Network


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

Hotspot


Dive into the research topics where Benjamin E. Paluch is active.

Publication


Featured researches published by Benjamin E. Paluch.


Journal of Cell Science | 2012

PTEN at a glance

Yuji Shi; Benjamin E. Paluch; Xinjiang Wang; Xuejun Jiang

Since its discovery in 1997 ([Li and Sun, 1997][1]; [Li et al., 1997][2]; [Steck et al., 1997][3]), the phosphatidylinositol (3,4,5)-trisphosphate [PtdIns(3,4,5) P 3] phosphatase and tensin homolog (PTEN) has been established as one of the most frequently mutated tumor suppressor genes in human


Leukemia Research | 2014

Immunomodulatory action of SGI-110, a hypomethylating agent, in acute myeloid leukemia cells and xenografts

Pragya Srivastava; Benjamin E. Paluch; Junko Matsuzaki; Smitha R. James; Golda Collamat-Lai; Julia Karbach; Michael J. Nemeth; Pietro Taverna; Adam R. Karpf; Elizabeth A. Griffiths

The mechanism of clinical action for the FDA approved hypomethylating drugs azacitidine and decitabine remains unresolved and in this context the potential immunomodulatory effect of these agents on leukemic cells is an area of active investigation. Induced expression of methylated Cancer Testis Antigen (CTA) genes has been demonstrated in leukemic cell lines following exposure to hypomethylating drugs in vitro. SGI-110 is a novel hypomethylating dinucleotide with prolonged in vivo exposure and clinical activity in patients with MDS and AML. We demonstrate that this agent, like decitabine, produces robust re-expression of the CTAs NY-ESO-1 and MAGE-A, both in vitro and in leukemia-bearing AML xenografts. Upregulation of these genes in vitro was sufficient to induce cytotoxicity by HLA-compatible CD8+ T-cells specific for NY-ESO-1, a well-recognized and immunogenic CTA. Additionally, exposure to SGI-110 enhances MHC class I and co-stimulatory molecule expression, potentially contributing to recognition of CTAs. SGI-110, like the parent compound decitabine, induces expression of CTAs and might modulate immune recognition of myeloid malignancy.


Epigenetics | 2015

Immunomodulatory action of the DNA methyltransferase inhibitor SGI-110 in epithelial ovarian cancer cells and xenografts

Pragya Srivastava; Benjamin E. Paluch; Junko Matsuzaki; Smitha R. James; Golda Collamat-Lai; Pietro Taverna; Adam R. Karpf; Elizabeth A. Griffiths

We aimed to determine the effect of SGI-110 on methylation and expression of the cancer testis antigens (CTAs) NY-ESO-1 and MAGE-A in epithelial ovarian cancer (EOC) cells in vitro and in vivo and to establish the impact of SGI-110 on expression of major histocompatibility (MHC) class I and Intracellular Adhesion Molecule 1 (ICAM-1) on EOC cells, and on recognition of EOC cells by NY-ESO-1-specific CD8+ T-cells. We also tested the impact of combined SGI-110 and NY-ESO-1-specific CD8+ T-cells on tumor growth and/or murine survival in a xenograft setting. EOC cells were treated with SGI-110 in vitro at various concentrations and as tumor xenografts with 3 distinct dose schedules. Effects on global methylation (using LINE-1), NY-ESO-1 and MAGE-A methylation, mRNA, and protein expression were determined and compared to controls. SGI-110 treated EOC cells were evaluated for expression of immune-modulatory genes using flow cytometry, and were co-cultured with NY-ESO-1 specific T-cell clones to determine immune recognition. In vivo administration of SGI-110 and CD8+ T-cells was performed to determine anti-tumor effects on EOC xenografts. SGI-110 treatment induced hypomethylation and CTA gene expression in a dose dependent manner both in vitro and in vivo, at levels generally superior to azacitidine or decitabine. SGI-110 enhanced the expression of MHC I and ICAM-1, and enhanced recognition of EOC cells by NY-ESO-1-specific CD8+ T-cells. Sequential SGI-110 and antigen-specific CD8+ cell treatment restricted EOC tumor growth and enhanced survival in a xenograft setting. SGI-110 is an effective hypomethylating agent and immune modulator and, thus, an attractive candidate for combination with CTA-directed vaccines in EOC.


