Network


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

Hotspot


Dive into the research topics where Sophie Paczesny is active.

Publication


Featured researches published by Sophie Paczesny.


Blood | 2008

A biomarker panel for acute graft-versus-host disease.

Sophie Paczesny; Oleg Krijanovski; Thomas M. Braun; Sung Won Choi; Shawn G. Clouthier; Rork Kuick; David E. Misek; Kenneth R. Cooke; Carrie L. Kitko; Angela C. Weyand; Daniel Bickley; Dawn Jones; Joel Whitfield; Pavan Reddy; John E. Levine; Samir M. Hanash; James L.M. Ferrara

No validated biomarkers exist for acute graft-versus-host disease (GVHD). We screened plasma with antibody microarrays for 120 proteins in a discovery set of 42 patients who underwent transplantation that revealed 8 potential biomarkers for diagnostic of GVHD. We then measured by enzyme-linked immunosorbent assay (ELISA) the levels of these biomarkers in samples from 424 patients who underwent transplantation randomly divided into training (n = 282) and validation (n = 142) sets. Logistic regression analysis of these 8 proteins determined a composite biomarker panel of 4 proteins (interleukin-2-receptor-alpha, tumor-necrosis-factor-receptor-1, interleukin-8, and hepatocyte growth factor) that optimally discriminated patients with and without GVHD. The area under the receiver operating characteristic curve distinguishing these 2 groups in the training set was 0.91 (95% confidence interval, 0.87-0.94) and 0.86 (95% confidence interval, 0.79-0.92) in the validation set. In patients with GVHD, Cox regression analysis revealed that the biomarker panel predicted survival independently of GVHD severity. A panel of 4 biomarkers can confirm the diagnosis of GVHD in patients at onset of clinical symptoms of GVHD and provide prognostic information independent of GVHD severity.


Journal of Immunology | 2008

Human Mesenchymal Stem Cells License Adult CD34+ Hemopoietic Progenitor Cells to Differentiate into Regulatory Dendritic Cells through Activation of the Notch Pathway

Yin Ping Li; Sophie Paczesny; Evelyne Lauret; Sonia Poirault; P. Bordigoni; Fatiha Mekhloufi; Olivier Hequet; Yves Bertrand; Jing Ping Ou-Yang; Jean-François Stoltz; Pierre Miossec; Assia Eljaafari

The mechanisms underlying the immunomodulatory functions of mesenchymal stem cells (MSC) on dendritic cells (DC) have been shown to involve soluble factors, such as IL-6 or TGF-β, or cell-cell contact, or both depending on the report referenced. In this study, we intend to clarify these mechanisms by examining the immunosuppressive effect of human adult MSC on adult DC differentiated from CD34+ hemopoietic progenitor cells (HPC). MSC have been shown to inhibit interstitial DC differentiation from monocytes and umbilical CD34+ HPC. In this study, we confirm that MSC not only halt interstitial DC but also Langerhans cell differentiation from adult CD34+ HPC, as assessed by the decreased expression of CD1a, CD14, CD86, CD80, and CD83 Ags on their cell surface. Accordingly, the functional capacity of CD34+ HPC-derived DC (CD34-DC) to stimulate alloreactive T cells was impaired. Furthermore, we showed that 1) MSC inhibited commitment of CD34+ HPC into immature DC, but not maturation of CD34-DC, 2) this inhibitory effect was reversible, and 3) DC generated in coculture with MSC (MSC-DC) induced the generation of alloantigen-specific regulatory T cells following secondary allostimulation. Conditioned medium from MSC cultures showed some inhibitory effect independent of IL-6, M-CSF, and TGF-β. In comparison, direct coculture of MSC with CD34+ HPC resulted in much stronger immunosuppressive effect and led to an activation of the Notch pathway as assessed by the overexpression of Hes1 in MSC-DC. Finally, DAPT, a γ-secretase inhibitor that inhibits Notch signaling, was able to overcome MSC-DC defects. In conclusion, our data suggest that MSC license adult CD34+ HPC to differentiate into regulatory DC through activation of the Notch pathway.


The New England Journal of Medicine | 2013

ST2 as a Marker for Risk of Therapy-Resistant Graft-versus-Host Disease and Death

Mark Vander Lugt; Thomas M. Braun; Samir M. Hanash; Jerome Ritz; Vincent T. Ho; Joseph H. Antin; Qing Zhang; Chee Hong Wong; Hong Wang; Alice Chin; Aurelie Gomez; Andrew C. Harris; John E. Levine; Sung W. Choi; Daniel R. Couriel; Pavan Reddy; James L.M. Ferrara; Sophie Paczesny

BACKGROUND No plasma biomarkers are associated with the response of acute graft-versus-host disease (GVHD) to therapy after allogeneic hematopoietic stem-cell transplantation. METHODS We compared 12 biomarkers in plasma obtained a median of 16 days after therapy initiation from 10 patients with a complete response by day 28 after therapy initiation and in plasma obtained from 10 patients with progressive GVHD during therapy. The lead biomarker, suppression of tumorigenicity 2 (ST2), was measured at the beginning of treatment for GVHD in plasma from 381 patients and during the first month after transplantation in three independent sets totaling 673 patients to determine the association of this biomarker with treatment-resistant GVHD and 6-month mortality after treatment or transplantation. RESULTS Of the 12 markers, ST2 had the most significant association with resistance to GVHD therapy and subsequent death without relapse. As compared with patients with low ST2 values at therapy initiation, patients with high ST2 values were 2.3 times as likely to have treatment-resistant GVHD (95% confidence interval [CI], 1.5 to 3.6) and 3.7 times as likely to die within 6 months after therapy (95% CI, 2.3 to 5.9). Patients with low ST2 values had lower mortality without relapse than patients with high ST2 values, regardless of the GVHD grade (11% vs. 31% among patients with grade I or II GVHD and 14% vs. 67% among patients with grade III or IV GVHD, P<0.001 for both comparisons). Plasma ST2 values at day 14 after transplantation were associated with 6-month mortality without relapse, regardless of the intensity of the conditioning regimen. CONCLUSIONS ST2 levels measured at the initiation of therapy for GVHD and during the first month after transplantation improved risk stratification for treatment-resistant GVHD and death without relapse after transplantation. (Funded by the National Institutes of Health.)


Blood | 2011

Regenerating islet-derived 3-alpha is a biomarker of gastrointestinal graft-versus-host disease

James L.M. Ferrara; Andrew C. Harris; Joel K. Greenson; Thomas M. Braun; Ernst Holler; Takanori Teshima; John E. Levine; Sung W.J. Choi; Elisabeth Huber; Karin Landfried; Koichi Akashi; Mark Vander Lugt; Pavan Reddy; Alice Chin; Qing Zhang; Samir M. Hanash; Sophie Paczesny

There are no plasma biomarkers specific for GVHD of the gastrointestinal (GI) tract, the GVHD target organ most associated with nonrelapse mortality (NRM) following hematopoietic cell transplantation (HCT). Using an unbiased, large-scale, quantitative proteomic discovery approach to identify candidate biomarkers that were increased in plasma from HCT patients with GI GVHD, 74 proteins were increased at least 2-fold; 5 were of GI origin. We validated the lead candidate, REG3α, by ELISA in samples from 1014 HCT patients from 3 transplantation centers. Plasma REG3α concentrations were 3-fold higher in patients at GI GVHD onset than in all other patients and correlated most closely with lower GI GVHD. REG3α concentrations at GVHD onset predicted response to therapy at 4 weeks, 1-year NRM, and 1-year survival (P ≤ .001). In a multivariate analysis, advanced clinical stage, severe histologic damage, and high REG3α concentrations at GVHD diagnosis independently predicted 1-year NRM, which progressively increased with higher numbers of onset risk factors present: 25% for patients with 0 risk factors to 86% with 3 risk factors present (P < .001). REG3α is a plasma biomarker of GI GVHD that can be combined with clinical stage and histologic grade to improve risk stratification of patients.


Bone Marrow Transplantation | 2010

New perspectives on the biology of acute GVHD

Sophie Paczesny; David A. Hanauer; Y Sun; Pavan Reddy

The use of allogeneic hematopoietic cell transplantation (HCT) has increased as new techniques have been developed for transplantation in patients who previously would not have been considered HCT candidates. However, its efficacy continued to be limited by the development of frequent and severe acute GVHD. The complex and intricate pathophysiology of acute GVHD is a consequence of interactions between the donor and host innate and adaptive immune responses. Multiple inflammatory molecules and cell types are implicated in the development of GVHD that can be categorized as: (1) triggers that initiate GVHD by therapy-induced tissue damage and the antigen disparities between host and graft tissue; (2) sensors that detect the triggers, that is, process and present alloantigens; (3) mediators such as T-cell subsets (naive, memory, regulatory, Th17 and natural killer T cells) and (4) the effectors and amplifiers that cause damage of the target organs. These multiple inflammatory molecules and cell types that are implicated in the development of GVHD have been described with models that use stepwise cascades. Herein, we provide a novel perspective on the immunobiology of acute GVHD and briefly discuss some of the outstanding questions and limitations of the model systems.


Science Translational Medicine | 2010

Elafin is a biomarker of graft-versus-host disease of the skin.

Sophie Paczesny; Thomas M. Braun; John E. Levine; Jason M. Hogan; Jeffrey Crawford; Bryan N. Coffing; Stephen H. Olsen; Sung W. Choi; Hong Wang; Vitor M. Faça; Sharon J. Pitteri; Qing Zhang; Alice Chin; Carrie L. Kitko; Shin Mineishi; Gregory A. Yanik; Edward Peres; David A. Hanauer; Ying Wang; Pavan Reddy; Samir M. Hanash; James L.M. Ferrara

Plasma elafin concentrations correlate with graft-versus-host disease of the skin and long-term survival. Progress toward biomarker commercialization requires the discovery, qualification, verification, optimization, and clinical validation of a candidate before it is incorporated into existing therapeutic diagnostic platforms. The tremendous value that could be derived from the advancement of methods to detect disease at earlier and more treatable stages puts this pipeline approach at the forefront of biomarker development. However, to date there are no clear success stories in which discovery proteomics has led to a deployed protein biomarker. There is no polymerase chain reaction equivalent available to detect, quantify, and amplify proteins. Rather, proteomics-based biomarker discovery across a wide assortment of diseases is enabled by technologies such as mass spectrometry to sift through a large span of complex analytes at variable concentrations. Now, Paczesny and colleagues use a mass spectrometry–based technique to unambiguously identify candidate plasma biomarkers of skin acute graft-versus-host disease (GVHD)—the primary cause of nonrelapse mortality after bone marrow transplantation (BMT). Rashes are common after BMT and can be caused by a variety of reasons, but because the consequences of GVHD are serious, physicians initiate treatment of suspected GVHD without a bona fide confirmed diagnosis. In the discovery set of this work, the authors examined plasma samples from patients who had received BMT with and without clinical diagnosis of skin GVHD, and found that in patients with skin GVHD, the concentration of one lead marker, elafin, was three times as high. In a follow-up independent validation of 492 BMT patients, skin biopsies stained with elafin stratified the patients consistently according to GVHD parameters, and elafin plasma concentrations were concordantly higher in patients with GVHD. The specificity and sensitivity of elafin relative to other markers revealed that it was the single best discriminator for the diagnosis of GVHD in BMT patients with a rash, and was correlated with the severity of the disease. Elafin concentrations also correlated with the eventual maximum grade of GVHD and with nonrelapse mortality. These results show that elafin concentrations may serve as a noninvasive diagnostic test as well as a prognostic marker in determining GVHD grading in the clinic. Graft-versus-host disease (GVHD), the major complication of allogeneic bone marrow transplantation, affects the skin, liver, and gastrointestinal tract. There are no plasma biomarkers specific for any acute GVHD target organ. We used a large-scale quantitative proteomic discovery procedure to identify biomarker candidates of skin GVHD and validated the lead candidate, elafin, with enzyme-linked immunosorbent assay in samples from 492 patients. Elafin was overexpressed in GVHD skin biopsies. Plasma concentrations of elafin were significantly higher at the onset of skin GVHD, correlated with the eventual maximum grade of GVHD, and were associated with a greater risk of death relative to other known risk factors (hazard ratio, 1.78). We conclude that elafin has significant diagnostic and prognostic value as a biomarker of skin GVHD.


Blood | 2012

Acute graft-versus-host disease biomarkers measured during therapy can predict treatment outcomes: a Blood and Marrow Transplant Clinical Trials Network study

John E. Levine; Brent R. Logan; Juan Wu; Amin M. Alousi; Javier Bolaños-Meade; James L.M. Ferrara; Vincent T. Ho; Daniel J. Weisdorf; Sophie Paczesny

Acute graft-versus-host disease (GVHD) is the primary limitation of allogeneic hematopoietic cell transplantation, and once it develops, there are no reliable diagnostic tests to predict treatment outcomes. We hypothesized that 6 previously validated diagnostic biomarkers of GVHD (IL-2 receptor-α; tumor necrosis factor receptor-1; hepatocyte growth factor; IL-8; elafin, a skin-specific marker; and regenerating islet-derived 3-α, a gastrointestinal tract-specific marker) could discriminate between therapy responsive and nonresponsive patients and predict survival in patients receiving GVHD therapy. We measured GVHD biomarker concentrations from samples prospectively obtained at the initiation of treatment, day 14, and day 28, on a multicenter, randomized, 4-arm phase 2 clinical trial for newly diagnosed acute GVHD. We found that at each of 3 time points, GVHD onset, 2 weeks into treatment, and 4 weeks into treatment, a 6-protein biomarker panel predicted for the important clinical outcomes of day 28 posttherapy nonresponse and mortality at day 180 from onset. GVHD biomarker panels can be used for early identification of patients at high or low risk for treatment nonresponsiveness or death, and they may provide opportunities for early intervention and improved survival after hematopoietic cell transplantation. The study was registered in clinicaltrials.gov as NCT00224874.


Blood | 2013

Discovery and Validation of Graft-versus-Host Disease Biomarkers

Sophie Paczesny

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the most effective tumor immunotherapy available. Although allo-HSCT provides beneficial graft-versus-tumor effects, acute GVHD (aGVHD) is the primary source of morbidity and mortality after HSCT. Diagnosis of aGVHD is typically based on clinical symptoms in one or more of the main target organs (skin, liver, gastrointestinal tract) and confirmed by biopsy. However, currently available diagnostic and staging tools often fail to identify patients at higher risk of GVHD progression, unresponsiveness to therapy, or death. In addition, there are shortcomings in the prediction of GVHD before clinical signs develop, indicating the urgent need for noninvasive and reliable laboratory tests. Through the continuing evolution of proteomics technologies seen in recent years, plasma biomarkers have been identified and validated as promising diagnostic tools for GVHD and prognostic tools for nonrelapse mortality. These biomarkers may facilitate timely and selective therapeutic intervention but should be more widely validated and incorporated into a new grading system for risk stratification of patients and better-customized treatment. This review identifies biomarkers for detecting GVHD, summarizes current information on aGVHD biomarkers, proposes future prospects for the blinded evaluation of these biomarkers, and discusses the need for biomarkers of chronic GVHD.


Biology of Blood and Marrow Transplantation | 2010

Frequency of CD4+CD25hiFOXP3+ Regulatory T Cells Has Diagnostic and Prognostic Value as a Biomarker for Acute Graft-versus-Host-Disease

John Magenau; Xuemei Qin; Isao Tawara; Clare E. Rogers; Carrie L. Kitko; Matthew Schlough; Daniel Bickley; Thomas M. Braun; Pil Sang Jang; Kathleen P. Lowler; Dawn Jones; Sung W. Choi; Pavan Reddy; Shin Mineishi; John E. Levine; James L.M. Ferrara; Sophie Paczesny

The relationship between regulatory T cells (Tregs) and acute graft-versus-host disease (aGVHD) in clinical allogeneic bone marrow transplantation (BMT) recipients is not well established. We conducted a prospective analysis of peripheral blood Tregs as determined by the frequency of CD4(+)CD25(hi)FOXP3(+) lymphocytes in 215 BMT patients. Autologous BMT patients (N = 90) and allogeneic BMT patients without GVHD (N = 65) had similar Treg frequencies, whereas allogeneic patients with GVHD (N = 60) had Treg frequencies that were 40% less than those without GVHD. Treg frequencies decreased linearly with increasing grades of GVHD at onset, and correlated with eventual maximum grade of GVHD (P < .001). In addition, frequency of Tregs at onset of GVHD predicted the response to GVHD treatment (P = .003). Patients with Treg frequencies less than the median had higher nonrelapse mortality (NRM) than patients with Tregs greater than the median, but experienced equivalent relapse mortality, resulting in an inferior survival at 2 years (38% versus 63%, P = .03). Treg frequency may therefore have important prognostic value as a biomarker of aGVHD.


Blood | 2008

Change in plasma tumor necrosis factor receptor 1 levels in the first week after myeloablative allogeneic transplantation correlates with severity and incidence of GVHD and survival

Sung W. Choi; Carrie L. Kitko; Thomas M. Braun; Sophie Paczesny; Gregory A. Yanik; Shin Mineishi; Oleg Krijanovski; Dawn Jones; Joel Whitfield; Kenneth R. Cooke; Raymond J. Hutchinson; James L.M. Ferrara; John E. Levine

Acute graft-versus-host disease (GVHD) remains a significant cause of mortality after hematopoietic cell transplantation (HCT). Tumor necrosis factor-alpha (TNF-alpha) mediates GVHD by amplifying donor immune responses to host tissues and by direct toxicity to target organs. We measured TNF receptor 1 (TNFR1) as a surrogate marker for TNF-alpha in 438 recipients of myeloablative HCT before transplantation and at day 7 after transplantation. Increases in TNFR1 levels more than or equal to 2.5 baseline correlated with eventual development of GVHD grade 2 to 4 (58% vs 32%, P < .001) and with treatment-related mortality (39% vs 17%, P < .001). In a multivariate analysis including age, degree of HLA match, donor type, recipient and donor sex, disease, and status at HCT, the increase in TNFR1 level at day 7 remained a significant predictor for outcome. Measurement of TNFR1 levels early after transplantation provides independent information in advance of important clinical outcomes, such as GVHD and death.

Collaboration


Dive into the Sophie Paczesny's collaboration.

Top Co-Authors

Avatar

John E. Levine

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James L.M. Ferrara

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Pavan Reddy

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Carrie L. Kitko

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qing Zhang

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Samir M. Hanash

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Shin Mineishi

Penn State Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge