Skip to main content
Top
Published in: Cancer Chemotherapy and Pharmacology 6/2018

Open Access 01-12-2018 | Original Article

Pharmacokinetic evaluation of the PNC disassembler metarrestin in wild-type and Pdx1-Cre;LSL-KrasG12D/+;Tp53R172H/+ (KPC) mice, a genetically engineered model of pancreatic cancer

Authors: Tomas Vilimas, Amy Q. Wang, Samarjit Patnaik, Emma A. Hughes, Marc D. Singleton, Zachary Knotts, Dandan Li, Kevin Frankowski, Jerome J. Schlomer, Theresa M. Guerin, Stephanie Springer, Catherine Drennan, Christopher Dextras, Chen Wang, Debra Gilbert, Noel Southall, Marc Ferrer, Sui Huang, Serguei Kozlov, Juan Marugan, Xin Xu, Udo Rudloff

Published in: Cancer Chemotherapy and Pharmacology | Issue 6/2018

Login to get access

Abstract

Purpose

Metarrestin is a first-in-class small molecule clinical candidate capable of disrupting the perinucleolar compartment, a subnuclear structure unique to metastatic cancer cells. This study aims to define the pharmacokinetic (PK) profile of metarrestin and the pharmacokinetic/pharmacodynamic relationship of metarrestin-regulated markers.

Methods

PK studies included the administration of single or multiple dose of metarrestin at 3, 10, or 25 mg/kg via intravenous (IV) injection, gavage (PO) or with chow to wild-type C57BL/6 mice and KPC mice bearing autochthonous pancreatic tumors. Metarrestin concentrations were analyzed by UPLC–MS/MS. Pharmacodynamic assays included mRNA expression profiling by RNA-seq and qRT-PCR for KPC mice.

Results

Metarrestin had a moderate plasma clearance of 48 mL/min/kg and a large volume of distribution of 17 L/kg at 3 mg/kg IV in C57BL/6 mice. The oral bioavailability after single-dose (SD) treatment was > 80%. In KPC mice treated with SD 25 mg/kg PO, plasma AUC0–∞ of 14400 ng h/mL, Cmax of 810 ng/mL and half-life (t1/2) of 8.5 h were observed. At 24 h after SD of 25 mg/kg PO, the intratumor concentration of metarrestin was high with a mean value of 6.2 µg/g tissue (or 13 µM), well above the cell-based IC50 of 0.4 µM. At multiple dose (MD) 25 mg/kg/day PO in KPC mice, mean tissue/plasma AUC0–24h ratio for tumor, spleen and liver was 37, 30 and 31, respectively. There was a good linear relationship of dosage to AUC0–24h and C24h. AUC0–24h MD to AUC0–24h SD ratios ranged from two for liver to five for tumor indicating additional accumulation in tumors. Dose-dependent normalization of FOXA1 and FOXO6 mRNA expression was observed in KPC tumors.

Conclusions

Metarrestin is an effective therapeutic candidate with a favorable PK profile achieving excellent intratumor tissue levels in a disease with known poor drug delivery.
Appendix
Available only for authorised users
Literature
1.
go back to reference Siegel RL, Miller KD, Jemal A (2016) Cancer statistics. CA Cancer J Clin 66(1):7–30CrossRef Siegel RL, Miller KD, Jemal A (2016) Cancer statistics. CA Cancer J Clin 66(1):7–30CrossRef
2.
go back to reference Tevaarwerk AJ et al (2013) Survival in patients with metastatic recurrent breast cancer after adjuvant chemotherapy: little evidence of improvement over the past 30 years. Cancer 119(6):1140–1148CrossRef Tevaarwerk AJ et al (2013) Survival in patients with metastatic recurrent breast cancer after adjuvant chemotherapy: little evidence of improvement over the past 30 years. Cancer 119(6):1140–1148CrossRef
3.
go back to reference Bernards N et al (2013) No improvement in median survival for patients with metastatic gastric cancer despite increased use of chemotherapy. Ann Oncol 24(12):3056–3060CrossRef Bernards N et al (2013) No improvement in median survival for patients with metastatic gastric cancer despite increased use of chemotherapy. Ann Oncol 24(12):3056–3060CrossRef
4.
5.
go back to reference Pollock C, Huang S (2009) The perinucleolar compartment. J Cell Biochem 107(2):189–193CrossRef Pollock C, Huang S (2009) The perinucleolar compartment. J Cell Biochem 107(2):189–193CrossRef
6.
go back to reference Norton JT et al (2009) The perinucleolar compartment is directly associated with DNA. J Biol Chem 284(7):4090–4101CrossRef Norton JT et al (2009) The perinucleolar compartment is directly associated with DNA. J Biol Chem 284(7):4090–4101CrossRef
7.
go back to reference Slusarczyk A et al (2010) Structure and function of the perinucleolar compartment in cancer cells. Cold Spring Harb Symp Quant Biol 75:599–605CrossRef Slusarczyk A et al (2010) Structure and function of the perinucleolar compartment in cancer cells. Cold Spring Harb Symp Quant Biol 75:599–605CrossRef
8.
go back to reference Wang C et al (2003) RNA polymerase III transcripts and the PTB protein are essential for the integrity of the perinucleolar compartment. Mol Biol Cell 14(6):2425–2435CrossRef Wang C et al (2003) RNA polymerase III transcripts and the PTB protein are essential for the integrity of the perinucleolar compartment. Mol Biol Cell 14(6):2425–2435CrossRef
9.
go back to reference Pollock C et al (2011) Characterization of MRP RNA-protein interactions within the perinucleolar compartment. Mol Biol Cell 22(6):858–867CrossRef Pollock C et al (2011) Characterization of MRP RNA-protein interactions within the perinucleolar compartment. Mol Biol Cell 22(6):858–867CrossRef
10.
go back to reference Norton JT, Huang S (2013) The perinucleolar compartment: RNA metabolism and cancer. Cancer Treat Res 158:139–152CrossRef Norton JT, Huang S (2013) The perinucleolar compartment: RNA metabolism and cancer. Cancer Treat Res 158:139–152CrossRef
11.
go back to reference Kamath RV et al (2005) Perinucleolar compartment prevalence has an independent prognostic value for breast cancer. Cancer Res 65(1):246–253PubMed Kamath RV et al (2005) Perinucleolar compartment prevalence has an independent prognostic value for breast cancer. Cancer Res 65(1):246–253PubMed
12.
go back to reference Norton JT et al (2008) Perinucleolar compartment prevalence is a phenotypic pancancer marker of malignancy. Cancer 113(4):861–869CrossRef Norton JT et al (2008) Perinucleolar compartment prevalence is a phenotypic pancancer marker of malignancy. Cancer 113(4):861–869CrossRef
13.
go back to reference Norton JT et al (2009) Automated high-content screening for compounds that disassemble the perinucleolar compartment. J Biomol Screen 14(9):1045–1053CrossRef Norton JT et al (2009) Automated high-content screening for compounds that disassemble the perinucleolar compartment. J Biomol Screen 14(9):1045–1053CrossRef
14.
go back to reference Frankowski K et al (2010) Discovery and development of small molecules that reduce PNC prevalence. In: Probe reports from the NIH molecular libraries program, Bethesda (MD) Frankowski K et al (2010) Discovery and development of small molecules that reduce PNC prevalence. In: Probe reports from the NIH molecular libraries program, Bethesda (MD)
15.
go back to reference Frankowski KJ et al (2018) Metarrestin, a potent perinucleolar compartment inhibitor, effectively suppresses metastasis. Sci Transl Med 10(441):eaap8307CrossRef Frankowski KJ et al (2018) Metarrestin, a potent perinucleolar compartment inhibitor, effectively suppresses metastasis. Sci Transl Med 10(441):eaap8307CrossRef
16.
go back to reference Tomlinson VA et al (2005) Translation elongation factor eEF1A2 is a potential oncoprotein that is overexpressed in two-thirds of breast tumours. BMC Cancer 5:113CrossRef Tomlinson VA et al (2005) Translation elongation factor eEF1A2 is a potential oncoprotein that is overexpressed in two-thirds of breast tumours. BMC Cancer 5:113CrossRef
17.
go back to reference Tomlinson VA et al (2007) Expression of eEF1A2 is associated with clear cell histology in ovarian carcinomas: overexpression of the gene is not dependent on modifications at the EEF1A2 locus. Br J Cancer 96(10):1613–1620CrossRef Tomlinson VA et al (2007) Expression of eEF1A2 is associated with clear cell histology in ovarian carcinomas: overexpression of the gene is not dependent on modifications at the EEF1A2 locus. Br J Cancer 96(10):1613–1620CrossRef
18.
go back to reference Scaggiante B et al (2012) Dissecting the expression of EEF1A1/2 genes in human prostate cancer cells: the potential of EEF1A2 as a hallmark for prostate transformation and progression. Br J Cancer 106(1):166–173CrossRef Scaggiante B et al (2012) Dissecting the expression of EEF1A1/2 genes in human prostate cancer cells: the potential of EEF1A2 as a hallmark for prostate transformation and progression. Br J Cancer 106(1):166–173CrossRef
19.
go back to reference Kawamura M et al (2014) The prognostic significance of eukaryotic elongation factor 1 alpha-2 in non-small cell lung cancer. Anticancer Res 34(2):651–658PubMed Kawamura M et al (2014) The prognostic significance of eukaryotic elongation factor 1 alpha-2 in non-small cell lung cancer. Anticancer Res 34(2):651–658PubMed
20.
go back to reference Zang W et al (2015) miR-663 attenuates tumor growth and invasiveness by targeting eEF1A2 in pancreatic cancer. Mol Cancer 14:37CrossRef Zang W et al (2015) miR-663 attenuates tumor growth and invasiveness by targeting eEF1A2 in pancreatic cancer. Mol Cancer 14:37CrossRef
21.
go back to reference Gross SR, Kinzy TG (2005) Translation elongation factor 1A is essential for regulation of the actin cytoskeleton and cell morphology. Nat Struct Mol Biol 12(9):772–778CrossRef Gross SR, Kinzy TG (2005) Translation elongation factor 1A is essential for regulation of the actin cytoskeleton and cell morphology. Nat Struct Mol Biol 12(9):772–778CrossRef
22.
go back to reference Shamovsky I et al (2006) RNA-mediated response to heat shock in mammalian cells. Nature 440(7083):556–560CrossRef Shamovsky I et al (2006) RNA-mediated response to heat shock in mammalian cells. Nature 440(7083):556–560CrossRef
23.
go back to reference Mateyak MK, Kinzy TG (2010) eEF1A: thinking outside the ribosome. J Biol Chem 285(28):21209–21213CrossRef Mateyak MK, Kinzy TG (2010) eEF1A: thinking outside the ribosome. J Biol Chem 285(28):21209–21213CrossRef
24.
go back to reference Huang HY, Hopper AK (2015) In vivo biochemical analyses reveal distinct roles of beta-importins and eEF1A in tRNA subcellular traffic. Genes Dev 29(7):772–783CrossRef Huang HY, Hopper AK (2015) In vivo biochemical analyses reveal distinct roles of beta-importins and eEF1A in tRNA subcellular traffic. Genes Dev 29(7):772–783CrossRef
25.
go back to reference Chuang SM et al (2005) Proteasome-mediated degradation of cotranslationally damaged proteins involves translation elongation factor 1A. Mol Cell Biol 25(1):403–413CrossRef Chuang SM et al (2005) Proteasome-mediated degradation of cotranslationally damaged proteins involves translation elongation factor 1A. Mol Cell Biol 25(1):403–413CrossRef
26.
go back to reference Xu C, Hu DM, Zhu Q (2013) eEF1A2 promotes cell migration, invasion and metastasis in pancreatic cancer by upregulating MMP-9 expression through Akt activation. Clin Exp Metastasis 30(7):933–944CrossRef Xu C, Hu DM, Zhu Q (2013) eEF1A2 promotes cell migration, invasion and metastasis in pancreatic cancer by upregulating MMP-9 expression through Akt activation. Clin Exp Metastasis 30(7):933–944CrossRef
27.
go back to reference Hingorani SR et al (2005) Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 7(5):469–483CrossRef Hingorani SR et al (2005) Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 7(5):469–483CrossRef
28.
go back to reference Olive KP et al (2009) Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 324(5933):1457–1461CrossRef Olive KP et al (2009) Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 324(5933):1457–1461CrossRef
29.
go back to reference Feig C et al (2012) The pancreas cancer microenvironment. Clin Cancer Res 18(16):4266–4276CrossRef Feig C et al (2012) The pancreas cancer microenvironment. Clin Cancer Res 18(16):4266–4276CrossRef
30.
go back to reference Provenzano PP et al (2012) Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 21(3):418–429CrossRef Provenzano PP et al (2012) Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 21(3):418–429CrossRef
31.
go back to reference Takeuchi H, Yamamoto H, Kawashima Y (2001) Mucoadhesive nanoparticulate systems for peptide drug delivery. Adv Drug Deliv Rev 47(1):39–54CrossRef Takeuchi H, Yamamoto H, Kawashima Y (2001) Mucoadhesive nanoparticulate systems for peptide drug delivery. Adv Drug Deliv Rev 47(1):39–54CrossRef
32.
go back to reference Rasenack N, Muller BW (2002) Dissolution rate enhancement by in situ micronization of poorly water-soluble drugs. Pharm Res 19(12):1894–1900CrossRef Rasenack N, Muller BW (2002) Dissolution rate enhancement by in situ micronization of poorly water-soluble drugs. Pharm Res 19(12):1894–1900CrossRef
33.
go back to reference Kerc J et al (1999) Micronization of drugs using supercritical carbon dioxide. Int J Pharm 182(1):33–39CrossRef Kerc J et al (1999) Micronization of drugs using supercritical carbon dioxide. Int J Pharm 182(1):33–39CrossRef
34.
go back to reference Mutalik S et al (2008) Enhancement of dissolution rate and bioavailability of aceclofenac: a chitosan-based solvent change approach. Int J Pharm 350(1–2):279–290CrossRef Mutalik S et al (2008) Enhancement of dissolution rate and bioavailability of aceclofenac: a chitosan-based solvent change approach. Int J Pharm 350(1–2):279–290CrossRef
35.
go back to reference Roe JS et al (2017) Enhancer reprogramming promotes pancreatic cancer metastasis. Cell 170(5):875–888 (e20) CrossRef Roe JS et al (2017) Enhancer reprogramming promotes pancreatic cancer metastasis. Cell 170(5):875–888 (e20) CrossRef
36.
go back to reference Hu HJ et al (2015) FoxO6 inhibits cell proliferation in lung carcinoma through up-regulation of USP7. Mol Med Rep 12(1):575–580CrossRef Hu HJ et al (2015) FoxO6 inhibits cell proliferation in lung carcinoma through up-regulation of USP7. Mol Med Rep 12(1):575–580CrossRef
37.
go back to reference Wolf J et al (2014) An in vivo RNAi screen identifies SALL1 as a tumor suppressor in human breast cancer with a role in CDH1 regulation. Oncogene 33(33):4273–4278CrossRef Wolf J et al (2014) An in vivo RNAi screen identifies SALL1 as a tumor suppressor in human breast cancer with a role in CDH1 regulation. Oncogene 33(33):4273–4278CrossRef
38.
go back to reference Abbas W, Kumar A, Herbein G (2015) The eEF1A proteins: at the crossroads of oncogenesis, apoptosis, and viral infections. Front Oncol 5:75CrossRef Abbas W, Kumar A, Herbein G (2015) The eEF1A proteins: at the crossroads of oncogenesis, apoptosis, and viral infections. Front Oncol 5:75CrossRef
39.
go back to reference LaBarge MA (2010) The difficulty of targeting cancer stem cell niches. Clin Cancer Res 16(12):3121–3129CrossRef LaBarge MA (2010) The difficulty of targeting cancer stem cell niches. Clin Cancer Res 16(12):3121–3129CrossRef
40.
go back to reference Neesse A et al (2014) SPARC independent drug delivery and antitumour effects of nab-paclitaxel in genetically engineered mice. Gut 63(6):974–983CrossRef Neesse A et al (2014) SPARC independent drug delivery and antitumour effects of nab-paclitaxel in genetically engineered mice. Gut 63(6):974–983CrossRef
41.
go back to reference Von Hoff DD et al (2013) Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 369(18):1691–1703CrossRef Von Hoff DD et al (2013) Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 369(18):1691–1703CrossRef
Metadata
Title
Pharmacokinetic evaluation of the PNC disassembler metarrestin in wild-type and Pdx1-Cre;LSL-KrasG12D/+;Tp53R172H/+ (KPC) mice, a genetically engineered model of pancreatic cancer
Authors
Tomas Vilimas
Amy Q. Wang
Samarjit Patnaik
Emma A. Hughes
Marc D. Singleton
Zachary Knotts
Dandan Li
Kevin Frankowski
Jerome J. Schlomer
Theresa M. Guerin
Stephanie Springer
Catherine Drennan
Christopher Dextras
Chen Wang
Debra Gilbert
Noel Southall
Marc Ferrer
Sui Huang
Serguei Kozlov
Juan Marugan
Xin Xu
Udo Rudloff
Publication date
01-12-2018
Publisher
Springer Berlin Heidelberg
Published in
Cancer Chemotherapy and Pharmacology / Issue 6/2018
Print ISSN: 0344-5704
Electronic ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-018-3699-0

Other articles of this Issue 6/2018

Cancer Chemotherapy and Pharmacology 6/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine