1
|
He Y, Chen S, Song W, Wu W, Rao Z, Wang X, Fang L, Shi J, Wang J. Targeted melanoma therapy: High-efficiency siRNA delivery with R8-PEI dissolvable microneedles. Int J Pharm 2025; 675:125516. [PMID: 40139450 DOI: 10.1016/j.ijpharm.2025.125516] [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: 12/05/2024] [Revised: 03/11/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025]
Abstract
Cutaneous melanoma, characterized by its high malignancy and propensity for early metastasis, has become one of the most lethal forms of skin cancer. RNA interference (RNAi) presents a promising therapeutic strategy for melanoma by using small interfering RNAs (siRNAs) to silence gene expression. However, the clinical application of RNAi is hindered by challenges associated with siRNA delivery, including degradation by nucleases, limited cellular uptake, and the barrier of the stratum corneum. In this study, we developed a carrier system comprising of polyethyleneimine (PEI) modified with octamer arginine (R8), a cell-penetrating peptide, and utilized this modified PEI to fabricate dissolvable microneedles loaded with siRNA (siRNA@R8-PEI/DMNs).The siRNA@R8-PEI/DMNs exhibited an excellent overall appearance, featuring a flat backing layer and a needle content of (93.92 ± 4.23)%. The microneedles demonstrated robust mechanical strength,with an overall force of (62.13 ± 8.93) N and a tip strength of (1.69 ± 0.37) N, facilitating effective penetration through the Parafilm®M membrane to a depth of approximately 380 μm. In vitro puncture tests on rat skin revealed a puncture rate exceeding 80%, indicating the system's strong capability to penetrate the skin. In vitro cellular experiments demonstrated that siRNA@R8-PEI effectively inhibited the proliferation of A375 cells by approximately 90% in a concentration-dependent manner. Additionally, the 24-hour cell migration rate was reduced to approximately 20%, and the gene silencing efficiency reached up to about 74%. In vivo studies further confirmed the therapeutic potential of siRNA@R8-PEI/DMNs, with a volumetric tumor inhibition rate of about 67%, inducing tumor cell apoptosis and reducing BRAF expression.siRNA@R8-PEI/DMNs offer a promising transdermal siRNA delivery system for cutaneous melanoma therapy, potentially providing novel insights into the development of safe and efficient transdermal siRNA delivery vectors. This study lays the groundwork for further exploration of RNAi-based therapeutics in dermatological oncology.
Collapse
Affiliation(s)
- Yaozhen He
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China; Guangdong High Education Institutes Engineering Research Center of Modified-released Pharmaceutical Products, Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China
| | - Songsen Chen
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China
| | - Wenyang Song
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China
| | - Wenhao Wu
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China; Guangdong High Education Institutes Engineering Research Center of Modified-released Pharmaceutical Products, Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China
| | - Zhiqi Rao
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China; Guangdong High Education Institutes Engineering Research Center of Modified-released Pharmaceutical Products, Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China
| | - Xiaodan Wang
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China; Guangdong High Education Institutes Engineering Research Center of Modified-released Pharmaceutical Products, Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China
| | - Linrong Fang
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China; Guangdong High Education Institutes Engineering Research Center of Modified-released Pharmaceutical Products, Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China
| | - Jun Shi
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China; Guangdong High Education Institutes Engineering Research Center of Modified-released Pharmaceutical Products, Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China.
| | - Jiu Wang
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China; Guangdong High Education Institutes Engineering Research Center of Modified-released Pharmaceutical Products, Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006 Guangdong, China.
| |
Collapse
|
2
|
Hoinoiu T, Dumitrascu V, Pit D, Schipor DA, Jabri-Tabrizi M, Hoinoiu B, Petreuș DE, Seiman C. Quercetin as a Potential Therapeutic Agent for Malignant Melanoma-A Review of Current Evidence and Future Directions. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:656. [PMID: 40282947 PMCID: PMC12028657 DOI: 10.3390/medicina61040656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/23/2025] [Accepted: 03/31/2025] [Indexed: 04/29/2025]
Abstract
Neoplastic disorders, particularly malignant carcinomas, are complex systemic diseases characterized by unregulated cellular proliferation, the invasion of adjacent tissues, and potential metastasis to distant bodily sites. Among the diverse spectrum of cancer subtypes, malignant melanoma is a highly aggressive form of cutaneous cancer originating in melanocytes, the pigment-producing cells resident in the skin. This malignancy is distinguished by its rapid and uncontrolled growth, as well as its propensity for metastasis to vital organs, thereby posing significant challenges to therapeutic intervention and prognostication. Early detection of melanoma is crucial for optimizing patient outcomes, as diagnosis at an advanced stage often yields a poor prognosis and limited treatment options. Diagnostic modalities for melanoma encompass comprehensive clinical evaluations by dermatologists; radiological imaging techniques such as ultrasonography, magnetic resonance imaging (MRI), computed tomography (CT) scans; and excisional biopsies for accurate histopathological assessment. Malignant melanoma is typically treated with surgery to remove the tumor, followed by immunotherapy to enhance the immune response, targeted therapy for tumors with specific genetic mutations, chemotherapy for advanced stages, radiation therapy to manage metastasis, and other adjunct therapies. This review presents the properties and possible adjunct therapeutic effects against malignant melanoma of quercetin found in the literature and explores, based on the observed physicochemical properties and biological activity, its potential development as a topical formulation for cutaneous application. Quercetin is a naturally occurring flavonoid compound abundant in various plant-based food sources, including apples, onions, berries, and citrus fruits, and has exhibited promising antiproliferative, antioxidant, and anticancer properties. Its distinctive biochemical structure enables quercetin to effectively neutralize reactive oxygen species and modulate key carcinogenic pathways, thereby rendering it a potential candidate for therapeutic intervention in managing malignant tumors, including melanoma.
Collapse
Affiliation(s)
- Teodora Hoinoiu
- Department of Clinical Practical Skills, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. Nr. 2, 300041 Timisoara, Romania; (T.H.); (M.J.-T.)
- Center for Advanced Research in Cardiovascular Pathology and Hemostaseology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Victor Dumitrascu
- Department of Pharmacology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Daniel Pit
- Department of Clinical Practical Skills, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. Nr. 2, 300041 Timisoara, Romania; (T.H.); (M.J.-T.)
- Center for Advanced Research in Cardiovascular Pathology and Hemostaseology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - David-Alexandru Schipor
- Faculty of Chemistry, Biology, Geography, West University of Timisoara, Pestalozzi 16, 300115 Timisoara, Romania;
| | - Madalina Jabri-Tabrizi
- Department of Clinical Practical Skills, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. Nr. 2, 300041 Timisoara, Romania; (T.H.); (M.J.-T.)
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Bogdan Hoinoiu
- Department of Oral Rehabilitation and Dental Emergencies, Faculty of Dentistry, “Victor Babes” University of Medicine and Pharmacy, P-ta Eftimie Murgu 2, 300041 Timisoara, Romania;
- Interdisciplinary Research Center for Dental Medical Research, Lasers and Innovative Technologies, 300070 Timisoara, Romania
| | - David Emanuel Petreuș
- Department of Chemistry, Faculty of Chemistry, Biology, Geography, West University of Timisoara, Pestalozzi 16, 300115 Timisoara, Romania
| | - Corina Seiman
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania;
| |
Collapse
|
3
|
de Groot S, Blommestein HM, Leeneman B, Uyl-de Groot CA, Haanen JBAG, Wouters MWJM, Aarts MJB, van den Berkmortel FWPJ, Blokx WAM, Boers-Sonderen MJ, van den Eertwegh AJM, de Groot JWB, Hospers GAP, Kapiteijn E, van Not OJ, van der Veldt AAM, Suijkerbuijk KPM, van Baal PHM. Development of a Decision Model to Estimate the Outcomes of Treatment Sequences in Advanced Melanoma. Med Decis Making 2025; 45:302-317. [PMID: 39985400 PMCID: PMC11894896 DOI: 10.1177/0272989x251319338] [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: 05/06/2024] [Accepted: 12/17/2024] [Indexed: 02/24/2025]
Abstract
BackgroundA decision model for patients with advanced melanoma to estimate outcomes of a wide range of treatment sequences is lacking.ObjectivesTo develop a decision model for advanced melanoma to estimate outcomes of treatment sequences in clinical practice with the aim of supporting decision making. The article focuses on methodology and long-term health benefits.MethodsA semi-Markov model with a lifetime horizon was developed. Transitions describing disease progression, time to next treatment, and mortality were estimated from real-world data (RWD) as a function of time since starting treatment or disease progression and patient characteristics. Transitions were estimated separately for melanoma with and without a BRAF mutation and for patients with favorable and intermediate prognostic factors. All transitions can be adjusted using relative effectiveness of treatments derived from a network meta-analysis of randomized controlled trials (RCTs). The duration of treatment effect can be adjusted to obtain outcomes under different assumptions.ResultsThe model distinguishes 3 lines of systemic treatment for melanoma with a BRAF mutation and 2 lines of systemic treatment for melanoma without a BRAF mutation. Life expectancy ranged from 7.8 to 12.0 years in patients with favorable prognostic factors and from 5.1 to 8.7 years in patients with intermediate prognostic factors when treated with sequences consisting of targeted therapies and immunotherapies. Scenario analyses illustrate how estimates of life expectancy depend on the duration of treatment effect.ConclusionThe model is flexible because it can accommodate different treatments and treatment sequences, and the duration of treatment effects and the transitions influenced by treatment can be adjusted. We show how using RWD and data from RCTs can harness advantages of both data sources, guiding the development of future decision models.HighlightsThe model is flexible because it can accommodate different treatments and treatment sequences, and the duration of treatment effects as well as the transitions that are influenced by treatment can be adjusted.The long-term health benefits of treatment sequences depend on the place of different therapies within a treatment sequence.Assumptions about the duration of relative treatment effects influence the estimates of long-term health benefits.We show how the use of real-world data and data from randomized controlled trials harness the advantages of both data sources, guiding the development of future decision models.
