首页 > 最新文献

Critical reviews in oncology/hematology最新文献

英文 中文
Cellular senescence in tumor immune escape: Mechanisms, implications, and therapeutic potential.
Pub Date : 2025-01-24 DOI: 10.1016/j.critrevonc.2025.104628
Li You, Qinghua Wu

Cellular senescence, a hallmark of aging, has emerged as a captivating area of research in tumor immunology with profound implications for cancer prevention and treatment. In the tumor microenvironment, senescent cells exhibit a dual role, simultaneously hindering tumor development through collaboration with immune cells and evading immune cell attacks by upregulating immunoinhibitory proteins. However, the intricate immune escape mechanism of cellular senescence in the tumor microenvironment remains a subject of intense investigation. Chronic inflammation is exacerbated by cellular senescence through the upregulation of pro-inflammatory factors such as interleukin-1β, thereby augmenting the risk of tumorigenesis. Additionally, the interplay between autophagy and cellular senescence adds another layer of complexity. Autophagy, known to slow down the aging process by reducing p53/p21 levels, may be downregulated by cellular senescence. To harness the therapeutic potential of cellular senescence, targeting its immunological aspects has gained significant attention. Strategies such as immune checkpoint inhibitors and T-cell senescence inhibition are being explored in the context of cellular senescence immunotherapy. In this comprehensive review, we provide a compelling overview of the regulation of cellular senescence and delve into the influencing factors, including chronic inflammation, autophagy, and circadian rhythms, associated with senescence in the tumor microenvironment. We specifically focus on unraveling the enigmatic dual role of cellular senescence in tumor immune escape. By deciphering the intricate nature of cellular senescence in the tumor microenvironment, this review aims to advance our understanding and pave the way for leveraging senescence as a promising target for tumor immunotherapy applications.

{"title":"Cellular senescence in tumor immune escape: Mechanisms, implications, and therapeutic potential.","authors":"Li You, Qinghua Wu","doi":"10.1016/j.critrevonc.2025.104628","DOIUrl":"10.1016/j.critrevonc.2025.104628","url":null,"abstract":"<p><p>Cellular senescence, a hallmark of aging, has emerged as a captivating area of research in tumor immunology with profound implications for cancer prevention and treatment. In the tumor microenvironment, senescent cells exhibit a dual role, simultaneously hindering tumor development through collaboration with immune cells and evading immune cell attacks by upregulating immunoinhibitory proteins. However, the intricate immune escape mechanism of cellular senescence in the tumor microenvironment remains a subject of intense investigation. Chronic inflammation is exacerbated by cellular senescence through the upregulation of pro-inflammatory factors such as interleukin-1β, thereby augmenting the risk of tumorigenesis. Additionally, the interplay between autophagy and cellular senescence adds another layer of complexity. Autophagy, known to slow down the aging process by reducing p53/p21 levels, may be downregulated by cellular senescence. To harness the therapeutic potential of cellular senescence, targeting its immunological aspects has gained significant attention. Strategies such as immune checkpoint inhibitors and T-cell senescence inhibition are being explored in the context of cellular senescence immunotherapy. In this comprehensive review, we provide a compelling overview of the regulation of cellular senescence and delve into the influencing factors, including chronic inflammation, autophagy, and circadian rhythms, associated with senescence in the tumor microenvironment. We specifically focus on unraveling the enigmatic dual role of cellular senescence in tumor immune escape. By deciphering the intricate nature of cellular senescence in the tumor microenvironment, this review aims to advance our understanding and pave the way for leveraging senescence as a promising target for tumor immunotherapy applications.</p>","PeriodicalId":93958,"journal":{"name":"Critical reviews in oncology/hematology","volume":" ","pages":"104628"},"PeriodicalIF":0.0,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143048890","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Future directions in the evaluation and management of newly diagnosed metastatic cancer.
Pub Date : 2025-01-24 DOI: 10.1016/j.critrevonc.2025.104631
Eric J Lehrer, Chachrit Khunsriraksakul, Sara Garrett, Daniel M Trifiletti, Jason P Sheehan, Matthias Guckenberger, Alexander V Louie, Shankar Siva, Piet Ost, Karyn A Goodman, Laura A Dawson, Leila T Tchelebi, Jonathan T Yang, Timothy N Showalter, Henry S Park, Daniel E Spratt, Amar U Kishan, Gaorav P Gupta, Chirag Shah, Stefano Fanti, Jeremie Calais, Ming Wang, Kathryn Schmitz, Dajiang Liu, John A Abraham, Robert T Dess, Irène Buvat, Benjamin Solomon, Nicholas G Zaorsky

There is a much debate regarding optimal selection in patients with metastatic cancer who should undergo local treatment (surgery or radiation treatment) to the primary tumor and/or metastases. Additionally, the optimal treatment of newly diagnosed metastatic cancer is largely unclear. Current prognostication systems to best inform these clinical scenarios are limited, as all metastatic patients are grouped together as having Stage IV disease without further incorporation of patient and disease-specific covariates that significantly impact patient outcomes. Therefore, improving current prognostic scoring systems and incorporation of these covariates is essential to best individualize treatment for patients with metastatic cancer. In this narrative review article, we provide a detailed review of prognostication systems that can be used for both the site of metastasis and primary site to best tailor treatment in these patients. Additionally, we discuss the incorporation and ongoing developments in radiographic, genomic, and biostatistical techniques that can be used as prognostication tools.

{"title":"Future directions in the evaluation and management of newly diagnosed metastatic cancer.","authors":"Eric J Lehrer, Chachrit Khunsriraksakul, Sara Garrett, Daniel M Trifiletti, Jason P Sheehan, Matthias Guckenberger, Alexander V Louie, Shankar Siva, Piet Ost, Karyn A Goodman, Laura A Dawson, Leila T Tchelebi, Jonathan T Yang, Timothy N Showalter, Henry S Park, Daniel E Spratt, Amar U Kishan, Gaorav P Gupta, Chirag Shah, Stefano Fanti, Jeremie Calais, Ming Wang, Kathryn Schmitz, Dajiang Liu, John A Abraham, Robert T Dess, Irène Buvat, Benjamin Solomon, Nicholas G Zaorsky","doi":"10.1016/j.critrevonc.2025.104631","DOIUrl":"https://doi.org/10.1016/j.critrevonc.2025.104631","url":null,"abstract":"<p><p>There is a much debate regarding optimal selection in patients with metastatic cancer who should undergo local treatment (surgery or radiation treatment) to the primary tumor and/or metastases. Additionally, the optimal treatment of newly diagnosed metastatic cancer is largely unclear. Current prognostication systems to best inform these clinical scenarios are limited, as all metastatic patients are grouped together as having Stage IV disease without further incorporation of patient and disease-specific covariates that significantly impact patient outcomes. Therefore, improving current prognostic scoring systems and incorporation of these covariates is essential to best individualize treatment for patients with metastatic cancer. In this narrative review article, we provide a detailed review of prognostication systems that can be used for both the site of metastasis and primary site to best tailor treatment in these patients. Additionally, we discuss the incorporation and ongoing developments in radiographic, genomic, and biostatistical techniques that can be used as prognostication tools.</p>","PeriodicalId":93958,"journal":{"name":"Critical reviews in oncology/hematology","volume":" ","pages":"104631"},"PeriodicalIF":0.0,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143048904","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Small cell lung cancer unveiled: Exploring the untapped resource of circulating tumor cells-derived organoids. 揭示小细胞肺癌:探索循环肿瘤细胞衍生类器官的未开发资源。
Pub Date : 2025-01-19 DOI: 10.1016/j.critrevonc.2025.104622
Jesús A Pérez-Cabello, Ana Artero-Castro, Sonia Molina-Pinelo

Small cell lung cancer (SCLC) remains a challenge in oncology due to its aggressive behavior and dismal prognosis. Despite advances in treatments, novel strategies are urgently needed. Enter liquid biopsy-a game-changer in SCLC management. This revolutionary non-invasive approach allows for the analysis of circulating tumor cells (CTCs), offering insights into tumor behavior and treatment responses. Our review focuses on a groundbreaking frontier: harnessing CTCs to create three-dimensional (3D) organoid models. These models, derived from CTCs that break away from the primary tumor or metastatic locations, hold immense potential for revolutionizing cancer research, especially in SCLC. We explore the essential conditions for successfully establishing CTC-derived organoids-a transformative approach with profound implications for personalized medicine. Our evaluation spans diverse isolation techniques, shedding light on their advantages and limitations. Furthermore, we uncover the critical factors governing the cultivation of 3D organoids from CTCs, meticulously mimicking the tumor microenvironment. This review comprehensively elucidates the molecular characterization of these organoids, showcasing their potential in identifying treatment targets and predicting responses. In essence, our review amalgamates cutting-edge methodologies for isolating CTCs, establishing transformative CTC-derived organoids, and characterizing their molecular landscape. This represents a promising frontier for advancing personalized medicine in the complex realm of SCLC management and holds significant implications for translational research.

小细胞肺癌(SCLC)由于其侵袭性行为和预后不佳,在肿瘤学中仍然是一个挑战。尽管在治疗方面取得了进展,但迫切需要新的策略。液体活检的出现改变了SCLC的管理。这种革命性的非侵入性方法允许对循环肿瘤细胞(ctc)进行分析,提供对肿瘤行为和治疗反应的见解。我们的综述集中在一个开创性的前沿:利用ctc来创建三维(3D)类器官模型。这些模型来源于脱离原发肿瘤或转移部位的ctc,具有巨大的癌症研究革命潜力,特别是在SCLC中。我们探索成功建立ctc衍生类器官的基本条件-一种对个性化医疗具有深远影响的变革性方法。我们的评估涵盖了不同的隔离技术,揭示了它们的优点和局限性。此外,我们揭示了控制ctc培养3D类器官的关键因素,精心模拟肿瘤微环境。这篇综述全面阐述了这些类器官的分子特征,展示了它们在识别治疗靶点和预测反应方面的潜力。从本质上讲,我们的综述融合了分离ctc,建立变革性ctc衍生类器官和表征其分子景观的尖端方法。这代表了在复杂的SCLC管理领域推进个性化医疗的一个有希望的前沿,并对转化研究具有重要意义。
{"title":"Small cell lung cancer unveiled: Exploring the untapped resource of circulating tumor cells-derived organoids.","authors":"Jesús A Pérez-Cabello, Ana Artero-Castro, Sonia Molina-Pinelo","doi":"10.1016/j.critrevonc.2025.104622","DOIUrl":"10.1016/j.critrevonc.2025.104622","url":null,"abstract":"<p><p>Small cell lung cancer (SCLC) remains a challenge in oncology due to its aggressive behavior and dismal prognosis. Despite advances in treatments, novel strategies are urgently needed. Enter liquid biopsy-a game-changer in SCLC management. This revolutionary non-invasive approach allows for the analysis of circulating tumor cells (CTCs), offering insights into tumor behavior and treatment responses. Our review focuses on a groundbreaking frontier: harnessing CTCs to create three-dimensional (3D) organoid models. These models, derived from CTCs that break away from the primary tumor or metastatic locations, hold immense potential for revolutionizing cancer research, especially in SCLC. We explore the essential conditions for successfully establishing CTC-derived organoids-a transformative approach with profound implications for personalized medicine. Our evaluation spans diverse isolation techniques, shedding light on their advantages and limitations. Furthermore, we uncover the critical factors governing the cultivation of 3D organoids from CTCs, meticulously mimicking the tumor microenvironment. This review comprehensively elucidates the molecular characterization of these organoids, showcasing their potential in identifying treatment targets and predicting responses. In essence, our review amalgamates cutting-edge methodologies for isolating CTCs, establishing transformative CTC-derived organoids, and characterizing their molecular landscape. This represents a promising frontier for advancing personalized medicine in the complex realm of SCLC management and holds significant implications for translational research.</p>","PeriodicalId":93958,"journal":{"name":"Critical reviews in oncology/hematology","volume":" ","pages":"104622"},"PeriodicalIF":0.0,"publicationDate":"2025-01-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143017903","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
« Augmented radiotherapy » in the management of high-risk prostate cancer (PCa): A systematic review. “增强放疗”在高危前列腺癌(PCa)治疗中的应用:一项系统综述。
Pub Date : 2025-01-18 DOI: 10.1016/j.critrevonc.2025.104623
Jennifer Le Guévelou, Vedang Murthy, Thomas Zilli, Luca Nicosia, Alberto Bossi, Leonard P Bokhorst, Eric Barret, Idir Ouzaid, Paul L Nguyen, Federica Ferrario, Cyrus Chargari, Stefano Arcangeli, Nicolas Magne, Paul Sargos

Background: In patients with high-risk (HR) prostate cancer (PCa) treated with radiotherapy and androgen deprivation therapy (ADT), intensification with androgen receptor pathway inhibitor (ARPI) improves overall survival (OS), at the cost of significant side-effects. We hypothesized that "augmented RT" schedules (defined as either dose-escalation on the prostate gland over 78 Gy and/or addition of whole pelvic radiotherapy (WPRT)), combined with long-term ADT can reach excellent prostate cancer specific survival (PCSS) in this population with little detrimental impact on quality of life.

Methods: We searched Pubmed database until February 8, 2024. Studies reporting both oncological and toxicity outcomes after "augmented RT" were deemed eligible. Studies without ADT or with ARPI intensification were deemed ineligible.

Results: Dose-escalation within the prostate gland at doses over 78 Gy halved the risk of biochemical recurrence at 5 years, with however no impact on PCSS. The addition of WPRT provides a 5-year disease-free survival (DFS) reaching 89.5 % at 5 years, with no significant increase in late grade≥ 2 genito-urinary (GU) or gastrointestinal (GI) toxicity. Combined approaches result in 9-year PCSS ranging between 96.1 % and 100 %. Most approaches demonstrated excellent safety profiles.

Conclusions: "Augmented RT" reached excellent oncological outcomes, with minimal additional toxicity. The development of biomarkers might lead to further treatment personalization, in the rapidly evolving landscape of systemic therapies.

背景:在接受放疗和雄激素剥夺治疗(ADT)的高危(HR)前列腺癌(PCa)患者中,雄激素受体途径抑制剂(ARPI)的强化治疗可提高总生存率(OS),但代价是显著的副作用。我们假设“增强放疗”计划(定义为前列腺剂量增加超过78Gy和/或增加全盆腔放疗(WPRT)),结合长期ADT可以在该人群中达到极好的前列腺癌特异性生存率(PCSS),对生活质量几乎没有不利影响。方法:检索Pubmed数据库至2024年2月8日。报告“增强型放射治疗”后肿瘤和毒性结果的研究被认为是合格的。没有ADT或ARPI增强的研究被认为不合格。结果:前列腺内剂量增加,剂量超过78Gy, 5年生化复发风险减半,但对PCSS没有影响。WPRT的加入使5年无病生存率(DFS)达到89.5%,晚期≥2级泌尿生殖系统(GU)或胃肠道(GI)毒性没有显著增加。综合方法的9年PCSS范围在96.1%至100%之间。大多数方法显示出良好的安全性。结论:“增强型放射治疗”达到了极好的肿瘤学结果,并且具有最小的额外毒性。在快速发展的全身治疗领域,生物标志物的发展可能会导致进一步的治疗个性化。
{"title":"« Augmented radiotherapy » in the management of high-risk prostate cancer (PCa): A systematic review.","authors":"Jennifer Le Guévelou, Vedang Murthy, Thomas Zilli, Luca Nicosia, Alberto Bossi, Leonard P Bokhorst, Eric Barret, Idir Ouzaid, Paul L Nguyen, Federica Ferrario, Cyrus Chargari, Stefano Arcangeli, Nicolas Magne, Paul Sargos","doi":"10.1016/j.critrevonc.2025.104623","DOIUrl":"10.1016/j.critrevonc.2025.104623","url":null,"abstract":"<p><strong>Background: </strong>In patients with high-risk (HR) prostate cancer (PCa) treated with radiotherapy and androgen deprivation therapy (ADT), intensification with androgen receptor pathway inhibitor (ARPI) improves overall survival (OS), at the cost of significant side-effects. We hypothesized that \"augmented RT\" schedules (defined as either dose-escalation on the prostate gland over 78 Gy and/or addition of whole pelvic radiotherapy (WPRT)), combined with long-term ADT can reach excellent prostate cancer specific survival (PCSS) in this population with little detrimental impact on quality of life.</p><p><strong>Methods: </strong>We searched Pubmed database until February 8, 2024. Studies reporting both oncological and toxicity outcomes after \"augmented RT\" were deemed eligible. Studies without ADT or with ARPI intensification were deemed ineligible.</p><p><strong>Results: </strong>Dose-escalation within the prostate gland at doses over 78 Gy halved the risk of biochemical recurrence at 5 years, with however no impact on PCSS. The addition of WPRT provides a 5-year disease-free survival (DFS) reaching 89.5 % at 5 years, with no significant increase in late grade≥ 2 genito-urinary (GU) or gastrointestinal (GI) toxicity. Combined approaches result in 9-year PCSS ranging between 96.1 % and 100 %. Most approaches demonstrated excellent safety profiles.</p><p><strong>Conclusions: </strong>\"Augmented RT\" reached excellent oncological outcomes, with minimal additional toxicity. The development of biomarkers might lead to further treatment personalization, in the rapidly evolving landscape of systemic therapies.</p>","PeriodicalId":93958,"journal":{"name":"Critical reviews in oncology/hematology","volume":" ","pages":"104623"},"PeriodicalIF":0.0,"publicationDate":"2025-01-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143018357","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Real-world evidence in extensive disease small cell lung cancer: The missing piece of the puzzle. 广泛疾病小细胞肺癌的真实证据:拼图缺失的那一块。
Pub Date : 2025-01-17 DOI: 10.1016/j.critrevonc.2025.104618
Paola Damiano, Alessio Stefani, Alice Avancini, Lorenzo Belluomini, Emilio Bria, Sara Pilotto

Small cell lung cancer (SCLC) is a highly aggressive disease, often diagnosed at an advanced stage and with limited treatment options. In recent years, immunotherapy has been approved in combination with chemotherapy in the first line setting of extensive stage disease (ES-SCLC). However, only 10-15 % of patients with ES-SCLC treated with chemoimmunotherapy (CT-IO) experience a long-term benefit. In addition, patients are often clinically frail due to advanced age, comorbidities, and disease-related symptoms, making SCLC a challenging condition. Real-world evidence (RWE) becomes particularly valuable in this scenario, not only to confirm the results of pivotal trials, but also to evaluate the outcomes of CT-IO in populations that are generally excluded from clinical trials. RWE could also define the role of integrative treatments such as thoracic consolidation radiotherapy and prophylactic cranial irradiation, which are used in selected patients in the clinical practice but were scarcely applied in pivotal trials. In this review, we focused on RWE in ES-SCLC, with the aim of improving clinical decision making. Notably, real-world data have largely confirmed the efficacy and safety of CT-IO observed in pivotal clinical trials, with a possible benefit even in more fragile patients. However, these studies also highlight that a significant proportion of the ES-SCLC population remains untreated due to poor clinical conditions.

小细胞肺癌(SCLC)是一种高度侵袭性的疾病,通常在晚期诊断出来,治疗选择有限。近年来,免疫治疗已被批准与化疗联合用于广泛分期疾病(ES-SCLC)的一线治疗。然而,只有10-15%的ES-SCLC患者接受了化学免疫疗法(CT-IO)的长期获益。此外,由于高龄、合并症和疾病相关症状,患者通常在临床上虚弱,使SCLC成为一种具有挑战性的疾病。在这种情况下,真实世界证据(RWE)变得特别有价值,不仅可以证实关键试验的结果,还可以评估通常被排除在临床试验之外的人群的CT-IO结果。RWE还可以确定综合治疗的作用,如胸部巩固放疗和预防性颅脑照射,这些治疗在临床实践中用于选定的患者,但很少用于关键试验。在这篇综述中,我们关注ES-SCLC的RWE,目的是改善临床决策。值得注意的是,现实世界的数据在很大程度上证实了关键临床试验中观察到的CT-IO的有效性和安全性,甚至在更脆弱的患者中也可能受益。然而,这些研究也强调,由于临床条件差,很大一部分ES-SCLC患者仍未得到治疗。
{"title":"Real-world evidence in extensive disease small cell lung cancer: The missing piece of the puzzle.","authors":"Paola Damiano, Alessio Stefani, Alice Avancini, Lorenzo Belluomini, Emilio Bria, Sara Pilotto","doi":"10.1016/j.critrevonc.2025.104618","DOIUrl":"10.1016/j.critrevonc.2025.104618","url":null,"abstract":"<p><p>Small cell lung cancer (SCLC) is a highly aggressive disease, often diagnosed at an advanced stage and with limited treatment options. In recent years, immunotherapy has been approved in combination with chemotherapy in the first line setting of extensive stage disease (ES-SCLC). However, only 10-15 % of patients with ES-SCLC treated with chemoimmunotherapy (CT-IO) experience a long-term benefit. In addition, patients are often clinically frail due to advanced age, comorbidities, and disease-related symptoms, making SCLC a challenging condition. Real-world evidence (RWE) becomes particularly valuable in this scenario, not only to confirm the results of pivotal trials, but also to evaluate the outcomes of CT-IO in populations that are generally excluded from clinical trials. RWE could also define the role of integrative treatments such as thoracic consolidation radiotherapy and prophylactic cranial irradiation, which are used in selected patients in the clinical practice but were scarcely applied in pivotal trials. In this review, we focused on RWE in ES-SCLC, with the aim of improving clinical decision making. Notably, real-world data have largely confirmed the efficacy and safety of CT-IO observed in pivotal clinical trials, with a possible benefit even in more fragile patients. However, these studies also highlight that a significant proportion of the ES-SCLC population remains untreated due to poor clinical conditions.</p>","PeriodicalId":93958,"journal":{"name":"Critical reviews in oncology/hematology","volume":" ","pages":"104618"},"PeriodicalIF":0.0,"publicationDate":"2025-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143017730","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Fibrinogen: a new player and target on the formation of pre-metastatic niche in tumor metastasis. 纤维蛋白原:肿瘤转移前生态位形成的新参与者和靶点。
Pub Date : 2025-01-16 DOI: 10.1016/j.critrevonc.2025.104625
Yuxin Zhang, Zelin Li, Jiamao Zhang, Tatenda Mafa, Jingyu Zhang, Hui Zhu, Lifang Chen, Zhen Zong, Lingling Yang

Tumor metastasis involves a series of complex and coordinated processes, which is the main cause of patient death and still a significant challenge in cancer treatment. Pre-metastatic niches (PMN), a specialized microenvironment that develops in distant organs prior to the arrival of metastatic cancer cells, plays a crucial role in driving tumor metastasis. The development of PMN depends on a complex series of cellular and molecular components including tumor-derived factors, bone marrow-derived cells, resident immune cells, and extracellular matrix. Fibrinogen, a key factor in the typical blood clotting process, is related to tumor metastasis and prognosis, according to a growing body of evidence in recent years. Fibrinogen has emerged as an important factor in mediating the formation of tumor microenvironment. Nevertheless, a clear and detailed mechanism by which fibrinogen promotes tumor metastasis remains unknown. In this review, we first explore the roles of fibrinogen in the development of PMN from four perspectives: immunosuppression, inflammation, angiogenesis, and extracellular matrix remodeling. We highlight the significance of fibrinogen in shaping PMN and discuss its potential therapeutic values, opening new avenues for targeting fibrinogen to prevent or treat metastasis.

肿瘤转移涉及一系列复杂而协调的过程,是导致患者死亡的主要原因,也是癌症治疗的一个重大挑战。转移前生态位(Pre-metastatic niche, PMN)是一种特异性的微环境,在肿瘤转移过程中起着至关重要的作用。PMN的发展取决于一系列复杂的细胞和分子成分,包括肿瘤源性因子、骨髓源性细胞、常驻免疫细胞和细胞外基质。近年来越来越多的证据表明,纤维蛋白原是典型凝血过程中的关键因子,与肿瘤转移和预后有关。纤维蛋白原已成为介导肿瘤微环境形成的重要因素。然而,纤维蛋白原促进肿瘤转移的具体机制尚不清楚。在本文中,我们首先从免疫抑制、炎症、血管生成和细胞外基质重塑四个方面探讨纤维蛋白原在PMN发生中的作用。我们强调纤维蛋白原在PMN形成中的重要性,并讨论其潜在的治疗价值,为靶向纤维蛋白原预防或治疗转移开辟了新的途径。
{"title":"Fibrinogen: a new player and target on the formation of pre-metastatic niche in tumor metastasis.","authors":"Yuxin Zhang, Zelin Li, Jiamao Zhang, Tatenda Mafa, Jingyu Zhang, Hui Zhu, Lifang Chen, Zhen Zong, Lingling Yang","doi":"10.1016/j.critrevonc.2025.104625","DOIUrl":"https://doi.org/10.1016/j.critrevonc.2025.104625","url":null,"abstract":"<p><p>Tumor metastasis involves a series of complex and coordinated processes, which is the main cause of patient death and still a significant challenge in cancer treatment. Pre-metastatic niches (PMN), a specialized microenvironment that develops in distant organs prior to the arrival of metastatic cancer cells, plays a crucial role in driving tumor metastasis. The development of PMN depends on a complex series of cellular and molecular components including tumor-derived factors, bone marrow-derived cells, resident immune cells, and extracellular matrix. Fibrinogen, a key factor in the typical blood clotting process, is related to tumor metastasis and prognosis, according to a growing body of evidence in recent years. Fibrinogen has emerged as an important factor in mediating the formation of tumor microenvironment. Nevertheless, a clear and detailed mechanism by which fibrinogen promotes tumor metastasis remains unknown. In this review, we first explore the roles of fibrinogen in the development of PMN from four perspectives: immunosuppression, inflammation, angiogenesis, and extracellular matrix remodeling. We highlight the significance of fibrinogen in shaping PMN and discuss its potential therapeutic values, opening new avenues for targeting fibrinogen to prevent or treat metastasis.</p>","PeriodicalId":93958,"journal":{"name":"Critical reviews in oncology/hematology","volume":" ","pages":"104625"},"PeriodicalIF":0.0,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143017628","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Challenges and prospects in HER2-positive breast cancer-targeted therapy. her2阳性乳腺癌靶向治疗的挑战和前景。
Pub Date : 2025-01-16 DOI: 10.1016/j.critrevonc.2025.104624
Xiyin Li, Xueying Zhang, Saige Yin, Jianyun Nie

Breast cancer remains the most prevalent malignancy among women globally and ranks as the leading cause of cancer-related mortality in this demographic. Approximately 13 %-15 % of all breast cancer cases are classified as HER2-positive, a subtype associated with a particularly unfavorable prognosis. A large number of patients with HER2-positive breast cancer continue to face disease progression after receiving standardized treatment. Given these challenges, a thorough exploration into the mechanisms underlying drug resistance in HER2-targeted therapy is imperative. This review focuses on the factors related to drug resistance in HER2-targeted therapy, including tumor heterogeneity, antibody-binding efficacy, variations in the tumor microenvironment, and abnormalities in signal activation and transmission. Additionally, corresponding strategies to counteract these resistance mechanisms are discussed, to advance therapeutic efficacy and clinical benefits in the management of HER2-positive breast cancer.

乳腺癌仍然是全球妇女中最普遍的恶性肿瘤,也是这一人口统计学中癌症相关死亡的主要原因。大约13%-15%的乳腺癌病例被归类为her2阳性,这是一种与特别不利的预后相关的亚型。大量her2阳性乳腺癌患者在接受标准化治疗后仍面临疾病进展。鉴于这些挑战,深入探索her2靶向治疗的耐药机制势在必行。本文综述了与her2靶向治疗耐药相关的因素,包括肿瘤异质性、抗体结合效果、肿瘤微环境变化、信号激活和传递异常等。此外,本文还讨论了对抗这些耐药机制的相应策略,以提高her2阳性乳腺癌的治疗效果和临床效益。
{"title":"Challenges and prospects in HER2-positive breast cancer-targeted therapy.","authors":"Xiyin Li, Xueying Zhang, Saige Yin, Jianyun Nie","doi":"10.1016/j.critrevonc.2025.104624","DOIUrl":"10.1016/j.critrevonc.2025.104624","url":null,"abstract":"<p><p>Breast cancer remains the most prevalent malignancy among women globally and ranks as the leading cause of cancer-related mortality in this demographic. Approximately 13 %-15 % of all breast cancer cases are classified as HER2-positive, a subtype associated with a particularly unfavorable prognosis. A large number of patients with HER2-positive breast cancer continue to face disease progression after receiving standardized treatment. Given these challenges, a thorough exploration into the mechanisms underlying drug resistance in HER2-targeted therapy is imperative. This review focuses on the factors related to drug resistance in HER2-targeted therapy, including tumor heterogeneity, antibody-binding efficacy, variations in the tumor microenvironment, and abnormalities in signal activation and transmission. Additionally, corresponding strategies to counteract these resistance mechanisms are discussed, to advance therapeutic efficacy and clinical benefits in the management of HER2-positive breast cancer.</p>","PeriodicalId":93958,"journal":{"name":"Critical reviews in oncology/hematology","volume":" ","pages":"104624"},"PeriodicalIF":0.0,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143017345","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Preclinical models of soft tissue sarcomas - generation and applications to enhance translational research. 软组织肉瘤发生的临床前模型及其应用,以加强转化研究。
Pub Date : 2025-01-15 DOI: 10.1016/j.critrevonc.2025.104621
Sandro Pasquali, David S Moura, Molly R Danks, Piotr J Manasterski, Nadia Zaffaroni, Silvia Stacchiotti, Jose L Mondaza-Hernandez, William G J Kerrison, Javier Martin-Broto, Paul H Huang, Valerie G Brunton

Soft tissue sarcomas (STS) represent a large group of rare and ultra-rare tumors distinguished by unique morphological, molecular and clinical features. Patients with such rare cancers are generally underrepresented in clinical trials which has limited the introduction of new treatment options and subsequent improvement of patient outcomes. Preclinical models of STS that recapitulate the human disease can aid progress in identifying new effective treatments. However, due to the rarity of these tumors there are limited STS models available. Here we review the existing preclinical models of STS, including patient-derived cell lines and organoids, patient-derived xenografts and genetically engineered mouse models. We discuss the advantages and disadvantages of the different models and describe to what extent they have aided clinical translation. Finally, we consider what can be done in the future to enhance their predictivity in the preclinical setting.

软组织肉瘤(STS)是一大类罕见和超罕见的肿瘤,具有独特的形态、分子和临床特征。患有这种罕见癌症的患者通常在临床试验中代表性不足,这限制了新治疗方案的引入和随后患者预后的改善。概括人类疾病的STS临床前模型可以帮助确定新的有效治疗方法。然而,由于这些肿瘤的罕见性,可用的STS模型有限。在这里,我们回顾了现有的STS临床前模型,包括患者来源的细胞系和类器官,患者来源的异种移植和基因工程小鼠模型。我们讨论了不同模型的优点和缺点,并描述了他们在多大程度上辅助临床翻译。最后,我们考虑了未来可以做些什么来提高他们在临床前的预测能力。
{"title":"Preclinical models of soft tissue sarcomas - generation and applications to enhance translational research.","authors":"Sandro Pasquali, David S Moura, Molly R Danks, Piotr J Manasterski, Nadia Zaffaroni, Silvia Stacchiotti, Jose L Mondaza-Hernandez, William G J Kerrison, Javier Martin-Broto, Paul H Huang, Valerie G Brunton","doi":"10.1016/j.critrevonc.2025.104621","DOIUrl":"https://doi.org/10.1016/j.critrevonc.2025.104621","url":null,"abstract":"<p><p>Soft tissue sarcomas (STS) represent a large group of rare and ultra-rare tumors distinguished by unique morphological, molecular and clinical features. Patients with such rare cancers are generally underrepresented in clinical trials which has limited the introduction of new treatment options and subsequent improvement of patient outcomes. Preclinical models of STS that recapitulate the human disease can aid progress in identifying new effective treatments. However, due to the rarity of these tumors there are limited STS models available. Here we review the existing preclinical models of STS, including patient-derived cell lines and organoids, patient-derived xenografts and genetically engineered mouse models. We discuss the advantages and disadvantages of the different models and describe to what extent they have aided clinical translation. Finally, we consider what can be done in the future to enhance their predictivity in the preclinical setting.</p>","PeriodicalId":93958,"journal":{"name":"Critical reviews in oncology/hematology","volume":"207 ","pages":"104621"},"PeriodicalIF":0.0,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143018157","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Role of the International Society of Liquid Biopsy (ISLB) in establishing quality control frameworks for clinical integration. 国际液体活检学会(ISLB)在建立临床整合的质量控制框架中的作用。
Pub Date : 2025-01-15 DOI: 10.1016/j.critrevonc.2025.104619
Nicola Fusco, Umberto Malapelle, Christian Rolfo

Liquid biopsy (LB) has revolutionized molecular pathology, offering non-invasive insights into tumor biology. However, widespread adoption is hindered by a lack of standardized protocols, requiring robust quality control and harmonized workflows. Large-scale studies are needed to establish effective standard operating procedures (SOPs), particularly for circulating tumor DNA (ctDNA) assays tailored to different disease stages. The International Society of Liquid Biopsy (ISLB) has addressed these challenges by forming a Quality Control and Accreditation Committee, focused on developing frameworks for pre-analytical, analytical, and interpretive processes. Key priorities include mitigating pre-analytical variability with stringent guidelines for blood handling and ensuring adherence to international standards like ISO 15189 and CLIA/CAP. ISLB emphasizes harmonized methodologies, with advanced techniques like droplet digital PCR and next-generation sequencing requiring unified workflows. Collaboration with global initiatives, including the European Society of Medical Oncology (ESMO), American Society of Clinical Oncology (ASCO), and International Liquid Biopsy Standardization Alliance (ILSA), supports the validation of ctDNA testing. These efforts are vital for integrating LB into clinical care, advancing precision oncology, and improving patient outcomes through reliable and standardized applications.

液体活检(LB)彻底改变了分子病理学,为肿瘤生物学提供了非侵入性的见解。然而,由于缺乏标准化协议,需要强有力的质量控制和协调的工作流程,因此阻碍了广泛采用。需要大规模的研究来建立有效的标准操作程序(sop),特别是针对不同疾病阶段的循环肿瘤DNA (ctDNA)测定。国际液体活检学会(ISLB)通过成立质量控制和认证委员会来应对这些挑战,重点是制定分析前、分析和解释过程的框架。关键优先事项包括通过严格的血液处理指南减轻分析前的可变性,并确保遵守ISO 15189和CLIA/CAP等国际标准。ISLB强调统一的方法,如液滴数字PCR和下一代测序等先进技术需要统一的工作流程。与全球倡议的合作,包括欧洲肿瘤医学学会(ESMO),美国临床肿瘤学会(ASCO)和国际液体活检标准化联盟(ILSA),支持ctDNA检测的验证。这些努力对于将LB整合到临床护理、推进精准肿瘤学以及通过可靠和标准化的应用程序改善患者预后至关重要。
{"title":"Role of the International Society of Liquid Biopsy (ISLB) in establishing quality control frameworks for clinical integration.","authors":"Nicola Fusco, Umberto Malapelle, Christian Rolfo","doi":"10.1016/j.critrevonc.2025.104619","DOIUrl":"https://doi.org/10.1016/j.critrevonc.2025.104619","url":null,"abstract":"<p><p>Liquid biopsy (LB) has revolutionized molecular pathology, offering non-invasive insights into tumor biology. However, widespread adoption is hindered by a lack of standardized protocols, requiring robust quality control and harmonized workflows. Large-scale studies are needed to establish effective standard operating procedures (SOPs), particularly for circulating tumor DNA (ctDNA) assays tailored to different disease stages. The International Society of Liquid Biopsy (ISLB) has addressed these challenges by forming a Quality Control and Accreditation Committee, focused on developing frameworks for pre-analytical, analytical, and interpretive processes. Key priorities include mitigating pre-analytical variability with stringent guidelines for blood handling and ensuring adherence to international standards like ISO 15189 and CLIA/CAP. ISLB emphasizes harmonized methodologies, with advanced techniques like droplet digital PCR and next-generation sequencing requiring unified workflows. Collaboration with global initiatives, including the European Society of Medical Oncology (ESMO), American Society of Clinical Oncology (ASCO), and International Liquid Biopsy Standardization Alliance (ILSA), supports the validation of ctDNA testing. These efforts are vital for integrating LB into clinical care, advancing precision oncology, and improving patient outcomes through reliable and standardized applications.</p>","PeriodicalId":93958,"journal":{"name":"Critical reviews in oncology/hematology","volume":" ","pages":"104619"},"PeriodicalIF":0.0,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143017743","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
STAT3 orchestrates immune dynamics in hepatocellular carcinoma: A pivotal nexus in tumor progression. STAT3在肝细胞癌中协调免疫动力学:肿瘤进展的关键联系
Pub Date : 2025-01-14 DOI: 10.1016/j.critrevonc.2025.104620
Chen Zhang, Songbai Hu, Chuanzheng Yin, Guoliang Wang, Pian Liu

Hepatocellular carcinoma (HCC) presents a formidable challenge in oncology, attributed to its association with chronic liver diseases and global prevalence. The immune microenvironment profoundly influences HCC progression, balancing immune suppression and antitumor responses. The Signal Transducer and Activator of Transcription 3 (STAT3) is central to this equilibrium, orchestrating immune dynamics and intertwining tumor progression with immune evasion mechanisms. Dysregulated STAT3 signaling, activated by various stimuli, including cytokines and growth factors, promotes an immunosuppressive milieu within HCC tumors, fostering tumor survival and proliferation while hindering immune surveillance. Non-coding RNAs and other molecules regulate this process, modulating STAT3 activity and influencing immune cell function. Moreover, therapeutic interventions targeting the STAT3 pathway, alongside advancements in radiotherapy, cancer vaccines, and diabetes-related drugs, offer promising strategies in HCC management. Integrating natural compounds with immunotherapy emerges as a novel approach, leveraging their ability to enhance antitumor immunity and counter immune evasion strategies. Understanding the multifaceted role of STAT3 and its interactions within the immune landscape of HCC is paramount for devising effective therapeutic interventions and improving patient outcomes.

肝细胞癌(HCC)由于其与慢性肝脏疾病和全球患病率的相关性,在肿瘤学领域提出了一个巨大的挑战。免疫微环境深刻影响HCC的进展,平衡免疫抑制和抗肿瘤反应。信号换能器和转录激活因子3 (STAT3)是这种平衡的核心,它协调免疫动力学并将肿瘤进展与免疫逃避机制交织在一起。失调的STAT3信号被各种刺激激活,包括细胞因子和生长因子,促进HCC肿瘤内的免疫抑制环境,促进肿瘤存活和增殖,同时阻碍免疫监视。非编码rna和其他分子调节这一过程,调节STAT3活性并影响免疫细胞功能。此外,针对STAT3通路的治疗干预,以及放疗、癌症疫苗和糖尿病相关药物的进展,为HCC治疗提供了有希望的策略。将天然化合物与免疫疗法结合起来是一种新的方法,利用它们增强抗肿瘤免疫和对抗免疫逃避策略的能力。了解STAT3的多方面作用及其在HCC免疫环境中的相互作用对于设计有效的治疗干预措施和改善患者预后至关重要。
{"title":"STAT3 orchestrates immune dynamics in hepatocellular carcinoma: A pivotal nexus in tumor progression.","authors":"Chen Zhang, Songbai Hu, Chuanzheng Yin, Guoliang Wang, Pian Liu","doi":"10.1016/j.critrevonc.2025.104620","DOIUrl":"10.1016/j.critrevonc.2025.104620","url":null,"abstract":"<p><p>Hepatocellular carcinoma (HCC) presents a formidable challenge in oncology, attributed to its association with chronic liver diseases and global prevalence. The immune microenvironment profoundly influences HCC progression, balancing immune suppression and antitumor responses. The Signal Transducer and Activator of Transcription 3 (STAT3) is central to this equilibrium, orchestrating immune dynamics and intertwining tumor progression with immune evasion mechanisms. Dysregulated STAT3 signaling, activated by various stimuli, including cytokines and growth factors, promotes an immunosuppressive milieu within HCC tumors, fostering tumor survival and proliferation while hindering immune surveillance. Non-coding RNAs and other molecules regulate this process, modulating STAT3 activity and influencing immune cell function. Moreover, therapeutic interventions targeting the STAT3 pathway, alongside advancements in radiotherapy, cancer vaccines, and diabetes-related drugs, offer promising strategies in HCC management. Integrating natural compounds with immunotherapy emerges as a novel approach, leveraging their ability to enhance antitumor immunity and counter immune evasion strategies. Understanding the multifaceted role of STAT3 and its interactions within the immune landscape of HCC is paramount for devising effective therapeutic interventions and improving patient outcomes.</p>","PeriodicalId":93958,"journal":{"name":"Critical reviews in oncology/hematology","volume":" ","pages":"104620"},"PeriodicalIF":0.0,"publicationDate":"2025-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143018056","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Critical reviews in oncology/hematology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
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
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1