1
|
Tauch S, Hey J, Kast B, Gengenbacher N, Weiß L, Sator‐Schmitt M, Lohr S, Brobeil A, Schirmacher P, Utikal J, Augustin HG, Plass C, Angel P. A Unique Signature for Cancer-Associated Fibroblasts in Melanoma Metastases. Pigment Cell Melanoma Res 2025; 38:e70002. [PMID: 39924882 PMCID: PMC11808227 DOI: 10.1111/pcmr.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/27/2024] [Accepted: 01/16/2025] [Indexed: 02/11/2025]
Abstract
Cancer-associated fibroblasts (CAFs) represent a central cell population of the tumor microenvironment (TME). Recently, single-cell RNA-sequencing (scRNA-seq) analyses of primary tumors of different cancer entities yielded different classifications of CAF subsets underscoring the heterogeneity of CAFs within the TME. Here, we analyzed the transcriptional signatures of approximately 8400 CAFs and normal fibroblasts by scRNA-seq and compared genetic profiles of CAFs from murine melanoma primary tumors to CAFs from corresponding melanoma lung metastases. This revealed distinct subsets for primary tumor and metastasis-specific CAF populations, respectively. Combined with the spatial characterization of metastasis CAFs at the RNA and protein level, scRNA analyses indicate tumor-dependent crosstalk between neutrophils and CAFs, mediated via SAA3 and IL1b-related signaling pathways, which can be recapitulated in vitro. Analyzing tissue sections of human patient samples, this interaction was found to be present in human melanoma metastasis. Taken together, our data highlight unique characteristics of metastasis CAFs with potential therapeutic impact for melanoma metastasis.
Collapse
Affiliation(s)
- Saskia Tauch
- Division Signal Transduction and Growth ControlGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Joschka Hey
- Division of Cancer EpigenomicsGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Bettina Kast
- Division Signal Transduction and Growth ControlGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
| | - Nicolas Gengenbacher
- Division of Vascular Oncology and MetastasisGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
- European Center for Angioscience (ECAS), Medical Faculty MannheimHeidelberg UniversityMannheimGermany
- DKFZ‐Hector Cancer Institute, University Medical Centre MannheimMannheimGermany
| | - Lena Weiß
- Division Signal Transduction and Growth ControlGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
| | - Melanie Sator‐Schmitt
- Division Signal Transduction and Growth ControlGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
| | - Sabrina Lohr
- Division Signal Transduction and Growth ControlGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
| | - Alexander Brobeil
- Institute of PathologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Peter Schirmacher
- Institute of PathologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Jochen Utikal
- DKFZ‐Hector Cancer Institute, University Medical Centre MannheimMannheimGermany
- Skin Cancer UnitGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Department of Dermatology, Venereology and AllergologyUniversity Medical Center Mannheim, Ruprecht‐Karl University of HeidelbergMannheimGermany
| | - Hellmut G. Augustin
- Division of Vascular Oncology and MetastasisGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
- European Center for Angioscience (ECAS), Medical Faculty MannheimHeidelberg UniversityMannheimGermany
- DKFZ‐Hector Cancer Institute, University Medical Centre MannheimMannheimGermany
| | - Christoph Plass
- Division of Cancer EpigenomicsGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Peter Angel
- Division Signal Transduction and Growth ControlGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
| |
Collapse
|
2
|
Marcovici I, Chioibas R, Zupko I, Pinzaru I, Moaca A, Ledeti A, Barbu-Tudoran L, Geamantan A, Predescu I, Dehelean CA. Preclinical pharmaco-toxicological screening of biomimetic melanin-like nanoparticles as a potential therapeutic strategy for cutaneous melanoma. Front Pharmacol 2025; 16:1487854. [PMID: 39981176 PMCID: PMC11839674 DOI: 10.3389/fphar.2025.1487854] [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: 08/28/2024] [Accepted: 01/20/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction Despite its rarity, cutaneous melanoma (CM) represents the deadliest skin cancer with a high mortality rate, an incidence on the rise, and limited therapeutic options at present. Melanin is a polymeric pigment naturally produced within melanocytes and CM cells that gained a noteworthy attention due to its pharmacological properties, and potential for the design of nanoplatforms with biomedical applications. Up to date, the utilization of melanin-like nanoparticles (MEL-NPs) in cancer treatment has been well-documented, although their efficacy in CM therapy remains scarcely investigated. The current study presents the preclinical evaluation of MEL-NPs as a potential nanomedicine for CM management. Methods MEL-NPs were produced through the oxidative polymerization of dopamine and characterized via electron microscopy and UV-VIS spectroscopy. The antioxidant activity was determined by using the DPPH method. The cytotoxic, anti-migratory, anti-clonogenic, pro-oxidant and pro-apoptotic properties of MEL-NPs were investigated in vitro by applying the MTT viability test, bright-field and immunofluorescence microscopy, DCFDA/H2DCFDA test, scratch assay, colony formation assay, and RT-qPCR. The irritant and anti-angiogenic effects were assessed in ovo on the vascularized chorioallantoic membrane (CAM). Results The as-made MEL-NPs presented a spherical morphology, an average size of 85.61 nm, a broad UV-VIS absorption spectrum, and a strong antioxidant activity. After a 24 h treatment, MEL-NPs exerted a selective cytotoxicity in SH-4 and B164A5 CM cells compared to HEMa, HaCaT, and JB6 Cl 41-5a healthy skin cells, except for the concentration of 100 µg/mL, at which their viability declined under 70%. Additionally, MEL-NPs accumulated within the intracellular space of CM cells, forming a perinuclear coating, inhibited their motility and clonogenic potential, increased intracellular oxidative stress, targeted the epithelial-to-mesenchymal transition, and induced apoptosis by altering cell morphology, nuclear aspect, F-actin and tubulin distribution, and by modulating the expression of pro- and anti-apoptotic markers. In ovo, MEL-NPs lacked irritant and vascular toxic effects, while exerting an angio-suppressive activity. Conclusion MEL-NPs demonstrated promising anti-melanoma properties, showing a selective cytotoxicity, a strong anti-invasive effect and a pro-apoptotic activity in CM cells, while inhibiting CAM angiogenesis, these novel findings contributing to future research on the potential application of this nanoplatform in CM therapy.
Collapse
Affiliation(s)
- Iasmina Marcovici
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy from Timisoara, Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy from Timisoara, Timisoara, Romania
| | - Raul Chioibas
- Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy from Timisoara, Timisoara, Romania
- CBS Medcom Hospital, Timisoara, Romania
| | - Istvan Zupko
- Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - Iulia Pinzaru
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy from Timisoara, Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy from Timisoara, Timisoara, Romania
| | - Alina Moaca
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy from Timisoara, Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy from Timisoara, Timisoara, Romania
| | - Adriana Ledeti
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy from Timisoara, Timisoara, Romania
- Advanced Instrumental Screening Center, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania
| | - Lucian Barbu-Tudoran
- Electron Microscopy Laboratory “Prof. C. Craciun”, Faculty of Biology and Geology, “Babes-Bolyai” University, Cluj-Napoca, Romania
- Electron Microscopy Integrated Laboratory, National Institute for R and D of Isotopic and Molecular Technologies, Cluj-Napoca, Romania
| | - Andreea Geamantan
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy from Timisoara, Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy from Timisoara, Timisoara, Romania
| | - Iasmina Predescu
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy from Timisoara, Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy from Timisoara, Timisoara, Romania
| | - Cristina Adriana Dehelean
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy from Timisoara, Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy from Timisoara, Timisoara, Romania
| |
Collapse
|
3
|
Gouveia E, de Sousa RT, Aguiar SI, Gírio A, Costa I, Dionísio MR, Moital I. Malignant melanoma in Portuguese adult population: a scoping review of the real-world evidence. Clin Transl Oncol 2025; 27:770-777. [PMID: 39012454 DOI: 10.1007/s12094-024-03579-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/17/2024] [Indexed: 07/17/2024]
Abstract
PURPOSE Malignant melanoma is an aggressive cancer, and there is a notable dearth on epidemiology, clinical and treatment characterization within the Portuguese population. We performed a scoping review to identify real-world evidence studies focused in Portuguese adult patients with malignant melanoma. METHODS A comprehensive search was conducted. After screening, we described the studies by design, sample size, geographics, setting, population, and outcomes reported. RESULTS The search yielded 54 studies, mainly retrospective (79.6%). The population assessed was heterogeneous varying from patients with melanoma in general to specific types of melanoma, or even more restricted to patients with specific conditions. The evidence found was mostly concerning clinical outcomes (n=46), patients' clinical profile (n=44) and demographic characterization (n=48). Treatment information was described in 30 studies whereas only 18 reported epidemiological parameters. Studies were mainly performed by the major oncology centers in Lisbon, Oporto and Coimbra, and only two evaluated the entire Portuguese population. To allow comparability, only studies including patients with cutaneous malignant melanoma were considered (13 of the 54) for outcomes evaluation analysis. Median OS varied from 18 to 36 months, assessed after melanoma treatment. Incidence was the most reported epidemiological parameter, confirming the increasing number of cutaneous malignant melanoma patients over the years. Only one study reported prevalence and four reported mortality rates. CONCLUSIONS The evidence found confirms the lack of information about malignant melanoma in Portugal, highlighting the need of real-world studies to assess melanoma prevalence and incidence rates, current treatment approaches, and clinical characterization of these patients.
Collapse
Affiliation(s)
- Emanuel Gouveia
- Department of Medical Oncology, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Rita Teixeira de Sousa
- Department of Medical Oncology, Centro Hospitalar Universitário Lisboa Norte-Hospital de Santa Maria, Lisbon, Portugal
| | - Sandra I Aguiar
- Novartis Innovative Medicines International, Produtos Farmacêuticos S.A., Porto Salvo, Portugal
| | - Ana Gírio
- Novartis Innovative Medicines International, Produtos Farmacêuticos S.A., Porto Salvo, Portugal.
| | - Inês Costa
- Novartis Innovative Medicines International, Produtos Farmacêuticos S.A., Porto Salvo, Portugal
| | - Maria Rita Dionísio
- Novartis Innovative Medicines International, Produtos Farmacêuticos S.A., Porto Salvo, Portugal
| | - Inês Moital
- Novartis Innovative Medicines International, Produtos Farmacêuticos S.A., Porto Salvo, Portugal
| |
Collapse
|
4
|
Bidram M, Ganjalikhany MR. Bioactive peptides from food science to pharmaceutical industries: Their mechanism of action, potential role in cancer treatment and available resources. Heliyon 2024; 10:e40563. [PMID: 39654719 PMCID: PMC11626046 DOI: 10.1016/j.heliyon.2024.e40563] [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: 02/08/2024] [Revised: 10/29/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024] Open
Abstract
Cancer is known as the main cause of mortality in the world, and every year, the rate of incidence and death due to cancer is increasing. Bioactive peptides are one of the novel therapeutic options that are considered a suitable alternative to toxic chemotherapy drugs because they limit side effects with their specific function. In fact, bioactive peptides are short amino acid sequences that obtain diverse physiological functions to maintain human health after being released from parent proteins. This group of biological molecules that can be isolated from different types of natural protein sources has attracted much attention in the field of pharmaceutical and functional foods production. The current article describes the therapeutic benefits of bioactive peptides and specifically and extensively reviews their role in cancer treatment, available sources for discovering anticancer peptides, mechanisms of action, production methods, and existing challenges.
Collapse
Affiliation(s)
- Maryam Bidram
- Department of Cell and Molecular Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mohamad Reza Ganjalikhany
- Department of Cell and Molecular Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
5
|
Shi Y, Mo R, Chen Y, Ma Z, Wen B, Tan Q. Establishment and Validation of Prognostic Nomograms for Nonmetastatic Melanoma of the Limbs-A SEER-Based Study. J INVEST SURG 2024; 37:2401125. [PMID: 39370138 DOI: 10.1080/08941939.2024.2401125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/23/2024] [Accepted: 08/31/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Malignant melanoma, a highly aggressive skin cancer, has remarkable incidence and mortality nowadays. This study aims to explore prognostic factors associated with nonmetastatic cutaneous melanoma of the limbs and to develop nomograms for predicting overall survival (OS) and cancer-specific survival (CSS). METHODS The study cohort was derived from the Surveillance, Epidemiology, and End Results database. Univariate Cox regression, Lasso regression, and multivariate Cox regression analyses were conducted to identify prognostic factors and construct nomograms. The receiver operating characteristic (ROC) curve, time-dependent C-index, calibration curve, decision curve analysis (DCA) and Kaplan-Meier method were used to evaluate the accuracy and clinical applicability of the nomograms. RESULTS A total of 15,606 patients were enrolled. Multivariate analysis identified several prognostic factors for OS and CSS including age, sex, histologic type, N stage, tumor thickness, depth of invasion, mitotic rate, ulceration, surgery of primary site, systemic therapy, race, and number of lymph nodes examined. A nomogram incorporating 12 independent predictors for OS was developed, with a C-index of 0.866 (95% confidence interval [CI]: 0.858-0.874) in the training cohort and 0.853 (95% CI: 0.839-0.867) in validation. For CSS, 10 independent predictors and one related factor were included, yielding a C-index of 0.913 (95% CI: 0.903-0.923) in the training cohort and 0.922 (95% CI: 0.908-0.936) in validation. The ROC curve, time-dependent C-index, calibration curve, DCA, and K-M plot demonstrated favorable discrimination, calibration, and clinical utility. CONCLUSION The developed nomograms provide a precise and personalized predictive tool for risk management of patients with nonmetastatic limb melanoma.
Collapse
Affiliation(s)
- Yutong Shi
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Ran Mo
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yutong Chen
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhouji Ma
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Bo Wen
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qian Tan
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
6
|
Ribeiro AB, de Melo MRS, de Melo Junqueira M, Rodrigues MGL, de Souza TO, Fernandes G, Santos MFC, Ambrósio SR, Bastos JK, Tavares DC. Efficacy and safety of guttiferone E in melanoma-bearing mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5265-5274. [PMID: 38270618 DOI: 10.1007/s00210-024-02962-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/15/2024] [Indexed: 01/26/2024]
Abstract
Melanoma, an aggressive and potentially fatal skin cancer, is constrained by immunosuppression, resistance, and high toxicity in its treatment. Consequently, there is an urgent need for innovative antineoplastic agents. Therefore, this study investigated the antimelanoma potential of guttiferone E (GE). In an allogeneic murine B16 melanoma model, GE was administered subcutaneously and intraperitoneally. Antitumor evaluation included tumor volume/weight measurements and histopathological and immunohistochemical analysis. Furthermore, the toxicity of the treatments was evaluated through body/organ weights, biochemical parameters, and genotoxicity. Subcutaneous administration of 20 mg/kg of GE resulted in a significant reduction in both tumor volume and weight, effectively suppressing melanoma cell proliferation as evidenced by a decrease in mitotic figures. The tumor growth inhibition rate was equivalent to 54%. This treatment upregulated cleaved caspase-3, indicating apoptosis induction. On the other hand, intraperitoneal administration of GE showed no antimelanoma effect. Remarkably, GE treatments exhibited no toxicity, evidenced by non-significant differences in body weight gain, as well as organ weight, biochemical parameters of nephrotoxicity and hepatotoxicity, and genotoxic damage. This study revealed, for the first time, the efficacy of subcutaneous administration of GE in reducing melanoma, in the absence of toxicity. Furthermore, it was observed that the apoptotic signaling pathway is involved in the antimelanoma property of GE. These findings offer valuable insights for further exploring GE's therapeutic applications in melanoma treatment.
Collapse
Affiliation(s)
- Arthur Barcelos Ribeiro
- University of Franca, Avenida Dr. Armando Salles Oliveira, 201, Parque Universitário, Franca, São Paulo, 14404-600, Brazil.
| | - Matheus Reis Santos de Melo
- University of Franca, Avenida Dr. Armando Salles Oliveira, 201, Parque Universitário, Franca, São Paulo, 14404-600, Brazil
| | - Marcela de Melo Junqueira
- University of Franca, Avenida Dr. Armando Salles Oliveira, 201, Parque Universitário, Franca, São Paulo, 14404-600, Brazil
| | - Mônica Garcia Leal Rodrigues
- University of Franca, Avenida Dr. Armando Salles Oliveira, 201, Parque Universitário, Franca, São Paulo, 14404-600, Brazil
| | - Thiago Olimpio de Souza
- University of Franca, Avenida Dr. Armando Salles Oliveira, 201, Parque Universitário, Franca, São Paulo, 14404-600, Brazil
| | - Gabriela Fernandes
- University of Franca, Avenida Dr. Armando Salles Oliveira, 201, Parque Universitário, Franca, São Paulo, 14404-600, Brazil
| | | | - Sérgio Ricardo Ambrósio
- University of Franca, Avenida Dr. Armando Salles Oliveira, 201, Parque Universitário, Franca, São Paulo, 14404-600, Brazil
| | - Jairo Kenupp Bastos
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n, Ribeirão Preto, São Paulo, 14040-903, Brazil
| | - Denise Crispim Tavares
- University of Franca, Avenida Dr. Armando Salles Oliveira, 201, Parque Universitário, Franca, São Paulo, 14404-600, Brazil.
| |
Collapse
|
7
|
Bunda S, Kálmán-Szabó I, Lihi N, Képes Z, Szikra D, Peline Szabo J, Timári I, Szücs D, May NV, Papp G, Trencsényi G, Kálmán FK. Diagnosis of Melanoma with 61Cu-Labeled PET Tracer. J Med Chem 2024; 67:9342-9354. [PMID: 38753457 DOI: 10.1021/acs.jmedchem.4c00479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Until the recent years, substances containing radioactive 61Cu were strongly considered as potential positron-emitting radiopharmaceuticals for use in positron emission tomography (PET) applications; however, due to their suitably long half-life, and generator-independent and cost-effective production, they seem to be economically viable for human imaging. Since malignant melanoma (MM) is a major public health problem, its early diagnosis is a crucial contributor to long-term survival, which can be achieved using radiolabeled α-melanocyte-stimulating hormone analog NAPamide derivatives. Here, we report on the physicochemical features of a new CB-15aneN5-based Cu(II) complex ([Cu(KFTGdiac)]-) and the ex vivo and in vivo characterization of its NAPamide conjugate. The rigid chelate possesses prompt complex formation and suitable inertness (t1/2 = 18.4 min in 5.0 M HCl at 50 °C), as well as excellent features in the diagnosis of B16-F10 melanoma tumors (T/M(SUVs) (in vivo): 12.7, %ID/g: 6.6 ± 0.3, T/M (ex vivo): 22).
