Targeting cell cycle and hormone receptor pathways in cancer

C E S Comstock, M A Augello, J F Goodwin, R de Leeuw, M J Schiewer, W F Ostrander Jr, R A Burkhart, A K McClendon, P A McCue, E J Trabulsi, C D Lallas, L G Gomella, M M Centenera, J R Brody, L M Butler, W D Tilley, K E Knudsen, C E S Comstock, M A Augello, J F Goodwin, R de Leeuw, M J Schiewer, W F Ostrander Jr, R A Burkhart, A K McClendon, P A McCue, E J Trabulsi, C D Lallas, L G Gomella, M M Centenera, J R Brody, L M Butler, W D Tilley, K E Knudsen

Abstract

The cyclin/cyclin-dependent kinase (CDK)/retinoblastoma (RB)-axis is a critical modulator of cell cycle entry and is aberrant in many human cancers. New nodes of therapeutic intervention are needed that can delay or combat the onset of malignancies. The antitumor properties and mechanistic functions of PD-0332991 (PD; a potent and selective CDK4/6 inhibitor) were investigated using human prostate cancer (PCa) models and primary tumors. PD significantly impaired the capacity of PCa cells to proliferate by promoting a robust G1-arrest. Accordingly, key regulators of the G1-S cell cycle transition were modulated including G1 cyclins D, E and A. Subsequent investigation demonstrated the ability of PD to function in the presence of existing hormone-based regimens and to cooperate with ionizing radiation to further suppress cellular growth. Importantly, it was determined that PD is a critical mediator of PD action. The anti-proliferative impact of CDK4/6 inhibition was revealed through reduced proliferation and delayed growth using PCa cell xenografts. Finally, first-in-field effects of PD on proliferation were observed in primary human prostatectomy tumor tissue explants. This study shows that selective CDK4/6 inhibition, using PD either as a single-agent or in combination, hinders key proliferative pathways necessary for disease progression and that RB status is a critical prognostic determinant for therapeutic efficacy. Combined, these pre-clinical findings identify selective targeting of CDK4/6 as a bona fide therapeutic target in both early stage and advanced PCa and underscore the benefit of personalized medicine to enhance treatment response.

Figures

Figure 1
Figure 1
CDK4/6-specific inhibition suppresses proliferation of androgen-dependent PCa cells. The impact of the CDK4/6-specific inhibitor (PD) on proliferation and cell cycle components was characterized in multiple androgen-dependent PCa cell model systems. (a) Bivariate flow cytometry analyses of: LNCaP (upper), LAPC4 (middle) and VCaP (lower) cells treated 24 h with 0.1% dimethylsulphoxide control (left column) or 0.5 μM PD (right column). Profiles are representative of three independent experiments. The x axis denotes relative DNA content as indicated by propidium iodide (PI) staining. The y axis denotes cells undergoing active S-phase as indicated by 2-h pulse-label of BrdU. Inset values: % BrdU incorporation (mean±s.d., from an experiment performed in biological triplicate). (b) Crystal violet staining at day 7 (d7) relative to plating at day 0 (d0) from LNCaP, LAPC4, and VCaP cells initially treated with control or 0.5 μM PD for 24 h. Data are representative of three independent experiments. (c) Immunoblot analyses, from parallel treated cells in panel (a) for the indicated prostate and cell cycle components (left panels) and quantified by LI-COR image analyses (right panels). Loading and quantification are relative to Ran. Grey and black bars=control and PD treatments, respectively. * ** *** indicates P-values: <0.05; 0.01; 0.001, respectively.