Collapse
Affiliation(s)
- Saskia de Groot
- Institute for Medical Technology Assessment (iMTA), Erasmus University Rotterdam, The Netherlands
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, The Netherlands
| | - Hedwig M. Blommestein
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, The Netherlands
| | - Brenda Leeneman
- Institute for Medical Technology Assessment (iMTA), Erasmus University Rotterdam, The Netherlands
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, The Netherlands
| | - Carin A. Uyl-de Groot
- Institute for Medical Technology Assessment (iMTA), Erasmus University Rotterdam, The Netherlands
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, The Netherlands
| | - John B. A. G. Haanen
- Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Michel W. J. M. Wouters
- Scientific Bureau, Dutch Institute for Clinical Auditing, Leiden, The Netherlands
- Department of Surgical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Amsterdam, The Netherlands
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Maureen J. B. Aarts
- Department of Medical Oncology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | | | - Willeke A. M. Blokx
- Department of Pathology, Division of Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marye J. Boers-Sonderen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alfons J. M. van den Eertwegh
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Geke A. P. Hospers
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ellen Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Olivier J. van Not
- Scientific Bureau, Dutch Institute for Clinical Auditing, Leiden, The Netherlands
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Astrid A. M. van der Veldt
- Department of Medical Oncology and Radiology & Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Pieter H. M. van Baal
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, The Netherlands
| |
Collapse
|
4
|
Zuo C, Jiang X, Hu J, Bu X. Molecular interactions between fibrinogen and 6-methoxydihydrosanguinarine and their modulation of anticancer activity in melanoma A375 cells. Int J Biol Macromol 2025; 307:142170. [PMID: 40120880 DOI: 10.1016/j.ijbiomac.2025.142170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
Natural benzophenanthridine alkaloids such as sanguinarine, chelerythrine, and 6-methoxydihydrosanguinarine (6-MDS) possess anticancer properties against various cancer cell types. However, the physical interactions of these compounds with blood proteins and the anticancer effects of the resulting complex remain unknown. Therefore, in this study, the interaction between 6-MDS and fibrinogen (Fb) was evaluated through experimental and theoretical studies. The binding analysis revealed that Fb's intrinsic fluorescence is quenched following a moderate interaction with 6-MDS, and a 1:1 complex predominantly forms via hydrogen bonds. Synchronous/ 8-Anilino-1-naphthalenesulfonic acid (ANS) fluorescence and circular dichroism (CD) studies, along with molecular dynamics simulation analysis, indicated that 6-MDS caused a partial rearrangement in the Fb conformation. Differential scanning calorimetry (DSC) measurements showed that upon the addition of 40 μM 6-MDS concentrations, Tm values of Fb slightly decreased, which is consistent with the UV-visible studies. Cellular data revealed IC50 concentrations of 2.85 and 2.65 μM for 6-MDS and the Fb-6-MDS complex against melanoma A375 cells, respectively. These values were significantly lower than those for normal primary human dermal fibroblast (NHDF) cells. It was then observed that the presence of Fb could enhance the anticancer effects of the 6-MDS against melanoma A375 cells via the upregulation of LDH release, ROS production, and caspase-3 activity. Finally, these findings clarify the interaction between Fb and 6-MDS, as well as the anticancer activity of the resulting complex.
Collapse
Affiliation(s)
- Chenchen Zuo
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Xulei Jiang
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Juncheng Hu
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xin Bu
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
5
|
Yang Y, Brenna A, Potenza DM, Sundaramoorthy S, Cheng X, Ming XF, Yang Z. Arginase-II promotes melanoma and lung cancer cell growth by regulating Sirt3-mtROS axis. Front Cell Dev Biol 2025; 13:1528972. [PMID: 40177131 PMCID: PMC11961885 DOI: 10.3389/fcell.2025.1528972] [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: 11/15/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Background Aberrant mitochondrial metabolism is a key source of massive mitochondrial reactive oxygen species (mtROS) in tumour cells. Arginase-II (Arg-II), a widely expressed mitochondrial metabolic enzyme, has recently been shown to enhance mtROS production and melanoma progression. However, how Arg-II enhances mtROS and whether mtROS is involved in stimulation of cancer cell proliferation and migration remain unclear. Methods and results Here, we show that ablation of arg-ii suppresses cell growth, migration, nuclear deformation, and DNA damage in melanoma cells. Vice versa, overexpression of arg-ii in melanoma cells promotes melanoma cell growth and migration accompanied by enhanced nuclear deformation and DNA damage. Ablation or overexpression of arg-ii reduces or enhances mtROS, respectively, accounting for the effects of Arg-II on melanoma growth, migration, and DNA damage. Further data demonstrate that Arg-II enhances mtROS through decreasing Sirtuin 3 (Sirt3) levels. Silencing sirt3 promotes melanoma growth, migration, nuclear deformation, and DNA damage through enhancing mtROS. In supporting of these findings, overexpression of sirt3 prevented Arg-II-induced mtROS production with concomitant prevention of Arg-II-induced cell growth, migration, nuclear deformation and DNA damage. Furthermore, we show that upregulation of Arg-II under hypoxia induces nuclear deformation and DNA damage through suppressing Sirt3. Similar results are obtained in A549 human lung carcinoma cells. In addition, analysis of publicly accessible datasets reveals that elevated arg-ii mRNA levels in human tumor samples including skin cutaneous melanoma and lung cancers associate with poorer prognosis. Conclusion Altogether, our findings demonstrate a critical role of Arg-II-Sirt3-mtROS cascade in promoting melanoma growth, migration, nuclear deformation, and DNA damage linking to melanoma progression and malignancy, which could be therapeutic targets for cancers such as melanoma and lung carcinoma.
Collapse
|
6
|
Leung L, Kirkwood J, Srinivasan S, Dyer M, Qian A, Pourrahmat MM, Kasireddy E, May J, Moshyk A, Kurt M. Challenges and opportunities of predicting overall survival benefit from improvements to recurrence-free survival in stage II/III melanoma: a correlation meta-analysis. IMMUNO-ONCOLOGY TECHNOLOGY 2025; 25:101042. [PMID: 40093596 PMCID: PMC11908549 DOI: 10.1016/j.iotech.2025.101042] [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] [Indexed: 03/19/2025]
Abstract
Background We evaluated the association between treatment effects on recurrence-free survival (RFS) and overall survival (OS) in randomized controlled trials (RCTs) studying resected stage II/III melanoma. Methods Hazard ratios (HRs) of RFS and OS were obtained from a literature review. Bivariate random-effects meta-analysis (BRMA) and weighted linear regression (WLR) models estimated correlations [95% confidence interval (CI)] between HRRFS and HROS. Slopes and intercepts of surrogacy equations were estimated. Surrogate threshold effect was derived from WLR for various sample sizes. Validity and predictive performance of WLR were assessed using leave-one-out cross-validation. Sensitivity analyses evaluated impact of RCTs violating proportional hazards assumption, publication year, treatments' mechanism of action, and cancer stage. Results Across 30 RCTs, treatments included interferon-α (n = 17), other immunotherapy-containing regimens (n = 10), immune checkpoint inhibitors (n = 3), and targeted therapies (n = 2). BRMA (0.68, 95% CI 0.45-0.82) and WLR (0.71, 95% CI 0.42-0.87) estimated moderate correlation between HRRFS and HROS. Surrogate threshold effect was 0.66/0.68 for studies with 800/1000 patients. Slope coefficients were statistically significant in both models (95% CI 0.09-0.61 BRMA; 95% CI 0.41-0.92 WLR). The 95% prediction intervals around the HROS predicted by WLR accurately contained 29/31 (93.5%) of observed HROS. Across sensitivity analyses correlations ranged between 0.69 and 0.84 (BRMA) and 0.55 and 0.77 (WLR). Conclusions Statistically meaningful correlation between HRRFS and HROS can assist earlier predictions of OS benefit from improvements in RFS for RCTs in resected stage II/III melanoma and provide insights for the earlier evaluation of emerging therapies. Primary model predictions should be approached with caution as nearly half of the evidence base comprised interferon-α trials.
Collapse
Affiliation(s)
- L. Leung
- Evidinno Outcomes Research Inc., Vancouver, Canada
| | - J.M. Kirkwood
- Department of Medicine, University of Pittsburgh, Pittsburgh, USA
| | | | - M. Dyer
- Bristol Myers Squibb, Uxbridge, UK
| | - A. Qian
- Evidinno Outcomes Research Inc., Vancouver, Canada
| | | | - E. Kasireddy
- Evidinno Outcomes Research Inc., Vancouver, Canada
| | - J.R. May
- Bristol Myers Squibb, Uxbridge, UK
| | - A. Moshyk
- Bristol Myers Squibb, Princeton, USA
| | - M. Kurt
- Bristol Myers Squibb, Princeton, USA
| |
Collapse
|
7
|
Connor C, Carr QL, Sweazy A, McMasters K, Hao H. Clinical Approaches for the Management of Skin Cancer: A Review of Current Progress in Diagnosis, Treatment, and Prognosis for Patients with Melanoma. Cancers (Basel) 2025; 17:707. [PMID: 40002300 PMCID: PMC11853469 DOI: 10.3390/cancers17040707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Melanoma represents a significant public health challenge due to its increasing incidence and potential for metastasis. This review will explore the current clinical approaches to the management of melanoma, focusing on advancements in diagnosis, treatment, and prognosis. Methods for early detection and accurate staging have been enhanced by new diagnostic strategies. Treatment modalities have expanded beyond traditional surgical excision to include targeted therapy and immunotherapy. Prognostic assessment has benefited from the development of novel biomarkers and genetic profiling. This review will highlight the progress made in the multidisciplinary management of melanoma, underscoring the importance of continuous research to improve patient outcomes.
Collapse
Affiliation(s)
- Colton Connor
- School of Medicine, University of Louisville, Louisville, KY 40202, USA; (C.C.); (Q.L.C.)
| | - Quinton L. Carr
- School of Medicine, University of Louisville, Louisville, KY 40202, USA; (C.C.); (Q.L.C.)
| | - Alisa Sweazy
- The Hiram C. Polk, Jr., MD Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (A.S.); (K.M.)
| | - Kelly McMasters
- The Hiram C. Polk, Jr., MD Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (A.S.); (K.M.)
| | - Hongying Hao
- The Hiram C. Polk, Jr., MD Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (A.S.); (K.M.)
| |
Collapse
|
8
|
Lopes J, Rodrigues CM, Godinho-Santos A, Coelho JMP, Cabaço LC, Barral DC, Faísca P, Catarino J, Nunes D, Fortunato E, Martins R, Rodrigues CMP, Gaspar MM, Reis CP. Combination of gold nanoparticles with near-infrared light as an alternative approach for melanoma management. Int J Pharm 2025; 668:124952. [PMID: 39547473 DOI: 10.1016/j.ijpharm.2024.124952] [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: 08/05/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Melanoma is the most aggressive type of skin cancer and recently approved drugs are often associated with resistance and significant adverse effects. Therefore, the design of more effective and safe options remains imperative. Photothermal therapy (PTT) using gold nanoparticles (AuNPs) presents a promising and innovative approach. In this work, the efficacy of combining a previously optimized formulation of AuNPs coated with a mixture of hyaluronic and oleic acids (HAOA-AuNPs) with near-infrared (NIR) laser irradiation in melanoma cell lines was explored. Coated and uncoated AuNPs formulations were characterized in physicochemical, morphological and elemental terms. Next, the cellular uptake efficiency as well as antiproliferative activity of the combination of each formulation with laser irradiation was evaluated. Subsequently, HAOA-AuNPs were selected to assess the underlying mechanism of combined therapy by cell cycle and Annexin V/PI assays. An in vivo syngeneic murine melanoma model was also conducted. In vitro studies demonstrated that 24 h after incubation and in the absence of laser, HAOA-AuNPs did not exhibit cytotoxic effects on the melanoma cell lines tested, similar to the laser alone. On the contrary, the combination therapy resulted in a large reduction in cell viability. Furthermore, it has been shown to promote S-phase cell cycle arrest and increase in the percentage of late apoptotic cells. Finally, the in vivo proof-of-concept showed that the intratumoral administration of HAOA-AuNPs followed by three laser irradiations impaired tumor progression. Collectively, AuNP-based PTT holds significant potential to improve treatment efficacy and safety, offering a versatile and potent tool against cancer.
Collapse
Affiliation(s)
- Joana Lopes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, Lisboa 1649-003, Portugal
| | - Carla M Rodrigues
- REQUIMTE - LAQV, Chemistry Department, NOVA School of Science and Technology, NOVA University Lisbon, Campus da Caparica Caparica 2829-516, Portugal
| | - Ana Godinho-Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, Lisboa 1649-003, Portugal
| | - João M P Coelho
- Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande 1749-016, Lisboa, Portugal
| | - Luís C Cabaço
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, Lisboa 1169-056, Portugal
| | - Duarte C Barral
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, Lisboa 1169-056, Portugal
| | - Pedro Faísca
- CECAV- Centro de Ciência Animal e Veterinária- Faculdade de Medicina, Veterinária de Lisboa- Universidade Lusófona-Centro Universitário de Lisboa, Portugal
| | - José Catarino
- Faculty of Veterinary Medicine, Universidade Lusófona-Centro Universitário de Lisboa, Portugal; School of Animal Health, Protection and Welfare, Lusophone Polytechnic Institute, Lisbon, Portugal
| | - Daniela Nunes
- Department of Materials Science, NOVA School of Science and Technology, Campus de Caparica, i3N/CENIMAT, Caparica 2829-516, Portugal
| | - Elvira Fortunato
- Department of Materials Science, NOVA School of Science and Technology, Campus de Caparica, i3N/CENIMAT, Caparica 2829-516, Portugal
| | - Rodrigo Martins
- Department of Materials Science, NOVA School of Science and Technology, Campus de Caparica, i3N/CENIMAT, Caparica 2829-516, Portugal
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, Lisboa 1649-003, Portugal
| | - Maria Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, Lisboa 1649-003, Portugal; Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande 1749-016, Lisboa, Portugal.
| | - Catarina Pinto Reis
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, Lisboa 1649-003, Portugal; Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande 1749-016, Lisboa, Portugal.
| |
Collapse
|
9
|
Yang L, Ding R, Tong X, Shen T, Jia S, Yan X, Zhang C, Wu L. Discovery of cloxiquine derivatives as potent HDAC inhibitors for the treatment of melanoma via activating PPARγ. Eur J Med Chem 2025; 281:117029. [PMID: 39522492 DOI: 10.1016/j.ejmech.2024.117029] [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: 09/14/2024] [Revised: 10/30/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
The combined treatment with histone deacetylase (HDAC) inhibitors with peroxisome proliferator-activated receptor γ (PPARγ) agonists has displayed significant anticancer efficacy. Based on these results, a series of cloxiquine derivatives were prepared as potent HDAC inhibitors for the treatment of melanoma. Among these compounds, CS4 exhibited excellent inhibitory effects on HDAC1 (IC50 = 38 nM) and HDAC6 (IC50 = 12 nM), and had good antiproliferative effects against A375 and SK-MEL-5 melanoma cells (IC50 values, 1.20 and 0.93 μM, respectively). Mechanism research indicated that CS4 inhibited SK-MEL-5 cell growth by promoting α-tubulin and histone 3 (H3) acetylation. At the metabolic level, treatment with BG11 activated PPARγ and blocked glycolysis in SK-MEL-5 cells, which mediated partial antimelanoma effects of CS4. In addition, CS4 also induced cell cycle arrest at G2, suppressed migration and facilitated apoptosis of SK-MEL-5 cells. More importantly, compound CS4 demonstrated significant in vivo anticancer effect compared with SAHA, and exhibited neglectable toxicity. Consequently, CS4 is the potent HDAC inhibitor, which may be developed as the candidate antimelanoma drug.
Collapse
Affiliation(s)
- Limin Yang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Ran Ding
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xiaojie Tong
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Tong Shen
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Shuting Jia
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China; Jincheng People's Hospital, Jincheng, 048026, China
| | - Xiqing Yan
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Chong Zhang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Liqiang Wu
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
10
|
Yuan Q, Jiang H, Zhu M, Chu Y, Zhang Y, Bie J, Li L. Lasalocid inhibits melanoma by down-regulating FOXM1 through PI3K/AKT and JNK/P38 MAPK pathways. J Cancer 2025; 16:765-783. [PMID: 39781349 PMCID: PMC11705059 DOI: 10.7150/jca.101798] [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: 08/02/2024] [Accepted: 10/28/2024] [Indexed: 01/12/2025] Open
Abstract
Melanoma poses a significant challenge to patients due to its aggressive nature and limited treatment options. Recent studies have suggested that lasalocid, a feed additive ionophore antibiotic, may have potential as an anticancer agent. However, the mechanism of lasalocid in melanoma is unclear. This study found that lasalocid could inhibit melanoma cell proliferation, migration, and invasion, while inducing cell cycle arrest and apoptosis. Transcriptome sequencing and bioinformatics analysis identified FOXM1 as the hub gene of lasalocid-mediated melanoma treatment. In vitro experiments confirmed that lasalocid regulates FOXM1 expression through the PI3K/AKT and JNK/P38 MAPK pathways. In vivo experiments showed that lasalocid effectively inhibited melanoma growth with acceptable safety. In summary, our study results emphasize lasalocid's potential as a melanoma therapeutic agent and elucidate its role in regulating FOXM1 through the PI3K/AKT and JNK/P38 MAPK pathways.
Collapse
Affiliation(s)
- Qiang Yuan
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan, China, 637003
- School of Pharmacy, North Sichuan Medical College, Nanchong, China, 637100
| | - Hangyu Jiang
- School of Pharmacy, North Sichuan Medical College, Nanchong, China, 637100
- Department of Pharmacy, The Affiliated Hospital of North Sichuan Medical College, Nanchong, China, 637002
| | - Maofei Zhu
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan, China, 637003
- School of Pharmacy, North Sichuan Medical College, Nanchong, China, 637100
| | - Yueming Chu
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan, China, 637003
- School of Pharmacy, North Sichuan Medical College, Nanchong, China, 637100
| | - Yan Zhang
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan, China, 637003
- Nanchong Key Laboratory of Individualized Drug Therapy, Nanchong, China, 637003
| | - Jun Bie
- Department of Oncology, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China, 637003
- Institute of Tissue Engineering and Stem Cells, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China, 637003
| | - Lin Li
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan, China, 637003
- School of Pharmacy, North Sichuan Medical College, Nanchong, China, 637100
- Institute of Tissue Engineering and Stem Cells, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China, 637003
- Nanchong Key Laboratory of Individualized Drug Therapy, Nanchong, China, 637003
| |
Collapse
|
11
|
Babaei S, Fadaee M, Abbasi-Kenarsari H, Shanehbandi D, Kazemi T. Exosome-based immunotherapy as an innovative therapeutic approach in melanoma. Cell Commun Signal 2024; 22:527. [PMID: 39482766 PMCID: PMC11526674 DOI: 10.1186/s12964-024-01906-1] [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: 08/07/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024] Open
Abstract
The malignant form of melanoma is one of the deadliest human cancers that accounts for almost all of the skin tumor-related fatalities in its later stages. Achieving an exhaustive understanding of reliable cancer-specific markers and molecular pathways can provide numerous practical techniques and direct the way toward the development of rational curative medicines to increase the lifespan of patients. Immunotherapy has significantly enhanced the treatment of metastatic and late-stage melanoma, resulting in an incredible increase in positive responses to therapy. Despite the increasing occurrence of melanoma, the median survival rate for patients with advanced, inoperable terminal disease has increased from around six months to almost six years. The current knowledge of the tumor microenvironment (TME) and its interaction with the immune system has resulted in the swift growth of innovative immunotherapy treatments. Exosomes are small extracellular vesicles (EVs), ranging from 30 to 150 nm in size, that the majority of cells released them. Exosomes possess natural advantages such as high compatibility with living organisms and low potential for causing immune reactions, making them practical for delivering therapeutic agents like chemotherapy drugs, nucleic acids, and proteins. This review highlights recent advancements in using exosomes as an approach to providing medications for the treatment of melanoma.
Collapse
Affiliation(s)
- Shabnam Babaei
- Immunology Research Center , Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran, P.O. Box: 5165683146
| | - Manouchehr Fadaee
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran, P.O. Box: 5165683146
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajar Abbasi-Kenarsari
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Dariush Shanehbandi
- Immunology Research Center , Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center , Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran, P.O. Box: 5165683146.
| |
Collapse
|
12
|
Rothe R, Xu Y, Wodtke J, Brandt F, Meister S, Laube M, Lollini PL, Zhang Y, Pietzsch J, Hauser S. Programmable Release of Chemotherapeutics from Ferrocene-Based Injectable Hydrogels Slows Melanoma Growth. Adv Healthc Mater 2024; 13:e2400265. [PMID: 39007274 DOI: 10.1002/adhm.202400265] [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: 01/23/2024] [Revised: 07/02/2024] [Indexed: 07/16/2024]
Abstract
Hydrogel-based injectable drug delivery systems provide temporally and spatially controlled drug release with reduced adverse effects on healthy tissues. Therefore, they represent a promising therapeutic option for unresectable solid tumor entities. In this study, a peptide-starPEG/hyaluronic acid-based physical hydrogel is modified with ferrocene to provide a programmable drug release orchestrated by matrix-drug interaction and local reactive oxygen species (ROS). The injectable ROS-responsive hydrogel (hiROSponse) exhibits adequate biocompatibility and biodegradability, which are important for clinical applications. HiROSponse is loaded with the two cytostatic drugs (hiROSponsedox/ptx) doxorubicin (dox) and paclitaxel (ptx). Dox is a hydrophilic compound and its release is mainly controlled by Fickian diffusion, while the hydrophobic interactions between ptx and ferrocene can control its release and thus be regulated by the oxidation of ferrocene to the more hydrophilic state of ferrocenium. In a syngeneic malignant melanoma-bearing mouse model, hiROSponsedox/ptx slows tumor growth without causing adverse side effects and doubles the relative survival probability. Programmable release is further demonstrated in a tumor model with a low physiological ROS level, where dox release, low dose local irradiation, and the resulting ROS-triggered ptx release lead to tumor growth inhibition and increased survival.
Collapse
Affiliation(s)
- Rebecca Rothe
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
- Technische Universität Dresden, Faculty of Chemistry and Food Chemistry, School of Science, Bergstrasse 66, 01069, Dresden, Germany
| | - Yong Xu
- B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, Tatzberg 41, 01307, Dresden, Germany
| | - Johanna Wodtke
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Florian Brandt
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
- Technische Universität Dresden, Faculty of Chemistry and Food Chemistry, School of Science, Bergstrasse 66, 01069, Dresden, Germany
| | - Sebastian Meister
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Markus Laube
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Pier-Luigi Lollini
- Alma Mater Studiorum, University of Bologna, Department of Medical and Surgical Sciences, Viale Filopanti 22, Bologna, 40126, Italy
| | - Yixin Zhang
- B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, Tatzberg 41, 01307, Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
- Technische Universität Dresden, Faculty of Chemistry and Food Chemistry, School of Science, Bergstrasse 66, 01069, Dresden, Germany
| | - Sandra Hauser
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
| |
Collapse
|
13
|
Almeida TC, Giannotti KC, Ribeiro Silva LM, Marques-Porto R, DeOcesano-Pereira C, Camargo L, Chudzinski-Tavassi AM, Reid P, Picolo G. Crotoxin induces cytotoxic effects in human malignant melanoma cells in both native and detoxified forms. Front Pharmacol 2024; 15:1425446. [PMID: 39114354 PMCID: PMC11303296 DOI: 10.3389/fphar.2024.1425446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction: Melanoma, a highly aggressive skin cancer originating in melanocytes, poses a significant threat due to its metastatic potential. While progress has been made in treating melanoma with targeted therapies and immunotherapies, challenges persist. Crotoxin (CTX), the principal toxin in Crotalus durissus terrificus snake venom, exhibits various biological activities, including anti-tumoral effects across multiple cancers. However, its clinical use is limited by toxicity. Thus, exploring alternatives to mitigate adverse effects is crucial. Methods and Results: This study investigates the antitumoral potential of CTX in its native and in a detoxified form, in melanoma cells. Firstly, we demonstrated that detoxified CTX presented reduced phospholipase activity. Both forms proved to be more cytotoxic to SK-MEL-28 and MeWo melanoma cells than non-tumoral cells. In SK-MEL-28 cells, where cytotoxic effects were more pronounced, native and detoxified CTX induced increased necrosis and apoptosis rates. We also confirmed the apoptosis death demonstrated by the activation of caspase-3 and 7, and the formation of apoptotic bodies. Furthermore, both CTX caused cell cycle arrest at the G2/M phase, interfering with melanoma cell proliferation. Cell migration and invasion were also suppressed by both CTX. These results confirm the antitumoral potential of CTX. Discussion: The maintenance of the antiproliferative effects in the detoxified version, with reduced enzymatic activity often liked to harm effects, supports further studies to identify active parts of the molecule responsible for the interesting effects without causing substantial toxic events, contributing to the future use of CTX-derived drugs with safety and efficacy.
Collapse
Affiliation(s)
| | | | | | | | | | - Lauren Camargo
- Centre of Excellence in New Target Discovering (CENTD), Butantan Institute, Sao Paulo, Brazil
| | | | - Paul Reid
- Celtic Biotech Ltd., Dublin, Ireland
| | - Gisele Picolo
- Laboratory of Pain and Signaling, Butantan Institute, Sao Paulo, Brazil
| |
Collapse
|
14
|
Mentucci FM, Romero Nuñez EA, Ercole A, Silvetti V, Dal Col J, Lamberti MJ. Impact of Genomic Mutation on Melanoma Immune Microenvironment and IFN-1 Pathway-Driven Therapeutic Responses. Cancers (Basel) 2024; 16:2568. [PMID: 39061208 PMCID: PMC11274745 DOI: 10.3390/cancers16142568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
The BRAFV600E mutation, found in approximately 50% of melanoma cases, plays a crucial role in the activation of the MAPK/ERK signaling pathway, which promotes tumor cell proliferation. This study aimed to evaluate its impact on the melanoma immune microenvironment and therapeutic responses, particularly focusing on immunogenic cell death (ICD), a pivotal cytotoxic process triggering anti-tumor immune responses. Through comprehensive in silico analysis of the Cancer Genome Atlas data, we explored the association between the BRAFV600E mutation, immune subtype dynamics, and tumor mutation burden (TMB). Our findings revealed that the mutation correlated with a lower TMB, indicating a reduced generation of immunogenic neoantigens. Investigation into immune subtypes reveals an exacerbation of immunosuppression mechanisms in BRAFV600E-mutated tumors. To assess the response to ICD inducers, including doxorubicin and Me-ALA-based photodynamic therapy (PDT), compared to the non-ICD inducer cisplatin, we used distinct melanoma cell lines with wild-type BRAF (SK-MEL-2) and BRAFV600E mutation (SK-MEL-28, A375). We demonstrated a differential response to PDT between the WT and BRAFV600E cell lines. Further transcriptomic analysis revealed upregulation of IFNAR1, IFNAR2, and CXCL10 genes associated with the BRAFV600E mutation, suggesting their involvement in ICD. Using a gene reporter assay, we showed that PDT robustly activated the IFN-1 pathway through cGAS-STING signaling. Collectively, our results underscore the complex interplay between the BRAFV600E mutation and immune responses, suggesting a putative correlation between tumors carrying the mutation and their responsiveness to therapies inducing the IFN-1 pathway, such as the ICD inducer PDT, possibly mediated by the elevated expression of IFNAR1/2 receptors.
Collapse
Affiliation(s)
- Fátima María Mentucci
- Departamento de Biología Molecular, INBIAS, Universidad Nacional de Río Cuarto, Río Cuarto X5800BIA, Argentina; (F.M.M.); (V.S.)
| | - Elisa Ayelén Romero Nuñez
- Departamento de Biología Molecular, INBIAS, Universidad Nacional de Río Cuarto, Río Cuarto X5800BIA, Argentina; (F.M.M.); (V.S.)
| | - Agustina Ercole
- Departamento de Biología Molecular, INBIAS, Universidad Nacional de Río Cuarto, Río Cuarto X5800BIA, Argentina; (F.M.M.); (V.S.)
| | - Valentina Silvetti
- Departamento de Biología Molecular, INBIAS, Universidad Nacional de Río Cuarto, Río Cuarto X5800BIA, Argentina; (F.M.M.); (V.S.)
| | - Jessica Dal Col
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84081 Baronissi, Italy;
| | - María Julia Lamberti
- Departamento de Biología Molecular, INBIAS, Universidad Nacional de Río Cuarto, Río Cuarto X5800BIA, Argentina; (F.M.M.); (V.S.)
| |
Collapse
|
15
|
Silverstein J, Goyal N, Tsai KK. For the Long Haul: Management of Long-Term Survivors after Melanoma Systemic Therapy. Curr Oncol Rep 2024; 26:804-817. [PMID: 38780676 DOI: 10.1007/s11912-024-01541-6] [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] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE OF REVIEW This review summarizes the latest advancements in survivorship care for patients with advanced melanoma who received systemic therapy and emphasizes the areas where more research is needed. RECENT FINDINGS Over the last decade there have been remarkable advances in the treatment of advanced and metastatic melanoma. Due to these novel treatments, including several immune checkpoint inhibitors and tyrosine kinase inhibitors, there are and will continue to be increasing numbers of long-term melanoma survivors who have been treated with systemic therapy. These patients will navigate new challenges are they are essentially among the first long term survivors after these novel therapies. Survivorship care focuses on improving the health-related quality of life of patients including the physical, emotional, social and functional effects of cancer that begin at diagnosis and continue through the end of life. Survivorship also includes screening for cancer recurrence and second cancers. As the number of melanoma survivors who received systemic therapy continues to grow, the survivorship care plan will become increasingly important for optimal care of patients even after their cancer treatments. Understanding the many domains of survivorship care for this group of patients is imperative for their care now and to identify unmet needs for future research.
Collapse
Affiliation(s)
- Jordyn Silverstein
- Department of Medicine, Division of Hematology/Oncology, University of California, Los Angeles (UCLA), 757 Westwood Plaza, Los Angeles, CA, 90095, USA.
| | - Neha Goyal
- Department of Psycho-Oncology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Katy K Tsai
- Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco (UCSF), San Francisco, CA, USA
| |
Collapse
|
16
|
Środa-Pomianek K, Barycka A, Gleńsk M, Rajbhandari M, Skonieczna M, Palko-Łabuz A, Wesołowska O. Pretreatment of Melanoma Cells with Aqueous Ethanol Extract from Madhuca longifolia Bark Strongly Potentiates the Activity of a Low Dose of Dacarbazine. Int J Mol Sci 2024; 25:7220. [PMID: 39000329 PMCID: PMC11241726 DOI: 10.3390/ijms25137220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Madhuca longifolia is an evergreen tree distributed in India, Nepal, and Sri Lanka. This tree is commonly known as Mahua and is used in traditional medicine. It was demonstrated that ethanol extract from the bark of M. longifolia possessed potent cytotoxic activity towards two melanoma cell lines, in contrast to aqueous extract that exhibited no activity. Apart from being selectively cytotoxic to cancer cells (with no activity towards non-cancerous fibroblasts), the studied extract induced apoptosis and increased reactive oxygen species generation in melanoma cells. Additionally, the use of the extract together with dacarbazine (both in non-toxic concentrations) resulted in the enhancement of their anticancer activity. Moreover, the pretreatment of melanoma cells with M. longifolia extract potentiated the activity of a low dose of dacarbazine to an even higher extent. It was concluded that ethanol extract of M. longifolia sensitized human melanoma cells to chemotherapeutic drugs. It can therefore be interesting as a promising source of compounds for prospective combination therapy.
Collapse
Affiliation(s)
- Kamila Środa-Pomianek
- Department of Biophysics and Neurobiology, Wroclaw Medical University, 50-369 Wrocław, Poland; (K.Ś.-P.); (A.B.)
| | - Anna Barycka
- Department of Biophysics and Neurobiology, Wroclaw Medical University, 50-369 Wrocław, Poland; (K.Ś.-P.); (A.B.)
| | - Michał Gleńsk
- Department of Pharmacognosy and Herbal Medicines, Wrocław Medical University, 50-367 Wrocław, Poland;
| | - Meena Rajbhandari
- Research Centre for Applied Science and Technology (RECAST), Kirtipur 44600, Kathmandu, Nepal;
| | - Magdalena Skonieczna
- Department of Systems Biology and Engineering, The Silesian University of Technology, 44-100 Gliwice, Poland;
- Biotechnology Centre, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Anna Palko-Łabuz
- Department of Biophysics and Neurobiology, Wroclaw Medical University, 50-369 Wrocław, Poland; (K.Ś.-P.); (A.B.)
| | - Olga Wesołowska
- Department of Biophysics and Neurobiology, Wroclaw Medical University, 50-369 Wrocław, Poland; (K.Ś.-P.); (A.B.)
| |
Collapse
|
17
|
Goodman RS, Jung S, Fletcher K, Burnette H, Mohyuddin I, Irlmeier R, Ye F, Johnson DB. Primary Tumor Characteristics as Biomarkers of Immunotherapy Response in Advanced Melanoma: A Retrospective Cohort Study. Cancers (Basel) 2024; 16:2355. [PMID: 39001417 PMCID: PMC11240575 DOI: 10.3390/cancers16132355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/16/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Identifying patients likely to benefit from immune checkpoint inhibitor (ICI) treatment remains a crucial goal for melanoma. The objective of this study is to assess the association between primary tumor features and immunotherapy response and survival in advanced melanoma patients. In this single-center retrospective cohort study, disease characteristics, response to immunotherapy, PFS, and OS were assessed among melanoma patients (excluding mucosal and uveal primaries) treated with ICI. Among 447 patients, 300 (67.1%) received anti-PD-1 monotherapy and 147 (32.9%) received ipilimumab/nivolumab. A total of 338 (75.6%) had cutaneous melanoma, 29 (6.5%) had acral melanoma, and 80 (17.9%) had melanoma of unknown primary. Ulceration and stage at initial presentation were associated with inferior outcomes on univariate analysis. However, on multivariate analysis, this result was not observed, but cutaneous melanoma and each of its subtypes (superficial spreading, nodular, other, unknown) were positively associated with response, longer PFS, and longer OS. Metastatic stage (M1c, M1d) at presentation (OR = 1.8, p < 0.05) and BRAFV600E mutation status (OR = 1.6, p < 0.001) were associated with shorter PFS. This study is limited by its retrospective and single-center design. Cutaneous melanoma and its subtypes were significantly associated with response, PFS, and OS compared with acral or unknown primary melanoma.
Collapse
Affiliation(s)
- Rachel S Goodman
- Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Seungyeon Jung
- Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Kylie Fletcher
- Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Hannah Burnette
- Department of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | | | - Rebecca Irlmeier
- Vanderbilt University School of Medicine, Nashville, TN 37240, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Fei Ye
- Vanderbilt University School of Medicine, Nashville, TN 37240, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Douglas B Johnson
- Department of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| |
Collapse
|
18
|
Gromek P, Senkowska Z, Płuciennik E, Pasieka Z, Zhao LY, Gielecińska A, Kciuk M, Kłosiński K, Kałuzińska-Kołat Ż, Kołat D. Revisiting the standards of cancer detection and therapy alongside their comparison to modern methods. World J Methodol 2024; 14:92982. [PMID: 38983668 PMCID: PMC11229876 DOI: 10.5662/wjm.v14.i2.92982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/15/2024] [Accepted: 04/28/2024] [Indexed: 06/13/2024] Open
Abstract
In accordance with the World Health Organization data, cancer remains at the forefront of fatal diseases. An upward trend in cancer incidence and mortality has been observed globally, emphasizing that efforts in developing detection and treatment methods should continue. The diagnostic path typically begins with learning the medical history of a patient; this is followed by basic blood tests and imaging tests to indicate where cancer may be located to schedule a needle biopsy. Prompt initiation of diagnosis is crucial since delayed cancer detection entails higher costs of treatment and hospitalization. Thus, there is a need for novel cancer detection methods such as liquid biopsy, elastography, synthetic biosensors, fluorescence imaging, and reflectance confocal microscopy. Conventional therapeutic methods, although still common in clinical practice, pose many limitations and are unsatisfactory. Nowadays, there is a dynamic advancement of clinical research and the development of more precise and effective methods such as oncolytic virotherapy, exosome-based therapy, nanotechnology, dendritic cells, chimeric antigen receptors, immune checkpoint inhibitors, natural product-based therapy, tumor-treating fields, and photodynamic therapy. The present paper compares available data on conventional and modern methods of cancer detection and therapy to facilitate an understanding of this rapidly advancing field and its future directions. As evidenced, modern methods are not without drawbacks; there is still a need to develop new detection strategies and therapeutic approaches to improve sensitivity, specificity, safety, and efficacy. Nevertheless, an appropriate route has been taken, as confirmed by the approval of some modern methods by the Food and Drug Administration.
Collapse
Affiliation(s)
- Piotr Gromek
- Department of Functional Genomics, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
| | - Zuzanna Senkowska
- Department of Functional Genomics, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
| | - Elżbieta Płuciennik
- Department of Functional Genomics, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
| | - Zbigniew Pasieka
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, Lodz 90-136, Lodzkie, Poland
| | - Lin-Yong Zhao
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Adrianna Gielecińska
- Department of Molecular Biotechnology and Genetics, University of Lodz, Lodz 90-237, Lodzkie, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, Lodz 90-237, Lodzkie, Poland
| | - Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, University of Lodz, Lodz 90-237, Lodzkie, Poland
| | - Karol Kłosiński
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, Lodz 90-136, Lodzkie, Poland
| | - Żaneta Kałuzińska-Kołat
- Department of Functional Genomics, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, Lodz 90-136, Lodzkie, Poland
| | - Damian Kołat
- Department of Functional Genomics, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, Lodz 90-136, Lodzkie, Poland
| |
Collapse
|
19
|
Natarelli N, Aleman SJ, Mark IM, Tran JT, Kwak S, Botto E, Aflatooni S, Diaz MJ, Lipner SR. A Review of Current and Pipeline Drugs for Treatment of Melanoma. Pharmaceuticals (Basel) 2024; 17:214. [PMID: 38399429 PMCID: PMC10892880 DOI: 10.3390/ph17020214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Malignant melanoma is the most aggressive form of skin cancer. Standard treatment options include surgery, radiation therapy, systemic chemotherapy, targeted therapy, and immunotherapy. Combining these modalities often yields better responses. Surgery is suitable for localized cases, sometimes involving lymph node dissection and biopsy, to assess the spread of the disease. Radiation therapy may be sometimes used as a standalone treatment or following surgical excision. Systemic chemotherapy, while having low response rates, is utilized as part of combination treatments or when other methods fail. The development of resistance to systemic chemotherapies and associated side effects have prompted further research and clinical trials for novel approaches. In the case of advanced-stage melanoma, a comprehensive approach may be necessary, incorporating targeted therapies and immunotherapies that demonstrate significant antitumor activity. Targeted therapies, including inhibitors targeting BRAF, MEK, c-KIT, and NRAS, are designed to block the specific molecules responsible for tumor growth. These therapies show promise, particularly in patients with corresponding mutations. Combination therapy, including BRAF and MEK inhibitors, has been evidenced to improve progression-free survival; however, concerns about resistance and cutaneous toxicities highlight the need for close monitoring. Immunotherapies, leveraging tumor-infiltrating lymphocytes and CAR T cells, enhance immune responses. Lifileucel, an FDA-approved tumor-infiltrating lymphocyte therapy, has demonstrated improved response rates in advanced-stage melanoma. Ongoing trials continue to explore the efficacy of CAR T-cell therapy for advanced melanoma. Checkpoint inhibitors targeting CTLA-4 and PD-1 have enhanced outcomes. Emerging IL-2 therapies boost dendritic cells, enhancing anticancer immunity. Oncolytic virus therapy, approved for advanced melanoma, augments treatment efficacy in combination approaches. While immunotherapy has significantly advanced melanoma treatment, its success varies, prompting research into new drugs and factors influencing outcomes. This review provides insights into current melanoma treatments and recent therapeutic advances.
Collapse
Affiliation(s)
- Nicole Natarelli
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Sarah J. Aleman
- School of Medicine, Louisiana State University, New Orleans, LA 70112, USA
| | - Isabella M. Mark
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jasmine T. Tran
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Sean Kwak
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Elizabeth Botto
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Shaliz Aflatooni
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Michael J. Diaz
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Shari R. Lipner
- Department of Dermatology, Weill Cornell Medicine, New York City, NY 10021, USA
| |
Collapse
|
20
|
Benesch MGK, Skitzki JJ. Impact of anesthesia choice in cutaneous melanoma surgery. Melanoma Res 2024; 34:16-21. [PMID: 37924527 PMCID: PMC10842619 DOI: 10.1097/cmr.0000000000000936] [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] [Indexed: 11/06/2023]
Abstract
Invasive cutaneous melanoma is the most lethal skin cancer, but fortunately, the vast majority can be surgically treated with wide local excision, and sometimes additionally with sentinel or index lymph node biopsy for prognostication. Melanomas are particularly immunogenic malignancies, and preclinical studies have demonstrated that use of volatile anesthetics and opioids, unlike local agents, can suppress the immune system during the perioperative period. Immunosuppression has implications for creating a potentially favorable microenvironment for the survival and propagation of residual melanoma cells or micro-metastases, which could lead to disease relapse, both in the local tumor bed and distally. Results from observational clinical studies are mixed, but the literature would suggest that patients are at risk of decreased melanoma-specific survival after undergoing general anesthesia compared to regional anesthesia and spinal blocks. With the safety of close observation now established rather than automatic completion or total lymph node dissection for patients with either a positive sentinel lymph node biopsy or significant clinical response to neoadjuvant immunotherapy after index node sampling, the indications for definitive surgery with local or regional anesthesia have increased tremendously in recent years. Therefore, cutaneous melanoma patients might benefit from avoidance of general anesthesia and other perioperative drugs that suppress cell-mediated immunity if the option to circumvent systemic anesthesia agents is feasible.
Collapse
Affiliation(s)
- Matthew G K Benesch
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | | |
Collapse
|
21
|
Saamarthy K, Ahlqvist K, Daams R, Balagunaseelan N, Rinaldo-Matthis A, Kazi JU, Sime W, Massoumi R. Discovery of a small molecule that inhibits Bcl-3-mediated cyclin D1 expression in melanoma cells. BMC Cancer 2024; 24:103. [PMID: 38238702 PMCID: PMC10795364 DOI: 10.1186/s12885-023-11663-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/21/2023] [Indexed: 01/22/2024] Open
Abstract
Molecular targeted therapy using a drug that suppresses the growth and spread of cancer cells via inhibition of a specific protein is a foundation of precision medicine and treatment. High expression of the proto-oncogene Bcl-3 promotes the proliferation and metastasis of cancer cells originating from tissues such as the colon, prostate, breast, and skin. The development of novel drugs targeting Bcl-3 alone or in combination with other therapies can cure these patients or prolong their survival. As a proof of concept, in the present study, we focused on metastatic melanoma as a model system. High-throughput screening and in vitro experiments identified BCL3ANT as a lead molecule that could interfere with Bcl-3-mediated cyclin D1 expression and cell proliferation and migration in melanoma. In experimental animal models of melanoma, it was demonstrated that the use of a Bcl-3 inhibitor can influence the survival of melanoma cells. Since there are no other inhibitors against Bcl-3 in the clinical pipeline for cancer treatment, this presents a unique opportunity to develop a highly specific drug against malignant melanoma to meet an urgent clinical need.
Collapse
Affiliation(s)
- Karunakar Saamarthy
- Department of Laboratory Medicine, Translational Cancer Research, Division of Molecular Tumor Pathology, Lund University, Medicon Village, 22383, Lund, Sweden
| | - Kristofer Ahlqvist
- Department of Laboratory Medicine, Translational Cancer Research, Division of Molecular Tumor Pathology, Lund University, Medicon Village, 22383, Lund, Sweden
| | - Renée Daams
- Department of Laboratory Medicine, Translational Cancer Research, Division of Molecular Tumor Pathology, Lund University, Medicon Village, 22383, Lund, Sweden
| | - Navisraj Balagunaseelan
- Department of Medical Biochemistry and Biophysics, Division of Chemistry II, Karolinska Institutet, Stockholm, Sweden
| | - Agnes Rinaldo-Matthis
- Department of Medical Biochemistry and Biophysics, Division of Chemistry II, Karolinska Institutet, Stockholm, Sweden
| | - Julhash U Kazi
- Department of Laboratory Medicine, Translational Cancer Research, Division of Molecular Tumor Pathology, Lund University, Medicon Village, 22383, Lund, Sweden
| | - Wondossen Sime
- Department of Laboratory Medicine, Translational Cancer Research, Division of Molecular Tumor Pathology, Lund University, Medicon Village, 22383, Lund, Sweden
| | - Ramin Massoumi
- Department of Laboratory Medicine, Translational Cancer Research, Division of Molecular Tumor Pathology, Lund University, Medicon Village, 22383, Lund, Sweden.
| |
Collapse
|
22
|
Wellman R, Jacobson D, Secrier M, Labbadia J. Distinct patterns of proteostasis network gene expression are associated with different prognoses in melanoma patients. Sci Rep 2024; 14:198. [PMID: 38167612 PMCID: PMC10761826 DOI: 10.1038/s41598-023-50640-0] [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: 09/05/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2024] Open
Abstract
The proteostasis network (PN) is a collection of protein folding and degradation pathways that spans cellular compartments and acts to preserve the integrity of the proteome. The differential expression of PN genes is a hallmark of many cancers, and the inhibition of protein quality control factors is an effective way to slow cancer cell growth. However, little is known about how the expression of PN genes differs between patients and how this impacts survival outcomes. To address this, we applied unbiased hierarchical clustering to gene expression data obtained from primary and metastatic cutaneous melanoma (CM) samples and found that two distinct groups of individuals emerge across each sample type. These patient groups are distinguished by the differential expression of genes encoding ATP-dependent and ATP-independent chaperones, and proteasomal subunits. Differences in PN gene expression were associated with increased levels of the transcription factors, MEF2A, SP4, ZFX, CREB1 and ATF2, as well as markedly different survival outcomes. However, surprisingly, similar PN alterations in primary and metastatic samples were associated with discordant survival outcomes in patients. Our findings reveal that the expression of PN genes demarcates CM patients and highlights several new proteostasis sub-networks that could be targeted for more effective suppression of CM within specific individuals.
Collapse
Affiliation(s)
- Rachel Wellman
- Division of Biosciences, Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, UK
- Division of Biosciences, Department of Genetics, Evolution and Environment, UCL Genetics Institute, University College London, London, UK
| | - Daniel Jacobson
- Division of Biosciences, Department of Genetics, Evolution and Environment, UCL Genetics Institute, University College London, London, UK
- UCL Cancer Institute, University College London, London, UK
| | - Maria Secrier
- Division of Biosciences, Department of Genetics, Evolution and Environment, UCL Genetics Institute, University College London, London, UK.
| | - John Labbadia
- Division of Biosciences, Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, UK.
| |
Collapse
|
23
|
Kozyra P, Pitucha M. Revisiting the Role of B-RAF Kinase as a Therapeutic Target in Melanoma. Curr Med Chem 2024; 31:2003-2020. [PMID: 37855341 DOI: 10.2174/0109298673258495231011065225] [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: 04/18/2023] [Revised: 07/19/2023] [Accepted: 08/17/2023] [Indexed: 10/20/2023]
Abstract
Malignant melanoma is the rarest but most aggressive and deadly skin cancer. Melanoma is the result of a malignant transformation of melanocytes, which leads to their uncontrolled proliferation. Mutations in the mitogen-activated protein kinase (MAPK) pathway, which are crucial for the control of cellular processes, such as apoptosis, division, growth, differentiation, and migration, are one of its most common causes. BRAF kinase, as one of the known targets of this pathway, has been known for many years as a prominent molecular target in melanoma therapy, and the following mini-review outlines the state-of-the-art knowledge regarding its structure, mutations and mechanisms.
Collapse
Affiliation(s)
- Paweł Kozyra
- Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, Lublin, PL, 20093, Poland
| | - Monika Pitucha
- Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, Lublin, PL-20093, Poland
| |
Collapse
|
24
|
Zheng S, Yu H, Zheng X, Wu UT, Ming WK, Huang H, Song J, Zhang X, Lyu J, Deng L. Analysis and prediction of 5-year survival in patients with cutaneous melanoma: a model-based period analysis. Front Endocrinol (Lausanne) 2023; 14:1238086. [PMID: 38125787 PMCID: PMC10731280 DOI: 10.3389/fendo.2023.1238086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 10/30/2023] [Indexed: 12/23/2023] Open
Abstract
Background The survival and prognosis of patients are significantly threatened by cutaneous melanoma (CM), which is a highly aggressive disease. It is therefore crucial to determine the most recent survival rate of CM. This study used population-based cancer registry data to examine the 5-year relative survival rate of CM in the US. Methods Period analysis was used to assess the relative survival rate and trends of patients with CM in the Surveillance, Epidemiology, and End Results (SEER) database during 2004-2018. And based on the data stratified by age, gender, race and subtype in the SEER database, a generalized linear model was 12established to predict the 5-year relative survival rate of CM patients from 2019 to 2023. Results The 5-year relative survival increased to various degrees for both total CM and CM subtypes during the observation period. The improvement was greatest for amelanotic melanoma, increasing from 69.0% to 81.5%. The 5-year overall relative survival rates of CM were 92.9%, 93.5%, and 95.6% for 2004-2008, 2009-2013, and 2014-2018, respectively. Females had a marginally higher survival rate than males for almost all subtypes, older people had lower survival rates than younger people, white patients had higher survival rates than nonwhite ones, and urban locations had higher rates of survival from CM than rural locations did. The survival rate of CM was significantly lower for distant metastasis. Conclusion The survival rate of patients with CM gradually improved overall during 2004-2018. With the predicted survival rate of 96.7% for 2019-2023, this trend will still be present. Assessing the changes experienced by patients with CM over the previous 15 years can help in predicting the future course of CM. It also provides a scientific foundation that associated departments can use to develop efficient tumor prevention and control strategies.
Collapse
Affiliation(s)
- Suzheng Zheng
- Department of Dermatology, The First Affiliated Hospital of Jinan University and Jinan University Institute of Dermatology, Guangzhou, China
| | - Hai Yu
- Department of Dermatology, The First Affiliated Hospital of Jinan University and Jinan University Institute of Dermatology, Guangzhou, China
| | - Xinkai Zheng
- Department of Dermatology, The First Affiliated Hospital of Jinan University and Jinan University Institute of Dermatology, Guangzhou, China
| | - U Tim Wu
- Meng Yi Centre Limited, Macao, Macao SAR, China
| | - Wai-kit Ming
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Hui Huang
- Department of Dermatology, The First Affiliated Hospital of Jinan University and Jinan University Institute of Dermatology, Guangzhou, China
| | - Jiaxin Song
- Department of Dermatology, The First Affiliated Hospital of Jinan University and Jinan University Institute of Dermatology, Guangzhou, China
| | - Xiaoxi Zhang
- Department of Dermatology, The First Affiliated Hospital of Jinan University and Jinan University Institute of Dermatology, Guangzhou, China
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, China
| | - Liehua Deng
- Department of Dermatology, The First Affiliated Hospital of Jinan University and Jinan University Institute of Dermatology, Guangzhou, China
- Department of Dermatology, The Fifth Affiliated Hospital of Jinan University, Heyuan, China
| |
Collapse
|
25
|
Soares PRO, Passos DCS, da Silva FM, da Silva-Giardini APB, Coelho NP, de Oliveira CMA, Kato L, da Silva CC, Guillo L. In vitro antiproliferative and apoptotic effects of thiosemicarbazones based on (-)-camphene and R-(+)-limonene in human melanoma cells. PLoS One 2023; 18:e0295012. [PMID: 38032914 PMCID: PMC10688736 DOI: 10.1371/journal.pone.0295012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/13/2023] [Indexed: 12/02/2023] Open
Abstract
A series of 38 thiosemicarbazone derivatives based on camphene and limonene were evaluated for their antiproliferative activity. Among them, 19 were synthesized and characterized using proton and carbon-13 nuclear magnetic resonance (1H and 13C NMR). For initial compound selection, human melanoma cells (SK-MEL-37) were exposed to a single concentration of a compound (100 μM) for 24, 48, and 72 hours, and cell detachment was visually observed. Cell viability was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method. Nineteen compounds (4, 6, 8, 11, 13, 14, 15, 16, 17, 18, 20, 22, 25, 26, 31, 3', 4', 6', and 9') yielded cell viability below 20%. Subsequently, IC50 values for these compounds were determined, ranging from 11.56 to 55.38 μM, after 72 hours of treatment. Compound 17 (o-hydroxybenzaldehyde (-)-camphene-based thiosemicarbazone) demonstrated the lowest IC50 value, followed by compound 4 (benzaldehyde (-) camphene-based thiosemicarbazone) at 12.84 μM. Regarding compound 4, we observed the induction of a characteristic ladder pattern of DNA fragmentation through gel electrophoresis. Furthermore, fluorescence, flow cytometry and scanning microscopy assays revealed morphological changes consistent with apoptosis induction. Additionally, the measurement of caspase 6 and 8 activity in cellular extracts after treatment for 2, 4, 6, and 24 hours suggested the potential involvement of the extrinsic apoptosis pathway in the mechanism of action of compound 4. Further investigations, including molecular docking studies, are required to fully explore the potential of compound 4 and the other selected compounds, highlighting their promising role in future melanoma therapy research.
Collapse
Affiliation(s)
- Paula Roberta Otaviano Soares
- Department of Biochemistry and Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Débora Cristina Souza Passos
- Department of Biochemistry and Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Francielly Moreira da Silva
- Laboratory of Natural Products and Organic Synthesis, Institute of Chemistry, Federal University of Goiás, Goiânia, Brazil
| | | | - Narcimário Pereira Coelho
- Department of Chemistry, Federal Institute of Mato Grosso do Sul, Nova Andradina, Mato Grosso do Sul, Brazil
| | | | - Lucília Kato
- Laboratory of Natural Products and Organic Synthesis, Institute of Chemistry, Federal University of Goiás, Goiânia, Brazil
| | | | - Lidia Guillo
- Department of Biochemistry and Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, Brazil
| |
Collapse
|
26
|
Kozyra P, Adamczuk G, Karczmarzyk Z, Matysiak J, Podkościelna B, Humeniuk E, Wysocki W, Korga-Plewko A, Senczyna B, Pitucha M. Novel phenoxyacetylthiosemicarbazide derivatives as novel ligands in cancer diseases. Toxicol Appl Pharmacol 2023; 475:116634. [PMID: 37482255 DOI: 10.1016/j.taap.2023.116634] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023]
Abstract
Numerous epidemiological studies report an increased risk of developing prostate cancer in patients with melanoma and an increased risk of developing melanoma in patients with prostate cancer. Based on our previous studies demonstrating the high anticancer activity of thiosemicarbazides with a phenoxy moiety, we designed nineteen phenoxyacetylthiosemicarbazide derivatives and four of them acting as potential dual-ligands for both cancers. All of the compounds were characterized by their melting points and 1H, 13C NMR and IR spectra. For selected compounds, X-ray investigations were carried out to confirm the synthesis pathway, identify the tautomeric form and intra- and intermolecular interaction in the crystalline state. The conformational preferences and electronic structure of molecules were investigated by theoretical calculation method. Lipophilicity of compounds (log kw) was determined using isocratic reversed phase/high pressure liquid chromatography RP-18. For the obtained compounds, in vitro tests were carried out on four melanoma cell lines (A375, G-361, SK-MEL2, SK-MEL28), four prostate cancer cell lines (PC-3, DU-145, LNCaP, VcaP) and a normal human fibroblast cell line (BJ). The most active compounds turned out to be F6. Cell cycle analysis, apoptosis detection, CellROX staining and mitochondrial membrane potential analysis were performed for the most sensitive cancer cells treated with most active compounds. DSC analysis was additionally performed for selected compounds to determine their purity, compatibility, and thermal stability. The process of prooxidation was proposed as a potential mechanism of anticancer activity.
Collapse
Affiliation(s)
- Paweł Kozyra
- Independent Radiopharmacy Unit, Medical University of Lublin, Chodzki 4a, PL-20093 Lublin, Poland.
| | - Grzegorz Adamczuk
- Independent Medical Biology Unit, Faculty of Pharmacy, Medical University of Lublin, PL-20093 Lublin, Poland
| | - Zbigniew Karczmarzyk
- Faculty of Science, Siedlce University of Natural Sciences and Humanities, 3 Maja 54, PL-08110 Siedlce, Poland
| | - Joanna Matysiak
- Department of Chemistry, University of Life Sciences in Lublin, Akademicka 15, PL-20950 Lublin, Poland
| | - Beata Podkościelna
- Department of Polymer Chemistry, Faculty of Chemistry, Maria Curie-Sklodowska University in Lublin, Gliniana 33, PL 20-400 Lublin, Poland
| | - Ewelina Humeniuk
- Independent Medical Biology Unit, Faculty of Pharmacy, Medical University of Lublin, PL-20093 Lublin, Poland
| | - Waldemar Wysocki
- Faculty of Science, Siedlce University of Natural Sciences and Humanities, 3 Maja 54, PL-08110 Siedlce, Poland
| | - Agnieszka Korga-Plewko
- Independent Medical Biology Unit, Faculty of Pharmacy, Medical University of Lublin, PL-20093 Lublin, Poland
| | - Bogusław Senczyna
- Department of Chemistry, University of Life Sciences in Lublin, Akademicka 15, PL-20950 Lublin, Poland
| | - Monika Pitucha
- Independent Radiopharmacy Unit, Medical University of Lublin, Chodzki 4a, PL-20093 Lublin, Poland
| |
Collapse
|
27
|
Ma W, Wu Z, Maghsoudloo M, Ijaz I, Dehghan Shasaltaneh M, Zhang Y, Weng Q, Fu J, Imani S, Wen QL. Dermokine mutations contribute to epithelial-mesenchymal transition and advanced melanoma through ERK/MAPK pathways. PLoS One 2023; 18:e0285806. [PMID: 37432950 PMCID: PMC10335698 DOI: 10.1371/journal.pone.0285806] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/29/2023] [Indexed: 07/13/2023] Open
Abstract
To discover vulnerabilities associated with dermokine (DMKN) as a new trigger of the epithelial-mesenchymal transition (EMT) -driven melanoma, we undertook a genome-wide genetic screening using transgenic. Here, we showed that DMKN expression could be constitutively increased in human malignant melanoma (MM) and that this correlates with poor overall survival in melanoma patients, especially in BRAF-mutated MM samples. Furthermore, in vitro, knockdown of DMKN inhibited the cell proliferation, migration, invasion, and apoptosis of MM cancer cells by the activation of ERK/MAPK signaling pathways and regulator of STAT3 in downstream molecular. By interrogating the in vitro melanoma dataset and characterization of advanced melanoma samples, we found that DMKN downregulated the EMT-like transcriptional program by disrupting EMT cortical actin, increasing the expression of epithelial markers, and decreasing the expression of mesenchymal markers. In addition, whole exome sequencing was presented with p.E69D and p.V91A DMKN mutations as a novel somatic loss of function mutations in those patients. Moreover, our purposeful proof-of-principle modeled the interaction of ERK with p.E69D and p.V91A DMKN mutations in the ERK-MAPK kinas signaling that may be naturally associated with triggering the EMT during melanomagenesis. Altogether, these findings provide preclinical evidence for the role of DMKN in shaping the EMT-like melanoma phenotype and introduced DMKN as a new exceptional responder for personalized MM therapy.
Collapse
Affiliation(s)
- Wenqiong Ma
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Zexiu Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Mazaher Maghsoudloo
- Faculty of Advanced Science and Technology, Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- The Center of Research and Training for Occupational Technical Safety and Health, Tehran, Iran
| | - Iqra Ijaz
- Sichuan Provincial Center for Gynecological and Breast Diseases, Southwest Medical University, Luzhou, Sichuan, China
| | | | - Yuqin Zhang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qiao Weng
- Department of Obstetrics & Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Saber Imani
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Qing Lian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
28
|
Guo YC, Fu ZY, Ding ZJ. Immune infiltration associated C1q acts as a novel prognostic biomarker of cutaneous melanoma. Medicine (Baltimore) 2023; 102:e33088. [PMID: 36897727 PMCID: PMC9997796 DOI: 10.1097/md.0000000000033088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 02/03/2023] [Indexed: 03/11/2023] Open
Abstract
C1q (complement C1q A chain, complement C1q B chain, and complement C1q C chain) is a recognized component of the classical complement pathway that influences the prognosis of various cancers. However, the effects of C1q on cutaneous melanoma (SKCM) outcomes and immune infiltration remain unknown. Gene expression profiling interactive analysis 2 and the human protein atlas were used to evaluate differential expression of C1q mRNA and protein. The relationship between C1q expression and clinicopathological features was also examined. The genetic alterations of C1q and their impact on survival were analyzed using the cbioportal database. The Kaplan-Meier approach was used to assess the significance of C1q in individuals with SKCM. The cluster profiler R package and the cancer single-cell state atlas database were used to investigate the function and mechanism of C1q in SKCM. The relationship between C1q and immune cell infiltration was estimated using single-sample gene set enrichment analysis. C1q expression was increased, and predicted a favorable prognosis. High C1q expression correlated with clinicopathological T stage, pathological stage, overall survival, and disease specific survival events. Moreover, C1q genetic alterations range from 2.7% to 4%, with no impact on prognosis. According to the enrichment analysis, C1q and immune-related pathways were closely connected. The link between complement C1q B chain and the functional state of inflammation was determined using the cancer single-cell state atlas database. In particular, C1q expression was significantly associated with infiltration of most immune cells and checkpoints PDCD1, CD274, and HAVCR2. The results of this study suggest that C1q is associated with prognosis and immune cell infiltration, supporting its value as a diagnostic and prognostic biomarker.
Collapse
Affiliation(s)
- Yi-Cheng Guo
- Dermatology Hospital of Jiangxi Province, Nanchang, China
- Jiangxi Province Clinical Research Center for Skin Diseases, Nanchang, China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Nanchang, Jiangxi, China
| | - Zhi-Yuan Fu
- Dermatology Hospital of Jiangxi Province, Nanchang, China
| | - Zhi-Jun Ding
- Jiangxi Province Clinical Research Center for Skin Diseases, Nanchang, China
| |
Collapse
|
29
|
Orecchini E, Belladonna ML, Pallotta MT, Volpi C, Zizi L, Panfili E, Gargaro M, Fallarino F, Rossini S, Suvieri C, Macchiarulo A, Bicciato S, Mondanelli G, Orabona C. The signaling function of IDO1 incites the malignant progression of mouse B16 melanoma. Oncoimmunology 2023; 12:2170095. [PMID: 36733497 PMCID: PMC9888476 DOI: 10.1080/2162402x.2023.2170095] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Indoleamine 2,3 dioxygenase 1 (IDO1), a leader tryptophan-degrading enzyme, represents a recognized immune checkpoint molecule. In neoplasia, IDO1 is often highly expressed in dendritic cells infiltrating the tumor and/or in tumor cells themselves, particularly in human melanoma. In dendritic cells, IDO1 does not merely metabolize tryptophan into kynurenine but, after phosphorylation of critical tyrosine residues in the non-catalytic small domain, it triggers a signaling pathway prolonging its immunoregulatory effects by a feed-forward mechanism. We here investigated whether the non-enzymatic function of IDO1 could also play a role in tumor cells by using B16-F10 mouse melanoma cells transfected with either the wild-type Ido1 gene (Ido1WT ) or a mutated variant lacking the catalytic, but not signaling activity (Ido1H350A ). As compared to the Ido1WT -transfected counterpart (B16WT), B16-F10 cells expressing Ido1H350A (B16H350A) were characterized by an in vitro accelerated growth mediated by increased Ras and Erk activities. Faster growth and malignant progression of B16H350A cells, also detectable in vivo, were found to be accompanied by a reduction in tumor-infiltrating CD8+ T cells and an increase in Foxp3+ regulatory T cells. Our data, therefore, suggest that the IDO1 signaling function can also occur in tumor cells and that alternative therapeutic approach strategies should be undertaken to effectively tackle this important immune checkpoint molecule.
Collapse
Affiliation(s)
- E Orecchini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - ML Belladonna
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - MT Pallotta
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - C Volpi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - L Zizi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - E Panfili
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - M Gargaro
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - F Fallarino
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - S Rossini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - C Suvieri
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - A Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - S Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - G Mondanelli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - C Orabona
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy,CONTACT C Orabona Department of Medicine and Surgery, University of Perugia, Piazza Severi, Perugia06129, Italy
| |
Collapse
|
30
|
Kahlon N, Doddi S, Yousif R, Najib S, Sheikh T, Abuhelwa Z, Burmeister C, Hamouda DM. Melanoma Treatments and Mortality Rate Trends in the US, 1975 to 2019. JAMA Netw Open 2022; 5:e2245269. [PMID: 36472871 PMCID: PMC9856246 DOI: 10.1001/jamanetworkopen.2022.45269] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
IMPORTANCE Melanoma accounts for most of the deaths due to skin cancer. In the past decade, effective US Food and Drug Administration (FDA)-approved therapies for melanoma have emerged. OBJECTIVE To review changes in the long-term melanoma mortality rate (MMR) trends in the US and determine whether they have any temporal association with the FDA approval of new agents. DESIGN, SETTING, AND PARTICIPANTS This cross-sectional study used population data from the Surveillance, Epidemiology, and End Results (SEER) database and retrospectively reviewed the age-adjusted MMR trends in adult patients (aged ≥18 years) from 1975 to 2019 in the US population. The timeline of the FDA approvals for melanoma treatment was also reviewed. Data were analyzed from March 15 to August 15, 2022. EXPOSURES Outcomes were assessed in association with FDA approval of drugs for the treatment of melanoma. MAIN OUTCOMES AND MEASURES Mortality rates are from the SEER database, reported per 100 000 population and age-adjusted to the 2000 US standard population. The annual percent change (APC) has been used to report long-term trends. RESULTS After the introduction of newer treatments in 2011 (most after 2013), a significant reduction in MMR was seen from 2013 to 2017 in the US for the first time in the past 40 years. Rates increased from 1975 to 1988 (APC, 1.65% [95% CI, 1.30%-2.00%]; P < .001). No statistically significant change in MMR was seen from 1988 to 2013 (APC, 0.01% [95% CI, -1.10% to 0.12%]; P = .85). The MMR decreased significantly from 2013 to 2017 (APC, -6.28% [95% CI, -8.52% to -3.97%]; P < .001). CONCLUSIONS AND RELEVANCE These findings suggest a benefit associated with the availability of effective therapies in the past decade and further suggest that the use of new pharmacological therapies is associated with decreased MMR in the US population. These data are very encouraging and support the continued development of such therapies. Additionally, the accessibility of these treatments and the associated health care costs need to be addressed.
Collapse
Affiliation(s)
- Navkirat Kahlon
- Division of Hematology and Oncology, Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Sishir Doddi
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Rame Yousif
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Sana Najib
- Division of Hematology and Oncology, Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Taha Sheikh
- Division of Hematology and Oncology, Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Ziad Abuhelwa
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Cameron Burmeister
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Danae M. Hamouda
- Division of Hematology and Oncology, Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| |
Collapse
|
31
|
Sosa Cuevas E, Valladeau-Guilemond J, Mouret S, Roubinet B, de Fraipont F, Landemarre L, Charles J, Bendriss-Vermare N, Chaperot L, Aspord C. Unique CLR expression patterns on circulating and tumor-infiltrating DC subsets correlated with clinical outcome in melanoma patients. Front Immunol 2022; 13:1040600. [PMID: 36353633 PMCID: PMC9638162 DOI: 10.3389/fimmu.2022.1040600] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/10/2022] [Indexed: 08/15/2023] Open
Abstract
Subversion of immunity by tumors is a crucial step for their development. Dendritic cells (DCs) are strategic immune cells that orchestrate anti-tumor immune responses but display altered functions in cancer. The bases for such DCs' hijacking are not fully understood. Tumor cells harbor unusual glycosylation patterns of surface glycoproteins and glycolipids. DCs express glycan-binding receptors, named C-type lectin receptors (CLR), allowing them to sense changes in glycan signature of their environment, and subsequently trigger a response. Recognition of tumor glycans by CLRs is crucial for DCs to shape antitumor immunity, and decisive in the orientation of the response. Yet the status of the CLR machinery on DCs in cancer, especially melanoma, remained largely unknown. We explored CLR expression patterns on circulating and tumor-infiltrating cDC1s, cDC2s, and pDCs of melanoma patients, assessed their clinical relevance, and further depicted the correlations between CLR expression profiles and DCs' features. For the first time, we highlighted that the CLR repertoire of circulating and tumor-infiltrating cDC1s, cDC2s, and pDCs was strongly perturbed in melanoma patients, with modulation of DCIR, CLEC-12α and NKp44 on circulating DCs, and perturbation of Dectin-1, CD206, DEC205, DC-SIGN and CLEC-9α on tumor-infiltrating DCs. Furthermore, melanoma tumor cells directly altered CLR expression profiles of healthy DC subsets, and this was associated with specific glycan patterns (Man, Fuc, GlcNAc) that may interact with DCs through CLR molecules. Notably, specific CLR expression profiles on DC subsets correlated with unique DCs' activation status and functionality and were associated with clinical outcome of melanoma patients. Higher proportions of DCIR-, DEC205-, CLEC-12α-expressing cDCs were linked with a better survival, whereas elevated proportions of CD206-, Dectin1-expressing cDCs and NKp44-expressing pDCs were associated with a poor outcome. Thus, melanoma tumor may shape DCs' features by exploiting the plasticity of the CLR machinery. Our study revealed that melanoma manipulates CLR pathways to hijack DC subsets and escape from immune control. It further paved the way to exploit glycan-lectin interactions for the design of innovative therapeutic strategies, which exploit DCs' potentialities while avoiding hijacking by tumor, to properly reshape anti-tumor immunity by manipulating the CLR machinery.
Collapse
Affiliation(s)
- Eleonora Sosa Cuevas
- Etablissement Français du Sang Auvergne-Rhône-Alpes, R&D Laboratory, Grenoble, France
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
| | - Jenny Valladeau-Guilemond
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Stephane Mouret
- Dermatology, Allergology & Photobiology Department, CHU Grenoble Alpes, Grenoble, France
| | | | - Florence de Fraipont
- Medical Unit of Molecular genetic (Hereditary Diseases and Oncology), Grenoble University Hospital, Grenoble, France
| | | | - Julie Charles
- Dermatology, Allergology & Photobiology Department, CHU Grenoble Alpes, Grenoble, France
| | - Nathalie Bendriss-Vermare
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Laurence Chaperot
- Etablissement Français du Sang Auvergne-Rhône-Alpes, R&D Laboratory, Grenoble, France
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
| | - Caroline Aspord
- Etablissement Français du Sang Auvergne-Rhône-Alpes, R&D Laboratory, Grenoble, France
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
| |
Collapse
|
32
|
Kozyra P, Krasowska D, Pitucha M. New Potential Agents for Malignant Melanoma Treatment-Most Recent Studies 2020-2022. Int J Mol Sci 2022; 23:6084. [PMID: 35682764 PMCID: PMC9180979 DOI: 10.3390/ijms23116084] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 02/05/2023] Open
Abstract
Malignant melanoma (MM) is the most lethal skin cancer. Despite a 4% reduction in mortality over the past few years, an increasing number of new diagnosed cases appear each year. Long-term therapy and the development of resistance to the drugs used drive the search for more and more new agents with anti-melanoma activity. This review focuses on the most recent synthesized anti-melanoma agents from 2020-2022. For selected agents, apart from the analysis of biological activity, the structure-activity relationship (SAR) is also discussed. To the best of our knowledge, the following literature review delivers the latest achievements in the field of new anti-melanoma agents.
Collapse
Affiliation(s)
- Paweł Kozyra
- Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Danuta Krasowska
- Department of Dermatology, Venerology and Pediatric Dermatology, Medical University of Lublin, 20-081 Lublin, Poland;
| | - Monika Pitucha
- Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland;
| |
Collapse
|