Network


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

Hotspot


Dive into the research topics where Elizabeth McMichael is active.

Publication


Featured researches published by Elizabeth McMichael.


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.


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.


Oncotarget | 2017

Targeting tissue factor as a novel therapeutic oncotarget for eradication of cancer stem cells isolated from tumor cell lines, tumor xenografts and patients of breast, lung and ovarian cancer

Zhiwei Hu; Jie Xu; Jijun Cheng; Elizabeth McMichael; Lianbo Yu; William E. Carson

Targeting cancer stem cell (CSC) represents a promising therapeutic approach as it can potentially fight cancer at its root. The challenge is to identify a surface therapeutic oncotarget on CSC. Tissue factor (TF) is known as a common yet specific surface target for cancer cells and tumor neovasculature in several solid cancers. However, it is unknown if TF is expressed by CSCs. Here we demonstrate that TF is constitutively expressed on CD133 positive (CD133+) or CD24-CD44+ CSCs isolated from human cancer cell lines, tumor xenografts from mice and breast tumor tissues from patients. TF-targeted agents, i.e., a factor VII (fVII)-conjugated photosensitizer (fVII-PS for targeted photodynamic therapy) and fVII-IgG1Fc (Immunoconjugate or ICON for immunotherapy), can eradicate CSC via the induction of apoptosis and necrosis and via antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity, respectively. In conclusion, these results demonstrate that TF is a novel surface therapeutic oncotarget for CSC, in addition to cancer cell TF and tumor angiogenic vascular endothelial TF. Moreover, this research highlights that TF-targeting therapeutics can effectively eradicate CSCs, without drug resistance, isolated from breast, lung and ovarian cancer with potential to translate into other most commonly diagnosed solid cancer, in which TF is also highly expressed.


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.


Melanoma Research | 2016

Folate-conjugated immunoglobulin targets melanoma tumor cells for NK cell effector functions.

Cassandra C. Skinner; Elizabeth McMichael; Alena Cristina Jaime-Ramirez; Zachary B. Abrams; Robert J. Lee; William E. Carson

The folate receptor (FR) is overexpressed on the vascular side of cancerous cells including those of the breast, ovaries, testes, and cervix. We hypothesized that a folate-conjugated immunoglobulin (F-IgG) would bind to the FR that is overexpressed on melanoma tumor cells to target these cells for lysis by natural killer (NK) cells. Folate receptor expression was confirmed in the Mel-39 (human melanoma) cell line by flow cytometry and immunoblot analysis using KB (human oral epithelial) and F01 (human melanoma) as a positive and a negative control, respectively. FR-positive and FR-negative cell lines were treated with F-IgG or control immunoglobulin G in the presence or absence of cytokines to determine NK cell ability to lyse FR-positive cell lines. NK cell activation was significantly upregulated and lysis of Mel 39 tumor cells increased following treatment with F-IgG compared with control immunoglobulin G at all effector : target (E : T) ratios (P<0.01). This trend further increased by NK cell stimulation with the activating cytokine interleukin-12. NK cell production of cytokines such as interferon-gamma, macrophage inflammatory protein 1&agr;, and regulated on activation normal T-cell expressed and secreted (RANTES) was also significantly increased in response to costimulation with interleukin-12 stimulation and F-IgG-coated Mel 39 target cells compared with controls (P<0.01). In contrast, F-IgG did not bind to the FR-negative cell line F01 and had no significant effect on NK cell lysis or cytokine production. This research indicates the potential use of F-IgG for its ability to induce an immune response from NK cells against FR-positive melanoma tumor cells, which can be further increased by the addition of cytokines.


Molecular Cancer Therapeutics | 2016

A Phase I Trial to Evaluate Antibody-Dependent Cellular Cytotoxicity of Cetuximab and Lenalidomide in Advanced Colorectal and Head and Neck Cancer

Erin M. Bertino; Elizabeth McMichael; Xiaokui Mo; Prashant Trikha; Melanie E. Davis; Bonnie Paul; Michael R. Grever; William E. Carson; Gregory A. Otterson

mAbs can induce antibody-dependent cellular cytotoxicity (ADCC) via the innate immune systems ability to recognize mAb-coated cancer cells and activate immune effector cells. Lenalidomide is an immunomodulatory agent with the capacity to stimulate immune cell cytokine production and ADCC activity. This phase I trial evaluated the combination of cetuximab with lenalidomide for the treatment of advanced colorectal and head and neck squamous cell cancers (HNSCC). This trial included patients with advanced colorectal cancer or HNSCC. Treatment consisted of cetuximab 500 mg/m2 i.v. every two weeks with lenalidomide given orally days 1–21 on a 28-day cycle. Three dose levels of lenalidomide were evaluated (15, 20, 25 mg). Correlative studies included measurement of ADCC, FcγRIIIA polymorphism genotyping, measurement of serum cytokine levels, and flow cytometric analysis of immune cell subtypes. Twenty-two patients were enrolled (19 colorectal cancer, 3 HNSCC). Fatigue was the only dose-limiting toxicity. One partial response was observed and 8 patients had stable disease at least 12 weeks. The recommended phase II dose is cetuximab 500 mg/m2 with lenalidomide 25 mg daily, days 1–21. Correlative studies demonstrated a dose-dependent increase in natural killer cytotoxic activity with increasing doses of lenalidomide. Cetuximab and lenalidomide were well tolerated. There was a lenalidomide dose-dependent increase in ADCC with higher activity in patients enrolled in cohort 3 than those enrolled in cohorts 1/2. Although response was not a primary endpoint, there was evidence of antitumor activity for the combination therapy. Further investigation of lenalidomide as an immunomodulator in solid tumors is warranted. Mol Cancer Ther; 15(9); 2244–50. ©2016 AACR.


Journal of Biological Chemistry | 2016

Interferon-γ promotes antibody-mediated fratricide of Acute Myeloid Leukemia cells

Kavin Fatehchand; Elizabeth McMichael; Brenda F. Reader; Huiqing Fang; Ramasamy Santhanam; Shalini Gautam; Saranya Elavazhagan; Payal Mehta; Nathaniel J. Buteyn; Giovanna Merchand-Reyes; Sumithira Vasu; Xiaokui Mo; Don M. Benson; James S. Blachly; William E. Carson; John C. Byrd; Jonathan P. Butchar; Susheela Tridandapani

Acute myeloid leukemia (AML) is characterized by the proliferation of immature myeloid lineage blasts. Due to its heterogeneity and to the high rate of acquired drug resistance and relapse, new treatment strategies are needed. Here, we demonstrate that IFNγ promotes AML blasts to act as effector cells within the context of antibody therapy. Treatment with IFNγ drove AML blasts toward a more differentiated state, wherein they showed increased expression of the M1-related markers HLA-DR and CD86, as well as of FcγRI, which mediates effector responses to therapeutic antibodies. Importantly, IFNγ was able to up-regulate CD38, the target of the therapeutic antibody daratumumab. Because the antigen (CD38) and effector receptor (FcγRI) were both simultaneously up-regulated on the AML blasts, we tested whether IFNγ treatment of the AML cell lines THP-1 and MV4-11 could stimulate them to target one another after the addition of daratumumab. Results showed that IFNγ significantly increased daratumumab-mediated cytotoxicity, as measured both by 51Cr release and lactate dehydrogenase release assays. We also found that the combination of IFNγ and activation of FcγR led to the release of granzyme B by AML cells. Finally, using a murine NSG model of subcutaneous AML, we found that treatment with IFNγ plus daratumumab significantly attenuated tumor growth. Taken together, these studies show a novel mechanism of daratumumab-mediated killing and a possible new therapeutic strategy for AML.


Cancer immunology research | 2016

NK Cell–Mediated Antitumor Effects of a Folate-Conjugated Immunoglobulin Are Enhanced by Cytokines

Alena Cristina Jaime-Ramirez; Elizabeth McMichael; Sri Vidya Kondadasula; Cassandra C. Skinner; Bethany L. Mundy-Bosse; Eric Luedke; Natalie B. Jones; Aruna Mani; Julie Roda; Volodymyr Karpa; Hong Li; Jilong Li; Saranya Elavazhagan; Krista La Perle; Alessandra C. Schmitt; Yanhui Lu; Xiaoli Zhang; Xueliang Pan; Hsaioyin Mao; Melanie E. Davis; David Jarjoura; Jonathan P. Butchar; Ming Poi; Mitch A. Phelps; Susheela Tridandapani; John C. Byrd; Michael A. Caligiuri; Robert J. Lee; William E. Carson

Many tumors express receptors for the vitamin folate (FRs). Folate-conjugated IgG, which can bind both FR+ tumors and NK-cell receptors for IgG, induced potent NK-cell antitumor responses that were further augmented by cytokine therapy. Optimally effective antitumor therapies would not only activate immune effector cells but also engage them at the tumor. Folate conjugated to immunoglobulin (F-IgG) could direct innate immune cells with Fc receptors to folate receptor–expressing cancer cells. F-IgG bound to human KB and HeLa cells, as well as murine L1210JF, a folate receptor (FR)–overexpressing cancer cell line, as determined by flow cytometry. Recognition of F-IgG by natural killer (NK) cell Fc receptors led to phosphorylation of the ERK transcription factor and increased NK cell expression of CD69. Lysis of KB tumor cells by NK cells increased by about 5-fold after treatment with F-IgG, an effect synergistically enhanced by treatment with IL2, IL12, IL15, or IL21 (P < 0.001). F-IgG also enhanced the lysis of chronic lymphocytic leukemia cells by autologous NK cells. NK cells significantly increased production of IFNγ, MIP-1α, and RANTES in response to F-IgG–coated KB target cells in the presence of the NK cell–activating cytokine IL12, and these coculture supernatants induced significant T-cell chemotaxis (P < 0.001). F-IgG-coated targets also stimulated FcR-mediated monocyte effector functions. Studies in a murine leukemia model confirmed the intratumoral localization and antitumor activity of F-IgG, as well as enhancement of its effects by IL12 (P = 0.05). The antitumor effect of this combination was dependent on NK cells and led to decreased tumor cell proliferation in vivo. Thus, F-IgG can induce an immune response against FR-positive tumor cells that is mediated by NK cells and can be augmented by cytokine therapy. Cancer Immunol Res; 4(4); 323–36. ©2016 AACR.


Journal for ImmunoTherapy of Cancer | 2015

Interleukin-21 activates natural killer cell activity against cetuximab-coated pancreatic tumor cells

Elizabeth McMichael; Alena Cristin Jaime-Ramirez; Kristan D. Guenterberg; Eric Luedke; Lakhvir S. Atwal; William E. Carson

Natural killer (NK) cells are large granular lymphocytes that play a critical role in anti-cancer mechanisms. These effector cells mediate antibody-dependent cellular cytotoxicity (ADCC) and produce IFN-γ in response to antibody-coated targets. The monoclonal antibody cetuximab targets the epidermal growth factor receptor (EGFR), which is over expressed in the majority of pancreatic cancers, but fails to effectively block MAP kinase activity when KRAS is concurrently mutated. Interleukin-21 (IL-21) is an immunomodulatory cytokine that stimulates the anti-tumor response of NK cells. We hypothesized that IL-21 treatment of NK cells would enhance their response to cetuximab-coated EGFR-positive pancreatic tumor cells, regardless of KRAS mutational status and thereby generate an effective anti-tumor response. Expression of EGFR was measured on six human pancreatic cancer cell lines. All six cell lines showed >85% expression of EGFR by flow cytometry, which was confirmed by immunoblot assay. NK cells from normal healthy donors and pancreatic cancer patients were treated overnight with IL-21 (10 ng/ml) and tested for their ability to lyse cetuximab-coated tumor cells in a standard 4-hour 51Cr release assay. NK cell killing of cetuximab-coated wild-type and mutant KRAS cell lines was significantly higher following NK cell IL-21 treatment, as compared to controls (p < 0.001). In response to cetuximab-coated pancreatic tumor cells, IL-21 treated NK cells secreted significantly higher levels of IFN-γ (p < 0.001), released higher levels of the chemokines IL-8, MIP1α/β, and RANTES (p < 0.001), induced greater chemotaxis of T cells (p < 0.01), and enhanced NK cell signal transduction via activation of ERK and STAT1. Treatment of mice bearing subcutaneous EGFR-positive/KRAS mutant pancreatic tumor xenographs with mIL-21 and cetuximab led to significant inhibition of tumor growth as compared to control conditions (p < 0.05). This result was confirmed in a second intraperitoneal model of pancreatic cancer. Treatment of tumor bearing mice with gemcitabine and cetuximab in combination led to only a modest reduction in tumor burden in vivo (as is seen in humans), but this effect was markedly enhanced by the addition of IL-21 (p < 0.05). Overall, this data suggests that cetuximab treatment in combination with IL-21 can greatly enhance NK cell effector functions and mediate a significant anti-tumor response in EGFR-positive pancreatic cancers in the presence of mutated KRAS.

Collaboration


Dive into the Elizabeth McMichael'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