Network


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

Hotspot


Dive into the research topics where Mariano Severgnini is active.

Publication


Featured researches published by Mariano Severgnini.


The New England Journal of Medicine | 2016

Ipilimumab for Patients with Relapse after Allogeneic Transplantation

Matthew S. Davids; Haesook T. Kim; Pavan Bachireddy; Caitlin Costello; Rebecca Liguori; Alexandra Savell; Alexander Lukez; David Avigan; Yi-Bin Chen; Peter A. McSweeney; Nicole R. LeBoeuf; Michael S. Rooney; Michaela Bowden; Chensheng W. Zhou; Scott R. Granter; Jason L. Hornick; Scott J. Rodig; Masahiro Hirakawa; Mariano Severgnini; F. Stephen Hodi; Catherine J. Wu; Vincent T. Ho; Corey Cutler; John Koreth; Edwin P. Alyea; Joseph H. Antin; Philippe Armand; Howard Streicher; Edward D. Ball; Jerome Ritz

BACKGROUND Loss of donor-mediated immune antitumor activity after allogeneic hematopoietic stem-cell transplantation (HSCT) permits relapse of hematologic cancers. We hypothesized that immune checkpoint blockade established by targeting cytotoxic T-lymphocyte-associated protein 4 with ipilimumab could restore antitumor reactivity through a graft-versus-tumor effect. METHODS We conducted a phase 1/1b multicenter, investigator-initiated study to determine the safety and efficacy of ipilimumab in patients with relapsed hematologic cancer after allogeneic HSCT. Patients received induction therapy with ipilimumab at a dose of 3 or 10 mg per kilogram of body weight every 3 weeks for a total of 4 doses, with additional doses every 12 weeks for up to 60 weeks in patients who had a clinical benefit. RESULTS A total of 28 patients were enrolled. Immune-related adverse events, including one death, were observed in 6 patients (21%), and graft-versus-host disease (GVHD) that precluded further administration of ipilimumab was observed in 4 patients (14%). No responses that met formal response criteria occurred in patients who received a dose of 3 mg per kilogram. Among 22 patients who received a dose of 10 mg per kilogram, 5 (23%) had a complete response, 2 (9%) had a partial response, and 6 (27%) had decreased tumor burden. Complete responses occurred in 4 patients with extramedullary acute myeloid leukemia and 1 patient with the myelodysplastic syndrome developing into acute myeloid leukemia. Four patients had a durable response for more than 1 year. Responses were associated with in situ infiltration of cytotoxic CD8+ T cells, decreased activation of regulatory T cells, and expansion of subpopulations of effector T cells in the blood. CONCLUSIONS Our early-phase data showed that administration of ipilimumab was feasible in patients with recurrent hematologic cancers after allogeneic HSCT, although immune-mediated toxic effects and GVHD occurred. Durable responses were observed in association with several histologic subtypes of these cancers, including extramedullary acute myeloid leukemia. (Funded by the National Institutes of Health and others; ClinicalTrials.gov number, NCT01822509.).


Cancer immunology research | 2017

Angiopoietin-2 as a Biomarker and Target for Immune Checkpoint Therapy

Xinqi Wu; Anita Giobbie-Hurder; Xiaoyun Liao; Courtney Connelly; Erin M. Connolly; Jingjing Li; Michael P. Manos; Donald P. Lawrence; David F. McDermott; Mariano Severgnini; Jun Zhou; Evisa Gjini; Ana Lako; Mikel Lipschitz; Christine Pak; Sara Abdelrahman; Scott J. Rodig; F. Stephen Hodi

Outcomes for metastatic melanoma patients treated with checkpoint blockade were poor when circulating Ang-2 was high. Ang-2 promoted recruitment of tumor macrophages and upregulated PD-L1, making it a predictive and/or prognostic biomarker and potential target to combine with checkpoint blockade. Immune checkpoint therapies targeting CTLA-4 and PD-1 have proven effective in cancer treatment. However, the identification of biomarkers for predicting clinical outcomes and mechanisms to overcome resistance remain as critical needs. Angiogenesis is increasingly appreciated as an immune modulator with potential for combinatorial use with checkpoint blockade. Angiopoietin-2 (ANGPT2) is an immune target in patients and is involved in resistance to anti-VEGF treatment with the monoclonal antibody bevacizumab. We investigated the predictive and prognostic value of circulating ANGPT2 in metastatic melanoma patients receiving immune checkpoint therapy. High pretreatment serum ANGPT2 was associated with reduced overall survival in CTLA-4 and PD-1 blockade–treated patients. These treatments also increased serum ANGPT2 in many patients early after treatment initiation, whereas ipilimumab plus bevacizumab treatment decreased serum concentrations. ANGPT2 increases were associated with reduced response and/or overall survival. Ipilimumab increased, and ipilimumab plus bevacizumab decreased, tumor vascular ANGPT2 expression in a subset of patients, which was associated with increased and decreased tumor infiltration by CD68+ and CD163+ macrophages, respectively. In vitro, bevacizumab blocked VEGF-induced ANGPT2 expression in tumor-associated endothelial cells, whereas ANGPT2 increased PD-L1 expression on M2-polarized macrophages. Treatments elicited long-lasting and functional antibody responses to ANGPT2 in a subset of patients receiving clinical benefit. Our findings suggest that serum ANGPT2 may be considered as a predictive and prognostic biomarker for immune checkpoint therapy and may contribute to treatment resistance via increasing proangiogenic and immunosuppressive activities in the tumor microenvironment. Targeting ANGPT2 provides a rational combinatorial approach to improve the efficacy of immune therapy. Cancer Immunol Res; 5(1); 17–28. ©2016 AACR.


Cancer | 2017

Immunotherapy with single agent nivolumab for advanced leiomyosarcoma of the uterus: Results of a phase 2 study

Eytan Ben-Ami; Constance Barysauskas; Sarah Solomon; Kadija Tahlil; Rita Malley; Melissa Hohos; Kathleen Polson; Margaret Loucks; Mariano Severgnini; Tara Patel; Amy Cunningham; Scott J. Rodig; F. Stephen Hodi; Jeffrey A. Morgan; Priscilla Merriam; Andrew J. Wagner; Geoffrey I. Shapiro; Suzanne George

Immunotherapy has changed the therapeutic landscape in oncology. Advanced uterine leiomyosarcoma (ULMS) remains an incurable disease in most cases, and despite new drug approvals, improvements in overall survival have been modest at best. The goal of this study was to evaluate programmed‐death 1 (PD‐1) inhibition with nivolumab in this patient population.


British Journal of Cancer | 2016

Definitive chemoradiation alters the immunologic landscape and immune checkpoints in head and neck cancer

Vishwajith Sridharan; Danielle N. Margalit; Stephanie A. Lynch; Mariano Severgnini; Jun Zhou; Nicole G. Chau; Guilherme Rabinowits; Jochen H. Lorch; Peter S. Hammerman; F. Stephen Hodi; Robert I. Haddad; Roy B. Tishler; Jonathan D. Schoenfeld

Background:Preclinical and clinical studies suggest potential synergy between high dose per fraction focal radiation and immunotherapy. However, conventionally fractionated radiation regimens in combination with concurrent chemotherapy are more commonly administered to patients as definitive treatment and may have both immune-stimulating and -suppressive effects.Methods:We prospectively collected longitudinal samples from head and neck squamous cell carcinoma patients receiving definitive radiation therapy. We quantified changes in populations of circulating immune cells and chemokines CXCL9, 10, and 16. Analyses of humoral and cellular immune responses were conducted in select patients via proteomic analysis and T-cell receptor sequencing.Results:Treatment not only increased circulating CD-8+ T-effector cells, but also myeloid-derived suppressor cells, regulatory T cells, and checkpoint receptor-expressing T cells, particularly PD-1+ T cells. Significant decreases in CXCL10 and increases in CXLC16 were noted. Treatment also increased the percentage of unique and dominant TCR clones, and increased humoral responses as measured by proteomic array.Conclusions:Our results suggest that fractionated chemoradiation leads to quantifiable effects in circulating immune mediators, including a balance of stimulatory and suppressive mechanisms. These results suggest future combinations with immune checkpoint blockade.


Journal for ImmunoTherapy of Cancer | 2018

Distinct predictive biomarker candidates for response to anti-CTLA-4 and anti-PD-1 immunotherapy in melanoma patients

Priyanka B. Subrahmanyam; Zhiwan Dong; Daniel Gusenleitner; Anita Giobbie-Hurder; Mariano Severgnini; Jun Zhou; Michael P. Manos; Lauren M. Eastman; Holden T. Maecker; F. Stephen Hodi

BackgroundWhile immune checkpoint blockade has greatly improved clinical outcomes in diseases such as melanoma, there remains a need for predictive biomarkers to determine who will likely benefit most from which therapy. To date, most biomarkers of response have been identified in the tumors themselves. Biomarkers that could be assessed from peripheral blood would be even more desirable, because of ease of access and reproducibility of sampling.MethodsWe used mass cytometry (CyTOF) to comprehensively profile peripheral blood of melanoma patients, in order to find predictive biomarkers of response to anti-CTLA-4 or anti-PD-1 therapy. Using a panel of ~ 40 surface and intracellular markers, we performed in-depth phenotypic and functional immune profiling to identify potential predictive biomarker candidates.ResultsImmune profiling of baseline peripheral blood samples using CyTOF revealed that anti-CTLA-4 and anti-PD-1 therapies have distinct sets of candidate biomarkers. The distribution of CD4+ and CD8+ memory/non-memory cells and other memory subsets was different between responders and non-responders to anti-CTLA-4 therapy. In anti-PD-1 (but not anti-CTLA-4) treated patients, we discovered differences in CD69 and MIP-1β expressing NK cells between responders and non-responders. Finally, multivariate analysis was used to develop a model for the prediction of response.ConclusionsOur results indicate that anti-CTLA-4 and anti-PD-1 have distinct predictive biomarker candidates. CD4+ and CD8+ memory T cell subsets play an important role in response to anti-CTLA-4, and are potential biomarker candidates. For anti-PD-1 therapy, NK cell subsets (but not memory T cell subsets) correlated with clinical response to therapy. These functionally active NK cell subsets likely play a critical role in the anti-tumor response triggered by anti-PD-1.


Journal for ImmunoTherapy of Cancer | 2016

Effects of definitive chemoradiation on circulating immunologic angiogenic cytokines in head and neck cancer patients

Vishwajith Sridharan; Danielle N. Margalit; Stephanie A. Lynch; Mariano Severgnini; F. Stephen Hodi; Robert I. Haddad; Roy B. Tishler; Jonathan D. Schoenfeld

BackgroundPreclinical studies suggest a synergistic effect between radiation, immunotherapy and anti-angiogenic therapy, although the mechanisms are unclear. Angiogenic cytokines are known to affect the immune system, and their levels may be associated with response to immunotherapy. Here, we assess changes in circulating VEGF, as well as angiogenic cytokines angiopoietin-1 and -2 (Ang1, Ang2), and placental growth factor (PLGF) that occur during definitive chemo-radiotherapy in HNSCC patients.MethodsWe prospectively collected blood samples from patients receiving definitive radiation with or without chemotherapy. Serum Ang1, Ang2, VEGF, and PLGF were measured via cytokine assays.ResultsThe majority of patients had advanced stage, node positive HPV-associated oropharyngeal cancer, and received radiation to a median dose of 70 Gy with concurrent cisplatin. Over the course of treatment, serum VEGF and Ang1 levels decreased in 20/24 (84 %, p < 0.0001) and 21/24 (88 %, p < 0.0001) patients, respectively, and Ang2 and PLGF levels increased in 20/24 (83 %, p < 0.0001) patients.ConclusionsWe find significant changes in angiogenic cytokines in the majority of HNSCC patients over the course of chemoradiation. Decreases in VEGF caused by radiation may represent one mechanism of potential synergy with immunotherapy. Increases in Ang2 and PLGF are interesting given their link to tumor associated angiogenesis and poor prognosis. Additional studies are needed to explore synergies between anti-angiogenic treatments, immunotherapy, and chemoradiation in HNSCC.


Cancer immunology research | 2016

Immune profiling of adenoid cystic carcinoma: PD-L2 Expression and Associations With Tumor Infiltrating Lymphocytes

Vishwajith Sridharan; Evisa Gjini; Xiaoyun Liao; Nicole G. Chau; Robert I. Haddad; Mariano Severgnini; Peter S. Hammerman; Adel K. El-Naggar; Gordon J. Freeman; F. Stephen Hodi; Scott J. Rodig; Glenn Dranoff; Jonathan D. Schoenfeld

PD-1 ligand expression and T-cell infiltration may predict responsiveness to PD-1 pathway inhibitors. ACC tumors expressed PD-L2 and the Wnt and PI3K pathways but had little immune infiltration. Chemoradiotherapy promoted antitumor responses, suggesting potential synergies with PD-1 blockade. Adenoid cystic carcinoma (ACC) is among the most lethal salivary gland tumors, with no treatments for metastatic disease that prolong survival. We examined tissue from 28 primary and metastatic ACC deposits obtained from 21 patients for infiltrating immune cells and PD-L1/PD-L2 expression and determined mRNA profiles of over 1,400 oncogenic and immune-related genes. We also assessed the effect of chemoradiation on immune mediators in a patient who had serial biopsies available. Most tumors expressed PD-L2 but had few infiltrating immune cells. Lack of immune-cell infiltrate was associated with expression of genes in the β-catenin/Wnt and PI3K pathways. Additionally, certain transcripts linked to growth and invasion were differentially expressed among primary and metastatic deposits. Chemoradiation appeared to increase CD8+ effector T cells, decrease regulatory T cells, and promote a systemic humoral response. These data suggest a potential role for PD-L2 inhibition and immune modulation as treatment for patients with ACC. Cancer Immunol Res; 4(8); 679–87. ©2016 AACR.


Cytotechnology | 2018

Development of an 8-color antibody panel for functional phenotyping of human CD8+ cytotoxic T cells from peripheral blood mononuclear cells

Tara Patel; Amy Cunningham; Martha Holland; John F. Daley; Suzan Lazo; F. Stephen Hodi; Mariano Severgnini

The study of CD8 positive cells in peripheral blood has become an essential part of research in the field of cancer immunotherapies, vaccine development, inflammation, autoimmune disease, etc. In this study, an 8-color flow cytometry panel, containing lineage and functional markers, was developed for the identification of CD8+ cytotoxic T cells in previously cryopreserved peripheral blood mononuclear cells from healthy human donors. By studying functional markers in naïve and CD3/CD28 activated T cells we demonstrate that the panel is capable of detecting protein markers corresponding to different T cell activation statuses. Data generated by flow cytometry were corroborated by different antibody based assay technologies to detect soluble cytokines. Our findings suggest that there is an inter donor variability in both baseline and activation responses. We have also successfully developed an antibody panel for flow cytometry that could be used to study cytotoxic function of CD8 T cells in clinical immunology research areas.


OncoImmunology | 2018

Anti-CTLA-4 based therapy elicits humoral immunity to galectin-3 in patients with metastatic melanoma

Xinqi Wu; Anita Giobbie-Hurder; Erin M. Connolly; Jingjing Li; Xiaoyun Liao; Mariano Severgnini; Jun Zhou; Scott J. Rodig; F. Stephen Hodi


Journal of Clinical Oncology | 2018

Phase II study of pembrolizumab or pembrolizumab plus bevacizumab for recurrent glioblastoma (rGBM) patients.

David A. Reardon; Lakshmi Nayak; Katherine B. Peters; Jennifer Leigh Clarke; Justin T. Jordan; John F. de Groot; Phioanh L. Nghiemphu; Thomas Kaley; Howard Colman; Sarah C. Gaffey; Victoria Caruso; Myriam Bednarek Debruyne; Chinmay Bhavsar; Annette M. Molinaro; Timothy R. Smith; Mariano Severgnini; Patrick Y. Wen

Collaboration


Dive into the Mariano Severgnini's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Scott J. Rodig

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge