrettably, among patients who withdrew from the study, two had prostate cancer and had SD for 325 and 72 days before any signs of disease progression were identified. In a separate clinical investigation (NCT02215161) involving metastatic debulking‐resistant prostate cancer patients (n = 14), the efficacy of selinexor in combination with enzalutamide and abiraterone was examined. 238 Due to poor drug tolerance, 21% (3/14) of the patients had to prematurely discontinue their participation in the study. Furthermore, severe adverse effects were reported by 36% (5/14) of the patients, leading to early termination of the study. Of the eight patients who could be evaluated, 50% had SD, 25% had PR and some had a decreasing trend in prostate‐specific antigen (PSA) levels after selinexor treatment. These findings underscore the necessity of continued investigation into the clinical implications of selinexor for the treatment of prostate cancer.

7.6. Gynaecologic oncology

In an initial clinical study (NCT01607905), the efficacy of selinexor on solid tumours was evaluated; one patient with ovarian cancer (1/8) and one patient with cervical cancer (1/7) achieved a PR as demonstrated by posttreatment imaging. 10 In a multicentre phase I study, the safety, tolerability and efficacy of selinexor were evaluated in patients with advanced platinum‐resistant ovarian cancer. 111 The findings showed that selinexor therapy hindered tumour growth in 60% (3/5) of the evaluated patients. In five evaluable patients, selinexor treatment inhibited tumour growth in 60% (3/5) of patients, with one showing a PR and the other two demonstrating SD and moderate tolerability of the drug. In a subsequent phase II study (NCT02025985), the efficacy and safety of selinexor were examined for recurrent gynaecologic tumours. Phase I included 25 ovarian cancer patients, 23 endometrial cancer (EC) patients and 25 cervical cancer patients, while phase II included 41 ovarian cancer patients. 239 Following phase I treatment, the DCR was 28% for ovarian cancer, 24% for cervical cancer and 35% for EC. After phase II treatment, the DCR was 33% for patients with ovarian cancer, with a selinexor treatment dose of 35 mg/m2 administered twice weekly and 50 mg/m2 once weekly. Additionally, eight patients achieved a PR throughout the treatment period. In summary, this research demonstrated that selinexor, when administered orally, has a modest degree of efficacy in the management of ovarian and ECs.

To enhance our understanding of the effectiveness and safety of selinexor combined with chemotherapy for treating gynaecological tumours, a study (NCT02419495) included 14 patients suffering from various gynaecological malignancies. 240 The group comprised patients with various gynaecological tumours, including ovarian cancer (n = 5), EC (n = 2) and single instances of both fallopian tube and vaginal cancer. Following treatment with a 60 mg dose of selinexor coupled with topotecan, a promising 46% of patients (6/13) exhibited SD and demonstrated moderate tolerance to the medication. These findings suggest the potential of this combined therapeutic approach for gynaecological cancer treatment. Furthermore, an additional phase I clinical study (NCT02269293) involving five patients with ovarian cancer and 18 patients with EC evaluated the safety and effectiveness of selinexor in combination with paclitaxel and carboplatin. 241 Although the sample size for this study was somewhat limited, the results were encouraging, with a notable treatment effect observed in 56.5% (13/23) of the patients. These clinical studies underscore the potential therapeutic effectiveness of selinexor combined with chemotherapy for treating gynaecological cancers. In a randomised, prospective, multicentre, double‐blind, placebo‐controlled, phase III study (NCT03555422), 263 patients with advanced or recurrent EC were randomly assigned to receive selinexor or placebo after combination chemotherapy. 242 Notably, patients with p53 WT EC had a median PFS of 13.7 months when treated with selinexor and 3.7 months when treated with placebo. These findings suggest that selinexor is important for maintenance therapy in p53 WT ECs, but this requires further investigation.

In a single‐centre, multiarm phase Ib clinical trial (NCT02419495), 28 patients with refractory or platinum‐resistant ovarian cancer were enrolled, along with one individual with cervical cancer. 231 Almost all participants experienced a treatment‐emergent adverse event (TEAE). However, these findings were all within an assessable range. The most common TEAEs of grade ≥ 3 included neutropenia (46%), anaemia (31%) and nausea (21%). Among the 24 patients with ovarian cancer that were evaluated, a response rate of 17% (4/24), a clinical benefit rate of 58% (14/24) and a median PFS of 6.8 months were observed. This evidence of the modest clinical efficacy and manageable toxicity of these agents suggests that further exploration of combination therapy for malignant gynaecologic tumours could be beneficial.

7.7. Sarcoma

In a phase Ib clinical trial (NCT01896505) designed to assess treatment efficacy and safety, 54 patients with advanced, refractory bone/soft tissue sarcoma were treated with various doses of selinexor (30, 50, or 60 mg/m2). 243 Despite the absence of CR or PR among the 52 evaluable patients, SD was observed for 4 months or longer in 33% (17/52) of patients. This included seven patients with dedifferentiated liposarcoma (out of 15), 40% (6/15) of whom exhibited significant antitumour effects. In a subsequent phase II‐III randomised, multicentre, double‐blind, placebo‐controlled study (NCT02606461), 285 patients with advanced refractory dedifferentiated liposarcoma were enrolled (188 on selinexor, 97 on placebo). 244 PFS increased by 30% after 12 or more weeks of selinexor treatment, compared to that in the placebo group, with an ORR of 2.7% (5/188). However, after a median follow‐up of 14.6 months, OS did not significantly differ between the selinexor group and the placebo group. These findings provide innovative therapeutic insights for the treatment of dedifferentiated liposarcoma.

In a phase Ib trial (NCT03042819), selinexor was tested in combination with adriamycin for treating patients with advanced soft tissue sarcoma. 245 A total of 25 patients were included in the trial. Eighty percent of these patients experienced grade 3 or higher treatment‐related adverse events (AEs). The most prevalent haematologic adverse events included anaemia, leukopenia, neutropenia and granulocytopenia, each affecting 92% of the participants. Despite these adverse effects, 21% of patients achieved a PR, while 63% achieved SD, leading to an overall DCR of 84%. This finding suggests the potential applicability of combining selinexor with adriamycin for soft tissue sarcoma treatment. Furthermore, the effects of the administration of 80 mg of selinexor in combination with adriamycin were investigated. This study also revealed possibilities for incorporating other drugs to mitigate the associated toxic side effects.

7.8. Melanoma

In an initial clinical trial (NCT01607905) investigating the use of selinexor for solid tumours (n = 189), CR was observed in one melanoma patient, while another demonstrated PR, as revealed by imaging. 10 A separate study (NCT02120222) focused on unresectable melanoma involving seven patients demonstrated that 57.1% (4/7) of patients achieved SD and 14.3% (1/7) experienced a minor response. This led to a DCR of 71.4% (5/7). 246 Due to the favourable tolerability of selinexor among melanoma patients, another trial was performed. The results from this subsequent melanoma study are anticipated (NCT02120222).

8. SAFETY AND ADVERSE EFFECTS IN PATIENTS WITH SOLID CANCERS

Given the remarkable therapeutic effect of selinexor on haematologic malignancies, the question remains whether selinexor has toxic effects on haematopoietic stem cells in solid tumours. In a phase I clinical trial (NCT01607905) of selinexor for advanced solid tumours, the most common haemosystem‐related AEs were thrombocytopenia (16%), anaemia (9%) and neutropenia (8%). 10 Most AEs, including fatigue, nausea, anorexia, vomiting and hyponatremia, were manageable with the indicated agents. Notably, the occurrence of hyponatremia includes multiple factors (such as comorbidities and medications) and is often associated with selinexor‐related gastrointestinal side effects (such as nausea and vomiting). 247 Other studies have shown that the unique mechanism of selinexor and its effective penetration into the central nervous system may lead to gastrointestinal AEs. 182 , 248 Recently, eltanexor has been tested in clinical trials for the treatment of haematologic malignancies. 249 Compared to selinexor, eltanexor reduced the incidence and severity of hyponatremia and decreased appetite, fatigue and nausea in MM patients. 250 Subsequent studies have shown that haematological AEs can be controlled by reducing selinexor doses or adding the indicated agents. 229 , 231 For example, inhibition of XPO1 expression leads to thrombocytopenia by inhibiting thrombopoietin (TPO) signalling and blocking the differentiation of stem cells into megakaryocytes. 251 TPO agonists reverse selinexor‐induced thrombocytopenia. In a phase 1 clinical trial regarding the use of selinexor combined with neoadjuvant chemoradiation for the treatment of patients with locally advanced rectal cancer, thrombocytopenia resolved within days of pausing treatment. 236

9. RESPONSE BIOMARKERS

Although XPO1 inhibitors have demonstrated potent antitumour effects, not all patients benefit from XPO1 inhibitors. XPO1 mutations, especially the XPO1 E571 mutation, occur in a variety of cancers. 36 , 252 The XPO1 E571 mutation promotes the development of B‐cell malignancies and increases therapeutic sensitivity to selinexor. 33 Similarly, the XPO1 E571 mutation was detected in plasma cell‐free DNA from Hodgkin's lymphoma patients and was associated with tumour regression or progression. Upregulation of neuregulin 1 (NRG1) and ERBB3 expression inhibits the antitumour effect of SINEs. 211 In MM, downregulation of the activity of the transforming growth factor‐β (TGFβ)/drosophila mothers against decapentaplegic protein 4 (SMAD4) pathway was associated with resistance to selinexor. 253 Inhibition of the RNA‐binding protein (hnRNPU) increased the sensitivity of selinexor treatment. 254 Interestingly, both ankyrin repeat and the suppressor of cytokine signalling (SOCS) box containing 8 overexpression and knockout sensitise cells to selinexor. 253 A study found that selinexor upregulates the expression of P2RY2 (a purinergic receptor), which leads to the activation of the AKT pathway. 255 Activation of the AKT pathway, which promotes FOXO3a nuclear export, limits the efficacy of selinexor, and the combination of AKT inhibitors and selinexor increases the efficacy. 256 A novel three‐gene expression signature (WNT10A, DUSP1 and ETV7) predicts selinexor response in MM patients. High expression of these three genes was associated with prolonged PFS in MM patients treated with selinexor, suggesting that the expression signatures of these three genes are potential biomarkers for predicting treatment outcomes. 257 A recent study showed that XPO1, nuclear factor kappa B (NF‐κB) (p65), myeloid cell leukemia‐1 (MCL‐1) and p53 protein levels in haematological tumours may be used as biomarkers of the response to XPO1 inhibitor therapy. 258 However, due to the small sample size, verification with a larger sample size is needed. Furthermore, p53 phosphorylation was found to be a key regulator of selinexor treatment efficacy. 256 Notably, at present, there are few therapeutic options for solid tumours treated with SINEs. The therapeutic predictors for solid tumours treated with SINEs need to be verified and expanded upon by further research (Figure 5).

FIGURE 5.

FIGURE 5

Schematic diagram of response biomarkers of selective inhibitor of nuclear exports (SINEs) in tumour therapy.

10. CONCLUSION

XPO1, a critical component of nuclear‐cytoplasmic transport, plays a pivotal role in tumour development and therapeutic resistance. The targeted disruption of the nuclear‐cytoplasmic transport functionality of XPO1 has led to promising outcomes in cancer therapy.

In clinical trials for solid tumours, nuclear export‐selective inhibitors have shown notable antitumour activity via multiple pathways. The potential of these agents coupled with targeted drugs, chemotherapeutic agents and cycle inhibitors enhances their effectiveness. Moreover, combination treatment for solid tumours could reverse drug resistance to a certain degree. Furthermore, the FDA approved selinexor, an XPO1 inhibitor, for MM and refractory diffuse large B‐cell lymphoma. Despite potential concerns surrounding the safety and feasibility of XPO1 inhibitors in solid tumours, preclinical studies have revealed remarkable antitumour activity. The combination of these inhibitors with various existing drugs has yielded encouraging results, indicating the need for a more thorough investigation into the use of XPO1 inhibitors for solid tumour treatment. Notably, nuclear export‐selective inhibitors bind covalently, causing XPO1 inhibition. However, Cys528 mutations have never been detected in humans treated with XPO1 inhibitors. Gene editing technology has revealed that the Cys528 mutation can inactivate these inhibitors. 259 , 260 This finding suggests the continuous need for the optimisation of nuclear export‐selective inhibitors.

In conclusion, XPO1 targeting holds considerable promise for solid tumour treatment. Nuclear export‐selective inhibitors such as selinexor have shown significant efficacy in treating solid tumours and reducing drug resistance, suggesting new therapeutic directions for future treatments. To optimise the effectiveness of these agents in managing solid tumours, nuclear export‐selective inhibitors should be further refined through extensive clinical trials.

AUTHOR CONTRIBUTIONS

Chuanxi Lai drafted the manuscript. Lingna Xu and Shengdai discussed and revised the manuscript. All the authors read and approved the final manuscript.

CONFLICT OF INTEREST STATEMENT

The authors declare no conflicts of interest.

ACKNOWLEDGEMENTS

Dai Sheng is supported by the National Natural Science Foundation of China (82072628 and 81703076) and the Key Research and Development Program of Zhejiang (2022C03032). Xu Lingna is supported by the National Natural Science Foundation of China (32200593) and the Natural Science Foundation of Zhejiang Province, China (LQ23H160040).

Lai C, Xu L, Dai S. The nuclear export protein exportin‐1 in solid malignant tumours: From biology to clinical trials. Clin Transl Med. 2024;14:e1684. 10.1002/ctm2.1684

Contributor Information

Lingna Xu, Email: xulingna@zju.edu.cn.

Sheng Dai, Email: daimd@zju.edu.cn.

DATA AVAILABILITY STATEMENT

No data were generated for the research described in the article.

REFERENCES

  • 1. Nigg EA. Nucleocytoplasmic transport: signals, mechanisms and regulation. Nature. 1997;386(6627):779‐787. [DOI] [PubMed] [Google Scholar]
  • 2. Knockenhauer KE, Schwartz TU. The nuclear pore complex as a flexible and dynamic gate. Cell. 2016;164(6):1162‐1171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Azmi AS, Mohammad RM. Targeting cancer at the nuclear pore. J Clin Oncol. 2016;34(34):4180‐4182. [DOI] [PubMed] [Google Scholar]
  • 4. Mahipal A, Malafa M. Importins and exportins as therapeutic targets in cancer. Pharmacol Ther. 2016;164:135‐143. [DOI] [PubMed] [Google Scholar]
  • 5. Wing CE, Fung HYJ, Chook YM. Karyopherin‐mediated nucleocytoplasmic transport. Nat Rev Mol Cell Biol. 2022;23(5):307‐3328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Cook A, Bono F, Jinek M, Conti E. Structural biology of nucleocytoplasmic transport. Annu Rev Biochem. 2007;76:647‐671. [DOI] [PubMed] [Google Scholar]
  • 7. Saulino DM, Younes PS, Bailey JM, Younes M. CRM1/XPO1 expression in pancreatic adenocarcinoma correlates with survivin expression and the proliferative activity. Oncotarget. 2018;9(30):21289‐21295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Subhash VV, Yeo MS, Wang LZ, et al. Anti‐tumor efficacy of Selinexor (KPT‐330) in gastric cancer is dependent on nuclear accumulation of p53 tumor suppressor. Sci Rep. 2018;8:12248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Aladhraei M, Kassem Al‐Thobhani A, Poungvarin N, Suwannalert P. Association of XPO1 Overexpression with NF‐kappaB and Ki67 in colorectal cancer. Asian Pac J Cancer Prev. 2019;20(12):3747‐3754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Abdul Razak AR, Mau‐Soerensen M, Gabrail NY, et al. First‐in‐class, first‐in‐human phase i study of selinexor, a selective inhibitor of nuclear export, in patients with advanced solid tumors. J Clin Oncol. 2016;34(34):4142‐4150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Jardin F, Pujals A, Pelletier L, et al. Recurrent mutations of the exportin 1 gene (XPO1) and their impact on selective inhibitor of nuclear export compounds sensitivity in primary mediastinal B‐cell lymphoma. Am J Hematol. 2016;91(9):923‐930. [DOI] [PubMed] [Google Scholar]
  • 12. Dolgin E. XPO1 inhibitor approved for multiple myeloma. Cancer Discov. 2019;9(9):1150‐1151. [DOI] [PubMed] [Google Scholar]
  • 13. Kasamon YL, Price LSL, Okusanya OO, et al. FDA approval summary: selinexor for relapsed or refractory diffuse large B‐cell lymphoma. Oncologist. 2021;26(10):879‐886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Adachi Y, Yanagida M. Higher‐order chromosome structure is affected by cold‐sensitive mutations in a Schizosaccharomyces‐pombe gene crm1+ which encodes a 115‐kD protein preferentially localized in the nucleus and at its periphery. J Cell Biol. 1989;108(4):1195‐1207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Stade K, Ford CS, Guthrie C, Weis K. Exportin 1 (Crm1p) is an essential nuclear export factor. Cell. 1997;90(6):1041‐1050. [DOI] [PubMed] [Google Scholar]
  • 16. OssarehNazari B, Bachelerie F, Dargemont C. Evidence for a role of CRM1 in signal‐mediated nuclear protein export. Science. 1997;278(5335):141‐144. [DOI] [PubMed] [Google Scholar]
  • 17. Fukuda M, Asano S, Nakamura T, et al. CRM1 is responsible for intracellular transport mediated by the nuclear export signal. Nature. 1997;390(6657):308‐311. [DOI] [PubMed] [Google Scholar]
  • 18. Hutten S, Kehlenbach RH. CRM1‐mediated nuclear export: to the pore and beyond. Trends Cell Biol. 2007;17(4):193‐201. [DOI] [PubMed] [Google Scholar]
  • 19. Ossareh‐Nazari B, Dargemont C. Domains of Crm1 involved in the formation of the Crm1, RanGTP, and leucine‐rich nuclear export sequences trimeric complex. Exp Cell Res. 1999;252(1):236‐241. [DOI] [PubMed] [Google Scholar]
  • 20. Macara IG. Transport into and out of the nucleus. Microbiol Mol Biol R. 2001;65(4):570‐594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Tan DS, Bedard PL, Kuruvilla J, Siu LL, Razak AR. Promising SINEs for embargoing nuclear‐cytoplasmic export as an anticancer strategy. Cancer Discov. 2014;4(5):527‐537. [DOI] [PubMed] [Google Scholar]
  • 22. Ishizawa J, Kojima K, Hail N, Jr. , Tabe Y, Andreeff M. Expression, function, and targeting of the nuclear exporter chromosome region maintenance 1 (CRM1) protein. Pharmacol Ther. 2015;153:25‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Nemergut ME, Lindsay ME, Brownawell AM, Macara IG. Ran‐binding protein 3 links Crm1 to the Ran guanine nucleotide exchange factor. J Biol Chem. 2002;277(20):17385‐17388. [DOI] [PubMed] [Google Scholar]
  • 24. Azmi AS, Uddin MH, Mohammad RM. The nuclear export protein XPO1—from biology to targeted therapy. Nat Rev Clin Oncol. 2021;18(3):152‐169. [DOI] [PubMed] [Google Scholar]
  • 25. Dong XH, Biswas A, Suel KE, et al. Structural basis for leucine‐rich nuclear export signal recognition by CRM1. Nature. 2009;461(7263):1136‐1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Andrade MA, Bork P. HEAT repeats in the Huntington's disease protein. Nat Genet. 1995;11(2):115‐116. [DOI] [PubMed] [Google Scholar]
  • 27. Petosa C, Schoehn G, Askjaer P, et al. Architecture of CRM1/exportin1 suggests how cooperativity is achieved during formation of a nuclear export complex. Mol Cell. 2004;16(5):761‐775. [DOI] [PubMed] [Google Scholar]
  • 28. Fornerod M, vanDeursen J, vanBaal S, et al. The human homologue of yeast CRM1 is in a dynamic subcomplex with CAN/Nup214 and a novel nuclear pore component Nup88. Embo J. 1997;16(4):807‐816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Wen W, Meinkoth JL, Tsien RY, Taylor SS. Identification of a signal for rapid export of proteins from the nucleus. Cell. 1995;82(3):463‐473. [DOI] [PubMed] [Google Scholar]
  • 30. Guttler T, Madl T, Neumann P, et al. NES consensus redefined by structures of PKI‐type and Rev‐type nuclear export signals bound to CRM1. Nat Struct Mol Biol. 2010;17(11):1367‐1376. [DOI] [PubMed] [Google Scholar]
  • 31. Kudo N, Matsumori N, Taoka H, et al. Leptomycin B inactivates CRM1/exportin 1 by covalent modification at a cysteine residue in the central conserved region. Proc Natl Acad Sci USA. 1999;96(16):9112‐9117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Yashiroda Y, Yoshida M. Nucleo‐cytoplasmic transport of proteins as a target for therapeutic drugs. Curr Med Chem. 2003;10(9):741‐748. [DOI] [PubMed] [Google Scholar]
  • 33. Taylor J, Sendino M, Gorelick AN, et al. Altered Nuclear export signal recognition as a driver of oncogenesis. Cancer Discov. 2019;9(10):1452‐1467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Nagasaka M, Asad MFB, Al Hallak MN, et al. Impact of XPO1 mutations on survival outcomes in metastatic non‐small cell lung cancer (NSCLC). Lung Cancer. 2021;160:92‐98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Garcia‐Santisteban I, Arregi I, Alonso‐Marino M, et al. A cellular reporter to evaluate CRM1 nuclear export activity: functional analysis of the cancer‐related mutant E571K. Cell Mol Life Sci. 2016;73(24):4685‐4699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Camus V, Stamatoullas A, Mareschal S, et al. Detection and prognostic value of recurrent exportin 1 mutations in tumor and cell‐free circulating DNA of patients with classical Hodgkin lymphoma. Haematologica. 2016;101(9):1094‐1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Forbes DJ, Travesa A, Nord MS, Bernis C. Reprint of “Nuclear transport factors: global regulation of mitosis”. Curr Opin Cell Biol. 2015;34:122‐134. [DOI] [PubMed] [Google Scholar]
  • 38. Fischer U, Huber J, Boelens WC, Mattaj IW, Luhrmann R. The Hiv‐1 Rev activation domain is a nuclear export signal that accesses an export pathway used by specific cellular RNAs. Cell. 1995;82(3):475‐483. [DOI] [PubMed] [Google Scholar]
  • 39. Fu SC, Huang HC, Horton P, Juan HF. ValidNESs: a database of validated leucine‐rich nuclear export signals. Nucleic Acids Res. 2013;41(D1):D338‐D343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Gravina GL, Mancini A, Sanita P, et al. KPT‐330, a potent and selective exportin‐1 (XPO‐1) inhibitor, shows antitumor effects modulating the expression of cyclin D1 and survivin in prostate cancer models. BMC Cancer. 2015;15:941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Mori K, Oshima Y, Nose K, Shibanuma M. Competitive nuclear export of cyclin D1 and Hic‐5 regulates anchorage‐dependence of cell growth and survival. Mol Biol Cell. 2009;20(1):218‐232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Martin APJ, Jacquemyn M, Lipecka J, et al. STK38 kinase acts as XPO1 gatekeeper regulating the nuclear export of autophagy proteins and other cargoes. Embo Rep. 2019;20(11):e48150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Taagepera S, McDonald D, Loeb JE, et al. Nuclear‐cytoplasmic shuttling of C‐ABL tyrosine kinase. Proc Natl Acad Sci USA. 1998;95(13):7457‐7462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Dominguez D, Montserrat‐Sentis B, Virgos‐Soler A, et al. Phosphorylation regulates the subcellular location and activity of the snail transcriptional repressor. Mol Cell Biol. 2003;23(14):5078‐5089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Azizian NG, Li YL. XPO1‐dependent nuclear export as a target for cancer therapy. J Hematol Oncol. 2020;13(1):61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Kirli K, Karaca S, Dehne HJ, et al. A deep proteomics perspective on CRM1‐mediated nuclear export and nucleocytoplasmic partitioning. Elife. 2015;4:e11466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Johnson AW, Lund E, Dahlberg J. Nuclear export of ribosomal subunits. Trends Biochem Sci. 2002;27(11):580‐585. [DOI] [PubMed] [Google Scholar]
  • 48. Muqbil I, Bao B, Abou‐Samra AB, Mohammad RM, Azmi AS. Nuclear export mediated regulation of microRNAs: potential target for drug intervention. Curr Drug Targets. 2013;14(10):1094‐1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Moy TI, Silver PA. Requirements for the nuclear export of the small ribosomal subunit. J Cell Sci. 2002;115(14):2985‐2995. [DOI] [PubMed] [Google Scholar]
  • 50. Thomas F, Kutay U. Biogenesis and nuclear export of ribosomal subunits in higher eukaryotes depend on the CRM1 export pathway. J Cell Sci. 2003;116(12):2409‐2419. [DOI] [PubMed] [Google Scholar]
  • 51. Tschochner H, Hurt E. Pre‐ribosomes on the road from the nucleolus to the cytoplasm. Trends Cell Biol. 2003;13(5):255‐263. [DOI] [PubMed] [Google Scholar]
  • 52. Will CL, Luhrmann R. Spliceosomal UsnRNP biogenesis, structure and function. Curr Opin Cell Biol. 2001;13(3):290‐301. [DOI] [PubMed] [Google Scholar]
  • 53. Merkwirth C, Jovaisaite V, Durieux J, et al. Two conserved histone demethylases regulate mitochondrial stress‐induced longevity. Cell. 2016;165(5):1209‐1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Sleeman J. A regulatory role for CRM1 in the multi‐directional trafficking of splicing snRNPs in the mammalian nucleus. J Cell Sci. 2007;120(9):1540‐1550. [DOI] [PubMed] [Google Scholar]
  • 55. Ohno M, Segref A, Bachi A, Wilm M, Mattaj IW. PHAX, a mediator of U snRNA nuclear export whose activity is regulated by phosphorylation. Cell. 2000;101(2):187‐198. [DOI] [PubMed] [Google Scholar]
  • 56. Fornerod M, Ohno M, Yoshida M, Mattaj IW. CRM1 is an export receptor for leucine‐rich nuclear export signals. Cell. 1997;90(6):1051‐1060. [DOI] [PubMed] [Google Scholar]
  • 57. Izaurralde E, Kutay U, vonKobbe C, Mattaj IW, Gorlich D. The asymmetric distribution of the constituents of the Ran system is essential for transport into and out of the nucleus. Embo J. 1997;16(21):6535‐6547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Izaurralde E, Lewis J, Gamberi C, Jarmolowski A, Mcguigan C, Mattaj IW. A cap‐binding protein complex mediating U snRNA export. Nature. 1995;376(6542):709‐712. [DOI] [PubMed] [Google Scholar]
  • 59. Herold A, Teixeira L, Izaurralde E. Genome‐wide analysis of nuclear mRNA export pathways in Drosophila. EMBO J. 2003;22(10):2472‐2483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Hodge CA, Colot HV, Stafford P, Cole CN. Rat8p/Dbp5p is a shuttling transport factor that interacts with Rat7p/Nup159p and Gle1p and suppresses the mRNA export defect of xpo1‐1 cells. Embo J. 1999;18(20):5778‐5788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Okamura M, Inose H, Masuda S. RNA export through the NPC in eukaryotes. Genes. 2015;6(1):124‐149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Yang J, Bogerd HP, Wang PJ, Page DC, Cullen BR. Two closely related human nuclear export factors utilize entirely distinct export pathways. Mol Cell. 2001;8(2):397‐406. [DOI] [PubMed] [Google Scholar]
  • 63. Siddiqui N, Borden KL. mRNA export and cancer. Wiley Interdiscip Rev RNA. 2012;3(1):13‐25. [DOI] [PubMed] [Google Scholar]
  • 64. Lee Y, Kim M, Han J, et al. MicroRNA genes are transcribed by RNA polymerase II. EMBO J. 2004;23(20):4051‐4060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Denli AM, Tops BB, Plasterk RH, Ketting RF, Hannon GJ. Processing of primary microRNAs by the microprocessor complex. Nature. 2004;432(7014):231‐235. [DOI] [PubMed] [Google Scholar]
  • 66. Gregory RI, Yan KP, Amuthan G, et al. The microprocessor complex mediates the genesis of microRNAs. Nature. 2004;432(7014):235‐240. [DOI] [PubMed] [Google Scholar]
  • 67. Han J, Lee Y, Yeom KH, Kim YK, Jin H, Kim VN. The Drosha‐DGCR8 complex in primary microRNA processing. Genes Dev. 2004;18(24):3016‐3027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Yi R, Qin Y, Macara IG, Cullen BR. Exportin‐5 mediates the nuclear export of pre‐microRNAs and short hairpin RNAs. Genes Dev. 2003;17(24):3011‐3016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Lund E, Guttinger S, Calado A, Dahlberg JE, Kutay U. Nuclear export of microRNA precursors. Science. 2004;303(5654):95‐98. [DOI] [PubMed] [Google Scholar]
  • 70. Martinez I, Hayes KE, Barr JA, et al. An Exportin‐1‐dependent microRNA biogenesis pathway during human cell quiescence. Proc Natl Acad Sci USA. 2017;114(25):E4961‐E4970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Xie M, Li M, Vilborg A, et al. Mammalian 5'‐capped microRNA precursors that generate a single microRNA. Cell. 2013;155(7):1568‐1580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Kudo N, Khochbin S, Nishi K, et al. Molecular cloning and cell cycle‐dependent expression of mammalian CRM1, a protein involved in nuclear export of proteins. J Biol Chem. 1997;272(47):29742‐29751. [DOI] [PubMed] [Google Scholar]
  • 73. Arnaoutov A, Azuma Y, Ribbeck K, et al. Crm1 is a mitotic effector of Ran‐GTP in somatic cells. Nat Cell Biol. 2005;7(6):626‐632. [DOI] [PubMed] [Google Scholar]
  • 74. Arnaoutov A, Dasso M. Ran‐GTP regulates kinetochore attachment in somatic cells. Cell Cycle. 2005;4(9):1161‐1165. [DOI] [PubMed] [Google Scholar]
  • 75. Blanvillain R, Boavida LC, McCormick S, Ow DW. EXPORTIN1 genes are essential for development and function of the gametophytes in Arabidopsis thaliana . Genetics. 2008;180(3):1493‐1500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Liu QY, Jiang Q, Zhang CM. A fraction of Crm1 locates at centrosomes by its CRIME domain and regulates the centrosomal localization of pericentrin. Biochem Biophys Res Commun. 2009;384(3):383‐388. [DOI] [PubMed] [Google Scholar]
  • 77. Noguchi E, Saitoh YH, Sazer S, Nishimoto T. Disruption of the YRB 2 gene retards nuclear protein export, causing a profound mitotic delay, and can be rescued by overexpression of XPO1/CRM1 . J Biochem. 1999;125(3):574‐585. [DOI] [PubMed] [Google Scholar]
  • 78. Funabiki H, Hagan I, Uzawa S, Yanagida M. Cell cycle‐dependent specific positioning and clustering of centromeres and telomeres in fission yeast. J Cell Biol. 1993;121(5):961‐976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Callanan M, Kudo N, Gout S, et al. Developmentally regulated activity of CRM1/XPO1 during early Xenopus embryogenesis . J Cell Sci. 2000;113(3):451‐459. [DOI] [PubMed] [Google Scholar]
  • 80. Nguyen KT, Holloway MP, Altura RA. The CRM1 nuclear export protein in normal development and disease. Int J Biochem Mol Biol. 2012;3(2):137‐151. [PMC free article] [PubMed] [Google Scholar]
  • 81. Yao H. The expression of CRM1 is associated with prognosis in human osteosarcoma. Oncol Rep. 1994;21(1):229‐235. [PubMed] [Google Scholar]
  • 82. Shen A, Wang Y, Zhao Y, Zou L, Sun L, Cheng C. Expression of CRM1 in human gliomas and its significance in p27 expression and clinical prognosis. Neurosurgery. 2009;65(1):153‐159; discussion 159‐160. [DOI] [PubMed] [Google Scholar]
  • 83. Gao W, Lu C, Chen L, Keohavong P. Overexpression of CRM1: a characteristic feature in a transformed phenotype of lung carcinogenesis and a molecular target for lung cancer adjuvant therapy. J Thorac Oncol. 2015;10(5):815‐825. [DOI] [PubMed] [Google Scholar]
  • 84. Noske A, Weichert W, Niesporek S, et al. Expression of the nuclear export protein chromosomal region maintenance/exportin 1/Xpo1 is a prognostic factor in human ovarian cancer. Cancer. 2008;112(8):1733‐1743. [DOI] [PubMed] [Google Scholar]
  • 85. Huang WY, Yue L, Qiu WS, Wang LW, Zhou XH, Sun YJ. Prognostic value of CRM1 in pancreas cancer. Clin Invest Med. 2009;32(6):E315. [PubMed] [Google Scholar]
  • 86. van der Watt PJ, Zemanay W, Govender D, Hendricks DT, Parker MI, Leaner VD. Elevated expression of the nuclear export protein, Crm1 (exportin 1), associates with human oesophageal squamous cell carcinoma. Oncol Rep. 2014;32(2):730‐738. [DOI] [PubMed] [Google Scholar]
  • 87. Zhou F, Qiu WS, Yao RY, et al. CRM1 is a novel independent prognostic factor for the poor prognosis of gastric carcinomas. Med Oncol. 2013;30(4):726. [DOI] [PubMed] [Google Scholar]
  • 88. Kosyna FK, Depping R. Controlling the gatekeeper: therapeutic targeting of nuclear transport. Cells. 2018;7(11):221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Sun QX, Chen XQ, Zhou Q, Burstein E, Yang SY, Jia D. Inhibiting cancer cell hallmark features through nuclear export inhibition. Signal Transduct Tar. 2016;1:16010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Gravina GL, Senapedis W, McCauley D, Baloglu E, Shacham S, Festuccia C. Nucleo‐cytoplasmic transport as a therapeutic target of cancer. J Hematol Oncol. 2014;7:85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Baek HB, Lombard AP, Libertini SJ, et al. XPO1 inhibition by selinexor induces potent cytotoxicity against high grade bladder malignancies. Oncotarget. 2018;9(77):34567‐34581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Zheng Y, Gery S, Sun HB, Shacham S, Kauffman M, Koeffler HP. KPT‐330 inhibitor of XPO1‐mediated nuclear export has anti‐proliferative activity in hepatocellular carcinoma. Cancer Chemother Pharmacol. 2014;74(3):487‐495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Yuan ZL, Guan YJ, Chatterjee D, Chin YE. Stat3 dimerization regulated by reversible acetylation of a single lysine residue. Science. 2005;307(5707):269‐273. [DOI] [PubMed] [Google Scholar]
  • 94. Wang R, Cherukuri P, Luo J. Activation of Stat3 sequence‐specific DNA binding and transcription by p300/CREB‐binding protein‐mediated acetylation. J Biol Chem. 2005;280(12):11528‐11534. [DOI] [PubMed] [Google Scholar]
  • 95. Cheng Y, Holloway MP, Nguyen K, et al. XPO1 (CRM1) inhibition represses STAT3 activation to drive a survivin‐dependent oncogenic switch in triple‐negative breast cancer. Mol Cancer Ther. 2014;13(3):675‐686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Golomb L, Bublik DR, Wilder S, et al. Importin 7 and exportin 1 link c‐Myc and p53 to regulation of ribosomal biogenesis. Mol Cell. 2012;45(2):222‐232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Pourdehnada M, Truitt ML, Siddiqi IN, Ducker GS, Shokat KM, Ruggero D. Myc and mTOR converge on a common node in protein synthesis control that confers synthetic lethality in Myc‐driven cancers. Proc Natl Acad Sci USA. 2013;110(29):11988‐11993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Wendel HG, De Stanchina E, Fridman JS, et al. Survival signalling by Akt and eIF4E in oncogenesis and cancer therapy. Nature. 2004;428(6980):332‐337. [DOI] [PubMed] [Google Scholar]
  • 99. Li S, Fu J, Walker CJ, Yang J, et al. Dual targeting of protein translation and nuclear protein export results in enhanced antimyeloma effects. Blood Adv. 2023;7(12):2926‐2937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Azmi AS, Li Y, Muqbil I, et al. Exportin 1 (XPO1) inhibition leads to restoration of tumor suppressor miR‐145 and consequent suppression of pancreatic cancer cell proliferation and migration. Oncotarget. 2017;8(47):82144‐82155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. El‐Tanani M, Dakir el H, Raynor B, Morgan R. Mechanisms of nuclear export in cancer and resistance to chemotherapy. Cancers. 2016;8(3):35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Conforti F, Wang Y, Rodriguez JA, Alberobello AT, Zhang YW, Giaccone G. Molecular pathways: anticancer activity by inhibition of nucleocytoplasmic shuttling. Clin Cancer Res. 2015;21(20):4508‐4513. [DOI] [PubMed] [Google Scholar]
  • 103. Santivasi WL, Wang H, Wang T, et al. Association between cytosolic expression of BRCA1 and metastatic risk in breast cancer. Brit J Cancer. 2015;113(3):453‐459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Wang Z, Pan B, Yao Y, et al. XPO1 intensifies sorafenib resistance by stabilizing acetylation of NPM1 and enhancing epithelial‐mesenchymal transition in hepatocellular carcinoma. Biomed Pharmacother. 2023;160:114402. [DOI] [PubMed] [Google Scholar]
  • 105. Turner JG, Sullivan DM. CRM1‐mediated nuclear export of proteins and drug resistance in cancer. Curr Med Chem. 2008;15(26):2648‐2655. [DOI] [PubMed] [Google Scholar]
  • 106. Yang JY, Hung MC. A new fork for clinical application: targeting forkhead transcription factors in cancer. Clin Cancer Res. 2009;15(3):752‐757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Yan R, Zhu H, Huang P, et al. Liquidambaric acid inhibits Wnt/beta‐catenin signaling and colon cancer via targeting TNF receptor‐associated factor 2. Cell Rep. 2022;38(5):110319. [DOI] [PubMed] [Google Scholar]
  • 108. Takenaka Y, Fukumori T, Yoshii T, et al. Nuclear export of phosphorylated galectin‐3 regulates its antiapoptotic activity in response to chemotherapeutic drugs. Mol Cell Biol. 2004;24(10):4395‐4406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Santos SN, Junqueira MS, Francisco G, et al. O‐glycan sialylation alters galectin‐3 subcellular localization and decreases chemotherapy sensitivity in gastric cancer. Oncotarget. 2016;7(50):83570‐83587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Fukumori T, Kanayama HO, Raz A. The role of galectin‐3 in cancer drug resistance. Drug Resist Updat. 2007;10(3):101‐108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Chen Y, Camacho SC, Silvers TR, et al. Inhibition of the nuclear export receptor XPO1 as a therapeutic target for platinum‐resistant ovarian cancer. Clin Cancer Res. 2017;23(6):1552‐1563. [DOI] [PubMed] [Google Scholar]
  • 112. Haudek KC, Spronk KJ, Voss PG, Patterson RJ, Wang JL, Arnoys EJ. Dynamics of galectin‐3 in the nucleus and cytoplasm. Biochim Biophys Acta. 2010;1800(2):181‐189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Gillespie KP, Pirnie R, Mesaros C, Blair IA. Cisplatin dependent secretion of immunomodulatory high mobility group box 1 (HMGB1) protein from lung cancer cells. Biomolecules. 2023;13(9):1335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Corno C, Stucchi S, De Cesare M, et al. FoxO‐1 contributes to the efficacy of the combination of the XPO1 inhibitor selinexor and cisplatin in ovarian carcinoma preclinical models. Biochem Pharmacol. 2018;147:93‐103. [DOI] [PubMed] [Google Scholar]
  • 115. Gao W, Liang J, Ye Y, et al. FUT4siRNA augments the chemosensitivity of non‐small cell lung cancer to cisplatin through activation of FOXO1‐induced apoptosis. BMC Cancer. 2020;20(1):895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Song Y, Zou X, Zhang D, Liu S, Duan Z, Liu L. Self‐enforcing HMGB1/NF‐kappaB/HIF‐1alpha feedback loop promotes cisplatin resistance in hepatocellular carcinoma cells. J Cancer. 2020;11(13):3893‐3902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Hazar‐Rethinam M, de Long LM, Gannon OM, et al. A novel E2F/sphingosine kinase 1 axis regulates anthracycline response in squamous cell carcinoma. Clin Cancer Res. 2015;21(2):417‐427. [DOI] [PubMed] [Google Scholar]
  • 118. Hazar‐Rethinam M, de Long LM, Gannon OM, et al. RacGAP1 is a novel downstream effector of E2F7‐dependent resistance to doxorubicin and is prognostic for overall survival in squamous cell carcinoma. Mol Cancer Ther. 2015;14(8):1939‐1950. [DOI] [PubMed] [Google Scholar]
  • 119. Saenz‐Ponce N, Pillay R, de Long LM, et al. Targeting the XPO1‐dependent nuclear export of E2F7 reverses anthracycline resistance in head and neck squamous cell carcinomas. Sci Transl Med. 2018;10(447):eaar7223. [DOI] [PubMed] [Google Scholar]
  • 120. Pyne NJ, El Buri A, Adams DR, Pyne S. Sphingosine 1‐phosphate and cancer. Adv Biol Regul. 2018;68:97‐106. [DOI] [PubMed] [Google Scholar]
  • 121. Johnson KR, Becker KP, Facchinetti MM, Hannun YA, Obeid LM. PKC‐dependent activation of sphingosine kinase 1 and translocation to the plasma membrane. Extracellular release of sphingosine‐1‐phosphate induced by phorbol 12‐myristate 13‐acetate (PMA). J Biol Chem. 2002;277(38):35257‐35262. [DOI] [PubMed] [Google Scholar]
  • 122. Yao C, Wu SJ, Li D, et al. Co‐administration phenoxodiol with doxorubicin synergistically inhibit the activity of sphingosine kinase‐1 (SphK1), a potential oncogene of osteosarcoma, to suppress osteosarcoma cell growth both and. Mol Oncol. 2012;6(4):392‐404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Ren X, Su C, Sphingosine kinase 1 contributes to doxorubicin resistance and glycolysis in osteosarcoma. Mol Med Rep. 2020;22(3):2183‐2190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Cervantes‐Badillo MG, Paredes‐Villa A, Gomez‐Romero V, et al. IFI27/ISG12 downregulates estrogen receptor alpha transactivation by facilitating its interaction with CRM1/XPO1 in breast cancer cells. Front Endocrinol. 2020;11:568375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Wrobel K, Zhao YC, Kulkoyluoglu E, et al. ERalpha‐XPO1 cross talk controls tamoxifen sensitivity in tumors by altering ERK5 cellular localization. Mol Endocrinol. 2016;30(10):1029‐1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Hosford SR, Miller TW. Clinical potential of novel therapeutic targets in breast cancer: CDK4/6, Src, JAK/STAT, PARP, HDAC, and PI3K/AKT/mTOR pathways. Pharmgenomics Pers Med. 2014;7:203‐215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Cui JJ, Germer K, Wu TY, et al. Cross‐talk between HER2 and MED1 regulates tamoxifen resistance of human breast cancer cells. Cancer Res. 2012;72(21):5625‐5634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Giri DK, Ali‐Seyed M, Li LY, et al. Endosomal transport of ErbB‐2: mechanism for nuclear entry of the cell surface receptor. Mol Cell Biol. 2005;25(24):11005‐11018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Ignatov A, Ignatov T, Weissenborn C, et al. G‐protein‐coupled estrogen receptor GPR30 and tamoxifen resistance in breast cancer. Breast Cancer Res Treat. 2011;128(2):457‐466. [DOI] [PubMed] [Google Scholar]
  • 130. Yu T, Yang G, Hou Y, et al. Cytoplasmic GPER translocation in cancer‐associated fibroblasts mediates cAMP/PKA/CREB/glycolytic axis to confer tumor cells with multidrug resistance. Oncogene. 2017;36(15):2131‐2145. [DOI] [PubMed] [Google Scholar]
  • 131. Tian Y, Chen ZH, Wu P, et al. MIR497HG‐derived miR‐195 and miR‐497 mediate tamoxifen resistance via PI3K/AKT signaling in breast cancer. Adv Sci. 2023;10(12):e2204819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Caffa I, Spagnolo V, Vernieri C, et al. Fasting‐mimicking diet and hormone therapy induce breast cancer regression. Nature. 2020;583(7817):620‐624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Kulkoyluoglu‐Cotul E, Smith BP, Wrobel K, et al. Combined targeting of estrogen receptor alpha and XPO1 prevent Akt activation, remodel metabolic pathways and induce autophagy to overcome tamoxifen resistance. Cancers. 2019;11(4):479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Cotul EK, Zuo Q, Santaliz‐Casiano A, et al. Combined targeting of estrogen receptor alpha and Exportin 1 in metastatic breast cancers. Cancers. 2020;12(9):2397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Chen W, Zheng R, Baade PD, et al. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66(2):115‐132. [DOI] [PubMed] [Google Scholar]
  • 136. Wu M, Yuan Y, Pan YY, Zhang Y. Combined gefitinib and pemetrexed overcome the acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non‐small cell lung cancer. Mol Med Rep. 2014;10(2):931‐938. [DOI] [PubMed] [Google Scholar]
  • 137. Sun C, Gao W, Liu J, Cheng H, Hao J. FGL1 regulates acquired resistance to Gefitinib by inhibiting apoptosis in non‐small cell lung cancer. Respir Res. 2020;21(1):210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Zhao G, Wang Q, Zhang Y, et al. DDX17 induces epithelial‐mesenchymal transition and metastasis through the miR‐149‐3p/CYBRD1 pathway in colorectal cancer. Cell Death Dis. 2023;14(1):1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Li K, Mo C, Gong D, et al. DDX17 nucleocytoplasmic shuttling promotes acquired gefitinib resistance in non‐small cell lung cancer cells via activation of beta‐catenin. Cancer Lett. 2017;400:194‐202. [DOI] [PubMed] [Google Scholar]
  • 140. Shin S, Rossow KL, Grande JP, Janknecht R. Involvement of RNA helicases p68 and p72 in colon cancer. Cancer Res. 2007;67(16):7572‐7578. [DOI] [PubMed] [Google Scholar]
  • 141. Liu J, Xiao Q, Xiao J, Niu C, et al. Wnt/beta‐catenin signalling: function, biological mechanisms, and therapeutic opportunities. Signal Transduct Target Ther. 2022;7(1):3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Guan S, Chen X, Chen Y, et al. FOXM1 variant contributes to Gefitinib resistance via activating wnt/beta‐catenin signal pathway in patients with non‐small cell lung cancer. Clin Cancer Res. 2022;28(17):3770‐3784. [DOI] [PubMed] [Google Scholar]
  • 143. Fang X, Gu P, Zhou C, et al. Beta‐catenin overexpression is associated with gefitinib resistance in non‐small cell lung cancer cells. Pulm Pharmacol Ther. 2014;28(1):41‐48. [DOI] [PubMed] [Google Scholar]
  • 144. Gu Z, Du Y, Zhao X, Wang C. Tumor microenvironment and metabolic remodeling in gemcitabine‐based chemoresistance of pancreatic cancer. Cancer Lett. 2021;521:98‐108. [DOI] [PubMed] [Google Scholar]
  • 145. Yang G, Guan W, Cao Z, et al. Integrative genomic analysis of gemcitabine resistance in pancreatic cancer by patient‐derived xenograft models. Clin Cancer Res. 2021;27(12):3383‐3396. [DOI] [PubMed] [Google Scholar]
  • 146. Kazim S, Malafa MP, Coppola D, et al. Selective nuclear export inhibitor KPT‐330 enhances the antitumor activity of gemcitabine in human pancreatic cancer. Mol Cancer Ther. 2015;14(7):1570‐1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Uddin MH, Al‐Hallak MN, Khan HY, et al. Molecular analysis of XPO1 inhibitor and gemcitabine‐nab‐paclitaxel combination in KPC pancreatic cancer mouse model. Clin Transl Med. 2023;13(12):e1513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Quint K, Tonigold M, Di Fazio P, et al. Pancreatic cancer cells surviving gemcitabine treatment express markers of stem cell differentiation and epithelial‐mesenchymal transition. Int J Oncol. 2012;41(6):2093‐2102. [DOI] [PubMed] [Google Scholar]
  • 149. Hosein AN, Brekken RA, Maitra A. Pancreatic cancer stroma: an update on therapeutic targeting strategies. Nat Rev Gastroenterol Hepatol. 2020;17(8):487‐505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Liu XL, Wang BB, Wang Y, et al. Unbiased screening reveals that blocking exportin 1 overcomes resistance to PI3Kalpha inhibition in breast cancer. Signal Transduct Target Ther. 2019;4:49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Khan HY, Ge J, Nagasaka M, et al. Targeting XPO1 and PAK4 in 8505C anaplastic thyroid cancer cells: putative implications for overcoming lenvatinib therapy resistance. Int J Mol Sci. 2019;21(1):237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Nishi K, Yoshida M, Fujiwara D, Nishikawa M, Horinouchi S, Beppu T. Leptomycin‐B targets a regulatory cascade of crm1, a fission yeast nuclear‐protein, involved in control of higher‐order chromosome structure and gene expression. J Biol Chem. 1994;269(9):6320‐6324. [PubMed] [Google Scholar]
  • 153. Wach JY, Guttinger S, Kutay U, Gademann K. The cytotoxic styryl lactone goniothalamin is an inhibitor of nucleocytoplasmic transport. Bioorg Med Chem Lett. 2010;20(9):2843‐2846. [DOI] [PubMed] [Google Scholar]
  • 154. Hamamoto T, Uozumi T, Beppu T. Leptomycins A and B, new antifungal antibiotics. III. Mode of action of leptomycin B on Schizosaccharomyces pombe . J Antibiot. 1985;38(11):1573‐1580. [DOI] [PubMed] [Google Scholar]
  • 155. Koster M, Lykke‐Andersen S, Elnakady YA, et al. Ratjadones inhibit nuclear export by blocking CRM1/exportin 1. Exp Cell Res. 2003;286(2):321‐331. [DOI] [PubMed] [Google Scholar]
  • 156. Ferreira BI, Cautain B, Grenho I, Link W. Small molecule inhibitors of CRM1. Front Pharmacol. 2020;11:625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Kudo N, Wolff B, Sekimoto T, et al. Leptomycin B inhibition of signal‐mediated nuclear export by direct binding to CRM1. Exp Cell Res. 1998;242(2):540‐547. [DOI] [PubMed] [Google Scholar]
  • 158. Lu CW, Shao CX, Cobos E, Singh KP, Gao WM. Chemotherapeutic sensitization of leptomycin B resistant lung cancer cells by pretreatment with doxorubicin. PLoS ONE. 2012;7(3):e32895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Newlands ES, Rustin GJS, Brampton MH. Phase I trial of elactocin. Brit J Cancer. 1996;74(4):648‐649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Hayakawa Y, Sohda KY, Shin‐Ya K, Hidaka T, Seto H. Anguinomycins C and D, new antitumor antibiotics with selective cytotoxicity against transformed cells. J Antibiot. 1995;48(9):954‐961. [DOI] [PubMed] [Google Scholar]
  • 161. Meissner T, Krause E, Vinkemeier U. Ratjadone and leptomycin B block CRM1‐dependent nuclear export by identical mechanisms. Febs Letters. 2004;576(1‐2):27‐30. [DOI] [PubMed] [Google Scholar]
  • 162. Gong LH, Chen XX, Wang H, et al. Piperlongumine induces apoptosis and synergizes with cisplatin or paclitaxel in human ovarian cancer cells. Oxid Med Cell Longev. 2014;2014:906804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Duan CQ, Zhang B, Deng C, et al. Piperlongumine induces gastric cancer cell apoptosis and G2/M cell cycle arrest both in vitro and in vivo. Tumor Biol. 2016;37(8):10793‐10804. [DOI] [PubMed] [Google Scholar]
  • 164. Roh JL, Kim EH, Park JY, Kim JW, Kwon M, Lee BH. Piperlongumine selectively kills cancer cells and increases cisplatin antitumor activity in head and neck cancer. Oncotarget. 2014;5(19):9227‐9238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Mosieniak G, Adamowicz M, Alster O, et al. Curcumin induces permanent growth arrest of human colon cancer cells: link between senescence and autophagy. Mech Ageing Dev. 2012;133(6):444‐455. [DOI] [PubMed] [Google Scholar]
  • 166. Howells LM, Iwuji COO, Irving GRB, et al. Curcumin combined with FOLFOX chemotherapy is safe and tolerable in patients with metastatic colorectal cancer in a randomized phase IIa trial. J Nutr. 2019;149(7):1133‐1139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Wang JY, Wang X, Wang XJ, et al. Curcumin inhibits the growth via Wnt/beta‐catenin pathway in non‐small‐cell lung cancer cells. Eur Rev Med Pharmacol Sci. 2018;22(21):7492‐7499. [DOI] [PubMed] [Google Scholar]
  • 168. Su YJ, Liu LG, Wang YL, et al. Curcumin inhibits the proliferation and invasion of MG‐63 cells through inactivation of the p‐JAK2/p‐STAT3 pathway. Oncotargets Ther. 2019;12:2011‐2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Sinha S, Pal K, Elkhanany A, et al. Plumbagin inhibits tumorigenesis and angiogenesis of ovarian cancer cells in vivo. Int J Cancer. 2013;132(5):1201‐1212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Yan W, Tu B, Liu YY, et al. Suppressive effects of plumbagin on invasion and migration of breast cancer cells via the inhibition of STAT3 signaling and down‐regulation of inflammatory cytokine expressions. Bone Res. 2013;1:362‐370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Fujita E, Nagao Y, Node M, Kaneko K, Nakazawa S, Kuroda H. Antitumor activity of the isodon diterpenoids: structural requirements for the activity. Experientia. 1976;32(2):203‐206. [DOI] [PubMed] [Google Scholar]
  • 172. Wang MY, Lin C, Zhang TM. Cytokinetic effects of oridonin on leukemia‐L1210 cells. Acta Pharmacol Sin. 1985;6(3):195‐198. [PubMed] [Google Scholar]
  • 173. Liu QQ, Chen K, Ye Q, Jiang XH, Sun YW. Oridonin inhibits pancreatic cancer cell migration and epithelial‐mesenchymal transition by suppressing Wnt/beta‐catenin signaling pathway. Cancer Cell Int. 2016;16:57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Mutka SC, Yang WQ, Dong SD, et al. Identification of nuclear export inhibitors with potent anticancer activity in vivo. Cancer Res. 2009;69(2):510‐517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Sakakibara K, Saito N, Sato T, et al. CBS9106 is a novel reversible oral CRM1 inhibitor with CRM1 degrading activity. Blood. 2011;118(14):3922‐3931. [DOI] [PubMed] [Google Scholar]
  • 176. Kalid O, Toledo Warshaviak D, Shechter S, Sherman W, Shacham S. Consensus Induced Fit Docking (cIFD): methodology, validation, and application to the discovery of novel Crm1 inhibitors. J Comput Aided Mol Des. 2012;26(11):1217‐1228. [DOI] [PubMed] [Google Scholar]
  • 177. Mendonca J, Sharma A, Kim HS, et al. Selective inhibitors of nuclear export (SINE) as novel therapeutics for prostate cancer. Oncotarget. 2014;5(15):6102‐6112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Sexton R, Mahdi Z, Chaudhury R, et al. Targeting nuclear exporter protein XPO1/CRM1 in gastric cancer. Int J Mol Sci. 2019;20(19):4826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Etchin J, Sun Q, Kentsis A, et al. Antileukemic activity of nuclear export inhibitors that spare normal hematopoietic cells. Leukemia. 2013;27(1):66‐74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Razak ARA, Mau‐Soerensen M, Gabrail NY, et al. First‐in‐class, first‐in‐human phase I study of selinexor, a selective inhibitor of nuclear export, in patients with advanced solid tumors. J Clin Oncol. 2016;34(34):4142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Alexander TB, Lacayo NJ, Choi JK, Ribeiro RC, Pui CH, Rubnitz JE. Phase I study of selinexor, a selective inhibitor of nuclear export, in combination with fludarabine and cytarabine, in pediatric relapsed or refractory acute leukemia. J Clin Oncol. 2016;34(34):4094‐4101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Green AL, Ramkissoon SH, McCauley D, et al. Preclinical antitumor efficacy of selective exportin 1 inhibitors in glioblastoma. Neuro Oncol. 2015;17(5):697‐707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Attiyeh EF, Maris JM, Lock R, et al. Pharmacodynamic and genomic markers associated with response to the XPO1/CRM1 inhibitor selinexor (KPT‐330): a report from the pediatric preclinical testing program. Pediatr Blood Cancer. 2016;63(2):276‐286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Zhu ZC, Liu JW, Yang C, Zhao M, Xiong ZQ. XPO1 inhibitor KPT‐330 synergizes with Bcl‐xL inhibitor to induce cancer cell apoptosis by perturbing rRNA processing and Mcl‐1 protein synthesis. Cell Death Dis. 2019;10(6):395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Shang EY, Zhang YR, Shu C, et al. Dual inhibition of Bcl‐2/Bcl‐xL and XPO1 is synthetically lethal in glioblastoma model systems. Sci Rep. 2018;8:15383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Wahba A, Rath BH, O'Neill JW, Camphausen K, Tofilon PJ. The XPO1 inhibitor selinexor inhibits translation and enhances the radiosensitivity of glioblastoma cells grown in vitro and in vivo. Mol Cancer Ther. 2018;17(8):1717‐1726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. DeSisto JA, Flannery P, Lemma R, et al. Exportin 1 inhibition induces nerve growth factor receptor expression to inhibit the NF‐kappaB pathway in preclinical models of pediatric high‐grade glioma. Mol Cancer Ther. 2020;19(2):540‐551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Sun H, Hattori N, Chien W, et al. KPT‐330 has antitumour activity against non‐small cell lung cancer. Brit J Cancer. 2014;111(2):281‐291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Wang J, Sun T, Meng Z, et al. XPO1 inhibition synergizes with PARP1 inhibition in small cell lung cancer by targeting nuclear transport of FOXO3a. Cancer Lett. 2021;503:197‐212. [DOI] [PubMed] [Google Scholar]
  • 190. Quintanal‐Villalonga A, Taniguchi H, Hao Y, et al. Inhibition of XPO1 sensitizes small cell lung cancer to first‐ and second‐line chemotherapy. Cancer Res. 2022;82(3):472‐483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Khan HY, Nagasaka M, Li Y, et al. Inhibitor of the nuclear transport protein XPO1 enhances the anticancer efficacy of KRAS G12C inhibitors in preclinical models of KRAS G12C‐mutant cancers. Cancer Res Commun. 2022;2(5):342‐352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Kim J, McMillan E, Kim HS, et al. XPO1‐dependent nuclear export is a druggable vulnerability in KRAS‐mutant lung cancer. Nature. 2016;538(7623):114‐117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Rosen JC, Weiss J, Pham NA, et al. Antitumor efficacy of XPO1 inhibitor Selinexor in KRAS‐mutant lung adenocarcinoma patient‐derived xenografts. Transl Oncol. 2021;14(10):101179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Depping R, von Fallois M, Landesman Y, Kosyna FK. The nuclear export inhibitor selinexor inhibits hypoxia signaling pathways and 3D spheroid growth of cancer cells. Onco Targets Ther. 2019;12:8387‐8399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195. Arango NP, Yuca E, Zhao M, et al. Selinexor (KPT‐330) demonstrates anti‐tumor efficacy in preclinical models of triple‐negative breast cancer. Breast Cancer Res. 2017;19(1):93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Miyake T, Pradeep S, Wu SY, et al. XPO1/CRM1 inhibition causes antitumor effects by mitochondrial accumulation of eIF5A. Clin Cancer Res. 2015;21(14):3286‐3297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197. Rashid NS, Hairr NS, Murray G, et al. Identification of nuclear export inhibitor‐based combination therapies in preclinical models of triple‐negative breast cancer. Transl Oncol. 2021;14(12):101235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Martini S, Figini M, Croce A, et al. Selinexor sensitizes TRAIL‐R2‐positive TNBC cells to the activity of TRAIL‐R2xCD3 bispecific antibody. Cells. 2020;9(10):2231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Marijon H, Gery S, Chang H, et al. Selinexor, a selective inhibitor of nuclear export, enhances the anti‐tumor activity of olaparib in triple negative breast cancer regardless of BRCA1 mutation status. Oncotarget. 2021;12(18):1749‐1762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Martini S, Zuco V, Tortoreto M, et al. miR‐34a‐mediated survivin inhibition improves the antitumor activity of selinexor in triple‐negative breast cancer. Pharmaceuticals. 2021;14(6):523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Shi Y, Xu S, Li S. Selinexor improves the anti‐cancer effect of tucidinostat on TP53 wild‐type breast cancer. Mol Cell Endocrinol. 2022;545:111558. [DOI] [PubMed] [Google Scholar]
  • 202. Gao J, Azmi AS, Aboukameel A, Kauffman M, Shacham S, Abou‐Samra AB, Mohammad RM. Nuclear retention of Fbw7 by specific inhibitors of nuclear export leads to Notch1 degradation in pancreatic cancer. Oncotarget. 2014;5(11):3444‐3454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203. Azmi AS, Khan HY, Muqbil I, et al. Preclinical assessment with clinical validation of selinexor with gemcitabine and nab‐paclitaxel for the treatment of pancreatic ductal adenocarcinoma. Clin Cancer Res. 2020;26(6):1338‐1348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Wu T, Chen W, Zhong Y, et al. Nuclear export of ubiquitinated proteins determines the sensitivity of colorectal cancer to proteasome inhibitor. Mol Cancer Ther. 2017;16(4):717‐728. [DOI] [PubMed] [Google Scholar]
  • 205. Inoue A, Robinson FS, Minelli R, et al. Sequential administration of XPO1 and ATR inhibitors enhances therapeutic response in TP53‐mutated colorectal cancer. Gastroenterology. 2021;161(1):196‐210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206. von Fallois M, Depping R. Radiation response enhanced by inhibition of XPO1 in preclinical rectal cancer models. Strahlenther Onkol. 2016;192(12):961‐962. [DOI] [PubMed] [Google Scholar]
  • 207. Ferreiro‐Neira I, Torres NE, Liesenfeld LF, et al. XPO1 inhibition enhances radiation response in preclinical models of rectal cancer. Clin Cancer Res. 2016;22(7):1663‐1673. [DOI] [PubMed] [Google Scholar]
  • 208. Seervi M, Sumi S, Chandrasekharan A, Sharma AK, SanthoshKumar TR. Molecular profiling of anastatic cancer cells: potential role of the nuclear export pathway. Cell Oncol. 2019;42(5):645‐661. [DOI] [PubMed] [Google Scholar]
  • 209. Alzahrani A, Natarajan U, Rathinavelu A. Enhancement of MDM2 inhibitory effects through blocking nuclear export mechanisms in ovarian cancer cells. Cancer Genet. 2022;266:57‐68. [DOI] [PubMed] [Google Scholar]
  • 210. Handley KF, Rodriguez‐Aguayo C, Ma S, et al. Rational combination of CRM1 inhibitor selinexor and olaparib shows synergy in ovarian cancer cell lines and mouse models. Mol Cancer Ther. 2021;20(12):2352‐2361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. Miyake TM, Pradeep S, Bayraktar E, et al. NRG1/ERBB3 pathway activation induces acquired resistance to XPO1 inhibitors. Mol Cancer Ther. 2020;19(8):1727‐1735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212. Gravina GL, Tortoreto M, Mancini A, et al. XPO1/CRM1‐Selective Inhibitors of Nuclear Export (SINE) reduce tumor spreading and improve overall survival in preclinical models of prostate cancer (PCa), J Hematol Oncol. 2014;7:46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213. Gravina GL, Mancini A, Colapietro A, et al. Correction: pharmacological treatment with inhibitors of nuclear export enhances the antitumor activity of docetaxel in human prostate cancer. Oncotarget. 2019;10(59):6393‐6395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Muqbil I, Li YW, Baloglu E, et al. Down‐regulation of AR splice variants through XPO1 suppression contributes to the inhibition of prostate cancer progression. Clin Cancer Res. 2018;78(13):35327‐35342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215. Uddin MH, Li Y, Khan HY, et al. Nuclear export inhibitor KPT‐8602 synergizes with PARP inhibitors in escalating apoptosis in castration resistant cancer cells. Int J Mol Sci. 2021;22(13):6676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216. Nakayama R, Zhang YX, Czaplinski JT, et al. Preclinical activity of selinexor, an inhibitor of XPO1, in sarcoma. Oncotarget. 2016;7(13):16581‐16592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. Meng W, Gao SJ. Targeting XPO1 enhances innate immune response and inhibits KSHV lytic replication during primary infection by nuclear stabilization of the p62 autophagy adaptor protein. Cell Death Dis. 2021;12(1):29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218. Breitbach JT, Louke DS, Tobin SJ, Watts MR, Davies AE, Fenger JM. The selective inhibitor of nuclear export (SINE) verdinexor exhibits biologic activity against canine osteosarcoma cell lines. Vet Comp Oncol. 2021;19(2):362‐373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219. Sun H, Lin DC, Cao Q, et al. CRM1 inhibition promotes cytotoxicity in Ewing sarcoma cells by repressing EWS‐FLI1‐dependent IGF‐1 signaling. Cancer Res. 2016;76(9):2687‐2697. [DOI] [PubMed] [Google Scholar]
  • 220. Nair JS, Musi E, Schwartz GK. Selinexor (KPT‐330) induces tumor suppression through nuclear sequestration of IkappaB and downregulation of survivin. Clin Cancer Res. 2017;23(15):4301‐4311. [DOI] [PubMed] [Google Scholar]
  • 221. Garg M, Kanojia D, Mayakonda A, et al. Molecular mechanism and therapeutic implications of selinexor (KPT‐330) in liposarcoma. Oncotarget. 2017;8(5):7521‐7532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222. Zuco V, Pasquali S, Tortoreto M, et al. Selinexor versus doxorubicin in dedifferentiated liposarcoma PDXs: evidence of greater activity and apoptotic response dependent on p53 nuclear accumulation and survivin down‐regulation. J Exp Clin Cancer Res. 2021;40(1):83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223. Jeitany M, Prabhu A, Dakle P, et al. Novel carfilzomib‐based combinations as potential therapeutic strategies for liposarcomas. Cell Mol Life Sci. 2021;78(4):1837‐1851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224. von Fallois M, Kosyna FK, Mandl M, Landesman Y, Dunst J, Depping R. Selinexor decreases HIF‐1alpha via inhibition of CRM1 in human osteosarcoma and hepatoma cells associated with an increased radiosensitivity. J Cancer Res Clin Oncol. 2021;147(7):2025‐2033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225. Yang J, Bill MA, Young GS, et al. Novel small molecule XPO1/CRM1 inhibitors induce nuclear accumulation of TP53, phosphorylated MAPK and apoptosis in human melanoma cells. PLoS One. 2014;9(7):e102983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226. Breit MN, Kisseberth WC, Bear MD, et al. Biologic activity of the novel orally bioavailable selective inhibitor of nuclear export (SINE) KPT‐335 against canine melanoma cell lines. BMC Vet Res. 2014;10:160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227. Tyler PM, Servos MM, de Vries RC, et al. Clinical dosing regimen of selinexor maintains normal immune homeostasis and t‐cell effector function in mice: implications for combination with immunotherapy. Mol Cancer Ther. 2017;16(3):428‐439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228. Farren MR, Hennessey RC, et al. The Exportin‐1 inhibitor selinexor exerts superior antitumor activity when combined with T‐cell checkpoint inhibitors. Mol Cancer Ther. 2017;16(3):417‐427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229. Lassman AB, Wen PY, van den Bent MJ, et al. A phase II study of the efficacy and safety of oral selinexor in recurrent glioblastoma. Clin Cancer Res. 2022;28(3):452‐460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230. Shafique M, Ismail‐Khan R, Extermann M, et al. A phase II trial of selinexor (KPT‐330) for metastatic triple‐negative breast cancer. Oncologist. 2019;24(7):887. e416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231. Westin SN, Fu S, Tsimberidou A, et al. Selinexor in combination with weekly paclitaxel in patients with metastatic solid tumors: results of an open label, single‐center, multi‐arm phase 1b study with expansion phase in ovarian cancer. Gynecol Oncol. 2023;168:76‐82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Heong VYM, Goh ML, Yong WP, et al. Phase Ib study of safety and tolerability of selinexor in Asian patients with advanced solid cancers. Dev Ther. 2018;29:VIII141. [Google Scholar]
  • 233. Azmi AS, Khan HY, Muqbil I, et al. Preclinical assessment with clinical validation of selinexor with gemcitabine and nab‐paclitaxel for the treatment of pancreatic ductal adenocarcinoma. Clin Cancer Res. 2020;26(6):1338‐1348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234. Heong VYM, Koe P, Yong WP, et al. RAS/AKT pathway mutations as predictive biomarkers in patients with colorectal cancer treated with the exportin 1 (XPO1) inhibitor selinexor (SEL)—inhibition of nuclear‐cytoplasmic translocation of p27 as a mechanism of anti‐tumour activity. Ann Oncol. 2016;27:VI122. [Google Scholar]
  • 235. Nilsson S, Stein A, Rolfo C, et al. Selinexor (KPT‐330), an oral selective inhibitor of nuclear export (SINE) compound, in combination with FOLFOX in patients with metastatic colorectal cancer (mCRC)—final results of the phase I trial SENTINEL. Curr Cancer Drug Targets. 2020;20(10):811‐817. [DOI] [PubMed] [Google Scholar]
  • 236. Lawrence YR, Shacham‐Shmueli E, et al. Nuclear export inhibition for radiosensitization: a proof‐of‐concept phase 1 clinical trial of selinexor (KPT‐330) combined with neoadjuvant chemoradiation in locally advanced rectal cancer. Int J Radiat Oncol Biol Phys. 2022;114(2):250‐255. [DOI] [PubMed] [Google Scholar]
  • 237. Golan T, Kutiel TS, Geva R, et al. Open‐label phase 1 study evaluating the tolerability and anti‐tumor activity of selinexor and pembrolizumab in colorectal cancer. J Clin Oncol. 2021;39(15):e15579. [Google Scholar]
  • 238. Wei XX, Siegel AP, Aggarwal R, et al. A phase II trial of selinexor, an oral selective inhibitor of nuclear export compound, in abiraterone‐ and/or enzalutamide‐refractory metastatic castration‐resistant prostate cancer. Oncologist. 2018;23(6):656. e64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239. Vergote IB, Lund B, Peen U, et al. Phase 2 study of the Exportin 1 inhibitor selinexor in patients with recurrent gynecological malignancies. Gynecol Oncol. 2020;156(2):308‐314. [DOI] [PubMed] [Google Scholar]
  • 240. Thein KZ, Piha‐Paul SA, Tsimberidou A, et al. Selinexor in combination with topotecan in patients with advanced or metastatic solid tumors: results of an open‐label, single‐center, multi‐arm phase Ib study. Invest New Drugs. 2021;39(5):1357‐1365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241. Rubinstein MM, Grisham RN, Cadoo K, et al. A phase I open‐label study of selinexor with paclitaxel and carboplatin in patients with advanced ovarian or endometrial cancers. Gynecol Oncol. 2021;160(1):71‐76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242. Vergote I, Perez‐Fidalgo JA, Hamilton EP, et al. Oral selinexor as maintenance therapy after first‐line chemotherapy for advanced or recurrent endometrial cancer. J Clin Oncol. 2023;41(35):5400‐5410. [DOI] [PubMed] [Google Scholar]
  • 243. Gounder MM, Zer A, Tap WD, et al. Phase IB Study of selinexor, a first‐in‐class inhibitor of nuclear export, in patients with advanced refractory bone or soft tissue sarcoma. J Clin Oncol. 2016;34(26):3166‐3174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244. Gounder MM, Razak AA, Somaiah N, et al. Selinexor in advanced, metastatic dedifferentiated liposarcoma: a multinational, randomized, double‐blind, placebo‐controlled trial. J Clin Oncol. 2022;40(22):2479‐2490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245. Lewin J, Malone E, Al‐Ezzi E, et al. A phase 1b trial of selinexor, a first‐in‐class selective inhibitor of nuclear export (SINE), in combination with doxorubicin in patients with advanced soft tissue sarcomas (STS). Eur J Cancer. 2021;144:360‐367. [DOI] [PubMed] [Google Scholar]
  • 246. Kendra KL, Watson R, Lesinski GB. Selinexor, a selective inhibitor of nuclear export (SINE), in patients with unresectable melanoma. J Clin Oncol. 2017;35:78883‐78895. [Google Scholar]
  • 247. Kala J, Mamlouk O, Jhaveri KD. Selinexor‐associated hyponatremia: single‐center, real‐world data. Kidney Int. 2020;98(3):789‐791. [DOI] [PubMed] [Google Scholar]
  • 248. Bobillo S, Abrisqueta P, Carpio C, et al. Promising activity of selinexor in the treatment of a patient with refractory diffuse large B‐cell lymphoma and central nervous system involvement. Haematologica. 2018;103(2):e92‐e93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249. Lee S, Mohan S, Knupp J, et al. Oral eltanexor treatment of patients with higher‐risk myelodysplastic syndrome refractory to hypomethylating agents. J Hematol Oncol. 2022;15(1):103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250. Cornell RF, Baz R, Richter JR, et al. A phase 1 clinical trial of oral eltanexor in patients with relapsed or refractory multiple myeloma. Am J Hematol. 2022;97(2):E54‐E58. [DOI] [PubMed] [Google Scholar]
  • 251. Machlus KR, Wu SK, Vijey P, et al. Selinexor‐induced thrombocytopenia results from inhibition of thrombopoietin signaling in early megakaryopoiesis. Blood. 2017;130(9):1132‐1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252. Lin DC, Hao JJ, Nagata Y, et al. Genomic and molecular characterization of esophageal squamous cell carcinoma. Nat Genet. 2014;46(5):467‐473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253. Kwanten B, Deconick T, Walker C, Wang F, Landesman Y, Daelemans D. E3 ubiquitin ligase ASB8 promotes selinexor‐induced proteasomal degradation of XPO1. Biomed Pharmacother. 2023;160:114305. [DOI] [PubMed] [Google Scholar]
  • 254. Wang X, Xu J, Li Q, et al. RNA‐binding protein hnRNPU regulates multiple myeloma resistance to selinexor. Cancer Lett. 2024;580:216486. [DOI] [PubMed] [Google Scholar]
  • 255. Lin KH, Rutter JC, Xie A, et al. P2RY2‐AKT activation is a therapeutically actionable consequence of XPO1 inhibition in acute myeloid leukemia. Nat Cancer. 2022;3(7):837‐851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256. Emdal KB, Palacio‐Escat N, Wigerup C, et al. Phosphoproteomics of primary AML patient samples reveals rationale for AKT combination therapy and p53 context to overcome selinexor resistance. Cell Rep. 2022;40(6):111177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257. Restrepo P, Bhalla S, Ghodke‐Puranik Y, et al. A three‐gene signature predicts response to selinexor in multiple myeloma. JCO Precis Oncol. 2022;6:e2200147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258. Totiger TM, Chaudhry S, Musi E, et al. Protein biomarkers for response to XPO1 inhibition in haematologic malignancies. J Cell Mol Med. 2023;27(4):587‐590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259. Neggers JE, Vercruysse T, Jacquemyn M, et al. Identifying drug‐target selectivity of small‐molecule CRM1/XPO1 inhibitors by CRISPR/Cas9 genome editing. Chem Biol. 2015;22(1):107‐116. [DOI] [PubMed] [Google Scholar]
