Sophie Paczesny

Permanent URI for this collection

Translating Novel Biomarkers of Transplant Immunity and Tolerance

Allogeneic hematopoietic stem cell transplantation (HSCT) is a major therapy for malignant diseases of the blood and bone marrow and the most potent form of immune therapy against these diseases through its graft-versus-leukemia/tumor (GVL/GVT) effect. However, the efficacy of allogeneic HSCT has been impeded by frequent and severe graft-versus- host disease (GVHD) that is tightly linked to the GVL/GVT effect. Both acute and chronic forms of GVHD exist. The immunology of GVHD and GVL responses are complex and cytokines and cellular effectors are critical. Dr. Paczesy’s laboratory focuses on understanding the role of cytokines and cellular effectors in the biology of GVHD/GVL by discovering and investigating biomarkers in the blood and tissue of patients following allogeneic HSCT.

The goal of Dr. Paczesy’s laboratory is to develop tests to predict the risk of developing CVHD, responsiveness to treatment, and patient survival by integrating both proteomic and cellular biomarkers for the diagnosis and prognosis of GVHD. They are interested in both acute and chronic GVHD, which has overlapping features of immunodeficiency and symptoms of naturally occurring autoimmune disorders. Indeed, a prominent clinical feature of chronic GVHD is a debilitating fibrosing skin disease whose gross and histologic features resemble scleroderma (SSc) and, less commonly, morphea. Because of these potential biological similarities between chronic GVHD and autoimmune diseases, her work is also relevant.

Dr. Paczesy’s work to predict the risk of developing CVHD is another example of how IUPUI faculty are TRANSLATING RESEARCH INTO PRACTICE.

Browse

Recent Submissions

Now showing 1 - 10 of 45
  • Item
    Post-haematopoietic cell transplantation outcomes: why ST2 became a ‘golden nugget’ biomarker
    (Wiley, 2021-03) Paczesny, Sophie; Pediatrics, School of Medicine
    Immunotherapies have emerged as highly promising approaches to treat cancer patients. Allogeneic haematopoietic cell transplantation (HCT) is the most validated tumour immunotherapy available to date but its clinical efficacy is limited by toxicities, such as graft-versus-host disease (GVHD) and treatment resistance leading to relapse. The problems with new cellular therapies and checkpoint inhibitors are similar. However, development of biomarkers post-HCT, particularly for toxicities, has taken off in the last decade and has expanded greatly. Thanks to the advances in genomics, transcriptomics, proteomics and cytomics technologies, blood biomarkers have been identified and validated in promising diagnostic tests, prognostic tests stratifying for future occurrence of GVHD, and predictive tests for responsiveness to GVHD therapy and non-relapse mortality. These biomarkers may facilitate timely and selective therapeutic intervention. This review outlines a path from biomarker discovery to first clinical correlation, focusing on soluble STimulation-2 (sST2) – the interleukin (IL)-33-decoy receptor – which is the most validated biomarker.
  • Item
    The IL-33 Receptor/ST2 acts as a positive regulator of functional mouse bone marrow hematopoietic stem and progenitor cells
    (Elsevier, 2020-09) Capitano, Maegan L.; Griesenauer, Brad; Guo, Bin; Cooper, Scott; Paczesny, Sophie; Broxmeyer, Hal E.; Microbiology and Immunology, School of Medicine
    There is a paucity of information on a potential role for the IL-33 receptor/ST2 in the regulation of mouse bone marrow (BM) hematopoietic stem (HSC) and progenitor (HPC) cells. Comparing the BM of st2-/- and wild type (WT) control mice using functional assays, it was found that st2-/- BM cells had poorer engrafting capacity than WT BM in a competitive repopulating assay using congenic mice, with no changes in reconstitution of B-, T- and myeloid cells following transplantation. The BM of st2-/- mice also had fewer granulocyte-macrophage, erythroid, and multipotential progenitors than that of WT BM and these st2-/- HPC were in a slow cycling state compared to that of the rapidly cycling HPC of the WT mice. While functional assessment of HSC and HPC demonstrated that ST2 has a positive influence on regulation of HSC, we could not pick up differences in st2-/- compared to WT BM using only phenotypic analysis of HSC and HPC populations prior to transplantation, again demonstrating that phenotypic analysis of HSC and HPC do not always recapitulate the functional assessments of these immature hematopoietic cells.
  • Item
    Ex vivo culture of mouse skin activates an interleukin 1 alpha-dependent inflammatory response
    (Wiley, 2020-01) Zhou, Hong-Ming; Slominski, Radomir M.; Seymour, Leroy J.; Bell, Maria C.; Dave, Priya; Atumonye, Joseph; Wright, William, III.; Dawes, Avery; Griesenauer, Brad; Paczesny, Sophie; Kaplan, Mark H.; Spandau, Dan F.; Turner, Matthew J.; Dermatology, School of Medicine
    Ex vivo culture of mouse and human skin causes an inflammatory response characterized by production of multiple cytokines. We used ex vivo culture of mouse tail skin specimens to investigate mechanisms of this skin culture-induced inflammatory response. Multiplex assays revealed production of interleukin 1 alpha (IL-1α), interleukin 1 beta (IL-1β), interleukin 6 (IL-6), chemokine C-X-C motif ligand 1 (CXCL1), granulocyte colony-stimulating factor (G-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF) during skin culture, and quantitative PCR revealed transcripts for these proteins were also increased. Ex vivo cultures of skin from myeloid differentiation primary response 88 deficient mice (Myd88-/- ) demonstrated significantly reduced expression of transcripts for the aforementioned cytokines. The same result was observed with skin from interleukin 1 receptor type 1 deficient mice (Il1r1-/- ). These data suggested the IL-1R1/MyD88 axis is required for the skin culture-induced inflammatory response and led us to investigate the role of IL-1α and IL-1β (the ligands for IL-1R1) in this process. Addition of IL-1α neutralizing antibody to skin cultures significantly reduced expression of Cxcl1, Il6 and Csf3. IL-1β neutralization did not reduce levels of these transcripts. These studies suggest that IL-1α promotes the skin the culture-induced inflammatory response.
  • Item
    ICOSL+ plasmacytoid dendritic cells as inducer of graft-versus-host disease, responsive to a dual ICOS/CD28 antagonist
    (American Association for the Advancement of Science, 2020-10-07) Adom, Djamilatou; Dillon, Stacey R.; Yang, Jinfeng; Liu, Hao; Ramadan, Abdulraouf; Kushekhar, Kushi; Hund, Samantha; Albright, Amanda; Kirksey, Maykala; Adeniyan, Titilayo; Lewis, Katherine E.; Evans, Lawrence; Wu, Rebecca; Levin, Steven D.; Mudri, Sherri; Yang, Jing; Rickel, Erika; Seaberg, Michelle; Henderson, Katherine; Gudgeon, Chelsea J.; Wolfson, Martin F.; Swanson, Ryan M.; Swiderek, Kristine M.; Peng, Stanford L.; Hippen, Keli L.; Blazar, Bruce R.; Paczesny, Sophie; Pediatrics, School of Medicine
    Acute graft-versus-host disease (aGVHD) remains a major complication of allogeneic hematopoietic cell transplantation (HCT). CD146 and CCR5 are proteins that mark activated T helper 17 (Th17) cells. The Th17 cell phenotype is promoted by the interaction of the receptor ICOS on T cells with ICOS ligand (ICOSL) on dendritic cells (DCs). We performed multiparametric flow cytometry in a cohort of 156 HCT recipients and conducted experiments with aGVHD murine models to understand the role of ICOSL+ DCs. We observed an increased frequency of ICOSL+ plasmacytoid DCs, correlating with CD146+CCR5+ T cell frequencies, in the 64 HCT recipients with gastrointestinal aGVHD. In murine models, donor bone marrow cells from ICOSL-deficient mice compared to those from wild-type mice reduced aGVHD-related mortality. Reduced aGVHD resulted from lower intestinal infiltration of pDCs and pathogenic Th17 cells. We transplanted activated human ICOSL+ pDCs along with human peripheral blood mononuclear cells into immunocompromised mice and observed infiltration of intestinal CD146+CCR5+ T cells. We found that prophylactic administration of a dual human ICOS/CD28 antagonist (ALPN-101) prevented aGVHD in this model better than did the clinically approved belatacept (CTLA-4-Fc), which binds CD80 (B7-1) and CD86 (B7-2) and interferes with the CD28 T cell costimulatory pathway. When started at onset of aGVHD signs, ALPN-101 treatment alleviated symptoms of ongoing aGVHD and improved survival while preserving antitumoral cytotoxicity. Our data identified ICOSL+-pDCs as an aGVHD biomarker and suggest that coinhibition of the ICOSL/ICOS and B7/CD28 axes with one biologic drug may represent a therapeutic opportunity to prevent or treat aGVHD.
  • Item
    Donor HLA-E Status Associates with Disease-Free Survival and Transplant-Related Mortality after Non In Vivo T Cell-Depleted HSCT for Acute Leukemia
    (Elsevier, 2019-12) Tsamadou, Chrysanthi; Fürst, Daniel; Wan, Tao; He, Naya; Lee, Stephanie J.; Spellman, Stephen R.; Fleischhauer, Katharina; Hsu, Katharine C.; Paczesny, Sophie; Verneris, Michael R.; Schrezenmeier, Hubert; Mytilineos, Joannis; Pediatrics, School of Medicine
    Previous studies have suggested that HLA-E may have a significant role in the outcome of matched unrelated hematopoietic stem cell transplantation (HSCT), especially for patients with acute leukemia. We used Center for International Blood and Marrow Transplant Research data and samples of 1840 adult patients with acute leukemia and their 10/10 HLA-matched unrelated donors to investigate the impact of HLA-E matching status as well as of donor/recipient (D/R) HLA-E genotype on post-HSCT outcome. Both patients and donors were HLA-E genotyped by next-generation sequencing. All patients received their first transplant in complete remission between 2000 and 2015. Median follow-up time was 90 months. Overall survival, disease-free survival (DFS), transplant-related mortality (TRM), and relapse incidence were primary endpoints with statistical significance set at .01. D/R HLA-E genotype analysis revealed a significant association of donor HLA-E*01:03/01:03 genotype with DFS (hazard ratio [HR] = 1.35, P = .0006) and TRM (HR= 1.41, P = .0058) in patients who received T cell replete (ie, without in vivo T cell depletion) transplants (n = 1297). As for D/R HLA-E matching, we did not identify any significant effect on any of the clinical outcome endpoints. In conclusion, this is the largest study to date reporting an improvement of DFS and TRM after matched unrelated HSCT by avoidance of HLA-E*01:03 homozygous donors in patients transplanted with T cell replete grafts for acute leukemia.
  • Item
    Clinical Proteomics for Post-Hematopoeitic Stem Cell Transplantation Outcomes
    (Wiley, 2019-03) Paczesny, Sophie; Metzger, Jochen; Pediatrics, School of Medicine
    Allogeneic hematopoietic stem cell transplantation (HSCT) is the most effective form of tumor immunotherapy available to date. However, while HSCT can induce beneficial graft-versus-leukemia (GVL) effect, the adverse effect of graft-versus-host disease (GVHD), which is closely linked to GVL, is the major source of morbidity and mortality following HSCT. Until recently, available diagnostic and staging tools frequently fail to identify those at higher risk of disease progression or death. Furthermore, there are shortcomings in the prediction of the need for therapeutic interventions or the response rates to different forms of therapy. The past decade has been characterized by an explosive evolution of proteomics technologies, largely due to important advances in high-throughput MS instruments and bioinformatics. Building on these opportunities, blood biomarkers have been identified and validated both as promising diagnostic tools, prognostic tools that risk-stratify patients before future occurrence of GVHD and as predictive tools for responsiveness to GVHD therapy and non-relapse mortality. These biomarkers might facilitate timely and selective therapeutic intervention. This review summarizes current information on clinical proteomics for GVHD as well as other complications following HSCT. Finally, it proposes future directions for the translation of clinical proteomics to discovery of new potential therapeutic targets to the development of drugs.
  • Item
    Granzyme A–producing T helper cells are critical for acute graft-versus-host disease
    (American Society for Clinical Investigation, 2020-08-18) Park, Sungtae; Griesenauer, Brad; Jiang, Hua; Adom, Djamilatou; Mehrpouya-Bahrami, Pegah; Chakravorty, Srishti; Kazemian, Majid; Imam, Tanbeena; Srivastava, Rajneesh; Hayes, Tristan A.; Pardo, Julian; Janga, Sarath Chandra; Paczesny, Sophie; Kaplan, Mark H.; Olson, Matthew R.; Microbiology and Immunology, School of Medicine
    Acute graft-versus-host disease (aGVHD) can occur after hematopoietic cell transplant in patients undergoing treatment for hematological malignancies or inborn errors. Although CD4+ T helper (Th) cells play a major role in aGVHD, the mechanisms by which they contribute, particularly within the intestines, have remained elusive. We have identified a potentially novel subset of Th cells that accumulated in the intestines and produced the serine protease granzyme A (GrA). GrA+ Th cells were distinct from other Th lineages and exhibited a noncytolytic phenotype. In vitro, GrA+ Th cells differentiated in the presence of IL-4, IL-6, and IL-21 and were transcriptionally unique from cells cultured with either IL-4 or the IL-6/IL-21 combination alone. In vivo, both STAT3 and STAT6 were required for GrA+ Th cell differentiation and played roles in maintenance of the lineage identity. Importantly, GrA+ Th cells promoted aGVHD-associated morbidity and mortality and contributed to crypt destruction within intestines but were not required for the beneficial graft-versus-leukemia effect. Our data indicate that GrA+ Th cells represent a distinct Th subset and are critical mediators of aGVHD.
  • Item
    GVHD risk assessment beyond current HLA evaluation
    (Elsevier, 2020-01) Paczesny, Sophie; Pediatrics, School of Medicine
    As one of the most clinically validated immunotherapies to date, allogeneic haemopoietic cell transplantation (HCT) is a potentially curative option for blood cancers via the graft-versus-leukaemia effect. However, T-cell reactivity to alloantigens in normal host tissues also gives rise to graft-versus-host disease (GVHD), particularly in transplantation recipients from unrelated donors, accounting for more than 20% of deaths in patients who have a HCT. To limit GVHD, clinicians have been matching the patients and the unrelated donor graft the best they can using HLA-A, HLA-B, HLA-C, HLA-DRB1, and HLA-DQB1. However, this matching is not always possible, particularly for patients of non-white backgrounds who are not well represented in the donor registry. HCT from donors with one HLA mismatch followed by standard GVHD prophylaxis can be done, but is often followed by severe acute GVHD. The question of why is addressed by Effie Petersdorf and colleagues in The Lancet Haematology.
  • Item
    ST2 as checkpoint target for colorectal cancer immunotherapy
    (American Society for Clinical Investigation, 2020-05-07) Jeught, Kevin Van der; Sun, Yifan; Fang, Yuanzhang; Zhou, Zhuolong; Jiang, Hua; Yu, Tao; Yang, Jinfeng; Kamocka, Malgorzata M.; So, Ka Man; Li, Yujing; Eyvani, Haniyeh; Sandusky, George E.; Frieden, Michael; Braun, Harald; Beyaert, Rudi; He, Xiaoming; Zhang, Xinna; Zhang, Chi; Paczesny, Sophie; Lu, Xiongbin; Pediatrics, School of Medicine
    Immune checkpoint blockade immunotherapy delivers promising clinical results in colorectal cancer (CRC). However, only a fraction of cancer patients develop durable responses. The tumor microenvironment (TME) negatively impacts tumor immunity and subsequently clinical outcomes. Therefore, there is a need to identify other checkpoint targets associated with the TME. Early-onset factors secreted by stromal cells as well as tumor cells often help recruit immune cells to the TME, among which are alarmins such as IL-33. The only known receptor for IL-33 is stimulation 2 (ST2). Here we demonstrated that high ST2 expression is associated with poor survival and is correlated with low CD8+ T cell cytotoxicity in CRC patients. ST2 is particularly expressed in tumor-associated macrophages (TAMs). In preclinical models of CRC, we demonstrated that ST2-expressing TAMs (ST2+ TAMs) were recruited into the tumor via CXCR3 expression and exacerbated the immunosuppressive TME; and that combination of ST2 depletion using ST2-KO mice with anti–programmed death 1 treatment resulted in profound growth inhibition of CRC. Finally, using the IL-33trap fusion protein, we suppressed CRC tumor growth and decreased tumor-infiltrating ST2+ TAMs. Together, our findings suggest that ST2 could serve as a potential checkpoint target for CRC immunotherapy.
  • Item
    Early high plasma ST2, the decoy IL-33 receptor, in children undergoing hematopoietic cell transplantation is associated with the development of post-transplant diabetes mellitus
    (Ferrata Storti Foundation, 2020-05) Rowan, Courtney M.; Teagarden, Alicia M.; Cater, Daniel T.; Moser, Elizabeth A.S.; Baykoyanni, Giorgos; Paczesny, Sophie; Pediatrics, School of Medicine