Weekly Digests
‹ Back to April

Targeting macrophages and neutrophils enhances response to chemotherapy

April 3, 2019

Previous research has shown that the immune system contributes to chemotherapy response; therefore, immunosuppressive mechanisms can be hypothesized to limit the cytotoxic effect. In a study recently published in Nature Cell Biology, Salvagno et al. demonstrated that targeting immunosuppressive macrophages and neutrophils enhances the efficacy of platinum-based chemotherapy in a mouse breast cancer model.

To find ways to enhance the efficacy of chemotherapy, researchers utilized the poorly immunogenic K14cre;Cdh1F/F;Trp53F/F (KEP) transgenic mouse model of spontaneous mammary tumorigenesis, which is similar to human invasive lobular carcinoma. They found that both cancer and host cells in KEP tumors express high levels of colony-stimulating factor-1 (CSF-1), which leads to high infiltration by macrophages. The CSF-1 receptor (CSF-1R) was found to be expressed on tumor-associated macrophages (TAMs) at high levels and on infiltrating monocytes and neutrophils at lower levels. Treating tumor-bearing KEP mice with anti-CSF-1R significantly reduced the TAM population, however, it did not affect tumor growth or metastasis formation. Anti-CSF-1R synergized in combination with cisplatin or oxaliplatin (platinum-based, DNA cross-linking chemotherapeutic agents) to improve tumor control and prolong survival compared to chemotherapy with a control antibody. Interestingly, such synergy did not occur between anti-CSF-1R and docetaxel (an antimitotic chemotherapeutic agent).

While anti-CSF-1R depleted the majority of TAMs, which were CD11bhi CD206lo with some expression of MHC-II, the CD11b+F4/80+ cells that remained in the tumor appeared to have an altered profile – they had increased expression of MHC-II, PD-L1, monocyte marker Ly6C, and costimulatory molecules CD80 and CD86, and decreased expression of chemokine receptors CCR2 and CX3CR1. The results of further experiments suggested that this cell population may have arisen from newly recruited circulating monocytes, which were increased in the tumors after anti-CSF-1R treatment.

Digging deeper to understand the mechanism behind the synergistic response of the anti-CSF-1R and cisplatin combination therapy, the researchers utilized RNAseq and found that the CD11b+F4/80+ cells remaining in the tumor after combination treatment were strongly enriched in type I IFN production and signaling genes. This cell population expressed lower levels of CSF-1R, potentially explaining resistance to anti-CSF-1R therapy. Mechanistically, anti-CSF-1R upreguled intracellular pattern recognition receptors that signal to induce type I IFN production, and this increase in intratumoral type I IFN signaling was found to be essential for the antitumor synergy of the anti-CSF-1R and cisplatin combination therapy. Further experiments demonstrated that type I IFNs also directly inhibited KEP cancer cells.

Interestingly, the efficacy of the combination treatment was not dependent on CD8+ T cells or any other components of the adaptive immune system, indicating that the anti-CSF-1R-mediated type I IFN-rich tumor microenvironment was not sufficient to induce an antitumor T cell response. Although the anti-CSF-1R led to enriched type I IFN signaling genes in neutrophils, Salvagno et al. suspected that this cell population was immunosuppressive in the context of the KEP tumors. Adding the neutrophil-depleting anti-Ly6G to anti-CSF-1R and cisplatin further improved tumor control and survival, and this additional benefit was dependent on the innate and adaptive immune response, including NK cells and CD8+ T cells.

Seeking to validate the preclinical results in patients, Salvagno et al. analyzed pre-treatment and on-treatment tumor biopsies from patients with advanced solid tumors who were treated with emactuzumab, a humanized anti-human CSF-1R monoclonal antibody. Concordant with the preclinical data, gene expression profiling of the biopsies indicated that treatment increased the expression of multiple type I IFN-stimulated genes.

Overall, Salvagno et al. demonstrated that in the KEP transgenic mouse model of breast cancer, targeting TAMs with anti-CSF-1R depleted most of the macrophages and altered the profile of the remaining CD11b+F4/80+ cells to induce the production and signaling of type I IFN, which was required for the synergy between anti-CSF-1R and platinum-based chemotherapy and led to improved survival. However, additional depletion of the immunosuppressive intratumoral neutrophils with anti-Ly6G was necessary to engage the endogenous antitumor response in order to further boost tumor control and prolong survival. The ability of anti-human CSF-1R treatment in patients with advanced solid tumors to increase the expression of type I IFN-stimulated genes indicates the potential clinical relevance of this combinatorial approach.

by Anna Scherer

References:

Salvagno C., Ciampricotti M., Tuit S., Hau C.S., van Weverwijk A., Coffelt S.B., Kersten K., Vrijland K., Kos K., Ulas T., Song J.Y., Ooi C.H., Rüttinger D., Cassier P.A., Jonkers J., Schultze J.L., Ries C.H., de Visser K.E. Therapeutic targeting of macrophages enhances chemotherapy efficacy by unleashing type I interferon response. Nat Cell Biol. 2019 Mar 18.

In the Spotlight...

KRAS-IRF2 Axis Drives Immune Suppression and Immune Therapy Resistance in Colorectal Cancer

Using a colorectal cancer (CRC) mouse model with an inducible oncogenic Kras allele, Liao et al. found that mutated KRAS inhibits antitumor immunity by suppressing the expression of IRF2 (a repressor of Cxcl3 expression). With low or no IRF2 expression, cancer cells express high levels of CXCL3, which recruits MDSCs to the tumor via CXCR2. Enforcing IRF2 expression reduced CXCL3 expression, MDSC infiltration, and tumor growth. Inhibiting the CXCL3-CXCR2 axis overcame resistance to anti-PD-1. Analysis of human CRC samples aligned with this mechanism, and higher IRF2 levels correlated with response to anti-PD-1 in a small cohort.

Secreted PD-L1 variants mediate resistance to PD-L1 blockade therapy in non-small cell lung cancer

Using RNAseq, Gong et al. identified secreted PD-L1 (sPD-L1) splicing variants lacking the transmembrane domain in NSCLC patients who relapsed after anti-PD-L1 treatment. Patients with these variants had elevated levels of soluble PD-L1 in the plasma and in pleural effusion fluid. sPD-L1 variants conferred resistance to anti-PD-L1 in mice bearing MC38 tumors by accumulating soluble PD-L1 in the plasma, which could trap anti-PD-L1 and inhibit its function, thus shortening survival. Anti-PD-1 blockade efficacy was not affected by soluble PD-L1, and may be an option for patients with sPD-L1 variants.

Engineered immune cells as highly sensitive cancer diagnostics

Finding that M2 macrophages form a major immune subset within solid tumors, Aalipour et al. engineered macrophages expressing secreted Gaussia luciferase (Gluc) downstream of the arginase-1 promoter as a cancer diagnostic. Bone marrow-derived and RAW264.7 macrophages migrated and upregulated Arg1 in response to tumor-conditioned media in vitro, and co-localized with tumors in vivo. Plasma Gluc from transferred macrophages detected metastases and subcutaneous tumors as small as 25mm3 with higher sensitivity than shed-tumor-antigen and cell-free DNA biosensors, and tracked wound healing in models of inflammation.

Contributed by Alex Najibi

REVIEW: Antibody-Cytokine Fusions: Versatile Products for the Modulation of Anticancer Immunity

In this review, Dario Neri discusses the current status and future directions for antibody-cytokine fusion proteins (immunocytokines), which aim to increase the therapeutic index of cytokines with anticancer activity by delivering them directly to the TME. Various factors affect the efficacy of immunocytokines, including the target antigen, the design (e.g. IgG vs. antibody fragment), the cytokine payload, whether off-tumor receptors act as immunocytokine sinks, and the dose and schedule of treatment. Reducing the systemic toxicity of immunocytokines, improving their efficacy, and designing rational combination therapies are identified as key goals.

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.