1
|
Ulisse S, Baldini E, Pironi D, Gagliardi F, Tripodi D, Lauro A, Carbotta S, Tarroni D, D’Armiento M, Morrone A, Forte F, Frattaroli F, Persechino S, Odorisio T, D’Andrea V, Lori E, Sorrenti S. Is Melanoma Progression Affected by Thyroid Diseases? Int J Mol Sci 2022; 23:ijms231710036. [PMID: 36077430 PMCID: PMC9456309 DOI: 10.3390/ijms231710036] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/25/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Clinical and epidemiological evidence indicate a relationship between thyroid diseases and melanoma. In particular, the hypothyroidism condition appears to promote melanoma spread, which suggests a protective role of thyroid hormones against disease progression. In addition, experimental data suggest that, in addition to thyroid hormones, other hormonal players of the hypothalamic–pituitary–thyroid (HPT) axis, namely the thyrotropin releasing hormone and the thyrotropin, are likely to affect melanoma cells behavior. This information warrants further clinical and experimental studies in order to build a precise pattern of action of the HPT hormones on melanoma cells. An improved knowledge of the involved molecular mechanism(s) could lead to a better and possibly personalized clinical management of these patients.
Collapse
Affiliation(s)
- Salvatore Ulisse
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
- Correspondence:
| | - Enke Baldini
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Daniele Pironi
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Federica Gagliardi
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Domenico Tripodi
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Augusto Lauro
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Sabino Carbotta
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Danilo Tarroni
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Matteo D’Armiento
- Scientific Direction, IRCCS San Gallicano Dermatological Institute, 00144 Rome, Italy
| | - Aldo Morrone
- Scientific Direction, IRCCS San Gallicano Dermatological Institute, 00144 Rome, Italy
| | - Flavio Forte
- Urology Department, M.G. Vannini Hospital, 00177 Rome, Italy
| | - Flaminia Frattaroli
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Severino Persechino
- Department of Neurosciences, Mental Health and Sensory Organs, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Teresa Odorisio
- Laboratory of Molecular and Cell Biology, Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00167 Rome, Italy
| | - Vito D’Andrea
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Eleonora Lori
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Salvatore Sorrenti
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| |
Collapse
|
2
|
The Sex-Related Interplay between TME and Cancer: On the Critical Role of Estrogen, MicroRNAs and Autophagy. Cancers (Basel) 2021; 13:cancers13133287. [PMID: 34209162 PMCID: PMC8267629 DOI: 10.3390/cancers13133287] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/17/2021] [Accepted: 06/25/2021] [Indexed: 01/18/2023] Open
Abstract
The interplay between cancer cells and the tumor microenvironment (TME) has a fundamental role in tumor progression and response to therapy. The plethora of components constituting the TME, such as stroma, fibroblasts, endothelial and immune cells, as well as macromolecules, e.g., hormones and cytokines, and epigenetic factors, such as microRNAs, can modulate the survival or death of cancer cells. Actually, the TME can stimulate the genetically regulated programs that the cell puts in place under stress: apoptosis or, of interest here, autophagy. However, the implication of autophagy in tumor growth appears still undefined. Autophagy mainly represents a cyto-protective mechanism that allows cell survival but, in certain circumstances, also leads to the blocking of cell cycle progression, possibly leading to cell death. Since significant sex/gender differences in the incidence, progression and response to cancer therapy have been widely described in the literature, in this review, we analyzed the roles played by key components of the TME, e.g., estrogen and microRNAs, on autophagy regulation from a sex/gender-based perspective. We focused our attention on four paradigmatic and different forms of cancers-colon cancer, melanoma, lymphoma, and lung cancer-concluding that sex-specific differences may exert a significant impact on TME/cancer interaction and, thus, tumor growth.
Collapse
|
3
|
Morgese F, Sampaolesi C, Torniai M, Conti A, Ranallo N, Giacchetti A, Serresi S, Onofri A, Burattini M, Ricotti G, Berardi R. Gender Differences and Outcomes in Melanoma Patients. Oncol Ther 2020; 8:103-114. [PMID: 32700073 PMCID: PMC7359998 DOI: 10.1007/s40487-020-00109-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Melanoma is one of the most common cancers in younger people. The incidence of cutaneous melanoma is increasing in patients of both sexes, with female patients generally living longer than their male counterparts. The aim of this retrospective study was to evaluate and confirm the sex-based difference in survival of melanoma patients and the relationship of this difference with pathological features. METHODS A total of 1023 patients who had been treated at the Department of Medical Oncology, Università Politecnica Marche (Ancona, Italy) and the INRCA-IRCCS Department of Dermatology (Ancona, Italy) between 1987 and 2014 were enrolled in the study. RESULTS In terms of stage of disease at onset, there was a significant difference in disease-free survival (DFS) and overall survival (OS) in favor of female patients in disease stage I (P = 0.001 and P = 0.01, respectively) and II (P = 0.02 and P = 0.009, respectively). Female patients also showed a significant improvement in 12-year DFS and 12-year OS adjusted for pathological features (Breslow thickness, ulceration, "absent" tumor-infiltrating lymphocyte (TIL) melanomas, "non-brisk" TIL pattern). Globally, female patients had an advantage over with male patients in both DFS and OS (P < 0.001). CONCLUSIONS Our results show that women have a survival benefit over with men after adjustment for many variables that can reduce mortality risk in female melanoma patients. In a future investigation we wish to examine possible biological sex differences in tumor-host interactions.
Collapse
Affiliation(s)
- Francesca Morgese
- Clinica Oncologica, Università Politecnica delle Marche-Azienda Ospedaliero-Universitaria Umberto I, Via Conca, Ancona, AN, Italy
| | - Caterina Sampaolesi
- Clinica Oncologica, Università Politecnica delle Marche-Azienda Ospedaliero-Universitaria Umberto I, Via Conca, Ancona, AN, Italy
| | - Mariangela Torniai
- Clinica Oncologica, Università Politecnica delle Marche-Azienda Ospedaliero-Universitaria Umberto I, Via Conca, Ancona, AN, Italy
| | - Alessandro Conti
- Department of Clinical and Specialist Sciences, Urology, Università Politecnica delle Marche-Azienda Ospedaliero-Universitaria Umberto I, Via Conca, Ancona, AN, Italy
| | - Nicoletta Ranallo
- Clinica Oncologica, Università Politecnica delle Marche-Azienda Ospedaliero-Universitaria Umberto I, Via Conca, Ancona, AN, Italy
| | - Alfredo Giacchetti
- L'Unità Operativa di Dermatologia, INRCA/IRCCS, Via Montagnola, Ancona, AN, Italy
| | - Stefano Serresi
- L'Unità Operativa di Dermatologia, INRCA/IRCCS, Via Montagnola, Ancona, AN, Italy
| | - Azzurra Onofri
- Clinica Oncologica, Università Politecnica delle Marche-Azienda Ospedaliero-Universitaria Umberto I, Via Conca, Ancona, AN, Italy
| | - Michela Burattini
- Clinica Oncologica, Università Politecnica delle Marche-Azienda Ospedaliero-Universitaria Umberto I, Via Conca, Ancona, AN, Italy
| | - Giuseppe Ricotti
- L'Unità Operativa di Dermatologia, INRCA/IRCCS, Via Montagnola, Ancona, AN, Italy
| | - Rossana Berardi
- Clinica Oncologica, Università Politecnica delle Marche-Azienda Ospedaliero-Universitaria Umberto I, Via Conca, Ancona, AN, Italy.
| |
Collapse
|
4
|
Ethinylestradiol and Levonorgestrel as Active Agents in Normal Skin, and Pathological Conditions Induced by UVB Exposure: In Vitro and In Ovo Assessments. Int J Mol Sci 2018; 19:ijms19113600. [PMID: 30441863 PMCID: PMC6275072 DOI: 10.3390/ijms19113600] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/08/2018] [Accepted: 11/10/2018] [Indexed: 11/22/2022] Open
Abstract
The link between melanoma development and the use of oral combined contraceptives is not fully elucidated, and the data concerning this issue are scarce and controversial. In the present study, we show that the components of oral contraceptives, ethinylestradiol (EE), levonorgestrel (LNG), and their combination (EE + LNG) ± UVB (ultraviolet B radiation) induced differential effects on healthy (human keratinocytes, fibroblasts, and primary epidermal melanocytes, and murine epidermis cells) and melanoma cells (human—A375 and murine—B164A5), as follows: (i) at low doses (1 µM), the hormones were devoid of significant toxicity on healthy cells, but in melanoma cells, they triggered cell death via apoptosis; (ii) higher doses (10 µM) were associated with cytotoxicity in all cells, the most affected being the melanoma cells; (iii) UVB irradiation proved to be toxic for all types of cells; (iv) UVB irradiation + hormonal stimulation led to a synergistic cytotoxicity in the case of human melanoma cells—A375 and improved viability rates of healthy and B164A5 cells. A weak irritant potential exerted by EE and EE + LNG (10 µM) was assessed by the means of a chick chorioallantoic membrane assay. Further studies are required to elucidate the hormones’ cell type-dependent antimelanoma effect and the role played by melanin in this context.
Collapse
|
5
|
Oliveira C, Lourenço GJ, Rinck-Junior JA, de Moraes AM, Lima CSP. Polymorphisms in apoptosis-related genes in cutaneous melanoma prognosis: sex disparity. Med Oncol 2017; 34:19. [PMID: 28050764 DOI: 10.1007/s12032-016-0868-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 12/07/2016] [Indexed: 01/18/2023]
Abstract
Cutaneous melanoma (CM) cells are resistant to apoptosis, and steroid hormones are involved in this process through regulation of TP53, MDM2, BAX, and BCL2 expression. We analyzed herein sex differences in outcomes of CM patients associated with TP53 c.215G>C, MDM2 c.309T>G, BAX c.-248G>A, and BCL2 c.-717C>A polymorphisms. DNA from 121 men and 116 women patients was analyzed by polymerase chain reaction and enzymatic digestion assays. At 60 months of follow-up, shorter progression-free survival (PFS) was seen in males with MDM2 GG + BCL2 AA (20.0 vs. 62.6%, P = 0.0008) genotype. Men carriers of the genotype had poor PFS (HR 3.78, 95% CI 1.30-11.0) than others. For women, shorter PFS was associated with TP53 GC or CC (61.4 vs. 80.8%, P = 0.01) and TP53 GC or CC + MDM2 TG or GG (59.1 vs. 85.4%, P = 0.01) genotypes at the same time. Women carriers of the genotypes had poor PFS (HR 2.46, 95% CI 1.19-5.09; HR 9.49, 95% CI 1.14-78.50) than others, respectively. Our data present, for the first time, preliminary evidence that inherited abnormalities on TP53, MDM2 and BCL2 genes, enrolled in apoptosis pathways, have a pivotal role in differences of outcomes in women and men with CM.
Collapse
Affiliation(s)
- Cristiane Oliveira
- Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Rua Alexander Fleming, 181, Cidade Universitária "Zeferino Vaz", Distrito de Barão Geraldo, Campinas, São Paulo, CEP 13083-970, Brazil
| | - Gustavo Jacob Lourenço
- Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Rua Alexander Fleming, 181, Cidade Universitária "Zeferino Vaz", Distrito de Barão Geraldo, Campinas, São Paulo, CEP 13083-970, Brazil
| | - José Augusto Rinck-Junior
- Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Rua Alexander Fleming, 181, Cidade Universitária "Zeferino Vaz", Distrito de Barão Geraldo, Campinas, São Paulo, CEP 13083-970, Brazil
| | - Aparecida Machado de Moraes
- Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Rua Alexander Fleming, 181, Cidade Universitária "Zeferino Vaz", Distrito de Barão Geraldo, Campinas, São Paulo, CEP 13083-970, Brazil
| | - Carmen Silvia Passos Lima
- Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Rua Alexander Fleming, 181, Cidade Universitária "Zeferino Vaz", Distrito de Barão Geraldo, Campinas, São Paulo, CEP 13083-970, Brazil.
| |
Collapse
|
6
|
Abstract
The skin is considered the mirror of the soul and is affected by neurohormonal triggers, especially stress. Hair follicles, keratinocytes, mast cells, melanocytes, and sebocytes all express sex and stress hormones implicating them in a local "hypothalamic-pituitary-adrenal axis." In particular, the peptides corticotropin-releasing hormone (CRH) and neurotensin (NT) have synergistic action stimulating mast cells and are uniquely elevated in the serum of patients with skin diseases exacerbated by stress. Addressing the neurohormonal regulation of skin function could lead to new targets for effective treatment of inflammatory skin diseases.
Collapse
Affiliation(s)
- Theoharis C Theoharides
- Department of Integrative Physiology and Pathobiology, Molecular Immunopharmacology and Drug Discovery Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Suite J304, Boston, MA, 02111, USA.
- Sackler School of Graduate Biomedical Sciences, Program in Pharmacology and Experimental Therapeutics, Tufts University, Boston, MA, USA.
- Department of Internal Medicine, Tufts University School of Medicine and Tufts Medical Center, Boston, MA, USA.
| | - Julia M Stewart
- Department of Integrative Physiology and Pathobiology, Molecular Immunopharmacology and Drug Discovery Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Suite J304, Boston, MA, 02111, USA
| | - Alexandra Taracanova
- Department of Integrative Physiology and Pathobiology, Molecular Immunopharmacology and Drug Discovery Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Suite J304, Boston, MA, 02111, USA
- Sackler School of Graduate Biomedical Sciences, Program in Pharmacology and Experimental Therapeutics, Tufts University, Boston, MA, USA
| | - Pio Conti
- Department of Graduate Medical Sciences, University of Chieti, Chieti, Italy
| | - Christos C Zouboulis
- Departments of Dermatology, Venereology, Allergology and Immunology, Dessau Medical Center, Dessau, Germany
| |
Collapse
|
7
|
Tamoxifen Resistance: Emerging Molecular Targets. Int J Mol Sci 2016; 17:ijms17081357. [PMID: 27548161 PMCID: PMC5000752 DOI: 10.3390/ijms17081357] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/10/2016] [Accepted: 08/16/2016] [Indexed: 12/12/2022] Open
Abstract
17β-Estradiol (E2) plays a pivotal role in the development and progression of breast cancer. As a result, blockade of the E2 signal through either tamoxifen (TAM) or aromatase inhibitors is an important therapeutic strategy to treat or prevent estrogen receptor (ER) positive breast cancer. However, resistance to TAM is the major obstacle in endocrine therapy. This resistance occurs either de novo or is acquired after an initial beneficial response. The underlying mechanisms for TAM resistance are probably multifactorial and remain largely unknown. Considering that breast cancer is a very heterogeneous disease and patients respond differently to treatment, the molecular analysis of TAM’s biological activity could provide the necessary framework to understand the complex effects of this drug in target cells. Moreover, this could explain, at least in part, the development of resistance and indicate an optimal therapeutic option. This review highlights the implications of TAM in breast cancer as well as the role of receptors/signal pathways recently suggested to be involved in the development of TAM resistance. G protein—coupled estrogen receptor, Androgen Receptor and Hedgehog signaling pathways are emerging as novel therapeutic targets and prognostic indicators for breast cancer, based on their ability to mediate estrogenic signaling in ERα-positive or -negative breast cancer.
Collapse
|
8
|
Poletini MO, de Assis LVM, Moraes MN, Castrucci AMDL. Estradiol differently affects melanin synthesis of malignant and normal melanocytes: a relationship with clock and clock-controlled genes. Mol Cell Biochem 2016; 421:29-39. [DOI: 10.1007/s11010-016-2781-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 08/05/2016] [Indexed: 12/20/2022]
|
9
|
Marzagalli M, Montagnani Marelli M, Casati L, Fontana F, Moretti RM, Limonta P. Estrogen Receptor β in Melanoma: From Molecular Insights to Potential Clinical Utility. Front Endocrinol (Lausanne) 2016; 7:140. [PMID: 27833586 PMCID: PMC5080294 DOI: 10.3389/fendo.2016.00140] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 10/12/2016] [Indexed: 12/14/2022] Open
Abstract
Cutaneous melanoma is an aggressive tumor; its incidence has been reported to increase fast in the past decades. Melanoma is a heterogeneous tumor, with most patients harboring mutations in the BRAF or NRAS oncogenes, leading to the overactivation of the MAPK/ERK and PI3K/Akt pathways. The current therapeutic approaches are based on therapies targeting mutated BRAF and the downstream pathway, and on monoclonal antibodies against the immune checkpoint blockade. However, treatment resistance and side effects are common events of these therapeutic strategies. Increasing evidence supports that melanoma is a hormone-related cancer. Melanoma incidence is higher in males than in females, and females have a significant survival advantage over men. Estrogens exert their effects through estrogen receptors (ERα and ERβ) that affect cancer growth in an opposite way: ERα is associated with a proliferative action and ERβ with an anticancer effect. ERβ is the predominant ER in melanoma, and its expression decreases in melanoma progression, supporting its role as a tumor suppressor. Thus, ERβ is now considered as an effective molecular target for melanoma treatment. 17β-estradiol was reported to inhibit melanoma cells proliferation; however, clinical trials did not provide the expected survival benefits. In vitro studies demonstrate that ERβ ligands inhibit the proliferation of melanoma cells harboring the NRAS (but not the BRAF) mutation, suggesting that ERβ activation might impair melanoma development through the inhibition of the PI3K/Akt pathway. These data suggest that ERβ agonists might be considered as an effective treatment strategy, in combination with MAPK inhibitors, for NRAS mutant melanomas. In an era of personalized medicine, pretreatment evaluation of the expression of ER isoforms together with the concurrent oncogenic mutations should be considered before selecting the most appropriate therapeutic intervention. Natural compounds that specifically bind to ERβ have been identified. These phytoestrogens decrease the proliferation of melanoma cells. Importantly, these effects are unrelated to the oncogenic mutations of melanomas, suggesting that, in addition to their ERβ activating function, these compounds might impair melanoma development through additional mechanisms. A better identification of the role of ERβ in melanoma development will help increase the therapeutic options for this aggressive pathology.
Collapse
Affiliation(s)
- Monica Marzagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Marina Montagnani Marelli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Lavinia Casati
- Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, Milano, Italy
| | - Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Roberta Manuela Moretti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
- *Correspondence: Patrizia Limonta,
| |
Collapse
|