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
. 2023 Jun 28;12(1):2230669.
doi: 10.1080/2162402X.2023.2230669. eCollection 2023.

Short-term immune-checkpoint inhibition partially rescues perturbed bone marrow hematopoiesis in mismatch-repair deficient tumors

Affiliations

Short-term immune-checkpoint inhibition partially rescues perturbed bone marrow hematopoiesis in mismatch-repair deficient tumors

Paula Krone et al. Oncoimmunology. .

Abstract

Wide-spread cancer-related immunosuppression often curtails immune-mediated antitumoral responses. Immune-checkpoint inhibitors (ICIs) have become a state-of-the-art treatment modality for mismatch repair-deficient (dMMR) tumors. Still, the impact of ICI-treatment on bone marrow perturbations is largely unknown. Using anti-PD1 and anti-LAG-3 ICI treatments, we here investigated the effect of bone marrow hematopoiesis in tumor-bearing Msh2loxP/loxP;TgTg(Vil1-cre) mice. The OS under anti-PD1 antibody treatment was 7.0 weeks (vs. 3.3 weeks and 5.0 weeks, control and isotype, respectively). In the anti-LAG-3 antibody group, OS was 13.3 weeks and thus even longer than in the anti-PD1 group (p = 0.13). Both ICIs induced a stable disease and reduced circulating and splenic regulatory T cells. In the bone marrow, a perturbed hematopoiesis was identified in tumor-bearing control mice, which was partially rescued by ICI treatment. In particular, B cell precursors and innate lymphoid progenitors were significantly increased upon anti-LAG-3 therapy to levels seen in tumor-free control mice. Additional normalizing effects of ICI treatment were observed for lin-c-Kit+IRF8+ hematopoietic stem cells, which function as a "master" negative regulator of the formation of polymorphonuclear-myeloid-derived suppressor cell generation. Accompanying immunofluorescence on the TME revealed significantly reduced numbers of CD206+F4/80+ and CD163+ tumor-associated M2 macrophages and CD11b+Gr1+ myeloid-derived suppressor cells especially upon anti-LAG-3 treatment. This study confirms the perturbed hematopoiesis in solid cancer. Anti-LAG-3 treatment partially restores normal hematopoiesis. The interference of anti-LAG-3 with suppressor cell populations in otherwise inaccessible niches renders this ICI very promising for subsequent clinical application.

Keywords: Hematopoietic progenitor cell; hypermutated tumor; immunotherapy; tumor microenvironment.

PubMed Disclaimer

Conflict of interest statement

No potential conflict of interest was reported by the author(s).

Figures

None
Graphical abstract
Figure 1.
Figure 1.
Workflow, 18F-FDG PET/CT imaging in vivo, and overall survival of Msh2loxP/loxP;TgTg(Vil1-cre) mice treated with ICIs.
Figure 2.
Figure 2.
Spectral flow cytometry of the peripheral blood.
Figure 3.
Figure 3.
Spectral flow cytometry and gene expression analysis of spleens.
Figure 4.
Figure 4.
Characterization of the TME using spectral flow cytometry and immunofluorescence.
Figure 5.
Figure 5.
Gene expression analysis in residual tumors and IRF5 detection by tumor immunofluorescence.
Figure 6.
Figure 6.
Altered murine haematopoiesis during tumor development.
Figure 7.
Figure 7.
Impact of ICI treatment on murine haematopoiesis.

References

    1. Seth S, Ager A, Arends MJ, Frayling IM.. Lynch syndrome - cancer pathways, heterogeneity and immune escape. J Pathol. 2018;246(2):129–14. doi:10.1002/PATH.5139. - DOI - PubMed
    1. Lim SM, Choi JG, Cho SH, Kang EJ, Hwang IG, Yun T, Choi JW, Lee KW, Chang H, Kim JH, et al. Investigating the feasibility of targeted next-generation sequencing to guide the treatment of head and neck squamous cell carcinoma. Cancer Res Treat. 2019;51(1):300–312. doi:10.4143/crt.2018.012. - DOI - PMC - PubMed
    1. Chalabi M, Fanchi LF, Dijkstra KK, Van den Berg JG, Aalbers AG, Sikorska K, Lopez-Yurda M, Grootscholten C, Beets GL, Snaebjornsson P, et al. Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers. Nat Med. 2020;26(4):566–576. 264. 2020. doi:10.1038/s41591-020-0805-8. - DOI - PubMed
    1. Trullas A, Delgado J, Genazzani A, Mueller-Berghaus J, Migali C, Müller-Egert S, Zander H, Enzmann H, Pignatti F.. The EMA assessment of pembrolizumab as monotherapy for the first-line treatment of adult patients with metastatic microsatellite instability-high or mismatch repair deficient colorectal cancer. ESMO Open. 2021;6(3):100145. doi:10.1016/j.esmoop.2021.100145. - DOI - PMC - PubMed
    1. André T, Shiu K-K, Kim TW, Jensen BV, Jensen LH, Punt C, Smith D, Garcia-Carbonero R, Benavides M, Gibbs P, et al. Pembrolizumab in microsatellite-instability–high advanced colorectal cancer. N Engl J Med. 2020;383(23):2207–2218. doi:10.1056/NEJMoa2017699. - DOI - PubMed

Publication types

Substances