scholarly journals Adoptive transfer of immature myeloid cells lacking NF‐κB p50 (p50‐IMC) impedes the growth of MHC‐matched high‐risk neuroblastoma

2021 ◽  
Author(s):  
Cheng Cui ◽  
Theresa Barberi ◽  
Rahul Suresh ◽  
Alan D. Friedman
2020 ◽  
Vol 8 (1) ◽  
pp. e000244 ◽  
Author(s):  
Rahul Suresh ◽  
David J Barakat ◽  
Theresa Barberi ◽  
Lei Zheng ◽  
Elizabeth Jaffee ◽  
...  

BackgroundMacrophages and dendritic cells lacking the transcription factor nuclear factor kappa B p50 are skewed toward a proinflammatory phenotype, with increased cytokine expression and enhanced T cell activation; additionally, murine melanoma, fibrosarcoma, colon carcinoma, and glioblastoma grow slower in p50−/−mice. We therefore evaluated the efficacy of p50-negative immature myeloid cells (p50-IMCs) adoptively transferred into tumor-bearing hosts. Immature cells were used to maximize tumor localization, and pretreatment with 5-fluorouracil (5FU) was examined due to its potential to impair marrow production of myeloid cells, to target tumor myeloid cells and to release tumor neoantigens.MethodsWild-type (WT)-IMC or p50-IMC were generated by culturing lineage-negative marrow cells from WT or p50−/−mice in media containing thrombopoietin, stem cell factor and Flt3 ligand for 6 days followed by monocyte colony-stimulating factor for 1 day on ultralow attachment plates. Mice inoculated with Hi-Myc prostate cancer (PCa) cells or K-RasG12Dpancreatic ductal carcinoma (PDC)-luciferase cells received 5FU followed 5 days later by three doses of 107immature myeloid cells (IMC) every 3–4 days.ResultsPCa cells grew slower in p50−/−mice, and absence of host p50 prolonged the survival of mice inoculated orthotopically with PDC cells. IMC from Cytomegalovirus (CMV)-luciferase mice localized to tumor, nodes, spleen, marrow, and lung. 5FU followed by p50-IMC slowed PCa and PDC tumor growth, ~3-fold on average, in contrast to 5FU followed by WT-IMC, 5FU alone or p50-IMC alone. Slowed tumor growth was evident for 93% of PCa but only 53% of PDC tumors; we therefore focused on PCa for additional IMC analyses. In PCa, p50-IMC matured into F4/80+macrophages, as well as CD11b+F4/80−CD11c+conventional dendritic cells (cDCs). In both tumor and draining lymph nodes, p50-IMC generated more macrophages and cDCs than WT-IMC. Activated tumor CD8+T cells were increased fivefold by p50-IMC compared with WT-IMC, and antibody-mediated CD8+T cell depletion obviated slower tumor growth induced by 5FU followed by p50-IMC.Conclusions5FU followed by p50-IMC slows the growth of murine prostate and pancreatic carcinoma and depends on CD8+T cell activation. Deletion of p50 in patient-derived marrow CD34+cells and subsequent production of IMC for adoptive transfer may contribute to the therapy of these and additional cancers.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A103-A103
Author(s):  
Cheng Cui ◽  
Theresa Barberi ◽  
Rahul Suresh ◽  
Ludovic Zimmerlin ◽  
Elias Zambidis ◽  
...  

