Network


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

Hotspot


Dive into the research topics where Michelle H. Nelson is active.

Publication


Featured researches published by Michelle H. Nelson.


Frontiers in Immunology | 2014

Th17 Cells in Cancer: The Ultimate Identity Crisis

Stefanie R. Bailey; Michelle H. Nelson; Richard A. Himes; Zihai Li; Shikhar Mehrotra; Chrystal M. Paulos

T helper 17 (Th17) cells play a complex and controversial role in tumor immunity and have been found to exhibit a fluctuating identity within the context of cancer. The recent, expanding literature on these cells attests to their puzzling nature, either promoting or suppressing tumor growth depending on the malignancy and course of therapeutic intervention investigated. This review addresses several newly appreciated factors that may help delineate Th17 cells’ immunological properties in the context of cancer. Several reports suggest that inflammatory signals induced in the tumor milieu regulate the functional fate and antitumor activity of Th17 cells. Recent findings also point to significant alterations in Th17 cells due to their interplay with regulatory T lymphocytes and cytotoxic CD8+ T cells within the tumor microenvironment. Finally, an appreciation for the stem cell-like properties of Th17 cells that augment their persistence and activity emerges from recent reports. The impact of these factors on Th17 cells’ antitumor efficacy and how these factors may be exploited to improve cancer therapies will be discussed.


Science immunology | 2017

Platelets subvert T cell immunity against cancer via GARP-TGFβ axis

Saleh Rachidi; Alessandra Metelli; Brian Riesenberg; Bill X. Wu; Michelle H. Nelson; Caroline Wallace; Chrystal M. Paulos; Mark P. Rubinstein; Elizabeth Garrett-Mayer; Mirko Hennig; Daniel W. Bearden; Yi Yang; Bei Liu; Zihai Li

T cell immunity against cancer in mice is enhanced with platelet-specific deletion of GARP and by platelet inhibitors. Cancer immunotherapy according to GARP Cancer, like microbes, can adapt to a single therapy, making combination therapies the approach of choice. Complementary therapies that decrease immunosuppression may boost the efficacy of immunotherapies. Now, Rachidi et al. report that targeting platelets improves adoptive T cell therapy of multiple cancers in mice. They found that transforming growth factor β (TGFβ) from platelets decrease T cell function largely through the expression of the TGFβ-docking receptor glycoprotein A repetitions predominant (GARP). These data suggest that combining immunotherapy with platelet inhibitors may be a complementary approach to cancer therapy. Cancer-associated thrombocytosis has long been linked to poor clinical outcome, but the underlying mechanism is enigmatic. We hypothesized that platelets promote malignancy and resistance to therapy by dampening host immunity. We show that genetic targeting of platelets enhances adoptive T cell therapy of cancer. An unbiased biochemical and structural biology approach established transforming growth factor β (TGFβ) and lactate as major platelet-derived soluble factors to obliterate CD4+ and CD8+ T cell functions. Moreover, we found that platelets are the dominant source of functional TGFβ systemically as well as in the tumor microenvironment through constitutive expression of the TGFβ-docking receptor glycoprotein A repetitions predominant (GARP) rather than secretion of TGFβ per se. Platelet-specific deletion of the GARP-encoding gene Lrrc32 blunted TGFβ activity at the tumor site and potentiated protective immunity against both melanoma and colon cancer. Last, this study shows that T cell therapy of cancer can be substantially improved by concurrent treatment with readily available antiplatelet agents. We conclude that platelets constrain T cell immunity through a GARP-TGFβ axis and suggest a combination of immunotherapy and platelet inhibitors as a therapeutic strategy against cancer.


Immunological Reviews | 2015

Novel immunotherapies for hematologic malignancies

Michelle H. Nelson; Chrystal M. Paulos

The immune system is designed to discriminate between self and tumor tissue. Through genetic recombination, there is fundamentally no limit to the number of tumor antigens that immune cells can recognize. Yet, tumors use a variety of immunosuppressive mechanisms to evade immunity. Insight into how the immune system interacts with tumors is expanding rapidly and has accelerated the translation of immunotherapies into medical breakthroughs. Herein, we appraise novel strategies that exploit the patients immune system to kill cancer. We review various forms of immune‐based therapies, which have shown significant promise in patients with hematologic malignancies, including (i) conventional monoclonal therapies like rituximab; (ii) engineered monoclonal antibodies called bispecific T‐cell engagers; (iii) monoclonal antibodies and pharmaceutical drugs that block inhibitory T‐cell pathways (i.e. PD‐1, CTLA‐4, and IDO); and (iv) adoptive cell transfer therapy with T cells engineered to express chimeric antigen receptors or T‐cell receptors. We also assess the idea of using these therapies in combination and conclude by suggesting multi‐prong approaches to improve treatment outcomes and curative responses in patients.


Journal of Immunology | 2015

The Inducible Costimulator Augments Tc17 Cell Responses to Self and Tumor Tissue

Michelle H. Nelson; Sreenath Kundimi; Jacob S. Bowers; Carolyn Rogers; Logan W. Huff; Kristina M. Schwartz; Krishnamurthy Thyagarajan; Elizabeth C. Little; Shikhar Mehrotra; David J. Cole; Mark P. Rubinstein; Chrystal M. Paulos

The inducible costimulator (ICOS) plays a key role in the development of Th17 cells, but its role in the development and antitumor activity of IL-17–producing CD8+ T cells (Tc17) remains unknown. We found that ICOS costimulation was important for the functional maintenance, but not differentiation, of Tc17 cells in vitro. Blocking the ICOS pathway using an antagonist mAb or by using recipient mice genetically deficient in the ICOS ligand reduced the antitumor activity of adoptively transferred Tc17 cells. Conversely, activating Tc17 cells with an ICOS agonist in vitro enhanced their capacity to eradicate melanoma and induce autoimmune vitiligo when infused into mice. However, ICOS stimulation did not augment the antitumor activity of IL-2 expanded T cells. Additional investigation revealed that ICOS stimulation not only increased IL-2Rα, CXCR3, and IL-23R expression on Tc17 cells, but also dampened their expression of suppressive molecule CD39. Although Tc17 cells activated with an ICOS agonist cosecreted heightened IL-17A, IL-9, and IFN-γ, their therapeutic effectiveness was critically dependent on IFN-γ production. Depletion of IL-17A and IL-9 had little impact on antitumor Tc17 cells activated with an ICOS agonist. Collectively, our work reveals that the ICOS pathway potentiates the antitumor activity of adoptively transferred Tc17 cells. This work has major implications for the design of vaccine, Ab and cell-based therapies for autoimmunity, infectious disease, and cancer.


Clinical Cancer Research | 2015

Dendritic Cells in Irradiated Mice Trigger the Functional Plasticity and Antitumor Activity of Adoptively Transferred Tc17 Cells via IL12 Signaling

Jacob S. Bowers; Michelle H. Nelson; Sreenath Kundimi; Stefanie R. Bailey; Logan W. Huff; Kristina M. Schwartz; David J. Cole; Mark P. Rubinstein; Chrystal M. Paulos

Purpose: The adoptive cell transfer (ACT) of CD8+ T cells is a promising treatment for advanced malignancies. Lymphodepletion before ACT enhances IFNγ+CD8+ T cell (Tc0)–mediated tumor regression. Yet, how lymphodepletion regulates the function and antitumor activity of IL17A+CD8+ T cells (Tc17) is unknown. Experimental Design: To address this question, pmel-1 CD8+ T cells were polarized to secrete either IL17A or IFNγ. These subsets were then infused into mice with B16F10 melanoma that were lymphoreplete [no total body irradiation (TBI)], or lymphodepleted with nonmyeloablative (5 Gy) or myeloablative (9 Gy with hematopoietic stem cell transplantation) TBI. The activation of innate immune cells and function of donor T-cell subsets were monitored in recipient mice. Results: Tc17 cells regress melanoma in myeloablated mice to a greater extent than in lymphoreplete or nonmyeloablated mice. TBI induced functional plasticity in Tc17 cells, causing conversion from IL17A to IFNγ producers. Additional investigation revealed that Tc17 plasticity and antitumor activity were mediated by IL12 secreted by irradiated host dendritic cells (DC). Neutralization of endogenous IL12 reduced the antitumor activity of Tc17 cells in myeloablated mice, whereas ex vivo priming with IL12 enhanced their capacity to regress melanoma in nonmyeloablated animals. This, coupled with exogenous administration of low-dose IL12, obviated the need for host preconditioning, creating curative responses in nonirradiated mice. Conclusions: Our findings indicate that TBI-induced IL12 augments Tc17 cell–mediated tumor immunity and underline the substantial implications of in vitro preparation of antitumor Tc17 cells with IL12 in the design of T-cell immunotherapies. Clin Cancer Res; 21(11); 2546–57. ©2015 AACR.


