Michael Quigley
Bristol-Myers Squibb
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Michael Quigley.
Cancer immunology research | 2013
Mark J. Selby; John J. Engelhardt; Michael Quigley; Karla A. Henning; Timothy Chen; Mohan Srinivasan; Alan J. Korman
The therapeutic potential of CTLA-4 blockade is evident in the ability of anti-CTLA-4 antibody to induce regression of established tumors. In an elegant set of experiments using a panel of murine immunoglobulin in various isotypes, Selby and colleagues delineated the mechanism of action of CTLA-4 blockade. Anti-CTLA-4 promotes antitumor activity by a selective reduction of intratumoral T-regulatory cells along with concomitant activation of T-effector cells. Antitumor activity of CTLA-4 antibody blockade is thought to be mediated by interfering with the negative regulation of T-effector cell (Teff) function resulting from CTLA-4 engagement by B7-ligands. In addition, a role for CTLA-4 on regulatory T cells (Treg), wherein CTLA-4 loss or inhibition results in reduced Treg function, may also contribute to antitumor responses by anti-CTLA-4 treatment. We have examined the role of the immunoglobulin constant region on the antitumor activity of anti-CTLA-4 to analyze in greater detail the mechanism of action of anti-CTLA-4 antibodies. Anti-CTLA-4 antibody containing the murine immunoglobulin G (IgG)2a constant region exhibits enhanced antitumor activity in subcutaneous established MC38 and CT26 colon adenocarcinoma tumor models compared with anti-CTLA-4 containing the IgG2b constant region. Interestingly, anti-CTLA-4 antibodies containing mouse IgG1 or a mutated mouse IgG1-D265A, which eliminates binding to all Fcγ receptors (FcγR), do not show antitumor activity in these models. Assessment of Teff and Treg populations at the tumor and in the periphery showed that anti-CTLA-4-IgG2a mediated a rapid and dramatic reduction of Tregs at the tumor site, whereas treatment with each of the isotypes expanded Tregs in the periphery. Expansion of CD8+ Teffs is observed with both the IgG2a and IgG2b anti-CTLA-4 isotypes, resulting in a superior Teff to Treg ratio for the IgG2a isotype. These data suggest that anti-CTLA-4 promotes antitumor activity by a selective reduction of intratumoral Tregs along with concomitant activation of Teffs. Cancer Immunol Res; 1(1); 32–42. ©2013 AACR.
PLOS ONE | 2016
Mark J. Selby; John J. Engelhardt; Robert J. Johnston; Li-Sheng Lu; Minhua Han; Kent B. Thudium; Dapeng Yao; Michael Quigley; Jose Valle; Changyu Wang; Bing Chen; Pina M. Cardarelli; Diann Blanset; Alan J. Korman
The monoclonal antibodies ipilimumab (anti-CTLA-4) and nivolumab (anti-PD-1) have shown remarkable antitumor activity in an increasing number of cancers. When combined, ipilimumab and nivolumab have demonstrated superior activity in patients with metastatic melanoma (CHECKMATE-067). Here we describe the preclinical development strategy that predicted these clinical results. Synergistic antitumor activity in mouse MC38 and CT26 colorectal tumor models was observed with concurrent, but not sequential CTLA-4 and PD-1 blockade. Significant antitumor activity was maintained using a fixed dose of anti-CTLA-4 antibody with decreasing doses of anti-PD-1 antibody in the MC38 model. Immunohistochemical and flow cytometric analyses confirmed that CD3+ T cells accumulated at the tumor margin and infiltrated the tumor mass in response to the combination therapy, resulting in favorable effector and regulatory T-cell ratios, increased pro-inflammatory cytokine secretion, and activation of tumor-specific T cells. Similarly, in vitro studies with combined ipilimumab and nivolumab showed enhanced cytokine secretion in superantigen stimulation of human peripheral blood lymphocytes and in mixed lymphocyte response assays. In a cynomolgus macaque toxicology study, dose-dependent immune-related gastrointestinal inflammation was observed with the combination therapy; this response had not been observed in previous single agent cynomolgus studies. Together, these in vitro assays and in vivo models comprise a preclinical strategy for the identification and development of highly effective antitumor combination immunotherapies.
Immunology and Cell Biology | 2017
Bryan C Barnhart; Michael Quigley
The use of antibody therapy for cancer has steadily increased in recent years and has become standard treatment for numerous tumor types. It is now appreciated that the clinical activity of these antibodies relies upon their specific interactions with Fc receptors in addition to the well‐studied target‐binding region. The interactions mediated by antibody Fc domains can strongly affect the functional outcome of antibody therapy. The Fc portion of an antibody defines its interaction with numerous immune cells and has become an intense area of research as selecting the optimal Fc can greatly enhance the activity as well as mechanism of action of therapeutic antibodies. Recent advances in antibody engineering have enabled the development of antibodies that have altered Fc receptor interactions to take advantage of these findings. Engineering the Fc can fulfill diverse functions such as enhancing effector function for killing of tumor cells or depletion of unwanted immune subsets, enhancing agonist receptor signaling on particular immune cells or eliminating interaction with Fc receptors to avoid cellular depletion or toxicity in normal tissues. This review highlights important data and studies examining the role of Fc–Fc receptor interactions in therapeutic antibodies with a considerations for the future of engineered antibody therapy.
Cancer immunology research | 2017
Karla R. Wiehagen; Natasha M. Girgis; Douglas H. Yamada; Andressa Smith; Szeman Ruby Chan; Iqbal S. Grewal; Michael Quigley; Raluca Verona
This study demonstrates the advantage of combining agonist anti-CD40 with CSF-1R blockade for maximal antitumor benefit and survival in a preclinical mouse model. The results provide rationale for the combination of two distinct immunotherapies to improve clinical outcome. Efficacious antitumor immune responses must overcome multiple suppressive mechanisms in the tumor microenvironment to control cancer progression. In this study, we demonstrate that dual targeting of suppressive myeloid populations by inhibiting CSF-1/CSF-1R signaling and activation of antigen-presenting cells with agonist anti-CD40 treatment confers superior antitumor efficacy and increased survival compared with monotherapy treatment in preclinical tumor models. Concurrent CSF-1R blockade and CD40 agonism lead to profound changes in the composition of immune infiltrates, causing an overall decrease in immunosuppressive cells and a shift toward a more inflammatory milieu. Anti-CD40/anti–CSF-1R–treated tumors contain decreased tumor-associated macrophages and Foxp3+ regulatory T cells. This combination approach increases maturation and differentiation of proinflammatory macrophages and dendritic cells and also drives potent priming of effector T cells in draining lymph nodes. As a result, tumor-infiltrating effector T cells exhibit improved responses to tumor antigen rechallenge. These studies show that combining therapeutic approaches may simultaneously remove inhibitory immune populations and sustain endogenous antitumor immune responses to successfully impair cancer progression. Cancer Immunol Res; 5(12); 1109–21. ©2017 AACR.
Archive | 2013
John J. Engelhardt; Alan J. Korman; Michael Quigley; Mark J. Selby; Changyu Wang
Archive | 2016
Zhehong Cai; Indrani Chakraborty; Marie-Michelle Navarro Garcia; Thomas D. Kempe; Alan J. Korman; Alexander Kozhich; Hadia Lemar; Mark Maurer; Christina Milburn; Michael Quigley; Maria Rodriguez; Xiang Shao; Mohan Srinivasan; Brenda L. Stevens; Kent B. Thudium; Susan Chien-Szu Wong; Jochem Gokemeijer; Xi-tao Wang; Han Chang; Christine Huang; Maria Jure-Kunkel; Zheng Yang; Yan Feng; Patrick Guirnalda; Nils Lonberg; Bryan C. Barnhart; Aaron P. Yamniuk; Karla A. Henning; Michelle Minhua Han; Ming Lei
Cancer Research | 2018
Rui Wang; Yan Feng; Ed Hilt; Xiling Yuan; Chan Gao; Xiao Shao; Yongliang Sun; Michael D'silva; Katherine S. Yang; Becky Penhallow; Rajesh Anand; Irene Pak; Danielle M. Greenawalt; Anke Klippel; Nataly Manjarrez-Orduño; Robert Neely; Michael Quigley; Michael Nathan Hedrick; Praveen Aanur; Z Cao
Cancer Research | 2018
Marie-Claude Gaudreau; Christina Milburn; Chan Gao; Alla Pritsker; Mark P. Fereshteh; Zheng Yang; Bryan C. Barnhart; Alan J. Korman; Michael Quigley
Archive | 2016
Praveen Aanur; Arvind Rajpal; Sandra V. Hatcher; Liang Schweizer; Ming Lei; Michelle Minhua Han; Karla A. Henning; Aaron P. Yamniuk; Bryan C. Barnhart; Mark Selby; Nils Lonberg; Patrick Guirnalda; Yan Feng; Maria Jure-Kunkel; Christine Huang; Han Chang; Xi Wang; Jochem Gokemeijer; Susan Wong; Kent B. Thudium; Brenda L. Stevens; Mohan Srinivasan; Xiang Shao; Mark Maurer; Hadia Lemar; Alexander Kozhich; Alan J. Korman; Thomas D. Kempe; Marie Navarro Garcia; Indrani Chakraborty
Archive | 2016
Zhehong Cai; Indrani Chakraborty; Marie-Michelle Navarro Garcia; Thomas D. Kempe; Alan J. Korman; Alexander Kozhich; Hadia Lemar; Mark Maurer; Christina Milburn; Michael Quigley; Maria Rodriguez; Xiang Shao; Mohan Srinivasan; Brenda L. Stevens; Kent B. Thudium; Susan Chien-Szu Wong; Jochem Gokemeijer; Xi-tao Wang; Han Chang; Christine Huang; Maria Jure-Kunkel; Zheng Yang; Yan Feng; Patrick Guirnalda; Nils Lonberg; Bryan C. Barnhart; Aaron P. Yamniuk; Karla A. Henning; Michelle Minhua Han; Ming Lei