Characterizing and Monitoring Antigen-Specific T and B Cells in Cancer Vaccine Development.
Over 20% of papers studying antigen-specific T cells using MHC Dextramer® are published in high impact Nature and Science journals.
This is testament to the importance of a detailed understanding of the cellular immune response.
Studies utilizing MHC Dextramer® technology significantly advance cancer research, infectious disease understanding, vaccine development, and T cell therapy by enabling highly sensitive and specific detection of antigen-specific T cells.
In this blog, we highlight some of the most impactful papers from 2025 which use MHC Dextramer®.
Area |
Featured Publication |
Summary |
Species |
Sample Type |
MHC Dextramer® used in study |
Cancer |
Glioblastoma-instructed astrocytes suppress tumour-specific T cell immunity.
Akl et al. Nature 2025. https://www.nature.com/articles/s41586-025-08997-x
|
Glioblastoma is an aggressive brain cancer with a highly immunosuppressive tumor microenvironment (TME) that limits therapeutic response. This study identified a subset of astrocytes in the glioblastoma TME that induce T cell apoptosis through tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). MHC Dextramer® was used for detection of HSV-1-specific CD8+ T cells by flow cytometry.
|
Mouse |
CNS-infiltrating T cells |
H-2Kb SSIEFARL (JD02670) |
Infectious Disease |
Intragenic viral silencer element regulates HTLV-1 latency via RUNX complex recruitment.
Sugata et al. Nature Microbiology 2025. https://www.nature.com/articles/s41564-025-02006-7
|
This study explores the role of an intragenic viral silencer element in regulating latency in Human T cell leukemia virus type 1 (HTLV-1) and its interaction with the RUNX transcription factor complex. The study found that HTLV-1 latency is maintained by transcriptional suppression from its 5′ LTR. ATAC-seq revealed open chromatin regions linking the RUNX1 silencer complex to reduced transcription. Mutations in RUNX1 increased p19 production, underscoring its critical role in latency maintenance. Overall, these findings may inform therapeutic strategies for retroviral infections. |
Human |
T cells |
HLA-A24–Tax301-309 |
T cell therapy |
SEED-Selection enables high-efficiency enrichment of primary T cells edited at multiple loci.
Chang et al. Nature Biotechnology 2025. https://www.nature.com/articles/s41587-024-02531-6
|
This research paper describes the development of a novel method called "SEED-Selection" to efficiently enrich for primary T cells edited at multiple loci. SEED integration disrupts the expression of target surface proteins, allowing edited cells to be negatively selected using immunomagnetic depletion. This enabled the isolation of up to 98% pure populations of cells with individual modifications and up to 90% pure populations with 3 knock-ins and 3 knockouts. Overall, SEED-Selection provides a versatile, drug-free method to isolate highly pure populations of gene-edited T cells, which could enable the development of more advanced cell therapies. |
Human |
Edited primary human T cells |
NY-ESO-1 (SLLMWITQV) WB03247.
MART-1 (ELAGIGILTV) WB02162. |
T cell therapy |
Autologous T cell therapy for PRAME+ advanced solid tumors in HLA-A*02+ patients: a phase 1 trial.
Wermke et al. Nature Medicine 2025. https://www.nature.com/articles/s41591-025-03650-6
|
This study evaluated the safety and efficacy of IMA203, an autologous T cell receptor (TCR)-engineered T cell therapy targeting the tumor antigen PRAME, in patients with recurrent and/or refractory solid tumors. In 40 treated patients, the overall response rate (unconfirmed or confirmed) was 52.5%, with a confirmed response rate of 28.9% and a median duration of response of 4.4 months across multiple tumor types. MHC Dextramer® was used to characterize the drug product and provide key critical quality attribute data from IMA203 infused patients (n=40). %Dex: dextramer-positive out of CD3+CD8+ T cells. |
Human |
TCR-T cell therapy drug product |
HLA-A*02, PRAME |
Cancer vaccines |
Endogenous viral elements constitute a complementary source of antigens for personalized cancer vaccines.
Garde et al. Nature Vaccines 2025. https://www.nature.com/articles/s41541-025-01107-y
|
The research paper explored Endogenous Retroviral Elements (EVEs) as tumor antigens for personalized cancer vaccines (PCVs) using the ObsERV computational pipeline. EVE expression in healthy tissues and solid cancer biopsies was analyzed, and ObsERV predicted EVE-derived peptides presented on tumor MHC molecules. Preclinical tests in mice showed that ObsERV-selected EVE epitopes triggered strong CD4+ and CD8+ T-cell responses and offered tumor protection. The study confirmed that EVE-derived peptides were processed and presented by MHC on tumor surfaces and could be accurately predicted in silico. It highlighted EVEs as a valuable antigen source for PCVs, particularly in cancers with low mutational burden, illustrating how ObsERV aids in designing vaccines targeting EVE epitopes. Overall, the study underscores the potential of Dextramer® to enhance the effectiveness of personalized cancer vaccines using EVE-derived antigens. MHC Dextramer® was used to quanitfy AH1-directed CD8+ T-cell recognition in the blood of the vaccinated mice. |
Mouse (CT26) |
Blood |
H2-Ld AH1 (SPSYVYHQF) JG3294 |
Cancer vaccines |
Repurposing anti-viral subunit and mRNA vaccines T cell immunity for intratumoral immunotherapy against solid tumors.
Sethi et al., Nature Vaccines 2025. https://www.nature.com/articles/s41541-025-01131-y
|
This research paper investigates the intratumoral delivery of licensed viral vaccines (Shingrix, Gardasil-9, and Spikevax) in prevaccinated mice using the murine tumor model TC-1 expressing HPV16 oncogenes E6 and E7. These results demonstrate the potential of licensed viral vaccines as platforms for intratumoral immunotherapy, either alone or combined with vaccine-or tumor-derived peptide epitopes. MHC Dextramer® were used to quantify circulating CD8+ T cells directed against the tumor-specific viral oncoprotein E7 and L1. |
Mouse (C57BL/6) |
Single cell suspensions |
H2-Db/E749-57
H2-Db/L1165-173 |
Cancer vaccines |
TEIPP-vaccination in checkpoint-resistant non-small cell lung cancer: a first-in-human phase I/II dose-escalation study.
Emmers et al., Nature Communications 2025. https://www.nature.com/articles/s41467-025-60281-8
|
A phase I/II dose-escalation study assessed the safety, tolerability, and immunogenicity of TEIPP24, a synthetic peptide vaccine targeting LRPAP1 in HLA-A*02:01-positive non-small cell lung cancer (NSCLC) patients who progressed post-checkpoint blockade. TEIPP24 was well tolerated in 26 patients, with no dose-limiting toxicities. LRPAP1-specific CD8+ T cells were found in 83% of evaluable patients; 62% showed CD4+ T cell responses. |
Human |
PBMCs |
HLA-A*0201 LRPAP121-30V and wild-type LRPAP121-30S, FluM158-66 (GILGFVFTL) and CMV pp65495-503 (NLVPMVATV) |
Cancer vaccines |
A cowpea mosaic virus adjuvant conjugated to liposomes loaded with tumor cell lysates as an ovarian cancer vaccine.
Zhao et al., Nature Communications 2025. https://www.nature.com/articles/s41467-025-60239-w
|
The research paper presents a novel nanotechnology-based personalized cancer vaccine aimed at preventing recurrent ovarian cancer during the remission stage following surgery and chemotherapy. The vaccine utilizes autologous tumor cell lysates (TCL) rich in neoantigens harvested from patients' tumor tissues. In this proof of concept study in mice, the TCL is encapsulated in liposomes (TCL-Lip) and coupled with cowpea mosaic virus (CPMV), which serves as an effective adjuvant to enhance immune response. Utilizing ovalbumin (OVA) as a model antigen, the OVA-Lip-CPMV vaccine effectively protected mice from post-surgical lung metastasis of B16F10-OVA tumors. MHC Dextramer® was used to quantify the number of antigen-specific T cells that could recognize the tumor antigen. |
Mouse (C57BL/6J) |
i.p. washes and spleen |
H-2 Kb SIINFEKL (JD02163) |
Infectious disease vaccines |
Combined immunization with SARS-CoV-2 spike and SARS-CoV nucleocapsid protects K18-hACE2 mice but increases lung pathology.
Kim et al., Nature Vaccines 2025. https://www.nature.com/articles/s41541-025-01085-1
|
The study explored the immunogenicity and protective efficacy of adenoviral constructs expressing the nucleocapsid (N) proteins of SARS-CoV (N1) and SARS-CoV-2 (N2) in K18-hACE2 mice. Utilizing a prime-boost immunization schedule with intramuscular inoculations, the research found that both Ad-N1 and Ad-N2 induced significant T cell and antibody responses. Mice immunized with these constructs showed reduced viral replication and lung pathology following SARS-CoV-2 challenge, compared to controls.
|
Mouse (K18-hACE2) |
Spleenocytes |
N220 |
Infectious disease vaccines |
Efficacy of emergency maternal MVA-ZIKV vaccination in a rapid challenge model of lethal Zika infection.
Volz et al., Nature Vaccines 2025. https://www.nature.com/articles/s41541-025-01094-0
|
This research focused on evaluating the protective efficacy of a recombinant Modified Vaccinia Ankara (MVA) vaccine expressing the ZIKV prM and E proteins (MVA-ZIKV) against Zika virus infection in a mouse model. A single dose of the MVA-ZIKV vaccine administered 2 days before lethal ZIKV challenge fully protected both non-pregnant and pregnant IFNAR-/- mice, preventing viral replication and disease. Early protection was associated with rapid induction of ZIKV-specific CD8+ T cell responses, as depletion of CD8+ T cells abrogated the protective efficacy of the vaccine. In contrast, neutralizing antibody (nAb) responses were induced later and did not contribute significantly to the rapid protection observed. These results suggest that the MVA-ZIKV vaccine can provide rapid, T cell-mediated protection against lethal ZIKV infection, including in the context of pregnancy, making it a promising candidate for emergency vaccination during outbreaks. |
Mouse (C57BL/6) |
Blood and spleenocytes |
IGVSNRDFV-Kb |
Characterizing and Monitoring Antigen-Specific T and B Cells in Cancer Vaccine Development.
Explore how Dextramer® technology can uncover antigen-specific immune responses and transform vaccine development.
Explore how Dextramer® reagents support the development and manufacturing of effective cellular immunotherapies.
Sensitively quantify target immune cells by flow cytometry with a strong signal-to-noise ratio.
Fill in the form and one of our dedicated specialists will get in touch with you shortly.