scholarly journals TPX2 Derived Vaccine to Inhibit Tumorigenesis and Metastasis in Triple Negative Breast Cancer 

Author(s):  
Yueqiang Jiang ◽  
Yan Liu ◽  
Ling Cheng ◽  
Xiaolong Tan ◽  
Jian Luo ◽  
...  

Abstract Background: triple negative breast cancer (TNBC) lacks of treatment approaches rather than other subtypes of breast cancer. However, it characterizes the highest the level of tumor infiltrating lymphocyte (TIL) than other subtypes, indicating the possibility of immunotherapy. Method: female BALB/c background mice are immunized with a TPX2 derived vaccine 4 consecutive times (once a week) from 6 weeks old, and then 4T1 cells are transplanted to the #4 mammary fatpad. Surface tumor volume and No. of lung metastasis were recorded. TIL and splenocyte was collected for T cell subgroup analysis by methods of flow cytometry and IFN-γ ELISPOT. Result: as a result, TPX2 derived vaccine shows moderate effect, shrinking the tumor volume from 804.4 to 504.5 mm3, and decreasing the number of lung metastasis from 16.6 to 6.0 in control group compared to vaccine group. CD8+ T cell ratio are obviously increased in TIL between vaccine group and control group (5.87% vs 3.37%, P=0.0012). And vaccine could induce strong immune response both in tumor site (87.6 vs 7.0, p=0.0004) and system (28.2 vs 3.8, p<0.0001) through IFN-γ ELISPOT. Conclusion: our result indicated that TPX2 derived vaccine may be an effective approach to inhibit TNBC tumorigenesis and metastasis.

Antibodies ◽  
2020 ◽  
Vol 9 (4) ◽  
pp. 51
Author(s):  
Ellinor I. Peerschke ◽  
Elisa de Stanchina ◽  
Qing Chang ◽  
Katia Manova-Todorova ◽  
Afsar Barlas ◽  
...  

gC1qR is highly expressed in breast cancer and plays a role in cancer cell proliferation. This study explored therapy with gC1qR monoclonal antibody 60.11, directed against the C1q binding domain of gC1qR, in a murine orthotopic xenotransplant model of triple negative breast cancer. MDA231 breast cancer cells were injected into the mammary fat pad of athymic nu/nu female mice. Mice were segregated into three groups (n = 5, each) and treated with the vehicle (group 1) or gC1qR antibody 60.11 (100 mg/kg) twice weekly, starting at day 3 post-implantation (group 2) or when the tumor volume reached 100 mm3 (group 3). At study termination (d = 35), the average tumor volume in the control group measured 895 ± 143 mm3, compared to 401 ± 48 mm3 and 701 ± 100 mm3 in groups 2 and 3, respectively (p < 0.05). Immunohistochemical staining of excised tumors revealed increased apoptosis (caspase 3 and TUNEL staining) in 60.11-treated mice compared to controls, and decreased angiogenesis (CD31 staining). Slightly decreased white blood cell counts were noted in 60.11-treated mice. Otherwise, no overt toxicities were observed. These data are the first to demonstrate an in vivo anti-tumor effect of 60.11 therapy in a mouse model of triple negative breast cancer.


Aging ◽  
2021 ◽  
Vol 13 (5) ◽  
pp. 7211-7227
Author(s):  
Jie Ma ◽  
Ying Yang ◽  
Lulu Wang ◽  
Xiaowei Jia ◽  
Tao Lu ◽  
...  

Molecules ◽  
2021 ◽  
Vol 26 (12) ◽  
pp. 3644
Author(s):  
Daeun You ◽  
Yisun Jeong ◽  
Sun Young Yoon ◽  
Sung A Kim ◽  
Eunji Lo ◽  
...  

Interleukin-1 (IL1) is a proinflammatory cytokine and promotes cancer cell proliferation and invasiveness in a diversity of cancers, such as breast and colon cancer. Here, we focused on the pharmacological effect of Entelon® (ETL) on the tumorigenesis of triple-negative breast cancer (TNBC) cells by IL1-alpha (IL1A). IL1A enhanced the cell growth and invasiveness of TNBC cells. We observed that abnormal IL1A induction is related with the poor prognosis of TNBC patients. IL1A also increased a variety of chemokines such as CCL2 and IL8. Interestingly, IL1A expression was reduced by the ETL treatment. Here, we found that ETL significantly decreased the MEK/ERK signaling pathway in TNBC cells. IL1A expression was reduced by UO126. Lastly, we studied the effect of ETL on the metastatic potential of TNBC cells. Our results showed that ETL significantly reduced the lung metastasis of TNBC cells. Our results showed that IL1A expression was regulated by the MEK/ERK- and PI3K/AKT-dependent pathway. Taken together, ETL inhibited the MEK/ERK and PI3K/AKT signaling pathway and suppressing the lung metastasis of TNBC cells through downregulation of IL1A. Therefore, we propose the possibility of ETL as an effective adjuvant for treating TNBC.


2021 ◽  
Vol 85 ◽  
pp. 104664
Author(s):  
Mohan Li ◽  
Kexin Zheng ◽  
Shiliang Ma ◽  
Pengpeng Hu ◽  
Bo Yuan ◽  
...  

2021 ◽  
Vol 360 ◽  
pp. 104262
Author(s):  
Pengxiang Yang ◽  
Xingjian Cao ◽  
Huilong Cai ◽  
Panfeng Feng ◽  
Xiang Chen ◽  
...  

Oncotarget ◽  
2015 ◽  
Vol 6 (28) ◽  
pp. 25356-25367 ◽  
Author(s):  
Claudia Paret ◽  
Petra Simon ◽  
Kirsten Vormbrock ◽  
Christian Bender ◽  
Anne Kölsch ◽  
...  

2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A644-A644
Author(s):  
Anita Mehta ◽  
Madeline Townsend ◽  
Madisson Oliwa ◽  
Patrice Lee ◽  
Nicholas Saccomano ◽  
...  

BackgroundPoly(ADP-ribose) polymerase inhibitors (PARPi) have improved the outcomes of BRCA-associated breast cancer; however, treatment responses are often not durable. Our preclinical studies demonstrated that PARPi activates the cGAS/STING pathway and recruitment of anti-tumor CD8+ T-cells that are required for tumor clearance [1]. These studies contributed to development of clinical trials testing PARPi plus immune checkpoint blockade (ICB). Unfortunately, early phase trials of PARPi + ICB have not yet suggested efficacy will be superior to PARPi monotherapy. Lack of demonstrated clinical synergy between PARPi + ICB underscores the need to study the tumor microenvironment (TME) during PARPi therapy to identify optimal strategies to enhance T-cell activation. We recently showed that PARPi induces CSF-1R+ suppressive tumor associated macrophages (TAMs) that restrict antitumor immune responses, contributing to PARPi resistance [2]. Removing TAMs with anti-CSF-1R therapy in combination with PARPi significantly enhanced overall survival (OS) compared to PARPi monotherapy in preclinical models [2]. Here, we investigate how modulating TAMs can enhance PARPi + ICB.MethodsMice bearing BRCA1-deficient TNBC (K14-Cre;Brca1f/f;p53f/f) tumors were treated for 98 days with PARPi (Talazoparib) ± small molecule inhibitor of CSF-1R (ARRAY-382; CSF-1Ri) ± anti-PD-1 and then followed for survival. Flow cytometry was employed to elucidate changes in the TME after treatment.ResultsPARPi conferred a significant survival advantage over vehicle treated mice (median OS 33 v. 14 days; p=0.0034) and 2/8 PARPi-treated mice experienced complete tumor clearance at day 98. PARPi + CSF-1Ri treated mice (median OS 140 days) remarkably cleared 7/10 tumors by day 98. The addition of anti-PD-1 to PARPi did not enhance OS compared to PARPi monotherapy. The triple combination of anti-PD-1 + PARPi + CSF-1Ri has not yet significantly enhanced the median OS compared to PARPi + CSF-1Ri (ongoing; 168 v. 140 days); nor did it increase clearance of tumor by day 98 (7/10). However, the triple combination led to superior long term tumor clearance. At day 161 the triple combination exhibited 5/10 tumor free mice compared to 2/10 treated with PARPi + CSF-1Ri. To elucidate how CSR-1Ri enhanced PARPi + ICB responses, flow cytometry was performed and revealed increased expression of the co-stimulatory molecule CD80, reduced tissue resident macrophages (CX3CR1+) and lower CSF-1R expression compared to PARPi + ICB.ConclusionsThese data suggest that targeting immunosuppressive macrophages may induce a favorable anti-tumor immune response and enhance responses to PARPi plus ICB. We are currently evaluating the adaptive immune response in this context.ReferencesPantelidou, C., et al., PARP inhibitor efficacy depends on CD8+ T cell recruitment via intratumoral STING pathway activation in BRCA-deficient models of triple-negative breast cancer. Cancer Discovery, 2019: p. CD-18-1218.Mehta, A.K., et al., Targeting immunosuppressive macrophages overcomes PARP inhibitor resistance in BRCA1-associated triple-negative breast cancer. Nat Cancer, 2021. 2(1): p. 66–82.


2021 ◽  
Vol 12 ◽  
Author(s):  
Gabriel Goncalves ◽  
Kerry A. Mullan ◽  
Divya Duscharla ◽  
Rochelle Ayala ◽  
Nathan P. Croft ◽  
...  

Peptide vaccination remains a viable approach to induce T-cell mediated killing of tumors. To identify potential T-cell targets for Triple-Negative Breast Cancer (TNBC) vaccination, we examined the effect of the pro-inflammatory cytokine interferon-γ (IFNγ) on the transcriptome, proteome, and immunopeptidome of the TNBC cell line MDA-MB-231. Using high resolution mass spectrometry, we identified a total of 84,131 peptides from 9,647 source proteins presented by human leukocyte antigen (HLA)-I and HLA-II alleles. Treatment with IFNγ resulted in a remarkable remolding of the immunopeptidome, with only a 34% overlap between untreated and treated cells across the HLA-I immunopeptidome, and expression of HLA-II only detected on treated cells. IFNγ increased the overall number, diversity, and abundance of peptides contained within the immunopeptidome, as well increasing the coverage of individual source antigens. The suite of peptides displayed under conditions of IFNγ treatment included many known tumor associated antigens, with the HLA-II repertoire sampling 17 breast cancer associated antigens absent from those sampled by HLA-I molecules. Quantitative analysis of the transcriptome (10,248 transcripts) and proteome (6,783 proteins) of these cells revealed 229 common proteins and transcripts that were differentially expressed. Most of these represented downstream targets of IFNγ signaling including components of the antigen processing machinery such as tapasin and HLA molecules. However, these changes in protein expression did not explain the dramatic modulation of the immunopeptidome following IFNγ treatment. These results demonstrate the high degree of plasticity in the immunopeptidome of TNBC cells following cytokine stimulation and provide evidence that under pro-inflammatory conditions a greater variety of potential HLA-I and HLA-II vaccine targets are unveiled to the immune system. This has important implications for the development of personalized cancer vaccination strategies.


Author(s):  
Rama Das ◽  
Parna Basu ◽  
Suman Ghosh ◽  
Debasish Guha

Introduction: Breast cancer continues to be the most common cancers among women in India. The Triple Negative Breast Cancer (TNBC) is a heterogeneous group of malignancy which is often aggressive and has a worse prognosis. Aim: The aim of this study was to assess the hormone receptor and HER2/neu status with platinum based chemotherapy in TNBC. Materials and Methods: The study was analysed retrospectively in a tertiary care centre of West Bengal from Januay 2017 to December 2019. Forty TNBC patients of Locally Advanced Breast Cancer (LABC) cases who received carboplatin along with neoadjuvant chemotherapy (study group) were compared with other group of 64 TNBC patients (control group) who did not receive any chemotherapy making a total of 104 cases of TNBC patients who were selected for the study. All the patients in both the groups had modified radical mastectomy. The study group of 40 TNBC patients who received chemotherapy also showed pathological partial response. Masterchart was prepared comprising patient’s age, menopausal status, family history, therapy history, histo-morphological features, hormone receptor and HER2/neu status after platinum added chemotherapy. Oestrogen Receptor (ER)/Progesterone Receptor (PR) were considered positive, if >1% tumour cell nuclei were immunoreactive and negative, if it was otherwise. HER-2/neu score of 3+ was taken as positive by Immunohistochemistry (IHC) method. Statistical analysis for descriptive purposes, percentages and mean were calculated. Comparison of both the groups was done by Pearson’s Chi-squared and Fisher’s-exact test. Significance level was considered at p-value <0.05. Results: TNBC patients (NACT group) showed hormone receptor positivity of 21 cases (52.50%) after chemotherapy along with carboplatin. HER2/neu positivity was detected in 9 (22.5%) cases. Non-NACT (64) cases were considered as control group for comparison. The effect of NACT in TNBC patients was found to be statistically significant with respect to change in HER2/neu (p=0.033, p<0.05) and ER status (p<0.05) while change in PR status was found to be statistically insignificant. Conclusion: The study showed significant alteration in hormonal and HER2/neu receptor status in TNBC patients receiving platinum added neoadjuvant chemotherapy. This study found statistical significance and justifies re-evaluation of these Hormone Receptor (HR) and HER2/neu markers in residual tumour after chemotherapy.


Sign in / Sign up

Export Citation Format

Share Document