Abstract

Extracorporeal photochemotherapy (ECP) is employed for the management of cutaneous T cell lymphoma (CTCL). ECP involves the extracorporeal exposure of white blood cells (WBCs) to a photosensitizer, 8-methoxypsoralen (8-MOP), in the context of ultraviolet A (UVA) radiation, followed by WBC reinfusion. Historically, the therapeutic activity of ECP has been attributed to selective cytotoxicity on circulating CTCL cells. However, only a fraction of WBCs is exposed to ECP, and 8-MOP is inactive in the absence of UVA light, implying that other mechanisms underlie the anticancer effects of ECP. Recently, ECP has been shown to enable the physiological differentiation of monocytes into dendritic cells (DCs) that efficiently cross-present tumor-associated antigens (TAAs) to CD8+ T lymphocytes to initiate cognate immunity. However, the source of TAAs and immunostimulatory signals for such DCs remains to be elucidated. Here, we demonstrate that 8-MOP plus UVA light reduces melanoma cell viability along with the emission of ICD-associated danger signals including calreticulin (CALR) exposure on the cell surface and secretion of ATP, high mobility group box 1 (HMGB1) and type I interferon (IFN). Consistently, melanoma cells succumbing to 8-MOP plus UVA irradiation are efficiently engulfed by monocytes, ultimately leading to cross-priming of CD8+ T cells against cancer. Moreover, malignant cells killed by 8-MOP plus UVA irradiation in vitro vaccinate syngeneic immunocompetent mice against living cancer cells of the same type, and such a protection is lost when cancer cells are depleted of calreticulin or HMGB1, as well as in the presence of an ATP-degrading enzyme or antibodies blocking type I IFN receptors. ECP induces bona fide ICD, hence simultaneously providing monocytes with abundant amounts of TAAs and immunostimulatory signals that are sufficient to initiate cognate anticancer immunity.

Highlights

  • The term extracorporeal photochemotherapy (ECP)refers to a therapeutic procedure in which cutaneous T cell lymphoma (CTCL) patients are subjected to leukapheresis followed by: (1) extracorporeal exposure or white blood cells (WBCs) to 8-methoxypsoralen (8-MOP) in the context of ultraviolet A (UVA) irradiation, and (2) WBC reinfusion[1]

  • We employed a fluorometric assay for cell number on ovalbumin (OVA)expressing mouse melanoma B16 (B16-OVA) cells exposed to 8-MOP plus UVA light at different therapeutically relevant doses (1J – 4J) for 48 h, as compared to (1) B16 cells receiving 4J UVA light in the absence of 8MOP; (2) B16 cells responding to the chemotherapeutic agent cisplatin (CDDP), which is known to mediate a non-immunogenic variant of regulated cell death (RCD);[26] and (3) B16 cells subjected to freeze–thawing cycles, which cause a rapid necrotic response with tolerogenic, rather than immunogenic, consequences[27,28]

  • We set to interrogate biochemical biomarkers of apoptotic cell death in B16-OVA cells exposed to 8-MOP plus 4J UVA light in vitro over time

Read more

Summary

Introduction

Refers to a therapeutic procedure in which cutaneous T cell lymphoma (CTCL) patients are subjected to leukapheresis followed by: (1) extracorporeal exposure or white blood cells (WBCs) to 8-methoxypsoralen (8-MOP) in the context of ultraviolet A (UVA) irradiation, and (2) WBC reinfusion[1]. 8-MOP can be finely titrated to cause sufficient amounts of photoadducts to overwhelm the DNA repair machinery, leading to a rather slow wave of regulated cell death (RCD) that develops over the 3–4 days after ECP2,4,5. Not all cell types exhibit comparable sensitivity to 8-MOP plus UVA light[6]. Circulating lymphocytes appear to be considerably more sensitive than monocytes to RCD driven by 8-MOP plus UVA irradiation[7].

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