Clinical & Developmental Immunology | 2015

Harnessing the Microbiome to Enhance Cancer Immunotherapy

Michelle H. Nelson; Marshall A. Diven; Logan W. Huff; Chrystal M. Paulos

The microbiota plays a key role in regulating the innate and adaptive immune system. Herein, we review the immunological aspects of the microbiota in tumor immunity in mice and man, with a focus on toll-like receptor (TLR) agonists, vaccines, checkpoint modulators, chemotherapy, and adoptive T cell transfer (ACT) therapies. We propose innovative treatments that may safely harness the microbiota to enhance T cell-based therapies in cancer patients. Finally, we highlight recent developments in tumor immunotherapy, particularly novel ways to modulate the microbiome and memory T cell responses to human malignancies.


Nature Communications | 2017

Human CD26 high T cells elicit tumor immunity against multiple malignancies via enhanced migration and persistence

Stefanie R. Bailey; Michelle H. Nelson; Kinga Majchrzak; Jacob S. Bowers; Megan M. Wyatt; Aubrey S. Smith; Lillian R. Neal; Keisuke Shirai; Carmine Carpenito; Carl H. June; Michael J. Zilliox; Chrystal M. Paulos

CD8+ T lymphocytes mediate potent immune responses against tumor, but the role of human CD4+ T cell subsets in cancer immunotherapy remains ill-defined. Herein, we exhibit that CD26 identifies three T helper subsets with distinct immunological properties in both healthy individuals and cancer patients. Although CD26neg T cells possess a regulatory phenotype, CD26int T cells are mainly naive and CD26high T cells appear terminally differentiated and exhausted. Paradoxically, CD26high T cells persist in and regress multiple solid tumors following adoptive cell transfer. Further analysis revealed that CD26high cells have a rich chemokine receptor profile (including CCR2 and CCR5), profound cytotoxicity (Granzyme B and CD107A), resistance to apoptosis (c-KIT and Bcl2), and enhanced stemness (β-catenin and Lef1). These properties license CD26high T cells with a natural capacity to traffic to, regress and survive in solid tumors. Collectively, these findings identify CD4+ T cell subsets with properties critical for improving cancer immunotherapy.The role of human CD4+ T cell subsets in cancer immunotherapy is still unclear. Here, the authors show that CD26 identifies three CD4+ T cell subsets with distinct immunological properties in both healthy individuals and cancer patients.


JCI insight | 2017

Th17 cells are refractory to senescence and retain robust antitumor activity after long-term ex vivo expansion

Jacob S. Bowers; Michelle H. Nelson; Kinga Majchrzak; Stefanie R. Bailey; Baerbel Rohrer; Andrew Kaiser; Carl Atkinson; Luca Gattinoni; Chrystal M. Paulos

Adoptive immunotherapy for solid tumors relies on infusing large numbers of T cells to mediate successful antitumor responses in patients. While long-term rapid-expansion protocols (REPs) produce sufficient numbers of CD8+ T cells for treatment, they also cause decline in the cells therapeutic fitness. In contrast, we discovered that IL-17-producing CD4+ T cells (Th17 cells) do not require REPs to expand 5,000-fold over 3 weeks. Also, unlike Th1 cells, Th17 cells do not exhibit hallmarks of senescence or apoptosis, retaining robust antitumor efficacy in vivo. Three-week-expanded Th17 cells eliminated melanoma as effectively as Th17 cells expanded for 1 week when infused in equal numbers into mice. However, treating mice with large recalcitrant tumors required the infusion of all cells generated after 2 or 3 weeks of expansion, while the cell yield obtained after 1-week expansion was insufficient. Long-term-expanded Th17 cells also protected mice from tumor rechallenge including lung metastasis. Importantly, 2-week-expanded human chimeric antigen receptor-positive (CAR+) Th17 cells also retained their ability to regress human mesothelioma, while CAR+ Th1 cells did not. Our results indicate that tumor-reactive Th17 cells are an effective cell therapy for cancer, remaining uncompromised when expanded for a long duration owing to their resistance to senescence.


Cancer Immunology, Immunotherapy | 2016

Exploiting IL-17-producing CD4+ and CD8+ T cells to improve cancer immunotherapy in the clinic.

Majchrzak K; Michelle H. Nelson; Bailey; Jacob S. Bowers; Xue-Zhong Yu; Mark P. Rubinstein; Richard A. Himes; Chrystal M. Paulos

Cancer immunotherapy is one the most effective approaches for treating patients with tumors, as it bolsters the generation and persistence of memory T cells. In preclinical work, it has been reported that adoptively transferred CD4+ and CD8+ lymphocytes that secrete IL-17A (i.e., Th17 and Tc17 cells) regress tumors to a greater extent than IFN-γ+Th1 or Tc1 cells in vivo. Herein, we review the mechanisms underlying how infused Th17 and Tc17 cells regress established malignancies in clinically relevant mouse models of cancer. We also discuss how unique signaling cues—such as co-stimulatory molecules (ICOS and 41BB), cytokines (IL-12 and IL-23) or pharmaceutical reagents (Akt inhibitors, etc.)—can be exploited to bolster the therapeutic potential of IL-17+ lymphocytes with an emphasis on using this knowledge to improve next-generation clinical trials for patients with cancer.


JCI insight | 2017

β -catenin and PI3K δ inhibition expands precursor Th17 cells with heightened stemness and antitumor activity

Kinga Majchrzak; Michelle H. Nelson; Jacob S. Bowers; Stefanie R. Bailey; Megan M. Wyatt; John Wrangle; Mark P. Rubinstein; Juan C. Varela; Zihai Li; Richard A. Himes; Sherine S.L. Chan; Chrystal M. Paulos

ICOS costimulation generates Th17 cells with durable memory responses to tumor. Herein, we found that ICOS induces PI3K/p110δ/Akt and Wnt/β-catenin pathways in Th17 cells. Coinhibiting PI3Kδ and β-catenin altered the biological fate of Th17 cells. Th17 cells inhibited of both pathways expressed less RORγt, which, in turn, reduced their ability to secrete IL-17. Unexpectedly, these cells were more effective (than uninhibited cells) at regressing tumor when infused into mice, leading to long-term curative responses. PI3Kδ inhibition expanded precursor Th17 cells with a central memory phenotype that expressed nominal regulatory properties (low FoxP3), while β-catenin inhibition enhanced Th17 multifunctionality in vivo. Remarkably, upon TCR restimulation, RORγt and IL-17 rebounded in Th17 cells treated with PI3Kδ and β-catenin inhibitors. Moreover, these cells regained β-catenin, Tcf7, and Akt expression, licensing them to secrete heightened IL-2, persist, and eradicate solid tumors without help from endogenous NK and CD8 T cells. This finding shines a light on ways to repurpose FDA-approved drugs to augment T cell-based cancer immunotherapies.

Collaboration


Dive into the Michelle H. Nelson's collaboration.

Top Co-Authors

Avatar

Chrystal M. Paulos

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Stefanie R. Bailey

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Jacob S. Bowers

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Mark P. Rubinstein

University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Logan W. Huff

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Kinga Majchrzak

University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Sreenath Kundimi

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Zihai Li

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

David J. Cole

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Megan M. Wyatt

Medical University of South Carolina

View shared research outputs
Researchain Logo
Decentralizing Knowledge