Weekly Digests
‹ Back to July

Depletion of tumor-associated macrophages with a CSF-1R kinase inhibitor enhances antitumor immunity and survival induced by DC immunotherapy

July 5, 2017

Malignant mesothelioma is a cancer characterized by a significant stromal component that is dominated by macrophages. Tumor-associated macrophages (TAMs) are supported by macrophage-colony stimulating factor (M-CSF), which not only enhances TAM survival and proliferation, but also their recruitment towards tumors and the expression of a pro-tumor phenotype. TAMs are linked to immune suppression, angiogenesis, and tumor aggressiveness in mesothelioma, which led Dammeijer et al. to the hypothesis that depletion of TAMs could improve the efficacy of a tumor lysate-pulsed dendritic cell (DC) vaccine.

For this study, the researchers used two orthotopic mesothelioma mouse models, BALB/c (AB1) and CBA/J (AC29). The CBA/J (AC29) model was selected for its more prominent presence of macrophages of the pro-tumor phenotype - likely due to an increased expression of M-CSF - and its tendency to form ascites, paralleling the presentation of mesothelioma in cancer patients. In an effort to inhibit the receptor for M-CSF and reduce the number of TAMs in vivo, the researchers used the clinically-tested small molecule tyrosine kinase inhibitor PLX3397 (pexidartinib).

PLX3397 monotherapy effectively reduced the number of TAMs, decreased the volume of ascites, reduced neo-angiogenesis and reduced the number of circulating nonclassical monocytes, which typically express PD-L1. PLX3397 alone, however, did not improve the overall survival of mesothelioma-bearing mice.

Vaccination with tumor lysate-pulsed DCs in isolation, on the other hand, increased survival in ~20% of the mice, increased the number of non-exhausted CD8+ T cells in the tumor, and improved the CD8+/ Treg cell ratio in the peripheral blood. Within the circulating CD8+ T cell population, DC vaccination raised the percentage of short-lived KLRG1+ effector cells from virtually 0% to about 20%.

The combination of the DC vaccine with PLX3397 treatment significantly enhanced survival. The number of circulating Treg cells decreased even further than it had with DC vaccination alone, and an increased proportion of CD8+ T cells was found to co-express CD4 (this double positivity has been associated with improved effector functions). With TAMs and PD-L1+ nonclassical monocytes mostly cleared, CD8+ T cell populations within the tumor showed reduced surface expression of PD-1, indicating less exhaustion. Overall, this suggests that the depletion of TAMs by PLX3397 helped by improving the phenotypes of tumor-infiltrating lymphocytes recruited following DC vaccination.

Three months after treatment with the combination or the DC vaccine alone, surviving animals were subjected to a tumor rechallenge and proved to be fully protected. Proliferation of CD8+ and CD4+ T cells was higher in combination-treated mice compared to mice treated with DC monotherapy, indicating a superior memory response.

These results show that the combination of a DC vaccine that efficiently induces an anti-tumor immune response, with an agent that effectively targets inhibitory macrophages in the tumor microenvironment, works in synergy and leads to robust and durable antitumor immunity.

by Lauren Hitchings

References:

Dammeijer F., Lievense L.A., Kaijen-Lambers M.E.H, van Nimwegen M., Bezemer K., Hegmans J.P., van Hall T., Hendriks R.W., Aerts J.G. Depletion of tumor-associated macrophages with a CSF-1R kinase inhibitor enhances antitumor immunity and survival induced by DC immunotherapy. Cancer Immunol Res. 2017 May 23.

In the Spotlight...

Co-administration of RANKL and CTLA4 antibodies enhances lymphocyte-mediated anti-tumor immunity in mice.

A mechanism of action study of a combination of anti-CTLA-4 and anti-RANKL antibodies in subcutaneous and metastatic murine tumor models revealed improved tumor control, increased cytokine production, and increased T cell recruitment compared with either monotherapy. The synergistic effect gained by blocking the interaction between RANKL+ TILs and RANK+ myeloid cells was depended on cross-presenting Batf3+ DCs.

Vaccination with High-Affinity Epitopes Impairs Antitumor Efficacy by Increasing PD-1 Expression on CD8+ T Cells.

Using a related set of epitope peptides which varied in MHC affinity and TCR interaction strength, Zahm et al. made the counterintuitive observation that epitopes with more moderate interaction strengths may be preferable over stronger epitopes due to lower and less persistent upregulation of exhaustion markers. This preference was reversible if anti-PD-1 therapy accompanied the vaccination.

Immature human DCs efficiently translocate endocytosed antigens into the cytosol for proteasomal processing.

Using cytochrome C protein (cyt c) as a marker, Baleeiro and Walden show that immature human DCs endocytose cyt c and translocate it to the cytoplasm intact, but that it is rapidly degraded by the proteasome, consistent with effective cross-presentation capability. Mature DCs and monocytes/macrophages/lymphocytes were poor or ineffective, respectively, in shuttling any protein to the cytoplasm.

Tissue-resident memory features are linked to the magnitude of cytotoxic T cell responses in human lung cancer.

Transcriptome profiling of purified CD8+ TILs from treatment-naive lung or head and neck cancer patients revealed commonalities in the expression of genes associated with exhaustion and activation, suggestive of continued stimulation, and novel heterogeneity in the expression of CD103, a marker of resident memory cells, which is linked to survival outcomes in lung cancer.

Cutting Edge: Activation of STING in T Cells Induces Type I IFN Responses and Cell Death.

Larkin et al. provide the first evidence that T cells, much like innate cells, can be activated by STING to produce Type I IFN. Unexpectedly, STING activation in T cells also activated cell stress and death pathways, and affected T cell proliferation (observed in vitro, but not in vivo), calling for evaluation of the effect STING therapies may have on the T cell compartment.

Selective replication of oncolytic virus M1 results in a bystander killing effect that is potentiated by Smac mimetics.

Cai et al. tested a combination therapy of Smac mimetic compounds, which enhance apoptosis, with the oncolytic alphavirus M1, which targets and kills tumor cells and induces a host immune response including cytokine production. The combination therapy enhanced viral replication and irreversible ER stress in infected tumor cells, and sensitized uninfected bystander tumor cells to killing by M1-induced cytokines.

In situ vaccination after accelerated hypofractionated radiation and surgery in a mesothelioma mouse model.

Optimal timing of non-ablative local radiation and subsequent surgery in a mesothelioma mouse model resulted in an increase of activated, antigen-specific CD8+ T cells in the tumor and a protective, long-term immunological response via memory CD4+ T cells. Radiation synergized with concomitant CTLA-4 blockade, eliciting a call for clinical evaluation of the combined therapies.

First-Line Nivolumab in Stage IV or Recurrent Non-Small-Cell Lung Cancer.

Unexpectedly, in an open-label phase 3 trial comparing first-line nivolumab with platinum-based chemotherapy in patients with PD-L1+ NSCLC, no significant difference in progression-free survival or overall survival was found. Only high mutation burden, an exploratory biomarker, was associated with improved outcomes in the nivolumab group.

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.