Network


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

Hotspot


Dive into the research topics where Alicja Copik is active.

Publication


Featured researches published by Alicja Copik.


Biology of Blood and Marrow Transplantation | 2015

Generation of Highly Cytotoxic Natural Killer Cells for Treatment of Acute Myelogenous Leukemia using a Feeder-Free, Particle-Based Approach

Jeremiah Oyer; Robert Y. Igarashi; Alexander R. Kulikowski; Dominic A. Colosimo; Melhem Solh; Ahmed Zakari; Yasser Khaled; Deborah A. Altomare; Alicja Copik

Natural killer (NK) cell immunotherapy as a cancer treatment shows promise, but expanding NK cells consistently from a small fraction (∼ 5%) of peripheral blood mononuclear cells (PBMCs) to therapeutic amounts remains challenging. Most current ex vivo expansion methods use co-culture with feeder cells (FC), but their use poses challenges for wide clinical application. We developed a particle-based NK cell expansion technology that uses plasma membrane particles (PM-particles) derived from K562-mbIL15-41BBL FCs. These PM-particles induce selective expansion of NK cells from unsorted PBMCs, with NK cells increasing 250-fold (median, 35; 10 donors; range, 94 to 1492) after 14 days of culture and up to 1265-fold (n = 14; range, 280 to 4426) typically after 17 days. The rate and efficiency of NK cell expansions with PM-particles and live FCs are comparable and far better than stimulation with soluble 41BBL, IL-15, and IL-2. Furthermore, NK cells expand selectively with PM-particles to 86% (median, 35; range, 71% to 99%) of total cells after 14 days. The extent of NK cell expansion and cell content was PM-particle concentration dependent. These NK cells were highly cytotoxic against several leukemic cell lines and also against patient acute myelogenous leukemia blasts. Phenotype analysis of these PM-particle-expanded NK cells was consistent with an activated cytotoxic phenotype. This novel NK cell expansion methodology has promising clinical therapeutic implications.


Cytokine | 2017

Cytokines in immunogenic cell death: Applications for cancer immunotherapy

Anne Showalter; Arati Limaye; Jeremiah Oyer; Robert Y. Igarashi; Christina Kittipatarin; Alicja Copik; Annette R. Khaled

&NA; Despite advances in treatments like chemotherapy and radiotherapy, metastatic cancer remains a leading cause of death for cancer patients. While many chemotherapeutic agents can efficiently eliminate cancer cells, long‐term protection against cancer is not achieved and many patients experience cancer recurrence. Mobilizing and stimulating the immune system against tumor cells is one of the most effective ways to protect against cancers that recur and/or metastasize. Activated tumor specific cytotoxic T lymphocytes (CTLs) can seek out and destroy metastatic tumor cells and reduce tumor lesions. Natural Killer (NK) cells are a front‐line defense against drug‐resistant tumors and can provide tumoricidal activity to enhance tumor immune surveillance. Cytokines like IFN‐&ggr; or TNF play a crucial role in creating an immunogenic microenvironment and therefore are key players in the fight against metastatic cancer. To this end, a group of anthracyclines or treatments like photodynamic therapy (PDT) exert their effects on cancer cells in a manner that activates the immune system. This process, known as immunogenic cell death (ICD), is characterized by the release of membrane‐bound and soluble factors that boost the function of immune cells. This review will explore different types of ICD inducers, some in clinical trials, to demonstrate that optimizing the cytokine response brought about by treatments with ICD‐inducing agents is central to promoting anti‐cancer immunity that provides long‐lasting protection against disease recurrence and metastasis. HighlightsShifting from inhibitory to activating cytokines is a challenge for immunotherapy.Cancer cells undergoing ICD act like “vaccines” to stimulate anti‐cancer immunity.Death induced by type I and II ICD inducers involve endoplasmic reticulum stress.Optimizing the cytokine response during ICD could yield new combination therapies.


Cytotherapy | 2016

Natural killer cells stimulated with PM21 particles expand and biodistribute in vivo: Clinical implications for cancer treatment

Jeremiah Oyer; Veethika Pandey; Robert Y. Igarashi; Srinivas S. Somanchi; Ahmed Zakari; Melhem Solh; Dean A. Lee; Deborah A. Altomare; Alicja Copik

BACKGROUND AIMS Natural killer (NK) cell immunotherapy for treatment of cancer is promising, but requires methods that expand cytotoxic NK cells that persist in circulation and home to disease site. METHODS We developed a particle-based method that is simple, effective and specifically expands cytotoxic NK cells from peripheral blood mononuclear cells (PBMCs) both ex vivo and in vivo. This method uses particles prepared from plasma membranes of K562-mb21-41BBL cells, expressing 41BBL and membrane bound interleukin-21 (PM21 particles). RESULTS Ex vivo, PM21 particles caused specific NK-cell expansion from PBMCs from healthy donors (mean 825-fold, range 163-2216, n = 13 in 14 days) and acute myeloid leukemia patients. The PM21 particles also stimulated in vivo NK cell expansion in NSG mice. Ex vivo pre-activation of PBMCs with PM21 particles (PM21-PBMC) before intraperitoneal (i.p.) injection resulted in 66-fold higher amounts of hNK cells in peripheral blood (PB) of mice compared with unactivated PBMCs on day 12 after injection. In vivo administration of PM21 particles resulted in a dose-dependent increase of PB hNK cells in mice injected i.p. with 2.0 × 10(6) PM21-PBMCs (11% NK cells). Optimal dose of 800 µg/injection of PM21 particles (twice weekly) with low-dose interleukin 2 (1000 U/thrice weekly) resulted in 470 ± 40 hNK/µL and 95 ± 2% of total hCD45(+) cells by day 12 in PB. Furthermore, hNK cells were found in marrow, spleen, lung, liver and brain (day 16 after i.p. PM21/PBMC injection), and mice injected with PM21 particles had higher amounts. CONCLUSIONS The extent of NK cells observed in PB, their persistence and the biodistribution would be relevant for cancer treatment.


Photodiagnosis and Photodynamic Therapy | 2015

Development and characterization of conducting polymer nanoparticles for photodynamic therapy in vitro

Mona Doshi; Alicja Copik; Andre J. Gesquiere

Conducting polymer nanoparticles (CPNPs), composed of the conducting polymer poly[2-methoxy-5-(2-ethylhexyl-oxy)-p-phenylenevinylene] (MEH-PPV) were studied for applications in biophotonics and therapeutics. The extent of cellular uptake, cytotoxicity, and effectiveness of these nanoparticles in photodynamic therapy (PDT) was investigated for four cell lines, namely TE-71, MDA-MB-231, A549 and OVCAR3. Confocal fluorescence imaging and flow cytometry show that CPNPs are taken up only in limited quantities by TE-71, while they are taken up extensively by the cancer cell lines. The uptake among the cancer cell lines was observed to vary with cell line, with CPNPs uptake increasing from MDA-MB-231 to A549 to OVCAR3. Fluorescence imaging experiments show that the CPNPs have high brightness and appear stable in the intracellular environment. No cytotoxicity of non-photoactivated CPNPs (in dark) was observed from MTT assay. After completion of PDT, the quantitative data on cell viability suggest that cell death scales across the cell lines with CPNP uptake, is light dose dependent, and is complete for OVCAR3. In addition, for OVCAR3 apoptotic cell death is observed after PDT. The reported work illustrates the potential of the intrinsically fluorescent and photoactivateable CPNPs for application in biophotonics and therapeutics.


Molecular Pharmaceutics | 2018

Difluoromethylornithine Combined with a Polyamine Transport Inhibitor Is Effective against Gemcitabine Resistant Pancreatic Cancer

Sarah B. Gitto; Veethika Pandey; Jeremiah Oyer; Alicja Copik; Frederick C. Hogan; Otto Phanstiel; Deborah A. Altomare

Pancreatic ductal adenocarcinoma (PDAC) is highly chemo-resistant and has an extremely poor patient prognosis, with a survival rate at five years of <8%. There remains an urgent need for innovative treatments. Targeting polyamine biosynthesis through inhibition of ornithine decarboxylase with difluoromethylornithine (DFMO) has had mixed clinical success due to tumor escape via an undefined transport system, which imports exogenous polyamines and sustains intracellular polyamine pools. Here, we tested DFMO in combination with a polyamine transport inhibitor (PTI), Trimer44NMe, against Gemcitabine-resistant PDAC cells. DFMO alone and with Trimer44NMe significantly reduced PDAC cell viability by inducing apoptosis or diminishing proliferation. DFMO alone and with Trimer44NMe also inhibited in vivo orthotopic PDAC growth and resulted in decreased c-Myc expression, a readout of polyamine pathway dysfunction. Moreover, dual inhibition significantly prolonged survival of tumor-bearing mice. Collectively, these studies demonstrate that targeting polyamine biosynthesis and import pathways in PDAC can lead to increased survival in pancreatic cancer.


Oncotarget | 2017

Ponatinib promotes a G 1 cell-cycle arrest of merlin/NF2-deficient human schwann cells

Alejandra M. Petrilli; Jeanine Garcia; Marga Bott; Stephani Klingeman Plati; Christine T. Dinh; Olena Bracho; Denise Yan; Bing Zou; Rahul Mittal; Fred F. Telischi; Xue Zhong Liu; Long-Sheng Chang; D. Bradley Welling; Alicja Copik; Cristina Fernandez-Valle

Neurofibromatosis type 2 (NF2) is a genetic syndrome that predisposes individuals to multiple benign tumors of the central and peripheral nervous systems, including vestibular schwannomas. Currently, there are no FDA approved drug therapies for NF2. Loss of function of merlin encoded by the NF2 tumor suppressor gene leads to activation of multiple mitogenic signaling cascades, including platelet-derived growth factor receptor (PDGFR) and SRC in Schwann cells. The goal of this study was to determine whether ponatinib, an FDA-approved ABL/SRC inhibitor, reduced proliferation and/or survival of merlin-deficient human Schwann cells (HSC). Merlin-deficient HSC had higher levels of phosphorylated PDGFRα/β, and SRC than merlin-expressing HSC. A similar phosphorylation pattern was observed in phospho-protein arrays of human vestibular schwannoma samples compared to normal HSC. Ponatinib reduced merlin-deficient HSC viability in a dose-dependent manner by decreasing phosphorylation of PDGFRα/β, AKT, p70S6K, MEK1/2, ERK1/2 and STAT3. These changes were associated with decreased cyclin D1 and increased p27Kip1levels, leading to a G1 cell-cycle arrest as assessed by Western blotting and flow cytometry. Ponatinib did not modulate ABL, SRC, focal adhesion kinase (FAK), or paxillin phosphorylation levels. These results suggest that ponatinib is a potential therapeutic agent for NF2-associated schwannomas and warrants further in vivo investigation.


OncoImmunology | 2018

PD-L1 blockade enhances anti-tumor efficacy of NK cells

Jeremiah Oyer; Sarah B. Gitto; Deborah A. Altomare; Alicja Copik

ABSTRACT Anti-PD-1/anti-PD-L1 therapies have shown success in cancer treatment but responses are limited to ~ 15% of patients with lymphocyte infiltrated, PD-L1 positive tumors. Hence, strategies that increase PD-L1 expression and tumor infiltration should make more patients eligible for PD-1/PD-L1 blockade therapy, thus improving overall outcomes. PD-L1 expression on tumors is induced by IFNγ, a cytokine secreted by NK cells. Therefore, we tested if PM21-particle expanded NK cells (PM21-NK cells) induced expression of PD-L1 on tumors and if anti-PD-L1 treatment enhanced NK cell anti-tumor efficacy in an ovarian cancer model. Studies here showed that PM21-NK cells secrete high amounts of IFNγ and that adoptively transferred PM21-NK cells induce PD-L1 expression on SKOV-3 cells in vivo. The induction of PD-L1 expression on SKOV-3 cells coincided with the presence of regulatory T cells (Tregs) in the abdominal cavity and within tumors. In in vitro experiments, anti-PD-L1 treatment had no direct effect on cytotoxicity or cytokine secretion by predominantly PD-1 negative PM21-NK cells in response to PD-L1+ targets. However, significant improvement of NK cell anti-tumor efficacy was observed in vivo when combined with anti-PD-L1. PD-L1 blockade also resulted in increased in vivo NK cell persistence and retention of their cytotoxic phenotype. These results support the use of anti-PD-L1 in combination with NK cell therapy regardless of initial tumor PD-L1 status and indicate that NK cell therapy would likely augment the applicability of anti-PD-L1 treatment.


Molecular Cancer Therapeutics | 2017

Combination therapy with c-Met and Src inhibitors induces caspase-dependent apoptosis of merlin-deficient Schwann cells and suppresses growth of schwannoma cells

Marisa A. Fuse; Stephani Klingeman Plati; Sarah S. Burns; Christine T. Dinh; Olena Bracho; Denise Yan; Rahul Mittal; Rulong Shen; Julia N. Soulakova; Alicja Copik; Xue Zhong Liu; Fred F. Telischi; Long-Sheng Chang; Maria Clara Franco; Cristina Fernandez-Valle

Neurofibromatosis type 2 (NF2) is a nervous system tumor disorder caused by inactivation of the merlin tumor suppressor encoded by the NF2 gene. Bilateral vestibular schwannomas are a diagnostic hallmark of NF2. Mainstream treatment options for NF2-associated tumors have been limited to surgery and radiotherapy; however, off-label uses of targeted molecular therapies are becoming increasingly common. Here, we investigated drugs targeting two kinases activated in NF2-associated schwannomas, c-Met and Src. We demonstrated that merlin-deficient mouse Schwann cells (MD-MSC) treated with the c-Met inhibitor, cabozantinib, or the Src kinase inhibitors, dasatinib and saracatinib, underwent a G1 cell-cycle arrest. However, when MD-MSCs were treated with a combination of cabozantinib and saracatinib, they exhibited caspase-dependent apoptosis. The combination therapy also significantly reduced growth of MD-MSCs in an orthotopic allograft mouse model by greater than 80% of vehicle. Moreover, human vestibular schwannoma cells with NF2 mutations had a 40% decrease in cell viability when treated with cabozantinib and saracatinib together compared with the vehicle control. This study demonstrates that simultaneous inhibition of c-Met and Src signaling in MD-MSCs triggers apoptosis and reveals vulnerable pathways that could be exploited to develop NF2 therapies. Mol Cancer Ther; 16(11); 2387–98. ©2017 AACR.


Blood | 2013

Relapse After Allogeneic Hematopoietic Stem Cell Transplantation (HCT) For Acute Myeloid Leukemia (AML)/Myelodysplastic Syndrome (MDS) Following Intravenous Busulfan/Fludarabine Based Conditioning: Outcomes and Monocyte Chemo-Attractant Protein -1( MCP-1) As a Predictive Marker Of Relapse

Abraham J. Matar; Melhem Solh; Alicja Copik; Sally Litherland; Alvin oliverasAlmodovar; Kristin Rathmann


Archive | 2018

Methods and Compositions High Scale Therapeutic Production of Memory NK Cells

Alicja Copik; Robert Y. Igarashi; Jeremiah Oyer; Deborah A. Altomare

Collaboration


Dive into the Alicja Copik's collaboration.

Top Co-Authors

Avatar

Jeremiah Oyer

University of Central Florida

View shared research outputs
Top Co-Authors

Avatar

Deborah A. Altomare

University of Central Florida

View shared research outputs
Top Co-Authors

Avatar

Robert Y. Igarashi

University of Central Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sarah B. Gitto

University of Central Florida

View shared research outputs
Top Co-Authors

Avatar

Veethika Pandey

University of Central Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Annette R. Khaled

University of Central Florida

View shared research outputs
Top Co-Authors

Avatar

Arati Limaye

University of Central Florida

View shared research outputs
Top Co-Authors

Avatar

Dean A. Lee

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge