Yusuke Nakamura’s research while affiliated with National Institute of Biomedical Innovation, Health and Nutrition and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (352)


Automated and Decentralized Genomic Profiling of Plasma Cell-Free DNA for Identification of Targetable and Resistance Alterations in Advanced Solid Tumors
  • Article

May 2025

·

6 Reads

Clinical Chemistry

·

Masumi Otaki

·

·

[...]

·

Background Existing circulating cell-free DNA (cfDNA) assays are primarily centralized, requiring specialized sample handling and transportation. Implementing a flexible, decentralized sequencing system at point of care, with minimal technical oversight, can enhance turnaround times and patient access to genomic profiling. In this study, we aimed to evaluate the clinical feasibility of an automated and decentralized cfDNA next-generation sequencing (NGS) assay for identifying actionable alterations in advanced solid tumors. Methods Genomic profiling of plasma cfDNA from 298 patients with advanced solid tumors was conducted using an automated NGS assay. We assessed concordance of tumor mutations detected in plasma cfDNA and patient-matched tumor tissues analyzed by an FDA-approved assay, investigating clinical factors influencing circulating tumor DNA (ctDNA) aberration (mut-ctDNA) detection sensitivity. Results Sequencing success rates for cfDNA genomic profiling was significantly higher than archived tumor tissue (99% vs 96%). Mut-ctDNA detection rates ranged from 20% to 67% across different solid tumors. Targetable or resistance alterations were found in 18% of the patients. About 72% of the patients showed concordant alterations from tissue and plasma. The level of concordance was associated with the cancer type, tumor burden, and metastatic location. Notably, 63 plasma-only alterations were identified in 18% of patients and were more frequently observed in those with prior targeted treatments (24%) compared to chemotherapy (10%). Conclusions This study underscores the clinical feasibility of an automated, decentralized cfDNA genomic profiling approach. It emphasizes the importance of considering confounding clinical factors when selecting plasma- or tissue-based profiling assay. Such an approach holds promise for enhancing patient access to timely genomic profiling and targeted therapy selection.


Figure 1 Induction of mp68 neoantigen expression in a progressively growing cancer causes infiltration of mp68-specific T cells that are unable to suppress tumor growth. (A) 8101 cancer cells harbor a serine (S) to phenylalanine (F) exchange in position 551 of the p68 protein that is caused by a point mutation (cytosine to thymidine transition). Electropherograms show the presence or absence of the point mutation (red) in 8101 (mp68 + , heterozygous) and 8101PRO (mp68 − ). (B) 8101PRO-derived 4E9 cancer cells are mp68 − (4E9 OFF ) and express the mp68-Thy1.1 fusion gene on incubation with doxycycline (4E9 ON ). Histograms show surface expression of Thy1.1 in indicated cells as determined by flow cytometry. (C) 1D9 TCR-Ts secrete IFN-γ when incubated with 4E9 ON but not 4E9 OFF cancer cells. Depicted values are technical replicates of an experiment that was repeated nine times showing comparable results. P/I, phorbol-12-myristate-13-acetate/ionomycin. (D) Induction of mp68 expression cannot prevent progression of 4E9 ON tumors in C57BL/6 mice. Data are compiled from two independent experiments and plotted on separate x-axes according to growth kinetics. Curves are shown for tumors isolated 3 weeks (upper left, mouse T1 (red) and T2 (cyan) are highlighted), 5 weeks (upper right, T3 in orange), and 14.5 weeks (lower left, T4 in purple) after inducing mp68 expression 21 days after inoculation. Other mice in the treatment group are shown as open circles. Growth of 4E9 OFF tumors is shown as control (lower right, gray squares). (E-F) 4E9 ON -infiltrating CD8 T cells show surface markers of exhaustion (E) and contain a fraction that binds mp68-tetramers (F). CD3 + CD8 + TILs from reisolated 4E9 ON tumors were analyzed by flow cytometry to assess expression of CD39 and PD-1 (E) and binding of mp68-tetramers (F) using material from three to nine animals, respectively. Mice T1-T4 are color-coded. Additional mice are shown as open circles. Representative FACS plots show TILs of mouse T3 (E) and T1 (F) gated on CD3 + CD8 + cells (left). A comparison with corresponding splenocytes (E) and TILs from 4E9 OFF tumors (F) is shown as a scatter plot (right). The number (n) indicates the sample size for each graph. P values result from paired t-test (E) or unpaired t-test with Welch's correction (F). FACS, Fluorescence-Activated Cell Sorting; IFN, interferon; P/I, phorbol-12-myristate-13-acetate/ionomycin; PD-1, Programmed Death-1; TIL, tumor-infiltrating lymphocyte. Protected by copyright, including for uses related to text and data mining, AI training, and similar technologies.
Figure 2 T cells that bind mp68-tetramers isolated from TILs of 4E9 ON tumors or immunized mice comprise a diverse repertoire of TCRs. (A-B) The frequency of TRAV-TRAJ (A) and TRBV-TRBJ (B) combinations found in TCR genes of TILs isolated from the 4E9 ON tumor of mouse T1 is shown as a heat map. Combinations with a frequency above 10% in the repertoire are indicated in red and their CDR3 usage is shown. (C-D) TCR sequences of single CD8 + T cells binding the mp68-tetramer were determined and the relative abundance of T-cell clonotypes is indicated in pie charts. T cells were isolated from 4E9 ON tumors of mice T1 (red), T2 (cyan), T3 (orange), and T4 (purple) (C) or from spleens of 4E9 ON -immunized mice V1 (blue) and V2 (green) (D). Expanded T-cell clonotypes are color-coded and the fraction of non-expanded clonotypes, which are represented in ≤10 (C) or ≤5 wells (C-D), are indicated in gray (C) and white (C-D). The number of clonotypes contained in the non-expanded fraction is given. TCR, T-cell receptor; TIL, tumor-infiltrating lymphocyte; TRAJ, TCR-α chain joining region; TRAV, TCR-α chain variable region; TRBJ, TCR-β chain joining region; TRBV, TCR-β chain variable region. Protected by copyright, including for uses related to text and data mining, AI training, and similar technologies.
Figure 3 Less than half of the TCRs isolated from mp68-tetramer-binding TILs or T cells of immunized mice are therapeutically effective. (A) The therapeutic quality of TCR-Ts was determined by TCR-therapy in mice bearing large established 8101PRO-mp68 tumors and is represented by their ability to destroy tumors. Shown are tumor growth curves of individual mice pooled from two to three independent experiments. The number of treated mice in each group is shown in the upper right of each graph. Injection of 8101PRO-mp68 cancer cells is indicated with an arrow. 2-3 weeks after cancer cell injection, TCR-Ts (colorcoded for each originating mouse and TCR clonotype) were adoptively transferred (day 0, dashed arrows). Mice receiving unmodified T cells (UT) are shown as control. The average tumor size at treatment start was 182±103 mm 3 . The number (n) indicates the sample size for each graph. (B) Loss of Thy1.1 expression on reisolated cancer cells of relapsing 8101PRO-mp68 tumors is indicative of treatment success. Reisolated cancer cells were adapted to culture and the percentage of Thy1.1 + cells was determined by flow cytometry. Lack of Thy1.1 + cells indicates the selection of mp68 − escape variants by the respective TCR-Ts. Each data point represents one tumor. Mean and SEM are shown. TCR-Ts, T-cell receptor-modified T cells; TIL, tumor-infiltrating lymphocyte.
Figure 4 Long-term, but not short-term in vitro assays are accurate predictors of in vivo efficacy of mp68-specific TCRTs. TCR-Ts expressing mp68-specific TCRs derived from TILs of 4E9 ON tumors or immunized mice were incubated with splenocytes loaded with graded amounts of mp68 peptide (A), 8101 cancer cells with ectopic (B, left) or native expression of mp68 (B, right and C). TCRs were classified as effective or failing based on their in vivo efficacy (figure 3) and are represented as single dots showing an average of three to eight independent experiments (detailed in online supplemental figure 5). (A) EC 50 is the concentration of mp68 peptide required to elicit half-maximal IFN-γ release by TCR-Ts when loaded on splenocytes in 24 hours co-cultures. (B) IFN-γ release by TCR-Ts was determined after 24 hours co-culture with 8101PRO-mp68 (left) or 8101 cancer cells (right). (C) TCR-Ts were incubated with 8101-12-GFP cancer cells and time to tumor outgrowth (≥10% confluence) was assessed by monitoring co-cultures using Incucyte SX5. Additional 8101-12-GFP cancer cells were added to the culture every 3 days. The range of the data set for therapeutically effective TCRs is indicated in gray. Failing TCRs that fall within this range are indicated. P values result from unpaired t-tests. IFN, interferon; TCR-Ts, T-cell receptor-modified T cells; TIL, tumorinfiltrating lymphocyte. Protected by copyright, including for uses related to text and data mining, AI training, and similar technologies.
Figure 5 Patient-derived TCRs that are specific for the neoantigen CDK4 R24L display optimal in vivo efficacy. TCR-Ts were generated using TCRs derived from patients (P1 (red), 14/35 (cyan)), third party donors (H1-1 (blue), H2-1 (green), H2-2 (light green)) or humanized mice (M1, magenta). (A) Human TCR-Ts were incubated with WM-902B+A2 cancer cells and time to tumor outgrowth (≥15% confluence) was assessed by monitoring co-cultures using Incucyte SX5. Additional WM-902B+A2 cancer cells were added to the culture every 3 days. TCRs are color-coded and symbols refer to six independent experiments pooled in the depicted scatter plot. The p value results from unpaired t-test comparing TCRs predicted to be therapeutically effective (P1, 14/35, H1-1, H2-1, (M1) or failing (H2-2). (B) Mouse TCR-Ts were generated to assess TCR quality in vivo by adoptive therapy of TNA2 mice bearing large established MC703-ALD tumors. Shown are tumor growth curves pooled from two to three independent experiments. The number of treated mice in each group is shown in the upper right of each graph (n). The number of mice that rejected the tumor on transfer of TCR-Ts is stated below (r). Injection of MC703-ALD cancer cells is indicated with an arrow. The time of injecting TCR-Ts (color-coded for each TCR) is shown with a dashed arrow and was between 3 and 4 weeks after cancer cell inoculation. Mice receiving no T cells (None) are shown as control. The average tumor size at treatment start was 219±218 mm 3 . TCR-Ts, T-cell receptor-modified T cells. Protected by copyright, including for uses related to text and data mining, AI training, and similar technologies.

+1

Selection of therapeutically effective T-cell receptors from the diverse tumor-bearing repertoire
  • Article
  • Full-text available

May 2025

·

33 Reads

Background The development of T-cell receptor (TCR)-based T-cell therapies is hampered by the difficulties in identifying therapeutically effective tumor-specific TCRs from the natural repertoire of a patient’s cancer-specific T cells. Methods Here, we mimic experimentally near-patient conditions to analyze the T-cell repertoire in euthymic tumor-bearing mice responding to the H-2K b -presented neoantigen p68 S551F (mp68). We temporarily separated the time point of mp68 expression from that of cancer cell transplantation to exclude the influence of injection-induced inflammation on T-cell priming. Thus, the mp68-specific T-cell response could only develop after the acute inflammatory phase had subsided. Results We found that mp68-specific TCRs isolated from either tumor-infiltrating T cells or spleens of mice immunized with mp68-expressing cancer cells are diverse and not inherently therapeutic when introduced into peripheral T cells and used for adoptive therapy of established tumors. While measuring short-term T-cell responses in vitro was unreliable for some TCRs in predicting their therapeutic failure, assessing the persistence of cancer cell destruction by TCR-modified T cells in long-term cultures accurately predicted therapeutic outcomes. A tumor-derived TCR with optimal function was also correctly identified with this approach when analyzing human TCRs that recognize the HLA-A2-presented neoantigen CDK4 R24L . Conclusions We show that a neoantigen-directed T-cell response in tumor-bearing hosts comprises a diverse repertoire. Infiltration and expansion of certain T-cell clonotypes in the tumor do not necessarily correlate with therapeutic efficacy of their TCRs in adoptive therapy. We propose that analysis of persistent rather than immediate responses of TCR-modified T cells in vitro serves as a reliable parameter to identify TCRs that are therapeutically effective in vivo.

Download

Abstract 3409: T cell receptors and antibodies targeting shared indel-derived neoantigens

April 2025

Cancer Research

T cells play a pivotal role in cancer immunotherapy by recognizing neoantigens presented on HLA molecules, which are specifically expressed by cancer cells. While neoantigens are generally unique to individual cancers, shared antigens that are common in a subset of cancer patients, represent promising immunotherapeutic targets. Since frameshift insertions or deletions (indels) are reported to generate highly immunogenic neoantigens through novel open reading frames. However, few studies have explored shared neoantigens from frameshift indels, which are shred among multiple cancer patients. In this study, we analyzed frameshift neoantigens from frameshift indels in TCGA exome sequencing data, identifying 760 recurrent frameshift mutation clusters (FSCs) that shared frameshifted open reading frames and premature stop codons. Among these, we focused on two FSCs in the APC gene, which generate potential neoantigens (AS3 and AS10) presented the HLA-A24 molecule. We successfully identified the T cell receptor (TCR) sequences for APC frameshift neoantigens from healthy donor’s blood. Genetically engineered T cells expressing these TCRs demonstrated recognition and cytotoxic activity against target cells presenting these APC frameshift neoantigens. Additionally, we explored the development of an antibody targeting a complex formed by the APC shared neoantigen AS10 and the HLA-A24 molecule. Using phage display library screening, we isolated single-chain variable fragments (scFvs) that specifically recognized the AS10-HLA-A24 complex. A bispecific antibody (BsAb) was then designed to connect T cells via an anti-CD3 scFv to the AS10 neoantigen on HLA-A24. We found that the AS10 BsAb specifically recognized both the AS10-HLA-A24 complex and CD3 protein, and showed significant IFN-γ release and cytotoxicity against target cells expressing frameshift neoantigens. By focusing on recurrent frameshift mutations and shared neoantigens, this study provides compelling evidence for the development of more effective and broadly applicable immunotherapeutic strategies. Citation Format Kazuma Kiyotani, Peng Zhao, Xiaojing Wu, Yusuke Nakamura. T cell receptors and antibodies targeting shared indel-derived neoantigens [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2025; Part 1 (Regular Abstracts); 2025 Apr 25-30; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2025;85(8_Suppl_1):Abstract nr 3409.


Neoantigen peptide-pulsed dendritic cell vaccine therapy after surgical treatment of pancreatic cancer: a retrospective study

April 2025

·

31 Reads

Introduction Pancreatic cancer shows very poor prognosis and high resistance to conventional standard chemotherapy and immunotherapy; therefore, the development of new breakthrough therapies is highly desirable. Method We retrospectively evaluated the safety and efficacy of neoantigen peptide-pulsed dendritic cell (Neo-P DC) vaccine therapy after surgical treatment of pancreatic cancer. Result The result showed induction of neoantigen-specific T cells in 13 (81.3%) of the 16 patients who received Neo-P DC vaccines. In survival analysis of the nine patients who received Neo-P DC vaccines after recurrence, longer overall survival was observed in patients with neoantigen-specific T cell induction than those without T cell induction. Notably, only one of the seven patients who received Neo-P DC vaccines as adjuvant setting developed recurrence, and no patient died during median follow-up 61 months after surgery (range, 25-70 months). Furthermore, TCR repertoire analyses were performed in a case treated with Neo-P DC vaccine combined with long and short peptides, and one significantly dominant clone induced by the long peptide was detected among CD4⁺ T cell populations. Discussion The present study suggests the feasibility and efficacy of Neo-P DC vaccine therapy after surgical treatment of pancreatic cancer in both postoperative recurrence cases and adjuvant setting. A case analysis suggests the importance of combination with long peptides targeting CD4⁺ T cell.



Fig. 1. Results of meta-analyses for genome-wide association studies for serum liver enzyme levels in the Ryukyu population. Manhattan plots (left) and Quantile-Quantile plots (right) of GWAS for serum (a) ALT, (b) AST, and (c) GGT levels. Red lines indicate the threshold for the genome-wide significance level (P = 5 × 10 −8 ). Green plots correspond to variants that attained genome-wide significance. A novel locus is shown by red letters.
Fig. 2. A regional plot for association of HMMR/MMMR-AS1 locus with serum ALT values in the Ryukyu population. The lead variant is shown as purple diamond. The other variants are colored according to the extent of LD estimated in 3256 Japanese WGS samples from the BBJ and 2504 individuals from the 1KGP phase 3.
A variant in HMMR/HMMR-AS1 is associated with serum alanine aminotransferase levels in the Ryukyu population

February 2025

·

23 Reads

The Ryukyu archipelago is located southwest of the Japanese islands, and people originally from this region, the Ryukyu population, have a unique genetic background distinct from that of other populations, including people from mainland Japan. However, few genetic studies have focused on the Ryukyu population. In this study, we performed genome-wide association studies (GWAS) on the serum levels of alanine aminotransferase (ALT, n = 15,224), aspartate aminotransferase (AST, n = 15,203), and gamma-glutamyl transferase (GGT, n = 14,496) in the Ryukyu population. We found 13 loci with a genome-wide significant association (P < 5 × 10-8), three for ALT, four for AST, and six for GGT, including one novel locus associated with ALT: rs117595134-A in HMMR/HMMR-AS1, ß = - 0.131, standard error = 0.024, P = 4.90 × 10-8. Rs117595134-A is common in the Japanese population but is not observed in other ethnic populations in the 1000 genomes database. Additionally, 77 of 80 loci derived from Korean GWAS and 541 of 716 loci from European GWAS showed the same directions of effect (P = 1.41 × 10-19, P = 2.50 × 10-44, binomial test), indicating that most of susceptibility loci are shared between the Ryukyu population and other ethnic populations.



Retrospective Analysis of HLA Class II-Restricted Neoantigen Peptide-Pulsed Dendritic Cell Vaccine for Breast Cancer

December 2024

·

19 Reads

·

1 Citation

Background/Objectives: Neoantigens have attracted attention as ideal therapeutic targets for anti-tumour immunotherapy because the T cells that respond to neoantigens are not affected by central immune tolerance. Recent findings have revealed that the activation of CD4-positive T cells plays a central role in antitumor immunity, and thus targeting human leukocyte antigen (HLA) class II-restricted neoantigens, which are targets of CD4-positive T cells, is of significance. However, there are very few detailed reports of neoantigen vaccine therapies that use an HLA class II-restricted long peptide. In the present study, we retrospectively analysed the ability of HLA class II-restricted neoantigen-pulsed dendritic cell vaccines to induce immune response in five breast cancer patients. Methods: We performed whole exome and RNA sequencing of breast cancer tissues and neoantigen prediction using an in silico pipeline. We then administered dendritic cells pulsed with synthesized an HLA class II-restricted long peptide containing an epitope with high affinity to HLA class I in the lymph node. Results: ELISPOT analysis confirmed that a T-cell response specific for the HLA class II-restricted neoantigen was induced in all cases. TCR repertoire analysis of peripheral blood mononuclear cells before and after treatment in three patients showed increases of specific T-cell clones in two of the three patients. Importantly, no recurrence was observed in all patients. Conclusions: Our analysis demonstrated the immunological efficacy of the HLA class II-restricted neoantigen peptide dendritic cell vaccine against breast cancer and provides useful information for the development of neoantigen vaccine therapy for breast cancer.


403 A four-cell type interaction between CD4 + T cells, CD8 + T cells, stromal and cancer cells as a potential mechanism for cancer eradication when one CD4TCR is combined with one CD8TCR

Background Even when tumors are infiltrated by T cells, progressive growth eventually kills the host. The use of immune checkpoint inhibitors (ICI) or adoptive transfer of tumor-infiltrating lymphocytes (TILs) have led to advanced immunotherapies which can be successful in some patients with certain types of cancers. Even though these immunotherapies have revolutionized modern cancer treatments, achieving tumor eradication remains a major challenge.1 2 When ICI and TIL therapies are effective, cancer-specific neoantigens seem to be the important immunological targets.3 4 An alternative approach isolates T cell receptors (TCRs) from neoantigen-specific T cells for adoptive transfer of TCR-engineered peripheral blood T cells (TCR-T cell therapy).5 6 We wanted to determine the requirements needed for TCR-T cell therapy to eradicate established solid tumors. Methods We used the UV-induced, primary and autochthonous cancer cell line 6132A and its well-studied neoantigen mL9.⁷ We isolated a TCR from mL9-specific CD4⁺ T cells (CD4TCR) and another TCR from 6132A-specific CD8⁺ T cells (CD8TCR) generated from the spleen of 6132A–immune mice. We cloned the CD4TCR and the CD8TCR into retroviral vectors and evaluated in vitro and in vivo responses of TCR-engineered CD4⁺ and CD8⁺ T cells. Results The CD4TCR recognized the immunosuppressive tumor stroma and caused destruction followed by long-term growth arrest of tumors formed by the MHC class II negative 6132A cancer cells. However, the CD4TCR failed to eradicate the tumors completely. Remarkably, the CD8TCR was ineffective on its own, yet large unmanipulated solid tumors were eradicated when the CD8TCR was combined with the CD4TCR. Thus, direct cancer cell recognition by the CD8TCR together with stromal recognition by the CD4TCR was needed for effective therapy. As underlying mechanism, in vitro analyses suggest a four-cell type interaction between CD4⁺ and CD8⁺ T cells together with stromal APCs and cancer cells leading to effective elimination of cancer cells.⁸ Conclusions CD8TCRs eradicated long-established solid tumors expressing unmanipulated mutant neoantigens only when combined with a CD4TCR. In vitro experiments suggest a four-cell interaction being essential for cancer cell eradication. References • Haslam A, V Prasad. Estimation of the percentage of us patients with cancer who are eligible for and respond to checkpoint inhibitor immunotherapy drugs. JAMA Netw Open, 2019; 2 (5):e192535. • Rosenberg SA, NP Restifo. Adoptive cell transfer as personalized immunotherapy for human cancer. Science, 2015; 348 (6230):62–8. • Tran E, PF Robbins, SA Rosenberg. ‘Final common pathway’ of human cancer immunotherapy: targeting random somatic mutations. Nat Immunol, 2017; 18 (3):255–262. • Gubin MM, et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature, 2014; 515 (7528):577–81. • Kim SP, et al. Adoptive cellular therapy with autologous tumor-infiltrating lymphocytes and T-cell receptor-engineered T cells targeting common p53 neoantigens in human solid tumors. Cancer Immunol Res, 2022:OF1-OF15. • Leidner R, et al. Neoantigen T-cell receptor gene therapy in pancreatic cancer. N Engl J Med, 2022; 386 (22):2112–2119. • Monach PA, et al. A unique tumor antigen produced by a single amino acid substitution. Immunity, 1995; 2 (1):45–59. • Wolf SP, et al. One CD4+TCR and One CD8+TCR targeting autochthonous neoantigens are essential and sufficient for tumor eradication. Clin Cancer Res 2024; 30 (8):1642–1654.



Citations (59)


... In addition, by whole exome and RNA sequencing, the researchers synthesized HLA class II-restricted long peptides containing epitopes with high affinity for HLA class I and prepared DC vaccines. Subsequently, specific T cell clones were generated in two of the three vaccinated patients, and no relapse was observed in all three patients (Morisaki et al., 2024). ...

Reference:

Promising future of breast cancer vaccine asking for multidisciplinary collaboration: a literature review
Retrospective Analysis of HLA Class II-Restricted Neoantigen Peptide-Pulsed Dendritic Cell Vaccine for Breast Cancer

... Instead, in the peripheral blood mononuclear cells (PBMCs) of MS patients, the IGHV3-23 and IGHV3-74 subfamilies are overrepresented when compared to PBMCs from OND patients. Nonetheless, this overrepresentation is insufficient to allow for statistically precise biomarker development [32,34]. Using mass spectrometry proteomics, we discovered that the IgG heavy chains IGHV1-3, IGHV3-43, and IGHV3-49 were present in the IgG aggregates for relapsing-remitting MS (RRMS) and secondary progressive MS (SPMS) but not in controls [35]. ...

Comparative analysis of the B cell receptor repertoire during relapse and remission in patients with multiple sclerosis
  • Citing Article
  • November 2024

Clinical Immunology

... We previously showed that convergent TCR recombination can be used as an indicator for the therapeutic efficacy of tumorderived TCRs isolated from CD4 T cells. 13 In the current study, we pursued a functional approach to predict the therapeutic efficacy of tumor-derived TCRs experimentally. We developed a model system with inducible expression of the H-2K bpresented NeoAg p68 S551F (mp68) 3 that mimics obstacles faced by T cells in naturally growing tumors, such as suboptimal priming conditions due to lack of acute inflammation. ...

CD4+ T cells with convergent TCR recombination reprogram stroma and halt tumor progression in adoptive therapy
  • Citing Article
  • September 2024

Science Immunology

... One key challenge is the inherent variability in immune responses among individuals. Immune cell composition and function can vary greatly depending on genetic, environmental, and lifestyle factors, making it difficult to generalize findings across diverse populations [97]. This variability introduces challenges in interpreting study results, as immune responses may not be uniform across individuals or even within the same individual at different stages of pregnancy [98]. ...

Common and rare genetic variants predisposing females to unexplained recurrent pregnancy loss

... They have gained increasing attention in the regulation of immune tolerance and in the management of autoimmune diseases. Research has extensively explored their roles and applications in diseases such as systemic lupus erythematosus, non-alcoholic fatty liver disease, and various cancers (2)(3)(4)(5)(6). ...

Characterization of double-negative T cells in colorectal cancers and their corresponding lymph nodes

... AIs as 'second readers'-by which we mean that they assist a first-reader radiologist, with the aim of improving overall diagnostic performance. This possibility of using AIs as second-readers has attracted significant scholarly interest (Bruno, 2019;Rodríguez-Ruiz et al., 2020;McKinney et al., 2020;Sharma et al., 2021;Yoo et al., 2021;Ng et al., 2023a, b;Nakai et al., 2024;Gefter et al., 2023b) but to our knowledge has not received careful epistemological attention. This paper aims to fill this gap. ...

Artificial intelligence as a second reader for screening mammography
  • Citing Article
  • May 2024

Radiology Advances

... When comparing the genes selected by DGE and Met-tleRNASeq, no overlaps were found in Skin, while only three genes overlapped in Bone: Igha, Ly6c2, and Rnf213, linked to processes like immune response, angiogenesis and cell proliferation, migration, and invasion Ye et al., 2023;Zhang et al., 2023;Chiablaem et al., 2024;Dechant and Valerius, 2001;Upadhyay, 2019). Notably, in Skin both approaches highlighted related pathways (AMPK and cAMP), connected through the Ataxia Telangiectasia Mutated (ATM) protein, and involved in radiation-induced DNA damage responses and cell death (Li et al., 2017;Cho et al., 2014). ...

Identification of RNF213 as a Potential Suppressor of Local Invasion in Intrahepatic Cholangiocarcinoma

Laboratory Investigation

... 5 7 We have shown that targeting multiple NeoAgs with TCR-Ts can reduce the risk of therapy-induced tumor escape. 8 However, even TCR therapy using three NeoAg-specific TCRs derived from CD8 T cells was rarely effective when targeting autochthonous NeoAgs because the enormous heterogeneity of established tumors allowed antigen-negative cancer cells to escape. 8 In contrast, we further showed that two TCRs, one isolated from CD4 T cells (reactive against a cancer-derived NeoAg presented on stromal cells) and one from CD8 T cells (targeting a NeoAg on cancer cells), are essential and sufficient to eliminate large heterogeneous cancers in mice. ...

One CD4+TCR and One CD8+TCR Targeting Autochthonous Neoantigens Are Essential and Sufficient for Tumor Eradication
  • Citing Article
  • January 2024

Clinical Cancer Research

... In the hybrid neoantigen-pulsed DC vaccine trial, researchers induced strong T-cell reactions in four patients. 69 Such hybrid/combination with DC vaccines could be a future direction. ...

Immunological analysis of hybrid neoantigen peptide encompassing class I/II neoepitope-pulsed dendritic cell vaccine

... Within the context of HPV-negative HNSCC, our group showed that SMYD3 functions as a master epigenetic regulator of immune-related genes and is significantly overexpressed in HPV-negative HNSCC compared to normal and dysplastic buccal epithelium 12 . Specifically, we demonstrated that SMYD3 depletion induced upregulation of type I IFN response and antigen presentation machinery genes in HPV-negative HNSCC cells. ...

SMYD3 represses tumor-intrinsic interferon response in HPV-negative squamous cell carcinoma of the head and neck

Cell Reports