Collapse
Affiliation(s)
- Szilvia Bunda
- Department of Physical Chemistry, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
| | - Ibolya Kálmán-Szabó
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Norbert Lihi
- HUN-REN-UD Mechanisms of Complex Homogeneous and Heterogeneous Chemical Reactions Research Group, Department of Inorganic and Analytical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Zita Képes
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Dezső Szikra
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Judit Peline Szabo
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - István Timári
- Department of Organic Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
- HUN-REN-UD Molecular Recognition and Interaction Research Group, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Dániel Szücs
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Nóra V May
- Centre for Structural Science, Research Centre for Natural Sciences, Hungarian Research Network (HUN-REN), Magyar tudósok körútja 2, H-1117 Budapest, Hungary
| | - Gábor Papp
- Department of Physical Chemistry, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
| | - György Trencsényi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Ferenc K Kálmán
- Department of Physical Chemistry, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
| |
Collapse
|
8
|
Zhang J, Wang Q, Qi S, Duan Y, Liu Z, Liu J, Zhang Z, Li C. An oncogenic enhancer promotes melanoma progression via regulating ETV4 expression. J Transl Med 2024; 22:547. [PMID: 38849954 PMCID: PMC11157841 DOI: 10.1186/s12967-024-05356-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Enhancers are important gene regulatory elements that promote the expression of critical genes in development and disease. Aberrant enhancer can modulate cancer risk and activate oncogenes that lead to the occurrence of various cancers. However, the underlying mechanism of most enhancers in cancer remains unclear. Here, we aim to explore the function and mechanism of a crucial enhancer in melanoma. METHODS Multi-omics data were applied to identify an enhancer (enh17) involved in melanoma progression. To evaluate the function of enh17, CRISPR/Cas9 technology were applied to knockout enh17 in melanoma cell line A375. RNA-seq, ChIP-seq and Hi-C data analysis integrated with luciferase reporter assay were performed to identify the potential target gene of enh17. Functional experiments were conducted to further validate the function of the target gene ETV4. Multi-omics data integrated with CUT&Tag sequencing were performed to validate the binding profile of the inferred transcription factor STAT3. RESULTS An enhancer, named enh17 here, was found to be aberrantly activated and involved in melanoma progression. CRISPR/Cas9-mediated deletion of enh17 inhibited cell proliferation, migration, and tumor growth of melanoma both in vitro and in vivo. Mechanistically, we identified ETV4 as a target gene regulated by enh17, and functional experiments further support ETV4 as a target gene that is involved in cancer-associated phenotypes. In addition, STAT3 acts as a transcription factor binding with enh17 to regulate the transcription of ETV4. CONCLUSIONS Our findings revealed that enh17 plays an oncogenic role and promotes tumor progression in melanoma, and its transcriptional regulatory mechanisms were fully elucidated, which may open a promising window for melanoma prevention and treatment.
Collapse
Affiliation(s)
- Junyou Zhang
- School of Engineering Medicine, Beihang University, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing, 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Qilin Wang
- School of Engineering Medicine, Beihang University, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing, 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Sihan Qi
- School of Engineering Medicine, Beihang University, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing, 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Yingying Duan
- School of Engineering Medicine, Beihang University, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing, 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Zhaoshuo Liu
- School of Engineering Medicine, Beihang University, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing, 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Jiaxin Liu
- School of Engineering Medicine, Beihang University, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing, 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Ziyi Zhang
- School of Engineering Medicine, Beihang University, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing, 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Chunyan Li
- School of Engineering Medicine, Beihang University, Beijing, 100191, China.
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing, 100191, China.
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China.
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, 100191, China.
| |
Collapse
|
9
|
Basson C, Phiri AE, Gandhi M, Anguelov R, Serem JC, Bipath P, Hlophe YN. In vitro effects and mathematical modelling of CTCE-9908 (a chemokine receptor 4 antagonist) on melanoma cell survival. Clin Exp Pharmacol Physiol 2024; 51:e13865. [PMID: 38692577 DOI: 10.1111/1440-1681.13865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/09/2024] [Accepted: 04/02/2024] [Indexed: 05/03/2024]
Abstract
CTCE-9908, a CXC chemokine receptor 4 (CXCR4) antagonist, prevents CXCR4 phosphorylation and inhibits the interaction with chemokine ligand 12 (CXCL12) and downstream signalling pathways associated with metastasis. This study evaluated the in vitro effects of CTCE-9908 on B16 F10 melanoma cells with the use of mathematical modelling. Crystal violet staining was used to construct a mathematical model of CTCE-9908 B16 F10 (melanoma) and RAW 264.7 (non-cancerous macrophage) cell lines on cell viability to predict the half-maximal inhibitory concentration (IC50). Morphological changes were assessed using transmission electron microscopy. Flow cytometry was used to assess changes in cell cycle distribution, apoptosis via caspase-3, cell survival via extracellular signal-regulated kinase1/2 activation, CXCR4 activation and CXCL12 expression. Mathematical modelling predicted IC50 values from 0 to 100 h. At IC50, similar cytotoxicity between the two cell lines and ultrastructural morphological changes indicative of cell death were observed. At a concentration 10 times lower than IC50, CTCE-9908 induced inhibition of cell survival (p = 0.0133) in B16 F10 cells but did not affect caspase-3 or cell cycle distribution in either cell line. This study predicts CTCE-9908 IC50 values at various time points using mathematical modelling, revealing cytotoxicity in melanoma and non-cancerous cells. CTCE-9908 significantly inhibited melanoma cell survival at a concentration 10 times lower than the IC50 in B16 F10 cells but not RAW 264.7 cells. However, CTCE-9908 did not affect CXCR4 phosphorylation, apoptosis,\ or cell cycle distribution in either cell line.
Collapse
Affiliation(s)
- Charlise Basson
- Department of Physiology, University of Pretoria, Pretoria, South Africa
| | - Avulundiah Edwin Phiri
- Department of Mathematics, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, South Africa
| | - Manjunath Gandhi
- Department of Mathematics, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, South Africa
| | - Roumen Anguelov
- Department of Mathematics, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, South Africa
| | | | - Priyesh Bipath
- Department of Physiology, University of Pretoria, Pretoria, South Africa
| | | |
Collapse
|
10
|
Li C, Wu N, Lin X, Zhou Q, Xu M. Integrated transcriptomic and immunological profiling reveals new diagnostic and prognostic models for cutaneous melanoma. Front Pharmacol 2024; 15:1389550. [PMID: 38863979 PMCID: PMC11165152 DOI: 10.3389/fphar.2024.1389550] [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: 02/21/2024] [Accepted: 05/13/2024] [Indexed: 06/13/2024] Open
Abstract
The mortality rate associated with cutaneous melanoma (SKCM) remains alarmingly high, highlighting the urgent need for a deeper understanding of its molecular underpinnings. In our study, we leveraged bulk transcriptome sequencing data from the SKCM cohort available in public databases such as TCGA and GEO. We utilized distinct datasets for training and validation purposes and also incorporated mutation and clinical data from TCGA, along with single-cell sequencing data from GEO. Through dimensionality reduction, we annotated cell subtypes within the single-cell data and analyzed the expression of tumor-related pathways across these subtypes. We identified differentially expressed genes (DEGs) in the training set, which were further refined using the Least Absolute Shrinkage and Selection Operator (LASSO) machine learning algorithm, employing tenfold cross-validation. This enabled the construction of a prognostic model, whose diagnostic efficacy we subsequently validated. We conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses on the DEGs, and performed immunological profiling on two risk groups to elucidate the relationship between model genes and the immune responses relevant to SKCM diagnosis, treatment, and prognosis. We also knocked down the GMR6 expression level in the melanoma cells and verified its effect on cancer through multiple experiments. The results indicate that the GMR6 gene plays a role in promoting the proliferation, invasion, and migration of cancer cells in human melanoma. Our findings offer novel insights and a theoretical framework that could enhance prognosis, treatment, and drug development strategies for SKCM, potentially leading to more precise therapeutic interventions.
Collapse
Affiliation(s)
- Changchang Li
- Department of Dermatology,Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, China
| | - Nanhui Wu
- Department of Dermatopathology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoqiong Lin
- Department of Dermatology,Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, China
| | - Qiaochu Zhou
- Department of Dermatology,Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, China
| | - Mingyuan Xu
- Department of Dermatopathology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Filograna A, De Tito S, Monte ML, Oliva R, Bruzzese F, Roca MS, Zannetti A, Greco A, Spano D, Ayala I, Liberti A, Petraccone L, Dathan N, Catara G, Schembri L, Colanzi A, Budillon A, Beccari AR, Del Vecchio P, Luini A, Corda D, Valente C. Identification and characterization of a new potent inhibitor targeting CtBP1/BARS in melanoma cells. J Exp Clin Cancer Res 2024; 43:137. [PMID: 38711119 PMCID: PMC11071220 DOI: 10.1186/s13046-024-03044-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 04/10/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND The C-terminal-binding protein 1/brefeldin A ADP-ribosylation substrate (CtBP1/BARS) acts both as an oncogenic transcriptional co-repressor and as a fission inducing protein required for membrane trafficking and Golgi complex partitioning during mitosis, hence for mitotic entry. CtBP1/BARS overexpression, in multiple cancers, has pro-tumorigenic functions regulating gene networks associated with "cancer hallmarks" and malignant behavior including: increased cell survival, proliferation, migration/invasion, epithelial-mesenchymal transition (EMT). Structurally, CtBP1/BARS belongs to the hydroxyacid-dehydrogenase family and possesses a NAD(H)-binding Rossmann fold, which, depending on ligands bound, controls the oligomerization of CtBP1/BARS and, in turn, its cellular functions. Here, we proposed to target the CtBP1/BARS Rossmann fold with small molecules as selective inhibitors of mitotic entry and pro-tumoral transcriptional activities. METHODS Structured-based screening of drug databases at different development stages was applied to discover novel ligands targeting the Rossmann fold. Among these identified ligands, N-(3,4-dichlorophenyl)-4-{[(4-nitrophenyl)carbamoyl]amino}benzenesulfonamide, called Comp.11, was selected for further analysis. Fluorescence spectroscopy, isothermal calorimetry, computational modelling and site-directed mutagenesis were employed to define the binding of Comp.11 to the Rossmann fold. Effects of Comp.11 on the oligomerization state, protein partners binding and pro-tumoral activities were evaluated by size-exclusion chromatography, pull-down, membrane transport and mitotic entry assays, Flow cytometry, quantitative real-time PCR, motility/invasion, and colony assays in A375MM and B16F10 melanoma cell lines. Effects of Comp.11 on tumor growth in vivo were analyzed in mouse tumor model. RESULTS We identify Comp.11 as a new, potent and selective inhibitor of CtBP1/BARS (but not CtBP2). Comp.11 directly binds to the CtBP1/BARS Rossmann fold affecting the oligomerization state of the protein (unlike other known CtBPs inhibitors), which, in turn, hinders interactions with relevant partners, resulting in the inhibition of both CtBP1/BARS cellular functions: i) membrane fission, with block of mitotic entry and cellular secretion; and ii) transcriptional pro-tumoral effects with significantly hampered proliferation, EMT, migration/invasion, and colony-forming capabilities. The combination of these effects impairs melanoma tumor growth in mouse models. CONCLUSIONS: This study identifies a potent and selective inhibitor of CtBP1/BARS active in cellular and melanoma animal models revealing new opportunities to study the role of CtBP1/BARS in tumor biology and to develop novel melanoma treatments.
Collapse
Affiliation(s)
- Angela Filograna
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Stefano De Tito
- Molecular Cell Biology of Autophagy, The Francis Crick Institute, London, UK. The Study Has Been Previously Performed at IEOS-CNR, Naples, Italy
| | - Matteo Lo Monte
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Rosario Oliva
- Department of Chemical Sciences, University of Naples Federico II, 80126, Naples, Italy
| | - Francesca Bruzzese
- Animal Facility Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Maria Serena Roca
- Experimental Pharmacology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, 80131, Italy
| | - Antonella Zannetti
- Institute of Biostructures and Bioimaging (IBB), National Research Council (CNR), Naples, 80145, Italy
| | - Adelaide Greco
- Interdepartmental Service Center of Veterinary Radiology, University of Naples Federico II, 80137, Naples, Italy
| | - Daniela Spano
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Inmaculada Ayala
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Assunta Liberti
- National Research Council (CNR), Piazzale Aldo Moro, 700185, Rome, Italy
- Biology and Evolution of Marine Organisms (BEOM), Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Luigi Petraccone
- Department of Chemical Sciences, University of Naples Federico II, 80126, Naples, Italy
| | - Nina Dathan
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Giuliana Catara
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), 80131, Naples, Italy
| | - Laura Schembri
- National Research Council (CNR), Piazzale Aldo Moro, 700185, Rome, Italy
- Department of Pharmacy, University of Naples Federico II, 80131, Naples, Italy
| | - Antonino Colanzi
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Alfredo Budillon
- Scientific Directorate, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | | | - Pompea Del Vecchio
- Department of Chemical Sciences, University of Naples Federico II, 80126, Naples, Italy
| | - Alberto Luini
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Daniela Corda
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy.
| | - Carmen Valente
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy.
- Present address: Dompé Farmaceutici S.P.A, L'Aquila, Italy.
| |
Collapse
|
12
|
Feng Q, Xu X, Zhang S. Nrf2 protein in melanoma progression, as a new means of treatment. Pigment Cell Melanoma Res 2024; 37:247-258. [PMID: 37777339 DOI: 10.1111/pcmr.13137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 10/02/2023]
Abstract
Melanoma is a potentially lethal form of skin cancer resulting from the unlimited proliferation of melanocytes. Melanocytic lineage appears to have a greater rate of reactive oxygen species (ROS) production, possibly as a result of exposure to ultraviolet (UV) light and the production of melanin. It has been established that nuclear factor erythroid 2-related factor 2 (Nrf2) serves as a master regulator of the cellular response to oxidative stresses. Recent research has shown that the Nrf2 and its critical negative regulator Kelch-like ECH-associated protein 1 (Keap1) are misregulated in melanoma, and the Keap1-Nrf2 pathway has emerged as a promising new target for treating and preventing melanoma. In melanoma, Nrf2 may either limit tumor growth or promote its development. This review covers a wide range of topics, including the dual functions played by the Keap1-Nrf2 signaling pathway in melanoma and the most recent targeting techniques of the Nrf2.
Collapse
Affiliation(s)
- Qun Feng
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130017, China
| | - Xiaolin Xu
- Cardiology Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Shoulin Zhang
- Nephrology Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, China
| |
Collapse
|
13
|
Wongso H, Kurniawan A, Setiadi Y, Kusumaningrum CE, Widyasari EM, Wibawa TH, Mahendra I, Febrian MB, Sriyani ME, Halimah I, Daruwati I, Gunawan R, Achmad A, Nugraha DH, Lesmana R, Nugraha AS. Translocator Protein 18 kDa (TSPO): A Promising Molecular Target for Image-Guided Surgery of Solid Cancers. Adv Pharm Bull 2024; 14:86-104. [PMID: 38585455 PMCID: PMC10997928 DOI: 10.34172/apb.2024.015] [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: 05/01/2023] [Revised: 08/26/2023] [Accepted: 10/08/2023] [Indexed: 04/09/2024] Open
Abstract
The translocator protein 18-kDa (TSPO) is a mitochondrial membrane protein that is previously identified as the peripheral benzodiazepine receptor (PBR). Furthermore, it plays a significant role in a diverse range of biochemical processes, including steroidogenesis, mitochondrial cholesterol transport, cell survival and death, cell proliferation, and carcinogenesis. Several investigations also reported its roles in various types of cancers, including colorectal, brain, breast, prostate, and lung cancers, as well as melanoma. According to a previous study, the expression of TSPO was upregulated in cancer cells, which corresponds to an aggressive phenotype and/or poor prognosis. Consequently, the potential for crafting diagnostic and prognostic tools with a focus on TSPO holds great potential. In this context, several radioligands designed to target this protein have been identified, and some of the candidates have advanced to clinical trials. In recent years, the use of hybrid probes with radioactive and fluorescence molecules for image-guided surgery has exhibited promising results in animal and human studies. This indicates that the approach can serve as a valuable surgical navigator during cancer surgery. The current hybrid probes are built from various molecular platforms, including small molecules, nanoparticles, and antibodies. Although several TSPO-targeted imaging probes have been developed, their development for image-guided surgery of cancers is still limited. Therefore, this review aims to highlight recent findings on the involvement of TSPO in carcinogenesis, as well as provide a new perspective on the potential application of TSPO-targeted hybrid probes for image-guided surgery.
Collapse
Affiliation(s)
- Hendris Wongso
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
| | - Ahmad Kurniawan
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Yanuar Setiadi
- Research Center for Environmental and Clean Technology, Research Organization for Life Sciences and Environment, National Research and Innovation Agency, Puspiptek, Banten 15314, Indonesia
| | - Crhisterra E. Kusumaningrum
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Eva M. Widyasari
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Teguh H.A. Wibawa
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Isa Mahendra
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
| | - Muhamad B. Febrian
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Maula E. Sriyani
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Iim Halimah
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Isti Daruwati
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
| | - Rudi Gunawan
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
| | - Arifudin Achmad
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
- Department of Nuclear Medicine and Molecular Theranostics, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161
- Oncology and Stem Cells Working Group, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161
| | | | - Ronny Lesmana
- Department of Biomedical Science, Faculty of Medicine, Universitas Padjadjaran, Jatinangor 45363, Indonesia
- Physiology Molecular, Division of Biological Activity, Central Laboratory, Universitas Padjadjaran, Jatinangor 45363, Indonesia
- Laboratory of Sciences, Graduate School, Universitas Padjadjaran, Bandung, Indonesia
| | - Ari S. Nugraha
- Drug Utilisation and Discovery Research Group, Faculty of Pharmacy, Universitas Jember, Jember 68121, Indonesia
- School of Chemistry and Molecular Biosciences, Molecular Horizons, University of Wollongong, Wollongong, New South Wales, 2522, Australia
| |
Collapse
|
14
|
He Z, Chen M, Luo Z. Identification of immune-related genes and integrated analysis of immune-cell infiltration in melanoma. Aging (Albany NY) 2024; 16:911-927. [PMID: 38217549 PMCID: PMC10817386 DOI: 10.18632/aging.205427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/04/2023] [Indexed: 01/15/2024]
Abstract
OBJECTIVE This study was conducted to screen out immune-related genes in connection with the prognosis of melanoma, construct a prognosis model and explore the relevant mechanisms. METHODS AND MATERIALS 1973 genes associated with immune system were derived from the Immport database, and RNA-seq data of melanoma and information of patients were searched from the Xena database. Cox univariate analysis, Lasso analysis and Cox multivariate analysis were used to screen out six genes to construct the model. Then the risk scores were estimated for patients based on our constructed prognosis model. Estimate was used to affirm that the model was about immune infiltration, and CIBERSORT was used to screen out immune cells associated with prognosis. TIDE was applied to predict the efficacy of immunotherapy. Finally, GSE65904 and GSE19234 were used to confirm the effectiveness of the model. RESULTS ADCYAP1R1, GPI, NTS might cause poor prognosis while IFITM1, KIR2DL4, LIF were more likely conductive to prognosis of melanoma patients and a model of prognosis was constructed on the basis of these six genes. The effectiveness of the model has been proven by the ROC curve, and the miRNAs targeting the screened genes were found out, suggesting that the immune system might impact on the prognosis of melanoma by T cell CD8+, T cell CD4+ memory and NK cells. CONCLUSIONS In this study, the screened six genes were associated with the prognosis of melanoma, which was conductive to clinical prognostic prediction and individualized treatment strategy.
