Phase I Study of ATR Inhibitor M6620 in Combination With Topotecan in Patients With Advanced Solid Tumors.

Purpose Our preclinical work identified depletion of ATR as a top candidate for topoisomerase 1 (TOP1) inhibitor synthetic lethality and showed that ATR inhibition sensitizes tumors to TOP1 inhibitors. We hypothesized that a combination of selective ATR inhibitor M6620 (previously VX-970) and topotecan, a selective TOP1 inhibitor, would be tolerable and active, particularly in tumors with high replicative stress. Patients and Methods This phase I study tested the combination of M6620 and topotecan in 3-week cycles using 3 + 3 dose escalation. The primary end point was the identification of the maximum tolerated dose of the combination. Efficacy and pharmacodynamics were secondary end points. Results Between September 2016 and February 2017, 21 patients enrolled. The combination was well tolerated, which allowed for dose escalation to the highest planned dose level (topotecan 1.25 mg/m2, days 1 to 5; M6620 210 mg/m2, days 2 and 5). One of six patients at this dose level experienced grade 4 thrombocytopenia that required transfusion, a dose-limiting toxicity. Most common treatment-related grade 3 or 4 toxicities were anemia, leukopenia, and neutropenia (19% each); lymphopenia (14%); and thrombocytopenia (10%). Two partial responses (≥ 18 months, ≥ 7 months) and seven stable disease responses ≥ 3 months (median, 9 months; range, 3 to 12 months) were seen. Three of five patients with small-cell lung cancer, all of whom had platinum-refractory disease, had a partial response or prolonged stable disease (10, ≥ 6, and ≥ 7 months). Pharmacodynamic studies showed preliminary evidence of ATR inhibition and enhanced DNA double-stranded breaks in response to the combination. Conclusion To our knowledge, this report is the first of an ATR inhibitor-chemotherapy combination. The maximum dose of topotecan plus M6620 is tolerable. The combination seems particularly active in platinum-refractory small-cell lung cancer, which tends not to respond to topotecan alone. Phase II studies with biomarker evaluation are ongoing.

[1]  M. Lawrence,et al.  APOBEC3A and APOBEC3B Activities Render Cancer Cells Susceptible to ATR Inhibition. , 2017, Cancer research.

[2]  M. Ellis,et al.  CDK4/6 inhibition triggers anti-tumor immunity , 2017, Nature.

[3]  A. Nicolas,et al.  The impact of replication stress on replication dynamics and DNA damage in vertebrate cells , 2017, Nature Reviews Genetics.

[4]  K. Ishii,et al.  DNA-Containing Exosomes Derived from Cancer Cells Treated with Topotecan Activate a STING-Dependent Pathway and Reinforce Antitumor Immunity , 2017, The Journal of Immunology.

[5]  S. Jackson,et al.  Targeting DNA Repair in Cancer: Beyond PARP Inhibitors. , 2017, Cancer discovery.

[6]  Samuel E. Jones,et al.  ATR inhibitors as a synthetic lethal therapy for tumours deficient in ARID1A , 2016, Nature Communications.

[7]  Y. Pommier,et al.  Small cell lung cancer: Time to revisit DNA-damaging chemotherapy , 2016, Science Translational Medicine.

[8]  N. Tunariu,et al.  Phase I trial of a first-in-class ATR inhibitor VX-970 as monotherapy (mono) or in combination (combo) with carboplatin (CP) incorporating pharmacodynamics (PD) studies. , 2016 .

[9]  O. Fernandez-Capetillo,et al.  A Genome-wide CRISPR Screen Identifies CDC25A as a Determinant of Sensitivity to ATR Inhibitors. , 2016, Molecular cell.

[10]  T. Stankovic,et al.  ATR inhibition induces synthetic lethality and overcomes chemoresistance in TP53- or ATM-defective chronic lymphocytic leukemia cells. , 2016, Blood.

