Abstract

IntroductionResponse to trastuzumab in metastatic breast cancer correlates with expression of the high binding variant (158V) of the activating Fcγ receptor IIIA (CD16A). We engineered MGAH22, a chimeric anti-HER2 monoclonal antibody with specificity and affinity similar to trastuzumab, with an Fc domain engineered for increased binding to both alleles of human CD16A.MethodsMGAH22 was compared to an identical anti-HER2 mAb except for a wild type Fc domain. Antibody-dependent cell cytotoxicity (ADCC) assays were performed with HER2-expressing cancer cells as targets and human PBMC or purified NK cells as effectors. Xenograft studies were conducted in mice with wild type murine FcγRs; in mice lacking murine CD16; or in mice lacking murine CD16 but transgenic for human CD16A-158F, the low-binding variant. The latter model reproduces the differential binding between wild type and the Fc-optimized mAb for human CD16A. The JIMT-1 human breast tumor line, derived from a patient that progressed on trastuzumab therapy, was used in these studies. Single and repeat dose toxicology studies with MGAH22 administered intravenously at high dose were conducted in cynomolgus monkeys.ResultsThe optimized Fc domain confers enhanced ADCC against all HER2-positive tumor cells tested, including cells resistant to trastuzumab's anti-proliferative activity or expressing low HER2 levels. The greatest improvement occurs with effector cells isolated from donors homozygous or heterozygous for CD16A-158F, the low-binding allele. MGAH22 demonstrates increased activity against HER2-expressing tumors in mice transgenic for human CD16A-158F. In single and repeat-dose toxicology studies in cynomolgus monkeys, a species with a HER2 expression pattern comparable to that in humans and Fcγ receptors that exhibit enhanced binding to the optimized Fc domain, MGAH22 was well tolerated at all doses tested (15-150 mg/kg) and exhibited pharmacokinetic parameters similar to that of other anti-HER2 antibodies. Induction of cytokine release by MGAH22 in vivo or in vitro was similar to that induced by the corresponding wild type mAb or trastuzumab.ConclusionsThe data support the clinical development of MGAH22, which may have utility in patients with low HER2 expressing tumors or carrying the CD16A low-binding allele.

Highlights

  • Response to trastuzumab in metastatic breast cancer correlates with expression of the high binding variant (158V) of the activating Fcg receptor IIIA (CD16A)

  • While the relationship between CD16A polymorphism and benefit is controversial for cetuximab [12,13,14,15], CD16-158V and CD32A-131H homozygosity appear to be associated with beneficial responses for rituximab and infliximab [16,17,18]

  • MGAH22 preserves HER2-binding and anti-proliferative properties of trastuzumab MGAH22 is a human/mouse chimeric immunoglobulin G 1 (IgG1) anti-HER2 antibody based on mouse clone 4D5, the precursor of trastuzumab [23] with an engineered Fc domain (MGFc0264) similar to the previously described Fc variant 18 [25], except that the V305I mutation was replaced by L235V to reduce CD32B binding

Read more

Summary

Introduction

Response to trastuzumab in metastatic breast cancer correlates with expression of the high binding variant (158V) of the activating Fcg receptor IIIA (CD16A). Trastuzumab acts against HER2-positive tumors by multiple mechanisms, including receptor internalization, receptor ‘shedding’, direct anti-proliferative activity, antibody-dependent cell-mediated cytotoxicity (ADCC), and presentation of antigenic determinants of opsonized cells to antigen-presenting cells [6] The latter mechanisms depend upon the interaction of the Fc domain of trastuzumab with Fc-gamma receptors (FcgRs) expressed by immune effector populations, such as natural killer (NK) cells or mononuclear phagocytes [7,8,9,10]. FcgR genotypes most frequently associated with greater beneficial responses occur in a minority of the population This provides a strong rationale for engineering the Fc domain of trastuzumab to better interact with low-binding alleles of activating FcgRs to expand, without regard to FcgR genotype, the benefit of treatment to patients

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.