Abstract

Abstract Background: Acute myeloid leukemia (AML) is an aggressive leukemia of myeloid lineage. CSF1R (colony-stimulating factor 1 receptor) regulates the proliferation, differentiation and polarization of cells of myeloid lineages, e.g. monocytes/macrophages. CSF1R expression is aberrantly upregulated in many AML, leading to a hypothesis that CSF1R is a putative leukemogenic driver in a subset of AML and thus could be a potential target for treating AML. Previous studies demonstrated that Narazaciclib (HX301 or ON123300), an investigational kinase inhibitor (TKi), exhibited high potency activity against CSF1R (IC50 0.285 nM) and inhibition of macrophage proliferation and AML tumor growth both in vitro/in vivo, implicating its potential use as an AML therapeutic1 (An et al., 2023 AACR Annual Meeting). In addition, narazaciclib has recently completed the dose escalation phase 1 study in advanced solid tumors. However, the therapeutic window of narazaciclib as a CSF1Ri for treating AML remains to be determined. The present study attempted to utilize both preclinical PK and in vivo efficacy models to predict the effective clinical dose of narazaciclib for guiding the future AML clinical trials. Methods: A single dose of narazaciclib at various of dose levels was orally administrated in NOD/Scid mice, and plasma samples were collected at designated timepoints. The concentrations of narazaciclib in plasma were analyzed by LC-MS. In a Ba/F3-ETV6-CSF1R in vivo xenograft tumor model in Nod/Scid mice, narazaciclib at various dose levels was orally administrated daily. Tumor volumes were measured twice weekly. Results: The mouse PK study showed that narazaciclib displayed overall similar absorption pattern as seen in unpublished human clinical data. Narazaciclib was absorbed rapidly after oral dosing with a mean Tmax ranging from 1 to 2 hours in mouse. However, narazaciclib was cleared faster in mouse, with a mean T1/2 ranging from 1.9 to 2.8 hours, than in human. In addition, the exposures of narazaciclib, as measured by Cmax and AUC in plasma, increased in a relatively dose-proportional manner, as seen in human patients. In the Ba/F3-ETV6-CSF1R xenograft model where the tumor cell growth is solely dependent on the CSF1R kinase activity, narazaciclib at 10 mg/kg did not induce any anti-tumor effects with a TGI of -14.61%. In contrast, narazaciclib at 50 mg/kg and 100 mg/kg exerted significant anti-tumor effects with a TGI of 85.49% and 94.55%, respectively. Based on the mouse PK results, narazaciclib at 50 mg/kg in mouse is estimated to have an AUC of approximate 2000 h*ng/mL, which is equivalent to the dose of 160 - 200 mg/d in human patients. Therefore, 160 mg dose level is predicted to be an effective dose for narazaciclib in AML patients. Conclusions: Narazaciclib at 160 mg/d is predicted to be an effective dose in AML patients. Citation Format: Tao Yang, Hang Ke, Jinping Liu, Kefeng Gong, Henry Qixiang Li. Preclinical pharmacology modeling (pharmacokinetics/efficacy) of narazaciclib (HX301 or ON123300) to predict its clinical effective dose [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 7161.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call