Figure 2
Figure 2
AR-directed therapies are effective in the presence of CDK4/6-specific inhibition. To assess AR activity, androgen-dependent PCa cells: (a) LNCaP (b) LAPC4 and (c) VCaP were cultured 72 h in media containing steroid-deprived serum (5% charcoal-dextran treated (CDT)) then stimulated 24 h with (or without) DHT (1 nM) in the presence of PD (0.5 μM), Csdx (10 μM) or combination of PD and Csdx. Relative mRNA expression normalized to glyceraldehyde 3-phosphate dehydrogenase was determined by quantitative PCR (qPCR) for the known AR-target genes: KLK3/PSA (left), TMPRSS2 (middle) and KLK2 (right). Indicated treatments for each gene are relative to non-DHT and non-drug treated cells. * ** *** indicates P-values: <0.05; 0.01; 0.001, respectively.
Figure 3
Figure 3
CDK4/6-specific inhibition cooperates with IR to attenuate PCa cell growth. IR was administered in conjunction with PD to evaluate the effect of combinatorial therapy, as described in the Material and methods section. (a) Cell growth analyses of LNCaP, LAPC4 and VCaP cells treated with single-dose IR (2 Gy), PD (0.5 μM) or a combination of both. Cell number, for the indicated times and treatments, was determined by Trypan blue exclusion and was normalized to the initial day of treatment (day 1). **Indicates a P-value <0.01 relative to individual treatments alone. Data shown are representative of three independent experiments. (b) Clonogenic assay using LNCaP cells treated with the indicated dose (Gy) of IR or in combination with PD (0.5 μM). Colonies were stained with crystal violet and counted 14 days post-treatment. Studies were performed in biological triplicate and are representative of three independent experiments. *Indicates a P-value <0.05 relative to IR treatment alone.
Figure 4
Figure 4
CRPC cells are sensitive to CDK4/6-specific inhibition, dependent on RB status. The relevance of PD treatment to advanced PCa was determined using multiple CRPC cell model systems. Flow cytometry was performed on CRPC cells: (a) PC3M; (b) LNCaP shRB1 (stable LNCaP cells depleted of the RB protein); (c) 22Rv1; and (d) C4-2 treated with control (left panels) or PD (right panels), as described in Figure 1a. Representative flow cytometry profiles for each cell model system are shown. The y axis denotes the number of fixed cells stained with propidium iodide (PI). Inset values: % PI-stained cells (mean±s.d., from an experiment performed in biological triplicate) in the G1-, S-, G2M-phases as determined using the cell cycle algorithm in FlowJo. Note: 22Rv1 and C4-2 cells were treated in media containing 5% charcoal-dextran treated (CDT) serum, as described in the Materials and methods section. (e) Gene expression analyses from C4-2 cells was performed, as described in Figure 2, to assess the impact of indicated therapies on AR activity. Representative AR target genes: KLK3/PSA (left) and TMPRSS2 (right) are shown. * ** indicates P-values: <0.05; 0.01, respectively.
Figure 5
Figure 5
CDK4/6-specific inhibition suppresses proliferation of xenografts in vivo and primary human prostate tissue ex vivo. The efficacy of PD on PCa was determined by: (a) Proliferative Ki-67 marker analysis from VCaP xenografts, grown in SCID mice, treated with control (n=4 mice) or PD (n=4 mice). Mice were treated with 150 mg/kg of PD, as described detail in the Materials and methods section. Representative images (20 × ) are shown (left panel) and Aperio-based quantification of % Ki-67 (right panel). *Indicates a P-value <0.05. (b) Representative images from histo- and immunohistological analyses of human prostate tissues used for ex vivo tissue explant culture: left panel, images (20 × ) from hematoxylin and eosin (H&E) and RB-stained tissue before processing for culture; right panel, H&E images (20 × , upper row) and Ki-67 images (20 × , lower row) from ex vivo explant tissues treated 6 days with control (left column) and 0.5 (middle column) or 1 μM PD (right column). (c) Aperio-based quantification of % Ki-67 from human explant tissues cultured ex vivo for a short (day 2) and long (day 6) treatment with control (C) or 0.5 or 1 μM PD. Bars represent (mean±s.e.m.) from five patient specimens. *Indicates a P-value <0.05.

References

    1. Knudsen KE, Kelly WK. Outsmarting androgen receptor: creative approaches for targeting aberrant androgen signaling in advanced prostate cancer. Expert Rev Endocrinol Metab. 2011;6:483–493.
    1. Udayakumar T, Shareef MM, Diaz DA, Ahmed MM, Pollack A. The E2F1/Rb and p53/MDM2 pathways in DNA repair and apoptosis: understanding the crosstalk to develop novel strategies for prostate cancer radiotherapy. Semin Radiat Oncol. 2010;20:258–266.
    1. Schiewer MJ, Augello MA, Knudsen KE. The AR dependent cell cycle: mechanisms and cancer relevance. Mol Cell Endocrinol. 2012;352:34–45.
    1. Collins I, Garrett MD. Targeting the cell division cycle in cancer: CDK and cell cycle checkpoint kinase inhibitors. Curr Opin Pharmacol. 2005;5:366–373.
    1. Diaz-Padilla I, Siu LL, Duran I. Cyclin-dependent kinase inhibitors as potential targeted anticancer agents. Invest New Drugs. 2009;27:586–594.
    1. Dickson MA, Schwartz GK. Development of cell-cycle inhibitors for cancer therapy. Curr Oncol. 2009;16:36–43.
    1. Wesierska-Gadek J, Maurer M, Zulehner N, Komina O. Whether to target single or multiple CDKs for therapy? That is the question. J Cell Physiol. 2011;226:341–349.
    1. Drees M, Dengler WA, Roth T, Labonte H, Mayo J, Malspeis L, et al. Flavopiridol (L86-8275): selective antitumor activity in vitro and activity in vivo for prostate carcinoma cells. Clin Cancer Res. 1997;3:273–279.
    1. Sedlacek H, Czech J, Naik R, Kaur G, Worland P, Losiewicz M, et al. Flavopiridol (L86 8275; NSC 649890), a new kinase inhibitor for tumor therapy. Int J Oncol. 1996;9:1143–1168.
    1. Senderowicz AM, Headlee D, Stinson SF, Lush RM, Kalil N, Villalba L, et al. Phase I trial of continuous infusion flavopiridol, a novel cyclin-dependent kinase inhibitor, in patients with refractory neoplasms. J Clin Oncol. 1998;16:2986–2999.
    1. Liu G, Gandara DR, Lara PN, Jr., Raghavan D, Doroshow JH, Twardowski P, et al. A Phase II trial of flavopiridol (NSC #649890) in patients with previously untreated metastatic androgen-independent prostate cancer. Clin Cancer Res. 2004;10:924–928.
    1. Toogood PL, Harvey PJ, Repine JT, Sheehan DJ, VanderWel SN, Zhou H, et al. Discovery of a potent and selective inhibitor of cyclin-dependent kinase 4/6. J Med Chem. 2005;48:2388–2406.
    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. Vaughn DJ, Flaherty K, Lal P, Gallagher M, O'Dwyer P, Wilner K, et al. Treatment of growing teratoma syndrome. N Engl J Med. 2009;360:423–424.
    1. Marzec M, Kasprzycka M, Lai R, Gladden AB, Wlodarski P, Tomczak E, et al. Mantle cell lymphoma cells express predominantly cyclin D1a isoform and are highly sensitive to selective inhibition of CDK4 kinase activity. Blood. 2006;108:1744–1750.
    1. Wang L, Wang J, Blaser BW, Duchemin AM, 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.
    1. Rodriguez-Otero P, Roman-Gomez J, Vilas-Zornoza A, Jose-Eneriz ES, Martin-Palanco V, Rifon J, et al. Deregulation of FGFR1 and CDK6 oncogenic pathways in acute lymphoblastic leukaemia harbouring epigenetic modifications of the MIR9 family. Br J Haematol. 2011;155:73–83.
    1. Menu E, Garcia J, Huang X, Di Liberto M, Toogood PL, Chen I, et al. A novel therapeutic combination using PD 0332991 and bortezomib: study in the 5T33MM myeloma model. Cancer Res. 2008;68:5519–5523.
    1. Baughn LB, Di Liberto M, Wu K, Toogood PL, Louie T, Gottschalk R, et al. A novel orally active small molecule potently induces G1 arrest in primary myeloma cells and prevents tumor growth by specific inhibition of cyclin-dependent kinase 4/6. Cancer Res. 2006;66:7661–7667.
    1. Finn RS, Dering J, Conklin D, Kalous O, Cohen DJ, Desai AJ, et al. PD 0332991, a selective cyclin D kinase 4/6 inhibitor, preferentially inhibits proliferation of luminal estrogen receptor-positive human breast cancer cell lines in vitro. Breast Cancer Res. 2009;11:R77.
    1. Miller TW, Balko JM, Fox EM, Ghazoui Z, Dunbier A, Anderson H, et al. ERalpha-dependent E2F transcription can mediate resistance to estrogen deprivation in human breast cancer. Cancer Discov. 2011;1:338–351.
    1. Roberts PJ, Bisi JE, Strum JC, Combest AJ, Darr DB, Usary JE, et al. Multiple roles of cyclin-dependent kinase 4/6 inhibitors in cancer therapy. J Natl Cancer Inst. 2012;104:476–487.
    1. Thangavel C, Dean JL, Ertel A, Knudsen KE, Aldaz CM, Witkiewicz AK, et al. Therapeutically activating RB: reestablishing cell cycle control in endocrine therapy-resistant breast cancer. Endocr Relat Cancer. 2011;18:333–345.
    1. Zhang C, Yan Z, Arango ME, Painter CL, Anderes K. Advancing bioluminescence imaging technology for the evaluation of anticancer agents in the MDA-MB-435-HAL-Luc mammary fat pad and subrenal capsule tumor models. Clin Cancer Res. 2009;15:238–246.
    1. Zhang CC, Yan Z, Li W, Kuszpit K, Painter CL, Zhang Q, et al. [18F]FLT-PET imaging does not always "light up" proliferating tumor cells. Clin Cancer Res. 2012;18:1303–1312.
    1. Sutherland JJ, Low J, Blosser W, Dowless M, Engler TA, Stancato LF. A robust high-content imaging approach for probing the mechanism of action and phenotypic outcomes of cell-cycle modulators. Mol Cancer Ther. 2011;10:242–254.
    1. Heijink DM, Fehrmann RS, de Vries EG, Koornstra JJ, Oosterhuis D, van der Zee AG, et al. A bioinformatical and functional approach to identify novel strategies for chemoprevention of colorectal cancer. Oncogene. 2011;30:2026–2036.
    1. Cole AM, Myant K, Reed KR, Ridgway RA, Athineos D, Van den Brink GR, et al. Cyclin D2-cyclin-dependent kinase 4/6 is required for efficient proliferation and tumorigenesis following Apc loss. Cancer Res. 2010;70:8149–8158.
    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.
    1. Ismail A, Bandla S, Reveiller M, Toia L, Zhou Z, Gooding WE, et al. Early G cyclin-dependent kinases as prognostic markers and potential therapeutic targets in esophageal adenocarcinoma. Clin Cancer Res. 2011;17:4513–4522.
    1. Wiedemeyer WR, Dunn IF, Quayle SN, Zhang J, Chheda MG, Dunn GP, et al. Pattern of retinoblastoma pathway inactivation dictates response to CDK4/6 inhibition in GBM. Proc Natl Acad Sci USA. 2010;107:11501–11506.
    1. Michaud K, Solomon DA, Oermann E, Kim JS, Zhong WZ, Prados MD, et al. Pharmacologic inhibition of cyclin-dependent kinases 4 and 6 arrests the growth of glioblastoma multiforme intracranial xenografts. Cancer Res. 2010;70:3228–3238.
    1. Schwartz GK, LoRusso PM, Dickson MA, Randolph SS, Shaik MN, Wilner KD, et al. Phase I study of PD 0332991, a cyclin-dependent kinase inhibitor, administered in 3-week cycles (schedule 2/1) Br J Cancer. 2011;104:1862–1868.
    1. Leonard JP, Lacasce AS, Smith MR, Noy A, Chirieac LR, Rodig SJ, et al. Selective CDK4/6 inhibition with tumor responses by PD0332991 in patients with mantle cell lymphoma. Blood. 2012;119:4597–4607.
    1. Flaherty KT, Lorusso PM, Demichele A, Abramson VG, Courtney R, Randolph SS, et al. Phase I, dose-escalation trial of the oral cyclin-dependent kinase 4/6 inhibitor PD 0332991, administered using a 21-day schedule in patients with advanced cancer. Clin Cancer Res. 2012;18:568–576.
    1. Dean JL, Thangavel C, McClendon AK, Reed CA, Knudsen ES. Therapeutic CDK4/6 inhibition in breast cancer: key mechanisms of response and failure. Oncogene. 2010;29:4018–4032.
    1. Konecny GE, Winterhoff B, Kolarova T, Qi J, Manivong K, Dering J, et al. Expression of p16 and retinoblastoma determines response to CDK4/6 inhibition in ovarian cancer. Clin Cancer Res. 2011;17:1591–1602.
    1. Burkhart DL, Sage J. Cellular mechanisms of tumour suppression by the retinoblastoma gene. Nat Rev Cancer. 2008;8:671–682.
    1. Knudsen ES, Knudsen KE. Tailoring to RB: tumour suppressor status and therapeutic response. Nat Rev Cancer. 2008;8:714–724.
    1. Freemantle SJ, Liu X, Feng Q, Galimberti F, Blumen S, Sekula D, et al. Cyclin degradation for cancer therapy and chemoprevention. J Cell Biochem. 2007;102:869–877.
    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.
    1. Jirawatnotai S, Hu Y, Michowski W, Elias JE, Becks L, Bienvenu F, et al. A function for cyclin D1 in DNA repair uncovered by protein interactome analyses in human cancers. Nature. 2011;474:230–234.
    1. Comstock CE, Augello MA, Schiewer MJ, Karch J, Burd CJ, Ertel A, et al. Cyclin D1 is a selective modifier of androgen-dependent signaling and androgen receptor function. J Biol Chem. 2011;286:8117–8127.
    1. Schiewer MJ, Den R, Hoang DT, Augello MA, Lawrence YR, Dicker AP, et al. mTOR is a selective effector of the radiation therapy response in androgen receptor-positive prostate cancer. Endocr Relat Cancer. 2012;19:1–12.
    1. Tzelepi V, Zhang J, Lu JF, Kleb B, Wu G, Wan X, et al. Modeling a lethal prostate cancer variant with small-cell carcinoma features. Clin Cancer Res. 2011;18:666–677.
    1. Sharma A, Yeow WS, Ertel A, Coleman I, Clegg N, Thangavel C, et al. The retinoblastoma tumor suppressor controls androgen signaling and human prostate cancer progression. J Clin Invest. 2010;120:4478–4492.
    1. Rubin MA, Maher CA, Chinnaiyan AM. Common gene rearrangements in prostate cancer. J Clin Oncol. 2011;29:3659–3668.
    1. Rivadeneira DB, Mayhew CN, Thangavel C, Sotillo E, Reed CA, Grana X, et al. Proliferative suppression by CDK4/6 inhibition: complex function of the retinoblastoma pathway in liver tissue and hepatoma cells. Gastroenterology. 2010;138:1920–1930.
