Abstract

Simple SummaryAMHRII, the anti-Müllerian hormone receptor, is selectively expressed in normal sexual organs in healthy adults but is also re-expressed in ovarian, colorectal and lung cancers. In this context, we developed murlentamab, a humanized glyco-engineered anti-AMHRII monoclonal antibody, currently in clinical trial. Preliminary data suggest that murlentamab anti-tumor activity involves immune response activation. Thus, in vitro experiments were performed to precisely characterize the murlentamab effect on the human immune system. We show that murlentamab treatment is associated with evidences of innate and adaptive immune cell activation in cancer patient samples. Moreover, we demonstrate that the murlentamab opsonization of AMHRII-expressing ovarian tumor cells promotes a polarization switch of both naïve and tumor-associated macrophages towards an anti-tumor M1-like phenotype. Our work also supports that, through macrophage reeducation, murlentamab activates an anti-tumor adaptive immune response. Finally, the combination of murlentamab with pembrolizumab confirmed novel clinical perspectives of murlentamab association with checkpoint inhibitors and other immuno-modulators.AMHRII, the anti-Müllerian hormone receptor, is selectively expressed in normal sexual organs but is also re-expressed in gynecologic cancers. Hence, we developed murlentamab, a humanized glyco-engineered anti-AMHRII monoclonal antibody currently in clinical trial. Low-fucosylated antibodies are known to increase the antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) potency of effector cells, but some preliminary results suggest a more global murlentamab-dependent activation of the immune system. In this context, we demonstrate here that the murlentamab opsonization of AMHRII-expressing ovarian tumor cells, in the presence of unstimulated- or tumor-associated macrophage (TAM)-like macrophages, significantly promotes macrophage-mediated ADCC and shifts the whole microenvironment towards a pro-inflammatory and anti-tumoral status, thus triggering anti-tumor activity. We also report that murlentamab orients both unstimulated- and TAM-like macrophages to an M1-like phenotype characterized by a strong expression of co-stimulation markers, pro-inflammatory cytokines and chemokines, favoring T cell recruitment and activation. Moreover, we show that murlentamab treatment shifts CD4+ Th1/Th2 balance towards a Th1 response and activates CD8+ T cells. Altogether, these results suggest that murlentamab, through naïve macrophage orientation and TAM reprogrammation, stimulates the anti-tumor adaptive immune response. Those mechanisms might contribute to the sustained clinical benefit observed in advanced cancer patients treated with murlentamab. Finally, the enhanced murlentamab activity in combination with pembrolizumab opens new therapeutic perspectives.

Highlights

  • The Anti-Müllerian hormone type II receptor (AMHRII), known as MIS type II receptor (MISRII or MISIIR), is a member of the transforming growth factor beta (TGF-β) receptor superfamily [1,2]

  • Phase I (C101) phase IIa (C201) studies were performed with murlentamab to determine recommended doses and treatment responses, characterized by overall survival (OS) and progression-free survival (PFS) (Figure S1)

  • We explored whether the murlentamab opsonization of ovarian tumor cells could trigger human Monocyte-Derived Macrophages (MDMs)-dependent antibody-dependent cell-mediated cytotoxicity (ADCC)

Read more

Summary

Introduction

The Anti-Müllerian hormone type II receptor (AMHRII), known as MIS type II receptor (MISRII or MISIIR), is a member of the transforming growth factor beta (TGF-β) receptor superfamily [1,2]. Several studies have confirmed the expression of AMHRII in gynecological cancer tissues [6,7,8,9,10] and, more recently, AMHRII was found expressed by certain non-gynecological cancer such as non-small cell lung cancer and colorectal cancer [11,12]. This set of consistent evidence led to the development of murlentamab, named GM102 or 3C23K, a humanized glyco-engineered anti-AMHRII monoclonal antibody. Following an extensive pharmacological profiling as well as toxicological studies in cynomolgus monkeys [12,13,14], a phase I study of murlentamab in gynecological cancers (www.clinicaltrial.gov/NCT02978755, accessed on: 20 June 2016) and a phase IIa in colorectal cancer (www.clinicaltrial.gov/NCT03799731, accessed on: 11 July 2018) are currently ongoing

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

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