Weekly Digests
‹ Back to January

Host expression of PD-L1 is essential for success of PD-L1 blockade

January 31, 2018

With the success of immune checkpoint blockade (ICB) therapies, the search is on for an understanding of the mechanisms and biomarkers to predict patient response. Lin et al. and Tang et al. tackled this problem head on by examining the role of PD-L1 expression on tumors versus host cells during ICB therapy, and separately came to the same surprising conclusion: host cell expression, and not tumor expression, of PD-L1 is essential for antitumor response during PD-L1/PD-1 blockade therapy.

To demonstrate that an intact host adaptive immune system is necessary for ICB efficacy, Lin et al. inoculated one of four cancer cell lines (MC38 colon cancer, ID8 ovarian cancer, B16-F10 melanoma, and LLC lung cancer) into mice with variable degrees of immunodeficiency: wild-type (WT), Rag-/- (no mature T, B cells), and NSG (no mature T, B, NK cells). Tang et al. performed similar experiments using Rag-/- mice with MC38 tumors. The researchers found that treatment of mice with anti-PD-L1 antibody reduced tumor growth and improved survival in WT mice with MC38, ID8, and B16-F10 tumors, and had no effect on LLC tumors. Meanwhile, PD-L1 blockade had no antitumor effect in NSG and Rag-/- mice in any tumor models, supporting the expected essential role of adaptive immunity for ICB efficacy.

Next, the researchers wanted to elucidate the role of host PD-L1 and PD-1 in tumor immunity. Lin et al. inoculated PD-L1-/- and PD-1-/- mice with MC38, ID8, or B16-F10 tumors and found that the therapeutic effect of anti-PD-L1 or anti-PD-1 antibody treatment was abrogated in both mouse models. Tang et al. also found that PD-1-/- mice with MC38 tumors had no response to anti-PD-L1. These results suggest that host PD-L1 and PD-1 expression may be necessary for response to PD-L1 or PD-1 blockade.

To isolate the effect of tumor-expressed PD-L1 from host PD-L1, both teams utilized CRISPR-Cas9 to knock out PD-L1 in ID8 and B16-F10 (Lin et al.), MC38 (Lin et al. and Tang et al.), and A20 B lymphoma (Tang et al.) tumor cells and inoculated them into mice. Surprisingly, the PD-L1-/- tumors responded to anti-PD-L1 therapy as well as WT tumors, reducing tumor growth and increasing survival, indicating that it is indeed the host’s, and not the tumor’s expression of PD-L1 that is essential for anti-PD-L1 treatment efficacy.

So, if anti-PD-L1 treatment is not exerting its effect by acting directly on the tumor, what is it actually doing? Lin et al. examined the T cells within the tumor microenvironment (TME) and in tumor-draining lymph nodes (dLNs) in anti-PD-L1-treated mice, and found that CD4+ and CD8+ T cells were activated not only in the tumor but also in the dLNs of WT mice, but that no activation occurred in either venue in PD-L1-/- or PD-1-/- mice. Tang et al. found similar effects in the dLNs and, using an inhibitor of lymphocyte trafficking out of dLNs, concluded that efficacy depended significantly on T cell extravasation from dLNs. These data imply that PD-L1 is involved not only in the T cell effector phase within the TME, but also, more importantly, in the T cell priming phase within dLNs. This suggests that blocking PD-L1 signaling in the lymph nodes could contribute to tumor control.

After all these experiments, the question remained: which host cells are the target of anti-PD-L1 therapy? Using whole-body PET/CT with radiolabeled anti-PD-L1 antibody injected into tumor-bearing mice, Tang et al. found that the antibody targeted the tumor tissue at similar levels in both WT and PD-L1-/- tumors. Furthermore, using bone marrow chimeras, they were able to directly implicate bone marrow-derived cells as a key host cell target. Both groups then analyzed PD-L1 expression and function in host immune cell subsets in tumor-bearing mice and found high levels of PD-L1 expression on macrophages, myeloid-derived suppressor cells (MDSCs), and dendritic cells (DCs) in the tumors and dLNs, eventually concluding that tumor-associated antigen-presenting cells (APCs) are PD-L1+, regulate T cell function, and are the main targets of anti-PD-L1 therapy.

To find out if these results are relevant in human cancers, Lin et al. cultured human T cells from peripheral blood with DCs, macrophages, and radiated ovarian cancer cells, and found that DCs and macrophages, but not tumor cells, efficiently activated the T cells, and this effect was further enhanced by anti-PD-L1 treatment. The researchers then used multi-color immunofluorescence on tissue sections from patients with locally advanced and metastatic melanoma who received PD-1 and CTLA-4 checkpoint blockade and found a positive correlation between the percentage of PD-L1+ DCs and macrophages within the tumor and complete clinical response. Lin et al. also observed a positive correlation between APC PD-L1 expression and general clinical response to PD-1 blockade in patients with ovarian cancer. Overall, the data suggests that PD-L1+ APCs may correlate with clinical efficacy of PD-1 blockade in melanoma and ovarian cancer patients.

Although neither research group excludes a role for tumor-expressed PD-L1 in anti-PD-L1 antitumor response, they both conclude that it is the host PD-L1+ APCs that play an essential part in mediating response to PD-L1 and PD-1 blockade, and that these cells may be the biomarker that predicts patient response to ICB therapy.

by Anna Scherer

References:

Lin H., Wei S., Hurt E.M., Green M.D., Zhao L., Vatan L., Szeliga W., Herbst R., Harms P.W., Fecher L.A., Vats P., Chinnaiyan A.M., Lao C.D., Lawrence T.S., Wicha M., Hamanishi J., Mandai M., Kryczek I., Zou W. Host expression of PD-L1 determines efficacy of PD-L1 pathway blockade-mediated tumor regression. J Clin Invest. 2018 Jan 16.

Tang H., Liang Y., Anders R.A., Taube J.M., Qiu X., Mulgaonkar A., Liu X., Harrington S.M., Guo J., Xin Y., Xiong Y., Nham K., Silvers W., Hao G., Sun X., Chen M., Hannan R., Qiao J., Dong H., Peng H., Fu Y.X. PD-L1 on host cells is essential for PD-L1 blockade-mediated tumor regression. J Clin Invest. 2018 Jan 16.

In the Spotlight...

Regulatory T cells trigger effector T cell DNA damage and senescence caused by metabolic competition

Liu et al. found that Treg cells outcompete responding T cells for glucose, triggering activation of ATM (a key regulator of the DNA damage response) in responding cells and resulting in T cell senescence. Senescence was found to be controlled by MAPK and STAT1/3 signaling and the cell cycle regulators p53 and p16. Senescent T cells are molecularly distinct from exhausted or anergic T cells and exhibit immunosuppressive activity. Inhibition of the ATM or STAT1/3 pathways reduced Treg-mediated T cell senescence in vivo, indicating a possible strategy for immunotherapy.

Early Assessment of Lung Cancer Immunotherapy Response via Circulating Tumor DNA

Goldberg et al. evaluated the peripheral blood of 28 patients with metastatic non-small cell lung cancer for circulating tumor DNA (ctDNA) as a possible indicator of efficacy in ongoing anti-PD-1 therapy. CtDNA response (a drop in ctDNA to >50% below baseline) agreed significantly with radiographic response, but was apparent sooner (24.5 vs 72.5 days). CtDNA response correlated with progression-free and overall survival and could be used to inform clinical decisions on whether to continue immunotherapy in individual patients.

Epithelial-mesenchymal transition leads to NK cell-mediated metastasis-specific immunosurveillance in lung cancer

Chockley et al. demonstrate in vitro and in vivo that during the epithelial-mesenchymal transition and associated metastasis, tumor cells become increasingly susceptible to NK cell killing due to increased expression of cell adhesion molecule 1 (CADM1, an activating NK ligand) and decreased expression of E-cadherin (an inhibitory NK ligand). Higher CADM1 expression correlated with improved patient survival and lower rate of metastasis, suggesting that boosting NK cell activity may be a therapeutic target that could help to prevent metastasis formation.

Enhancing CAR T cell persistence through ICOS and 4-1BB costimulation

To improve the persistence of CAR T cells in vivo, Guedan et al. modified specific intracellular signaling domains (ICDs) to induce preferred phenotypes. The ICOS ICD in CD4+ CAR T cells drove them towards a Th/Th17 phenotype and enhanced in vivo persistence, which in turn improved the persistence of co-transferred CD8+ CAR T cells with CD28 or 4-1BB ICDs. Third generation CARs incorporating ICOS and 4-1BB ICDs showed enhanced persistence and efficacy, but only when the ICOS ICD was proximal to the ICOS transmembrane domain.

Signaling by the Epstein-Barr virus LMP1 protein induces potent cytotoxic CD4(+) and CD8(+) T cell responses

In Epstein-Barr virus (EBV)-related lymphomas, EBV oncoprotein LMP1 upregulates MHC class I and class II molecules and costimulatory ligands CD70, OX40L, and 4-1BBL on the cancer cell surface, allowing LMP1+ B cells to act as antigen-presenting cells and directly prime cytolytic CD4+ and CD8+ T cell responses. Further, CD70 and OX40L drive the differentiation of CD4+ T cells towards an Eomes-programmed (Granzyme-B producing) cytotoxic state, while CD70, OX40L, and 4-1BBL all contribute to the Eomes-programming of CD8+ T cells.

The Cancer Immunotherapy Revolution: Mechanistic Insights

The Journal of Immunology opened 2018 with a special topical review on cancer immunotherapy, assembling more than 10 reviews on topics critical to translating tumor immunology to clinical success; topics include immunosurveillance, mutated antigens as T cell targets, vaccine strategies, metabolism, CAR T cells, cellular and molecular forms of immunosuppression, and strategies to effectively overcome that suppression.

Everything New this Week In...

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.