2024-01-20 2024, Volume 14 Issue 1

  • Select all
  • LETTER TO THE JOURNAL
    Chong Wei , Wei Wang , Wanying Li , Yan Zhang , Danqing Zhao , Wei Zhang , Daobin Zhou
    2024, 14(1): e1491. https://doi.org/10.1002/ctm2.1491
  • LETTER TO THE JOURNAL
    Yan Dang , Shutian Zhang
    2024, 14(1): e1496. https://doi.org/10.1002/ctm2.1496
  • REVIEW
    David Pellerin , Matt C. Danzi , Mathilde Renaud , Henry Houlden , Matthis Synofzik , Stephan Zuchner , Bernard Brais
    2024, 14(1): e1504. https://doi.org/10.1002/ctm2.1504

    Hereditary ataxias, especially when presenting sporadically in adulthood, present a particular diagnostic challenge owing to their great clinical and genetic heterogeneity. Currently, up to 75% of such patients remain without a genetic diagnosis. In an era of emerging disease-modifying gene-stratified therapies, the identification of causative alleles has become increasingly important. Over the past few years, the implementation of advanced bioinformatics tools and long-read sequencing has allowed the identification of a number of novel repeat expansion disorders, such as the recently described spinocerebellar ataxia 27B (SCA27B) caused by a (GAA)•(TTC) repeat expansion in intron 1 of the fibroblast growth factor 14 (FGF14) gene. SCA27B is rapidly gaining recognition as one of the most common forms of adult-onset hereditary ataxia, with several studies showing that it accounts for a substantial number (9-61%) of previously undiagnosed cases from different cohorts. First natural history studies and multiple reports have already outlined the progression and core phenotype of this novel disease, which consists of a late-onset slowly progressive pan-cerebellar syndrome that is frequently associated with cerebellar oculomotor signs, such as downbeat nystagmus, and episodic symptoms. Furthermore, preliminary studies in patients with SCA27B have shown promising symptomatic benefits of 4-aminopyridine, an already marketed drug. This review describes the current knowledge of the genetic and molecular basis, epidemiology, clinical features and prospective treatment strategies in SCA27B.

  • LETTER TO THE JOURNAL
    Yang Yuan , Xiaona Dong , Shengju Yin , Guoliang Zhang , Haiyan Zhou , Guohui Li , Yan Tang , Xiaofan Wei , Hongquan Zhang
    2024, 14(1): e1517. https://doi.org/10.1002/ctm2.1517
  • COMMENTARY
    Jingwen Wang , Conglin Xu , Zhen Zhang , Fei Xavier Chen
    2024, 14(1): e1519. https://doi.org/10.1002/ctm2.1519
  • REVIEW
    Xinxin Ren , Xiang Wang , Guowan Zheng , Shanshan Wang , Qiyue Wang , Mengnan Yuan , Tong Xu , Jiajie Xu , Ping Huang , Minghua Ge
    2024, 14(1): e1521. https://doi.org/10.1002/ctm2.1521

    Background: One-carbon (1C) metabolism is a metabolic network that plays essential roles in biological reactions. In 1C metabolism, a series of nutrients are used to fuel metabolic pathways, including nucleotide metabolism, amino acid metabolism, cellular redox defence and epigenetic maintenance. At present, 1C metabolism is considered the hallmark of cancer. The 1C units obtained from the metabolic pathways increase the proliferation rate of cancer cells. In addition, anticancer drugs, such as methotrexate, which target 1C metabolism, have long been used in the clinic. In terms of immunotherapy, 1C metabolism has been used to explore biomarkers connected with immunotherapy response and immune-related adverse events in patients.

    Methods: We collected numerous literatures to explain the roles of one-carbon metabolism in cancer immunotherapy.

    Results: In this review, we focus on the important pathways in 1C metabolism and the function of 1C metabolism enzymes in cancer immunotherapy. Then, we summarise the inhibitors acting on 1C metabolism and their potential application on cancer immunotherapy. Finally, we provide a viewpoint and conclusion regarding the opportunities and challenges of targeting 1C metabolism for cancer immunotherapy in clinical practicability in the future.

    Conclusion: Targeting one-carbon metabolism is useful for cancer immunotherapy.

  • LETTER TO THE JOURNAL
    Li Li , Jinghao Duan , Yongsheng Gao , Ying Yin , Fengchang Yang , Wenjie Tang , Xiaoyu Song , Tao Hu , Jinfeng Cui , Jinming Yu , Shuanghu Yuan
    2024, 14(1): e1522. https://doi.org/10.1002/ctm2.1522
  • RESEARCH ARTICLE
    Fanfan Xie , Wenjie Guo , Xingguo Wang , Kaixiang Zhou , Shanshan Guo , Yang Liu , Tianlei Sun , Shengjing Li , Zhiyang Xu , Qing Yuan , Huanqin Zhang , Xiwen Gu , Jinliang Xing , Shujuan Liu
    2024, 14(1): e1523. https://doi.org/10.1002/ctm2.1523

    Background: Epithelial ovarian cancer (EOC) heavily relies on oxidative phosphorylation (OXPHOS) and exhibits distinct mitochondrial metabolic reprogramming. Up to now, the evolutionary pattern of somatic mitochondrial DNA (mtDNA) mutations in EOC tissues and their potential roles in metabolic remodelling have not been systematically elucidated.

    Methods: Based on a large somatic mtDNA mutation dataset from private and public EOC cohorts (239 and 118 patients, respectively), we most comprehensively characterised the EOC-specific evolutionary pattern of mtDNA mutations and investigated its biological implication.

    Results: Mutational profiling revealed that the mitochondrial genome of EOC tissues was highly unstable compared with non-cancerous ovary tissues. Furthermore, our data indicated the delayed heteroplasmy accumulation of mtDNA control region (mtCTR) mutations and near-complete absence of mtCTR non-hypervariable segment (non-HVS) mutations in EOC tissues, which is consistent with stringent negative selection against mtCTR mutation. Additionally, we observed a bidirectional and region-specific evolutionary pattern of mtDNA coding region mutations, manifested as significant negative selection against mutations in complex V (ATP6/ATP8) and tRNA loop regions, and potential positive selection on mutations in complex III (MT-CYB). Meanwhile, EOC tissues showed higher mitochondrial biogenesis compared with non-cancerous ovary tissues. Further analysis revealed the significant association between mtDNA mutations and both mitochondrial biogenesis and overall survival of EOC patients.

    Conclusions: Our study presents a comprehensive delineation of EOC-specific evolutionary patterns of mtDNA mutations that aligned well with the specific mitochondrial metabolic remodelling, conferring novel insights into the functional roles of mtDNA mutations in EOC tumourigenesis and progression.

  • COMMENTARY
    Alessandro Cherubini , Elia Casirati , Serena Pelusi , Luca Valenti
    2024, 14(1): e1524. https://doi.org/10.1002/ctm2.1524
  • LETTER TO THE JOURNAL
    Andreas Neueder , Philipp Nitzschner , Ronja Wagner , Julia Hummel , Franziska Hoschek , Maximilian Wagner , Alshaimaa Abdelmoez , Björn von Einem , G. Bernhard Landwehrmeyer , Sarah J. Tabrizi , Michael Orth
    2024, 14(1): e1525. https://doi.org/10.1002/ctm2.1525
  • COMMENTARY
    Lin Lin , Hans Clevers
    2024, 14(1): e1526. https://doi.org/10.1002/ctm2.1526
  • LETTER TO THE JOURNAL
    Xin Huang , Lutong Wang , Haoyu Guo , Weiyue Zhang
    2024, 14(1): e1527. https://doi.org/10.1002/ctm2.1527
  • COMMENTARY
    Shanhe Yu , Shijun Chen , Jiang Zhu , Jieming Qu
    2024, 14(1): e1530. https://doi.org/10.1002/ctm2.1530
  • RESEARCH ARTICLE
    Yi Cai , Minfeng Chen , Yuchen Gong , Guyu Tang , Zhiwei Shu , Jiaxian Chen , Hengfeng Zhou , Yao He , Zhi Long , Yu Gan
    2024, 14(1): e1531. https://doi.org/10.1002/ctm2.1531

    Background: Prostate cancer (PCa) initially shows satisfactory response to therapies targeting the androgen receptor (AR). However, progression to a castration-resistant stage indicates poor prognosis in PCa patients. AR signalling still plays a central role in most castration-resistant prostate cancers (CRPC). Therefore, unveiling the mechanisms of AR reactivation under androgen-deprived conditions is imperative to discover novel therapeutic targets for CRPC.

    Methods: Using an integrative analysis of the transcriptomics of three independent PCa cohorts and a published landscape of AR-regulated long non-coding RNA (lncRNA), lncRNA LINC01126 was selected as a candidate gene that could drive CRPC progression for further study. Quantitative reverse transcription polymerase chain reaction, in situ hybridisation (ISH) and fluorescent ISH were performed to detect LINC01126 in PCa tissues and cells. The functional role and mechanism of LINC01126 were further investigated using in vitro and in vivo gain and loss of function assays.

    Results: LINC01126, identified as an AR-repressed lncRNA, was significantly upregulated after AR-targeted therapies. In addition, we found that LINC01126 was upregulated in CRPC and was associated with poor prognosis. We also proved that LINC01126 stabilised AR protein and enhanced AR nuclear translocation and transactivation by promoting the transition from O-GlcNAcylation at threonine 80 to phosphorylation at serine 81 (S81) within the AR protein. Mechanism analysis revealed that LINC01126 facilitates the interaction of CDK9 with AR and impedes the binding of O-linked N-acetylglucosamine (O-GlcNAc) transferase to AR. Consequently, LINC01126 expression was sufficient to activate AR signalling without androgen. LINC01126 overexpression increased, whereas LINC01126 knockdown decreased castration resistance traits in PCa cells in vitro and in vivo. Furthermore, our data showed that LINC01126-targeting antisense oligonucleotides (ASO) substantially inhibited CRPC cells in vitro.

    Conclusions: Our research expands the functions of AR-regulated lncRNA in sustaining androgen-independent AR activity and promoting CRPC progression and reveals that LINC01126 may be a new therapeutic target for PCa.

  • LETTER TO THE JOURNAL
    Yi-Ge Shen , Meng-Meng Ji , Hong-Mei Yi , Rong Shen , Di Fu , Shu Cheng , Chuan-Xin Huang , Li Wang , Peng-Peng Xu , Hong-Jing Dou , Wei-Li Zhao
    2024, 14(1): e1532. https://doi.org/10.1002/ctm2.1532
  • COMMENTARY
    Hongyi Zhang , Bo Li
    2024, 14(1): e1533. https://doi.org/10.1002/ctm2.1533

    Recent studies revealed a new biological process that malignant cancer cells hijack mitochondria from nearby T cells, providing another potential mechanism for immune evasion. We further confirmed this process at the single-cell genomic level through MERCI, a novel algorithm for tracking mitochondrial (MT) transfer. Applied to human cancer samples, MERCI identified a new cancer phenotype linked to MT hijacking, correlating with rapid tumour proliferation and poor patient survival. This discovery offers insights into the limitations of current cancer immunotherapies and suggests new therapeutic avenues targeting MT transfer to enhance cancer treatment efficacy.

  • COMMENTARY
    Yuwei Li , Chaoxiong Wang , Zhigang Lu , Min Luo
    2024, 14(1): e1534. https://doi.org/10.1002/ctm2.1534
  • RESEARCH ARTICLE
    Yafei Deng , Canlin Li , Lanlan Huang , Peiwen Xiong , Yana Li , Yongjie Liu , Songyang Li , Weijian Chen , Qiang Yin , Yong Li , Qinglan Yang , Hongyan Peng , Shuting Wu , Xiangyu Wang , Qin Tong , Hongjuan Ouyang , Die Hu , Xinjia Liu , Liping Li , Jieyu You , Zhiyi Sun , Xiulan Lu , Zhenghui Xiao , Youcai Deng , Hongmei Zhao
    2024, 14(1): e1535. https://doi.org/10.1002/ctm2.1535

    Background: The understanding of the heterogeneous cellular microenvironment of colonic polyps in paediatric patients with solitary juvenile polyps (SJPs), polyposis syndrome (PJS) and Peutz-Jeghers syndrome (PJS) remains limited.

    Methods: We conducted single-cell RNA sequencing and multiplexed immunohistochemistry (mIHC) analyses on both normal colonic tissue and different types of colonic polyps obtained from paediatric patients.

    Results: We identified both shared and disease-specific cell subsets and expression patterns that played important roles in shaping the unique cellular microenvironments observed in each polyp subtype. As such, increased myeloid, endothelial and epithelial cells were the most prominent features of SJP, JPS and PJS polyps, respectively. Noticeably, memory B cells were increased, and a cluster of epithelial-mesenchymal transition (EMT)-like colonocytes existed across all polyp subtypes. Abundant neutrophil infiltration was observed in SJP polyps, while CX3CR1hi CD8+ T cells and regulatory T cells (Tregs) were predominant in SJP and JPS polyps, while GZMAhi natural killer T cells were predominant in PJS polyps. Compared with normal colonic tissues, myeloid cells exhibited specific induction of genes involved in chemotaxis and interferon-related pathways in SJP polyps, whereas fibroblasts in JPS polyps had upregulation of myofiber-associated genes and epithelial cells in PJS polyps exhibited induction of a series of nutrient absorption-related genes. In addition, the TNF-α response was uniformly upregulated in most cell subsets across all polyp subtypes, while endothelial cells and fibroblasts separately showed upregulated cell adhesion and EMT signalling in SJP and JPS polyps. Cell-cell interaction network analysis showed markedly enhanced intercellular communication, such as TNF, VEGF, CXCL and collagen signalling networks, among most cell subsets in polyps, especially SJP and JPS polyps.

    Conclusion: These findings strengthen our understanding of the heterogeneous cellular microenvironment of polyp subtypes and identify potential therapeutic approaches to reduce the recurrence of polyps in children.

  • EDITORIAL
    Iker Ausejo-Mauleon , Sara Nuin , Marta M. Alonso
    2024, 14(1): e1536. https://doi.org/10.1002/ctm2.1536
  • COMMENTARY
    Hao Fan , Freya Q. Zhang , Jing Chen
    2024, 14(1): e1537. https://doi.org/10.1002/ctm2.1537
  • COMMENTARY
    Qingguo Du , Wen-Xue Li
    2024, 14(1): e1538. https://doi.org/10.1002/ctm2.1538
  • RESEARCH ARTICLE
    Yuan Tan , Jiao Qiao , Shuo Yang , Qingchen Wang , Hongchao Liu , Qi Liu , Weimin Feng , Boxin Yang , Zhongxin Li , Liyan Cui
    2024, 14(1): e1539. https://doi.org/10.1002/ctm2.1539

    • The H3K4me3 mark and chromatin accessibility at the ARID5B promoter are increased in vitro model mimicked APS.

    • ARID5B-mediated LINC01128 induces pyroptosis and apoptosis via p-STAT3 by binding to BTF3.

    • ARID5B is high- expressed in patients with primary APS and positively correlated with LINC01128 expression.

    • OICR-9429 treatment mitigates pyroptosis and related inflammation in vivo and in vitro models mimicked APS.

  • COMMENTARY
    Alina Kurolap , Chofit Chai Gadot , David Zahler , Jacob N Ablin , Hagit Baris Feldman
    2024, 14(1): e1540. https://doi.org/10.1002/ctm2.1540
  • LETTER TO THE JOURNAL
    Carmen González-Olmedo , Francisco José García-Verdejo , Antonio Reguera-Teba , Carmen Rosa-Garrido , José Antonio López-López , Leticia Díaz-Beltrán , Patricia Mena García , Natalia Luque-Caro , Fernando Gálvez-Montosa , Ana Laura Ortega-Granados , María Ruiz-Sanjuan , Antonio Cózar-Ibáñez , Juan Sainz , Juan Antonio Marchal , José Camacho , José Pérez del Palacio , Rosario Fernández-Godino , Caridad Díaz , Pedro Sánchez-Rovira
    2024, 14(1): e1541. https://doi.org/10.1002/ctm2.1541
  • COMMENTARY
    Giovanna Giacca , Luigi Naldini , Mario Leonardo Squadrito
    2024, 14(1): e1542. https://doi.org/10.1002/ctm2.1542
  • REVIEW
    Jesús Fuentes-Antrás , Philippe L. Bedard , David W. Cescon
    2024, 14(1): e1544. https://doi.org/10.1002/ctm2.1544

    Breast cancer arises from a series of molecular alterations that disrupt cell cycle checkpoints, leading to aberrant cell proliferation and genomic instability. Targeted pharmacological inhibition of cell cycle regulators has long been considered a promising anti-cancer strategy. Initial attempts to drug critical cell cycle drivers were hampered by poor selectivity, modest efficacy and haematological toxicity. Advances in our understanding of the molecular basis of cell cycle disruption and the mechanisms of resistance to CDK4/6 inhibitors have reignited interest in blocking specific components of the cell cycle machinery, such as CDK2, CDK4, CDK7, PLK4, WEE1, PKMYT1, AURKA and TTK. These targets play critical roles in regulating quiescence, DNA replication and chromosome segregation. Extensive preclinical data support their potential to overcome CDK4/6 inhibitor resistance, induce synthetic lethality or sensitise tumours to immune checkpoint inhibitors. This review provides a biological and drug development perspective on emerging cell cycle targets and novel inhibitors, many of which exhibit favourable safety profiles and promising activity in clinical trials.

  • REVIEW
    Xingpeng Di , Jiawei Chen , Ya Li , Menghua Wang , Jingwen Wei , Tianyue Li , Banghua Liao , Deyi Luo
    2024, 14(1): e1545. https://doi.org/10.1002/ctm2.1545

    Background: The impact of fibroblasts on the immune system provides insight into the function of fibroblasts. In various tissue microenvironments, multiple fibroblast subtypes interact with immunocytes by secreting growth factors, cytokines, and chemokines, leading to wound healing, fibrosis, and escape of cancer immune surveillance. However, the specific mechanisms involved in the fibroblast-immunocyte interaction network have not yet been fully elucidated.

    Main body and conclusion: Therefore, we systematically reviewed the molecular mechanisms of fibroblast-immunocyte interactions in fibrosis, from the history of cellular evolution and cell subtype divisions to the regulatory networks between fibroblasts and immunocytes. We also discuss how these communications function in different tissue and organ statuses, as well as potential therapies targeting the reciprocal fibroblast-immunocyte interplay in fibrosis. A comprehensive understanding of these functional cells under pathophysiological conditions and the mechanisms by which they communicate may lead to the development of effective and specific therapies targeting fibrosis.

  • RESEARCH ARTICLE
    Minxiao Yi , Ye Yuan , Li Ma , Long Li , Wan Qin , Bili Wu , Bolong Zheng , Xin Liao , Guangyuan Hu , Bo Liu
    2024, 14(1): e1546. https://doi.org/10.1002/ctm2.1546

    • Activated TGFβ1 has a superior capacity in predicting radiation pneumonitis (RP) risk and plays a vital role in the development of radiation-induced pulmonary fibrosis (RIPF).

    • Conditional knock out Itgav in myofibroblasts prevented mice from developing RIPF.

    • Cilengitide alleviated the development of RIPF by inhibiting αv integrinmediated TGFβ1 activation and may be used in targeted approaches for preventing RIPF.

  • LETTER TO THE JOURNAL
    Ji Hye Shin , Mi-Jeong Kim , Ji Young Kim , Yeeun Kang , Duk-Hwan Kim , Soo-Kyung Jeong , Eunyoung Chun , Ki-Young Lee
    2024, 14(1): e1547. https://doi.org/10.1002/ctm2.1547
  • RESEARCH ARTICLE
    Ryohei Yoshitake , Hitomi Mori , Desiree Ha , Xiwei Wu , Jinhui Wang , Xiaoqiang Wang , Kohei Saeki , Gregory Chang , Hyun Jeong Shim , Yin Chan , Shiuan Chen
    2024, 14(1): e1548. https://doi.org/10.1002/ctm2.1548

    Background: Intratumour heterogeneity is a hallmark of most solid tumours, including breast cancers. We applied spatial transcriptomics and single-cell RNA-sequencing on patient-derived xenografts (PDXs) to profile spatially resolved cell populations within oestrogen receptor-positive (ER+) breast cancer and to elucidate their importance in oestrogen-dependent tumour growth.

    Methods: Two PDXs of ‘ER-high’ breast cancers with opposite oestrogen-mediated growth responses were investigated: oestrogen-suppressed GS3 (80-100% ER) and oestrogen-dependent SC31 (40-90% ER) models. The observation was validated via single-cell analyses on an ‘ER-low’ PDX, GS1 (5% ER). The results from our spatial and single-cell analyses were further supported by a public ER+ breast cancer single-cell dataset and protein-based dual immunohistochemistry (IHC) of SC31 examining important luminal cancer markers (i.e., ER, progesterone receptor and Ki67). The translational implication of our findings was assessed by clinical outcome analyses on publicly available cohorts.

    Results: Our space-gene-function study revealed four spatially distinct compartments within ER+ breast cancers. These compartments showed functional diversity (oestrogen-responsive, proliferative, hypoxia-induced and inflammation-related). The ‘proliferative’ population, rather than the ‘oestrogen-responsive’ compartment, was crucial for oestrogen-dependent tumour growth, leading to the acquisition of luminal B-like features. The cells expressing typical oestrogen-responsive genes like PGR were not directly linked to oestrogen-dependent proliferation. Dual IHC analyses demonstrated the distinct contribution of the Ki67+ proliferative cells toward oestrogen-mediated growth and their response to a CDK4/6 inhibitor. The gene signatures derived from the proliferative, hypoxia-induced and inflammation-related compartments were significantly correlated with worse clinical outcomes, while patients with the oestrogen-responsive signature showed better prognoses, suggesting that this compartment would not be directly associated with oestrogen-dependent tumour progression.

    Conclusions: Our study identified the gene signature in our ‘proliferative’ compartment as an important determinant of luminal cancer subtypes. This ‘proliferative’ cell population is a causative feature of luminal B breast cancer, contributing toward its aggressive behaviours.

  • LETTER TO THE JOURNAL
    Wei Ouyang , Ran Xu , Hanyu Yao , Shusuan Jiang , Qiang Lu , Cai Lv , Pei Li , Genming Xu , Jianye Liu , Long Wang
    2024, 14(1): e1549. https://doi.org/10.1002/ctm2.1549
  • LETTER TO THE JOURNAL
    So Yun Lim , Jineui Kim , Ji-Soo Kwon , Sung-Woon Kang , Seung-Beom Kim , Woori Kim , Ju Yeon Son , Choi Young Jang , Heedo Park , Jeonghun Kim , Sohyun Lee , Kyung Taek Kim , Jaeuk Choi , Ji Yeun Kim , Joon Seo Lim , Euijin Chang , Seongman Bae , Jiwon Jung , Min Jae Kim , Yong Pil Chong , Sang-Oh Lee , Sang-Ho Choi , Yang Soo Kim , Man-Seong Park , Sung-Han Kim
    2024, 14(1): e1551. https://doi.org/10.1002/ctm2.1551
  • LETTER TO THE JOURNAL
    Qintao Ge , Zhijie Zhao , Xiao Li , Feixiang Yang , Meng Zhang , Zongyao Hao , Chaozhao Liang , Jialin Meng
    2024, 14(1): e1552. https://doi.org/10.1002/ctm2.1552
  • RESEARCH ARTICLE
    Baohong Luo , Yu Sun , Qinru Zhan , Yuting Luo , Yu Chen , Tongze Fu , Tiantian Yang , Lijuan Ren , Zhongpeng Xie , Xiaohua Situ , Bixia Liu , Kejing Tang , Zunfu Ke
    2024, 14(1): e1553. https://doi.org/10.1002/ctm2.1553

    Background: T-cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) is an immune checkpoint molecule that suppresses CD8+ T-cell function in cancer. However, the expression profile and functional significance of TIGIT in the immune microenvironment of lung adenocarcinoma (LUAD) remain elusive. Interleukin (IL)-15 has emerged as a promising candidate for enhancing CD8+ T-cell mediated tumour eradication. Exploring therapeutic strategies that combine IL-15 with TIGIT blockade in LUAD is warranted.

    Methods: We investigated the regulatory network involving coinhibitory TIGIT and CD96, as well as costimulatory CD226 in LUAD using clinical samples. The potential role of TIGIT in regulating the pathogenesis of LUAD was addressed through a murine model with transplanted tumours constructed in Tigit−/− mice. The therapeutic strategy that combines TIGIT blockade with IL-15 stimulation was verified using a transplanted tumour murine model and a patient-derived organoid (PDO) model.

    Results: The frequency of TIGIT+CD8+ T cells was significantly increased in LUAD. Increased TIGIT expression indicated poorer prognosis in LUAD patients. Furthermore, the effector function of TIGIT+CD8+ tumour-infiltrating lymphocytes (TILs) was impaired in LUAD patients and TIGIT inhibited antitumour immune response of CD8+ TILs in tumour-bearing mice. Mechanistically, IL-15 enhanced the effector function of CD8+ TILs but stimulated the expression of TIGIT on CD8+ TILs concomitantly. The application of IL-15 combined with TIGIT blockade showed additive effects in enhancing the cytotoxicity of CD8+ TILs and thus further increased the antitumour immune response in LUAD.

    Conclusions: Our findings identified TIGIT as a promising therapeutic target for LUAD. LUAD could benefit more from the combined therapy of IL-15 stimulation and TIGIT blockade.

  • LETTER TO THE JOURNAL
    Li-Juan Jiang , Song-Bin Guo , Zhao-Hui Zhou , Zhi-Yong Li , Fang-Jian Zhou , Chun-Ping Yu , Mei Li , Wei-Juan Huang , Zhuo-Wei Liu , Xiao-Peng Tian
    2024, 14(1): e1555. https://doi.org/10.1002/ctm2.1555
  • RESEARCH ARTICLE
    Shengming Xu , Chaoji Shi , Rong Zhou , Yong Han , NianNian Li , Chuxiang Qu , Ronghui Xia , Chunye Zhang , Yuhua Hu , Zhen Tian , Shuli Liu , Lizhen Wang , Jiang Li , Zhiyuan Zhang
    2024, 14(1): e1556. https://doi.org/10.1002/ctm2.1556

    Background: Human papillomavirus (HPV) integration into the host genome is an important factor in HPV(+)OPSCC carcinogenesis, in conjunction with HPV oncoproteins E6/E7. However, a well-studied investigation about virus-host interaction still needs to be completed. Our objective is to characterise HPV integration to investigate potential mechanisms of tumourigenesis independent of E6/E7 oncoproteins.

    Materials and methods: High-throughput viral integration detection was performed on 109 HPV(+)OPSCC tumours with relevant clinicopathological information. Of these tumours, 38 tumours underwent targeted gene sequencing, 29 underwent whole exome sequencing and 26 underwent RNA sequencing.

    Results: HPV integration was detected in 94% of tumours (with a mean integration count of 337). Tumours occurring at the tonsil/oropharyngeal wall that exhibit higher PD-L1 expression demonstrated increased integration sites (p = .024). HPV exhibited a propensity for integration at genomic sites located within specific fragile sites (FRA19A) or genes associated with functional roles such as cell proliferation and differentiation (PTEN, AR), immune evasion (CD274) and glycoprotein biosynthesis process (FUT8). The viral oncogenes E2, E4, E6 and E7 tended to remain intact. HPV fragments displayed enrichment within host copy number variation (CNV) regions. However, insertions into genes related to altered homologous recombination repair were infrequent. Genes with integration had distinct expression levels. Fifty-nine genes whose expression level was affected by viral integration were identified, for example, EPHB1, which was reported to be involved in cellular protein metabolic process.

    Conclusions: HPV can promote oncogenesis through recurrent integration into functional host genome regions, leading to subsequent genomic aberrations and gene expression disruption. This study characterises viral integrations and virus-host interactions, enhancing our understanding of HPV-related carcinogenesis mechanisms.

  • LETTER TO THE JOURNAL
    Li Guo , Daoliang Xia , Jiaming Jin , Shizheng Xiong , Xinru Xu , Lulu Luo , Xueni Yang , Xinmiao Zhao , Dekang Ren , Jiafeng Yu , Tingming Liang
    2024, 14(1): e1557. https://doi.org/10.1002/ctm2.1557
  • COMMENTARY
    Bin Xiao , Eng-King Tan
    2024, 14(1): e1559. https://doi.org/10.1002/ctm2.1559
  • COMMENTARY
    Rashad Hussain , Maiken Nedergaard
    2024, 14(1): e1562. https://doi.org/10.1002/ctm2.1562
  • RESEARCH ARTICLE
    Chen Su , Haoquan Zhang , Jie Mo , Zhibin Liao , Bixiang Zhang , Peng Zhu
    2024, 14(1): e1563. https://doi.org/10.1002/ctm2.1563

    Background: Hepatocellular carcinoma (HCC) continues to pose a significant threat to patient survival. Emerging evidence underscores the pivotal involvement of long non-coding RNAs (lncRNAs) in the cancer process. Nevertheless, our understanding of the roles and processes of lncRNAs in HCC remains limited.

    Methods: The expression level of USP27X-AS1 was assessed in an HCC patient cohort through a combination of bioinformatics analysis and qRT-PCR. Subsequent biological experiments were conducted to delve into the functional aspects of USP27X-AS1. Additional molecular biology techniques, including RNA pulldown and RNA immunoprecipitation (RIP), were employed to elucidate the potential mechanisms involving USP27X-AS1 in HCC. Finally, CUT-RUN assay and other investigations were carried out to determine the factors contributing to the heightened expression of USP27X-AS1 in HCC.

    Results: High expression of the novel oncogene USP27X-AS1 predicted poor prognosis in HCC patients. Further investigation confirmed that USP27X-AS1 promoted the proliferation and metastasis of HCC by enabling USP7 to interact with AKT, which reduced level of AKT poly-ubiquitylation and enhanced AKT protein stability, which improves protein stabilisation of AKT and promotes the progression of HCC. Moreover, we also revealed that SP1 binds to USP27X-AS1 promoter to activate its transcription.

    Conclusions: Novel oncogenic lncRNA USP27X-AS1 promoted HCC progression via recruiting USP7 to deubiquitinate AKT. SP1 transcriptionally activated USP27X-AS1 expression. These findings shed light on HCC and pointed to USP27X-AS1 as a potential predictive biomarker and treatment target for the malignancy.

  • COMMENTARY
    Yuhei Hosokawa , Manuel Maestre-Reyna
    2024, 14(1): e1566. https://doi.org/10.1002/ctm2.1566