Molecular Biology of Ovarian Cancer: From Mechanisms of Intraperitoneal Metastasis to Therapeutic Opportunities

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

Deadline for manuscript submissions: closed (30 November 2020) | Viewed by 43344

Special Issue Editor


E-Mail Website
Guest Editor
Immunology and Cellular Biology Department, Molecular Biology Centre Severo Ochoa, Madrid, Spain
Interests: mesothelial cells; peritoneal metastasis; mesothelial to mesenchymal transition

Special Issue Information

Dear Colleagues,

Ovarian cancer is still the most lethal malignancy of the female genital tract. It is diagnosed usually at advanced stages of the disease, mostly in patients with peritoneal carcinomatosis. To the largest extent, the severity of the disease is associated with late diagnosis, resulting from the lack of valuable and specific markers of ovarian cancer cells. Another problem is the proximity of the ovaries with the prime site of the cancer metastasis – the peritoneum. A therapy of ovarian cancer largely depends on the stage of the disease. The best outcomes are associated with the radical cytoreduction, followed by the chemotherapy based on platins combined with taxanes. Unfortunately, however, the relapse of the disease is often, and mechanisms of this phenomenon are elusive.  

The proposal welcomes original research and review article that will illustrate and stimulate the rapid advances that are taking place in the area of ovarian cancer research. We are especially interested in articles describing molecular mechanisms of the development of intraperitoneal ovarian cancer metastasis and studies providing new targets for personalized therapy of the disease. 

Potential subjects include but are not limited to the following:

  • molecular features of ovarian cancer cells promoting their growth and dissemination
  • small and long non-coding RNAs in ovarian cancer biology
  • the role of reactive stroma in ovarian cancer progression
  • mechanisms of ovarian cancer relapse
  • ovarian cancer stem cells
  • malignant ascites in ovarian cancer development
  • EMT, MET, and MMT in ovarian cancer progression
  • molecular targets for therapy of ovarian cancer
  • personalized approaches to combat ovarian cancer

Prof. Krzysztof Ksiązek
Prof. Manuel Lopez-Cabrera
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

  • ovarian cancer
  • personalized therapy
  • tumor microenvironment

Published Papers (13 papers)

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

Editorial

Jump to: Research, Review, Other

4 pages, 185 KiB  
Editorial
Molecular Biology of Ovarian Cancer: From Mechanisms of Intraperitoneal Metastasis to Therapeutic Opportunities
by Krzysztof Książek
Cancers 2021, 13(7), 1661; https://doi.org/10.3390/cancers13071661 - 01 Apr 2021
Cited by 3 | Viewed by 1639
Abstract
Ovarian cancer (OC) is one of the most frequent malignancies of the female genital tract, and is still the leading cause of death from gynecological tumors [...] Full article

Research

Jump to: Editorial, Review, Other

15 pages, 3949 KiB  
Article
FOXM1 Inhibition in Ovarian Cancer Tissue Cultures Affects Individual Treatment Susceptibility Ex Vivo
by Luzie Brückner, Annika Reinshagen, Ngoc Anh Hoang, Anne Kathrin Höhn, Florian Lordick, Ingo Bechmann, Bahriye Aktas, Ivonne Nel and Sonja Kallendrusch
Cancers 2021, 13(5), 956; https://doi.org/10.3390/cancers13050956 - 25 Feb 2021
Cited by 11 | Viewed by 2658
Abstract
Diagnosis in an advanced state is a major hallmark of ovarian cancer and recurrence after first line treatment is common. With upcoming novel therapies, tumor markers that support patient stratification are urgently needed to prevent ineffective therapy. Therefore, the transcription factor FOXM1 is [...] Read more.
Diagnosis in an advanced state is a major hallmark of ovarian cancer and recurrence after first line treatment is common. With upcoming novel therapies, tumor markers that support patient stratification are urgently needed to prevent ineffective therapy. Therefore, the transcription factor FOXM1 is a promising target in ovarian cancer as it is frequently overexpressed and associated with poor prognosis. In this study, fresh tissue specimens of 10 ovarian cancers were collected to investigate tissue cultures in their ability to predict individual treatment susceptibility and to identify the benefit of FOXM1 inhibition. FOXM1 inhibition was induced by thiostrepton (3 µM). Carboplatin (0.2, 2 and 20 µM) and olaparib (10 µM) were applied and tumor susceptibility was analyzed by tumor cell proliferation and apoptosis in immunofluorescence microscopy. Resistance mechanisms were investigated by determining the gene expression of FOXM1 and its targets BRCA1/2 and RAD51. Ovarian cancer tissue was successfully maintained for up to 14 days ex vivo, preserving morphological characteristics of the native specimen. Thiostrepton downregulated FOXM1 expression in tissue culture. Individual responses were observed after combined treatment with carboplatin or olaparib. Thus, we successfully implemented a complex tissue culture model to ovarian cancer and showed potential benefit of combined FOXM1 inhibition. Full article
Show Figures

Figure 1

16 pages, 786 KiB  
Article
Remission-Stage Ovarian Cancer Cell Vaccine with Cowpea Mosaic Virus Adjuvant Prevents Tumor Growth
by Courtney T. Stump, Gregory Ho, Chenkai Mao, Frank A. Veliz, Veronique Beiss, Jennifer Fields, Nicole F. Steinmetz and Steven Fiering
Cancers 2021, 13(4), 627; https://doi.org/10.3390/cancers13040627 - 05 Feb 2021
Cited by 14 | Viewed by 3177
Abstract
Ovarian cancer is the deadliest gynecological malignancy. Though most patients enter remission following initial interventions, relapse is common and often fatal. Accordingly, there is a substantial need for ovarian cancer therapies that prevent relapse. Following remission generated by surgical debulking and chemotherapy, but [...] Read more.
Ovarian cancer is the deadliest gynecological malignancy. Though most patients enter remission following initial interventions, relapse is common and often fatal. Accordingly, there is a substantial need for ovarian cancer therapies that prevent relapse. Following remission generated by surgical debulking and chemotherapy, but prior to relapse, resected and inactivated tumor tissue could be used as a personalized vaccine antigen source. The patient’s own tumor contains relevant antigens and, when combined with the appropriate adjuvant, could generate systemic antitumor immunity to prevent relapse. Here, we model this process in mice to investigate the optimal tumor preparation and vaccine adjuvant. Cowpea mosaic virus (CPMV) has shown remarkable efficacy as an immunostimulatory cancer therapy in ovarian cancer mouse models, so we use CPMV as an adjuvant in a prophylactic vaccine against a murine ovarian cancer model. Compared to its codelivery with tumor antigens prepared in three other ways, we show that CPMV co-delivered with irradiated ovarian cancer cells constitutes an effective prophylactic vaccine against a syngeneic model of ovarian cancer in C57BL/6J mice. Following two vaccinations, 72% of vaccinated mice reject tumor challenges, and all those mice survived subsequent rechallenges, demonstrating immunologic memory formation. This study supports remission-stage vaccines using irradiated patient tumor tissue as a promising option for treating ovarian cancer, and validates CPMV as an antitumor vaccine adjuvant for that purpose. Full article
Show Figures

