Targeting Cancer Metastasis (Volume II)

A special issue of Cancers (ISSN 2072-6694). This special issue belongs to the section "Cancer Metastasis".

Deadline for manuscript submissions: closed (31 March 2023) | Viewed by 16818

Special Issue Editors


E-Mail Website
Guest Editor
Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
Interests: pharmacokinetics; natural compounds; anticancer agents; drug–drug interactions; cancer biomarkers
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
St. Jude Children's Research Hospital, Memphis, TN, USA
Interests: myelofibrosis; cancer metabolism; cancer therapeutics
Special Issues, Collections and Topics in MDPI journals
The Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
Interests: cancer immunology; cancer stem cells; metabolism; leukemia; pluripotent stem cells; hematopoiesis

Special Issue Information

Dear Colleagues,

This collection is the second edition of the previous "Targeting Cancer Metastasis" at https://www.mdpi.com/journal/cancers/special_issues/TCM.

Tumor metastasis, one of the crucial hallmarks of cancer, is the major cause of treatment failure in cancer patients, leading to 90% of cancer-related deaths. It is now widely recognized that metastatic lesions are distinct from the primary tumors which have traditionally been targeted. Thus, the development of novel therapeutic approaches to effectively prevent metastasis, as well as target metastatic lesions, is an unmet clinical need.

Metastasis is an inherently complex, heterogeneous, and dynamic pathophysiological process that is highly dependent on the interaction of the tumor with the tumor microenvironment (TME). Multiple biological factors work synergistically to create pro-metastatic niches, such as hypoxia, reprogrammed anaerobic glycolysis (Warburg effect), angiogenesis, vascular permeability, lymphatic vasculature, cancer stem cells, regulatory and suppressive immune cells, cancer-associated fibroblasts (CAFs), extracellular matrix, and tumor-derived exosomes (TDEs). Therefore, novel therapeutic strategies to normalize TME and clinical tests to precisely predict cancer metastasis can greatly improve the survival outcome of cancer patients.

This Special Issue highlights new frontiers in the treatment of metastatic cancers, covering preclinical studies identifying novel druggable targets and discovering potent drug candidates, as well as clinical management of cancer metastasis in combination with predictive biomarkers to improve the outcome of cancer patients.

Dr. Lingzhi Wang
Dr. Qiang Jeremy Wen
Dr. Lei Han
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cancers is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • cancer therapeutics
  • metastasis
  • hypoxia
  • Warburg effect
  • blood vessel disruption
  • tumor-derived exosomes
 

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

18 pages, 2805 KiB  
Article
ICAM-1-suPAR-CD11b Axis Is a Novel Therapeutic Target for Metastatic Triple-Negative Breast Cancer
by Dong Li, Hami Hemati, Younhee Park, Rokana Taftaf, Youbin Zhang, Jinpeng Liu, Massimo Cristofanilli and Xia Liu
Cancers 2023, 15(10), 2734; https://doi.org/10.3390/cancers15102734 - 12 May 2023
Cited by 3 | Viewed by 2087
Abstract
Accumulating evidence demonstrates that circulating tumor cell (CTC) clusters have higher metastatic ability than single CTCs and negatively correlate with cancer patient outcomes. Along with homotypic CTC clusters, heterotypic CTC clusters (such as neutrophil–CTC clusters), which have been identified in both cancer mouse [...] Read more.
Accumulating evidence demonstrates that circulating tumor cell (CTC) clusters have higher metastatic ability than single CTCs and negatively correlate with cancer patient outcomes. Along with homotypic CTC clusters, heterotypic CTC clusters (such as neutrophil–CTC clusters), which have been identified in both cancer mouse models and cancer patients, lead to more efficient metastasis formation and worse patient outcomes. However, the mechanism by which neutrophils bind to CTCs remains elusive. In this study, we found that intercellular adhesion molecule-1 (ICAM-1) on triple-negative breast cancer (TNBC) cells and CD11b on neutrophils mediate tumor cell–neutrophil binding. Consequently, CD11b deficiency inhibited tumor cell–neutrophil binding and TNBC metastasis. Furthermore, CD11b mediated hydrogen peroxide (H2O2) production from neutrophils. Moreover, we found that ICAM-1 in TNBC cells promotes tumor cells to secrete suPAR, which functions as a chemoattractant for neutrophils. Knockdown of uPAR in ICAM-1+ TNBC cells reduced lung-infiltrating neutrophils and lung metastasis. Bioinformatics analysis confirmed that uPAR is highly expressed in TNBCs, which positively correlates with higher neutrophil infiltration and negatively correlates with breast cancer patient survival. Collectively, our findings provide new insight into how neutrophils bind to CTC to facilitate metastasis and discover a novel potential therapeutic strategy by blocking the ICAM-1-suPAR-CD11b axis to inhibit TNBC metastasis. Full article
(This article belongs to the Special Issue Targeting Cancer Metastasis (Volume II))
Show Figures

