Systematic analysis of markers for T-cell differentiation revealing CD95 as a novel biomarker for prognosis and immunotherapeutic efficacy in colon cancer

Yuxin Shi, Jie Mei, Rui Hou, Hao Wang, Junli Ding, Junying Xu
{"title":"Systematic analysis of markers for T-cell differentiation revealing CD95 as a novel biomarker for prognosis and immunotherapeutic efficacy in colon cancer","authors":"Yuxin Shi,&nbsp;Jie Mei,&nbsp;Rui Hou,&nbsp;Hao Wang,&nbsp;Junli Ding,&nbsp;Junying Xu","doi":"10.1002/mog2.57","DOIUrl":null,"url":null,"abstract":"<p>Colon cancer is the third most frequently diagnosed cancer worldwide. Considerable progress has been made in the therapeutic strategies and the accuracy of predicting the patients' prognosis. In the past decade, immunotherapy, represented by programmed cell death 1 (PD-1) and programmed cell death 1 ligand 1 (PD-L1) signaling inhibitors have made remarkable advances in many solid tumors, but limited effect in colon cancer. In particular, colon cancer demonstrates a remarkably poor response to immunotherapy compared with other cancers, with a notable exception of tumors harboring high microsatellite instability (MSI-H) or deficient mismatch repair (dMMR).<span><sup>1</sup></span> MSI-H or dMMR tumors are characterized by high mutational/neo-antigen burden, and an inflammatory tumor microenvironment with abundant tumor-infiltrating lymphocytes (TILs).<span><sup>2</sup></span> Although better efficacy could be achieved in a small group of patients with MSI-H or dMMR tumors, a proportion of other patients could benefit from immunotherapy. Thus, more appropriate biomarkers are needed to be discovered to predict the patients' prognosis and immunotherapeutic efficacy.</p><p>Different T-cell subsets in the tumor immune microenvironment (TIME) are closely related to the antitumor response and the prognosis of tumor patients. Tumor-specific CD8<sup>+</sup> T cells are the core cellular components that exert antitumor effects in TIME, by recognizing specific receptors on the tumor surface and secreting cytokines or via the Fas/FsaL pathway. Multiple T-cell subsets are characterized by respective differentiation markers. Currently, with the deepening research of T-cell subsets, the detection of relevant immune indicators and the formulation of therapeutic schedules have attracted increasing clinical attention. By monitoring the expression of T-cell differentiation markers in patients with colon cancer, the benefit of patients receiving immunotherapy can be further evaluated, thus providing a theoretical basis for judging the clinical prognosis and designing more feasible treatment plans.</p><p>In this research, we systematically analyzed the prognostic values of multiple markers for different T-cell subsets<span><sup>3</sup></span> using the Cancer Genome Atlas (TCGA) data set. Among all markers for different T cell subsets, we found that CD95 expression was correlated with better overall survival (OS) and progression-free survival (PFS), while BCL2 and CD122 were only correlated with PFS (Figure 1A,B). Therefore, CD95 was speculated to be associated with prognosis in colon cancer and was selected for further investigation and validation. The analysis of single-cell RNA-sequencing data set confirmed that CD95 was highly expressed in immune cells, especially in various T-cell subtypes (Figure S1A–C). CD95 is a member of the tumor necrosis factor receptor (TNF-R) superfamily and treated as characteristic marker of memeroy cells. It could also be used as the most classical death receptor to induce apoptosis. The CD95-induced apoptosis pathway is a powerful way for the immune system to clear virus-infected cells or tumor cells, therefore CD95 was initially identified as a tumor suppressor gene.</p><p>Next, to understand the potential biological role of CD95 in colon cancer, we applied Gene Set Enrichment Analysis (GSEA) in the TCGA data set. The results showed that CD95 was significantly related to multiple vital immune-related processes, such as allograft rejection, inflammatory response, and interferon-gamma response (Figure 1C and Table S1). Given the tight correlation between CD95 and immune-related processes in colon cancer, we next assessed the correlation between CD95 and comprehensive immunomodulators consisting of chemokines, receptors, MHC molecules, immunoinhibitors, and immunostimulators, which were associated with features of TIME and the responses to immunotherapy. The results showed that CD95 was positively correlated with most immunomodulators (Figure 1D and Table S2). In addition, we also analyzed the correlation between CD95 and T-cell subsets calculated by the ImmuCellAI tool, and the results showed that the strongest positive correlation was observed between CD95 and cytotoxic T cells (Table S3). All findings suggested CD95 was highly correlated with the inflamed TIME. In particular, substantial evidence suggests that patients with MSI-H/dMMR colon cancer benefit more from immunotherapy and have an optimistic immunotherapeutic efficacy compared with patients with proficient MMR (pMMR). Subsequently, we used three validated cohorts, namely the TCGA cohort, the GSE39582 cohort, and the CPTAC2 cohort, and the results indicated that CD95 was upregulated in tumors with MSI-H/dMMR status (Figure S2A–C). Moreover, the correlations between CD95 and several common mutations associated with immunotherapeutic response, including NF1, POLE, ARID1A, and BRAF, were also assessed. The results exhibited that tumor with gene mutations expressed higher CD95 (Figure S3A–D). Furthermore, a small-scale immunotherapy cohort consisting of 29 colon cancer specimens receiving immunotherapy was recruited to further validate the association between CD95 expression and immunotherapeutic response. Immunofluorescence exhibited that CD95 was expressed in immune cells and tumor cells (Figure 1E). Given the function of CD95 in T-cell differentiation, only CD95-positive immune cells were calculated. The results suggested that patients with well immunotherapeutic responses expressed high CD95 compared with those with poor response (Figure 1E). Finally, survival analysis showed patients with high CD95<sup>+</sup> immune cell infiltration exhibited well prognosis than that with low CD95<sup>+</sup> cell infiltration (Figure 1F). To sum up, these results supported that CD95 could serve as a novel and promising biomarker of benefit to immunotherapy in colon cancer.</p><p>CD95 and CD95L are widely expressed in colon cancer, previous studies in colon cancer showed that switching CD95 signaling from apoptotic to nonapoptotic ERK/NF-κB signaling facilitates tumor incidence, local growth, and the ability to metastasize to the liver, which depends on expression of CD95L. Furthermore, existing evidence suggests that CD95 mainly implements nonapoptotic signaling pathways such as NF-κB, MAPK, and PI3K that are involved in cell migration, differentiation, survival, and cytokine secretion. Not only in colon cancer, but in lung cancer, glioblastoma, ovarian cancer, and liver cancer, CD95 expression in some conditions conversely activates its nonapoptotic pathway, which plays an important role in tumorigenesis and progression.<span><sup>4</sup></span> In addition, the latest research showed that targeting CD95 in melanoma increased the levels of cytotoxic T cells and dendritic cells.<span><sup>5</sup></span> We hypothesized that CD95 may mediate the immunogenic death of tumor cells. However, the concrete effects of CD95 on tumor biological behavior and their relevance to immunotherapy efficacy are unclear. Based on the above finding that CD95 was positively associated with multiple immune-related processes and immunomodulators in the TIME, we speculated that in colon cancer CD95 was associated with inflamed TIME and identified immuno-hot tumors which favored antitumor immunity. Moreover, we also found that CD95 was upregulated in tumors with MSI-H/dMMR status. Colon tumors with dMMR status have association with a higher density of CD8<sup>+</sup> TILs and higher tumor mutation burden (TMB), which are qualities that seem to be biomarkers of immunotherapeutic response and demonstrated a positive prognostic value. Considering the high incidence and mortality of colon cancer and the dilemma in obtaining satisfactory treatment results, CD95 as an appropriate biomarker is needed to help identify candidates that could benefit from immunotherapy and can appraise clinical outcomes and therapeutic responses, which may make up for the deficiency of existing immunotherapy strategy.</p><p>In conclusion, the current study identified potential biomarkers to predict well clinical outcomes and response to immunotherapy in colon cancer. After systematic analysis, CD95, a memory marker of T cells, might be an appropriate and accurate biomarker in clinical practice to predict the prognosis and immunotherapeutic responses. Scientifically, the identification of novel predictive biomarkers will greatly expand our understanding of the mechanisms of colon cancer immunity, providing information on the decision of treatment strategies in individuals.</p><p><b>Yuxin Shi</b>: Data curation (equal); formal analysis (equal); methodology (equal); resources (equal); visualization (equal); writing—original draft (equal). <b>Jie Mei</b>: Data curation (equal); formal analysis (equal); investigation (equal); methodology (equal); validation (equal); writing—original draft (equal). <b>Rui Hou</b>: Formal analysis (supporting); validation (supporting). <b>Hao Wang</b>: Formal analysis (supporting); resources (supporting). <b>Junli Ding</b>: Conceptualization (equal); funding acquisition (supporting); project administration (equal); writing—review and editing (equal). <b>Junying Xu</b>: Conceptualization (equal); funding acquisition (lead); project administration (equal); writing—review and editing (equal). All authors have read and approved the final manuscript.</p><p>The authors declare no conflict of interest.</p><p>Ethical approval was granted by the Clinical Research Ethics Committee of Nanjing Medical University (2022-383). This is a retrospective study, and trial registration number is not needed. Written informed consent was obtained from all participants.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 4","pages":""},"PeriodicalIF":0.0000,"publicationDate":"2023-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.57","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"MedComm – Oncology","FirstCategoryId":"1085","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/mog2.57","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Colon cancer is the third most frequently diagnosed cancer worldwide. Considerable progress has been made in the therapeutic strategies and the accuracy of predicting the patients' prognosis. In the past decade, immunotherapy, represented by programmed cell death 1 (PD-1) and programmed cell death 1 ligand 1 (PD-L1) signaling inhibitors have made remarkable advances in many solid tumors, but limited effect in colon cancer. In particular, colon cancer demonstrates a remarkably poor response to immunotherapy compared with other cancers, with a notable exception of tumors harboring high microsatellite instability (MSI-H) or deficient mismatch repair (dMMR).1 MSI-H or dMMR tumors are characterized by high mutational/neo-antigen burden, and an inflammatory tumor microenvironment with abundant tumor-infiltrating lymphocytes (TILs).2 Although better efficacy could be achieved in a small group of patients with MSI-H or dMMR tumors, a proportion of other patients could benefit from immunotherapy. Thus, more appropriate biomarkers are needed to be discovered to predict the patients' prognosis and immunotherapeutic efficacy.

Different T-cell subsets in the tumor immune microenvironment (TIME) are closely related to the antitumor response and the prognosis of tumor patients. Tumor-specific CD8+ T cells are the core cellular components that exert antitumor effects in TIME, by recognizing specific receptors on the tumor surface and secreting cytokines or via the Fas/FsaL pathway. Multiple T-cell subsets are characterized by respective differentiation markers. Currently, with the deepening research of T-cell subsets, the detection of relevant immune indicators and the formulation of therapeutic schedules have attracted increasing clinical attention. By monitoring the expression of T-cell differentiation markers in patients with colon cancer, the benefit of patients receiving immunotherapy can be further evaluated, thus providing a theoretical basis for judging the clinical prognosis and designing more feasible treatment plans.

In this research, we systematically analyzed the prognostic values of multiple markers for different T-cell subsets3 using the Cancer Genome Atlas (TCGA) data set. Among all markers for different T cell subsets, we found that CD95 expression was correlated with better overall survival (OS) and progression-free survival (PFS), while BCL2 and CD122 were only correlated with PFS (Figure 1A,B). Therefore, CD95 was speculated to be associated with prognosis in colon cancer and was selected for further investigation and validation. The analysis of single-cell RNA-sequencing data set confirmed that CD95 was highly expressed in immune cells, especially in various T-cell subtypes (Figure S1A–C). CD95 is a member of the tumor necrosis factor receptor (TNF-R) superfamily and treated as characteristic marker of memeroy cells. It could also be used as the most classical death receptor to induce apoptosis. The CD95-induced apoptosis pathway is a powerful way for the immune system to clear virus-infected cells or tumor cells, therefore CD95 was initially identified as a tumor suppressor gene.

Next, to understand the potential biological role of CD95 in colon cancer, we applied Gene Set Enrichment Analysis (GSEA) in the TCGA data set. The results showed that CD95 was significantly related to multiple vital immune-related processes, such as allograft rejection, inflammatory response, and interferon-gamma response (Figure 1C and Table S1). Given the tight correlation between CD95 and immune-related processes in colon cancer, we next assessed the correlation between CD95 and comprehensive immunomodulators consisting of chemokines, receptors, MHC molecules, immunoinhibitors, and immunostimulators, which were associated with features of TIME and the responses to immunotherapy. The results showed that CD95 was positively correlated with most immunomodulators (Figure 1D and Table S2). In addition, we also analyzed the correlation between CD95 and T-cell subsets calculated by the ImmuCellAI tool, and the results showed that the strongest positive correlation was observed between CD95 and cytotoxic T cells (Table S3). All findings suggested CD95 was highly correlated with the inflamed TIME. In particular, substantial evidence suggests that patients with MSI-H/dMMR colon cancer benefit more from immunotherapy and have an optimistic immunotherapeutic efficacy compared with patients with proficient MMR (pMMR). Subsequently, we used three validated cohorts, namely the TCGA cohort, the GSE39582 cohort, and the CPTAC2 cohort, and the results indicated that CD95 was upregulated in tumors with MSI-H/dMMR status (Figure S2A–C). Moreover, the correlations between CD95 and several common mutations associated with immunotherapeutic response, including NF1, POLE, ARID1A, and BRAF, were also assessed. The results exhibited that tumor with gene mutations expressed higher CD95 (Figure S3A–D). Furthermore, a small-scale immunotherapy cohort consisting of 29 colon cancer specimens receiving immunotherapy was recruited to further validate the association between CD95 expression and immunotherapeutic response. Immunofluorescence exhibited that CD95 was expressed in immune cells and tumor cells (Figure 1E). Given the function of CD95 in T-cell differentiation, only CD95-positive immune cells were calculated. The results suggested that patients with well immunotherapeutic responses expressed high CD95 compared with those with poor response (Figure 1E). Finally, survival analysis showed patients with high CD95+ immune cell infiltration exhibited well prognosis than that with low CD95+ cell infiltration (Figure 1F). To sum up, these results supported that CD95 could serve as a novel and promising biomarker of benefit to immunotherapy in colon cancer.

CD95 and CD95L are widely expressed in colon cancer, previous studies in colon cancer showed that switching CD95 signaling from apoptotic to nonapoptotic ERK/NF-κB signaling facilitates tumor incidence, local growth, and the ability to metastasize to the liver, which depends on expression of CD95L. Furthermore, existing evidence suggests that CD95 mainly implements nonapoptotic signaling pathways such as NF-κB, MAPK, and PI3K that are involved in cell migration, differentiation, survival, and cytokine secretion. Not only in colon cancer, but in lung cancer, glioblastoma, ovarian cancer, and liver cancer, CD95 expression in some conditions conversely activates its nonapoptotic pathway, which plays an important role in tumorigenesis and progression.4 In addition, the latest research showed that targeting CD95 in melanoma increased the levels of cytotoxic T cells and dendritic cells.5 We hypothesized that CD95 may mediate the immunogenic death of tumor cells. However, the concrete effects of CD95 on tumor biological behavior and their relevance to immunotherapy efficacy are unclear. Based on the above finding that CD95 was positively associated with multiple immune-related processes and immunomodulators in the TIME, we speculated that in colon cancer CD95 was associated with inflamed TIME and identified immuno-hot tumors which favored antitumor immunity. Moreover, we also found that CD95 was upregulated in tumors with MSI-H/dMMR status. Colon tumors with dMMR status have association with a higher density of CD8+ TILs and higher tumor mutation burden (TMB), which are qualities that seem to be biomarkers of immunotherapeutic response and demonstrated a positive prognostic value. Considering the high incidence and mortality of colon cancer and the dilemma in obtaining satisfactory treatment results, CD95 as an appropriate biomarker is needed to help identify candidates that could benefit from immunotherapy and can appraise clinical outcomes and therapeutic responses, which may make up for the deficiency of existing immunotherapy strategy.

In conclusion, the current study identified potential biomarkers to predict well clinical outcomes and response to immunotherapy in colon cancer. After systematic analysis, CD95, a memory marker of T cells, might be an appropriate and accurate biomarker in clinical practice to predict the prognosis and immunotherapeutic responses. Scientifically, the identification of novel predictive biomarkers will greatly expand our understanding of the mechanisms of colon cancer immunity, providing information on the decision of treatment strategies in individuals.

Yuxin Shi: Data curation (equal); formal analysis (equal); methodology (equal); resources (equal); visualization (equal); writing—original draft (equal). Jie Mei: Data curation (equal); formal analysis (equal); investigation (equal); methodology (equal); validation (equal); writing—original draft (equal). Rui Hou: Formal analysis (supporting); validation (supporting). Hao Wang: Formal analysis (supporting); resources (supporting). Junli Ding: Conceptualization (equal); funding acquisition (supporting); project administration (equal); writing—review and editing (equal). Junying Xu: Conceptualization (equal); funding acquisition (lead); project administration (equal); writing—review and editing (equal). All authors have read and approved the final manuscript.

The authors declare no conflict of interest.

Ethical approval was granted by the Clinical Research Ethics Committee of Nanjing Medical University (2022-383). This is a retrospective study, and trial registration number is not needed. Written informed consent was obtained from all participants.

Abstract Image

t细胞分化标志物的系统分析揭示CD95作为结肠癌预后和免疫治疗疗效的新生物标志物
结肠癌是世界上第三大最常见的癌症。在治疗策略和预测患者预后的准确性方面取得了长足的进步。在过去的十年中,以程序性细胞死亡1 (PD-1)和程序性细胞死亡1配体1 (PD-L1)信号抑制剂为代表的免疫治疗在许多实体肿瘤中取得了显著进展,但在结肠癌中的效果有限。特别是,与其他癌症相比,结肠癌对免疫治疗的反应明显较差,但具有高微卫星不稳定性(MSI-H)或错配修复缺陷(dMMR)的肿瘤除外MSI-H或dMMR肿瘤的特点是高突变/新抗原负荷,肿瘤微环境具有丰富的肿瘤浸润淋巴细胞(til)虽然在一小部分MSI-H或dMMR肿瘤患者中可以获得更好的疗效,但也有一部分其他患者可以从免疫治疗中获益。因此,需要发现更合适的生物标志物来预测患者的预后和免疫治疗效果。肿瘤免疫微环境(TIME)中不同的t细胞亚群与肿瘤患者的抗肿瘤反应和预后密切相关。肿瘤特异性CD8+ T细胞通过识别肿瘤表面的特异性受体并分泌细胞因子或通过Fas/FsaL通路,是TIME中发挥抗肿瘤作用的核心细胞成分。多个t细胞亚群由各自的分化标记物表征。目前,随着t细胞亚群研究的不断深入,相关免疫指标的检测和治疗方案的制定越来越受到临床的重视。通过监测结肠癌患者t细胞分化标志物的表达,可以进一步评估患者接受免疫治疗的获益,从而为判断临床预后和设计更可行的治疗方案提供理论依据。在这项研究中,我们使用癌症基因组图谱(TCGA)数据集系统地分析了多个标记物对不同t细胞亚群的预后价值。在不同T细胞亚群的所有标记物中,我们发现CD95表达与更好的总生存期(OS)和无进展生存期(PFS)相关,而BCL2和CD122仅与PFS相关(图1A,B)。因此,我们推测CD95与结肠癌的预后相关,并选择CD95进行进一步的研究和验证。单细胞rna测序数据集分析证实,CD95在免疫细胞中高度表达,特别是在各种t细胞亚型中(图S1A-C)。CD95是肿瘤坏死因子受体(TNF-R)超家族的成员,被认为是记忆细胞的特征标志物。它也可以作为诱导细胞凋亡的最经典的死亡受体。CD95诱导的细胞凋亡途径是免疫系统清除病毒感染细胞或肿瘤细胞的有力途径,因此CD95最初被确定为肿瘤抑制基因。接下来,为了了解CD95在结肠癌中的潜在生物学作用,我们在TCGA数据集中应用了基因集富集分析(GSEA)。结果显示,CD95与多种重要的免疫相关过程显著相关,如同种异体移植排斥反应、炎症反应和干扰素- γ反应(图1C和表S1)。鉴于CD95与结肠癌免疫相关过程之间的密切相关性,我们接下来评估了CD95与综合免疫调节剂(包括趋化因子、受体、MHC分子、免疫抑制剂和免疫刺激剂)之间的相关性,这些免疫调节剂与TIME特征和免疫治疗反应相关。结果显示,CD95与大多数免疫调节剂呈正相关(图1D和表S2)。此外,我们还分析了CD95与ImmuCellAI工具计算的T细胞亚群之间的相关性,结果显示CD95与细胞毒性T细胞之间存在最强的正相关性(表S3)。所有的研究结果都表明CD95与发炎的TIME高度相关。特别是,大量证据表明,与熟练MMR (pMMR)患者相比,MSI-H/dMMR结肠癌患者从免疫治疗中获益更多,且免疫治疗效果乐观。随后,我们使用了三个经过验证的队列,即TCGA队列、GSE39582队列和CPTAC2队列,结果表明CD95在MSI-H/dMMR状态的肿瘤中上调(图S2A-C)。此外,还评估了CD95与几种与免疫治疗反应相关的常见突变(包括NF1、POLE、ARID1A和BRAF)之间的相关性。结果显示,基因突变的肿瘤表达更高的CD95(图S3A-D)。 此外,招募了一个由29个接受免疫治疗的结肠癌标本组成的小规模免疫治疗队列,以进一步验证CD95表达与免疫治疗反应之间的关联。免疫荧光显示CD95在免疫细胞和肿瘤细胞中均有表达(图1E)。考虑到CD95在t细胞分化中的作用,我们只计算CD95阳性的免疫细胞。结果表明,免疫治疗反应良好的患者与反应较差的患者相比,CD95表达较高(图1E)。最后,生存分析显示,CD95+免疫细胞浸润高的患者比CD95+免疫细胞浸润低的患者预后好(图1F)。综上所述,这些结果支持CD95可以作为一种新的有前景的生物标志物,对结肠癌的免疫治疗有益。CD95和CD95L在结肠癌中广泛表达,以往在结肠癌中的研究表明,将CD95信号从凋亡转换为非凋亡的ERK/NF-κB信号有利于肿瘤的发生、局部生长和向肝脏转移的能力,这些都依赖于CD95L的表达。此外,已有证据表明CD95主要实现NF-κB、MAPK和PI3K等非凋亡信号通路,参与细胞迁移、分化、存活和细胞因子分泌。不仅在结肠癌中,在肺癌、胶质母细胞瘤、卵巢癌和肝癌中,CD95在某些情况下的表达反过来激活其非凋亡通路,在肿瘤的发生和进展中发挥重要作用此外,最新的研究表明,靶向CD95在黑色素瘤中增加了细胞毒性T细胞和树突状细胞的水平我们假设CD95可能介导肿瘤细胞的免疫原性死亡。然而,CD95对肿瘤生物学行为的具体影响及其与免疫治疗疗效的相关性尚不清楚。基于上述发现CD95与TIME中多个免疫相关过程和免疫调节剂呈正相关,我们推测在结肠癌中CD95与炎症性TIME相关,并鉴定出有利于抗肿瘤免疫的免疫热肿瘤。此外,我们还发现CD95在MSI-H/dMMR状态的肿瘤中表达上调。具有dMMR状态的结肠肿瘤与较高的CD8+ TILs密度和较高的肿瘤突变负荷(TMB)相关,这些特性似乎是免疫治疗反应的生物标志物,并显示出积极的预后价值。考虑到结肠癌的高发病率和死亡率以及难以获得令人满意的治疗结果,需要CD95作为一种合适的生物标志物来帮助识别可能受益于免疫治疗的候选人,并可以评估临床结果和治疗反应,这可能弥补现有免疫治疗策略的不足。总之,目前的研究确定了潜在的生物标志物来预测结肠癌的临床结果和对免疫治疗的反应。经系统分析,CD95作为T细胞的记忆标记物,在临床应用中可能是预测预后和免疫治疗反应的合适、准确的生物标志物。科学地说,新的预测性生物标志物的鉴定将极大地扩展我们对结肠癌免疫机制的理解,为个体治疗策略的决策提供信息。史宇昕:数据策展(equal);形式分析(相等);方法(平等);资源(平等);可视化(平等);写作-原稿(同等)。杰梅:数据管理(平等);形式分析(相等);调查(平等);方法(平等);验证(平等);写作-原稿(同等)。侯睿:形式分析(支持);验证(支持)。王浩:形式分析(支持);资源(支持)。丁俊丽:概念化(平等);资金获取(支持);项目管理(同等);写作—评审与编辑(同等)。许俊英:概念化(平等);获得资金(牵头);项目管理(同等);写作—评审与编辑(同等)。所有作者都阅读并批准了最终稿件。作者声明无利益冲突。经南京医科大学临床研究伦理委员会(2022-383)伦理批准。这是一项回顾性研究,不需要试验注册号。所有参与者均获得书面知情同意。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
自引率
0.00%
发文量
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信