Pub Date : 2024-01-01Epub Date: 2024-07-20DOI: 10.1016/bs.acr.2024.07.001
Pedro P Gonçalves, Cláudia L da Silva, Nuno Bernardes
Cancer remains as one of the highest challenges to human health. However, anticancer drugs exhibit one of the highest attrition rates compared to other therapeutic interventions. In part, this can be attributed to a prevalent use of in vitro models with limited recapitulative potential of the in vivo settings. Three dimensional (3D) models, such as tumor spheroids and organoids, offer many research opportunities to address the urgent need in developing models capable to more accurately mimic cancer biology and drug resistance profiles. However, their wide adoption in high-throughput pre-clinical studies is dependent on scalable manufacturing to support large-scale therapeutic drug screenings and multi-omic approaches for their comprehensive cellular and molecular characterization. Extracellular vesicles (EVs), which have been emerging as promising drug delivery systems (DDS), stand to significantly benefit from such screenings conducted in realistic cancer models. Furthermore, the integration of these nanomedicines with 3D cancer models and omics profiling holds the potential to deepen our understanding of EV-mediated anticancer effects. In this chapter, we provide an overview of the existing 3D models used in cancer research, namely spheroids and organoids, the innovations in their scalable production and discuss how omics can facilitate the implementation of these models at different stages of drug testing. We also explore how EVs can advance drug delivery in cancer therapies and how the synergy between 3D cancer models and omics approaches can benefit in this process.
癌症仍然是人类健康面临的最大挑战之一。然而,与其他治疗干预措施相比,抗癌药物的损耗率最高。部分原因在于体外模型的普遍使用,对体内环境的再现潜力有限。三维(3D)模型,如肿瘤球体和有机体,提供了许多研究机会,以满足开发能够更准确地模拟癌症生物学和耐药性特征的模型的迫切需要。然而,它们在高通量临床前研究中的广泛应用取决于可扩展的制造工艺,以支持大规模治疗药物筛选和多组学方法对其进行全面的细胞和分子表征。细胞外囊泡 (EV) 作为一种新兴的药物递送系统 (DDS),将从在真实癌症模型中进行的此类筛选中受益匪浅。此外,将这些纳米药物与三维癌症模型和全息图谱分析相结合,有可能加深我们对 EV 介导的抗癌作用的理解。在本章中,我们将概述癌症研究中使用的现有三维模型(即球形体和有机体)、其可扩展生产中的创新,并讨论全局组学如何促进这些模型在药物测试不同阶段的实施。我们还探讨了EV如何推动癌症疗法中的药物输送,以及三维癌症模型和omics方法之间的协同作用如何在这一过程中获益。
{"title":"Advancing cancer therapeutics: Integrating scalable 3D cancer models, extracellular vesicles, and omics for enhanced therapy efficacy.","authors":"Pedro P Gonçalves, Cláudia L da Silva, Nuno Bernardes","doi":"10.1016/bs.acr.2024.07.001","DOIUrl":"https://doi.org/10.1016/bs.acr.2024.07.001","url":null,"abstract":"<p><p>Cancer remains as one of the highest challenges to human health. However, anticancer drugs exhibit one of the highest attrition rates compared to other therapeutic interventions. In part, this can be attributed to a prevalent use of in vitro models with limited recapitulative potential of the in vivo settings. Three dimensional (3D) models, such as tumor spheroids and organoids, offer many research opportunities to address the urgent need in developing models capable to more accurately mimic cancer biology and drug resistance profiles. However, their wide adoption in high-throughput pre-clinical studies is dependent on scalable manufacturing to support large-scale therapeutic drug screenings and multi-omic approaches for their comprehensive cellular and molecular characterization. Extracellular vesicles (EVs), which have been emerging as promising drug delivery systems (DDS), stand to significantly benefit from such screenings conducted in realistic cancer models. Furthermore, the integration of these nanomedicines with 3D cancer models and omics profiling holds the potential to deepen our understanding of EV-mediated anticancer effects. In this chapter, we provide an overview of the existing 3D models used in cancer research, namely spheroids and organoids, the innovations in their scalable production and discuss how omics can facilitate the implementation of these models at different stages of drug testing. We also explore how EVs can advance drug delivery in cancer therapies and how the synergy between 3D cancer models and omics approaches can benefit in this process.</p>","PeriodicalId":94294,"journal":{"name":"Advances in cancer research","volume":"163 ","pages":"137-185"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142305454","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-01-01Epub Date: 2024-04-25DOI: 10.1016/bs.acr.2024.04.003
Yuqian Gao, Hyeyoon Kim, Reta Birhanu Kitata, Tai-Tu Lin, Adam C Swensen, Tujin Shi, Tao Liu
Prostate cancer (PCa) is the most common non-skin cancer among men in the United States. However, the widely used protein biomarker in PCa, prostate-specific antigen (PSA), while useful for initial detection, its use alone cannot detect aggressive PCa and can lead to overtreatment. This chapter provides an overview of PCa protein biomarker development. It reviews the state-of-the-art liquid chromatography-mass spectrometry-based proteomics technologies for PCa biomarker development, such as enhancing the detection sensitivity of low-abundance proteins through antibody-based or antibody-independent protein/peptide enrichment, enriching post-translational modifications such as glycosylation as well as information-rich extracellular vesicles, and increasing accuracy and throughput using advanced data acquisition methodologies. This chapter also summarizes recent PCa biomarker validation studies that applied those techniques in diverse specimen types, including cell lines, tissues, proximal fluids, urine, and blood, developing novel protein biomarkers for various clinical applications, including early detection and diagnosis, prognosis, and therapeutic intervention of PCa.
{"title":"Multiplexed quantitative proteomics in prostate cancer biomarker development.","authors":"Yuqian Gao, Hyeyoon Kim, Reta Birhanu Kitata, Tai-Tu Lin, Adam C Swensen, Tujin Shi, Tao Liu","doi":"10.1016/bs.acr.2024.04.003","DOIUrl":"10.1016/bs.acr.2024.04.003","url":null,"abstract":"<p><p>Prostate cancer (PCa) is the most common non-skin cancer among men in the United States. However, the widely used protein biomarker in PCa, prostate-specific antigen (PSA), while useful for initial detection, its use alone cannot detect aggressive PCa and can lead to overtreatment. This chapter provides an overview of PCa protein biomarker development. It reviews the state-of-the-art liquid chromatography-mass spectrometry-based proteomics technologies for PCa biomarker development, such as enhancing the detection sensitivity of low-abundance proteins through antibody-based or antibody-independent protein/peptide enrichment, enriching post-translational modifications such as glycosylation as well as information-rich extracellular vesicles, and increasing accuracy and throughput using advanced data acquisition methodologies. This chapter also summarizes recent PCa biomarker validation studies that applied those techniques in diverse specimen types, including cell lines, tissues, proximal fluids, urine, and blood, developing novel protein biomarkers for various clinical applications, including early detection and diagnosis, prognosis, and therapeutic intervention of PCa.</p>","PeriodicalId":94294,"journal":{"name":"Advances in cancer research","volume":"161 ","pages":"31-69"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141736295","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-01-01Epub Date: 2024-05-22DOI: 10.1016/bs.acr.2024.05.001
Gunjan Purohit, Polash Ghosh, Oleh Khalimonchuk
Our understanding of the roles that mitochondria play in cellular physiology has evolved drastically-from a mere cellular energy supplier to a crucial regulator of metabolic and signaling processes, particularly in the context of development and progression of human diseases such as cancers. The present review examines the role of OMA1, a conserved, redox-sensitive metallopeptidase in cancer biology. OMA1's involvement in mitochondrial quality control, redox activity, and stress responses underscores its potential as a novel target in cancer diagnosis and treatment. However, our incomplete understanding of OMA1's regulation and structural detail presents ongoing challenges to target OMA1 for therapeutic purposes. Further exploration of OMA1 holds promise in uncovering novel insights into cancer mechanisms and therapeutic strategies. In this chapter, we briefly summarize our current knowledge about OMA1, its redox-regulation, and emerging role in certain cancers.
{"title":"Mitochondrial metallopeptidase OMA1 in cancer.","authors":"Gunjan Purohit, Polash Ghosh, Oleh Khalimonchuk","doi":"10.1016/bs.acr.2024.05.001","DOIUrl":"10.1016/bs.acr.2024.05.001","url":null,"abstract":"<p><p>Our understanding of the roles that mitochondria play in cellular physiology has evolved drastically-from a mere cellular energy supplier to a crucial regulator of metabolic and signaling processes, particularly in the context of development and progression of human diseases such as cancers. The present review examines the role of OMA1, a conserved, redox-sensitive metallopeptidase in cancer biology. OMA1's involvement in mitochondrial quality control, redox activity, and stress responses underscores its potential as a novel target in cancer diagnosis and treatment. However, our incomplete understanding of OMA1's regulation and structural detail presents ongoing challenges to target OMA1 for therapeutic purposes. Further exploration of OMA1 holds promise in uncovering novel insights into cancer mechanisms and therapeutic strategies. In this chapter, we briefly summarize our current knowledge about OMA1, its redox-regulation, and emerging role in certain cancers.</p>","PeriodicalId":94294,"journal":{"name":"Advances in cancer research","volume":"162 ","pages":"75-97"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141790737","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-01-01Epub Date: 2024-05-03DOI: 10.1016/bs.acr.2024.04.001
Elias S J Arnér, Edward E Schmidt
Cysteine is required for synthesis of glutathione (GSH), coenzyme A, other sulfur-containing metabolites, and most proteins. In most cells, cysteine comes from extracellular disulfide sources including cystine, glutathione-disulfide, and peptides. The thioredoxin reductase-1 (TrxR1)- or glutathione-disulfide reductase (GSR)-driven enzymatic systems can fuel cystine reduction via thioredoxins, glutaredoxins, or other thioredoxin-fold proteins. Free cystine enters cells thorough the cystine-glutamate antiporter, xCT, but systemically, plasma glutathione-disulfide might predominate as a cystine source. Erastin, inhibiting both xCT and voltage-dependent anion channels, induces ferroptotic cell death, so named because this type of cell death is antagonized by iron-chelators. Many cancer cells seem to be predisposed to ferroptosis, which has been proposed as a targetable cancer liability. Ferroptosis is associated with lipid peroxidation and loss of either glutathione peroxidase-4 (GPX4) or ferroptosis suppressor protein-1 (FSP1), which each prevent accumulation of lipid peroxides. It has been suggested that an xCT inhibition-induced cellular cysteine-deficiency lowers GSH levels, starving GPX4 for reducing power and allowing membrane lipid peroxides to accumulate, thereby causing ferroptosis. Aspects of ferroptosis are however not fully understood and need to be further scrutinized, for example that neither disruption of GSH synthesis, loss of GSH, nor disruption of glutathione disulfide reductase (GSR), triggers ferroptosis in animal models. Here we reevaluate the relationships between Erastin, xCT, GPX4, cellular cysteine and GSH, RSL3 or ML162, and ferroptosis. We conclude that, whereas both Cys and ferroptosis are potential liabilities in cancer, their relationship to each other remains insufficiently understood.
{"title":"Unresolved questions regarding cellular cysteine sources and their possible relationships to ferroptosis.","authors":"Elias S J Arnér, Edward E Schmidt","doi":"10.1016/bs.acr.2024.04.001","DOIUrl":"10.1016/bs.acr.2024.04.001","url":null,"abstract":"<p><p>Cysteine is required for synthesis of glutathione (GSH), coenzyme A, other sulfur-containing metabolites, and most proteins. In most cells, cysteine comes from extracellular disulfide sources including cystine, glutathione-disulfide, and peptides. The thioredoxin reductase-1 (TrxR1)- or glutathione-disulfide reductase (GSR)-driven enzymatic systems can fuel cystine reduction via thioredoxins, glutaredoxins, or other thioredoxin-fold proteins. Free cystine enters cells thorough the cystine-glutamate antiporter, xCT, but systemically, plasma glutathione-disulfide might predominate as a cystine source. Erastin, inhibiting both xCT and voltage-dependent anion channels, induces ferroptotic cell death, so named because this type of cell death is antagonized by iron-chelators. Many cancer cells seem to be predisposed to ferroptosis, which has been proposed as a targetable cancer liability. Ferroptosis is associated with lipid peroxidation and loss of either glutathione peroxidase-4 (GPX4) or ferroptosis suppressor protein-1 (FSP1), which each prevent accumulation of lipid peroxides. It has been suggested that an xCT inhibition-induced cellular cysteine-deficiency lowers GSH levels, starving GPX4 for reducing power and allowing membrane lipid peroxides to accumulate, thereby causing ferroptosis. Aspects of ferroptosis are however not fully understood and need to be further scrutinized, for example that neither disruption of GSH synthesis, loss of GSH, nor disruption of glutathione disulfide reductase (GSR), triggers ferroptosis in animal models. Here we reevaluate the relationships between Erastin, xCT, GPX4, cellular cysteine and GSH, RSL3 or ML162, and ferroptosis. We conclude that, whereas both Cys and ferroptosis are potential liabilities in cancer, their relationship to each other remains insufficiently understood.</p>","PeriodicalId":94294,"journal":{"name":"Advances in cancer research","volume":"162 ","pages":"1-44"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141790741","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-01-01Epub Date: 2024-06-22DOI: 10.1016/bs.acr.2024.06.001
Sheeraz Un Nazir, Juhi Mishra, Shailendra Kumar Maurya, Negin Ziamiavaghi, Sanika Bodas, Benjamin A Teply, Samikshan Dutta, Kaustubh Datta
Prostate cancer, one of the most frequently diagnosed cancers in men, leads to significant mortality worldwide. Its study is important due to the complexity and diversity in its progression, highlighting the urgent need for improved therapeutic strategies. This chapter probes into the genetic and epigenetic factors influencing prostate cancer progression, underscoring the importance of understanding the disease's molecular fundamentals for the development of targeted therapies. It specifically reviews the role of key genetic mutations in genes such as Androgen Receptor, TP53, SPOP, FOXA1 and PTEN which are crucial for the disease onset and a progression. Furthermore, it examines the impact of epigenetic modifications, including DNA methylation and histone modification, which contribute to the cancer's progression by affecting gene expression and cellular behavior. Further, in this chapter we delve into the underlying signaling mechanism, the advancements in targeting genetic and epigenetic alterations in prostate cancer. These findings have revealed promising targets for therapeutic advancements, aiming to understand and identify promising avenues for future therapies. This chapter improves our current understanding of prostate cancer genetic and epigenetic landscape, emphasizing the necessity of advancing our knowledge to refine and expand treatment options for prostate cancer patients.
前列腺癌是最常见的男性癌症之一,在全球范围内导致大量死亡。由于前列腺癌进展的复杂性和多样性,对前列腺癌的研究显得尤为重要,这也凸显了改进治疗策略的迫切需要。本章探讨了影响前列腺癌进展的遗传和表观遗传因素,强调了了解该疾病的分子基础对开发靶向疗法的重要性。它特别回顾了雄激素受体、TP53、SPOP、FOXA1 和 PTEN 等基因中关键遗传突变的作用,这些基因对疾病的发生和发展至关重要。此外,本章还探讨了表观遗传修饰(包括 DNA 甲基化和组蛋白修饰)的影响,这些修饰通过影响基因表达和细胞行为促进癌症进展。此外,本章还深入探讨了前列腺癌的基本信号转导机制、针对基因和表观遗传学改变的研究进展。这些发现揭示了有前景的治疗目标,旨在了解和确定未来治疗的可行途径。本章加深了我们目前对前列腺癌基因和表观遗传学状况的了解,强调了我们有必要增进知识,以完善和扩大前列腺癌患者的治疗方案。
{"title":"Deciphering the genetic and epigenetic architecture of prostate cancer.","authors":"Sheeraz Un Nazir, Juhi Mishra, Shailendra Kumar Maurya, Negin Ziamiavaghi, Sanika Bodas, Benjamin A Teply, Samikshan Dutta, Kaustubh Datta","doi":"10.1016/bs.acr.2024.06.001","DOIUrl":"https://doi.org/10.1016/bs.acr.2024.06.001","url":null,"abstract":"<p><p>Prostate cancer, one of the most frequently diagnosed cancers in men, leads to significant mortality worldwide. Its study is important due to the complexity and diversity in its progression, highlighting the urgent need for improved therapeutic strategies. This chapter probes into the genetic and epigenetic factors influencing prostate cancer progression, underscoring the importance of understanding the disease's molecular fundamentals for the development of targeted therapies. It specifically reviews the role of key genetic mutations in genes such as Androgen Receptor, TP53, SPOP, FOXA1 and PTEN which are crucial for the disease onset and a progression. Furthermore, it examines the impact of epigenetic modifications, including DNA methylation and histone modification, which contribute to the cancer's progression by affecting gene expression and cellular behavior. Further, in this chapter we delve into the underlying signaling mechanism, the advancements in targeting genetic and epigenetic alterations in prostate cancer. These findings have revealed promising targets for therapeutic advancements, aiming to understand and identify promising avenues for future therapies. This chapter improves our current understanding of prostate cancer genetic and epigenetic landscape, emphasizing the necessity of advancing our knowledge to refine and expand treatment options for prostate cancer patients.</p>","PeriodicalId":94294,"journal":{"name":"Advances in cancer research","volume":"161 ","pages":"191-221"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141736277","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Worldwide, prostate cancer (PCa) remains a leading cause of death in men. Histologically, the majority of PCa cases are classified as adenocarcinomas, which are mainly composed of androgen receptor-positive luminal cells. PCa is initially driven by the androgen receptor axis, where androgen-mediated activation of the receptor is one of the primary culprits for disease progression. Therefore, in advanced stage PCa, patients are generally treated with androgen deprivation therapies alone or in combination with androgen receptor pathway inhibitors. However, after an initial decrease, the cancer recurs for majority patients. At this stage, cancer is known as castration-resistant prostate cancer (CRPC). Majority of CRPC tumors still depend on androgen receptor axis for its progression to metastasis. However, in around 20-30% of cases, CRPC progresses via an androgen receptor-independent pathway and is often presented as neuroendocrine cancer (NE). This NE phenotype is highly aggressive with poor overall survival as compared to CRPC adenocarcinoma. NE cancers are resistant to standard taxane chemotherapies, which are often used to treat metastatic disease. Pathologically and morphologically, NE cancers are highly diverse and often co-exist with adenocarcinoma. Due to the lack of proper biomarkers, it is often difficult to make an early diagnosis of this lethal disease. Moreover, increased tumor heterogeneity and admixtures of adeno and NE subtypes in the same tumor make early detection of NE tumors very difficult. With the advancement of our knowledge and sequencing technology, we are now able to better understand the molecular mediators of this transformation pathway. This current study will give an update on how various molecular regulators are involved in these lineage transformation processes and what challenges we are still facing to detect and treat this cancer.
在全球范围内,前列腺癌(PCa)仍然是导致男性死亡的主要原因。从组织学角度看,大多数 PCa 病例属于腺癌,主要由雄激素受体阳性的管腔细胞组成。PCa 最初由雄激素受体轴驱动,雄激素介导的受体激活是疾病进展的罪魁祸首之一。因此,对于晚期 PCa 患者,一般采用雄激素剥夺疗法单独治疗或与雄激素受体通路抑制剂联合治疗。然而,大多数患者的病情在初步缓解后又会复发。在这一阶段,癌症被称为阉割抵抗性前列腺癌(CRPC)。大多数 CRPC 肿瘤仍依赖雄激素受体轴进行转移。不过,约有 20%-30% 的 CRPC 会通过雄激素受体依赖性途径发展,通常表现为神经内分泌癌(NE)。与 CRPC 腺癌相比,这种 NE 表型具有高度侵袭性,总生存率较低。NE 癌症对标准的类固醇化疗具有抗药性,而类固醇化疗通常用于治疗转移性疾病。从病理和形态上看,NE 癌具有高度多样性,并经常与腺癌并存。由于缺乏适当的生物标志物,通常很难对这种致命疾病做出早期诊断。此外,肿瘤异质性的增加以及同一肿瘤中腺癌和 NE 亚型的混合,也使 NE 肿瘤的早期检测变得非常困难。随着知识和测序技术的进步,我们现在能够更好地了解这一转化途径的分子介质。本研究将介绍各种分子调控因子是如何参与这些细胞系转化过程的,以及我们在检测和治疗这种癌症方面仍面临哪些挑战。
{"title":"Understanding the molecular regulators of neuroendocrine prostate cancer.","authors":"Sreyashi Bhattacharya, Avery Stillahn, Kaitlin Smith, Michael Muders, Kaustubh Datta, Samikshan Dutta","doi":"10.1016/bs.acr.2024.04.006","DOIUrl":"https://doi.org/10.1016/bs.acr.2024.04.006","url":null,"abstract":"<p><p>Worldwide, prostate cancer (PCa) remains a leading cause of death in men. Histologically, the majority of PCa cases are classified as adenocarcinomas, which are mainly composed of androgen receptor-positive luminal cells. PCa is initially driven by the androgen receptor axis, where androgen-mediated activation of the receptor is one of the primary culprits for disease progression. Therefore, in advanced stage PCa, patients are generally treated with androgen deprivation therapies alone or in combination with androgen receptor pathway inhibitors. However, after an initial decrease, the cancer recurs for majority patients. At this stage, cancer is known as castration-resistant prostate cancer (CRPC). Majority of CRPC tumors still depend on androgen receptor axis for its progression to metastasis. However, in around 20-30% of cases, CRPC progresses via an androgen receptor-independent pathway and is often presented as neuroendocrine cancer (NE). This NE phenotype is highly aggressive with poor overall survival as compared to CRPC adenocarcinoma. NE cancers are resistant to standard taxane chemotherapies, which are often used to treat metastatic disease. Pathologically and morphologically, NE cancers are highly diverse and often co-exist with adenocarcinoma. Due to the lack of proper biomarkers, it is often difficult to make an early diagnosis of this lethal disease. Moreover, increased tumor heterogeneity and admixtures of adeno and NE subtypes in the same tumor make early detection of NE tumors very difficult. With the advancement of our knowledge and sequencing technology, we are now able to better understand the molecular mediators of this transformation pathway. This current study will give an update on how various molecular regulators are involved in these lineage transformation processes and what challenges we are still facing to detect and treat this cancer.</p>","PeriodicalId":94294,"journal":{"name":"Advances in cancer research","volume":"161 ","pages":"403-429"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141736301","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-01-01Epub Date: 2024-07-09DOI: 10.1016/bs.acr.2024.06.007
Rishabh Maurya, Isha Chug, Vignesh Vudatha, António M Palma
Cancer is a complex disease intrinsically associated with cellular processes and gene expression. With the development of techniques such as single-cell sequencing and sequential fluorescence in situ hybridization (seqFISH), it was possible to map the location of cells based on their gene expression with more precision. Moreover, in recent years, many tools have been developed to analyze these extensive datasets by integrating machine learning and artificial intelligence in a comprehensive manner. Since these tools analyze sequencing data, they offer the chance to analyze any tissue regardless of its origin. By applying this to cancer settings, spatial transcriptomic analysis based on artificial intelligence may help us understand cell-cell communications within the tumor microenvironment. Another advantage of this analysis is the identification of new biomarkers and therapeutic targets. The integration of such analysis with other omics data and with routine exams such as magnetic resonance imaging can help physicians with the earlier diagnosis of tumors as well as establish a more personalized treatment for pancreatic cancer patients. In this review, we give an overview description of pancreatic cancer, describe how spatial transcriptomics and artificial intelligence have been used to study pancreatic cancer and provide examples of how integrating these tools may help physicians manage pancreatic cancer in a more personalized approach.
{"title":"Applications of spatial transcriptomics and artificial intelligence to develop integrated management of pancreatic cancer.","authors":"Rishabh Maurya, Isha Chug, Vignesh Vudatha, António M Palma","doi":"10.1016/bs.acr.2024.06.007","DOIUrl":"https://doi.org/10.1016/bs.acr.2024.06.007","url":null,"abstract":"<p><p>Cancer is a complex disease intrinsically associated with cellular processes and gene expression. With the development of techniques such as single-cell sequencing and sequential fluorescence in situ hybridization (seqFISH), it was possible to map the location of cells based on their gene expression with more precision. Moreover, in recent years, many tools have been developed to analyze these extensive datasets by integrating machine learning and artificial intelligence in a comprehensive manner. Since these tools analyze sequencing data, they offer the chance to analyze any tissue regardless of its origin. By applying this to cancer settings, spatial transcriptomic analysis based on artificial intelligence may help us understand cell-cell communications within the tumor microenvironment. Another advantage of this analysis is the identification of new biomarkers and therapeutic targets. The integration of such analysis with other omics data and with routine exams such as magnetic resonance imaging can help physicians with the earlier diagnosis of tumors as well as establish a more personalized treatment for pancreatic cancer patients. In this review, we give an overview description of pancreatic cancer, describe how spatial transcriptomics and artificial intelligence have been used to study pancreatic cancer and provide examples of how integrating these tools may help physicians manage pancreatic cancer in a more personalized approach.</p>","PeriodicalId":94294,"journal":{"name":"Advances in cancer research","volume":"163 ","pages":"107-136"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142305455","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-01-01Epub Date: 2024-07-02DOI: 10.1016/bs.acr.2024.06.003
Padmanabhan Mannangatti, Praveen Bhoopathi, Amit Kumar, Swadesh K Das, Luni Emdad, Paul B Fisher
Focused ultrasound (FUS) combined with microbubble (MB) treatment is a promising strategy capable of accurately delivering molecular medicines and gene therapy to treat various disease states. The rapid progression and use of FUS technology, from its inception to applications in contemporary medicine, exemplifies the significance and expanding potential of this technology. FUS for drug delivery in the brain can overcome challenging obstacles posed by the blood-brain barrier (BBB) in treating central nervous system (CNS) disorders. Both FUS and magnetic resonance imaging-guided FUS are non-invasive techniques for effectively opening the BBB and enhancing the transportation of molecular medicines and imaging agents into the brain. By integrating MBs into this process, it is possible to disrupt the BBB, facilitating delivery of therapeutic compounds including neuropeptides, proteins, antibodies, chemotherapeutic drugs and recently viruses accurately into the CNS. The safety and versatility of ultrasound makes it an attractive approach for administering molecular medicines, with potential applications extending beyond neurological disorders to include cancer treatment and other medical fields. Preclinical and clinical studies confirm that FUS is safe and efficient in enhancing drug administration, particularly where delivery to a precise location in the CNS is required. Combination therapies that utilize FUS and MBs also provide synergistic responses in cancer therapy. Further refining FUS and MB approaches both from a mechanical and reagent perspective will be forthcoming in the future and prove valuable in precisely defining targets and broadening therapeutic applications. Continued development and applications of FUS and MB technologies will improve therapeutic outcomes and advance patient care in multiple diseases states. This will elevate FUS and MBs from infrequently used medical options to mainstream medical applications.
{"title":"Precision medicine focus on the central nervous system: Non-invasive therapeutic agent delivery with focused ultrasound and microbubbles.","authors":"Padmanabhan Mannangatti, Praveen Bhoopathi, Amit Kumar, Swadesh K Das, Luni Emdad, Paul B Fisher","doi":"10.1016/bs.acr.2024.06.003","DOIUrl":"https://doi.org/10.1016/bs.acr.2024.06.003","url":null,"abstract":"<p><p>Focused ultrasound (FUS) combined with microbubble (MB) treatment is a promising strategy capable of accurately delivering molecular medicines and gene therapy to treat various disease states. The rapid progression and use of FUS technology, from its inception to applications in contemporary medicine, exemplifies the significance and expanding potential of this technology. FUS for drug delivery in the brain can overcome challenging obstacles posed by the blood-brain barrier (BBB) in treating central nervous system (CNS) disorders. Both FUS and magnetic resonance imaging-guided FUS are non-invasive techniques for effectively opening the BBB and enhancing the transportation of molecular medicines and imaging agents into the brain. By integrating MBs into this process, it is possible to disrupt the BBB, facilitating delivery of therapeutic compounds including neuropeptides, proteins, antibodies, chemotherapeutic drugs and recently viruses accurately into the CNS. The safety and versatility of ultrasound makes it an attractive approach for administering molecular medicines, with potential applications extending beyond neurological disorders to include cancer treatment and other medical fields. Preclinical and clinical studies confirm that FUS is safe and efficient in enhancing drug administration, particularly where delivery to a precise location in the CNS is required. Combination therapies that utilize FUS and MBs also provide synergistic responses in cancer therapy. Further refining FUS and MB approaches both from a mechanical and reagent perspective will be forthcoming in the future and prove valuable in precisely defining targets and broadening therapeutic applications. Continued development and applications of FUS and MB technologies will improve therapeutic outcomes and advance patient care in multiple diseases states. This will elevate FUS and MBs from infrequently used medical options to mainstream medical applications.</p>","PeriodicalId":94294,"journal":{"name":"Advances in cancer research","volume":"164 ","pages":"191-240"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142305480","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-01-01DOI: 10.1016/S0065-230X(24)00038-1
O John Semmes, Julius O Nyalwidhe, Swadesh K Das, Paul B Fisher
{"title":"Preface.","authors":"O John Semmes, Julius O Nyalwidhe, Swadesh K Das, Paul B Fisher","doi":"10.1016/S0065-230X(24)00038-1","DOIUrl":"https://doi.org/10.1016/S0065-230X(24)00038-1","url":null,"abstract":"","PeriodicalId":94294,"journal":{"name":"Advances in cancer research","volume":"161 ","pages":"xvii-xx"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141736296","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Prostate cancer is one of the most common malignancies among men worldwide. Besides genetic alterations, epigenetic modulations including DNA methylation, histone modifications and miRNA mediated alteration of gene expression are the key driving forces for the prostate tumor development and cancer progression. Aberrant expression and/or the activity of the epigenetic modifiers/enzymes, results in aberrant expression of genes involved in DNA repair, cell cycle regulation, cell adhesion, apoptosis, autophagy, tumor suppression and hormone response and thereby disease progression. Altered epigenome is associated with prostate cancer recurrence, progression, aggressiveness and transition from androgen-dependent to androgen-independent phenotype. These epigenetic modifications are reversible and various compounds/drugs targeting the epigenetic enzymes have been developed that are effective in cancer treatment. This chapter focuses on the epigenetic alterations in prostate cancer initiation and progression, listing different epigenetic biomarkers for diagnosis and prognosis of the disease and their potential as therapeutic targets. This chapter also summarizes different epigenetic drugs approved for prostate cancer therapy and the drugs available for clinical trials.
前列腺癌是全球男性最常见的恶性肿瘤之一。除基因改变外,包括 DNA 甲基化、组蛋白修饰和 miRNA 介导的基因表达改变在内的表观遗传修饰是前列腺肿瘤发生和癌症进展的主要驱动力。表观遗传修饰因子/酶的异常表达和/或活性导致参与 DNA 修复、细胞周期调控、细胞粘附、细胞凋亡、自噬、肿瘤抑制和激素反应的基因异常表达,从而导致疾病进展。表观基因组的改变与前列腺癌的复发、进展、侵袭性以及从依赖雄激素表型向不依赖雄激素表型的转变有关。这些表观遗传修饰是可逆的,目前已开发出多种针对表观遗传酶的化合物/药物,可有效治疗癌症。本章重点介绍前列腺癌发生和发展过程中的表观遗传学改变,列出用于诊断和预后的不同表观遗传学生物标志物及其作为治疗靶点的潜力。本章还总结了已批准用于前列腺癌治疗的不同表观遗传药物以及可用于临床试验的药物。
{"title":"Epigenetic regulation of androgen dependent and independent prostate cancer.","authors":"Jagdish Mishra, Subhajit Chakraborty, Piyasa Nandi, Soumen Manna, Tirthankar Baral, Niharika, Ankan Roy, Prahallad Mishra, Samir Kumar Patra","doi":"10.1016/bs.acr.2024.05.007","DOIUrl":"https://doi.org/10.1016/bs.acr.2024.05.007","url":null,"abstract":"<p><p>Prostate cancer is one of the most common malignancies among men worldwide. Besides genetic alterations, epigenetic modulations including DNA methylation, histone modifications and miRNA mediated alteration of gene expression are the key driving forces for the prostate tumor development and cancer progression. Aberrant expression and/or the activity of the epigenetic modifiers/enzymes, results in aberrant expression of genes involved in DNA repair, cell cycle regulation, cell adhesion, apoptosis, autophagy, tumor suppression and hormone response and thereby disease progression. Altered epigenome is associated with prostate cancer recurrence, progression, aggressiveness and transition from androgen-dependent to androgen-independent phenotype. These epigenetic modifications are reversible and various compounds/drugs targeting the epigenetic enzymes have been developed that are effective in cancer treatment. This chapter focuses on the epigenetic alterations in prostate cancer initiation and progression, listing different epigenetic biomarkers for diagnosis and prognosis of the disease and their potential as therapeutic targets. This chapter also summarizes different epigenetic drugs approved for prostate cancer therapy and the drugs available for clinical trials.</p>","PeriodicalId":94294,"journal":{"name":"Advances in cancer research","volume":"161 ","pages":"223-320"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141736278","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}