Figure 1

21 pages, 1928 KiB  
Article
The TLK1–MK5 Axis Regulates Motility, Invasion, and Metastasis of Prostate Cancer Cells
by Md Imtiaz Khalil and Arrigo De Benedetti
Cancers 2022, 14(23), 5728; https://doi.org/10.3390/cancers14235728 - 22 Nov 2022
Cited by 2 | Viewed by 1977
Abstract
Background: Metastatic dissemination of prostate cancer (PCa) accounts for the majority of PCa-related deaths. However, the exact mechanism of PCa cell spread is still unknown. We uncovered a novel interaction between two unrelated promotility factors, tousled-like kinase 1 (TLK1) and MAPK-activated protein [...] Read more.
Background: Metastatic dissemination of prostate cancer (PCa) accounts for the majority of PCa-related deaths. However, the exact mechanism of PCa cell spread is still unknown. We uncovered a novel interaction between two unrelated promotility factors, tousled-like kinase 1 (TLK1) and MAPK-activated protein kinase 5 (MK5), that initiates a signaling cascade promoting metastasis. In PCa, TLK1–MK5 signaling might be crucial, as androgen deprivation therapy (ADT) leads to increased expression of both TLK1 and MK5 in metastatic patients, but in this work, we directly investigated the motility, invasive, and metastatic capacity of PCa cells following impairment of the TLK1 > MK5 axis. Results: We conducted scratch wound repair and transwell invasion assays with LNCaP and PC3 cells to determine if TLK1 and MK5 can regulate motility and invasion. Both genetic depletion and pharmacologic inhibition of TLK1 and MK5 resulted in reduced migration and invasion through a Matrigel plug. We further elucidated the potential mechanisms underlying these effects and found that this is likely due to the reorganization of the actin fibers at lamellipodia and the focal adhesions network, in conjunction with increased expression of some MMPs that can affect penetration through the ECM. PC3, a highly metastatic cell line when assayed in xenografts, was further tested in a tail-vein injection/lung metastasis model, and we showed that, following inoculation, treatment with GLPG0259 (MK5 specific inhibitor) or J54 (TLK1 inhibitor) resulted in the lung tumor nodules being greatly diminished in number, and for J54, also in size. Conclusion: Our data support that the TLK1–MK5 axis is functionally involved in driving PCa cell metastasis and clinical aggressiveness; hence, disruption of this axis may inhibit the metastatic capacity of PCa. Full article
(This article belongs to the Special Issue Targeting Cancer Metastasis (Volume II))
Show Figures

Figure 1

16 pages, 2855 KiB  
Article
H3F3A K27M Mutation Promotes the Infiltrative Growth of High-Grade Glioma in Adults by Activating β-Catenin/USP1 Signaling
by Zhiyuan Sun, Yufu Zhu, Xia Feng, Xiaoyun Liu, Kunlin Zhou, Qing Wang, Hengzhu Zhang and Hengliang Shi
Cancers 2022, 14(19), 4836; https://doi.org/10.3390/cancers14194836 - 03 Oct 2022
Viewed by 1836
Abstract
H3F3A K27M (H3.3K27M) is a newly identified molecular pathological marker in glioma and is strongly correlated with the malignancy of diffuse intrinsic pontine glioma (DIPG). In recent years, accumulating evidence has revealed that other types of glioma also contain the H3.3K27M mutation. However, [...] Read more.
H3F3A K27M (H3.3K27M) is a newly identified molecular pathological marker in glioma and is strongly correlated with the malignancy of diffuse intrinsic pontine glioma (DIPG). In recent years, accumulating evidence has revealed that other types of glioma also contain the H3.3K27M mutation. However, the role of H3.3K27M in high-grade adult glioma, the most malignant glioma, has not been investigated. In this study, we focused on exploring the expression and function of H3.3K27M in high-grade glioma in adults. We found that H3.3K27M was highly expressed at high levels in some high-grade glioma tissues. Then, we introduced H3.3K27M into H3.3 wild-type glioma cells, U87 cells and LN229 cells. We found that H3.3K27M did not affect the growth of glioma cells in vitro and in vivo; however, the survival of mice with transplanted tumors was significantly reduced. Further investigation revealed that H3.3K27M expression mainly promoted the migration and invasion of glioma cells. Moreover, we confirmed that H3.3K27M overexpression increased the levels of the β-catenin and p-β-catenin (Ser675) proteins, the ubiquitin-specific protease 1 (USP1) mRNA and protein levels, and the enhancer of zeste homolog 2 (EZH2) protein level. In addition, the β-catenin inhibitor XAV-939 significantly attenuated the upregulation of the aforementioned proteins and inhibited the increased migration and invasion caused by the H3.3K27M mutation. Overall, the H3.3K27M mutation in high-grade glioma is a potential biomarker for poor prognosis mainly due to the infiltration of glioma cells that is at least partially mediated by the β-catenin/USP1/EZH2 pathway. Full article
(This article belongs to the Special Issue Targeting Cancer Metastasis (Volume II))
Show Figures

Figure 1

13 pages, 3616 KiB  
Article
SIPA1 Regulates LINC01615 to Promote Metastasis in Triple-Negative Breast Cancer
by Yuan Xiang, Lingyun Feng, Hui Liu, Yuhuan Liu, Jiapeng Li, Li Su and Xinghua Liao
Cancers 2022, 14(19), 4815; https://doi.org/10.3390/cancers14194815 - 01 Oct 2022
Cited by 6 | Viewed by 1466
Abstract
Long non-coding RNAs (lncRNAs) are reported to play an important regulatory effect in carcinogenesis and malignancy. We found by high-throughput sequencing that LINC01615 is upregulated in breast cancer patients and reduces patients’ overall survival. In vivo and in vitro experiments, we clarified that [...] Read more.
Long non-coding RNAs (lncRNAs) are reported to play an important regulatory effect in carcinogenesis and malignancy. We found by high-throughput sequencing that LINC01615 is upregulated in breast cancer patients and reduces patients’ overall survival. In vivo and in vitro experiments, we clarified that overexpression of LINC01615 can promote breast cancer cell metastasis ability. The expression of LINC01615 is regulated by the transcriptional activator SIPA1, thereby promoting carcinogenesis in breast cancer cells. Our research clarified that LINC01615 can act as an oncogenic factor in promoting the development of breast cancer. Full article
(This article belongs to the Special Issue Targeting Cancer Metastasis (Volume II))
Show Figures

Figure 1

Review

Jump to: Research

25 pages, 1454 KiB  
Review
A Multi-Omics Overview of Colorectal Cancer to Address Mechanisms of Disease, Metastasis, Patient Disparities and Outcomes
by Guang Yang, Xi (Richard) Yu, Daniel J. Weisenberger, Tao Lu and Gangning Liang
Cancers 2023, 15(11), 2934; https://doi.org/10.3390/cancers15112934 - 26 May 2023
Cited by 2 | Viewed by 1923
Abstract
Human colorectal cancer (CRC) is one of the most common malignancies in men and women across the globe, albeit CRC incidence and mortality shows a substantial racial and ethnic disparity, with the highest burden in African American patients. Even with effective screening tools [...] Read more.
Human colorectal cancer (CRC) is one of the most common malignancies in men and women across the globe, albeit CRC incidence and mortality shows a substantial racial and ethnic disparity, with the highest burden in African American patients. Even with effective screening tools such as colonoscopy and diagnostic detection assays, CRC remains a substantial health burden. In addition, primary tumors located in the proximal (right) or distal (left) sides of the colorectum have been shown to be unique tumor types that require unique treatment schema. Distal metastases in the liver and other organ systems are the major causes of mortality in CRC patients. Characterizing genomic, epigenomic, transcriptomic and proteomic (multi-omics) alterations has led to a better understanding of primary tumor biology, resulting in targeted therapeutic advancements. In this regard, molecular-based CRC subgroups have been developed that show correlations with patient outcomes. Molecular characterization of CRC metastases has highlighted similarities and differences between metastases and primary tumors; however, our understanding as to how to improve patient outcomes based on metastasis biology is lagging and remains a major obstacle to improving CRC patient outcomes. In this review, we will summarize the multi-omics features of primary CRC tumors and their metastases across racial and ethnic groups, the differences in proximal and distal tumor biology, molecular-based CRC subgroups, treatment strategies and challenges for improving patient outcomes. Full article
(This article belongs to the Special Issue Targeting Cancer Metastasis (Volume II))
Show Figures

Figure 1

23 pages, 2473 KiB  
Review
Inhibitory Potential of Resveratrol in Cancer Metastasis: From Biology to Therapy
by Baohong Song, Wei Wang, Xuemei Tang, Robby Miguel Wen-Jing Goh, Win Lwin Thuya, Paul Chi Lui Ho, Lu Chen and Lingzhi Wang
Cancers 2023, 15(10), 2758; https://doi.org/10.3390/cancers15102758 - 14 May 2023
Cited by 5 | Viewed by 2389
Abstract
Cancer metastasis is a significant challenge in cancer treatment, and most existing drugs are designed to inhibit tumor growth but are often ineffective in treating metastatic cancer, which is the leading cause of cancer-related deaths. Resveratrol, a polyphenol found in grapes, berries, and [...] Read more.
Cancer metastasis is a significant challenge in cancer treatment, and most existing drugs are designed to inhibit tumor growth but are often ineffective in treating metastatic cancer, which is the leading cause of cancer-related deaths. Resveratrol, a polyphenol found in grapes, berries, and peanuts, has shown potential in preclinical studies as an anticancer agent to suppress metastasis. However, despite positive results in preclinical studies, little progress has been made in clinical trials. To develop resveratrol as an effective anticancer agent, it is crucial to understand its cellular processes and signaling pathways in tumor metastasis. This review article evaluates the current state and future development strategies of resveratrol to enhance its potency against cancer metastasis within its therapeutic dose. In addition, we critically evaluate the animal models used in preclinical studies for cancer metastasis and discuss novel techniques to accelerate the translation of resveratrol from bench to bedside. The appropriate selection of animal models is vital in determining whether resveratrol can be further developed as an antimetastatic drug in cancer therapy. Full article
(This article belongs to the Special Issue Targeting Cancer Metastasis (Volume II))
Show Figures

Figure 1

19 pages, 1941 KiB  
Review
Theranostic Applications of Glycosaminoglycans in Metastatic Renal Cell Carcinoma
by San Hue Hua, Maximillian Viera, George W. Yip and Boon Huat Bay
Cancers 2023, 15(1), 266; https://doi.org/10.3390/cancers15010266 - 30 Dec 2022
Cited by 4 | Viewed by 2122
Abstract
Renal cell carcinoma (RCC) makes up the majority of kidney cancers, with a poor prognosis for metastatic RCC (mRCC). Challenges faced in the management of mRCC, include a lack of reliable prognostic markers and biomarkers for precise monitoring of disease treatment, together with [...] Read more.
Renal cell carcinoma (RCC) makes up the majority of kidney cancers, with a poor prognosis for metastatic RCC (mRCC). Challenges faced in the management of mRCC, include a lack of reliable prognostic markers and biomarkers for precise monitoring of disease treatment, together with the potential risk of toxicity associated with more recent therapeutic options. Glycosaminoglycans (GAGs) are a class of carbohydrates that can be categorized into four main subclasses, viz., chondroitin sulfate, hyaluronic acid, heparan sulfate and keratan sulfate. GAGs are known to be closely associated with cancer progression and modulation of metastasis by modification of the tumor microenvironment. Alterations of expression, composition and spatiotemporal distribution of GAGs in the extracellular matrix (ECM), dysregulate ECM functions and drive cancer invasion. In this review, we focus on the clinical utility of GAGs as biomarkers for mRCC (which is important for risk stratification and strategizing effective treatment protocols), as well as potential therapeutic targets that could benefit patients afflicted with advanced RCC. Besides GAG-targeted therapies that holds promise in mRCC, other potential strategies include utilizing GAGs as drug carriers and their mimetics to counter cancer progression, and enhance immunotherapy through binding and transducing signals for immune mediators. Full article
(This article belongs to the Special Issue Targeting Cancer Metastasis (Volume II))
Show Figures

Figure 1

25 pages, 1188 KiB  
Review
Non-Coding RNAs of Extracellular Vesicles: Key Players in Organ-Specific Metastasis and Clinical Implications
by Qian Jiang, Xiao-Ping Tan, Cai-Hua Zhang, Zhi-Yuan Li, Du Li, Yan Xu, Yu Xuan Liu, Lingzhi Wang and Zhaowu Ma
Cancers 2022, 14(22), 5693; https://doi.org/10.3390/cancers14225693 - 19 Nov 2022
Cited by 3 | Viewed by 1842
Abstract
Extracellular vesicles (EVs) are heterogeneous membrane-encapsulated vesicles released by most cells. They act as multifunctional regulators of intercellular communication by delivering bioactive molecules, including non-coding RNAs (ncRNAs). Metastasis is a major cause of cancer-related death. Most cancer cells disseminate and colonize a specific [...] Read more.
Extracellular vesicles (EVs) are heterogeneous membrane-encapsulated vesicles released by most cells. They act as multifunctional regulators of intercellular communication by delivering bioactive molecules, including non-coding RNAs (ncRNAs). Metastasis is a major cause of cancer-related death. Most cancer cells disseminate and colonize a specific target organ via EVs, a process known as “organ-specific metastasis”. Mounting evidence has shown that EVs are enriched with ncRNAs, and various EV-ncRNAs derived from tumor cells influence organ-specific metastasis via different mechanisms. Due to the tissue-specific expression of EV-ncRNAs, they could be used as potential biomarkers and therapeutic targets for the treatment of tumor metastasis in various types of cancer. In this review, we have discussed the underlying mechanisms of EV-delivered ncRNAs in the most common organ-specific metastases of liver, bone, lung, brain, and lymph nodes. Moreover, we summarize the potential clinical applications of EV-ncRNAs in organ-specific metastasis to fill the gap between benches and bedsides. Full article
(This article belongs to the Special Issue Targeting Cancer Metastasis (Volume II))
Show Figures

Figure 1

Back to TopTop