A phase 1 dose escalation and expansion study of Tarextumab (OMP-59R5) in patients with solid tumors

David C Smith, Rashmi Chugh, Amita Patnaik, Kyriakos P Papadopoulos, Min Wang, Ann M Kapoun, Lu Xu, Jakob Dupont, Robert J Stagg, Anthony Tolcher, David C Smith, Rashmi Chugh, Amita Patnaik, Kyriakos P Papadopoulos, Min Wang, Ann M Kapoun, Lu Xu, Jakob Dupont, Robert J Stagg, Anthony Tolcher

Abstract

Purpose This Phase I trial evaluated the maximum tolerated dose, safety, pharmacokinetics, pharmacodynamics and preliminary efficacy of tarextumab (OMP-5948), a novel cross-reactive antibody which binds and selectively inhibits signaling via both Notch2 and Notch3, in adult patients with advanced malignancies. Methods Standard 3 + 3 design with tarextumab 0.5, 1, 2.5, or 5 mg/kg weekly, or 5, 7.5, or 10 mg/kg every other week, or 7.5 mg every 3 weeks. Dose-limiting toxicities (DLT) were assessed during the first 28 days. Results Forty-two patients received tarextumab (21 weekly, 15 every other week, 6 every three weeks). 2/6 subjects at the 5 mg/kg weekly dose, 2/3 at 10 mg/kg every other week, and 0/6 at 7.5 mg/kg every three weeks had a DLT. The maximum tolerated dose (MTD) was 2.5 mg/kg weekly, and 7.5 mg/kg on the every other and every three week schedules. Gastrointestinal (GI) toxicity was the most common adverse event with diarrhea (81%), fatigue (48%), nausea (45%), anorexia (38%), and vomiting (38%) and abdominal pain and constipation (24% each). Biomarker analysis showed regulation of stem cell and Notch gene signaling. Conclusion Tarextumab was generally well-tolerated at doses <2.5 mg weekly and 7.5 mg/kg every other and every third week. Diarrhea was dose-limiting above these levels, but relatively easily managed at lower doses. Inhibition of Notch pathway signaling was demonstrated at these doses. ClinicalTrials.gov Identifier: NCT01277146.

Keywords: Notch inhibition; Phase 1; Tarextumab.

Conflict of interest statement

At the time of the study Drs. Wang, Kapoun, Xu, Dupont and Stagg were employees of OncoMed. Dr. Dupont is currently employed by Genentech. Drs. Smith and Chugh are employees of the University of Michigan which has licensed intellectual property to OncoMed Pharmaceuticals and may receive royalties. Drs. Smith, Chugh, Patnaik, Papadopoulos and Tolcher reported that the research was funded at their institutions by OncoMed.

Figures

Fig. 1
Fig. 1
Group mean concentration-time profiles. Data organized by nominal time; sample tested below LLOQ (lower limit of quantitation) were imputed to 1 μg/mL
Fig. 2
Fig. 2
Stem cell and differentiation genes regulated in hair follicles by tarextumab treatment. Tarextumab significantly affected genes associated with cellular fate (KITLG, RGS14, ADAM23). RNAs were isolated from hair follicles (n = 19 pt). The y-axis represents gene expression at baseline and 28 days following treatment initiation. The pts. represented were dosed as follows: 0.5 mg/kg QW (n = 3), 1.0 mg/kg QW (n = 3), 2.5 mg/kg QW (n = 5), 5.0 mg/kg QW (n = 8)
Fig. 3
Fig. 3
Effects of tarextumab treatment on Notch target gene expression in whole blood. Tarextumab significantly down-regulated expression of Notch pathway genes, HES1 and NEURL in whole blood. RNAs were isolated from whole blood from each of 38 patients. The y-axis represents gene expression at baseline (Day 0) and at various time points following treatment initiation (indicated along x-axis). The patients represented were dosed as follows: 0.5 mg/kg QW (n = 3), 1.0 mg/kg QW (n = 3), 2.5 mg/kg QW (n = 6), 5.0 mg/kg QW (n = 9)
Fig. 4
Fig. 4
Tarextumab reduced Notch and stem cell signatures in biopsied tumors. Gene Set Enrichment Analysis (GSEA) showed tarextumab significantly affected gene expression associated with Notch signaling and stem cell genes in 3 paired tumor biopsies. a Heat map of fold change of gene expression ratios comparing post-treatment (day 35) with pre- treatment (day 0) samples. Gene set enrichment analysis demonstrated that (b) Notch target and (c) consensus cancer stem cell genes (CSC, Pubmed#21169407) were significantly down-regulated in the treated tumors post tarextumab treatment

