ijms-logo

Journal Browser

Journal Browser

Gynecologic Oncology: From Molecular Mechanisms to Targeted Therapies 2018

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Pathology, Diagnostics, and Therapeutics".

Deadline for manuscript submissions: closed (31 December 2018) | Viewed by 36940

Special Issue Editors


E-Mail Website
Guest Editor
Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
Interests: rare ovarian cancer; cancer mouse model; KRAS; p53 genes; targeted therapy
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Co-Guest Editor
Discipline Obstetrics and Gynaecology, Group Leader Reproductive Cancer Research, Robinson Research Institute, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
Interests: ovarian cancer; tumour microenvironment; extracellular matrix; metastasis
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

This Special Issue calls for a joint collaborative effort to illustrate how the understanding of the molecular mechanisms of gynecological cancers may lead to potential targeted therapies.

Considerable progress has been made in deciphering the molecular mechanisms for gynecologic cancers in the past decade. Integrated genomic analyses of large cohorts of ovarian cancer and uterine cancer, through The Cancer Genome Atlas (TCGA) effort, have generated a large amount of data regarding early mutation events and dysregulated pathways in carcinogenesis. Various clinical trials, based on specific molecular features, are being developed or in early phase clinical studies. Targeting cancer metabolism, tumor microenvironment, and immunotherapy are also in the horizon.

We encourage and invite researchers with related experiences in ovarian cancer, uterine cancer, cervical cancer, or other rare gynecological cancers to contribute original research articles or review articles.

Prof. Dr. Kwong-Kwok Wong
Dr. Carmela Ricciardelli
Guest Editor

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. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. 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
  • uterine cancer
  • cervical cancer
  • tyrosine kinases
  • growth factor receptors
  • DNA repair
  • cell cycle
  • cancer metabolism
  • resistance to targeted therapies
  • immunotherapies

Published Papers (8 papers)

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

Research

Jump to: Review, Other

21 pages, 2846 KiB  
Article
Single-Domain Antibodies Represent Novel Alternatives to Monoclonal Antibodies as Targeting Agents against the Human Papillomavirus 16 E6 Protein
by Melissa Togtema, Greg Hussack, Guillem Dayer, Megan R. Teghtmeyer, Shalini Raphael, Jamshid Tanha and Ingeborg Zehbe
Int. J. Mol. Sci. 2019, 20(9), 2088; https://doi.org/10.3390/ijms20092088 - 28 Apr 2019
Cited by 10 | Viewed by 4338
Abstract
Approximately one fifth of all malignancies worldwide are etiologically associated with a persistent viral or bacterial infection. Thus, there is a particular interest in therapeutic molecules which use components of a natural immune response to specifically inhibit oncogenic microbial proteins, as it is [...] Read more.
Approximately one fifth of all malignancies worldwide are etiologically associated with a persistent viral or bacterial infection. Thus, there is a particular interest in therapeutic molecules which use components of a natural immune response to specifically inhibit oncogenic microbial proteins, as it is anticipated they will elicit fewer off-target effects than conventional treatments. This concept has been explored in the context of human papillomavirus 16 (HPV16)-related cancers, through the development of monoclonal antibodies and fragments thereof against the viral E6 oncoprotein. Challenges related to the biology of E6 as well as the functional properties of the antibodies themselves appear to have precluded their clinical translation. Here, we addressed these issues by exploring the utility of the variable domains of camelid heavy-chain-only antibodies (denoted as VHHs). Through construction and panning of two llama, immune VHH phage display libraries, a pool of potential VHHs was isolated. The interactions of these with recombinant E6 were further characterized using an enzyme-linked immunosorbent assay (ELISA), Western blotting under denaturing and native conditions, and surface plasmon resonance. Three VHHs were identified that bound recombinant E6 with nanomolar affinities. Our results lead the way for subsequent studies into the ability of these novel molecules to inhibit HPV16-infected cells in vitro and in vivo. Full article
Show Figures

Figure 1

14 pages, 2110 KiB  
Article
Potential Interplay of the Gatipotuzumab Epitope TA-MUC1 and Estrogen Receptors in Ovarian Cancer
by Sabine Heublein, Sabina Page, Doris Mayr, Elisa Schmoeckel, Fabian Trillsch, Frederik Marmé, Sven Mahner, Udo Jeschke and Aurelia Vattai
Int. J. Mol. Sci. 2019, 20(2), 295; https://doi.org/10.3390/ijms20020295 - 12 Jan 2019
Cited by 7 | Viewed by 3990
Abstract
Anti-tumor efficacy of Gatipotuzumab, a therapeutic antibody targeting Tumor-Associated Mucin-1 (TA-MUC1), in relapsed ovarian cancer (OC) appeared to be rather heterogeneous. Whether adding a second anti-neoplastic drug may augment response towards Gatipotuzumab, has not been elucidated so far. Since it is known that [...] Read more.
Anti-tumor efficacy of Gatipotuzumab, a therapeutic antibody targeting Tumor-Associated Mucin-1 (TA-MUC1), in relapsed ovarian cancer (OC) appeared to be rather heterogeneous. Whether adding a second anti-neoplastic drug may augment response towards Gatipotuzumab, has not been elucidated so far. Since it is known that anti-MUC1 antibodies may alter estrogen receptor activity in breast cancer, this potential interplay was investigated in OC. The correlation between TA-MUC1, estrogen receptors (ERs) and another 12 protein markers as well as their correlation with clinico-pathological parameters in 138 ovarian cancer cases was studied. Finally, Gatipotuzumab and 4-Hydroxy-TTamoxifen (4-OHT) as well as the combination of both was tested for its impact on cell viability in COV318, OV-90, OVCAR-3, and SKOV-3 cells. A strong positive correlation between TA-MUC1 and ERs was detected in OC tissue. Those cases missing ERs but staining positive for TA-MUC1 had significantly reduced overall survival. The combination of 4-OHT and Gatipotuzumab significantly reduced cell viability and was more effective than treatment with Gatipotuzumab alone. Co-stimulation with Gatipotuzumab enhanced the efficacy of 4-OHT in OVCAR-3 and SKOV-3. The data suggest an interplay of TA-MUC1 and ERs in OC. Whether the combination of Gatipotuzumab and TTamoxifen may enhance efficacy of either of the two drugs in vivo, or may even translate into a clinically relevant benefit over the respective monotherapies, remains to be investigated. Full article
Show Figures

Figure 1

10 pages, 4039 KiB  
Article
The Epithelial-Mesenchymal Transition Initiated by Malignant Ascites Underlies the Transmesothelial Invasion of Ovarian Cancer Cells
by Martyna Pakuła, Justyna Mikuła-Pietrasik, Anna Witucka, Katarzyna Kostka-Jeziorny, Paweł Uruski, Rafał Moszyński, Eryk Naumowicz, Stefan Sajdak, Andrzej Tykarski and Krzysztof Książek
Int. J. Mol. Sci. 2019, 20(1), 137; https://doi.org/10.3390/ijms20010137 - 02 Jan 2019
Cited by 14 | Viewed by 3466
Abstract
The role of the epithelial-mesenchymal transition (EMT) in ovarian cancer cell progression is unquestioned. In this report, we describe that malignant ascites, fluid that accumulates in the peritoneal cavity in a large group of patients with ovarian cancer, stimulate EMT in two representative [...] Read more.
The role of the epithelial-mesenchymal transition (EMT) in ovarian cancer cell progression is unquestioned. In this report, we describe that malignant ascites, fluid that accumulates in the peritoneal cavity in a large group of patients with ovarian cancer, stimulate EMT in two representative ovarian cancer cell lines (A2780, SKOV-3). In addition, we identify the ascites-derived mediators of EMT and signaling pathways initiated in the cancer cells that underlie this phenomenon. Finally, we demonstrate that EMT induced in the cancer cells in response to the malignant ascites contributes to their increased transmesothelial invasion. Altogether, our study provides new insight into the mechanistic aspects of the malignant ascites-dependent exacerbation of the intraperitoneal progression of ovarian cancer. Full article
Show Figures

Figure 1

21 pages, 4157 KiB  
Article
Key Immunological Functions Involved in the Progression of Epithelial Ovarian Serous Carcinoma Discovered by the Gene Ontology-Based Immunofunctionome Analysis
by Cheng-Chang Chang, Kuo-Min Su, Kai-Hsi Lu, Chi-Kang Lin, Peng-Hui Wang, Hsin-Yang Li, Mong-Lien Wang, Cheng-Kuo Lin, Mu-Hsien Yu and Chia-Ming Chang
Int. J. Mol. Sci. 2018, 19(11), 3311; https://doi.org/10.3390/ijms19113311 - 24 Oct 2018
Cited by 14 | Viewed by 2740
Abstract
Serous carcinoma (SC) is the most common and lethal subtype of epithelial ovarian carcinoma; immunotherapy is a potential treatment for SC, however, the global immunological functions of SC as well as their change during the progression of SC have not been investigated in [...] Read more.
Serous carcinoma (SC) is the most common and lethal subtype of epithelial ovarian carcinoma; immunotherapy is a potential treatment for SC, however, the global immunological functions of SC as well as their change during the progression of SC have not been investigated in detail till now. We conducted a genome-wide integrative analysis to investigate the immunofunctionomes of SC at four tumor stages by quantifying the immunological functions defined by the Gene Ontology gene sets. DNA microarray gene expression profiles of 1100 SCs and 136 normal ovarian tissue controls were downloaded from the Gene Expression Omnibus database and converted to the functionome. Then the immunofunctionomes were reconstructed by extracting the offspring from the functionome for the four SC staging groups. The key immunological functions extracted from immunofunctionomes with a series of filters revealed that the immunopathy of SC consisted of a group of deregulated functions with the core members including B cell activation and differentiation, regulation of leukocyte chemotaxis/cellular extravasation, antigen receptor mediated signaling pathway, T helper mediated immunity and macrophage activation; and the auxiliary elements included leukocyte mediated immunity, regulation of inflammatory response, T cell differentiation, mononuclear cell migration, megakaryocyte differentiation, complement activation and cytokine production. These deregulated immunological functions reveal the candidates to target in the immunotherapy. Full article
Show Figures

Figure 1

Review

Jump to: Research, Other

17 pages, 926 KiB  
Review
Therapeutic Targeting of Collective Invasion in Ovarian Cancer
by Laura Moffitt, Nazanin Karimnia, Andrew Stephens and Maree Bilandzic
Int. J. Mol. Sci. 2019, 20(6), 1466; https://doi.org/10.3390/ijms20061466 - 22 Mar 2019
Cited by 44 | Viewed by 7163
Abstract
Ovarian cancer is the seventh most commonly diagnosed cancer amongst women and has the highest mortality rate of all gynaecological malignancies. It is a heterogeneous disease attributed to one of three cell types found within the reproductive milieu: epithelial, stromal, and germ cell. [...] Read more.
Ovarian cancer is the seventh most commonly diagnosed cancer amongst women and has the highest mortality rate of all gynaecological malignancies. It is a heterogeneous disease attributed to one of three cell types found within the reproductive milieu: epithelial, stromal, and germ cell. Each histotype differs in etiology, pathogenesis, molecular biology, risk factors, and prognosis. Furthermore, the origin of ovarian cancer remains unclear, with ovarian involvement secondary to the contribution of other gynaecological tissues. Despite these complexities, the disease is often treated as a single entity, resulting in minimal improvement to survival rates since the introduction of platinum-based chemotherapy over 30 years ago. Despite concerted research efforts, ovarian cancer remains one of the most difficult cancers to detect and treat, which is in part due to the unique mode of its dissemination. Ovarian cancers tend to invade locally to neighbouring tissues by direct extension from the primary tumour, and passively to pelvic and distal organs within the peritoneal fluid or ascites as multicellular spheroids. Once at their target tissue, ovarian cancers, like most epithelial cancers including colorectal, melanoma, and breast, tend to invade as a cohesive unit in a process termed collective invasion, driven by specialized cells termed “leader cells”. Emerging evidence implicates leader cells as essential drivers of collective invasion and metastasis, identifying collective invasion and leader cells as a viable target for the management of metastatic disease. However, the development of targeted therapies specifically against this process and this subset of cells is lacking. Here, we review our understanding of metastasis, collective invasion, and the role of leader cells in ovarian cancer. We will discuss emerging research into the development of novel therapies targeting collective invasion and the leader cell population. Full article
Show Figures

Figure 1

13 pages, 239 KiB  
Review
Strengthening the AntiTumor NK Cell Function for the Treatment of Ovarian Cancer
by Marco Greppi, Giovanna Tabellini, Ornella Patrizi, Simona Candiani, Andrea Decensi, Silvia Parolini, Simona Sivori, Silvia Pesce, Laura Paleari and Emanuela Marcenaro
Int. J. Mol. Sci. 2019, 20(4), 890; https://doi.org/10.3390/ijms20040890 - 19 Feb 2019
Cited by 31 | Viewed by 4958
Abstract
The crosstalk between cancer cells and host cells is a crucial prerequisite for tumor growth and progression. The cells from both the innate and adaptive immune systems enter into a perverse relationship with tumor cells to create a tumor-promoting and immunosuppressive tumor microenvironment [...] Read more.
The crosstalk between cancer cells and host cells is a crucial prerequisite for tumor growth and progression. The cells from both the innate and adaptive immune systems enter into a perverse relationship with tumor cells to create a tumor-promoting and immunosuppressive tumor microenvironment (TME). Epithelial ovarian cancer (EOC), the most lethal of all gynecological malignancies, is characterized by a unique TME that paves the way to the formation of metastasis and mediates therapy resistance through the deregulation of immune surveillance. A characteristic feature of the ovarian cancer TME is the ascites/peritoneal fluid, a malignancy-associated effusion occurring at more advanced stages, which enables the peritoneal dissemination of tumor cells and the formation of metastasis. The standard therapy for EOC involves a combination of debulking surgery and platinum-based chemotherapy. However, most patients experience disease recurrence. New therapeutic strategies are needed to improve the prognosis of patients with advanced EOC. Harnessing the body’s natural immune defenses against cancer in the form of immunotherapy is emerging as an innovative treatment strategy. NK cells have attracted attention as a promising cancer immunotherapeutic target due to their ability to kill malignant cells and avoid healthy cells. Here, we will discuss the recent advances in the clinical application of NK cell immunotherapy in EOC. Full article
Show Figures

Graphical abstract

14 pages, 3529 KiB  
Review
Interstitial Fluid in Gynecologic Tumors and Its Possible Application in the Clinical Practice
by Blendi Ura, Giovanni Di Lorenzo, Federico Romano, Lorenzo Monasta, Giuseppe Mirenda, Federica Scrimin and Giuseppe Ricci
Int. J. Mol. Sci. 2018, 19(12), 4018; https://doi.org/10.3390/ijms19124018 - 12 Dec 2018
Cited by 9 | Viewed by 4336
Abstract
Gynecologic cancers are an important cause of worldwide mortality. The interstitium consists of solid and fluid phases, situated between the blood vessels and cells. The interstitial fluid (IF), or fluid phase, is an extracellular fluid bathing and surrounding the tissue cells. The TIF [...] Read more.
Gynecologic cancers are an important cause of worldwide mortality. The interstitium consists of solid and fluid phases, situated between the blood vessels and cells. The interstitial fluid (IF), or fluid phase, is an extracellular fluid bathing and surrounding the tissue cells. The TIF (tumor interstitial fluid) is a dynamic fluid rich in lipids, proteins and enzyme-derived substances. The molecules found in the IF may be associated with pathological changes in tissues leading to cancer growth and metastatization. Proteomic techniques have allowed an extensive study of the composition of the TIF as a source of biomarkers for gynecologic cancers. In our review, we analyze the composition of the TIF, its formation process, the sampling methods, the consequences of its accumulation and the proteomic analyses performed, that make TIF valuable for monitoring different types of cancers. Full article
Show Figures

Figure 1

Other

Jump to: Research, Review

16 pages, 1974 KiB  
Perspective
S100A10 and Cancer Hallmarks: Structure, Functions, and its Emerging Role in Ovarian Cancer
by Tannith M. Noye, Noor A. Lokman, Martin K. Oehler and Carmela Ricciardelli
Int. J. Mol. Sci. 2018, 19(12), 4122; https://doi.org/10.3390/ijms19124122 - 19 Dec 2018
Cited by 30 | Viewed by 5529
Abstract
S100A10, which is also known as p11, is located in the plasma membrane and forms a heterotetramer with annexin A2. The heterotetramer, comprising of two subunits of annexin A2 and S100A10, activates the plasminogen activation pathway, which is involved in cellular repair of [...] Read more.
S100A10, which is also known as p11, is located in the plasma membrane and forms a heterotetramer with annexin A2. The heterotetramer, comprising of two subunits of annexin A2 and S100A10, activates the plasminogen activation pathway, which is involved in cellular repair of normal tissues. Increased expression of annexin A2 and S100A10 in cancer cells leads to increased levels of plasmin—which promotes the degradation of the extracellular matrix—increased angiogenesis, and the invasion of the surrounding organs. Although many studies have investigated the functional role of annexin A2 in cancer cells, including ovarian cancer, S100A10 has been less studied. We recently demonstrated that high stromal annexin A2 and high cytoplasmic S100A10 expression is associated with a 3.4-fold increased risk of progression and 7.9-fold risk of death in ovarian cancer patients. Other studies have linked S100A10 with multidrug resistance in ovarian cancer; however, no functional studies to date have been performed in ovarian cancer cells. This article reviews the current understanding of S100A10 function in cancer with a particular focus on ovarian cancer. Full article
Show Figures

Figure 1

Back to TopTop