Potentiation of tumor responses to DNA damaging therapy by the selective ATR inhibitor VX-970

Amy B Hall, Dave Newsome, Yuxin Wang, Diane M Boucher, Brenda Eustace, Yong Gu, Brian Hare, Mac A Johnson, Sean Milton, Cheryl E Murphy, Darin Takemoto, Crystal Tolman, Mark Wood, Peter Charlton, Jean-Damien Charrier, Brinley Furey, Julian Golec, Philip M Reaper, John R Pollard, Amy B Hall, Dave Newsome, Yuxin Wang, Diane M Boucher, Brenda Eustace, Yong Gu, Brian Hare, Mac A Johnson, Sean Milton, Cheryl E Murphy, Darin Takemoto, Crystal Tolman, Mark Wood, Peter Charlton, Jean-Damien Charrier, Brinley Furey, Julian Golec, Philip M Reaper, John R Pollard

Abstract

Platinum-based DNA-damaging chemotherapy is standard-of-care for most patients with lung cancer but outcomes remain poor. This has been attributed, in part, to the highly effective repair network known as the DNA-damage response (DDR). ATR kinase is a critical regulator of this pathway, and its inhibition has been shown to sensitize some cancer, but not normal, cells in vitro to DNA damaging agents. However, there are limited in vivo proof-of-concept data for ATR inhibition. To address this we profiled VX-970, the first clinical ATR inhibitor, in a series of in vitro and in vivo lung cancer models and compared it with an inhibitor of the downstream kinase Chk1. VX-970 markedly sensitized a large proportion of a lung cancer cell line and primary tumor panel in vitro to multiple DNA damaging drugs with clear differences to Chk1 inhibition observed. In vivo VX-970 blocked ATR activity in tumors and dramatically enhanced the efficacy of cisplatin across a panel of patient derived primary lung xenografts. The combination led to complete tumor growth inhibition in three cisplatin-insensitive models and durable tumor regression in a cisplatin-sensitive model. These data provide a strong rationale for the clinical evaluation of VX-970 in lung cancer patients.

Figures

Figure 1. VX-970 is a potent and…
Figure 1. VX-970 is a potent and selective inhibitor of ATR
(A) Chemical structure of VX-970. (B) Exponentially growing H2009 cells were treated overnight (17 h) with the indicated concentrations of VX-970 alone (lanes 1-6) or in combination with 20 μM cisplatin (lanes 7-12). Cells were then harvested and analyzed for the expression of P-Chk1-S345, P-H2AX-S139 and P-KAP1-S824 by immunoblotting.
Figure 2. ATR inhibition sensitizes lung cancer…
Figure 2. ATR inhibition sensitizes lung cancer cells to DNA damaging agents
(A) Analysis of shifts in the concentration of DNA damaging agent required to inhibit cell viability by 50% (IC50) was used to determine the synergistic or antagonistic effects of a DDR inhibitor. Heat map representing the IC50 shift with the largest absolute value observed with VX-970 or AZD7762 in combination with cisplatin, etoposide, gemcitabine, oxaliplatin and irinotecan across a panel of 36 lung cell lines at 96 h. The colors represent a shift range from −10 (antagonism-blue) to +10 (synergy-red). (B) Histograms showing the percentage of cell lines with > 3-fold (top panel) or 10-fold (bottom panel) synergy with VX-970 (ATR) or AZD7762 (Chk1/2) in combination with cisplatin, oxaliplatin, irinotecan, gemcitabine and etoposide. (C) Impact of p53 on response to VX-970 in A549 cells. Histogram depicts maximum IC50 shift in vector control and p53 knockdown cells observed with VX-970 in combination with cisplatin, etoposide, gemcitabine, oxaliplatin and irinotecan.
Figure 3. VX-970 synergizes with cisplatin across…
Figure 3. VX-970 synergizes with cisplatin across a range of human primary lung tumor models in vitro
Dissociated tumor cells were treated in triplicate in vitro with a matrix of VX-970 and cisplatin concentrations, and synergy or antagonism was analyzed at the 95% confidence interval with MacSynergy II software. Degree of synergy, shown as positive log volume, and antagonism, shown as negative log volume, are shown.
Figure 4. VX-970 enhances the therapeutic efficacy…
Figure 4. VX-970 enhances the therapeutic efficacy of cisplatin in patient-derived lung tumor xenografts
(A-G) Human primary tumor tissues were passaged in SCID mice. Treatment started when the average tumor size was approximately 200 mm3. Tumor bearing mice were treated with vehicle, VX-970 (30 mg/kg in all models except 60 mg/kg in OD26749 and OD26131) PO, 4 consecutive days a week, alone and in combination with cisplatin (3 mg/kg IP, q7d), and cisplatin alone. Tumor volume and body weight were measured twice a week. Studies were terminated one or two days after the final dose of VX-970. Points show the mean tumor volume (mm3) for each treatment group (n=5-10).
Figure 5. VX-970 inhibits ATR and promotes…
Figure 5. VX-970 inhibits ATR and promotes sustained regression in a human primary lung tumor xenograft
(A) Mice bearing OD26749 tumors were treated with a single dose of VX-970 (60 mg/kg PO) and cisplatin (3 mg/kg IP) either as monotherapy or in combination. P-Chk1 and P-H2AX were analyzed by western blot 4 and 48 h after treatment, respectively, and were corrected to total H2AX levels. (B) Representative PET/CT images of mice at baseline (top row) and six days following the indicated treatment (bottom row). Concentration of 18FLT is indicated by the intensity of the orange color in the PET image. An X-ray CT image of the skeletal system (white) is superimposed for anatomical reference. The tumor is circled (green). Treatment with VX-970 alone had no discernable effect on PET signal (image not shown). (C) Average standard uptake value (SUV) of 18FLT. (D) Tumor size changes as determined by CT. (E) Examination of prognostic value of 18FLT imaging. (F) Assessment of the durability of response to VX-970. OD26749 tumors were passaged in SCID mice to P4. Treatment started when the average tumor size was approximately 200 mm3. Tumor bearing mice were treated with vehicle, VX-970 alone (60 mg/kg PO, 4 consecutive days a week), cisplatin alone (3 mg/kg, IP, q7d) and the combination, for two weeks. Vehicle and VX-970 groups were terminated after the final dose of VX-970. Following treatment discontinuation, tumor growth in the cisplatin alone and combination groups was followed twice a week until the average tumor volume reached 1000 mm3. The arrowhead on the graph marks the end of treatment. Error bars are standard errors for all graphs.

References

    1. Longley DB, Johnston PG. Molecular mechanisms of drug resistance. The Journal of Pathology. 2005;205:275–292.
    1. Olaussen KA, Dunant A, Fouret P, Brambilla E, Andre F, Haddad V, Taranchon E, Filipits M, Pirker R, Popper HH, Stahel R, Sabatier L, Pignon JP, Tursz T, Le Chevalier T, Soria JC, et al. DNA repair by ERCC1 in non-small-cell lung cancer and cisplatin-based adjuvant chemotherapy. The New England Journal of Medicine. 2006;355:983–991.
    1. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, Kros JM, Hainfellner JA, Mason W, Mariani L, Bromberg JE, Hau P, Mirimanoff RO, Cairncross JG, Janzer RC, Stupp R. MGMT gene silencing and benefit from temozolomide in glioblastoma. The New England Journal of Medicine. 2005;352:997–1003.
    1. Norquist B, Wurz KA, Pennil CC, Garcia R, Gross J, Sakai W, Karlan BY, Taniguchi T, Swisher EM. Secondary somatic mutations restoring BRCA1/2 predict chemotherapy resistance in hereditary ovarian carcinomas. Journal of Clinical Oncology: official journal of the American Society of Clinical Oncology. 2011;29:3008–3015.
    1. Jackson SP, Bartek J. The DNA-damage response in human biology and disease. Nature. 2009;461:1071–1078.
    1. Cimprich KA, Cortez D. ATR: an essential regulator of genome integrity. Nature Reviews Molecular Cell Biology. 2008;9:616–627.
    1. Kastan MB, Bartek J. Cell-cycle checkpoints and cancer. Nature. 2004;432:316–323.
    1. Reaper PM, Griffiths MR, Long JM, Charrier JD, Maccormick S, Charlton PA, Golec JM, Pollard JR. Selective killing of ATM- or p53-deficient cancer cells through inhibition of ATR. Nature Chemical Biology. 2011;7:428–430.
    1. Cancer Genome Atlas N. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70.
    1. Ding L, Getz G, Wheeler DA, Mardis ER, McLellan MD, Cibulskis K, Sougnez C, Greulich H, Muzny DM, Morgan MB, Fulton L, Fulton RS, Zhang Q, Wendl MC, Lawrence MS, Larson DE, et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature. 2008;455:1069–1075.
    1. Greenman C, Stephens P, Smith R, Dalgliesh GL, Hunter C, Bignell G, Davies H, Teague J, Butler A, Stevens C, Edkins S, O'Meara S, Vastrik I, Schmidt EE, Avis T, Barthorpe S, et al. Patterns of somatic mutation in human cancer genomes. Nature. 2007;446:153–158.
    1. Jiang H, Reinhardt HC, Bartkova J, Tommiska J, Blomqvist C, Nevanlinna H, Bartek J, Yaffe MB, Hemann MT. The combined status of ATM and p53 link tumor development with therapeutic response. Genes & Development. 2009;23:1895–1909.
    1. Halazonetis TD, Gorgoulis VG, Bartek J. An oncogene-induced DNA damage model for cancer development. Science. 2008;319:1352–1355.
    1. Toledo LI, Murga M, Zur R, Soria R, Rodriguez A, Martinez S, Oyarzabal J, Pastor J, Bischoff JR, Fernandez-Capetillo O. A cell-based screen identifies ATR inhibitors with synthetic lethal properties for cancer-associated mutations. Nature Structural & Molecular Biology. 2011;18:721–727.
    1. Murga M, Bunting S, Montana MF, Soria R, Mulero F, Canamero M, Lee Y, McKinnon PJ, Nussenzweig A, Fernandez-Capetillo O. A mouse model of ATR-Seckel shows embryonic replicative stress and accelerated aging. Nature Genetics. 2009;41:891–898.
    1. Nghiem P, Park PK, Kim Ys YS, Desai BN, Schreiber SL. ATR is not required for p53 activation but synergizes with p53 in the replication checkpoint. The Journal of Biological Chemistry. 2002;277:4428–4434.
    1. Sangster-Guity N, Conrad BH, Papadopoulos N, Bunz F. ATR mediates cisplatin resistance in a p53 genotype-specific manner. Oncogene. 2011;30:2526–2533.
    1. Fokas E, Prevo R, Pollard JR, Reaper PM, Charlton PA, Cornelissen B, Vallis KA, Hammond EM, Olcina MM, Gillies McKenna W, Muschel RJ, Brunner TB. Targeting ATR in vivo using the novel inhibitor VE-822 results in selective sensitization of pancreatic tumors to radiation. Cell Death & Disease. 2012;3:e441.
    1. Nghiem P, Park PK, Kim Y, Vaziri C, Schreiber SL. ATR inhibition selectively sensitizes G1 checkpoint-deficient cells to lethal premature chromatin condensation. Proceedings of the National Academy of Sciences of the United States of America. 2001;98:9092–9097.
    1. Gilad O, Nabet BY, Ragland RL, Schoppy DW, Smith KD, Durham AC, Brown EJ. Combining ATR suppression with oncogenic Ras synergistically increases genomic instability, causing synthetic lethality or tumorigenesis in a dosage-dependent manner. Cancer Research. 2010;70:9693–9702.
    1. Schoppy DW, Ragland RL, Gilad O, Shastri N, Peters AA, Murga M, Fernandez-Capetillo O, Diehl JA, Brown EJ. Oncogenic stress sensitizes murine cancers to hypomorphic suppression of ATR. The Journal of Clinical Investigation. 2012;122:241–252.
    1. Murga M, Campaner S, Lopez-Contreras AJ, Toledo LI, Soria R, Montana MF, D'Artista L, Schleker T, Guerra C, Garcia E, Barbacid M, Hidalgo M, Amati B, Fernandez-Capetillo O. Exploiting oncogene-induced replicative stress for the selective killing of Myc-driven tumors. Nature Structural & Molecular Biology. 2011;18:1331–1335.
    1. Pires IM, Olcina MM, Anbalagan S, Pollard JR, Reaper PM, Charlton PA, McKenna WG, Hammond EM. Targeting radiation-resistant hypoxic tumour cells through ATR inhibition. British Journal of Cancer. 2012;107:291–299.
    1. Hammond EM, Dorie MJ, Giaccia AJ. Inhibition of ATR leads to increased sensitivity to hypoxia/reoxygenation. Cancer Research. 2004;64:6556–6562.
    1. Lesnock JL, Darcy KM, Tian C, Deloia JA, Thrall MM, Zahn C, Armstrong DK, Birrer MJ, Krivak TC. BRCA1 expression and improved survival in ovarian cancer patients treated with intraperitoneal cisplatin and paclitaxel: a Gynecologic Oncology Group Study. British Journal of Cancer. 2013;108:1231–1237.
    1. Carser JE, Quinn JE, Michie CO, O'Brien EJ, McCluggage WG, Maxwell P, Lamers E, Lioe TF, Williams AR, Kennedy RD, Gourley C, Harkin DP. BRCA1 is both a prognostic and predictive biomarker of response to chemotherapy in sporadic epithelial ovarian cancer. Gynecologic Oncology. 2011;123:492–498.
    1. Travis WD. Pathology of lung cancer. Clinics in Chest Medicine. 2011;32:669–692.
    1. Cufer T, Ovcaricek T, O'Brien ME. Systemic therapy of advanced non-small cell lung cancer: major-developments of the last 5-years. European Journal of Cancer. 2013;49:1216–1225.
    1. Simos D, Sajjady G, Sergi M, Liew MS, Califano R, Ho C, Leighl N, White S, Summers Y, Petrcich W, Wheatley-Price P. Third-line chemotherapy in small-cell lung cancer: an international analysis. Clinical Lung Cancer. 2014;15:110–118.
    1. Raparia K, Villa C, DeCamp MM, Patel JD, Mehta MP. Molecular profiling in non-small cell lung cancer: a step toward personalized medicine. Archives of Pathology & Laboratory Medicine. 2013;137:481–491.
    1. Cancer Genome Atlas Research N. Comprehensive genomic characterization of squamous cell lung cancers. Nature. 2012;489:519–525.
    1. Smith J, Tho LM, Xu N, Gillespie DA. The ATM-Chk2 and ATR-Chk1 pathways in DNA damage signaling and cancer. Advances in Cancer Research. 2010;108:73–112.
    1. Huntoon CJ, Flatten KS, Wahner Hendrickson AE, Huehls AM, Sutor SL, Kaufmann SH, Karnitz LM. ATR Inhibition Broadly Sensitizes Ovarian Cancer Cells to Chemotherapy Independent of BRCA Status. Cancer Research. 2013;73:3683–3691.
    1. Zabludoff SD, Deng C, Grondine MR, Sheehy AM, Ashwell S, Caleb BL, Green S, Haye HR, Horn CL, Janetka JW, Liu D, Mouchet E, Ready S, Rosenthal JL, Queva C, Schwartz GK, et al. AZD7762, a novel checkpoint kinase inhibitor, drives checkpoint abrogation and potentiates DNA-targeted therapies. Molecular Cancer Therapeutics. 2008;7:2955–2966.
    1. Peasland A, Wang LZ, Rowling E, Kyle S, Chen T, Hopkins A, Cliby WA, Sarkaria J, Beale G, Edmondson RJ, Curtin NJ. Identification and evaluation of a potent novel ATR inhibitor, NU6027, in breast and ovarian cancer cell lines. British Journal of Cancer. 2011;105:372–381.
    1. Tehrani OS, Shields AF. PET imaging of proliferation with pyrimidines. Journal of nuclear medicine: official publication, Society of Nuclear Medicine. 2013;54:903–912.
    1. Lord CJ, Ashworth A. The DNA damage response and cancer therapy. Nature. 2012;481:287–294.
    1. O'Connor MJ, Martin NM, Smith GC. Targeted cancer therapies based on the inhibition of DNA strand break repair. Oncogene. 2007;26:7816–7824.
    1. Chanoux RA, Yin B, Urtishak KA, Asare A, Bassing CH, Brown EJ. ATR and H2AX cooperate in maintaining genome stability under replication stress. The Journal of Biological Chemistry. 2009;284:5994–6003.
    1. Wagner JM, Karnitz LM. Cisplatin-induced DNA damage activates replication checkpoint signaling components that differentially affect tumor cell survival. Molecular Pharmacology. 2009;76:208–214.
    1. Wang Q, Fan S, Eastman A, Worland PJ, Sausville EA, O'Connor PM. UCN-01: a potent abrogator of G2 checkpoint function in cancer cells with disrupted p53. Journal of the National Cancer Institute. 1996;88:956–965.
    1. Blackwood E, Epler J, Yen I, Flagella M, O'Brien T, Evangelista M, Schmidt S, Xiao Y, Choi J, Kowanetz K, Ramiscal J, Wong K, Jakubiak D, Yee S, Cain G, Gazzard L, et al. Combination drug scheduling defines a “window of opportunity” for chemopotentiation of gemcitabine by an orally bioavailable, selective CHK1 inhibitor, GNE-900. Molecular Cancer Therapeutics. 2013
    1. Origanti S, Cai SR, Munir AZ, White LS, Piwnica-Worms H. Synthetic lethality of Chk1 inhibition combined with p53 and/or p21 loss during a DNA damage response in normal and tumor cells. Oncogene. 2013;32:577–588.
    1. Andreassen PR, D'Andrea AD, Taniguchi T. ATR couples FANCD2 monoubiquitination to the DNA-damage response. Genes & development. 2004;18:1958–1963.
    1. Shigechi T, Tomida J, Sato K, Kobayashi M, Eykelenboom JK, Pessina F, Zhang Y, Uchida E, Ishiai M, Lowndes NF, Yamamoto K, Kurumizaka H, Maehara Y, Takata M. ATR-ATRIP kinase complex triggers activation of the Fanconi anemia DNA repair pathway. Cancer Research. 2012;72:1149–1156.
    1. Singh TR, Ali AM, Paramasivam M, Pradhan A, Wahengbam K, Seidman MM, Meetei AR. ATR-Dependent Phosphorylation of FANCM at Serine 1045 Is Essential for FANCM Functions. Cancer Research. 2013;73:4300–4310.
    1. Ritz C, Streibig JC. Bioassay Analysis Using R. Journal of Statistical Software. 2005;12:1–22.
    1. Andreotti PE, Cree IA, Kurbacher CM, Hartmann DM, Linder D, Harel G, Gleiberman I, Caruso PA, Ricks SH, Untch M, et al. Chemosensitivity testing of human tumors using a microplate adenosine triphosphate luminescence assay: clinical correlation for cisplatin resistance of ovarian carcinoma. Cancer Research. 1995;55:5276–5282.

Source: PubMed

3
Tilaa