1
|
Jing F, Pazhava A, Shachaf C, Meves A. The Role of Stromal Tumor Environment in Advancing Skin Cancer Diagnosis and Treatment. Int J Dermatol 2025. [PMID: 40369720 DOI: 10.1111/ijd.17827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/13/2025] [Accepted: 04/18/2025] [Indexed: 05/16/2025]
Abstract
The stromal tumor environment (STE) plays a vital yet often underrecognized role in cancer progression, shaping tumor development. It influences complex interactions among stromal cells, immune cells, extracellular matrix (ECM) components, and signaling molecules. Stromal cells regulate integrin signaling and are involved in remodeling the tumor microenvironment, driving critical processes such as cell migration, survival, angiogenesis, and proliferation. Advances in understanding the STE are creating new opportunities for early cancer detection and treatment. Innovations in molecular imaging, particularly for melanoma, are transforming cancer diagnostics by providing deeper insights into tumor biology, cellular interactions, and angiogenesis, paving the way for more personalized and timely therapeutic interventions. Among these advancements, in vivo skin fluorescent imaging (SFI) stands out as a powerful tool, offering real-time molecular insights at the point of care with the ability to improve patient management.
Collapse
Affiliation(s)
- Frank Jing
- Department of Dermatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ani Pazhava
- Department of Dermatology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Alexander Meves
- Department of Dermatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
2
|
DU JUNWEI, ZHANG QIANG, ZHANG JING, MAIHEMUTI MAIERDANJIANG, HE HAIYANG, JIANG RENBING. Melanoma cell line-derived exosomal miR-424-5p: a key promoter of angiogenesis through LATS2 interaction. Oncol Res 2025; 33:357-367. [PMID: 39866229 PMCID: PMC11753990 DOI: 10.32604/or.2024.050878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/20/2024] [Indexed: 01/28/2025] Open
Abstract
Objectives Melanoma is a highly aggressive and metastatic form of cancer, and the role of exosomal microRNAs (miRNAs) in its progression remains largely unexplored. This study aimed to investigate the effects of melanoma cell-derived exosomal miR-424-5p on angiogenesis and its underlying mechanisms. Methods Exosomes were isolated from melanoma cell lines A375 and A2058, and their effects on the proliferation, migration, and angiogenesis of human umbilical vein endothelial cells (HUVECs) were examined. The interaction between miR-424-5p and its target gene, large tumor suppressor kinase 2 (LATS2), was analyzed using luciferase reporter assays and functional experiments. In vivo, tumor growth and angiogenesis were studied in a xenograft model using nude mice. Results Melanoma cell-derived exosomes could be internalized by HUVECs, which promoted proliferation, migration, and angiogenesis. miR-424-5p was highly expressed in melanoma cells and their exosomes, and its inhibition in exosomes suppressed HUVEC proliferation, migration, and angiogenesis. LATS2 was identified as a direct target of miR-424-5p, and its silencing reversed the inhibitory effects of miR-424-5p inhibition on HUVEC functions. In vivo, exosomes derived from miR-424-5p-inhibited melanoma cells suppressed tumor growth and angiogenesis in xenograft models. Conclusions Melanoma cell-derived exosomal miR-424-5p promotes angiogenesis by targeting LATS2, contributing to melanoma progression. Targeting the exosomal miR-424-5p/LATS2 axis could be a potential therapeutic strategy for melanoma.
Collapse
Affiliation(s)
- JUNWEI DU
- Department of Bone and Soft Tissue Tumors and Melanoma, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830000, China
| | - QIANG ZHANG
- Department of Bone and Soft Tissue Tumors and Melanoma, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830000, China
| | - JING ZHANG
- Department of Bone and Soft Tissue Tumors and Melanoma, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830000, China
| | - MAIERDANJIANG MAIHEMUTI
- Department of Bone and Soft Tissue Tumors and Melanoma, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830000, China
| | - HAIYANG HE
- Department of Bone and Soft Tissue Tumors and Melanoma, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830000, China
| | - RENBING JIANG
- Department of Bone and Soft Tissue Tumors and Melanoma, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830000, China
| |
Collapse
|
3
|
Comes MC, Fucci L, Strippoli S, Bove S, Cazzato G, Colangiuli C, Risi ID, Roma ID, Fanizzi A, Mele F, Ressa M, Saponaro C, Soranno C, Tinelli R, Guida M, Zito A, Massafra R. An artificial intelligence-based model exploiting H&E images to predict recurrence in negative sentinel lymph-node melanoma patients. J Transl Med 2024; 22:838. [PMID: 39267101 PMCID: PMC11391752 DOI: 10.1186/s12967-024-05629-2] [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: 03/07/2024] [Accepted: 08/18/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Risk stratification and treatment benefit prediction models are urgent to improve negative sentinel lymph node (SLN-) melanoma patient selection, thus avoiding costly and toxic treatments in patients at low risk of recurrence. To this end, the application of artificial intelligence (AI) could help clinicians to better calculate the recurrence risk and choose whether to perform adjuvant therapy. METHODS We made use of AI to predict recurrence-free status (RFS) within 2-years from diagnosis in 94 SLN- melanoma patients. In detail, we detected quantitative imaging information from H&E slides of a cohort of 71 SLN- melanoma patients, who registered at Istituto Tumori "Giovanni Paolo II" in Bari, Italy (investigational cohort, IC). For each slide, two expert pathologists firstly annotated two Regions of Interest (ROIs) containing tumor cells alone (TUMOR ROI) or with infiltrating cells (TUMOR + INF ROI). In correspondence of the two kinds of ROIs, two AI-based models were developed to extract information directly from the tiles in which each ROI was automatically divided. This information was then used to predict RFS. Performances of the models were computed according to a 5-fold cross validation scheme. We further validated the prediction power of the two models on an independent external validation cohort of 23 SLN- melanoma patients (validation cohort, VC). RESULTS The TUMOR ROIs have revealed more informative than the TUMOR + INF ROIs. An Area Under the Curve (AUC) value of 79.1% and 62.3%, a sensitivity value of 81.2% and 76.9%, a specificity value of 70.0% and 43.3%, an accuracy value of 73.2% and 53.4%, were achieved on the TUMOR and TUMOR + INF ROIs extracted for the IC cohort, respectively. An AUC value of 76.5% and 65.2%, a sensitivity value of 66.7% and 41.6%, a specificity value of 70.0% and 55.9%, an accuracy value of 70.0% and 56.5%, were achieved on the TUMOR and TUMOR + INF ROIs extracted for the VC cohort, respectively. CONCLUSIONS Our approach represents a first effort to develop a non-invasive prognostic method to better define the recurrence risk and improve the management of SLN- melanoma patients.
Collapse
Affiliation(s)
- Maria Colomba Comes
- Laboratorio di Biostatistica e Bioinformatica, I.R.C.C.S. Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - Livia Fucci
- Unità Operativa Complessa di Anatomia Patologica, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Sabino Strippoli
- Unità Operativa Tumori Rari e Melanoma, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Samantha Bove
- Laboratorio di Biostatistica e Bioinformatica, I.R.C.C.S. Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - Gerardo Cazzato
- Dipartimento di Medicina di Precisione e Rigenerativa e Area Jonica, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Carmen Colangiuli
- Laboratorio di Biostatistica e Bioinformatica, I.R.C.C.S. Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - Ivana De Risi
- Unità Operativa Tumori Rari e Melanoma, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Ileana De Roma
- Unità Operativa Tumori Rari e Melanoma, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Annarita Fanizzi
- Laboratorio di Biostatistica e Bioinformatica, I.R.C.C.S. Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - Fabio Mele
- Unità Operativa Complessa di Anatomia Patologica, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Maurizio Ressa
- Unità Operativa Complessa di Chirurgica Plastica e Ricostruttiva, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Concetta Saponaro
- Unità Operativa Complessa di Anatomia Patologica, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | | | - Rosita Tinelli
- Unità Operativa Complessa di Anatomia Patologica, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Michele Guida
- Unità Operativa Tumori Rari e Melanoma, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy.
| | - Alfredo Zito
- Unità Operativa Complessa di Anatomia Patologica, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Raffaella Massafra
- Laboratorio di Biostatistica e Bioinformatica, I.R.C.C.S. Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| |
Collapse
|
4
|
Tian X, Si Q, Liu M, Shi J, Zhao R, Xiong Y, Yu L, Cui H, Guan H. Advance in vasculogenic mimicry in ovarian cancer (Review). Oncol Lett 2023; 26:456. [PMID: 37736556 PMCID: PMC10509778 DOI: 10.3892/ol.2023.14043] [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: 02/19/2023] [Accepted: 08/21/2023] [Indexed: 09/23/2023] Open
Abstract
Ovarian cancer (OC) is a common and highly prevalent malignant tumor in women, associated with a high mortality rate, easy recurrence and easy metastasis, which is predominantly at an advanced stage when detected in patients. This renders the cancer more difficult to treat, and consequently it is also associated with a low survival rate, being the malignancy with the highest mortality rate among the various gynecological tumors. As an important factor affecting the development and metastasis of OC, understanding the underlying mechanism(s) through which it is formed and developed is crucial in terms of its treatment. At present, the therapeutic methods of angiogenic mimicry for OC remain in the preliminary stages of exploration and have not been applied in actual clinical practice. In the present review, various signaling pathways and factors affecting angiogenic mimicry in OC were described, and the chemical synthetic drugs, natural compound extracts, small-molecule protein antibodies and their associated targets, and so on, that target angiogenic mimicry in the treatment of OC, were discussed. The purpose of this review was to provide new research ideas and potential theoretical support for the discovery of novel therapeutic targets for OC that may be applied in the clinic, with the aim of effectively reducing its metastasis and recurrence rates.
Collapse
Affiliation(s)
- Xinyuan Tian
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010107, P.R. China
| | - Qin Si
- Scientific Research Department, Inner Mongolia Cancer Hospital and Affiliated People's Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010020, P.R. China
| | - Menghe Liu
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010107, P.R. China
| | - Jianping Shi
- School of Traditional Chinese Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010107, P.R. China
| | - Rongwei Zhao
- Department of Obstetrics and Gynecology, Inner Mongolia Medical University, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
| | - Yang Xiong
- Department of Hepatobiliary Surgery, General Surgery Department of Ordos Central Hospital, Ordos, Inner Mongolia Autonomous Region 017000, P.R. China
| | - Lei Yu
- Department of Pharmacy, Traditional Chinese Medicine Hospital of Inner Mongolia Autonomous Region, Hohhot, Inner Mongolia Autonomous Region 010020, P.R. China
| | - Hongwei Cui
- Scientific Research Department, Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
| | - Haibin Guan
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010107, P.R. China
| |
Collapse
|
5
|
Michielon E, López González M, Stolk DA, Stolwijk JGC, Roffel S, Waaijman T, Lougheed SM, de Gruijl TD, Gibbs S. A Reconstructed Human Melanoma-in-Skin Model to Study Immune Modulatory and Angiogenic Mechanisms Facilitating Initial Melanoma Growth and Invasion. Cancers (Basel) 2023; 15:2849. [PMID: 37345186 DOI: 10.3390/cancers15102849] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/08/2023] [Accepted: 05/17/2023] [Indexed: 06/23/2023] Open
Abstract
Invasion, immune modulation, and angiogenesis are crucial in melanoma progression. Studies based on animals or two-dimensional cultures poorly recapitulate the tumor-microenvironmental cross-talk found in humans. This highlights a need for more physiological human models to better study melanoma features. Here, six melanoma cell lines (A375, COLO829, G361, MeWo, RPMI-7951, and SK-MEL-28) were used to generate an in vitro three-dimensional human melanoma-in-skin (Mel-RhS) model and were compared in terms of dermal invasion and immune modulatory and pro-angiogenic capabilities. A375 displayed the most invasive phenotype by clearly expanding into the dermal compartment, whereas COLO829, G361, MeWo, and SK-MEL-28 recapitulated to different extent the initial stages of melanoma invasion. No nest formation was observed for RPMI-7951. Notably, the integration of A375 and SK-MEL-28 cells into the model resulted in an increased secretion of immune modulatory factors (e.g., M-CSF, IL-10, and TGFβ) and pro-angiogenic factors (e.g., Flt-1 and VEGF). Mel-RhS-derived supernatants induced endothelial cell sprouting in vitro. In addition, observed A375-RhS tissue contraction was correlated to increased TGFβ release and α-SMA expression, all indicative of differentiation of fibroblasts into cancer-associated fibroblast-like cells and reminiscent of epithelial-to-mesenchymal transition, consistent with A375's most prominent invasive behavior. In conclusion, we successfully generated several Mel-RhS models mimicking different stages of melanoma progression, which can be further tailored for future studies to investigate individual aspects of the disease and serve as three-dimensional models to assess efficacy of therapeutic strategies.
Collapse
Affiliation(s)
- Elisabetta Michielon
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location Vrije Universiteit, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
| | - Marta López González
- Amsterdam Institute for Infection and Immunity, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit, 1105 AZ Amsterdam, The Netherlands
| | - Dorian A Stolk
- Amsterdam Institute for Infection and Immunity, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit, 1105 AZ Amsterdam, The Netherlands
| | - Joeke G C Stolwijk
- Amsterdam Institute for Infection and Immunity, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit, 1105 AZ Amsterdam, The Netherlands
| | - Sanne Roffel
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location Vrije Universiteit, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, 1105 AZ Amsterdam, The Netherlands
| | - Taco Waaijman
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location Vrije Universiteit, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, 1105 AZ Amsterdam, The Netherlands
| | - Sinéad M Lougheed
- Amsterdam Institute for Infection and Immunity, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit, 1105 AZ Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Amsterdam Institute for Infection and Immunity, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit, 1105 AZ Amsterdam, The Netherlands
| | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location Vrije Universiteit, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, 1105 AZ Amsterdam, The Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
6
|
Shen Y, Li M, Liao L, Gao S, Wang Y. Plasma exosome-derived connexin43 as a promising biomarker for melanoma patients. BMC Cancer 2023; 23:242. [PMID: 36918803 PMCID: PMC10012581 DOI: 10.1186/s12885-023-10705-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/06/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND To examine the levels of exosome-derived connexin 43 (Cx43) in plasma and estimate its forecast value in patients with melanoma. METHODS We measured the plasma exosome-derived Cx43 levels in the plasma of 112 melanoma patients and 50 healthy controls. RESULTS The plasma exosome-derived Cx43 levels in patients with melanoma were substantially downregulated as opposed to the levels in healthy controls (P < 0.001). Kaplan-Meier analysis indicated that overall survival (OS) and disease-free survival (DFS) were poorer in patients with melanoma who exhibited lower levels of plasma exosome-derived Cx43 (both P < 0.001). The levels of plasma exosome-derived Cx43 were considerably elevated in patients with melanoma whose tumor was situated in the skin, tumor size < 10 cm, with Clark level I-III, TNM stages IIb-IV, and had no lymph node metastasis as opposed to patients whose tumor was situated in the viscera or mucosa, tumor size ≥ 10 cm, Clark level IV-V, TNM stages IIb-IV and had lymph node metastasis (all P < 0.05). The receiver operating characteristic (ROC) of plasma exosome-derived Cx43 for forecasting 5-year DFS in patients with melanoma was 0.78 (95% confidence interval (CI): 0.70-0.86), with a specificity of 77.78% and a sensitivity of 81.55%. The ROC of plasma exosome-derived Cx43 for forecasting 5-year OS of patients with melanoma was 0.77 (95% CI: 0.68-0.84), with a specificity of 80.0% and sensitivity of 65.98%. CONCLUSION The overall findings indicated that the levels of plasma exosome-derived Cx43 in patients with melanoma were considerably downregulated. It can therefore be inferred that the levels of plasma exosome-derived Cx43 might be a prospective prognostic indicator for 5 5-year OS and 5-year DFS of patients with melanoma.
Collapse
Affiliation(s)
- Yue Shen
- Department of Dermatology, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, 242 Guangji Road, Suzhou, Jiangsu, 215008, China
| | - Ming Li
- Department of Pathology, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, 242 Guangji Road, Suzhou, Jiangsu, 215008, China
| | - Li Liao
- Department of Dermatology, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, 242 Guangji Road, Suzhou, Jiangsu, 215008, China
| | - Suyue Gao
- Department of Dermatology, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, 242 Guangji Road, Suzhou, Jiangsu, 215008, China
| | - Yongzhen Wang
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, 26 Daoqian Street, Suzhou, Jiangsu, 215008, China.
