Pub Date : 2024-08-29DOI: 10.1016/j.ccell.2024.08.001
Luigi Perelli, Giannicola Genovese, Giulio F. Draetta
In this issue of Cancer Cell, McIntyre et al. show that specific mutations in the KRAS proto-oncogene shape clinical progression of pancreatic ductal adenocarcinoma (PDAC). Importantly, they find that the KRASG12R mutation is enriched in early-stage PDAC, and it is characterized by distinctly activated molecular programs.
{"title":"The KRAS mutational spectrum and its clinical implications in pancreatic cancer","authors":"Luigi Perelli, Giannicola Genovese, Giulio F. Draetta","doi":"10.1016/j.ccell.2024.08.001","DOIUrl":"https://doi.org/10.1016/j.ccell.2024.08.001","url":null,"abstract":"<p>In this issue of <em>Cancer Cell</em>, McIntyre et al. show that specific mutations in the <em>KRAS</em> proto-oncogene shape clinical progression of pancreatic ductal adenocarcinoma (PDAC). Importantly, they find that the <em>KRAS</em><sup><em>G12R</em></sup> mutation is enriched in early-stage PDAC, and it is characterized by distinctly activated molecular programs.</p>","PeriodicalId":9670,"journal":{"name":"Cancer Cell","volume":"28 1","pages":""},"PeriodicalIF":50.3,"publicationDate":"2024-08-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142090508","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 : 2024-08-29DOI: 10.1016/j.ccell.2024.08.010
Vivek Subbiah, Razelle Kurzrock
Precision oncology endeavors to tailor therapies based on individual patient and tumor characteristics. This rapidly evolving field has transformed cancer treatment across all age groups. In this commentary, we review the application of precision oncology across different age groups, specifically in children, adolescents, and young adults, and emphasize that precision medicine is age and tissue agnostic.
{"title":"Precision oncology across the ages: Impact on children, adolescents, and young adults","authors":"Vivek Subbiah, Razelle Kurzrock","doi":"10.1016/j.ccell.2024.08.010","DOIUrl":"https://doi.org/10.1016/j.ccell.2024.08.010","url":null,"abstract":"<p>Precision oncology endeavors to tailor therapies based on individual patient and tumor characteristics. This rapidly evolving field has transformed cancer treatment across all age groups. In this commentary, we review the application of precision oncology across different age groups, specifically in children, adolescents, and young adults, and emphasize that precision medicine is age and tissue agnostic.</p>","PeriodicalId":9670,"journal":{"name":"Cancer Cell","volume":"50 1","pages":""},"PeriodicalIF":50.3,"publicationDate":"2024-08-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142090435","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 : 2024-08-29DOI: 10.1016/j.ccell.2024.08.005
Itay Tirosh, Mario L. Suva
Human tumors are intricate ecosystems composed of diverse genetic clones and malignant cell states that evolve in a complex tumor micro-environment. Single-cell RNA-sequencing (scRNA-seq) provides a compelling strategy to dissect this intricate biology and has enabled a revolution in our ability to understand tumor biology over the last ten years. Here we reflect on this first decade of scRNA-seq in human tumors and highlight some of the powerful insights gleaned from these studies. We first focus on computational approaches for robustly defining cancer cell states and their diversity and highlight some of the most common patterns of gene expression intra-tumor heterogeneity (eITH) observed across cancer types. We then discuss ambiguities in the field in defining and naming such eITH programs. Finally, we highlight critical developments that will facilitate future research and the broader implementation of these technologies in clinical settings.
{"title":"Cancer cell states: Lessons from ten years of single-cell RNA-sequencing of human tumors","authors":"Itay Tirosh, Mario L. Suva","doi":"10.1016/j.ccell.2024.08.005","DOIUrl":"https://doi.org/10.1016/j.ccell.2024.08.005","url":null,"abstract":"<p>Human tumors are intricate ecosystems composed of diverse genetic clones and malignant cell states that evolve in a complex tumor micro-environment. Single-cell RNA-sequencing (scRNA-seq) provides a compelling strategy to dissect this intricate biology and has enabled a revolution in our ability to understand tumor biology over the last ten years. Here we reflect on this first decade of scRNA-seq in human tumors and highlight some of the powerful insights gleaned from these studies. We first focus on computational approaches for robustly defining cancer cell states and their diversity and highlight some of the most common patterns of gene expression <em>intra</em>-tumor heterogeneity (eITH) observed across cancer types. We then discuss ambiguities in the field in defining and naming such eITH programs. Finally, we highlight critical developments that will facilitate future research and the broader implementation of these technologies in clinical settings.</p>","PeriodicalId":9670,"journal":{"name":"Cancer Cell","volume":"41 1","pages":""},"PeriodicalIF":50.3,"publicationDate":"2024-08-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142090446","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 : 2024-08-29DOI: 10.1016/j.ccell.2024.08.003
Ashwin Somasundaram, Hanna K. Sanoff
Neoadjuvant chemoradiotherapy has been a mainstay of the treatment of locally advanced rectal cancer. Programmed cell death protein 1 (PD-1) blockade therapy has demonstrated efficacy in combination with radiation. In this issue, Xiao et al. demonstrate promising efficacy with the addition of PD-1 blockade to neoadjuvant therapy for mismatch-repair proficient rectal cancer.
{"title":"PD-1 blockade is a promising therapeutic addition to neoadjuvant chemoradiation in locally advanced rectal cancer","authors":"Ashwin Somasundaram, Hanna K. Sanoff","doi":"10.1016/j.ccell.2024.08.003","DOIUrl":"https://doi.org/10.1016/j.ccell.2024.08.003","url":null,"abstract":"<p>Neoadjuvant chemoradiotherapy has been a mainstay of the treatment of locally advanced rectal cancer. Programmed cell death protein 1 (PD-1) blockade therapy has demonstrated efficacy in combination with radiation. In this issue, Xiao et al. demonstrate promising efficacy with the addition of PD-1 blockade to neoadjuvant therapy for mismatch-repair proficient rectal cancer.</p>","PeriodicalId":9670,"journal":{"name":"Cancer Cell","volume":"7 1","pages":""},"PeriodicalIF":50.3,"publicationDate":"2024-08-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142090445","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 : 2024-08-29DOI: 10.1016/j.ccell.2024.08.002
Caitlin A. McIntyre, Adrien Grimont, Jiwoon Park, Yinuo Meng, Whitney J. Sisso, Kenneth Seier, Gun Ho Jang, Henry Walch, Victoria G. Aveson, David J. Falvo, William B. Fall, Christopher W. Chan, Andrew Wenger, Brett L. Ecker, Alessandra Pulvirenti, Rebecca Gelfer, Maria Paz Zafra, Nikolaus Schultz, Wungki Park, Eileen M. O’Reilly, Rohit Chandwani
KRAS mutations in pancreatic ductal adenocarcinoma (PDAC) are suggested to vary in oncogenicity but the implications for human patients have not been explored in depth. We examined 1,360 consecutive PDAC patients undergoing surgical resection and find that KRASG12R mutations are enriched in early-stage (stage I) disease, owing not to smaller tumor size but increased node-negativity. KRASG12R tumors are associated with decreased distant recurrence and improved survival as compared to KRASG12D. To understand the biological underpinnings, we performed spatial profiling of 20 patients and bulk RNA-sequencing of 100 tumors, finding enhanced oncogenic signaling and epithelial-mesenchymal transition (EMT) in KRASG12D and increased nuclear factor κB (NF-κB) signaling in KRASG12R tumors. Orthogonal studies of mouse KrasG12R PDAC organoids show decreased migration and improved survival in orthotopic models. KRAS alterations in PDAC are thus associated with distinct presentation, clinical outcomes, and biological behavior, highlighting the prognostic value of mutational analysis and the importance of articulating mutation-specific PDAC biology.
{"title":"Distinct clinical outcomes and biological features of specific KRAS mutants in human pancreatic cancer","authors":"Caitlin A. McIntyre, Adrien Grimont, Jiwoon Park, Yinuo Meng, Whitney J. Sisso, Kenneth Seier, Gun Ho Jang, Henry Walch, Victoria G. Aveson, David J. Falvo, William B. Fall, Christopher W. Chan, Andrew Wenger, Brett L. Ecker, Alessandra Pulvirenti, Rebecca Gelfer, Maria Paz Zafra, Nikolaus Schultz, Wungki Park, Eileen M. O’Reilly, Rohit Chandwani","doi":"10.1016/j.ccell.2024.08.002","DOIUrl":"https://doi.org/10.1016/j.ccell.2024.08.002","url":null,"abstract":"<p><em>KRAS</em> mutations in pancreatic ductal adenocarcinoma (PDAC) are suggested to vary in oncogenicity but the implications for human patients have not been explored in depth. We examined 1,360 consecutive PDAC patients undergoing surgical resection and find that <em>KRAS</em><sup><em>G12R</em></sup> mutations are enriched in early-stage (stage I) disease, owing not to smaller tumor size but increased node-negativity. <em>KRAS</em><sup><em>G12R</em></sup> tumors are associated with decreased distant recurrence and improved survival as compared to <em>KRAS</em><sup><em>G12D</em></sup>. To understand the biological underpinnings, we performed spatial profiling of 20 patients and bulk RNA-sequencing of 100 tumors, finding enhanced oncogenic signaling and epithelial-mesenchymal transition (EMT) in <em>KRAS</em><sup><em>G12D</em></sup> and increased nuclear factor κB (NF-κB) signaling in <em>KRAS</em><sup><em>G12R</em></sup> tumors. Orthogonal studies of mouse <em>Kras</em><sup><em>G12R</em></sup> PDAC organoids show decreased migration and improved survival in orthotopic models. <em>KRAS</em> alterations in PDAC are thus associated with distinct presentation, clinical outcomes, and biological behavior, highlighting the prognostic value of mutational analysis and the importance of articulating mutation-specific PDAC biology.</p>","PeriodicalId":9670,"journal":{"name":"Cancer Cell","volume":"57 1","pages":""},"PeriodicalIF":50.3,"publicationDate":"2024-08-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142090447","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 : 2024-08-29DOI: 10.1016/j.ccell.2024.08.007
Kevin Wang, Paulina Coutifaris, David Brocks, Guanning Wang, Tarek Azar, Sabrina Solis, Ajeya Nandi, Shaneaka Anderson, Nicholas Han, Sasikanth Manne, Evgeny Kiner, Chirag Sachar, Minke Lucas, Sangeeth George, Patrick K. Yan, Melanie W. Kier, Amy I. Laughlin, Shawn Kothari, Josephine Giles, Divij Mathew, Alexander C. Huang
Combination checkpoint blockade with anti-PD-1 and anti-CTLA-4 antibodies has shown promising efficacy in melanoma. However, the underlying mechanism in humans remains unclear. Here, we perform paired single-cell RNA and T cell receptor (TCR) sequencing across time in 36 patients with stage IV melanoma treated with anti-PD-1, anti-CTLA-4, or combination therapy. We develop the algorithm Cyclone to track temporal clonal dynamics and underlying cell states. Checkpoint blockade induces waves of clonal T cell responses that peak at distinct time points. Combination therapy results in greater magnitude of clonal responses at 6 and 9 weeks compared to single-agent therapies, including melanoma-specific CD8+ T cells and exhausted CD8+ T cell (TEX) clones. Focused analyses of TEX identify that anti-CTLA-4 induces robust expansion and proliferation of progenitor TEX, which synergizes with anti-PD-1 to reinvigorate TEX during combination therapy. These next generation immune profiling approaches can guide the selection of drugs, schedule, and dosing for novel combination strategies.
抗PD-1和抗CTLA-4抗体联合检查点阻断疗法在黑色素瘤中显示出了良好的疗效。然而,人类的潜在机制仍不清楚。在这里,我们对36名接受抗PD-1、抗CTLA-4或联合疗法治疗的IV期黑色素瘤患者进行了成对单细胞RNA和T细胞受体(TCR)跨时间测序。我们开发了 Cyclone 算法来跟踪克隆的时间动态和潜在的细胞状态。检查点阻断诱导了一波波克隆 T 细胞反应,这些反应在不同的时间点达到峰值。与单药疗法相比,联合疗法能在6周和9周时产生更大规模的克隆反应,包括黑色素瘤特异性CD8+ T细胞和衰竭CD8+ T细胞(TEX)克隆。对TEX的集中分析表明,抗CTLA-4能诱导祖细胞TEX的强力扩增和增殖,它与抗PD-1协同作用,在联合疗法中为TEX注入新的活力。这些新一代免疫分析方法可以指导新型联合疗法的药物选择、疗程安排和剂量。
{"title":"Combination anti-PD-1 and anti-CTLA-4 therapy generates waves of clonal responses that include progenitor-exhausted CD8+ T cells","authors":"Kevin Wang, Paulina Coutifaris, David Brocks, Guanning Wang, Tarek Azar, Sabrina Solis, Ajeya Nandi, Shaneaka Anderson, Nicholas Han, Sasikanth Manne, Evgeny Kiner, Chirag Sachar, Minke Lucas, Sangeeth George, Patrick K. Yan, Melanie W. Kier, Amy I. Laughlin, Shawn Kothari, Josephine Giles, Divij Mathew, Alexander C. Huang","doi":"10.1016/j.ccell.2024.08.007","DOIUrl":"https://doi.org/10.1016/j.ccell.2024.08.007","url":null,"abstract":"<p>Combination checkpoint blockade with anti-PD-1 and anti-CTLA-4 antibodies has shown promising efficacy in melanoma. However, the underlying mechanism in humans remains unclear. Here, we perform paired single-cell RNA and T cell receptor (TCR) sequencing across time in 36 patients with stage IV melanoma treated with anti-PD-1, anti-CTLA-4, or combination therapy. We develop the algorithm <em>Cyclone</em> to track temporal clonal dynamics and underlying cell states. Checkpoint blockade induces waves of clonal T cell responses that peak at distinct time points. Combination therapy results in greater magnitude of clonal responses at 6 and 9 weeks compared to single-agent therapies, including melanoma-specific CD8<sup>+</sup> T cells and exhausted CD8<sup>+</sup> T cell (T<sub>EX</sub>) clones. Focused analyses of T<sub>EX</sub> identify that anti-CTLA-4 induces robust expansion and proliferation of progenitor T<sub>EX</sub>, which synergizes with anti-PD-1 to reinvigorate T<sub>EX</sub> during combination therapy. These next generation immune profiling approaches can guide the selection of drugs, schedule, and dosing for novel combination strategies.</p>","PeriodicalId":9670,"journal":{"name":"Cancer Cell","volume":"379 1","pages":""},"PeriodicalIF":50.3,"publicationDate":"2024-08-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142090513","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 : 2024-08-12DOI: 10.1016/j.ccell.2024.07.006
Tertiary lymphoid structures (TLSs) are associated with enhanced immunity in tumors. However, their formation and functions in colorectal cancer liver metastasis (CRLM) remain unclear. Here, we reveal that intra- and peri-tumor mature TLSs (TLS+) are associated with improved clinical outcomes than TLS− tumors. Using single-cell-RNA-sequencing and spatial-enhanced-resolution-omics-sequencing (Stereo-seq), we reveal that TLS+ tumors are enriched with IgG+ plasma cells (PCs), while TLS− tumors are characterized with IgA+ PCs. By generating TLS-associated PC-derived monoclonal antibodies in vitro, we show that TLS-PCs secrete tumor-targeting antibodies. As the proof-of-concept, we demonstrate the anti-tumor activities of TLS-PC-mAb6 antibody in humanized mouse model of colorectal cancer. We identify a fibroblast lineage secreting CCL19 that facilitates lymphocyte trafficking to TLSs. CCL19 treatment promotes TLS neogenesis and prevents tumor growth in mice. Our data uncover the central role of CCL19+ fibroblasts in TLS formation, which in turn generates therapeutic antibodies to restrict CRLM.
{"title":"CCL19-producing fibroblasts promote tertiary lymphoid structure formation enhancing anti-tumor IgG response in colorectal cancer liver metastasis","authors":"","doi":"10.1016/j.ccell.2024.07.006","DOIUrl":"https://doi.org/10.1016/j.ccell.2024.07.006","url":null,"abstract":"<p>Tertiary lymphoid structures (TLSs) are associated with enhanced immunity in tumors. However, their formation and functions in colorectal cancer liver metastasis (CRLM) remain unclear. Here, we reveal that intra- and peri-tumor mature TLSs (TLS+) are associated with improved clinical outcomes than TLS− tumors. Using single-cell-RNA-sequencing and spatial-enhanced-resolution-omics-sequencing (Stereo-seq), we reveal that TLS+ tumors are enriched with IgG<sup>+</sup> plasma cells (PCs), while TLS− tumors are characterized with IgA<sup>+</sup> PCs. By generating TLS-associated PC-derived monoclonal antibodies <em>in vitro</em>, we show that TLS-PCs secrete tumor-targeting antibodies. As the proof-of-concept, we demonstrate the anti-tumor activities of TLS-PC-mAb6 antibody in humanized mouse model of colorectal cancer. We identify a fibroblast lineage secreting CCL19 that facilitates lymphocyte trafficking to TLSs. CCL19 treatment promotes TLS neogenesis and prevents tumor growth in mice. Our data uncover the central role of CCL19<sup>+</sup> fibroblasts in TLS formation, which in turn generates therapeutic antibodies to restrict CRLM.</p>","PeriodicalId":9670,"journal":{"name":"Cancer Cell","volume":"83 1","pages":""},"PeriodicalIF":50.3,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141918674","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 : 2024-08-12DOI: 10.1016/j.ccell.2024.07.009
No Abstract
无摘要
{"title":"Development of Peptidomimetic Inhibitors of the ERG Gene Fusion Product in Prostate Cancer","authors":"","doi":"10.1016/j.ccell.2024.07.009","DOIUrl":"https://doi.org/10.1016/j.ccell.2024.07.009","url":null,"abstract":"No Abstract","PeriodicalId":9670,"journal":{"name":"Cancer Cell","volume":"19 1","pages":""},"PeriodicalIF":50.3,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141918685","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 : 2024-08-12DOI: 10.1016/j.ccell.2024.07.005
Changes in plasma and fecal metabolomes in colorectal cancer (CRC) progression (normal-adenoma-CRC) remain unclear. Here, plasma and fecal samples were collected from four independent cohorts of 1,251 individuals (422 CRC, 399 colorectal adenoma [CRA], and 430 normal controls [NC]). By metabolomic profiling, signature plasma and fecal metabolites with consistent shift across NC, CRA, and CRC are identified, including CRC-enriched oleic acid and CRC-depleted allocholic acid. Oleic acid exhibits pro-tumorigenic effects in CRC cells, patient-derived organoids, and two murine CRC models, whereas allocholic acid has opposing effects. By integrative analysis, we found that oleic acid or allocholic acid directly binds to α-enolase or farnesoid X receptor-1 in CRC cells, respectively, to modulate cancer-associated pathways. Clinically, we establish a panel of 17 plasma metabolites that accurately diagnoses CRC in a discovery and three validation cohorts (AUC = 0.848–0.987). Overall, we characterize metabolite signatures, mechanistic significance, and diagnostic potential of plasma and fecal metabolomes in CRC.
{"title":"Integrative plasma and fecal metabolomics identify functional metabolites in adenoma-colorectal cancer progression and as early diagnostic biomarkers","authors":"","doi":"10.1016/j.ccell.2024.07.005","DOIUrl":"https://doi.org/10.1016/j.ccell.2024.07.005","url":null,"abstract":"<p>Changes in plasma and fecal metabolomes in colorectal cancer (CRC) progression (normal-adenoma-CRC) remain unclear. Here, plasma and fecal samples were collected from four independent cohorts of 1,251 individuals (422 CRC, 399 colorectal adenoma [CRA], and 430 normal controls [NC]). By metabolomic profiling, signature plasma and fecal metabolites with consistent shift across NC, CRA, and CRC are identified, including CRC-enriched oleic acid and CRC-depleted allocholic acid. Oleic acid exhibits pro-tumorigenic effects in CRC cells, patient-derived organoids, and two murine CRC models, whereas allocholic acid has opposing effects. By integrative analysis, we found that oleic acid or allocholic acid directly binds to α-enolase or farnesoid X receptor-1 in CRC cells, respectively, to modulate cancer-associated pathways. Clinically, we establish a panel of 17 plasma metabolites that accurately diagnoses CRC in a discovery and three validation cohorts (AUC = 0.848–0.987). Overall, we characterize metabolite signatures, mechanistic significance, and diagnostic potential of plasma and fecal metabolomes in CRC.</p>","PeriodicalId":9670,"journal":{"name":"Cancer Cell","volume":"43 1","pages":""},"PeriodicalIF":50.3,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141918641","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}