Journal of Experimental & Clinical Cancer Research (Sep 2024)

Cell therapy using ex vivo reprogrammed macrophages enhances antitumor immune responses in melanoma

  • Satish kumar Reddy Noonepalle,
  • Maria Gracia-Hernandez,
  • Nima Aghdam,
  • Michael Berrigan,
  • Hawa Coulibaly,
  • Xintang Li,
  • Christian Zevallos-Delgado,
  • Andrew Pletcher,
  • Bryan Weselman,
  • Erica Palmer,
  • Tessa Knox,
  • Eduardo Sotomayor,
  • Katherine B. Chiappinelli,
  • Duncan Wardrop,
  • Anelia Horvath,
  • Brett A. Shook,
  • Norman Lee,
  • Anatoly Dritschilo,
  • Rohan Fernandes,
  • Karthik Musunuri,
  • Maho Shibata,
  • Alejandro Villagra

DOI
https://doi.org/10.1186/s13046-024-03182-w
Journal volume & issue
Vol. 43, no. 1
pp. 1 – 21

Abstract

Read online

Abstract Background Macrophage-based cell therapies have shown modest success in clinical trials, which can be attributed to their phenotypic plasticity, where transplanted macrophages get reprogrammed towards a pro-tumor phenotype. In most tumor types, including melanoma, the balance between antitumor M1-like and tumor-promoting M2-like macrophages is critical in defining the local immune response with a higher M1/M2 ratio favoring antitumor immunity. Therefore, designing novel strategies to increase the M1/M2 ratio in the TME has high clinical significance and benefits macrophage-based cell therapies. Methods In this study, we reprogrammed antitumor and proinflammatory macrophages ex-vivo with HDAC6 inhibitors (HDAC6i). We administered the reprogrammed macrophages intratumorally as an adoptive cell therapy (ACT) in the syngeneic SM1 murine melanoma model and patient-derived xenograft bearing NSG-SGM3 humanized mouse models. We phenotyped the tumor-infiltrated immune cells by flow cytometry and histological analysis of tumor sections for macrophage markers. We performed bulk RNA-seq profiling of murine bone marrow-derived macrophages treated with vehicle or HDAC6i and single-cell RNA-seq profiling of SM1 tumor-infiltrated immune cells to determine the effect of intratumor macrophage ACT on the tumor microenvironment (TME). We further analyzed the single-cell data to identify key cell-cell interactions and trajectory analysis to determine the fate of tumor-associated macrophages post-ACT. Results Macrophage ACT resulted in diminished tumor growth in both mouse models. We also demonstrated that HDAC6 inhibition in macrophages suppressed the polarization toward tumor-promoting phenotype by attenuating STAT3-mediated M2 reprogramming. Two weeks post-transplantation, ACT macrophages were viable, and inhibition of HDAC6 rendered intratumor transplanted M1 macrophages resistant to repolarization towards protumor M2 phenotype in-vivo. Further characterization of tumors by flow cytometry, single-cell transcriptomics, and single-cell secretome analyses revealed a significant enrichment of antitumor M1-like macrophages, resulting in increased M1/M2 ratio and infiltration of CD8 effector T-cells. Computational analysis of single-cell RNA-seq data for cell-cell interactions and trajectory analyses indicated activation of monocytes and T-cells in the TME. Conclusions In summary, for the first time, we demonstrated the potential of reprogramming macrophages ex-vivo with HDAC6 inhibitors as a viable macrophage cell therapy to treat solid tumors.