Network


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

Hotspot


Dive into the research topics where Annkristin Heine is active.

Publication


Featured researches published by Annkristin Heine.


Blood | 2013

The JAK-inhibitor ruxolitinib impairs dendritic cell function in vitro and in vivo

Annkristin Heine; Stefanie Andrea Erika Held; Solveig Nora Daecke; Stephanie Wallner; Sowmya Parampalli Yajnanarayana; Christian Kurts; Dominik Wolf; Peter Brossart

The Janus kinase (JAK)-inhibitor ruxolitinib decreases constitutional symptoms and spleen size of myelofibrosis (MF) patients by mechanisms distinct from its anticlonal activity. Here we investigated whether ruxolitinib affects dendritic cell (DC) biology. The in vitro development of monocyte-derived DCs was almost completely blocked when the compound was added throughout the differentiation period. Furthermore, when applied solely during the final lipopolysaccharide-induced maturation step, ruxolitinib reduced DC activation as demonstrated by decreased interleukin-12 production and attenuated expression of activation markers. Ruxolitinib also impaired both in vitro and in vivo DC migration. Dysfunction of ruxolitinib-exposed DCs was further underlined by their impaired induction of allogeneic and antigen-specific T-cell responses. Ruxolitinib-treated mice immunized with ovalbumin (OVA)/CpG induced markedly reduced in vivo activation and proliferation of OVA-specific CD8⁺ T cells compared with vehicle-treated controls. Finally, using an adenoviral infection model, we show that ruxolitinib-exposed mice exhibit delayed adenoviral clearance. Our results demonstrate that ruxolitinib significantly affects DC differentiation and function leading to impaired T-cell activation. DC dysfunction may result in increased infection rates in ruxolitinib-treated patients. However, our findings may also explain the outstanding anti-inflammatory and immunomodulating activity of JAK inhibitors currently used in the treatment of MF and autoimmune diseases.


Blood | 2013

Ruxolitinib is a potent immunosuppressive compound: is it time for anti-infective prophylaxis?

Annkristin Heine; Peter Brossart; Dominik Wolf

To the editor: Treatment with the Janus-activated kinase (JAK) 1 and 2 inhibitor ruxolitinib decreases constitutional symptoms and spleen size in myelofibrosis. However, accumulating evidence suggests that the drug also exerts substantial immunosuppressive activity.[1][1],[2][2] A very recent


Cancer Research | 2015

JAK Inhibition Impairs NK Cell Function in Myeloproliferative Neoplasms

Kathrin Schönberg; Janna Rudolph; Maria Vonnahme; Sowmya Parampalli Yajnanarayana; Isabelle Cornez; Maryam Hejazi; Angela R. Manser; Markus Uhrberg; W. Verbeek; Steffen Koschmieder; Tim H. Brümmendorf; Peter Brossart; Annkristin Heine; Dominik Wolf

Ruxolitinib is a small-molecule inhibitor of the JAK kinases, which has been approved for the treatment of myelofibrosis, a rare myeloproliferative neoplasm (MPN), but clinical trials are also being conducted in inflammatory-driven solid tumors. Increased infection rates have been reported in ruxolitinib-treated patients, and natural killer (NK) cells are immune effector cells known to eliminate both virus-infected and malignant cells. On this basis, we sought to compare the effects of JAK inhibition on human NK cells in a cohort of 28 MPN patients with or without ruxolitinib treatment and 24 healthy individuals. NK cell analyses included cell frequency, receptor expression, proliferation, immune synapse formation, and cytokine signaling. We found a reduction in NK cell numbers in ruxolitinib-treated patients that was linked to the appearance of clinically relevant infections. This reduction was likely due to impaired maturation of NK cells, as reflected by an increased ratio in immature to mature NK cells. Notably, the endogenous functional defect of NK cells in MPN was further aggravated by ruxolitinib treatment. In vitro data paralleled these in vivo results, showing a reduction in cytokine-induced NK cell activation. Further, reduced killing activity was associated with an impaired capacity to form lytic synapses with NK target cells. Taken together, our findings offer compelling evidence that ruxolitinib impairs NK cell function in MPN patients, offering an explanation for increased infection rates and possible long-term side effects associated with ruxolitinib treatment.


British Journal of Haematology | 2015

JAK1/2 inhibition impairs T cell function in vitro and in patients with myeloproliferative neoplasms.

Sowmya Parampalli Yajnanarayana; Thomas Stübig; Isabelle Cornez; Haefaa Alchalby; Kathrin Schönberg; Janna Rudolph; Ioanna Triviai; Christine Wolschke; Annkristin Heine; Peter Brossart; Nicolaus Kröger; Dominik Wolf

Ruxolitinib (INCB018424) is the first JAK1/JAK2 inhibitor approved for treatment of myelofibrosis. JAK/STAT‐signalling is known to be involved in the regulation of CD4+ T cells, which critically orchestrate inflammatory responses. To better understand how ruxolitinib modulates CD4+ T cell responses, we undertook an in‐depth analysis of CD4+ T cell function upon ruxolitinib exposure. We observed a decrease in total CD3+ cells after 3 weeks of ruxolitinib treatment in patients with myeloproliferative neoplasms. Moreover, we found that the number of regulatory T cells (Tregs), pro‐inflammatory T‐helper cell types 1 (Th1) and Th17 were reduced, which were validated by in vitro studies. In line with our in vitro data, we found that inflammatory cytokines [tumour necrosis factor‐α (TNF), interleukin (IL)5, IL6, IL1B] were also downregulated in T cells from patients (all P < 0·05). Finally, we showed that ruxolitinib does not interfere with the T cell receptor signalling pathway, but impacts IL2‐dependent STAT5 activation. These data provide a rationale for testing JAK inhibitors in diseases triggered by hyperactive CD4+ T cells, such as autoimmune diseases. In addition, they also provide a potential explanation for the increased infection rates (i.e. viral reactivation and urinary tract infection) seen in ruxolitinib‐treated patients.


Leukemia | 2011

Immunomodulatory effects of anti-angiogenic drugs

Annkristin Heine; Stefanie Ae Held; Anita Bringmann; Tobias A. W. Holderried; Peter Brossart

Much progress and significant therapeutic changes have been made in the field of tumor therapy in the past decades. Besides chemotherapy and radiotherapy, a special focus was laid on targeted therapies such as small molecule tyrosine kinase inhibitors (TKIs) and other immunomodulatory drugs, which have become standard therapies and important combination partners in a variety of malignancies. In contrast to the widely established use of these often anti-angiogenic drugs, many functional molecular mechanisms are yet not completely understood. Recent analyses focused not only on their direct anti-tumor responses, but also on their influence on tumor microenvironment, as well as on their effects on malignant and healthy cells. Different anti-angiogenic compounds targeting the vascular endothelial growth factor (VEGF) or platelet-derived growth factor pathways seem to be capable of modulating immune responses, in a positive, as well as apparently harmful manner. For an optimal clinical anti-cancer treatment, a better understanding of these immunomodulatory effects is necessary. Here we summarize recent reports on the immunomodulatory function of lately introduced clinically applied anti-angiogenic compounds, such as the humanized monoclonal antibody against VEGF bevacizumab, the small molecule TKIs sunitinib, sorafenib, imatinib, dasatinib, nilotinib and the proteasome inhibitor bortezomib.


BioMed Research International | 2010

RNA Vaccines in Cancer Treatment

Anita Bringmann; Stefanie Andrea Erika Held; Annkristin Heine; Peter Brossart

The Cancer Report from the World Health Organization states that in the year 2000 12% of all death cases worldwide were caused by cancer. In the western world, the cancer death rates are often devastating, being at about 25%. This fact stresses the urgency to find effective cures against malignant diseases. New approaches in the treatment of cancer focus on the development of immunotherapies to fight the disease. Besides other methods, the usage of tumor-specific RNA as part of vaccines is investigated lately. RNA, administered alone or used for transfection of dendritic cells, shows several advantages as a vaccine including feasibility, applicability, safeness, and effectiveness when it comes to the generation of immune responses. This review concentrates on results from in vitro experiments and recent trials using RNA vaccines to present an overview about this specific strategy.


Clinical Cancer Research | 2008

Matrilysin (MMP-7) Is a Novel Broadly Expressed Tumor Antigen Recognized by Antigen-Specific T Cells

Yuko Yokoyama; Frank Grünebach; Susanne M. Schmidt; Annkristin Heine; Maik Häntschel; Stefan Stevanovic; Hans-Georg Rammensee; Peter Brossart

Purpose: A prerequisite for the development of vaccination strategies is the identification and characterization of relevant tumor-associated antigen. Using microarray and reverse transcription-PCR analysis, we found matrix metalloproteinase (MMP)-7 to be extensively up-regulated in renal cell carcinomas and expressed in a broad variety of malignant cells. MMP-7 can promote cancer invasion and angiogenesis by proteolytic cleavage of extracellular matrix and basement membrane proteins, thus making it a promising target in the context of immunotherapies. Experimental Design: To analyze the possible use of MMP-7 as a tumor-associated antigen, specific CTLs were induced using monocyte-derived dendritic cells electroporated with MMP-7-mRNA. In addition, to better characterize the fine specificity of these CTLs, MMP-7 MHC class I ligands were isolated and characterized in renal cell carcinoma tissue, which overexpressed MMP-7, by mass spectrometry–based peptide sequencing. Using this approach, we identified a novel HLA-A3–binding antigenic MMP-7 peptide. CTLs generated from healthy donors by in vitro priming with dendritic cells, pulsed with the novel peptide, were used as effectors in 51Cr-release assays. Results: The induced CTLs elicited an antigen-specific and HLA-restricted cytolytic activity against tumor cells endogenously expressing the MMP-7 protein. Furthermore, we were able to induce MMP-7–specific CTLs using peripheral blood mononuclear cells from a patient with acute lymphoblastic leukemia capable of recognizing the autologous leukemic blasts while sparing nonmalignant cells. Conclusions: Our study describes the identification of a novel broadly expressed T-cell epitope derived from the MMP-7 protein that represents an interesting candidate to be applied in immunotherapies of human malignancies targeting both tumor cells and neovascularization.