References

    1. Pannuti A, Foreman K, Rizzo P, Osips C, Golde T, Osborne B, Miele L (2010) Targeting notch to target Cancer stem cells. CCR Focus. 10.1158/078-0432.CCR-09-2823
    1. Kopan R, Ilagan MX. The canonical notch signaling pathway: unfolding the activation mechanism. Cell. 2009;137:216–233. doi: 10.1016/j.cell.2009.03.045.
    1. Yen WC, Fischer MM, Axelrod F, Bond C, Cain J, Cancilla B, Henner WR, Meisner R, Sato A, Shah J, Tang T, Wallace B, Wang M, Zhang C, Kapoun AM, Lewicki J, Gurney A, Hoey T. Targeting notch signaling with a Notch2/Notch3 antagonist (tarextumab) inhibits tumor growth and decreases tumor-initiating cell frequency. Clin Cancer Res. 2015;21:2084–2095. doi: 10.1158/1078-0432.CCR-14-2808.
    1. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, Rubinstein L, Shankar L, Dodd L, Kaplan R, Lacombe D, Verweij J. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45:228–247. doi: 10.1016/j.ejca.2008.10.026.
    1. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47. doi: 10.1093/nar/gkv007.
    1. DiCiccio TJ, Efron B. Bootstrap confidence intervals. Stat Sci. 1996;11:189–212. doi: 10.1214/ss/1032280214.
    1. Efron B, Tibshirani R. Bootstrap confidence intervals, confidence intervals and other methods of statistical accuracy. Stat Sci. 1986;1:54–75. doi: 10.1214/ss/1177013815.
    1. Krop I, Demuth T, Guthrie T, Wen PY, Mason WP, Chinnaiyan P, Butowski N, Groves MD, Kesari S, Freedman SJ, Blackman S, Watters J, Loboda A, Podtelezhnikov A, Lunceford J, Chen C, Giannotti M, Hing J, Beckman R, LoRusso P. Phase I pharmacologic and pharmacodynamic study of the gamma secretase (notch) inhibitor MK-0752 in adult patients with advanced solid tumors. J Clin Oncol. 2012;30:2307–2313. doi: 10.1200/JCO.2011.39.1540.
    1. Messersmith WA, Shapiro GI, Cleary JM, Jimeno A, Dasari A, Huang B, Shaik MN, Cesari R, Zheng X, Reynolds JM, English PA, McLachlan KR, Kern KA, LoRusso PM. A phase I, dose-finding study in patients with advanced solid malignancies of the oral gamma-secretase inhibitor PF-03084014. Clin Cancer Res. 2015;21:60–67. doi: 10.1158/1078-0432.CCR-14-0607.
    1. Fre S, Huyghe M, Mourikis P, Robine S, Louvard D, Artavanis-Tsakonas S. Notch signals control the fate of immature progenitor cells in the intestine. Nature. 2005;435:964–968. doi: 10.1038/nature03589.
    1. van Es JH, van Gijn ME, Riccio O, van den Born M, Vooijs M, Begthel H, et al. Notch/gamma-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells. Nature. 2005;435:959–963. doi: 10.1038/nature03659.
    1. Wei P, Walls M, Qiu M, Ding R, Denlinger RH, Wong A, Tsaparikos K, Jani JP, Hosea N, Sands M, Randolph S, Smeal T. Evaluation of selective gamma-secretase inhibitor PF-03084014 for its antitumor efficacy and gastrointestinal safety to guide optimal clinical trial design. Mol Cancer Ther. 2010;9:1618–1628. doi: 10.1158/1535-7163.MCT-10-0034.
    1. Schott AF, Landis MD, Dontu G, Griffith KA, Layman RM, Krop I, Paskett LA, Wong H, Dobrolecki LE, Lewis MT, Froehlich AM, Paranilam J, Hayes DF, Wicha MS, Chang JC. Preclinical and clinical studies of gamma secretase inhibitors with docetaxel on human breast tumors. Clin Cancer Res. 2013;19:1512–1524. doi: 10.1158/1078-0432.CCR-11-3326.
    1. Tolcher AW, Messersmith WA, Mikulski SM, Papadopoulos KP, Kwak EL, Gibbon DG, Patnaik A, Falchook GS, Dasari A, Shapiro GI, Boylan JF, Xu ZX, Wang K, Koehler A, Song J, Middleton SA, Deutsch J, DeMario M, Kurzrock R, Wheler JJ. Phase I study of RO4929097, a gamma secretase inhibitor of notch signaling, in patients with refractory metastatic or locally advanced solid tumors. J Clin Oncol. 2012;30:2348–2353. doi: 10.1200/JCO.2011.36.8282.

Source: PubMed

3
S'abonner