Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2025 Aug 26;9(16):4217-4231.
doi: 10.1182/bloodadvances.2025016432.

IGSF9-targeted therapy inhibits the progression of acute myeloid leukemia

Affiliations

IGSF9-targeted therapy inhibits the progression of acute myeloid leukemia

Lijun Hui et al. Blood Adv. .

Abstract

Previously, we reported that targeting immunoglobulin superfamily member 9 (IGSF9) could enhance antitumor T-cell activity and sensitivity to anti-PD-1 immunotherapy, although the detailed mechanism remains unclear. In this study, we find that, similar to the regulation of PD-L1 expression, interferon gamma (IFN-γ) also induces the expression of IGSF9 in acute myeloid leukemia (AML). The small interfering RNA specifically targeting JAK1 and a STAT1 inhibitor blocking IFN-γ signal pathway significantly inhibit the expression of IGSF9 and PD-L1. As a tumor-specific immune checkpoint molecule, IGSF9 plays a significant role in promoting tumor escape. The induction of both PD-L1 and IGSF9 by IFN-γ in the tumor microenvironment explains why IGSF9 is highly expressed in tumors and tumor-infiltrating immune cells. This induction also underpins the strong synergistic effects when combining anti-IGSF9 and anti-PD-1 therapies. Additionally, IGSF9 also mediates the extramedullary infiltration of AML cells, which can be inhibited by depletion of IGSF9 or anti-IGSF9. The binding epitopes of anti-IGSF9 are located within the immunoglobulin G2 and fibronectin type-III-2 domains of IGSF9. Based on these findings, we develop an antibody-drug conjugate (ADC) targeting IGSF9 (anti-IGSF9-linker-DXd). This ADC exhibits 99.7% purity, and primarily exists in monomeric form, demonstrating excellent homogeneity (drug-to-antibody ratio, 8-10) and specificity. Anti-IGSF9-linker-DXd effectively kills IGSF9-positive tumor cells and exhibits a potent bystander effect. In vivo, anti-IGSF9-linker-DXd almost completely eliminates early- and mid-stage tumors and significantly inhibits the progression of advanced tumors. In summary, our findings underscore the potential of IGSF9 as a novel therapeutic target for AML treatment, highlighting its role in disease progression and the efficacy of targeted therapies.

PubMed Disclaimer

Conflict of interest statement

Conflict-of-interest disclosure: Anti-IGSF9 and anti–IGSF9-linker-DXd have been patented (no. ZL202211000509.7 and 202311326074.X), which covers anti-IGSF9, anti–IGSF9-linker-DXd, and their application for treating tumors; and Z.L., Juan Zhang, Z.Z., S.J., and H.W. are listed as inventors. The remaining authors declare no competing financial interests.

Figures

None
Graphical abstract
Figure 1.
Figure 1.
IGSF9 level is detected by flow cytometry. (A-E) IGSF9 levels are detected by flow cytometry in T cells, B cells, and myeloid cells from healthy donors (n = 10), CD33+/CD13+ leukemia cells (n = 27), CD34+/CD117+ leukemia stem cells (n = 15), umbilical cord blood (n = 5), and AML cell lines including THP-1, MV4-11, Molm13, and U937 cells. APC, allophycocyanin; FITC, fluorescein isothiocyanate; PBMC, peripheral blood mononuclear cell.
Figure 2.
Figure 2.
IGSF9 is induced by IFNγ. (A) Mouse bone marrow cells are differentiated into macrophages. (B) Mouse IGSF9 expression is assessed in bone marrow cells, M0, M1, and M2 macrophages. (C-F) IFN-γ induces the expression of IGSF9 and PD-L1 in THP-1 and MV4-11 cells. APC, allophycocyanin; BMNC, bone marrow mononuclear cells.
Figure 3.
Figure 3.
Silencing JAK1 inhibits the expressions of IGSF9 and PD-L1. (A) The siRNA targeting JAK1 blocks the IFN-γ signal pathway, (B-E) leading to reduced levels of IGSF9 and PD-L1 when JAK1 is silenced by siRNA. si, small interfering.
Figure 4.
Figure 4.
The characterization of anti–IGSF9-DXd. (A) The basic structure of anti–IGSF9-DXd. (B) The purity of anti–IGSF9-DXd is assessed by size exclusion chromatography, and (C) the DAR values are determined by hydrophobic interaction chromatography. (D,E) The binding activities and serum half-life of anti-IGSF9 and anti–IGSF9-DXd are determined.
Figure 5.
Figure 5.
Internalization and cytotoxic assay. (A) The antigen-antibody complexes are endocytosed, and anti–IGSF9-DXd is fluorescently labeled with green, and anti–LAMP-1 is fluorescently labeled with red. (B-E) The different concentrations of anti–IGSF9-DXd are used to treat THP-1-IGSF9-WT, -KO, MV4-11, and U937 cells. (F-H) The cleaved PARP, pCHK1, and pH2A are detected by western blotting in THP-1 and MV4-11 cells. Ab, antibody; Con, concentration.
Figure 6.
Figure 6.
Anti–IGSF9-DXd treats the early and mid-stage AML models. About 1 × 106 MV4-11-luc cells are injected into NSG mice (n = 10). Treatment with anti-IGSF9 as a control and anti–IGSF9-DXd is initiated the following day and administered every 5 days thereafter. (A) Disease progression is monitored by bioluminescence imaging. (B) The GFP+ cell percentages in bone marrow, spleen and lungs are detected by flow cytometry, and (C) the tumors in all organs are shown by bioluminescence imaging. About 1 × 106 THP-1-luc cells are injected into NSG mice (n = 10), (D) and anti-IGSF9 or anti–IGSF9-DXd are administered on days 7 and 14, and (E) the survival curves are drawn.
Figure 7.
Figure 7.
Anti–IGSF9-DXd–treated the advanced AML models. A total of 1 × 106 THP-1-luc cells are injected into NSG mice via the tail vein. (A) At 15 days after injection administration, treatment is initiated using isotype control, anti-IGSF9, DXd, and anti–IGSF9-DXd, and administered every 4 days. (B,C) The bioluminescence imaging is used to detect disease progression, and the survival curve is drawn. (D) H&E staining is used to observe the tumors in lungs and liver, and tissue structure and cell morphology of kidney, heart, and brain.

Similar articles

References

    1. Eckardt JN, Stölzel F, Kunadt D, et al. Molecular profiling and clinical implications of patients with acute myeloid leukemia and extramedullary manifestations. J Hematol Oncol. 2022;15(1) - PMC - PubMed
    1. Byrd JC, Mrózek K, Dodge RK, et al. Cancer and Leukemia Group B (CALGB 8461) Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute myeloid leukemia: results from Cancer and Leukemia Group B (CALGB 8461) Blood. 2002;100(13):4325–4336. - PubMed
    1. Yang LX, Zhang CT, Yang MY, et al. C1Q labels a highly aggressive macrophage-like leukemia population indicating extramedullary infiltration and relapse. Blood. 2023;141(7):766–786. - PMC - PubMed
    1. Ganzel C, Manola J, Douer D, et al. Extramedullary disease in adult acute myeloid leukemia is common but lacks independent significance: analysis of patients in ECOG-ACRIN Cancer Research Group Trials, 1980-2008 [published correction appears in J Clin Oncol. 2017;35(2):263] J Clin Oncol. 2016;34(29):3544–3553. - PMC - PubMed
    1. Xu J, Zhang W, Yan XJ, et al. DNMT3A mutation leads to leukemic extramedullary infiltration mediated by TWIST1. J Hematol Oncol. 2016;9(1) - PMC - PubMed

MeSH terms