Blood | 2011

Regulation of dectin-1–mediated dendritic cell activation by peroxisome proliferator–activated receptor-gamma ligand troglitazone

Grethe Kock; Anita Bringmann; Stefanie Andrea Erika Held; Solveig Nora Daecke; Annkristin Heine; Peter Brossart

Dectin-1 is the major receptor for fungal β-glucans. The activation of Dectin-1 leads to the up-regulation of surface molecules on dendritic cells (DCs) and cytokine secretion. Furthermore, Dectin-1 is important for the recruitment of leukocytes and the production of inflammatory mediators. Peroxisome proliferator-activated receptor-γ (PPAR-γ) and its ligands, cyclopentenone prostaglandins or thiazolidinediones, have modulatory effects on B-cell, T-cell, and DC function. In the present study, we analyzed the effects of troglitazone (TGZ), a high-affinity synthetic PPAR-γ ligand, on the Dectin-1-mediated activation of monocyte-derived human DCs. Dectin-1-mediated activation of DCs was inhibited by TGZ, as shown by down-regulation of costimulatory molecules and reduced secretion of cytokines and chemokines involved in T-lymphocyte activation. Furthermore, TGZ inhibited the T-cell-stimulatory capacity of DCs. These effects were not due to a diminished expression of Dectin-1 or to a reduced phosphorylation of spleen tyrosine kinase; they were mediated by the inhibition of downstream signaling molecules such as mitogen-activated protein kinases and nuclear factor-κB. Furthermore, curdlan-mediated accumulation of caspase recruitment domain 9 (CARD9) in the cytosol was inhibited by TGZ. Our data demonstrate that the PPAR-γ ligand TGZ inhibits Dectin-1-mediated activation by interfering with CARD9, mitogen-activated protein kinase, and nuclear factor-κB signaling pathways. This confirms their important role as negative-feedback regulators of potentially harmful inflammatory responses.


Cancer Immunology, Immunotherapy | 2008

Generation of antigen-specific CTL responses using RGS1 mRNA transfected dendritic cells

Frank Grünebach; Stefanie Erndt; Maik Häntschel; Annkristin Heine; Peter Brossart

Advances in tumor immunology and Identification of tumor-associated antigens (TAAs) provide a basis for the development of novel immunotherapies to treat malignant diseases. In order to identify novel TAAs, we performed comparative microarray analysis of (heterogeneous) tissues and found regulator of G protein-signaling 1 (RGS1) extensively up-regulated in renal cell carcinoma (RCC) tissues. To examine the possible function of this molecule as a novel, broadly applicable TAA, synthetic full-length RGS1-mRNA was synthesized for the transfection of monocyte-derived dendritic cells (DCs). These modified antigen-presenting cells (APCs) were then used to induce RGS1-specific cytotoxic T cells (CTLs) in vitro. The CTLs generated from several healthy donors and a patient with chronic lymphocytic leukemia (CLL) elicited an antigen-specific and HLA-A2- and -A3-restricted cytolytic activity against tumor cells endogenously expressing the RGS1 protein including renal cell carcinomas (RCCs), melanoma, ovarian carcinoma and the primary autologous CLL-blasts. In conclusion, our study demonstrates that the in vitro induction of RGS1-specific CTLs by RNA-transfected DCs is feasible and highly effective. Since this molecule is (over-) expressed in a broad variety of malignancies it might represent an interesting novel TAA in the context of cancer vaccines designed to target RGS1 expressing tumor cells.


Case Reports in Oncology | 2016

Development of Hemolytic Anemia in a Nivolumab-Treated Patient with Refractory Metastatic Squamous Cell Skin Cancer and Chronic Lymphatic Leukemia

K.S. Schwab; Annkristin Heine; T. Weimann; G. Kristiansen; Peter Brossart

Management of patients with metastatic squamous cell skin cancer, refractory to initial therapy with standard chemotherapy and radiation protocols, remains difficult with poor overall prognosis and limited therapeutic options. Recently, promising response rates with nivolumab, a programmed death receptor-1-blocking antibody, in squamous cancer of the head and neck have been demonstrated. Considering the similar histological patterns of squamous cell cancer of the skin and squamous cell cancer of the head and neck, we assumed that nivolumab could also be effective in our patients with refractory metastatic squamous cell cancer of the skin. So far, there have been no clinical data on the therapeutic efficacy of nivolumab in squamous cell skin cancer. We here present a case of a patient with metastatic squamous cell skin cancer refractory to previous therapies, who showed a good response to nivolumab over a period of 5 months, but developed a serious hemolytic crisis under nivolumab treatment after eight applications.

Collaboration


Dive into the Annkristin Heine's collaboration.

Top Co-Authors

Avatar

Peter Brossart

University Hospital Bonn

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dominik Wolf

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dominik Wolf

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Janna Rudolph

University Hospital Bonn

View shared research outputs
Researchain Logo
Decentralizing Knowledge