Network


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

Hotspot


Dive into the research topics where Amanda Campbell is active.

Publication


Featured researches published by Amanda Campbell.


Cancer Research | 2016

Myeloid-derived suppressor cells express Bruton's tyrosine kinase and can be depleted in tumor bearing hosts by ibrutinib treatment

Andrew Stiff; Prashant Trikha; Robert Wesolowski; Kari Kendra; Vincent Hsu; Sarvani Uppati; Elizabeth McMichael; Megan C. Duggan; Amanda Campbell; Karen Keller; Ian Landi; Yiming Zhong; Jason A. Dubovsky; John Harrison Howard; Lianbo Yu; Bonnie K. Harrington; Matthew Old; Sean D. Reiff; Thomas A. Mace; Susheela Tridandapani; Natarajan Muthusamy; Michael A. Caligiuri; John C. Byrd; William E. Carson

Myeloid-derived suppressor cells (MDSC) are a heterogeneous group of immature myeloid cells that expand in tumor-bearing hosts in response to soluble factors produced by tumor and stromal cells. MDSC expansion has been linked to loss of immune effector cell function and reduced efficacy of immune-based cancer therapies, highlighting the MDSC population as an attractive therapeutic target. Ibrutinib, an irreversible inhibitor of Brutons tyrosine kinase (BTK) and IL2-inducible T-cell kinase (ITK), is in clinical use for the treatment of B-cell malignancies. Here, we report that BTK is expressed by murine and human MDSCs, and that ibrutinib is able to inhibit BTK phosphorylation in these cells. Treatment of MDSCs with ibrutinib significantly impaired nitric oxide production and cell migration. In addition, ibrutinib inhibited in vitro generation of human MDSCs and reduced mRNA expression of indolamine 2,3-dioxygenase, an immunosuppressive factor. Treatment of mice bearing EMT6 mammary tumors with ibrutinib resulted in reduced frequency of MDSCs in both the spleen and tumor. Ibrutinib treatment also resulted in a significant reduction of MDSCs in wild-type mice bearing B16F10 melanoma tumors, but not in X-linked immunodeficiency mice (XID) harboring a BTK mutation, suggesting that BTK inhibition plays an important role in the observed reduction of MDSCs in vivo Finally, ibrutinib significantly enhanced the efficacy of anti-PD-L1 (CD274) therapy in a murine breast cancer model. Together, these results demonstrate that ibrutinib modulates MDSC function and generation, revealing a potential strategy for enhancing immune-based therapies in solid malignancies. Cancer Res; 76(8); 2125-36. ©2016 AACR.


Journal of Biological Chemistry | 2016

Analysis of the Effects of the Bruton's tyrosine kinase (Btk) Inhibitor Ibrutinib on Monocyte Fcγ Receptor (FcγR) Function.

Li Ren; Amanda Campbell; Huiqing Fang; Shalini Gautam; Saranya Elavazhagan; Kavin Fatehchand; Payal Mehta; Andrew Stiff; Brenda F. Reader; Xiaokui Mo; John C. Byrd; William E. Carson; Jonathan P. Butchar; Susheela Tridandapani

The irreversible Brutons tyrosine kinase (Btk) inhibitor ibrutinib has shown efficacy against B-cell tumors such as chronic lymphocytic leukemia and B-cell non-Hodgkin lymphoma. Fcγ receptors (FcγR) on immune cells such as macrophages play an important role in tumor-specific antibody-mediated immune responses, but many such responses involve Btk. Here we tested the effects of ibrutinib on FcγR-mediated activities in monocytes. We found that ibrutinib did not affect monocyte FcγR-mediated phagocytosis, even at concentrations higher than those achieved physiologically, but suppressed FcγR-mediated cytokine production. We confirmed these findings in macrophages from Xid mice in which Btk signaling is defective. Because calcium flux is a major event downstream of Btk, we tested whether it was involved in phagocytosis. The results showed that blocking intracellular calcium flux decreased FcγR-mediated cytokine production but not phagocytosis. To verify this, we measured activation of the GTPase Rac, which is responsible for actin polymerization. Results showed that ibrutinib did not inhibit Rac activation, nor did the calcium chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid tetrakis(acetoxymethyl ester). We next asked whether the effect of ibrutinib on monocyte FcγR-mediated cytokine production could be rescued by IFNγ priming because NK cells produce IFNγ in response to antibody therapy. Pretreatment of monocytes with IFNγ abrogated the effects of ibrutinib on FcγR-mediated cytokine production, suggesting that IFNγ priming could overcome this Btk inhibition. Furthermore, in monocyte-natural killer cell co-cultures, ibrutinib did not inhibit FcγR-mediated cytokine production despite doing so in single cultures. These results suggest that combining ibrutinib with monoclonal antibody therapy could enhance chronic lymphocytic leukemia cell killing without affecting macrophage effector function.


Clinical Cancer Research | 2017

IL-21 Enhances Natural Killer Cell Response to Cetuximab-Coated Pancreatic Tumor Cells.

Elizabeth McMichael; Alena Cristina Jaime-Ramirez; Kristan D. Guenterberg; Eric Luedke; Lakhvir S. Atwal; Amanda Campbell; Zhiwei Hu; Armika S. Tatum; Sri Vidya Kondadasula; Xiaokui Mo; Susheela Tridandapani; Mark Bloomston; E. Christopher Ellison; Terence M. Williams; Tanios Bekaii-Saab; William E. Carson

Purpose: Alternative strategies to EGFR blockage by mAbs is necessary to improve the efficacy of therapy in patients with locally advanced or metastatic pancreatic cancer. One such strategy includes the use of NK cells to clear cetuximab-coated tumor cells, as need for novel therapeutic approaches to enhance the efficacy of cetuximab is evident. We show that IL-21 enhances NK cell-mediated effector functions against cetuximab-coated pancreatic tumor cells irrespective of KRAS mutation status. Experimental Design: NK cells from normal donors or donors with pancreatic cancer were used to assess ADCC, IFN-γ release, and T-cell chemotaxis toward human pancreatic cancer cell lines. The in vivo efficacy of IL-21 in combination with cetuximab was evaluated in a subcutaneous and intraperitoneal model of pancreatic cancer. Results: NK cell lysis of cetuximab-coated wild-type and mutant kras pancreatic cancer cell lines were significantly higher following NK cell IL-21 treatment. In response to cetuximab-coated pancreatic tumor cells, IL-21–treated NK cells secreted significantly higher levels of IFN-γ and chemokines, increased chemotaxis of T cells, and enhanced NK cell signal transduction via activation of ERK and STAT1. Treatment of mice bearing subcutaneous or intraperitoneal EGFR-positive pancreatic tumor xenografts with mIL-21 and cetuximab led to significant inhibition of tumor growth, a result further enhanced by the addition of gemcitabine. Conclusions: These results suggest that cetuximab treatment in combination with IL-21 adjuvant therapy in patients with EGFR-positive pancreatic cancer results in significant NK cell activation, irrespective of KRAS mutation status, and may be a potential therapeutic strategy. Clin Cancer Res; 23(2); 489–502. ©2016 AACR.


Journal of Immunology | 2015

The Raf Kinase Inhibitor Sorafenib Inhibits JAK–STAT Signal Transduction in Human Immune Cells

Sara Martin del Campo; Kala M. Levine; Bethany L. Mundy-Bosse; Valerie P. Grignol; Ene T. Fairchild; Amanda Campbell; Prashant Trikha; Thomas A. Mace; Bonnie Paul; Alena Cristina Jaime-Ramirez; Joseph Markowitz; Sri Vidya Kondadasula; Kristan D. Guenterberg; Susan McClory; Volodymyr Karpa; Xueliang Pan; Thomas Olencki; J. Paul Monk; Amir Mortazavi; Susheela Tridandapani; Gregory B. Lesinski; John C. Byrd; Michael A. Caligiuri; Manisha H. Shah; William E. Carson

Sorafenib is an oral multikinase inhibitor that was originally developed as a Raf kinase inhibitor. We hypothesized that sorafenib would also have inhibitory effects on cytokine signaling pathways in immune cells. PBMCs from normal donors were treated with varying concentrations of sorafenib and stimulated with IFN-α or IL-2. Phosphorylation of STAT1 and STAT5 was measured by flow cytometry and confirmed by immunoblot analysis. Changes in IFN-α– and IL-2–stimulated gene expression were measured by quantitative PCR, and changes in cytokine production were evaluated by ELISA. Cryopreserved PBMCs were obtained from cancer patients before and after receiving 400 mg sorafenib twice daily. Patient PBMCs were thawed, stimulated with IL-2 or IFN-α, and evaluated for phosphorylation of STAT1 and STAT5. Pretreatment of PBMCs with 10 μM sorafenib decreased STAT1 and STAT5 phosphorylation after treatment with IFN-α or IL-2. This inhibitory effect was observed in PBMCs from healthy donors over a range of concentrations of sorafenib (5–20 μM), IL-2 (2–24 nM), and IFN-α (101–106 U/ml). This effect was observed in immune cell subsets, including T cells, B cells, NK cells, regulatory T cells, and myeloid-derived suppressor cells. Pretreatment with sorafenib also inhibited PBMC expression of IFN-α– and IL-2–regulated genes and inhibited NK cell production of IFN-γ, RANTES, MIP1-α, and MIG in response to IFN-α stimulation. PBMCs from patients receiving sorafenib therapy showed decreased responsiveness to IL-2 and IFN-α treatment. Sorafenib is a Raf kinase inhibitor that could have off-target effects on cytokine-induced signal transduction in immune effector cells.


Cancer immunology research | 2017

MICA-Expressing Monocytes Enhance Natural Killer Cell Fc Receptor-Mediated Antitumor Functions

Amanda Campbell; Megan C. Duggan; Lorena P. Suarez-Kelly; Neela Bhave; Kallan S. Opheim; Elizabeth McMichael; Prashant Trikha; Robin Parihar; Eric Luedke; Adrian Lewis; Bryant C. Yung; Robert J. Lee; David H. Raulet; Susheela Tridandapani; Veronika Groh; Lianbo Yu; Vedat O. Yildiz; John C. Byrd; Michael A. Caligiuri; William E. Carson

Natural killer (NK) cells secrete immunostimulatory factors like IFNγ in response to tumors. Engagement of monocyte MICA and NK cell NKG2D promoted and enhanced the NK response to HER2+ breast tumors treated with mAb to HER2 in a murine model. Natural killer (NK) cells are large granular lymphocytes that promote the antitumor response via communication with other cell types in the tumor microenvironment. Previously, we have shown that NK cells secrete a profile of immune stimulatory factors (e.g., IFNγ, MIP-1α, and TNFα) in response to dual stimulation with the combination of antibody (Ab)-coated tumor cells and cytokines, such as IL12. We now demonstrate that this response is enhanced in the presence of autologous monocytes. Monocyte enhancement of NK cell activity was dependent on cell-to-cell contact as determined by a Transwell assay. It was hypothesized that NK cell effector functions against Ab-coated tumor cells were enhanced via binding of MICA on monocytes to NK cell NKG2D receptors. Strategies to block MICA–NKG2D interactions resulted in reductions in IFNγ production. Depletion of monocytes in vivo resulted in decreased IFNγ production by murine NK cells upon exposure to Ab-coated tumor cells. In mice receiving trastuzumab and IL12 therapy, monocyte depletion resulted in significantly greater tumor growth in comparison to mock-depleted controls (P < 0.05). These data suggest that NK cell–monocyte interactions enhance NK cell antitumor activity in the setting of monoclonal Ab therapy for cancer. Cancer Immunol Res; 5(9); 778–89. ©2017 AACR.


Clinical Cancer Research | 2018

Nitric Oxide Production by Myeloid Derived Suppressor Cells Plays a Role in Impairing Fc Receptor-Mediated Natural Killer Cell Function.

Andrew Stiff; Prashant Trikha; Bethany L. Mundy-Bosse; Elizabeth McMichael; Thomas A. Mace; Brooke Benner; Kari Kendra; Amanda Campbell; Shalini Gautam; David Abood; Ian Landi; Vincent Hsu; Megan C. Duggan; Robert Wesolowski; Matthew Old; John Harrison Howard; Lianbo Yu; Nancy Stasik; Thomas Olencki; Natarajan Muthusamy; Susheela Tridandapani; John C. Byrd; Michael A. Caligiuri; William E. Carson

Purpose: mAbs are used to treat solid and hematologic malignancies and work in part through Fc receptors (FcRs) on natural killer cells (NK). However, FcR-mediated functions of NK cells from patients with cancer are significantly impaired. Identifying the mechanisms of this dysfunction and impaired response to mAb therapy could lead to combination therapies and enhance mAb therapy. Experimental Design: Cocultures of autologous NK cells and MDSC from patients with cancer were used to study the effect of myeloid-derived suppressor cells (MDSCs) on NK-cell FcR-mediated functions including antibody-dependent cellular cytotoxicity, cytokine production, and signal transduction in vitro. Mouse breast cancer models were utilized to study the effect of MDSCs on antibody therapy in vivo and test the efficacy of combination therapies including a mAb and an MDSC-targeting agent. Results: MDSCs from patients with cancer were found to significantly inhibit NK-cell FcR-mediated functions including antibody-dependent cellular cytotoxicity, cytokine production, and signal transduction in a contact-independent manner. In addition, adoptive transfer of MDSCs abolished the efficacy of mAb therapy in a mouse model of pancreatic cancer. Inhibition of iNOS restored NK-cell functions and signal transduction. Finally, nonspecific elimination of MDSCs or inhibition of iNOS in vivo significantly improved the efficacy of mAb therapy in a mouse model of breast cancer. Conclusions: MDSCs antagonize NK-cell FcR-mediated function and signal transduction leading to impaired response to mAb therapy in part through nitric oxide production. Thus, elimination of MDSCs or inhibition of nitric oxide production offers a strategy to improve mAb therapy. Clin Cancer Res; 24(8); 1891–904. ©2018 AACR.


BMC Medical Genomics | 2015

Gene expression profiling of the human natural killer cell response to Fc receptor activation: unique enhancement in the presence of interleukin-12

Amanda Campbell; Kelly Regan; Neela Bhave; Arka Pattanayak; Robin Parihar; Andrew Stiff; Prashant Trikha; Steven D. Scoville; Sandya Liyanarachchi; Sri Vidya Kondadasula; Omkar Lele; Ramana V. Davuluri; Philip R. O. Payne; William E. Carson

BackgroundTraditionally, the CD56dimCD16+ subset of Natural Killer (NK) cells has been thought to mediate cellular cytotoxicity with modest cytokine secretion capacity. However, studies have suggested that this subset may exert a more diverse array of immunological functions. There exists a lack of well-developed functional models to describe the behavior of activated NK cells, and the interactions between signaling pathways that facilitate effector functions are not well understood. In the present study, a combination of genome-wide microarray analyses and systems-level bioinformatics approaches were utilized to elucidate the transcriptional landscape of NK cells activated via interactions with antibody-coated targets in the presence of interleukin-12 (IL-12).MethodsWe conducted differential gene expression analysis of CD56dimCD16+ NK cells following FcR stimulation in the presence or absence of IL-12. Next, we functionally characterized gene sets according to patterns of gene expression and validated representative genes using RT-PCR. IPA was utilized for biological pathway analysis, and an enriched network of interacting genes was generated using GeneMANIA. Furthermore, PAJEK and the HITS algorithm were employed to identify important genes in the network according to betweeness centrality, hub, and authority node metrics.ResultsAnalyses revealed that CD56dimCD16+ NK cells co-stimulated via the Fc receptor (FcR) and IL-12R led to the expression of a unique set of genes, including genes encoding cytotoxicity receptors, apoptotic proteins, intracellular signaling molecules, and cytokines that may mediate enhanced cytotoxicity and interactions with other immune cells within inflammatory tissues. Network analyses identified a novel set of connected key players, BATF, IRF4, TBX21, and IFNG, within an integrated network composed of differentially expressed genes in NK cells stimulated by various conditions (immobilized IgG, IL-12, or the combination of IgG and IL-12).ConclusionsThese results are the first to address the global mechanisms by which NK cells mediate their biological functions when encountering antibody-coated targets within inflammatory sites. Moreover, this study has identified a set of high-priority targets for subsequent investigation into strategies to combat cancer by enhancing the anti-tumor activity of CD56dimCD16+ NK cells.


Proceedings of SPIE | 2016

Fluorescent nanodiamonds and their use in biomedical research

Lorena P. Suarez-Kelly; Isaac Rampersaud; Charles E. Moritz; Amanda Campbell; Zhiwei Hu; Masfer H. Alkahtani; Fahad Alghannam; Phillip Hemmer; William E. Carson; Arfaan A. Rampersaud

Nanodiamonds containing color-centers produce non-quenching fluorescence that is easily detected. This makes them useful for cellular, proteomic and genomic applications. However, fluorescent nanodiamonds have yet to become popular in the biomedical research community as labeling reagents. We discuss production of nanodiamonds with distinct color-centers and assess their biocompatibility and techniques for bioconjugation. Fluorescent diamonds were fabricated by electron irradiation of high-pressure, high-temperature micron-sized diamonds which generated diamonds with vacancy-related defects (V). These diamonds were annealed to create nitrogen vacancy (NV)-centers then following a milling step were fractionated into nanoparticle sizes of 30, 60, and 95 nm. Optical characterization of Vand NV-center diamonds demonstrated fluorescence in two distinct green and red channels, respectively. In vitro studies demonstrated that these nanodiamonds are biocompatible and readily taken up by murine macrophage cells. Quantification of NV-center nanodiamond uptake by flow cytometry, showed that uptake was independent of nanodiamond size. Confocal microscopy demonstrated that NV-center nanodiamonds accumulate within the cytoplasm of these cells. NV-center nanodiamonds were then conjugated with streptavidin using a short polyethylene chain as linker. Conjugation was confirmed via a catalytic assay employing biotinylated-horseradish peroxidase. We present a technique for large-scale production of biocompatible conjugated V- or NV-center nanodiamonds. Functional testing is essential for standardization of fluorescent nanodiamond bioconjugates and quality control. Large-scale production of bioconjugated fluorescent nanodiamonds is crucial to their development as novel tools for biological and medical applications.


OncoImmunology | 2018

Co-stimulation of the fc receptor and interleukin-12 receptor on human natural killer cells leads to increased expression of cd25

Megan C. Duggan; Amanda Campbell; Elizabeth McMichael; Kallan S. Opheim; Kala M. Levine; Neela Bhave; Michelle C. Culbertson; Tiffany Noel; Lianbo Yu; We Carson rd

ABSTRACT Natural killer (NK) cells serve a critical role in the immune response against microbes and developing tumors. We have demonstrated that NK cells produce stimulatory cytokines (e.g., IFN-γ) in response to potent stimulation via immobilized IgG (to engage Fc receptors) and interleukin (IL)-12. CD25 is a component of the high-affinity IL-2R, which promotes NK cell activation in response to low doses of IL-2 such as those released by activated T cells. We hypothesized that stimulation of NK cells via IgG and IL-12 would enhance CD25 expression and promote NK cell anti-tumor activity in response to low-dose IL-2. It was confirmed that this dual stimulation strategy significantly enhanced NK cell CD25 expression compared to unstimulated cells or cells treated with IgG or IL-12 alone. Dual stimulated NK cells also were more responsive to low-dose IL-2. Dual stimulated NK cells subsequently treated with low-dose IL-2 (10 pg/mL) displayed enhanced intracellular signaling as indicated by increased pSTAT5 levels. IFN-γ production and cytotoxicity against K562 cells by NK cells stimulated with low-dose IL-2 was comparable to that of cells treated with high-dose IL-2 (10 ng/mL). Importantly, cells isolated from head and neck cancer patients receiving the mAb cetuximab and IL-12 on a clinical trial displayed increased CD25 expression following combination therapy compared to baseline. Altogether, these findings suggest that FcR and IL-12R co-stimulation induces expression of the high-affinity IL-2R and promotes NK cell anti-tumor activity.


OncoImmunology | 2017

Activation of the FcgammaReceptorIIIa on human natural killer cells leads to increased expression of functional interleukin-21 receptor

Elizabeth McMichael; Nicholas B. Courtney; Megan C. Duggan; Robert Wesolowski; Dionisia Quiroga; Sri Vidya Kondadasula; Lakhvir S. Atwal; Neela Bhave; Eric Luedke; Alena Cristina Jaime-Ramirez; Amanda Campbell; Xiaokui Mo; John C. Byrd; William E. Carson

ABSTRACT Natural killer (NK) cells are innate immune effector cells that play a crucial role in immune surveillance and the destruction of cancer cells. NK cells express a low-affinity receptor for the Fc or constant region of immunoglobulin G (FcγRIIIa) and multiple cytokine receptors that respond to antibody-coated targets and cytokines in the tumor microenvironment. In the present work, microarray gene expression analysis revealed that the IL-21 receptor (IL-21R) was strongly upregulated following FcR stimulation. The IL-21R was found to be upregulated on FcR-stimulated NK cells at the transcript level as determined by reverse transcription polymerase chain reaction (RT-PCR). Immunoblot analysis revealed that protein expression of the IL-21R peaked at 8 h post-stimulation of the FcR. Inhibition of the mitogen-activated protein kinase (MAPK) pathway downstream of the FcR blocked the induction of IL-21R expression. Increased expression of the IL-21R sensitized NK cells to IL-21 stimulation, as treatment of FcR-stimulated NK cells led to significantly increased phosphorylation of STAT1 and STAT3, as measured by intracellular flow cytometry and immunoblot analysis. Following FcR-stimulation, IL-21-activated NK cells were better able to mediate the lysis of trastuzumab-coated human epidermal growth factor receptor 2 (HER2+) SK-BR-3 tumor cells as compared to control-treated cells. Likewise, IL-21-induced NK cell secretion of IFNγ following exposure to antibody-coated tumor cells was enhanced following FcR-stimulation. The analysis of NK cells from patients receiving trastuzumab therapy for HER2+ cancer exhibited increased levels of the IL-21R following the administration of antibody suggesting that the presence of monoclonal antibody-coated tumor cells in vivo can stimulate the increased expression of IL-21R on NK cells.

Collaboration


Dive into the Amanda Campbell's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lianbo Yu

Ohio State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zhiwei Hu

Ohio State University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge