Network


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

Hotspot


Dive into the research topics where Patricia Simms is active.

Publication


Featured researches published by Patricia Simms.


Scientific Reports | 2016

Tumor-derived exosomes regulate expression of immune function-related genes in human T cell subsets.

Laurent Muller; Masato Mitsuhashi; Patricia Simms; William E. Gooding; Theresa L. Whiteside

Tumor cell-derived exosomes (TEX) suppress functions of immune cells. Here, changes in the gene profiles of primary human T lymphocytes exposed in vitro to exosomes were evaluated. CD4+ Tconv, CD8+ T or CD4+ CD39+ Treg were isolated from normal donors’ peripheral blood and co-incubated with TEX or exosomes isolated from supernatants of cultured dendritic cells (DEX). Expression levels of 24–27 immune response-related genes in these T cells were quantified by qRT-PCR. In activated T cells, TEX and DEX up-regulated mRNA expression levels of multiple genes. Multifactorial data analysis of ΔCt values identified T cell activation and the immune cell type, but not exosome source, as factors regulating gene expression by exosomes. Treg were more sensitive to TEX-mediated effects than other T cell subsets. In Treg, TEX-mediated down-regulation of genes regulating the adenosine pathway translated into high expression of CD39 and increased adenosine production. TEX also induced up-regulation of inhibitory genes in CD4+ Tconv, which translated into a loss of CD69 on their surface and a functional decline. Exosomes are not internalized by T cells, but signals they carry and deliver to cell surface receptors modulate gene expression and functions of human T lymphocytes.


Cancer Immunology, Immunotherapy | 2016

TCR gene-modified T cells can efficiently treat established hepatitis C-associated hepatocellular carcinoma tumors

Timothy T. Spear; Glenda G. Callender; Jeffrey J. Roszkowski; Kelly Moxley; Patricia Simms; Kendra C. Foley; David C. Murray; Gina Scurti; Mingli Li; Justin T. Thomas; Alexander Langerman; Elizabeth Garrett-Mayer; Yi Zhang; Michael I. Nishimura

Abstract The success in recent clinical trials using T cell receptor (TCR)-genetically engineered T cells to treat melanoma has encouraged the use of this approach toward other malignancies and viral infections. Although hepatitis C virus (HCV) infection is being treated with a new set of successful direct anti-viral agents, potential for virologic breakthrough or relapse by immune escape variants remains. Additionally, many HCV+ patients have HCV-associated disease, including hepatocellular carcinoma (HCC), which does not respond to these novel drugs. Further exploration of other approaches to address HCV infection and its associated disease are highly warranted. Here, we demonstrate the therapeutic potential of PBL-derived T cells genetically engineered with a high-affinity, HLA-A2-restricted, HCV NS3:1406-1415-reactive TCR. HCV1406 TCR-transduced T cells can recognize naturally processed antigen and elicit CD8-independent recognition of both peptide-loaded targets and HCV+ human HCC cell lines. Furthermore, these cells can mediate regression of established HCV+ HCC in vivo. Our results suggest that HCV TCR-engineered antigen-reactive T cells may be a plausible immunotherapy option to treat HCV-associated malignancies, such as HCC.


Cancer Research | 2014

Promoting Thiol Expression Increases the Durability of Antitumor T-cell Functions

Pravin Kesarwani; Amir A. Al-Khami; Gina Scurti; Krishnamurthy Thyagarajan; Navtej Kaur; Shahid Husain; Quan Fang; Osama Naga; Patricia Simms; Gyda C. Beeson; Christina Voelkel-Johnson; Elizabeth Garrett-Mayer; Craig Beeson; Michael I. Nishimura; Shikhar Mehrotra

Ex vivo-expanded CD8(+) T cells used for adoptive immunotherapy generally acquire an effector memory-like phenotype (TEM cells). With regard to therapeutic applications, two undesired features of this phenotype in vivo are limited persistence and reduced antitumor efficacy, relative to CD8(+) T cells with a central memory-like phenotype (TCM cells). Furthermore, there is incomplete knowledge about all the differences between TEM and TCM cells that may influence tumor treatment outcomes. Given that TCM cells survive relatively longer in oxidative tumor microenvironments, we investigated the hypothesis that TCM cells possess relatively greater antioxidative capacity than TEM cells. Here, we report that TCM cells exhibit a relative increase compared with TEM cells in the expression of cell surface thiols, a key target of cellular redox controls, along with other antioxidant molecules. Increased expression of redox regulators in TCM cells inversely correlated with the generation of reactive oxygen and nitrogen species, proliferative capacity, and glycolytic enzyme levels. Notably, T-cell receptor-transduced T cells pretreated with thiol donors, such as N-acetyl cysteine or rapamycin, upregulated thiol levels and antioxidant genes. A comparison of antitumor CD8(+) T-cell populations on the basis of surface thiol expression showed that thiol-high cells persisted longer in vivo and exerted superior tumor control. Our results suggest that higher levels of reduced cell surface thiols are a key characteristic of T cells that can control tumor growth and that profiling this biomarker may have benefits to adoptive T-cell immunotherapy protocols.


