Preclinical Assessment with Clinical Validation of Selinexor with Gemcitabine and Nab-Paclitaxel for the Treatment of Pancreatic Ductal Adenocarcinoma

Asfar S Azmi, Husain Yar Khan, Irfana Muqbil, Amro Aboukameel, Jasper E Neggers, Dirk Daelemans, Amit Mahipal, Gregory Dyson, Mandana Kamgar, Mohammad Najeeb Al-Hallak, Anteneh Tesfaye, Steve Kim, Vinod Shidham, Ramzi M Mohammad, Philip A Philip, Asfar S Azmi, Husain Yar Khan, Irfana Muqbil, Amro Aboukameel, Jasper E Neggers, Dirk Daelemans, Amit Mahipal, Gregory Dyson, Mandana Kamgar, Mohammad Najeeb Al-Hallak, Anteneh Tesfaye, Steve Kim, Vinod Shidham, Ramzi M Mohammad, Philip A Philip

Abstract

Purpose: Pancreatic ductal adenocarcinoma (PDAC) remains a deadly disease urgently requiring new treatments. Overexpression of the protein transporter exportin-1 (XPO1) leads to mislocalization of tumor-suppressor proteins (TSP) and their inactivation. Earlier, we showed that blocking XPO1 by CRISPR/Cas9 validated Selective Inhibitor of Nuclear Export (SINE) compounds (selinexor and analogs) restores the antitumor activity of multiple TSPs leading to suppression of PDAC in vitro and in orthotopic models.

Experimental design: We evaluate the synergy between SINE compounds and standard-of-care treatments in preclinical models and in a PDAC Phase Ib trial.

Results: SINE compounds synergize with gemcitabine (GEM) and nanoparticle albumin-bound (nab)-paclitaxel leading to suppression of PDAC cellular growth and cancer stem cell (CSC) spheroids disintegration. Label-free quantitative proteome profiling with nuclear and cytoplasmic enrichment showed superior enhancement in nuclear protein fraction in combination treatment. Selinexor inhibited the growth of PDAC CSC and two patient-derived (PDX) subcutaneous xenografts. Selinexor-GEM-nab-paclitaxel blocked PDX and orthotopic tumor growth. In a phase 1b study (NCT02178436), 9 patients were exposed to selinexor (60 mg oral) with GEM (1,000 mg/m2 i.v.) and nab-paclitaxel (125 mg/m2 i.v.) on days 1, 8, and 15 of 28-day cycle. Two patients showed partial response, and 2 had stable disease. An outstanding, durable objective response was observed in one of the responders with progression-free survival of 16 months and overall survival of 22 months.

Conclusions: Our preclinical and ongoing clinical study lends support to the use of selinexor-GEM-nab-paclitaxel as an effective therapy for metastatic PDAC.

Conflict of interest statement

Conflict of interest statement: None to declare.

©2019 American Association for Cancer Research.

Figures

Figure 1.. Selinexor synergizes with GEM and…
Figure 1.. Selinexor synergizes with GEM and nab-paclitaxel in PDAC cellular models.
MiaPaCa-2 [A] or L3.6pl [B] cells growing in 96 well plates (5,000 cells/well) were exposed to vehicle, selinexor, GEM-nab-paclitaxel or selinexor-GEM-nab-paclitaxel for 72 hrs and standard MTT assay was performed. Isobologram analysis was performed and combination index (CI) was calculated using GraphPad Prism software. Data is representative of 3 independent experiments. MiaPaCa-2 [C] or L3.6pl [D] cells growing in 6 well plates (50,000 cells/well) for 24 hrs and exposed to selinexor (300 nM), GEM (90 nM), nab-paclitaxel (1.5 nM), selinexor+GEM, selinexor+nab-paclitaxel, GEM+nab-paclitaxel or a triple combination of selinexor, GEM and nab-paclitexal for 72 hrs. The cells were trypsinized and collected. 1000 cells from each treatment group were reseeded in 100 mm petri dishes. Clonogenic assay was performed. The plates were photographed under a light microscope (Nikon). L3.6pl [E] or MiaPaCa-2 [F] cells were grown in 6 well plates in duplicate and exposed to different drugs (at doses mimicking clonogenic assay) for 72 hrs. At the end of the treatment period, cells were trypsinized and 10,000 cells were subjected to Annexin V FITC analysis (Biovision USA).
Figure 2.. Selinexor synergizes with GEM-nab-paclitaxel in…
Figure 2.. Selinexor synergizes with GEM-nab-paclitaxel in PDAC CSCs.
[A and B] Equal number of Panc-1 or MiaPaCa-2 CSC spheroids were seeded in ultra-low attachment plates for 1 week. Spheroids were exposed to KPT-330, GEM, nab-paclitaxel (100 nM) each or their combination for 1 week. Spheroids were counted and photographed under an inverted light microscope fitted with camera (at 100X magnification). [C] MiaPaCa-2 cells growing in 6 well plate (50,000 cells/well) in duplicate were exposed to different SINE analogs (100 nM each) for 72 hrs. RNA was isolated using standard methods. The RNA was subjected to RT-PCR using standard methods. The expression of CSC markers CD24, vimentin and CD44 was normalized to GAPDH (*p<0.05 and **p<0.001). Spheroid data is representative of two independent experiments.
Figure 3.. Selinexor-GEM-nab-paclitaxel retains TSPs in the…
Figure 3.. Selinexor-GEM-nab-paclitaxel retains TSPs in the nuclei of PDAC cellular models.
L3.6pl or MiaPaCa-2 cells grown on slides (3000 cells/well) and were exposed to vehicle or a combination of selinexor (300 nM), GEM (150 nM) and nab-paclitaxel (1.5 nM) for 24 hrs followed by IF analysis for nuclear TSPs (FOXO3a, Hsp90 and PAR-4).
Figure 4.. Proteomic profiling for nuclear retention…
Figure 4.. Proteomic profiling for nuclear retention of proteins.
MiaPaCa-2 cells were exposed to 1 μM selinexor, 1 μM selinexor plus 300 nM GEM and 3 nM nab-paclitaxel for 24 hours. The cell pellets were collected and analyzed by Label-Free Quantitative (LFQ) Proteome Profiling with nuclear and cytoplasmic enrichment using Thermo Q-Exactive HF-X Orbitrap Mass Spectrometer at Bioproximity. The intensity value of total 27528 proteins within 3617 protein sets in each samples was read. [A] Protein intensity value changes across samples. [B] Percentage of up-regulated and down regulated genes in nuclei of MiaPaCa-2 cells post selinexor single agent treatment. [C] Annotation of molecular function of proteins up-regulated in nucleus after single agent selinexor treatment. [D] Annotation of the protein class up-regulated in the nucleus post selinexor single agent treatment. [E] Percentage of nuclear proteins up-regulated post selinexor-GEM-nab-paclitaxel treatment.
Figure 5.. XPO1 CRISPRCas9 genome editing abrogates…
Figure 5.. XPO1 CRISPRCas9 genome editing abrogates the activity of selinexor-GEM-nab-paclitaxel.
[A] Schema of CRISPR/Cas9-induced homologous recombination of XPO1 (adopted from our previous publication (Chem and Biol 22 (1) 107–116, 2015). [B] HEK-293 wild-type or HEK-XPO1C528S mutant cells were grown at a density of 5,000 cells/well in chambered slides. The next day, cells were exposed to indicated concentrations of either vehicle (DMSO) or selinexor for 2 hrs. After the treatment was over, IF analysis was performed using RanBP1 antibody (Cell Signaling USA). [C] HEK-293 wild-type or [D] HEK-XPO1C528S mutant cells were grown at a density of 5,000 cells/well in 96 well plates. Cells were treated with selinexor or GEM-nab-paclitaxel or their combination for additional 72 hrs at indicated doses. MTT assay was performed using standard methods. [E] HEK-293 wild-type or [F] HEK-XPO1C528S mutant cells were grown at a density of 50,000 cells/well in six well plates and exposed to selinexor (150 nM), GEM (75 nM) and nab-paclitaxel (0.75 nM) for 72 hrs. Apoptosis analysis was performed using Annexin V FITC analysis. [G] HEK-293 wild-type or HEK-XPO1C528S mutant cells were grown in chambered slides (5,000 cells per well) in duplicate and were exposed to selinexor (150 nM), GEM (75 nM) and nab-paclitaxel (0.75 nM) for 24 hrs. IF assay was performed using Hsp90 antibody.
Figure 6.. Selinexor-GEM-nab-paclitaxel anti-tumor activity in PDAC…
Figure 6.. Selinexor-GEM-nab-paclitaxel anti-tumor activity in PDAC CSC and patient derived xenograft.
MiaPaCa-2 cells were flow sorted for CSC markers (CD44, CD133, EpCAM). These CSCs form tumor from 20,000 cells [A] Twelve mice were injected with CSCs. When palpable tumors were formed, mice were divided into two groups of five mice each and exposed to selinexor as single agent (used at 15 mg/kg every other day x3 weeks). Tumor weight was recorded every three days. [B] 10 mice bearing CSCs were divided into two groups. Group I vehicle and group II selinexor at sub-MTD (5 mg/kg twice a weekx3weeks) in combination with GEM (50 mg/kg i.v.) and nab-paclitaxel (30 mg/kg i.v.) (**p<0.01). [C] Anti-tumor activity of selinexor or KPT-8602 (Eltanexor) in PDX sub-cutaneous xenograft. [D] Photograph of residual tumors post treatment. [E] Anti-tumor activity of selinexor in combination with GEM-nab-paclitaxel (triple combination) compared to selinexor (single agent) or vehicle control in PDAC Pdx. Selinexor was administered orally (5 mg/kg), while gemcitabine (GEM) and nab-paclitaxel (PAC) were given i.v. 30 mg/kg and 20 mg/kg respectively. [F] Photographs showing reduction in tumor size in the combination treatment on day 53 post MiaPACa-2 orthotopic implantation. [G] Dot Graph representing gross tumor weights 72 hours post last treatment. Evaluable response to selinexor-GEM-nab-paclitaxel based treatment in patient with metastatic PDAC. [H] Before and after treatment CT-Scan showing the changes in the liver metastatic disease. [I] Serial measurements of the tumor marker CA19-9 in the patient with PDAC treated with a selinexor based regimen. Serum CA19-9 showed a precipitous drop after the first few treatments and a lower, steady concentration thereafter.

