programmed cell death 1 receptor

程序性细胞死亡 1 受体
  • 文章类型: Journal Article
    抗血管生成药物和免疫检查点抑制剂(ICIs)在肿瘤治疗中的组合正在成为改善ICIs抗性肿瘤治疗的一种方法。此外,肠道微生物(GM)参与肿瘤微环境中的血管生成,也与免疫检查点抑制剂的抗肿瘤功能有关。然而,目前尚不清楚在抗血管生成药物和免疫检查点抑制剂联合治疗癌症时,肠道微生物是否在抗肿瘤功能中发挥作用.内皮抑素,血管生成抑制剂,已被广泛用作癌症的抗血管生成疗法。我们表明与编码人内皮抑素的腺病毒联合治疗,名为Ad-E,和PD-1阻断显著消除MC38肿瘤生长。联合处理后小鼠肠道微生物结构发生改变。我们发现,消除肠道微生物后,联合治疗的抗肿瘤功能受到抑制。在微生物群耗尽的小鼠中,脆弱拟杆菌的口服灌胃在一定程度上挽救了Ad-E和αPD-1单克隆抗体(mAb)的抗肿瘤作用。Further,脆弱拟杆菌可以改善CD3+T细胞,NK细胞,和IFNγ+CD8+T细胞在肿瘤微环境中抑制肿瘤生长。此外,脆弱拟杆菌可能通过下调异丁酸(IBA)恢复抗肿瘤功能。我们的结果表明,GM可能参与Ad-E和αPD-1mAb的联合治疗癌症,这对肿瘤生长动力学和癌症免疫监视具有肿瘤学意义。
    The combination of anti-angiogenic drugs and immune checkpoint inhibitors (ICIs) in the treatment of tumors is emerging as a way to improve ICIs-resistant tumor therapy. In addition, gut microbes (GMs) are involved in angiogenesis in the tumor microenvironment and are also associated with the antitumor function of immune checkpoint inhibitors. However, it is unclear whether gut microbes have a role in anti-tumor function in the combination of anti-angiogenic drugs and immune checkpoint inhibitors for cancer treatment. Endostatin, an angiogenesis inhibitor, has been widely used as an antiangiogenic therapy for cancer. We showed that combined therapy with an adenovirus encoding human endostatin, named Ad-E, and PD-1 blockade dramatically abrogated MC38 tumor growth. The structure of intestinal microbes in mice was changed after combination treatment. We found that the antitumor function of combination therapy was inhibited after the elimination of intestinal microbes. In mice with depleted microbiota, oral gavage of Bacteroides fragilis salvaged the antitumor effects of combination Ad-E and αPD-1 monoclonal antibody (mAb) to a certain extent. Further, Bacteroides fragilis could improve CD3+T cells, NK cells, and IFNγ+CD8+ T cells in the tumor microenvironment to inhibit tumor growth. Besides, Bacteroides fragilis might restore antitumor function by down-regulating isobutyric acid (IBA). Our results suggested that GMs may be involved in the combination of Ad-E and αPD-1 mAb for cancer treatment, which has oncological implications for tumor growth dynamics and cancer immune surveillance.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    前列腺癌表现为免疫学上的“冷”恶性肿瘤,其特征是大多数患者对免疫疗法缺乏反应。前列腺肿瘤代谢功能障碍被认为是免疫逃避的关键因素。导致免疫治疗干预措施的有效性降低。尽管有这种意识,前列腺癌代谢失调的确切分子机制及其与免疫逃避的复杂关系仍未完全阐明。在这项研究中,我们介绍了多药耐药蛋白ABCC4/MRP4作为在前列腺癌中显著表达的关键角色,在抑制肿瘤内CD8+T细胞的活性中发挥关键作用。前列腺癌细胞中ABCC4的耗尽阻止了前列腺素E2(PGE2)的释放,一种减少CD8+T细胞数量并降低其细胞毒性的分子。相反,抑制CD8+T细胞中PGE2信号的激活有效地提高了PD-1阻断前列腺癌治疗的疗效.在这个过程中,JAK1-STAT3通路的下调和线粒体去极化是导致T细胞无反应的关键因素。总的来说,我们的研究将ABCC4-PGE2轴确定为逆转肿瘤浸润淋巴细胞(TIL)内功能障碍和增强前列腺癌对免疫疗法的次优反应性的有前景的靶标.
    Prostate cancer presents as an immunologically \"cold\" malignancy, characterized by a lack of response to immunotherapy in the majority of patients. The dysfunction of prostate tumor metabolism is recognized as a critical factor in immune evasion, resulting in reduced effectiveness of immunotherapeutic interventions. Despite this awareness, the precise molecular mechanisms underpinning metabolic dysregulation in prostate cancer and its intricate relationship with immune evasion remain incompletely elucidated. In this study, we introduce the multi-drug resistance protein ABCC4/MRP4 as a key player prominently expressed in prostate cancer, exerting a pivotal role in suppressing the activity of intratumoral CD8+ T cells. Depletion of ABCC4 in prostate cancer cells halts the release of prostaglandin E2 (PGE2), a molecule that diminishes the population of CD8+ T cells and curtails their cytotoxic capabilities. Conversely, constraining the activation of PGE2 signaling in CD8+ T cells effectively improved the efficacy of prostate cancer treatment with PD-1 blockade. During this process, downregulation of the JAK1-STAT3 pathway and depolarization of mitochondria emerge as crucial factors contributing to T cell anergy. Collectively, our research identifies the ABCC4-PGE2 axis as a promising target for reversing dysfunction within tumor-infiltrating lymphocytes (TILs) and augmenting the suboptimal responsiveness to immunotherapy in prostate cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    光疗是一种有前途的抗肿瘤方式,其中包括光热疗法(PTT)和光动力疗法(PDT)。然而,肿瘤局部缺氧极大地阻碍了光疗的疗效,吲哚胺2,3-双加氧酶(IDO)和程序性细胞死亡配体-1(PD-L1)在肿瘤细胞上的过表达。为了解决这些问题,自组装多功能聚合物胶束(RIMNA)被开发用于共同递送光敏剂吲哚菁绿(ICG),氧合器MnO2,IDO抑制剂NLG919和toll样受体4激动剂单磷酰脂质A(MPLA)。值得注意的是,RIMNA聚合物胶束具有良好的稳定性,均匀的形态,优越的生物相容性,并加强PTT/PDT效应。更重要的是,RIMNA介导的IDO抑制与程序性死亡受体1(PD-1)/PD-L1阻断相结合,显着改善了免疫抑制并促进了免疫激活。基于RIMNA的光免疫疗法与PD-1抗体协同作用可显著抑制原发肿瘤增殖,以及刺激免疫力,极大地抑制肺转移和远处肿瘤的生长。这项研究提供了一种有效的方法来加强光疗的疗效和减轻免疫抑制,从而为癌症治疗带来临床益处。
    Phototherapy is a promising antitumor modality, which consists of photothermal therapy (PTT) and photodynamic therapy (PDT). However, the efficacy of phototherapy is dramatically hampered by local hypoxia in tumors, overexpression of indoleamine 2,3-dioxygenase (IDO) and programmed cell death ligand-1 (PD-L1) on tumor cells. To address these issues, self-assembled multifunctional polymeric micelles (RIMNA) were developed to co-deliver photosensitizer indocyanine green (ICG), oxygenator MnO2, IDO inhibitor NLG919, and toll-like receptor 4 agonist monophosphoryl lipid A (MPLA). It is worth noting that RIMNA polymeric micelles had good stability, uniform morphology, superior biocompatibility, and intensified PTT/PDT effect. What\'s more, RIMNA-mediated IDO inhibition combined with programmed death receptor-1 (PD-1)/PD-L1 blockade considerably improved immunosuppression and promoted immune activation. RIMNA-based photoimmunotherapy synergized with PD-1 antibody could remarkably inhibit primary tumor proliferation, as well as stimulate the immunity to greatly suppress lung metastasis and distant tumor growth. This study offers an efficient method to reinforce the efficacy of phototherapy and alleviate immunosuppression, thereby bringing clinical benefits to cancer treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    目的:了解食管腺癌(OAC)的CD8+肿瘤浸润淋巴细胞(TIL)区隔与淋巴细胞耗竭的关系,组织驻留,并确定对治疗的不同反应背后的可能原因。
    方法:通过流式细胞术评估了44例接受OAC根治性切除术的患者的肿瘤样本中是否存在抗原经历的TIL以及激活和耗尽的标志物。对PD-1和CD39阳性OACTIL的群体进行排序,和使用改进的SmartSeq2方案进行的批量RNA测序。完成功能的流式细胞术评估。
    结果:经历CD8+OACTILs的抗原比例较高与手术后生存率提高有关;而,在这些TIL中,PD-1和CD39的高双阳性(DP)也与结局显著相关.这些DPTIL具有少数群体,其对耗竭标记TIM3和LAG3呈阳性。对PD-1和CD39DPTIL的转录组学评估表明,与其他癌症生存率提高相关的组织常驻记忆T淋巴细胞(TRM)表型富集。通过流式细胞术对典型TRM标记CD103的阳性增强。这个群体在他们的转录组学谱中都表现出维持的功能能力,在流式细胞术评估中,以及保留的增殖能力。
    结论:切除的OAC被PD-1和CD39DPTIL不同程度地浸润,淋巴细胞中的丰富与存活率的提高有关。这个DP人口增加了,但仍然谦虚,与DN相比,TIM3和LAG3阳性的频率,并且与功能性有能力的TRM表型保持一致。
    OBJECTIVE: To understand the CD8+ tumour infiltrating lymphocyte (TIL) compartment of oesophageal adenocarcinoma (OAC) with regards to markers of lymphocyte exhaustion, tissue residency and to identify possible reasons behind differential responses to therapy.
    METHODS: Tumour samples from 44 patients undergoing curative resection for OAC were assessed by flow cytometry for presence of antigen-experienced TILs and markers of activation and exhaustion. Populations of PD-1 and CD39 positive OAC TILs were sorted, and bulk RNA sequencing undertaken using a modified SmartSeq2 protocol. Flow cytometric assessment of functionality was completed.
    RESULTS: A higher proportion of antigen experienced CD8+ OAC TILs was associated with improved survival following surgery; while, high double positivity (DP) for PD-1 and CD39 among these TILs also correlated significantly with outcome. These DP TILs possess a minority population which is positive for the markers of exhaustion TIM3 and LAG3. Transcriptomic assessment of the PD-1 and CD39 DP TILs demonstrated enrichment for a tissue resident memory T lymphocyte (TRM) phenotype associated with improved survival in other cancers, reinforced by positivity for the canonical TRM marker CD103 by flow cytometry. This population demonstrated maintained functional capacity both in their transcriptomic profile, and on flow cytometric assessment, as well as preserved proliferative capacity.
    CONCLUSIONS: Resected OAC are variably infiltrated by PD-1 and CD39 DP TILs, an abundance of which among lymphocytes is associated with improved survival. This DP population has an increased, but still modest, frequency of TIM3 and LAG3 positivity compared to DN, and is in keeping with a functionally competent TRM phenotype.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:尽管ICI单药治疗效果良好,大多数非小细胞肺癌(NSCLC)患者无反应.因此,确定能够从ICI治疗中最佳获益的患者仍然是一个挑战.
    方法:在183名接受ICI单药治疗的晚期或复发NSCLC患者中,我们分析了110例治疗前和治疗后血浆样本可用的患者.在ICI开始和6周后测量73种可溶性免疫介质。为了识别有用的生物标志物,我们分析了治疗前水平和治疗中可溶性免疫介质的变化与患者生存率的关系.治疗前或治疗中的生物标志物与irAE发展的关联,PD-L1表达,CD8+TIL密度,还分析了中性粒细胞与淋巴细胞比率(NLR)。
    结果:单变量分析表明,治疗前生物标志物包括6种免疫介质,而治疗中的生物标志物包括8种免疫介质.多因素分析显示,治疗前生物标志物包括4种免疫介质(CCL19、CCL21、CXCL5、CXCL10),而治疗中的生物标志物包括5种免疫介质(CCL7,CCL19,CCL23,CCL25,IL-32).IrAE的发展与CCL23的治疗改变有关。PD-L1表达与TNFSF13B的治疗前水平和CCL25的治疗中变化相关。CD8+TIL密度与治疗前CXCL10水平相关,而NLR与治疗前CCL13和CCL17水平相关。
    结论:我们确定了几种可溶性免疫介质作为ICI单药治疗的NSCLC患者治疗前和治疗中的生存生物标志物。这些生物标志物中的一些与其他可能的预测因子相关,包括IRAE开发,PD-L1表达,CD8+TIL密度和NLR。需要进一步的大规模研究来建立接受ICI单药治疗的NSCLC患者的生物标志物。
    BACKGROUND: Despite the favorable therapeutic efficacy observed with ICI monotherapy, the majority of non-small cell lung cancer (NSCLC) patients do not respond. Therefore, identifying patients who could optimally benefit from ICI treatment remains a challenge.
    METHODS: Among 183 patients with advanced or recurrent NSCLC who received ICI monotherapy, we analyzed 110 patients whose pre- and post-treatment plasma samples were available. Seventy-three soluble immune mediators were measured at ICI initiation and 6 weeks later. To identify useful biomarkers, we analyzed the association of pre-treatment levels and on-treatment changes of soluble immune mediators with survival of patients. The associations of pre-treatment or on-treatment biomarkers with irAE development, PD-L1 expression, CD8+ TIL density, and neutrophil to lymphocyte ratio (NLR) were also analyzed.
    RESULTS: Univariate analysis showed that pre-treatment biomarkers included 6 immune mediators, whereas on-treatment biomarkers included 8 immune mediators. Multivariate analysis showed that pre-treatment biomarkers included 4 immune mediators (CCL19, CCL21, CXCL5, CXCL10), whereas on-treatment biomarkers included 5 immune mediators (CCL7, CCL19, CCL23, CCL25, IL-32). IrAE development was associated with on-treatment change in CCL23. PD-L1 expression was associated with the pre-treatment levels of TNFSF13B and the on-treatment change in CCL25. CD8+ TIL density was associated with the pre-treatment CXCL10 level, whereas NLR was correlated with pre-treatment levels of CCL13 and CCL17.
    CONCLUSIONS: We identified several soluble immune mediators as pre-treatment and on-treatment biomarkers of survival in patients with NSCLC treated with ICI monotherapy. Some of these biomarkers were associated with other possible predictors, including irAE development, PD-L1 expression, CD8+ TIL density and NLR. Further large-scale studies are needed to establish biomarkers for patients with NSCLC who received ICI monotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    淋巴细胞活化基因(Lag)-3是癌症的抑制性共受体和免疫检查点抑制剂(ICI)治疗的靶标。分析了Lag-3在T细胞激活后的免疫突触处的动态行为以阐明Lag-3抑制机制。Lag-3形成簇并且在T细胞活化后与T细胞受体微簇(TCR-MC)共定位,类似于PD-1。Lag-3阻断抗体(Abs)抑制Lag-3和TCR-MC之间的共定位而不抑制Lag-3簇形成。Lag-3还抑制MHC-II非依赖性刺激和Lag-3Ab,不阻断MHC-II结合的化合物仍然可以阻断Lag-3的抑制功能,这表明Lag-3Ab通过解离TCR-MC和Lag-3簇的共组装来阻断Lag-3抑制信号。与PD-1和Lag-3Ab增强T细胞反应的组合益处一致,双特异性Lag-3/PD-1拮抗剂有效抑制簇形成和PD-1和Lag-3与TCR-MC的共定位。因此,Lag-3抑制TCR-MC的T细胞活化,Lag-3ICI的目标是解离Lag-3与TCR-MC的共定位。
    Lymphocyte activation gene (Lag)-3 is an inhibitory co-receptor and target of immune checkpoint inhibitor (ICI) therapy for cancer. The dynamic behavior of Lag-3 was analyzed at the immune synapse upon T-cell activation to elucidate the Lag-3 inhibitory mechanism. Lag-3 formed clusters and co-localized with T-cell receptor microcluster (TCR-MC) upon T-cell activation similar to PD-1. Lag-3 blocking antibodies (Abs) inhibited the co-localization between Lag-3 and TCR-MC without inhibiting Lag-3 cluster formation. Lag-3 also inhibited MHC-II-independent stimulation and Lag-3 Ab, which did not block MHC-II binding could still block Lag-3\'s inhibitory function, suggesting that the Lag-3 Ab blocks the Lag-3 inhibitory signal by dissociating the co-assembly of TCR-MC and Lag-3 clusters. Consistent with the combination benefit of PD-1 and Lag-3 Abs to augment T-cell responses, bispecific Lag-3/PD-1 antagonists effectively inhibited both cluster formation and co-localization of PD-1 and Lag-3 with TCR-MC. Therefore, Lag-3 inhibits T-cell activation at TCR-MC, and the target of Lag-3 ICI is to dissociate the co-localization of Lag-3 with TCR-MC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    皮肤癌是全世界广泛关注的健康问题,表现在许多亚型,如鳞状细胞癌,基底细胞癌,还有黑色素瘤.虽然所有这些类型经常发生,它们通常缺乏被治愈的可能性,强调早期发现和治疗的重要性。这项综合研究探讨了程序性细胞死亡蛋白1(PD-1)在皮肤癌中的作用,关注其在免疫调节中的分子机制及其在肿瘤免疫逃避中的关键作用,同时也阐明了癌症发病机制中免疫检查点的复杂性。它严格评估PD-1抑制剂的临床应用,突出他们在治疗皮肤癌方面的治疗潜力,同时也解决了抵抗的重大挑战。这项工作进一步讨论了针对PD-1抑制剂的耐药机制的演变,并提出了缓解这些问题的潜在方法。从而提高这些疗法的有效性。该研究进一步强调了PD-1靶向治疗的现状,并为未来的研究奠定了基础,旨在优化这些治疗方法以获得更好的皮肤癌临床结果。
    Skin cancer is a widespread worldwide health concern, manifesting in many subtypes such as squamous cell carcinoma, basal cell carcinoma, and melanoma. Although all these types occur frequently, they generally lack the possibility of being cured, emphasizing the importance of early discovery and treatment. This comprehensive study explores the role of programmed cell death protein 1 (PD-1) in skin cancer, focusing on its molecular mechanisms in immune regulation and its critical role in tumor immune evasion, while also clarifying the complexities of immune checkpoints in cancer pathogenesis. It critically evaluates the clinical applications of PD-1 inhibitors, spotlighting their therapeutic potential in treating skin cancer, while also addressing the significant challenge of resistance. This work further discusses the evolution of resistance mechanisms against PD-1 inhibitors and suggests potential approaches to mitigate these issues, thereby enhancing the effectiveness of these therapies. The study further highlights the current state of PD-1 targeted therapies and sets the stage for future research aimed at optimizing these treatments for better clinical outcomes in skin cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    血管免疫母细胞性T细胞淋巴瘤(AITL)是成熟外周T细胞淋巴瘤(PTCL)的常见亚型,其发病机制尚未被充分阐明。因其具有较大的异质性导致患者的治疗面临着巨大的挑战,总体预后较差。程序性死亡受体-1(PD-1)是一种重要的免疫抑制分子,是AITL的诊断标志物。越来越多的研究表明PD-1与AITL的预后相关并提出针对PD-1的靶向治疗。该文就PD-1在AITL中的研究进展作一综述。.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    随着免疫检查点抑制剂(ICIs)在非小细胞肺癌中的革命性进展,确定谁将受益于ICI的癌症患者已变得至关重要和紧迫.这里,我们报道了蛋白酪氨酸磷酸酶受体T型(PTPRT)丢失作为一种精确和方便的预测标志物,不依赖于抗PD-1/PD-L1轴治疗的PD-L1表达.抗PD-1/PD-L1轴治疗显著增加PTPRT缺陷型肿瘤患者的无进展生存期。PTPRT缺陷型肿瘤表现出累积的DNA损伤,增加胞质DNA释放,和更高的肿瘤突变负担。此外,STING的酪氨酸残基240被鉴定为PTPRT的直接底物。PTPRT损失在Y240提高了STING的磷酸化,从而抑制了其蛋白酶体介导的降解。PTPRT缺陷型肿瘤释放更多的IFN-β,CCL5和CXCL10通过激活STING通路而增加免疫细胞浸润,特别是CD8T细胞和自然杀伤细胞,最终增强抗PD-1治疗在多个皮下和原位肿瘤小鼠模型中的疗效。PTPRT缺陷型肿瘤对抗PD-1治疗的反应取决于肿瘤固有的STING途径。总之,我们的研究结果揭示了PTPRT缺陷型肿瘤如何对抗PD-1治疗变得敏感的机制,并强调了PTPRT在先天免疫中的生物学功能.考虑到PTPRT突变和阴性表达的患病率,本研究对患者分层和临床决策具有重要价值。
    With the revolutionary progress of immune checkpoint inhibitors (ICIs) in non-small cell lung cancer, identifying patients with cancer who would benefit from ICIs has become critical and urgent. Here, we report protein tyrosine phosphatase receptor type T (PTPRT) loss as a precise and convenient predictive marker independent of PD-L1 expression for anti-PD-1/PD-L1 axis therapy. Anti-PD-1/PD-L1 axis treatment markedly increased progression-free survival in patients with PTPRT-deficient tumors. PTPRT-deficient tumors displayed cumulative DNA damage, increased cytosolic DNA release, and higher tumor mutation burden. Moreover, the tyrosine residue 240 of STING was identified as a direct substrate of PTPRT. PTPRT loss elevated phosphorylation of STING at Y240 and thus inhibited its proteasome-mediated degradation. PTPRT-deficient tumors released more IFN-β, CCL5, and CXCL10 by activation of STING pathway and increased immune cell infiltration, especially of CD8 T cells and natural killer cells, ultimately enhancing the efficacy of anti-PD-1 therapy in multiple subcutaneous and orthotopic tumor mouse models. The response of PTPRT-deficient tumors to anti-PD-1 therapy depends on the tumor-intrinsic STING pathway. In summary, our findings reveal the mechanism of how PTPRT-deficient tumors become sensitive to anti-PD-1 therapy and highlight the biological function of PTPRT in innate immunity. Considering the prevalence of PTPRT mutations and negative expression, this study has great value for patient stratification and clinical decision-making.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    程序性细胞死亡蛋白1(PD-1,CD279)是许多肿瘤疾病的重要治疗靶标。这种检查点蛋白抑制T淋巴细胞攻击体内的其他细胞,从而阻断它提高了免疫系统对肿瘤细胞的清除。虽然已经有多种FDA批准的抗PD-1抗体,包括nivolumab(百时美施贵宝公司的Opdivo®)和pembrolizumab(默克公司的Keytruda®),目前正在努力发现新的和改进的检查点抑制剂疗法.在这项研究中,我们提出了多个抗PD-1抗体片段,这些片段是使用蛋白质扩散计算得出的,并通过我们的可扩展评估,在硅管道。在此,我们提出了9种合成Fv结构,由于期望的预测结合性能,其适合于进一步经验测试其抗PD-1活性。
    The programmed cell death protein 1 (PD-1, CD279) is an important therapeutic target in many oncological diseases. This checkpoint protein inhibits T lymphocytes from attacking other cells in the body and thus blocking it improves the clearance of tumor cells by the immune system. While there are already multiple FDA-approved anti-PD-1 antibodies, including nivolumab (Opdivo® from Bristol-Myers Squibb) and pembrolizumab (Keytruda® from Merck), there are ongoing efforts to discover new and improved checkpoint inhibitor therapeutics. In this study, we present multiple anti-PD-1 antibody fragments that were derived computationally using protein diffusion and evaluated through our scalable, in silico pipeline. Here we present nine synthetic Fv structures that are suitable for further empirical testing of their anti-PD-1 activity due to desirable predicted binding performance.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号