BackgroundHigh-risk neuroblastomas, including those with MYCN gene amplification, harbor abundant myeloid cells that suppress anti-tumor immunity and favor tumor growth. Macrophages lacking the inhibitory NF-κB p50 subunit adopt an M1-polarized, T-cell-activating phenotype. Multiple cancers grow slower in mice lacking p50, and colon cancer grows slower in mice lacking p50 specifically in myeloid cells.MethodsTumor growth was monitored in mice inoculated subcutaneously with 9464D cells. To generate p50-IMC from p50-/-mice, marrow cells were lineage-depleted and then expanded in media with SCF, FL, and TPO, followed by transfer to M-CSF for one day. To generate p50-IMC from wild-type mice, marrow cells were nucleofected with a p50 sgRNA:Cas9 complex, followed by expansion in serum-free media. Mice received 5-fluorouracil on day 27, followed five days later by three doses of 1E7 p50-IMC via tail vein every three to four days. PD-1 antibody was administered twice weekly for four doses, starting on day 32. Azacytidine was administered 5 days per week alternating with ITF-2357 5 days per week, again starting on day 32. CD4 and CD8 antibody was given twice weekly starting on day 21. Tumor T cells were analyzed by flow cytometry.ResultsWe have now found that murine 9464D neuroblastoma cells, expressing high levels of exogenous human MYCN, also grow slower in syngeneic C57BL/6 (B6) p50(f/f);Lys-Cre mice, lacking p50 in macrophages and neutrophils, compared with p50(f/f) littermates. Slowed tumor growth in p50(f/f);Lys-Cre mice was associated with increased total and activated tumor CD4+ and CD8+ T cells, and depletion of both CD4+ and CD8+ T cells accelerated tumor growth. PD-1 checkpoint blockade or DNA methyltransferase and histone deacetylase inhibition further slowed tumor growth in p50(f/f);Lys-Cre mice. In addition, adoptive transfer of p50-IMC, generated either from the bone marrow of p50-/- B6 mice or via nucleofection of a p50 sgRNA:Cas9 complex into wild-type B6 hematopoietic progenitors, also slowed neuroblastoma tumor growth, following a dose of myelo-depleting 5-fluorouracil.ConclusionsThese findings using a neuroblastoma model further validate the utility of targeting myeloid NF-κB p50 as an immunotherapy strategy for cancer therapy and demonstrate activity of p50-IMC generated by gene editing of syngeneic marrow cells, a cell product relevant to clinical translation. We have also developed means to efficiently gene edit p50 in human marrow CD34 cells, and have demonstrated the feasibility of generating p50-IMC from human induced pluripotent stem cells. We are currently evaluating the efficacy of these gene-edited human cells against human neuroblastoma in immune-deficient mice.


Cells ◽  
2021 ◽  
Vol 10 (5) ◽  
pp. 968
Author(s):  
Tyler J. Wildes ◽  
Bayli DiVita Dean ◽  
Catherine T. Flores

Our understanding of the relationship between the immune system and cancers has undergone significant discovery recently. Immunotherapy with T cell therapies and checkpoint blockade has meaningfully changed the oncology landscape. While remarkable clinical advances in adaptive immunity are occurring, modulation of innate immunity has proven more difficult. The myeloid compartment, including macrophages, neutrophils, and dendritic cells, has a significant impact on the persistence or elimination of tumors. Myeloid cells, specifically in the tumor microenvironment, have direct contact with tumor tissue and coordinate with tumor-reactive T cells to either stimulate or antagonize cancer immunity. However, the myeloid compartment comprises a broad array of cells in various stages of development. In addition, hematopoietic stem and progenitor cells at various stages of myelopoiesis in distant sites undergo significant modulation by tumors. Understanding how tumors exert their influence on myeloid progenitors is critical to making clinically meaningful improvements in these pathways. Therefore, this review will cover recent developments in our understanding of how solid tumors modulate myelopoiesis to promote the formation of pro-tumor immature myeloid cells. Then, it will cover some of the potential avenues for capitalizing on these mechanisms to generate antitumor immunity.


2002 ◽  
Vol 51 (6) ◽  
pp. 293-298 ◽  
Author(s):  
Sergei Kusmartsev ◽  
Dmitry Gabrilovich

2017 ◽  
Vol 7 (2) ◽  
pp. 1227-1230
Author(s):  
Meghashree Vishwanath ◽  
Purnima S Rao ◽  
Muktha R Pai

Myeloid sarcoma is a rare tumour composed of immature myeloid cells. Its occurrence in childhood is rare. Only a few cases of intraoral myeloid sarcoma have been reported in literature. We present a case of myeloid sarcoma with simultaneous involvement of intraoral soft tissues and lymph node. Here, importance is given to suspect this frequently misdiagnosed disease. 


2018 ◽  
Vol 218 (1) ◽  
pp. S21
Author(s):  
Ofer Fainaru ◽  
Ola Gutzeit ◽  
Rivka Hertz ◽  
Yuval Ginsberg ◽  
Ron Beloosesky ◽  
...  

2012 ◽  
Vol 27 (1) ◽  
pp. 151-162 ◽  
Author(s):  
Lothar C. Dieterich ◽  
Petter Schiller ◽  
Hua Huang ◽  
Eric F. Wawrousek ◽  
Angelica Loskog ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document