Vai al contenuto principale

Dott.ssa Silvia Carloni

  • Dottorato: 26° ciclo
  • Matricola: 743965

Tesi di dottorato

INTRODUCTION

      Acute myeloid leukemia (AML) is the most common form of acute leukemia in adults (Siegel et al., 2012)and current treatments remain unsatisfactory. Serious infections, resistance to therapy and relapse are the main causes of mortality among  patients (Pagano et al., 2012; Estey, 2012; Romani et al., 2011). Modulation of the immune system can help to control AML, as occurs in hematopoietic stem cell transplantation (Schmid et al., 2007) and a large percentage of patients who are probably destined to die from severe infections, could be saved by a better understanding of immunosuppressive mechanisms working in the disease and by their complete restoration. TH17 cells and their respective cytokines play a key role in the inflammatory response and autoimmune disorders (Ouyang et al., 2008; Luger et al., 2008; Monteleone et al., 2012), and also coordinate the defense against bacterial and fungal infections (gastrointestinal tract, skin, airways and lung) (Khader et al., 2009). Moreover, the remarkable epigenetic plasticity of TH17 cells in tumors has recently been demonstrated, together with their ability to transdifferentiate into T helper 1-like cells (secreting IFN-g and endowed with tumor suppressor activity) (Muranski and Restifo, 2009; Zou and Restifo, 2010) or into T regulatory (Treg)-like cells (secreting IL-10, probably with immunosuppressive functions and tumor promoter activity) (Martin et al., 2012; Zielinski et al., 2012; Ye et al., 2011; Ustun et al., 2011; Koenen et al., 2008). Furthermore, TH17 cells are long-lived cells with a stem-like molecular signature (Muranski et al., 2011) and so their suppressor or promoter capacity against tumors may be powerful and more durable (Muranski et al., 2008). However, their real role in several tumor types is controversial (Muranski et al., 2009; Zou et al., 2010; Martin et al., 2012). In particular, their involvement in hematological malignancies, especially AML, and in the infections (especially fungal) to which AML patients are susceptible, remains to be defined (Wu et al., 2009; Tian et al., 2013; Ersvaer et al., 2010).

 

METHODS

Blood samples and PBMCs collection. After obtaining the patient’s informed consent and the approval of  the local ethics committee, in accordance with the Declaration of Helsinki, samples of peripheral blood (15-20 ml) were collected from 30 newly diagnosed AML patients without infections before any treatment was started and from 30 age-matched (+/- 10 years) healthy volunteers (HV). AML patients were diagnosed according to the French American-British (FAB) classification system (Bennett et al., 1976). Patient and HV characteristics are reported in Table 1. Blood samples were collected in sterile EDTA tubes and mononuclear cells (PBMCs) were separated by density gradient centrifugation using Lymphosep (Biowest) and frozen in 90% heat inactivated fetal bovine serum (FBS) (PAA) and 10% dimethylsulfoxide (Sigma Aldrich).

CD4+ cell isolation and culture. PBMCs were thawed and human CD4+ T helper cells were isolated by negative depletion of CD8+, CD14+, CD15+, CD16+, CD19+, CD36+, CD56+, CD123+, TCR y/d and CD235a+, using the CD4+ T cell isolation kit (Miltenyi Biotec). Cells were cultured in RPMI 1640 medium (PAA) supplemented with 10% heat inactivated FBS, 2 mM L-glutamine (Euroclone), penicillin (100 U/ml) and streptomycin (100 μg/ml) (PAA). CD4+ cells were primed for 24 h at 37°C with IL-6 (25 μg/ml) (Miltenyi Biotec) or TGF-b (10 ng/ml) (Abcam) or a combination of IL-6 and TGF-b. T cells were then incubated for 5 h at 37°C with phorbol 12-myristate-13-acetate (PMA, 50 ng/ml) and ionomycin (1 μg/ml) (Invitrogen) in the presence of GolgiStop Protein Transport Inhibitor (BD Pharmingen). An unstimulated control prepared by incubating CD4+ cells with GolgiStop Protein Transport Inhibitor only was included for each experiment.

Immunophenotypic analysis of T cells. After stimulation, cells were fixed and permeabilized with Cytofix/Cytoperm (BD Biosciences) after which then immunophenotyping for intracellular IFN-γ, IL-4 and IL-17A expression was performed using the human TH1/TH2/TH17 phenotyping kit (BD Pharmingen) following the manufacturer's protocol.For Treg analysis, naïve PBMCs were stained with       anti-human FITC CD4 (0.6 μg/ml, clone SK3; BD Biosciences) and anti-human            APC-Cy7 CD25 (2.5 μg/ml, clone M-A251; BD Biosciences) for 10 min at 4°C in the dark. After incubation, cells were fixed and permeabilized and then stained with anti-human APC FoxP3 (1:11, clone 3G3; Miltenyi Biotec) for 30 min at 4°C in the dark. Appropriate isotype controls were included for each sample.

Cytokine secretion analysis. Stimulated CD4+ cells were washed with cold PBS containing 0.5% (v/v) bovine serum albumin (BSA) (Sigma Aldrich) and 2 mM of EDTA and analyzed using human IL-17 and IL-10 secretion assay - detection kits (Miltenyi Biotec). Briefly, cells were stained with IL-17 and IL-10 catch reagents for    5 min on ice, incubated for 45 min at 37°C to allow cytokine secretion and then with anti-human PE IL-17A, anti-human APC IL-10 and anti-human FITC CD4 for 10 min on ice, according to the manufacturer’s instructions. Samples were washed and suspended for flow cytometric analysis.

Direct and indirect allogeneic co-cultures. For direct co-cultures, leukemic CD33+ blast cells magnetically isolated with CD33 MicroBeads (Miltenyi Biotec) from          15 AML patients and allogeneic CD4+ T cells obtained from 15 HV as previously reported were co-seeded in 1:1, 1:5 and 1:10 ratios. For indirect co-cultures, purified CD4+cells were seeded in the bottom part of the 6-well plates of transwell cell culture system (pore size 0.4 μm; Costar Corp.), whereas CD33+ cells were seeded in the corresponding transwell cell culture inserts. In addition, each cell type was seeded individually in 6-well plates for single culture as control. All samples were cultured in complete medium and stimulated as previously described. At the end of stimulation,     T cell immunophenotypic and cytokine secretion analysis was performed.

Immunofluorescence assays. T cells (1 x 106) isolated from healthy donors were stained with Cell Tracker Blue CMAC (7-amino-4 chloromethylcoumarin) (Molecular Probes) for 45 min at 37°C following the manufacturer’s instruction. CD4+ cells were co-seeded with an equal number of CD33+ cells isolated from AML patients for 24 h at 37°C and then fixed for 15 min at room temperature with 2% formaldehyde in PBS. Cells were permeabilized and stained with 1:40 Phalloidin Alexa Fluor 594 (Life Technologies) in Inside Perm (Miltenyi Biotec) for 20 min at room temperature.After labeling, specimens were washed and mounted on Prolong Gold Antifade Reagent (Invitrogen). The specificity of staining was optimized using isotype-control antibody. Fluorescence was acquired with a Evos FL Microscope (AMG, Washington, USA) equipped with Light Cube for DAPI, GFP and RFP.

T cell activation with C. Albicans and isolation of IL-17-secreting cells. CD4+ cells (2.5 x 106) were stimulated for 24 h at 37°C with 1 μg/ml of C. Albicans peptides (JPT, Berlin, Germany). During the last 5 h of incubation, cells were maintained in the presence of GolgiStop Protein Transport Inhibitor (BD Pharmingen).Samples were centrifuged at 4°C, incubated with 2mM of EDTA in PBS for 10 min at 37°C, washed with 0.5% BSA and 0.1% sodium azide in PBS. Cells were then depleted of IL-17- secreting cells using the IL-17 Secretion Assay-Cell Enrichment and Detection Kit (Miltenyi Biotec). The IL-17 specific catch reagent was attached to the cell surface as previously described, after which cells were labeled with anti-human PE IL-17A and stained with anti-PE microbeads. IL-17-secreting cells were separated through two consecutive column runs, according to the manufacturer’s instructions.Negative fraction was cultured for a further 24 h in complete medium supplemented with 1μg/ml of C. Albicans peptides and then analyzed for intracellular IFN-γ expression using the human TH1/TH2/TH17 phenotyping kit (BD Pharmingen). A sample stimulated with C. Albicans for 48 h without depletion of IL-17-secreting cells was added as control.

Flow cytometry. Flow cytometric analysis were performed using a FACSCanto flow cytometer (Becton Dickinson) equipped with 488 nm (blue) and 633 (red) lasers and 50,000 events were recorded for each sample. The acquisition and analysis gates were set on lymphocytes based on forward (FSC) and side scatter (SSC) properties of cells. FSC and SSC were set in a linear scale. For more extensive analysis, gates were set on CD4+ T cell subsets. Flow cytometry data were analyzed with Diva Software (Becton Dickinson).

Statistical analysis. Data were summarized by descriptive statistics (mean ± standard deviation for continued variables and frequency and percentage for categorical variables). Statistical analyses were carried out using the paired and unpaired two-tailed Student’s t tests and confirmed with the non parametric Wilcoxon signed-rank test. P values < 0.05 were considered as significant.


RESULTS AND DISCUSSION

      In order to examine the relationship between TH17 cells, leukemic cells, infections and immunocompetence, we first focused on the frequency of CD4+ T cells (TH1, TH2, Tregs and TH17 cells) in the peripheral blood of 30 newly diagnosed, untreated AML patients and 30 sex- and age-matched  (+/- 10 years) healthy volunteers (HV). AML patients were diagnosed according to the French American-British classification system (Bennett et al., 1976)and characterized by karyotype and molecular biology mutations (Table 1). Given the controversies over the different TH17 polarization methods (Ghoreschi et al., 2010; Acosta-Rodriguez et al., 2007; Yang et al., 2008; Ganjalikhani et al., 2011), we stimulated CD4+ cells isolated through a negative immunomagnetic system in serum-containing media with IL-6, TGF-β or  IL-6+TGF-β. As no significant differences were observed (Fig. S1), TH1, TH2 and TH17 analyses were performed with IL-6 alone, thereby reducing the risk of the TGF-b-mediated induction or inhibition of other cytokines (Ghoreschi et al., 2010; Acosta-Rodriguez et al., 2007; Yang et al., 2008; Ganjalikhani et al., 2011). The frequency of circulating Treg cells was assessed in unstimulated PBMCs.

      Flow cytometric analysis revealed that the frequencies of T CD4+ populations were altered in AML patients (Fig.1, A-C). In particular, CD4+ cells showed a decreased capacity to produce IFN-g and IL-4, whereas the frequency of IL-17+ cells was higher in AML patients than in HV. In addition, the percentage of circulating CD4+ CD25highFoxP3+ Tregs was higher in AML patients (Fig. 1 D-E; Fig. S2). All the observed differences were statistically significant. However, in view of the singular infectious susceptibility of these patients and to the physiological protective role of TH17 cells, we also decided to evaluate the ability of TH17 cells to simultaneously produce or secrete other cytokines, focusing on IL-10 and IFN-γ(Zielinski et al., 2012; Ye et al., 2011). Intriguingly, we observed a strong, statistically significant increase in the frequency of CD4+ IL-17A/IL-10-secreting cells in AML patients compared to HV (0.6±0.5 and 0.075±0.6 respectively; Fig. 1, F-G), whereas the percentage of  IL-17A+/IFN-γ+ cells remained unchanged (Fig. S3). No correlation was observed with specific karyotype or molecular biology mutations. Having identified an alteration that probably leads to an immunosuppressed state, we proceeded to confirm this and to evaluate its effect on the immune response. We stimulated CD4+ cells from HV and untreated AML patients with an infectious fungal antigen from Candida Albicans (C. Albicans). The samples were then depleted of IL-17-secreting cells, cultured for a further 24 h with C. Albicans peptides and analyzed for intracellular IFN-γ expression. Other samples from HV and AML patients stimulated with C. Albicans as above but not depleted of IL-17-secreting cells were used as control. A significant reduction in the frequency of IFN-g positive cells was observed in control samples from AML patients compared to HV. Interestingly, after removal of the IL-17-secreting cells, the IFN-g production has returned elevated in patients (Fig. 2, A-B). Conversely, in HV, the depletion of IL-17-secreting cells did not induce significant changes in the production of IFN-g.

      Finally, to investigate the role of leukemic cells in such changes, we performed direct and indirect co-cultures of CD4+ T cells obtained from 15 HV and CD33+ blast cells magnetically isolated from 15 AML patients. After 24 h, T cell immunophenotypic and cytokine secretion analyses were performed. A cytokine pattern similar to that found in AML patients was observed in CD33+ and CD4+ co-cultured directly at a    1:1 ratio, with a reduction in IFN-g and IL-4-positive cells and a strong increase in the percentage of CD4+ IL-17A/IL-10-secreting cells (Fig. 3 A). Moreover, cells stained with fluorescent phalloidin revealed a cellular distribution of F-actin and, in particular, cytoskeletal remodeling at the point of contact between blast and T cells, suggesting a role of cell-cell contact in the changes (Fig. 3 B). However, the observed cytokine alterations were also present when blasts were physically separated from CD4+ cells by a membrane (Fig. 3 A). In both co-culture methods, the described alterations were not observed at CD33+ and CD4+ ratios of 1:5 and 1:10 (Fig. S4), indicating that, in vitro, the number of CD33+ circulating blasts played an important role in inducing morphological and functional alteration in T helper cells. Notably, when patient CD4+ T cells were depleted of CD33+ blasts, the former regained a capacity to produce IFN-g and IL-4 similar to that of HV (Fig. 3, C-D). Furthermore, IL-17/IL-10 producing cells significantly decreased after CD33+ removal, indicating that blasts play a part in the control of this population.

      In conclusion, in addition to the known pathologic mechanisms of AML (Curti et al., 2010; Zhou et al., 2010; Szczepanski et al., 2009; Mothy et al., 2001) and to the changes in Tregs (Ustun et al., 2011)which differentiate into FOXP3+ IL-17A-secreting cells under specific stimuli (Koenen et al., 2008), our results identified a novel process in this disease that induces a substantial percentage of TH17 cells to change into IL-17/IL-10-secreting cells with immunosuppressive activity. We also observed a dual mechanism of action mediated by contact and soluble factors that can be held responsible for these changes. Furthermore, the immunosuppressive action of TH17 cells observed in our study would seem to lead to an alteration in the physiological role of these cells in AML patients, favoring infections and probably promoting immune escape of the disease. Further studies are needed, nevertheless the long-lasting activity and plasticity of TH17 cells and the opportunity to restore and to change them in tumor suppressor cells, makes TH17 an optimal new target to enhance immunotherapy in AML.

 

 

REFERENCES

Acosta-Rodriguez, E.V., Napolitani, G., Lanzavecchia, A., and Sallusto, F. 2007. Interleukins 1beta and 6 but not transforming growth factor-beta are essential for the differentiation of interleukin 17-producing human T helper cells. Nat. Immunol. 8:942-949. http://doi:10.1038/ni1496

Bennett, J.M., Catovsky, D., Daniel, M.T., Flandrin, G., Galton, D.A., Gralnick, H.R., and Sultan, C.1976. Proposals for the classification of the acute leukaemias. French-American-British (FAB) co-operative group. Br. J. Haematol. 33:       451-458.

Curti, A., Trabanelli, S., Onofri, C., Aluigi, M., Salvestrini, V., Ocadlikova, D., Evangelisti, C., Rutella, S., De Cristofaro, R., Ottaviani, E., Baccarani, M., and Lemoli, R.M. 2010.Indoleamine 2,3-dioxygenase-expressing leukemic dendritic cells impair a leukemia-specific immune response by inducing potent T regulatory cells. Haematologica  95:2022-2030. http://doi:10.3324/ haematol. 2010.025924

Ersvaer, E., Liseth, K., Skavland, J., Gjertsen, B.T., and Bruserud, Ø. 2010. Intensive chemotherapy for acute myeloid leukemia differentially affects circulating TC1, TH1, TH17 and TREG cells. BMC. Immunol. 11:38. http://doi:10.1186/         1471-2172-11-38

Estey, E.H. 2013. Acute myeloid leukemia: 2013 update on risk-stratification and management. Am. J. Hematol. 88:318-327. http://doi:10.1002/ajh.23404

Ganjalikhani Hakemi, M., Ghaedi, K., Andalib, A., Hosseini, M., and Rezaei, A. Optimization of human Th17 cell differentiation in vitro: evaluating different polarizing factors. In Vitro Cell. Dev. Biol. Anim. 47:581-592. http://doi:10.1007/s11626-011-9444-1

Ghoreschi, K., Laurence, A., Yang, X.P., Tato, C.M., McGeachy, M.J., Konkel, J.E., Ramos, H.L., Wei, L., Davidson, T.S., Bouladoux, N., Grainger, J.R., Chen, Q., Kanno, Y., Watford, W.T., Sun, H.W., Eberl, G., Shevach, E.M., Belkaid, Y., Cua, D.J., Chen, W., and O'Shea, J.J. 2010. Generation of pathogenic T(H)17 cells in the absence of TGF-β signalling. Nature 467:967-971. http://doi:10.1038/nature09447

Khader, S.A., Gaffen, S.L., and Kolls, J.K. 2009. Th17 cells at the crossroads of innate and adaptive immunity against infectious diseases at the mucosa. Mucosal. Immunol. 2:403-411. http://doi:10.1038/mi.2009.100.

Koenen, H.J., Smeets, R.L., Vink, P.M., van Rijssen, E., Boots, A.M., and Joosten, I. 2008. Human CD25highFoxp3pos regulatory T cells differentiate into IL-17-producing cells. Blood  112:2340-2352. http://doi:10.1182/blood-2008-01-133967

Luger, D., Silver, P.B., Tang, J., Cua, D., Chen, Z., Iwakura, Y., Bowman, E.P., Sgambellone, N.M., Chan, C.C., and Caspi, R.R. 2008. Either a Th17 or a Th1 effector response can drive autoimmunity: conditions of disease induction affect dominant effector category. J. Exp. Med. 205: 799-810. http://doi:10.1084/jem.20071258

Martin, F., Apetoh, L., and Ghiringhelli, F. 2012. Controversies on the role of Th17 in cancer: a TGF-β-dependent immunosuppressive activity? Trends. Mol. Med. 18:742-749. http://doi:10.1016/j.molmed.2012.09.007

Mohty, M., Jarrossay, D., Lafage-Pochitaloff, M., Zandotti, C., Brière, F., de Lamballeri, X.N., Isnardon, D., Sainty, D., Olive, D., and Gaugler, B. 2001.Circulating blood dendritic cells from myeloid leukemia patients display quantitative and cytogenetic abnormalities as well as functional impairment. Blood  98:3750-3756. http://doi:10.1182/blood.V98.13.3750

Monteleone, I., Sarra, M., Pallone, F., and Monteleone, G. 2012. Th17-related cytokines in inflammatory bowel diseases: friends or foes? Curr. Mol. Med. 12:592-597. http:/7doi: 10.2174/156652412800620066

Muranski, P., and Restifo, N.P. 2009. Does IL-17 promote tumor growth? Blood 114:231-232. http://doi:10.1182/blood-2009-04-215541

Muranski, P., Boni, A., Antony, P.A., Cassard, L., Irvine, K.R., Kaiser, A., Paulos, C.M., Palmer, D.C., Touloukian, C.E., Ptak, K., Gattinoni, L., Wrzesinski, C., Hinrichs, C.S., Kerstann, K.W., Feigenbaum, L., Chan, C.C., and Restifo, N.P. Tumor- specific Th17-polarized cells eradicate large established melanoma. Blood  112:362-373. http://doi:10.1182/blood-2007-11-120998

Muranski, P., Borman, Z.A., Kerkar, S.P., Klebanoff, C.A., Ji, Y., Sanchez-Perez, L., Sukumar, M., Reger, R.N., Yu, Z., Kern, S.J., Roychoudhuri, R., Ferreyra, G.A., Shen, W., Durum, S.K., Feigenbaum, L., Palmer, D.C., Antony, P.A., Chan, C.C., Laurence, A., Danner, R.L., Gattinoni, L., and Restifo, N.P. Th17 cells are long lived and retain a stem cell-like molecular signature. Immunity. 35:972-985. http://doi:10.1016/j.immuni.2011.09.019

Ouyang, W.J., Kolls, J.K. and Zheng, Y. 2008.The biological functions of T helper      17 cell effector cytokines in inflammation. Immunity. 28:454-467. http://doi: 10.1016/j.immuni.2008.03.004

Pagano, L., Caira, M., Rossi, G., Tumbarello, M., Fanci, R., Garzia, M.G., Vianelli, N., Filardi, N., De Fabritiis, P., Beltrame, A., Musso, M., Piccin, A., Cuneo, A., Cattaneo, C., Aloisi, T., Riva, M., Rossi, G., Salvadori, U., Brugiatelli, M., Sannicolò, S., Morselli, M., Bonini, A., Viale, P., Nosari, A., Aversa, F. and Hema e-Chart Group, Italy. 2012. A prospective survey of febrile events in hematological malignancies. Ann. Hematol. 91:767-774. http://doi: 10.1007/s00277-011-1373-2

Romani, L. 2011. Immunity to fungal infections. Nat. Rev. Immunol. 11:275-288. http://doi:  10.1038/nri2939. 

Schmid, C., Labopin, M., Nagler, A., Bornhäuser, M., Finke, J., Fassas, A., Volin, L., Gürman, G., Maertens, J., Bordigoni, P., Holler, E., Ehninger, G., Polge, E., Gorin, N.C., Kolb, H.J., Rocha, V. and EBMT Acute Leukemia Working Party. 2007.Donor lymphocyte infusion in the treatment of first hematological relapse after allogeneic stem-cell transplantation in adults with acute myeloid leukemia:   a retrospective risk factors analysis and comparison with other strategies by the EBMT Acute Leukemia Working Party. J. Clin. Oncol. 25:4938-4945. http://doi:10.1200/JCO.2007.11.6053

Siegel, R., Naishadham, D., and Jemal, A. 2012. Cancer statistics, 2012. CA. Cancer J. Clin. 62,10-29. http://doi:10.3322/caac.20138

Szczepanski, M.J., Szajnik, M., Czystowska, M., Mandapathil, M., Strauss, L., Welsh, A., Foon, K.A., Whiteside, T.L., and Boyiadzis, M.2009.Increased frequency and suppression by regulatory T cells in patients with acute myelogenous leukemia. Clin. Cancer Res.15:3325-3332. http://doi: 10.1158/1078-0432.CCR-08-3010

Tian, T., Yu, S., Wang, M., Yuan, C., Zhang, H., Ji, C., and Ma, D. 2013. Aberrant T helper 17 cells and related cytokines in bone marrow microenvironment of patients with acute myeloid leukemia. Clin. Dev. Immunol. 2013:915873. http://doi:10.1155/2013/915873

Ustun, C., Miller, J.S., Munn, D.H., Weisdorf, D.J., and Blazar, B.R. 2011. Regulatory T cells in acute myelogenous leukemia: is it time for immunomodulation? Blood  118:5084-5095.  http://doi:10.1182/blood-2011-07-365817

Wu, C., Wang, S., Wang, F., Chen, Q., Peng, S., Zhang, Y., Qian, J., Jin, J., and Xu, H. 2009. Increased frequencies of T helper type 17 cells in the peripheral blood of patients with acute myeloid leukaemia. Clin. Exp. Immunol. 158:199-204. http://doi: 10.1111/j.1365-2249. 2009.04011.x

Yang, L., Anderson, D.E., Baecher-Allan, C., Hastings, W.D., Bettelli, E., Oukka, M., Kuchroo, V.K., and Hafler, D.A. 2008. IL-21 and TGF-beta are required for differentiation of human T(H)17 cells. Nature 454:350-352. http://doi:10.1038/nature07021

Ye, J., Su, X., Hsueh, E.C., Zhang, Y., Koenig, J.M., Hoft, D.F., and Peng, G. 2011. Human tumor- infiltrating Th17 cells have the capacity to differentiate into IFN-g + and FOXP3 + T cells with potent suppressive function. Eur. J. Immunol. 41:936-951. http://doi: 10.1002/eji.201040682

Zhou, Q., Munger, M.E., Highfill, S.L., Tolar, J., Weigel, B.J., Riddle, M., Sharpe, A.H., Vallera, D.A., Azuma, M., Levine, B.L., June, C.H., Murphy, W.J., Munn, D.H., and Blazar, B.R. 2010.Program death-1 signaling and regulatory T cells collaborate to resist the function of adoptively transferred cytotoxic T lymphocytes in advanced acute myeloid leukemia. Blood  116:2484-2493. http://doi:10.1182/blood-2010-03-275446

Zielinski, C.E., Mele, F., Aschenbrenner, D., Jarrossay, D., Ronchi, F., Gattorno, M., Monticelli, S., Lanzavecchia, A., and Sallusto, F. 2012. Pathogen-induced human TH17 cells produce IFN-γ or IL-10 and are regulated by IL-1β. Nature 484:     514-518. http://doi:10.1038/nature10957

Zou, W., and Restifo, N.P. 2010. T(H)17 cells in tumour immunity and immunotherapy. Nat. Rev. Immunol. 10:248-256. http://doi:10.1038/nri2742

 

Attività di ricerca

 Fabbri F, Zoli W, Carloni S, Ulivi P, Arienti C, Brigliadori G, Montanari M, Tesei A, Silvestrini R, Amadori D. Activity of Different Anthracycline Formulations in Hormone-Refractory Prostate Cancer Cell Lines: Role of Golgi Apparatus. J Cell Physiol. 2011 Feb 1.

Tesei A, Brigliadori G, Carloni S, Fabbri F, Ulivi P, Arienti C, Sparatore A, Del Soldato P, Pasini A, Amadori D, Silvestrini R, Zoli W. Organosulfur derivatives of the HDAC inhibitor valproic acid sensitize human lung cancer cell lines to apoptosis and to cisplatin cytotoxicity. J Cell Physiol. 2012 Oct.

Ibrahim T, Mercatali L, Sacanna E, Tesei A, Carloni S, Ulivi P, Liverani C, Fabbri F, Zanoni M, Zoli W, Amadori D. Inhibition of breast cancer cell proliferation in repeated and non-repeated treatment with zoledronic acid. Cancer Cell Int. 2012 Nov 22.  

Montanari M, Fabbri F, Rondini E, Frassineti GL, Mattioli R, Carloni S, Scarpi E, Zoli W, Amadori D, Cruciani G. Phase II trial of non-pegylated liposomal doxorubicin and low-dose prednisone in second-line chemotherapy for hormone-refractory prostate cancer. Tumori. 2012 Nov;98(6):696-701.

Arienti C, Tesei A, Carloni S, Ulivi P, Romeo A, Ghigi G, Menghi E, Sarnelli A, Parisi E, Silvestrini R, Zoli W. SLUG silencing increases radiosensitivity of melanoma cells in vitro. Cell Oncol. 2013 Apr.

Fabbri F, Carloni S, Zoli W, Ulivi P, Gallerani G, Fici P, Chiadini E, Passardi A, Frassineti GL, Ragazzini A, Amadori D. Detection and recovery of circulating colon cancer cells using a dielectrophoresis-based device: KRAS mutation status in pure CTCs. Cancer Lett. 2013 Feb 15.

Lucchesi A, Carloni S, Cangini D, Frassineti GL, Casadei Gardini A. Acute Oxaliplatin-Induced Thrombotic Thrombocytopenic Purpura: A Case Report and Results From a Cytoflourimetric Assay of Platelet Fibrinogen Receptor. J Clin Oncol. 2013 Apr 22.

Casadei Gardini A, Aquilina M, Oboldi D, Lucchesi A, Carloni S, Tenti E, Burgio MA, Amadori D, Frassineti GL. Separate episodes of capillary leak syndrome and pulmonary hypertension after adjuvant gemcitabine and three years later after nab-paclitaxel for metastatic disease. BMC Cancer. 2013 Nov 12.

Arienti C, Zoli W, Pignatta S, Carloni S, Paganelli G, Ulivi P, Romeo A, Menghi E, Sarnelli A, Medri L, Polico R, Silvestrini R, Tesei A. Efficacy of different sequences of radio- and chemotherapy in experimental models of human melanoma. J Cell Physiol. 2014 Oct;229(10):1548-56.

Guerrini A, Tesei A, Ferroni C, Paganelli G, Zamagni A, Carloni S, Di Donato M, Castoria G, Leonetti C, Porru M, De Cesare M, Zaffaroni N, Beretta GL, Del Rio A, Varchi G. A New Avenue toward Androgen Receptor Pan-antagonists: C2 Sterically Hindered Substitution of Hydroxy-propanamides. J Med Chem. 2014 Aug 28. 

Preliminary investigation of circulating NSCLC cells using dielectrophoresis-based instrumentation Silvia Carloni, Francesco Fabbri, Paola Ulivi, Dino Amadori, Wainer Zoli AACR 102nd Annual Meeting April 2-6 2011 (Orlando, 2-6 Aprile 2011)


Isolation of living circulating tumor cells using dielectrophoresis-based instrumentation S. Carloni, F. Fabbri, G. Brigliadori, D. Amadori, W. Zoli Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Meldola, Italy 39th Congress of the International Society of Oncology and Biomarkers (Firenze, 15-19 Ottobre 2011)

Isolation of living circulating tumor cells using dielectrophoresis-based instrumentation S. Carloni, F. Fanini, G. Gallerani, E. Bandini, S. Pignatta, F. Fabbri, G. Brigliadori, D. Amadori, W. Zoli Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Meldola, Italy Corso “CTCs from invention to diagnostics” (San Diego, 1 Febbraio 2012)

Convegno “Cellule tumorali circolanti: la rivoluzione microscopica” (Roma, 26 Novembre 2010 )

  • AACR 102nd Annual Meeting April 2-6 2011 (Orlando, 2-6 Aprile 2011) con presentazione di un poster dal titolo “Preliminary investigation of circulating NSCLC cells using dielectrophoresis-based instrumentation”
  • Corso di aggiornamento di Radioprotezione (Meldola, Settembre 2011)
  • Corso “Lavorare in sicurezza” (Formazione a distanza, Settembre 2011)
  • Corso di analisi di database (Università di Torino, Settembre 2011)
  • 39th Congress of the International Society of Oncology and Biomarkers (Firenze, 15-19 Ottobre 2011) con presentazione di un poster dal titolo “Isolation of living circulating tumor cells using dielectrophoresis-based instrumentation”
  • Corso NIBIT di formazione sulla immunobioterapia dei tumori umani (Meldola, 18 Novembre 11)
  • Corso “CTCs from invention to diagnostics” (San Diego, 1 Febbraio 2012) con presentazione di un poster dal titolo “Isolation of living circulating tumor cells using dielectrophoresis-based instrumentation”
  • Congresso “Cell Culture 2012” (San Diego, 2-3 Febbraio 2012)
  • Corso di “Statistica per l’analisi di dati di interesse biologico” (Torino, 27-28 Febbraio 2012)
  • Congresso "Immunoterapia: Nouva frontiera della terapia Oncologica" (Meldola, 15 gennaio 2013)
  • Meeting "La sanità in prima pagina in Italia e nel mondo" (Meldola, 11 Marzo 2013)
  • Congresso "Indolent Lymphomas" (Meldola, 6 Maggio 2013)
  • Congresso: "Terapie innovative nel melanoma e nel carcinoma baso-cellulare" (Meldola, 17 Settembre 2013)
  • Convegno "Farmacovigilanza in oncologia e in ematologia: esperienze a confronto" (Meldola, 27 Settembre 2013)
  • Workshop "La ricerca in rete per l'Europa" (Meldola, 1 Ottobre 2013)
  • 4° Workshop Nazionale SIES "Ematologia Traslazionale" (Bologna, 14-15 Novembre 2013)
  • Corso Nazionale SISET (Roma, 28-29 Novembre 2013)
  • 3 International Conference "Translational Research in Oncology" (Forlì, 6-9 Maggio 2014)
  • Workshop: Progettazione europea - Horizon 2020: opportunità e scenari applicativi (Meldola, 13 Maggio 2014)
  • XIII Congresso nazionale SIES (Rimini, 15-17 Ottobre 2014)

Meet the Professor: Marco Danova. “Lo studio delle cellule tumorali circolanti: aspetti metodologici e possibili ricadute cliniche” (Meldola 06 Marzo 2012)

Meet the Professor: Khalid Shah. “Targeting primary and metastatic brain tumors with therapeutic stem cells” (Meldola 20 Marzo 2012)

Meet the Professor: Luigi Naldi. "Tossicità dei farmaci oncologici mirati: incidenza, pattern clinic, trattamento, prevenzione, modelli biologici" (Meldola 22 Gennaio 2013)

Meet the Professor: Angelo L. Vescovi. "Cellule staminali cerebrali: un nuovo modello per lo studio dei glioblastomi umani e recenti sviluppi pre-clinici e clinici nei tumori solidi" (Meldola 02 Febbraio 2013)

Meet the Professor: Gaetano Guglielmi: La valutazione scientifica degli IRCCS e il sistema di valutazione dei progetti del bando di ricerca finalizzata" (Meldola 5 Maggio 2013)

Meet the Professor: Massimo Federico. "La ricerca clinica sui linfomi in Italia: più di ieri e, speriamo, meno di domani" (9 Aprile 2013)

Meet the Professor: Sergio Siragusa. Microangiopatie trombotiche: dalla clinica alla terapia" (Meldola, 14 Maggio 2013)

Meet the Professor: Daniela Maria Cardinale. "Cardioncologia: una nuova specialità medica" (Meldola, 26 Novembre 2013)

Meet the Professor: Maria Paola Costi. "An integrated Mass Spectometry proteomic-bioinformatic approach to study biomarkers in oncology" (Meldola 21 Febbraio 2014)

 

Meet the Professor: Valerio De Stefano: "Patogenesi e trattamento delle complicanze trombotiche nelle neoplasie mieloproliferative Ph-negative" (Meldola, 1 Luglio 2014)

2011, 1 anno presso Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Meldola, Forlì 

Ultimo aggiornamento: 13/05/2015 10:45
Non cliccare qui!