Network


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

Hotspot


Dive into the research topics where Amy M. Beebe is active.

Publication


Featured researches published by Amy M. Beebe.


Cytokine & Growth Factor Reviews | 2002

The role of interleukin-10 in autoimmune disease: systemic lupus erythematosus (SLE) and multiple sclerosis (MS).

Amy M. Beebe; Daniel J. Cua; Rene de Waal Malefyt

Interleukin-10 (IL-10) is an immunoregulatory cytokine that plays a crucial role in inflammatory and immune reactions. It has potent anti-inflammatory and immunosuppressive activities on myeloid cell functions which forms a solid basis for its use in acute and chronic inflammatory diseases. Here, we discuss the role of IL-10 in autoimmune diseases and examine its beneficial effects in cellular-based autoimmune diseases such as multiple sclerosis (MS) or its involvement in humoral-based autoimmune diseases such as systemic lupus erythematosus (SLE). Inhibition of the immune stimulatory activities of IL-10 may provide novel approaches in the treatment of humoral autoimmune diseases, infectious diseases and cancer.


Immunity | 1997

Serial backcross mapping of multiple loci associated with resistance to Leishmania major in mice.

Amy M. Beebe; Smita Mauze; Nicholas J. Schork; Robert L. Coffman

Resistance or susceptibility of inbred mouse strains to the parasite Leishmania major correlates with CD4+ T cell responses of the Th1 or Th2 subsets, respectively. To evaluate the genetic basis for this difference, resistant B10.D2 mice were backcrossed onto susceptible BALB/c mice for five generations with selection for resistance. Candidate resistance loci were identified by high frequency of heterozygosity in resistant N5 backcross mice. Loci on chromosomes 6, 7, 10, 11, 15, and 16 were associated with resistance, demonstrating the multigenic nature of this phenotype. The presence of all six loci was not necessary to confer resistance and no single locus was required. Rather, a variety of combinations of these loci may be capable of interacting to confer resistance.


Journal of Immunology | 2012

Blockade of IL-10 Signaling during Bacillus Calmette-Guérin Vaccination Enhances and Sustains Th1, Th17, and Innate Lymphoid IFN-γ and IL-17 Responses and Increases Protection to Mycobacterium tuberculosis Infection

Jonathan M. Pitt; Evangelos Stavropoulos; Paul S. Redford; Amy M. Beebe; Gregory J. Bancroft; Douglas B. Young; Anne O'Garra

Vaccination with Mycobacterium bovis bacillus Calmette-Guérin (BCG) remains the only prophylactic vaccine against tuberculosis, caused by Mycobacterium tuberculosis, but gives variable protection against pulmonary disease. The generation of host Th1 responses following BCG vaccination is accepted as the major mechanism of protection against M. tuberculosis infection. Early production of IL-17 in the lungs following M. tuberculosis challenge of mice previously vaccinated with M. tuberculosis peptides in adjuvant has been shown to be required for efficient Th1 cell recruitment. IL-10 regulates various processes involved in generation of Th1 and Th17 responses. Previous studies have shown IL-10 as a negative regulator of the immune response to primary M. tuberculosis infection, with Il10−/− mice having reduced lung bacterial loads. In this study we show that inhibition of IL-10 signaling during BCG vaccination enhances host-generated Ag-specific IFN-γ and IL-17A responses, and that this regimen gives significantly greater protection against aerogenic M. tuberculosis challenge in both susceptible and relatively resistant strains of mice. In M. tuberculosis-susceptible CBA/J mice, Ab blockade of IL-10R specifically during BCG vaccination resulted in additional protection against M. tuberculosis challenge of >1-log10 compared with equivalent isotype-treated controls. The protection observed following BCG vaccination concurrent with anti–IL-10R mAb treatment was sustained through chronic M. tuberculosis infection and correlated with enhanced lung Th1 and Th17 responses and increased IFN-γ and IL-17A production by γδ T cells and an innate-like Thy1.2+CD3− lymphoid population. We show that IL-10 inhibits optimal BCG-elicited protection, therefore suggesting that antagonists of IL-10 may be of great benefit as adjuvants in preventive vaccination against tuberculosis.


Cancer Research | 2017

Dual roles for regulatory T cell depletion and co-stimulatory signaling in agonistic GITR targeting for tumor immunotherapy

Ashley E. Mahne; Smita Mauze; Barbara Joyce-Shaikh; Jane Xia; Edward P. Bowman; Amy M. Beebe; Daniel J. Cua; Renu Jain

Agonistic monoclonal antibodies (mAb) targeting the T-cell receptor coregulatory molecule GITR exert potent therapeutic activities in preclinical tumor models. Although anti-GITR mAb are thought to act by depleting and destabilizing the intratumoral T regulatory cell (Treg) population, the precise mechanism of action is obscure. Here, we addressed this issue using a Treg fate-mapping approach, which revealed that Treg loss was primarily due to cell depletion, with minimal evidence of Treg conversion to a non-Foxp3-expressing population. Further characterization of persisting Tregs following anti-GITR mAb treatment showed that a highly activated subpopulation of CD44hiICOShi intratumoral Tregs were preferentially targeted for elimination, with the remaining Tregs exhibiting a less suppressive phenotype. With these changes in the Treg population, intratumoral CD8+ T cells acquired a more functional phenotype characterized by downregulation of the exhaustion markers PD-1 and LAG-3. This reversal of CD8+ T-cell exhaustion was dependent on both agonistic GITR signaling and Treg depletion, as neither mechanism by itself could fully rescue the exhaustion phenotype. Tests of anti-human GITR antibody MK-4166 in a humanized mouse model of cancer mimicked many of the effects of anti-mouse GITR mAb in syngeneic tumor models, decreasing both Treg numbers and immune suppressor phenotype while enhancing effector responsiveness. Overall, our results show how anti-GITR mAb shifts Treg populations to enable immune attack on tumors, with clinical implications for molecular markers to modify emerging treatments. Cancer Res; 77(5); 1108-18. ©2016 AACR.


Blood | 2014

Anaphylaxis caused by repetitive doses of a GITR agonist monoclonal antibody in mice

Judith T. Murphy; Andre P. Burey; Amy M. Beebe; Danling Gu; Leonard G. Presta; Taha Merghoub; Jedd D. Wolchok

Immunotherapy for cancer using antibodies to enhance T-cell function has been successful in recent clinical trials. Many molecules that improve activation and effector function of T cells have been investigated as potential new targets for immunomodulatory antibodies, including the tumor necrosis factor receptor superfamily members GITR and OX40. Antibodies engaging GITR or OX40 result in significant tumor protection in preclinical models. In this study, we observed that the GITR agonist antibody DTA-1 causes anaphylaxis in mice upon repeated intraperitoneal dosing. DTA-1-induced anaphylaxis requires GITR, CD4(+) T cells, B cells, and interleukin-4. Transfer of serum antibodies from DTA-1-treated mice, which contain high levels of DTA-1-specific immunoglobulin G1 (IgG1), can induce anaphylaxis in naive mice upon administration of an additional dose of DTA-1, suggesting that anaphylaxis results from anti-DTA-1 antibodies. Depletion of basophils and blockade of platelet-activating factor, the key components of the IgG1 pathway of anaphylaxis, rescues the mice from DTA-1-induced anaphylaxis. These results demonstrate a previously undescribed lethal side effect of repetitive doses of an agonist immunomodulatory antibody as well as insight into the mechanism of toxicity, which may offer a means of preventing adverse effects in future clinical trials using anti-GITR or other agonist antibodies as immunotherapies.


Cancer Research | 2017

Characterization of MK-4166, a clinical agonistic antibody that targets human GITR and inhibits the generation and suppressive effects of T regulatory cells

Selvakumar Sukumar; Douglas C. Wilson; Ying Yu; Jerelyn Wong; Saraswathi Naravula; Grigori Ermakov; Romina Riener; Bhagyashree Bhagwat; Antoaneta S. Necheva; Jeff Grein; Tatyana Churakova; Ruban Mangadu; Peter Georgiev; Denise Manfra; Elaine M. Pinheiro; Venkataraman Sriram; Wendy J. Bailey; Danuta J. Herzyk; Terrill K. McClanahan; Aarron Willingham; Amy M. Beebe; Svetlana Sadekova

GITR is a T-cell costimulatory receptor that enhances cellular and humoral immunity. The agonist anti-mouse GITR antibody DTA-1 has demonstrated efficacy in murine models of cancer primarily by attenuation of Treg-mediated immune suppression, but the translatability to human GITR biology has not been fully explored. Here, we report the potential utility of MK-4166, a humanized GITR mAb selected to bind to an epitope analogous to the DTA-1 epitope, which enhances the proliferation of both naïve and tumor-infiltrating T lymphocytes (TIL). We also investigated the role of GITR agonism in human antitumor immune responses and report here the preclinical characterization and toxicity assessment of MK-4166, which is currently being evaluated in a phase I clinical study. Expression of human GITR was comparable with that of mouse GITR in tumor-infiltrating Tregs despite being drastically lower in other human TILs and in many human peripheral blood populations. MK-4166 decreased induction and suppressive effects of Tregsin vitro In human TIL cultures, MK-4166 induced phosphorylation of NFκB and increased expression of dual specificity phosphatase 6 (DUSP6), indicating that MK-4166 activated downstream NFκB and Erk signaling pathways. Furthermore, MK-4166 downregulated FOXP3 mRNA in human tumor infiltrating Tregs, suggesting that, in addition to enhancing the activation of TILs, MK-4166 may attenuate the Treg-mediated suppressive tumor microenvironment. Cancer Res; 77(16); 4378-88. ©2017 AACR.


Advances in Experimental Medicine and Biology | 1998

GENETIC CONTROL OF THE T CELL RESPONSE TO LEISHMANIA MAJOR INFECTION

Robert L. Coffman; Amy M. Beebe

Cutaneous infection of most inbred mouse strains with the protozoan parasite, Leishmania major, leads to a localized lesion that is contained and resolved primarily by CD4+ Th1 cells (1,2). Th1 cells secrete the potent macrophage activator IFN-γ, thus stimulating effective killing of this parasite (1,3). A few inbred mouse strains, such as BALB/c and SWR, fail to control parasite replication. This failure results in progressive lesion development, spread to visceral organs and eventual death. Highly susceptible BALB/c mice have been shown to make a strong immune response, but one that is dominated by CD4+ Th2 cells (4), which inhibit macrophage activation by producing IL-4, IL-10 and IL-13 (5). This sharp delineation of Th1 and Th2 responses has made L. major infection a useful model for studying the control of functional differentiation in CD4+ T cells. The clear strain differences in the response to L. major also offer the opportunity to study the genetic factors that determine whether an animal can mount a protective or a pathogenic immune response to this infection.


Journal of Pharmacology and Experimental Therapeutics | 2016

The Role of Anti-Drug Antibodies in the Pharmacokinetics, Disposition, Target Engagement, and Efficacy of a GITR Agonist Monoclonal Antibody in Mice

Nicholas D. Brunn; Smita Mauze; Danling Gu; Derek Wiswell; Roanna Ueda; Douglas Hodges; Amy M. Beebe; Shuli Zhang; Enrique Escandón

Administration of biologics to enhance T-cell function is part of a rapidly growing field of cancer immunotherapy demonstrated by the unprecedented clinical success of several immunoregulatory receptor targeting antibodies. While these biologic agents confer significant anti-tumor activity through targeted immune response modulation, they can also elicit broad immune responses potentially including the production of anti-drug antibodies (ADAs). DTA-1, an agonist monoclonal antibody against GITR, is a highly effective anti-tumor treatment in preclinical models. We demonstrate that repeated dosing with murinized DTA-1 (mDTA-1) generates ADAs with corresponding reductions in drug exposure and engagement of GITR on circulating CD3+ CD4+ T cells, due to rapid hepatic drug uptake and catabolism. Mice implanted with tumors after induction of preexisting mDTA-1 ADA show no anti-tumor efficacy when given 3 mg/kg mDTA-1, an efficacious dose in naive mice. Nonetheless, increasing mDTA-1 treatment to 30 mg/kg in ADA-positive mice restores mDTA-1 exposure and GITR engagement on circulating CD3+ CD4+ T cells, thereby partially restoring anti-tumor efficacy. Formation of anti-mDTA-1 antibodies and changes in drug exposure and disposition does not occur in GITR−/− mice, consistent with a role for GITR agonism in humoral immunity. Finally, the administration of muDX400, a murinized monoclonal antibody against the checkpoint inhibitor PD-1, dosed alone or combined with mDTA-1 did not result in reduced muDX400 exposure, nor did it change the nature of the anti-mDTA-1 response. This indicates that anti-GITR immunogenicity may not necessarily impact the pharmacology of coadministered monoclonal antibodies, supporting combination immunomodulatory strategies.


Cancer Research | 2015

Abstract 4521: Evaluation of the relationship between serum exposure, receptor (GITR) availability and tumor suppression following administration of the anti-GITR antibody DX400 in mouse syngeneic tumor models

Ayse Meric Ovacik; Natalie Shinsky-Bjorde; Douglas Hodges; Svetlana Antonenko; Roanna Ueda; Smita Mauze; Danling Gu; Derek Wiswell; Shuli Zhang; Amy M. Beebe; Mohammad Tabrizi

GITR is a type I transmembrane protein of the tumor necrosis factor receptor superfamily which is expressed primarily on T lymphocytes and natural killer cells. Ligation of GITR on activated T cells provides a costimulatory signal that positively modulates antigen-specific T cell responses, leading to enhanced cellular and humoral immunity. The anti-GITR antibody DX400 is a murinized agonistic monoclonal antibody that targets mouse GITR, and has shown tumor growth inhibition in mouse syngeneic tumor models. In this study we examined the pharmacokinetic/pharmacodynamics (PK/PD) properties of DX400. Studies were conducted to examine the potential relationships between anti-GITR antibody serum exposure (PK) and receptor availability (PD) on the relevant T-cell subsets. The concentrations of DX400 in serum were determined using an electrochemiluminescence (ECL) assay, and the availability of receptor (GITR) was determined using flow cytometry. Non-linear PK properties for DX400 were observed over the dose range examined. In line with changes in serum concentration-time profiles for the antibody, a dose dependent effect in receptor availability was also observed. The relationships between receptor availability, serum concentrations of DX400 and tumor suppression were described by a mechanistic PK/PD model. This analysis provided an estimated potency (EC50) value of 4.2 ng/mL for blood receptor engagement on T-cell subsets. Citation Format: Ayse Meric Ovacik, Natalie Shinsky-Bjorde, Douglas Hodges, Svetlana Antonenko, Roanna Ueda, Smita Mauze, Danling Gu, Derek Wiswell, Shuli Zhang, Amy Beebe, Mohammad Tabrizi. Evaluation of the relationship between serum exposure, receptor (GITR) availability and tumor suppression following administration of the anti-GITR antibody DX400 in mouse syngeneic tumor models. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 4521. doi:10.1158/1538-7445.AM2015-4521


Cancer Cell | 2011

IL-10 Elicits IFNγ-Dependent Tumor Immune Surveillance

John B. Mumm; Jan Emmerich; Xueqing Zhang; Ivan S. F. Chan; Lingling Wu; Smita Mauze; Steven J. Blaisdell; Beth Basham; Jie Dai; Jeff Grein; Catherine Sheppard; Kyu Hong; Collette Cutler; Scott Turner; Drake LaFace; Melanie A. Kleinschek; Michael Judo; Gulesi Ayanoglu; John Langowski; Danling Gu; Brittany Paporello; Erin Murphy; Venkataraman Sriram; Saraswathi Naravula; Bela Desai; Satya Medicherla; Wolfgang Seghezzi; Terrill K. McClanahan; Susan Cannon-Carlson; Amy M. Beebe

Collaboration


Dive into the Amy M. Beebe's collaboration.

Researchain Logo
Decentralizing Knowledge