1
|
Siskin M, Economides MP, Wise DR. Cyclin-Dependent Kinase Inhibition in Prostate Cancer: Past, Present, and Future. Cancers (Basel) 2025; 17:774. [PMID: 40075623 PMCID: PMC11898528 DOI: 10.3390/cancers17050774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Despite significant progress, prostate cancer remains a leading cause of death. Cyclin-dependent kinase (CDK) 4/6 inhibitors, which are already approved for the treatment of hormone receptor-positive breast cancer, are undergoing extensive testing as monotherapy and in various combinations as a potentially valuable treatment modality in prostate cancer patients. Thus far, a limited number of these studies have published results, which have been largely disappointing. AREAS COVERED In this review, we describe the biologic rationale for the use of CDK4/6 inhibitors in prostate cancer, the existing clinical data describing their use in prostate cancer, and ongoing clinical trials of CDK4/6 inhibitors as monotherapy and in combination for the treatment of prostate cancer. In particular, we focus on possible resistance mechanisms that may be particularly relevant in prostate cancer patients, leading to de novo and acquired resistance, and we highlight novel strategies that can overcome this resistance. CONCLUSIONS Current clinical trials are actively working to (1) refine the role of CDK4/6 inhibitors in prostate cancer patients; (2) develop new inhibitors of other cell-cycle targets, such as CDK2 and CDK7; and (3) explore novel combination therapies with inhibitors of other relevant pathways, such as PI3K or MAPK. Further genomic subtyping of advanced prostate cancer will likely shed light on the subsets of patients most likely to benefit from cell-cycle-targeted agents.
Collapse
Affiliation(s)
| | | | - David R. Wise
- Genitourinary Medical Oncology Service, Perlmutter Cancer Center, NYU Langone Heath Center, New York, NY 10016, USA; (M.S.); (M.P.E.)
| |
Collapse
|
2
|
Fu F, Yu Y, Zou B, Long Y, Wu L, Yin J, Zhou Q. Role of actin-binding proteins in prostate cancer. Front Cell Dev Biol 2024; 12:1430386. [PMID: 39055653 PMCID: PMC11269120 DOI: 10.3389/fcell.2024.1430386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
The molecular mechanisms driving the onset and metastasis of prostate cancer remain poorly understood. Actin, under the control of actin-binding proteins (ABPs), plays a crucial role in shaping the cellular cytoskeleton, which in turn supports the morphological alterations in normal cells, as well as the invasive spread of tumor cells. Previous research indicates that ABPs of various types serve distinct functions, and any disruptions in their activities could predispose individuals to prostate cancer. These ABPs are intricately implicated in the initiation and advancement of prostate cancer through a complex array of intracellular processes, such as severing, linking, nucleating, inducing branching, assembling, facilitating actin filament elongation, terminating elongation, and promoting actin molecule aggregation. As such, this review synthesizes existing literature on several ABPs linked to prostate cancer, including cofilin, filamin A, and fascin, with the aim of shedding light on the molecular mechanisms through which ABPs influence prostate cancer development and identifying potential therapeutic targets. Ultimately, this comprehensive examination seeks to contribute to the understanding and management of prostate diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qing Zhou
- Department of Andrology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
3
|
Giovannelli P, Di Donato M, Licitra F, Sabbatino E, Tutino V, Castoria G, Migliaccio A. Filamin A in triple negative breast cancer. Steroids 2024; 205:109380. [PMID: 38311094 DOI: 10.1016/j.steroids.2024.109380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/06/2024]
Abstract
Triple-negative breast cancer is a rare but highly heterogeneous breast cancer subtype with a limited choice of specific treatments. Chemotherapy remains the only efficient treatment, but its side effects and the development of resistance consolidate the urgent need to discover new targets. In TNBC, filamin A expression correlates to grade and TNM stage. Accordingly, this protein could constitute a new target for this BC subtype. Even if most of the data indicates its direct involvement in cancer progression, some contrasting results underline the need to deepen the studies. To elucidate a possible function of this protein as a TNBC marker, we summarized the main characteristic of filamin A and its involvement in physiological and pathological processes such as cancer. Lastly, we scrutinized its actions in triple-negative breast cancer and highlighted the need to increase the number of studies useful to better clarify the role of this versatile protein as a marker and target in TNBC, alone or in "collaboration" with other proteins with a relevant role in this BC subgroup.
Collapse
Affiliation(s)
- Pia Giovannelli
- Department of Precision Medicine, University of Campania "L.Vanvitelli", Via L. De Crecchio, 7-80138 Naples, Italy.
| | - Marzia Di Donato
- Department of Precision Medicine, University of Campania "L.Vanvitelli", Via L. De Crecchio, 7-80138 Naples, Italy
| | - Fabrizio Licitra
- Department of Precision Medicine, University of Campania "L.Vanvitelli", Via L. De Crecchio, 7-80138 Naples, Italy
| | - Emilia Sabbatino
- Department of Precision Medicine, University of Campania "L.Vanvitelli", Via L. De Crecchio, 7-80138 Naples, Italy
| | - Viviana Tutino
- Department of Precision Medicine, University of Campania "L.Vanvitelli", Via L. De Crecchio, 7-80138 Naples, Italy
| | - Gabriella Castoria
- Department of Precision Medicine, University of Campania "L.Vanvitelli", Via L. De Crecchio, 7-80138 Naples, Italy
| | - Antimo Migliaccio
- Department of Precision Medicine, University of Campania "L.Vanvitelli", Via L. De Crecchio, 7-80138 Naples, Italy
| |
Collapse
|
4
|
Liang K, Ke Z, Huang J, Zhang X. Expression and clinical value of NLRP1 and NLRC4 inflammasomes in prostate cancer. Oncol Lett 2023; 26:385. [PMID: 37559581 PMCID: PMC10407840 DOI: 10.3892/ol.2023.13971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/08/2023] [Indexed: 08/11/2023] Open
Abstract
The present study explored the clinical value of the protein expression levels of nucleotide binding oligomerization-like receptor family pyrin domain containing 1 (NLRP1) and nucleotide-binding oligomerization domain leucine-rich repeat and caspase recruitment domain-containing 4 (NLRC4) inflammasomes in the diagnosis and treatment of prostate cancer. A total of 54 patients with prostatic hyperplasia and 58 patients with prostate cancer were recruited at The First People's Hospital of Pinghu between January and May 2022. Immunohistochemical staining was used to determine the protein expression levels of the NLRP1 and NLRC4 inflammasomes in addition to the proinflammatory cytokines IL-18 and IL-1β in the two groups of patients. The protein expression levels of NLRP1 and NLRC4 inflammasome were significantly increased in patients with prostate cancer compared with patients with prostate hyperplasia. The differences in expression of NLRP1 and NLRC4 inflammatory vesicles in prostate cancer of different stages were also compared based on data from The Cancer Genome Atlas. The protein expression level of NLRP1 demonstrated a significant positive correlation with IL-1β and IL-18 expression, and the protein expression level of the NLRC4 inflammasome was significantly positively correlated with IL-18 expression. The protein expression levels of both NLRP1 and NLRC4 demonstrated a significant positive correlation with the Gleason score of prostate cancer. The expression of NLRP1 in tumor (T)3/T4 was significantly higher compared with T1 and expression of the NLRC4 inflammasome in T2 and T3/T4 was significantly higher compared with T1. Expression of the NLRP1 and NLRC4 inflammasomes was significantly higher in patients with prostate cancer, compared with patients with prostatic hyperplasia. Therefore, expression of NLRP1 and NLRC4 may promote tumorigenesis by promoting the maturation and release of proinflammatory cytokines IL-1β and IL-18. Expression of the NLRP1 and NLRC4 inflammasomes demonstrated a significant positive correlation with the risk of prostate cancer. Expression of the NLRP1 and NLRC4 inflammasomes in middle- and advanced-stage tumors was higher compared with early-stage tumors. These results suggested that inflammasome expression may serve a significant role in the progression of tumors and could provide a fixed value for the risk assessment and prognosis prediction of prostate cancer.
Collapse
Affiliation(s)
- Ke Liang
- Department of Urology, The First People's Hospital of Pinghu, Pinghu, Zhejiang 314200, P.R. China
| | - Zunjin Ke
- Department of Urology, The First People's Hospital of Pinghu, Pinghu, Zhejiang 314200, P.R. China
| | - Jianhong Huang
- Department of Urology, The First People's Hospital of Pinghu, Pinghu, Zhejiang 314200, P.R. China
| | - Xijiong Zhang
- Department of Pathology, The First People's Hospital of Pinghu, Pinghu, Zhejiang 314200, P.R. China
| |
Collapse
|
5
|
Di Donato M, Giovannelli P, Migliaccio A, Castoria G. The nerve growth factor-delivered signals in prostate cancer and its associated microenvironment: when the dialogue replaces the monologue. Cell Biosci 2023; 13:60. [PMID: 36941697 PMCID: PMC10029315 DOI: 10.1186/s13578-023-01008-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 03/06/2023] [Indexed: 03/22/2023] Open
Abstract
Prostate cancer (PC) represents the most diagnosed and the second most lethal cancer in men worldwide. Its development and progression occur in concert with alterations in the surrounding tumor microenvironment (TME), made up of stromal cells and extracellular matrix (ECM) that dynamically interact with epithelial PC cells affecting their growth and invasiveness. PC cells, in turn, can functionally sculpt the TME through the secretion of various factors, including neurotrophins. Among them, the nerve growth factor (NGF) that is released by both epithelial PC cells and carcinoma-associated fibroblasts (CAFs) triggers the activation of various intracellular signaling cascades, thereby promoting the acquisition of a metastatic phenotype. After many years of investigation, it is indeed well established that aberrations and/or derangement of NGF signaling are involved not only in neurological disorders, but also in the pathogenesis of human proliferative diseases, including PC. Another key feature of cancer progression is the nerve outgrowth in TME and the concept of nerve dependence related to perineural invasion is currently emerging. NGF released by cancer cells can be a driver of tumor neurogenesis and nerves infiltrated in TME release neurotransmitters, which might stimulate the growth and sustainment of tumor cells.In this review, we aim to provide a snapshot of NGF action in the interactions between TME, nerves and PC cells. Understanding the molecular basis of this dialogue might expand the arsenal of therapeutic strategies against this widespread disease.
Collapse
Affiliation(s)
- Marzia Di Donato
- Department of Precision Medicine, University of Campania "L.Vanvitelli", 80138, Naples, Italy.
| | - Pia Giovannelli
- Department of Precision Medicine, University of Campania "L.Vanvitelli", 80138, Naples, Italy.
| | - Antimo Migliaccio
- Department of Precision Medicine, University of Campania "L.Vanvitelli", 80138, Naples, Italy
| | - Gabriella Castoria
- Department of Precision Medicine, University of Campania "L.Vanvitelli", 80138, Naples, Italy
| |
Collapse
|
6
|
Abikar A, Saimon C, Ranganathan P. To block or not to block-hormonal signaling in the treatment of cancers. Front Endocrinol (Lausanne) 2023; 14:1129332. [PMID: 36891053 PMCID: PMC9986485 DOI: 10.3389/fendo.2023.1129332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023] Open
Abstract
The breast and prostate glands are the two major organs that are highly dependent on the gonadal steroid hormones for their development and homeostasis. The cancers of these organs also show a large dependence on steroid hormones and have formed the basis of endocrine therapy. Estrogen deprivation by oophorectomy has been in active practice since the 1970s, and androgen deprivation therapy for prostate cancer was a major breakthrough in medicine in 1941. Since then, several improvisations have happened in these modes of therapy. However, the development of resistance to this deprivation and the emergence of hormone independence are major problems in both cancers. The lessons learned from rodent models have made it clear that the male hormone has a role in females and vice versa. Also, the metabolic products of these hormones may have unintentional effects including proliferative conditions in both sexes. Hence, administering estrogen as a method of chemical castration in males and administering DHT in females may not be the ideal scenario. It would be important to consider the status of the opposite sex hormone signaling and its effects and come up with a combinatorial regime to strike a balance between androgen and estrogen signaling. This review summarizes the current understanding and developments in this field in the context of prostate cancer.
Collapse
|
7
|
Di Donato M, Giovannelli P, Migliaccio A, Bilancio A. Inhibition of Vps34 and p110δ PI3K Impairs Migration, Invasion and Three-Dimensional Spheroid Growth in Breast Cancer Cells. Int J Mol Sci 2022; 23:9008. [PMID: 36012280 PMCID: PMC9409264 DOI: 10.3390/ijms23169008] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/07/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Breast cancer is a heterogeneous disease that represents the most common cancer around the world; it comprises 12% of new cases according to the World Health Organization. Despite new approaches in early diagnosis and current treatment, breast cancer is still the leading cause of death for cancer mortality. New targeted therapies against key signalling transduction molecules are required. Phosphoinositide 3-kinase (PI3K) regulates multiple biological functions such as proliferation, survival, migration, and growth. It is well established that PI3K isoform-selective inhibitors show fewer toxic side effects compared to broad spectrum inhibition of PI3K (pan-PI3K inhibitors). Therefore, we tested the PI3K p110δ-selective inhibitor, IC87114, and Vps34-selective inhibitor, Vps34-IN1, on the breast cancer cell lines MCF-7 and MDA-MB-231, representing hormone-responsive and triple-negative breast cancer cells, respectively. Our data show that both inhibitors decreased migration of MCF-7 and MDA-MB-231 cells, and Vps34 also significantly impacted MCF-7 cell proliferation. Three-dimensional (3D) in vitro culture models show that IC87114 and Vps34-IN1 treatment reduced the growth of MCF-7 and MDA-MB-231 cells in 3D tumour spheroid cultures. This study identifies IC87114 and Vps34-IN1 as potential therapeutic approaches in breast cancer.
Collapse
Affiliation(s)
| | | | | | - Antonio Bilancio
- Department of Medicine Precision, “Luigi Vanvitelli”, Affiliation University of Campania, Via L. De Crecchio 7, 80138 Naples, Italy
| |
Collapse
|
8
|
Relevance of Emerging Metabolomics-Based Biomarkers of Prostate Cancer: A Systematic Review. Expert Rev Mol Med 2022; 24:e25. [PMID: 35730322 DOI: 10.1017/erm.2022.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
9
|
Pollution Characteristics and Risk Prediction of Endocrine Disruptors in Lakes of Wuhan. TOXICS 2022; 10:toxics10020093. [PMID: 35202278 PMCID: PMC8880694 DOI: 10.3390/toxics10020093] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 02/14/2022] [Indexed: 02/05/2023]
Abstract
As a new and ubiquitous trace organic pollutant, endocrine-disrupting compounds (EDCs) can cause endocrine-disrupting effects on organisms even at low levels. However, little information is available on the resource and assessment of EDC risks in the water environment. The study area was selected based on the paucity of information on the pollution status of inland lakes. Wuhan has numerous and diverse types of lakes which receive micropollutants from different pathways. In this study, the spatial distribution, occurrence, quantity and ecological risks of EDCs in 12 lakes were investigated. Five EDCs, including 17-alpha-ethinylestradiol (17α-EE2), estrone (E1), β-estradiol (β-E2), estriol (E3) and bisphenol A (BPA) were detected in surface waters. The distribution of EDC content in the lakes was ordered as follows: exurban zone < suburban area < urban areas. The pollution sources in remote lakes mainly included agricultural and aquaculture wastewater, while those in suburban and urban areas included domestic or industrial wastewater. Areas with higher EDC content were frequently related to agricultural activities, aquaculture water or dense populations. Water quality parameters, including dissolved oxygen, pH and water temperature, were significantly related to the occurrence and distribution of EDCs in the lakes. Risk assessment demonstrated that the occurrence of EDCs posed minimum to medium risk to aquatic organisms in the lakes. The results showed that the lakes faced a threat hormone pollution though it was at lower doses and, thus, the ecological risk of EDCs should be considered in future environmental policies and decisions in China.
Collapse
|
10
|
Rapid Androgen-Responsive Proteome Is Involved in Prostate Cancer Progression. Biomedicines 2021; 9:biomedicines9121877. [PMID: 34944692 PMCID: PMC8698566 DOI: 10.3390/biomedicines9121877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 01/20/2023] Open
Abstract
Androgen exerts its functions by binding with an androgen receptor (AR). It can activate many signaling pathways that are important to the progression of castration-resistant prostate cancer (CRPC). Here, we characterized the rapid proteomic changes seen at 5, 15, 30, and 60 min after the androgen treatment of VCaP cells via the tandem mass tag (TMT) labeling strategy. A total of 5529 proteins were successfully identified and quantified. Dynamic time profiling of protein expression patterns allowed us to identify five protein clusters involved in various stages of androgen-initiated signal transmission and processing. More details of protein functions and localization patterns, and our elucidation of an AR-interacting protein network, were obtained. Finally, we validated the expression level of AR-regulated proteins known to be significantly regulated in CRPC patients using the mouse xenograft model and patient samples. Our work offers a systematic analysis of the rapid proteomic changes induced by androgen and provides a global view of the molecular mechanisms underlying CRPC progression.
Collapse
|
11
|
Gevaert T, Van Eycke YR, Vanden Broeck T, Van Poppel H, Salmon I, Rorive S, Muilwijk T, Claessens F, De Ridder D, Joniau S, Decaestecker C. The potential of tumour microenvironment markers to stratify the risk of recurrence in prostate cancer patients. PLoS One 2020; 15:e0244663. [PMID: 33370412 PMCID: PMC7769484 DOI: 10.1371/journal.pone.0244663] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/15/2020] [Indexed: 12/21/2022] Open
Abstract
The tumour micro-environment (TME) plays a crucial role in the onset and progression of prostate cancer (PCa). Here we studied the potential of a selected panel of TME-markers to predict clinical recurrence (CLR) in PCa. Patient cohorts were matched for the presence or absence of CLR 5 years post-prostatectomy. Tissue micro-arrays (TMA) were composed with both prostate non-tumour (PNT) and PCa tissue and subsequently processed for immunohistochemistry (IHC). The IHC panel included markers for cancer activated fibroblasts (CAFs), blood vessels and steroid hormone receptors ((SHR): androgen receptor (AR), progesterone receptor (PR) and estrogen receptor (ER)). Stained slides were digitalised, selectively annotated and analysed for percentage of marker expression with standardized and validated image analysis algorithms. A univariable analysis identified several TME markers with significant impact on CR: expression of CD31 (vascular marker) in PNT stroma, expression of alpha smooth muscle actin (αSMA) in PCa stroma, and PR expression ratio between PCa stroma and PNT stroma. A multivariable model, which included CD31 expression (vascular marker) in PNT stroma and PR expression ratio between PCa stroma and PNT stroma, could significantly stratify patients for CLR, with the identification of a low risk and high-risk subgroup. If validated and confirmed in an independent prospective series, this subgroup might have clinical potential for PCa patient stratification.
Collapse
Affiliation(s)
- Thomas Gevaert
- Department of Urology, UZ Leuven, Leuven, Belgium
- Organ Systems, KU Leuven, Leuven, Belgium
- Department of Pathology, AZ Klina, Brasschaat, Belgium
- P.E.A.R.L. (ProstatE cAncer Research Leuven), Leuven, Belgium
| | - Yves-Rémi Van Eycke
- Laboratories of Image, Synthesis and Analysis (LISA), Brussels School of Engineering/École polytechnique de Bruxelles, ULB, Brussels, Belgium
- DIAPath-Center for Microscopy and Molecular Imaging (CMMI), ULB, Gosselies, Belgium
| | - Thomas Vanden Broeck
- Department of Urology, UZ Leuven, Leuven, Belgium
- P.E.A.R.L. (ProstatE cAncer Research Leuven), Leuven, Belgium
- Department of Molecular and Cellular Medicine, KU Leuven, Leuven, Belgium
| | - Hein Van Poppel
- Department of Urology, UZ Leuven, Leuven, Belgium
- Organ Systems, KU Leuven, Leuven, Belgium
- P.E.A.R.L. (ProstatE cAncer Research Leuven), Leuven, Belgium
| | - Isabelle Salmon
- DIAPath-Center for Microscopy and Molecular Imaging (CMMI), ULB, Gosselies, Belgium
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Centre Universitaire Inter Régional d'Expertise en Anatomie Pathologique Hospitalière (CurePath), Jumet, Belgium
| | - Sandrine Rorive
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Centre Universitaire Inter Régional d'Expertise en Anatomie Pathologique Hospitalière (CurePath), Jumet, Belgium
| | - Tim Muilwijk
- Department of Urology, UZ Leuven, Leuven, Belgium
- Organ Systems, KU Leuven, Leuven, Belgium
| | - Frank Claessens
- P.E.A.R.L. (ProstatE cAncer Research Leuven), Leuven, Belgium
- Department of Molecular and Cellular Medicine, KU Leuven, Leuven, Belgium
| | - Dirk De Ridder
- Department of Urology, UZ Leuven, Leuven, Belgium
- Organ Systems, KU Leuven, Leuven, Belgium
- P.E.A.R.L. (ProstatE cAncer Research Leuven), Leuven, Belgium
| | - Steven Joniau
- Department of Urology, UZ Leuven, Leuven, Belgium
- Organ Systems, KU Leuven, Leuven, Belgium
- P.E.A.R.L. (ProstatE cAncer Research Leuven), Leuven, Belgium
| | - Christine Decaestecker
- Laboratories of Image, Synthesis and Analysis (LISA), Brussels School of Engineering/École polytechnique de Bruxelles, ULB, Brussels, Belgium
- DIAPath-Center for Microscopy and Molecular Imaging (CMMI), ULB, Gosselies, Belgium
| |
Collapse
|
12
|
Kase AM, Copland III JA, Tan W. Novel Therapeutic Strategies for CDK4/6 Inhibitors in Metastatic Castrate-Resistant Prostate Cancer. Onco Targets Ther 2020; 13:10499-10513. [PMID: 33116629 PMCID: PMC7576355 DOI: 10.2147/ott.s266085] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/18/2020] [Indexed: 12/15/2022] Open
Abstract
The majority of patients with castrate-resistant prostate cancer will have metastatic disease at the time of diagnosis. Investigative efforts on new therapeutics for this patient population have improved with the development of androgen signaling inhibitors, such as abiraterone and enzalutamide, and PARP inhibitors, such as rucaparib and olaparib, to accompany the previously FDA-approved docetaxel, cabazitaxel, sipuleucel-T, and Radium 223. However, new therapeutic strategies are necessary to prolong survival as progression after these agents is inevitable. CDK4/6 inhibitors have advanced the field of estrogen receptor positive breast cancer treatment and are being investigated in prostate cancer given the role of androgen receptor signaling effects on the cell cycle. Response to CDK4/6 inhibitors may be predicted by the tumors' genomic profile and may provide insight into combinatory therapy with CDK4/6 inhibitors in order to delay resistance or provide synergistic effects. Here, we review the use of CDK4/6 inhibitors in prostate cancer and potential combinations based on known resistance mechanisms to CDK4/6 inhibitors, prostate cancer regulatory pathways, and prostate-cancer-specific genomic alterations.
Collapse
Affiliation(s)
- Adam M Kase
- Mayo Clinic Florida Division of Hematology Oncology, Jacksonville, FL32224, USA
| | - John A Copland III
- Mayo Clinic Florida Department of Cancer Biology, Jacksonville, FL32224, USA
| | - Winston Tan
- Mayo Clinic Florida Division of Hematology Oncology, Jacksonville, FL32224, USA
| |
Collapse
|
13
|
Domínguez-López I, Yago-Aragón M, Salas-Huetos A, Tresserra-Rimbau A, Hurtado-Barroso S. Effects of Dietary Phytoestrogens on Hormones throughout a Human Lifespan: A Review. Nutrients 2020; 12:E2456. [PMID: 32824177 PMCID: PMC7468963 DOI: 10.3390/nu12082456] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/31/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023] Open
Abstract
Dietary phytoestrogens are bioactive compounds with estrogenic activity. With the growing popularity of plant-based diets, the intake of phytoestrogen-rich legumes (especially soy) and legume-derived foods has increased. Evidence from preclinical studies suggests these compounds may have an effect on hormones and health, although the results of human trials are unclear. The effects of dietary phytoestrogens depend on the exposure (phytoestrogen type, matrix, concentration, and bioavailability), ethnicity, hormone levels (related to age, sex, and physiological condition), and health status of the consumer. In this review, we have summarized the results of human studies on dietary phytoestrogens with the aim of assessing the possible hormone-dependent outcomes and health effects of their consumption throughout a lifespan, focusing on pregnancy, childhood, adulthood, and the premenopausal and postmenopausal stages. In pregnant women, an improvement of insulin metabolism has been reported in only one study. Sex hormone alterations have been found in the late stages of childhood, and goitrogenic effects in children with hypothyroidism. In premenopausal and postmenopausal women, the reported impacts on hormones are inconsistent, although beneficial goitrogenic effects and improved glycemic control and cardiovascular risk markers have been described in postmenopausal individuals. In adult men, different authors report goitrogenic effects and a reduction of insulin in non-alcoholic fatty liver patients. Further carefully designed studies are warranted to better elucidate the impact of phytoestrogen consumption on the endocrine system at different life stages.
Collapse
Affiliation(s)
- Inés Domínguez-López
- Department of Nutrition, Food Science and Gastronomy, XaRTA, INSA, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (I.D.-L.); (M.Y.-A.); (S.H.-B.)
| | - Maria Yago-Aragón
- Department of Nutrition, Food Science and Gastronomy, XaRTA, INSA, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (I.D.-L.); (M.Y.-A.); (S.H.-B.)
| | - Albert Salas-Huetos
- Andrology and IVF Laboratory, Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT 84108, USA;
| | - Anna Tresserra-Rimbau
- Department of Nutrition, Food Science and Gastronomy, XaRTA, INSA, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (I.D.-L.); (M.Y.-A.); (S.H.-B.)
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unitat de Nutrició, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, 43204 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43201 Reus, Spain
| | - Sara Hurtado-Barroso
- Department of Nutrition, Food Science and Gastronomy, XaRTA, INSA, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (I.D.-L.); (M.Y.-A.); (S.H.-B.)
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
14
|
Stromal Cell Signature Associated with Response to Neoadjuvant Chemotherapy in Locally Advanced Breast Cancer. Cells 2019; 8:cells8121566. [PMID: 31817155 PMCID: PMC6953077 DOI: 10.3390/cells8121566] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/27/2019] [Accepted: 10/31/2019] [Indexed: 01/26/2023] Open
Abstract
Breast cancer stromal compartment, may influence responsiveness to chemotherapy. Our aim was to detect a stromal cell signature (using a direct approach of microdissected stromal cells) associated with response to neoadjuvant chemotherapy (neoCT) in locally advanced breast cancer (LABC). The tumor samples were collected from 44 patients with LABC (29 estrogen receptor (ER) positive and 15 ER negative) before the start of any treatment. Neoadjuvant chemotherapy consisted of doxorubicin and cyclophosphamide, followed by paclitaxel. Response was defined as downstaging to maximum ypT1a-b/ypN0. The stromal cells, mainly composed of fibroblast and immune cells, were microdissected from fresh frozen tumor samples and gene expression profile was determined using Agilent SurePrint G3 Human Gene Expression microarrays. Expression levels were compared using MeV (MultiExperiment Viewer) software, applying SAM (significance analysis of microarrays). To classify samples according to tumor response, the order of median based on confidence statements (MedOr) was used, and to identify gene sets correlated with the phenotype downstaging, gene set enrichment analysis (GSEA). Nine patients presented disease downstaging. Eleven sequences (FDR 17) were differentially expressed, all of which (except H2AFJ) more expressed in responsive tumors, including PTCHD1 and genes involved in abnormal cytotoxic T cell physiology, TOX, LY75, and SH2D1A. The following four pairs of markers could correctly classify all tumor samples according to response: PTCHD1/PDXDC2P, LOC100506731/NEURL4, SH2D1A/ENST00000478672, and TOX/H2AFJ. Gene sets correlated with tumor downstaging (FDR < 0.01) were mainly involved in immune response or lymphocyte activation, including CD47, LCK, NCK1, CD24, CD3E, ZAP70, FOXP3, and CD74, among others. In locally advanced breast cancer, stromal cells may present specific features of immune response that may be associated with chemotherapy response.
Collapse
|
15
|
Giovannelli P, Di Donato M, Galasso G, Di Zazzo E, Medici N, Bilancio A, Migliaccio A, Castoria G. Breast cancer stem cells: The role of sex steroid receptors. World J Stem Cells 2019; 11:594-603. [PMID: 31616537 PMCID: PMC6789191 DOI: 10.4252/wjsc.v11.i9.594] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/06/2019] [Accepted: 08/21/2019] [Indexed: 02/06/2023] Open
Abstract
Breast cancer (BC) is the most common cancer among women, and current available therapies often have high success rates. Nevertheless, BC might acquire drug resistance and sometimes relapse. Current knowledge about the most aggressive forms of BC points to the role of specific cells with stem properties located within BC, the so-called “BC stem cells” (BCSCs). The role of BCSCs in cancer formation, growth, invasiveness, therapy resistance and tumor recurrence is becoming increasingly clear. The growth and metastatic properties of BCSCs are regulated by different pathways, which are only partially known. Sex steroid receptors (SSRs), which are involved in BC etiology and progression, promote BCSC proliferation, dedifferentiation and migration. However, in the literature, there is incomplete information about their roles. Particularly, there are contrasting conclusions about the expression and role of the classical BC hormonal biomarkers, such as estrogen receptor alpha (ERα), together with scant, albeit promising information concerning ER beta (ERβ) and androgen receptor (AR) properties that control different transduction pathways in BCSCs. In this review, we will discuss the role that SRs expressed in BCSCs play to BC progression and recurrence and how these findings have opened new therapeutic possibilities.
Collapse
Affiliation(s)
- Pia Giovannelli
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Naples 80138, Italy
| | - Marzia Di Donato
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Naples 80138, Italy
| | - Giovanni Galasso
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Naples 80138, Italy
| | - Erika Di Zazzo
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Naples 80138, Italy
| | - Nicola Medici
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Naples 80138, Italy
| | - Antonio Bilancio
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Naples 80138, Italy
| | - Antimo Migliaccio
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Naples 80138, Italy
| | - Gabriella Castoria
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Naples 80138, Italy
| |
Collapse
|
16
|
A Rich Array of Prostate Cancer Molecular Biomarkers: Opportunities and Challenges. Int J Mol Sci 2019; 20:ijms20081813. [PMID: 31013716 PMCID: PMC6515282 DOI: 10.3390/ijms20081813] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 01/30/2023] Open
Abstract
Prostate cancer is the most prevalent non-skin cancer in men and is the leading cause of cancer-related death. Early detection of prostate cancer is largely determined by a widely used prostate specific antigen (PSA) blood test and biopsy is performed for definitive diagnosis. Prostate cancer is asymptomatic in the early stage of the disease, comprises of diverse clinico-pathologic and progression features, and is characterized by a large subset of the indolent cancer type. Therefore, it is critical to develop an individualized approach for early detection, disease stratification (indolent vs. aggressive), and prediction of treatment response for prostate cancer. There has been remarkable progress in prostate cancer biomarker discovery, largely through advancements in genomic technologies. A rich array of prostate cancer diagnostic and prognostic tests has emerged for serum (4K, phi), urine (Progensa, T2-ERG, ExoDx, SelectMDx), and tumor tissue (ConfirmMDx, Prolaris, Oncoytype DX, Decipher). The development of these assays has created new opportunities for improving prostate cancer diagnosis, prognosis, and treatment decisions. While opening exciting opportunities, these developments also pose unique challenges in terms of selecting and incorporating these assays into the continuum of prostate cancer patient care.
Collapse
|
17
|
Park SR, Cho A, Kim JW, Lee HY, Hong IS. A Novel Endogenous Damage Signal, CSF-2, Activates Multiple Beneficial Functions of Adipose Tissue-Derived Mesenchymal Stem Cells. Mol Ther 2019; 27:1087-1100. [PMID: 30962162 DOI: 10.1016/j.ymthe.2019.03.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 03/08/2019] [Accepted: 03/12/2019] [Indexed: 12/15/2022] Open
Abstract
The major challenges of current mesenchymal stem cell (MSC)-based therapeutics are their low differentiation potential into specialized cell types and their homing ability to sites of injury. Therefore, many researchers have directed their efforts toward finding a novel stimulatory factor that can significantly enhance the therapeutic effects of MSCs. Colony-stimulating factor 2 (CSF-2) is previously known as a hematopoietic growth factor involved in the differentiation of various myeloid cells from hematopoietic progenitor cells. In addition to this canonical hematopoietic function, we identified for the first time that CSF-2 is actively secreted by stem cells, in response to various types of injuries, as an endogenous damage signal that promotes the therapeutic effects of MSCs by enhancing their multi-lineage differentiation and migratory capacities, possibly through its receptor CD116. Our results also revealed that CSF-2 exerts its stimulatory effects on MSCs via PI3K/Akt- and/or FAK/ERK1/2-signaling pathways. More importantly, we also found that MSCs stimulated with CSF-2 show markedly enhanced differentiation and migratory capacities and subsequent in vivo therapeutic effects in an endometrial ablation animal model. Collectively, our findings provide compelling evidence for a novel non-hematopoietic function of CSF-2 in promoting multiple beneficial functions of MSCs via a non-canonical mechanism as an endogenous damage signal.
Collapse
Affiliation(s)
- Se-Ra Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea; Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| | - Ara Cho
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea; Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| | - Jae-Wan Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea; Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| | - Hwa-Yong Lee
- Department of Biomedical Science, Jungwon University, 85 Goesan-eup, Munmu-ro, Goesan-gun, Chungcheongbuk-do 367-700, Republic of Korea.
| | - In-Sun Hong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea; Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea.
| |
Collapse
|
18
|
Kdadra M, Höckner S, Leung H, Kremer W, Schiffer E. Metabolomics Biomarkers of Prostate Cancer: A Systematic Review. Diagnostics (Basel) 2019; 9:E21. [PMID: 30791464 PMCID: PMC6468767 DOI: 10.3390/diagnostics9010021] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 12/27/2022] Open
Abstract
Prostate cancer (PCa) diagnosis with current biomarkers is difficult and often results in unnecessary invasive procedures as well as over-diagnosis and over-treatment, highlighting the need for novel biomarkers. The aim of this review is to provide a summary of available metabolomics PCa biomarkers, particularly for clinically significant disease. A systematic search was conducted on PubMed for publications from July 2008 to July 2018 in accordance with PRISMA guidelines to report biomarkers with respect to their application in PCa diagnosis, progression, aggressiveness, recurrence, and treatment response. The vast majority of studies report biomarkers with the ability to distinguish malignant from benign prostate tissue with a few studies investigating biomarkers associated with disease progression, treatment response or tumour recurrence. In general, these studies report high dimensional datasets and the number of analysed metabolites often significantly exceeded the number of available samples. Hence, observed multivariate differences between case and control samples in the datasets might potentially also be associated with pre-analytical, technical, statistical and confounding factors. Giving the technical and methodological hurdles, there are nevertheless a number of metabolites and pathways repeatedly reported across various technical approaches, cohorts and sample types that appear to play a predominant role in PCa tumour biology, progression and recurrence.
Collapse
Affiliation(s)
| | | | - Hing Leung
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK.
- CRUK Beatson Institute, Bearsden, Glasgow G61 1BD, UK.
| | - Werner Kremer
- Institute of Biophysics and Physical Biochemistry, University of Regensburg, 93053 Regensburg, Germany.
| | - Eric Schiffer
- Numares AG, Am BioPark 9, 93053 Regensburg, Germany.
| |
Collapse
|
19
|
Rossi V, Di Zazzo E, Galasso G, De Rosa C, Abbondanza C, Sinisi AA, Altucci L, Migliaccio A, Castoria G. Estrogens Modulate Somatostatin Receptors Expression and Synergize With the Somatostatin Analog Pasireotide in Prostate Cells. Front Pharmacol 2019; 10:28. [PMID: 30828298 PMCID: PMC6384260 DOI: 10.3389/fphar.2019.00028] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 01/11/2019] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer (PC) is one of the most frequently diagnosed cancers and a leading cause of cancer-related deaths in Western society. Current PC therapies prevalently target the functions of androgen receptor (AR) and may only be effective within short time periods, beyond which the majority of PC patients progress to castration-resistant PC (CRPC) and metastatic disease. The role of estradiol/estradiol receptor (ER) axis in prostate transformation and PC progression is well established. Further, considerable efforts have been made to investigate the mechanism by which somatostatin (SST) and somatostatin receptors (SSTRs) influence PC growth and progression. A number of therapeutic strategies, such as the combination of SST analogs with other drugs, show, indeed, strong promise. However, the effect of the combined treatment of SST analogs and estradiol on proliferation, epithelial mesenchyme transition (EMT) and migration of normal- and cancer-derived prostate cells has not been investigated so far. We now report that estradiol plays anti-proliferative and pro-apoptotic effect in non-transformed EPN prostate cells, which express both ERα and ERβ. A weak apoptotic effect is observed in transformed CPEC cells that only express low levels of ERβ. Estradiol increases, mainly through ERα activation, the expression of SSTRs in EPN, but not CPEC cells. As such, the hormone enhances the anti-proliferative effect of the SST analog, pasireotide in EPN, but not CPEC cells. Estradiol does not induce EMT and the motility of EPN cells, while it promotes EMT and migration of CPEC cells. Addition of pasireotide does not significantly modify these responses. Altogether, our results suggest that pasireotide may be used, alone or in combination with other drugs, to limit the growth of prostate proliferative diseases, provided that both ER isoforms (α and β) are present. Further investigations are needed to better define the cross talk between estrogens and SSTRs as well as its role in PC.
Collapse
Affiliation(s)
- Valentina Rossi
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Erika Di Zazzo
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Giovanni Galasso
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Caterina De Rosa
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Ciro Abbondanza
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Antonio A. Sinisi
- Dipartimento di Scienze Mediche, Chirurgiche, Neurologiche, Metaboliche e dell’Invecchiamento, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Lucia Altucci
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Antimo Migliaccio
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Gabriella Castoria
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| |
Collapse
|
20
|
Rodríguez-Lozano DC, Piña-Medina AG, Hansberg-Pastor V, Bello-Alvarez C, Camacho-Arroyo I. Testosterone Promotes Glioblastoma Cell Proliferation, Migration, and Invasion Through Androgen Receptor Activation. Front Endocrinol (Lausanne) 2019; 10:16. [PMID: 30778332 PMCID: PMC6369181 DOI: 10.3389/fendo.2019.00016] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/10/2019] [Indexed: 01/04/2023] Open
Abstract
Glioblastomas (GBM) are the most frequent and aggressive human brain tumors due to their high capacity to migrate and invade normal brain tissue. Epidemiological data report that GBM occur in a greater proportion in men than in women (3:2), suggesting the participation of sex hormones in the development of these tumors. It has been reported an increase in testosterone (T) levels in patients with GBM. In addition, androgen receptor (AR) is overexpressed in human GBM, and genetic silencing of AR, and its pharmacological inhibition, induce GBM cell death in vivo and in vitro. However, the role of T in proliferation, migration and invasion in human GBM cell lines has not been evaluated. We observed that T increased the number of U87, U251, and D54 cells derived from human GBM due to an increase in cell proliferation. This induction was blocked with flutamide, an antagonist of AR. T also induced migration and invasion of GBM cells that flutamide partially blocked. These data suggest that T through AR contributes to the progression of GBM by promoting proliferation, migration, and invasion.
Collapse
Affiliation(s)
- Dulce Carolina Rodríguez-Lozano
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Ana Gabriela Piña-Medina
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Claudia Bello-Alvarez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|
21
|
Anti-Cancer Effects of Green Tea Polyphenols Against Prostate Cancer. Molecules 2019; 24:molecules24010193. [PMID: 30621039 PMCID: PMC6337309 DOI: 10.3390/molecules24010193] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/03/2019] [Accepted: 01/03/2019] [Indexed: 12/17/2022] Open
Abstract
Prostate cancer is the most common cancer among men. Green tea consumption is reported to play an important role in the prevention of carcinogenesis in many types of malignancies, including prostate cancer; however, epidemiological studies show conflicting results regarding these anti-cancer effects. In recent years, in addition to prevention, many investigators have shown the efficacy and safety of green tea polyphenols and combination therapies with green tea extracts and anti-cancer agents in in vivo and in vitro studies. Furthermore, numerous studies have revealed the molecular mechanisms of the anti-cancer effects of green tea extracts. We believe that improved understanding of the detailed pathological roles at the molecular level is important to evaluate the prevention and treatment of prostate cancer. Therefore, in this review, we present current knowledge regarding the anti-cancer effects of green tea extracts in the prevention and treatment of prostate cancer, with a particular focus on the molecular mechanisms of action, such as influencing tumor growth, apoptosis, androgen receptor signaling, cell cycle, and various malignant behaviors. Finally, the future direction for the use of green tea extracts as treatment strategies in patients with prostate cancer is introduced.
Collapse
|
22
|
IL-8 Secreted from M2 Macrophages Promoted Prostate Tumorigenesis via STAT3/MALAT1 Pathway. Int J Mol Sci 2018; 20:ijms20010098. [PMID: 30591689 PMCID: PMC6337597 DOI: 10.3390/ijms20010098] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/21/2022] Open
Abstract
Prostate cancer (PCa) is a major health problem in males. Metastasis-associated with lung adenocarcinoma transcript-1 (MALAT1), which is overexpressed in PCa tissue, is associated with physiological and pathological conditions of PCa. M2 macrophages are major immune cells abundant in the tumor microenvironment. However, it remains unknown whether M2 macrophages are involved in the effects or not, and molecular mechanisms of MALAT1 on PCa progression have not yet been comprehensively explored. Here we reported that, M2 macrophages (PMA/IL-4 treated THP1) induced MALAT1 expression in PCa cell lines. Knockdown MALAT1 expression level in PCa cell lines inhibited cellular proliferation, invasion, and tumor formation. Further mechanistic dissection revealed that M2 macrophages secreted IL-8 was sufficient to drive up MALAT1 expression level via activating STAT3 signaling pathway. Additional chromatin immunoprecipitation (ChIP) and luciferase reporter assays displayed that STAT3 could bind to the MALAT1 promoter region and transcriptionally stimulate the MALAT1 expression. In summary, our present study identified the IL-8/STAT3/MALAT1 axis as key regulators during prostate tumorigenesis and therefore demonstrated a new mechanism for the MALAT1 transcriptional regulation.
Collapse
|
23
|
Bleach R, McIlroy M. The Divergent Function of Androgen Receptor in Breast Cancer; Analysis of Steroid Mediators and Tumor Intracrinology. Front Endocrinol (Lausanne) 2018; 9:594. [PMID: 30416486 PMCID: PMC6213369 DOI: 10.3389/fendo.2018.00594] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 09/19/2018] [Indexed: 12/16/2022] Open
Abstract
Androgen receptor (AR) is the most widely expressed steroid receptor protein in normal breast tissue and is detectable in approximately 90% of primary breast cancers and 75% of metastatic lesions. However, the role of AR in breast cancer development and progression is mired in controversy with evidence suggesting it can either inhibit or promote breast tumorigenesis. Studies have shown it to antagonize estrogen receptor alpha (ERα) DNA binding, thereby preventing pro-proliferative gene transcription; whilst others have demonstrated AR to take on the mantle of a pseudo ERα particularly in the setting of triple negative breast cancer. Evidence for a potentiating role of AR in the development of endocrine resistant breast cancer has also been mounting with reports associating high AR expression with poor response to endocrine treatment. The resurgence of interest into the function of AR in breast cancer has resulted in various emergent clinical trials evaluating anti-AR therapy and selective androgen receptor modulators in the treatment of advanced breast cancer. Trials have reported varied response rates dependent upon subtype with overall clinical benefit rates of ~19-29% for anti-androgen monotherapy, suggesting that with enhanced patient stratification AR could prove efficacious as a breast cancer therapy. Androgens and AR have been reported to facilitate tumor stemness in some cancers; a process which may be mediated through genomic or non-genomic actions of the AR, with the latter mechanism being relatively unexplored in breast cancer. Steroidogenic ligands of the AR are produced in females by the gonads and as sex-steroid precursors secreted from the adrenal glands. These androgens provide an abundant reservoir from which all estrogens are subsequently synthesized and their levels are undiminished in the event of standard hormonal therapeutic intervention in breast cancer. Steroid levels are known to be altered by lifestyle factors such as diet and exercise; understanding their potential role in dictating the function of AR in breast cancer development could therefore have wide-ranging effects in prevention and treatment of this disease. This review will outline the endogenous biochemical drivers of both genomic and non-genomic AR activation and how these may be modulated by current hormonal therapies.
Collapse
Affiliation(s)
| | - Marie McIlroy
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
24
|
Ito S, Ueno A, Ueda T, Nakagawa H, Taniguchi H, Kayukawa N, Fujihara-Iwata A, Hongo F, Okihara K, Ukimura O. CNPY2 inhibits MYLIP-mediated AR protein degradation in prostate cancer cells. Oncotarget 2018; 9:17645-17655. [PMID: 29707137 PMCID: PMC5915145 DOI: 10.18632/oncotarget.24824] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 03/01/2018] [Indexed: 12/02/2022] Open
Abstract
The androgen receptor (AR) is a ligand-dependent transcription factor that promotes prostate cancer (PC) cell growth through control of target gene expression. This report suggests that Canopy FGF signaling regulator 2 (CNPY2) controls AR protein levels in PC cells. We found that AR was ubiquitinated by an E3 ubiquitin ligase, myosin regulatory light chain interacting protein (MYLIP) and then degraded through the ubiquitin-proteasome pathway. CNPY2 decreased the ubiquitination activity of MYLIP by inhibition of interaction between MYLIP and UBE2D1, an E2 ubiquitin ligase. CNPY2 up-regulated gene expression of AR target genes such as KLK3 gene which encodes the prostate specific antigen (PSA) and promoted cell growth of PC cells. The cell growth inhibition by CNPY2 knockdown was rescued by AR overexpression. Furthermore, positive correlation of expression levels between CNPY2 and AR/AR target genes was observed in tissue samples from human prostate cancer patients. Together, these results suggested that CNPY2 promoted cell growth of PC cells by inhibition of AR protein degradation through MYLIP-mediated AR ubiquitination.
Collapse
Affiliation(s)
- Saya Ito
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto-City, Kyoto 602-8566, Japan
| | - Akihisa Ueno
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto-City, Kyoto 602-8566, Japan
| | - Takashi Ueda
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto-City, Kyoto 602-8566, Japan.,Department of Urology, Uji Takeda Hospital, Uji-City, Kyoto 611-0021, Japan
| | - Hideo Nakagawa
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto-City, Kyoto 602-8566, Japan
| | - Hidefumi Taniguchi
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto-City, Kyoto 602-8566, Japan
| | - Naruhiro Kayukawa
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto-City, Kyoto 602-8566, Japan
| | - Atsuko Fujihara-Iwata
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto-City, Kyoto 602-8566, Japan
| | - Fumiya Hongo
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto-City, Kyoto 602-8566, Japan
| | - Koji Okihara
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto-City, Kyoto 602-8566, Japan
| | - Osamu Ukimura
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto-City, Kyoto 602-8566, Japan
| |
Collapse
|
25
|
Di Zazzo E, Galasso G, Giovannelli P, Di Donato M, Castoria G. Estrogens and Their Receptors in Prostate Cancer: Therapeutic Implications. Front Oncol 2018; 8:2. [PMID: 29404276 PMCID: PMC5778111 DOI: 10.3389/fonc.2018.00002] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/04/2018] [Indexed: 12/21/2022] Open
Abstract
A major challenge in clinical management of prostate cancer (PC) is to limit tumor growth and prevent metastatic spreading. Considerable efforts have been made to discover new compounds for PC therapy and recent years have seen promising progress in this field. Pharmacological approaches have been designed to achieve benefits in PC treatment and avoid the negative side effects resulting from administration of antagonists or agonists or new drugs. Nonetheless, the currently available therapies frequently induce resistance and PC progresses toward castration-resistant forms that can be caused by the androgen receptor reactivation and/or mutations, or derangement of signaling pathways. Preclinical and clinical findings have also shown that other nuclear receptors are frequently altered in PC. In this review, we focus on the role of estradiol/estradiol receptor (ER) axis, which controls PC growth and progression. Selective targeting of ER subtypes (α or β) may be an attractive way to limit the growth and spreading of prostatic cancer cells.
Collapse
Affiliation(s)
- Erika Di Zazzo
- Department of Biochemistry, Biophysics and General Pathology, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giovanni Galasso
- Department of Biochemistry, Biophysics and General Pathology, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Pia Giovannelli
- Department of Biochemistry, Biophysics and General Pathology, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Marzia Di Donato
- Department of Biochemistry, Biophysics and General Pathology, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Gabriella Castoria
- Department of Biochemistry, Biophysics and General Pathology, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
26
|
Castoria G, Auricchio F, Migliaccio A. Extranuclear partners of androgen receptor: at the crossroads of proliferation, migration, and neuritogenesis. FASEB J 2016; 31:1289-1300. [PMID: 28031322 DOI: 10.1096/fj.201601047r] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/19/2016] [Indexed: 01/11/2023]
Abstract
In this review, we focus on the role played by the protein partners of ligand-activated extranuclear androgen receptor (AR) in the final effects of hormone action, such as proliferation, migration, and neuritogenesis. The choice of AR partner, at least in part, depends on cell type. Androgen-activated receptor directly associates with cytoplasmic Src tyrosine kinase in epithelial cells, whereas in mesenchymal and neuronal cells, it prevalently interacts with filamin A. In the former, proliferation represents the final hormonal outcome, whereas in the latter, either migration or neuritogenesis, respectively, occurs. Furthermore, AR partner filamin A is replaced with Src when mesenchymal cells are stimulated with very low androgen concentrations. Consequently, the migratory effect is replaced by mitogenesis. Use of peptides that prevent receptor/partner assembly abolishes the effects that are dependent on their association and offers new therapeutic approaches to AR-related diseases. Perturbation of migration is often associated with metastatic spreading in cancer. In turn, cell cycle aberration causes tumors to grow faster, whereas toxic signaling triggers neurodegenerative events in the CNS. Here, we provide examples of new tools that interfere in rapid androgen effects, including migration, proliferation, and neuronal differentiation, together with their potential therapeutic applications in AR-dependent diseases-mainly prostate cancer and neurodegenerative disorders.-Castoria, G., Auricchio, F., Migliaccio, A. Extranuclear partners of androgen receptor: at the crossroads of proliferation, migration, and neuritogenesis.
Collapse
Affiliation(s)
- Gabriella Castoria
- Department of Biochemistry, Biophysics, and General Pathology, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Ferdinando Auricchio
- Department of Biochemistry, Biophysics, and General Pathology, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Antimo Migliaccio
- Department of Biochemistry, Biophysics, and General Pathology, University of Campania "Luigi Vanvitelli," Naples, Italy
| |
Collapse
|
27
|
Di Zazzo E, Galasso G, Giovannelli P, Di Donato M, Di Santi A, Cernera G, Rossi V, Abbondanza C, Moncharmont B, Sinisi AA, Castoria G, Migliaccio A. Prostate cancer stem cells: the role of androgen and estrogen receptors. Oncotarget 2016; 7:193-208. [PMID: 26506594 PMCID: PMC4807992 DOI: 10.18632/oncotarget.6220] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/30/2015] [Indexed: 12/22/2022] Open
Abstract
Prostate cancer is one of the most commonly diagnosed cancers in men, and androgen deprivation therapy still represents the primary treatment for prostate cancer patients. This approach, however, frequently fails and patients develop castration-resistant prostate cancer, which is almost untreatable. Cancer cells are characterized by a hierarchical organization, and stem/progenitor cells are endowed with tumor-initiating activity. Accumulating evidence indicates that prostate cancer stem cells lack the androgen receptor and are, indeed, resistant to androgen deprivation therapy. In contrast, these cells express classical (α and/or β) and novel (GPR30) estrogen receptors, which may represent new putative targets in prostate cancer treatment. In the present review, we discuss the still-debated mechanisms, both genomic and non-genomic, by which androgen and estradiol receptors (classical and novel) mediate the hormonal control of prostate cell stemness, transformation, and the continued growth of prostate cancer. Recent preclinical and clinical findings obtained using new androgen receptor antagonists, anti-estrogens, or compounds such as enhancers of androgen receptor degradation and peptides inhibiting non-genomic androgen functions are also presented. These new drugs will likely lead to significant advances in prostate cancer therapy.
Collapse
Affiliation(s)
- Erika Di Zazzo
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | - Giovanni Galasso
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | - Pia Giovannelli
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | - Marzia Di Donato
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | - Annalisa Di Santi
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | - Gustavo Cernera
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | - Valentina Rossi
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | - Ciro Abbondanza
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | | | - Antonio Agostino Sinisi
- Endocrinology Section, Department of Cardio-Thoracic and Respiratory Diseases, II University of Naples, Naples, Italy
| | - Gabriella Castoria
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | - Antimo Migliaccio
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| |
Collapse
|