Abstract

The immune checkpoint programmed cell death protein 1 (PD-1) plays a major role in T cell exhaustion in cancer and chronic HIV infection. The inhibitor of apoptosis protein antagonist Debio 1143 (D1143) enhances tumor cell death and synergizes with anti-PD-1 agents to promote tumor immunity and displayed HIV latency reversal activity in vitro. We asked in this study whether D1143 would stimulate the potency of an anti-human PD-1 monoclonal antibody (mAb) to reduce HIV loads in humanized mice. Anti-PD-1 mAb treatment decreased PD-1+ CD8+ cell population by 32.3% after interruption of four weeks treatment, and D1143 co-treatment further reduced it from 32.3 to 73%. Anti-PD-1 mAb administration reduced HIV load in blood by 94%, and addition of D1143 further enhanced this reduction from 94 to 97%. D1143 also more profoundly promoted with the anti-PD-1-mediated reduction of HIV loads in all tissues analyzed including spleen (71 to 96.4%), lymph nodes (64.3 to 80%), liver (64.2 to 94.4), lung (64.3 to 80.1%) and thymic organoid (78.2 to 98.2%), achieving a >5 log reduction of HIV loads in CD4+ cells isolated from tissues 2 weeks after drug treatment interruption. Ex vivo anti-CD3/CD28 stimulation increased the ability to activate exhausted CD8+ T cells in infected mice having received in vivo anti-PD-1 treatment by 7.9-fold (5 to 39.6%), and an additional increase by 1.7-fold upon D1143 co-treatment (39.6 to 67.3%). These findings demonstrate for the first time that an inhibitor of apoptosis protein antagonist enhances in a statistically manner the effects of an immune check point inhibitor on antiviral immunity and on HIV load reduction in tissues of humanized mice, suggesting that the combination of two distinct classes of immunomodulatory agents constitutes a promising anti-HIV immunotherapeutic approach.

Highlights

  • WHO and UNAIDS estimated that 40 million people live with HIV

  • We recently reported that the inhibitor of apoptosis protein antagonist (IAPa) Debio 1143 (D1143) modulates the non-canonical NF-kB pathway by rapidly degrading a repressor of this important signaling pathway—the baculoviral inhibitor of apoptosis proteins (IAP) repeat-containing 2 (BIRC2) [36]

  • D1143 was obtained from Debiopharm International S.A., anti-human PD-1 monoclonal antibody (mAb) used for BLT mouse treatment was obtained from Bio X Cell (Clone J116), anti-human PD-1 mAb (Clone EH12-1540-29C9) used for cell surface staining was obtained from Synagis [50], and anti-human CD8 (Clone RPA T8), CD3 (Clone UCHT1), CD4, CD45 (Clone HI30) and IFNγ (Clone 4S.B3) antibodies used for cell staining were obtained from BioLegend

Read more

Summary

Introduction

The Centers for Disease Control and Prevention estimated that 38,500 people were newly infected with HIV in the United States in 2015, and 2.1 million worldwide [1]. T cells have a critical function in constraining viremia during acute and chronic HIV infection. CD8+ T cells are responsible for the rapid decrease of viremia during acute HIV infection [2,3,4]. CD8+ T cells inhibit HIV replication in vitro [5], and CD8+ T cell depletion in SIV-infected primates resulted in a loss of viremia control during infection [6]. AIDS progression during sustained chronic infection often leads to impairment and exhaustion of effector and memory CD8+ T cells, resulting in a boost of viremia [13]. Dysfunctional CD8+ T cells were found in humans during chronic HIV, hepatitis B virus (HBV), hepatitis C virus (HCV) and human T lymphotropic virus (HTLV) infections as well as in primates during chronic SIV infection [14]

Methods
Results
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