Immunobiology | 2000

Comparison of cytokines and CD80 for enhancement of immunogenicity of cervical cancer cells.

Andreas Kaufmann; Lutz Gissmann; Patricia Simms; Carola Schreckenberger; Liang Qiao

Tumor cells fail to activate specific cytotoxic T lymphocytes due to lack of costimulatory molecules e.g. CD80 (B7.1). We were able to render cervical carcinoma cells immunogenic by introduction of the CD80 gene into the tumor cells. In order to enhance the efficiency of T cell activation we investigated whether addition of interleukins would augment immunostimulation by CD80. To this end, allogeneic T cells were stimulated with CD80-expressing HeLa cells or CaSki cells in the absence or presence of IL-2, IL-7, IL-12, or combinations thereof. The proliferative response of the T cells was determined. CD80-transduced HeLa or CaSki cells induced a stronger proliferative response in allogeneic T cells than parental or mock transfected control cells. All three interleukins enhanced the proliferative response of allogeneic T cells to CD80-expressing tumor cells. IL-2 or IL-7 had stronger effects in expanding the T cells than IL-12. Combination of IL-2 and IL-7 resulted in best T cell expansion. The proliferating T cells were mainly CD8+ cells with MHC class I restricted and unrestricted cytotoxic activity. Stimulation with CD80 alone or in combination with IL-7 induced mainly cytotoxic T lymphocytes. IL-2, IL-12 or the combination of IL-2 and IL-7 induced natural killer cell-like activity and specific cytolytic activity against parental and CD80-positive tumor cells. Our data suggest that the expression of both CD80 and IL-2 plus IL-7 can enhance the efficacy of tumor vaccines.


Journal of Leukocyte Biology | 2016

Hepatitis C virus‐cross‐reactive TCR gene‐modified T cells: a model for immunotherapy against diseases with genomic instability

Timothy T. Spear; Timothy P. Riley; Gretchen E. Lyons; Glenda G. Callender; Jeffrey J. Roszkowski; Yuan Wang; Patricia Simms; Gina Scurti; Kendra C. Foley; David C. Murray; Lance M. Hellman; Rachel H. McMahan; Makio Iwashima; Elizabeth Garrett-Mayer; Hugo R. Rosen; Brian M. Baker; Michael I. Nishimura

A major obstacle hindering the development of effective immunity against viral infections, their associated disease, and certain cancers is their inherent genomic instability. Accumulation of mutations can alter processing and presentation of antigens recognized by antibodies and T cells that can lead to immune escape variants. Use of an agent that can intrinsically combat rapidly mutating viral or cancer‐associated antigens would be quite advantageous in developing effective immunity against such disease. We propose that T cells harboring cross‐reactive TCRs could serve as a therapeutic agent in these instances. With the use of hepatitis C virus, known for its genomic instability as a model for mutated antigen recognition, we demonstrate cross‐reactivity against immunogenic and mutagenic nonstructural protein 3:1406‐1415 and nonstructural protein 3:1073‐1081 epitopes in PBL‐derived, TCR‐gene‐modified T cells. These single TCR‐engineered T cells can CD8‐independently recognize naturally occurring and epidemiologically relevant mutant variants. TCR‐peptide MHC modeling data allow us to rationalize how TCR structural properties accommodate recognition of certain mutated epitopes and how these substitutions impact the requirement of CD8 affinity enhancement for recognition. A better understanding of such TCRs’ promiscuous behavior may allow for exploitation of these properties to develop novel, adoptive T cell‐based therapies for viral infections and cancers exhibiting similar genomic instability.


Cancer Research | 2016

Efficacy of Adoptive T-cell Therapy Is Improved by Treatment with the Antioxidant N-Acetyl Cysteine, Which Limits Activation-Induced T-cell Death

Matthew J. Scheffel; Gina Scurti; Patricia Simms; Elizabeth Garrett-Mayer; Shikhar Mehrotra; Michael I. Nishimura; Christina Voelkel-Johnson

Although adoptive transfer of autologous tumor antigen-specific T-cell immunotherapy can produce remarkable clinical efficacy, most patients do not achieve durable complete responses. We hypothesized that reducing susceptibility of T cells to activation-induced cell death (AICD), which increases during the rapid in vitro expansion of therapeutic T cells before their infusion, might improve the persistence of adoptively transferred cells. Our investigations revealed that repetitive stimulation of the T-cell receptor (TCR) induced AICD, as a result of activating the DNA damage response pathway through ATM-mediated Ser15 phosphorylation of p53. Activation of this DNA damage response pathway also occurred upon antigen-specific restimulation in TCR-transduced TIL1383I T cells prepared for adoptive transfer to patients as part of a clinical trial. Notably, treatment with the antioxidant N-acetyl cysteine (NAC) significantly reduced upregulation of the DNA damage marker γH2AX, subsequent ATM activation, and cell death. In the Pmel mouse model of melanoma, the presence of NAC during ex vivo T-cell expansion improved the persistence of adoptively transferred cells, reduced tumor growth, and increased survival. Taken together, our results offer a preclinical proof of concept for the addition of NAC to current therapeutic T-cell expansion protocols, offering immediate potential to improve the quality and therapeutic efficacy of adoptive T-cell therapeutics infused into patients. Cancer Res; 76(20); 6006-16. ©2016 AACR.


Journal of Immunology | 2015

FOXO3-NF-κB RelA Protein Complexes Reduce Proinflammatory Cell Signaling and Function.

Matthew Thompson; Michelle Larson; Amy Vidrine; Kelly Barrios; Flor Navarro; Kaitlyn Meyers; Patricia Simms; Kushal Prajapati; Lennox Chitsike; Lance M. Hellman; Brian M. Baker; Stephanie K. Watkins

Tumor-associated myeloid cells, including dendritic cells (DCs) and macrophages, are immune suppressive. This study demonstrates a novel mechanism involving FOXO3 and NF-κB RelA that controls myeloid cell signaling and impacts their immune-suppressive nature. We find that FOXO3 binds NF-κB RelA in the cytosol, impacting both proteins by preventing FOXO3 degradation and preventing NF-κB RelA nuclear translocation. The location of protein–protein interaction was determined to be near the FOXO3 transactivation domain. In turn, NF-κB RelA activation was restored upon deletion of the same sequence in FOXO3 containing the DNA binding domain. We have identified for the first time, to our knowledge, a direct protein–protein interaction between FOXO3 and NF-κB RelA in tumor-associated DCs. These detailed biochemical interactions provide the foundation for future studies to use the FOXO3–NF-κB RelA interaction as a target to enhance tumor-associated DC function to support or enhance antitumor immunity.


Journal of Leukocyte Biology | 2017

Comparative exploration of multidimensional flow cytometry software: a model approach evaluating T cell polyfunctional behavior

Timothy T. Spear; Michael I. Nishimura; Patricia Simms

Advancement in flow cytometry reagents and instrumentation has allowed for simultaneous analysis of large numbers of lineage/functional immune cell markers. Highly complex datasets generated by polychromatic flow cytometry require proper analytical software to answer investigators’ questions. A problem among many investigators and flow cytometry Shared Resource Laboratories (SRLs), including our own, is a lack of access to a flow cytometry‐knowledgeable bioinformatics team, making it difficult to learn and choose appropriate analysis tool(s). Here, we comparatively assess various multidimensional flow cytometry software packages for their ability to answer a specific biologic question and provide graphical representation output suitable for publication, as well as their ease of use and cost. We assessed polyfunctional potential of TCR‐transduced T cells, serving as a model evaluation, using multidimensional flow cytometry to analyze 6 intracellular cytokines and degranulation on a per‐cell basis. Analysis of 7 parameters resulted in 128 possible combinations of positivity/negativity, far too complex for basic flow cytometry software to analyze fully. Various software packages were used, analysis methods used in each described, and representative output displayed. Of the tools investigated, automated classification of cellular expression by nonlinear stochastic embedding (ACCENSE) and coupled analysis in Pestle/simplified presentation of incredibly complex evaluations (SPICE) provided the most user‐friendly manipulations and readable output, evaluating effects of altered antigen‐specific stimulation on T cell polyfunctionality. This detailed approach may serve as a model for other investigators/SRLs in selecting the most appropriate software to analyze complex flow cytometry datasets. Further development and awareness of available tools will help guide proper data analysis to answer difficult biologic questions arising from incredibly complex datasets.


Cancer Immunology, Immunotherapy | 2017

Critical biological parameters modulate affinity as a determinant of function in T-cell receptor gene-modified T-cells

Timothy T. Spear; Yuan Wang; Kendra C. Foley; David C. Murray; Gina Scurti; Patricia Simms; Elizabeth Garrett-Mayer; Lance M. Hellman; Brian M. Baker; Michael I. Nishimura

T-cell receptor (TCR)-pMHC affinity has been generally accepted to be the most important factor dictating antigen recognition in gene-modified T-cells. As such, there is great interest in optimizing TCR-based immunotherapies by enhancing TCR affinity to augment the therapeutic benefit of TCR gene-modified T-cells in cancer patients. However, recent clinical trials using affinity-enhanced TCRs in adoptive cell transfer (ACT) have observed unintended and serious adverse events, including death, attributed to unpredicted off-tumor or off-target cross-reactivity. It is critical to re-evaluate the importance of other biophysical, structural, or cellular factors that drive the reactivity of TCR gene-modified T-cells. Using a model for altered antigen recognition, we determined how TCR–pMHC affinity influenced the reactivity of hepatitis C virus (HCV) TCR gene-modified T-cells against a panel of naturally occurring HCV peptides and HCV-expressing tumor targets. The impact of other factors, such as TCR–pMHC stabilization and signaling contributions by the CD8 co-receptor, as well as antigen and TCR density were also evaluated. We found that changes in TCR–pMHC affinity did not always predict or dictate IFNγ release or degranulation by TCR gene-modified T-cells, suggesting that less emphasis might need to be placed on TCR–pMHC affinity as a means of predicting or augmenting the therapeutic potential of TCR gene-modified T-cells used in ACT. A more complete understanding of antigen recognition by gene-modified T-cells and a more rational approach to improve the design and implementation of novel TCR-based immunotherapies is necessary to enhance efficacy and maximize safety in patients.


Clinical Cancer Research | 2018

Inhibition of HER2 Increases JAGGED1-dependent Breast Cancer Stem Cells: Role for Membrane JAGGED1

Deep Shah; Debra Wyatt; Andrew Baker; Patricia Simms; Daniel S. Peiffer; Michelle L Fernandez; Emad A. Rakha; Andrew R. Green; Aleksandra Filipovic; Lucio Miele; Clodia Osipo

Purpose: HER2-positive breast cancer is driven by cells possessing stem-like properties of self-renewal and differentiation, referred to as cancer stem cells (CSC). CSCs are implicated in radiotherapy, chemotherapy resistance, and tumor recurrence. NOTCH promotes breast CSC survival and self-renewal, and overexpression of NOTCH1 and the NOTCH ligand JAGGED1 predict poor outcome. Resistance to anti-HER2 therapy in HER2+ breast cancer requires NOTCH1, and that combination of trastuzumab and a gamma secretase inhibitor (GSI) prevents tumor relapse in xenograft models. Experimental Design: The current study investigates mechanisms by which HER2 tyrosine kinase activity regulates NOTCH-dependent CSC survival and tumor initiation. Results: Lapatinib-mediated HER2 inhibition shifts the population of HER2+ breast cancer cells from low membrane JAGGED1 expression to higher levels, independent of sensitivity to anti-HER2 treatment within the bulk cell population. This increase in membrane JAGGED1 is associated with higher NOTCH receptor expression, activation, and enrichment of CSCs in vitro and in vivo. Importantly, lapatinib treatment results in growth arrest and cell death of JAGGED1 low-expressing cells while the JAGGED1 high-expressing cells continue to cycle. High membrane JAGGED1 protein expression predicts poor overall cumulative survival in women with HER2+ breast cancer. Conclusions: These results indicate that higher membrane JAGGED1 expression may be used to either predict response to anti-HER2 therapy or for detection of NOTCH-sensitive CSCs posttherapy. Sequential blockade of HER2 followed by JAGGED1 or NOTCH could be more effective than simultaneous blockade to prevent drug resistance and tumor progression. Clin Cancer Res; 24(18); 4566–78. ©2018 AACR.

Collaboration


Dive into the Patricia Simms's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elizabeth Garrett-Mayer

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Gina Scurti

Loyola University Chicago

View shared research outputs
Top Co-Authors

Avatar

Thomas M. Ellis

Loyola University Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian M. Baker

University of Notre Dame

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David C. Murray

Loyola University Chicago

View shared research outputs
Top Co-Authors

Avatar

Kendra C. Foley

Loyola University Chicago

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge