Network


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

Hotspot


Dive into the research topics where Dina Stroopinsky is active.

Publication


Featured researches published by Dina Stroopinsky.


Cancer Immunology, Immunotherapy | 2013

Lenalidomide enhances anti-myeloma cellular immunity

Katarina Luptakova; Jacalyn Rosenblatt; Brett Glotzbecker; Heidi Mills; Dina Stroopinsky; Turner Kufe; Baldev Vasir; Jon Arnason; Dimitri Tzachanis; Jeffrey I. Zwicker; Robin Joyce; James D. Levine; Kenneth C. Anderson; Donald Kufe; David Avigan

Lenalidomide is an effective therapeutic agent for multiple myeloma that exhibits immunomodulatory properties including the activation of T and NK cells. The use of lenalidomide to reverse tumor-mediated immune suppression and amplify myeloma-specific immunity is currently being explored. In the present study, we examined the effect of lenalidomide on T-cell activation and its ability to amplify responses to a dendritic cell-based myeloma vaccine. We demonstrate that exposure to lenalidomide in the context of T-cell expansion with direct ligation of CD3/CD28 complex results in polarization toward a Th1 phenotype characterized by increased IFN-γ, but not IL-10 expression. In vitro exposure to lenalidomide resulted in decreased levels of regulatory T cells and a decrease in T-cell expression of the inhibitory marker, PD-1. Lenalidomide also enhanced T-cell proliferative responses to allogeneic DCs. Most significantly, lenalidomide treatment potentiated responses to the dendritic cell/myeloma fusion vaccine, which were characterized by increased production of inflammatory cytokines and increased cytotoxic lymphocyte-mediated lysis of autologous myeloma targets. These findings indicate that lenalidomide enhances the immunologic milieu in patients with myeloma by promoting T-cell proliferation and suppressing inhibitory factors, and thereby augmenting responses to a myeloma-specific tumor vaccine.


Science Translational Medicine | 2016

Individualized vaccination of AML patients in remission is associated with induction of antileukemia immunity and prolonged remissions

Jacalyn Rosenblatt; Richard Stone; Lynne Uhl; Donna Neuberg; Robin Joyce; James D. Levine; Jon Arnason; Malgorzata McMasters; Katarina Luptakova; Salvia Jain; Jeffrey I. Zwicker; Ayad Hamdan; Vassiliki A. Boussiotis; David P. Steensma; Daniel J. DeAngelo; Ilene Galinsky; Poorvi Somaiya Dutt; Emma Logan; Mary Paty Bryant; Dina Stroopinsky; Lillian Werner; Kristen Palmer; Max Coll; Abigail Washington; Leandra Cole; Donald Kufe; David Avigan

A personalized DC/AML fusion cell vaccine promotes the expansion of leukemia-specific T cells and prolonged remission in patients. Immune cells join leukemia then beat it Acute myeloid leukemia (AML) is an aggressive hematologic cancer. The only curative treatment available for this disease is hematopoietic stem cell transplantation, which can result in donor immune cells helping to eradicate the cancer. Unfortunately, this procedure is not always effective and is itself associated with numerous complications and risk of death. Rosenblatt et al. have identified a potentially better way to stimulate an immune response against AML by fusing patients’ own leukemia cells with dendritic cells. The resulting fusion cells were very effective at presenting tumor antigens to T cells, resulting in a strong antitumor T cell response and prolonged survival in human patients. We developed a personalized cancer vaccine in which patient-derived acute myeloid leukemia (AML) cells are fused with autologous dendritic cells, generating a hybridoma that potently stimulates broad antitumor responses. We report results obtained from the first 17 AML patients, who achieved remission after chemotherapy and were then serially vaccinated to target minimal residual disease and prevent relapse. Vaccination was well tolerated and induced inflammatory responses at the site of administration, characterized by the dense infiltration of T cells. Vaccination was also associated with a marked rise in circulating T cells recognizing whole AML cells and leukemia-specific antigens that persisted for more than 6 months. Twelve of 17 vaccinated patients (71%; 90% confidence interval, 52 to 89%) remain alive without recurrence at a median follow-up of 57 months. The results demonstrate that personalized vaccination of AML patients in remission induces the expansion of leukemia-specific T cells and may be protective against disease relapse.


Cancer Research | 2013

MUC1 Is a Potential Target for the Treatment of Acute Myeloid Leukemia Stem Cells

Dina Stroopinsky; Jacalyn Rosenblatt; Keisuke Ito; Heidi Mills; Li Yin; Hasan Rajabi; Baldev Vasir; Turner Kufe; Katarina Luptakova; Jon Arnason; Caterina Nardella; James D. Levine; Robin Joyce; Ilene Galinsky; Yoram Reiter; Richard Stone; Pier Paolo Pandolfi; Donald Kufe; David Avigan

Acute myeloid leukemia (AML) is a malignancy of stem cells with an unlimited capacity for self-renewal. MUC1 is a secreted, oncogenic mucin that is expressed aberrantly in AML blasts, but its potential uses to target AML stem cells have not been explored. Here, we report that MUC1 is highly expressed on AML CD34(+)/lineage(-)/CD38(-) cells as compared with their normal stem cell counterparts. MUC1 expression was not restricted to AML CD34(+) populations as similar results were obtained with leukemic cells from patients with CD34(-) disease. Engraftment of AML stem cell populations that highly express MUC1 (MUC1(high)) led to development of leukemia in NOD-SCID IL2Rgamma(null) (NSG) immunodeficient mice. In contrast, MUC1(low) cell populations established normal hematopoiesis in the NSG model. Functional blockade of the oncogenic MUC1-C subunit with the peptide inhibitor GO-203 depleted established AML in vivo, but did not affect engraftment of normal hematopoietic cells. Our results establish that MUC1 is highly expressed in AML stem cells and they define the MUC1-C subunit as a valid target for their therapeutic eradication.


European Journal of Immunology | 2009

Allogeneic induced human FOXP3+IFN‐γ+ T cells exhibit selective suppressive capacity

Dina Stroopinsky; Irit Avivi; Jacob M. Rowe; David Avigan; Tamar Katz

Human induced CD4+CD25+ T cells have been shown to express FOXP3, similar to naturally occurring Treg cells (nTreg). However, the suppressive capacity of these cells is still under debate. The current study was designed to investigate functional characteristics of CD25+FOXP3+ derived from CD25− T cells. Stimulation of CD25− PBMC with allogeneic PBMC resulted in production of CD4+CD25high T cells. This process was more rapid and prominent when highly mature DC were used for stimulation. The resultant CD4+CD25high population concurrently exhibited regulatory markers FOXP3, CTLA‐4, GITR, and inflammatory cytokines IL‐2 and IFN‐γ. These human‐induced FOXP3+IFN‐γ+ T cells were shown, for the first time, to markedly inhibit alloreactive T‐cell expansion, similar to nTreg. However, in contrast to nTreg, the induced CD4+CD25+FOXP3+ cells did not suppress proliferation against a third party donor stimulus or CMV. This suggested that the cell population possessed a more selective suppressive capacity targeted against the original stimulus only. The induced human CD4+CD25+FOXP3+ subset derived from CD25− T cells, while expressing inflammatory cytokines, exhibits a suppressive cell contact‐dependent effect, restricted against T cells responding to the original stimulus. Such unique properties suggest that these cells are potentially ideal for the use as post‐transplant GVH disease prophylaxis.


Blood Reviews | 2013

Anti-CD20 monoclonal antibodies: Beyond B-cells

Irit Avivi; Dina Stroopinsky; Tamar Katz

Anti-CD20 monoclonal antibodies (MoAbs), employed in treating CD20⁺ lymphomas and autoimmune diseases, appear to have broader functions than just eradicating malignant B-cells and decreasing autoantibody production. Rituximab-induced T-cell inactivation, reported both in-vitro and in-vivo, may contribute to the increased risk of T-cell-dependent infections, observed in patients receiving this therapy. T-cell polarization into a suppressive phenotype, often observed in patients receiving rituximab for autoimmune disorders, was reported to be associated with prolonged remissions. Elimination of B-cells serving as antigen-presenting cells, thereby causing impaired T-cell activation, could play a significant role in induction of these changes. Direct binding of rituximab to a CD20dim T-cell population, inducing its depletion, may contribute to the decreased T-cell activation following rituximab therapy. Further investigation of the complex network through which rituximab and new anti-CD20 MoAbs act, would advance the employment of these agents in different clinical settings.


Blood | 2015

Mucin 1 is a potential therapeutic target in cutaneous T-cell lymphoma

Salvia Jain; Dina Stroopinsky; Li Yin; Jacalyn Rosenblatt; Maroof Alam; Parul Bhargava; Rachael A. Clark; Thomas S. Kupper; Kristen Palmer; Maxwell Douglas Coll; Hasan Rajabi; Athalia Rachel Pyzer; Michal Bar-Natan; Katarina Luptakova; Jon Arnason; Robin Joyce; Donald Kufe; David Avigan

Cutaneous T-cell lymphoma (CTCL) is an aggressive neoplasm with limited treatments for patients with advanced disease. The mucin 1 C-terminal subunit (MUC1-C) oncoprotein plays a critical role in regulating cell proliferation, apoptosis, and protection from cytotoxic injury mediated by reactive oxygen species (ROS). Although CTCL cells exhibit resistance to ROS-induced apoptosis, the expression and functional significance of MUC1 in CTCL have not been previously investigated. Present studies demonstrate that MUC1-C is overexpressed in CTCL cell lines and primary CTCL cells but is absent in resting T cells from healthy donors and B-cell lymphoma cells. We have developed a cell-penetrating peptide that disrupts homodimerization of the MUC1-C subunit necessary for its nuclear translocation and downstream signaling. We show that treatment of CTCL cells with the MUC1-C inhibitor is associated with downregulation of the p53-inducible regulator of glycolysis and apoptosis and decreases in reduced NAD phosphate and glutathione levels. In concert with these results, targeting MUC1-C in CTCL cells increased ROS and, in turn, induced ROS-mediated late apoptosis/necrosis. Targeting MUC1-C in CTCL tumor xenograft models demonstrated significant decreases in disease burden. These findings indicate that MUC1-C maintains redox balance in CTCL cells and is thereby a novel target for the treatment of patients with CTCL.


Oncotarget | 2016

MUC1-C induces DNA methyltransferase 1 and represses tumor suppressor genes in acute myeloid leukemia

Ashujit Tagde; Hasan Rajabi; Dina Stroopinsky; Reddy Gali; Maroof Alam; Audrey Bouillez; Surender Kharbanda; Richard Stone; David Avigan; Donald Kufe

Aberrant DNA methylation is a hallmark of acute myeloid leukemia (AML); however, the regulation of DNA methyltransferase 1 (DNMT1), which is responsible for maintenance of DNA methylation patterns, has largely remained elusive. MUC1-C is a transmembrane oncoprotein that is aberrantly expressed in AML stem-like cells. The present studies demonstrate that targeting MUC1-C with silencing or a pharmacologic inhibitor GO-203 suppresses DNMT1 expression. In addition, MUC1 expression positively correlates with that of DNMT1 in primary AML cells, particularly the CD34+/CD38− population. The mechanistic basis for this relationship is supported by the demonstration that MUC1-C activates the NF-κB p65 pathway, promotes occupancy of the MUC1-C/NF-κB complex on the DNMT1 promoter and drives DNMT1 transcription. We also show that targeting MUC1-C substantially reduces gene promoter-specific DNA methylation, and derepresses expression of tumor suppressor genes, including CDH1, PTEN and BRCA1. In support of these results, we demonstrate that combining GO-203 with the DNMT1 inhibitor decitabine is highly effective in reducing DNMT1 levels and decreasing AML cell survival. These findings indicate that (i) MUC1-C is an attractive target for the epigentic reprogramming of AML cells, and (ii) targeting MUC1-C in combination with decitabine is a potentially effective clinical approach for the treatment of AML.


Leukemia | 2017

MUC1 inhibition leads to decrease in PD-L1 levels via upregulation of miRNAs

Athalia Rachel Pyzer; Dina Stroopinsky; Jacalyn Rosenblatt; Eleni Anastasiadou; Hasan Rajabi; Abigail Washington; Ashujit Tagde; Jen-Hwa Chu; Maxwell Douglas Coll; Alan L Jiao; Lt Tsai; De Tenen; Leandra Cole; Kristen Palmer; Adam Ephraim; Rebecca Karp Leaf; Myrna R. Nahas; Arie Apel; Michal Bar-Natan; Salvia Jain; Malgorzata McMasters; Lourdes Mendez; Jon Arnason; Benjamin Alexander Raby; Frank J. Slack; Donald Kufe; David Avigan

The PD-L1/PD-1 pathway is a critical component of the immunosuppressive tumor microenvironment in acute myeloid leukemia (AML), but little is known about its regulation. We investigated the role of the MUC1 oncoprotein in modulating PD-L1 expression in AML. Silencing of MUC1 in AML cell lines suppressed PD-L1 expression without a decrease in PD-L1 mRNA levels, suggesting a post-transcriptional mechanism of regulation. We identified the microRNAs miR-200c and miR-34a as key regulators of PD-L1 expression in AML. Silencing of MUC1 in AML cells led to a marked increase in miR-200c and miR-34a levels, without changes in precursor microRNA, suggesting that MUC1 might regulate microRNA-processing. MUC1 signaling decreased the expression of the microRNA-processing protein DICER, via the suppression of c-Jun activity. NanoString (Seattle, WA, USA) array of MUC1-silenced AML cells demonstrated an increase in the majority of probed microRNAs. In an immunocompetent murine AML model, targeting of MUC1 led to a significant increase in leukemia-specific T cells. In concert, targeting MUC1 signaling in human AML cells resulted in enhanced sensitivity to T-cell-mediated lysis. These findings suggest MUC1 is a critical regulator of PD-L1 expression via its effects on microRNA levels and represents a potential therapeutic target to enhance anti-tumor immunity.


Blood | 2017

MUC1 mediated induction of myeloid-derived suppressor cells in patients with acute myeloid leukemia.

Athalia Rachel Pyzer; Dina Stroopinsky; Hasan Rajabi; Abigail Washington; Ashujit Tagde; Maxwell Douglas Coll; Jacqueline Fung; Mary Paty Bryant; Leandra Cole; Kristen Palmer; Poorvi Somaiya; Rebecca Karp Leaf; Myrna R. Nahas; Arie Apel; Salvia Jain; Malgorzata McMasters; Lourdes Mendez; James D. Levine; Robin Joyce; Jon Arnason; Pier Paolo Pandolfi; Donald Kufe; Jacalyn Rosenblatt; David Avigan

Myeloid-derived suppressor cells (MDSCs) play a critical role in promoting immune tolerance and disease growth. The mechanism by which tumor cells evoke the expansion of MDSCs in acute myeloid leukemia (AML) has not been well described. We have demonstrated that patients with AML exhibit increased presence of MDSCs in their peripheral blood, in comparison with normal controls. Cytogenetic studies demonstrated that MDSCs in patients with AML may be derived from leukemic or apparently normal progenitors. Engraftment of C57BL/6 mice with TIB-49 AML led to an expansion of CD11b+ Gr1+ MDSCs in bone marrow and spleen. Coculture of the AML cell lines MOLM-4, THP-1 or primary AML cells with donor peripheral blood mononuclear cells elicited a cell contact-dependent expansion of MDSCs. MDSCs were suppressive of autologous T-cell responses as evidenced by reduced T-cell proliferation and a switch from a Th1 to a Th2 phenotype. We hypothesized that the expansion of MDSCs in AML is accomplished by tumor-derived extracellular vesicles (EVs). Using tracking studies, we demonstrated that AML EVs are taken-up myeloid progenitor cells, resulting in the selective proliferation of MDSCs in comparison with functionally competent antigen-presenting cells. The MUC1 oncoprotein was subsequently identified as the critical driver of EV-mediated MDSC expansion. MUC1 induces increased expression of c-myc in EVs that induces proliferation in the target MDSC population via downstream effects on cell cycle proteins. Moreover, we demonstrate that the microRNA miR34a acts as the regulatory mechanism by which MUC1 drives c-myc expression in AML cells and EVs.


Blood | 2014

MUC1-C oncoprotein promotes FLT3 receptor activation in acute myeloid leukemia cells

Suiyang Liu; Li Yin; Dina Stroopinsky; Hasan Rajabi; Alexandre Puissant; Kimberly Stegmaier; David Avigan; Surender Kharbanda; Donald Kufe; Richard Stone

Blasts from approximately one-third of patients with acute myeloid leukemia (AML) harbor activating mutations in the FMS-like tyrosine kinase 3 (FLT3) receptor tyrosine kinase that confer a poor prognosis. The Mucin 1-C-terminal subunit (MUC1-C) oncoprotein is aberrantly expressed in AML blasts and stem cells; however, there is no known interaction between MUC1-C and FLT3. The present studies demonstrate that MUC1-C associates with wild-type and mutant FLT3 in AML cells. Targeting MUC1-C with the cell-penetrating peptide inhibitor GO-203 disrupts MUC1-C/FLT3 complexes and downregulates FLT3 activation. GO-203 treatment of AML cells was also associated with inhibition of the FLT3 downstream effectors AKT, extracellular signal-regulated kinase, and STAT5. The results further show that AML cells with FLT3-activating mutations and resistant to the FLT3 inhibitor midostaurin/PKC412 are sensitive to GO-203-induced growth arrest and death. Moreover, GO-203 increases sensitivity of mutant FLT3 AML cells to FLT3 inhibitor treatment. These results indicate that MUC1-C contributes to FLT3 activation in AML cells and that targeting MUC1-C inhibits the FLT3 signaling pathway. Our findings support the development of MUC1-C inhibitors alone and in combination with agents that target FLT3 for the treatment of wild-type and mutant FLT3 AML.

Collaboration


Dive into the Dina Stroopinsky's collaboration.

Top Co-Authors

Avatar

David Avigan

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jacalyn Rosenblatt

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jon Arnason

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Katarina Luptakova

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

James D. Levine

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Robin Joyce

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Salvia Jain

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Athalia Rachel Pyzer

Beth Israel Deaconess Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge