Abstract

Abstract Background: INO-VATE (NCT01564784) demonstrated higher rates of remission, MRD-negativity, and improved overall survival (hazard ratio 0.75, one-sided P=0.01) for pts with R/R ALL who received InO vs standard chemotherapy (SC). Here we explore the potential association between leukemic molecular profiles and efficacy of InO. Methods: Pts with CD22+ ALL due to receive salvage treatment were randomized to InO or SC; clinical methods and results were published previously (Kantarjian HM, et al. N Engl J Med 2016; PMID: 27292104). Bone marrow samples collected at screening post protocol amendment 2 underwent RNA-sequencing with analysis of gene expression profile, fusions, and genomic alterations. Data cutoff: Jan 4, 2017. Results: Of 326 pts in the INO-VATE intent-to-treat (ITT) population, 91 (InO, 43; SC, 48) had samples evaluable for molecular profiling. Baseline characteristics were similar in the evaluable pts vs the ITT population. Most common (≥5%) ALL subtypes were BCR-ABL1-positive (14%), low hypodiploidy (12%), Philadelphia chromosome (Ph)-like (11%), KMT2A-rearranged (9%), and DUX4-rearranged (6%); 29% (26/91) of pts could not be subtyped due to low blast counts. Most common (≥5%) non-BCR-ABL1 oncogenic fusions detected were IGH-CRLF2 (8%), and KMT2A-AFF1 (6%). Among non-fusion genomic alterations, those involving chromatin modifying genes featured prominently (e.g. KMT2D, 18%; CREBBP, 12%; KMT2C, 11%; ARID2, 9%; ARID1A, 8%; SET2D, 7%; EP300, 6%). Other commonly altered (≥9%) genes included TP53 (14%), NOTCH1 (11%), KRAS (9%), and PAX5 (9%). Similar to the overall findings of INO-VATE, complete remission (CR)/CR with incomplete count recovery (CRi) rates were numerically higher with InO vs SC across ALL subtypes, including but not limited to, low hypodiploidy (67% [2/3] vs 13% [1/8], P=0.15), Ph-like (83% [5/6] vs 0% [0/4], P=0.02), and KMT2A (100% [2/2] vs 17% [1/6], P=0.11), although not in BCR-ABL1-positive (67% [4/6] vs 57% [4/7], P=0.59). Within pts bearing common non-BCR-ABL1 oncogenic fusions, CR/CRi was seen for InO including for IGH-CRLF2 (50% [1/2] vs 20% [1/5] for SC), KMT2A-AFF1 (100% [1/1] vs 25% [1/4] for SC), and IGH-DUX4 (100% [3/3] vs 0% [0/1] for SC). Consistent with a previous report (Yang X, et al. Blood 2019; 134 [Suppl 1]: 3888) CR/CRi rates were high for InO in pts with TP53 alterations (100% [5/5] vs 13% [1/8] for SC, P=0.0047). Conclusions: This retrospective, exploratory analysis of INO-VATE demonstrated potential for benefit with InO for ALL pts across leukemic subtypes, including Ph-like, and for those bearing diverse fusions. Further exploration of Ph-like subtype or TP53 alterations as candidate predictive biomarkers for InO in pts with R/R ALL is warranted. Citation Format: Yaqi Zhao, A Douglas Laird, Kathryn Roberts, Rolla Yafawi, Hagop Kantarjian, Daniel J. DeAngelo, Matthias Stelljes, Michaela Liedtke, Wendy Stock, Nicola Gökbuget, Susan O'Brien, Elias Jabbour, Ryan D. Cassaday, Melanie R. Loyd, Scott Olsen, Geoffrey Neale, Xueli Liu, Erik Vandendries, Charles G. Mullighan, Anjali Advani. Exploration of association of leukemic molecular profile with efficacy in patients (pts) with relapsed/refractory acute lymphoblastic leukemia (ALL) treated with inotuzumab ozogamicin (InO) in the phase 3 INO-VATE trial [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr CT027.

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