Abstract
Nuclear-cytoplasmic trafficking of proteins is a significant factor in the development of cancer and drug resistance. Subcellular localization of exported proteins linked to cancer development include those involved in cell growth and proliferation, apoptosis, cell cycle regulation, transformation, angiogenesis, cell adhesion, invasion, and metastasis. Here, we examined the basic mechanisms involved in the export of proteins from the nucleus to the cytoplasm. All proteins over 40 kDa use the nuclear pore complex to gain entry or exit from the nucleus, with the primary nuclear export molecule involved in these processes being chromosome region maintenance 1 (CRM1, exportin 1 or XPO1). Proteins exported from the nucleus must possess a hydrophobic nuclear export signal (NES) peptide that binds to a hydrophobic groove containing an active-site Cys528 in the CRM1 protein. CRM1 inhibitors function largely by covalent modification of the active site Cys528 and prevent binding to the cargo protein NES. In the absence of a CRM1 inhibitor, CRM1 binds cooperatively to the NES of the cargo protein and RanGTP, forming a trimer that is actively transported out of the nucleus by facilitated diffusion. Nuclear export can be blocked by CRM1 inhibitors, NES peptide inhibitors or by preventing post-translational modification of cargo proteins. Clinical trials using the classic CRM1 inhibitor leptomycin B proved too toxic for patients; however, a new generation of less toxic small molecule inhibitors are being used in clinical trials in patients with both hematological malignancies and solid tumors. Additional trials are being initiated using small-molecule CRM1 inhibitors in combination with chemotherapeutics such as pegylated liposomal doxorubicin. In this review, we present evidence that combining the new CRM1 inhibitors with other classes of therapeutics may prove effective in the treatment of cancer. Potential combinatorial therapies discussed include the use of CRM1 inhibitors and the addition of alkylating agents (melphalan), anthracyclines (doxorubicin and daunomycin), BRAF inhibitors, platinum drugs (cisplatin and oxaliplatin), proteosome inhibitors (bortezomib and carfilzomib), or tyrosine-kinase inhibitors (imatinib). Also, the sequence of treatment may be important for combination therapy. We found that the most effective treatment regimen involved first priming the cancer cells with the CRM1 inhibitor followed by doxorubicin, bortezomib, carfilzomib, or melphalan. This order sensitized both de novo and acquired drug-resistant cancer cell lines.
Keywords: CRM1, topoisomerase IIα, nuclear pore complex, drug resistance, multiple myeloma, nuclear export
1. Nuclear export
1.1 Nuclear-pore complex structure and function
The cell nucleus is a sequestered environment due to the highly selective transport of proteins greater than 40 kDa both into and out of the nucleus through the nuclear pore complex (NPC). The NPC is one of the largest protein complex structures (125 MDa) in the cell, containing at least 30 different proteins called nucleoporins [1; 2]. The NPC has a central transporter region containing a nucleoporin matrix that assists movement of macromolecules, eight fibril proteins that extend into the cytoplasm, and a nuclear basket structure. Movement of molecules through the NPC is bidirectional; movement in or out of the nucleus is dependent on the type of receptor protein and the molecule associated with it.
For nuclear export or import of a large protein (>40 kDa) to occur, it must be bound to a nuclear export receptor molecule. The majority of these NPC transport molecules are members of the karyopherin-ß family of proteins. There are approximately 19 members of karyopherin-ß receptor family proteins, with each recognizing a specific group of cargo proteins or RNA [3]. CRM1 (chromosome region maintenance 1) is a ubiquitous transport receptor protein that binds its cargo via a hydrophobic nuclear export signal (NES) peptide sequence (see Section 1.2). Export complexes are formed in the nucleus and are made up of three components or trimer consisting of, CRM1, RanGTP, and the cargo or exported substrate. Binding of CRM1 to the cargo is weak; however, when the cargo protein and RanGTP bind to CRM1 cooperatively, the affinity of CRM1 to both RanGTP and the cargo substrate is increased 500- to 1,000-fold [4; 5]. Figure 1A shows the trimer formation of CRM1, RanGTP, and the cargo protein (in this case topoisomerase IIα (topo IIα)) in the nucleus at the NPC and transport through the NPC. The energy required for transport is provided by RanGTP. RanGTP is maintained in high concentrations in the nucleus by the presence of RCC1, a guanine nucleotide exchange factor (GEF). The high RanGTP concentration gradient from the nucleus to the cytoplasm provides the energy required for facilitated nuclear export. After the CRM1/RanGTP/cargo trimer arrives in the cytoplasm, RanGTP is hydrolyzed to RanGDP by a RanGAP (Figure 1B), causing dissociation of the trimer and release of the cargo protein into the cytoplasm. CRM1 and RanGDP are then recycled back into the nucleus through the NPC for another export cycle (Figure 1C).
Figure 1. Intracellular export of proteins.
A CRM1/RanGTP/Cargo protein (topo IIα) complex is transported through the NPC to the cytoplasm where phosphorylated topo IIα is released. The nuclear export of topo IIα can be blocked by 1) CK2 inhibitors that prevent phosphorylation and binding of topo IIα to CRM1; 2) covalent binding of CRM1 inhibitors to CRM1, preventing binding of topo IIα NES, and 3) specific binding of the NES by small molecule inhibitors. Blocking the nuclear export of topo IIα increases the amount in the nucleus, which is able to cleave DNA in the presence of topo II inhibitors, thus overcoming this de novo resistance. Figure 1A shows the trimer formation of CRM1, RanGTP, and the cargo protein (topo IIα)) in the nucleus at the NPC and transport through the NPC. Figure 1B, RanGTP is hydrolyzed to RanGDP by a RanGAP in the cytoplasm (Figure 1B), causing dissociation of the trimer and release of the cargo protein. Figure 1C, CRM1 and RanGDP are then recycled back into the nucleus through the NPC for another export cycle.CK2, casein kinase 2; CRM1, chromosome maintenance protein 1; NES, nuclear export signal; NPC, nuclear pore complex.
1.2 CRM1 and nuclear export signals
CRM1 is a ubiquitous nuclear export receptor molecule that binds to a cargo substrate (protein or RNA) containing a hydrophobic NES. At last count (January 2014), the “NESdb” database of NES-containing CRM1 cargoes published that there are 241 known macromolecules that bind to and are exported by CRM1 [6; 7]. The hydrophobic NES of the cargo protein binds to a hydrophobic groove of CRM1 containing an active site Cys528 [8]. NES were originally characterized as leucine-rich; however, characterization of the NES may be better described as not specifically leucine-rich but that it possesses a general hydrophobic pattern. NES peptides are 8–15 residues long and conform loosely to a consensus sequence of Φ1-X2,3-Φ2-X2,3-Φ3-XΦ4 (Φn represents Leu, Val, Ile, Phe, or Met and X can be any amino acid), as reviewed in Xu et al [7].
2. Blocking nuclear export of proteins
Figure 1 demonstrates three potential means of attenuating nuclear export of the cancer drug target topo IIα: 1) CRM1 inhibitors, 2) NES small molecule inhibitors, and 3) casein kinase 2 inhibitors, with the last preventing post-translational phosphorylation of topo IIα.
2.1 CRM1 inhibitors
Direct inhibition of nuclear export is most commonly achieved with CRM1 inhibitors. There is a large cadre of nuclear export inhibitors from both natural and synthetic sources, as recently reviewed by Turner and Sullivan et al [9] and Hill and Link et al [10]. These reviews include more detailed information than will be covered here, including chemical structures. The majority of these inhibitors bind to the active site Cys528 contained within a hydrophobic groove of CRM1 and irreversibly modify it by a Michael-type covalent addition. Site-directed mutagenesis of Cys528 to Ser528 will prevent binding of CRM1 by leptomycin B [11], ratjadone [12] and other small molecule CRM1 inhibitors [13]. Exceptions to this include the CRM1 inhibitor CBS9106 developed by Sakakibara and Kawabe et al [14]. CBS9106 associates with the active site Cys528, but the binding is reversible. In addition, this compound does not induce CRM1 mRNA expression, whereas other CRM1 inhibitors, including the classic inhibitor of CRM1, leptomycin B, and newer generation N-azolylacrylate analogs now referred to as SINE (selective inhibitors of nuclear export) increase CRM1 mRNA in a dose-dependent manner [15]. It is interesting that leptomycin B and SINE inhibitors increase CRM1 mRNA, but only the SINE compounds and CBS9106 lead to proteasome-mediated degradation of CRM1 [15]. SINEs are selective for the export molecule CRM1 and do not appear to inhibit other karyopherin-ß (import or export) proteins (unpublished observations, Landesman and Shacham et al (Karyopharm Therapeutics)). Currently, only KPT-330 is in clinical trials (see Section 3).
2.2 Nuclear Export Signal Small Molecule Inhibitors
Since CRM1 is a ubiquitous exporter of nuclear proteins (>241 proteins), truly selective inhibitors that bind to the NES and prevent export may produce a more targeted therapy (Figure 1). We have explored this idea by developing inhibitors that bind to a single specific NES peptide and prevent export of a topo IIα. We have shown that topo IIα has a NES at amino acids 1017-1028 (site A: DILRDFFELRLK) and at amino acids 1054-1066 (site B: FILEKIDGKIIIE) [16]. Disrupting the binding between topo IIα-NES and CRM1 could prevent CRM1-mediated nuclear export of topo IIα and result in the sensitization of MM cells to topo II inhibitors. We identified, by a virtual screening method, a series of inhibitors of the interaction of CRM1 and topo IIα which block the nuclear export of topo IIα [17]. A homology model of human topo IIα based on S. cerevisiae topo II was used for molecular docking, These NES inhibitors are designed to bind specifically to the NES of topo IIα and prevent its export. We found that they did not prevent the export of other proteins such as p53. When NES inhibitors are combined with the topo II poison doxorubicin, they sensitize cancer cells to doxorubicin treatment and synergistically induce apoptosis and inhibit proliferation [17].
2.3 Post-translational modification of protein cargoes
Post-translational modification of cargo proteins include phosphorylation or dephosphorylation [18; 19; 20], sumoylation [18; 21], ubiquitination [18; 21], acetylation [18], or protein binding [22; 23; 24]; in effect any biochemical event that will prevent CRM1/cargo protein binding by either masking a NES or by exposing a nuclear localization signal peptide. Examples of proteins that are modified by phosphorylation before import or export include p53 [25], Nrf2 [26], topo IIα [27], and PPAR-gamma [28]. We investigated the phosphorylation status of topo IIα, isolated from nuclear and cytoplasmic extracts of human myeloma cells by immunoprecipitation [29]. The purified topo IIα was digested with trypsin and analyzed with an electrospray ion trap mass spectrometer and liquid chromatography. Comparing the phosphorylation sites of nuclear and cytoplasmic topo II we found that cytoplasmic topo IIα but not nuclear topo was highly phosphorylated at serine 1524, a published casein kinase II (CK2) motif [29]. Using site-directed mutagenesis we converted serine 1524 to an alanine and found that mutated recombinant FLAG-tagged TOP2A export was reduced, as compared to wild-type FLAG-tagged topo IIα. This was observed by immunofluorescence microscopy in human myeloma cell lines. Serine 1524 is a CK2 phosphorylation site, therefore we used the CK2 inhibitor 4,5,6,7-tetrabromobenzotriazole (TBB) and found by immunofluorescence microscopy, that topo IIα nuclear export was blocked in high-density cells. The TBB data was duplicated using a CK2 specific siRNA to knockdown CK2 expression (90% knockdown). In addition, we found that blocking nuclear export of topo IIα with TBB or CK2-specific siRNA sensitized drug-resistant myeloma cells to topo II poisons doxorubicin and VP-16 [29].
It may therefore be possible to block nuclear export of a specific cargo by inhibition of a kinase or other specific post-translation event.
3. Clinical Experience with CRM 1 inhibitors
3.1 Leptomycin B Clinical Trial
The first CRM1 inhibitor to enter the clinical arena was leptomycin B. Newlands et al reported on a phase I trial of elactocin (leptomycin B) in patients with various solid tumors in 1996 [30]. This was based on preclinical evidence of efficacy of this agent in various cell lines and the suggestion that it inhibits DNA synthesis and DNA polymerase. Thirty-three patients were enrolled in the phase I trial, and several schedules of administration were studied in order to mitigate the toxicity: 1 hour infusion every 3 weeks, 1 hour weekly infusion, 1 hour daily infusion for 5 days, a 24-hour continuous infusion, and a continuous 5-day infusion [30]. Regardless of the infusion schedule, the dose-limiting toxicities were nausea, vomiting, and profound anorexia and malaise, often lasting more than 1 week with the need for intravenous hydration. No responses were noted in this trial, and the authors recommended no further investigations given the side effects and lack of benefits noted [30].
3.2 Selinexor or KPT-330 Clinical Trials
More recently, Selinexor (KPT-330), an oral CRM1 inhibitor, has been administered to patients with advanced hematologic malignancies and solid tumors in two phase I clinical trials (KPT-330-001 and KPT-330-002, respectively) [31; 32; 33; 34; 35]. Over 100 patients have been treated as part of those two studies. In the initial design, Selinexor was administered orally three times per week alternating with twice per week (10 doses in a 4-week cycle). Subsequently, Selinexor was administered twice weekly with improvement in tolerability [33]. The maximum tolerated dose of the 10 dose schedule was 30 mg/m2, but dose escalation continued on the 8 dose schedule at doses above 30 mg/m2 [34]. The elimination half-life was independent of the dose and was about 7.0 hours [31; 32; 33; 34; 35]. In addition, evaluation of tumor tissues confirmed nuclear localization of multiple tumor suppressor protein after therapy with Selinexor [31; 32; 33; 34; 35]. Gastrointestinal adverse events represent the predominant toxicity, which included nausea, vomiting, anorexia, and fatigue. Other notable toxicities included thrombocytopenia, which was seen in patients with advanced hematologic malignancies [33]. Evidence of anti-tumor activity was noted in patients with various tumors and particularly in patients with advanced hematologic malignancies, including patients with acute myeloid leukemia (4 patients of 14 evaluable patients achieved a complete response with or without complete count recovery) [31], non-Hodgkin lymphoma/chronic lymphocytic leukemia (3 patients of 15 evaluable achieved more than 50% reduction in lymph nodes) [33], and multiple myeloma (1 patients achieved a partial response and 6 a minimal response out of 15 evaluable patients) [32]. Solid tumor responses were noted in patients with colorectal cancer (4 of 12 patients had a CEA decrease and 1 patient had a partial response, with 6 patients having stable disease) [34], whereas patients with cervical cancer and endometrial stromal sarcoma had prolonged disease stabilization [35].
3.3 Combination of Doxorubicin and Selinexor in Patients with Multiple Myeloma
Doxorubicin is commonly used in the treatment of patients with multiple myeloma, and pegylated liposomal doxorubicin is approved in combination with bortezomib for the treatment of patients with relapsed and refractory myeloma [36]. KPT-330-001 has demonstrated clinical activity for Selinexor in patients with advanced myeloma [32]. Our group has shown that high-density human myeloma cell lines and patient myeloma cells export DNA topoisomerase IIα using CRM1 from the nucleus to the cytoplasm, thus rendering the myeloma cells resistant to the cytotoxic effects of topoisomerase II inhibitors (for example, doxorubicin) [16; 37]. In addition, CRM1 inhibitors (leptomycin B and ratjadones) and CRM1 siRNA block the nuclear export of topoisomerase IIα and re-sensitize these cells to doxorubicin [38]. Based on these findings, we plan a phase I/II trial of Selinexor in combination with pegylated liposomal doxorubicin and dexamethasone in patients with relapsed and refractory myeloma. Specifically, eligible patients will receive a loading dose of Selinexor orally twice weekly for 2 weeks in combination with weekly dexamethasone. After the loading dose, patients will receive escalating doses of weekly Selinexor in combination with pegylated liposomal doxorubicin on day 1 and weekly dexamethasone. After 8 cycles, patients will receive maintenance therapy with weekly Selinexor and dexamethasone until disease progression. The primary endpoint of the phase I portion is determination of the maximum tolerated dose of Selinexor in this combination, and the primary endpoint for the phase II portion will be the overall response rate of the combination. Ex vivo drug sensitivity and nuclear localization of topoisomerase IIα will be evaluated as part of planned correlative studies.
4. Combination Studies: In vitro, In vivo Xenografts, and Ex vivo
In studies done with CRM1 inhibitors (see table 1), single-agent CRM1 inhibitors induced apoptosis in cancer cell lines, slowed tumor growth in xenograft mice, and improved survival. However, in most cancers, single-agent CRM1 inhibitors were less likely to eliminate established tumors or reduce tumor burden. In addition, the CRM1 inhibitor's ability to induce apoptosis or anti-proliferative effects in cell lines improved synergistically when combined with chemotherapeutics such as ABT-737, AraC, bevacizumab, BRAF inhibitors, bortezomib, carfilzomib, cisplatin, daunomycin, decitabine, docetaxel, doxorubicin, imatinib, melphalan, oxaliplatin, paclitaxel, SN-38, nutlin-3a, or topotecan. CRM1 inhibitors have been shown to sensitize cancer cells both in vitro, in xenografts, or ex vivo to these chemotherapeutics and in some cases reverse drug resistance.
Table 1.
Combination Therapy with CRM1 inhibitors, AACR, ASH and ASCO 2011–2013 Abstracts
| Cancer | Drugs Used | Anti-tumor Drug Effects, In vitro and In vivo Studies | Reference |
|---|---|---|---|
| AML | KPT-185 +/− Nutlin-3a | KPT-185/Nutlin-3a synergistically induced apoptosis and cytotoxicity. p53 knockdown abrogated these effects. No synergy in normal cord blood cells. | Kojima et al. Blood: 2012; 120 Abst # 870 |
| AML | KPT-330 +/− AraC | KPT-330/AraC showed additive antiproliferative effects in acute promyelocytic leukemia cell lines. In vivo KPT-330/AraC prolonged survival compared to monotherapy (P < 0.0001) in mice. | Rettig et al. Blood: 2013; 122 Abstr #237 |
| AML | KPT-330 +/− Decitabine | KPT-330/Dct increased cytotoxicity compared to monotherapy in AML cells. In a mouse xenograft model, treatment with Dct followed by KPT-330 improved survival compared to KPT-330 alone (47 vs 36.5 days) p=0.008. | Ranganathan et al. Blood: 2013; 122 Abstr #1453 |
| CLL | KPT-185 +/− CPG, CD40L, BAFF, TNF-α, IL-6, IL-4 | KPT-185 abrogated protection induced by soluble factors in CLL cells. | Lapalombella et al. Blood: 2011; 118 Abst #232 |
| CML | KPT-330 +/− Imatinib | KPT-330 +/− Imatinib reduced colony formation significantly compared to Imatinib alone in primary CML cells. | Sorouri et al. Blood: 2013; 122 Abst #2707 |
| MM & AML | KOS-2464 +/−Dox, Daun | KOS-2464 sensitized MM and AML cells to Dox (CI 0.268) and Daun as shown by apoptosis and cell viability assays. | Turner et al. Can Res: 2012; 72 Abst #1771 |
| Multiple Myeloma | KPT-185 +/− Dex, Len, Mel, Btz | KPT-185/Dex, Len, or Btz synergistically (CI <1.0) induced cytotoxicity in MM cell lines. KPT-185/Mel induced additive effects. | Kong et al. Blood: 2011; 118 Abst #2913 |
| Multiple Myeloma | KPT-185, KPT-276 +/− Btz, Dex | KPT/Btz and/or Dex increased the cellular cytotoxicity in MM lines. | Kandarpa et al. Blood: 2011; 118 Abst #1852 |
| Multiple Myeloma | KOS-2464, KPT-185, KPT-249, KPT-330 +/− Btz, Cfz | CRM1 inhibitors sensitized both MM cell lines and MM patient cells (ex vivo) synergistically (CI <0.5) to Btz and Cfz as shown by apoptosis and cell viability assays. | Turner et al. Can Res: 2013; 73 Abstr #2066 |
| Multiple Myeloma | KPT-330 +/− Cfz | KPT-330/CFZ synergistically increased cell death (CI 0.2−0.6) by both apoptosis and autophagy in MM cell lines. In a xenograft mouse KPT-330/CFZ was more effective than monotherapy, and high dose completely impaired tumor growth with good tolerability. | Rosebecket al. Blood; 2013 122 Abst #279 |
| Multiple Myeloma | KPT-185, KPT-276 +/− Btz, Dex | KPT-185/Btz markedly increased apoptosis in MM1R cells vs monotherapy. Dex/KPT-185 or KPT-276 induced synergistic cytotoxicity in cell lines. | Tai et al. Can Res: 2013; 73 Abstr #2142 |
| Multiple Myeloma | NCI-9138 +/−Dox | NCI-9138 sensitized human myeloma cells H929 drug-resistant to Dox but did not affect normal fibroblast cell lines or human PBMCs. | Turner etal. Blood; 2013 122 Abst #1925 |
| NHL | KPT-330 +/− Cfz | KPT-330/Cfz induced strong synergy (CI<1.0) in aggressive NHL cell lines using MTS assay and Annexin-V apoptosis assays. | Lopez et al. Blood: 2013; 122 Abst #5147 |
| Colon | KPT-185 +/− SN38 | KPT185/SN38 resulted in synergistic cell death in colon cancer cells (CI<1). CDDE assay showed KPT-post increased apoptosis (4.3-fold) compared with KPT-pre (4.2-fold), concurrent (3.8-fold) and SN38 alone (1-fold). | Chung etal. J Clin Oncol 2012; 30 Abstr #609 |
| Colon | KPT-251 +/− SN38 | KPT-251/SN3S induced synergism in both CPT-11/SN3S-sensitive and CPT-11/SN38-resistant colon cancers cells in vitro and in vivo. | Chung et al. J Clin Oncol 2012; 30 Abstr #396 |
| Colon Pancreatic | KPT-185 +/− oxaliplatin | KPT-185/oxaliplatin synergistically enhanced apoptosis in all the cell lines. | Azmi et al. J Clin Oncol 2012; 30 Abstr #245 |
| NSCLC | LMB +/− Dox | Pre-treatment with Dox improved the cytotoxic effects of LMB 2-fold on A549 cells (P<0.05) as compared to LMB alone. LMB then Dox or concurrent did not change the cytotoxic effects of either LMB or Dox. | Lu et al. Can Res: 2011; 71 Abst #3515 |
| NSCLC | KPT-185 +/− ABT-737 | KPT-185/ABT-737 induced synergistic cytotoxicity in the SINE-resistant A549 cell line & modest killing in the sensitive cell line H-226. | McCauley et al. Can Res: 2012; 72 Abstr #1831 |
| NSCLC | KPT-185,KPT-330 +/− RT | KPT-185/RT clonogenic assays showed an additive effect for SINE-resistant A549 and a dose-related effect for SINE-sensitive H1299 cell lines. | Rashal et al. Can Res: 2013; 73 Abstr #2075 |
| Melanoma | novel CRM1 inhibitor +/− PLX4032 | CRM1 inhibiton and BRAF inhibition were strongly synergistic in BRAF mutant cell lines with a statistically significant decrease in cell proliferation and increased apoptosis. | Fragomeni et al. Can Res: 2012; 72 Abstr #1914 |
| Neuroblastoma | KPT-330 +/− Irt, Tpt, Cisplat, Dox | Combination treatment with KPT-330/Irinotecan, Topotecan, Cisplatin, and Doxorubicin showed additive effects in growth inhibition. | Attiyeh et al. Can Res: 2013; 73 Abstr #2756 |
| Ovarian | KPT-185, KPT-330 +/− Cisplatin | In vitro KPT/Cisplat showed additive effect on cell death, overcoming cisplatinum resistance in the isogenic cell lines A2780 and CP70. | Chen et al. Can Res: 2013; 73 Abstr #2163 |
| Ovarian | KPT-185 +/− Tpt, Cisplat, LipoDox, Paclitaxel, Bevacizumab | KPT-185/Tpt, Cisplat and LipoDox show synergistic (CI<1) reduction in cell viability in human ovarian cancer cell lines. In the A2780 mouse model, KPT-185/Tpt reduced tumor growth by 98% (p<0.01). Significant tumor growth inhibition also was seen with KPT-330/Paclitaxel or Bevacizumab. | Miyake et al. Can Res: 2013; 73 Abstr #5541 |
| Prostate | KPT-185, KPT-251 +/− Cisplatin, Docetaxel | Synergy was seen with KPT in combination with Cisplatin and Docetaxel in vitro and in vivo. | Gravina etal. Can Res: 2012; 72 Abstr #1841 |
| Renal | KPT-330 +/−ABT-737 | KPT-330/ABT-737 showed synergism in RCC cell lines and a normal human kidney cell line in vitro. | Inoue et al. J Clin Oncol 2013; 31 Abstr #411 |
Abbreviations:
AML, Acute Myelocytic Leukemia; BAFF, B-cell activating factor; Btz, Bortezomib; CDDE, Cell Death Detection ELISA assay; CD40L, Cluster of Differentiation 40 ligand; Cfz, Carfilzomib; CLL, Chronic Lymphocytic Leukemia; Daun, Daunorubicin; Dct, Decitabine; Dex, Dexamethasone; Dox, Doxorubicin; IL-6 & IL-4, Interleukin 6 and 4; Len, Lenalidomide; LipoDox, Liposomal Doxorubicin; LMB, Leptomycin B; Mel, Melphalan; MM, Multiple Myeloma; MTS; Microculture Tetrazolium Assay; NHL, Non-Hodgkins Lymphoma; NSCLC, Non-small cell lung cancer; PARP, Poly (ADP-ribose) polymerase; PBMC, peripheral blood mononuclear cells; RT, Radiation Therapy; TNF-α, Tumor necrosis Factor alpha
Listed in this section are several studies where CRM1 inhibitors were used in combination with other cancer therapeutics and in drug-resistant/relapsed cancers. A table of current abstracts (Table 1) summarizes the latest findings in CRM1 combination therapies. In addition, we include a table on drug sequencing in the treatment of multiple myeloma (Table 2).
