Preclinical characterization of the CDK4/6 inhibitor LY2835219: in-vivo cell cycle-dependent/independent anti-tumor activities alone/in combination with gemcitabine

Lawrence M Gelbert, Shufen Cai, Xi Lin, Concepcion Sanchez-Martinez, Miriam Del Prado, Maria Jose Lallena, Raquel Torres, Rose T Ajamie, Graham N Wishart, Robert Steven Flack, Blake Lee Neubauer, Jamie Young, Edward M Chan, Philip Iversen, Damien Cronier, Emiko Kreklau, Alfonso de Dios, Lawrence M Gelbert, Shufen Cai, Xi Lin, Concepcion Sanchez-Martinez, Miriam Del Prado, Maria Jose Lallena, Raquel Torres, Rose T Ajamie, Graham N Wishart, Robert Steven Flack, Blake Lee Neubauer, Jamie Young, Edward M Chan, Philip Iversen, Damien Cronier, Emiko Kreklau, Alfonso de Dios

Abstract

The G1 restriction point is critical for regulating the cell cycle and is controlled by the Rb pathway (CDK4/6-cyclin D1-Rb-p16/ink4a). This pathway is important because of its inactivation in a majority of human tumors. Transition through the restriction point requires phosphorylation of retinoblastoma protein (Rb) by CDK4/6, which are highly validated cancer drug targets. We present the identification and characterization of a potent CDK4/6 inhibitor, LY2835219. LY2835219 inhibits CDK4 and CDK6 with low nanomolar potency, inhibits Rb phosphorylation resulting in a G1 arrest and inhibition of proliferation, and its activity is specific for Rb-proficient cells. In vivo target inhibition studies show LY2835219 is a potent inhibitor of Rb phosphorylation, induces a complete cell cycle arrest and suppresses expression of several Rb-E2F-regulated proteins 24 hours after a single dose. Oral administration of LY2835219 inhibits tumor growth in human tumor xenografts representing different histologies in tumor-bearing mice. LY2835219 is effective and well tolerated when administered up to 56 days in immunodeficient mice without significant loss of body weight or tumor outgrowth. In calu-6 xenografts, LY2835219 in combination with gemcitabine enhanced in vivo antitumor activity without a G1 cell cycle arrest, but was associated with a reduction of ribonucleotide reductase expression. These results suggest LY2835219 may be used alone or in combination with standard-of-care cytotoxic therapy. In summary, we have identified a potent, orally active small-molecule inhibitor of CDK4/6 that is active in xenograft tumors. LY2835219 is currently in clinical development.

Trial registration: ClinicalTrials.gov NCT01394016 NCT01739309 NCT02014129 NCT02057133 NCT02079636 NCT02117648.