Collapse
Affiliation(s)
- Zhenghao He
- Department of Plastic Surgery, Zhongshan City People's Hospital, Zhongshan 528403, Guangdong, China
| | - Manli Chen
- Department of Plastic Surgery, Zhongshan City People's Hospital, Zhongshan 528403, Guangdong, China
| | - Zhijun Luo
- Department of Plastic Surgery, Zhongshan City People's Hospital, Zhongshan 528403, Guangdong, China
| |
Collapse
|
15
|
Almufareh MF. Unveiling the Spectrum of UV-Induced DNA Damage in Melanoma: Insights From AI-Based Analysis of Environmental Factors, Repair Mechanisms, and Skin Pigment Interactions. IEEE ACCESS 2024; 12:64837-64860. [DOI: 10.1109/access.2024.3395988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Affiliation(s)
- Maram Fahaad Almufareh
- Department of Information Systems, College of Computer and Information Sciences, Jouf University, Al Jouf, Saudi Arabia
| |
Collapse
|
16
|
Yeh YW, Hsu TW, Su YH, Wang CH, Liao PH, Chiu CF, Tseng PC, Chen TM, Lee WR, Tzeng YS. Silencing of Dicer enhances dacarbazine resistance in melanoma cells by inhibiting ADSL expression. Aging (Albany NY) 2023; 15:12873-12889. [PMID: 37976135 DOI: 10.18632/aging.205207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/15/2023] [Indexed: 11/19/2023]
Abstract
Dacarbazine (DTIC) is the primary first-line treatment for advanced-stage metastatic melanoma; thus, DTIC resistance is poses a major challenge. Therefore, investigating the mechanism underlying DTIC resistance must be investigated. Dicer, a type III cytoplasmic endoribonuclease, plays a pivotal role in the maturation of miRNAs. Aberrant Dicer expression may contribute to tumor progression, clinical aggressiveness, and poor prognosis in various tumors. Dicer inhibition led to a reduction in DTIC sensitivity and an augmentation in stemness in melanoma cells. Clinical analyses indicated a low Dicer expression level as a predictor of poor prognosis factor. Metabolic alterations in tumor cells may interfere with drug response. Adenylosuccinate lyase (ADSL) is a crucial enzyme in the purine metabolism pathway. An imbalance in ADSL may interfere with the therapeutic efficacy of drugs. We discovered that DTIC treatment enhanced ADSL expression and that Dicer silencing significantly reduced ADSL expression in melanoma cells. Furthermore, ADSL overexpression reversed Dicer silencing induced DTIC resistance and cancer stemness. These findings indicate that Dicer-mediated ADSL regulation influences DTIC sensitivity and stemness in melanoma cells.
Collapse
Affiliation(s)
- Yu-Wen Yeh
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan
- Division of Dermatology, Tri-Service General Hospital Songshan Branch, National Defense Medical Center, Taipei 105, Taiwan
| | - Tung-Wei Hsu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, Taipei 235, Taiwan
| | - Yen-Hao Su
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, Taipei 235, Taiwan
- Department of General Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Chih-Hsin Wang
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Po-Hsiang Liao
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, Taipei 235, Taiwan
| | - Ching-Feng Chiu
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Po-Chen Tseng
- Department of Ophthalmology, Taipei City Hospital, Renai Branch, Taipei 106, Taiwan
- Department of Ophthalmology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Tim-Mo Chen
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Woan-Ruoh Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yuan-Sheng Tzeng
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Department of Surgery, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung 813, Taiwan
| |
Collapse
|
17
|
Zhong L, Qian W, Gong W, Zhu L, Zhu J. Development of anoikis-related long non-coding RNA signature associated with prognosis and immune landscape in cutaneous melanoma patients. Aging (Albany NY) 2023; 15:7655-7672. [PMID: 37543428 PMCID: PMC10457054 DOI: 10.18632/aging.204932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/19/2023] [Indexed: 08/07/2023]
Abstract
BACKGROUND Anoikis is involved in many critical biological processes in tumors; however, function in CM is still unknown. In this study, the relevance between Anoikis-related lncRNAs (ARLs) and the clinicopathological characteristics of patients with CM was comprehensively assessed. METHODS Through analysis of TCGA dataset, ARLs were identified by using TCGA dataset. Based on the ARLs, a risk model was established to anticipate the prognosis of patients with CM, besides, the prediction accuracy of the model was evaluated. The immune infiltration landscape of patients with CM was assessed comprehensively, and the correlation between ARLs and immunity was elucidated. Immunotherapy and drug sensitivity analyses were applied to analyze the treatment response in patients with CM with diverse risk scores. Different subgroups were distinguished among the patients using consensus cluster analysis. RESULTS A risk model based on six ARLs was set up to obtain an accurate prediction of the prognosis of patients with CM. There were distinctions in the immune landscape among CM patients with diverse risk scores and subgroups. Six prognosis-related ARLs were highly correlated with the number of immune cells. Patients with CM with different risk scores have various sensitivities to immunotherapy and antitumor drug treatments. CONCLUSION Our newly risk model associated with ARLs has considerable prognostic value for patients with CM. Not only has the risk model high prediction accuracy but it also indicates the immune status of CM patients, which will provide a new direction for the individualized therapy of patients with CM.
Collapse
Affiliation(s)
- Like Zhong
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| | - Wenkang Qian
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| | - Wangang Gong
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| | - Li Zhu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| | - Junfeng Zhu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| |
Collapse
|
18
|
Riazi Esfahani P, Mazboudi P, Reddy AJ, Farasat VP, Guirgus ME, Tak N, Min M, Arakji GH, Patel R. Leveraging Machine Learning for Accurate Detection and Diagnosis of Melanoma and Nevi: An Interdisciplinary Study in Dermatology. Cureus 2023; 15:e44120. [PMID: 37750114 PMCID: PMC10518209 DOI: 10.7759/cureus.44120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2023] [Indexed: 09/27/2023] Open
Abstract
This study explores the application of machine learning and deep learning algorithms to facilitate the accurate diagnosis of melanoma, a type of malignant skin cancer, and benign nevi. Leveraging a dataset of 793 dermatological images from the Kaggle online platform (Google LLC, Mountain View, California, United States), we developed a model that can accurately differentiate between these lesions based on their distinctive features. The dataset was divided into training (80%), validation (10%), and testing (10%) sets to optimize model performance and ensure its generalizability. Our findings demonstrate the potential of machine learning algorithms in enhancing the efficiency and accuracy of melanoma and nevi detection, with the developed model exhibiting robust performance metrics. Nonetheless, limitations exist due to the potential lack of comprehensive representation of melanoma and nevi cases in the dataset, and variations in image quality and acquisition methods, which may influence the model's performance in real-world clinical settings. Therefore, further research, validation studies, and integration into clinical practice are necessary to ensure the reliability and generalizability of these models. This study underscores the promise of artificial intelligence in advancing dermatologic diagnostics, aiming to improve patient outcomes by supporting early detection and treatment initiation for melanoma.
Collapse
Affiliation(s)
| | - Pasha Mazboudi
- Medicine, California University of Science and Medicine, Colton, USA
| | - Akshay J Reddy
- Medicine, California University of Science and Medicine, Colton, USA
| | | | - Monica E Guirgus
- Medicine, California University of Science and Medicine, Colton, USA
| | - Nathaniel Tak
- Medicine, Midwestern University Arizona College of Osteopathic Medicine, Glendale, USA
| | - Mildred Min
- Dermatology, California Northstate University College of Medicine, Elk Grove, USA
| | - Gordon H Arakji
- Health Sciences, California Northstate University, Rancho Cordova, USA
| | - Rakesh Patel
- Internal Medicine, East Tennessee State University Quillen College of Medicine, Johnson City, USA
| |
Collapse
|
19
|
Hamid O, Hassel JC, Shoushtari AN, Meier F, Bauer TM, Salama AKS, Kirkwood JM, Ascierto PA, Lorigan PC, Mauch C, Orloff M, Evans TRJ, Holland C, Edukulla R, Abedin SE, Middleton MR. Tebentafusp in combination with durvalumab and/or tremelimumab in patients with metastatic cutaneous melanoma: a phase 1 study. J Immunother Cancer 2023; 11:e006747. [PMID: 37286303 PMCID: PMC10254987 DOI: 10.1136/jitc-2023-006747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2023] [Indexed: 06/09/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors have significantly improved outcomes in first line cutaneous melanoma. However, there is a high unmet need for patients who progress on these therapies and combination therapies are being explored to improve outcomes. Tebentafusp is a first-in-class gp100×CD3 ImmTAC bispecific that demonstrated overall survival (OS) benefit (HR 0.51) in metastatic uveal melanoma despite a modest overall response rate of 9%. This phase 1b trial evaluated the safety and initial efficacy of tebentafusp in combination with durvalumab (anti-programmed death ligand 1 (PDL1)) and/or tremelimumab (anti-cytotoxic T lymphocyte-associated antigen 4) in patients with metastatic cutaneous melanoma (mCM), the majority of whom progressed on prior checkpoint inhibitors. METHODS In this open-label, multicenter, phase 1b, dose-escalation trial, HLA-A*02:01-positive patients with mCM received weekly intravenous tebentafusp with increasing monthly doses of durvalumab and/or tremelimumab starting day 15 of each cycle. The primary objective was to identify the maximum tolerated dose (MTD) or recommended phase 2 dose for each combination. Efficacy analyses were performed in all tebentafusp with durvalumab±tremelimumab treated patients with a sensitivity analysis in those who progressed on prior anti-PD(L)1 therapy. RESULTS 85 patients were assigned to receive tebentafusp in combination with durvalumab (n=43), tremelimumab (n=13), or durvalumab and tremelimumab (n=29). Patients were heavily pretreated with a median of 3 prior lines of therapy, including 76 (89%) who received prior anti-PD(L)1. Maximum target doses of tebentafusp (68 mcg) alone or in combination with durvalumab (20 mg/kg) and tremelimumab (1 mg/kg) were tolerated; MTD was not formally identified for any arm. Adverse event profile was consistent with each individual therapy and there were no new safety signals nor treatment-related deaths. In the efficacy subset (n=72), the response rate was 14%, tumor shrinkage rate was 41% and 1-year OS rate was 76% (95% CI: 70% to 81%). The 1-year OS for triplet combination (79%; 95% CI: 71% to 86%) was similar to tebentafusp plus durvalumab (74%; 95% CI: 67% to 80%). CONCLUSION At maximum target doses, the safety of tebentafusp with checkpoint inhibitors was consistent with safety of each individual therapy. Tebentafusp with durvalumab demonstrated promising efficacy in heavily pretreated patients with mCM, including those who progressed on prior anti-PD(L)1. TRIAL REGISTRATION NUMBER NCT02535078.
Collapse
Affiliation(s)
- Omid Hamid
- The Angeles Clinic and Research Institute, a Cedars-Sinai Affiliate, Los Angeles, California, USA
| | | | - Alexander N Shoushtari
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medical College, New York, New York, USA
| | - Friedegund Meier
- Skin Cancer Center at the National Center for Tumor Diseases and University Cancer Centre, Dresden, Germany
- Department of Dermatology, University Hospital Carl Gustav Carus, Dresden, Germany
| | | | | | - John M Kirkwood
- University of Pittsburgh Medical Center Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | | | | | - Marlana Orloff
- Sidney Kimmel Cancer Center, Jefferson University, Philadelphia, Pennsylvania, USA
| | | | | | | | | | - Mark R Middleton
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, UK
| |
Collapse
|
20
|
Zhu ML, Wang LY, Bai XQ, Wu C, Liu XY. Primary malignant melanoma of the esophagus combined with squamous cell carcinoma: A case report. World J Gastrointest Surg 2023; 15:287-293. [PMID: 36896304 PMCID: PMC9988636 DOI: 10.4240/wjgs.v15.i2.287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/13/2022] [Accepted: 01/10/2023] [Indexed: 02/27/2023] Open
Abstract
BACKGROUND Primary malignant melanoma of the esophagus is a rare malignant tumor of the esophagus, and its combination with squamous cell carcinoma is also rare. Here, we report the diagnosis and treatment of a case of primary esophageal malignant melanoma combined with squamous cell carcinoma.
CASE SUMMARY A middle-aged man underwent gastroscopy for dysphagia. Gastroscopy revealed multiple bulging esophageal lesions, and after pathologic and immunohistochemical analyses, the patient was finally diagnosed with "malignant melanoma with squamous cell carcinoma". This patient received comprehensive treatment. After one year of follow-up, the patient was in good condition, and the esophageal lesions seen on gastroscopy were controlled, but unfortunately, liver metastasis occurred.
CONCLUSION When multiple esophageal lesions are present, the possibility of multiple pathological sources should be considered. This patient was diagnosed with primary esophageal malignant melanoma combined with squamous cell carcinoma.
Collapse
Affiliation(s)
- Mei-Lin Zhu
- Department of Geriatrics, Jining No. 1 People's Hospital, Jining 272000, Sahndong Province, China
| | - Ling-Yun Wang
- Department of Gastroenterology, Jining No. 1 People's Hospital, Jining 272000, Shandong Province, China
| | - Xue-Qin Bai
- Department of Pathology, Jining No. 1 People's Hospital, Jining 272000, Sahndong Province, China
| | - Chen Wu
- Department of Gastroenterology, Jining No. 1 People's Hospital, Jining 272000, Shandong Province, China
- College of Clinical Medicine, Jining Medical University, Jining 272013, Shandong Province, China
| | - Xiang-Yu Liu
- Department of Gastroenterology, Jining No. 1 People's Hospital, Jining 272000, Shandong Province, China
| |
Collapse
|
21
|
Sapozhnikov KV, Sokolova VD, Sableva NA, Tolkacheva DG. Efficacy of immunotherapy (Prolgolimab) and targeted therapy (Trametinib and Dabrafenib, Cobimetinib and Vemurafenib) in adult patients with metastatic or unresectable skin melanoma: matching-adjusted indirect comparison. JOURNAL OF MODERN ONCOLOGY 2023. [DOI: 10.26442/18151434.2022.4.202034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Aim. To assess the comparative clinical efficacy of Prolgolimab monotherapy versus combination therapy with BRAF/MEK inhibitors (Dabrafenib and Trametinib, Vemurafenib and Cobimetinb) in adult patients with metastatic or unresectable skin melanoma implementing a matching-adjusted indirect comparison (MAIC).
Materials and methods. We conducted a systematic search for randomized clinical trials of Prolgolimab, combinations of Dabrafenib and Trametinib, Cobimetinib and Vemurafenib. Unanchored MAIC was applied due to the absence of common comparator between trials. We determined effect modifiers based on an expert survey. The population from Prolgolimab studies was weighted using defined effect modifiers, followed by the approximation of survival curves.
Results. Systematic literature search revealed 4 RCTs that met the inclusion criteria: MIRACULUM, coBRIM, combi-v and combi-d. To increase the power of prolgolimab comparison, data from the observational study FORA were included in evidence synthesis and combined with data from MIRACULUM. We selected M staging and the proportion of patients with elevated LDH levels as effect modifiers. No significant differences (all p0.05) were established between Prolgolimab and combination therapy with BRAF/MEK inhibitors for both OS after 1 year and PFS outcomes after 2 years from initiation.
Discussion. Despite the inclusion of observational data and the limitations of adjusted indirect comparison method, the results of this analyses are consistent with both other comparisons of anti-PD1 inhibitors with BRAF/MEK inhibitors, and with real world data. It is necessary to recompare targeted therapy and immunotherapy after five-year follow-up period due to peculiarities of time of onset of their effect with the presence of a primary failure with a gradual exit to a long plateau on anti-PD1 inhibitors therapy.
Conclusion. In these unanchored MAICs, Prolgolimab monotherapy showed comparable efficacy with combinations of BRAF/MEK inhibitors (Dabrafenib + Trametinib, Vemurafenib + Cobimetinib) in first line therapy of patients with metastatic or unresectable melanoma. This analysis may be relevant for clinical decision-making about the choice of the first line therapy for patients with BRAF mutation.
Collapse
|
22
|
Design-of-Experiments (DoE)-Assisted Fabrication of Quercetin-Loaded Nanoemulgel and Its Evaluation against Human Skin Cancer Cell Lines. Pharmaceutics 2022; 14:pharmaceutics14112517. [PMID: 36432708 PMCID: PMC9692577 DOI: 10.3390/pharmaceutics14112517] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
Background: Quercetin (QCT) is a natural polyphenolic flavonoid showing great potential in the treatment of skin cancer. However, its use is limited owing to its poor water solubility, poor absorption, quick metabolism and excretion, as well as low stability. Preparation of nanoemulgel has been proven to be an effective approach to deliver the drugs topically due to various advantages associated with it. Objectives: This study aimed to prepare stable nanoemulgel of QCT using a Design-of-Experiments (DoE) tool for optimization, to characterize and to assess its in vivo toxicity and efficacy against human cancer cell lines in vitro. Methods: An ultrasonication emulsification method was used for the preparation of QCT-loaded nanoemulsion (QCT@NE). Box-Behnken design was used for the optimization of developed nanoemulgel. Then, in vitro characterization of prepared nanoemulsion was performed using Fourier Transform-Infra Red (FT-IR) spectroscopy, Scanning Electron Microscopy (SEM), particle size analysis, determination of zeta potential and entrapment efficiency (%EE). Further, the developed QCT-loaded nanoemulgel (QCT@NG) was characterized in vitro using texture profile analysis, viscosity and pH determination. Eventually, the cell cytotoxicity studies of the prepared nanoemulgel were performed on the skin cancer cell lines A431 followed by an acute toxicity and skin irritation study on male wistar rats in vivo. Results: The developed QCT@NE was found to be nanometric in size (173.1 ± 1.2 nm) with low polydispersity index (0.353 ± 0.13), zeta potential (-36.1 ± 5.9 mV), and showed good %EE (90.26%). The QCT@NG was found to be substantially more effective against the human skin carcinoma (A431) cell lines as compared to plain QCT with IC50 values of 108.5 and 579.0 µM, respectively. Skin irritation study showed no sign of toxicity and ensured safety for topical application. Hematological analysis revealed no significant differences between the treatment and control group in any biochemical parameter. In the nanoemulgel treatment group, there were no discernible differences in the liver enzymes, bilirubin, hemoglobin, total leukocyte and platelet counts as compared to the control group. Conclusions: The optimized QCT@NG was found to be an ideal and promising formulation for the treatment of skin cancer without showing skin irritation and organ toxicity.
Collapse
|
23
|
Andreucci E, Ruzzolini J, Bianchini F, Versienti G, Biagioni A, Lulli M, Guasti D, Nardini P, Serratì S, Margheri F, Laurenzana A, Nediani C, Peppicelli S, Calorini L. miR-214-Enriched Extracellular Vesicles Released by Acid-Adapted Melanoma Cells Promote Inflammatory Macrophage-Dependent Tumor Trans-Endothelial Migration. Cancers (Basel) 2022; 14:cancers14205090. [PMID: 36291876 PMCID: PMC9599952 DOI: 10.3390/cancers14205090] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 11/22/2022] Open
Abstract
Simple Summary Cutaneous melanoma is the most aggressive form of skin cancer with high-metastatic ability. Despite the recent advancements in melanoma treatments, the prognosis of metastatic patients remains very poor. A better understanding of the molecular mechanisms leading to melanoma dissemination is urgently needed in order to develop novel therapeutical strategies to ameliorate patients’ outcomes. Extracellular vesicles (EV) released by tumor cells are key players in metastasis development: by conveying bioactive molecules with oncogenic activity, they can modulate the surrounding—and even the distant—microenvironment and reprogram recipient cells to facilitate the metastatic cascade. Here, we show that melanoma cells release a higher amount of miR-214-enriched EV when adapted to extracellular acidosis, which promote a macrophage activation state, capable of facilitating the trans-endothelial migration of melanoma cells. Thus, we disclose a new molecular mechanism to prevent melanoma intravasation based on miR-214 targeting. Abstract The understanding of the molecular mechanisms leading to melanoma dissemination is urgently needed in view of the identification of new targets and the development of innovative strategies to improve patients’ outcomes. Within the complexity of tumor intercellular communications leading to metastatic dissemination, extracellular vesicles (EV) released by tumor cells are central players. Indeed, the ability to travel through the circulatory system conveying oncogenic bioactive molecules even at distant sites makes EV capable of modulating recipient cells to facilitate metastatic dissemination. The dynamic remodeling of the tumor microenvironment might influence, along with a number of other events, tumoral EV release. We observed that, in melanoma, extracellular acidosis increases the release of EV enriched in miR-214, an onco-miRNA involved in melanoma metastasis. Then, miR-214-enriched EV were found to induce a state of macrophage activation, leading to an overproduction of proinflammatory cytokines and nitric oxide. Such an inflammatory microenvironment was able to alter the endothelial cell permeability, thereby facilitating the trans-endothelial migration of melanoma cells, a crucial step in the metastatic cascade. The use of synthetic miR-214 inhibitors and miR-214 overexpression allowed us to demonstrate the key role of miR-214 in the EV-dependent induction of macrophage activation. Overall, our in vitro study reveals that the release of tumor miR-214-enriched EV, potentiated by adapting tumor cells to extracellular acidosis, drives a macrophage-dependent trans-endothelial migration of melanoma cells. This finding points to miR-214 as a potential new therapeutic target to prevent melanoma intravasation.