Tables 2. Combination Index of concurrent or sequential treatment of MM cells.
Combination index (CI) values of high density human multiple myeloma cell line NCI-H929 treated sequentially or concurrently with CRM1 inhibitors and either proteasome inhibitors (table 2a), doxorubicin (table 2b), or other chemotherapy agents (table 2c). Cells were placed at high density (3.0 × 106/ml) and incubated with the first drug for 24 hours followed by the second drug for an additional 24 hours for sequential treatment. For concurrent treatment cells were treated with both drugs for 48 hrs. Cytotoxicity was determined by a high-throughput CellTiter-Blue (Promega Corp.) cell viability assay. The combination index value was calculated using the Chou and Talalay method with CI <1 representing synergy, =1 additive, and <1, antagonistic results.
| a. Combination Index (CI) Values for CRM1 Inhibitors + Proteasome Inhibitors in High-density H929 MM Cells by CT-Blue Viability Assay | |||||
|---|---|---|---|---|---|
| Drug Administration: concurrent (+) or sequential (→) | Concentration range: Drug 1 | Concentration range: Drug 2 | Combination Index (mean) | Combination Index (SEM) | n |
| KOS-2464 + Bortezomib | 9.1 nM – 1 μM | 1.8 nM – 200 nM | 0.711 | 0.184 | 2 |
| KPT 185 + Bortezomib | 390.6 nM – 100 μM | 39 pM – 50 nM | 0.922 | 0.057 | 2 |
| KPT 249 + Bortezomib | 390.6 nM – 100 μM | 39 pM – 10 nM | 0.887 | 0.006 | 2 |
| KPT 330 + Bortezomib | 780.4 nM – 20 μM | 1.8 nM – 50 nM | 0.953 | 0.065 | 3 |
| KOS-2464 → Bortezomib | 3.9 nM – 5.0 μM | 39 pM – 100 nM | 0.415 | 0.134 | 3 |
| KPT-185 → Bortezomib | 390.6 nM – 100 μM | 39 pM – 50 nM | 0.41 | 0.019 | 3 |
| KPT-249 → Bortezomib | 390.6 nM – 100 μM | 39 pM – 10 nM | 1.098 | 0.259 | 2 |
| KPT-330 → Bortezomib | 780.4 nM – 20 μM | 1.8 nM – 50 nM | 0.651 | 0.144 | 4 |
| Bortezomib → KPT 330 | 4.5 nM – 50 nM | 1.8 μM – 20 μM | 0.8 | 0.072 | 2 |
| KOS-2464 + Carfilzomib | 9.1 nM – 100 nM | 1.8 nM – 20 nM | 0.713 | 0.141 | 2 |
| KPT 185 + Carfilzomib | 1.8 μM – 20 μM | 1.8 nM – 20 nM | 0.408 | 0.083 | 3 |
| KPT 249 + Carfilzomib | 1.8 μM – 20 μM | 1.8 nM – 50 nM | 0.469 | 0.089 | 2 |
| KPT 330 + Carfilzomib | 1.8 μM – 20 μM | 1.8 nM – 50 nM | 0.552 | 0.045 | 2 |
| KOS-2464 → Carfilzomib | 9.1 nM – 1 μM | 1.8 nM – 200 nM | 0.224 | 0.021 | 3 |
| KPT-185 → Carfilzomib | 780.4 nM – 20 μM | 1.8 nM – 50 nM | 0.322 | 0.136 | 3 |
| KPT-249 → Carfilzomib | 780.4 nM – 20 μM | 1.8 nM – 100 nM | 0.426 | 0.084 | 4 |
| KPT-330 → Carfilzomib | 780.4 nM – 20 μM | 1.8 nM – 50 nM | 0.388 | 0.08 | 3 |
| Carfilzomib → KPT-249 | 4.5 nM – 50 nM | 1.8 μM – 20 μM | 1.269 | 1 | |
| Carfilzomib → KPT-330 | 4.5 nM – 50 nM | 1.8 μM – 20 μM | 1.153 | 0.341 | 2 |
| b. Combination Index (CI) Values for CRM1 Inhibitors + Topoisomerase II Inhibitor in High-density H929 MM Cells by CT-Blue Assay | |||||
|---|---|---|---|---|---|
| Drug Administration: concurrent (+) or sequential (→) | Concentration range: Drug 1 | Concentration range: Drug 2 | Combination Index (mean) | Combination Index (SEM) | n |
| KOS-2464 + Doxorubicin | 3.9 nM – 5 μM | 19.5 nM – 100 μM | 0.674 | 0.147 | 2 |
| KPT-185 + Doxorubicin | 195.3 nM – 100 μM | 7.8 nM – 10 μM | 0.807 | 0.165 | 4 |
| KPT-249 + Doxorubicin | 195.3 nM – 100 μM | 7.8 nM – 10 μM | 0.735 | 0.094 | 4 |
| KPT-330 + Doxorubicin | 195.3 nM – 100 μM | 7.8 nM – 10 μM | 0.696 | 0.155 | 5 |
| KOS-2464 → Doxorubicin | 3.9 nM – 5 μM | 19.5 nM – 100 μM | 0.278 | 0.065 | 7 |
| KPT-185 → Doxorubicin | 391 nM – 100 μM | 390.6 nM – 100 μM | 0.377 | 0.087 | 4 |
| KPT-249 → Doxorubicin | 3.9 nM – 100 μM | 39.1 nM – 100 μM | 0.628 | 0.053 | 7 |
| KPT-276 → Doxorubicin | 39.1 nM – 100 μM | 390.6 nM – 100 μM | 0.340 | 0.134 | 7 |
| KPT-330 → Doxorubicin | 78.1 nM – 20 μM | 390.6 nM – 200 μM | 0.092 | 0.025 | 3 |
| Doxorubicin → KOS-2464 | 97.66 nM – 25 μM | 195.31 pM – 50 nM | 1.003 | 0.010 | 2 |
| Doxorubicin → KPT 185 | 97.66 nM – 25 μM | 195.31 nM – 50 μM | 1.197 | 0.259 | 3 |
| Doxorubicin → KPT 249 | 97.66 nM – 25 μM | 195.31 nM – 50 μM | 1.064 | 0.249 | 2 |
| Doxorubicin → KPT 330 | 97.66 nM – 25 μM | 195.31 nM – 50 μM | 1.024 | 0.233 | 4 |
| c. Combination Index (CI) Values for CRM1 Inhibitors + Other Drugs in High-density H929 MM Cells by CT-Blue Assay | |||||
|---|---|---|---|---|---|
| Drug Administration: sequential (→) | Concentration range: Drug 1 | Concentration range: Drug 2 | Combination Index (mean) | Combination Index (SEM) | n |
| KOS-2464 → Melphalan | 3.9 nM – 5 μM | 390.6 nM – 100 μM | 0.362 | 0.216 | 3 |
| KPT-185 → Melphalan | 78.1 nM – 100 μM | 390.6 nM – 200 μM | 0.849 | 0.104 | 4 |
| KPT-249 → Melphalan | 390.6 nM – 100 μM | 39.1 nM – 10 μM | 0.979 | 0.066 | 3 |
| KPT-330 → Melphalan | 78.1 nM – 20 μM | 390.6 nM – 200 μM | 0.549 | 0.117 | 3 |
| KPT-249 → Dexamethasone | 390.6 nM – 100 μM | 39.1 nM – 200 μM | 1.263 | 0.238 | 4 |
| KPT-249 → Lenalidomide | 390.6 nM – 100 μM | 39.1 nM – 10 μM | 1.098 | 0.11 | 3 |
4.1 BRAF inhibitors
Metastatic melanoma is a highly aggressive tumor with generally poor prognosis. Over half the patients with metastatic melanoma have a constitutively activated BRAF kinase driving proliferation of the cancer [39]. Drugs that target the mutated BRAF kinase have been shown to significantly improve overall survival of metastatic melanoma patients, emphasizing the role of this oncogene in melanoma biology. BRAF inhibitors block melanoma cell growth signals and subsequent proliferation and have shown good clinical results with low toxicity. However, resistance to BRAF inhibitor therapy eventually develops, and subsequent recurrences or relapses regularly occur within a short period after BRAF inhibitor treatment.
CRM1 is over-expressed in malignant melanoma and may prove to be a negative prognostic indicator [40]. Melanoma cells treated with the CRM1 inhibitor leptomycin B had high levels of apoptosis without negatively affecting normal melanocytes or primary lung fibroblasts. Cell death involved both intrinsic and extrinsic apoptotic pathways and included nuclear retention (entrapment) of anti-proliferative factors p53 and p21 and the down-regulation of the anti-apoptotic factor survivin. CRM1 inhibitor-treated melanoma cells went into G1 cell-cycle arrest, and wild-type p53 expression was increased [40]. These data indicate the potential of new therapies using BRAF and CRM1 inhibitors to overcome both de novo resistance and prevent the development of acquired resistance in melanoma.