Oncotarget | 2016

Induction of cancer testis antigen expression in circulating acute myeloid leukemia blasts following hypomethylating agent monotherapy

Pragya Srivastava; Benjamin E. Paluch; Junko Matsuzaki; Smitha R. James; Golda Collamat-Lai; Nadja Blagitko-Dorfs; Laurie A. Ford; Rafeh Naqash; Michael Lübbert; Adam R. Karpf; Michael J. Nemeth; Elizabeth A. Griffiths

Cancer testis antigens (CTAs) are promising cancer associated antigens in solid tumors, but in acute myeloid leukemia, dense promoter methylation silences their expression. Leukemia cell lines exposed to HMAs induce expression of CTAs. We hypothesized that AML patients treated with standard of care decitabine (20mg/m2 per day for 10 days) would demonstrate induced expression of CTAs. Peripheral blood blasts serially isolated from AML patients treated with decitabine were evaluated for CTA gene expression and demethylation. Induction of NY-ESO-1 and MAGEA3/A6, were observed following decitabine. Re-expression of NY-ESO-1 and MAGEA3/A6 was associated with both promoter specific and global (LINE-1) hypomethylation. NY-ESO-1 and MAGEA3/A6 mRNA levels were increased irrespective of clinical response, suggesting that these antigens might be applicable even in patients who are not responsive to HMA therapy. Circulating blasts harvested after decitabine demonstrate induced NY-ESO-1 expression sufficient to activate NY-ESO-1 specific CD8+ T-cells. Induction of CTA expression sufficient for recognition by T-cells occurs in AML patients receiving decitabine. Vaccination against NY-ESO-1 in this patient population is feasible.


Journal of Biological Chemistry | 2013

Agonist-induced down-regulation of endogenous protein kinase c α through an endolysosomal mechanism.

Michelle A. Lum; Krista E. Pundt; Benjamin E. Paluch; Adrian R. Black; Jennifer D. Black

Background: Mechanisms of endogenous PKC signal termination remain to be fully characterized. Results: Activated endogenous PKCα undergoes dynamin-dependent and -independent endocytic uptake and traffics through early and late endosomes for processing by lysosomes. Conclusion: Lysosomal degradation represents a novel mechanism of desensitizing PKC-mediated signaling. Significance: Multiple degradation mechanisms ensure strict control of the duration of PKC signaling in cells. Protein kinase C (PKC) isozymes undergo down-regulation upon sustained stimulation. Previous studies have pointed to the existence of both proteasome-dependent and -independent pathways of PKCα processing. Here we demonstrate that these down-regulation pathways are engaged in different subcellular compartments; proteasomal degradation occurs mainly at the plasma membrane, whereas non-proteasomal processing occurs in the perinuclear region. Using cholesterol depletion, pharmacological inhibitors, RNA interference, and dominant-negative mutants, we define the mechanisms involved in perinuclear accumulation of PKCα and identify the non-proteasomal mechanism mediating its degradation. We show that intracellular accumulation of PKCα involves at least two clathrin-independent, cholesterol/lipid raft-mediated pathways that do not require ubiquitination of the protein; one is dynamin-dependent and likely involves caveolae, whereas the other is dynamin- and small GTPase-independent. Internalized PKCα traffics through endosomes and is delivered to the lysosome for degradation. Supportive evidence includes (a) detection of the enzyme in EEA1-positive early endosomes, Rab7-positive late endosomes/multivesicular bodies, and LAMP1-positive lysosomes and (b) inhibition of its down-regulation by lysosome-disrupting agents and leupeptin. Only limited dephosphorylation of PKCα occurs during trafficking, with fully mature enzyme being the main target for lysosomal degradation. These studies define a novel and widespread mechanism of desensitization of PKCα signaling that involves endocytic trafficking and lysosome-mediated degradation of the mature, fully phosphorylated protein.


Oncotarget | 2017

Robust detection of immune transcripts in FFPE samples using targeted RNA sequencing

Benjamin E. Paluch; Sean T. Glenn; Jeffrey Conroy; Antonios Papanicolau-Sengos; Wiam Bshara; Angela Omilian; Elizabeth Brese; Mary Nesline; Blake Burgher; Jonathan Andreas; Kunle Odunsi; Kevin H. Eng; Ji He; Maochun Qin; Mark Gardner; Lorenzo Galluzzi; Carl Morrison

Current criteria for identifying cancer patients suitable for immunotherapy with immune checkpoint blockers (ICBs) are subjective and prone to misinterpretation, as they mainly rely on the visual assessment of CD274 (best known as PD-L1) expression levels by immunohistochemistry (IHC). To address this issue, we developed a RNA sequencing (RNAseq)-based approach that specifically measures the abundance of immune transcripts in formalin-fixed paraffin embedded (FFPE) specimens. Besides exhibiting superior sensitivity as compared to whole transcriptome RNAseq, our assay requires little starting material, implying that it is compatible with RNA degradation normally caused by formalin. Here, we demonstrate that a targeted RNAseq panel reliably profiles mRNA expression levels in FFPE samples from a cohort of ovarian carcinoma patients. The expression profile of immune transcripts as measured by targeted RNAseq in FFPE versus freshly frozen (FF) samples from the same tumor was highly concordant, in spite of the RNA quality issues associated with formalin fixation. Moreover, the results of targeted RNAseq on FFPE specimens exhibited a robust correlation with mRNA expression levels as measured on the same samples by quantitative RT-PCR, as well as with protein abundance as determined by IHC. These findings demonstrate that RNAseq profiling on archival FFPE tissues can be used reliably in studies assessing the efficacy of cancer immunotherapy.


Clinical Cancer Research | 2017

NY-ESO-1 Vaccination in Combination with Decitabine Induces Antigen-Specific T-lymphocyte Responses in Patients with Myelodysplastic Syndrome

Elizabeth A. Griffiths; Pragya Srivastava; Junko Matsuzaki; Zachary Brumberger; Eunice S. Wang; Justin Kocent; Austin Miller; Gregory W. Roloff; Hong Yuen Wong; Benjamin E. Paluch; Linda G. Lutgen-Dunckley; Brandon L. Martens; Kunle Odunsi; Adam R. Karpf; Christopher S. Hourigan; Michael J. Nemeth

Purpose: Treatment options are limited for patients with high-risk myelodysplastic syndrome (MDS). The azanucleosides, azacitidine and decitabine, are first-line therapy for MDS that induce promoter demethylation and gene expression of the highly immunogenic tumor antigen NY-ESO-1. We demonstrated that patients with acute myeloid leukemia (AML) receiving decitabine exhibit induction of NY-ESO-1 expression in circulating blasts. We hypothesized that vaccinating against NY-ESO-1 in patients with MDS receiving decitabine would capitalize upon induced NY-ESO-1 expression in malignant myeloid cells to provoke an NY-ESO-1–specific MDS-directed cytotoxic T-cell immune response. Experimental Design: In a phase I study, 9 patients with MDS received an HLA-unrestricted NY-ESO-1 vaccine (CDX-1401 + poly-ICLC) in a nonoverlapping schedule every four weeks with standard-dose decitabine. Results: Analysis of samples serially obtained from the 7 patients who reached the end of the study demonstrated induction of NY-ESO-1 expression in 7 of 7 patients and NY-ESO-1–specific CD4+ and CD8+ T-lymphocyte responses in 6 of 7 and 4 of 7 of the vaccinated patients, respectively. Myeloid cells expressing NY-ESO-1, isolated from a patient at different time points during decitabine therapy, were capable of activating a cytotoxic response from autologous NY-ESO-1–specific T lymphocytes. Vaccine responses were associated with a detectable population of CD141Hi conventional dendritic cells, which are critical for the uptake of NY-ESO-1 vaccine and have a recognized role in antitumor immune responses. Conclusions: These data indicate that vaccination against induced NY-ESO-1 expression can produce an antigen-specific immune response in a relatively nonimmunogenic myeloid cancer and highlight the potential for induced antigen-directed immunotherapy in a group of patients with limited options. Clin Cancer Res; 24(5); 1019–29. ©2017 AACR. See related commentary by Fuchs, p. 991


Blood Reviews | 2016

Epigenetics: A primer for clinicians

Benjamin E. Paluch; Abdul Rafeh Naqash; Zachary Brumberger; Michael J. Nemeth; Elizabeth A. Griffiths


Blood | 2015

NY-ESO-1 Vaccination in Combination with Decitabine for Patients with MDS Induces CD4+ and CD8+ T-Cell Responses

Pragya Srivastava; Junko Matsuzaki; Benjamin E. Paluch; Zachary Brumberger; Stephanie Kaufman; Adam R. Karpf; Kunle Odunsi; Austin Miller; Justin Kocent; Eunice S. Wang; Michael J. Nemeth; Elizabeth A. Griffiths


Blood | 2016

Vaccination with NY-ESO-1 in Combination with Decitabine for Patients with MDS

Pragya Srivastava; Junko Matsuzaki; Benjamin E. Paluch; Stephanie Kaufman; Adam R. Karpf; Kunle Odunsi; Austin Miller; Justin Kocent; Eunice S. Wang; Michael J. Nemeth; Elizabeth A. Griffiths

Collaboration


Dive into the Benjamin E. Paluch's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pragya Srivastava

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Michael J. Nemeth

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Adam R. Karpf

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Junko Matsuzaki

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Kunle Odunsi

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Smitha R. James

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Austin Miller

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Eunice S. Wang

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Golda Collamat-Lai

Roswell Park Cancer Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge