Tiziana Schioppa
Queen Mary University of London
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Tiziana Schioppa.
Cancer Research | 2006
Alessandra Saccani; Tiziana Schioppa; Chiara Porta; Subhra K. Biswas; Manuela Nebuloni; Luca Vago; Barbara Bottazzi; Mario P. Colombo; Alberto Mantovani; Antonio Sica
Tumor-associated macrophages (TAM) are a major inflammatory infiltrate in tumors and a major component of the protumor function of inflammation. TAM in established tumors generally have an M2 phenotype with defective production of interleukin-12 (IL-12) and high IL-10. Here, we report that defective responsiveness of TAM from a murine fibrosarcoma and human ovarian carcinoma to M1 activation signals was associated with a massive nuclear localization of the p50 nuclear factor-kappaB (NF-kappaB) inhibitory homodimer. p50 overexpression inhibited IL-12 expression in normal macrophages. TAM isolated from p50(-/-) mice showed normal production of M1 cytokines, associated with reduced growth of transplanted tumors. Bone marrow chimeras showed that p50 inactivation in hematopoietic cells was sufficient to result in reduced tumor growth. Thus, p50 NF-kappaB overexpression accounts for the inability of TAM to mount an effective M1 antitumor response capable of inhibiting tumor growth.
Clinical Cancer Research | 2011
Jermaine Coward; Hagen Kulbe; Probir Chakravarty; David Leader; Vessela Vassileva; D. Andrew Leinster; Richard G. Thompson; Tiziana Schioppa; Jeffery Nemeth; Jessica Vermeulen; Naveena Singh; Norbert Avril; Jeff Cummings; Elton Rexhepaj; Karin Jirström; William M. Gallagher; Donal J. Brennan; Iain A. McNeish; Frances R. Balkwill
Purpose: We investigated whether inhibition of interleukin 6 (IL-6) has therapeutic activity in ovarian cancer via abrogation of a tumor-promoting cytokine network. Experimental Design: We combined preclinical and in silico experiments with a phase 2 clinical trial of the anti-IL-6 antibody siltuximab in patients with platinum-resistant ovarian cancer. Results: Automated immunohistochemistry on tissue microarrays from 221 ovarian cancer cases showed that intensity of IL-6 staining in malignant cells significantly associated with poor prognosis. Treatment of ovarian cancer cells with siltuximab reduced constitutive cytokine and chemokine production and also inhibited IL-6 signaling, tumor growth, the tumor-associated macrophage infiltrate and angiogenesis in IL-6–producing intraperitoneal ovarian cancer xenografts. In the clinical trial, the primary endpoint was response rate as assessed by combined RECIST and CA125 criteria. One patient of eighteen evaluable had a partial response, while seven others had periods of disease stabilization. In patients treated for 6 months, there was a significant decline in plasma levels of IL-6–regulated CCL2, CXCL12, and VEGF. Gene expression levels of factors that were reduced by siltuximab treatment in the patients significantly correlated with high IL-6 pathway gene expression and macrophage markers in microarray analyses of ovarian cancer biopsies. Conclusion: IL-6 stimulates inflammatory cytokine production, tumor angiogenesis, and the tumor macrophage infiltrate in ovarian cancer and these actions can be inhibited by a neutralizing anti-IL-6 antibody in preclinical and clinical studies. Clin Cancer Res; 17(18); 6083–96. ©2011 AACR.
Proceedings of the National Academy of Sciences of the United States of America | 2011
Tiziana Schioppa; R. L. Moore; Richard G. Thompson; Elizabeth C. Rosser; Hagen Kulbe; Sergei A. Nedospasov; Claudia Mauri; Lisa M. Coussens; Frances R. Balkwill
The inflammatory cytokine TNF-α has been recognized as a critical tumor promoter, but the effector cells that mediate its action have not been fully characterized. Because B cells regulate squamous and prostate carcinogenesis, and Tnf−/− mice harbor B-cell defects, we investigated the hypothesis that B cells are important effector cells for TNF-α–mediated promotion of cancer development. Using an adoptive transfer strategy and the 7,12-dimethylbenz[α]anthracene/terephthalic acid (DMBA/TPA) two-stage model of skin carcinogenesis, we found that both B cells and TNF-α are critical for the development of DMBA/TPA-induced papilloma. Transfer of B cells from DMBA/TPA-treated wild-type mice to Tnf−/− mice rescued papilloma development to a wild-type level, a result not observed when B cells from Tnf−/− mice were transferred to Rag2−/− mice or when TNF-α was eliminated selectively in B cells. Resistance to papilloma development in Tnf−/− mice was associated with increased IFN-γ and CD8+ T cells in skin and a significant reduction in IL-10–producing B regulatory cells alongside an increase in IFN-γ–producing CD8+ T cells in the spleen. These data indicate that during DMBA/TPA-induced squamous carcinogenesis TNF-α mediates tumor-promoting activity via regulatory B cells that repress antitumor immunity.
Journal of Immunology | 2010
Kisato Nosaka; Yuetsu Tanaka; Tiziana Schioppa; Frances R. Balkwill; Graham P. Taylor; Charles R. M. Bangham
We recently reported that human T-lymphotropic virus type 1 (HTLV-1) infection is accompanied by a high frequency of CD4+FoxP3+ cells in the circulation. In asymptomatic carriers of HTLV-1 and in patients with HTLV-1–associated inflammatory and malignant diseases, a high FoxP3+ cell frequency correlated with inefficient cytotoxic T cell-mediated killing of HTLV-1–infected cells. In adult T cell leukemia/lymphoma (ATLL), the FoxP3+ population was distinct from the leukemic T cell clones. However, the cause of the increase in FoxP3+ cell frequency in HTLV-1 infection was unknown. In this study, we report that the plasma concentration of the chemokine CCL22 is abnormally high in HTLV-1–infected subjects and that the concentration is strongly correlated with the frequency of FoxP3+ cells, which express the CCL22 receptor CCR4. Further, we show that CCL22 is produced by cells that express the HTLV-1 transactivator protein Tax, and that the increased CCL22 enhances the migration and survival of FoxP3+ cells in vitro. Finally, we show that FoxP3+ cells inhibit the proliferation of ex vivo, autologous leukemic clones from patients with ATLL. We conclude that HTLV-1–induced CCL22 causes the high frequency of FoxP3+ cells observed in HTLV-1 infection; these FoxP3+ cells may both retard the progression of ATLL and HTLV-1–associated inflammatory diseases and contribute to the immune suppression seen in HTLV-1 infection, especially in ATLL.
Journal of Clinical Investigation | 2017
Chiara Berlato; Moddasar N. Khan; Tiziana Schioppa; Richard B. Thompson; Eleni Maniati; Anne Montfort; Maryam Jangani; Monica Canosa; Hagen Kulbe; Urs Beat Hagemann; Alexander R. Duncan; Laura Fletcher; Robert W. Wilkinson; Thomas Powles; Sergio A. Quezada; Frances R. Balkwill
Elevated expression of the chemokine receptor CCR4 in tumors is associated with poor prognosis in several cancers. Here, we have determined that CCR4 was highly expressed in human renal cell carcinoma (RCC) biopsies and observed abnormal levels of CCR4 ligands in RCC patient plasma. An antagonistic anti-CCR4 antibody had antitumor activity in the RENCA mouse model of RCC. CCR4 inhibition did not reduce the proportion of infiltrating leukocytes in the tumor microenvironment but altered the phenotype of myeloid cells, increased NK cell and Th1 cytokine levels, and reduced immature myeloid cell infiltrate and blood chemokine levels. In spite of prominent changes in the myeloid compartment, the anti-CCR4 antibody did not affect RENCA tumors in T cell–deficient mice, and treatment with an anti–class II MHC antibody abrogated its antitumor activity. We concluded that the effects of the anti-CCR4 antibody required the adaptive immune system and CD4+ T cells. Moreover, CCL17-induced IFN-&ggr; production was reduced when Th1-polarized normal CD4+ T cells were exposed to the CCR4 ligand, evidencing the involvement of CCR4 in Th1/Th2 regulation. The anti-CCR4 antibody, alone or in combination with other immune modulators, is a potential treatment approach to human solid cancers with high levels of CCR4-expressing tumor-infiltrating leukocytes and abnormal plasma CCR4 ligand levels.
Cancer Research | 2014
Chiara Berlato; Moddasar N. Kahn; Tiziana Schioppa; Richard B. Thompson; Eleni Maniati; Monica Canosa; Hagen Kulbe; Chris Sheldon; Keith Wreggett; Urs B. Hagemann; Alexander R. Duncan; Laura Fletcher; Robert W. Wilkinson; Thomas Powles; Sergio A. Quezada; Frances R. Balkwill
Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Tumor microenvironments posses complex chemokine networks that contribute to the extent and phenotype of the host infiltrate. Malignant cells may gain functional chemokine receptors, often as a consequence of oncogenic mutations, allowing them to respond to distant chemokine gradients during metastasis. The chemokine receptor CCR4 was highly expressed in human renal cell carcinoma, RCC, biopsies and RCC patient plasma had abnormal levels of CCR4 ligands. However, during pre-clinical evaluation of CCR4 as a target in RCC, we found that both a small molecule CCR4 inhibitor and an anti-CCR4 antagonistic antibody had unexpected and novel anti-tumor activity in the mouse RCC RENCA model. CCR4 antagonists did not reduce the proportion of infiltrating leukocytes in the tumor microenvironment but altered the phenotype of myeloid cells, increased NK cells and Th1 cytokine levels, as well as reducing splenic MDSC infiltrate, and blood chemokine levels. Although the most prominent changes were in the myeloid compartment, anti-tumor activity was lost in athymic mice, supporting a role for the adaptive immune system in CCR4 antagonist action. CCR4 antagonists, alone, or in combination with other immune modulators, may be of interest in human cancers with high levels of tumor CCR4 and abnormal plasma CCR4 ligand levels. Citation Format: Chiara Berlato, Moddasar N. Kahn, Tiziana Schioppa, Richard Thompson, Eleni Maniati, Monica Canosa, Hagen Kulbe, Chris Sheldon, Keith Wreggett, Urs Hagemann, Alexander Duncan, Laura Fletcher, Robert W. Wilkinson, Thomas Powles, Sergio Quezada, Frances Balkwill. Antagonists of the chemokine receptor CCR4 reverse the tumor-promoting microenvironment of renal cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1076. doi:10.1158/1538-7445.AM2014-1076
Blood | 2006
Subhra K. Biswas; Lisa Gangi; Saki Paul; Tiziana Schioppa; Alessandra Saccani; Marina Sironi; Barbara Bottazzi; Andrea Doni; Bronte Vincenzo; Fabio Pasqualini; Luca Vago; Manuela Nebuloni; Alberto Mantovani; Antonio Sica
Cancer Research | 2012
Hagen Kulbe; Probir Chakravarty; D. Andrew Leinster; Kellie A. Charles; Joseph Kwong; Richard G. Thompson; Jermaine Coward; Tiziana Schioppa; Stephen C. Robinson; William M. Gallagher; Laura Galletta; Michael A. Salako; John F. Smyth; Thorsten Hagemann; Donal J. Brennan; David Bowtell; Frances R. Balkwill
Novartis Foundation symposium | 2004
Alberto Mantovani; Silvano Sozzani; Massimo Locati; Tiziana Schioppa; Alessandra Saccani; Paola Allavena; Antonio Sica
Archive | 2008
Frances Balkwill; Violet Slettenaar; Julia Wilson; Yaohe Wang; Tiziana Schioppa