Network


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

Hotspot


Dive into the research topics where Marina Moskalenko is active.

Publication


Featured researches published by Marina Moskalenko.


Cancer Immunology, Immunotherapy | 2008

Local secretion of anti-CTLA-4 enhances the therapeutic efficacy of a cancer immunotherapy with reduced evidence of systemic autoimmunity

Andrew Simmons; Marina Moskalenko; Jennifer Creson; Jianmin Fang; Saili Yi; Melinda VanRoey; James P. Allison; Karin Jooss

Promising anti-tumor responses have been observed in the clinic using monoclonal antibodies (mAbs) that block immune checkpoints. One concern with these therapeutic agents remains the potential induction of immune breakthrough events (IBEs) resulting from the disruption of T cell homeostasis or the breaking of tolerance to self antigens. As an approach to maintaining anti-tumor responses but decreasing the likelihood of these events, the local expression of a mAb in combination with a GM-CSF-secreting cancer immunotherapy was evaluated. Using anti-cytotoxic T lymphocyte antigen (CTLA)-4 as a model antibody to test this hypothesis, tumor cell lines were generated that expressed the full-length mAb in addition to GM-CSF. Evaluation of these cell lines in two therapeutic tumor models revealed that local, cell-mediated delivery of anti-CTLA-4 from a GM-CSF-secreting tumor cell immunotherapy activated potent anti-tumor responses and prolonged overall survival at significantly lower serum mAb levels in the host. Furthermore, lowering the systemic exposure of the host to the immune modulatory mAb correlated with reduced evidence of systemic autoimmunity. This approach has broad utility for the delivery of mAbs or proteins locally from cellular immunotherapies to minimize IBEs while retaining the potent therapeutic effects of such combination treatments.


Cancer Immunology, Immunotherapy | 2007

GM-CSF-secreting cancer immunotherapies: preclinical analysis of the mechanism of action

Andrew Simmons; Melissa Gonzalez-Edick; Carol Lin; Marina Moskalenko; Thomas Du; Jennifer Creson; Melinda VanRoey; Karin Jooss

Granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting tumor cell immunotherapies have demonstrated long-lasting, and specific anti-tumor immune responses in animal models. The studies reported here specifically evaluate two aspects of the immune response generated by such immunotherapies: the persistence of irradiated tumor cells at the immunization site, and the breadth of the immune response elicited to tumor associated antigens (TAA) derived from the immunotherapy. To further define the mechanism of GM-CSF-secreting cancer immunotherapies, immunohistochemistry studies were performed using the B16F10 melanoma tumor model. In contrast to previous reports, our data revealed that the irradiated tumor cells persisted and secreted high levels of GM-CSF at the injection site for more than 21 days. Furthermore, dense infiltrates of dendritic cells were observed only in mice treated with GM-CSF-secreting B16F10 cells, and not in mice treated with unmodified B16F10 cells with or without concurrent injection of rGM-CSF. In addition, histological studies also revealed enhanced neutrophil and CD4+ T cell infiltration, as well as the presence of apoptotic cells, at the injection site of mice treated with GM-CSF-secreting tumor cells. To evaluate the scope of the immune response generated by GM-CSF-secreting cancer immunotherapies, several related B16 melanoma tumor cell subclones that exist as a result of genetic drift in the original cell line were used to challenge mice previously immunized with GM-CSF-secreting B16F10 cells. These studies revealed that GM-CSF-secreting cancer immunotherapies elicit T cell responses that effectively control growth of related but antigenically distinct tumors. Taken together, these studies provide important new insights into the mechanism of action of this promising novel cancer immunotherapy.


Melanoma Research | 2013

Elevated rates of transaminitis during ipilimumab therapy for metastatic melanoma.

