Network


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

Hotspot


Dive into the research topics where Karla A. Henning is active.

Publication


Featured researches published by Karla A. Henning.


Cancer immunology research | 2013

Anti-CTLA-4 Antibodies of IgG2a Isotype Enhance Antitumor Activity through Reduction of Intratumoral Regulatory T Cells

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.


Clinical Cancer Research | 2013

BMS-936564/MDX-1338: A Fully Human Anti-CXCR4 Antibody Induces Apoptosis In Vitro and Shows Antitumor Activity In Vivo in Hematologic Malignancies

Michelle Kuhne; Tanya Mulvey; Blake Belanger; Sharline Chen; Chin Pan; Colin Chong; Fei Cao; Wafa Niekro; Tom Kempe; Karla A. Henning; Lewis J. Cohen; Alan J. Korman; Pina M. Cardarelli

Purpose: CXCR4 has been identified as a prognostic marker for acute myeloid leukemia (AML) and other malignancies. We describe the development and characterization of a fully human antibody to CXCR4 and its application for therapy of AML, non–Hodgkin lymphoma (NHL), chronic lymphoid leukemia (CLL), and multiple myeloma. Experimental Design: Human transgenic mice were immunized with CXCR4-expressing cells, and antibodies reactive with CXCR4 were analyzed for apoptosis induction and ability to interfere with CXCL12-induced migration and calcium flux. In vivo efficacy was determined in multiple AML, NHL, and multiple myeloma xenograft tumors in severe combined immunodeficient mice. Results: BMS-936564/MDX-1338 is a fully human IgG4 monoclonal antibody that specifically recognizes human CXCR4. In vitro studies show that MDX-1338 binds to CXCR4-expressing cells with low nanomolar affinity, blocks CXCL12 binding to CXCR4-expressing cells, and inhibits CXCL12-induced migration and calcium flux with low nanomolar EC50 values. When given as monotherapy, MDX-1338 exhibits antitumor activity in established tumors including AML, NHL, and multiple myeloma xenograft models. In addition, we show that MDX-1338 induced apoptosis on a panel of cell lines and propose that antibody-induced apoptosis is one of the mechanisms of tumor growth inhibition. Conclusions: BMS-936564/MDX-1338 is a potent CXCR4 antagonist which is efficacious as monotherapy in tumor-bearing mice and is currently in phase I for the treatment of relapsed/refractory AML, NHL, CLL, and multiple myeloma. Clin Cancer Res; 19(2); 357–66. ©2012 AACR.


Blood Advances | 2017

PD-1 blockade enhances elotuzumab efficacy in mouse tumor models

Natalie Bezman; Amy Jhatakia; Alper Y. Kearney; Ty Brender; Mark Maurer; Karla A. Henning; Misty R. Jenkins; Amy J. Rogers; Paul Neeson; Alan J. Korman; Michael Robbins; Robert F. Graziano

Elotuzumab, a humanized monoclonal antibody that binds human signaling lymphocytic activation molecule F7 (hSLAMF7) on myeloma cells, was developed to treat patients with multiple myeloma (MM). Elotuzumab has a dual mechanism of action that includes the direct activation of natural killer (NK) cells and the induction of NK cell-mediated antibody-dependent cellular cytotoxicity. This study aimed to characterize the effects of elotuzumab on NK cells in vitro and in patients with MM and to determine whether elotuzumab antitumor activity was improved by programmed death receptor-1 (PD-1) blockade. Elotuzumab promoted NK cell activation when added to a coculture of human NK cells and SLAMF7-expressing myeloma cells. An increased frequency of activated NK cells was observed in bone marrow aspirates from elotuzumab-treated patients. In mouse tumor models expressing hSLAMF7, maximal antitumor efficacy of a murine immunoglobulin G2a version of elotuzumab (elotuzumab-g2a) required both Fcγ receptor-expressing NK cells and CD8+ T cells and was significantly enhanced by coadministration of anti-PD-1 antibody. In these mouse models, elotuzumab-g2a and anti-PD-1 combination treatment promoted tumor-infiltrating NK and CD8+ T-cell activation, as well as increased intratumoral cytokine and chemokine release. These observations support the rationale for clinical investigation of elotuzumab/anti-PD-1 combination therapy in patients with MM.


Archive | 2015

Antibodies against glucocorticoid-induced tumor necrosis factor receptor (gitr) and uses thereof

Changyu Wang; Nils Lonberg; Alan J. Korman; Mark J. Selby; Mohan Srinivasan; Karla A. Henning; Michelle Minhua Han; Guodong Chen; Richard Y.-C. Huang; Indrani Chakraborty; Haichun Huang; Susan Wong; Huiming Li


OncoImmunology | 2017

The anti-SLAMF7 antibody elotuzumab mediates NK cell activation through both CD16-dependent and –independent mechanisms

Tatiana Pazina; Ashley M. James; Alexander W. MacFarlane; Natalie Bezman; Karla A. Henning; Christine Bee; Robert F. Graziano; Michael Robbins; Adam D. Cohen; Kerry S. Campbell


Archive | 2015

Antibodies against cd73 and uses thereof

Nils Lonberg; Alan J. Korman; Bryan C. Barnhart; Aaron P. Yamniuk; Mohan Srinivasan; Karla A. Henning; Ming Lei; Emanuela Sega; Angela Goodenough; Maria Jure-Kunkel; Guodong Chen; John S. Sack; Richard Y.-C. Huang; Martin J. Corbett; Joseph E. Myers; Liang Schweizer; Sandra V. Hatcher; Haichun Huang; Pingping Zhang


Blood | 2011

BMS-936564/MDX-1338: A Fully Human Anti-CXCR4 Antibody Induces Apoptosis In Vitro and Shows Anti Tumor Activity In Vivo

Michelle Kuhne; Tanya Mulvey; Sharline Chen; Chin Pan; Colin Chong; Wafa Niekro; Tom Kempe; Karla A. Henning; Lewis J. Cohen; Alan J. Korman; Pina M. Cardarelli


Archive | 2016

Antibodies comprising modified heavy constant regions

Nils Lonberg; Korman Alan J; Selby Mark J; Bryan C. Barnhart; Yamniuk, Aaron, P.; Mohan Srinivasan; Karla A. Henning; Han, Michelle, Minhua; Ming Lei; Liang Schweizer; Hatcher, Sandra, V.; Arvind Rajpal


Archive | 2016

ANTIBODIES AGAINST OX40 AND USES THEREOF

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


Archive | 2017

ANTICUERPOS CONTRA CD73 Y SUS USOS

Sandra V. Hatcher; Liang Schweizer; Joseph E. Myers; Martin J. Corbett; Richard Y.-C. Huang; John S. Sack; Guodong Chen; Maria Jure-Kunkel; Angela Goodenough; Emanuela Sega; Ming Lei; Karla A. Henning; Mohan Srinivasan; Aaron P. Yamniuk; Bryan C. Barnhart; Alan J. Korman; Nils Lonberg

Collaboration


Dive into the Karla A. Henning's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ming Lei

Bristol-Myers Squibb

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge