Network


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

Hotspot


Dive into the research topics where Jeffrey W. Leong is active.

Publication


Featured researches published by Jeffrey W. Leong.


Blood | 2012

Cytokine activation induces human memory-like NK cells

Rizwan Romee; Stephanie Schneider; Jeffrey W. Leong; Julie M. Chase; Catherine R. Keppel; Ryan P. Sullivan; Megan A. Cooper; Todd A. Fehniger

Natural killer (NK) cells are lymphocytes that play an important role in the immune response to infection and malignancy. Recent studies in mice have shown that stimulation of NK cells with cytokines or in the context of a viral infection results in memory-like properties. We hypothesized that human NK cells exhibit such memory-like properties with an enhanced recall response after cytokine preactivation. In the present study, we show that human NK cells preactivated briefly with cytokine combinations including IL-12, IL-15, and IL-18 followed by a 7- to 21-day rest have enhanced IFN-γ production after restimulation with IL-12 + IL-15, IL-12 + IL-18, or K562 leukemia cells. This memory-like phenotype was retained in proliferating NK cells. In CD56(dim) NK cells, the memory-like IFN-γ response was correlated with the expression of CD94, NKG2A, NKG2C, and CD69 and a lack of CD57 and KIR. Therefore, human NK cells have functional memory-like properties after cytokine activation, which provides a novel rationale for integrating preactivation with combinations of IL-12, IL-15, and IL-18 into NK cell immunotherapy strategies.


Science Translational Medicine | 2016

Cytokine-induced memory-like natural killer cells exhibit enhanced responses against myeloid leukemia

Rizwan Romee; Maximillian Rosario; Melissa M. Berrien-Elliott; Julia A. Wagner; Brea A. Jewell; Timothy Schappe; Jeffrey W. Leong; Sara Abdel-Latif; Stephanie Schneider; Sarah Willey; Carly Neal; Liyang Yu; Stephen T. Oh; Yi Shan Lee; Arend Mulder; Frans H.J. Claas; Megan A. Cooper; Todd A. Fehniger

Cytokine-induced differentiation of memory-like natural killer cells enhances antileukemia responses. Natural killers of leukemia Natural killer cells, part of the innate immune system, play a role in immune responses against exogenous pathogens as well as cancer. Recent studies have identified the existence of memory-like characteristics in some natural killer cells, and Romee et al. investigated these memory cells’ potential as a cancer therapy. The authors compared human natural killer memory cells to non-memory control cells, then demonstrated their effectiveness against myeloid leukemia models in vitro and in mice. They also performed a clinical trial in human patients with acute myeloid leukemia, where the natural killer memory-like cells again demonstrated antileukemia effects, some of which produced clinical remissions. Natural killer (NK) cells are an emerging cellular immunotherapy for patients with acute myeloid leukemia (AML); however, the best approach to maximize NK cell antileukemia potential is unclear. Cytokine-induced memory-like NK cells differentiate after a brief preactivation with interleukin-12 (IL-12), IL-15, and IL-18 and exhibit enhanced responses to cytokine or activating receptor restimulation for weeks to months after preactivation. We hypothesized that memory-like NK cells exhibit enhanced antileukemia functionality. We demonstrated that human memory-like NK cells have enhanced interferon-γ production and cytotoxicity against leukemia cell lines or primary human AML blasts in vitro. Using mass cytometry, we found that memory-like NK cell functional responses were triggered against primary AML blasts, regardless of killer cell immunoglobulin-like receptor (KIR) to KIR-ligand interactions. In addition, multidimensional analyses identified distinct phenotypes of control and memory-like NK cells from the same individuals. Human memory-like NK cells xenografted into mice substantially reduced AML burden in vivo and improved overall survival. In the context of a first-in-human phase 1 clinical trial, adoptively transferred memory-like NK cells proliferated and expanded in AML patients and demonstrated robust responses against leukemia targets. Clinical responses were observed in five of nine evaluable patients, including four complete remissions. Thus, harnessing cytokine-induced memory-like NK cell responses represents a promising translational immunotherapy approach for patients with AML.


Biology of Blood and Marrow Transplantation | 2014

Preactivation with IL-12, IL-15, and IL-18 induces CD25 and a functional high-affinity IL-2 receptor on human cytokine-induced memory-like natural killer cells.

Jeffrey W. Leong; Julie M. Chase; Rizwan Romee; Stephanie Schneider; Ryan P. Sullivan; Megan A. Cooper; Todd A. Fehniger

Natural killer (NK) cells are effector lymphocytes that are under clinical investigation for the adoptive immunotherapy of hematologic malignancies, especially acute myeloid leukemia. Recent work in mice has identified innate memory-like properties of NK cells. Human NK cells also exhibit memory-like properties, and cytokine-induced memory-like (CIML) NK cells are generated via brief preactivation with IL-12, IL-15, and IL-18, which later exhibit enhanced functionality upon restimulation. However, the optimal cytokine receptors and signals for maintenance of enhanced function and homeostasis after preactivation remain unclear. Here, we show that IL-12, IL-15, and IL-18 preactivation induces a rapid and prolonged expression of CD25, resulting in a functional high-affinity IL-2 receptor (IL-2Rαβγ) that confers responsiveness to picomolar concentrations of IL-2. The expression of CD25 correlated with STAT5 phosphorylation in response to picomolar concentrations of IL-2, indicating the presence of a signal-competent IL-2Rαβγ. Furthermore, picomolar concentrations of IL-2 acted synergistically with IL-12 to costimulate IFN-γ production by preactivated NK cells, an effect that was CD25 dependent. Picomolar concentrations of IL-2 also enhanced NK cell proliferation and cytotoxicity via the IL-2Rαβγ. Further, after adoptive transfer into immunodeficient NOD-SCID-γc(-/-) mice, human cytokine-preactivated NK cells expand preferentially in response to exogenous IL-2. Collectively, these data demonstrate that human CIML NK cells respond to IL-2 via IL-2Rαβγ with enhanced survival and functionality, and they provide additional rationale for immunotherapeutic strategies that include brief cytokine preactivation before adoptive NK cell transfer, followed by low-dose IL-2 therapy.


Scientifica | 2014

Utilizing Cytokines to Function-Enable Human NK Cells for the Immunotherapy of Cancer

Rizwan Romee; Jeffrey W. Leong; Todd A. Fehniger

Natural killer (NK) cells are innate lymphoid cells important for host defense against pathogens and mediate antitumor immunity. Cytokine receptors transduce important signals that regulate proliferation, survival, activation status, and trigger effector functions. Here, we review the roles of major cytokines that regulate human NK cell development, survival, and function, including IL-2, IL-12, IL-15, IL-18, and IL-21, and their translation to the clinic as immunotherapy agents. We highlight a recent development in NK cell biology, the identification of innate NK cell memory, and focus on cytokine-induced memory-like (CIML) NK cells that result from a brief, combined activation with IL-12, IL-15, and IL-18. This activation results in long lived NK cells that exhibit enhanced functionality when they encounter a secondary stimulation and provides a new approach to enable NK cells for enhanced responsiveness to infection and cancer. An improved understanding of the cellular and molecular aspects of cytokine-cytokine receptor signals has led to a resurgence of interest in the clinical use of cytokines that sustain and/or activate NK cell antitumor potential. In the future, such strategies will be combined with negative regulatory signal blockade and enhanced recognition to comprehensively enhance NK cells for immunotherapy.


Journal of Immunology | 2012

MicroRNA-Deficient NK Cells Exhibit Decreased Survival but Enhanced Function

Ryan P. Sullivan; Jeffrey W. Leong; Stephanie Schneider; Catherine R. Keppel; Elizabeth Germino; Anthony R. French; Todd A. Fehniger

NK cells are innate immune lymphocytes important for early host defense against infectious pathogens and malignant transformation. MicroRNAs (miRNAs) are small RNA molecules that regulate a wide variety of cellular processes, typically by specific complementary targeting of the 3′UTR of mRNAs. The Dicer1 gene encodes a conserved enzyme essential for miRNA processing, and Dicer1 deficiency leads to a global defect in miRNA biogenesis. In this study, we report a mouse model of lymphocyte-restricted Dicer1 disruption to evaluate the role of Dicer1-dependent miRNAs in the development and function of NK cells. As expected, Dicer1-deficient NK cells had decreased total miRNA content. Furthermore, miRNA-deficient NK cells exhibited reduced survival and impaired maturation defined by cell surface phenotypic markers. However, Dicer1-deficient NK cells exhibited enhanced degranulation and IFN-γ production in vitro in response to cytokines, tumor target cells, and activating NK cell receptor ligation. Moreover, a similar phenotype of increased IFN-γ was evident during acute MCMV infection in vivo. miRs-15a/15b/16 were identified as abundant miRNAs in NK cells that directly target the murine IFN-γ 3′UTR, thereby providing a potential mechanism for enhanced IFN-γ production. These data suggest that the function of miRNAs in NK cell biology is complex, with an important role in NK cell development, survival, or homeostasis, while tempering peripheral NK cell activation. Further study of individual miRNAs in an NK cell specific fashion will provide insight into these complex miRNA regulatory effects in NK cell biology.


Clinical Cancer Research | 2016

The IL-15-Based ALT-803 Complex Enhances FcγRIIIa-Triggered NK Cell Responses and in Vivo Clearance of B Cell Lymphomas

Maximillian Rosario; Bai Liu; Lin Kong; Lynne Collins; Stephanie Schneider; Xiaoyue Chen; Kaiping Han; Emily K. Jeng; Peter R. Rhode; Jeffrey W. Leong; Timothy Schappe; Brea A. Jewell; Catherine R. Keppel; Keval Shah; Brian Hess; Rizwan Romee; David Piwnica-Worms; Amanda F. Cashen; Nancy L. Bartlett; Hing C. Wong; Todd A. Fehniger

Purpose: Anti-CD20 monoclonal antibodies (mAb) are an important immunotherapy for B-cell lymphoma, and provide evidence that the immune system may be harnessed as an effective lymphoma treatment approach. ALT-803 is a superagonist IL-15 mutant and IL-15Rα–Fc fusion complex that activates the IL-15 receptor constitutively expressed on natural killer (NK) cells. We hypothesized that ALT-803 would enhance anti–CD20 mAb-directed NK-cell responses and antibody-dependent cellular cytotoxicity (ADCC). Experimental Design: We tested this hypothesis by adding ALT-803 immunostimulation to anti-CD20 mAb triggering of NK cells in vitro and in vivo. Cell lines and primary human lymphoma cells were utilized as targets for primary human NK cells. Two complementary in vivo mouse models were used, which included human NK-cell xenografts in NOD/SCID-γc−/− mice. Results: We demonstrate that short-term ALT-803 stimulation significantly increased degranulation, IFNγ production, and ADCC by human NK cells against B-cell lymphoma cell lines or primary follicular lymphoma cells. ALT-803 augmented cytotoxicity and the expression of granzyme B and perforin, providing one potential mechanism for this enhanced functionality. Moreover, in two distinct in vivo B-cell lymphoma models, the addition of ALT-803 to anti-CD20 mAb therapy resulted in significantly reduced tumor cell burden and increased survival. Long-term ALT-803 stimulation of human NK cells induced proliferation and NK-cell subset changes with preserved ADCC. Conclusions: ALT-803 represents a novel immunostimulatory drug that enhances NK-cell antilymphoma responses in vitro and in vivo, thereby supporting the clinical investigation of ALT-803 plus anti-CD20 mAbs in patients with indolent B-cell lymphoma. Clin Cancer Res; 22(3); 596–608. ©2015 AACR.


Frontiers in Immunology | 2013

MicroRNA regulation of natural killer cells

Ryan P. Sullivan; Jeffrey W. Leong; Todd A. Fehniger

Natural killer (NK) cells are innate immune lymphocytes critical for host defense against viral infection and surveillance against malignant transformation. MicroRNAs (miRNAs) are a family of small, non-coding RNAs that regulate a wide variety of cellular processes. Recent advances have highlighted the importance of miRNA-mediated post-transcriptional regulation in NK cell development, maturation, and function. This review focuses on several facets of this regulatory mechanism in NK cells: (1) the expressed NK cell miRNA transcriptome; (2) the impact of total miRNA deficiency on NK cells; (3) the role of specific miRNAs regulating NK cell development, survival, and maturation; (4) the intrinsic role of miRNAs regulating NK cell function, including cytokine production, proliferation, and cytotoxicity; and (5) the role of NK cell miRNAs in disease. Currently our knowledge of how miRNAs regulate NK cell biology is limited, and thus we also explore key open questions in the field, as well as approaches and techniques to ascertain the role of individual miRNAs as important molecular regulators.


Journal of Immunology | 2013

MicroRNA-155 Tunes Both the Threshold and Extent of NK Cell Activation via Targeting of Multiple Signaling Pathways

Ryan P. Sullivan; Leslie A. Fogel; Jeffrey W. Leong; Stephanie Schneider; Rachel Wong; Rizwan Romee; To-Ha Thai; Veronika Sexl; Scot J. Matkovich; Gerald W. Dorn; Anthony R. French; Todd A. Fehniger

NK cells are innate lymphocytes important for host defense against viral infections and malignancy. However, the molecular programs orchestrating NK cell activation are incompletely understood. MicroRNA-155 (miR-155) is markedly upregulated following cytokine activation of human and mouse NK cells. Surprisingly, mature human and mouse NK cells transduced to overexpress miR-155, NK cells from mice with NK cell–specific miR-155 overexpression, and miR-155−/− NK cells all secreted more IFN-γ compared with controls. Investigating further, we found that activated NK cells with miR-155 overexpression had increased per-cell IFN-γ with normal IFN-γ+ percentages, whereas greater percentages of miR-155−/− NK cells were IFN-γ+. In vivo murine CMV–induced IFN-γ expression by NK cells in these miR-155 models recapitulated the in vitro phenotypes. We performed unbiased RNA-induced silencing complex sequencing on wild-type and miR-155−/− NK cells and found that mRNAs targeted by miR-155 were enriched in NK cell activation signaling pathways. Using specific inhibitors, we confirmed these pathways were mechanistically involved in regulating IFN-γ production by miR-155−/− NK cells. These data indicate that miR-155 regulation of NK cell activation is complex and that miR-155 functions as a dynamic tuner for NK cell activation via both setting the activation threshold as well as controlling the extent of activation in mature NK cells. In summary, miR-155−/− NK cells are more easily activated, through increased expression of proteins in the PI3K, NF-κB, and calcineurin pathways, and miR-155−/− and 155-overexpressing NK cells exhibit increased IFN-γ production through distinct cellular mechanisms.


Journal of Immunology | 2015

MicroRNA-15/16 Antagonizes Myb To Control NK Cell Maturation

Ryan P. Sullivan; Jeffrey W. Leong; Stephanie Schneider; Aaron R. Ireland; Melissa M. Berrien-Elliott; Anvita Singh; Timothy Schappe; Brea A. Jewell; Sexl; Todd A. Fehniger

NK cells develop in the bone marrow and complete their maturation in peripheral organs, but the molecular events controlling maturation are incompletely understood. The miR-15/16 family of microRNA regulates key cellular processes and is abundantly expressed in NK cells. In this study, we identify a critical role for miR-15/16 in the normal maturation of NK cells using a mouse model of NK-specific deletion, in which immature NK cells accumulate in the absence of miR-15/16. The transcription factor c-Myb (Myb) is expressed preferentially by immature NK cells, is a direct target of miR-15/16, and is increased in 15a/16-1 floxed knockout NK cells. Importantly, maturation of 15a/16-1 floxed knockout NK cells was rescued by Myb knockdown. Moreover, Myb overexpression in wild-type NK cells caused a defective NK cell maturation phenotype similar to deletion of miR-15/16, and Myb overexpression enforces an immature NK cell transcriptional profile. Thus, miR-15/16 regulation of Myb controls the NK cell maturation program.


European Journal of Immunology | 2014

microRNA management of NK-cell developmental and functional programs

Jeffrey W. Leong; Ryan P. Sullivan; Todd A. Fehniger

NK cells are innate lymphoid cells that are critical for host defense against infection, and mediate anti‐tumor responses. MicroRNAs (miRNAs) are a large family of small noncoding RNAs that target the 3′ untranslated region (UTR) of mRNAs, thereby attenuating protein translation. The expression of miRNAs within human peripheral blood and mouse splenic NK cells has been cataloged, with the majority of the miRNA sequence pool represented in the top 60 most abundantly expressed miRNAs. Global miRNA deficiency within NK cells has confirmed their critical role in NK‐cell biology, including defects in NK‐cell development and altered functionality. Studies using gain‐ and loss‐of‐function of individual miRNAs in NK cells have demonstrated the role of specific miRNAs in regulating NK‐cell development, maturation, and activation. miRNAs also regulate fundamental NK‐cell processes including cytokine production, cytotoxicity, and proliferation. This review provides an update on the intrinsic miRNA regulation of NK cells, including miRNA expression profiles, as well as their impact on NK‐cell biology. Additional profiling is needed to better understand miRNA expression within NK‐cell developmental intermediates, subsets, tissues, and in the setting of disease. Furthermore, key open questions in the field as well as technical challenges in the study of miRNAs in NK cells are highlighted.

Collaboration


Dive into the Jeffrey W. Leong's collaboration.

Top Co-Authors

Avatar

Todd A. Fehniger

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Ryan P. Sullivan

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Stephanie Schneider

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Rizwan Romee

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Brea A. Jewell

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Catherine R. Keppel

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Timothy Schappe

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Julia A. Wagner

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Maximillian Rosario

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Melissa M. Berrien-Elliott

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge