首页 > 最新文献

Current Opinion in Oncology最新文献

英文 中文
Recent advances in immunotherapy for small cell lung cancer. 小细胞肺癌免疫疗法的最新进展。
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2025-01-01 Epub Date: 2024-11-02 DOI: 10.1097/CCO.0000000000001105
Ziyuan Ren, Shijie Shang, Dawei Chen

Purpose of review: This review aims to provide an overview of recent advances in immunotherapy for small cell lung cancer (SCLC), with a focus on the current status of immune checkpoint inhibitors (ICIs), novel combination strategies, and key biomarkers.

Recent findings: The integration of ICIs into standard chemotherapy has established them as the first-line treatment for extensive-stage SCLC (ES-SCLC). The ADRIATIC trial further demonstrated the efficacy of ICI maintenance therapy in limited-stage SCLC. Additionally, combining radiotherapy with ICIs has shown promising synergistic effects, including the abscopal and radscopal effects. Ongoing investigations into the combination of ICIs with targeted therapies, such as antiangiogenic agents and DNA damage response inhibitors, have yielded encouraging preliminary results. Notably, the novel therapeutic agent tarlatamab, the first bispecific DLL3-directed CD3 T-cell engager, has recently received FDA approval for second-line treatment of ES-SCLC. Advances in omics technologies have shed light on the intra-tumor and inter-tumor heterogeneity of SCLC, leading to the identification of new molecular subtypes and biomarkers, thereby paving the way for precision medicine.

Summary: Despite the improved outcomes associated with immunotherapy in SCLC, the overall clinical benefit remains modest. Further preclinical and clinical studies are essential to identify optimal treatment regimens and enhance therapeutic efficacy.

综述目的:本综述旨在概述小细胞肺癌(SCLC)免疫疗法的最新进展,重点关注免疫检查点抑制剂(ICIs)的现状、新型联合策略和关键生物标志物:将 ICIs 纳入标准化疗已使 ICIs 成为广泛期 SCLC(ES-SCLC)的一线治疗药物。ADRIATIC试验进一步证明了ICI维持疗法对局限期SCLC的疗效。此外,放疗与 ICIs 的联合应用也显示出良好的协同效应,包括缺灶效应和射灶效应。ICIs 与抗血管生成药物和 DNA 损伤反应抑制剂等靶向疗法的结合研究正在进行中,已取得了令人鼓舞的初步结果。值得注意的是,新型治疗药物塔拉他单抗(tarlatamab)是第一种双特异性 DLL3 定向 CD3 T 细胞吸引剂,最近已获得 FDA 批准用于 ES-SCLC 的二线治疗。Omics技术的进步揭示了SCLC的瘤内和瘤间异质性,从而确定了新的分子亚型和生物标志物,为精准医疗铺平了道路。进一步的临床前和临床研究对于确定最佳治疗方案和提高疗效至关重要。
{"title":"Recent advances in immunotherapy for small cell lung cancer.","authors":"Ziyuan Ren, Shijie Shang, Dawei Chen","doi":"10.1097/CCO.0000000000001105","DOIUrl":"10.1097/CCO.0000000000001105","url":null,"abstract":"<p><strong>Purpose of review: </strong>This review aims to provide an overview of recent advances in immunotherapy for small cell lung cancer (SCLC), with a focus on the current status of immune checkpoint inhibitors (ICIs), novel combination strategies, and key biomarkers.</p><p><strong>Recent findings: </strong>The integration of ICIs into standard chemotherapy has established them as the first-line treatment for extensive-stage SCLC (ES-SCLC). The ADRIATIC trial further demonstrated the efficacy of ICI maintenance therapy in limited-stage SCLC. Additionally, combining radiotherapy with ICIs has shown promising synergistic effects, including the abscopal and radscopal effects. Ongoing investigations into the combination of ICIs with targeted therapies, such as antiangiogenic agents and DNA damage response inhibitors, have yielded encouraging preliminary results. Notably, the novel therapeutic agent tarlatamab, the first bispecific DLL3-directed CD3 T-cell engager, has recently received FDA approval for second-line treatment of ES-SCLC. Advances in omics technologies have shed light on the intra-tumor and inter-tumor heterogeneity of SCLC, leading to the identification of new molecular subtypes and biomarkers, thereby paving the way for precision medicine.</p><p><strong>Summary: </strong>Despite the improved outcomes associated with immunotherapy in SCLC, the overall clinical benefit remains modest. Further preclinical and clinical studies are essential to identify optimal treatment regimens and enhance therapeutic efficacy.</p>","PeriodicalId":10893,"journal":{"name":"Current Opinion in Oncology","volume":" ","pages":"17-26"},"PeriodicalIF":2.8,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142616411","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Neoadjuvant immunotherapy in melanoma: pathological response as a surrogate endpoint?
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-12-30 DOI: 10.1097/CCO.0000000000001119
Michel Meyers, Oumnia Mouna, Mireille Langouo

Purpose of review: This review evaluates by analyzing recent studies whether pathological complete response (pCR) can be used as a reliable surrogate marker for overall survival (OS) in melanoma treated with neoadjuvant immunotherapy.

Recent findings: Trials like Neo-Combi, Neo-Trio and COMBI-Neo show that pCR is crucial for long-term success in targeted therapy for melanoma, while studies like OpACIN-neo and SWOG S1801 demonstrate that immunotherapy can provide durable benefits even with partial responses. Findings from NADINA and the INMC analysis highlight that immunotherapy achieves higher pathologic response rates and improved survival outcomes, offering broader benefits compared to the pCR-dependent outcomes of targeted therapy.

Summary: pCR serves as a critical prognostic biomarker across cancer types, with strong validation in breast cancer where it predicts long-term survival, particularly in aggressive subtypes like triple-negative and HER2-positive breast cancer. In melanoma, pCR is gaining prominence as a surrogate marker in neoadjuvant therapies, with its predictive value varying between targeted treatments - where achieving pCR is essential - and immunotherapies, which provide durable survival benefits even with partial responses. Despite its potential, the role of pCR as a universally reliable surrogate endpoint in melanoma requires further validation through cancer-specific studies to ensure accurate treatment guidance and personalized care.

{"title":"Neoadjuvant immunotherapy in melanoma: pathological response as a surrogate endpoint?","authors":"Michel Meyers, Oumnia Mouna, Mireille Langouo","doi":"10.1097/CCO.0000000000001119","DOIUrl":"https://doi.org/10.1097/CCO.0000000000001119","url":null,"abstract":"<p><strong>Purpose of review: </strong>This review evaluates by analyzing recent studies whether pathological complete response (pCR) can be used as a reliable surrogate marker for overall survival (OS) in melanoma treated with neoadjuvant immunotherapy.</p><p><strong>Recent findings: </strong>Trials like Neo-Combi, Neo-Trio and COMBI-Neo show that pCR is crucial for long-term success in targeted therapy for melanoma, while studies like OpACIN-neo and SWOG S1801 demonstrate that immunotherapy can provide durable benefits even with partial responses. Findings from NADINA and the INMC analysis highlight that immunotherapy achieves higher pathologic response rates and improved survival outcomes, offering broader benefits compared to the pCR-dependent outcomes of targeted therapy.</p><p><strong>Summary: </strong>pCR serves as a critical prognostic biomarker across cancer types, with strong validation in breast cancer where it predicts long-term survival, particularly in aggressive subtypes like triple-negative and HER2-positive breast cancer. In melanoma, pCR is gaining prominence as a surrogate marker in neoadjuvant therapies, with its predictive value varying between targeted treatments - where achieving pCR is essential - and immunotherapies, which provide durable survival benefits even with partial responses. Despite its potential, the role of pCR as a universally reliable surrogate endpoint in melanoma requires further validation through cancer-specific studies to ensure accurate treatment guidance and personalized care.</p>","PeriodicalId":10893,"journal":{"name":"Current Opinion in Oncology","volume":" ","pages":""},"PeriodicalIF":2.8,"publicationDate":"2024-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143045828","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
From basic to clinical and therapeutic insights: news on actinic keratosis and skin squamous cell carcinoma.
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-12-24 DOI: 10.1097/CCO.0000000000001115
Carmen Orte Cano, Mariano Suppa, Ievgenia Pastushenko, Véronique Del Marmol

Purpose of review: Squamous cell carcinoma (SCC) is the second most common skin cancer, with an increasing incidence. This review highlights this past year's advances regarding the understanding of its pathogenesis, newly introduced diagnostic methods and updates in prevention and treatment.

Recent findings: While the pathogenesis of SCC progression remains unclear, new sequencing techniques are helping to better characterize these tumours at the molecular level. Recently introduced noninvasive imaging techniques are rapidly transforming SCC diagnosis and follow-up. Although nicotinamide has not demonstrated significant benefit in reducing SCC incidence among transplant recipients, larger studies are needed to achieve statistical power. Tirbanibulin, a new field treatment for actinic keratosis is now available and well tolerated for use in areas up to 100 cm2. Surgery remains the cornerstone of SCC treatment and can now be complemented with cemiplimab, when advanced.

Summary: Recent years have seen a diagnostic revolution in dermatology, driven by noninvasive imaging and artificial intelligence; however, the physiopathogenesis of SCC progression remains poorly understood. In treatment, immune checkpoint inhibitors have shown good survival outcomes for advanced SCC. Research continues in the neoadjuvant setting and among transplanted patients, with encouraging preliminary results.

{"title":"From basic to clinical and therapeutic insights: news on actinic keratosis and skin squamous cell carcinoma.","authors":"Carmen Orte Cano, Mariano Suppa, Ievgenia Pastushenko, Véronique Del Marmol","doi":"10.1097/CCO.0000000000001115","DOIUrl":"https://doi.org/10.1097/CCO.0000000000001115","url":null,"abstract":"<p><strong>Purpose of review: </strong>Squamous cell carcinoma (SCC) is the second most common skin cancer, with an increasing incidence. This review highlights this past year's advances regarding the understanding of its pathogenesis, newly introduced diagnostic methods and updates in prevention and treatment.</p><p><strong>Recent findings: </strong>While the pathogenesis of SCC progression remains unclear, new sequencing techniques are helping to better characterize these tumours at the molecular level. Recently introduced noninvasive imaging techniques are rapidly transforming SCC diagnosis and follow-up. Although nicotinamide has not demonstrated significant benefit in reducing SCC incidence among transplant recipients, larger studies are needed to achieve statistical power. Tirbanibulin, a new field treatment for actinic keratosis is now available and well tolerated for use in areas up to 100 cm2. Surgery remains the cornerstone of SCC treatment and can now be complemented with cemiplimab, when advanced.</p><p><strong>Summary: </strong>Recent years have seen a diagnostic revolution in dermatology, driven by noninvasive imaging and artificial intelligence; however, the physiopathogenesis of SCC progression remains poorly understood. In treatment, immune checkpoint inhibitors have shown good survival outcomes for advanced SCC. Research continues in the neoadjuvant setting and among transplanted patients, with encouraging preliminary results.</p>","PeriodicalId":10893,"journal":{"name":"Current Opinion in Oncology","volume":" ","pages":""},"PeriodicalIF":2.8,"publicationDate":"2024-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143045809","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Long-term immune related adverse events.
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-12-17 DOI: 10.1097/CCO.0000000000001113
Leo Plaçais, Olivier Lambotte

Purpose of review: Long-term immune related adverse events (irAEs) are an increasing reality in cancer patients treated with immune checkpoint inhibitors (ICIs) but remain under-reported. With the number of ICI treated patients increasing, we here aimed to expose the current evidence on their clinical presentations and diagnosis criteria.

Recent findings: First described in ICI validation trials, long term irAEs were further characterized through retrospective studies, providing clues on their frequency, clinical spectrum, risk factors and impact on patient's quality of life. Recent recommendations proposed consensual definitions to delineate their different subtypes, from delayed/late-onset irAE to chronic active and inactive irAE, paving the way to future prospective studies.

Summary: Knowledge of the different long-term irAE subtypes risk and clinical presentation is required to increase their detection and diagnosis, to better weight therapeutic decisions and to improve cancer patient's care.

{"title":"Long-term immune related adverse events.","authors":"Leo Plaçais, Olivier Lambotte","doi":"10.1097/CCO.0000000000001113","DOIUrl":"https://doi.org/10.1097/CCO.0000000000001113","url":null,"abstract":"<p><strong>Purpose of review: </strong>Long-term immune related adverse events (irAEs) are an increasing reality in cancer patients treated with immune checkpoint inhibitors (ICIs) but remain under-reported. With the number of ICI treated patients increasing, we here aimed to expose the current evidence on their clinical presentations and diagnosis criteria.</p><p><strong>Recent findings: </strong>First described in ICI validation trials, long term irAEs were further characterized through retrospective studies, providing clues on their frequency, clinical spectrum, risk factors and impact on patient's quality of life. Recent recommendations proposed consensual definitions to delineate their different subtypes, from delayed/late-onset irAE to chronic active and inactive irAE, paving the way to future prospective studies.</p><p><strong>Summary: </strong>Knowledge of the different long-term irAE subtypes risk and clinical presentation is required to increase their detection and diagnosis, to better weight therapeutic decisions and to improve cancer patient's care.</p>","PeriodicalId":10893,"journal":{"name":"Current Opinion in Oncology","volume":" ","pages":""},"PeriodicalIF":2.8,"publicationDate":"2024-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143045824","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Prophylactic cranial irradiation for small cell lung cancer in the era of immunotherapy and molecular subtypes. 免疫治疗和分子亚型时代小细胞肺癌的预防性颅脑照射。
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-12-03 DOI: 10.1097/CCO.0000000000001111
Veronika Pozonec, Maria Dorothea Pozonec, Clemens Aigner, Joachim Widder, Kristiina Boettiger, Zsolt Megyesfalvi, Balazs Dome

Purpose of review: Small cell lung cancer (SCLC) is an aggressive disease with a poor prognosis, whereas its metastatic capacity carries a predilection for the brain. Although prophylactic cranial irradiation (PCI) has been used to address this problem, upcoming alternatives might necessitate reflection of its application in SCLC treatment.

Recent findings: The addition of immunotherapy to treatment guidelines has provided a new strategy for the management of brain metastases. Complementation of immunotherapy with active MRI surveillance could potentially replace PCI and avoid irradiation-related cognitive side effects. SCLC's molecular profile is heterogeneous, with differential response to treatment modalities between subgroups. Investigation of these variances might be essential to improve therapeutic outcomes in SCLC patients.

Summary: The role of PCI in SCLC treatment must be examined in light of immunotherapy. We summarize recent results, bearing SCLC subtypes and therapeutic vulnerabilities in mind, to derive tailored treatment strategies for SCLC patients in future settings.

综述目的:小细胞肺癌(SCLC)是一种预后不良的侵袭性疾病,其转移能力倾向于脑部。虽然预防性颅脑照射(PCI)已被用于解决这一问题,但未来的替代方案可能需要反思其在SCLC治疗中的应用。最近的发现:在治疗指南中加入免疫疗法为脑转移的治疗提供了一种新的策略。免疫治疗与主动MRI监测的补充可能潜在地取代PCI,并避免辐射相关的认知副作用。SCLC的分子特征是异质性的,不同亚组对治疗方式的反应不同。研究这些差异可能对改善SCLC患者的治疗效果至关重要。总结:PCI在SCLC治疗中的作用必须结合免疫治疗来考察。我们总结了最近的研究结果,考虑到SCLC亚型和治疗脆弱性,以便在未来的环境中为SCLC患者提供量身定制的治疗策略。
{"title":"Prophylactic cranial irradiation for small cell lung cancer in the era of immunotherapy and molecular subtypes.","authors":"Veronika Pozonec, Maria Dorothea Pozonec, Clemens Aigner, Joachim Widder, Kristiina Boettiger, Zsolt Megyesfalvi, Balazs Dome","doi":"10.1097/CCO.0000000000001111","DOIUrl":"https://doi.org/10.1097/CCO.0000000000001111","url":null,"abstract":"<p><strong>Purpose of review: </strong>Small cell lung cancer (SCLC) is an aggressive disease with a poor prognosis, whereas its metastatic capacity carries a predilection for the brain. Although prophylactic cranial irradiation (PCI) has been used to address this problem, upcoming alternatives might necessitate reflection of its application in SCLC treatment.</p><p><strong>Recent findings: </strong>The addition of immunotherapy to treatment guidelines has provided a new strategy for the management of brain metastases. Complementation of immunotherapy with active MRI surveillance could potentially replace PCI and avoid irradiation-related cognitive side effects. SCLC's molecular profile is heterogeneous, with differential response to treatment modalities between subgroups. Investigation of these variances might be essential to improve therapeutic outcomes in SCLC patients.</p><p><strong>Summary: </strong>The role of PCI in SCLC treatment must be examined in light of immunotherapy. We summarize recent results, bearing SCLC subtypes and therapeutic vulnerabilities in mind, to derive tailored treatment strategies for SCLC patients in future settings.</p>","PeriodicalId":10893,"journal":{"name":"Current Opinion in Oncology","volume":" ","pages":""},"PeriodicalIF":2.8,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142767105","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Chimeric antigen receptor adoptive immunotherapy in central nervous system tumors: state of the art on clinical trials, challenges, and emerging strategies to addressing them. 中枢神经系统肿瘤的嵌合抗原受体采纳性免疫疗法:临床试验、挑战和新兴应对策略的最新进展。
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-11-01 Epub Date: 2024-07-11 DOI: 10.1097/CCO.0000000000001076
Giada Del Baldo, Andrea Carai, Angela Mastronuzzi

Purpose of review: Central nervous system (CNS) tumors represent a significant unmet medical need due to their enduring burden of high mortality and morbidity. Chimeric antigen receptor (CAR) T-cell therapy emerges as a groundbreaking approach, offering hope for improved treatment outcomes. However, despite its successes in hematological malignancies, its efficacy in solid tumors, including CNS tumors, remains limited. Challenges such as the intricate tumor microenvironment (TME), antigenic heterogeneity, and CAR T-cell exhaustion hinder its effectiveness. This review aims to explore the current landscape of CAR T-cell therapy for CNS tumors, highlighting recent advancements and addressing challenges in achieving therapeutic efficacy.

Recent findings: Innovative strategies aim to overcome the barriers posed by the TME and antigen diversity, prevent CAR T-cell exhaustion through engineering approaches and combination therapies with immune checkpoint inhibitors to improving treatment outcomes.

Summary: Researchers have been actively working to address these challenges. Moreover, addressing the unique challenges associated with neurotoxicity in CNS tumors requires specialized management strategies. These may include the development of grading systems, monitoring devices, alternative cell platforms and incorporation of suicide genes. Continued research efforts and clinical advancements are paramount to overcoming the existing challenges and realizing the full potential of CAR T-cell therapy in treating CNS tumors.

审查目的:中枢神经系统(CNS)肿瘤死亡率和发病率居高不下,是一项尚未得到满足的重大医疗需求。嵌合抗原受体(CAR)T 细胞疗法作为一种突破性方法出现,为改善治疗效果带来了希望。然而,尽管这种疗法在血液恶性肿瘤中取得了成功,但在实体瘤(包括中枢神经系统肿瘤)中的疗效仍然有限。复杂的肿瘤微环境(TME)、抗原异质性和CAR T细胞衰竭等挑战阻碍了它的有效性。本综述旨在探讨中枢神经系统肿瘤CAR T细胞疗法的现状,重点介绍最新进展,并探讨实现疗效所面临的挑战:创新策略旨在克服TME和抗原多样性带来的障碍,通过工程方法和与免疫检查点抑制剂的联合疗法防止CAR T细胞衰竭,以改善治疗效果:研究人员一直在积极应对这些挑战。此外,应对中枢神经系统肿瘤神经毒性相关的独特挑战需要专门的管理策略。这些策略可能包括开发分级系统、监测设备、替代细胞平台和加入自杀基因。要克服现有挑战,充分发挥 CAR T 细胞疗法治疗中枢神经系统肿瘤的潜力,持续的研究努力和临床进展至关重要。
{"title":"Chimeric antigen receptor adoptive immunotherapy in central nervous system tumors: state of the art on clinical trials, challenges, and emerging strategies to addressing them.","authors":"Giada Del Baldo, Andrea Carai, Angela Mastronuzzi","doi":"10.1097/CCO.0000000000001076","DOIUrl":"10.1097/CCO.0000000000001076","url":null,"abstract":"<p><strong>Purpose of review: </strong>Central nervous system (CNS) tumors represent a significant unmet medical need due to their enduring burden of high mortality and morbidity. Chimeric antigen receptor (CAR) T-cell therapy emerges as a groundbreaking approach, offering hope for improved treatment outcomes. However, despite its successes in hematological malignancies, its efficacy in solid tumors, including CNS tumors, remains limited. Challenges such as the intricate tumor microenvironment (TME), antigenic heterogeneity, and CAR T-cell exhaustion hinder its effectiveness. This review aims to explore the current landscape of CAR T-cell therapy for CNS tumors, highlighting recent advancements and addressing challenges in achieving therapeutic efficacy.</p><p><strong>Recent findings: </strong>Innovative strategies aim to overcome the barriers posed by the TME and antigen diversity, prevent CAR T-cell exhaustion through engineering approaches and combination therapies with immune checkpoint inhibitors to improving treatment outcomes.</p><p><strong>Summary: </strong>Researchers have been actively working to address these challenges. Moreover, addressing the unique challenges associated with neurotoxicity in CNS tumors requires specialized management strategies. These may include the development of grading systems, monitoring devices, alternative cell platforms and incorporation of suicide genes. Continued research efforts and clinical advancements are paramount to overcoming the existing challenges and realizing the full potential of CAR T-cell therapy in treating CNS tumors.</p>","PeriodicalId":10893,"journal":{"name":"Current Opinion in Oncology","volume":" ","pages":"545-553"},"PeriodicalIF":2.8,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11460750/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141579229","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Modeling the management of patients with human epidermal growth factor receptor 2-positive breast cancer with liquid biopsy: the future of precision medicine. 利用液体活检建立人类表皮生长因子受体 2 阳性乳腺癌患者管理模型:精准医疗的未来。
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-11-01 Epub Date: 2024-07-15 DOI: 10.1097/CCO.0000000000001082
Eleonora Nicolò, Caterina Gianni, Giuseppe Curigliano, Carolina Reduzzi, Massimo Cristofanilli

Purpose of review: In the evolving landscape of human epidermal growth factor receptor 2 (HER2)-positive breast cancer (BC) management, liquid biopsy offers unprecedented opportunities for guiding clinical decisions. Here, we review the most recent findings on liquid biopsy applications in HER2-positive BC and its potential role in addressing challenges specific to this BC subtype.

Recent findings: Recent studies have highlighted the significance of liquid biopsy analytes, primarily circulating tumor DNA (ctDNA) and circulating tumor cells (CTCs), in stratifying patients' prognosis, predicting treatment response, and monitoring tumor evolution in both early and advanced stages of BC. Liquid biopsy holds promise in studying minimal residual disease to detect and potentially treat disease recurrence before it manifests clinically. Additionally, liquid biopsy may have significant implication in the management of brain metastasis, a major challenge in HER2-positive BC, and could redefine parameters for determining HER2 positivity. Combining ctDNA and CTCs is crucial for a comprehensive understanding of HER2-positive tumors, as they provide complementary insights.

Summary: Research efforts are needed to address analytical challenges, validate, and broaden the application of liquid biopsy in HER2-positive BC. This effort will ultimately facilitate its integration into clinical practice, optimizing the care of patients with HER2-positive tumors.

综述的目的:在人类表皮生长因子受体 2 (HER2) 阳性乳腺癌 (BC) 的治疗中,液体活检为指导临床决策提供了前所未有的机会。在此,我们回顾了液体活检在HER2阳性乳腺癌中应用的最新发现,以及它在应对这一乳腺癌亚型特有挑战方面的潜在作用:最近的研究强调了液体活检分析物(主要是循环肿瘤DNA(ctDNA)和循环肿瘤细胞(CTCs))在对患者预后进行分层、预测治疗反应以及监测BC早期和晚期肿瘤演变方面的重要性。液体活检有望用于研究微小残留病,以便在临床表现出疾病复发之前发现并治疗疾病复发。此外,液体活检可能对脑转移(HER2 阳性 BC 的一大挑战)的治疗具有重要意义,并能重新定义确定 HER2 阳性的参数。将ctDNA和CTC结合起来对全面了解HER2阳性肿瘤至关重要,因为它们能提供互补的见解:需要努力开展研究,以解决分析难题、验证并扩大液体活检在 HER2 阳性 BC 中的应用。这项工作最终将促进液体活检与临床实践的结合,优化对HER2阳性肿瘤患者的治疗。
{"title":"Modeling the management of patients with human epidermal growth factor receptor 2-positive breast cancer with liquid biopsy: the future of precision medicine.","authors":"Eleonora Nicolò, Caterina Gianni, Giuseppe Curigliano, Carolina Reduzzi, Massimo Cristofanilli","doi":"10.1097/CCO.0000000000001082","DOIUrl":"10.1097/CCO.0000000000001082","url":null,"abstract":"<p><strong>Purpose of review: </strong>In the evolving landscape of human epidermal growth factor receptor 2 (HER2)-positive breast cancer (BC) management, liquid biopsy offers unprecedented opportunities for guiding clinical decisions. Here, we review the most recent findings on liquid biopsy applications in HER2-positive BC and its potential role in addressing challenges specific to this BC subtype.</p><p><strong>Recent findings: </strong>Recent studies have highlighted the significance of liquid biopsy analytes, primarily circulating tumor DNA (ctDNA) and circulating tumor cells (CTCs), in stratifying patients' prognosis, predicting treatment response, and monitoring tumor evolution in both early and advanced stages of BC. Liquid biopsy holds promise in studying minimal residual disease to detect and potentially treat disease recurrence before it manifests clinically. Additionally, liquid biopsy may have significant implication in the management of brain metastasis, a major challenge in HER2-positive BC, and could redefine parameters for determining HER2 positivity. Combining ctDNA and CTCs is crucial for a comprehensive understanding of HER2-positive tumors, as they provide complementary insights.</p><p><strong>Summary: </strong>Research efforts are needed to address analytical challenges, validate, and broaden the application of liquid biopsy in HER2-positive BC. This effort will ultimately facilitate its integration into clinical practice, optimizing the care of patients with HER2-positive tumors.</p>","PeriodicalId":10893,"journal":{"name":"Current Opinion in Oncology","volume":" ","pages":"503-513"},"PeriodicalIF":2.8,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141619600","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Editorial introductions. 编辑介绍。
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-11-01 Epub Date: 2024-10-10 DOI: 10.1097/CCO.0000000000001095
{"title":"Editorial introductions.","authors":"","doi":"10.1097/CCO.0000000000001095","DOIUrl":"https://doi.org/10.1097/CCO.0000000000001095","url":null,"abstract":"","PeriodicalId":10893,"journal":{"name":"Current Opinion in Oncology","volume":"36 6","pages":"vii-viii"},"PeriodicalIF":2.8,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142892253","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Novel PET tracers in breast cancer for treatment optimization: clinical utility and future perspectives. 用于乳腺癌治疗优化的新型 PET 示踪剂:临床实用性和未来展望。
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-11-01 Epub Date: 2024-06-12 DOI: 10.1097/CCO.0000000000001057
Laura Gilardi, Lighea Simona Airò Farulla, Elena Bonatto, Francesco Ceci

Purpose of review: There is a critical need for timely and accurate decisions in breast cancer management. This narrative review aims to clarify the potential role of novel PET tracers in optimizing treatment strategies for different breast cancer subtypes.

Recent findings: 2-deoxy-2-[18F]-fluoro-D-glucose PET/computed tomography (FDG PET/CT) has a pivotal role in response assessment in metastatic breast cancer, despite its limitations in certain histological and molecular subtypes. Further PET radiopharmaceuticals have been proposed to address these clinical needing.

Summary: FES PET/CT demonstrates its usefulness in assessing ER expression and predicting response to therapy in luminal breast cancer, with implications for treatment optimization and monitoring. In HER2-positive and HER2-low breast cancer, HER2-targeted PET tracers show potential in assessing HER2 status, while their accuracy in predicting response to targeted therapies is still debated. PARP-targeted PET imaging holds potential for selecting patients for PARP inhibitors treatments, particularly in triple-negative breast cancer (TNBC), where imaging tools are crucial due to the absence of specific targets. Immunotherapy and antibody-drug conjugates (ADCs) are emerging treatment options for TNBC, and PET imaging targeting immune checkpoints could aid in treatment selection and response monitoring. The dynamic role of PET/CT imaging in tailoring breast cancer treatments requires further multidisciplinary research to validate the clinical utility of targeted tracers.

审查目的:乳腺癌治疗亟需及时、准确的决策。最新发现:2-脱氧-2-[18F]-氟-D-葡萄糖 PET/计算机断层扫描(FDG PET/CT)在转移性乳腺癌的反应评估中具有关键作用,尽管它在某些组织学和分子亚型中存在局限性。为了满足这些临床需求,人们提出了更多的 PET 放射性药物:FES PET/CT 证明了它在评估ER表达和预测腔隙性乳腺癌治疗反应方面的作用,并对治疗优化和监测产生了影响。在 HER2 阳性和 HER2 低的乳腺癌中,HER2 靶向 PET 示踪剂显示出评估 HER2 状态的潜力,但其预测靶向治疗反应的准确性仍存在争议。PARP靶向PET成像在选择PARP抑制剂治疗患者方面具有潜力,特别是在三阴性乳腺癌(TNBC)中,由于缺乏特异性靶点,成像工具至关重要。免疫疗法和抗体药物共轭物(ADCs)是 TNBC 的新兴治疗方法,针对免疫检查点的 PET 成像有助于治疗选择和反应监测。PET/CT 成像在定制乳腺癌治疗中的动态作用需要进一步的多学科研究来验证靶向示踪剂的临床效用。
{"title":"Novel PET tracers in breast cancer for treatment optimization: clinical utility and future perspectives.","authors":"Laura Gilardi, Lighea Simona Airò Farulla, Elena Bonatto, Francesco Ceci","doi":"10.1097/CCO.0000000000001057","DOIUrl":"10.1097/CCO.0000000000001057","url":null,"abstract":"<p><strong>Purpose of review: </strong>There is a critical need for timely and accurate decisions in breast cancer management. This narrative review aims to clarify the potential role of novel PET tracers in optimizing treatment strategies for different breast cancer subtypes.</p><p><strong>Recent findings: </strong>2-deoxy-2-[18F]-fluoro-D-glucose PET/computed tomography (FDG PET/CT) has a pivotal role in response assessment in metastatic breast cancer, despite its limitations in certain histological and molecular subtypes. Further PET radiopharmaceuticals have been proposed to address these clinical needing.</p><p><strong>Summary: </strong>FES PET/CT demonstrates its usefulness in assessing ER expression and predicting response to therapy in luminal breast cancer, with implications for treatment optimization and monitoring. In HER2-positive and HER2-low breast cancer, HER2-targeted PET tracers show potential in assessing HER2 status, while their accuracy in predicting response to targeted therapies is still debated. PARP-targeted PET imaging holds potential for selecting patients for PARP inhibitors treatments, particularly in triple-negative breast cancer (TNBC), where imaging tools are crucial due to the absence of specific targets. Immunotherapy and antibody-drug conjugates (ADCs) are emerging treatment options for TNBC, and PET imaging targeting immune checkpoints could aid in treatment selection and response monitoring. The dynamic role of PET/CT imaging in tailoring breast cancer treatments requires further multidisciplinary research to validate the clinical utility of targeted tracers.</p>","PeriodicalId":10893,"journal":{"name":"Current Opinion in Oncology","volume":" ","pages":"514-520"},"PeriodicalIF":2.8,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141619601","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Endocrine therapy for early breast cancer in the era of oral selective estrogen receptor degraders: challenges and future perspectives. 口服选择性雌激素受体降解剂时代的早期乳腺癌内分泌治疗:挑战与未来展望。
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-11-01 Epub Date: 2024-08-12 DOI: 10.1097/CCO.0000000000001085
Liliana Ascione, Grazia Castellano, Giuseppe Curigliano, Paola Zagami

Purpose of review: Growth and survival of hormone receptor positive breast cancer cells are dependent on circulating hormones (e.g., estrogen and progesterone). Endocrine therapy improved outcomes in both early and advanced hormone receptor positive breast cancer. These treatments include drugs with different mechanisms of action, namely selective estrogen receptor modulators (SERM), aromatase inhibitors, and selective estrogen receptor degraders (SERDs). SERDs represent estrogen receptor antagonists, favoring its degradation and thus interfering with proliferation genes transcription and activation. Fulvestrant is the first approved SERD, administered intramuscularly for treating advanced breast cancer.

Recent findings: Oral SERDs have been tested to overcome the limitation of the intramuscular administration, and to increase SERD bioavailability. Recently, an oral SERD, Elacestrant, has been approved by the Food and Drug Administration (FDA) for patients carrying an ESR1 mutation. In fact, oral SERDs seem to be effective in tumors harboring ESR1 mutations, a well known mechanism of resistance to endocrine therapy (especially aromatase inhibitors).

Summary: More recently, oral SERDs have been tested in patients with early hormone receptor positive breast cancer, although their impact on survival and in this curative setting compared to standard endocrine therapy still needs to be elucidated. The best timing and duration of SERD administration and specific biomarkers in (neo)adjuvant setting remain largely unknown.

综述的目的:激素受体阳性乳腺癌细胞的生长和存活依赖于循环激素(如雌激素和孕酮)。内分泌治疗可改善早期和晚期激素受体阳性乳腺癌的治疗效果。这些疗法包括具有不同作用机制的药物,即选择性雌激素受体调节剂(SERM)、芳香化酶抑制剂和选择性雌激素受体降解剂(SERDs)。选择性雌激素受体降解剂是雌激素受体拮抗剂,有利于雌激素受体的降解,从而干扰增殖基因的转录和激活。氟维司群是首个获得批准的 SERD,通过肌肉注射治疗晚期乳腺癌:为了克服肌肉注射的局限性,并提高 SERD 的生物利用度,人们对口服 SERD 进行了测试。最近,美国食品和药物管理局(FDA)批准了一种口服 SERD 药物 Elacestrant,用于治疗携带 ESR1 基因突变的患者。事实上,口服 SERDs 似乎对携带 ESR1 突变的肿瘤有效,而 ESR1 突变是众所周知的内分泌治疗(尤其是芳香化酶抑制剂)的耐药机制。摘要:最近,口服 SERDs 已在早期激素受体阳性乳腺癌患者中进行了测试,但与标准内分泌治疗相比,口服 SERDs 对患者生存期和治愈率的影响仍有待阐明。在(新)辅助治疗中,服用SERD的最佳时机和持续时间以及特定的生物标志物在很大程度上仍是未知数。
{"title":"Endocrine therapy for early breast cancer in the era of oral selective estrogen receptor degraders: challenges and future perspectives.","authors":"Liliana Ascione, Grazia Castellano, Giuseppe Curigliano, Paola Zagami","doi":"10.1097/CCO.0000000000001085","DOIUrl":"10.1097/CCO.0000000000001085","url":null,"abstract":"<p><strong>Purpose of review: </strong>Growth and survival of hormone receptor positive breast cancer cells are dependent on circulating hormones (e.g., estrogen and progesterone). Endocrine therapy improved outcomes in both early and advanced hormone receptor positive breast cancer. These treatments include drugs with different mechanisms of action, namely selective estrogen receptor modulators (SERM), aromatase inhibitors, and selective estrogen receptor degraders (SERDs). SERDs represent estrogen receptor antagonists, favoring its degradation and thus interfering with proliferation genes transcription and activation. Fulvestrant is the first approved SERD, administered intramuscularly for treating advanced breast cancer.</p><p><strong>Recent findings: </strong>Oral SERDs have been tested to overcome the limitation of the intramuscular administration, and to increase SERD bioavailability. Recently, an oral SERD, Elacestrant, has been approved by the Food and Drug Administration (FDA) for patients carrying an ESR1 mutation. In fact, oral SERDs seem to be effective in tumors harboring ESR1 mutations, a well known mechanism of resistance to endocrine therapy (especially aromatase inhibitors).</p><p><strong>Summary: </strong>More recently, oral SERDs have been tested in patients with early hormone receptor positive breast cancer, although their impact on survival and in this curative setting compared to standard endocrine therapy still needs to be elucidated. The best timing and duration of SERD administration and specific biomarkers in (neo)adjuvant setting remain largely unknown.</p>","PeriodicalId":10893,"journal":{"name":"Current Opinion in Oncology","volume":" ","pages":"465-473"},"PeriodicalIF":2.8,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11460762/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142153365","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Current Opinion in Oncology
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