Sebastian Bernardo; Marina Moskalenko; Michael Pan; Shaily Shah; Harleen Sidhu; Serge Sicular; Sara Harcharik; Rui Chang; Philip Friedlander; Yvonne Saenger

Melanoma is the deadliest form of skin cancer. Ipilimumab, a novel immunotherapy, is the first treatment shown to improve survival in patients with metastatic melanoma in large randomized controlled studies. The most concerning side effects reported in clinical studies of ipilimumab fall into the category of immune-related adverse events, which include enterocolitis, dermatitis, thyroiditis, hepatitis, hypophysitis, uveitis, and others. During the course of routine clinical care at Mount Sinai Medical Center, frequent hepatotoxicity was noted when ipilimumab was administered at a dose of 3 mg/kg according to Food and Drug Administration (FDA) guidelines. To better characterize these adverse events, we conducted a retrospective review of the first 11 patients with metastatic melanoma treated with ipilimumab at the Mount Sinai Medical Center after FDA approval. Aspartate aminotransferase (AST) and alanine aminotransferase (ALT) elevation, as defined by the National Cancer Institute’s Common Terminology Criteria for Adverse Events, each occurred in six of 11 cases (≥grade 1), a notably higher frequency than could be expected on the basis of the FDA licensing study where elevations were reported in 0.8 and 1.5% of patients for AST and ALT, respectively. Grade 3 elevations in AST occurred in three of 11 patients as compared with 0% in the licensing trial. All cases of transaminitis resolved when ipilimumab was temporarily withheld without administration of immunosuppressive medication. During routine clinical care of late-stage melanoma patients with ipilimumab, physicians should monitor patients closely for hepatotoxicity and be aware that toxicity rates may differ across populations during ipilimumab therapy.


Journal of Investigative Dermatology | 2014

Dissection of Immune Gene Networks in Primary Melanoma Tumors Critical for Antitumor Surveillance of Patients with Stage II–III Resectable Disease

Shanthi Sivendran; Rui Chang; Lisa Pham; Robert G. Phelps; Sara Harcharik; Lawrence D. Hall; Sebastian Bernardo; Marina Moskalenko; Meera Sivendran; Yichun Fu; Ellen H. de Moll; Michael Pan; Jee Young Moon; Sonali Arora; Ariella Cohain; Analisa DiFeo; Tammie Ferringer; Mikhail Tismenetsky; Cindy L. Tsui; Philip Friedlander; Michael K. Parides; Jacques Banchereau; Damien Chaussabel; Mark Lebwohl; Jedd D. Wolchok; Nina Bhardwaj; Steven J. Burakoff; William Oh; Karolina Palucka; Miriam Merad

Patients with resected stage II-III cutaneous melanomas remain at high risk for metastasis and death. Biomarker development has been limited by the challenge of isolating high-quality RNA for transcriptome-wide profiling from formalin-fixed and paraffin-embedded (FFPE) primary tumor specimens. Using NanoString technology, RNA from 40 stage II-III FFPE primary melanomas was analyzed and a 53-immune-gene panel predictive of non-progression (area under the curve (AUC)=0.920) was defined. The signature predicted disease-specific survival (DSS P<0.001) and recurrence-free survival (RFS P<0.001). CD2, the most differentially expressed gene in the training set, also predicted non-progression (P<0.001). Using publicly available microarray data from 46 primary human melanomas (GSE15605), a coexpression module enriched for the 53-gene panel was then identified using unbiased methods. A Bayesian network of signaling pathways based on this data identified driver genes. Finally, the proposed 53-gene panel was confirmed in an independent test population of 48 patients (AUC=0.787). The gene signature was an independent predictor of non-progression (P<0.001), RFS (P<0.001), and DSS (P=0.024) in the test population. The identified driver genes are potential therapeutic targets, and the 53-gene panel should be tested for clinical application using a larger data set annotated on the basis of prospectively gathered data.


Clinical Immunology | 2009

Allogeneic GM-CSF-secreting tumor cell immunotherapies generate potent anti-tumor responses comparable to autologous tumor cell immunotherapies

Andrew Simmons; Thomas Du; Carol Lin; Marina Moskalenko; Melissa Gonzalez-Edick; Melinda VanRoey; Karin Jooss

Clinical studies of cell-based immunotherapies have included both patient-specific (autologous) and non-patient-specific (allogeneic) approaches. Major concerns in using allogeneic immunotherapies are that the induced immune responses may be predominantly directed against the allogeneic HLA molecules of the cellular immunotherapy and not against its potential tumor antigens and that only the allogeneic responses will be enhanced when the immunotherapies are combined with immune checkpoint regulators in an effort to enhance overall immunotherapy potency. To evaluate these possibilities, studies were performed using the GM-CSF-secreting B16F1 cell line as autologous immunotherapy (Auto) and the same cell line modified to over-express the MHC molecule K(d) to generate an immunotherapy that expresses an allogeneic component (Allo) when injected into C57/Bl6 mice. The goal was to compare the specific anti-tumor immune responses induced by these two immunotherapies, which share an identical antigen repertoire, with the exception of the allogeneic MHC class I molecule expressed by the Allo cells, and have identical GM-CSF-secretion levels. Both immunotherapies provided similar therapeutic benefit to tumor-bearing animals with a trend towards a more pronounced tumor growth delay in animals injected with the Allo immunotherapy. This correlated with a significant increase in the number of activated DCs and T-cells in the DLN of Allo-treated animals. In addition, persistent infiltration of effector CD8(+) T-cells was detected in the tumors of animals treated with the Allo immunotherapy, which correlated with a trend towards a greater antigen-specific T-cell response in these animals. When combined with the immune checkpoint regulator anti-PD-1, tumor-specific and allogeneic immune responses were equally enhanced. Thus, the ability of an allogeneic tumor cell immunotherapy to induce a therapeutic anti-tumor immune response is comparable, if not superior, to an autologous tumor cell immunotherapy and its anti-tumor potency can be enhanced when combined with immunomodulatory compounds.


Clinical Immunology | 2013

Depletion of regulatory T cells by targeting folate receptor 4 enhances the potency of a GM-CSF-secreting tumor cell immunotherapy

Spencer Liang; Marina Moskalenko; Melinda Van Roey; Karin Jooss

In this report, a Treg-depleting anti-FR4 antibody is combined with a GM-CSF-secreting tumor cell immunotherapy (GVAX) for treatment of melanoma-bearing animals. Median survival time (MST) of animals treated with GVAX was 41 days, compared to a MST of 32 days in untreated animals. Anti-FR4 monotherapy had no effect on MST. Combination of anti-FR4 and GVAX significantly prolonged MST to 55 days, suggesting that these two agents can function cooperatively. Combination therapy increased expression of IFN-γ and granzyme B by CD8 T cells. In contrast to anti-CD25-mediated Treg depletion, administration of anti-FR4 after GVAX did not reduce efficacy, suggesting that anti-FR4 does not deplete effector cells induced by GVAX. Triple combination of a blocking CTLA4 antibody with GVAX and anti-FR4 further enhanced overall survival and reduced growth of well-established melanomas. Considered together, anti-FR4 antibody and GVAX may be a promising approach for the treatment of patients with cancer.


Cancer immunology research | 2015

Requirement for Innate Immunity and CD90+ NK1.1− Lymphocytes to Treat Established Melanoma with Chemo-Immunotherapy

Marina Moskalenko; Michael Pan; Yichun Fu; Ellen H. de Moll; Daigo Hashimoto; Arthur Mortha; Marylene Leboeuf; Padmini Jayaraman; Sebastian Bernardo; Andrew G. Sikora; Jedd D. Wolchok; Nina Bhardwaj; Miriam Merad; Yvonne Saenger

Moskalenko, Pan, and colleagues show in a B16 melanoma model that tumor clearance from the combined regimen of cytotoxic doses of cyclophosphamide and an antibody targeting melanoma differentiation antigen tyrosine-related protein 1 requires Fcγ receptors and innate CD90+NK1.1− lymphocytes, not classical NK cells. We sought to define cellular immune mechanisms of synergy between tumor-antigen–targeted monoclonal antibodies and chemotherapy. Established B16 melanoma in mice was treated with cytotoxic doses of cyclophosphamide in combination with an antibody targeting tyrosinase-related protein 1 (αTRP1), a native melanoma differentiation antigen. We find that Fcγ receptors are required for efficacy, showing that antitumor activity of combination therapy is immune mediated. Rag1−/− mice deficient in adaptive immunity are able to clear tumors, and thus innate immunity is sufficient for efficacy. Furthermore, previously treated wild-type mice are not significantly protected against tumor reinduction, as compared with mice inoculated with irradiated B16 alone, consistent with a primarily innate immune mechanism of action of chemo-immunotherapy. In contrast, mice deficient in both classical natural killer (NK) lymphocytes and nonclassical innate lymphocytes (ILC) due to deletion of the IL2 receptor common gamma chain IL2γc−/−) are refractory to chemo-immunotherapy. Classical NK lymphocytes are not critical for treatment, as depletion of NK1.1+ cells does not impair antitumor effect. Depletion of CD90+NK1.1− lymphocytes, however, both diminishes therapeutic benefit and decreases accumulation of macrophages within the tumor. Tumor clearance during combination chemo-immunotherapy with monoclonal antibodies against native antigen is mediated by the innate immune system. We highlight a novel potential role for CD90+NK1.1− ILCs in chemo-immunotherapy. Cancer Immunol Res; 3(3); 296–304. ©2015 AACR.