    1. Centenera MM, Gillis JL, Hanson A, Jindal S, Taylor RA, Risbridger G, et al. Evidence for efficacy of new Hsp90 inhibitors revealed by ex vivo culture of human prostate tumors. Clin Cancer Res. 2012;18:3562–3570.
    1. Thomas JP, Tutsch KD, Cleary JF, Bailey HH, Arzoomanian R, Alberti D, et al. Phase I clinical and pharmacokinetic trial of the cyclin-dependent kinase inhibitor flavopiridol. Cancer Chemother Pharmacol. 2002;50:465–472.
    1. Halvorsen OJ, Hostmark J, Haukaas S, Hoisaeter PA, Akslen LA. Prognostic significance of p16 and CDK4 proteins in localized prostate carcinoma. Cancer. 2000;88:416–424.
    1. Martin JM, Supiot S, Berthold DR. Pharmacotherapeutic management of locally advanced prostate cancer: current status. Drugs. 2011;71:1019–1041.
    1. Lolli G, Lowe ED, Brown NR, Johnson LN. The crystal structure of human CDK7 and its protein recognition properties. Structure. 2004;12:2067–2079.
    1. De Bondt HL, Rosenblatt J, Jancarik J, Jones HD, Morgan DO, Kim SH. Crystal structure of cyclin-dependent kinase 2. Nature. 1993;363:595–602.
    1. Day PJ, Cleasby A, Tickle IJ, O'Reilly M, Coyle JE, Holding FP, et al. Crystal structure of human CDK4 in complex with a D-type cyclin. Proc Natl Acad Sci USA. 2009;106:4166–4170.
    1. Brotherton DH, Dhanaraj V, Wick S, Brizuela L, Domaille PJ, Volyanik E, et al. Crystal structure of the complex of the cyclin D-dependent kinase Cdk6 bound to the cell-cycle inhibitor p19INK4d. Nature. 1998;395:244–250.
    1. Lu H, Schulze-Gahmen U. Toward understanding the structural basis of cyclin-dependent kinase 6 specific inhibition. J Med Chem. 2006;49:3826–3831.
    1. Lim JT, Mansukhani M, Weinstein IB. Cyclin-dependent kinase 6 associates with the androgen receptor and enhances its transcriptional activity in prostate cancer cells. Proc Natl Acad Sci USA. 2005;102:5156–5161.
    1. Aparicio A, Den RB, Knudsen KE. Time to stratify? The retinoblastoma protein in castrate-resistant prostate cancer. Nat Rev Urol. 2011;8:562–568.
    1. Wang Q, Li W, Zhang Y, Yuan X, Xu K, Yu J, et al. Androgen receptor regulates a distinct transcription program in androgen-independent prostate cancer. Cell. 2009;138:245–256.
    1. Mostaghel EA, Plymate S. New hormonal therapies for castration-resistant prostate cancer. Endocrinol Metab Clin North Am. 2011;40:625–642.
    1. Johnson N, Shapiro GI. Cyclin-dependent kinases (cdks) and the DNA damage response: rationale for cdk inhibitor-chemotherapy combinations as an anticancer strategy for solid tumors. Expert Opin Ther Targets. 2010;14:1199–1212.
    1. Knudsen KE, Arden KC, Cavenee WK. Multiple G1 regulatory elements control the androgen-dependent proliferation of prostatic carcinoma cells. J Biol Chem. 1998;273:20213–20222.
    1. Korzynska A, Zychowicz M. A method of estimation of the cell doubling time on the basis of cell culture monitoring data. Biocybernet Biomed Engineer. 2008;28:75–82.
    1. Wetherill YB, Hess-Wilson JK, Comstock CE, Shah SA, Buncher CR, Sallans L, et al. Bisphenol A facilitates bypass of androgen ablation therapy in prostate cancer. Mol Cancer Ther. 2006;5:3181–3190.

Source: PubMed

3
Subscribe