The synergistic potential of CRM1 and BRAF inhibition was explored in a recent study by Fragomeni et al [41]. Multiple small molecule CRM1 inhibitors obtained from Karyopharm Therapeutics (KPT-185, KPT-251, KPT-276, and KPT-330 (the clinical compound)) were used in combination with BRAF inhibitor analogs PLX-4720 and PLX-4032 in this study. It was found that proliferation of UACC-903, A375, and Mewo melanoma cell lines was inhibited synergistically when CRM1 inhibitors and BRAF inhibitors were used, as measured by the MTT assay. These results were independent of whether the cells had a mutant (UACC-903/A375 cells) or wild-type (Mewo cells) BRAF gene. The CRM1/BRAF inhibitor combination also synergistically induced apoptosis when measured by caspase 3/7 activity. Synergy was very strong with a combinatorial index (CI) of <0.3. The level of synergy is reflected by the CI values. CI values <1 are synergistic, values equal to 1 are additive, and those >1 are antagonistic. In an A375 melanoma BRAF V600E tumor xenograft model, both CRM1 inhibitor KPT-276 and BRAF inhibitor PLX-4720 slowed tumor growth as single agents when compared to untreated controls. However, complete and total tumor regression was observed when CRM1 and BRAF inhibitors were used in combination. The mechanism of synergy was attributed to increased levels of p53 and retinoblastoma proteins and a decrease in survivin by CRM1 inhibition, as well as to abrogation of extracellular signal-regulated kinase phosphorylation normally associated with CRM1 inhibition by the BRAF inhibitors. In conclusion, the authors found that CRM1 and BRAF inhibition synergizes and induces melanoma regression in metastatic melanoma cell lines and mouse xenografts.
4.2 Tyrosine kinase and CRM1 inhibitors in leukemia
Chronic myelogenous leukemia (CML) is a clonal disease of hematopoietic progenitor cells. In the chronic phase, CML is characterized by the abnormal expansion and accumulation of granulocytes in the circulation. However, the disease eventually progresses into a blast crisis phase, where acute leukemias of lymphoid or myeloid cells expand, resulting in patient mortality. Imatinib (STI571 or Gleevec), a tyrosine kinase inhibitor, targets Philadelphia (Ph+) chromosome-positive leukemias (both CML and ALL); however, over time, patients generally develop drug resistance. The Ph+ genetic mutation produces a chimeric BCR-ABL oncoprotein that is primarily localized in the cytoplasm where it activates proliferative and anti-apoptotic signaling pathways [42; 43]. BCR-ABL can inhibit apoptosis by activating PI3-kinase and Akt. However, when the Abl kinase protein is present in the nucleus, whether alone or in the chimeric form (BCR-ABL), it is activated by the presence of DNA damage and will induce a functional homolog of the tumor suppressor p53, p73. Nuclear activation of Abl kinase and p73 will result in DNA damage-induced apoptosis [44; 45]. In a proof of concept paper by Vigneri and Wang [46], they found that combining the CRM1 inhibitor leptomycin B and the BCL-ABL tyrosine kinase inhibitor imatinib induces nuclear localization (entrapment) of BCR-ABL and promotes apoptosis in CML cells in vitro and in BCR-ABL-transformed mouse bone marrow cells in vivo, sparing normal bone marrow stem cells. Combinatorial treatment was synergistic and resulted in the irreversible and complete killing of all of the BCR-ABL expressing cells, whereas single-agent treatment with the CRM1 inhibitor or imatinib only produced transient and reversible anti-CML effects.
In a follow-up study [47], the efficacy of the leptomycin B and imatinib mesylate combination was evaluated in human cells. CD34-positive normal cells from 6 healthy donors and myeloid progenitor cells from 35 CML patients were collected for the study. Sequential treatment of CD34+ cells with imatinib followed by leptomycin B induced apoptosis specifically in BCR-ABL-positive CML cells but had little effect on CD34/BCR-ABL double-negative cells from normal donors and normal myeloid progenitors derived from CML patients. Detection of BCR-ABL positive and negative cell colonies was based on molecular assay (RT-PCR). Since leptomycin B or imatinib, when used separately did not kill BCR-ABL CML cells, this would indicate that a combination approach may be of significant therapeutic value for newly diagnosed and imatinib-resistant CML patients.
A recent study by Walker et al [48] in Ph+ CML and ALL demonstrated that the CRM1 inhibitor KPT-330 induced apoptosis in leukemic but not normal CD34+ progenitor cells and increased survival in mice challenged with BCR-ABL+ cells. Survival was 50% after 15 weeks of KPT-330 treatment and BCR-ABL+ cells were significantly reduced in vivo. In addition, it was found that the CRM1 inhibitor potentiated the effect of the tyrosine kinase inhibitor imatinib in BCR-ABL+ cells after 24 hours. It was not determined whether this effect was statistically synergistic; however, combinatorial therapy was clearly more effective than either CRM1 inhibitor or tyrosine kinase inhibitor alone.
4.3 Melphalan
Melphalan is an alkylating agent that binds to DNA, inducing DNA damage and resulting in cancer cell death. Melphalan is still commonly used in standard dose induction therapy for the treatment of multiple myeloma and is the backbone for high-dose chemotherapy with autologous stem cell transplantation [49]. Melphalan, when used in combination with the CRM1 inhibitors KOS-2464, KPT-185, KPT-249 or the clinical compound KPT-330, synergistically induced cytotoxicity in human myeloma cell lines [15] (Table 2c). Synergy was stronger with sequential treatment of the CRM1 inhibitor followed 24 hours later by melphalan. The myeloma drugs lenalidomide and dexamethasone did not synergize with CRM1 inhibitors [15] (Table 2c).
4.4 Cisplatin/Oxaliplatin
Cisplatin is a DNA cross-linking agent that is used as a single agent or in combination with other therapeutics to treat numerous cancers, especially advanced or relapsed cancers and cancers that cannot be treated with radiation or surgery. Malignancies treated with cisplatin include bladder cancer, cervical cancer, malignant mesothelioma, non-small cell lung cancer, ovarian cancer, squamous cell carcinoma of the head and neck, neuroblastoma, and testicular cancer. Combination therapy using cisplatin and the classic inhibitor of CRM1 mediated export (leptomycin B) have been shown to induce apoptosis in a galectin-3 mutant and galectin-3 wild-type breast cancer cell line (BT-549) in a study by Takenaka et al [50]. Galactin-3 over-expression and sub-cellular localization have been linked to the development of cancer, including cancer cell growth and proliferation, transformation, apoptosis, angiogenesis, cell adhesion, invasion, and metastasis. The wide range of galectin-3 activities in cancer cells is due to its multiple inter- and sub-cellular localizations and interaction with a wide range of various binding partners [51]. When cancer cells are treated with a pro-apoptotic agent, such as cisplatin, galectin-3 is phosphorylated by a post-translational event and then exported from the nucleus to the cytoplasm. Phosphorylated cytoplasmic galactin-3 activates ERK and JNK, resulting in inhibition of both cytochrome c release and caspase activation with subsequent suppression of apoptosis in cancerous cells. CRM1 inhibition by leptomycin B prevents nuclear export of galectin-3, circumventing the anti-apoptotic function of galectin-3 and restoring cisplatin-induced apoptosis in breast cancer cells [50]. In 2013, three abstracts were published in Cancer Research on the use of small molecule CRM1 inhibitors (SINE) in combination with cisplatin (Table 1). In a recent abstract by Azmi et al [52] (Table 1), the platinum drug oxaliplatin synergistically induced apoptosis when used in combination with KPT-185 in a panel of human pancreatic and colon cancer cell lines. All cell lines tested exhibited synergy with the KPT-185/oxaliplatin combination, and cell death was mediated by nuclear PAR-4 expression.
4.5 Daunomycin
In our laboratory, we have tested the use of daunomycin in combination with the CRM1 inhibitors KOS-2464, KPT-185, KPT-249, KPT-276, or the clinical compound KPT-330 in acute myelogenous leukemia (AML). Human AML cell lines and ex vivo samples from bone marrow aspirates obtained from newly diagnosed and relapsed patients were treated with both CRM1 inhibitors and daunomycin and assayed for apoptosis by flow cytometry. Combined therapy induced apoptosis synergistically in both human AML cell lines [53] and patient bone marrow biopsies (unpublished data).
4.6 Doxorubicin
Significant progress has been made over the past several years in the treatment of multiple myeloma [54]. Patient median survival rates have increased from approximately 2 years to over 7 years due to improved therapies, including bortezomib/carfilzomib and lenalidomide/pomalidomide-based therapies and high-dose chemotherapy with autologous stem cell transplant [54]. However, patients eventually develop drug resistance, become refractory to all anti-multiple myeloma therapies, and die from progressive disease [55]. The incurable nature of multiple myeloma clearly demonstrates the need for novel agents and treatments. Therapies for patients with newly diagnosed or relapsed multiple myeloma often include treatment with pegylated liposomal doxorubicin or proteosome inhibitors bortezomib and carfilzomib [56; 57]. A very complete picture has emerged for the use of the topoisomerase II (topo II) inhibitor doxorubicin in combination with CRM1 inhibitors. Original observations published by our laboratory and others have strongly indicated that topo IIα is exported from the nucleus of drug-resistant high-density multiple myeloma cells (cell lines and patient cells) [37; 58; 59]. High-density multiple myeloma cells (conditions that approximate the bone marrow environment) demonstrate de novo resistance to topo II inhibitors [16; 37; 58; 59] (Figure 2A) because they traffic topo IIα to the cytoplasm (Figure 2B and 2C), thus sequestering topo IIα from the nuclear DNA and rendering it ineffective in generating lethal DNA strand breaks in the presence of a topo II inhibitor. The nuclear export of topo IIα depends on the binding of specific NES on topo IIα by CRM1 [16]. Nuclear proteins with a molecular mass >40 kDa are unable to passively diffuse across the nuclear membrane [9]. Those with a hydrophobic amino acid NES bind CRM1 and, in the presence of RanGTP, are transported through the NPC to the cytoplasm and released [9; 60]. We have shown that topo IIα has a NES at amino acids 1017-1028 (DILRDFFELRLK) and at amino acids 1054-1066 (FILEKIDGKIIIE) [16]. Thus, disrupting the binding between topo IIα-NES and CRM1 prevents the CRM1-mediated nuclear export of topo IIα and results in the sensitization of multiple myeloma cells to topo II inhibitors. CRM1 binding to topo II can be inhibited by CRM1 siRNA, CRM1 inhibitors, and topo IIα-specific NES inhibitors [16]. We found that CRM1 inhibition/knockdown prevented nuclear export of p53 and topo IIα in myeloma cells. In vitro studies in human myeloma cell lines determined that CRM1 inhibitors, including ratjadone C or small molecule inhibitors from Karyopharm Therapeutics (KPT-185, KPT-249, and KPT-330) or CRM1 siRNA sensitized drug resistant multiple myeloma cells to doxorubicin [38]. KPT-185T, the inactive trans-isomer of KPT-185, did not synergize with doxorubicin. Ex vivo studies using myeloma cells obtained from both newly diagnosed and relapsed patient bone marrow aspirates treated with SINE drugs and doxorubicin synergistically induced apoptosis [15] (Figure 3). Mononuclear cell fractions isolated from aspirates were treated with the drug combination and myeloma cells were identified by flow cytometry by staining with light-chain and CD138 antibodies. Non-myeloma cells, defined as light-chain/CD138 negative/negative, were unaffected by CRM1 inhibitor/doxorubicin treatment (Figure 3). Nuclear p53 and topo IIα proteins were increased by CRM1 inhibition; therefore, the addition of DNA damage by doxorubicin is likely to have produced a synergistic effect. A phase I/II clinical trial of the CRM1 inhibitor KPT-330 (Selinexor) in combination with pegylated liposomal doxorubicin and dexamethasone in patients with relapsed and refractory myeloma is being initiated at the Moffitt Cancer Center.
Figure 2. (A) Intracellular trafficking of topo IIα in low- and high-density myeloma cells.
Human H929, 8226, and U266 MM cells were grown for 16 hours at low or high density and treated with 2 μM doxorubicin for 4 hours (n = 2). Apoptosis was determined by caspase 3 staining. Log cells that maintained nuclear topo IIα were significantly more sensitive to topo IIα-targeted chemotherapy. (B) NCI-H929 topo IIα immunofluorescence. H929 human MM cells were grown at low and high densities and stained for cytoplasm (phalloidin, green), topo IIα (red), and DNA (DAPI, blue). Results indicate that topo IIα is located in the nucleus of low-density cells and is exported from the nucleus to the cytoplasm in high-density cells. (C) Myeloma cells export topo IIα in patient bone marrow aspirates. Plasma cells in a bone marrow aspirate from a relapsed myeloma patient were identified by kappa-light chain antibody (green) (b & e). Cells were also stained for topo IIα (c) (red), CRM1(f) (red), DNA (blue; dapi) (a & d). Figure reprinted in part by from Cancer Research 2009; 69:6899–905.