Figures

Fig. 1
Fig. 1
a Structure of LY2835219. b KINOMEscan dendogram for biochemical kinase selectivity profile against 456 kinases. single point binding at 200 nM (left) and 2 μM (right) LY2835219
Fig. 2
Fig. 2
a Activity of LY2835219 in colo-205 and MCF10A cells using a multiplexed assay measuring p-Rb serine 780 (green), cells in G1 (2 N DNA content, red) and total cell number (black). after 24-hour exposure LY2835219 shows a dose-dependent inhibition of p-Rb (ser780) and a corresponding sustained G1 arrest. b MV4-11 acute myelocytic leukemia (AML) cells treated with increasing concentrations of LY2835219 for 24 h and cell cycle activity profiled by flow cytometry. c cell cycle activity of LY2835219 in cells with and without functional Rb. LY2835219 gives a G1 cell cycle arrest in Rb-proficient MDA-MB-231 breast cancer cells, but not in Rb-deficient MDA-MB-468 breast cancer cells
Fig. 3
Fig. 3
In vivo target inhibition by LY2835219. a tumors harvested 24 h after dosing for analysis with the markers topoIIα (S phase-specific), p-Rb (pRb ser 780, G1-specific), pHH3 (M-specific), and the Rb/E2F regulated proteins DHFR, RRM1 and RRM2. GAPDH was used as a loading control. statistical significance (p <0.05) is indicated above each dose by an asterisk for pRb, Topollα, and pHH3, respectively. b time course of LY2835219. tumor-bearing animals were dosed orally with 50 mg/kg of LY2835219 and tumors harvested at the indicated times (n = 5 per treatment, n = 8 for vehicle). the percent change in band intensity was calculated using vehicle-treated bands as 100 %. mean (±SD) plasma concentrations of the compound are shown in the lower right panel
Fig. 4
Fig. 4
In vivo antitumor activity of LY2835219 in subcutaneous human tumor xenografts. Tumors were implanted in the rear flank of athymic mice and randomized for treatment when the mean tumor volume reached 150 to 200 mm3. LY2835219 and PD0332991 were administered at the indicated dose and schedule. treatment period is indicated by the horizontal black bar along the X-axis, body weight shown for each experiment is shown in the upper left corner. a effect of 25, 50, or 100 mg/kg of LY2835219 and 100 mg/kg PD0332991 on colo-205 xenografts. b inhibition of cell cycle markers in colo-205 xenografts at the end of treatment with 100 mg/kg LY2835219. c effect of 25, 50, or 100 mg/kg LY2835219 and 50 mg/kg PD0332991 in MV4-11 xenografts. d LY2835219 inhibits tumor growth and is well tolerated in mice bearing colo-205 xenografts when dosed 56 days with 50 mg/kg continuously or intermittently
Fig. 5
Fig. 5
In vivo antitumor activity of LY2835219 and gemcitabine. a Calu-6 lung xenografts were treated with LY2835219 orally, with gemcitabine by intraperitoneal injection, LY2835219 followed by gemcitabine, or both compounds together. treatment period is indicated by horizontal black bar along the X-axis and body weight is shown in the upper left corner. b results of target inhibition studies

References

    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013.
    1. Pardee AB. A restriction point for control of normal animal cell proliferation. Proc Natl Acad Sci U S A. 1974;71:1286–1290. doi: 10.1073/pnas.71.4.1286.
    1. Blagosklonny MV, Pardee AB. The restriction point of the cell cycle. Cell Cycle. 2002;1:103–110.
    1. Ortega S, Malumbres M, Barbacid M. Cyclin D-dependent kinases, INK4 inhibitors and cancer. Biochim Biophys Acta. 2002;1602:73–87.
    1. Dyson N. The regulation of E2F by pRB-family proteins. Genes Dev. 1998;12:2245–2262. doi: 10.1101/gad.12.15.2245.
    1. Gladden AB, Diehl JA. Location, location, location: the role of cyclin D1 nuclear localization in cancer. J Cell Biochem. 2005;96:906–913. doi: 10.1002/jcb.20613.
    1. Harbour JW, Luo RX, Santi AD, Postigo AA, Dean DC. Cdk Phosphorylation triggers sequential intramolecular interactions that progressively block Rb functions as cells move through G1. Cell. 1999;98:859–869. doi: 10.1016/S0092-8674(00)81519-6.
    1. Lundberg AS, Weinberg RA. Functional inactivation of the retinoblastoma protein requires sequential modification by at least two distinct cyclin-cdk complexes. Mol Cell Biol. 1998;18:753–761. doi: 10.1128/MCB.18.2.753.
    1. Ishida S, Huang E, Zuzan H, Spang R, Leone G, West M, et al. Role for E2F in control of both DNA replication and mitotic functions as revealed from DNA microarray analysis. Mol Cell Biol. 2001;21:4684–4699. doi: 10.1128/MCB.21.14.4684-4699.2001.
    1. Anders L, Ke N, Hydbring P, Choi YJ, Widlund HR, Chick JM, et al. A systematic screen for CDK4/6 substrates links FOXM1 phosphorylation to senescence suppression in cancer cells. Cancer Cell. 2011;20:620–634. doi: 10.1016/j.ccr.2011.10.001.
    1. Fagan R, Flint KJ, Jones N. Phosphorylation of E2F-1 modulates its interaction with the retinoblastoma gene product and the adenoviral E4 19 kDa protein. Cell. 1994;78:799–811. doi: 10.1016/S0092-8674(94)90522-3.
    1. Donnellan R, Chetty R. Cyclin D1 and human neoplasia. Mol Pathol. 1998;51:1–7. doi: 10.1136/mp.51.1.1.
    1. Fernandez V, Hartmann E, Ott G, Campo E, Rosenwald A. Pathogenesis of mantle-cell lymphoma: all oncogenic roads lead to dysregulation of cell cycle and DNA damage response pathways. J Clin Oncol. 2005;23:6364–6369. doi: 10.1200/JCO.2005.05.019.
    1. Wang L, Wang J, Blaser BW, Duchemin A-M, Kusewitt DF, Liu T, et al. Pharmacologic inhibition of CDK4/6: mechanistic evidence for selective activity or acquired resistance in acute myeloid leukemia. Blood. 2007;110:2075–2083. doi: 10.1182/blood-2007-02-071266.
    1. Barretina J, Taylor BS, Banerji S, Ramos AH, Lagos-Quintana M, Decarolis PL, et al. Subtype-specific genomic alterations define new targets for soft-tissue sarcoma therapy. Nat Genet. 2010;42:715–721. doi: 10.1038/ng.619.
    1. Reifenberger G, Reifenberger J, Ichimura K, Peter CV. Amplification at 12q13-14 in human malignant gliomas is frequently accompanied by loss of heterozygosity at loci proximal and distal to the amplification site. Cancer Res. 1995;55:731–734.
    1. Yu J, Deshmukh H, Payton JE, Dunham C, Scheithauer BW, Tihan T, et al. Array-based comparative genomic hybridization identifies CDK4 and FOXM1 alterations as independent predictors of survival in malignant peripheral nerve sheath tumor. Clin Cancer Res. 2011;17:1924–1934. doi: 10.1158/1078-0432.CCR-10-1551.
    1. Chin L. The genetics of malignant melanoma: lessons from mouse and man. Nat Rev Cancer. 2003;3:559–570. doi: 10.1038/nrc1145.
    1. Horowitz JM, Park SH, Bogenmann E, Cheng JC, Yandell DW, Kaye FJ, et al. Frequent inactivation of the retinoblastoma anti-oncogene is restricted to a subset of human tumor cells. Proc Natl Acad Sci U S A. 1990;87:2775–2779. doi: 10.1073/pnas.87.7.2775.
    1. Ruas M, Peters G. The p16INK4a/CDKN2A tumor suppressor and its relatives. Biochim Biophys Acta. 1998;1378:F115–F177.
    1. Hughes A. CDK inhibitors in 3D: Problems with the drugs, their development plans or their linkage to disease? Gene Ther Mol Biol. 2006;10:41–54.
    1. Fry DW, Harvey PJ, Keller PR, Elliott WL, Meade M, Trachet E, et al. Specific inhibition of cyclin-dependent kinase 4/6 by PD 0332991 and associated antitumor activity in human tumor xenografts. Mol Cancer Ther. 2004;3:1427–1438.
    1. Oelgeschläger T. Regulation of RNA polymerase II activity by CTD phosphorylation and cell cycle control. J Cell Physiol. 2002;190:160–169. doi: 10.1002/jcp.10058.
    1. Kitagawa M, Higashi H, Jung HK, Suzuki-Takahashi I, Ikeda M, Tamai K, et al. The consensus motif for phosphorylation by cyclin D1-Cdk4 is different from that for phosphorylation by cyclin A/E-Cdk2. EMBO J. 1996;15:7060–7069.
    1. Goswami PC, Roti Roti JL, Hunt CR. The cell cycle-coupled expression of topoisomerase IIalpha during S phase is regulated by mRNA stability and is disrupted by heat shock or ionizing radiation. Mol Cell Biol. 1996;16:1500–1508. doi: 10.1128/MCB.16.4.1500.
    1. Goto H, Tomono Y, Ajiro K, Kosako H, Fujita M, Sakurai M, et al. Identification of a novel phosphorylation site on histone H3 coupled with mitotic chromosome condensation. J Biol Chem. 1999;274:25543–25549. doi: 10.1074/jbc.274.36.25543.
    1. Musgrove EA, Lilischkis R, Cornish AL, Lee CS, Setlur V, Seshadri R, et al. Expression of the cyclin-dependent kinase inhibitors p16INK4, p15INK4B and p21WAF1/CIP1 in human breast cancer. Int J Cancer. 1995;63:584–591. doi: 10.1002/ijc.2910630420.
    1. Angus SP, Wheeler LJ, Ranmal SA, Zhang X, Markey MP, Mathews CK, et al. Retinoblastoma tumor suppressor targets dNTP metabolism to regulate DNA replication. J Biol Chem. 2002;277:44376–44384. doi: 10.1074/jbc.M205911200.
    1. Knight ZA, Lin H, Shokat KM. Targeting the cancer kinome through polypharmacology. Nat Rev Cancer. 2010;10:130–137. doi: 10.1038/nrc2787.
    1. Deep G, Agarwal R. New combination therapies with cell-cycle agents. Curr Opin Investig Drugs. 2008;9:591–604.
    1. Merriman RL, Hertel LW, Schultz RM, Houghton PJ, Houghton JA, Rutherford PG, et al. Comparison of the antitumor activity of gemcitabine and ara-C in a panel of human breast, colon, lung and pancreatic xenograft models. Investig New Drugs. 1996;14:243–247. doi: 10.1007/BF00194526.
    1. Malumbres M, Barbacid M. To cycle or not to cycle: a critical decision in cancer. Nat Rev Cancer. 2001;1:222–231. doi: 10.1038/35106065.
    1. Malumbres M, Barbacid M. Is Cyclin D1-CDK4 kinase a bona fide cancer target? Cancer Cell. 2006;9:2–4. doi: 10.1016/j.ccr.2005.12.026.
    1. Shapiro GI. Cyclin-dependent kinase pathways as targets for cancer treatment. J Clin Oncol. 2006;24:1770–1783. doi: 10.1200/JCO.2005.03.7689.
    1. Guha M. Cyclin-dependent kinase inhibitors move into Phase III. Nat Rev Drug Discov. 2012;11:892–894. doi: 10.1038/nrd3908.
    1. Lin X, Gelbert L, Chu S, Medina D, Garcia-Paredes C, De Dios A (2007) High throughput multiplexing phospho retinoblastoma tumor suppressor protein (Rb) and cell cycle analysis using acumen technology. In: Proceedings of the AACR Annual Meeting; Apr 15; Los Angeles, CA. Abstract nr 4372
    1. Tate SC, Cai S, Ajamie RT, Burke T, Beckmann RP, Chan EM, et al (2014)Semi-mechanistic Pharmacokinetic/Pharmacodynamic Modeling of the Anti-Tumor Activity of LY2835219, a New Cyclin-Dependent Kinase 4/6 Inhibitor, in Mice Bearing Human Tumor Xenografts. Clin Can Res doi:10.1158/1078-0432.CCR-13-2846
