Radiotherapy increases the permissiveness of established mammary tumors to rejection by immunomodulatory antibodies

I Verbrugge, J Hagekyriakou, LL Sharp, M Galli, A West… - Cancer research, 2012 - AACR
I Verbrugge, J Hagekyriakou, LL Sharp, M Galli, A West, NM McLaughlin, H Duret, H Yagita…
Cancer research, 2012AACR
It is becoming increasingly evident that radiotherapy may benefit from coincident or
subsequent immunotherapy. In this study, we examined whether the antitumor effects of
radiotherapy, in established triple-negative breast tumors could be enhanced with
combinations of clinically relevant monoclonal antibodies (mAb), designed to stimulate
immunity [anti-(α)-CD137, α-CD40] or relieve immunosuppression [α-programmed death
(PD)-1]. While the concomitant targeting of the costimulatory molecules CD137 and CD40 …
Abstract
It is becoming increasingly evident that radiotherapy may benefit from coincident or subsequent immunotherapy. In this study, we examined whether the antitumor effects of radiotherapy, in established triple-negative breast tumors could be enhanced with combinations of clinically relevant monoclonal antibodies (mAb), designed to stimulate immunity [anti-(α)-CD137, α-CD40] or relieve immunosuppression [α-programmed death (PD)-1]. While the concomitant targeting of the costimulatory molecules CD137 and CD40 enhanced the antitumor effects of radiotherapy and promoted the rejection of subcutaneous BALB/c-derived 4T1.2 tumors, this novel combination was noncurative in mice bearing established C57BL/6-derived AT-3 tumors. We identified PD-1 signaling within the AT-3 tumors as a critical limiting factor to the therapeutic efficacy of α-CD137 therapy, alone and in combination with radiotherapy. Strikingly, all mice bearing established orthotopic AT-3 mammary tumors were cured when α-CD137 and α-PD-1 mAbs were combined with single- or low-dose fractionated radiotherapy. CD8+ T cells were essential for curative responses to this combinatorial regime. Interestingly, CD137 expression on tumor-associated CD8+ T cells was largely restricted to a subset that highly expressed PD-1. These CD137+PD-1High CD8+ T cells, persisted in irradiated AT-3 tumors, expressed Tim-3, granzyme B and Ki67 and produced IFN-γ ex vivo in response to phorbol 12-myristate 13-acetate (PMA) and ionomycin stimulation. Notably, radiotherapy did not deplete, but enriched tumors of functionally active, tumor-specific effector cells. Collectively, these data show that concomitant targeting of immunostimulatory and inhibitory checkpoints with immunomodulatory mAbs can enhance the curative capacity of radiotherapy in established breast malignancy. Cancer Res; 72(13); 3163–74. ©2012 AACR.
AACR