Antigen escape in CAR-T therapy: A tumor microenvironment perspective from hematological to solid tumors.

Int J Cancer

Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China.

Published: August 2025


Category Ranking

98%

Total Visits

921

Avg Visit Duration

2 minutes

Citations

20

Article Abstract

Chimeric antigen receptor T-cell (CAR-T) therapy has shown remarkable efficacy in hematologic malignancies, but antigen escape remains a major challenge, especially in solid tumors, where the tumor microenvironment (TME) exacerbates the problem. Mechanisms of antigen escape include antigen loss, epitope masking, lineage switching, and trogocytosis-mediated CAR dysfunction. The TME promotes immune evasion through physical barriers, immunosuppressive cells, and metabolic competition. To overcome these challenges, multi-targeted CAR-Ts, gene editing, epigenetic interventions, and combination therapies have been developed to enhance CAR-T efficacy. Emerging strategies-such as microbial-guided antigen labeling, nanotechnology for metabolic normalization, armored CAR-T secreting TME-modulating agents, and adaptive CAR systems responsive to TME signals-offer new solutions to target "cold" tumors. Future breakthroughs will rely on synergizing dynamic CAR systems for broad antigen coverage, achieved via multi-targeting and non-canonical antigen recognition, with engineered TME remodeling driven by microbial, viral, and immune cell allies, as well as armored CAR-T cells secreting immunomodulators. Combined with metabolic engineering and interdisciplinary innovation, this integrated approach will effectively enable CAR-T cells to orchestrate a multi-faceted anti-tumor ecosystem rather than functioning in isolation.

Download full-text PDF

Source
http://dx.doi.org/10.1002/ijc.70117DOI Listing

Publication Analysis

Top Keywords

antigen escape
12
antigen
8
car-t therapy
8
tumor microenvironment
8
solid tumors
8
armored car-t
8
car systems
8
car-t cells
8
car-t
6
escape car-t
4

Similar Publications

The presentation of peptides on HLA molecules is essential to CD8 T cell responses. Here, we show that loss of uL14 significantly downregulates the expression of antigen processing and presentation (APP) components in melanoma cell lines. Peptides generated following knockdown show different characteristics, with altered peptide charge, and differences in anchor residue positions.

View Article and Find Full Text PDF

Advances in Tumor Microenvironment and Immunotherapeutic Strategies for Hepatocellular Carcinoma.

Oncol Res

September 2025

Department of Biliary-Pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.

Hepatocellular carcinoma (HCC) is a highly aggressive malignancy, largely driven by an immunosuppressive tumor microenvironment (TME) that facilitates tumor growth, immune escape, and resistance to therapy. Although immunotherapy-particularly immune checkpoint inhibitors (ICIs)-has transformed the therapeutic landscape by restoring T cell-mediated anti-tumor responses, their clinical benefit as monotherapy remains suboptimal. This limitation is primarily attributed to immunosuppressive components within the TME, including tumor-associated macrophages, regulatory T cells (Tregs), and myeloid-derived suppressor cells (MDSCs).

View Article and Find Full Text PDF

Cancer vaccines in hematologic malignancy: A systematic review of the rational and evidence for clinical use.

Best Pract Res Clin Haematol

September 2025

Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA; Dana-Farber/Harvard Cancer Center, Harvard Medical School, Boston, USA.

Immunotherapy, including immune checkpoint blockade, CART cells and bispecific antibodies have resulted in dramatic improvements in outcomes for patients with hematological malignancies, demonstrating the unique potency of the immune system in targeting malignant cells. The development of cancer vaccines aims to evoke an activated effector cell population and a memory response to provide long term immune surveillance to protect from relapse. Developing a potent cancer vaccine relies on identifying appropriate antigen targets, enhancing antigen presentation, and overcoming the immune suppressive milieu of the micro-environment.

View Article and Find Full Text PDF

Chimeric antigen receptor (CAR) T-cell therapy has changed how we treat blood cancers but hasn't worked as well for solid tumors like pancreatic ductal adenocarcinoma (PDAC), mainly because these tumors are very aggressive and resistant to regular treatments. This review critically examines peer-reviewed studies to chart the evolution of immunotherapy in PDAC, emphasizing the unique barriers to effective CAR T-cell treatment and emerging strategies to overcome them. CAR T-cells that focus on tumor-related markers like mesothelin, HER2, and MUC1 have shown promise in early research models.

View Article and Find Full Text PDF

Background: Although immune checkpoint inhibitors (ICIs) have markedly improved first-line management of non-small cell lung cancer (NSCLC), many tumors eventually escape control after anti-PD-(L)1 therapy, leaving a clear therapeutic gap. Preclinical studies and preliminary clinical data suggest that coupling ICIs with anti-angiogenesis therapy can yield complementary antitumor effects. Consequently, we launched this investigation to evaluate the therapeutic benefit and tolerability of sintilimab, a PD-(L)1-blocking monoclonal antibody, together with the oral multi-target anti-angiogenesis agent anlotinib in metastatic NSCLC individuals experiencing progression after first-line PD-(L)1 inhibition.

View Article and Find Full Text PDF