    1. Shapiro G, Rosen LS, Tolcher, AW, Goldman JW, Gandhi L, Papadopoulos KP, et al A first-in-human phase I study o fthe CDK4/6 inhibitor, LY2835219, for patients with advanced cancer. J Clin Oncol 31, 2013 (Suppl; abstr 2500).
    1. Patnaik A, Rosen LS, Tolaney SM, Tolcher AW, Goldman JW, Gandhi L, et al Clinical activity of LY2835219, a novel cell cycle inhibitor selective for CDK4 and CDK6, in patients with metastatic breast cancer [abstract]. AACR 2014 Apr 7; Abstr CT-232.
    1. Kim KT, Baird K, Ahn JY, Meltzer P, Lilly M, Levis M, et al. Pim-1 is up-regulated by constitutively activated FLT3 and plays a role in FLT3-mediated cell survival. Blood. 2005;105:1759–1767. doi: 10.1182/blood-2004-05-2006.
    1. Kim KT, Levis M, Small D. Constitutively activated FLT3 phosphorylates BAD partially through pim-1. Br J Haematol. 2006;134:500–509. doi: 10.1111/j.1365-2141.2006.06225.x.
    1. de Vos S, Krug U, Hofmann WK, Pinkus GS, Swerdlow SH, Wachsman W, et al. Cell cycle alterations in the blastoid variant of mantle cell lymphoma (MCL-BV) as detected by gene expression profiling of mantle cell lymphoma (MCL) and MCL-BV. Diagn Mol Pathol. 2003;12:35–43. doi: 10.1097/00019606-200303000-00005.
    1. Zhu Y, Hollmen J, Raty R, Aalto Y, Nagy B, Elonen E, et al. Investigatory and analytical approaches to differential gene expression profiling in mantle cell lymphoma. Br J Haematol. 2002;119:905–915. doi: 10.1046/j.1365-2141.2002.03931.x.
    1. Sandler AB, Nemunaitis J, Denham C, von Pawel J, Cormier Y, Gatzemeier U, et al. Phase III trial of gemcitabine plus cisplatin versus cisplatin alone in patients with locally advanced or metastatic non–small-cell lung cancer. J Clin Oncol. 2000;18:122–130.
    1. Bepler G, Kusmartseva I, Sharma S, Gautam A, Cantor A, Sharma A, et al. RRM1 modulated in vitro and in vivo efficacy of gemcitabine and platinum in non–small-cell lung cancer. J Clin Oncol. 2006;24:4731–4737. doi: 10.1200/JCO.2006.06.1101.
    1. Davidson JD, Ma L, Flagella M, Geeganage S, Gelbert LM, Slapak CA. An increase in the expression of ribonucleotide reductase large subunit 1 is associated with gemcitabine resistance in non-small cell lung cancer cell lines. Cancer Res. 2004;64:3761–3766. doi: 10.1158/0008-5472.CAN-03-3363.
    1. Barton KL, Misuraca K, Cordero F, Dobrikova E, Min HD, Gromeier M, et al. PD-0332991, a CDK4/6 inhibitor, significantly prolongs survival in a genetically engineered mouse model of brainstem glioma. PLoS One. 2013;8:e77639. doi: 10.1371/journal.pone.0077639.
    1. Puyol M, Martin A, Dubus P, Mulero F, Pizcueta P, Khan G, et al. A synthetic lethal interaction between K-Ras oncogenes and Cdk4 unveils a therapeutic strategy for non-small cell lung carcinoma. Cancer Cell. 2010;18:63–73. doi: 10.1016/j.ccr.2010.05.025.
    1. Dempsey JA, Chan EM, Burke TF, Beckmann RP (2013) LY2835219, a selective inhibitor of CDK4 and CDK6, inhibits growth in preclinical models of human cancer. Cancer Res ; 73(8Suppl): LB-122.
    1. The Cancer Genome Atlas Research Network Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455:1061–1068. doi: 10.1038/nature07385.
    1. Steeg PS, Camphausen KA, Smith QR. Brain metastases as preventive and therapeutic targets. Nat Rev Cancer. 2011;11:352–363. doi: 10.1038/nrc3053.
    1. Patchell RA. The management of brain metastases. Cancer Treat Rev. 2003;29:533. doi: 10.1016/S0305-7372(03)00105-1.
    1. Pelletier EM, Shim B, Goodman S, Amonkar MM. Epidemiology and economic burden of brain metastases among patients with primary breast cancer: results from a US claims data analysis. Breast Cancer Res Treat. 2008;108:297–305. doi: 10.1007/s10549-007-9601-0.

Source: PubMed

3
Abonner