Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Suzanne Fioravanti is active.

Publication


Featured researches published by Suzanne Fioravanti.


Immunology | 2012

S100A9 a new marker for monocytic human myeloid‐derived suppressor cells

Fei Zhao; Bastian Hoechst; Austin Duffy; Jaba Gamrekelashvili; Suzanne Fioravanti; Michael P. Manns; Tim F. Greten; Firouzeh Korangy

Myeloid‐derived suppressor cells (MDSC) are a heterogeneous population of cells that negatively regulate the immune response during tumour progression, inflammation and infection. Only limited data are available on human MDSC because of the lack of specific markers. We have identified members of the S100 protein family – S100A8, S100A9 and S100A12 – specifically expressed in CD14+ HLA‐DR−/low MDSC. S100A9 staining in combination with anti‐CD14 could be used to identify MDSC in whole blood from patients with colon cancer. An increase in the population of CD14+ S100A9high MDSC was observed in the peripheral blood from colon cancer patients in comparison with healthy controls. Finally, nitric oxide synthase expression, a hallmark of MDSC, was induced in CD14+ S100A9high upon lipopolysaccharide/interferon‐γ stimulation. We propose S100 proteins as useful markers for the analysis and further characterization of human MDSC.


Cancer Immunology, Immunotherapy | 2013

Comparative analysis of monocytic and granulocytic myeloid-derived suppressor cell subsets in patients with gastrointestinal malignancies

Austin Duffy; Fei Zhao; Lydia Haile; Jaba Gamrekelashvili; Suzanne Fioravanti; Chi Ma; Tamar Kapanadze; Kathryn Compton; William D. Figg; Tim F. Greten

Myeloid-derived suppressor cells (MDSC) are a heterogenous population of cells comprising myeloid progenitor cells and immature myeloid cells, which have the ability to suppress the effector immune response. In humans, MDSC have not been well characterized owing to the lack of specific markers, although it is possible to broadly classify the MDSC phenotypes described in the literature as being predominantly granulocytic (expressing markers such as CD15, CD66, CD33) or monocytic (expressing CD14). In this study, we set out to perform a direct comparative analysis across both granulocytic and monocytic MDSC subsets in terms of their frequency, absolute number, and function in the peripheral blood of patients with advanced GI cancer. We also set out to determine the optimal method of sample processing given that this is an additional source of heterogeneity. Our findings demonstrate consistent changes across sample processing methods for monocytic MDSC, suggesting that reliance upon cryopreserved PBMC is acceptable. Although we did not see an increase in the population of granulocytic MDSC, these cells were found to be more suppressive than their monocytic counterparts.


Proceedings of the National Academy of Sciences of the United States of America | 2015

90Y-daclizumab, an anti-CD25 monoclonal antibody, provided responses in 50% of patients with relapsed Hodgkin’s lymphoma

John E. Janik; John C. Morris; Deirdre O'Mahony; Stefania Pittaluga; Elaine S. Jaffe; Christophe E. Redon; William M. Bonner; Martin W. Brechbiel; Chang H. Paik; Millie Whatley; Clara C. Chen; Jae-Ho Lee; Thomas A. Fleisher; Maggie Brown; Jeffrey D. White; Donn M. Stewart; Suzanne Fioravanti; Cathryn C. Lee; Carolyn K. Goldman; Bonita R. Bryant; Richard P. Junghans; Jorge A. Carrasquillo; Tatyana Worthy; Erin Corcoran; Kevin C. Conlon; Thomas A. Waldmann

Significance Despite advances, a significant proportion of patients with Hodgkin’s lymphoma (HL) will not respond or will relapse. We demonstrated that up to seven infusions of 90Y-daclizumab, an anti-CD25–directed monoclonal antibody, provided responses in 50% of patients with relapsed HL. The daclizumab was directed primarily not at tumor cells themselves but toward nonmalignant T cells rosetting around the Reed–Sternberg cells. 90Y provided strong β emissions that killed antigen-nonexpressing tumor cells at a distance by a crossfire effect. Furthermore, the strong β irradiation killed normal cells in the tumor microenvironment that nurture the malignant cells in the lymphomatous mass. Therefore 90Y-daclizumab infusions provide meaningful therapy for select HL patients. Despite significant advances in the treatment of Hodgkin’s lymphoma (HL), a significant proportion of patients will not respond or will subsequently relapse. We identified CD25, the IL-2 receptor alpha subunit, as a favorable target for systemic radioimmunotherapy of HL. The scientific basis for the clinical trial was that, although most normal cells with exception of Treg cells do not express CD25, it is expressed by a minority of Reed–Sternberg cells and by most polyclonal T cells rosetting around Reed–Sternberg cells. Forty-six patients with refractory and relapsed HL were evaluated with up to seven i.v. infusions of the radiolabeled anti-CD25 antibody 90Y-daclizumab. 90Y provides strong β emissions that kill tumor cells at a distance by a crossfire effect. In 46 evaluable HL patients treated with 90Y-daclizumab there were 14 complete responses and nine partial responses; 14 patients had stable disease, and nine progressed. Responses were observed both in patients whose Reed–Sternberg cells expressed CD25 and in those whose neoplastic cells were CD25− provided that associated rosetting T cells expressed CD25. As assessed using phosphorylated H2AX (γ-H2AX) as a bioindicator of the effects of radiation exposure, predominantly nonmalignant cells in the tumor microenvironment manifested DNA damage, as reflected by increased expression of γ-H2AX. Toxicities were transient bone-marrow suppression and myelodysplastic syndrome in six patients who had not been evaluated with bone-marrow karyotype analyses before therapy. In conclusion, repeated 90Y-daclizumab infusions directed predominantly toward nonmalignant T cells rosetting around Reed–Sternberg cells provided meaningful therapy for select HL patients.


United European gastroenterology journal | 2015

A phase II study of TRC105 in patients with hepatocellular carcinoma who have progressed on sorafenib

Austin Duffy; Susanna Varkey Ulahannan; Liang Cao; Osama E. Rahma; Oxana V. Makarova-Rusher; David E. Kleiner; Suzanne Fioravanti; Melissa Walker; S Carey; Yunkai Yu; Aradhana M. Venkatesan; Baris Turkbey; Peter L. Choyke; Jane B. Trepel; Kc Bollen; Seth M. Steinberg; William D. Figg; Tim F. Greten

Background Endoglin is an endothelial cell membrane receptor essential for angiogenesis and highly expressed on the vasculature of many tumor types, including hepatocellular carcinoma (HCC). TRC105 is a chimeric IgG1 anti-CD105 monoclonal antibody that inhibits angiogenesis and tumor growth by endothelial cell growth inhibition, ADCC and apoptosis, and complements VEGF inhibitors. Objective The aim of this phase II study was to evaluate the efficacy of anti-endoglin therapy with TRC105 in patients with advanced HCC, post-sorafenib. Methods Patients with HCC and compensated liver function (Childs-Pugh A/B7), ECOG 0/1, were enrolled to a single-arm, phase II study of TRC105 15 mg/kg IV every two weeks. Patients must have progressed on or been intolerant of prior sorafenib. A Simon optimal two-stage design was employed with a 50% four-month PFS target for progression to the second stage. Correlative biomarkers evaluated included DCE-MRI as well as plasma levels of angiogenic biomarkers and soluble CD105. Results A total accrual of 27 patients was planned. However, because of lack of efficacy and in accordance with the Simon two-stage design, 11 patients were enrolled. There were no grade 3/4 treatment-related toxicities. Most frequent toxicities were headache (G2; N = 3) and epistaxis (G1; N = 4). One patient had a confirmed partial response by standard RECIST criteria and biologic response on DCE-MRI but the four-month PFS was insufficient to proceed to the second stage of the study. Conclusions: TRC105 was well tolerated in this HCC population following sorafenib. Although there was evidence of clinical activity, this did not meet prespecified criteria to proceed to the second stage. TRC105 development in HCC continues as combination therapy with sorafenib.


International Journal of Cancer | 2016

Modulation of tumor eIF4E by antisense inhibition: A phase I/II translational clinical trial of ISIS 183750 – an antisense oligonucleotide against eIF4E – in combination with irinotecan in solid tumors and irinotecan‐refractory colorectal cancer

Austin Duffy; Oxana V. Makarova-Rusher; Susanna Varkey Ulahannan; Osama E. Rahma; Suzanne Fioravanti; Melissa Walker; S. Abdullah; Mark Raffeld; Victoria L. Anderson; Nadine Abi-Jaoudeh; E. Levy; Bradford J. Wood; Su Jae Lee; Yasushi Tomita; Jane B. Trepel; Seth M. Steinberg; A.S. Revenko; A.R. MacLeod; Cody J. Peer; William D. Figg; Tim F. Greten

The eukaryotic translation initiation factor 4E (eIF4E) is a potent oncogene that is found to be dysregulated in 30% of human cancer, including colorectal carcinogenesis (CRC). ISIS 183750 is a second‐generation antisense oligonucleotide (ASO) designed to inhibit the production of the eIF4E protein. In preclinical studies we found that EIF4e ASOs reduced expression of EIF4e mRNA and inhibited proliferation of colorectal carcinoma cells. An additive antiproliferative effect was observed in combination with irinotecan. We then performed a clinical trial evaluating this combination in patients with refractory cancer. No dose‐limiting toxicities were seen but based on pharmacokinetic data and tolerability the dose of irinotecan was reduced to 160 mg/m2 biweekly. Efficacy was evaluated in 15 patients with irinotecan‐refractory colorectal cancer. The median time of disease control was 22.1 weeks. After ISIS 183750 treatment, peripheral blood levels of eIF4E mRNA were decreased in 13 of 19 patients. Matched pre‐ and posttreatment tumor biopsies showed decreased eIF4E mRNA levels in five of nine patients. In tumor tissue, the intracellular and stromal presence of ISIS 183750 was detected by IHC in all biopsied patients. Although there were no objective responses stable disease was seen in seven of 15 (47%) patients who were progressing before study entry, six of whom were stable at the time of the week 16 CT scan. We were also able to confirm through mandatory pre‐ and posttherapy tumor biopsies penetration of the ASO into the site of metastasis.


Journal for ImmunoTherapy of Cancer | 2014

Paired tumor biopsy analysis and safety data from a pilot study evaluating Tremelimumab - a monoclonal antibody against CTLA-4 - in combination with ablative therapy in patients with hepatocellular carcinoma (HCC)

Austin Duffy; Sid P. Kerkar; David E. Kleiner; Susanna Varkey Ulahannan; Metin Kurtoglu; Oxana Rusher; Suzanne Fioravanti; Melissa Walker; William D. Figg; Kathryn Compton; Aradhana M. Venkatesan; Nadine Abi-Jaoudeh; Brad J. Wood; Tim F. Greten

Meeting abstracts Tremelimumab is a fully human monoclonal antibody that binds to CTLA-4 expressed on the surface of activated T lymphocytes and results in inhibition of B7-CTLA-4-mediated down regulation of T cell activation. Both transcatheter arterial chemoembolization (TACE) and radiofrequency


Investigational New Drugs | 2016

A phase I study of selumetinib (AZD6244/ARRY-142866), a MEK1/2 inhibitor, in combination with cetuximab in refractory solid tumors and KRAS mutant colorectal cancer

Dustin A. Deming; Ludmila L. Cavalcante; Sam J. Lubner; Daniel Mulkerin; Noelle K. LoConte; Jens C. Eickhoff; Jill M. Kolesar; Suzanne Fioravanti; Tim F. Greten; Kathryn Compton; Austin Doyle; George Wilding; Austin G. Duffy; Glenn Liu


Journal of Clinical Oncology | 2016

Tremelimumab, a monoclonal antibody against CTLA-4, in combination with subtotal ablation (trans-catheter arterial chemoembolization [TACE], radiofrequency ablation [RFA] or cryoablation) in patients with hepatocellular carcinoma (HCC) and biliary tract carcinoma (BTC).

Austin Duffy; Oxana V. Makarova-Rusher; Drew Pratt; David E. Kleiner; Suzanne Fioravanti; Melissa Walker; Stephanie Carey; William D. Figg; Seth M. Steinberg; Victoria L. Anderson; E. Levy; Venkatesh Krishnasamy; Bradford J. Wood; Tim F. Greten


Journal of Clinical Oncology | 2010

Phase II trial of daclizumab in human T-cell lymphotropic virus type-1 (HTLV-1)-associated adult T-cell leukemia/lymphoma (ATL).

J. L. Berkowitz; John E. Janik; Donn M. Stewart; Suzanne Fioravanti; Elaine S. Jaffe; Thomas A. Fleisher; N. Urquhart; G. H. Wharfe; Thomas A. Waldmann; John C. Morris


Journal of Clinical Oncology | 2016

A pilot study of AMP-224, a PD-L2 Fc fusion protein, in combination with stereotactic body radiation therapy (SBRT) in patients with metastatic colorectal cancer.

Austin G. Duffy; Oxana V. Makarova-Rusher; Drew Pratt; David E. Kleiner; Suzanne Fioravanti; Melissa Walker; Stephanie Carey; William D. Figg; Seth M. Steinberg; Victoria L. Anderson; Elliot Levy; Venkatesh Krishnasamy; Bradford J. Wood; J Jones; Deborah Citrin; Tim F. Greten

Collaboration


Dive into the Suzanne Fioravanti's collaboration.

Top Co-Authors

Avatar

Tim F. Greten

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

William D. Figg

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Austin G. Duffy

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Melissa Walker

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David E. Kleiner

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kathryn Compton

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Seth M. Steinberg

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aradhana M. Venkatesan

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge