Network


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

Hotspot


Dive into the research topics where Myrna R. Nahas is active.

Publication


Featured researches published by Myrna R. Nahas.


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.


British Journal of Haematology | 2017

Bone marrow stroma protects myeloma cells from cytotoxic damage via induction of the oncoprotein MUC1.

Michal Bar-Natan; Dina Stroopinsky; Katarina Luptakova; Maxwell Douglas Coll; Arie Apel; Hasan Rajabi; Athalia Rachel Pyzer; Kristen Palmer; Michaela R. Reagan; Myrna R. Nahas; Rebecca Karp Leaf; Salvia Jain; Jon Arnason; Irene M. Ghobrial; Kenneth C. Anderson; Donald Kufe; Jacalyn Rosenblatt; David Avigan

Multiple myeloma (MM) is a lethal haematological malignancy that arises in the context of a tumour microenvironment that promotes resistance to apoptosis and immune escape. In the present study, we demonstrate that co‐culture of MM cells with stromal cells results in increased resistance to cytotoxic and biological agents as manifested by decreased rates of cell death following exposure to alkylating agents and the proteosome inhibitor, bortezomib. To identify the mechanism of increased resistance, we examined the effect of the co‐culture of MM cells with stroma cells, on expression of the MUC1 oncogene, known to confer tumour cells with resistance to apoptosis and necrosis. Co‐culture of stroma with MM cells resulted in increased MUC1 expression by tumour cells. The effect of stromal cell co‐culture on MUC1 expression was not dependent on cell contact and was therefore thought to be due to soluble factors secreted by the stromal cells into the microenvironment. We demonstrated that MUC1 expression was mediated by interleukin‐6 and subsequent up‐regulation of the JAK‐STAT pathway. Interestingly, the effect of stromal cell co‐culture on tumour resistance was partially reversed by silencing of MUC1 in MM cells, consistent with the potential role of MUC1 in mediating resistance to cytotoxic‐based therapies.


Journal of Immunotherapy | 2017

DCOne as an Allogeneic Cell-based Vaccine for Multiple Myeloma

Rebecca Karp Leaf; Dina Stroopinsky; Athalia Rachel Pyzer; Ada M. Kruisbeek; Sandra van Wetering; Abigail Washington; Adam Ephraim; Leandra Cole; Adam Morin; Salvia Jain; Myrna R. Nahas; Arik Apel; Jon Arnason; Ayad Hamdan; Jacalyn Rosenblatt; David Avigan

Multiple myeloma (MM) is characterized by progressive immune dysregulation, loss of myeloma-specific immunity, and an immunosuppressive milieu that fosters disease growth and immune escape. Accordingly, cancer vaccines that reverse tumor-associated immune suppression represent a promising therapeutic avenue of investigation. We examined the potential of an allogeneic cellular vaccine to generate immune responses against MM tumor cells. The DCOne vaccine is comprised of a human myeloid leukemia cell line differentiated into a fully functional dendritic cell, expressing a range of tumor-associated antigens that are also known targets in MM. We found that the myeloma-specific antigens expressed by the DCOne vaccine can traffic via extracellular vesicles to surrounding antigen-presenting cells, thus stimulating autologous T-cell responses. Indeed, coculture of peripheral blood mononuclear cells from patients with MM with the DCOne vaccine resulted in the expansion of activated CD8+ T cells expressing interferon-&ggr; and perforin, with no significant change in the percentage of CD4+ T cells producing interleukin-10. Further, coculture of patient’s tumor cells with peripheral blood mononuclear cells and DCOne induced cytotoxic T-lymphocyte-mediated killing of autologous MM cells. These findings demonstrate that the allogeneic DCOne vaccine can induce T-cell activation and myeloma-specific immunity via cross presentation of antigens by native antigen-presenting cells.


Blood Reviews | 2018

Anti-cancer vaccine therapy for hematologic malignancies: An evolving era

Myrna R. Nahas; Jacalyn Rosenblatt; Hillard M. Lazarus; David Avigan

The potential promise of therapeutic vaccination as effective therapy for hematologic malignancies is supported by the observation that allogeneic hematopoietic cell transplantation is curative for a subset of patients due to the graft-versus-tumor effect mediated by alloreactive lymphocytes. Tumor vaccines are being explored as a therapeutic strategy to re-educate host immunity to recognize and target malignant cells through the activation and expansion of effector cell populations. Via several mechanisms, tumor cells induce T cell dysfunction and senescence, amplifying and maintaining tumor cell immunosuppressive effects, resulting in failure of clinical trials of tumor vaccines and adoptive T cell therapies. The fundamental premise of successful vaccine design involves the introduction of tumor-associated antigens in the context of effective antigen presentation so that tolerance can be reversed and a productive response can be generated. With the increasing understanding of the role of both the tumor and tumor microenvironment in fostering immune tolerance, vaccine therapy is being explored in the context of immunomodulatory therapies. The most effective strategy may be to use combination therapies such as anti-cancer vaccines with checkpoint blockade to target critical aspects of this environment in an effort to prevent the re-establishment of tumor tolerance while limiting toxicity associated with autoimmunity.


