scholarly journals The Absence or Overexpression of IL-15 Drastically Alters Breast Cancer Metastasis via Effects on NK Cells, CD4 T Cells, and Macrophages

2014 ◽  
Vol 193 (12) ◽  
pp. 6184-6191 ◽  
Author(s):  
Amy Gillgrass ◽  
Navkiran Gill ◽  
Artem Babian ◽  
Ali A. Ashkar
2011 ◽  
Vol 71 (10) ◽  
pp. 3505-3515 ◽  
Author(s):  
Purevdorj B. Olkhanud ◽  
Bazarragchaa Damdinsuren ◽  
Monica Bodogai ◽  
Ronald E. Gress ◽  
Ranjan Sen ◽  
...  

2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Jing Li ◽  
Shengqi Wang ◽  
Neng Wang ◽  
Yifeng Zheng ◽  
Bowen Yang ◽  
...  

Abstract Background Metastasis represents the leading cause of death in patients with breast cancer. Traditional Chinese medicine is particularly appreciated for metastatic diseases in Asian countries due to its benefits for survival period prolongation and immune balance modulation. However, the underlying molecular mechanisms remain largely unknown. This study aimed to explore the antimetastatic effect and immunomodulatory function of a clinical formula Aiduqing (ADQ). Methods Naive CD4+ T cells, regulatory T cells (Tregs), and CD8+ T cells were sorted by flow cytometry. Then, breast cancer cells and these immune cells were co-cultured in vitro or co-injected into mice in vivo to simulate their coexistence. Flow cytometry, ELISA, qPCR, double luciferase reporter gene assay, and chromatin immunoprecipitation assay were conducted to investigate the immunomodulatory and antimetastatic mechanisms of ADQ. Results ADQ treatment by oral gavage significantly suppressed 4T1-Luc xenograft growth and lung metastasis in the orthotopic breast cancer mouse model, without noticeable hepatotoxicity, nephrotoxicity, or hematotoxicity. Meanwhile, ADQ remodeled the immunosuppressive tumor microenvironment (TME) by increasing the infiltration of tumor-infiltrating lymphocytes (TILs) and cytotoxic CD8+ T cells, and decreasing the infiltration of Tregs, naive CD4+ T cells, and tumor-associated macrophages (TAMs). Molecular mechanism studies revealed that ADQ remarkably inhibited CXCL1 expression and secretion from TAMs and thus suppressed the chemotaxis and differentiation of naive CD4+ T cells into Tregs, leading to the enhanced cytotoxic effects of CD8+ T cells. Mechanistically, TAM-derived CXCL1 promoted the differentiation of naive CD4+ T cells into Tregs by transcriptionally activating the NF-κB/FOXP3 signaling. Lastly, mouse 4T1-Luc xenograft experiments validated that ADQ formula inhibited breast cancer immune escape and lung metastasis by suppressing the TAM/CXCL1/Treg pathway. Conclusions This study not only provides preclinical evidence supporting the application of ADQ in inhibiting breast cancer metastasis but also sheds novel insights into TAM/CXCL1/NF-κB/FOXP3 signaling as a promising therapeutic target for Treg modulation and breast cancer immunotherapy.


2021 ◽  
Author(s):  
Inga Hansine Rye ◽  
Kanutte Huse ◽  
Sarah E. Josefsson ◽  
Wanja Kildal ◽  
Håvard E. Danielsen ◽  
...  

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A125-A125
Author(s):  
Erica Goddard ◽  
Shivani Srivastava ◽  
Stanley Riddell ◽  
Cyrus Ghajar

BackgroundA significant fraction of breast cancer survivors develop metastases years or even decades after initial diagnosis.1–3 Mounting evidence suggests these late recurrences arise from dormant disseminated tumor cells (DTCs).4–6 However, no therapy currently exists for targeting DTCs for the purpose of metastasis prevention. Immunotherapy represents a promising avenue to target dormant DTCs. Yet, a functional relationship between adaptive immunity and dormant DTCs has not been established.MethodsHere, we have utilized a bone marrow organotypic microvascular niche co-culture model and immunocompetent murine models of breast cancer dormancy to study the relationship between the adaptive immune response and dormant DTCs and to develop immunotherapies for the purpose of eliminating dormant DTCs and preventing breast cancer metastasis.ResultsOur data suggest that breast cancer cells downregulate MHC class I antigen presentation upon dormancy induction, identifying one mechanism of immune evasion. Strikingly, outgrowing metastases re-express MHC I and presumably upregulate antigen presentation. These data suggest that MHC-dependent T cell-based immunotherapies may not effectively kill dormant DTCs, but that MHC-independent chimeric antigen receptor (CAR) T cells may be more applicable. Using the organotypic bone marrow microvascular niche co-culture system, we have shown that CAR T cells kill both proliferating and dormant tumor cells independent of tumor cell localization in the niche and independent of tumor cell cycle status. Further, we have established preclinical immunocompetent murine models of breast cancer dormancy to compare efficacy of engineered T cell receptor (TCR) and CAR T cells in eliminating dormant DTCs. From these models of breast cancer dormancy, we have found that CAR T cells eliminate both overt metastases and DTCs in the lung and bone marrow of mice. In contrast, preliminary data suggest that TCR T cells clear overt metastases but are less effective in clearing dormant disease, lending support that MHC I downregulation during dormancy may impact the efficacy of various T cell-based immunotherapies.ConclusionsOur findings identify CAR T cells as one potential immunotherapy to eradicate dormant disease, while simultaneously identifying both CAR and TCR T cells as effective treatments for the clearance of overt metastases. In sum, our findings lay the groundwork for developing adoptive cell therapies to eliminate dormant disease and prevent death from breast cancer metastasis.ReferencesPan H, Gray R, et al. 20-Year Risks of Breast-Cancer Recurrence after Stopping Endocrine Therapy at 5 Years. N Engl J Med 2017;377, 1836–1846.Karrison T, Ferguson D, Meier P. Dormancy of mammary carcinoma after mastectomy. J Natl Cancer Inst. 1999;91, 80–85.Goss PE, & Chambers AF. Does tumour dormancy offer a therapeutic target?Nat Rev Cancer 2010;10, 871–877.Aguirre-Ghiso JA. Models, mechanisms and clinical evidence for cancer dormancy. Nat Rev Cancer 2007;7, 834–846.Klein CA. Framework models of tumor dormancy from patient-derived observations. Curr Opin Genet Dev 2011;21, 42–49.Demicheli R, Abbattista A, Miceli R, Valagussa P, & Bonadonna G. Time distribution of the recurrence risk for breast cancer patients undergoing mastectomy: further support about the concept of tumor dormancy. Breast Cancer Res Treat 1996;41, 177–185.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A475-A475
Author(s):  
Lauren Milling ◽  
Darrell Irvine

BackgroundCyclic dinucleotides (CDN) – agonists of stimulator of interferon genes – can initiate potent anti-tumor immunity by activating antigen presenting cells which prime CD8+ T cells.1 Recent studies have also highlighted CDN activation of NK cells via IL-15 in T cell-resistant tumors.2 Thus far, limited analysis has been made of the impact of CDN-based therapies on cancer metastasis. We employed a surgical resection model of metastatic mammary carcinoma to examine the effects of surgery – a predominant breast cancer intervention – and lung metastasis on neoadjuvant therapy with CDNs combined with other clinically-relevant immunotherapies including IL-2 and anti-PD-1.3Methods4T1-luciferase cells were inoculated in the mammary fat pad, palpable tumors were treated with immunotherapy starting eight days later, any remaining primary tumor was surgically resected on day 17, and metastases were monitored by luciferase imaging. Combinations of intratumoral bisphosphorothioate 2’3’ c-di-AMP (CDN), intraperitoneal (i.p.) albumin-IL2 fusion protein (Alb-IL2), and i.p. anti-PD-1 were tested in this model by measuring primary tumor growth and monitoring overall survival. CD8+ T cells, CD4+ T cells, or NK cells were depleted using anti-CD8 (2.43), anti-CD4 (GK1.5), and anti-asialo-GM1 antibodies, respectively, administered i.p. every 3 days beginning one day prior to treatment initiation. Immunophenotyping of primary tumors and lungs was conducted at several timepoints after starting therapy.ResultsIn mice bearing orthotopic 4T1-luciferase tumors, administration of three doses of CDN resulted in no cures in the absence of surgical resection. When administered prior to surgical resection CDN monotherapy yields a 20% cure rate and enhanced median overall survival compared to untreated mice (median survival 44.5 days vs 38 days, p=0.0026). Combination of CDN with Alb-IL2 and anti-PD-1 substantially improved survival, with 60% of mice surviving long-term. Through cellular depletions we determined that neither CD8+ nor CD4+ T cells were required for efficacy in this neoadjuvant therapy model, while NK cell depletion decreased survival rate by approximately 50%. Lung immunophenotying of CDN/Alb-IL-2/anti-PD-1-treated mice revealed a near doubling of the absolute NK cell count compared to untreated controls. More strikingly, lung infiltrating NK cells in the CDN/Alb-IL2/anti-PD-1 cohort exhibited prolonged granzyme B production compared to CDN monotherapy (6.24x higher after 6 days) and Alb-IL2 monotherapy (25x higher after 6 days) cohorts.ConclusionsOur findings suggest that combining intratumoral CDN with systemic Alb-IL2 and anti-PD-1 can delay the growth of primary breast tumors and limit metastatic outgrowth in the lungs. Efficacy is attributed to sustained cytotoxicity of NK cells.Ethics ApprovalAll mouse experiments were approved by MIT’s Committee on Animal Care, protocol #0720-070-23.ReferencesCorrales L, et al. Direct activation of STING in the tumor microenvironment leads to potent and systemic tumor regression and immunity. Cell Reports 2015; 11:1018–30.Nicolai CJ, et al. NK cells mediate clearance of CD8+ T cell-resistant tumors in response to STING agonists. Science Immunology 2020; 5:eaaz2738.Al-Sahaf O, et al. Surgical injury enhances the expression of genes that mediate breast cancer metastasis to the lung. Ann Surg 2010; 252:1037–43.


2012 ◽  
Vol 189 (3) ◽  
pp. 1162-1172 ◽  
Author(s):  
Mercedes Tkach ◽  
Lorena Coria ◽  
Cinthia Rosemblit ◽  
Martín A. Rivas ◽  
Cecilia J. Proietti ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document