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin 2016;66:7–30.
    1. Kitano H Cancer robustness: tumour tactics. Nature 2003;426:125.
    1. Turner JG, Dawson J, Sullivan DM. Nuclear export of proteins and drug resistance in cancer. Biochem Pharmacol 2012;83:1021–32.
    1. Turner JG, Sullivan DM. CRM1-mediated nuclear export of proteins and drug resistance in cancer. Curr Med Chem 2008;15:2648–55.
    1. Moroianu J Distinct nuclear import and export pathways mediated by members of the karyopherin beta family. J Cell Biochem 1998;70:231–9.
    1. Azmi AS. The evolving role of nuclear transporters in cancer. Semin Cancer Biol 2014.
    1. Hutten S, Kehlenbach RH. CRM1-mediated nuclear export: to the pore and beyond. Trends Cell Biol 2007;17:193–201.
    1. Fukuda M, Asano S, Nakamura T, et al. CRM1 is responsible for intracellular transport mediated by the nuclear export signal. Nature 1997;390:308–11.
    1. Shaulsky G, Goldfinger N, Tosky MS, Levine AJ, Rotter V. Nuclear localization is essential for the activity of p53 protein. Oncogene 1991;6:2055–65.
    1. Turner JG, Dawson J, Cubitt CL, Baz R, Sullivan DM. Inhibition of CRM1-dependent nuclear export sensitizes malignant cells to cytotoxic and targeted agents. Semin Cancer Biol 2014;27:62–73.
    1. Azmi AS, Mohammad RM. Targeting Cancer at the Nuclear Pore. J Clin Oncol 2016;34:4180–2.
    1. 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.
    1. Yashiroda Y, Yoshida M. Nucleo-cytoplasmic transport of proteins as a target for therapeutic drugs. Curr Med Chem 2003;10:741–8.
    1. Lapalombella R, Sun Q, Williams K, et al. Selective inhibitors of nuclear export show that CRM1/XPO1 is a target in chronic lymphocytic leukemia. Blood 2012;120:4621–34.
    1. Etchin J, Sun Q, Kentsis A, et al. Antileukemic activity of nuclear export inhibitors that spare normal hematopoietic cells. Leukemia 2013;27:66–74.
    1. Ranganathan P, Yu X, Na C, et al. Preclinical activity of a novel CRM1 inhibitor in acute myeloid leukemia. Blood 2012;120:1765–73.
    1. Kalid O, Toledo WD, 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;26:1217–28.
    1. Inoue H, Kauffman M, Shacham S, et al. CRM1 Blockade by Selective Inhibitors of Nuclear Export (SINE) attenuates Kidney Cancer Growth. J Urol 2012.
    1. Zhang K, Wang M, Tamayo AT, et al. Novel selective inhibitors of nuclear export CRM1 antagonists for therapy in mantle cell lymphoma. Exp Hematol 2013;41:67–78.
    1. 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 2012.
    1. Tan DS, Bedard PL, Kuruvilla J, Siu LL, Razak AR. Promising SINEs for Embargoing Nuclear-Cytoplasmic Export as an Anticancer Strategy. Cancer Discov 2014;4:527–37.
    1. 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.
    1. Senapedis WT, Baloglu E, Landesman Y. Clinical translation of nuclear export inhibitors in cancer. Semin Cancer Biol 2014.
    1. Von Hoff DD, Goldstein D, Renschler MF. Albumin-bound paclitaxel plus gemcitabine in pancreatic cancer. N Engl J Med 2014;370:479–80.
    1. 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;14:1570–81.
    1. 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;22:107–16.
    1. Aboukameel A, Muqbil I, Senapedis W, et al. Novel p21-Activated Kinase 4 (PAK4) Allosteric Modulators Overcome Drug Resistance and Stemness in Pancreatic Ductal Adenocarcinoma. Mol Cancer Ther 2017;16:76–87.
    1. Azmi AS, Muqbil I, Wu J, et al. Targeting the Nuclear Export Protein XPO1/CRM1 Reverses Epithelial to Mesenchymal Transition. Sci Rep 2015;5:16077.
    1. 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;5:3444–54.
    1. Mettu NB, Abbruzzese JL. Clinical Insights Into the Biology and Treatment of Pancreatic Cancer. J Oncol Pract 2016;12:17–23.
    1. Rasheed ZA, Matsui W, Maitra A. Pathology of pancreatic stroma in PDAC. 2012.
    1. ngi-Garimella S, Krantz SB, Shields MA, Grippo PJ, Munshi HG. Epithelial-mesenchymal transition and pancreatic cancer progression. 2012.
    1. Sheikh R, Walsh N, Clynes M, O’Connor R, McDermott R. Challenges of drug resistance in the management of pancreatic cancer. Expert Rev Anticancer Ther 2010;10:1647–61.
    1. 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.
    1. Tai YT, Landesman Y, Acharya C, et al. CRM1 inhibition induces tumor cell cytotoxicity and impairs osteoclastogenesis in multiple myeloma: molecular mechanisms and therapeutic implications. Leukemia 2014;28:155–65.
    1. 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;16:417–27.
    1. Etchin J, Montero J, Berezovskaya A, et al. Activity of a selective inhibitor of nuclear export, selinexor (KPT-330), against AML-initiating cells engrafted into immunosuppressed NSG mice. Leukemia 2016;30:190–9.
    1. Hamamoto T, Gunji S, Tsuji H, Beppu T. Leptomycins A and B, new antifungal antibiotics. I. Taxonomy of the producing strain and their fermentation, purification and characterization. J Antibiot (Tokyo) 1983;36:639–45.
    1. Newlands ES, Rustin GJ, Brampton MH. Phase I trial of elactocin. Br J Cancer 1996;74:648–9.
    1. 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.
    1. 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;34:3166–74.
    1. 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;34:4094–101.

Source: PubMed

3
Předplatit