  • 260. Neggers JE, Vanstreels E, Baloglu E, Shacham S, Landesman Y, Daelemans D. Heterozygous mutation of cysteine in XPO1 is sufficient for resistance to selective inhibitors of nuclear export. Oncotarget. 2016;7(42):68842‐68850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261. Rosin‐Arbesfeld R, Townsley F, Bienz M. The APC tumour suppressor has a nuclear export function. Nature. 2000;406(6799):1009‐1012. [DOI] [PubMed] [Google Scholar]
  • 262. Aladhraei M, Al‐Salami E, Poungvarin N, Suwannalert P. The roles of p53 and XPO1 on colorectal cancer progression in Yemeni patients. J Gastrointest Oncol. 2019;10(3):437‐444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263. Zhao C, Yang ZY, Zhang J, et al. Inhibition of XPO1 with KPT‐330 induces autophagy‐dependent apoptosis in gallbladder cancer by activating the p53/mTOR pathway. J Transl Med. 2022;20(1):434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264. Pan LJ, Cheng C, Duan PW, Chen K, Wu YM, Wu ZX. XPO1/CRM1 is a promising prognostic indicator for neuroblastoma and represented a therapeutic target by selective inhibitor verdinexor. J Exp Clin Canc Res. 2021;40(1):255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265. Gupta A, Saltarski JM, White MA, Scaglioni PP, Gerber DE. Therapeutic targeting of nuclear export inhibition in lung cancer. J Thorac Oncol. 2017;12(9):1446‐1450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266. Currier AW, Kolb EA, Gorlick RG, Roth ME, Gopalakrishnan V, Sampson VB. p27/Kip1 functions as a tumor suppressor and oncoprotein in osteosarcoma. Sci Rep. 2019;9:6161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267. Luo J, Chen Y, Li Q, Wang B, Zhou Y, Lan H. CRM‐1 knockdown inhibits extrahepatic cholangiocarcinoma tumor growth by blocking the nuclear export of p27Kip1. Int J Mol Med. 2016;38(2):381‐390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268. Miyake T, Pradeep S, Wu SY, et al. XPO1/CRM1 inhibition causes antitumor effects by mitochondrial accumulation of eIF5A. Clin Cancer Res. 2015;21(14):3286‐3297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269. Pauty J, Couturier AM, Rodrigue A, et al. Cancer‐causing mutations in the tumor suppressor PALB2 reveal a novel cancer mechanism using a hidden nuclear export signal in the WD40 repeat motif. Nucleic Acids Res. 2017;45(5):2644‐2657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270. Jones S, Hruban RH, Kamiyama M, et al. Exomic sequencing identifies PALB2 as a pancreatic cancer susceptibility gene. Science. 2009;324(5924):217‐217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271. Barazeghi E, Prabhawa S, Norlen O, Hellman P, Stalberg P, Westin G. Decrease of 5‐hydroxymethylcytosine and TET1 with nuclear exclusion of TET2 in small intestinal neuroendocrine tumors. BMC Cancer. 2018;18(1):764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272. Azmi AS, Aboukameel A, Bao B, et al. Selective inhibitors of nuclear export block pancreatic cancer cell proliferation and reduce tumor growth in mice. Gastroenterology. 2013;144(2):447‐456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273. Shin D, Jeon JH, Jeong M, et al. VDUP1 mediates nuclear export of HIF1alpha via CRM1‐dependent pathway. Biochim Biophys Acta. 2008;1783(5):838‐848. [DOI] [PubMed] [Google Scholar]
  • 274. Chen‐Shtoyerman R, Theodor L, Harmati E, et al. Genetic analysis of familial colorectal cancer in Israeli Arabs. Hum Mutat. 2003;21(4):446‐447. [DOI] [PubMed] [Google Scholar]
  • 275. Cho YG, Kim CJ, Park CH, et al. Genetic alterations of the KLF6 gene in gastric cancer. Oncogene. 2005;24(28):4588‐4590. [DOI] [PubMed] [Google Scholar]
  • 276. Narla G, Heath KE, Reeves HL, et al. KLF6, a candidate tumor suppressor gene mutated in prostate cancer. Science. 2001;294(5551):2563‐2566. [DOI] [PubMed] [Google Scholar]
  • 277. Kumari G, Mahalingam S. Extracellular signal‐regulated kinase 2 (ERK‐2) mediated phosphorylation regulates nucleo‐cytoplasmic shuttling and cell growth control of Ras‐associated tumor suppressor protein, RASSF2. Exp Cell Res. 2009;315(16):2775‐2790. [DOI] [PubMed] [Google Scholar]
  • 278. O'Malley S, Su H, Zhang T, Ng C, Ge H, Tang CK. TOB suppresses breast cancer tumorigenesis. Int J Cancer. 2009;125(8):1805‐1813. [DOI] [PubMed] [Google Scholar]
  • 279. Gravina GL, Mancini A, Sanita P, et al. KPT‐330, a potent and selective exportin‐1 (XPO‐1) inhibitor, shows antitumor effects modulating the expression of cyclin D1 and survivin [corrected] in prostate cancer models. BMC Cancer. 2015;15:941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280. Chesi M, Bergsagel PL, Brents LA, Smith CM, Gerhard DS, Kuehl WM. Dysregulation of cyclin D1 by translocation into an IgH gamma switch region in two multiple myeloma cell lines. Blood. 1996;88(2):674‐681. [PubMed] [Google Scholar]
  • 281. Deng ZH, Gomez TS, Osborne DG, Phillips‐Krawczak CA, Zhang JS, Billadeau DD. Nuclear FAM21 participates in NF‐kappaB‐dependent gene regulation in pancreatic cancer cells. J Cell Sci. 2015;128(2):373‐384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282. Saji M, Vasko V, Kada F, Allbritton EH, Burman KD, Ringel MD. Akt1 contains a functional leucine‐rich nuclear export sequence. Biochem Biophys Res Commun. 2005;332(1):167‐173. [DOI] [PubMed] [Google Scholar]
  • 283. Hahn SA, Schutte M, Hoque ATMS, et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science. 1996;271(5247):350‐353. [DOI] [PubMed] [Google Scholar]
  • 284. Sjoblom T, Jones S, Wood LD, et al. The consensus coding sequences of human breast and colorectal cancers. Science. 2006;314(5797):268‐274. [DOI] [PubMed] [Google Scholar]
  • 285. Falini B, Brunetti L, Sportoletti P, Martelli MP. NPM1‐mutated acute myeloid leukemia: from bench to bedside. Blood. 2020;136(15):1707‐1721. [DOI] [PubMed] [Google Scholar]
  • 286. Zhang J, Xu X, Chen Y, Guan X, Zhu H, Qi Y. The abnormal expression of chromosomal region maintenance 1 (CRM1)‐survivin axis in ovarian cancer and its related mechanisms regulating proliferation and apoptosis of ovarian cancer cells. Bioengineered. 2022;13(1):624‐633. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 287. Gravina GL, Marampon F, Sherris D, et al. Torc1/Torc2 inhibitor, Palomid 529, enhances radiation response modulating CRM1‐mediated survivin function and delaying DNA repair in prostate cancer models. Prostate. 2014;74(8):852‐868. [DOI] [PubMed] [Google Scholar]
  • 288. Pockwinse SM, Rajgopal A, Young DW, et al. Microtubule‐dependent nuclear‐cytoplasmic shuttling of Runx2. J Cell Physiol. 2006;206(2):354‐362. [DOI] [PubMed] [Google Scholar]
  • 289. Chuang LSH, Matsuo J, Douchi D, Bte Mawan NA, Ito Y. RUNX3 in stem cell and cancer biology. Cells. 2023;12(3):408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290. Liu X, Chong Y, Liu H, Han Y, Niu M. Novel reversible selective inhibitor of CRM1 for targeted therapy in ovarian cancer. J Ovarian Res. 2015;8:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291. Nie D, Xiao X, Chen J, et al. Prognostic and therapeutic significance of XPO1 in T‐cell lymphoma. Exp Cell Res. 2022;416(2):113180. [DOI] [PubMed] [Google Scholar]
  • 292. Xu Z, Pan B, Miao Y, et al. Prognostic value and therapeutic targeting of XPO1 in chronic lymphocytic leukemia. Clin Exp Med. 2023;23(6):2651‐2662. [DOI] [PubMed] [Google Scholar]
  • 293. Turner JG, Kashyap T, Dawson JL, et al. XPO1 inhibitor combination therapy with bortezomib or carfilzomib induces nuclear localization of IkBa and overcomes acquired proteasome inhibitor resistance in human multiple myeloma. Oncotarget. 2016;7(48):78896‐78909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294. Wang XZ, Feng Y, Han YF, et al. Budlein A methylacrylate demonstrates potent activity against triple‐negative breast cancer by targeting IkappaBalpha kinase and exportin‐1. Toxicol Appl Pharmacol. 2020;408:115263. [DOI] [PubMed] [Google Scholar]
  • 295. Kudo N, Matsumori N, Taoka H, et al. Leptomycin B inactivates CRM1/exportin 1 by covalent modification at a cysteine residue in the central conserved region. Proc Natl Acad Sci USA. 1999;96(16):9112‐9117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296. Meissner T, Krause E, Vinkemeier U. Ratjadone and leptomycin B block CRM1‐dependent nuclear export by identical mechanisms. FEBS Lett. 2004;576(1‐2):27‐30. [DOI] [PubMed] [Google Scholar]
  • 297. Bonazzi S, Eidam O, Guttinger S, et al. Anguinomycins and derivatives: total syntheses, modeling, and biological evaluation of the inhibition of nucleocytoplasmic transport. J Am Chem Soc. 2010;132(4):1432‐1442. [DOI] [PubMed] [Google Scholar]
  • 298. Niu MS, Xu XY, Shen YL, et al. Piperlongumine is a novel nuclear export inhibitor with potent anticancer activity. Chem‐Biol Interact. 2015;237:66‐72. [DOI] [PubMed] [Google Scholar]
  • 299. Niu MS, Wu SJ, Mao L, Yang YL. CRM1 is a cellular target of curcumin: new insights for the myriad of biological effects of an ancient spice. Traffic. 2013;14(10):1042‐1052. [DOI] [PubMed] [Google Scholar]
  • 300. Liu XJ, Niu MS, Xu XY, et al. CRM1 is a direct cellular target of the natural anti‐cancer agent plumbagin. J Pharmacol Sci. 2014;124(4):486‐493. [DOI] [PubMed] [Google Scholar]
  • 301. Newlands ES, Rustin GJ, Brampton MH. Phase I trial of elactocin. Br J Cancer. 1996;74(4):648‐649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302. Kalesse M, Christmann M, Bhatt U, et al. The chemistry and biology of ratjadone. Chembiochem. 2001;2(9):709‐714. [DOI] [PubMed] [Google Scholar]
  • 303. Pei S, Minhajuddin M, Callahan KP, et al. Targeting aberrant glutathione metabolism to eradicate human acute myelogenous leukemia cells. J Biol Chem. 2013;288(47):33542‐33558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304. Heidari H, Bagherniya M, Majeed M, Sathyapalan T, Jamialahmadi T, Sahebkar A. Curcumin‐piperine co‐supplementation and human health: a comprehensive review of preclinical and clinical studies. Phytother Res. 2023;37(4):1462‐1487. [DOI] [PubMed] [Google Scholar]
  • 305. Nelson KM, Dahlin JL, Bisson J, et al. The essential medicinal chemistry of curcumin. J Med Chem. 2017;60(5):1620‐1637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306. Thakor N, Janathia B. Plumbagin: a potential candidate for future research and development. Curr Pharm Biotechnol. 2022;23(15):1800‐1812. [DOI] [PubMed] [Google Scholar]
  • 307. Li FF, Yi S, Wen L, et al. Oridonin induces NPM mutant protein translocation and apoptosis in NPM1c+ acute myeloid leukemia cells in vitro. Acta Pharmacol Sin. 2014;35(6):806‐813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308. Li X, Zhang CT, Ma W, Xie X, Huang Q. Oridonin: a review of its pharmacology, pharmacokinetics and toxicity. Front Pharmacol. 2021;12:645824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309. Gravina GL, Mancini A, et al. Erratum to: KPT‐330, a potent and selective exportin‐1 (XPO‐1) inhibitor, shows antitumor effects modulating the expression of cyclin D1 and survivin in prostate cancer models. BMC Cancer. 2016;16:8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310. Wei N, Song Y, Zhang F, Sun Z, Zhang X. Transcriptome profiling of acquired gefitinib resistant lung cancer cells reveals dramatically changed transcription programs and new treatment targets. Front Oncol. 2020;10:1424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311. Wang S, Han X, Wang J, Yao J, Shi Y. Antitumor effects of a novel chromosome region maintenance 1 (CRM1) inhibitor on non‐small cell lung cancer cells in vitro and in mouse tumor xenografts. PLoS One. 2014;9(3):e89848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312. Soung YH, Kashyap T, Nguyen T, et al. Selective inhibitors of nuclear export (SINE) compounds block proliferation and migration of triple negative breast cancer cells by restoring expression of ARRDC3. Oncotarget. 2017;8(32):52935‐52947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313. Subhash VV, Yeo MS, Wang L, et al. Anti‐tumor efficacy of selinexor (KPT‐330) in gastric cancer is dependent on nuclear accumulation of p53 tumor suppressor. Sci Rep. 2018;8(1):12248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314. Gao J, Azmi AS, Aboukameel A, et al. Nuclear retention of Fbw7 by specific inhibitors of nuclear export leads to Notch1 degradation in pancreatic cancer. Oncotarget. 2014;5(11):3444‐3454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315. Chen Y, Camacho SC, Silvers TR, et al. Inhibition of the nuclear export receptor XPO1 as a therapeutic target for platinum‐resistant ovarian cancer. Clin Cancer Res. 2017;23(6):1552‐1563. [DOI] [PubMed] [Google Scholar]