Collapse
Affiliation(s)
- Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
- Correspondence: (E.A.); (L.C.)
| | - Jessica Ruzzolini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Giampaolo Versienti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Daniele Guasti
- Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Patrizia Nardini
- Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Simona Serratì
- Laboratory of Nanotecnology, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Chiara Nediani
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
- Correspondence: (E.A.); (L.C.)
| |
Collapse
|
24
|
Romano D, García-Gutiérrez L, Aboud N, Duffy DJ, Flaherty KT, Frederick DT, Kolch W, Matallanas D. Proteasomal down-regulation of the proapoptotic MST2 pathway contributes to BRAF inhibitor resistance in melanoma. Life Sci Alliance 2022; 5:5/10/e202201445. [PMID: 36038253 PMCID: PMC9434705 DOI: 10.26508/lsa.202201445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 11/25/2022] Open
Abstract
The loss of MST2 pathway protein expression in BRAF inhibitor resistant melanoma cells is due to ubiquitination and subsequent proteasomal degradation and prevents MST2-mediated apoptosis. The RAS-RAF-MEK-ERK pathway is hyperactivated in most malignant melanomas, and mutations in BRAF or NRAS account for most of these cases. BRAF inhibitors (BRAFi) are highly efficient for treating patients with BRAFV600E mutations, but tumours frequently acquire resistance within a few months. Multiple resistance mechanisms have been identified, due to mutations or network adaptations that revive ERK signalling. We have previously shown that RAF proteins inhibit the MST2 proapoptotic pathway in a kinase-independent fashion. Here, we have investigated the role of the MST2 pathway in mediating resistance to BRAFi. We show that the BRAFV600E mutant protein, but not the wild-type BRAF protein, binds to MST2 inhibiting its proapoptotic signalling. Down-regulation of MST2 reduces BRAFi-induced apoptosis. In BRAFi-resistant cell lines, MST2 pathway proteins are down-regulated by ubiquitination and subsequent proteasomal degradation rendering cells refractory to MST2 pathway–induced apoptosis. Restoration of apoptosis can be achieved by increasing MST2 pathway protein expression using proteasome inhibitors. In summary, we show that the MST2 pathway plays a role in the acquisition of BRAFi resistance in melanoma.
Collapse
Affiliation(s)
- David Romano
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | | | - Nourhan Aboud
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | - David J Duffy
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland.,Department of Biology/Whitney Laboratory for Marine Bioscience, University of Florida, Gainesville, FL, USA
| | | | | | - Walter Kolch
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland .,Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - David Matallanas
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
25
|
Lopes J, Rodrigues CMP, Gaspar MM, Reis CP. Melanoma Management: From Epidemiology to Treatment and Latest Advances. Cancers (Basel) 2022; 14:4652. [PMID: 36230575 PMCID: PMC9562203 DOI: 10.3390/cancers14194652] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022] Open
Abstract
Melanoma is the deadliest skin cancer, whose morbidity and mortality indicators show an increasing trend worldwide. In addition to its great heterogeneity, melanoma has a high metastatic potential, resulting in very limited response to therapies currently available, which were restricted to surgery, radiotherapy and chemotherapy for many years. Advances in knowledge about the pathophysiological mechanisms of the disease have allowed the development of new therapeutic classes, such as immune checkpoint and small molecule kinase inhibitors. However, despite the incontestable progress in the quality of life and survival rates of the patients, effectiveness is still far from desired. Some adverse side effects and resistance mechanisms are the main barriers. Thus, the search for better options has resulted in many clinical trials that are now investigating new drugs and/or combinations. The low water solubility of drugs, low stability and rapid metabolism limit the clinical potential and therapeutic use of some compounds. Thus, the research of nanotechnology-based strategies is being explored as the basis for the broad application of different types of nanosystems in the treatment of melanoma. Future development focus on challenges understanding the mechanisms that make these nanosystems more effective.
Collapse
Affiliation(s)
- Joana Lopes
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Cecília M. P. Rodrigues
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Maria Manuela Gaspar
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Catarina Pinto Reis
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
- Instituto de Biofísica e Engenharia Biomédica, IBEB, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|
26
|
How to Treat Melanoma? The Current Status of Innovative Nanotechnological Strategies and the Role of Minimally Invasive Approaches like PTT and PDT. Pharmaceutics 2022; 14:pharmaceutics14091817. [PMID: 36145569 PMCID: PMC9504126 DOI: 10.3390/pharmaceutics14091817] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 12/13/2022] Open
Abstract
Melanoma is the most aggressive type of skin cancer, the incidence and mortality of which are increasing worldwide. Its extensive degree of heterogeneity has limited its response to existing therapies. For many years the therapeutic strategies were limited to surgery, radiotherapy, and chemotherapy. Fortunately, advances in knowledge have allowed the development of new therapeutic strategies. Despite the undoubted progress, alternative therapies are still under research. In this context, nanotechnology is also positioned as a strong and promising tool to develop nanosystems that act as drug carriers and/or light absorbents to potentially improve photothermal and photodynamic therapies outcomes. This review describes the latest advances in nanotechnology field in the treatment of melanoma from 2011 to 2022. The challenges in the translation of nanotechnology-based therapies to clinical applications are also discussed. To sum up, great progress has been made in the field of nanotechnology-based therapies, and our understanding in this field has greatly improved. Although few therapies based on nanoparticulate systems have advanced to clinical trials, it is expected that a large number will come into clinical use in the near future. With its high sensitivity, specificity, and multiplexed measurement capacity, it provides great opportunities to improve melanoma treatment, which will ultimately lead to enhanced patient survival rates.
Collapse
|
27
|
The Impact of SARS-CoV-2 Pandemic on Patients with Malignant Melanoma at a Romanian Academic Center: A Four-Year Retrospective Analysis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19148499. [PMID: 35886351 PMCID: PMC9317187 DOI: 10.3390/ijerph19148499] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022]
Abstract
Considering cancer patients may be at an increased risk of severe COVID-19 disease, their oncologic treatment cannot be delayed without risking their oncologic outcomes. Considering this, a comprehensive evaluation is required for the management of malignant diseases such as melanoma. The current study aimed to assess the impact of the COVID-19 pandemic on the delivery of cancer care services for patients diagnosed with malignant melanoma in Romania; to document the difference in patients’ addressability and melanoma staging between the pandemic and pre-pandemic periods; as well as to determine the risk factors responsible for disease progression during the pandemic. We developed a retrospective analysis using a monocentric hospital database to compare the final 24 months of the pre-pandemic era to the first 24 months of the COVID-19 pandemic. All outpatients and inpatients with a diagnosis of malignant melanoma were screened during the study period and included in the analysis if matching the inclusion criteria. A total of 301 patients were included in the study, with 163 cases identified in the 24 months before the COVID-19 pandemic and 138 patients during the first 24 months of the pandemic. It was observed during the first two lockdown periods from March to May 2020, and, respectively, from October to December 2020, that significantly fewer patients with malignant melanoma presented for specialized medical care, while there was a statistically significantly lower proportion of outpatients due to COVID-19 restrictions (18.1% vs. 42.9%). The average Breslow depth was 1.1 mm before the pandemic, compared with 1.8 mm during the pandemic (p-value < 0.001). Third-stage patients were the most prevalent during both study periods, although with a statistically significant difference during the pandemic, with an increase from 90 (55.2%) patients to 94 (68.1%) (p-value < 0.001). The significant risk factors for disease progression were advanced AJCC stage (HR = 3.48), high Breslow index (HR = 3.19), postponed treatment (HR = 2.46), missed appointments (HR = 2.31), anemia at presentation (HR = 1.60), and patient’s age (HR = 1.57). After the pandemic limitations are brought to an end, a broad skin-cancer-screening campaign is warranted to detect the missed cases during COVID-19.
Collapse
|
28
|
Fractal Dimension Analysis of Melanocytic Nevi and Melanomas in Normal and Polarized Light-A Preliminary Report. LIFE (BASEL, SWITZERLAND) 2022; 12:life12071008. [PMID: 35888097 PMCID: PMC9318244 DOI: 10.3390/life12071008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/22/2022] [Accepted: 06/30/2022] [Indexed: 11/21/2022]
Abstract
Clinical diagnosis of pigmented lesions can be a challenge in everyday practice. Benign and dysplastic nevi and melanomas may have similar clinical presentations, but completely different prognoses. Fractal dimensions of shape and texture can describe the complexity of the pigmented lesion structure. This study aims to apply fractal dimension analysis to differentiate melanomas, dysplastic nevi, and benign nevi in polarized and non-polarized light. A total of 87 Eighty-four patients with 97 lesions were included in this study. All examined lesions were photographed under polarized and non-polarized light, surgically removed, and examined by a histopathologist to establish the correct diagnosis. The obtained images were then processed and analyzed. Area, perimeter, and fractal dimensions of shape and texture were calculated for all the lesions under polarized and non-polarized light. The fractal dimension of shape in polarized light enables differentiating melanomas, dysplastic nevi, and benign nevi. It also makes it possible to distinguish melanomas from benign and dysplastic nevi under non-polarized light. The fractal dimension of texture allows distinguishing melanomas from benign and dysplastic nevi under polarized light. All examined parameters of shape and texture can be used for developing an automatic computer-aided diagnosis system. Polarized light is superior to non-polarized light for imaging texture details.
Collapse
|
29
|
Light-emitting diode photomodulation of uterine adenocarcinoma cells inhibited angiogenesis capacity via the regulation of exosome biogenesis. Lasers Med Sci 2022; 37:3193-3201. [PMID: 35727394 DOI: 10.1007/s10103-022-03597-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/13/2022] [Indexed: 10/18/2022]
Abstract
This study was conducted to investigate the inhibitory effects of light-emitting diodes (LEDs) on exosome biogenesis and angiogenesis capacity in Ishikawa endometrial cancer cells. To this end, cells were exposed to different energy densities (fluences) of 4, 8, 16, 32, and 64 J/cm2 for 5 days (once every 24 h), and cell viability was determined using an MTT assay. Based on data from the MTT panel, cells were exposed to 4 and 16 J/cm2 for subsequent analyses. Exosome biogenesis was also monitored via monitoring the expression of CD63, ALIX, and Rab27a and b. The size and morphology of exosomes in the supernatant were measured using scanning electron microscopy (SEM), and dynamic light scattering (DLS). Using Transwell insert, the migration capacity of these cells was studied. The angiogenic effects of irradiated Ishikawa cell secretome at different fluences were monitored on human endothelial cells using in vitro tubulogenesis. Results indicated LED can reduce the viability of Ishikawa cells in a dose-dependent manner. According to our data, 4 and 64 J/cm2 groups exhibited minimum and maximum cytotoxic effects compared to the control cells. Data revealed a close proportional relationship between the power of laser and exosome average size compared to the non-treated control cells (p < 0.05). Real-time PCR analysis showed the suppression of Rab27b and up-regulation of Rab27a in irradiated cells exposed to 4 and 16 J/cm2 (p < 0.05). These effects were evident in the 16 J/cm2 group. Likewise, LED can inhibit the migration of Ishikawa cells in a dose-dependent manner (p < 0.05). Tubulogenesis activity of endothelial cells was suppressed after incubation with the secretome of irradiated Ishikawa cells (p < 0.05). These data showed tumoricidal properties of LED irradiation on human adenocarcinoma Ishikawa cells via the inhibition of exosome biogenesis and suppression of angiogenesis capacity.
Collapse
|
30
|
Fine GC, Covington MF, Koppula BR, Salem AE, Wiggins RH, Hoffman JM, Morton KA. PET-CT in Clinical Adult Oncology-VI. Primary Cutaneous Cancer, Sarcomas and Neuroendocrine Tumors. Cancers (Basel) 2022; 14:2835. [PMID: 35740501 PMCID: PMC9221374 DOI: 10.3390/cancers14122835] [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: 04/29/2022] [Revised: 06/03/2022] [Accepted: 06/06/2022] [Indexed: 11/16/2022] Open
Abstract
PET-CT is an advanced imaging modality with many oncologic applications, including staging, therapeutic assessment, restaging and surveillance for recurrence. The goal of this series of six review articles is to provide practical information to providers and imaging professionals regarding the best use of PET-CT for specific oncologic indications, the potential pitfalls and nuances that characterize these applications, and guidelines for image interpretation. Tumor-specific clinical information and representative PET-CT images are provided. The current, sixth article in this series addresses PET-CT in an evaluation of aggressive cutaneous malignancies, sarcomas and neuroendocrine tumors. A discussion of the role of FDG PET for all types of tumors in these categories is beyond the scope of this review. Rather, this article focuses on the most common malignancies in adult patients encountered in clinical practice. It also focuses on Food and Drug Agency (FDA)-approved and clinically available radiopharmaceuticals rather than research tracers or those requiring a local cyclotron. This information will serve as a guide to primary providers for the appropriate role of PET-CT in managing patients with cutaneous malignancies, sarcomas and neuroendocrine tumors. The nuances of PET-CT interpretation as a practical guide for imaging providers, including radiologists, nuclear medicine physicians and their trainees, are also addressed.
Collapse
Affiliation(s)
- Gabriel C. Fine
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (G.C.F.); (M.F.C.); (B.R.K.); (A.E.S.); (R.H.W.); (J.M.H.)
| | - Matthew F. Covington
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (G.C.F.); (M.F.C.); (B.R.K.); (A.E.S.); (R.H.W.); (J.M.H.)
| | - Bhasker R. Koppula
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (G.C.F.); (M.F.C.); (B.R.K.); (A.E.S.); (R.H.W.); (J.M.H.)
| | - Ahmed Ebada Salem
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (G.C.F.); (M.F.C.); (B.R.K.); (A.E.S.); (R.H.W.); (J.M.H.)
- Faculty of Medicine, Department of Radiodiagnosis and Intervention, Alexandria University, Alexandria 21526, Egypt
| | - Richard H. Wiggins
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (G.C.F.); (M.F.C.); (B.R.K.); (A.E.S.); (R.H.W.); (J.M.H.)
| | - John M. Hoffman
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (G.C.F.); (M.F.C.); (B.R.K.); (A.E.S.); (R.H.W.); (J.M.H.)
| | - Kathryn A. Morton
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (G.C.F.); (M.F.C.); (B.R.K.); (A.E.S.); (R.H.W.); (J.M.H.)
- Intermountain Healthcare Hospitals, Summit Physician Specialists, Murray, UT 84123, USA
| |
Collapse
|
31
|
Rok J, Rzepka Z, Kowalska J, Banach K, Beberok A, Wrześniok D. The Anticancer Potential of Doxycycline and Minocycline-A Comparative Study on Amelanotic Melanoma Cell Lines. Int J Mol Sci 2022; 23:ijms23020831. [PMID: 35055021 PMCID: PMC8775630 DOI: 10.3390/ijms23020831] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/02/2022] [Accepted: 01/11/2022] [Indexed: 12/04/2022] Open
Abstract
Malignant melanoma is still a serious medical problem. Relatively high mortality, a still-growing number of newly diagnosed cases, and insufficiently effective methods of therapy necessitate melanoma research. Tetracyclines are compounds with pleiotropic pharmacological properties. Previously published studies on melanotic melanoma cells ascertained that minocycline and doxycycline exerted an anti-melanoma effect. The purpose of the study was to assess the anti-melanoma potential and mechanisms of action of minocycline and doxycycline using A375 and C32 human amelanotic melanoma cell lines. The obtained results indicate that the tested drugs inhibited proliferation, decreased cell viability, and induced apoptosis in amelanotic melanoma cells. The treatment caused changes in the cell cycle profile and decreased the intracellular level of reduced thiols and mitochondrial membrane potential. The exposure of A375 and C32 cells to minocycline and doxycycline triggered the release of cytochrome c and activated initiator and effector caspases. The anti-melanoma effect of analyzed drugs appeared to be related to the up-regulation of ERK1/2 and MITF. Moreover, it was noticed that minocycline and doxycycline increased the level of LC3A/B, an autophagy marker, in A375 cells. In summary, the study showed the pleiotropic anti-cancer action of minocycline and doxycycline against amelanotic melanoma cells. Considering all results, it could be concluded that doxycycline was a more potent drug than minocycline.
Collapse
Affiliation(s)
- Jakub Rok
- Correspondence: ; Tel.: +48-32-364-15-47
| | | | | | | | | | | |
Collapse
|
32
|
Catalani E, Giovarelli M, Zecchini S, Perrotta C, Cervia D. Oxidative Stress and Autophagy as Key Targets in Melanoma Cell Fate. Cancers (Basel) 2021; 13:cancers13225791. [PMID: 34830947 PMCID: PMC8616245 DOI: 10.3390/cancers13225791] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/10/2021] [Accepted: 11/17/2021] [Indexed: 01/18/2023] Open
Abstract
Melanoma originates from the malignant transformation of melanocytes and is one of the most aggressive forms of cancer. The recent approval of several drugs has increased the chance of survival although a significant subset of patients with metastatic melanoma do not show a long-lasting response to these treatments. The complex cross-talk between oxidative stress and the catabolic process autophagy seems to play a central role in all aspects of melanoma pathophysiology, from initiation to progression and metastasis, including drug resistance. However, determining the fine role of autophagy in cancer death and in response to redox disruption is still a fundamental challenge in order to advance both basic and translational aspects of this field. In order to summarize the interactions among reactive oxygen and nitrogen species, autophagy machinery and proliferation/growth/death/apoptosis/survival, we provide here a narrative review of the preclinical evidence for drugs/treatments that modulate oxidative stress and autophagy in melanoma cells. The significance and the potential for pharmacological targeting (also through multiple and combination approaches) of these two different events, which can contribute independently or simultaneously to the fate of melanoma, may help to define new processes and their interconnections underlying skin cancer biology and unravel new reliable approaches.
Collapse
Affiliation(s)
- Elisabetta Catalani
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, Largo dell’Università snc, 01100 Viterbo, Italy;
| | - Matteo Giovarelli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, Via G.B. Grassi 74, 20157 Milano, Italy; (M.G.); (S.Z.)
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, Via G.B. Grassi 74, 20157 Milano, Italy; (M.G.); (S.Z.)
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, Via G.B. Grassi 74, 20157 Milano, Italy; (M.G.); (S.Z.)
- Correspondence: (C.P.); (D.C.)
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, Largo dell’Università snc, 01100 Viterbo, Italy;
- Correspondence: (C.P.); (D.C.)
| |
Collapse
|