Attention Perception & Psychophysics | 2013

Modulation sensitivity in the perceptual organization of speech

Robert E. Remez; Emily F. Thomas; Kathryn R. Dubowski; Stavroula M. Koinis; Natalie A. C. Porter; Nina U. Paddu; Marina Moskalenko; Yael S. Grossman

In a spoken utterance, a talker expresses linguistic constituents in serial order. A listener resolves these linguistic properties in the rapidly fading auditory sample. Classic measures agree that auditory integration occurs at a fine temporal grain. In contrast, recent studies have proposed that sensory integration of speech occurs at a coarser grain, approximate to the syllable, on the basis of indirect and relatively insensitive perceptual measures. Evidence from cognitive neuroscience and behavioral primatology has also been adduced to support the claim of sensory integration at the pace of syllables. In the present investigation, we used direct performance measures of integration, applying an acoustic technique to isolate the contribution of short-term acoustic properties to the assay of modulation sensitivity. In corroborating the classic finding of a fine temporal grain of integration, these functional measures can inform theory and speculation in accounts of speech perception.


The Clinical Teacher | 2014

Instructional video for teaching venepuncture.

Michael Pan; Sara Harcharik; Adam J. Luber; Sebastian Bernardo; Jacob Levitt; Marina Moskalenko

Safe venepuncture technique is a critical skill for health care professionals, to avoid accidental occupational injury. This study investigates whether watching an instructional video improves medical students’ ability to perform venepuncture safely.


Journal of The American Academy of Dermatology | 2014

Defining the role of CD2 in disease progression and overall survival among patients with completely resected stage-II to -III cutaneous melanoma

Sara Harcharik; Sebastian Bernardo; Marina Moskalenko; Michael Pan; Meera Sivendran; Heather Bell; Lawrence D. Hall; Mireia Castillo-Martin; Kelly Fox; Carlos Cordon-Cardo; Rui Chang; Shanthi Sivendran; Robert G. Phelps; Yvonne Saenger

BACKGROUND Accurate assessment of prognosis remains clinically challenging in stage II to III cutaneous melanoma. Studies have implicated CD2 in immune surveillance, T-cell activation, and antitumor immunity, but its role in melanoma progression warrants further investigation. OBJECTIVE We sought to investigate the prognostic role of CD2 in primary cutaneous melanoma. METHODS Patients with American Joint Committee on Cancer stage II and III cutaneous melanoma were identified by retrospective review of dermatopathology databases from 2001 to 2010 at Mount Sinai Medical Center and Geisinger Medical Center. Additional patients were provided by New York University Medical Center based on retrospective review and tissue availability. Immunohistochemistry was performed on tumors from 90 patients with known recurrence status and documented follow-up. RESULTS Primary tumors from patients who developed recurrent disease had fewer CD2(+) cells (P = .0003). In multivariable analyses including standard clinicopathologic predictors, CD2 was an independent predictor of disease recurrence (P = .008) and overall survival (P = .007). CD2 count correlated with characterization of tumor-infiltrating lymphocytes (P = .0004). Among the intermediate prognosis group of patients with nonbrisk tumor-infiltrating lymphocytes, CD2 count was predictive of disease recurrence (P = .0006) and overall survival (P = .0318). LIMITATIONS Our retrospective design may have resulted in incomplete representation of patients lacking documented follow-up. CONCLUSIONS CD2 may be an independent predictor of disease recurrence and overall survival among patients with primary cutaneous melanoma.

Collaboration


Dive into the Marina Moskalenko's collaboration.

Top Co-Authors

Avatar

Sebastian Bernardo

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Michael Pan

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Yvonne Saenger

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Sara Harcharik

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert G. Phelps

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Rui Chang

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Shanthi Sivendran

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge