CD4(+)CD25(+) regulatory T cells (Tregs) can promote the growth of some tumors, but it is unknown whether this is true for all tumors, including malignant pleural mesothelioma. We have previously shown that the existence of Tregs was associated with poor survival in patients with malignant pleural mesothelioma. In this study, using an intrathoracic murine model of malignant mesothelioma (MM), we provide evidence suggesting that Treg blockade could enhance survival when combined with pemetrexed in established tumor. AC29 murine MM cells were injected into the right pleural cavity of CBA mice for tumor development. Four days after the tumor injection, tumor-bearing mice were then treated with pemetrexed alone, Treg blockade alone, or a combination of pemetrexed and Treg blockade. We observed a synergistic antitumor effect of Treg blockade combined with pemetrexed resulting in prolonged survival. The combination of Treg blockade and pemetrexed was associated with decreased tumor-infiltrating Tregs, increased IL-2 production, dendritic cell maturation, and increased CD3(+)CD8(+)IFN-gamma(+) tumor-infiltrating T cells when compared with mice treated with pemetrexed alone or Treg blockade alone. The survival benefit was abrogated if anti-CD8 mAb was administered simultaneously. Likewise, the survival benefit resulting from the combined Treg blockade with pemetrexed was not observed when immunodeficient mice were used. Therefore, this study suggests that Treg blockade combined with pemetrexed can suppress mesothelioma growth in established tumor in vivo through an immune-mediated process. This study also validates a new intrathoracic tumor model of pleural effusion to explore the role of antitumor immunity in murine MM.

Download full-text PDF

Source
http://dx.doi.org/10.4049/jimmunol.0900437DOI Listing

Publication Analysis

Top Keywords

treg blockade
32
combined pemetrexed
16
blockade combined
12
pemetrexed treg
12
blockade
9
pemetrexed
9
synergistic antitumor
8
malignant mesothelioma
8
malignant pleural
8
pleural mesothelioma
8

Similar Publications

Biomimetic calcium-chelation nanoparticles reprogram tumor metabolism to enhance antitumor immunity.

J Control Release

January 2025

NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China. Electronic address:

Metabolic reprogramming within the tumor microenvironment poses a significant obstacle to the therapeutic efficacy of antitumor immunity. Here, inspired by the diverse programme of cholesterol metabolism between tumor and immune cells, a biocompatible carboxy-modified cyclodextrin carrier equipped with a biomimetic surface was developed to encapsulate FX11 and Avasimibe (RM-CDC@FX11&Ava) for synergistic antitumor metabolic therapy and immunotherapy. Through the manipulation of calcium levels using poly-carboxylic compounds to initiate cholesterol biosynthesis, RM-CDC@FX11&Ava dynamically regulates glycolysis and blocks cholesterol esterification to navigate metabolic reprogramming.

View Article and Find Full Text PDF

Septic arthritis (SA) caused by Staphylococcus aureus is a severe inflammatory joint disease, characterized by synovitis accompanied with cartilage destruction and bone erosion. The available antibiotic treatment alone is insufficient to resolve the inflammation that leads to high rates of morbidity and mortality. Among the CD4 T helper lymphocytes, the Th17 and Tregs are key regulators of immune homeostasis.

View Article and Find Full Text PDF

Tumor initiating cells escape tumor immunity via CCL8 from tumor-associated macrophages in mice.

J Clin Invest

January 2025

Department of Medical Oncology; Department of Pancreato-Biliary Surgery; De, Center for Translational Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.

Tumor-initiating cells (TICs) play a key role in cancer progression and immune escape. However, how TICs evade immune elimination remains poorly characterized. Combining single-cell RNA sequencing (scRNA-seq), dual-recombinase-based lineage tracing, and other approaches, we identified a WNT-activated subpopulation of malignant cells that act as TICs in vivo.

View Article and Find Full Text PDF

Background: Treatment with immunotherapy can elicit varying responses across cancer types, and the mechanistic underpinnings that contribute to response vrsus progression remain poorly understood. However, to date there are few preclinical models that accurately represent these disparate disease scenarios.

Methods: Using combinatorial radio-immunotherapy consisting of PD-1 blockade, IL2Rβγ biased signaling, and OX40 agonism we were able to generate preclinical tumor models with conflicting responses, where head and neck squamous cell carcinoma (HNSCC) models respond and pancreatic ductal adenocarcinoma (PDAC) progresses.

View Article and Find Full Text PDF

Unleashing the Power of immune Checkpoints: A new strategy for enhancing Treg cells depletion to boost antitumor immunity.

Int Immunopharmacol

January 2025

Department of Oral Biology, School and Hospital of Stomatology, Jilin University, Changchun, China; Key Laboratory of Tooth Development and Bone Remodeling of Jilin Province, School and Hospital of Stomatology, Jilin University, Changchun, China. Electronic address:

Article Synopsis
  • Regulatory T (Treg) cells are a type of immunosuppressive CD4 T cells that can hinder anti-tumor immune responses, creating challenges in cancer treatment.
  • Various immune checkpoints (like PD-1/PD-L1, CTLA-4) play critical roles in managing Treg cell activity and proliferation in tumors, impacting overall tumor immunity.
  • Targeting these checkpoints and combining therapies, such as immune checkpoint blockade and CCR8-targeted treatments, may enhance anti-tumor immunity and improve results for cancer patients.
View Article and Find Full Text PDF

Want AI Summaries of new PubMed Abstracts delivered to your In-box?

Enter search terms and have AI summaries delivered each week - change queries or unsubscribe any time!