Expert Opinion on Biological Therapy | 2016

Challenges in vaccine therapy in hematological malignancies and strategies to overcome them

Myrna R. Nahas; David Avigan

ABSTRACT Introduction: Hematological malignancies (HM) are a promising platform for immunotherapy when considering the marked durable remissions historically achieved through allogeneic stem cell transplantation in select patients. Both non-cell and cell-based vaccine models have been utilized to elicit T cell tumor-specific eradication of malignant cells with resultant striking immunologic effects, but only modest clinical outcomes. In the last decade, the field of oncology has garnered greater insight into the complex mechanisms underpinning immune dysregulation in HM. Areas covered: This review addresses the development of vaccine strategies for HM examining the challenges of effectively inducing tumor-specific immunity and overcoming the barriers created by the tumor microenvironment. Expert opinion: Through a better understanding of the tumor immunosuppressive milieu and immunobiology of HM, rational immunotherapeutic combination therapies can be designed incorporating the potency of vaccine therapy to stimulate native immune responses. In current practice, the use of combinatorial immunotherapies in the treatment of HM is becoming more recognized. This strategy, with vaccines as the backbone, will likely lead to paradigm-changing therapeutic regimens in the decades to come, affording HM patients with durable remissions and improved quality of life.


Journal of Cellular and Molecular Medicine | 2018

MUC1‐C drives myeloid leukaemogenesis and resistance to treatment by a survivin‐mediated mechanism

Dina Stroopinsky; Hasan Rajabi; Myrna R. Nahas; Jacalyn Rosenblatt; Maryam Rahimian; Athalia Rachel Pyzer; Ashujit Tagde; Akriti Kharbanda; Salvia Jain; Turner Kufe; Rebecca Karp Leaf; Eleni Anastasiadou; Michal Bar-Natan; Shira Orr; Maxwell Douglas Coll; Kristen Palmer; Adam Ephraim; Leandra Cole; Abigail Washington; Donald Kufe; David Avigan

Acute myeloid leukaemia (AML) is an aggressive haematological malignancy with an unmet need for improved therapies. Responses to standard cytotoxic therapy in AML are often transient because of the emergence of chemotherapy‐resistant disease. The MUC1‐C oncoprotein governs critical pathways of tumorigenesis, including self‐renewal and survival, and is aberrantly expressed in AML blasts and leukaemia stem cells (LSCs). However, a role for MUC1‐C in linking leukaemogenesis and resistance to treatment has not been described. In this study, we demonstrate that MUC1‐C overexpression is associated with increased leukaemia initiating capacity in an NSG mouse model. In concert with those results, MUC1‐C silencing in multiple AML cell lines significantly reduced the establishment of AML in vivo. In addition, targeting MUC1‐C with silencing or pharmacologic inhibition with GO‐203 led to a decrease in active β‐catenin levels and, in‐turn, down‐regulation of survivin, a critical mediator of leukaemia cell survival. Targeting MUC1‐C was also associated with increased sensitivity of AML cells to Cytarabine (Ara‐C) treatment by a survivin‐dependent mechanism. Notably, low MUC1 and survivin gene expression were associated with better clinical outcomes in patients with AML. These findings emphasize the importance of MUC1‐C to myeloid leukaemogenesis and resistance to treatment by driving survivin expression. Our findings also highlight the potential translational relevance of combining GO‐203 with Ara‐C for the treatment of patients with AML.


Blood | 2018

Phase 1 clinical trial evaluating abatacept in patients with steroid-refractory chronic graft-versus-host disease

Myrna R. Nahas; Robert J. Soiffer; Haesook T. Kim; Edwin P. Alyea; Jon Arnason; Robin Joyce; Joseph H. Antin; Vincent T. Ho; Dina Stroopinsky; Shuli Li; James D. Levine; Malgorzata McMasters; Salvia Jain; Ayad Hamdan; Dimitrios Tzachanis; Mary Paty Bryant; Emma Logan; Josie Bazemore; Jeremy G. Stewart; Amy Joyce; Susan Stephenson; Abigail Washington; Leandra Cole; Athalia Rachel Pyzer; Rebecca Karp Leaf; David Avigan; Jacalyn Rosenblatt

Steroid-refractory chronic graft-versus-host disease (SR-cGVHD) remains a major cause of morbidity and mortality after allogeneic stem cell transplantation. Innovative immunotherapeutic strategies are urgently needed for the treatment of SR-cGVHD. We conducted a phase 1 clinical trial to evaluate the safety, efficacy, and immune effects of abatacept, a novel immunomodulatory drug that acts as an inhibitor of T-cell activation via costimulatory blockade, in the treatment of SR-cGVHD. The study followed a 3+3 design with 2 escalating abatacept doses: 3 mg/kg and 10 mg/kg, with an expansion cohort treated at 10 mg/kg. Abatacept was well-tolerated with no dose-limiting toxicities. Of the 16 evaluable patients, 44% achieved a clinical partial response per 2005 National Institutes of Health Consensus Criteria. Importantly, abatacept resulted in a 51.3% reduction in prednisone usage in clinical responders (mean baseline, 27 vs 14 mg; P = .01). Increased PD-1 expression on circulating CD4 (P = .009) and CD8 (P = .007) T cells was observed in clinical responders. In summary, abatacept was safe and led to a marked improvement in National Institutes of Health cGVHD scores and a significant reduction in prednisone use. In this cohort of heavily pretreated patients, the results suggest abatacept may be a promising therapeutic agent for SR-cGVHD, and a phase 2 trial has been initiated. This trial was registered at www.clinicaltrials.gov as #NCT01954979.


ACG Case Reports Journal | 2018

Posttransplant Lymphoproliferative Disorder Isolated to the Adrenal Gland in a Liver Transplant Patient

Tara T Ghaziani; Joy Liu; Zhenghui G. Jiang; Imad Nasser; Khalid Khwaja; Robert A. Fisher; Myrna R. Nahas; Michael P. Curry

Posttransplant lymphoproliferative disorder (PTLD) is a serious complication that accounts for up to 20% of malignancies after solid organ transplantation. We describe a rare case of isolated PTLD in the adrenal gland occurring 7 months after liver transplant in a patient who developed a primary Epstein-Barr virus infection. He was treated with rituximab and his immunosuppression regimen was minimized. We review the incidence, pathogenesis, presentation, and management of PTLD in the liver-transplant population. Our case highlights the variation in the presentation of PTLD and the importance of a high index of suspicion among the at-risk group.


Molecular Cancer Therapeutics | 2017

Decitabine Priming Enhances Mucin 1 Inhibition Mediated Disruption of Redox Homeostasis in Cutaneous T-cell Lymphoma

Salvia Jain; Abigail Washington; Rebecca Karp Leaf; Parul Bhargava; Rachael A. Clark; Thomas S. Kupper; Dina Stroopinsky; Athalia Rachel Pyzer; Leandra Cole; Myrna R. Nahas; Arie Apel; Jacalyn Rosenblatt; Jon Arnason; Donald Kufe; David Avigan

Cutaneous T-cell lymphoma (CTCL) is a heterogeneous neoplasm and patients with relapsed/refractory disease exhibit resistance to standard therapies. We have previously demonstrated that the Mucin 1 C-terminal subunit (MUC1-C) plays a critical role in protection from oxidative stress in CTCL cells. Targeting of MUC1-C with a pharmacologic inhibitor, GO-203, was associated with apoptosis in CTCL. However, disease responses were incomplete underscoring the need for combinatorial strategies that could exploit the vulnerability of CTCL cells to oxidative signals. Cell lines, primary samples, and xenograft models of CTCL were used to assess synergy of GO-203 with decitabine, a hypomethylating agent. Present studies demonstrate that exposure of CTCL cells to decitabine in combination with GO-203, increased the generation of reactive oxygen species (ROS) levels and decreased levels of scavenger molecules, NADP, NADPH, glutathione, and TIGAR, critical to intracellular redox homeostasis. Dual exposure to GO-203 and decitabine resulted in marked downregulation of DNA methyl transferases demonstrating significant synergy of these agents in inducing global and gene specific hypomethylation. Accordingly, treatment with decitabine and GO-203 upregulated the ROS generating enzymes, NADPH oxidase 4 and dual oxidase 2 potentially due to their effect on epigenomic regulation of these proteins. In concert with these findings, exposure to decitabine and GO-203 resulted in heightened apoptotic death in CTCL cell lines, patient-derived primary samples and in a murine xenograft model. These findings indicate that decitabine intensifies MUC1-C inhibition induced redox imbalance and provides a novel combination of targeted and epigenetic agents for patients with CTCL. Mol Cancer Ther; 16(10); 2304–14. ©2017 AACR.

Collaboration


Dive into the Myrna R. Nahas'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

Athalia Rachel Pyzer

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

Dina Stroopinsky

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jon Arnason

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rebecca Karp Leaf

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Abigail Washington

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Leandra Cole

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge