首页 > 最新文献

Anti-cancer agents in medicinal chemistry最新文献

英文 中文
A Comprehensive Overview of the Therapeutic Potential of Pyrimidine Analogues in the Management of Colorectal Cancer. 嘧啶类似物在结直肠癌治疗中的潜力综述。
IF 3 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-07-28 DOI: 10.2174/0118715206381536250709201255
Sathishkumar M, Dhunmati K, Ramalakshmi N, Nalini C N, Dibenthiran S, Indrakumar S

Colorectal cancer is the fourth most prevalent cause of cancer-related fatalities, and cancer is still one of the major causes of death globally. Although pyrimidine analogues can target abnormal cellular growth, they have demonstrated potential as therapeutic treatments for a variety of malignancies, including colon cancer. A vital nitrogen-containing aromatic heterocyclic molecule, pyrimidine is an important target for cancer treatments since it is involved in the construction of DNA and RNA. Recent research has investigated the synthesis and assessment of a number of pyrimidine derivatives, demonstrating their capacity to block particular enzymes and pathways linked to the development of cancer. According to the review, a number of pyrimidine-based chemical families, such as 1H-pyrazolo, diarylpyrazolo, and bromo-pyrimidine derivatives, have shown strong anti-cancer properties in preclinical models, especially against lung and colon cancer cell lines. Moreover, these compounds' structure-activity relationship (SAR) studies show that changes made at important positions on the pyrimidine scaffold improve their anticancer efficacy. These findings highlight the significance of further study into pyrimidine analogues as promising candidates for cancer therapy.

结直肠癌是癌症相关死亡的第四大最普遍原因,癌症仍然是全球主要死亡原因之一。虽然嘧啶类似物可以靶向异常细胞生长,但它们已被证明具有治疗多种恶性肿瘤的潜力,包括结肠癌。嘧啶是一种重要的含氮芳香杂环分子,因其参与DNA和RNA的构建而成为癌症治疗的重要靶点。最近的研究调查了一些嘧啶衍生物的合成和评估,证明了它们能够阻断与癌症发展有关的特定酶和途径。根据综述,一些以嘧啶为基础的化学家族,如1h -吡唑啉、二芳基吡唑啉和溴嘧啶衍生物,在临床前模型中显示出很强的抗癌特性,特别是对肺癌和结肠癌细胞系。此外,这些化合物的构效关系(SAR)研究表明,在嘧啶支架上重要位置的改变提高了它们的抗癌功效。这些发现强调了进一步研究嘧啶类似物作为癌症治疗有希望的候选者的重要性。
{"title":"A Comprehensive Overview of the Therapeutic Potential of Pyrimidine Analogues in the Management of Colorectal Cancer.","authors":"Sathishkumar M, Dhunmati K, Ramalakshmi N, Nalini C N, Dibenthiran S, Indrakumar S","doi":"10.2174/0118715206381536250709201255","DOIUrl":"https://doi.org/10.2174/0118715206381536250709201255","url":null,"abstract":"<p><p>Colorectal cancer is the fourth most prevalent cause of cancer-related fatalities, and cancer is still one of the major causes of death globally. Although pyrimidine analogues can target abnormal cellular growth, they have demonstrated potential as therapeutic treatments for a variety of malignancies, including colon cancer. A vital nitrogen-containing aromatic heterocyclic molecule, pyrimidine is an important target for cancer treatments since it is involved in the construction of DNA and RNA. Recent research has investigated the synthesis and assessment of a number of pyrimidine derivatives, demonstrating their capacity to block particular enzymes and pathways linked to the development of cancer. According to the review, a number of pyrimidine-based chemical families, such as 1H-pyrazolo, diarylpyrazolo, and bromo-pyrimidine derivatives, have shown strong anti-cancer properties in preclinical models, especially against lung and colon cancer cell lines. Moreover, these compounds' structure-activity relationship (SAR) studies show that changes made at important positions on the pyrimidine scaffold improve their anticancer efficacy. These findings highlight the significance of further study into pyrimidine analogues as promising candidates for cancer therapy.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-07-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144741008","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
The Potential of Next-generation Multi-functional Nanoplatforms for Breast Cancer. 下一代多功能乳腺癌纳米平台的潜力。
IF 3 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-07-24 DOI: 10.2174/0118715206392103250715115020
Shreya Gupta, Tanmay J Urs, Navya Aggarwal, Shinjini Sen, Banashree Bondhopadhyay

The next-generation nanoparticles overcome the drawbacks of early nanoplatforms by integrating multiple functions, such as drug delivery, controlled drug release, and combination therapy, into a single system. This study examines the biomedical applications of quantum dots, carbon nanotubes, superparamagnetic iron oxide nanoparticles, and layered double hydroxides for the delivery of breast cancer drugs. They are termed as "nextgeneration" nanoparticles, as they are advanced nanocarriers that offer a comprehensive and alternative approach towards breast cancer treatment, providing enhanced specificity and efficacy compared to their predecessors. The development of these nanoplatforms has significantly enhanced drug bioavailability and reduced toxicity. A comprehensive analysis of a nanotechnology-based drug delivery system was conducted. The keywords used for this review were "Breast Cancer", "Targeted Drug Delivery", "Quantum Dots", "Carbon Nanotubes", "Layer Double Hydroxides", and "Superparamagnetic Iron Oxide Nanoparticles". The inclusion criteria consisted of studies focusing on breast cancer, targeted drug delivery, and therapeutic applications of these nanocarriers. In contrast, exclusion criteria included studies focusing on the synthesis of nanocarriers and the diagnostic applications of these nanostructures. The study underscores their mechanisms, limitations, and future development directions. Additionally, the study tracks the evolution of the nanocarriers since their early discovery. Next-generation nanocarriers (QDs, CNTs, SPIONs, and LDHs) have strong therapeutic potential owing to their precisely engineered properties, such as size, shape, morphology, and surface modifications. Their trigger-initiated drug release mechanisms enable targeted delivery with a better rate of tumor penetration, while their ability to co-deliver multiple therapeutic agents addresses drug resistance issues and provides synergistic effects. Comparative analyses have revealed that these advanced nanoplatforms significantly outperform early-generation carriers in terms of bioavailability, reduced toxicity, and treatment efficacy across various breast cancer types. Next-generation nanoplatforms offer unprecedented opportunities for targeted and efficient cancer treatment. Continued research and innovation are necessary to address existing challenges and to optimize their therapeutic potential for clinical applications.

新一代纳米粒子克服了早期纳米平台的缺点,将多种功能(如药物传递、药物控制释放和联合治疗)集成到一个单一系统中。本研究探讨了量子点、碳纳米管、超顺磁性氧化铁纳米粒子和层状双氢氧化物在乳腺癌药物输送中的生物医学应用。它们被称为“下一代”纳米颗粒,因为它们是先进的纳米载体,为乳腺癌治疗提供了一种全面的替代方法,与它们的前辈相比,提供了更高的特异性和有效性。这些纳米平台的发展显著提高了药物的生物利用度,降低了毒性。对基于纳米技术的给药系统进行了综合分析。本综述使用的关键词是“乳腺癌”、“靶向药物递送”、“量子点”、“碳纳米管”、“层双氢氧化物”和“超顺磁性氧化铁纳米颗粒”。纳入标准包括关注乳腺癌、靶向药物递送和这些纳米载体的治疗应用的研究。相比之下,排除标准包括纳米载体的合成和这些纳米结构的诊断应用的研究。研究强调了它们的作用机制、局限性和未来的发展方向。此外,该研究还追踪了纳米载体自早期发现以来的演变过程。下一代纳米载体(量子点、碳纳米管、SPIONs和LDHs)由于其精确的工程特性,如尺寸、形状、形态和表面修饰,具有强大的治疗潜力。它们的触发启动药物释放机制使靶向递送具有更好的肿瘤穿透率,同时它们共同递送多种治疗药物的能力解决了耐药性问题并提供了协同效应。对比分析表明,这些先进的纳米平台在生物利用度、降低毒性和治疗各种乳腺癌类型方面明显优于早期载体。下一代纳米平台为靶向和有效的癌症治疗提供了前所未有的机会。持续的研究和创新是解决现有挑战和优化其临床应用的治疗潜力的必要条件。
{"title":"The Potential of Next-generation Multi-functional Nanoplatforms for Breast Cancer.","authors":"Shreya Gupta, Tanmay J Urs, Navya Aggarwal, Shinjini Sen, Banashree Bondhopadhyay","doi":"10.2174/0118715206392103250715115020","DOIUrl":"https://doi.org/10.2174/0118715206392103250715115020","url":null,"abstract":"<p><p>The next-generation nanoparticles overcome the drawbacks of early nanoplatforms by integrating multiple functions, such as drug delivery, controlled drug release, and combination therapy, into a single system. This study examines the biomedical applications of quantum dots, carbon nanotubes, superparamagnetic iron oxide nanoparticles, and layered double hydroxides for the delivery of breast cancer drugs. They are termed as \"nextgeneration\" nanoparticles, as they are advanced nanocarriers that offer a comprehensive and alternative approach towards breast cancer treatment, providing enhanced specificity and efficacy compared to their predecessors. The development of these nanoplatforms has significantly enhanced drug bioavailability and reduced toxicity. A comprehensive analysis of a nanotechnology-based drug delivery system was conducted. The keywords used for this review were \"Breast Cancer\", \"Targeted Drug Delivery\", \"Quantum Dots\", \"Carbon Nanotubes\", \"Layer Double Hydroxides\", and \"Superparamagnetic Iron Oxide Nanoparticles\". The inclusion criteria consisted of studies focusing on breast cancer, targeted drug delivery, and therapeutic applications of these nanocarriers. In contrast, exclusion criteria included studies focusing on the synthesis of nanocarriers and the diagnostic applications of these nanostructures. The study underscores their mechanisms, limitations, and future development directions. Additionally, the study tracks the evolution of the nanocarriers since their early discovery. Next-generation nanocarriers (QDs, CNTs, SPIONs, and LDHs) have strong therapeutic potential owing to their precisely engineered properties, such as size, shape, morphology, and surface modifications. Their trigger-initiated drug release mechanisms enable targeted delivery with a better rate of tumor penetration, while their ability to co-deliver multiple therapeutic agents addresses drug resistance issues and provides synergistic effects. Comparative analyses have revealed that these advanced nanoplatforms significantly outperform early-generation carriers in terms of bioavailability, reduced toxicity, and treatment efficacy across various breast cancer types. Next-generation nanoplatforms offer unprecedented opportunities for targeted and efficient cancer treatment. Continued research and innovation are necessary to address existing challenges and to optimize their therapeutic potential for clinical applications.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-07-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144741009","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
The Role of Kinase Inhibitors in Cancer Neuroscience: Mechanisms, Therapeutic Potential, and Future Directions. 激酶抑制剂在癌症神经科学中的作用:机制、治疗潜力和未来方向。
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-07-21 DOI: 10.2174/0118715206387712250711132221
Manos C Vlasiou

Introduction: Cancer progression is increasingly understood to be influenced by neural mechanisms, including neurotransmitter signaling, neurotrophic factor activity, neuroinflammation, and neurogenic inflammation. These neurobiological interactions contribute to tumor proliferation, angiogenesis, and metastasis. Kinase inhibitors, a class of targeted therapies that block dysregulated kinase activity, have demonstrated promise not only in direct tumor suppression but also in modulating neural pathways associated with cancer progression.

Methods: This review examines the role of kinase inhibitors in modulating cancer-associated neural mechanisms. A comprehensive literature search was conducted to identify studies exploring the effects of kinase inhibition on: (1) neurotransmitter signaling pathways; (2) neurotrophic factors such as nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF); (3) neuroinflammation through glial cell modulation; and (4) neurogenic inflammation. Additionally, we assessed the impact of kinase inhibitors on tumor-induced axonogenesis and stress-related signaling. Clinical relevance was evaluated through analysis of preclinical models, human case studies, and outcomes from relevant clinical trials.

Results: Kinase inhibitors were found to significantly modulate neural factors that facilitate tumor growth. Specifically, they can suppress neurotrophic signaling (e.g., NGF/TrkA, BDNF/TrkB), inhibit glial activation, reduce pro-inflammatory cytokine production, and block neurotransmitter-induced proliferation. Inhibition of stress-responsive kinases such as p38 MAPK and JNK also disrupted tumor-associated axonogenesis and inflammation. Clinical trials demonstrate improved outcomes in cancers such as glioblastoma, breast cancer, and pancreatic cancer when kinase inhibitors are employed with consideration of neural mechanisms.

Discussion: These findings support the emerging concept of targeting the neural tumor microenvironment as a therapeutic strategy. Kinase inhibitors represent a dual-action approach, suppressing both cancer cell intrinsic growth pathways and the neural factors that sustain them. However, several challenges persist, including resistance mechanisms, variability in patient neural profiles, and off-target effects. Future research should focus on the development of neural-specific kinase inhibitors, the use of neural biomarkers for therapy selection, and the integration of neuro-oncology into personalized treatment plans.

Conclusion: Kinase inhibitors offer a promising frontier in cancer treatment by targeting neural mechanisms that contribute to tumor progression. While current evidence is encouraging, further investigation is required to optimize their use within neuro-oncology. Personalized approaches and novel targets within the neural-cancer axis will be essential for translating thi

导言:癌症的进展越来越被理解为受神经机制的影响,包括神经递质信号、神经营养因子活性、神经炎症和神经源性炎症。这些神经生物学的相互作用促进了肿瘤的增殖、血管生成和转移。激酶抑制剂是一类阻断失调激酶活性的靶向疗法,不仅在直接抑制肿瘤方面有前景,而且在调节与癌症进展相关的神经通路方面也有前景。方法:本文综述了激酶抑制剂在调节癌症相关神经机制中的作用。我们进行了全面的文献检索,以确定探讨激酶抑制对:(1)神经递质信号通路的影响的研究;(2)神经营养因子,如神经生长因子(NGF)和脑源性神经营养因子(BDNF);(3)神经胶质细胞介导的神经炎症;(4)神经源性炎症。此外,我们评估了激酶抑制剂对肿瘤诱导的轴突发生和应激相关信号的影响。临床相关性通过分析临床前模型、人体病例研究和相关临床试验的结果来评估。结果:发现激酶抑制剂能显著调节促进肿瘤生长的神经因子。具体来说,它们可以抑制神经营养信号(如NGF/TrkA, BDNF/TrkB),抑制胶质细胞激活,减少促炎细胞因子的产生,并阻断神经递质诱导的增殖。抑制应激反应激酶如p38 MAPK和JNK也会破坏肿瘤相关的轴突发生和炎症。临床试验表明,在考虑神经机制的情况下使用激酶抑制剂,可以改善胶质母细胞瘤、乳腺癌和胰腺癌等癌症的治疗效果。讨论:这些发现支持了靶向神经肿瘤微环境作为治疗策略的新兴概念。激酶抑制剂代表了一种双重作用的方法,既抑制癌细胞内在的生长途径,又抑制维持它们的神经因子。然而,一些挑战仍然存在,包括耐药机制、患者神经谱的可变性和脱靶效应。未来的研究应侧重于开发神经特异性激酶抑制剂,使用神经生物标志物进行治疗选择,以及将神经肿瘤学纳入个性化治疗计划。结论:激酶抑制剂通过靶向促进肿瘤进展的神经机制,为癌症治疗提供了一个有前景的前沿。虽然目前的证据令人鼓舞,但需要进一步的研究来优化它们在神经肿瘤学中的应用。个性化的方法和神经-癌症轴内的新靶点对于将这一策略转化为临床实践和改善患者的长期预后至关重要。
{"title":"The Role of Kinase Inhibitors in Cancer Neuroscience: Mechanisms, Therapeutic Potential, and Future Directions.","authors":"Manos C Vlasiou","doi":"10.2174/0118715206387712250711132221","DOIUrl":"https://doi.org/10.2174/0118715206387712250711132221","url":null,"abstract":"<p><strong>Introduction: </strong>Cancer progression is increasingly understood to be influenced by neural mechanisms, including neurotransmitter signaling, neurotrophic factor activity, neuroinflammation, and neurogenic inflammation. These neurobiological interactions contribute to tumor proliferation, angiogenesis, and metastasis. Kinase inhibitors, a class of targeted therapies that block dysregulated kinase activity, have demonstrated promise not only in direct tumor suppression but also in modulating neural pathways associated with cancer progression.</p><p><strong>Methods: </strong>This review examines the role of kinase inhibitors in modulating cancer-associated neural mechanisms. A comprehensive literature search was conducted to identify studies exploring the effects of kinase inhibition on: (1) neurotransmitter signaling pathways; (2) neurotrophic factors such as nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF); (3) neuroinflammation through glial cell modulation; and (4) neurogenic inflammation. Additionally, we assessed the impact of kinase inhibitors on tumor-induced axonogenesis and stress-related signaling. Clinical relevance was evaluated through analysis of preclinical models, human case studies, and outcomes from relevant clinical trials.</p><p><strong>Results: </strong>Kinase inhibitors were found to significantly modulate neural factors that facilitate tumor growth. Specifically, they can suppress neurotrophic signaling (e.g., NGF/TrkA, BDNF/TrkB), inhibit glial activation, reduce pro-inflammatory cytokine production, and block neurotransmitter-induced proliferation. Inhibition of stress-responsive kinases such as p38 MAPK and JNK also disrupted tumor-associated axonogenesis and inflammation. Clinical trials demonstrate improved outcomes in cancers such as glioblastoma, breast cancer, and pancreatic cancer when kinase inhibitors are employed with consideration of neural mechanisms.</p><p><strong>Discussion: </strong>These findings support the emerging concept of targeting the neural tumor microenvironment as a therapeutic strategy. Kinase inhibitors represent a dual-action approach, suppressing both cancer cell intrinsic growth pathways and the neural factors that sustain them. However, several challenges persist, including resistance mechanisms, variability in patient neural profiles, and off-target effects. Future research should focus on the development of neural-specific kinase inhibitors, the use of neural biomarkers for therapy selection, and the integration of neuro-oncology into personalized treatment plans.</p><p><strong>Conclusion: </strong>Kinase inhibitors offer a promising frontier in cancer treatment by targeting neural mechanisms that contribute to tumor progression. While current evidence is encouraging, further investigation is required to optimize their use within neuro-oncology. Personalized approaches and novel targets within the neural-cancer axis will be essential for translating thi","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-07-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144688613","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
Innovative Nanocarriers: Magnetosomes in the Fight against Cancer. 创新纳米载体:抗癌中的磁小体。
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-07-17 DOI: 10.2174/0118715206377167250709062942
Shivani Yadav, Manoj Kumar Mishra

Recent advancements in medication formulations and drug delivery systems over the past two decades have improved patient adherence and pharmacological responses. Efficient, target-specific medication delivery remains challenging, with many current systems designed to minimize drug loss and degradation. Magnetosomes, as nanocarriers, show promise for delivering antibodies, vaccine DNA, and siRNA, enhancing the stability of chemotherapeutics, and enabling targeted delivery to malignant tumors. Targeted drug delivery is crucial in cancer treatment, as anticancer drugs often cannot differentiate between healthy and malignant cells, causing side effects and systemic toxicity. Magnetosome-based drug delivery offers a potential solution, minimizing adverse effects and promoting drug accumulation at the target site. This review covers the design, development, and advancements in magnetosome-based drug delivery for cancer therapy.

在过去二十年中,药物配方和药物输送系统的最新进展改善了患者的依从性和药理学反应。有效的、针对特定靶点的药物递送仍然具有挑战性,目前许多系统的设计都是为了尽量减少药物损失和降解。磁小体作为纳米载体,在递送抗体、疫苗DNA和siRNA、增强化疗药物的稳定性以及实现对恶性肿瘤的靶向递送方面显示出前景。靶向药物递送在癌症治疗中至关重要,因为抗癌药物通常无法区分健康细胞和恶性细胞,从而导致副作用和全身毒性。基于磁小体的药物递送提供了一种潜在的解决方案,可以最大限度地减少副作用并促进药物在目标部位的积累。本文综述了以磁小体为基础的肿瘤药物传递的设计、开发和进展。
{"title":"Innovative Nanocarriers: Magnetosomes in the Fight against Cancer.","authors":"Shivani Yadav, Manoj Kumar Mishra","doi":"10.2174/0118715206377167250709062942","DOIUrl":"https://doi.org/10.2174/0118715206377167250709062942","url":null,"abstract":"<p><p>Recent advancements in medication formulations and drug delivery systems over the past two decades have improved patient adherence and pharmacological responses. Efficient, target-specific medication delivery remains challenging, with many current systems designed to minimize drug loss and degradation. Magnetosomes, as nanocarriers, show promise for delivering antibodies, vaccine DNA, and siRNA, enhancing the stability of chemotherapeutics, and enabling targeted delivery to malignant tumors. Targeted drug delivery is crucial in cancer treatment, as anticancer drugs often cannot differentiate between healthy and malignant cells, causing side effects and systemic toxicity. Magnetosome-based drug delivery offers a potential solution, minimizing adverse effects and promoting drug accumulation at the target site. This review covers the design, development, and advancements in magnetosome-based drug delivery for cancer therapy.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144673782","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
Polyamines in Cancer: Mechanisms, Metabolic Targets, and Therapeutic Opportunities. 多胺在癌症中的作用:机制、代谢靶点和治疗机会。
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-07-17 DOI: 10.2174/0118715206390332250711060007
Azmi Yerlikaya

Introduction: Polyamine metabolism is essential for cancer cell growth, with enzymes like ornithine decarboxylase (ODC) and S-adenosylmethionine decarboxylase (AdoMetDC) playing key roles in polyamine (PA) biosynthesis. These polyamines (putrescine, spermidine, and spermine) regulate vital cellular processes, including DNA replication, protein synthesis, and cell cycle progression. Dysregulated polyamine metabolism is common in cancer, making ODC and AdoMetDC attractive therapeutic targets. This review highlights polyamines' role in cancer and explores combination therapies targeting polyamine metabolism and critical signaling pathways for improved clinical outcomes.

Methods: A comprehensive analysis of both historical and recent literature on polyamine metabolism in cancer was performed using PubMed, which provides access to over 37 million citations from biomedical literature. Expression data for key polyamine biosynthetic enzymes, ODC and AdoMetDC, were obtained from the UALCAN portal - an interactive web resource for the analysis of cancer OMICS data. The IUPAC names of drugs and inhibitors targeting the polyamine pathway were retrieved from the PubChem database and used to generate molecular structures using the BIOVIA Draw 2025 program. Additionally, the ClinicalTrials.gov database was explored to identify ongoing and completed clinical research studies, as well as to gather detailed information on therapeutic agents targeting polyamine metabolism.

Results: Aberrant polyamine metabolism in cancer is driven by oncogenic pathways like MYC, Akt, and mTOR. MYC upregulates ODC1, promoting polyamine dysregulation. Defects in enzymes such as MTA phosphorylase (MTAP) enhance cancer cell sensitivity to inhibitors of purine/pyrimidine synthesis and the ubiquitin-proteasome pathway, suggesting alternative therapeutic strategies.

Discussion: Therapeutic strategies combining polyamine biosynthesis inhibition with targeting nucleotide synthesis or proteasome function have shown synergistic potential. However, the dual nature of polyamines - supporting both, tumor growth and ferroptotic cell death - poses a therapeutic challenge. Balancing these effects is key to designing effective interventions. Advancing this field requires not only selective inhibitors but also a deeper understanding of context-dependent polyamine functions in tumor biology.

Conclusion: Developing more potent inhibitors with improved drug-like properties is crucial for advancing polyamine- targeted therapies and positioning this field at the forefront of cancer research.

多胺代谢对癌细胞生长至关重要,鸟氨酸脱羧酶(ODC)和s -腺苷蛋氨酸脱羧酶(AdoMetDC)等酶在多胺(PA)的生物合成中起着关键作用。这些多胺(腐胺、亚精胺和精胺)调节重要的细胞过程,包括DNA复制、蛋白质合成和细胞周期进程。多胺代谢失调在癌症中很常见,使ODC和AdoMetDC成为有吸引力的治疗靶点。这篇综述强调了多胺在癌症中的作用,并探讨了针对多胺代谢和关键信号通路的联合治疗以改善临床结果。方法:使用PubMed对癌症中多胺代谢的历史和近期文献进行综合分析,PubMed提供了超过3700万次生物医学文献的引用。关键多胺生物合成酶ODC和AdoMetDC的表达数据来自UALCAN门户网站——一个用于分析癌症组学数据的交互式网络资源。针对多胺途径的药物和抑制剂的IUPAC名称从PubChem数据库中检索,并使用BIOVIA Draw 2025程序生成分子结构。此外,还探索了ClinicalTrials.gov数据库,以确定正在进行和已完成的临床研究,以及收集针对多胺代谢的治疗剂的详细信息。结果:肿瘤多胺代谢异常是由MYC、Akt和mTOR等致癌途径驱动的。MYC上调ODC1,促进多胺失调。MTA磷酸化酶(MTAP)等酶的缺陷增强了癌细胞对嘌呤/嘧啶合成抑制剂和泛素-蛋白酶体途径的敏感性,提示了其他治疗策略。讨论:结合多胺生物合成抑制与靶向核苷酸合成或蛋白酶体功能的治疗策略显示出协同潜力。然而,多胺的双重性质——支持肿瘤生长和铁细胞死亡——给治疗带来了挑战。平衡这些影响是设计有效干预措施的关键。推进这一领域不仅需要选择性抑制剂,还需要更深入地了解肿瘤生物学中上下文依赖的多胺功能。结论:开发更有效的抑制剂,改善药物样特性,对于推进多胺靶向治疗和将该领域定位于癌症研究的前沿至关重要。
{"title":"Polyamines in Cancer: Mechanisms, Metabolic Targets, and Therapeutic Opportunities.","authors":"Azmi Yerlikaya","doi":"10.2174/0118715206390332250711060007","DOIUrl":"https://doi.org/10.2174/0118715206390332250711060007","url":null,"abstract":"<p><strong>Introduction: </strong>Polyamine metabolism is essential for cancer cell growth, with enzymes like ornithine decarboxylase (ODC) and S-adenosylmethionine decarboxylase (AdoMetDC) playing key roles in polyamine (PA) biosynthesis. These polyamines (putrescine, spermidine, and spermine) regulate vital cellular processes, including DNA replication, protein synthesis, and cell cycle progression. Dysregulated polyamine metabolism is common in cancer, making ODC and AdoMetDC attractive therapeutic targets. This review highlights polyamines' role in cancer and explores combination therapies targeting polyamine metabolism and critical signaling pathways for improved clinical outcomes.</p><p><strong>Methods: </strong>A comprehensive analysis of both historical and recent literature on polyamine metabolism in cancer was performed using PubMed, which provides access to over 37 million citations from biomedical literature. Expression data for key polyamine biosynthetic enzymes, ODC and AdoMetDC, were obtained from the UALCAN portal - an interactive web resource for the analysis of cancer OMICS data. The IUPAC names of drugs and inhibitors targeting the polyamine pathway were retrieved from the PubChem database and used to generate molecular structures using the BIOVIA Draw 2025 program. Additionally, the ClinicalTrials.gov database was explored to identify ongoing and completed clinical research studies, as well as to gather detailed information on therapeutic agents targeting polyamine metabolism.</p><p><strong>Results: </strong>Aberrant polyamine metabolism in cancer is driven by oncogenic pathways like MYC, Akt, and mTOR. MYC upregulates <i>ODC1</i>, promoting polyamine dysregulation. Defects in enzymes such as MTA phosphorylase (MTAP) enhance cancer cell sensitivity to inhibitors of purine/pyrimidine synthesis and the ubiquitin-proteasome pathway, suggesting alternative therapeutic strategies.</p><p><strong>Discussion: </strong>Therapeutic strategies combining polyamine biosynthesis inhibition with targeting nucleotide synthesis or proteasome function have shown synergistic potential. However, the dual nature of polyamines - supporting both, tumor growth and ferroptotic cell death - poses a therapeutic challenge. Balancing these effects is key to designing effective interventions. Advancing this field requires not only selective inhibitors but also a deeper understanding of context-dependent polyamine functions in tumor biology.</p><p><strong>Conclusion: </strong>Developing more potent inhibitors with improved drug-like properties is crucial for advancing polyamine- targeted therapies and positioning this field at the forefront of cancer research.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144673783","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
Design, Synthesis and Biological Evaluation of New 4-(4-(Methylsulfonyl) Phenyl)-6-Phenylpyrimidin-2-Amine Derivatives as Selective Cyclooxygenase (COX-2) Inhibitors. 新型4-(4-(甲基磺酰基)苯基)-6-苯基嘧啶-2-胺衍生物选择性环氧合酶抑制剂的设计、合成及生物学评价
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-07-16 DOI: 10.2174/0118715206380378250709112246
Shabnam Farzaneh, Mohammad Saeed Kordi, Mahsa Azami Movahed, Maryam Bayanati, Afshin Zarghi

Introduction: Cyclooxygenase, an enzyme that occurs in at least two distinct variants (COX-1 and COX-2), is the target of classical inhibitors, which lack selectivity and inhibit both types of COX. However, a recent approach focuses explicitly on inhibiting COX-2, commonly found in inflamed tissue, resulting in fewer adverse effects than COX-1 inhibitors.

Methods: A series of 4-(4-(methylsulfonyl)phenyl)-6-phenylpyrimidin-2-amine derivatives were synthesized through a two-step process. First, 4-substituted acetophenones underwent base-catalyzed Claisen-Schmidt condensation with 4-(methylsulfonyl)benzaldehyde to yield chalcones, which were then cyclized with guanidine hydrochloride under basic reflux conditions. Molecular docking was performed using AutoDock Vina software. The inhibitory activities of COX-1 and COX-2 were evaluated using enzymatic assays. Antiplatelet aggregation was measured via a turbidimetric method, and antiproliferative activity was assessed using the MTT assay.

Results: The in vitro experiments on COX inhibition revealed that a substantial number of the synthesized compounds presented a strong suppressive effect against COX-2. The assessment of antiplatelet aggregation activity indicated that most of the derivatives effectively inhibited ADP-induced platelet aggregation. Compound 4i exhibited the most potent antiproliferative activity, comparable to cisplatin. The docking studies and molecular modeling results demonstrated that the designed compounds, except for 4b, exhibited a binding behavior comparable to that of celecoxib. In addition, the insertion of the SO2Me moiety within the secondary binding site of COX-2 was observed.

Discussion: These findings suggest that the structural modifications introduced in the synthesized derivatives contribute significantly to their selective COX-2 inhibition and antiplatelet properties. The correlation between docking results and biological assays supports the rationale behind the design of the compound.

Conclusion: The 4-(4-(methylsulfonyl)phenyl)-6-phenylpyrimidin-2-amine exhibits unique properties as a COX-2 inhibitor, displaying effective inhibition of COX-2 while showing minimal interaction with the COX-1 enzyme. Furthermore, our study revealed that most of these compounds exhibited inhibitory effects on ADP-induced platelet aggregation.

环加氧酶是一种至少存在两种不同变体(COX-1和COX-2)的酶,是经典抑制剂的目标,缺乏选择性并抑制两种类型的COX。然而,最近的一种方法明确地关注于抑制COX-2,它通常存在于炎症组织中,比COX-1抑制剂产生更少的不良反应。方法:采用两步法合成一系列4-(4-(甲基磺酰基)苯基)-6-苯基嘧啶-2-胺衍生物。首先,4-取代苯乙酮与4-(甲基磺酰基)苯甲醛进行碱催化Claisen-Schmidt缩合反应,得到查尔酮,然后在碱性回流条件下与盐酸胍环化。使用AutoDock Vina软件进行分子对接。酶法测定COX-1和COX-2的抑制活性。通过浊度法测量抗血小板聚集,使用MTT法评估抗增殖活性。结果:体外COX抑制实验显示,大量合成的化合物对COX-2具有较强的抑制作用。抗血小板聚集活性评估表明,大多数衍生物能有效抑制adp诱导的血小板聚集。化合物4i表现出最有效的抗增殖活性,与顺铂相当。对接研究和分子建模结果表明,所设计的化合物除4b外,具有与塞来昔布相当的结合行为。此外,还观察到在COX-2的二级结合位点插入了SO2Me片段。讨论:这些发现表明,在合成的衍生物中引入的结构修饰对其选择性COX-2抑制和抗血小板性能有重要贡献。对接结果和生物分析之间的相关性支持了该化合物设计背后的基本原理。结论:4-(4-(甲基磺酰基)苯基)-6-苯基嘧啶-2-胺作为COX-2抑制剂具有独特的性质,对COX-2具有有效的抑制作用,同时与COX-1酶的相互作用最小。此外,我们的研究表明,大多数这些化合物对adp诱导的血小板聚集具有抑制作用。
{"title":"Design, Synthesis and Biological Evaluation of New 4-(4-(Methylsulfonyl) Phenyl)-6-Phenylpyrimidin-2-Amine Derivatives as Selective Cyclooxygenase (COX-2) Inhibitors.","authors":"Shabnam Farzaneh, Mohammad Saeed Kordi, Mahsa Azami Movahed, Maryam Bayanati, Afshin Zarghi","doi":"10.2174/0118715206380378250709112246","DOIUrl":"https://doi.org/10.2174/0118715206380378250709112246","url":null,"abstract":"<p><strong>Introduction: </strong>Cyclooxygenase, an enzyme that occurs in at least two distinct variants (COX-1 and COX-2), is the target of classical inhibitors, which lack selectivity and inhibit both types of COX. However, a recent approach focuses explicitly on inhibiting COX-2, commonly found in inflamed tissue, resulting in fewer adverse effects than COX-1 inhibitors.</p><p><strong>Methods: </strong>A series of 4-(4-(methylsulfonyl)phenyl)-6-phenylpyrimidin-2-amine derivatives were synthesized through a two-step process. First, 4-substituted acetophenones underwent base-catalyzed Claisen-Schmidt condensation with 4-(methylsulfonyl)benzaldehyde to yield chalcones, which were then cyclized with guanidine hydrochloride under basic reflux conditions. Molecular docking was performed using AutoDock Vina software. The inhibitory activities of COX-1 and COX-2 were evaluated using enzymatic assays. Antiplatelet aggregation was measured via a turbidimetric method, and antiproliferative activity was assessed using the MTT assay.</p><p><strong>Results: </strong>The in vitro experiments on COX inhibition revealed that a substantial number of the synthesized compounds presented a strong suppressive effect against COX-2. The assessment of antiplatelet aggregation activity indicated that most of the derivatives effectively inhibited ADP-induced platelet aggregation. Compound 4i exhibited the most potent antiproliferative activity, comparable to cisplatin. The docking studies and molecular modeling results demonstrated that the designed compounds, except for 4b, exhibited a binding behavior comparable to that of celecoxib. In addition, the insertion of the SO2Me moiety within the secondary binding site of COX-2 was observed.</p><p><strong>Discussion: </strong>These findings suggest that the structural modifications introduced in the synthesized derivatives contribute significantly to their selective COX-2 inhibition and antiplatelet properties. The correlation between docking results and biological assays supports the rationale behind the design of the compound.</p><p><strong>Conclusion: </strong>The 4-(4-(methylsulfonyl)phenyl)-6-phenylpyrimidin-2-amine exhibits unique properties as a COX-2 inhibitor, displaying effective inhibition of COX-2 while showing minimal interaction with the COX-1 enzyme. Furthermore, our study revealed that most of these compounds exhibited inhibitory effects on ADP-induced platelet aggregation.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144658177","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
Investigating the Therapeutic Potential of Cisplatin- and Rutin-Loaded Nanoliposomes against Colorectal Cancer Cells. 研究负载顺铂和芦丁的纳米脂质体对结直肠癌细胞的治疗潜力。
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-07-16 DOI: 10.2174/0118715206388843250709121645
Abdulazeez M Al-Mashhadani, Ali Al-Samydai, Ibrahim Al-Deeb, Simone Carradori, Hanan Azzam, Hussein S Janabi, Dana Al Qudah, Walhan Alshaer, M R Mozafari

Introduction: Colorectal cancer is an important cause of cancer-related mortality, necessitating innovative therapies to improve efficacy and reduce side effects. This study explores the potential of Cisplatin and Rutin-loaded nanoliposomes (Cis-NLs and Rut-NLs) for anti-colorectal cancer activity.

Methods: Cis-NLs and Rut-NLs were prepared using thin-film hydration, achieving encapsulation efficiencies of 95.5% and 62.5%, respectively. Drug release studies revealed controlled profiles, with Cis-NLs showing a complete release (100%) and Rut-NLs reaching 23.48% over 48 hours. Stability assessments demonstrated minimal changes in size, polydispersity index (PDI), and zeta potential over three months. Encapsulation efficiency decreased slightly for Cis-NLs (92.87%) and significantly for Rut-NLs (26.55%). Several tests were performed to evaluate the biological activity of this combination on colorectal cancer cells and HDF cells to check its selectivity.

Results: In vitro cytotoxicity studies on HT29 colorectal cancer cells revealed IC50 values of 1.72 μg/mL for free Cisplatin, 2.35 μg/mL for Cis-NLs, >100 μg/mL for free Rutin, and 63.33 μg/mL for Rut-NLs. A combination of Cis-NLs and Rut-NLs reduced the IC50 to 2.2 μg/mL. Selective toxicity evaluation using human dermal fibroblasts showed an IC50 of 79.24 μM for cisplatin, reduced to 63.3 μM in Cis-NLs, with Rut-NLs demonstrating negligible toxicity.

Discussion: Wound healing assays confirmed significant inhibition of cell migration, with wound closure reduced from 62.41% in controls to 34.35% in treated groups. Utilizing nanotechnology, liposomal formulations were synthesized to enhance drug delivery and therapeutic synergy.

Conclusion: These results highlight the potential of Cisplatin and Rutin-loaded nanoliposomes as a combination therapy for colorectal cancer.

导语:结直肠癌是癌症相关死亡的重要原因,需要创新的治疗方法来提高疗效和减少副作用。本研究探讨了顺铂和负载芦丁的纳米脂质体(Cis-NLs和Rut-NLs)抗结直肠癌活性的潜力。方法:采用薄膜水合法制备顺式nls和Rut-NLs,包封率分别为95.5%和62.5%。药物释放研究显示,顺式nls在48小时内完全释放(100%),而鲁特nls达到23.48%。稳定性评估表明,在三个月内,尺寸、多分散性指数(PDI)和zeta电位的变化很小。顺式nls的包封率略有下降(92.87%),而鲁特- nls的包封率显著下降(26.55%)。我们进行了几项试验来评估该组合对结直肠癌细胞和HDF细胞的生物活性,以检查其选择性。结果:体外对HT29结直肠癌细胞的细胞毒性研究显示,游离顺铂的IC50值为1.72 μg/mL,顺- nls的IC50值为2.35 μg/mL,游离芦丁的IC50值为100 μg/mL,游离芦丁的IC50值为63.33 μg/mL。Cis-NLs和Rut-NLs联合使用可使IC50降至2.2 μg/mL。使用人真皮成纤维细胞进行的选择性毒性评估显示,顺铂的IC50为79.24 μM,顺- nls的IC50降至63.3 μM,而Rut-NLs的毒性可以忽略。讨论:伤口愈合实验证实了细胞迁移的显著抑制,伤口愈合从对照组的62.41%降低到治疗组的34.35%。利用纳米技术,合成了脂质体制剂,以增强药物传递和治疗协同作用。结论:这些结果突出了顺铂和负载芦丁的纳米脂质体作为结直肠癌联合治疗的潜力。
{"title":"Investigating the Therapeutic Potential of Cisplatin- and Rutin-Loaded Nanoliposomes against Colorectal Cancer Cells.","authors":"Abdulazeez M Al-Mashhadani, Ali Al-Samydai, Ibrahim Al-Deeb, Simone Carradori, Hanan Azzam, Hussein S Janabi, Dana Al Qudah, Walhan Alshaer, M R Mozafari","doi":"10.2174/0118715206388843250709121645","DOIUrl":"https://doi.org/10.2174/0118715206388843250709121645","url":null,"abstract":"<p><strong>Introduction: </strong>Colorectal cancer is an important cause of cancer-related mortality, necessitating innovative therapies to improve efficacy and reduce side effects. This study explores the potential of Cisplatin and Rutin-loaded nanoliposomes (Cis-NLs and Rut-NLs) for anti-colorectal cancer activity.</p><p><strong>Methods: </strong>Cis-NLs and Rut-NLs were prepared using thin-film hydration, achieving encapsulation efficiencies of 95.5% and 62.5%, respectively. Drug release studies revealed controlled profiles, with Cis-NLs showing a complete release (100%) and Rut-NLs reaching 23.48% over 48 hours. Stability assessments demonstrated minimal changes in size, polydispersity index (PDI), and zeta potential over three months. Encapsulation efficiency decreased slightly for Cis-NLs (92.87%) and significantly for Rut-NLs (26.55%). Several tests were performed to evaluate the biological activity of this combination on colorectal cancer cells and HDF cells to check its selectivity.</p><p><strong>Results: </strong>In vitro cytotoxicity studies on HT29 colorectal cancer cells revealed IC50 values of 1.72 μg/mL for free Cisplatin, 2.35 μg/mL for Cis-NLs, >100 μg/mL for free Rutin, and 63.33 μg/mL for Rut-NLs. A combination of Cis-NLs and Rut-NLs reduced the IC50 to 2.2 μg/mL. Selective toxicity evaluation using human dermal fibroblasts showed an IC50 of 79.24 μM for cisplatin, reduced to 63.3 μM in Cis-NLs, with Rut-NLs demonstrating negligible toxicity.</p><p><strong>Discussion: </strong>Wound healing assays confirmed significant inhibition of cell migration, with wound closure reduced from 62.41% in controls to 34.35% in treated groups. Utilizing nanotechnology, liposomal formulations were synthesized to enhance drug delivery and therapeutic synergy.</p><p><strong>Conclusion: </strong>These results highlight the potential of Cisplatin and Rutin-loaded nanoliposomes as a combination therapy for colorectal cancer.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144658178","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
The Safety and Efficacy of Anti-LAG-3 for Patients with Melanoma: A Systematic Review and Meta-analysis Study. 抗lag -3治疗黑色素瘤患者的安全性和有效性:一项系统回顾和荟萃分析研究。
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-07-11 DOI: 10.2174/0118715206375121250701073343
Negar Nejati, Behrouz Robat-Jazi, Kianmehr Saleh, Mohsen Dashti, Ali Zand, Parsa Lorestani, Shaghayegh Karami, Majed Bahri Najafi, Parvaneh Rastgou, Fatemeh Rahimikia, Mohammad Amin Habibi, Maryam Barkhordar, Farhad Jadidi-Niaragh

Introduction: Melanoma, an aggressive skin cancer, has seen treatment advancements with immune checkpoint inhibitors (ICIs) like ipilimumab and nivolumab. Despite improved survival rates, resistance remains a challenge. The recent focus on lymphocyte activation gene-3 (LAG-3) inhibitors, such as relatlimab, shows promise in combination therapies, potentially improving outcomes with fewer adverse effects. This review evaluates the safety and efficacy of anti-LAG-3 antibodies in melanoma treatment.

Methods: This systematic review and meta-analysis, following the PRISMA guidelines and registered in PROSPERO (CRD42024565756), assessed anti-LAG-3 antibodies in melanoma treatment. A thorough search across PubMed, Embase, Scopus, and Web of Science up to January 2024 yielded relevant studies. Data on study characteristics, patient demographics, disease characteristics, treatment details, and clinical outcomes were extracted. Quality assessment was performed using the MINOR criteria. The meta-analysis, using STATA and random- effects models, addressed heterogeneity to determine safety and efficacy outcomes.

Results: We examined the clinical benefit of this combination therapeutic approach by measuring several primary endpoints and running a meta-analysis to determine the pooled estimate of 6-month progression-free survival (PFS), 1-year PFS, 6-month duration of response (DoR), 1-year DoR, 1-year overall survival (OS), 2-year OS, partial response (PR), complete response (CR), objective response rate (ORR), disease control rate (DCR), stable disease (SD), and progressive disease (PD) for patients diagnosed with melanoma. Our analysis showed 66% of any grade treatment-related adverse events (trAEs) (95% CI: 51%-81%), 19% of grade ≥ 3 trAEs (95% CI: 11%- 27%), 12% of any grade AEs leading to discontinuation (95% CI: 9%-14%), and 8% of grade ≥ 3 AEs leading to discontinuation (95% CI: 6%-10%). 76% of any grade overall AEs (95% CI: 34%-100%), and 33% of grade ≥ 3 overall AEs (95% CI: 15%-50%). The most common AEs were fatigue, pneumonitis, rash, pruritus, colitis, hepatitis, diarrhea, hypothyroidism, thyroiditis, and adrenal insufficiency.

Discussion: This systematic review and meta-analysis provide comprehensive evidence regarding the safety and efficacy of anti-LAG-3 antibodies in melanoma therapy. Pooled data reveals encouraging outcomes across several key endpoints, including PFS, OS, and ORR. While trAEs were common (66% for any grade and 19% for grade ≥3), most were manageable.

Conclusion: Anti-LAG-3 therapy is an active and safe treatment that shows promising results in melanoma treatment.

黑色素瘤是一种侵袭性皮肤癌,免疫检查点抑制剂(ICIs)如ipilimumab和nivolumab的治疗取得了进展。尽管存活率有所提高,但耐药性仍然是一个挑战。最近对淋巴细胞活化基因3 (LAG-3)抑制剂的关注,如relatlimab,显示出联合治疗的希望,可能改善结果,减少不良反应。本文综述了抗lag -3抗体在黑色素瘤治疗中的安全性和有效性。方法:本系统综述和荟萃分析,遵循PRISMA指南并在PROSPERO注册(CRD42024565756),评估抗lag -3抗体在黑色素瘤治疗中的作用。在PubMed, Embase, Scopus和Web of Science上进行全面搜索,直到2024年1月,得出了相关研究。提取有关研究特征、患者人口统计学、疾病特征、治疗细节和临床结果的数据。使用MINOR标准进行质量评估。荟萃分析使用STATA和随机效应模型,解决了异质性,以确定安全性和有效性结果。结果:我们通过测量几个主要终点并进行meta分析来检查这种联合治疗方法的临床获益,以确定6个月无进展生存期(PFS)、1年PFS、6个月反应持续时间(DoR)、1年DoR、1年总生存期(OS)、2年OS、部分缓解(PR)、完全缓解(CR)、客观缓解率(ORR)、疾病控制率(DCR)、疾病稳定(SD)、诊断为黑色素瘤的患者的进行性疾病(PD)。我们的分析显示,66%的任何级别的治疗相关不良事件(trAEs) (95% CI: 51%-81%), 19%的≥3级trAEs (95% CI: 11%- 27%), 12%的任何级别ae导致停药(95% CI: 9%-14%), 8%的≥3级ae导致停药(95% CI: 6%-10%)。任何级别总ae的76% (95% CI: 34%-100%),≥3级总ae的33% (95% CI: 15%-50%)。最常见的ae是疲劳、肺炎、皮疹、瘙痒、结肠炎、肝炎、腹泻、甲状腺功能减退、甲状腺炎和肾上腺功能不全。讨论:本系统综述和荟萃分析为抗lag -3抗体在黑色素瘤治疗中的安全性和有效性提供了全面的证据。汇集的数据显示了几个关键终点的令人鼓舞的结果,包括PFS、OS和ORR。虽然trae很常见(任何级别为66%,≥3级为19%),但大多数是可控的。结论:抗lag -3治疗是一种积极、安全的治疗方法,在黑色素瘤治疗中显示出良好的效果。
{"title":"The Safety and Efficacy of Anti-LAG-3 for Patients with Melanoma: A Systematic Review and Meta-analysis Study.","authors":"Negar Nejati, Behrouz Robat-Jazi, Kianmehr Saleh, Mohsen Dashti, Ali Zand, Parsa Lorestani, Shaghayegh Karami, Majed Bahri Najafi, Parvaneh Rastgou, Fatemeh Rahimikia, Mohammad Amin Habibi, Maryam Barkhordar, Farhad Jadidi-Niaragh","doi":"10.2174/0118715206375121250701073343","DOIUrl":"https://doi.org/10.2174/0118715206375121250701073343","url":null,"abstract":"<p><strong>Introduction: </strong>Melanoma, an aggressive skin cancer, has seen treatment advancements with immune checkpoint inhibitors (ICIs) like ipilimumab and nivolumab. Despite improved survival rates, resistance remains a challenge. The recent focus on lymphocyte activation gene-3 (LAG-3) inhibitors, such as relatlimab, shows promise in combination therapies, potentially improving outcomes with fewer adverse effects. This review evaluates the safety and efficacy of anti-LAG-3 antibodies in melanoma treatment.</p><p><strong>Methods: </strong>This systematic review and meta-analysis, following the PRISMA guidelines and registered in PROSPERO (CRD42024565756), assessed anti-LAG-3 antibodies in melanoma treatment. A thorough search across PubMed, Embase, Scopus, and Web of Science up to January 2024 yielded relevant studies. Data on study characteristics, patient demographics, disease characteristics, treatment details, and clinical outcomes were extracted. Quality assessment was performed using the MINOR criteria. The meta-analysis, using STATA and random- effects models, addressed heterogeneity to determine safety and efficacy outcomes.</p><p><strong>Results: </strong>We examined the clinical benefit of this combination therapeutic approach by measuring several primary endpoints and running a meta-analysis to determine the pooled estimate of 6-month progression-free survival (PFS), 1-year PFS, 6-month duration of response (DoR), 1-year DoR, 1-year overall survival (OS), 2-year OS, partial response (PR), complete response (CR), objective response rate (ORR), disease control rate (DCR), stable disease (SD), and progressive disease (PD) for patients diagnosed with melanoma. Our analysis showed 66% of any grade treatment-related adverse events (trAEs) (95% CI: 51%-81%), 19% of grade ≥ 3 trAEs (95% CI: 11%- 27%), 12% of any grade AEs leading to discontinuation (95% CI: 9%-14%), and 8% of grade ≥ 3 AEs leading to discontinuation (95% CI: 6%-10%). 76% of any grade overall AEs (95% CI: 34%-100%), and 33% of grade ≥ 3 overall AEs (95% CI: 15%-50%). The most common AEs were fatigue, pneumonitis, rash, pruritus, colitis, hepatitis, diarrhea, hypothyroidism, thyroiditis, and adrenal insufficiency.</p><p><strong>Discussion: </strong>This systematic review and meta-analysis provide comprehensive evidence regarding the safety and efficacy of anti-LAG-3 antibodies in melanoma therapy. Pooled data reveals encouraging outcomes across several key endpoints, including PFS, OS, and ORR. While trAEs were common (66% for any grade and 19% for grade ≥3), most were manageable.</p><p><strong>Conclusion: </strong>Anti-LAG-3 therapy is an active and safe treatment that shows promising results in melanoma treatment.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144635960","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
Lifileucel Therapy for Metastatic Melanoma: Advancements in Tumor-infiltrating Lymphocyte-based Immunotherapy. 转移性黑色素瘤的Lifileucel治疗:肿瘤浸润性淋巴细胞免疫治疗的进展。
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-07-04 DOI: 10.2174/0118715206380598250622182719
Issac V Cherian, Md Mustahidul Islam, Mamta Bishnoi, Sakshi Priya, Balak Das Kurmi, Sourabh Koshey, Preeti Patel

Metastatic melanoma is an aggressive malignancy with limited treatment options at advanced stages. Lifileucel, an FDA-approved autologous Tumor-Infiltrating Lymphocyte (TIL) therapy, marks a major advancement in immunotherapy, particularly for patients who fail conventional treatments like immune checkpoint inhibitors and targeted therapies. The mechanism of lifileucel involves the ex vivo expansion of patient-derived TILs to boost immune responses against melanoma cells. These expanded TILs are re-infused into patients, enhancing tumor-specific cytotoxicity and modulating the tumor microenvironment for sustained immune activation. Clinical trials have demonstrated its efficacy, with the overall response rate (ORR) reaching up to 36% in heavily pretreated populations, offering durable responses and improved progression-free survival compared to traditional therapies. The personalized approach of lifileucel, leveraging the patient's own T-cell repertoire, highlights its potential for precision oncology by targeting individual tumor profiles. Its integration with combination therapies, particularly immune checkpoint inhibitors, shows promising synergistic effects, broadening its clinical applicability. In addition to clinical success, the role of lifileucel in influencing the melanogenesis pathway offers insights into optimizing therapeutic strategies for melanoma. Ongoing research focuses on enhancing TIL functionality, overcoming challenges like tumor-induced immune suppression, and extending the applicability of lifileucel to other solid tumors. This breakthrough therapy not only addresses a critical unmet need in melanoma treatment but also represents a paradigm shift toward personalized medicine in oncology. Lifileucel underscores the potential of TILbased approaches to revolutionize cancer care, setting the stage for future advancements in immunotherapy.

转移性黑色素瘤是一种侵袭性恶性肿瘤,晚期治疗选择有限。Lifileucel是一种fda批准的自体肿瘤浸润淋巴细胞(TIL)治疗药物,标志着免疫治疗的重大进步,特别是对于免疫检查点抑制剂和靶向治疗等常规治疗失败的患者。lifileucel的机制涉及患者源性til的体外扩增,以增强对黑色素瘤细胞的免疫反应。这些扩展后的TILs被重新输注到患者体内,增强肿瘤特异性细胞毒性,调节肿瘤微环境,以实现持续的免疫激活。临床试验已经证明了它的有效性,在大量预处理的人群中,总缓解率(ORR)高达36%,与传统疗法相比,提供了持久的缓解和改善的无进展生存期。lifileucel的个性化方法,利用患者自身的t细胞库,突出了其针对个体肿瘤特征的精确肿瘤学潜力。它与联合疗法,特别是免疫检查点抑制剂的结合,显示出有希望的协同作用,扩大了其临床适用性。除了临床成功,lifileucel在影响黑色素形成途径中的作用为优化黑色素瘤的治疗策略提供了见解。正在进行的研究重点是增强TIL功能,克服肿瘤诱导的免疫抑制等挑战,并将lifileucel的适用性扩展到其他实体肿瘤。这一突破性疗法不仅解决了黑色素瘤治疗中一个关键的未满足的需求,而且代表了肿瘤学向个性化医疗的范式转变。Lifileucel强调了基于till的癌症治疗方法的潜力,为未来免疫治疗的进步奠定了基础。
{"title":"Lifileucel Therapy for Metastatic Melanoma: Advancements in Tumor-infiltrating Lymphocyte-based Immunotherapy.","authors":"Issac V Cherian, Md Mustahidul Islam, Mamta Bishnoi, Sakshi Priya, Balak Das Kurmi, Sourabh Koshey, Preeti Patel","doi":"10.2174/0118715206380598250622182719","DOIUrl":"https://doi.org/10.2174/0118715206380598250622182719","url":null,"abstract":"<p><p>Metastatic melanoma is an aggressive malignancy with limited treatment options at advanced stages. Lifileucel, an FDA-approved autologous Tumor-Infiltrating Lymphocyte (TIL) therapy, marks a major advancement in immunotherapy, particularly for patients who fail conventional treatments like immune checkpoint inhibitors and targeted therapies. The mechanism of lifileucel involves the ex vivo expansion of patient-derived TILs to boost immune responses against melanoma cells. These expanded TILs are re-infused into patients, enhancing tumor-specific cytotoxicity and modulating the tumor microenvironment for sustained immune activation. Clinical trials have demonstrated its efficacy, with the overall response rate (ORR) reaching up to 36% in heavily pretreated populations, offering durable responses and improved progression-free survival compared to traditional therapies. The personalized approach of lifileucel, leveraging the patient's own T-cell repertoire, highlights its potential for precision oncology by targeting individual tumor profiles. Its integration with combination therapies, particularly immune checkpoint inhibitors, shows promising synergistic effects, broadening its clinical applicability. In addition to clinical success, the role of lifileucel in influencing the melanogenesis pathway offers insights into optimizing therapeutic strategies for melanoma. Ongoing research focuses on enhancing TIL functionality, overcoming challenges like tumor-induced immune suppression, and extending the applicability of lifileucel to other solid tumors. This breakthrough therapy not only addresses a critical unmet need in melanoma treatment but also represents a paradigm shift toward personalized medicine in oncology. Lifileucel underscores the potential of TILbased approaches to revolutionize cancer care, setting the stage for future advancements in immunotherapy.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144574687","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
Anticancer Compounds from Myxobacteria: Current Scenario and Future Perspectives. 黏菌中的抗癌化合物:现状和未来展望。
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-07-04 DOI: 10.2174/0118715206384510250625004749
Swati Sihag, Shweta Sinha, Ramandeep Kaur

Natural products and their derivatives have played a dominant role in the development of therapeutic agents. Traditionally, most of the natural products developed for the effective treatment of different diseases have been sourced from plants. Natural product discovery has seen a shift of focus towards microorganisms due to the chemical diversity of bioactive products they synthesize. Myxobacteria produce a large variety of novel chemical entities with diverse structures and varied bioactivities. In the last few decades, secondary metabolites from different genera of myxobacteria have been recognized as harbouring potent anticancer activity. Several analogs of these anticancer compounds have been prepared to address the limitations such as, poor solubility, high toxicity and low production yield, in order to obtain the compounds in higher quantities with better pharmacological properties and target selectivity. For example, a semi-synthetic derivative of epothilone obtained from a strain of myxobacterium has been approved for clinical use against taxane-resistant breast cancer. The anticancer compounds from myxobacteria target microtubules, the cytoskeleton, vacuolar ATPase, methionine aminopeptidase, exportin, the proteasome or translation elongation factor to exert anticancer activity. The focus of this review is on the promising anticancer compounds produced by myxobacteria, their targets and their mechanisms of action in cancer cells.

天然产物及其衍生物在治疗剂的发展中起着主导作用。传统上,为有效治疗不同疾病而开发的大多数天然产品都来自植物。由于微生物合成的生物活性产物的化学多样性,天然产物的发现已将重点转向微生物。黏菌产生大量具有不同结构和不同生物活性的新型化学实体。在过去的几十年里,来自不同属的黏菌次生代谢物被认为具有强大的抗癌活性。针对这些抗癌化合物溶解度差、毒性大、产率低等缺点,制备了几种类似物,以期获得量大、药理学性质好、靶向选择性好的化合物。例如,从一种黏菌菌株中获得的一种半合成衍生物埃泊霉素已被批准用于临床治疗紫杉烷耐药乳腺癌。来自黏菌的抗癌化合物以微管、细胞骨架、空泡atp酶、蛋氨酸氨基肽酶、输出蛋白、蛋白酶体或翻译延伸因子为靶点发挥抗癌活性。本文就黏菌产生的抗癌化合物及其在癌细胞中的作用机制和作用靶点作一综述。
{"title":"Anticancer Compounds from Myxobacteria: Current Scenario and Future Perspectives.","authors":"Swati Sihag, Shweta Sinha, Ramandeep Kaur","doi":"10.2174/0118715206384510250625004749","DOIUrl":"https://doi.org/10.2174/0118715206384510250625004749","url":null,"abstract":"<p><p>Natural products and their derivatives have played a dominant role in the development of therapeutic agents. Traditionally, most of the natural products developed for the effective treatment of different diseases have been sourced from plants. Natural product discovery has seen a shift of focus towards microorganisms due to the chemical diversity of bioactive products they synthesize. Myxobacteria produce a large variety of novel chemical entities with diverse structures and varied bioactivities. In the last few decades, secondary metabolites from different genera of myxobacteria have been recognized as harbouring potent anticancer activity. Several analogs of these anticancer compounds have been prepared to address the limitations such as, poor solubility, high toxicity and low production yield, in order to obtain the compounds in higher quantities with better pharmacological properties and target selectivity. For example, a semi-synthetic derivative of epothilone obtained from a strain of myxobacterium has been approved for clinical use against taxane-resistant breast cancer. The anticancer compounds from myxobacteria target microtubules, the cytoskeleton, vacuolar ATPase, methionine aminopeptidase, exportin, the proteasome or translation elongation factor to exert anticancer activity. The focus of this review is on the promising anticancer compounds produced by myxobacteria, their targets and their mechanisms of action in cancer cells.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144574686","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
期刊
Anti-cancer agents in medicinal chemistry
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:604180095
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