Figure 3. KPT-185, KPT-249, and KPT-330 sensitize patient myeloma cells to doxorubicin, bortezomib, and carfilzomib.
Bone marrow mononuclear cells were isolated and treated with SINE (300 nM) +/− doxorubicin (2 μM), bortezomib (10 nM), carfilzomib (20 nM), or DMSO vehicle control (VC) for 20 hours. Treated cells were fluorescently labeled with antibodies against activated caspase 3, CD138, and light chain (kappa or lambda). Results for patient multiple myeloma cells (n=12 patient samples) (A–C) and patient non-myeloma cells (n=12) (D–F) are shown. Apoptosis was induced when SINE was co-incubated with doxorubicin (P = 0.0004), bortezomib (P = 0.003), or carfilzomib (P = 0.002) in CD138/light-chain double-positive myeloma cell populations (A–C) but not in non-myeloma CD138/light-chain double-negative cells (P > 0.25) (D–F). This indicates that SINE may specifically inhibit neoplastic cells in MM patients. Figure reprinted by permission from Journal of Cancer 2013; 4:614–625.
4.7 Proteosome inhibitors
Proteosome inhibitors, bortezomib and carfilzomib, are used extensively for the treatment of multiple myeloma. In a study by Turner et al [15], bortezomib and carfilzomib were tested in combination with the CRM1 inhibitors KPT-185T, KPT-185, and KPT-249 and the clinical compound KPT-330 in human myeloma cell lines and ex vivo bone marrow aspirates. Ex vivo cells were obtained from both relapsed and newly diagnosed patients. Ficoll-isolated mononuclear cell fractions were treated with individual agents and in combination, with myeloma cells identified via flow cytometry by staining with light-chain and CD138 antibodies. Myeloma cells treated with the CRM1/proteasome inhibitor combination showed a synergistic decrease in proliferation and increased apoptosis as compared to single agents (Figure 3). Non-myeloma cells, defined as light-chain/CD138 negative/negative, were unaffected by CRM1 inhibitor/doxorubicin treatment (Figure 3). The CRM1 inhibitor KPT-185 has been found to increase IkB and p53 protein in the nucleus of chronic lymphocytic leukemia cells [61]. IkB down-regulates NF-kB, a constitutively active protein in myeloma that stimulates proliferation and cell survival. P53 is a critical regulator of apoptosis that is suppressed in myeloma and other cancers. We found that KPT-185, KPT-249, KPT-330, leptomycin B, and KOS-2464 increased IkB and p53 levels in multiple myeloma cells (unpublished results). Proteasome inhibitors are also known to increase expression of p53 and IkB levels [62]. The overall increase in nuclear p53 inducing apoptosis and down-regulation of NF-kB may result in enhanced cell death of the myeloma cells while not affecting normal lymphocyte populations.
4.8 Sequential versus concurrent treatment (Table 2)
We have studied the sequencing of drugs in the treatment of multiple myeloma as summarized in Table 2. Drugs connected by plus signs indicate concurrent treatment or simultaneous administration of both drugs to cell cultures and incubation for 48 hours. Drugs connected by arrows indicate that the drug listed first was added for 24 hours followed by the addition of the second drug for an additional 24 hours. The level of synergy is reflected by the CI values. CI values <1 are synergistic, values equal to 1 are additive, and those >1 are antagonistic. Table 2a is a summary of CRM1 inhibitors and proteasome inhibitors bortezomib or carfilzomib. In general, we found that sequential treatment (where the CRM1 inhibitor was given first followed by the proteasome inhibitor) was more effective than concurrent treatment or treatment in the reverse order (proteasome inhibitors followed by CRM1 inhibitor). Table 2b shows that sequential treatment with CRM1 inhibitor followed by doxorubicin is highly synergistic and that sequential treatment of CRM1 inhibitor followed by melphalan is synergistic (Table 2c). Dexamethasone and lenalidomide were not synergistic with the CRM1 inhibitor KPT-249.
We believe that drug sequencing analyses indicate that it is very likely that the CRM1 inhibitor restores, in addition to nuclear topo IIα, the function of p53, IkB, and other antiproliferative factors. This primes the cells for treatment by proteasome inhibitors or DNA damage induced by doxorubicin and melphalan.
5. Discussion
Cancer development is a multi-step process whereby many tumor suppressive proteins or cell cycle inhibitors are either silenced or compartmentalized within the cell where they are no longer effective at detecting DNA damage and/or promoting cell death [63]. CRM1 over-expression has been shown to be a causal factor, whereby oncogenes, tumor suppressive factors, and cell cycle inhibitors are mislocalized from the nucleus into the cytoplasm, resulting in promotion of the cancerous phenotype. Examples of mislocalized factors in cancer include APC [20; 64; 65; 66], BCR-ABL [46; 47; 48], Bok [67], BRCA1 [68; 69; 70], estradiol receptor [71; 72], FOXO [18; 73], galectin-3 [50; 74], HSP90 [75], Inf1/hSNF [21], Merlin [76], nucleophosmin [77; 78; 79; 80], N-WASP/FAK [81; 82; 83], p21 [84; 85; 86; 87; 88], p27 [89; 90; 91; 92; 93; 94], p53 [25; 95; 96; 97; 98], RASSF2 [99], survivin [100; 101], Tob [102; 103], DNA topoisomerase I [104; 105], and topo IIα [9; 15; 16; 106; 107]. Recent studies have shown that many cancer types, including AML [108], CLL [61], CML [48], glioma [109], mantle cell lymphoma [110], melanoma [40], multiple myeloma [111], non-Hodgkin lymphoma [112], osteosarcoma [113], ovarian cancer [114], and pancreatic cancer [115; 116], over-express CRM1. CRM1 protein expression may become a significant biomarker for disease progression in specific malignancies. In the past, we and others have proposed the use of targeting CRM1 in the treatment of cancer [9; 38; 68; 106; 117; 118; 119]; however, only until recent developments of less toxic therapeutics has the potential use of CRM1 inhibitors come closer to being realized. These less toxic small-molecule CRM1 inhibitors, though reasonably effective against cancer cell lines and slowing the growth of xenograft tumors in mice, may need additional help in the form of combinatorial therapies. Potential combinatorial therapies include the use CRM1 inhibitors and the addition of alkylating agents (melphalan), anthracyclines (doxorubicin and daunomycin), BRAF inhibitors, platinum drugs (cisplatin and oxaliplatin), proteosome inhibitors (bortezomib and carfilzomib), SN-38, or tyrosine-kinase inhibitors (imatinib). The idea behind these therapies is that blocking nuclear export by CRM1 small-molecule inhibitors may restore the apoptotic pathways of the tumor suppressive factors listed above and synergistically induce cancer cell death.
Acknowledgements
We thank Rasa Hamilton (Moffitt Cancer Center) for editorial assistance.
Funding Our study received valuable assistance from the Flow Cytometry Core Facility at the H. Lee Moffitt Cancer Center & Research Institute; an NCI designated Comprehensive Cancer Center, supported under NIH grant P30-CA76292. We especially thank Jodi Kroeger for her expert assistance with flow cytometry. This work was partially supported by the State of Florida Bankhead-Coley Team Science Project Grant 2BT03-43424. Experimental data presented in table 2 contains lab results that were supported by Karyopharm Therapeutics (Natick, MA).
Dr. R. Baz received research funding from Karyopharm for a phase I clinical trial.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Competing Interests The other authors declare that there are no conflicts of interest.
References
- [1].Reichelt R, Holzenburg A, Buhle EL, Jr, Jarnik M, Engel A, Aebi U. Correlation between structure and mass distribution of the nuclear pore complex and of distinct pore complex components. J Cell Bio. 1990;110:883–894. doi: 10.1083/jcb.110.4.883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Lim RY, Aebi U, Fahrenkrog B. Towards reconciling structure and function in the nuclear pore complex. Histochem Cell Biol. 2008;129:105–16. doi: 10.1007/s00418-007-0371-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Cook A, Bono F, Jinek M, Conti E. Structural biology of nucleocytoplasmic transport. Annu Rev Biochem. 2007;76:647–671. doi: 10.1146/annurev.biochem.76.052705.161529. [DOI] [PubMed] [Google Scholar]
- [4].Monecke T, Guttler T, Neumann P, Dickmanns A, Gorlich D, Ficner R. Crystal structure of the nuclear export receptor CRM1 in complex with Snurportin1 and RanGTP. Science. 2009;324:1087–1091. doi: 10.1126/science.1173388. [DOI] [PubMed] [Google Scholar]
- [5].Dong X, Biswas A, Chook YM. Structural basis for assembly and disassembly of the CRM1 nuclear export complex. Nat Struct Mol Biol. 2009;16:558–560. doi: 10.1038/nsmb.1586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Xu D, Grishin NV, Chook YM. NESdb: a database of NES-containing CRM1 cargoes. Mol Biol Cell. 2012;23:3673–3676. doi: 10.1091/mbc.E12-01-0045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Xu D, Farmer A, Collett G, Grishin NV, Chook YM. Sequence and structural analyses of nuclear export signals in the NESdb database. Mol Biol Cell. 2012;23:3677–3693. doi: 10.1091/mbc.E12-01-0046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Sun Q, Carrasco YP, Hu Y, Guo X, Mirzaei H, Macmillan J, Chook YM. Nuclear export inhibition through covalent conjugation and hydrolysis of Leptomycin B by CRM1. Proc Natl Acad Sci U S A. 2013;110:1303–1308. doi: 10.1073/pnas.1217203110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Turner JG, Dawson J, Sullivan DM. Nuclear export of proteins and drug resistance in cancer. Biochem Pharmacol. 2012;83:1021–1032. doi: 10.1016/j.bcp.2011.12.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Hill R, Cautain B, de Pedro N, Link W. Targeting nucleocytoplasmic transport in cancer therapy. Oncotarget. 2013 doi: 10.18632/oncotarget.1457. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Kudo N, Matsumori N, Taoka H, Fujiwara D, Schreiner EP, Wolff B, Yoshida M, Horinouchi S. Leptomycin B inactivates CRM1/exportin 1 by covalent modification at a cysteine residue in the central conserved region. Proc Natl Acad Sci U S A. 1999;96:9112–9117. doi: 10.1073/pnas.96.16.9112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Meissner T, Krause E, Vinkemeier U. Ratjadone and leptomycin B block CRM1-dependent nuclear export by identical mechanisms. FEBS Lett. 2004;576:27–30. doi: 10.1016/j.febslet.2004.08.056. [DOI] [PubMed] [Google Scholar]
- [13].Shacham S, Nir R, Daelemans D, Shechter S, Vancurova I, Juvekar A, Lau W, Higgins G, Draetta G, Kauffman M. KPT0127, a novel, potent and selective inhibitor of Crm1-mediated nuclear export shows selective cytoxicity for neoplastic over normal cells and is well tolerated in animals. Clin Can Res. 2010;16:A19. Abstract. [Google Scholar]
- [14].Sakakibara K, Saito N, Sato T, Suzuki A, Hasegawa Y, Friedman JM, Kufe DW, Vonhoff DD, Iwami T, Kawabe T. CBS9106 is a novel reversible oral CRM1 inhibitor with CRM1 degrading activity. Blood. 2011;118:3922–3931. doi: 10.1182/blood-2011-01-333138. [DOI] [PubMed] [Google Scholar]
- [15].Turner JG, Dawson J, Emmons MF, Cubitt CL, Kauffman M, Shacham S, Hazlehurst LA, Sullivan DM. CRM1 Inhibition Sensitizes Drug Resistant Human Myeloma Cells to Topoisomerase II and Proteasome Inhibitors both In Vitro and Ex Vivo. J Cancer. 2013;4:614–625. doi: 10.7150/jca.7080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Turner JG, Engel R, Derderian JA, Jove R, Sullivan DM. Human topoisomerase IIalpha nuclear export is mediated by two CRM-1-dependent nuclear export signals. J Cell Sci. 2004;117:3061–3071. doi: 10.1242/jcs.01147. [DOI] [PubMed] [Google Scholar]
- [17].Turner JG, Dawson J, Sullivan DM. Targeting the nuclear export signal in multiple myeloma. Blood. 2013;122:1925. Abstract. [Google Scholar]
- [18].Vogt PK, Jiang H, Aoki M. Triple layer control: phosphorylation, acetylation and ubiquitination of FOXO proteins. Cell Cycle. 2005;4:908–913. doi: 10.4161/cc.4.7.1796. [DOI] [PubMed] [Google Scholar]
- [19].Craig E, Zhang ZK, Davies KP, Kalpana GV. A masked NES in INI1/hSNF5 mediates hCRM1-dependent nuclear export: implications for tumorigenesis. Embo J. 2002;21:31–42. doi: 10.1093/emboj/21.1.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Powell SM, Zilz N, Beazer-Barclay Y, Bryan TM, Hamilton SR, Thibodeau SN, Vogelstein B, Kinzler KW. APC mutations occur early during colorectal tumorigenesis. Nature. 1992;359:235–237. doi: 10.1038/359235a0. [DOI] [PubMed] [Google Scholar]
- [21].Pichler A, Melchior F. Ubiquitin-related modifier SUMO1 and nucleocytoplasmic transport. Traffic. 2002;3:381–387. doi: 10.1034/j.1600-0854.2002.30601.x. [DOI] [PubMed] [Google Scholar]
- [22].Kutay U, Guttinger S. Leucine-rich nuclear-export signals: born to be weak. Trends Cell Biol. 2005;15:121–124. doi: 10.1016/j.tcb.2005.01.005. [DOI] [PubMed] [Google Scholar]
- [23].Yoneda Y, Hieda M, Nagoshi E, Miyamoto Y. Nucleocytoplasmic protein transport and recycling of Ran. Cell Struct Funct. 1999;24:425–433. doi: 10.1247/csf.24.425. [DOI] [PubMed] [Google Scholar]
- [24].Poon IK, Jans DA. Regulation of nuclear transport: central role in development and transformation? Traffic. 2005;6:173–186. doi: 10.1111/j.1600-0854.2005.00268.x. [DOI] [PubMed] [Google Scholar]
- [25].Zhang Y, Xiong Y. A p53 amino-terminal nuclear export signal inhibited by DNA damage-induced phosphorylation. Science. 2001;292:1910–1915. doi: 10.1126/science.1058637. [DOI] [PubMed] [Google Scholar]
- [26].Jain AK, Jaiswal AK. Phosphorylation of tyrosine 568 controls nuclear export of Nrf2. J Biol Chem. 2006;281:12132–12142. doi: 10.1074/jbc.M511198200. [DOI] [PubMed] [Google Scholar]
- [27].Turner JG, Gump J, Ferraro B, Sullivan D. Nuclear and cytoplasmic topoisomerase II alpha are differentially phosphorylated and nuclear export is blocked by CRM1 antisense oligonucleotides. AACR Meeting Abstracts. 2005;46:5945. [Google Scholar]
- [28].von Knethen A, Tzieply N, Jennewein C, Brune B. Casein-kinase-II-dependent phosphorylation of PPARgamma provokes CRM1-mediated shuttling of PPARgamma from the nucleus to the cytosol. J Cell Sci. 2010;123:192–201. doi: 10.1242/jcs.055475. [DOI] [PubMed] [Google Scholar]
- [29].Turner JG, Dawson JL, Sullivan DM. Casein Kinase II Modulates Topoisomerase II Alpha Nuclear Export and Drug Sensitivity in Multiple Myeloma. Blood. 2012;120 Abstract. [Google Scholar]
- [30].Newlands ES, Rustin GJ, Brampton MH. Phase I trial of elactocin. Br J Cancer. 1996;74:648–649. doi: 10.1038/bjc.1996.415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Garzon R, Brown P.d.N., Yee K, Lancet JE, Gutierrez M, Gabrail N, Mau-Sorensen M, Baz R, Byrd JC, Kuruvilla J, Siegel DS, Shacham S, Rashal T, Yau CY, CCRP. McCauley D, Saint-Martin J-R, McCartney J, Landesman Y, Klebanov B, Pond G, Oza AM, Kauffman M, Mirza MR, Stone RM. Phase i trial of selinexor (KPT-330), a first-in-class oral selective inhibitor of nuclear export (SINE) in patients (pts) with advanced acute myelogenous leukemia (AML) Blood. 2013;122:1440. Abstract. [Google Scholar]
- [32].Gutierrez M, de Nully Brown P, Gabrail N, Baz R, Reece DE, Savona M, Trudel S, Siegel DS, Mau-Sorensen M, Kuruvilla J, Byrd JC, Shacham S, Rashal T, Yau CY, CCRP. McCauley D, Saint-Martin J-R, McCartney J, Landesman Y, Klebanov B, Pond G, Oza AM, Kauffman M, Mirza MR. Anti tumor activity of selinexor (KPT-330), a first-in-class oral selective inhibitor of nuclear export (SINE) XPO1/CRM1 antagonist in patients (pts) with relapsed / refractory multiple myeloma (MM) or Waldenstrom's macroglobulinemia (WM) Blood. 2013;122:1942. Abstract. [Google Scholar]
- [33].Gutierrez M, Shah BD, Gabrail NY, de Nully Brown P, Stone RM, Garzon R, Savona M, Siegel DS, Baz R, Mau-Sorensen M, Davids MS, Byrd JC, Shacham S, Rashal T, Yau CY, McCauley D, Saint-Martin J-R, McCartney J, Landesman Y, Klebanov B, Pond G, Oza AM, Kauffman M, Mirza MR. Preliminary evidence of anti tumor activity of selinexor (KPT-330) In a phase i trial of a first-in-class oral selective inhibitor of nuclear export (SINE) in patients (pts) with relapsed / refractory non-Hodgkin's lymphoma (NHL) and chronic lymphocytic leukemia (CLL) Blood. 2013;122:90. Abstract. [Google Scholar]
- [34].Mau-Soerensen M, Razak ARA, Mahipal A, Mahaseth H, Gerecitano JF, Shacham S, Yau CYF, Lassen U, Shields AF, McCauley D, Cooksey J, Tan DSP, Rashal T, Shacham E, Landesman Y, Pond G, Oza AM, Kauffman M, Siu L, Mirza MR. Safety and antitumor activity of selinexor (KPT-330), a first-in-class, oral XPO1 selective inhibitor of nuclear export: A phase I study expanded with colon cancer cohort. ASCO Meeting Abstracts. 2014;32:482. [Google Scholar]
- [35].Razak ARA, Soerensen MM, Mahipal A, Shacham S, Yau CYF, Lassen UN, McCauley D, Cooksey J, Tan DSP, Saint-Martin J-R, Landesman Y, Pond G, Oza AM, Kauffman M, Siu LL, Mirza MR. First-in-class, first-in-human phase I trial of KPT-330, a selective inhibitor of nuclear export (SINE) in patients (pts) with advanced solid tumors. ASCO Meeting Abstracts. 2013;31:2505. [Google Scholar]
- [36].Orlowski RZ, Nagler A, Sonneveld P, Blade J, Hajek R, Spencer A, San Miguel J, Robak T, Dmoszynska A, Horvath N, Spicka I, Sutherland HJ, Suvorov AN, Zhuang SH, Parekh T, Xiu L, Yuan Z, Rackoff W, Harousseau JL. Randomized phase III study of pegylated liposomal doxorubicin plus bortezomib compared with bortezomib alone in relapsed or refractory multiple myeloma: combination therapy improves time to progression. J Clin Oncol. 2007;25:3892–3901. doi: 10.1200/JCO.2006.10.5460. [DOI] [PubMed] [Google Scholar]
- [37].Engel R, Valkov NI, Gump JL, Hazlehurst L, Dalton WS, Sullivan DM. The cytoplasmic trafficking of DNA topoisomerase IIalpha correlates with etoposide resistance in human myeloma cells. Exp Cell Res. 2004;295:421–431. doi: 10.1016/j.yexcr.2004.01.012. [DOI] [PubMed] [Google Scholar]
- [38].Turner JG, Marchion DC, Dawson JL, Emmons MF, Hazlehurst LA, Washausen P, Sullivan DM. Human multiple myeloma cells are sensitized to topoisomerase II inhibitors by CRM1 inhibition. Cancer Res. 2009;69:6899–6905. doi: 10.1158/0008-5472.CAN-09-0484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Jakob JA, Bassett RL, Jr, Ng CS, Curry JL, Joseph RW, Alvarado GC, Rohlfs ML, Richard J, Gershenwald JE, Kim KB, Lazar AJ, Hwu P, Davies MA. NRAS mutation status is an independent prognostic factor in metastatic melanoma. Cancer. 2012;118:4014–4023. doi: 10.1002/cncr.26724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Pathria G, Wagner C, Wagner SN. Inhibition of CRM1-mediated nucleocytoplasmic transport: triggering human melanoma cell apoptosis by perturbing multiple cellular pathways. J Invest Dermatol. 2012;132:2780–2790. doi: 10.1038/jid.2012.233. [DOI] [PubMed] [Google Scholar]
- [41].Salas Fragomeni RA, Chung HW, Landesman Y, Senapedis W, Saint-Martin JR, Tsao H, Flaherty KT, Shacham S, Kauffman M, Cusack JC. CRM1 and BRAF inhibition synergize and induce tumor regression in BRAF-mutant melanoma. Mol Cancer Ther. 2013;12:1171–1179. doi: 10.1158/1535-7163.MCT-12-1171. [DOI] [PubMed] [Google Scholar]
- [42].Faderl S, Talpaz M, Estrov Z, Kantarjian HM. Chronic myelogenous leukemia: biology and therapy. Ann Intern Med. 1999;131:207–219. doi: 10.7326/0003-4819-131-3-199908030-00008. [DOI] [PubMed] [Google Scholar]
- [43].Faderl S, Talpaz M, Estrov Z, O'Brien S, Kurzrock R, Kantarjian HM. The biology of chronic myeloid leukemia. N Engl J Med. 1999;341:164–172. doi: 10.1056/NEJM199907153410306. [DOI] [PubMed] [Google Scholar]
- [44].Wang JY. Regulation of cell death by the Abl tyrosine kinase. Oncogene. 2000;19:5643–5650. doi: 10.1038/sj.onc.1203878. [DOI] [PubMed] [Google Scholar]
- [45].Gong JG, Costanzo A, Yang HQ, Melino G, Kaelin WG, Jr, Levrero M, Wang JY. The tyrosine kinase c-Abl regulates p73 in apoptotic response to cisplatin-induced DNA damage. Nature. 1999;399:806–809. doi: 10.1038/21690. [DOI] [PubMed] [Google Scholar]
- [46].Vigneri P, Wang JY. Induction of apoptosis in chronic myelogenous leukemia cells through nuclear entrapment of BCR-ABL tyrosine kinase. Nature Med. 2001;7:228–234. doi: 10.1038/84683. [DOI] [PubMed] [Google Scholar]
- [47].Aloisi A, Di Gregorio S, Stagno F, Guglielmo P, Mannino F, Sormani MP, Bruzzi P, Gambacorti-Passerini C, Saglio G, Venuta S, Giustolisi R, Messina A, Vigneri P. BCR-ABL nuclear entrapment kills human CML cells: ex vivo study on 35 patients with the combination of imatinib mesylate and leptomycin B. Blood. 2006;107:1591–1598. doi: 10.1182/blood-2005-05-2123. [DOI] [PubMed] [Google Scholar]
- [48].Walker CJ, Oaks JJ, Santhanam R, Neviani P, Harb JG, Ferenchak G, Ellis JJ, Landesman Y, Eisfeld AK, Gabrail NY, Smith CL, Caligiuri MA, Hokland P, Roy DC, Reid A, Milojkovic D, Goldman JM, Apperley J, Garzon R, Marcucci G, Shacham S, Kauffman MG, Perrotti D. Preclinical and clinical efficacy of XPO1/CRM1 inhibition by the karyopherin inhibitor KPT-330 in Ph+ leukemias. Blood. 2013;122:3034–3044. doi: 10.1182/blood-2013-04-495374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Lonial S, Miguel JF. Induction therapy for newly diagnosed multiple myeloma. J Natl Compr Canc Netw. 2013;11:19–28. doi: 10.6004/jnccn.2013.0005. [DOI] [PubMed] [Google Scholar]
- [50].Takenaka Y, Fukumori T, Yoshii T, Oka N, Inohara H, Kim HR, Bresalier RS, Raz A. Nuclear export of phosphorylated galectin-3 regulates its antiapoptotic activity in response to chemotherapeutic drugs. Mol Cell Biol. 2004;24:4395–4406. doi: 10.1128/MCB.24.10.4395-4406.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Newlaczyl AU, Yu LG. Galectin-3--a jack-of-all-trades in cancer. Cancer Lett. 2011;313:123–128. doi: 10.1016/j.canlet.2011.09.003. [DOI] [PubMed] [Google Scholar]
- [52].Azmi AS, Kauffman M, McCauley D, Shacham S, Mohammad RM. Novel small-molecule CRM-1 inhibitor for GI cancer therapy. J Clin Oncol. 2012;30 Abstract. [Google Scholar]
- [53].Turner JG, Dawson J, Sullivan D. KOS-2464, a novel CRM1 inhibitor and a potential drug for multiple myeloma and leukemia. Cancer Res. 2012;72 Abstract. [Google Scholar]
- [54].Alexanian R, Delasalle K, Wang M, Thomas S, Weber D. Curability of multiple myeloma. Bone Marrow Res. 2012;2012:916479. doi: 10.1155/2012/916479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Kumar SK, Lee JH, Lahuerta JJ, Morgan G, Richardson PG, Crowley J, Haessler J, Feather J, Hoering A, Moreau P, LeLeu X, Hulin C, Klein SK, Sonneveld P, Siegel D, Blade J, Goldschmidt H, Jagannath S, Miguel JS, Orlowski R, Palumbo A, Sezer O, Rajkumar SV, Durie BG. Risk of progression and survival in multiple myeloma relapsing after therapy with IMiDs and bortezomib: a multicenter international myeloma working group study. Leukemia. 2012;26:149–157. doi: 10.1038/leu.2011.196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Zervas K, Mihou D, Katodritou E, Pouli A, Mitsouli CH, Anagnostopoulos A, Delibasi S, Kyrtsonis MC, Anagnostopoulos N, Terpos E, Zikos P, Maniatis A, Dimopoulos MA. VAD-doxil versus VAD-doxil plus thalidomide as initial treatment for multiple myeloma: results of a multicenter randomized trial of the Greek Myeloma Study Group. Ann Oncol. 2007;18:1369–1375. doi: 10.1093/annonc/mdm178. [DOI] [PubMed] [Google Scholar]
- [57].Jakubowiak AJ, Kendall T, Al-Zoubi A, Khaled Y, Mineishi S, Ahmed A, Campagnaro E, Brozo C, Braun T, Talpaz M, Kaminski MS. Phase II trial of combination therapy with bortezomib, pegylated liposomal doxorubicin, and dexamethasone in patients with newly diagnosed myeloma. J Clin Oncol. 2009;27:5015–5022. doi: 10.1200/JCO.2008.19.5370. [DOI] [PubMed] [Google Scholar]
- [58].Valkov NI, Gump JL, Engel R, Sullivan DM. Cell density-dependent VP-16 sensitivity of leukaemic cells is accompanied by the translocation of topoisomerase IIalpha from the nucleus to the cytoplasm. Br J Haematol. 2000;108:331–345. doi: 10.1046/j.1365-2141.2000.01832.x. [DOI] [PubMed] [Google Scholar]
- [59].Valkov NI, Sullivan DM. Drug resistance to DNA topoisomerase I and II inhibitors in human leukemia, lymphoma, and multiple myeloma. Semin Hematol. 1997;34:48–62. [PubMed] [Google Scholar]
- [60].Dong X, Biswas A, Suel KE, Jackson LK, Martinez R, Gu H, Chook YM. Structural basis for leucine-rich nuclear export signal recognition by CRM1. Nature. 2009;458:1136–1141. doi: 10.1038/nature07975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Lapalombella R, Sun Q, Williams K, Tangeman L, Jha S, Zhong Y, Goettl V, Mahoney E, Berglund C, Gupta S, Farmer A, Mani R, Johnson AJ, Lucas D, Mo X, Daelemans D, Sandanayaka V, Shechter S, McCauley D, Shacham S, Kauffman M, Chook YM, Byrd JC. Selective inhibitors of nuclear export show that CRM1/XPO1 is a target in chronic lymphocytic leukemia. Blood. 2012;120:4621–4634. doi: 10.1182/blood-2012-05-429506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Crawford LJ, Walker B, Irvine AE. Proteasome inhibitors in cancer therapy. J Cell Commun Signal. 2011;5:101–110. doi: 10.1007/s12079-011-0121-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- [64].Senda T, Iizuka-Kogo A, Onouchi T, Shimomura A. Adenomatous polyposis coli (APC) plays multiple roles in the intestinal and colorectal epithelia. Med Mol Morphol. 2007;40:68–81. doi: 10.1007/s00795-006-0352-5. [DOI] [PubMed] [Google Scholar]
- [65].Neufeld KL, Nix DA, Bogerd H, Kang Y, Beckerle MC, Cullen BR, White RL. Adenomatous polyposis coli protein contains two nuclear export signals and shuttles between the nucleus and cytoplasm. Proc Natl Acad Sci USA. 2000;97:12085–12090. doi: 10.1073/pnas.220401797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Rosin-Arbesfeld R, Cliffe A, Brabletz T, Bienz M. Nuclear export of the APC tumour suppressor controls beta-catenin function in transcription. Embo J. 2003;22:1101–1113. doi: 10.1093/emboj/cdg105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Bartholomeusz G, Wu Y, Ali Seyed M, Xia W, Kwong KY, Hortobagyi G, Hung MC. Nuclear translocation of the pro-apoptotic Bcl-2 family member Bok induces apoptosis. Mol Carcinogenesis. 2006;45:73–83. doi: 10.1002/mc.20156. [DOI] [PubMed] [Google Scholar]
- [68].Fabbro M, Henderson BR. Regulation of tumor suppressors by nuclear-cytoplasmic shuttling. Exp Cell Res. 2003;282:59–69. doi: 10.1016/s0014-4827(02)00019-8. [DOI] [PubMed] [Google Scholar]
- [69].Fabbro M, Rodriguez JA, Baer R, Henderson BR. BARD1 induces BRCA1 intranuclear foci formation by increasing RING-dependent BRCA1 nuclear import and inhibiting BRCA1 nuclear export. J Biol Chem. 2002;277:21315–21324. doi: 10.1074/jbc.M200769200. [DOI] [PubMed] [Google Scholar]
- [70].Rodriguez JA, Henderson BR. Identification of a functional nuclear export sequence in BRCA1. J Biol Chem. 2000;275:38589–38596. doi: 10.1074/jbc.M003851200. [DOI] [PubMed] [Google Scholar]
- [71].Castoria G, Migliaccio A, Auricchio F. Signaling-dependent nuclear export of estradiol receptor controls cell cycle progression in breast cancer cells. Mol Cellular Endocrinol. 2009;308:26–31. doi: 10.1016/j.mce.2009.01.006. [DOI] [PubMed] [Google Scholar]
- [72].Castoria G, Migliaccio A, Giovannelli P, Auricchio F. Cell proliferation regulated by estradiol receptor: Therapeutic implications. Steroids. 2010;75:524–527. doi: 10.1016/j.steroids.2009.10.007. [DOI] [PubMed] [Google Scholar]
- [73].Nakamura N, Ramaswamy S, Vazquez F, Signoretti S, Loda M, Sellers WR. Forkhead transcription factors are critical effectors of cell death and cell cycle arrest downstream of PTEN. Mol Cell Biol. 2000;20:8969–8982. doi: 10.1128/mcb.20.23.8969-8982.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].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;1800:181–189. doi: 10.1016/j.bbagen.2009.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Diehl MC, Idowu MO, Kimmelshue K, York TP, Elmore LW, Holt SE. Elevated expression of nuclear Hsp90 in invasive breast tumors. Cancer Biol Ther. 2009;8:1952–1961. doi: 10.4161/cbt.8.20.9639. [DOI] [PubMed] [Google Scholar]
- [76].Muranen T, Gronholm M, Renkema GH, Carpen O. Cell cycle-dependent nucleocytoplasmic shuttling of the neurofibromatosis 2 tumour suppressor merlin. Oncogene. 2005;24:1150–1158. doi: 10.1038/sj.onc.1208283. [DOI] [PubMed] [Google Scholar]
- [77].Falini B, Bolli N, Shan J, Martelli MP, Liso A, Pucciarini A, Bigerna B, Pasqualucci L, Mannucci R, Rosati R, Gorello P, Diverio D, Roti G, Tiacci E, Cazzaniga G, Biondi A, Schnittger S, Haferlach T, Hiddemann W, Martelli MF, Gu W, Mecucci C, Nicoletti I. Both carboxy-terminus NES motif and mutated tryptophan(s) are crucial for aberrant nuclear export of nucleophosmin leukemic mutants in NPMc+ AML. Blood. 2006;107:4514–4523. doi: 10.1182/blood-2005-11-4745. [DOI] [PubMed] [Google Scholar]
- [78].Karhemo PR, Rivinoja A, Lundin J, Hyvonen M, Chernenko A, Lammi J, Sihto H, Lundin M, Heikkila P, Joensuu H, Bono P, Laakkonen P. An extensive tumor array analysis supports tumor suppressive role for nucleophosmin in breast cancer. Am J Pathol. 2011;179:1004–1014. doi: 10.1016/j.ajpath.2011.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Khandelwal N, Simpson J, Taylor G, Rafique S, Whitehouse A, Hiscox J, Stark LA. Nucleolar NF-kappaB/RelA mediates apoptosis by causing cytoplasmic relocalization of nucleophosmin. Cell Death Diff. 2011;18:1889–902. doi: 10.1038/cdd.2011.79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Falini B, Gionfriddo I, Cecchetti F, Ballanti S, Pettirossi V, Martelli MP. Acute myeloid leukemia with mutated nucleophosmin (NPM1): any hope for a targeted therapy? Blood Rev. 2011;25:247–254. doi: 10.1016/j.blre.2011.06.001. [DOI] [PubMed] [Google Scholar]
- [81].Wang WS, Zhong HJ, Xiao DW, Huang X, Liao LD, Xie ZF, Xu XE, Shen ZY, Xu LY, Li EM. The expression of CFL1 and N-WASP in esophageal squamous cell carcinoma and its correlation with clinicopathological features. Disease Esophagus. 2010;23:512–521. doi: 10.1111/j.1442-2050.2009.01035.x. [DOI] [PubMed] [Google Scholar]
- [82].Sanchez AM, Flamini MI, Baldacci C, Goglia L, Genazzani AR, Simoncini T. Estrogen receptor-alpha promotes breast cancer cell motility and invasion via focal adhesion kinase and N-WASP. Mol Endocrin. 2010;24:2114–2125. doi: 10.1210/me.2010-0252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Bourguignon LY, Peyrollier K, Gilad E, Brightman A. Hyaluronan-CD44 interaction with neural Wiskott-Aldrich syndrome protein (N-WASP) promotes actin polymerization and ErbB2 activation leading to beta-catenin nuclear translocation, transcriptional up-regulation, and cell migration in ovarian tumor cells. J Biol Chem. 2007;282:1265–1280. doi: 10.1074/jbc.M604672200. [DOI] [PubMed] [Google Scholar]
- [84].Zhou BP, Liao Y, Xia W, Spohn B, Lee MH, Hung MC. Cytoplasmic localization of p21Cip1/WAF1 by Akt-induced phosphorylation in HER-2/neu-overexpressing cells. Nat Cell Biol. 2001;3:245–252. doi: 10.1038/35060032. [DOI] [PubMed] [Google Scholar]
- [85].Asada M, Yamada T, Ichijo H, Delia D, Miyazono K, Fukumuro K, Mizutani S. Apoptosis inhibitory activity of cytoplasmic p21(Cip1/WAF1) in monocytic differentiation. Embo J. 1999;18:1223–1234. doi: 10.1093/emboj/18.5.1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Hwang CY, Kim IY, Kwon KS. Cytoplasmic localization and ubiquitination of p21(Cip1) by reactive oxygen species. Biochem Biophys Res Commun. 2007;358:219–225. doi: 10.1016/j.bbrc.2007.04.120. [DOI] [PubMed] [Google Scholar]
- [87].Rodriguez-Vilarrupla A, Jaumot M, Abella N, Canela N, Brun S, Diaz C, Estanyol JM, Bachs O, Agell N. Binding of calmodulin to the carboxy-terminal region of p21 induces nuclear accumulation via inhibition of protein kinase C-mediated phosphorylation of Ser153. Mol Cell Biol. 2005;25:7364–7374. doi: 10.1128/MCB.25.16.7364-7374.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Keeshan K, Cotter TG, McKenna SL. Bcr-Abl upregulates cytosolic p21WAF-1/CIP-1 by a phosphoinositide-3-kinase (PI3K)-independent pathway. Br J Haematol. 2003;123:34–44. doi: 10.1046/j.1365-2141.2003.04538.x. [DOI] [PubMed] [Google Scholar]
- [89].Susaki E, Nakayama KI. Multiple mechanisms for p27(Kip1) translocation and degradation. Cell Cycle. 2007;6:3015–3020. doi: 10.4161/cc.6.24.5087. [DOI] [PubMed] [Google Scholar]
- [90].Ishida N, Hara T, Kamura T, Yoshida M, Nakayama K, Nakayama KI. Phosphorylation of p27Kip1 on serine 10 is required for its binding to CRM1 and nuclear export. J Biol Chem. 2002;277:14355–14358. doi: 10.1074/jbc.C100762200. [DOI] [PubMed] [Google Scholar]
- [91].Foster JS, Fernando RI, Ishida N, Nakayama KI, Wimalasena J. Estrogens down-regulate p27Kip1 in breast cancer cells through Skp2 and through nuclear export mediated by the ERK pathway. J Biol Chem. 2003;278:41355–41366. doi: 10.1074/jbc.M302830200. [DOI] [PubMed] [Google Scholar]
- [92].Shin I, Rotty J, Wu FY, Arteaga CL. Phosphorylation of p27Kip1 at Thr-157 interferes with its association with importin alpha during G1 and prevents nuclear re-entry. J Biol Chem. 2005;280:6055–6063. doi: 10.1074/jbc.M412367200. [DOI] [PubMed] [Google Scholar]
- [93].Min YH, Cheong JW, Kim JY, Eom JI, Lee ST, Hahn JS, Ko YW, Lee MH. Cytoplasmic mislocalization of p27Kip1 protein is associated with constitutive phosphorylation of Akt or protein kinase B and poor prognosis in acute myelogenous leukemia. Cancer Res. 2004;64:5225–5231. doi: 10.1158/0008-5472.CAN-04-0174. [DOI] [PubMed] [Google Scholar]
- [94].Connor MK, Kotchetkov R, Cariou S, Resch A, Lupetti R, Beniston RG, Melchior F, Hengst L, Slingerland JM. CRM1/Ran-mediated nuclear export of p27(Kip1) involves a nuclear export signal and links p27 export and proteolysis. Mol Biol Cell. 2003;14:201–213. doi: 10.1091/mbc.E02-06-0319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Nikolaev AY, Li M, Puskas N, Qin J, Gu W. Parc: a cytoplasmic anchor for p53. Cell. 2003;112:29–40. doi: 10.1016/s0092-8674(02)01255-2. [DOI] [PubMed] [Google Scholar]
- [96].Wadhwa R, Yaguchi T, Hasan MK, Mitsui Y, Reddel RR, Kaul SC. Hsp70 family member, mot-2/mthsp70/GRP75, binds to the cytoplasmic sequestration domain of the p53 protein. Exp Cell Res. 2002;274:246–253. doi: 10.1006/excr.2002.5468. [DOI] [PubMed] [Google Scholar]
- [97].Stommel JM, Marchenko ND, Jimenez GS, Moll UM, Hope TJ, Wahl GM. A leucine-rich nuclear export signal in the p53 tetramerization domain: regulation of subcellular localization and p53 activity by NES masking. Embo J. 1999;18:1660–1672. doi: 10.1093/emboj/18.6.1660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].O'Brate A, Giannakakou P. The importance of p53 location: nuclear or cytoplasmic zip code? Drug Resist Updat. 2003;6:313–322. doi: 10.1016/j.drup.2003.10.004. [DOI] [PubMed] [Google Scholar]
- [99].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;315:2775–2790. doi: 10.1016/j.yexcr.2009.06.013. [DOI] [PubMed] [Google Scholar]
- [100].Knauer SK, Bier C, Habtemichael N, Stauber RH. The Survivin-Crm1 interaction is essential for chromosomal passenger complex localization and function. EMBO. 2006;7:1259–1265. doi: 10.1038/sj.embor.7400824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Knauer SK, Kramer OH, Knosel T, Engels K, Rodel F, Kovacs AF, Dietmaier W, Klein-Hitpass L, Habtemichael N, Schweitzer A, Brieger J, Rodel C, Mann W, Petersen I, Heinzel T, Stauber RH. Nuclear export is essential for the tumor-promoting activity of survivin. FASEB J. 2007;21:207–216. doi: 10.1096/fj.06-5741com. [DOI] [PubMed] [Google Scholar]
- [102].Maekawa M, Yamamoto T, Nishida E. Regulation of subcellular localization of the antiproliferative protein Tob by its nuclear export signal and bipartite nuclear localization signal sequences. Exp Cell Res. 2004;295:59–65. doi: 10.1016/j.yexcr.2003.12.016. [DOI] [PubMed] [Google Scholar]
- [103].O'Malley S, Su H, Zhang T, Ng C, Ge H, Tang CK. TOB suppresses breast cancer tumorigenesis. Int J Cancer. 2009;125:1805–1813. doi: 10.1002/ijc.24490. [DOI] [PubMed] [Google Scholar]
- [104].Danks MK, Garrett KE, Marion RC, Whipple DO. Subcellular redistribution of DNA topoisomerase I in anaplastic astrocytoma cells treated with topotecan. Cancer Res. 1996;56:1664–1673. [PubMed] [Google Scholar]
- [105].Santos A, Calvet L, Terrier-Lacombe MJ, Larsen A, Benard J, Pondarre C, Aubert G, Morizet J, Lavelle F, Vassal G. In vivo treatment with CPT-11 leads to differentiation of neuroblastoma xenografts and topoisomerase I alterations. Cancer Res. 2004;64:3223–3229. doi: 10.1158/0008-5472.can-03-2915. [DOI] [PubMed] [Google Scholar]
- [106].Turner JG, Sullivan DM. CRM1-mediated nuclear export of proteins and drug resistance in cancer. Curr Med Chem. 2008;15:2648–2655. doi: 10.2174/092986708786242859. [DOI] [PubMed] [Google Scholar]
- [107].Oloumi A, MacPhail SH, Johnston PJ, Banath JP, Olive PL. Changes in subcellular distribution of topoisomerase IIalpha correlate with etoposide resistance in multicell spheroids and xenograft tumors. Cancer Res. 2000;60:5747–5753. [PubMed] [Google Scholar]
- [108].Kojima K, Kornblau SM, Ruvolo V, Dilip A, Duvvuri S, Davis RE, Zhang M, Wang Z, Coombes KR, Zhang N, Qiu YH, Burks JK, Kantarjian H, Shacham S, Kauffman M, Andreeff M. Prognostic impact and targeting of CRM1 in acute myeloid leukemia. Blood. 2013;121:4166–4174. doi: 10.1182/blood-2012-08-447581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].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;65:153–159. doi: 10.1227/01.NEU.0000348550.47441.4B. discussion 159–160. [DOI] [PubMed] [Google Scholar]
- [110].Zhang K, Wang M, Tamayo AT, Shacham S, Kauffman M, Lee J, Zhang L, Ou Z, Li C, Sun L, Ford RJ, Pham LV. Novel selective inhibitors of nuclear export CRM1 antagonists for therapy in mantle cell lymphoma. Exp Hematol. 2013;41:67–78. e64. doi: 10.1016/j.exphem.2012.09.002. [DOI] [PubMed] [Google Scholar]
- [111].Schmidt J, Braggio E, Kortuem KM, Egan JB, Zhu YX, Xin CS, Tiedemann RE, Palmer SE, Garbitt VM, McCauley D, Kauffman M, Shacham S, Chesi M, Bergsagel PL, Stewart AK. Genome-wide studies in multiple myeloma identify XPO1/CRM1 as a critical target validated using the selective nuclear export inhibitor KPT-276. Leukemia. 2013;27:2357–2365. doi: 10.1038/leu.2013.172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Azmi AS, Al-Katib A, Aboukameel A, McCauley D, Kauffman M, Shacham S, Mohammad RM. Selective inhibitors of nuclear export for the treatment of non-Hodgkin's lymphomas. Haematologica. 2013;98:1098–1106. doi: 10.3324/haematol.2012.074781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Yao Y, Dong Y, Lin F, Zhao H, Shen Z, Chen P, Sun YJ, Tang LN, Zheng SE. The expression of CRM1 is associated with prognosis in human osteosarcoma. Oncol Rep. 2009;21:229–235. [PubMed] [Google Scholar]
- [114].Noske A, Weichert W, Niesporek S, Roske A, Buckendahl AC, Koch I, Sehouli J, Dietel M, Denkert C. Expression of the nuclear export protein chromosomal region maintenance/exportin 1/Xpo1 is a prognostic factor in human ovarian cancer. Cancer. 2008;112:1733–43. doi: 10.1002/cncr.23354. [DOI] [PubMed] [Google Scholar]
- [115].Azmi AS, Aboukameel A, Bao B, Sarkar FH, Philip PA, Kauffman M, Shacham S, Mohammad RM. Selective inhibitors of nuclear export block pancreatic cancer cell proliferation and reduce tumor growth in mice. Gastroenterology. 2013;144:447–456. doi: 10.1053/j.gastro.2012.10.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].Huang WY, Yue L, Qiu WS, Wang LW, Zhou XH, Sun YJ. Prognostic value of CRM1 in pancreas cancer. Clin Invest Med. 2009;32:E315–E321. [PubMed] [Google Scholar]
- [117].Yashiroda Y, Yoshida M. Nucleo-cytoplasmic transport of proteins as a target for therapeutic drugs. Curr Med Chem. 2003;10:741–748. doi: 10.2174/0929867033457791. [DOI] [PubMed] [Google Scholar]
- [118].Zaidi SK, Young DW, Javed A, Pratap J, Montecino M, van Wijnen A, Lian JB, Stein JL, Stein GS. Nuclear microenvironments in biological control and cancer. Nat Rev Cancer. 2007;7:454–463. doi: 10.1038/nrc2149. [DOI] [PubMed] [Google Scholar]
- [119].Kau TR, Way JC, Silver PA. Nuclear transport and cancer: from mechanism to intervention. Nat Rev Cancer. 2004;4:106–117. doi: 10.1038/nrc1274. [DOI] [PubMed] [Google Scholar]