Figure 1

22 pages, 4845 KiB  
Article
Ovulatory Follicular Fluid Facilitates the Full Transformation Process for the Development of High-Grade Serous Carcinoma
by Che-Fang Hsu, Pao-Chu Chen, Vaishnavi Seenan, Dah-Ching Ding and Tang-Yuan Chu
Cancers 2021, 13(3), 468; https://doi.org/10.3390/cancers13030468 - 26 Jan 2021
Cited by 15 | Viewed by 3130
Abstract
Background: High-grade serous carcinoma (HGSC) is mainly derived from the stepwise accumulation of driver mutations in the fallopian tube epithelium (FTE), and it subsequently metastasizes to the ovary and peritoneum that develops into a clinically evident ovarian carcinoma. The developmental process involves [...] Read more.
Background: High-grade serous carcinoma (HGSC) is mainly derived from the stepwise accumulation of driver mutations in the fallopian tube epithelium (FTE), and it subsequently metastasizes to the ovary and peritoneum that develops into a clinically evident ovarian carcinoma. The developmental process involves cell proliferation/clonal expansion, cell migration, anoikis resistance, anchorage-independent growth (AIG), peritoneum attachment, and cell invasion. Previously, we discovered FTE could be transformed by follicular fluid (FF) released from ovulation, the most crucial risk factor of ovarian cancer, and IGF axis proteins in FF confers stemness activation and clonal expansion via IGF-1R/AKT pathway. However, whether other phenotypes in advanced cancer development are involved is unknown. Methods: A panel of FTE and ovarian HGSC cell lines with different severity of transformation were treated with FF with or without IGF-1R and AKT inhibitors and analyzed for the transformation phenotypes in vitro, ex vivo, and in vivo. Results: FF largely promotes (by order of magnitude) cell migration, AIG, cell invasion, peritoneum attachment, anoikis resistance, and cell proliferation. Most of these activities worked in the full panel of cell lines. The AIG activity largely depends on IGF-1R/AKT phosphorylation, and the proliferation activity depends on an AKT phosphorylation not mediated by IGF-1R. In contrast, both AKT- and non-AKT-mediated signals are responsible for the other transformation activities. Conclusions: Our data demonstrate an extensive transformation activity of FF in the full journey of carcinogenesis, and endorsed ovulation-inhibition for the prevention and AKT-inhibition for the treatment of ovarian HGSC. Full article
Show Figures

Graphical abstract

19 pages, 3137 KiB  
Article
Changes in the Tumor Immune Microenvironment during Disease Progression in Patients with Ovarian Cancer
by Marie Christine Wulff Westergaard, Katy Milne, Magnus Pedersen, Thomas Hasselager, Lars Rønn Olsen, Michael S. Anglesio, Troels Holz Borch, Mia Kennedy, Gillian Briggs, Stacey Ledoux, Caroline Kreuzinger, Isabel von der Decken, Marco Donia, Dan Cacsire Castillo-Tong, Brad H. Nelson and Inge Marie Svane
Cancers 2020, 12(12), 3828; https://doi.org/10.3390/cancers12123828 - 18 Dec 2020
Cited by 19 | Viewed by 3270
Abstract
Anti-PD1/PDL1 therapy has proven efficacious against many cancers but only reached modest objective response rates against recurrent ovarian cancer. A deeper understanding of the tumor microenvironment (TME) may reveal other immunosuppressive mechanisms that warrant investigation as immunotherapeutic targets for this challenging disease. Matched [...] Read more.
Anti-PD1/PDL1 therapy has proven efficacious against many cancers but only reached modest objective response rates against recurrent ovarian cancer. A deeper understanding of the tumor microenvironment (TME) may reveal other immunosuppressive mechanisms that warrant investigation as immunotherapeutic targets for this challenging disease. Matched primary and recurrent tumors from patients with high-grade serous ovarian carcinoma (HGSC) were analyzed by multicolor immunohistochemistry/immunofluorescence for the presence of T cells, B cells, macrophages, and for the expression of immunosuppressive and HLA molecules. Cancer- and immune-related gene expression was assessed by NanoString analysis. Recurrent tumors showed increased infiltration by immune cells, displayed higher expression of PDL1, IDO, and HLA molecules, and contained more stromal tissue. NanoString analysis demonstrated increased expression of gene signatures related to chemokines and T cell functions in recurrent tumors. The ovarian tumors showed high gene expression of LAG3 and HAVCR2 (TIM3) and enhanced levels of TIGIT and CTLA4 in recurrent tumors compared to primary tumors. The majority of HGSC developed into a more inflamed phenotype during progression from primary to recurrent disease, including indications of adaptive immune resistance. This suggests that recurrent tumors may be particularly sensitive to inhibition of adaptive immune resistance mechanisms. Full article
Show Figures

Graphical abstract

19 pages, 6109 KiB  
Article
CD83, a Novel MAPK Signaling Pathway Interactor, Determines Ovarian Cancer Cell Fate
by Aalia Batool, Hao Liu, Yi-Xun Liu and Su-Ren Chen
Cancers 2020, 12(8), 2269; https://doi.org/10.3390/cancers12082269 - 13 Aug 2020
Cited by 11 | Viewed by 3197
Abstract
Ovarian cancer is a leading cause of death from gynecologic malignancies worldwide. Although CD83 is widely described as a solid marker for mature dendritic cells, emerging pieces of evidence indicate the expression of membrane protein CD83 by various tumor cells, including ovarian cancer [...] Read more.
Ovarian cancer is a leading cause of death from gynecologic malignancies worldwide. Although CD83 is widely described as a solid marker for mature dendritic cells, emerging pieces of evidence indicate the expression of membrane protein CD83 by various tumor cells, including ovarian cancer cells. However, the potential role of CD83 in ovarian cancer cell properties and development remains absolutely unknown. By using human CD83 stable overexpression and knockdown sublines of several ovarian cancer cells, we observed that CD83 advanced the growth proliferation, colony formation ability, spheroid formation, and in vivo tumorigenicity of ovarian cancer cells; surprisingly, CD83 limited their migration and invasion potentials. Positive regulation of proliferation/stemness factors (e.g., cyclin-CDKs and KIT/CD44) but negative regulation of matrix metallopeptidases (e.g., MMP1 and 7) by CD83 were revealed by the integrated analysis of transcriptome and proteome. Furthermore, immunoprecipitation-mass spectrometry (IP-MS) and co-immunoprecipitation (Co-IP) first identified the association of CD83 with MAP3K7 (also known as TAK1) and MAP3K7-binding protein TAB1 on the cell membrane. Moreover, CD83 functions through the activation of MAP3K7-MEK1/2-ERK1/2 cascades to further regulate downstream FOXO1/p21/CDK2/CCNB1 and STAT3/DKK1 signaling pathways, thus activating proliferation and spheroid formation of ovarian cancer cells, respectively. Collectively, our findings define a CD83-MAPK pathway in the regulation of proliferation and stemness in ovarian cancer cells, with potential therapeutic applications in blocking their progression. Full article
Show Figures

Figure 1

22 pages, 4159 KiB  
Article
EpCAM-Binding DARPins for Targeted Photodynamic Therapy of Ovarian Cancer
by Dirk van den Brand, Sanne A. M. van Lith, Jelske M. de Jong, Mark A. J. Gorris, Valentina Palacio-Castañeda, Stijn T. Couwenbergh, Mark R. G. Goldman, Inge Ebisch, Leon F. Massuger, William P. J. Leenders, Roland Brock and Wouter P. R. Verdurmen
Cancers 2020, 12(7), 1762; https://doi.org/10.3390/cancers12071762 - 02 Jul 2020
Cited by 18 | Viewed by 3833
Abstract
Ovarian cancer is the most lethal gynecological malignancy due to late detection associated with dissemination throughout the abdominal cavity. Targeted photodynamic therapy (tPDT) aimed at epithelial cell adhesion molecule (EpCAM), overexpressed in over 90% of ovarian cancer metastatic lesions, is a promising novel [...] Read more.
Ovarian cancer is the most lethal gynecological malignancy due to late detection associated with dissemination throughout the abdominal cavity. Targeted photodynamic therapy (tPDT) aimed at epithelial cell adhesion molecule (EpCAM), overexpressed in over 90% of ovarian cancer metastatic lesions, is a promising novel therapeutic modality. Here, we tested the specificity and activity of conjugates of EpCAM-directed designed ankyrin repeat proteins (DARPins) with the photosensitizer IRDye 700DX in in vitro and in vivo ovarian cancer models. EpCAM-binding DARPins (Ec1: Kd = 68 pM; Ac2: Kd = 130 nM) and a control DARPin were site-specifically functionalized with fluorophores or IRDye 700DX. Conjugation of anti-EpCAM DARPins with fluorophores maintained EpCAM-specific binding in cell lines and patient-derived ovarian cancer explants. Penetration of DARPin Ec1 into tumor spheroids was slower than that of Ac2, indicative of a binding site barrier effect for Ec1. DARPin-IRDye 700DX conjugates killed EpCAM-expressing cells in a highly specific and illumination-dependent fashion in 2D and 3D cultures. Furthermore, they effectively homed to EpCAM-expressing subcutaneous OV90 xenografts in mice. In conclusion, the high activity and specificity observed in preclinical ovarian cancer models, combined with a high specificity in patient material, warrant a further investigation of EpCAM-targeted PDT for ovarian cancer. Full article
Show Figures

Figure 1

19 pages, 5282 KiB  
Article
Neutrophil Extracellular Trap Formation Correlates with Favorable Overall Survival in High Grade Ovarian Cancer
by Besnik Muqaku, Dietmar Pils, Johanna C. Mader, Stefanie Aust, Andreas Mangold, Liridon Muqaku, Astrid Slany, Giorgia Del Favero and Christopher Gerner
Cancers 2020, 12(2), 505; https://doi.org/10.3390/cancers12020505 - 21 Feb 2020
Cited by 32 | Viewed by 4689
Abstract
It is still a question of debate whether neutrophils, often found in the tumor microenvironment, mediate tumor-promoting or rather tumor-inhibiting activities. The present study focuses on the involvement of neutrophils in high grade serous ovarian cancer (HGSOC). Macroscopic features classify two types of [...] Read more.
It is still a question of debate whether neutrophils, often found in the tumor microenvironment, mediate tumor-promoting or rather tumor-inhibiting activities. The present study focuses on the involvement of neutrophils in high grade serous ovarian cancer (HGSOC). Macroscopic features classify two types of peritoneal tumor spread in HGSOC. Widespread and millet sized lesions characterize the miliary type, while non-miliary metastases are larger and associated with better prognosis. Multi-omics and FACS data were generated from ascites samples. Integrated data analysis demonstrates a significant increase of neutrophil extracellular trap (NET)-associated molecules in non-miliary ascites samples. A co-association network analysis performed with the ascites data further revealed a striking correlation between NETosis-associated metabolites and several eicosanoids. The congruence of data generated from primary neutrophils with ascites analyses indicates the predominance of NADPH oxidase 2 (NOX)-independent NETosis. NETosis is associated with protein S100A8/A9 release. An increase of the S100A8/CRP abundance ratio was found to correlate with favorable survival of HGSOC patients. The analysis of additional five independent proteome studies with regard to S100A8/CRP ratios confirmed this observation. In conclusion, NET formation seems to relate with better cancer patient outcome. Full article
Show Figures

Graphical abstract

20 pages, 3828 KiB  
Article
Deciphering the Molecular Mechanism of Spontaneous Senescence in Primary Epithelial Ovarian Cancer Cells
by Martyna Pakuła, Ewa Mały, Paweł Uruski, Anna Witucka, Małgorzata Bogucka, Natalia Jaroszewska, Nicoletta Makowska, Arkadiusz Niklas, Rafał Moszyński, Stefan Sajdak, Andrzej Tykarski, Justyna Mikuła-Pietrasik and Krzysztof Książek
Cancers 2020, 12(2), 296; https://doi.org/10.3390/cancers12020296 - 27 Jan 2020
Cited by 17 | Viewed by 3732 | Correction
Abstract
Spontaneous senescence of cancer cells remains a puzzling and poorly understood phenomenon. Here we comprehensively characterize this process in primary epithelial ovarian cancer cells (pEOCs). Analysis of tumors from ovarian cancer patients showed an abundance of senescent cells in vivo. Further, serially passaged [...] Read more.
Spontaneous senescence of cancer cells remains a puzzling and poorly understood phenomenon. Here we comprehensively characterize this process in primary epithelial ovarian cancer cells (pEOCs). Analysis of tumors from ovarian cancer patients showed an abundance of senescent cells in vivo. Further, serially passaged pEOCs become senescent after a few divisions. These senescent cultures display trace proliferation, high expression of senescence biomarkers (SA-β-Gal, γ-H2A.X), growth-arrest in the G1 phase, increased level of cyclins D1, D2, decreased cyclin B1, up-regulated p16, p21, and p53 proteins, eroded telomeres, reduced activity of telomerase, predominantly non-telomeric DNA damage, activated AKT, AP-1, and ERK1/2 signaling, diminished JNK, NF-κB, and STAT3 pathways, increased formation of reactive oxygen species, unchanged activity of antioxidants, increased oxidative damage to DNA and proteins, and dysfunctional mitochondria. Moreover, pEOC senescence is inducible by normal peritoneal mesothelium, fibroblasts, and malignant ascites via the paracrine activity of GRO-1, HGF, and TGF-β1. Collectively, pEOCs undergo spontaneous senescence in a mosaic, telomere-dependent and telomere-independent manner, plausibly in an oxidative stress-dependent mechanism. The process may also be activated by extracellular stimuli. The biological and clinical significance of pEOC senescence remains to be explored. Full article
Show Figures

Figure 1

17 pages, 2765 KiB  
Article
L1 Cell Adhesion Molecule Confers Radioresistance to Ovarian Cancer and Defines a New Cancer Stem Cell Population
by Nastassja Terraneo, Francis Jacob, Claudia Peitzsch, Anna Dubrovska, Christiane Krudewig, Yen-Lin Huang, Viola Heinzelmann-Schwarz, Roger Schibli, Martin Béhé and Jürgen Grünberg
Cancers 2020, 12(1), 217; https://doi.org/10.3390/cancers12010217 - 15 Jan 2020
Cited by 22 | Viewed by 3739
Abstract
Many solid tumors, including ovarian cancer, contain small populations of cancer stem cells (CSCs). These cells are usually resistant against conventional cancer therapies and play a role in disease recurrence. We demonstrated that the L1 cell adhesion molecule (L1CAM) is a new CSC [...] Read more.
Many solid tumors, including ovarian cancer, contain small populations of cancer stem cells (CSCs). These cells are usually resistant against conventional cancer therapies and play a role in disease recurrence. We demonstrated that the L1 cell adhesion molecule (L1CAM) is a new CSC target in ovarian cancer, triggering radioresistance. Using fluorescence-activated cell sorting, specific cell populations expressing L1CAM alone or in combination with the established CSC marker CD133 were isolated from three ovarian cancer cell lines. Double-positive L1CAM+/CD133+ cells displayed higher spherogenic and clonogenic properties in comparison to L1CAM−/CD133− cells. Furthermore, L1CAM+/CD133+ cells retained highest clonogenic capacity after irradiation and exhibited up-regulation of some CSC-specific genes, enhanced tumor-initiating capacity, self-renewal and higher tumor take rate in nude mice when compared with other cell populations. Superior radioresistance by L1CAM expression was confirmed by deletion of L1CAM using CRISPR-Cas9 technology. Moreover, we found expression signatures associated with epithelial-to-mesenchymal transition phenotype in L1CAM deleted cells. These results indicate that L1CAM in combination with CD133 defines a new cancer cell population of ovarian tumor-initiating cells with the implication of targeting L1CAM as a novel therapeutic approach for ovarian CSCs. Full article
Show Figures

Figure 1

Review

Jump to: Editorial, Research, Other

25 pages, 1742 KiB  
Review
Lipid Regulatory Proteins as Potential Therapeutic Targets for Ovarian Cancer in Obese Women
by Jing Yang and M. Sharon Stack
Cancers 2020, 12(11), 3469; https://doi.org/10.3390/cancers12113469 - 21 Nov 2020
Cited by 19 | Viewed by 4027
Abstract
Obesity has become a recognized global epidemic that is associated with numerous comorbidities including type II diabetes, cardiovascular disease, hypertension, and cancer incidence and progression. Ovarian cancer (OvCa) has a unique mechanism of intra-peritoneal metastasis, already present in 80% of women at the [...] Read more.
Obesity has become a recognized global epidemic that is associated with numerous comorbidities including type II diabetes, cardiovascular disease, hypertension, and cancer incidence and progression. Ovarian cancer (OvCa) has a unique mechanism of intra-peritoneal metastasis, already present in 80% of women at the time of diagnosis, making it the fifth leading cause of death from gynecological malignancy. Meta-analyses showed that obesity increases the risk of OvCa progression, leads to enhanced overall and organ-specific tumor burden, and adversely effects survival of women with OvCa. Recent data discovered that tumors grown in mice fed on a western diet (40% fat) have elevated lipid levels and a highly increased expression level of sterol regulatory element binding protein 1 (SREBP1). SREBP1 is a master transcription factor that regulates de novo lipogenesis and lipid homeostasis, and induces lipogenic reprogramming of tumor cells. Elevated SREBP1 levels are linked to cancer cell proliferation and metastasis. This review will summarize recent findings to provide a current understanding of lipid regulatory proteins in the ovarian tumor microenvironment with emphasis on SREBP1 expression in the obese host, the role of SREBP1 in cancer progression and metastasis, and potential therapeutic targeting of SREBPs and SREBP-pathway genes in treating cancers, particularly in the context of host obesity. Full article
Show Figures

Figure 1

14 pages, 1195 KiB  
Review
Selectins: An Important Family of Glycan-Binding Cell Adhesion Molecules in Ovarian Cancer
by Ayon A. Hassan, Margarita Artemenko, Maggie K.S. Tang and Alice S.T. Wong
Cancers 2020, 12(8), 2238; https://doi.org/10.3390/cancers12082238 - 10 Aug 2020
Cited by 17 | Viewed by 4557
Abstract
Ovarian cancer is the most lethal gynecological malignancy worldwide. Unlike most other tumor types that metastasize via the vasculature, ovarian cancer metastasizes predominantly via the transcoelomic route within the peritoneal cavity. As cancer metastasis accounts for the majority of deaths, there is an [...] Read more.
Ovarian cancer is the most lethal gynecological malignancy worldwide. Unlike most other tumor types that metastasize via the vasculature, ovarian cancer metastasizes predominantly via the transcoelomic route within the peritoneal cavity. As cancer metastasis accounts for the majority of deaths, there is an urge to better understand its determinants. In the peritoneal cavity, tumor-mesothelial adhesion is an important step for cancer dissemination. Selectins are glycan-binding molecules that facilitate early steps of this adhesion cascade by mediating heterotypic cell-cell interaction under hydrodynamic flow. Here, we review the function and regulation of selectins in peritoneal carcinomatosis of ovarian cancer, and highlight how dysregulation of selectin ligand biogenesis affects disease outcome. Further, we will introduce the latest tools in studying selectin-glycan interaction. Finally, an overview of potential therapeutic intervention points that may lead to the development of efficacious therapies for ovarian cancer is provided. Full article
Show Figures

Figure 1

Other

3 pages, 1628 KiB  
Correction
Correction: Pakuła et al. Deciphering the Molecular Mechanism of Spontaneous Senescence in Primary Epithelial Ovarian Cancer Cells. Cancers 2020, 12, 296
by Martyna Pakuła, Ewa Mały, Paweł Uruski, Anna Witucka, Małgorzata Bogucka, Natalia Jaroszewska, Nicoletta Makowska, Arkadiusz Niklas, Rafał Moszyński, Stefan Sajdak, Andrzej Tykarski, Justyna Mikuła-Pietrasik and Krzysztof Książek
Cancers 2023, 15(3), 937; https://doi.org/10.3390/cancers15030937 - 02 Feb 2023
Viewed by 792
Abstract
In the original publication [...] Full article
Show Figures

Figure 5

Back to TopTop