Your browser doesn't support javascript.
loading
Cell therapy using ex vivo reprogrammed macrophages enhances antitumor immune responses in melanoma.
Noonepalle, Satish Kumar Reddy; Gracia-Hernandez, Maria; Aghdam, Nima; Berrigan, Michael; Coulibaly, Hawa; Li, Xintang; Zevallos-Delgado, Christian; Pletcher, Andrew; Weselman, Bryan; Palmer, Erica; Knox, Tessa; Sotomayor, Eduardo; Chiappinelli, Katherine B; Wardrop, Duncan; Horvath, Anelia; Shook, Brett A; Lee, Norman; Dritschilo, Anatoly; Fernandes, Rohan; Musunuri, Karthik; Shibata, Maho; Villagra, Alejandro.
Affiliation
  • Noonepalle SKR; Lombardi Comprehensive Cancer Center, Georgetown University, 3970 Reservoir Road, NW, E416 Research Bldg, Washington, DC, 20057, USA.
  • Gracia-Hernandez M; The George Washington University, Washington, DC, USA.
  • Aghdam N; Lombardi Comprehensive Cancer Center, Georgetown University, 3970 Reservoir Road, NW, E416 Research Bldg, Washington, DC, 20057, USA.
  • Berrigan M; The George Washington University, Washington, DC, USA.
  • Coulibaly H; Lombardi Comprehensive Cancer Center, Georgetown University, 3970 Reservoir Road, NW, E416 Research Bldg, Washington, DC, 20057, USA.
  • Li X; Lombardi Comprehensive Cancer Center, Georgetown University, 3970 Reservoir Road, NW, E416 Research Bldg, Washington, DC, 20057, USA.
  • Zevallos-Delgado C; The George Washington University, Washington, DC, USA.
  • Pletcher A; The George Washington University, Washington, DC, USA.
  • Weselman B; Lombardi Comprehensive Cancer Center, Georgetown University, 3970 Reservoir Road, NW, E416 Research Bldg, Washington, DC, 20057, USA.
  • Palmer E; The George Washington University, Washington, DC, USA.
  • Knox T; The George Washington University, Washington, DC, USA.
  • Sotomayor E; Tampa General Hospital, Tampa, FL, USA.
  • Chiappinelli KB; The George Washington University, Washington, DC, USA.
  • Wardrop D; University of Illinois at Chicago, Chicago, IL, USA.
  • Horvath A; The George Washington University, Washington, DC, USA.
  • Shook BA; The George Washington University, Washington, DC, USA.
  • Lee N; The George Washington University, Washington, DC, USA.
  • Dritschilo A; Lombardi Comprehensive Cancer Center, Georgetown University, 3970 Reservoir Road, NW, E416 Research Bldg, Washington, DC, 20057, USA.
  • Fernandes R; The George Washington University, Washington, DC, USA.
  • Musunuri K; Avstera Therapeutics, Malvern, PA, USA.
  • Shibata M; The George Washington University, Washington, DC, USA.
  • Villagra A; Lombardi Comprehensive Cancer Center, Georgetown University, 3970 Reservoir Road, NW, E416 Research Bldg, Washington, DC, 20057, USA. Alejandro.villagra@georgetown.edu.
J Exp Clin Cancer Res ; 43(1): 263, 2024 Sep 14.
Article in En | MEDLINE | ID: mdl-39272209
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.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Macrophages / Melanoma Limits: Animals / Humans Language: En Journal: J Exp Clin Cancer Res Year: 2024 Document type: Article Affiliation country: Country of publication:

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Macrophages / Melanoma Limits: Animals / Humans Language: En Journal: J Exp Clin Cancer Res Year: 2024 Document type: Article Affiliation country: Country of publication: