Pedro P Gonçalves, Cláudia L da Silva, Nuno Bernardes
{"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}
Yuqian Gao, Hyeyoon Kim, Reta Birhanu Kitata, Tai-Tu Lin, Adam C Swensen, Tujin Shi, Tao Liu
{"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}
{"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}
{"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}
Sheeraz Un Nazir, Juhi Mishra, Shailendra Kumar Maurya, Negin Ziamiavaghi, Sanika Bodas, Benjamin A Teply, Samikshan Dutta, Kaustubh Datta
{"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}
{"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}
Rishabh Maurya, Isha Chug, Vignesh Vudatha, António M Palma
{"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}
Padmanabhan Mannangatti, Praveen Bhoopathi, Amit Kumar, Swadesh K Das, Luni Emdad, Paul B Fisher
{"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}
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}
{"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}