[11]  N. Horita,et al.  Topotecan for Relapsed Small-cell Lung Cancer: Systematic Review and Meta-Analysis of 1347 Patients , 2015, Scientific Reports.

[12]  T. Stankovic,et al.  Synthetic lethality in chronic lymphocytic leukaemia with DNA damage response defects by targeting the ATR pathway , 2015, The Lancet.

[13]  H. Wakimoto,et al.  Alternative lengthening of telomeres renders cancer cells hypersensitive to ATR inhibitors , 2015, Science.

[14]  Y. Pommier,et al.  ATR inhibitors VE-821 and VX-970 sensitize cancer cells to topoisomerase i inhibitors by disabling DNA replication initiation and fork elongation responses. , 2014, Cancer research.

[15]  Abdelghani Mazouzi,et al.  DNA replication stress: causes, resolution and disease. , 2014, Experimental cell research.

[16]  P. Reaper,et al.  Potentiation of tumor responses to DNA damaging therapy by the selective ATR inhibitor VX-970 , 2014, Oncotarget.

[17]  D. Cortez,et al.  ATR pathway inhibition is synthetically lethal in cancer cells with ERCC1 deficiency. , 2014, Cancer research.

[18]  K. Cimprich,et al.  Causes and consequences of replication stress , 2013, Nature Cell Biology.

[19]  C. Seedhouse,et al.  Ataxia Telangiectasia Mutated and Rad3 Related (ATR) Protein Kinase Inhibition Is Synthetically Lethal in XRCC1 Deficient Ovarian Cancer Cells , 2013, PloS one.

[20]  B. Cornelissen,et al.  Targeting ATR in vivo using the novel inhibitor VE-822 results in selective sensitization of pancreatic tumors to radiation , 2012, Cell Death and Disease.

[21]  W. Mckenna,et al.  Targeting radiation-resistant hypoxic tumour cells through ATR inhibition , 2012, British Journal of Cancer.

[22]  O. Fernandez-Capetillo,et al.  Oncogenic stress sensitizes murine cancers to hypomorphic suppression of ATR. , 2012, The Journal of clinical investigation.

[23]  M. Barbacid,et al.  Exploiting oncogene-induced replicative stress for the selective killing of Myc-driven tumors , 2011, Nature Structural &Molecular Biology.

[24]  P. Reaper,et al.  Selective killing of ATM- or p53-deficient cancer cells through inhibition of ATR. , 2011, Nature chemical biology.

[25]  James R Bischoff,et al.  A cell-based screen identifies ATR inhibitors with synthetic lethal properties for cancer-associated mutations , 2011, Nature Structural &Molecular Biology.

[26]  A. Nakamura,et al.  Recent developments in the use of γ -H2AX as a quantitative DNA double-strand break biomarker , 2011, Aging.

[27]  B. Nabet,et al.  Combining ATR suppression with oncogenic Ras synergistically increases genomic instability, causing synthetic lethality or tumorigenesis in a dosage-dependent manner. , 2010, Cancer research.

[28]  A. Ashworth,et al.  Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. , 2009, The New England journal of medicine.

[29]  Y. Pommier Topoisomerase I inhibitors: camptothecins and beyond , 2006, Nature Reviews Cancer.

[30]  A. Pettitt,et al.  The ATR-p53 pathway is suppressed in noncycling normal and malignant lymphocytes , 2004, Oncogene.

[31]  Kai Rothkamm,et al.  Evidence for a lack of DNA double-strand break repair in human cells exposed to very low x-ray doses , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[32]  Junjie Chen,et al.  Histone H2AX Is Phosphorylated in an ATR-dependent Manner in Response to Replicational Stress* , 2001, The Journal of Biological Chemistry.

[33]  D. Lawrence,et al.  Protection of normal proliferating cells against chemotherapy by staurosporine-mediated, selective, and reversible G(1) arrest. , 2000, Journal of the National Cancer Institute.

[34]  M. Ratain,et al.  Topoisomerase I Inhibitors. , 1997, The oncologist.