scholarly journals Cryo-thermal therapy induces macrophage polarization for durable anti-tumor immunity

2019 ◽  
Vol 10 (3) ◽  
Author(s):  
Kun He ◽  
Shengguo Jia ◽  
Yue Lou ◽  
Ping Liu ◽  
Lisa X. Xu
2021 ◽  
Vol 22 (13) ◽  
pp. 7010
Author(s):  
Shicheng Wang ◽  
Man Cheng ◽  
Peng Peng ◽  
Yue Lou ◽  
Aili Zhang ◽  
...  

Macrophages play critical roles in both innate and adaptive immunity and are known for their high plasticity in response to various external signals. Macrophages are involved in regulating systematic iron homeostasis and they sequester iron by phagocytotic activity, which triggers M1 macrophage polarization and typically exerts antitumor effects. We previously developed a novel cryo-thermal therapy that can induce the mass release of tumor antigens and damage-associated molecular patterns (DAMPs), promoting M1 macrophage polarization. However, that study did not examine whether iron released after cryo-thermal therapy induced M1 macrophage polarization; this question still needed to be addressed. We hypothesized that cryo-thermal therapy would cause the release of a large quantity of iron to augment M1 macrophage polarization due to the disruption of tumor cells and blood vessels, which would further enhance antitumor immunity. In this study, we investigated iron released in primary tumors, the level of iron in splenic macrophages after cryo-thermal therapy and the effect of iron on macrophage polarization and CD4+ T cell differentiation in metastatic 4T1 murine mammary carcinoma. We found that a large amount of iron was released after cryo-thermal therapy and could be taken up by splenic macrophages, which further promoted M1 macrophage polarization by inhibiting ERK phosphorylation. Moreover, iron promoted DC maturation, which was possibly mediated by iron-induced M1 macrophages. In addition, iron-induced M1 macrophages and mature DCs promoted the differentiation of CD4+ T cells into the CD4 cytolytic T lymphocytes (CTL) subset and inhibited differentiation into Th2 and Th17 cells. This study explains the role of iron in cryo-thermal therapy-induced antitumor immunity from a new perspective.


2016 ◽  
Vol 6 (1) ◽  
Author(s):  
Jun Zhu ◽  
Yan Zhang ◽  
Aili Zhang ◽  
Kun He ◽  
Ping Liu ◽  
...  

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 135-135
Author(s):  
Claudine Graf ◽  
Petra Wilgenbus ◽  
Maki Kitano ◽  
Stephan Macher-Göppinger ◽  
Wolfram Ruf

Abstract Coagulation signaling through protease activated receptors (PARs) participates in inflammation and immunity. In cancer, tissue factor (TF) driven signaling via PAR2 promotes tumor progression, but effective pharmacological strategies to inhibit the PAR2 activating proteases for clinical anti-cancer benefit are currently unknown. To gain a better understanding of signaling by coagulation proteases, we generated PAR2 mouse strains with mutations that abolish canonical proteolysis by all proteases including FVIIa (PAR2 R38E) or create specific resistance to cleavage by the TF-FVIIa-Xa signaling complex (PAR2 G37I) that requires the endothelial cell protein C receptor (EPCR, Procr). As expected from delayed breast cancer development in PAR2-deficient polyoma middle T (PyMT) mice, cancer progression was impaired in completely cleavage-resistant PAR2 R38E relative to wild-type (WT) mice. In contrast, FXa-resistant PAR2 G37I mice displayed normal tumor initiation, but unexpectedly tumor growth was also markedly attenuated compared to WT mice. A similar reduction in transplanted syngeneic tumor growth in mutant PAR2 R38E and PAR2 G37I mice indicated that impaired FXa-PAR2 signaling in the tumor microenvironment (TME), but not by tumor cells, impaired tumor expansion in FXa-resistant PAR2 G37I mice. Macrophages, but not the studied tumor models, expressed FVII and FX, along with other components of the TF pathway. Tumor-associated macrophage (TAM) phenotypes are determined by complex immune cell alterations in the TME. Exposure of macrophage to tumor cell supernatant resulted in upregulation of immune-suppressive mediators; this effect was significantly attenuated in PAR2 G37I relative to WT macrophages. Accordingly, TAM isolated from spontaneous PyMT breast cancer demonstrated reduced expression of immune modulatory chemokines, as well as immune-suppressive, pro-angiogenic and pro-metastatic mediators in PAR2 mutant relative to WT mice. Thus, PAR2 signaling directly regulates TAM immune-evasive properties. In addition, macrophage-specific deletion of FX in F10flfl-LysMcre mice prevented macrophage polarization in vitro. Consistently, macrophage FX deficiency was sufficient to diminish tumor growth without reducing plasma FX levels in control or tumor-bearing littermate mice. TAM phenotypic changes in macrophage FX-deficient mice recapitulated the anti-tumor effects seen in PAR2 mutant mice and similarly resulted in increased tumor infiltration with cytotoxic anti-tumor T cells, demonstrating that PAR2 activation by cell autonomously synthesized FXa impairs anti-tumor immunity. We hypothesized that small molecule direct FXa inhibitors with peripheral tissue penetration could interfere with extravascular macrophage FXa signaling in the TME. Akin to FXa-PAR2 signaling-deficient macrophages, therapeutic concentrations of the direct FXa inhibitor rivaroxaban reduced macrophage expression of immune-suppressive mediators. Oral anticoagulation of PyMT mice achieving therapeutic plasma levels of rivaroxaban similarly altered TAM polarization in vivo and improved anti-tumor immunity specifically in macrophage FX-expressing mice. Thus, macrophage synthesized FXa is the primary target for oral FXa anticoagulants in reprogramming TAM phenotypes. These data show that cell autonomous PAR2 signaling dependent on extrahepatic synthesis of coagulation FX by macrophages controls the immune-evasive phenotype of TAM. While targeting intravascular thrombosis with heparin, the current standard anticoagulant therapy of cancer patients, has limited clinical anti-cancer efficacy, reprogramming of macrophages by direct oral anticoagulants with favorable tissue distribution may enhance cancer immunotherapy and provide anti-inflammatory benefit in other diseases. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 11 ◽  
Author(s):  
Marina Natoli ◽  
Petra Herzig ◽  
Elham Pishali Bejestani ◽  
Melanie Buchi ◽  
Reto Ritschard ◽  
...  

Reprogramming tumor infiltrating myeloid cells to elicit pro-inflammatory responses is an exciting therapeutic maneouver to improve anti-tumor responses. We recently demonstrated that a distinct microtubule-targeting drug, plinabulin—a clinical-stage novel agent—modulates dendritic cell maturation and enhances anti-tumor immunity. Here, we investigated the effects of plinabulin on macrophage polarization in vitro and in vivo. Plinabulin monotherapy induced significant tumor growth inhibition in mice bearing subcutaneous MC38 colon cancer. Importantly, the regressing tumors were characterized by an increase in M1-like/M2-like tumor-associated macrophages (TAM) ratio. The efficacy of plinabulin remained unaltered in T cell-deficient Rag2−/− mice, suggesting an important role of macrophages in driving the drug's anti-tumor effect. Exposure of murine and healthy human macrophages to plinabulin induced polarization toward the M1 phenotype, including increased expression of co-stimulatory molecules CD80, CD86 and pro-inflammatory cytokines IL-1β, IL-6, and IL-12. M2-associated immunosuppressive cytokines IL-10 and IL-4 were reduced. This pro-inflammatory M1-like skewing of TAMs in response to plinabulin was dependent on the JNK pathway. Functionally, plinabulin-polarized human M1 macrophages directly killed HuT 78 tumor cells in vitro. Importantly, plinabulin induced a functional M1-like polarization of tumor infiltrating macrophages in murine tumors as well as in tumor samples from ovarian cancer patients, by preferentially triggering M1 proliferation. Our study uncovers a novel immunomodulatory effect of plinabulin in directly triggering M1 polarization and proliferation as well as promoting TAM anti-tumoral effector functions.


2019 ◽  
Vol 9 (1) ◽  
Author(s):  
Shengguo Jia ◽  
Wentao Li ◽  
Ping Liu ◽  
Lisa X. Xu

Abstract Previous, we established a novel therapeutic approach to tumour of cryo-thermal therapy, which can induce durable anti-tumour memory immunity mediated by CD4+ T cell, and contribute to prolonged survival in B16F10 murine melanoma model and 4T1 murine mammary carcinoma. It has become apparent that innate immune cells are involved in the regulation of adaptive T cell immunity. Our previous studies revealed that cryo-thermal therapy induced M1 macrophage polarization and DCs maturation were required for the shaping of systemic long-lived T cell mediated anti-tumour memory immunity. Eosinophils are multifunctional innate effector cells and there is lack of knowledge on the role of eosinophils in cryo-thermal-induced anti-tumour immunity. This study revealed that cryo-thermal therapy activated eosinophils in spleen at early stage following the treatment. Furthermore, cryo-thermal-activated eosinophils exerted versatile immunologic regulation from innate immunity to anti-tumour adaptive immunity, such as M1 macrophage polarization, DCs maturation, differentiation of CD4-CTL subtypes and enhanced cytotoxicity of CD8+ T cells. Our study indicated that the cryo-thermal-activated eosinophils was essential for the shaping of durable anti-tumour memory immunity. Thus, our results present a new concept for eosinophils mediated anti-tumour immunity after cryo-thermal therapy.


Sign in / Sign up

Export Citation Format

Share Document