  • 316. Inoue H, Kauffman M, Shacham S, et al. CRM1 blockade by selective inhibitors of nuclear export attenuates kidney cancer growth. J Urol. 2013;189(6):2317‐2326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317. Salas Fragomeni RA, Chung HW, Landesman Y, et al. CRM1 and BRAF inhibition synergize and induce tumor regression in BRAF‐mutant melanoma. Mol Cancer Ther. 2013;12(7):1171‐1179. [DOI] [PubMed] [Google Scholar]
  • 318. Gravina GL, Mancini A, Colapietro A, et al. Pharmacological treatment with inhibitors of nuclear export enhances the antitumor activity of docetaxel in human prostate cancer. Oncotarget. 2017;8(67):111225‐111245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 319. De Cesare M, Cominetti D, Doldi V, et al. Anti‐tumor activity of selective inhibitors of XPO1/CRM1‐mediated nuclear export in diffuse malignant peritoneal mesothelioma: the role of survivin. Oncotarget. 2015;6(15):13119‐13132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320. Shang E, Zhang Y, Shu C, et al. Dual inhibition of Bcl‐2/Bcl‐xL and XPO1 is synthetically lethal in glioblastoma model systems. Sci Rep. 2018;8(1):15383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 321. Wang LH, Wei S, Yuan Y, et al. KPT330 promotes the sensitivity of glioblastoma to olaparib by retaining SQSTM1 in the nucleus and disrupting lysosomal function. Autophagy. 2024;20(2):295‐310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322. Quintanal‐Villalonga A, Durani V, Sabet A, et al. Exportin 1 inhibition prevents neuroendocrine transformation through SOX2 down‐regulation in lung and prostate cancers. Sci Transl Med. 2023;15(707):eadf7006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 323. Xu S, Shi Y, Li S. Enhanced anticancer synergy of LOM612 in combination with selinexor: FOXO1 nuclear translocation‐mediated inhibition of Wnt/beta‐catenin signaling pathway in breast cancer. Cancer Chemother Pharmacol. 2024;93(3):191‐202. [DOI] [PubMed] [Google Scholar]
  • 324. Wen T, Geng M, Bai E, et al. KPT‐330 and Y219 exert a synergistic antitumor effect in triple‐negative breast cancer through inhibiting NF‐kappaB signaling. FEBS Open Bio. 2023;13(4):751‐762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 325. Stiff PJ, Mehrotra S, Potkul RK, Banerjee S, Walker C, Drakes ML. Selinexor in combination with decitabine attenuates ovarian cancer in mice. Cancers. 2023;15(18):4541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 326. Corno C, Stucchi S, De Cesare M, et al. FoxO‐1 contributes to the efficacy of the combination of the XPO1 inhibitor selinexor and cisplatin in ovarian carcinoma preclinical models. Biochem Pharmacol. 2018;147:93‐103. [DOI] [PubMed] [Google Scholar]
  • 327. Handley KF, Rodriguez‐Aguayo C, et al. Rational Combination of CRM1 inhibitor selinexor and olaparib shows synergy in ovarian cancer cell lines and mouse models. Mol Cancer Ther. 2021;20(12):2352‐2361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328. Kashyap T, Argueta C, Aboukameel A, et al. Selinexor, a selective inhibitor of nuclear export (SINE) compound, acts through NF‐κB deactivation and combines with proteasome inhibitors to synergistically induce tumor cell death. Oncotarget. 2016;7(48):78883‐78895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 329. Jeitany M, Prabhu A, Dakle P, et al. Novel carfilzomib‐based combinations as potential therapeutic strategies for liposarcomas. Cell Mol Life Sci. 2021;78(4):1837‐1851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330. von Fallois M, Kosyna FK, Mandl M, Landesman Y, Dunst J, Depping R. Selinexor decreases HIF‐1α via inhibition of CRM1 in human osteosarcoma and hepatoma cells associated with an increased radiosensitivity. J Cancer Res Clin. 2021;147(7):2025‐2033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 331. Mittal S, Kadamberi IP, Chang H, et al. Preclinical activity of selinexor in combination with eribulin in uterine leiomyosarcoma. Exp Hematol Oncol. 2023;12(1):78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332. Hong AL, Tseng YY, Cowley GS, et al. Integrated genetic and pharmacologic interrogation of rare cancers. Nat Commun. 2016;7:11987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 333. Farren MR, Hennessey RC, Shakya R, et al. The exportin‐1 inhibitor selinexor exerts superior antitumor activity when combined with T‐cell checkpoint inhibitors. Mol Cancer Ther. 2017;16(3):417‐427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 334. Ou L, Wang X, Cheng S, et al. Verdinexor, a selective inhibitor of nuclear exportin 1, inhibits the proliferation and migration of esophageal cancer via XPO1/c‐Myc/FOSL1 axis. Int J Biol Sci. 2022;18(1):276‐291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 335. Wang Z, Pan B, Yao Y, Qiu J, Zhang X, Wu X, Tang N. XPO1 intensifies sorafenib resistance by stabilizing acetylation of NPM1 and enhancing epithelial‐mesenchymal transition in hepatocellular carcinoma. Biomed Pharmacother. 2023;160:114402. [DOI] [PubMed] [Google Scholar]
  • 336. Uddin MH, Li Y, Khan HY, Muqbil I, Aboukameel A, Sexton RE, Reddy S, Landesman Y, Kashyap T, Azmi AS, Heath EI. Nuclear export inhibitor KPT‐8602 synergizes with PARP inhibitors in escalating apoptosis in castration resistant cancer cells. Int J Mol Sci. 2021;22(13):6676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 337. Gounder MM, Zer A, Tap WD, et al. Phase IB study of selinexor, a first‐in‐class inhibitor of nuclear export, in patients with advanced refractory bone or soft tissue sarcoma. J Clin Oncol. 2016;34(26):3166‐3174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 338. Wei XX, Siegel AP, Aggarwal R, et al. A phase II trial of selinexor, an oral selective inhibitor of nuclear export compound, in abiraterone‐ and/or enzalutamide‐refractory metastatic castration‐resistant prostate cancer. Oncologist. 2018;23(6):656. e64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 339. Thein KZ, Karp DD, Tsimberidou A, et al. Selinexor in combination with carboplatin and paclitaxel in patients with advanced solid tumors: results of a single‐center, multi‐arm phase Ib study. Invest New Drugs. 2022;40(2):290‐299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 340. Lewin J, Malone E, Al‐Ezzi E, et al. A phase 1b trial of selinexor, a first‐in‐class selective inhibitor of nuclear export (SINE), in combination with doxorubicin in patients with advanced soft tissue sarcomas (STS). Eur J Cancer. 2021;144:360‐367. [DOI] [PubMed] [Google Scholar]
  • 341. Azmi AS, Khan HY, Muqbil I, et al. Preclinical assessment with clinical validation of selinexor with gemcitabine and nab‐paclitaxel for the treatment of pancreatic ductal adenocarcinoma. Clin Cancer Res. 2020;26(6):1338‐1348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 342. Abdul Razak AR, Mau‐Soerensen M, Gabrail NY, et al. First‐in‐class, first‐in‐human phase I study of selinexor, a selective inhibitor of nuclear export, in patients with advanced solid tumors. J Clin Oncol. 2016;34(34):4142‐4150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 343. Rubinstein MM, Grisham RN, Cadoo K, et al. A phase I open‐label study of selinexor with paclitaxel and carboplatin in patients with advanced ovarian or endometrial cancers. Gynecol Oncol. 2021;160(1):71‐76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 344. Thein KZ, Piha‐Paul SA, Tsimberidou A, et al. Selinexor in combination with topotecan in patients with advanced or metastatic solid tumors: results of an open‐label, single‐center, multi‐arm phase Ib study. Invest New Drug. 2021;39:1357‐1365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 345. Vergote IB, Lund B, Peen U, et al. Phase 2 study of the Exportin 1 inhibitor selinexor in patients with recurrent gynecological malignancies. Gynecol Oncol. 2020;156(2):308‐314. [DOI] [PubMed] [Google Scholar]
  • 346. Westin SN, Fu S, Tsimberidou A, et al. Selinexor in combination with weekly paclitaxel in patients with metastatic solid tumors: results of an open label, single‐center, multi‐arm phase 1b study with expansion phase in ovarian cancer. Gynecol Oncol. 2023;168:76‐82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 347. Gounder MM, Razak AA, Somaiah N, et al. Selinexor in advanced, metastatic dedifferentiated liposarcoma: a multinational, randomized, double‐blind, placebo‐controlled trial. J Clin Oncol. 2022;40(22):2479‐2490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 348. Vergote I, Perez‐Fidalgo JA, Hamilton EP, et al. Oral selinexor as maintenance therapy after first‐line chemotherapy for advanced or recurrent endometrial cancer. J Clin Oncol. 2023;41(35):5400. [DOI] [PubMed] [Google Scholar]
  • 349. Nelson BE, Saleem S, Damodaran S, et al. Phase 1b study of combined selinexor and eribulin for the treatment of advanced solid tumors and triple‐negative breast cancer. Cancer. 2023;129(14):2201‐2213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 350. Nilsson S, Stein A, Rolfo C, et al. Selinexor (KPT‐330), an oral selective inhibitor of nuclear export (SINE) compound, in combination with FOLFOX in patients with metastatic colorectal cancer (mCRC)—final results of the phase I trial SENTINEL. Curr Cancer Drug Targets. 2020;20(10):811‐817. [DOI] [PubMed] [Google Scholar]
  • 351. Lawrence YR, Shacham‐Shmueli E, Yarom N, et al. Nuclear export inhibition for radiosensitization: a proof‐of‐concept phase 1 clinical trial of selinexor (KPT‐330) combined with neoadjuvant chemoradiation in locally advanced rectal cancer. Int J Radiat Oncol Biol Phys. 2022;114(2):250‐255. [DOI] [PubMed] [Google Scholar]
  • 352. Shafique M, Ismail‐Khan R, Extermann M, et al. A phase II trial of selinexor (KPT‐330) for metastatic triple‐negative breast cancer. Oncologist. 2019;24(7):887. e416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353. Lassman AB, Wen PY, van den Bent MJ, et al. A phase II study of the efficacy and safety of oral selinexor in recurrent glioblastoma. Clin Cancer Res. 2022;28(3):452‐460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 354. Heong VYM, Koe P, Yong WP, et al. RAS/AKT pathway mutations as predictive biomarkers in patients with colorectal cancer treated with the exportin 1 (XPO1) inhibitor selinexor (SEL)—inhibition of nuclear‐cytoplasmic translocation of p27 as a mechanism of anti‐tumour activity. Ann Oncol. 2016;27:VI122. [Google Scholar]
  • 355. Kendra KL, Watson R, Lesinski GB. Selinexor, a selective inhibitor of nuclear export (SINE), in patients with unresectable melanoma. J Clin Oncol. 2017;35:78883‐78895. [Google Scholar]
  • 356. Golan T, Kutiel TS, Geva R, et al. Open‐label phase 1 study evaluating the tolerability and anti‐tumor activity of selinexor and pembrolizumab in colorectal cancer. J Clin Oncol. 2021;39(15):e15579‐e15579. [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No data were generated for the research described in the article.


Articles from Clinical and Translational Medicine are provided here courtesy of John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics

RESOURCES