The Induction of Dendritic Cells with IFN-alpha and GM-CSF from Bone Marrow Mononuclear Cells from Patients with Chronic Myeloid Leukemia.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4873-4873
Author(s):  
Shuier Zhen ◽  
Jie Jin ◽  
Xiangmin Tong

Abstract Chronic myelogenous leukemia (CML) is a malignant myeloproliferative disease arising from the clonal expansion of a stem cell with the typical Philadelphia (Ph) chromosome cytogenetic abnormality. IFN-a has been proven to be effective for patients in the chronic phase of myelogenous leukemia (CML), yet the mechanisms of the antitumor action of these cytokines are still a matter of debate. Dendritc cells (DCs) are potent antigen-presenting cells that prime effective T-cell response aginst tumour antigens. Recent studies have shown that IFN-a can exert a variety of effects on dendritic cells (DCs), which may play an important role in the induction of an antitumor immunity. Human DCs can be generated in vitro from peripheral blood(PB) monocytes or from CD34+ haematopoietic precursor cells in culture medium containing human granulocyte macrophage-colony stimulating factor (GM-CSF), IL-4 and some other cytokines. Previous studies have shown a new effective protocol for the generation of human DCs from unseparated BM aspirate cells with excellent functional capacity of antigen uptake and of stimulating naive and memory T cell responses superior to that of DCs from peripheral blood(PB) monocytes. We, therefore, explored whether treatment with IFN-a may influence the CML bone marrow mononuclear cells(BMMNCs) derived DCs in vitro. Treatment BMMNCs of 12 patients with CML in chronic phase with IFN-a+rhGM-CSF(IFN-a-DC) generated DCs with more mature phenotype properties expressing higher of CD80,CD86,HLA-DR,CD83 compared to the CML- BMMNCs treated with rhGM-CSF+IL-4(IL-4-DC). And in parallel with phenotypes, IFN-a-DC also showed more effective than IL-4-DC in eliciting an allogeneic mixed lymphocyte reaction by MTT assay. FISH confirmed the DCs of both groups were leukemic origin. These findings demonstrate that IFN-a promotes the differentiation/maturation of DCs derived from BMMNCs of patients with CML in vitro, these studies also broaden the clinical scope of IFN-a as a promising agent in the immunotherapy of CML.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 213-213 ◽  
Author(s):  
Margaret V. Ragni ◽  
Wenhu Wu ◽  
Xiaoyan Liang ◽  
Lina Lu

Abstract Inhibitor formation is a severe complication of hemophilia, occurring in up to 25% and associated with poor response to factor replacement, uncontrolled bleeding, and high morbidity. Preventing inhibitor formation is, thus, a major goal of hemophilia management. The role of dendritic cells (DC) in regulating immune response has been increasingly recognized: immature DC (imDC) induce T regulatory cells in vitro and promote Ag-specific tolerance in vivo. We, therefore, studied the role of imDC propagated from bone marrow with GM-CSF + TGFβ to prevent inhibitor formation in the hemophilia A murine model. Following tail vein injection of recombinant F.VIII (Advate, Baxter) 2.5 U (0.2 μg) on days 0, 2, and 4 in hemophilia A exon 16 KO C57Bl/6 mice, anti-VIII antibodies were detected by semi-quantitative APTT (scored 1-4), peaking on day 6. On rechallenge with F.VIII 2.5 U on days 12, 14, and 16, anti-VIII was detected, peaking on day 17. Anti-VIII production was associated with high level splenic T cell proliferation in response to F.VIII stimulation in vitro, measured by 3H-thymidine incorporation in mixed lymphocyte reaction (MLR). By contrast, there was no antibody formation in F.VIII-treated Wt C57Bl/6 mice: the latter was associated with low T cell response to F.VIII in vitro. Functionally immature DC (imDC) were propagated from the bone marrow of hemophilia A mice with GM-CSF (4ng/ml) and TGFβ (0.2ng/ml). For comparison, functionally mature dendritic cells (mDC) were propagated with GM-CSF (4ng/ml) and IL-4 (1000U/ml).The former (imDC) demonstrated deficient NF-kB binding activity in nuclear protein as detected by gel shifting assay and expressed low level of costimulatory molecules CD80, CD86; by contrast, the latter (mDC) demonstrated enhanced NF-kB binding activity and high levels of co-stimulatory molecules. Administration of 2x106 F.VIII-pulsed imDC (20U/ml x 24h) 7 days before F.VIII dosing on days 0, 2, and 4, led to reduction in inhibitor formation on day 6 (score 1.6 vs. 2.3 in control group) which was further reduced on day 8 (score 1.0 vs. 2.0 in control group). The inhibitor could not be detected on day 8 in 2 of 4 mice pretreated with F.VIII-pulsed imDC. By contrast, high levels of inhibitor were detected in mice pretreated with F.VIII-pulsed mDC (score 3.3). Rechallenge with F.VIII on day 10 in imDC-treated mice resulted in no increase in the reduced or absent anti-VIII effect on day 12. Splenic T cells (CD3+) from the imDC-pretreated mice showed lower proliferative capacity when restimulated in vitro with F.VIII, suggesting that imDC induced F.VIII unresponsiveness. These studies show that FVIII-pulsed imDC reduce the intensity of inhibitor formation, and suggest the potential role of modified DC in preventing or reducing F.VIII inhibitor formation.


PEDIATRICS ◽  
1986 ◽  
Vol 77 (3) ◽  
pp. 330-335
Author(s):  
Kevin Shannon ◽  
Gabriel Nunez ◽  
Lois W. Dow ◽  
Arthur G. Weinberg ◽  
Yuichi Sato ◽  
...  

Cells from three children with juvenile chronic myelogenous leukemia were studied using culture in semisolid media, cytogenetic analysis, and surface staining with the monocyte-specific monoclonal antibodies 61D3 and 63D3. The percentage of bone marrow mononuclear cells that were 61D3- and 63D3-positive was markedly increased in all three patients. Bone marrow and peripheral blood mononuclear cells exhibited exceptionally bright immunofluorescence with these antibodies. The presence of monocyte-specific antigens on the surface of juvenile chronic myelogenous leukemia cells suggests that they are derived from a precursor with monocytic characteristics. A specific chromosomal abnormality (47, XY+21) was present in fresh bone marrow cells from one patient; in contrast, 50 metaphases from phytohemagglutinin-stimulated peripheral blood contained a normal karyotype. The chromosomal abnormality was also identified in myeloid colonies grown in vitro from this patient. Granulocytic elements were demonstrated in tissue sections and in cultured myeloid colonies from this child. Our data suggest that malignant transformation in juvenile chronic myelogenous leukemia involves a myeloid progenitor population capable of differentiation in vitro to cells with monocytic or granulocytic characteristics.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4874-4874
Author(s):  
Jie Jin ◽  
Shuier Zheng ◽  
Xiangmin Tong

Abstract Mature dendritc cells(DCs) are potent antigen-presenting cells that prime effective T-cell response aginst tumour antigens. However, in the tumour microenviroment the differentiation and maturation of DCs is always suppressed by many factors, such as VEGF, IL-10,TGF-βet al. Though DC derived from CML patients have been shown to process and present endogenous BCR-ABL to CD4+ T cells, the function of DC may be abnormal in CML, which have been shown differences in cytoskeletal organisation and impaired migration, and a reduced capacity to capture and present nonleukaemic antigen, and less effective than normal DC in eliciting an allogeneic mixed lymphocyte reaction. STI571(trade name Glivec or Gleevec) is a specific inhibitor of Abl tyrosine kinase and has been a first- line agent for the treatment of chronic myelogenous leukemia (CML). Previous studies have found that defective blood dendritic cells in chronic myeloid leukemia correlate with high plasmatic VEGF and can be partially normalized by STI571. We, therefore, explored whether treatment with STI571 may influence the CML -derived DC in vitro. Treatment bone marrow mononuclear cells(BMMNCs) of 11 patients with CML in chronic phase with STI571+rhGM-CSF+IL-4(STI571-DC) generated DCs with more mature phenotype properties expressing higher of CD80,CD86,HLA-DR,CD83 compared to the CML- BMMNCs treated with rhGM-CSF+IL-4(IL-4-DC). And in parallel with phenotypes, STI571-DC also showed more effective than IL-DC in eliciting an allogeneic mixed lymphocyte reaction by MTT assay. FISH confirmed the DCs of both groups were leukemic origin. To explore whether the positive effects of STI571 on CML-DC may attributed to negative factors of DC differentiation/maturation suppressed by STI571, media were tested for VEGF concentrations by using the sandwich enzyme-linked immunosorbent assay (ELISA). As a result, the concentration of VEGF was dramatically reduced in STI571-DC. These findings demonstrate that treatment with STI571 promotes maturation and enhances the antigen-presenting capabilities of CML- BMMNCs derived DCs, these studies also offer a more comprehensive understanding of the role of STI571 in the immunotherapy of CML.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3196-3196
Author(s):  
Anne Barra ◽  
Lucie Brault ◽  
Ali G Turhan ◽  
Lydia Roy ◽  
François Guilhot ◽  
...  

Abstract It is currently established that the immune system plays a critical role in the control of chronic myelogenous leukemia (CML). Several cell populations including T cells, dendritic cells (Dc) and NK cells have been shown to exhibit potential anti-leukemic activities. We have previously shown that plasmacytoid dendritic cells (pDc) differentiation and IFN-a production was impaired along the chronic phase (CP) of CML and that the Tyrosine- Kinase (TK) activity of BCR-ABL was responsible for this impairment. Moreover, patients in complete cytogenetic remission (CCyR) after imatinib mesylate (IM) or IFN-a therapy present a partial restoration of the pDc compartment and IFN-a production. Some recent results (Esashi et al., Immunity, 2008) indicate that, in mice, STAT-5 but not STAT-3, block the expression of IRF-8, a transcription factor required for the differentiation of pDc and that the GM-CSF-induced STAT5 activation interferes with pDc differentiation. As STAT3 and STAT5 proteins are important targets of BCR-ABL TK activity, we postulate that their recruitment could be responsible for the CML pDc impairment and could be involved in the persistent pDc dysfunction in CR patients. We decided to investigate the effect of BCR-ABL or GM-CSF on STAT-3, STAT-5 and IRF8 status ex vivo, on CMN of patients who have achieved sustained CCyR and major molecular response (MMR) after IFN-a therapy and in whom treatment has been discontinued (n=8) compared to CMN of healthy subjects (HS) (n=5). The same investigations were also performed in vitro with the model of pDc differentiation of CD34+ haematopoietic progenitors from CP patients and HSs, using the combination of Stem Cell Factor, Flt3-Ligand and Thrombopoietin,. Flow cytometry analysis showed that the homeostasis of the IFN-a production was altered in CCyR patients : pDc (determined by the coexpression of CD303, CD123 and CD4), represent 34.6 ± 14.9 % of the IFN-a producing cells after stimulation by Influenza virus and intracellular detection of IFN-a, whereas in healthy donors (HD) IFN-a producing cells were principally pDc (72.1 ± 20.5 %of IFN-a+ cells, p<0.05 in comparison with the CCyR patients group). The IRF-8 expression analysed by flow cytometry was also deregulated as low levels of IRF8 were detected in these cells when compared with pDc of HD (based on mean fluorescence analysis of the IRF-8 staining). Moreover, higher levels of P-STAT5 were observed in CMN and pDc from CCyR patients (65.1 ± 18 % of pDc was P-STAT5 +) than from HD (0.75 ± 0.66 % of pDc are P-STAT5 +, p<0.01 in comparison with the CR patients group). The involvement of Human STAT5 in the repression of pDc differentiation was confirmed in our in vitro model. We showed that GM-CSF blocked the pDc differentiation of normal CD34+ cells and enhanced the P-STAT5 levels. We showed higher levels of P-STAT5 along differentiation of CD34+ cells of CML patients in CP, in correlation with the lack of differentiation and maturation of pDc. Altogether, our results suggest that in Human STAT5 is a target for BCR-ABL and GM-CSF and is responsible for the defect of pDc differentiation and IFN-a production. After CCyR, the pDc defect is only partially restored, principally because a STAT5 recruitment persists, possibly induced by soluble factors produced by the residual disease.


2021 ◽  
Vol 9 (Suppl 1) ◽  
pp. A3.2-A4
Author(s):  
J Grün ◽  
I Piseddu ◽  
C Perleberg ◽  
N Röhrle ◽  
S Endres ◽  
...  

BackgroundUnmethylated CpG-DNA is a potent ligand for the endosomal Toll-like-receptor-9, important for the immune activation to pathogen-associated molecules.1 CpG and other TLR-ligands show effective immunotherapeutic capacities in cancer treatment by inducing an antitumorigenic immunity.2 They are able to reduce tumor progression by reduction of intratumoral secretion of the immunoregulating chemokine CCL223 and subsequent recruitment of immunosuppressive regulatory T cells (Treg), which express CCR4 the only so far known receptor for CCL22.4 Our recent work has shown that CCL22 secretion by dendritic cells (DC) in the lymph node, mediates tolerance by inducing DC-Treg contacts.5 Indeed, in the absence of CCL22, immune responses to vaccination were stronger and resulted in tumor rejection.6 Therefore, we are aiming to investigate the effects of TLR-ligands on systemic CCL22 levels, elucidating all involved mechanisms to identify new targets for cancer immunotherapy.Materials and MethodsT, B and CD11c+ DCs of wildtype (wt) and RAG1-/- mice were isolated from splenocytes by magnetic-activated cell sorting for in vitro assays. Different co-cultures were incubated with CpG and GM-CSF, known as an CCL22 inducer.5 For in vivo experiments, wt mice were treated with CpG, R484 or poly(I:C) alone and in combination with GM-CSF. CCL22-levels in a number of organs were analyzed.ResultsAnalyzing the different immune cell compartments in vitro, we found that DCs in whole splenocytes secrete CCL22 during culture while DC cultured alone showed no CCL22 secretion. When treated with CpG, CCL22-levels were reduced in splenocytes, while it was induced in DC culture alone. The same results were seen when RAG splenocytes, that lack functional B and T cells, were cultured with CpG. CpG treated B cells were able to suppress CCL22 secretion by DC unlike T cells alone. Co-cultures of T and B cells treated with CpG, however, induced the strongest CCL22 suppression in DC. In vivo, we could show that all TLR ligands tested reduced CCL22 in a number of organs significantly. Furthermore, CpG showed the strongest suppression of CCL22 even in the presence of the CCL22 inducer GM-CSF.5ConclusionsWe could show that B cells with T cells mediate CCL22 suppression by TLR ligands. The fact that CpG was able to reduce CCL22 levels even in the presence of the inducer GM-CSF demonstrates the potent CCL22 suppressive capacity of TLR ligands.ReferencesO’Neill LA, et al. The history of toll-like receptors – redefining innate immunity. Nat Rev Immunol 2013;13(6):453–60.Rothenfusser S, et al. Recent advances in immunostimulatory CpG oligonucleotides. Curr Opin Mol Ther 2003;5(2):98–106.Wang S, et al. Intratumoral injection of a CpG oligonucleotide reverts resistance to PD-1 blockade by expanding multifunctional CD8+ T cells. Proc Natl Acad Sci U S A 2016;113(46): E7240–E7249.Rapp M, et al. CCL22 controls immunity by promoting regulatory T cell communication with dendritic cells in lymph nodes. J Exp Med 2019;216(5):1170–1181.Piseddu I, et al. Constitutive expression of CCL22 is mediated by T cell-derived GM-CSF. J Immunol 2020;205(8):2056–2065.Anz D, et al. Suppression of intratumoral CCL22 by type i interferon inhibits migration of regulatory T cells and blocks cancer progression. Cancer Res 2015;75(21):4483–93.Disclosure InformationJ. Grün: None. I. Piseddu: None. C. Perleberg: None. N. Röhrle: None. S. Endres: None. D. Anz: None.


2020 ◽  
Author(s):  
Melisa Gorosito Serrán ◽  
Facundo Fiocca Vernengo ◽  
Laura Almada ◽  
Cristian G Beccaria ◽  
Pablo F Canete ◽  
...  

ABSTRACTDuring infections with protozoan parasites or virus, T cell immunosuppression is generated simultaneously with a high B cell activation. Here, we show that in T. cruzi infection, all plasmablasts detected had higher surface expression of PD-L1, than other mononuclear cells. PD-L1hi plasmablasts were induced in vivo in an antigen-specific manner and required help from Bcl-6+CD4+T cells. PD-L1hi expression was not a characteristic of all antibody-secreting cells since plasma cells found during the chronic phase of infection express PD-L1 but at lower levels. PD-L1hi plasmablasts were also present in mice infected with Plasmodium or with lymphocytic choriomeningitis virus, but not in mice with autoimmune disorders or immunized with T cell-dependent antigens. PD-L1hi plasmablasts suppressed T cell response, via PD-L1, in vitro and in vivo. Thus, this study reveals that extrafollicular PD-L1hi plasmablasts, which precede the germinal center (CG) response, are a suppressive population in infections that may influence T cell response.Brief summaryPathogens develop different strategies to settle in the host. We identified a plasmablats population induced by pathogens in acute infections which suppress T cell response.


The Lancet ◽  
2002 ◽  
Vol 360 (9350) ◽  
pp. 2083 ◽  
Author(s):  
Shoichi Inaba ◽  
Kensuke Egashira ◽  
Kimihiro Komori

Sign in / Sign up

Export Citation Format

Share Document