| |
Collapse
|
7
|
Ziogas DC, Theocharopoulos C, Koutouratsas T, Haanen J, Gogas H. Mechanisms of resistance to immune checkpoint inhibitors in melanoma: What we have to overcome? Cancer Treat Rev 2023; 113:102499. [PMID: 36542945 DOI: 10.1016/j.ctrv.2022.102499] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022]
Abstract
Marching into the second decade after the approval of ipilimumab, it is clear that immune checkpoint inhibitors (ICIs) have dramatically improved the prognosis of melanoma. Although the current edge is already high, with a 4-year OS% of 77.9% for adjuvant nivolumab and a 6.5-year OS% of 49% for nivolumab/ipilimumab combination in the metastatic setting, a high proportion of patients with advanced melanoma have no benefit from immunotherapy, or experience an early disease relapse/progression in the first few months of treatment, surviving much less. Reasonably, the primary and acquired resistance to ICIs has entered into the focus of clinical research with positive (e.g., nivolumab and relatlimab combination) and negative feedbacks (e.g., nivolumab with pegylated-IL2, pembrolizumab with T-VEC, nivolumab with epacadostat, and combinatorial triplets of BRAF/MEK inhibitors with immunotherapy). Many intrinsic (intracellular or intra-tumoral) but also extrinsic (systematic) events are considered to be involved in the development of this resistance to ICIs: i) melanoma cell immunogenicity (e.g., tumor mutational burden, antigen-processing machinery and immunogenic cell death, neoantigen affinity and heterogeneity, genomic instability, melanoma dedifferentiation and phenotypic plasticity), ii) immune cell trafficking, T-cell priming, and cell death evasion, iii) melanoma neovascularization, cellular TME components(e.g., Tregs, CAFs) and extracellular matrix modulation, iv) metabolic antagonism in the TME(highly glycolytic status, upregulated CD39/CD73/adenosine pathway, iDO-dependent tryptophan catabolism), v) T-cell exhaustion and negative immune checkpoints, and vi) gut microbiota. In the present overview, we discuss how these parameters compromise the efficacy of ICIs, with an emphasis on the lessons learned by the latest melanoma studies; and in parallel, we describe the main ongoing approaches to overcome the resistance to immunotherapy. Summarizing this information will improve the understanding of how these complicated dynamics contribute to immune escape and will help to develop more effective strategies on how anti-tumor immunity can surpass existing barriers of ICI-refractory melanoma.
Collapse
Affiliation(s)
- Dimitrios C Ziogas
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Charalampos Theocharopoulos
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Tilemachos Koutouratsas
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - John Haanen
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| |
Collapse
|
8
|
Zarin B, Nedaeinia R, Laher I, Manian M, Javanmard SH. The effects of ALK5 inhibition and simultaneous inhibition or activation of HIF-1α in melanoma tumor growth and angiogenesis. Tumour Biol 2023; 45:111-126. [PMID: 37927290 DOI: 10.3233/tub-220020] [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] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND Hypoxia is the most common signature of the tumor microenvironment that drives tumorigenesis through the complex crosstalk of a family of transcription factors called hypoxia-inducible factors (HIFs), with other intercellular signaling networks. Hypoxia increases transforming growth factor-beta (TGF-β) expression. TGF-β and HIF-1α play critical roles in several malignancies and their interactions in melanoma progression remain unknown. Therefore, the aim of this study was to assess the impact of inhibiting activin receptor-like kinase-5 (ALK5), a TGF-β receptor, on the response to HIF-1α activation or inhibition in melanoma tumor progression. MATERIALS AND METHODS Tumors were induced in C57BL/6J mice by subcutaneous inoculation with B16F10 melanoma cells. Mice were divided into HIF-1α inhibitor, ALK5 inhibitor (1 mg/kg) and HIF-1α inhibitor (100 mg/kg), ALK5 inhibitor, HIF-1α activator (1000 mg/kg), HIF-1α activator and ALK5 inhibitor, and control groups to receive inhibitors and activators through intraperitoneal injection. The expression of E-cadherin was evaluated by RT-qPCR. Vessel density and platelet-derived growth factor receptor beta (PDGFR)-β+ cells around the vessels were investigated using immunohistochemistry. RESULTS The groups receiving HIF-1α inhibitor and activator showed lower and higher tumor growth compared to the control group, respectively. E-cadherin expression decreased in all groups compared to the control group, illustrating the dual function of E-cadherin in the tumor microenvironment. Vascular density was reduced in the groups given HIF-1α inhibitor, ALK5 inhibitor, and ALK5 and HIF-1α inhibitor simultaneously. The percentage of PDGFR-β+ cells was reduced in the presence of HIF-1α inhibitor, ALK5 inhibitor, HIF-1α and ALK5 inhibitors, and upon simultaneous treatment with HIF-1α activator and ALK5 inhibitor. CONCLUSION Despite increased expression and interaction between TGF-β and HIF-1α pathways in some cancers, in melanoma, inhibition of either pathway alone may have a stronger effect on tumor inhibition than simultaneous inhibition of both pathways. The synergistic effects may be context-dependent and should be further evaluated in different cancer types.
Collapse
Affiliation(s)
- Bahareh Zarin
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Mostafa Manian
- Isfahan Neurosciences Research Center, Alzahra Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
9
|
Iacopetta D, Fazio A, La Torre C, Barbarossa A, Ceramella J, Francomano F, Saturnino C, El-Kashef H, Alcaro S, Sinicropi MS. Annona cherimola Mill. Leaf Extracts Affect Melanoma Cells Growth and Progression. Foods 2022; 11:foods11162420. [PMID: 36010420 PMCID: PMC9407337 DOI: 10.3390/foods11162420] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 11/24/2022] Open
Abstract
Cancer represents one of the major causes of mortality worldwide; indeed, 19.3 million new cases and almost 10.0 million deaths were estimated last year. Among the different type of cancers, malignant melanoma represents the most aggressive and deadly skin cancer. Unfortunately, the long-term efficacy of melanoma treatments is limited by the lack of clinical efficacy, onset of side effects and resistance. The latter is a major obstacle for the success of the melanoma therapy; thus, the exploration of new potent and safer anticancer agents is of great importance. Recently, numerous plant species, used for therapeutic purposes and containing various non-toxic nutraceuticals have been widely studied. Herein, we investigated the antioxidant and anticancer properties on melanoma cells of the ethanolic, methanolic and aqueous Annona cherimola leaf extracts (ACE, ACM and ACW, respectively). The ethanolic extract showed higher anticancer activity, mostly against the malignant A2058 melanoma cell line (IC50 = 5.6 ± 0.8 ng/mL), together with a very low activity on the normal cells. It blocks the melanoma cells migration process, and induces a clear disorganization of cytoskeleton, triggering cell apoptosis. Finally, some bioactive compounds were identified in the studied extracts.
Collapse
Affiliation(s)
- Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata, di Rende, Italy
| | - Alessia Fazio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata, di Rende, Italy
| | - Chiara La Torre
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata, di Rende, Italy
| | - Alexia Barbarossa
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70126 Bari, Italy
| | - Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata, di Rende, Italy
- Correspondence: ; Tel.: +39-0984493200
| | - Fabrizio Francomano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata, di Rende, Italy
| | - Carmela Saturnino
- Department of Science, University of Basilicata, 85100 Potenza, Italy
| | - Hussein El-Kashef
- Chemistry Department, Faculty of Science, Assiut University, Assiut 17516, Egypt
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università “Magna Græcia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
- Net4Science SRL, Academic Spinoff, Università “Magna Græcia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
- Associazione CRISEA-Centro di Ricerca e Servizi Avanzati per l’Innovazione Rurale, Belcastro, 88055 Catanzaro, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata, di Rende, Italy
| |
Collapse
|
10
|
Hsu MJ, Chen HK, Lien JC, Huang YH, Huang SW. Suppressing VEGF-A/VEGFR-2 Signaling Contributes to the Anti-Angiogenic Effects of PPE8, a Novel Naphthoquinone-Based Compound. Cells 2022; 11:cells11132114. [PMID: 35805198 PMCID: PMC9266117 DOI: 10.3390/cells11132114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 06/30/2022] [Accepted: 07/02/2022] [Indexed: 02/05/2023] Open
Abstract
Natural naphthoquinones and their derivatives exhibit a broad spectrum of pharmacological activities and have thus attracted much attention in modern drug discovery. However, it remains unclear whether naphthoquinones are potential drug candidates for anti-angiogenic agents. The aim of this study was to evaluate the anti-angiogenic properties of a novel naphthoquinone derivative, PPE8, and explore its underlying mechanisms. Determined by various assays including BrdU, migration, invasion, and tube formation analyses, PPE8 treatment resulted in the reduction of VEGF-A-induced proliferation, migration, and invasion, as well as tube formation in human umbilical vein endothelial cells (HUVECs). We also used an aorta ring sprouting assay, Matrigel plug assay, and immunoblotting analysis to examine PPE8’s ex vivo and in vivo anti-angiogenic activities and its actions on VEGF-A signaling. It has been revealed that PPE8 inhibited VEGF-A-induced micro vessel sprouting and was capable of suppressing angiogenesis in in vivo models. In addition, PPE8 inhibited VEGF receptor (VEGFR)-2, Src, FAK, ERK1/2, or AKT phosphorylation in HUVECs exposed to VEGF-A, and it also showed significant decline in xenograft tumor growth in vivo. Taken together, these observations indicated that PPE8 may target VEGF-A–VEGFR-2 signaling to reduce angiogenesis. It also supports the role of PPE8 as a potential drug candidate for the development of therapeutic agents in the treatment of angiogenesis-related diseases including cancer.
Collapse
Affiliation(s)
- Ming-Jen Hsu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Han-Kun Chen
- Department of General Surgery, Chi Mei Medical Center, Tainan 71067, Taiwan;
| | - Jin-Cherng Lien
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan;
- Department of Medical Research, Hospital of China Medical University, Taichung 40402, Taiwan
| | - Yu-Han Huang
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Shiu-Wen Huang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Medical Research, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Research Center of Thoracic Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Correspondence: ; Tel.: +886-2-27361661 (ext. 3198)
| |
Collapse
|
11
|
Hu F, Fong KO, Cheung MPL, Liu JA, Liang R, Li TW, Sharma R, IP PP, Yang X, Cheung M. DEPDC1B Promotes Melanoma Angiogenesis and Metastasis through Sequestration of Ubiquitin Ligase CDC16 to Stabilize Secreted SCUBE3. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105226. [PMID: 35088579 PMCID: PMC8981904 DOI: 10.1002/advs.202105226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/11/2022] [Indexed: 05/28/2023]
Abstract
The ability of melanoma to acquire metastasis through the induction of angiogenesis is one of the major causes of skin cancer death. Here, it is found that high transcript levels of DEP domain containing 1B (DEPDC1B) in cutaneous melanomas are significantly associated with a poor prognosis. Tissue microarray analysis indicates that DEPDC1B expression is positively correlated with SOX10 in the different stages of melanoma. Consistently, DEPDC1B is both required and sufficient for melanoma growth, metastasis, angiogenesis, and functions as a direct downstream target of SOX10 to partly mediate its oncogenic activity. In contrast to other tumor types, the DEPDC1B-mediated enhancement of melanoma metastatic potential is not dependent on the activities of RHO GTPase signaling and canonical Wnt signaling, but is acquired through secretion of signal peptide, CUB domain and EGF like domain containing 3 (SCUBE3), which is crucial for promoting angiogenesis in vitro and in vivo. Mechanistically, DEPDC1B regulates SCUBE3 protein stability through the competitive association with ubiquitin ligase cell division cycle 16 (CDC16) to prevent SCUBE3 from undergoing degradation via the ubiquitin-proteasome pathway. Importantly, expression of SOX10, DEPDC1B, and SCUBE3 are positively correlated with microvessel density in the advanced stage of melanomas. In conclusion, it is revealed that a SOX10-DEPDC1B-SCUBE3 regulatory axis promotes melanoma angiogenesis and metastasis, which suggests that targeting secreted SCUBE3 can be a therapeutic strategy against metastatic melanoma.
Collapse
Affiliation(s)
- Feng Hu
- School of Biomedical SciencesLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Ki On Fong
- School of Biomedical SciencesLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - May Pui Lai Cheung
- School of Biomedical SciencesLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Jessica Aijia Liu
- Department of NeuroscienceCity University of Hong KongTat Chee AvenueHong KongChina
| | - Rui Liang
- School of Biomedical SciencesLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Tsz Wai Li
- School of Biomedical SciencesLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Rakesh Sharma
- Centre for PanorOmic SciencesProteomics and Metabolomics Core FacilityLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Philip Pun‐Ching IP
- Department of PathologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Xintao Yang
- School of Biomedical SciencesLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Martin Cheung
- School of Biomedical SciencesLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| |
Collapse
|
12
|
Filimon A, Preda IA, Boloca AF, Negroiu G. Interleukin-8 in Melanoma Pathogenesis, Prognosis and Therapy-An Integrated View into Other Neoplasms and Chemokine Networks. Cells 2021; 11:120. [PMID: 35011682 PMCID: PMC8750532 DOI: 10.3390/cells11010120] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/17/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
Cutaneous melanoma accounts for only about 7% of skin cancers but is causing almost 90% of deaths. Melanoma cells have a distinct repertoire of mutations from other cancers, a high plasticity and degree of mimicry toward vascular phenotype, stemness markers, versatility in evading and suppress host immune control. They exert a significant influence on immune, endothelial and various stromal cells which form tumor microenvironment. The metastatic stage, the leading cause of mortality in this neoplasm, is the outcome of a complex, still poorly understood, cross-talk between tumor and other cell phenotypes. There is accumulating evidence that Interleukin-8 (IL-8) is emblematic for advanced melanomas. This work aimed to present an updated status of IL-8 in melanoma tumor cellular complexity, through a comprehensive analysis including data from other chemokines and neoplasms. The multiple processes and mechanisms surveyed here demonstrate that IL-8 operates following orchestrated programs within signaling webs in melanoma, stromal and vascular cells. Importantly, the yet unknown molecularity regulating IL-8 impact on cells of the immune system could be exploited to overturn tumor fate. The molecular and cellular targets of IL-8 should be brought into the attention of even more intense scientific exploration and valorization in the therapeutical management of melanoma.
Collapse
Affiliation(s)
| | | | | | - Gabriela Negroiu
- Group of Molecular Cell Biology, Institute of Biochemistry of the Romanian Academy, 060031 Bucharest, Romania; (A.F.); (I.A.P.); (A.F.B.)
| |
Collapse
|
13
|
Shevtsov M, Kaesler S, Posch C, Multhoff G, Biedermann T. Magnetic nanoparticles in theranostics of malignant melanoma. EJNMMI Res 2021; 11:127. [PMID: 34905138 PMCID: PMC8671576 DOI: 10.1186/s13550-021-00868-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/28/2021] [Indexed: 12/14/2022] Open
Abstract
Malignant melanoma is an aggressive tumor with a tendency to metastasize early and with an increasing incidence worldwide. Although in early stage, melanoma is well treatable by excision, the chances of cure and thus the survival rate decrease dramatically after metastatic spread. Conventional treatment options for advanced disease include surgical resection of metastases, chemotherapy, radiation, targeted therapy and immunotherapy. Today, targeted kinase inhibitors and immune checkpoint blockers have for the most part replaced less effective chemotherapies. Magnetic nanoparticles as novel agents for theranostic purposes have great potential in the treatment of metastatic melanoma. In the present review, we provide a brief overview of treatment options for malignant melanoma with different magnetic nanocarriers for theranostics. We also discuss current efforts of designing magnetic particles for combined, multimodal therapies (e.g., chemotherapy, immunotherapy) for malignant melanoma.
Collapse
Affiliation(s)
- Maxim Shevtsov
- Central Institute for Translational Cancer Research (TranslaTUM), Radiation Immuno-Oncology Group, Klinikum rechts der Isar, School of Medicine, Technical University Munich (TUM), Einstein Str. 25, 81675, Munich, Germany
- Laboratory of Biomedical Cell Technologies, Far Eastern Federal University, Primorsky Krai, 690091, Vladivostok, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str, Saint Petersburg, Russian Federation, 197341
| | - Susanne Kaesler
- Department of Dermatology and Allergology, Klinikum rechts der Isar, School of Medicine, Technical University Munich (TUM), Biedersteinerstrasse 29, 80802, Munich, Germany
| | - Christian Posch
- Department of Dermatology and Allergology, Klinikum rechts der Isar, School of Medicine, Technical University Munich (TUM), Biedersteinerstrasse 29, 80802, Munich, Germany
| | - Gabriele Multhoff
- Central Institute for Translational Cancer Research (TranslaTUM), Radiation Immuno-Oncology Group, Klinikum rechts der Isar, School of Medicine, Technical University Munich (TUM), Einstein Str. 25, 81675, Munich, Germany
- Department of Radiation Oncology, Klinikum rechts der Isar, School of Medicine, Technical University Munich (TUM), Ismaninger Str. 22, 81675, Munich, Germany
| | - Tilo Biedermann
- Department of Dermatology and Allergology, Klinikum rechts der Isar, School of Medicine, Technical University Munich (TUM), Biedersteinerstrasse 29, 80802, Munich, Germany.
| |
Collapse
|
14
|
Costa PAC, Silva WN, Prazeres PHDM, Picoli CC, Guardia GDA, Costa AC, Oliveira MA, Guimarães PPG, Gonçalves R, Pinto MCX, Amorim JH, Azevedo VAC, Resende RR, Russo RC, Cunha TM, Galante PAF, Mintz A, Birbrair A. Chemogenetic modulation of sensory neurons reveals their regulating role in melanoma progression. Acta Neuropathol Commun 2021; 9:183. [PMID: 34784974 PMCID: PMC8594104 DOI: 10.1186/s40478-021-01273-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/10/2021] [Indexed: 02/08/2023] Open
Abstract
Sensory neurons have recently emerged as components of the tumor microenvironment. Nevertheless, whether sensory neuronal activity is important for tumor progression remains unknown. Here we used Designer Receptors Exclusively Activated by a Designer Drug (DREADD) technology to inhibit or activate sensory neurons' firing within the melanoma tumor. Melanoma growth and angiogenesis were accelerated following inhibition of sensory neurons' activity and were reduced following overstimulation of these neurons. Sensory neuron-specific overactivation also induced a boost in the immune surveillance by increasing tumor-infiltrating anti-tumor lymphocytes, while reducing immune-suppressor cells. In humans, a retrospective in silico analysis of melanoma biopsies revealed that increased expression of sensory neurons-related genes within melanoma was associated with improved survival. These findings suggest that sensory innervations regulate melanoma progression, indicating that manipulation of sensory neurons' activity may provide a valuable tool to improve melanoma patients' outcomes.
Collapse
Affiliation(s)
- Pedro A C Costa
- Departamento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Walison N Silva
- Departamento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Pedro H D M Prazeres
- Departamento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Caroline C Picoli
- Departamento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | | | - Alinne C Costa
- Departamento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Mariana A Oliveira
- Departamento de Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Pedro P G Guimarães
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Ricardo Gonçalves
- Departamento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Mauro C X Pinto
- Departamento de Farmacologia, Universidade Federal de Goiás, Goiânia, GO, Brasil
| | - Jaime H Amorim
- Centro das Ciências Biológicas e da Saúde, Universidade Federal do Oeste da Bahia, Barreiras, BA, Brasil
| | - Vasco A C Azevedo
- Departamento de Genetica, Ecologia e Evolucao, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Rodrigo R Resende
- Departamento de Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Remo C Russo
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Thiago M Cunha
- Departamento de Farmacologia, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Pedro A F Galante
- Centro de Oncologia Molecular, Hospital Sirio-Libanes, Sao Paulo, SP, Brasil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Departamento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil.
- Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
15
|
Matias M, Pinho JO, Penetra MJ, Campos G, Reis CP, Gaspar MM. The Challenging Melanoma Landscape: From Early Drug Discovery to Clinical Approval. Cells 2021; 10:3088. [PMID: 34831311 PMCID: PMC8621991 DOI: 10.3390/cells10113088] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/02/2021] [Accepted: 11/06/2021] [Indexed: 02/06/2023] Open
Abstract
Melanoma is recognized as the most dangerous type of skin cancer, with high mortality and resistance to currently used treatments. To overcome the limitations of the available therapeutic options, the discovery and development of new, more effective, and safer therapies is required. In this review, the different research steps involved in the process of antimelanoma drug evaluation and selection are explored, including information regarding in silico, in vitro, and in vivo experiments, as well as clinical trial phases. Details are given about the most used cell lines and assays to perform both two- and three-dimensional in vitro screening of drug candidates towards melanoma. For in vivo studies, murine models are, undoubtedly, the most widely used for assessing the therapeutic potential of new compounds and to study the underlying mechanisms of action. Here, the main melanoma murine models are described as well as other animal species. A section is dedicated to ongoing clinical studies, demonstrating the wide interest and successful efforts devoted to melanoma therapy, in particular at advanced stages of the disease, and a final section includes some considerations regarding approval for marketing by regulatory agencies. Overall, considerable commitment is being directed to the continuous development of optimized experimental models, important for the understanding of melanoma biology and for the evaluation and validation of novel therapeutic strategies.
Collapse
Affiliation(s)
- Mariana Matias
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.M.); (J.O.P.); (M.J.P.)
| | - Jacinta O. Pinho
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.M.); (J.O.P.); (M.J.P.)
| | - Maria João Penetra
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.M.); (J.O.P.); (M.J.P.)
| | - Gonçalo Campos
- CICS–UBI–Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6201-506 Covilhã, Portugal;
| | - Catarina Pinto Reis
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.M.); (J.O.P.); (M.J.P.)
| | - Maria Manuela Gaspar
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.M.); (J.O.P.); (M.J.P.)
| |
Collapse
|
16
|
Jęśkowiak I, Wiatrak B, Szeląg A, Mączyński M. Preclinical Study of Immunological Isoxazole Derivatives as a Potential Support for Melanoma Chemotherapy. Int J Mol Sci 2021; 22:ijms222010920. [PMID: 34681580 PMCID: PMC8535817 DOI: 10.3390/ijms222010920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/03/2021] [Accepted: 10/07/2021] [Indexed: 02/07/2023] Open
Abstract
(1) Background: Melanoma is an aggressive neoplasm derived from melanocyte precursors with a high metastatic potential. Responses to chemotherapy and immunotherapy for melanoma remain weak, underlining the urgent need to develop new therapeutic strategies for the treatment of melanoma. (2) Methods: The viability of NHDF and A375 cell cultures after the administration of the tested isoxazole derivatives was assessed after 24-h and 48-h incubation periods with the test compounds in the MTT test. ROS and NO scavenging analyses, a glycoprotein-P activity analysis, a migration assay, a test of apoptosis, and a multiple-criteria decision analysis were also performed. (3) Results: All compounds that were tested resulted in a slower migration of melanoma neoplastic cells. The mechanism of the antitumor activity of the tested compounds was confirmed-i.e., the pro-apoptotic activity of the compounds in A375 cell cultures. Compound O7K qualified for further research. (4) Conclusions: All the tested compounds inhibited the formation of melanoma metastases and demonstrated the ability to reduce the risk of developing drug resistance in the tumor. The MCDA results showed that O7K showed the strongest antitumor activity.
Collapse
Affiliation(s)
- Izabela Jęśkowiak
- Department of Pharmacology, Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wrocław, Poland; (B.W.); (A.S.)
- Correspondence:
| | - Benita Wiatrak
- Department of Pharmacology, Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wrocław, Poland; (B.W.); (A.S.)
| | - Adam Szeląg
- Department of Pharmacology, Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wrocław, Poland; (B.W.); (A.S.)
| | - Marcin Mączyński
- Department of Organic Chemistry, Faculty of Pharmacy, Wroclaw Medical University, 211A Borowska Str., 50-556 Wrocław, Poland;
| |
Collapse
|
17
|
Xu K, Han H, Luo Y, Ye H, Lin H, Ni L. The Angiotensin-Converting Enzyme Inhibitory State Promotes the Transformation of Non-Small Cell Lung Cancer Blood Supply Pattern Toward Vasculogenic Mimicry Formation. Front Oncol 2021; 11:663671. [PMID: 34221978 PMCID: PMC8242235 DOI: 10.3389/fonc.2021.663671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 05/27/2021] [Indexed: 11/13/2022] Open
Abstract
Tumor microenvironment plays an important role in tumor proliferation, metastasis, and angiogenesis. Local RAS is a key factor to tumor proliferation and metastasis in NSCLC microenvironment, but its role on angiogenesis and VM formation remains unclear. Although overwhelming majority of previous studies suggested that VM is well established in aggressive tumor and facilitates tumor growth and metastasis, we put forward different views from another angle. We proved that status of tumor blood supply patterns, including VM channels and endothelial vessels, can dynamically exchange with each other along with local RAS fluctuations in microenvironment. Quantitatively, ACE2/ACEI promotes VM formation via Nodal/Notch4 activation; while structurally, ACE2/ACEI leads to a strong and solid structure of VM via inhibition of VE-cadherin internalization. These changes induced by ACE2/ACEI relate to relatively low metastasis rate and comforting prognoses of NSCLC patients.
Collapse
Affiliation(s)
- Kandi Xu
- Department of Respiration and Critical Care Diseases, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Institute of Respiratory Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huize Han
- Respiratory and Critical Care Center, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Yexin Luo
- First Clinical Medical College, Anhui Medical University, Anhui, China
| | - Hong Ye
- School of Foreign Studies, Anhui University, Anhui, China
| | - Hongxia Lin
- Department of Respiration and Critical Care Diseases, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Institute of Respiratory Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Ni
- Department of Respiration and Critical Care Diseases, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Institute of Respiratory Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
18
|
Böhm M. In search of the needle in a haystack: Finding a suitable serum biomarker for monitoring disease activity of systemic sclerosis. Exp Dermatol 2021; 30:880-886. [PMID: 34121239 DOI: 10.1111/exd.14403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Markus Böhm
- Department of Dermatology, University of Münster, Münster, Germany
| |
Collapse
|
19
|
D'Aguanno S, Mallone F, Marenco M, Del Bufalo D, Moramarco A. Hypoxia-dependent drivers of melanoma progression. J Exp Clin Cancer Res 2021; 40:159. [PMID: 33964953 PMCID: PMC8106186 DOI: 10.1186/s13046-021-01926-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
Hypoxia, a condition of low oxygen availability, is a hallmark of tumour microenvironment and promotes cancer progression and resistance to therapy. Many studies reported the essential role of hypoxia in regulating invasiveness, angiogenesis, vasculogenic mimicry and response to therapy in melanoma. Melanoma is an aggressive cancer originating from melanocytes located in the skin (cutaneous melanoma), in the uveal tract of the eye (uveal melanoma) or in mucosal membranes (mucosal melanoma). These three subtypes of melanoma represent distinct neoplasms in terms of biology, epidemiology, aetiology, molecular profile and clinical features.In this review, the latest progress in hypoxia-regulated pathways involved in the development and progression of all melanoma subtypes were discussed. We also summarized current knowledge on preclinical studies with drugs targeting Hypoxia-Inducible Factor-1, angiogenesis or vasculogenic mimicry. Finally, we described available evidence on clinical studies investigating the use of Hypoxia-Inducible Factor-1 inhibitors or antiangiogenic drugs, alone or in combination with other strategies, in metastatic and adjuvant settings of cutaneous, uveal and mucosal melanoma.Hypoxia-Inducible Factor-independent pathways have been also reported to regulate melanoma progression, but this issue is beyond the scope of this review.As evident from the numerous studies discussed in this review, the increasing knowledge of hypoxia-regulated pathways in melanoma progression and the promising results obtained from novel antiangiogenic therapies, could offer new perspectives in clinical practice in order to improve survival outcomes of melanoma patients.
Collapse
Affiliation(s)
- Simona D'Aguanno
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Fabiana Mallone
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Marco Marenco
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| | | |
Collapse
|
20
|
Thinking Small: Small Molecules as Potential Synergistic Adjuncts to Checkpoint Inhibition in Melanoma. Int J Mol Sci 2021; 22:ijms22063228. [PMID: 33810078 PMCID: PMC8005112 DOI: 10.3390/ijms22063228] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/17/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022] Open
Abstract
Metastatic melanoma remains the deadliest form of skin cancer. Immune checkpoint inhibition (ICI) immunotherapy has defined a new age in melanoma treatment, but responses remain inconsistent and some patients develop treatment resistance. The myriad of newly developed small molecular (SM) inhibitors of specific effector targets now affords a plethora of opportunities to increase therapeutic responses, even in resistant melanoma. In this review, we will discuss the multitude of SM classes currently under investigation, current and prospective clinical combinations of ICI and SM therapies, and their potential for synergism in melanoma eradication based on established mechanisms of immunotherapy resistance.
Collapse
|