Pub Date : 2024-10-22DOI: 10.1136/gutjnl-2024-333702
Elisa Espinet, Gioacchino Natoli
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, with a dismal 5-year survival rate of less than 10%.1 Despite significant advances in cancer treatment over the past decades, PDAC has stubbornly defied progress, with most patients showing limited response to standard chemotherapy regimens. Perhaps most disappointingly, the revolutionary success of immunotherapy in various cancer types has not translated to PDAC, with checkpoint inhibitors and other immunomodulatory strategies showing dismally low response rates.2 This lack of efficacy is thought to be largely due to the characteristic PDAC tumour microenvironment (TME) that contributes to make this cancer immunologically ‘cold’.3 Characterised by a dense stromal compartment, low T cell infiltration and an overall immunosuppressive microenvironment, PDACs present a formidable barrier to the immune system that ultimately explains the inefficiency of current immunotherapeutic approaches. Whether and to what extent strategies can be identified to convert these ‘cold’ tumours into ‘hot’ ones and ultimately improve immune checkpoint blockade (ICB) efficacy in PDAC remains to be determined. One such strategy leverages the impact of epigenetic regulators in the control of genomic transposable elements, which comprise retrotransposons (or retroelements, including endogenous retroviruses, ERVs) and DNA transposons.4 Indeed, one …
胰腺导管腺癌(PDAC)仍然是最致命的恶性肿瘤之一,5 年生存率不到 10%,令人沮丧。1 尽管过去几十年来癌症治疗取得了重大进展,但 PDAC 顽固地拒绝进步,大多数患者对标准化疗方案的反应有限。也许最令人失望的是,免疫疗法在各种癌症类型中取得的革命性成功并没有应用于 PDAC,检查点抑制剂和其他免疫调节策略的反应率低得令人沮丧。3 PDAC 的特点是致密的基质区、低 T 细胞浸润和整体免疫抑制性微环境,这对免疫系统构成了巨大的障碍,最终导致目前的免疫治疗方法效率低下。是否能找到将这些 "冷 "肿瘤转化为 "热 "肿瘤的策略,并在多大程度上最终提高免疫检查点阻断疗法(ICB)在 PDAC 中的疗效,仍有待确定。其中一种策略是利用表观遗传调节因子在控制基因组转座元件方面的影响,转座元件包括逆转录转座子(或逆位子,包括内源性逆转录病毒 ERV)和 DNA 转座子。
{"title":"Leveraging the tug-of-war with genomic retroelements to enhance immunotherapy of pancreatic cancer","authors":"Elisa Espinet, Gioacchino Natoli","doi":"10.1136/gutjnl-2024-333702","DOIUrl":"https://doi.org/10.1136/gutjnl-2024-333702","url":null,"abstract":"Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, with a dismal 5-year survival rate of less than 10%.1 Despite significant advances in cancer treatment over the past decades, PDAC has stubbornly defied progress, with most patients showing limited response to standard chemotherapy regimens. Perhaps most disappointingly, the revolutionary success of immunotherapy in various cancer types has not translated to PDAC, with checkpoint inhibitors and other immunomodulatory strategies showing dismally low response rates.2 This lack of efficacy is thought to be largely due to the characteristic PDAC tumour microenvironment (TME) that contributes to make this cancer immunologically ‘cold’.3 Characterised by a dense stromal compartment, low T cell infiltration and an overall immunosuppressive microenvironment, PDACs present a formidable barrier to the immune system that ultimately explains the inefficiency of current immunotherapeutic approaches. Whether and to what extent strategies can be identified to convert these ‘cold’ tumours into ‘hot’ ones and ultimately improve immune checkpoint blockade (ICB) efficacy in PDAC remains to be determined. One such strategy leverages the impact of epigenetic regulators in the control of genomic transposable elements, which comprise retrotransposons (or retroelements, including endogenous retroviruses, ERVs) and DNA transposons.4 Indeed, one …","PeriodicalId":12825,"journal":{"name":"Gut","volume":null,"pages":null},"PeriodicalIF":24.5,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142487503","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-22DOI: 10.1136/gutjnl-2024-332755
Victoria Wilkinson-Smith, Mark Scott, Alex Menys, Lukasz Wiklendt, Luca Marciani, David Atkinson, Stefano Sansone, Ausra Zdanaviciene, Carol Coupland, Charles H Knowles, Philip Dinning, Stuart A Taylor, Penny Gowland, Caroline Louise Hoad, Maura Corsetti, Robin C Spiller
Background Colonic motility in constipation can be assessed non-invasively using MRI. Objective To compare MRI with high-resolution colonic manometry (HRCM) for predicting treatment response. Design Part 1: 44 healthy volunteers (HVs), 43 patients with irritable bowel syndrome with constipation (IBS-C) and 37 with functional constipation (FC) completed stool diaries and questionnaires and underwent oral macrogol (500–1000 mL) challenge. Whole gut transit time (WGTT), segmental colonic volumes (CV), MRI-derived Motility Index and chyme movement by ‘tagging’ were assessed using MRI and time to defecation after macrogol recorded. Left colonic HRCM was recorded before and after a 700 kcal meal. Patients then proceeded to Part 2: a randomised cross-over study of 10-days bisacodyl 10 mg daily versus hyoscine 20 mg three times per day, assessing daily pain and constipation. Results Part 1: Total CVs median (range) were significantly greater in IBS-C (776 (595–1033)) and FC (802 (633–951)) vs HV (645 (467–780)), p<0.001. Patients also had longer WGTT and delayed evacuation after macrogol. IBS-C patients showed significantly reduced tagging index and less propagated pressure wave (PPW) activity during HRCM versus HV. Compared with FC, IBS-C patients were more anxious and reported more pain. Abnormally large colons predicted significantly delayed evacuation after macrogol challenge (p<0.02), impaired manometric meal response and reduced pain with bisacodyl (p<0.05). Part 2: Bisacodyl compared with hyoscine increased bowel movements but caused more pain in both groups (p<0.03). Conclusion An abnormally large colon is an important feature in constipation which predicts impaired manometric response to feeding and treatment responses. HRCM shows that IBS-C patients have reduced PPW activity. Trial registration number The study was preregistered on ClinicalTrials.gov, Reference: [NCT03226145][1]. Data are available on reasonable request. [1]: /lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT03226145&atom=%2Fgutjnl%2Fearly%2F2024%2F10%2F22%2Fgutjnl-2024-332755.atom
{"title":"Combined MRI, high-resolution manometry and a randomised trial of bisacodyl versus hyoscine show the significance of an enlarged colon in constipation: the RECLAIM study","authors":"Victoria Wilkinson-Smith, Mark Scott, Alex Menys, Lukasz Wiklendt, Luca Marciani, David Atkinson, Stefano Sansone, Ausra Zdanaviciene, Carol Coupland, Charles H Knowles, Philip Dinning, Stuart A Taylor, Penny Gowland, Caroline Louise Hoad, Maura Corsetti, Robin C Spiller","doi":"10.1136/gutjnl-2024-332755","DOIUrl":"https://doi.org/10.1136/gutjnl-2024-332755","url":null,"abstract":"Background Colonic motility in constipation can be assessed non-invasively using MRI. Objective To compare MRI with high-resolution colonic manometry (HRCM) for predicting treatment response. Design Part 1: 44 healthy volunteers (HVs), 43 patients with irritable bowel syndrome with constipation (IBS-C) and 37 with functional constipation (FC) completed stool diaries and questionnaires and underwent oral macrogol (500–1000 mL) challenge. Whole gut transit time (WGTT), segmental colonic volumes (CV), MRI-derived Motility Index and chyme movement by ‘tagging’ were assessed using MRI and time to defecation after macrogol recorded. Left colonic HRCM was recorded before and after a 700 kcal meal. Patients then proceeded to Part 2: a randomised cross-over study of 10-days bisacodyl 10 mg daily versus hyoscine 20 mg three times per day, assessing daily pain and constipation. Results Part 1: Total CVs median (range) were significantly greater in IBS-C (776 (595–1033)) and FC (802 (633–951)) vs HV (645 (467–780)), p<0.001. Patients also had longer WGTT and delayed evacuation after macrogol. IBS-C patients showed significantly reduced tagging index and less propagated pressure wave (PPW) activity during HRCM versus HV. Compared with FC, IBS-C patients were more anxious and reported more pain. Abnormally large colons predicted significantly delayed evacuation after macrogol challenge (p<0.02), impaired manometric meal response and reduced pain with bisacodyl (p<0.05). Part 2: Bisacodyl compared with hyoscine increased bowel movements but caused more pain in both groups (p<0.03). Conclusion An abnormally large colon is an important feature in constipation which predicts impaired manometric response to feeding and treatment responses. HRCM shows that IBS-C patients have reduced PPW activity. Trial registration number The study was preregistered on ClinicalTrials.gov, Reference: [NCT03226145][1]. Data are available on reasonable request. [1]: /lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT03226145&atom=%2Fgutjnl%2Fearly%2F2024%2F10%2F22%2Fgutjnl-2024-332755.atom","PeriodicalId":12825,"journal":{"name":"Gut","volume":null,"pages":null},"PeriodicalIF":24.5,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142487501","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-22DOI: 10.1136/gutjnl-2023-330390
Mitsuhiro Shimura, Junichi Matsuo, ShuChin Pang, Nawaphat Jangphattananont, Aashiq Hussain, Muhammad Bakhait Rahmat, Jung-Won Lee, Daisuke Douchi, Jasmine Jie Lin Tong, Khine Myint, Supriya Srivastava, Ming Teh, Vivien Koh, Wei Peng Yong, Jimmy Bok Yan So, Patrick Tan, Khay-Guan Yeoh, Michiaki Unno, Linda Shyue Huey Chuang, Yoshiaki Ito
Background The elevation of IQGAP3 expression in diverse cancers indicates a key role for IQGAP3 in carcinogenesis. Although IQGAP3 was established as a proliferating stomach stem cell factor and a regulator of the RAS-ERK pathway, how it drives cancer growth remains unclear. Objective We define the function of IQGAP3 in gastric cancer (GC) development and progression. Design We studied the phenotypic changes caused by IQGAP3 knockdown in three molecularly diverse GC cell lines by RNA-sequencing. In vivo tumorigenesis and lung metastasis assays corroborated IQGAP3 as a mediator of oncogenic signalling. Spatial analysis was performed to evaluate the intratumoral transcriptional and functional differences between control tumours and IQGAP3 knockdown tumours. Results Transcriptomic profiling showed that IQGAP3 inhibition attenuates signal transduction networks, such as KRAS signalling, via phosphorylation blockade. IQGAP3 knockdown was associated with significant inhibition of MEK/ERK signalling-associated growth factors, including TGFβ1, concomitant with gene signatures predictive of impaired tumour microenvironment formation and reduced metastatic potential. Xenografts involving IQGAP3 knockdown cells showed attenuated tumorigenesis and lung metastasis in immunodeficient mice. Accordingly, immunofluorescence staining revealed significant reductions of TGFβ/SMAD signalling and αSMA-positive stromal cells; digital spatial analysis indicated that IQGAP3 is indispensable for the formation of two phenotypically diverse cell subpopulations, which played crucial but distinct roles in promoting oncogenic functions. Conclusion IQGAP3 knockdown suppressed the RAS-TGFβ signalling crosstalk, leading to a significant reduction of the tumour microenvironment. In particular, IQGAP3 maintains functional heterogeneity of cancer cells to enhance malignant growth. IQGAP3 is thus a highly relevant therapy target in GC. Data are available upon reasonable request. All data relevant to the study are included in the article or uploaded as supplementary information. Raw data of RNA sequence is available on GEO: GSE231642.
{"title":"IQGAP3 signalling mediates intratumoral functional heterogeneity to enhance malignant growth","authors":"Mitsuhiro Shimura, Junichi Matsuo, ShuChin Pang, Nawaphat Jangphattananont, Aashiq Hussain, Muhammad Bakhait Rahmat, Jung-Won Lee, Daisuke Douchi, Jasmine Jie Lin Tong, Khine Myint, Supriya Srivastava, Ming Teh, Vivien Koh, Wei Peng Yong, Jimmy Bok Yan So, Patrick Tan, Khay-Guan Yeoh, Michiaki Unno, Linda Shyue Huey Chuang, Yoshiaki Ito","doi":"10.1136/gutjnl-2023-330390","DOIUrl":"https://doi.org/10.1136/gutjnl-2023-330390","url":null,"abstract":"Background The elevation of IQGAP3 expression in diverse cancers indicates a key role for IQGAP3 in carcinogenesis. Although IQGAP3 was established as a proliferating stomach stem cell factor and a regulator of the RAS-ERK pathway, how it drives cancer growth remains unclear. Objective We define the function of IQGAP3 in gastric cancer (GC) development and progression. Design We studied the phenotypic changes caused by IQGAP3 knockdown in three molecularly diverse GC cell lines by RNA-sequencing. In vivo tumorigenesis and lung metastasis assays corroborated IQGAP3 as a mediator of oncogenic signalling. Spatial analysis was performed to evaluate the intratumoral transcriptional and functional differences between control tumours and IQGAP3 knockdown tumours. Results Transcriptomic profiling showed that IQGAP3 inhibition attenuates signal transduction networks, such as KRAS signalling, via phosphorylation blockade. IQGAP3 knockdown was associated with significant inhibition of MEK/ERK signalling-associated growth factors, including TGFβ1, concomitant with gene signatures predictive of impaired tumour microenvironment formation and reduced metastatic potential. Xenografts involving IQGAP3 knockdown cells showed attenuated tumorigenesis and lung metastasis in immunodeficient mice. Accordingly, immunofluorescence staining revealed significant reductions of TGFβ/SMAD signalling and αSMA-positive stromal cells; digital spatial analysis indicated that IQGAP3 is indispensable for the formation of two phenotypically diverse cell subpopulations, which played crucial but distinct roles in promoting oncogenic functions. Conclusion IQGAP3 knockdown suppressed the RAS-TGFβ signalling crosstalk, leading to a significant reduction of the tumour microenvironment. In particular, IQGAP3 maintains functional heterogeneity of cancer cells to enhance malignant growth. IQGAP3 is thus a highly relevant therapy target in GC. Data are available upon reasonable request. All data relevant to the study are included in the article or uploaded as supplementary information. Raw data of RNA sequence is available on GEO: GSE231642.","PeriodicalId":12825,"journal":{"name":"Gut","volume":null,"pages":null},"PeriodicalIF":24.5,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142487247","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-15DOI: 10.1136/gutjnl-2024-332820
Giulio Antonelli, Diogo Libanio, Albert Jeroen De Groof, Fons van der Sommen, Pietro Mascagni, Pieter Sinonquel, Mohamed Abdelrahim, Omer Ahmad, Tyler Berzin, Pradeep Bhandari, Michael Bretthauer, Miguel Coimbra, Evelien Dekker, Alanna Ebigbo, Tom Eelbode, Leonardo Frazzoni, Seth A Gross, Ryu Ishihara, Michal Filip Kaminski, Helmut Messmann, Yuichi Mori, Nicolas Padoy, Sravanthi Parasa, Nastazja Dagny Pilonis, Francesco Renna, Alessandro Repici, Cem Simsek, Marco Spadaccini, Raf Bisschops, Jacques J G H M Bergman, Cesare Hassan, Mario Dinis Ribeiro
Artificial intelligence (AI) holds significant potential for enhancing quality of gastrointestinal (GI) endoscopy, but the adoption of AI in clinical practice is hampered by the lack of rigorous standardisation and development methodology ensuring generalisability. The aim of the Quality Assessment of pre-clinical AI studies in Diagnostic Endoscopy (QUAIDE) Explanation and Checklist was to develop recommendations for standardised design and reporting of preclinical AI studies in GI endoscopy. The recommendations were developed based on a formal consensus approach with an international multidisciplinary panel of 32 experts among endoscopists and computer scientists. The Delphi methodology was employed to achieve consensus on statements, with a predetermined threshold of 80% agreement. A maximum three rounds of voting were permitted. Consensus was reached on 18 key recommendations, covering 6 key domains: data acquisition and annotation (6 statements), outcome reporting (3 statements), experimental setup and algorithm architecture (4 statements) and result presentation and interpretation (5 statements). QUAIDE provides recommendations on how to properly design (1. Methods, statements 1–14), present results (2. Results, statements 15–16) and integrate and interpret the obtained results (3. Discussion, statements 17–18). The QUAIDE framework offers practical guidance for authors, readers, editors and reviewers involved in AI preclinical studies in GI endoscopy, aiming at improving design and reporting, thereby promoting research standardisation and accelerating the translation of AI innovations into clinical practice.
{"title":"QUAIDE - Quality assessment of AI preclinical studies in diagnostic endoscopy","authors":"Giulio Antonelli, Diogo Libanio, Albert Jeroen De Groof, Fons van der Sommen, Pietro Mascagni, Pieter Sinonquel, Mohamed Abdelrahim, Omer Ahmad, Tyler Berzin, Pradeep Bhandari, Michael Bretthauer, Miguel Coimbra, Evelien Dekker, Alanna Ebigbo, Tom Eelbode, Leonardo Frazzoni, Seth A Gross, Ryu Ishihara, Michal Filip Kaminski, Helmut Messmann, Yuichi Mori, Nicolas Padoy, Sravanthi Parasa, Nastazja Dagny Pilonis, Francesco Renna, Alessandro Repici, Cem Simsek, Marco Spadaccini, Raf Bisschops, Jacques J G H M Bergman, Cesare Hassan, Mario Dinis Ribeiro","doi":"10.1136/gutjnl-2024-332820","DOIUrl":"https://doi.org/10.1136/gutjnl-2024-332820","url":null,"abstract":"Artificial intelligence (AI) holds significant potential for enhancing quality of gastrointestinal (GI) endoscopy, but the adoption of AI in clinical practice is hampered by the lack of rigorous standardisation and development methodology ensuring generalisability. The aim of the Quality Assessment of pre-clinical AI studies in Diagnostic Endoscopy (QUAIDE) Explanation and Checklist was to develop recommendations for standardised design and reporting of preclinical AI studies in GI endoscopy. The recommendations were developed based on a formal consensus approach with an international multidisciplinary panel of 32 experts among endoscopists and computer scientists. The Delphi methodology was employed to achieve consensus on statements, with a predetermined threshold of 80% agreement. A maximum three rounds of voting were permitted. Consensus was reached on 18 key recommendations, covering 6 key domains: data acquisition and annotation (6 statements), outcome reporting (3 statements), experimental setup and algorithm architecture (4 statements) and result presentation and interpretation (5 statements). QUAIDE provides recommendations on how to properly design (1. Methods, statements 1–14), present results (2. Results, statements 15–16) and integrate and interpret the obtained results (3. Discussion, statements 17–18). The QUAIDE framework offers practical guidance for authors, readers, editors and reviewers involved in AI preclinical studies in GI endoscopy, aiming at improving design and reporting, thereby promoting research standardisation and accelerating the translation of AI innovations into clinical practice.","PeriodicalId":12825,"journal":{"name":"Gut","volume":null,"pages":null},"PeriodicalIF":24.5,"publicationDate":"2024-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142440132","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-14DOI: 10.1136/gutjnl-2024-332837
Henrike Salié, Lara Wischer, Antonio D'Alessio, Ira Godbole, Yuan Suo, Patricia Otto-Mora, Juergen Beck, Olaf Neumann, Albrecht Stenzinger, Peter Schirmacher, Claudia A M Fulgenzi, Andreas Blaumeiser, Melanie Boerries, Natascha Roehlen, Michael Schultheiß, Maike Hofmann, Robert Thimme, David J Pinato, Thomas Longerich, Bertram Bengsch
Background: The determinants of the response to checkpoint immunotherapy in hepatocellular carcinoma (HCC) remain poorly understood. The organisation of the immune response in the tumour microenvironment (TME) is expected to govern immunotherapy outcomes but spatial immunotypes remain poorly defined.
Objective: We hypothesised that the deconvolution of spatial immune network architectures could identify clinically relevant immunotypes in HCC.
Design: We conducted highly multiplexed imaging mass cytometry on HCC tissues from 101 patients. We performed in-depth spatial single-cell analysis in a discovery and validation cohort to deconvolute the determinants of the heterogeneity of HCC immune architecture and develop a spatial immune classification that was tested for the prediction of immune checkpoint inhibitor (ICI) therapy.
Results: Bioinformatic analysis identified 23 major immune, stroma, parenchymal and tumour cell types in the HCC TME. Unsupervised neighbourhood detection based on the spatial interaction of immune cells identified three immune architectures with differing involvement of immune cells and immune checkpoints dominated by either CD8 T-cells, myeloid immune cells or B- and CD4 T-cells. We used these to define three major spatial HCC immunotypes that reflect a higher level of intratumour immune cell organisation: depleted, compartmentalised and enriched. Progression-free survival under ICI therapy differed significantly between the spatial immune types with improved survival of enriched patients. In patients with intratumour heterogeneity, the presence of one enriched area governed long-term survival.
{"title":"Spatial single-cell profiling and neighbourhood analysis reveal the determinants of immune architecture connected to checkpoint inhibitor therapy outcome in hepatocellular carcinoma.","authors":"Henrike Salié, Lara Wischer, Antonio D'Alessio, Ira Godbole, Yuan Suo, Patricia Otto-Mora, Juergen Beck, Olaf Neumann, Albrecht Stenzinger, Peter Schirmacher, Claudia A M Fulgenzi, Andreas Blaumeiser, Melanie Boerries, Natascha Roehlen, Michael Schultheiß, Maike Hofmann, Robert Thimme, David J Pinato, Thomas Longerich, Bertram Bengsch","doi":"10.1136/gutjnl-2024-332837","DOIUrl":"10.1136/gutjnl-2024-332837","url":null,"abstract":"<p><strong>Background: </strong>The determinants of the response to checkpoint immunotherapy in hepatocellular carcinoma (HCC) remain poorly understood. The organisation of the immune response in the tumour microenvironment (TME) is expected to govern immunotherapy outcomes but spatial immunotypes remain poorly defined.</p><p><strong>Objective: </strong>We hypothesised that the deconvolution of spatial immune network architectures could identify clinically relevant immunotypes in HCC.</p><p><strong>Design: </strong>We conducted highly multiplexed imaging mass cytometry on HCC tissues from 101 patients. We performed in-depth spatial single-cell analysis in a discovery and validation cohort to deconvolute the determinants of the heterogeneity of HCC immune architecture and develop a spatial immune classification that was tested for the prediction of immune checkpoint inhibitor (ICI) therapy.</p><p><strong>Results: </strong>Bioinformatic analysis identified 23 major immune, stroma, parenchymal and tumour cell types in the HCC TME. Unsupervised neighbourhood detection based on the spatial interaction of immune cells identified three immune architectures with differing involvement of immune cells and immune checkpoints dominated by either CD8 T-cells, myeloid immune cells or B- and CD4 T-cells. We used these to define three major spatial HCC immunotypes that reflect a higher level of intratumour immune cell organisation: depleted, compartmentalised and enriched. Progression-free survival under ICI therapy differed significantly between the spatial immune types with improved survival of enriched patients. In patients with intratumour heterogeneity, the presence of one enriched area governed long-term survival.</p>","PeriodicalId":12825,"journal":{"name":"Gut","volume":null,"pages":null},"PeriodicalIF":23.0,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142345116","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: The potency of T cell-mediated responses is a determinant of immunotherapy effectiveness in treating malignancies; however, the clinical efficacy of T-cell therapies has been limited in hepatocellular carcinoma (HCC) owing to the extensive immunosuppressive microenvironment.
Objective: Here, we aimed to investigate the key genes contributing to immune escape in HCC and raise a new therapeutic strategy for remoulding the HCC microenvironment.
Design: The genome-wide in vivo clustered regularly interspaced short palindromic repeats (CRISPR) screen library was conducted to identify the key genes associated with immune tolerance. Single-cell RNA-seq (scRNA-seq), flow cytometry, HCC mouse models, chromatin immunoprecipitation and coimmunoprecipitation were used to explore the function and mechanism of adenylate cyclase 7 (ADCY7) in HCC immune surveillance.
Results: Here, a genome-wide in vivo CRISPR screen identified a novel immune modulator-ADCY7. The transmembrane protein ADCY7 undergoes subcellular translocation via caveolae-mediated endocytosis and then translocates to the nucleus with the help of leucine-rich repeat-containing protein 59 (LRRC59) and karyopherin subunit beta 1 (KPNB1). In the nucleus, it functions as a transcription cofactor of CCAAT/enhancer binding protein alpha (CEBPA) to induce CCL5 transcription, thereby increasing CD8+ T cell infiltration to restrain HCC progression. Furthermore, ADCY7 can be secreted as exosomes and enter neighbouring tumour cells to promote CCL5 induction. Exosomes with high ADCY7 levels promote intratumoural infiltration of CD8+ T cells and suppress HCC tumour growth.
Conclusion: We delineate the unconventional function and subcellular location of ADCY7, highlighting its pivotal role in T cell-mediated immunity in HCC and its potential as a promising treatment target.
背景:T细胞介导的反应效力是免疫疗法治疗恶性肿瘤有效性的决定因素;然而,由于肝细胞癌(HCC)具有广泛的免疫抑制微环境,T细胞疗法的临床疗效受到了限制。目的:在此,我们旨在研究导致HCC免疫逃逸的关键基因,并提出重塑HCC微环境的新治疗策略:设计:我们在全基因组范围内建立了体内簇状规则间隔短回文重复序列(CRISPR)筛选文库,以确定与免疫耐受相关的关键基因。利用单细胞RNA-seq(scRNA-seq)、流式细胞术、HCC小鼠模型、染色质免疫沉淀和共免疫沉淀等方法探讨了腺苷酸环化酶7(ADCY7)在HCC免疫监视中的功能和机制:结果:一项全基因组的体内CRISPR筛选发现了一种新型免疫调节剂--ADCY7。跨膜蛋白ADCY7通过洞穴介导的内吞作用进行亚细胞转运,然后在富含亮氨酸重复序列蛋白59(LRRC59)和karyopherin亚基β1(KPNB1)的帮助下转运至细胞核。在细胞核中,它作为 CCAAT/增强子结合蛋白 alpha(CEBPA)的转录辅助因子,诱导 CCL5 转录,从而增加 CD8+ T 细胞浸润,抑制 HCC 的进展。此外,ADCY7 还可以作为外泌体分泌,进入邻近的肿瘤细胞,促进 CCL5 的诱导。ADCY7含量高的外泌体可促进CD8+ T细胞的瘤内浸润,抑制HCC肿瘤的生长:我们描述了 ADCY7 的非常规功能和亚细胞位置,强调了它在 HCC 中 T 细胞介导的免疫中的关键作用,以及作为治疗靶点的潜力。
{"title":"Nuclear translocation of plasma membrane protein ADCY7 potentiates T cell-mediated antitumour immunity in HCC.","authors":"Jianan Chen, Youhai Jiang, Minghui Hou, Chunliang Liu, Erdong Liu, Yali Zong, Xiang Wang, Zhengyuan Meng, Mingye Gu, Yu Su, Hongyang Wang, Jing Fu","doi":"10.1136/gutjnl-2024-332902","DOIUrl":"10.1136/gutjnl-2024-332902","url":null,"abstract":"<p><strong>Background: </strong>The potency of T cell-mediated responses is a determinant of immunotherapy effectiveness in treating malignancies; however, the clinical efficacy of T-cell therapies has been limited in hepatocellular carcinoma (HCC) owing to the extensive immunosuppressive microenvironment.</p><p><strong>Objective: </strong>Here, we aimed to investigate the key genes contributing to immune escape in HCC and raise a new therapeutic strategy for remoulding the HCC microenvironment.</p><p><strong>Design: </strong>The genome-wide in vivo clustered regularly interspaced short palindromic repeats (CRISPR) screen library was conducted to identify the key genes associated with immune tolerance. Single-cell RNA-seq (scRNA-seq), flow cytometry, HCC mouse models, chromatin immunoprecipitation and coimmunoprecipitation were used to explore the function and mechanism of adenylate cyclase 7 (ADCY7) in HCC immune surveillance.</p><p><strong>Results: </strong>Here, a genome-wide in vivo CRISPR screen identified a novel immune modulator-ADCY7. The transmembrane protein ADCY7 undergoes subcellular translocation via caveolae-mediated endocytosis and then translocates to the nucleus with the help of leucine-rich repeat-containing protein 59 (LRRC59) and karyopherin subunit beta 1 (KPNB1). In the nucleus, it functions as a transcription cofactor of CCAAT/enhancer binding protein alpha (CEBPA) to induce <i>CCL5</i> transcription, thereby increasing CD8<sup>+</sup> T cell infiltration to restrain HCC progression. Furthermore, ADCY7 can be secreted as exosomes and enter neighbouring tumour cells to promote CCL5 induction. Exosomes with high ADCY7 levels promote intratumoural infiltration of CD8<sup>+</sup> T cells and suppress HCC tumour growth.</p><p><strong>Conclusion: </strong>We delineate the unconventional function and subcellular location of ADCY7, highlighting its pivotal role in T cell-mediated immunity in HCC and its potential as a promising treatment target.</p>","PeriodicalId":12825,"journal":{"name":"Gut","volume":null,"pages":null},"PeriodicalIF":23.0,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142345115","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-14DOI: 10.1136/gutjnl-2024-333611
Yiwen Yuan, Xi Fu, Yiqun Deng, Yu Sun
{"title":"Exploring the role of genetics, gut microbiota and blood metabolites in IBD.","authors":"Yiwen Yuan, Xi Fu, Yiqun Deng, Yu Sun","doi":"10.1136/gutjnl-2024-333611","DOIUrl":"10.1136/gutjnl-2024-333611","url":null,"abstract":"","PeriodicalId":12825,"journal":{"name":"Gut","volume":null,"pages":null},"PeriodicalIF":23.0,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142375387","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-10DOI: 10.1136/gutjnl-2024-332695
Anke M Onnekink, Myrte Gorris, Noor Lh Bekkali, Philip Bos, Paul Didden, J Enrique Dominguez-Muñoz, Pieter Friederich, Emo E van Halsema, Wouter L Hazen, Nadine C van Huijgevoort, Akin Inderson, Maarten Ajm Jacobs, Jan J Koornstra, Sjoerd Kuiken, Bob Ch Scheffer, Hilbert Sloterdijk, Ellert J van Soest, Niels G Venneman, Rogier P Voermans, Thomas R de Wijkerslooth, Janneke Wonders, Roeland Zoutendijk, Serge Jlb Zweers, Paul Fockens, Robert C Verdonk, Roy L J van Wanrooij, Jeanin E Van Hooft
Background: Endoscopic retrograde cholangiopancreatography (ERCP) with fully covered self-expandable metal stent (FCSEMS) placement is the preferred approach for biliary drainage in patients with suspected distal malignant biliary obstruction (MBO). However, FCSEMS placement is associated with a high risk of post-ERCP pancreatitis (PEP). Endoscopic sphincterotomy prior to FCSEMS placement may reduce PEP risk.
Objective: To compare endoscopic sphincterotomy to no sphincterotomy prior to FCSEMS placement.
Design: This multicentre, randomised, superiority trial was conducted in 17 hospitals and included patients with suspected distal MBO. Patients were randomised during ERCP to receive either endoscopic sphincterotomy (sphincterotomy group) or no sphincterotomy (control group) prior to FCSEMS placement. The primary outcome was PEP within 30 days. Secondary outcomes included procedure-related complications and 30-day mortality. An interim analysis was performed after 50% of patients (n=259) had completed follow-up.
Results: Between May 2016 and June 2023, 297 patients were included in the intention-to-treat analysis, with 156 in the sphincterotomy group and 141 in the control group. After the interim analysis, the study was terminated prematurely due to futility. PEP did not differ between groups, occurring in 26 patients (17%) in the sphincterotomy group compared with 30 patients (21%) in the control group (relative risk 0.78, 95% CI 0.49 to 1.26, p=0.37). There were no significant differences in bleeding, perforation, cholangitis, cholecystitis or 30-day mortality.
Conclusion: This trial found that endoscopic sphincterotomy was not superior to no sphincterotomy in reducing PEP in patients with distal MBO. Therefore, there was insufficient evidence to recommend routine endoscopic sphincterotomy prior to FCEMS placement.
{"title":"Endoscopic sphincterotomy to prevent post-ERCP pancreatitis after self-expandable metal stent placement for distal malignant biliary obstruction (SPHINX): a multicentre, randomised controlled trial.","authors":"Anke M Onnekink, Myrte Gorris, Noor Lh Bekkali, Philip Bos, Paul Didden, J Enrique Dominguez-Muñoz, Pieter Friederich, Emo E van Halsema, Wouter L Hazen, Nadine C van Huijgevoort, Akin Inderson, Maarten Ajm Jacobs, Jan J Koornstra, Sjoerd Kuiken, Bob Ch Scheffer, Hilbert Sloterdijk, Ellert J van Soest, Niels G Venneman, Rogier P Voermans, Thomas R de Wijkerslooth, Janneke Wonders, Roeland Zoutendijk, Serge Jlb Zweers, Paul Fockens, Robert C Verdonk, Roy L J van Wanrooij, Jeanin E Van Hooft","doi":"10.1136/gutjnl-2024-332695","DOIUrl":"https://doi.org/10.1136/gutjnl-2024-332695","url":null,"abstract":"<p><strong>Background: </strong>Endoscopic retrograde cholangiopancreatography (ERCP) with fully covered self-expandable metal stent (FCSEMS) placement is the preferred approach for biliary drainage in patients with suspected distal malignant biliary obstruction (MBO). However, FCSEMS placement is associated with a high risk of post-ERCP pancreatitis (PEP). Endoscopic sphincterotomy prior to FCSEMS placement may reduce PEP risk.</p><p><strong>Objective: </strong>To compare endoscopic sphincterotomy to no sphincterotomy prior to FCSEMS placement.</p><p><strong>Design: </strong>This multicentre, randomised, superiority trial was conducted in 17 hospitals and included patients with suspected distal MBO. Patients were randomised during ERCP to receive either endoscopic sphincterotomy (sphincterotomy group) or no sphincterotomy (control group) prior to FCSEMS placement. The primary outcome was PEP within 30 days. Secondary outcomes included procedure-related complications and 30-day mortality. An interim analysis was performed after 50% of patients (n=259) had completed follow-up.</p><p><strong>Results: </strong>Between May 2016 and June 2023, 297 patients were included in the intention-to-treat analysis, with 156 in the sphincterotomy group and 141 in the control group. After the interim analysis, the study was terminated prematurely due to futility. PEP did not differ between groups, occurring in 26 patients (17%) in the sphincterotomy group compared with 30 patients (21%) in the control group (relative risk 0.78, 95% CI 0.49 to 1.26, p=0.37). There were no significant differences in bleeding, perforation, cholangitis, cholecystitis or 30-day mortality.</p><p><strong>Conclusion: </strong>This trial found that endoscopic sphincterotomy was not superior to no sphincterotomy in reducing PEP in patients with distal MBO. Therefore, there was insufficient evidence to recommend routine endoscopic sphincterotomy prior to FCEMS placement.</p><p><strong>Trial registration number: </strong>NL5130.</p>","PeriodicalId":12825,"journal":{"name":"Gut","volume":null,"pages":null},"PeriodicalIF":23.0,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142400035","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-08DOI: 10.1136/gutjnl-2024-332526
David Z Pan, Cameron M Soulette, Abhishek Aggarwal, Dong Han, Nicholas van Buuren, Peiwen Wu, Becket Feierbach, Jaw-Town Lin, Cheng-Hao Tseng, Chi-Yi Chen, Bryan Downie, Hongmei Mo, Lauri Diehl, Li Li, Simon P Fletcher, Scott Balsitis, Ricardo Ramirez, Vithika Suri, Yao-Chun Hsu
Background The impact of nucleos(t)ide analogues on intrahepatic viral burden and immune microenvironment in patients with chronic hepatitis B (CHB) is not clear. Objective We aimed to characterise the effects of tenofovir disoproxil fumarate (TDF) on intrahepatic viral burden and the liver immune microenvironment in patients with CHB. Design Core liver biopsies were collected at baseline and year 3 from patients with CHB with minimally raised serum alanine aminotransferase in a double-blind placebo-controlled trial ([NCT01522625][1]). Paired biopsies were analysed by RNA-sequencing (n=119 pairs), a custom multiplex immunofluorescence assay (n=30 pairs), and HBV-targeted long-read DNA sequencing (n=49 pairs). Results Both non-integrated and integrated HBV DNA were present in all patients at baseline, with >65% having interchromosomal translocations. Treatment significantly reduced the frequency of HBV core+ hepatocytes and intrahepatic (integrated and non-integrated) HBV DNA, but had no effect on HBsAg+ hepatocytes. Clonally expanded integrations were enriched for HBsAg coding regions and showed dysregulation of nearby genes. At baseline, there was significant enrichment of intrahepatic CD8+ T cell proximity to HBV core+ hepatocytes, but not to HBsAg+ cells. The densities of T cells and B cells were significantly reduced by TDF. Transcriptomic analyses found TDF induced widespread downregulation of immune-related genes including inhibitory and regulatory genes. Conclusion TDF significantly reduced intrahepatic integrated and non-integrated HBV DNA, exerting disparate effects on HBV core+ and HBsAg+ cells and on different immune cell subsets. Our data suggest there may be differential cytotoxic T cell-mediated killing of HBV core+ versus HBsAg+ hepatocytes, providing insights for HBV cure strategies. Data are available upon reasonable request. Data collected for this study, including deidentified individual participant data and data dictionaries defining fields in the datasets, will be made available on request to with qualified external researchers based on submitted curriculum vitae and reflecting non conflict of interest as well as laws and regulations regarding patient privacy. Approval of such requests is dependent on the nature of the request, the merit of the research proposed, the availability of the data and the intended use of the data. Data requests should be sent to the corresponding author at holdenhsu@gmail.com. [1]: /lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT01522625&atom=%2Fgutjnl%2Fearly%2F2024%2F10%2F08%2Fgutjnl-2024-332526.atom
{"title":"Effects of tenofovir disoproxil fumarate on intrahepatic viral burden and liver immune microenvironment in patients with chronic hepatitis B","authors":"David Z Pan, Cameron M Soulette, Abhishek Aggarwal, Dong Han, Nicholas van Buuren, Peiwen Wu, Becket Feierbach, Jaw-Town Lin, Cheng-Hao Tseng, Chi-Yi Chen, Bryan Downie, Hongmei Mo, Lauri Diehl, Li Li, Simon P Fletcher, Scott Balsitis, Ricardo Ramirez, Vithika Suri, Yao-Chun Hsu","doi":"10.1136/gutjnl-2024-332526","DOIUrl":"https://doi.org/10.1136/gutjnl-2024-332526","url":null,"abstract":"Background The impact of nucleos(t)ide analogues on intrahepatic viral burden and immune microenvironment in patients with chronic hepatitis B (CHB) is not clear. Objective We aimed to characterise the effects of tenofovir disoproxil fumarate (TDF) on intrahepatic viral burden and the liver immune microenvironment in patients with CHB. Design Core liver biopsies were collected at baseline and year 3 from patients with CHB with minimally raised serum alanine aminotransferase in a double-blind placebo-controlled trial ([NCT01522625][1]). Paired biopsies were analysed by RNA-sequencing (n=119 pairs), a custom multiplex immunofluorescence assay (n=30 pairs), and HBV-targeted long-read DNA sequencing (n=49 pairs). Results Both non-integrated and integrated HBV DNA were present in all patients at baseline, with >65% having interchromosomal translocations. Treatment significantly reduced the frequency of HBV core+ hepatocytes and intrahepatic (integrated and non-integrated) HBV DNA, but had no effect on HBsAg+ hepatocytes. Clonally expanded integrations were enriched for HBsAg coding regions and showed dysregulation of nearby genes. At baseline, there was significant enrichment of intrahepatic CD8+ T cell proximity to HBV core+ hepatocytes, but not to HBsAg+ cells. The densities of T cells and B cells were significantly reduced by TDF. Transcriptomic analyses found TDF induced widespread downregulation of immune-related genes including inhibitory and regulatory genes. Conclusion TDF significantly reduced intrahepatic integrated and non-integrated HBV DNA, exerting disparate effects on HBV core+ and HBsAg+ cells and on different immune cell subsets. Our data suggest there may be differential cytotoxic T cell-mediated killing of HBV core+ versus HBsAg+ hepatocytes, providing insights for HBV cure strategies. Data are available upon reasonable request. Data collected for this study, including deidentified individual participant data and data dictionaries defining fields in the datasets, will be made available on request to with qualified external researchers based on submitted curriculum vitae and reflecting non conflict of interest as well as laws and regulations regarding patient privacy. Approval of such requests is dependent on the nature of the request, the merit of the research proposed, the availability of the data and the intended use of the data. Data requests should be sent to the corresponding author at holdenhsu@gmail.com. [1]: /lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT01522625&atom=%2Fgutjnl%2Fearly%2F2024%2F10%2F08%2Fgutjnl-2024-332526.atom","PeriodicalId":12825,"journal":{"name":"Gut","volume":null,"pages":null},"PeriodicalIF":24.5,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142385581","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}