1
|
Képes Z, Szabó JP, Kálmán-Szabó I, Sass T, Esze R, Opposits G, Jószai I, Szikra D, Fenyvesi F, Hajdu I, Trencsényi G. 52Mn-labelled Beta-cyclodextrin for Melanoma Imaging: A Proof-of-concept Preclinical Study. In Vivo 2024; 38:2591-2600. [PMID: 39477386 PMCID: PMC11535897 DOI: 10.21873/invivo.13735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 11/07/2024]
Abstract
BACKGROUND/AIM As prostaglandin E2 (PGE2) and its receptors (EP2) are over-expressed on tumor cells and microenvironment, radiolabeled cyclodextrins targeting such biomolecules are valuable vector candidates in molecular cancer diagnostics. Using experimental melanoma models, we evaluated the in vivo imaging behavior of novel Manganese-52-labeled (52Mn) randomly methylated beta-cyclodextrin ([52Mn]Mn-DOTAGA-RAMEB) and compared it with the following well-established tumor-specific probes: melanocortin-1 receptor (MC1-R)-affine [68Ga]Ga-DOTA-NAPamide and PGE2 selective [68Ga]Ga-DOTAGA-RAMEB cyclodextrin. MATERIALS AND METHODS Post-injection of [68Ga]Ga-DOTA-NAPamide, [68Ga]Ga-DOTAGA-RAMEB, and [52Mn]Mn-DOTAGA-RAMEB into MC1-R positive B16F10 melanoma-bearing mice, tumor radio-pharmaceutical uptake was quantified in vivo and ex vivo using preclinical positron emission tomography (PET) and high-performance gamma counter. RESULTS Although all tracers performed well in tumor identification, the highest standardized uptake values were detected in the [68Ga]Ga-DOTA-NAPamide scans. Corresponding to the ex vivo data, meaningful [52Mn]Mn-DOTAGA-RAMEB accumulation 1 h post-injection confirmed the tumor-targeting potential of the tracer. Temporal changes in PGE2/EP2 expression of the neoplasms may explain the significant differences observed between the tumor uptake of the two cyclodextrin probes and that of the 52Mn-labelled compound measured 1 h, 4 h, and 3 days post-injection (p≤0.01, p≤0.05). CONCLUSION Although further pharmacokinetical optimization may be required, 52Mn-labelled cyclodextrin holds potential in melanoma diagnostics and the PET-based longitudinal assessment of tumor-associated PGE2/EP2 expression.
Collapse
Affiliation(s)
- Zita Képes
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary;
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Judit P Szabó
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ibolya Kálmán-Szabó
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Sass
- Department of Surgery, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Regina Esze
- Division of Metabolic Diseases, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Opposits
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Jószai
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dezső Szikra
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ferenc Fenyvesi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - István Hajdu
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - György Trencsényi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
2
|
Gonzalez-Melero L, Santos-Vizcaino E, Varela-Calvino R, Gomez-Tourino I, Asumendi A, Boyano MD, Igartua M, Hernandez RM. PLGA-PEI nanoparticle covered with poly(I:C) for personalised cancer immunotherapy. Drug Deliv Transl Res 2024; 14:2788-2803. [PMID: 38427275 PMCID: PMC11525302 DOI: 10.1007/s13346-024-01557-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2024] [Indexed: 03/02/2024]
Abstract
Melanoma is the main cause of death among skin cancers and its incidence worldwide has been experiencing an appalling increase. However, traditional treatments lack effectiveness in advanced or metastatic patients. Immunotherapy, meanwhile, has been shown to be an effective treatment option, but the rate of cancers responding remains far from ideal. Here we have developed a personalized neoantigen peptide-based cancer vaccine by encapsulating patient derived melanoma neoantigens in polyethylenimine (PEI)-functionalised poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) and coating them with polyinosinic:polycytidylic acid (poly(I:C)). We found that PLGA NPs can be effectively modified to be coated with the immunoadjuvant poly(I:C), as well as to encapsulate neoantigens. In addition, we found that both dendritic cells (DCs) and lymphocytes were effectively stimulated. Moreover, the developed NP was found to have a better immune activation profile than NP without poly(I:C) or without antigen. Our results demonstrate that the developed vaccine has a high capacity to activate the immune system, efficiently maturing DCs to present the antigen of choice and promoting the activity of lymphocytes to exert their cytotoxic function. Therefore, the immune response generated is optimal and specific for the elimination of melanoma tumour cells.
Collapse
Affiliation(s)
- Lorena Gonzalez-Melero
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain
| | - Ruben Varela-Calvino
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Santiago de Compostela, Santiago, Spain
| | - Iria Gomez-Tourino
- Centre for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Santiago, Spain
- Health Research Institute of Santiago de Compostela (IDIS), Santiago, Spain
| | - Aintzane Asumendi
- Biocruces Bizkaia Health Research Institute, 48903, Barakaldo, Spain
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain
| | - Maria Dolores Boyano
- Biocruces Bizkaia Health Research Institute, 48903, Barakaldo, Spain
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain
| | - Manoli Igartua
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain.
| | - Rosa Maria Hernandez
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
3
|
Kakish H, Pawar O, Bhatty M, Doh S, Mulligan KM, Rothermel LD, Bordeaux JS, Mangla A, Hoehn RS. Disparities in the Receipt of Systemic Treatment in Metastatic Melanoma. Am J Clin Oncol 2024; 47:239-245. [PMID: 38251734 DOI: 10.1097/coc.0000000000001083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
BACKGROUND In 2011, immunotherapy and targeted therapy revolutionized melanoma treatment. However, inequities in their use may limit the benefits seen by certain patients. METHODS We performed a retrospective review of patients in the National Cancer Database for patients with stage IV melanoma from 2 time periods: 2004-2010 and 2016-2020, distinguishing between those who received systemic therapy and those who did not. We investigated the rates and factors associated with treatment omission. We employed Kaplan-Meier analysis to explore the impact of treatment on overall survival. RESULTS A total of 19,961 patients met the inclusion criteria: 7621 patients were diagnosed in 2004-2010 and 12,340 patients in 2016-2020, of whom 54.9% and 28.3% did not receive systemic treatment, respectively. The rate of "no treatment" has decreased to a plateau of ∼25% in 2020. Median overall survival was improved with treatment in both time periods (2004-2010: 8.8 vs. 5.6 mo [ P <0.05]; and 2016-2020: 25.9 vs. 4.3 mo [ P <0.05]). Nonmedical factors associated with the omission of treatment in both periods included low socioeconomic status, Medicaid or no health insurance, and treatment at low-volume centers. In the period from 2016 to 2020, patients treated at nonacademic programs were also less likely to receive treatment. CONCLUSIONS Systemic therapies significantly improve survival for patients with metastatic melanoma, but significant disparities exist with their receipt. Local efforts are needed to ensure all patients benefit from these revolutionary treatments.
Collapse
Affiliation(s)
- Hanna Kakish
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center
| | - Omkar Pawar
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center
| | - Maira Bhatty
- School of Medicine, Case Western Reserve University School of Medicine
| | - Susan Doh
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center
| | | | - Luke D Rothermel
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center
| | - Jeremy S Bordeaux
- Department of Dermatology, University Hospitals Cleveland Medical Center, Case Western Reserve University
| | - Ankit Mangla
- Department of Medicine, Division of Hematology and Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH
| | - Richard S Hoehn
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center
| |
Collapse
|
4
|
Chen S, Luo Y, He Y, Li M, Liu Y, Zhou X, Hou J, Zhou S. In-situ-sprayed therapeutic hydrogel for oxygen-actuated Janus regulation of postsurgical tumor recurrence/metastasis and wound healing. Nat Commun 2024; 15:814. [PMID: 38280861 PMCID: PMC10821930 DOI: 10.1038/s41467-024-45072-x] [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: 06/30/2023] [Accepted: 01/12/2024] [Indexed: 01/29/2024] Open
Abstract
Surgery is the mainstay of treatment modality for malignant melanoma. However, the deteriorative hypoxic microenvironment after surgery is recognized as a stemming cause for tumor recurrence/metastasis and delayed wound healing. Here we design and construct a sprayable therapeutic hydrogel (HIL@Z/P/H) encapsulating tumor-targeted nanodrug and photosynthetic cyanobacteria (PCC 7942) to prevent tumor recurrence/metastasis while promote wound healing. In a postsurgical B16F10 melanoma model in female mice, the nanodrug can disrupt cellular redox homeostasis via the photodynamic therapy-induced cascade reactions within tumor cells. Besides, the photosynthetically generated O2 by PCC 7942 can not only potentiate the oxidative stress-triggered cell death to prevent local recurrence of residual tumor cells, but also block the signaling pathway of hypoxia-inducible factor 1α to inhibit their distant metastasis. Furthermore, the long-lasting O2 supply and PCC 7942-secreted extracellular vesicles can jointly promote angiogenesis and accelerate the wound healing process. Taken together, the developed HIL@Z/P/H capable of preventing tumor recurrence/metastasis while promoting wound healing shows great application potential for postsurgical cancer therapy.
Collapse
Affiliation(s)
- Shuiling Chen
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yang Luo
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yang He
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Ming Li
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yongjian Liu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Xishen Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jianwen Hou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China.
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Shaobing Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China.
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
5
|
Lombrea A, Watz CG, Bora L, Dehelean CA, Diaconeasa Z, Dinu S, Turks M, Lugiņina J, Peipiņš U, Danciu C. Enhanced Cytotoxicity and Antimelanoma Activity of Novel Semisynthetic Derivatives of Betulinic Acid with Indole Conjugation. PLANTS (BASEL, SWITZERLAND) 2023; 13:36. [PMID: 38202344 PMCID: PMC10780819 DOI: 10.3390/plants13010036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/08/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024]
Abstract
The prevalence and severity of skin cancer, specifically malignant melanoma, among Caucasians remains a significant concern. Natural compounds from plants have long been explored as potential anticancer agents. Betulinic acid (BI) has shown promise in its therapeutic properties, including its anticancer effects. However, its limited bioavailability has hindered its medicinal applications. To address this issue, two recently synthesized semisynthetic derivatives, N-(2,3-indolo-betulinoyl)diglycylglycine (BA1) and N-(2,3-indolo-betulinoyl)glycylglycine (BA2), were compared with previously reported compounds N-(2,3-indolo-betulinoyl)glycine (BA3), 2,3-indolo-betulinic acid (BA4), and BI. These compounds were evaluated for their effects on murine melanoma cells (B164A5) using various in vitro assays. The introduction of an indole framework at the C2 position of BI resulted in an increased cytotoxicity. Furthermore, the cytotoxicity of compound BA4 was enhanced by conjugating its carboxylic group with an amino acid residue. BA2 and BA3, with glycine and glycylglycine residues at C28, exhibited approximately 2.20-fold higher inhibitory activity compared to BA4. The safety assessment of the compounds on human keratinocytes (HaCaT) has revealed that concentrations up to 10 µM slightly reduced cell viability, while concentrations of 75 µM resulted in lower cell viability rates. LDH leakage assays confirmed cell membrane damage in B164A5 cells when exposed to the tested compounds. BA2 and BA3 exhibited the highest LDH release, indicating their strong cytotoxicity. The NR assay revealed dose-dependent lysosome disruption for BI and 2,3-indolo-betulinic acid derivatives, with BA1, BA2, and BA3 showing the most cytotoxic effects. Scratch assays demonstrated concentration-dependent inhibition of cell migration, with BA2 and BA3 being the most effective. Hoechst 3342 staining revealed that BA2 induced apoptosis, while BA3 induced necrosis at lower concentrations, confirming their anti-melanoma properties. In conclusion, the semisynthetic derivatives of BI, particularly BA2 and BA3, show promise as potential candidates for further research in developing effective anti-cancer therapies.
Collapse
Affiliation(s)
- Adelina Lombrea
- Department of Pharmacognosy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania; (A.L.); (L.B.); (C.D.)
- Research Center for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Claudia Geanina Watz
- Research Center for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
- Department of Pharmaceutical Physics, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy of Timisoara, 300041 Timisoara, Romania
| | - Larisa Bora
- Department of Pharmacognosy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania; (A.L.); (L.B.); (C.D.)
- Research Center for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Cristina Adriana Dehelean
- Research Center for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
- Department of Toxicology and Drug Industry, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
| | - Zorita Diaconeasa
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania;
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
| | - Stefania Dinu
- Department of Pedodontics, Faculty of Dental Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 9 No., Revolutiei Bv., 300041 Timisoara, Romania;
- Pediatric Dentistry Research Center, Faculty of Dental Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 9 No., Revolutiei Bv., 300041 Timisoara, Romania
| | - Māris Turks
- Institute of Technology of Organic Chemistry, Faculty of Materials Science and Applied Chemistry, Riga Technical University, P. Valdena Str. 3, LV-1048 Riga, Latvia; (M.T.); (J.L.); (U.P.)
| | - Jevgeņija Lugiņina
- Institute of Technology of Organic Chemistry, Faculty of Materials Science and Applied Chemistry, Riga Technical University, P. Valdena Str. 3, LV-1048 Riga, Latvia; (M.T.); (J.L.); (U.P.)
| | - Uldis Peipiņš
- Institute of Technology of Organic Chemistry, Faculty of Materials Science and Applied Chemistry, Riga Technical University, P. Valdena Str. 3, LV-1048 Riga, Latvia; (M.T.); (J.L.); (U.P.)
- Nature Science Technologies Ltd., Rupnicu Str. 4, LV-2114 Olaine, Latvia
| | - Corina Danciu
- Department of Pharmacognosy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania; (A.L.); (L.B.); (C.D.)
- Research Center for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
| |
Collapse
|
6
|
Malfanti A, Bausart M, Vanvarenberg K, Ucakar B, Préat V. Hyaluronic acid-antigens conjugates trigger potent immune response in both prophylactic and therapeutic immunization in a melanoma model. Drug Deliv Transl Res 2023; 13:2550-2567. [PMID: 37040031 DOI: 10.1007/s13346-023-01337-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2023] [Indexed: 04/12/2023]
Abstract
Immunotherapy of advanced melanoma has encountered significant hurdles in terms of clinical efficacy. Here, we designed a clinically translatable hyaluronic acid (HA)-based vaccine delivering a combination of major histocompatibility complex (MHC) class I- and class II-restricted melanoma antigens (TRP2 and Gp100, respectively) conjugated to HA. HA-nanovaccine (HA-TRP2-Gp100 conjugate) exhibited tropism in the lymph nodes and promoted stimulation of the immune response (2.3-fold higher than the HA+TRP2+Gp100). HA-nanovaccine significantly delayed the growth of B16F10 melanoma and extended survival in both the prophylactic and therapeutic settings (median survival of 22 and 27, respectively, vs 17 days of the untreated group). Moreover, mice prophylactically treated with the HA-nanovaccine displayed significantly higher CD8+ and CD4+ T-cell/Treg ratios in both the spleen and tumor at day 16, suggesting that the HA-nanovaccine overcame the immunosuppressive tumor microenvironment. Superior infiltration of active CD4+ and CD8+ T cells was observed at the endpoint. This study supports the conclusion that HA potentiates the effect of a combination of MHC I and MHC II antigens via a potent immune response against melanoma.
Collapse
Affiliation(s)
- Alessio Malfanti
- Advanced Drug Delivery and Biomaterials, UCLouvain, Louvain Drug Research Institute, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium.
| | - Mathilde Bausart
- Advanced Drug Delivery and Biomaterials, UCLouvain, Louvain Drug Research Institute, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Kevin Vanvarenberg
- Advanced Drug Delivery and Biomaterials, UCLouvain, Louvain Drug Research Institute, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Bernard Ucakar
- Advanced Drug Delivery and Biomaterials, UCLouvain, Louvain Drug Research Institute, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Véronique Préat
- Advanced Drug Delivery and Biomaterials, UCLouvain, Louvain Drug Research Institute, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium.
| |
Collapse
|
7
|
Augustyn K, Joseph J, Patel AB, Razmandi A, Ali AN, Tawbi HA. Treatment experience with encorafenib plus binimetinib for BRAF V600-mutant metastatic melanoma: management insights for clinical practice. Melanoma Res 2023; 33:406-416. [PMID: 37534686 PMCID: PMC10470431 DOI: 10.1097/cmr.0000000000000891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 02/24/2023] [Indexed: 08/04/2023]
Abstract
For patients with locally advanced or metastatic melanoma who have BRAF V600 activating mutations, combination therapy with BRAF and MEK inhibitors is now the standard of care. The combination of encorafenib, a highly selective adenosine triphosphate-competitive BRAF inhibitor, plus binimetinib, a potent, selective, allosteric, non-adenosine triphosphate-competitive MEK1/2 inhibitor, was approved by the US Food and Drug Administration for unresectable or metastatic melanoma with BRAF V600E or V600K mutations based on data from the phase III COLUMBUS study (NCT01909453). Clinical data evaluating BRAF and MEK inhibitor combinations in advanced melanoma indicate a specific profile of adverse events that includes serious retinopathy, skin disorders, and cardiovascular toxicities. Here we provide an overview of the rationale for combining BRAF and MEK inhibitors for the treatment of melanoma, long-term safety results from COLUMBUS, and guidance on managing the most common adverse events associated with this combination based on clinical experience. Proactive and appropriate management of adverse events can allow for longer treatment durations and may result in better treatment outcomes.
Collapse
Affiliation(s)
- Kourtney Augustyn
- Department of Melanoma Medical Oncology, Division of Cancer Medicine
| | | | | | - Azadeh Razmandi
- Department of Head and Neck Surgery, Division of Ophthalmology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Amatul Noor Ali
- Department of Head and Neck Surgery, Division of Ophthalmology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hussein A. Tawbi
- Department of Melanoma Medical Oncology, Division of Cancer Medicine
| |
Collapse
|
8
|
Inamdar S, Suresh AP, Mangal JL, Ng ND, Sundem A, Wu C, Lintecum K, Thumsi A, Khodaei T, Halim M, Appel N, Jaggarapu MMCS, Esrafili A, Yaron JR, Curtis M, Acharya AP. Rescue of dendritic cells from glycolysis inhibition improves cancer immunotherapy in mice. Nat Commun 2023; 14:5333. [PMID: 37660049 PMCID: PMC10475105 DOI: 10.1038/s41467-023-41016-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 08/21/2023] [Indexed: 09/04/2023] Open
Abstract
Inhibition of glycolysis in immune cells and cancer cells diminishes their activity, and thus combining immunotherapies with glycolytic inhibitors is challenging. Herein, a strategy is presented where glycolysis is inhibited in cancer cells using PFK15 (inhibitor of PFKFB3, rate-limiting step in glycolysis), while simultaneously glycolysis and function is rescued in DCs by delivery of fructose-1,6-biphosphate (F16BP, one-step downstream of PFKFB3). To demonstrate the feasibility of this strategy, vaccine formulations are generated using calcium-phosphate chemistry, that incorporate F16BP, poly(IC) as adjuvant, and phosphorylated-TRP2 peptide antigen and tested in challenging and established YUMM1.1 tumours in immunocompetent female mice. Furthermore, to test the versatility of this strategy, adoptive DC therapy is developed with formulations that incorporate F16BP, poly(IC) as adjuvant and mRNA derived from B16F10 cells as antigens in established B16F10 tumours in immunocompetent female mice. F16BP vaccine formulations rescue DCs in vitro and in vivo, significantly improve the survival of mice, and generate cytotoxic T cell (Tc) responses by elevating Tc1 and Tc17 cells within the tumour. Overall, these results demonstrate that rescuing glycolysis of DCs using metabolite-based formulations can be utilized to generate immunotherapy even in the presence of glycolytic inhibitor.
Collapse
Affiliation(s)
- Sahil Inamdar
- Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA
| | | | - Joslyn L Mangal
- Biological Design, Arizona State University, Tempe, AZ, 85281, USA
| | - Nathan D Ng
- Molecular Biosciences and Biotechnology, The College of Liberal Arts and Sciences, Arizona State University, Tempe, AZ, 85281, USA
| | - Alison Sundem
- Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA
| | - Christopher Wu
- Department of Biomedical Engineering, School of Biological and Health System Engineering, Arizona State University, Tempe, AZ, 85281, USA
| | - Kelly Lintecum
- Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA
| | - Abhirami Thumsi
- Biological Design, Arizona State University, Tempe, AZ, 85281, USA
| | - Taravat Khodaei
- Department of Biomedical Engineering, School of Biological and Health System Engineering, Arizona State University, Tempe, AZ, 85281, USA
| | - Michelle Halim
- Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA
| | - Nicole Appel
- Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA
- Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ, 85281, USA
| | | | - Arezoo Esrafili
- Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA
| | - Jordan R Yaron
- Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA
| | - Marion Curtis
- Department of Cancer Biology, Mayo Clinic, Scottsdale, AZ, 85259 8, USA
- College of Medicine and Science, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - Abhinav P Acharya
- Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA.
- Biological Design, Arizona State University, Tempe, AZ, 85281, USA.
- Department of Biomedical Engineering, School of Biological and Health System Engineering, Arizona State University, Tempe, AZ, 85281, USA.
- Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ, 85281, USA.
- Materials Science and Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA.
- Biodesign Center for Biomaterials Innovation and Translation, Arizona State University, Tempe, AZ, 85281, USA.
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, USA.
| |
Collapse
|
9
|
Karimi H, Moskal P, Żak A, Stępień EŁ. 3D melanoma spheroid model for the development of positronium biomarkers. Sci Rep 2023; 13:7648. [PMID: 37169794 PMCID: PMC10175546 DOI: 10.1038/s41598-023-34571-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 05/03/2023] [Indexed: 05/13/2023] Open
Abstract
It was recently demonstrated that newly invented positronium imaging may be used for improving cancer diagnostics by providing additional information about tissue pathology with respect to the standardized uptake value currently available in positron emission tomography (PET). Positronium imaging utilizes the properties of positronium atoms, which are built from the electrons and positrons produced in the body during PET examinations. We hypothesized that positronium imaging would be sensitive to the in vitro discrimination of tumor-like three-dimensional structures (spheroids) built of melanoma cell lines with different cancer activities and biological properties. The lifetime of ortho-positronium (o-Ps) was evaluated in melanoma spheroids from two cell lines (WM266-4 and WM115) differing in the stage of malignancy. Additionally, we considered parameters such as the cell number, spheroid size and melanoma malignancy to evaluate their relationship with the o-Ps lifetime. We demonstrate pilot results for o-Ps lifetime measurement in extracellular matrix-free spheroids. With the statistical significance of two standard deviations, we demonstrated that the higher the degree of malignancy and the rate of proliferation of neoplastic cells, the shorter the lifetime of ortho-positronium. In particular, we observed the following indications encouraging further research: (i) WM266-4 spheroids characterized by a higher proliferation rate and malignancy showed a shorter o-Ps lifetime than WM115 spheroids characterized by a lower growth rate. (ii) Both cell lines showed a decrease in the lifetime of o-Ps after spheroid generation on day 8 compared to day 4 in culture, and the mean o-Ps lifetime was longer for spheroids formed from WM115 cells than for those formed from WM266-4 cells, regardless of spheroid age. The results of this study revealed that positronium is a promising biomarker that may be applied in PET diagnostics for the assessment of the degree of cancer malignancy.
Collapse
Affiliation(s)
- Hanieh Karimi
- Department of Medical Physics, M. Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Łojasiewicza 11 Street, 30-348, Kraków, Poland
- Department of Biochemistry, University of Missouri, Columbia, USA
| | - Paweł Moskal
- Department of Experimental Particle Physics and Applications, M. Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Agata Żak
- Faculty of Chemistry, Jagiellonian University, Kraków, Poland
| | - Ewa Ł Stępień
- Department of Medical Physics, M. Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Łojasiewicza 11 Street, 30-348, Kraków, Poland.
- Center for Theranostics, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
10
|
Liszkay G, Benedek A, Polgár C, Oláh J, Holló P, Emri G, Csejtei A, Kenessey I, Polányi Z, Knollmajer K, Várnai M, Vokó Z, Nagy B, Rokszin G, Fábián I, Barcza Z, Gyulai R, Kiss Z. Significant improvement in melanoma survival over the last decade: A Hungarian nationwide study between 2011 and 2019. J Eur Acad Dermatol Venereol 2023; 37:932-940. [PMID: 36785988 DOI: 10.1111/jdv.18960] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/02/2023] [Indexed: 02/15/2023]
Abstract
BACKGROUND Recent real-world studies have reported significant improvements in the survival of malignant melanoma in the past few years, mainly as a result of modern therapies. However, long-term survival data from Central Eastern European countries such as Hungary are currently lacking. METHODS This nationwide, retrospective study examined melanoma survival in Hungary between 2011-2019 using the databases of the National Health Insurance Fund (NHIF) and Central Statistical Office (CSO) of Hungary. Crude overall survival and age-standardized 5-year net survival as well as the association between age, sex and survival were calculated. RESULTS Between 2011 and 2019, 22,948 newly diagnosed malignant melanoma cases were recorded in the NHIF database (47.89% male, mean age: 60.75 years (SD: ±16.39)). Five-year overall survival was 75.40% (women: 80.78%; men: 69.52%). Patients diagnosed between 2017-2019 had a 20% lower risk of mortality compared to patients diagnosed between 2011-2012 (HR 0.80, 95% CI 0.73-0.89; p < 0.0001). Age-standardized 5-year net survival rates in 2011-2014 and 2015-2019 were 90.6% and 95.8%, respectively (women: 93.1% and 98.4%, men: 87.8% and 92.7%, respectively). The highest age-standardized 5-year net survival rates were found in the 0-39 age cohort (94.6% in the 2015-2019 period). CONCLUSION Hungary has similar melanoma survival rates to Western European countries. Based on net survival, the risk of dying of melanoma within 5 years was cut by more than half (55%) during the study period, which coincides with the successful implementation of awareness campaigns and the wide availability of modern therapies.
Collapse
Affiliation(s)
- Gabriella Liszkay
- Department of Molecular Immunology and Toxicology and the National Tumorbiology Laboratory, National Institute of Oncology, Budapest, Hungary
| | | | - Csaba Polgár
- Department of Molecular Immunology and Toxicology and the National Tumorbiology Laboratory, National Institute of Oncology, Budapest, Hungary
| | - Judit Oláh
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - Péter Holló
- Department of Dermatology, Venereology and Dermatooncology, Semmelweis University, Budapest, Hungary
| | - Gabriella Emri
- Department of Dermatology, University of Debrecen, Debrecen, Hungary
| | - András Csejtei
- Department of Oncoradiology, Markusovszky University Teaching Hospital, Szombathely, Hungary
| | - István Kenessey
- Department of Molecular Immunology and Toxicology and the National Tumorbiology Laboratory, National Institute of Oncology, Budapest, Hungary
| | | | | | - Máté Várnai
- MSD Pharma Hungary Ltd., Budapest, Hungary
- Center for Health Technology Assessment, Semmelweis University, Budapest, Hungary
| | - Zoltán Vokó
- Center for Health Technology Assessment, Semmelweis University, Budapest, Hungary
| | - Balázs Nagy
- Center for Health Technology Assessment, Semmelweis University, Budapest, Hungary
| | | | - Ibolya Fábián
- RxTarget Ltd., Szolnok, Hungary
- University of Veterinary Medicine, Budapest, Hungary
| | - Zsófia Barcza
- Syntesia Medical Communications Ltd., Budapest, Hungary
| | - Rolland Gyulai
- Department of Dermatology, Venereology and Oncodermatology, Faculty of Medicine, University of Pécs, Pécs, Hungary
| | | |
Collapse
|
11
|
Álvaro-Martins MJ, Railean V, Martins F, Machuqueiro M, Pacheco R, Santos S. Synthesis and the In Vitro Evaluation of Antitumor Activity of Novel Thiobenzanilides. Molecules 2023; 28:molecules28041877. [PMID: 36838864 PMCID: PMC9963285 DOI: 10.3390/molecules28041877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/31/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Cancer is a generic term for a large group of diseases that are the second-leading cause of death worldwide, accounting for nearly 10 million deaths in 2020. Melanoma is a highly aggressive skin tumor with an increasing incidence and poor prognosis in the metastatic stage. Breast cancer still stands as one of the major cancer-associated deaths among women, and diagnosed cases are increasing year after year worldwide. Despite the recent therapeutic advances for this type of cancer, novel drugs and treatment strategies are still urgently needed. In this paper, the synthesis of 18 thiobenzanilide derivatives (17 of them new) is described, and their cytotoxic potential against melanoma cells (A375) and hormone-dependent breast cancer (MCF-7) cells is evaluated using the MTT assay. In the A375 cell line, most of the tested thiobenzanilides derivatives showed EC50 values in the order of μM. Compound 17 was the most promising, with an EC50 (24 h) of 11.8 μM. Compounds 8 and 9 are also interesting compounds that deserve to be further improved. The MCF-7 cell line, on the other hand, was seen to be less susceptible to these thiobenzanilides indicating that these compounds show different selectivity towards skin and breast cancer cells. Compound 15 showed the highest cytotoxic potential for MCF-7 cells, with an EC50 (24 h) of 43 μM, a value within the range of the EC50 value determined for tamoxifen (30.0 μM). ADME predictions confirm the potential of the best compounds. Overall, this work discloses a new set of thiobenzanilides that are worth being considered as new scaffolds for the further development of anticancer agents.
Collapse
Affiliation(s)
- Maria João Álvaro-Martins
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Violeta Railean
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
- BioISI-Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Filomena Martins
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Miguel Machuqueiro
- BioISI-Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Rita Pacheco
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
- Departamento de Engenharia Química, Instituto Superior de Engenharia de Lisboa, 1959-007 Lisboa, Portugal
- Correspondence: (R.P.); (S.S.); Tel.: +351-217-500-000 (ext. 28532) (R.P.); +351-217-500-000 (ext. 28513) (S.S.)
| | - Susana Santos
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
- Correspondence: (R.P.); (S.S.); Tel.: +351-217-500-000 (ext. 28532) (R.P.); +351-217-500-000 (ext. 28513) (S.S.)
| |
Collapse
|
12
|
Surman M, Jankowska U, Wilczak M, Przybyło M. Similarities and Differences in the Protein Composition of Cutaneous Melanoma Cells and Their Exosomes Identified by Mass Spectrometry. Cancers (Basel) 2023; 15:cancers15041097. [PMID: 36831440 PMCID: PMC9954195 DOI: 10.3390/cancers15041097] [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: 12/21/2022] [Revised: 01/31/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023] Open
Abstract
Intercellular transport of proteins mediated by extracellular vesicles (EVs)-exosomes and ectosomes-is one of the factors facilitating carcinogenesis. Therefore, the research on protein cargo of melanoma-derived EVs may provide a better understanding of the mechanisms involved in melanoma progression and contribute to the development of alternative biomarkers. Proteomic data on melanoma-derived EVs are very limited. The shotgun nanoLC-MS/MS approach was applied to analyze the protein composition of primary (WM115, WM793) and metastatic (WM266-4, WM1205Lu) cutaneous melanoma cells and exosomes released by them. All cells secreted homogeneous populations of exosomes that shared a characteristic set of proteins. In total, 3514 and 1234 unique proteins were identified in melanoma cells and exosomes, respectively. Gene ontology analysis showed enrichment in several cancer-related categories, including cell proliferation, migration, negative regulation of apoptosis, and angiogenesis. The obtained results broaden our knowledge on the role of selected proteins in exosome biology, as well as their functional role in the development and progression of cutaneous melanoma. The results may also inspire future studies on the clinical potential of exosomes.
Collapse
Affiliation(s)
- Magdalena Surman
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland
| | - Urszula Jankowska
- Proteomics and Mass Spectrometry Core Facility, Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Magdalena Wilczak
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348 Krakow, Poland
| | - Małgorzata Przybyło
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland
- Correspondence: ; Tel.: +48-12-664-6462
| |
Collapse
|
13
|
Chiu Y, Weng H, Lin Y, Lin Y, Yeh Y, Perng C, Ma H, Tsai S, Chou T. Genomic profiling with whole‐exome sequencing revealed distinct mutations and novel pathways in Asian melanoma. J Dermatol 2022; 49:1299-1309. [DOI: 10.1111/1346-8138.16579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/04/2022] [Accepted: 08/25/2022] [Indexed: 12/12/2022]
Affiliation(s)
- Yu‐Jen Chiu
- Institute of Clinical Medicine National Yang Ming Chiao Tung University Taipei Taiwan
- Division of Plastic and Reconstructive Surgery, Department of Surgery Taipei Veterans General Hospital Taipei Taiwan
- Department of Surgery, School of Medicine National Yang Ming Chiao Tung University Taipei Taiwan
| | - Hui‐Ying Weng
- Biomedical Industry PhD Program National Yang Ming Chiao Tung University Taipei Taiwan
| | - Yen‐Yu Lin
- Department of Pathology and Laboratory Medicine Taipei Veterans General Hospital Taipei Taiwan
| | - Yung‐Feng Lin
- Institute of Molecular and Genomic Medicine National Health Research Institutes Miaoli Taiwan
| | - Yi‐Chen Yeh
- Department of Pathology and Laboratory Medicine Taipei Veterans General Hospital Taipei Taiwan
| | - Chern‐Kang Perng
- Division of Plastic and Reconstructive Surgery, Department of Surgery Taipei Veterans General Hospital Taipei Taiwan
- Department of Surgery, School of Medicine National Yang Ming Chiao Tung University Taipei Taiwan
| | - Hsu Ma
- Division of Plastic and Reconstructive Surgery, Department of Surgery Taipei Veterans General Hospital Taipei Taiwan
- Department of Surgery, School of Medicine National Yang Ming Chiao Tung University Taipei Taiwan
- Department of Surgery National Defense Medical Center Taipei Taiwan
| | - Shih‐Feng Tsai
- Institute of Molecular and Genomic Medicine National Health Research Institutes Miaoli Taiwan
| | - Teh‐Ying Chou
- Institute of Clinical Medicine National Yang Ming Chiao Tung University Taipei Taiwan
- Department of Pathology and Laboratory Medicine Taipei Veterans General Hospital Taipei Taiwan
| |
Collapse
|
14
|
Lopes J, Rodrigues CMP, Gaspar MM, Reis CP. Melanoma Management: From Epidemiology to Treatment and Latest Advances. Cancers (Basel) 2022; 14:4652. [PMID: 36230575 PMCID: PMC9562203 DOI: 10.3390/cancers14194652] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022] Open
Abstract
Melanoma is the deadliest skin cancer, whose morbidity and mortality indicators show an increasing trend worldwide. In addition to its great heterogeneity, melanoma has a high metastatic potential, resulting in very limited response to therapies currently available, which were restricted to surgery, radiotherapy and chemotherapy for many years. Advances in knowledge about the pathophysiological mechanisms of the disease have allowed the development of new therapeutic classes, such as immune checkpoint and small molecule kinase inhibitors. However, despite the incontestable progress in the quality of life and survival rates of the patients, effectiveness is still far from desired. Some adverse side effects and resistance mechanisms are the main barriers. Thus, the search for better options has resulted in many clinical trials that are now investigating new drugs and/or combinations. The low water solubility of drugs, low stability and rapid metabolism limit the clinical potential and therapeutic use of some compounds. Thus, the research of nanotechnology-based strategies is being explored as the basis for the broad application of different types of nanosystems in the treatment of melanoma. Future development focus on challenges understanding the mechanisms that make these nanosystems more effective.
Collapse
Affiliation(s)
- Joana Lopes
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Cecília M. P. Rodrigues
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Maria Manuela Gaspar
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Catarina Pinto Reis
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
- Instituto de Biofísica e Engenharia Biomédica, IBEB, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|
15
|
How to Treat Melanoma? The Current Status of Innovative Nanotechnological Strategies and the Role of Minimally Invasive Approaches like PTT and PDT. Pharmaceutics 2022; 14:pharmaceutics14091817. [PMID: 36145569 PMCID: PMC9504126 DOI: 10.3390/pharmaceutics14091817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 12/13/2022] Open
Abstract
Melanoma is the most aggressive type of skin cancer, the incidence and mortality of which are increasing worldwide. Its extensive degree of heterogeneity has limited its response to existing therapies. For many years the therapeutic strategies were limited to surgery, radiotherapy, and chemotherapy. Fortunately, advances in knowledge have allowed the development of new therapeutic strategies. Despite the undoubted progress, alternative therapies are still under research. In this context, nanotechnology is also positioned as a strong and promising tool to develop nanosystems that act as drug carriers and/or light absorbents to potentially improve photothermal and photodynamic therapies outcomes. This review describes the latest advances in nanotechnology field in the treatment of melanoma from 2011 to 2022. The challenges in the translation of nanotechnology-based therapies to clinical applications are also discussed. To sum up, great progress has been made in the field of nanotechnology-based therapies, and our understanding in this field has greatly improved. Although few therapies based on nanoparticulate systems have advanced to clinical trials, it is expected that a large number will come into clinical use in the near future. With its high sensitivity, specificity, and multiplexed measurement capacity, it provides great opportunities to improve melanoma treatment, which will ultimately lead to enhanced patient survival rates.
Collapse
|
16
|
Smith R, Wafa EI, Geary SM, Ebeid K, Alhaj-Suliman SO, Salem AK. Cationic nanoparticles enhance T cell tumor infiltration and antitumor immune responses to a melanoma vaccine. SCIENCE ADVANCES 2022; 8:eabk3150. [PMID: 35857851 PMCID: PMC9299550 DOI: 10.1126/sciadv.abk3150] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 06/03/2022] [Indexed: 06/15/2023]
Abstract
In clinical settings, cancer vaccines as monotherapies have displayed limited success compared to other cancer immunotherapeutic treatments. Nanoscale formulations have the ability to increase the efficacy of cancer vaccines by combatting the immunosuppressive nature of the tumor microenvironment. Here, we have synthesized a previously unexplored cationic polymeric nanoparticle formulation using polyamidoamine dendrimers and poly(d,l-lactic-co-glycolic acid) that demonstrate adjuvant properties in vivo. Tumor-challenged mice vaccinated with an adenovirus-based cancer vaccine [encoding tumor-associated antigen (TAA)] and subsequently treated with this nanoparticulate formulation showed significant increases in TAA-specific T cells in the peripheral blood, reduced tumor burden, protection against tumor rechallenge, and a significant increase in median survival. An investigation into cell-based pathways suggests that administration of the nanoformulation at the site of the developing tumor may have created an inflammatory environment that attracted activated TAA-specific CD8+ T cells to the vicinity of the tumor, thus enhancing the efficacy of the vaccine.
Collapse
Affiliation(s)
| | | | | | - Kareem Ebeid
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Suhaila O. Alhaj-Suliman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | | |
Collapse
|
17
|
Fan KS, Fan KH, Tse PL, Ding H, Su R, Kwok HT. Analysis of top-cited articles on melanoma. ACTA DERMATOVENEROLOGICA ALPINA PANNONICA ET ADRIATICA 2022. [DOI: 10.15570/actaapa.2022.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
18
|
Matias M, Pinho JO, Penetra MJ, Campos G, Reis CP, Gaspar MM. The Challenging Melanoma Landscape: From Early Drug Discovery to Clinical Approval. Cells 2021; 10:3088. [PMID: 34831311 PMCID: PMC8621991 DOI: 10.3390/cells10113088] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/02/2021] [Accepted: 11/06/2021] [Indexed: 02/06/2023] Open
Abstract
Melanoma is recognized as the most dangerous type of skin cancer, with high mortality and resistance to currently used treatments. To overcome the limitations of the available therapeutic options, the discovery and development of new, more effective, and safer therapies is required. In this review, the different research steps involved in the process of antimelanoma drug evaluation and selection are explored, including information regarding in silico, in vitro, and in vivo experiments, as well as clinical trial phases. Details are given about the most used cell lines and assays to perform both two- and three-dimensional in vitro screening of drug candidates towards melanoma. For in vivo studies, murine models are, undoubtedly, the most widely used for assessing the therapeutic potential of new compounds and to study the underlying mechanisms of action. Here, the main melanoma murine models are described as well as other animal species. A section is dedicated to ongoing clinical studies, demonstrating the wide interest and successful efforts devoted to melanoma therapy, in particular at advanced stages of the disease, and a final section includes some considerations regarding approval for marketing by regulatory agencies. Overall, considerable commitment is being directed to the continuous development of optimized experimental models, important for the understanding of melanoma biology and for the evaluation and validation of novel therapeutic strategies.
Collapse
Affiliation(s)
- Mariana Matias
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.M.); (J.O.P.); (M.J.P.)
| | - Jacinta O. Pinho
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.M.); (J.O.P.); (M.J.P.)
| | - Maria João Penetra
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.M.); (J.O.P.); (M.J.P.)
| | - Gonçalo Campos
- CICS–UBI–Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6201-506 Covilhã, Portugal;
| | - Catarina Pinto Reis
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.M.); (J.O.P.); (M.J.P.)
| | - Maria Manuela Gaspar
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (M.M.); (J.O.P.); (M.J.P.)
| |
Collapse
|
19
|
Shigdar S, Schrand B, Giangrande PH, de Franciscis V. Aptamers: Cutting edge of cancer therapies. Mol Ther 2021; 29:2396-2411. [PMID: 34146729 PMCID: PMC8353241 DOI: 10.1016/j.ymthe.2021.06.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 06/06/2021] [Accepted: 06/09/2021] [Indexed: 02/07/2023] Open
Abstract
The development of an aptamer-based therapeutic has rapidly progressed following the first two reports in the 1990s, underscoring the advantages of aptamer drugs associated with their unique binding properties. In 2004, the US Food and Drug Administration (FDA) approved the first therapeutic aptamer for the treatment of neovascular age-related macular degeneration, Macugen developed by NeXstar. Since then, eleven aptamers have successfully entered clinical trials for various therapeutic indications. Despite some of the pre-clinical and clinical successes of aptamers as therapeutics, no aptamer has been approved by the FDA for the treatment of cancer. This review highlights the most recent and cutting-edge approaches in the development of aptamers for the treatment of cancer types most refractory to conventional therapies. Herein, we will review (1) the development of aptamers to enhance anti-cancer immunity and as delivery tools for inducing the expression of immunogenic neoantigens; (2) the development of the most promising therapeutic aptamers designed to target the hard-to-treat cancers such as brain tumors; and (3) the development of "carrier" aptamers able to target and penetrate tumors and metastasis, delivering RNA therapeutics to the cytosol and nucleus.
Collapse
Affiliation(s)
- Sarah Shigdar
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, 75 Pigdons Road, Waurn Ponds, VIC 3216, Australia
| | - Brett Schrand
- TCR(2) Therapeutics, Inc., 100 Binney Street, Cambridge, MA 02142, USA
| | - Paloma H Giangrande
- Internal Medicine, University of Iowa, Iowa City, IA 52242, USA; VP Platform Discovery Sciences, Biology, Wave Life Sciences, Cambridge, MA 02138, USA
| | - Vittorio de Franciscis
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan, Italy; Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
20
|
Serra F, Barruscotti S, Dominioni T, Zuccarini A, Pedrazzoli P, Chiellino S. Treatment Following Progression in Metastatic Melanoma: the State of the Art from Scientific Literature to Clinical Need. Curr Oncol Rep 2021; 23:84. [PMID: 34009481 DOI: 10.1007/s11912-021-01065-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 12/28/2022]
Abstract
INTRODUCTION In the last few years, the advent of targeted therapy and immunotherapy has improved the management and the prognosis of metastatic melanoma, but the spread of resistance mechanisms can lead to disease progression. The clinical management in this setting can be challenging because the oncologist has to decide what is the best treatment strategy among therapy beyond progression (TBP), therapy change, and the rechallenge approach. This review of the relevant scientific literature is intended to clarify which patients with progressing metastatic melanoma will benefit from continuation of ongoing therapy and which ones will not. The data are based on a total of about 4300 patients coming from the main retrospective studies in the chosen field. The article body is divided into four sections which analyze respectively the targeted therapy beyond progression, the immunotherapy beyond progression, the possible treatment sequences, and finally the rechallenge strategy. RECENT FINDINGS Despite the possible approaches of TBP or rechallenge, the patient may not have an optimal response and may need new therapy, which is currently missing. To broaden the pharmacological offer in the fight against melanoma, cancer research is studying new disease targets, like the NRAS, PI3K, and cKIT pathways or combination treatment of targeted therapy plus immunotherapy. Despite the limitations of this work, mainly due to the limited number of studies, their retrospective nature and the lack of comparative studies, the analysis performed allows us to draw some important conclusions: therapy beyond progression, both targeted therapy and immunotherapy, represents a valid treatment option with positive effects on disease control and survival outcomes for patients with low clinical risk, expressed as low disease burden, normal LDH levels, and good performance status; moreover, the prognosis and quality of life of these patients improve when TBP is associated with locoregional treatments. In patients with progressive metastatic melanoma and high clinical risk (high disease burden, high LDH levels, and poor performance status), it is recommended to change therapy, without ever forgetting the possibility of enrolling the patient in a clinical trial. Finally, an efficacious treatment alternative is the rechallenge strategy; this approach consists in a re-treatment with the same drug after a variable interval of discontinuation. Preliminary studies seem to have demonstrated that patients retreated with targeted therapy achieved a greater benefit if they had a low clinical risk and if the drug doublet (BRAF + MEK inhibitors) was used. On the side of immunotherapy, the rechallenge strategy produced a major benefit in patients who prior experienced a severe toxic episode.
Collapse
Affiliation(s)
- F Serra
- Medical Oncology Unit, IRCCS Policlinico San Matteo, Pavia, Italy
| | - S Barruscotti
- Dermatology Unit, IRCCS Policlinico San Matteo, Pavia, Italy
| | - T Dominioni
- General Surgery Unit, IRCCS Policlinico San Matteo, Pavia, Italy
| | - A Zuccarini
- Medical Oncology Unit, IRCCS Policlinico San Matteo, Pavia, Italy
| | - P Pedrazzoli
- Medical Oncology Unit, IRCCS Policlinico San Matteo, Pavia, Italy
| | - S Chiellino
- Medical Oncology Unit, IRCCS Policlinico San Matteo, Pavia, Italy.
| |
Collapse
|
21
|
Coricovac D, Dehelean CA, Pinzaru I, Mioc A, Aburel OM, Macasoi I, Draghici GA, Petean C, Soica C, Boruga M, Vlaicu B, Muntean MD. Assessment of Betulinic Acid Cytotoxicity and Mitochondrial Metabolism Impairment in a Human Melanoma Cell Line. Int J Mol Sci 2021; 22:ijms22094870. [PMID: 34064489 PMCID: PMC8125295 DOI: 10.3390/ijms22094870] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/26/2021] [Accepted: 05/01/2021] [Indexed: 11/30/2022] Open
Abstract
Melanoma represents one of the most aggressive and drug resistant skin cancers with poor prognosis in its advanced stages. Despite the increasing number of targeted therapies, novel approaches are needed to counteract both therapeutic resistance and the side effects of classic therapy. Betulinic acid (BA) is a bioactive phytocompound that has been reported to induce apoptosis in several types of cancers including melanomas; however, its effects on mitochondrial bioenergetics are less investigated. The present study performed in A375 human melanoma cells was aimed to characterize the effects of BA on mitochondrial bioenergetics and cellular behavior. BA demonstrated a dose-dependent inhibitory effect in both mitochondrial respiration and glycolysis in A375 melanoma cells and at sub-toxic concentrations (10 μM) induced mitochondrial dysfunction by eliciting a decrease in the mitochondrial membrane potential and changes in mitochondria morphology and localization. In addition, BA triggered a dose-dependent cytotoxic effect characterized by apoptotic features: morphological alterations (nuclear fragmentation, apoptotic bodies) and the upregulation of pro-apoptotic markers mRNA expression (Bax, Bad and Bak). BA represents a viable therapeutic option via a complex modulatory effect on mitochondrial metabolism that might be useful in advanced melanoma or as reliable strategy to counteract resistance to standard therapy.
Collapse
Affiliation(s)
- Dorina Coricovac
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (D.C.); (C.A.D.); (I.M.); (G.A.D.); (C.P.); (C.S.)
- Research Center for Pharmaco-toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania
| | - Cristina Adriana Dehelean
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (D.C.); (C.A.D.); (I.M.); (G.A.D.); (C.P.); (C.S.)
- Research Center for Pharmaco-toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania
| | - Iulia Pinzaru
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (D.C.); (C.A.D.); (I.M.); (G.A.D.); (C.P.); (C.S.)
- Research Center for Pharmaco-toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania
- Correspondence: (I.P.); (A.M.); Tel.: +40-256-494-604
| | - Alexandra Mioc
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (D.C.); (C.A.D.); (I.M.); (G.A.D.); (C.P.); (C.S.)
- Research Center for Pharmaco-toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania
- Correspondence: (I.P.); (A.M.); Tel.: +40-256-494-604
| | - Oana-Maria Aburel
- Faculty of Medicine “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (O.-M.A.); (M.B.); (B.V.); (M.D.M.)
- Center for Translational Research and Systems Medicine, Faculty of Medicine,” Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Sq. no. 2, RO-300041 Timișoara, Romania
| | - Ioana Macasoi
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (D.C.); (C.A.D.); (I.M.); (G.A.D.); (C.P.); (C.S.)
- Research Center for Pharmaco-toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania
| | - George Andrei Draghici
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (D.C.); (C.A.D.); (I.M.); (G.A.D.); (C.P.); (C.S.)
- Research Center for Pharmaco-toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania
| | - Crina Petean
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (D.C.); (C.A.D.); (I.M.); (G.A.D.); (C.P.); (C.S.)
| | - Codruta Soica
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (D.C.); (C.A.D.); (I.M.); (G.A.D.); (C.P.); (C.S.)
- Research Center for Pharmaco-toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania
| | - Madalina Boruga
- Faculty of Medicine “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (O.-M.A.); (M.B.); (B.V.); (M.D.M.)
| | - Brigitha Vlaicu
- Faculty of Medicine “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (O.-M.A.); (M.B.); (B.V.); (M.D.M.)
| | - Mirela Danina Muntean
- Faculty of Medicine “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (O.-M.A.); (M.B.); (B.V.); (M.D.M.)
- Center for Translational Research and Systems Medicine, Faculty of Medicine,” Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Sq. no. 2, RO-300041 Timișoara, Romania
| |
Collapse
|
22
|
Alrabadi NN, Abushukair HM, Ababneh OE, Syaj SS, Al-Horani SS, Qarqash AA, Darabseh OA, Al-Sous MM, Al-Aomar SR, Ahmed YB, Haddad R, Al Qarqaz FA. Systematic review and meta-analysis efficacy and safety of immune checkpoint inhibitors in advanced melanoma patients with anti-PD-1 progression: a systematic review and meta-analysis. Clin Transl Oncol 2021; 23:1885-1904. [PMID: 33877531 DOI: 10.1007/s12094-021-02598-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/15/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND More than half of melanoma patients taking first-line anti-PD-1 therapy either express transient or no response at all. The efficacy and safety of secondary treatments for these patients are still not well established. Here, we evaluate the efficacy and safety of different melanoma FDA-approved ICI modalities used in post-anti-PD-1 refractory settings. MATERIALS AND METHODS We searched the PubMed database and the ASCO meetings library for studies on advanced melanoma patients with cancer progression on anti-PD-1 therapy and were then treated with ipilimumab, nivolumab/ipilimumab combination, or retreated with anti-PD-1. Primary and secondary endpoints were efficacy and toxicity, respectively. Pooled estimates for each treatment group were obtained using a random or fixed effects model according to detected heterogeneity. RESULTS Fourteen studies, of which 10 on ipilimumab, 2 on anti-PD-1 treatment, and 6 on combination therapies, were included, involving a total of 1460 patients. Twelve studies reported objective response rates (ORRs) and nine of them reported immune-related adverse events (irAEs). As for ORR, patients experienced a response that was inferior compared to the same therapy in treatment -naïve patients, with combination therapy having the best ORR of a pooled 23.08% (95% CI: 16.75% to 30.03%), followed by ipilimumab with a pooled ORR of 8.19% (95% CI: 5.78% to 10.92%). Survival data were also inferior in the ipilimumab cohort (mOS: 5.1 to 7.4 months) compared to ipilimumab in anti-PD-1 naive patients. As for grade 3/4 irAE occurrence, the ipilimumab cohort showed an estimate of 43.77% (95% CI 22.55% to 66.19%). CONCLUSION Our findings provide the best current evidence that patients who progress on anti-PD-1 can still respond to different ICI modalities (ipilimumab with or without nivolumab, and retreatment or continuation beyond progression with anti-PD-1) with tolerable grade 3/4 irAEs. However, more prospective clinical trials are needed to confirm these results.
Collapse
Affiliation(s)
- N N Alrabadi
- Department of Pharmacology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan.
| | - H M Abushukair
- Facullty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - O E Ababneh
- Facullty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - S S Syaj
- Facullty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - S S Al-Horani
- Facullty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - A A Qarqash
- Facullty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - O A Darabseh
- Facullty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - M M Al-Sous
- Facullty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - S R Al-Aomar
- Facullty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Y B Ahmed
- Facullty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - R Haddad
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - F A Al Qarqaz
- Department of Dermatology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| |
Collapse
|
23
|
Kempf E, Zalcman G, Lebbe C. National early access programs and clinical trials: What opportunities for early access to therapeutic innovations for patients with malignant melanoma? Cancer 2021; 127:2181-2183. [PMID: 33788953 DOI: 10.1002/cncr.33495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 02/01/2021] [Indexed: 12/19/2022]
Affiliation(s)
- Emmanuelle Kempf
- Department of Medical Oncology, Galilee Early Phase Clinical Trials Center (CLIP), Henri Mondor-Albert Chenevier University Hospital, Public Hospital Network of Paris (AP-HP), University of Paris-East, Creteil, France
| | - Gérard Zalcman
- Thoracic Oncology Department, Clinical Investigation Center INSERM Unit 1425-CLIP, Bichat-Claude Bernard University Hospital, AP-HP, University of Paris-North, Paris, France
| | - Céleste Lebbe
- Université de Paris, AP-HP Dermatology, INSERM U976, Saint Louis Hospital, Paris, France
| | | |
Collapse
|
24
|
Pacheco-Torres J, Penet MF, Mironchik Y, Krishnamachary B, Bhujwalla ZM. The PD-L1 metabolic interactome intersects with choline metabolism and inflammation. Cancer Metab 2021; 9:10. [PMID: 33608051 PMCID: PMC7893974 DOI: 10.1186/s40170-021-00245-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/08/2021] [Indexed: 12/14/2022] Open
Abstract
Background Harnessing the power of the immune system by using immune checkpoint inhibitors has resulted in some of the most exciting advances in cancer treatment. The full potential of this approach has, however, not been fully realized for treating many cancers such as pancreatic and breast cancer. Cancer metabolism influences many aspects of cancer progression including immune surveillance. An expanded understanding of how cancer metabolism can directly impact immune checkpoints may allow further optimization of immunotherapy. We therefore investigated, for the first time, the relationship between the overexpression of choline kinase-α (Chk-α), an enzyme observed in most cancers, and the expression of the immune checkpoint PD-L1. Methods We used small interfering RNA to downregulate Chk-α, PD-L1, or both in two triple-negative human breast cancer cell lines (MDA-MB-231 and SUM-149) and two human pancreatic ductal adenocarcinoma cell lines (Pa09C and Pa20C). The effects of the downregulation were studied at the genomic, proteomic, and metabolomic levels. The findings were compared with the results obtained by the analysis of public data from The Cancer Genome Atlas Program. Results We identified an inverse dependence between Chk-α and PD-L1 at the genomic, proteomic, and metabolomic levels. We also found that prostaglandin-endoperoxide synthase 2 (COX-2) and transforming growth factor beta (TGF-β) play an important role in this relationship. We independently confirmed this relationship in human cancers by analyzing data from The Cancer Genome Atlas Program. Conclusions Our data identified previously unknown roles of PD-L1 in cancer cell metabolic reprogramming, and revealed the immunosuppressive increased PD-L1 effect of Chk-α downregulation. These data suggest that PD-L1 regulation of metabolism may be mediated through Chk-α, COX-2, and TGF-β. The observations provide new insights that can be applied to the rational design of combinatorial therapies targeting immune checkpoints and cancer metabolism. Supplementary Information The online version contains supplementary material available at 10.1186/s40170-021-00245-w.
Collapse
Affiliation(s)
- Jesus Pacheco-Torres
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Rm 208C Traylor Building, Baltimore, MD, 21205, USA
| | - Marie-France Penet
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Rm 208C Traylor Building, Baltimore, MD, 21205, USA.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yelena Mironchik
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Rm 208C Traylor Building, Baltimore, MD, 21205, USA
| | - Balaji Krishnamachary
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Rm 208C Traylor Building, Baltimore, MD, 21205, USA
| | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Rm 208C Traylor Building, Baltimore, MD, 21205, USA. .,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
25
|
Stępień EŁ, Kamińska A, Surman M, Karbowska D, Wróbel A, Przybyło M. Fourier-Transform InfraRed (FT-IR) spectroscopy to show alterations in molecular composition of EV subpopulations from melanoma cell lines in different malignancy. Biochem Biophys Rep 2021; 25:100888. [PMID: 33458258 PMCID: PMC7797365 DOI: 10.1016/j.bbrep.2020.100888] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/10/2020] [Accepted: 12/18/2020] [Indexed: 12/16/2022] Open
Abstract
Background Melanoma cells release extracellular vesicles (EVs) subpopulations which differ in size, phenotype and molecular content. Melanoma derived EVs play a role in the development and progression of cancer by delivering surface receptors and bioactive (proteins, lipids, nucleic acids) or signaling molecules to target cells. Methods We applied Fourier Transform Infrared Spectroscopy (FTIR) to compare infrared spectra of absorption for different subpopulations of EVs originating from normal human melanocytes, primary cutaneous melanoma (WM115) and metastatic cutaneous melanoma (WM266-4). Results FTIR results showed that exosome and ectosome populations differ in content of protein and lipid components. We obtained higher lipid to protein ratio for ectosomes in comparison with exosomes what confirms that exosomes are very densely packed with protein cargo. We identified the lowest value of saturated fatty acids/unsaturated fatty acids parameter in the metastatic WM266-4 cell line and ectosomes derived from WM266-4 cell line in comparison with normal melanocytes and the primary WM115 cell line. We identified the alterations in the content of secondary structures of proteins present in EV subpopulations originating from melanocytes and melanoma cells in different malignancy. Conclusions Obtained results revealed differences in the molecular composition of melanoma derived EVs subtypes, including protein secondary structure, and showed progressive structural changes during cancer development. Fourier-Transformed Infrared spectroscopy allows recognition lipid and protein content in extracellular vesicles (EVs). Subpopulations of (EVs) from human melanocytes and melanoma cells contain distinct lipid composition and protein structure. Ectosomes from malignant human melanoma are rich in saturated fatty acids and random coiled proteins. Exosomes from malignant human melanoma are bigger in compare to those from melanocytes and have higher lipid to amid ratio.
Collapse
Affiliation(s)
- Ewa Ł Stępień
- Department of Medical Physics, Marian Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348, Kraków, Poland
| | - Agnieszka Kamińska
- Department of Medical Physics, Marian Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348, Kraków, Poland
| | - Magdalena Surman
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387, Kraków, Poland
| | - Dagmara Karbowska
- Department of Medical Physics, Marian Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348, Kraków, Poland
| | - Andrzej Wróbel
- Department of Medical Physics, Marian Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348, Kraków, Poland
| | - Małgorzata Przybyło
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387, Kraków, Poland
| |
Collapse
|
26
|
Sheth M, Ko J. Exploring the relationship between Overall Survival (OS), Progression Free Survival (PFS) and Objective Response Rate (ORR) in patients with advanced melanoma. Cancer Treat Res Commun 2020; 26:100272. [PMID: 33338851 DOI: 10.1016/j.ctarc.2020.100272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 01/02/2023]
Abstract
PURPOSE From 2011 to 2016, 13 randomized clinical trials with active controls were submitted to U.S. Food and Drug Administration (FDA) for the treatment of advanced melanoma. While regular approval is generally granted due to a substantial improvement in overall survival (OS), or a large, clinically meaningful improvement in progression-free survival (PFS), accelerated approval can be granted based on incremental change in a surrogate end point reasonably likely to predict clinical benefit, such as objective response (ORR) of large magnitude and long duration. However, the relationship between objective response rate and progression free survival or objective response rate and overall survival in advanced melanoma has not been established. PATIENTS AND METHODS We conducted analyses to assess the correlation of objective response rate with progression free survival as well as overall survival by examining all advanced melanoma trials submitted to the FDA between 2011 and 2016. In order to examine these relationships, associations between trial-level hazard ratio (HR) of progression free survival, hazard ration of survival and odds ratio of objective response rate were analyzed using a weighted linear regression model. Patient-level responder analyses comparing progression free survival and overall survival between patients with and without an objective response were performed using pooled data from all studies. RESULTS In the trial-level analysis, the linear relationships between progression free survival and objective response rate, and overall survival and objective response rate were weak (R²adj = 0.019 and 0.093, respectively). The linear relationship between overall survival and progression free survival (R²adj = 0.075) was also weak. In the patient-level responder analyses, patients who achieved a response had better progression free survival and overall survival compared with non-responders in both the control drug treatment and the experimental drug treatment. CONCLUSION Based on this analysis, use of objective response rate as a surrogate endpoint for overall survival or progression free survival in this population appears not appropriate. However, due to the nature of heterogeneity, interpretation needs to be cautious.
Collapse
|
27
|
Shofolawe-Bakare OT, Stokes LD, Hossain M, Smith AE, Werfel TA. Immunostimulatory biomaterials to boost tumor immunogenicity. Biomater Sci 2020; 8:5516-5537. [PMID: 33049007 PMCID: PMC7837217 DOI: 10.1039/d0bm01183e] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy is exhibiting great promise as a new therapeutic modality for cancer treatment. However, immunotherapies are limited by the inability of some tumors to provoke an immune response. These tumors with a 'cold' immunological phenotype are characterized by low numbers of tumor-infiltrating lymphocytes, high numbers of immunosuppressive leukocytes (e.g. regulatory T cells, tumor-associated macrophages), and high production of immune-dampening signals (e.g. IL-10, TGF-β, IDO-1). Strategies to boost the aptitude of tumors to initiate an immune response (i.e. boost tumor immunogenicity) will turn 'cold' tumors 'hot' and augment the anti-tumor efficacy of current immunotherapies. Approaches to boost tumor immunogenicity already show promise; however, multifaceted delivery and immunobiology challenges exist. For instance, systemic delivery of many immune-stimulating agents causes off-target toxicity and/or the development of autoimmunity, limiting the administrable dose below the threshold needed to achieve efficacy. Moreover, once administered in vivo, molecules such as the nucleic acid-based agonists for many pattern recognition receptors are either rapidly cleared or degraded, and don't efficiently traffic to the intracellular compartments where the receptors are located. Thus, these nucleic acid-based drugs are ineffective without a delivery system. Biomaterials-based approaches aim to enhance current strategies to boost tumor immunogenicity, enable novel strategies, and spare dose-limiting toxicities. Here, we review recent progress to improve cancer immunotherapies by boosting immunogenicity within tumors using immunostimulatory biomaterials.
Collapse
|
28
|
Alternative Options for Skin Cancer Therapy via Regulation of AKT and Related Signaling Pathways. Int J Mol Sci 2020; 21:ijms21186869. [PMID: 32962182 PMCID: PMC7560163 DOI: 10.3390/ijms21186869] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/11/2020] [Accepted: 09/17/2020] [Indexed: 12/25/2022] Open
Abstract
Global environmental pollution has led to human exposure to ultraviolet (UV) radiation due to the damaged ozone layer, thereby increasing the incidence and death rate of skin cancer including both melanoma and non-melanoma. Overexpression and activation of V-akt murine thymoma viral oncogene homolog (AKT, also known as protein kinase B) and related signaling pathways are major factors contributing to many cancers including lung cancer, esophageal squamous cell carcinoma and skin cancer. Although BRAF inhibitors are used to treat melanoma, further options are needed due to treatment resistance and poor efficacy. Depletion of AKT expression and activation, and related signaling cascades by its inhibitors, decreases the growth of skin cancer and metastasis. Here we have focused the effects of AKT and related signaling (PI3K/AKT/mTOR) pathways by regulators derived from plants and suggest the need for efficient treatment in skin cancer therapy.
Collapse
|
29
|
Luo C, Balsa E, Perry EA, Liang J, Tavares CD, Vazquez F, Widlund HR, Puigserver P. H3K27me3-mediated PGC1α gene silencing promotes melanoma invasion through WNT5A and YAP. J Clin Invest 2020; 130:853-862. [PMID: 31929186 DOI: 10.1172/jci130038] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 10/30/2019] [Indexed: 12/14/2022] Open
Abstract
Oncogene-targeted and immune checkpoint therapies have revolutionized the clinical management of malignant melanoma and now offer hope to patients with advanced disease. Intimately connected to patients' overall clinical risk is whether the initial primary melanoma lesion will metastasize and cause advanced disease, but underlying mechanisms are not entirely understood. A subset of melanomas display heightened peroxisome proliferator-activated receptor γ coactivator 1-α (PGC1α) expression that maintains cell survival cues by promoting mitochondrial function, but also suppresses metastatic spread. Here, we show that PGC1α expression in melanoma cells was silenced by chromatin modifications that involve promoter H3K27 trimethylation. Pharmacological EZH2 inhibition diminished H3K27me3 histone markers, increased PGC1α expression, and functionally suppressed invasion within PGC1α-silenced melanoma cells. Mechanistically, PGC1α silencing activated transcription factor 12 (TCF12), to increase expression of WNT5A, which in turn stabilized YAP protein levels to promote melanoma migration and metastasis. Accordingly, inhibition of components of this transcription-signaling axis, including TCF12, WNT5A, or YAP, blocked melanoma migration in vitro and metastasis in vivo. These results indicate that epigenetic control of melanoma metastasis involved altered expression of PGC1α and an association with the inherent metabolic state of the tumor.
Collapse
Affiliation(s)
- Chi Luo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Eduardo Balsa
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Elizabeth A Perry
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Jiaxin Liang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Clint D Tavares
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Hans R Widlund
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
30
|
Cinobufagin Suppresses Melanoma Cell Growth by Inhibiting LEF1. Int J Mol Sci 2020; 21:ijms21186706. [PMID: 32933177 PMCID: PMC7554883 DOI: 10.3390/ijms21186706] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/09/2020] [Accepted: 09/11/2020] [Indexed: 02/07/2023] Open
Abstract
Constitutive activation of the β-catenin dependent canonical Wnt signaling pathway, which enhances tumor growth and progression in multiple types of cancer, is commonly observed in melanoma. LEF1 activates β-catenin/TCF4 transcriptional activity, promoting tumor growth and progression. Although several reports have shown that LEF1 is highly expressed in melanoma, the functional role of LEF1 in melanoma growth is not fully understood. While A375, A2058, and G361 melanoma cells exhibit abnormally high LEF1 expression, lung cancer cells express lower LEF1 levels. A luciferase assay-based high throughput screening (HTS) with a natural compound library showed that cinobufagin suppressed β-catenin/TCF4 transcriptional activity by inhibiting LEF1 expression. Cinobufagin decreases LEF1 expression in a dose-dependent manner and Wnt/β-catenin target genes such as Axin-2, cyclin D1, and c-Myc in melanoma cell lines. Cinobufagin sensitively attenuates cell viability and induces apoptosis in LEF1 expressing melanoma cells compared to LEF1-low expressing lung cancer cells. In addition, ectopic LEF1 expression is sufficient to attenuate cinobufagin-induced apoptosis and cell growth retardation in melanoma cells. Thus, we suggest that cinobufagin is a potential anti-melanoma drug that suppresses tumor-promoting Wnt/β-catenin signaling via LEF1 inhibition.
Collapse
|
31
|
Combination therapy with TiO 2 nanoparticles and cisplatin enhances chemotherapy response in murine melanoma models. Clin Transl Oncol 2020; 23:738-749. [PMID: 32734535 DOI: 10.1007/s12094-020-02463-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/17/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Despite recent progressions in the treatment of melanoma, the response to conventional therapies and the long-term survival in melanoma patients still remain poor. Recently, the use of nanoparticles (NPs) has been highlighted for promoting the chemotherapeutic effects of cytotoxic drugs in melanoma. The aim of this study is to mechanistically evaluate the potential of titanium dioxide (TiO2) nanoparticles (NPs) for enhancing chemotherapy effects in in vitro and in vivo models of murine melanoma. METHODS The F10 melanoma cells were exposed to different concentrations of TiO2 NPs and/or cisplatin, then cell growth, cell viability, and cell death were evaluated. In parallel, C57BL/6 syngeneic melanoma mice were treated by TiO2 NPs and/or cisplatin, and then drug responses, tumor size and mice's organs were studied pathologically. Autophagy was examined by evaluating the formation of autophagosomes and gene expression levels of autophagy markers (ATG5 and ATG6) by fluorescent microscopy and qPCR, respectively. RESULTS Nontoxic concentrations of TiO2 NPs (50 µg/ml) promote anti-proliferative and cytotoxic effects of cisplatin in F10 melanoma cells, which is mediated through the induction of autophagy and necrotic cell death. Whereas TiO2 NPs have no cytotoxic or metastatic effects in melanoma mice, its combination with cisplatin enhances drug responses (up to 50%), leading to higher inhibition of tumor growth compared with each monotherapy. CONCLUSION The combination of TiO2 NP with cisplatin enhances chemotherapy response in both in vitro and in vivo melanoma models. In addition, autophagy plays an essential role during sensitizing melanoma cells to chemotherapy.
Collapse
|
32
|
Tang Y, Chen M, Xie Q, Li L, Zhu L, Ma Q, Gao S. Construction and evaluation of hyaluronic acid-based copolymers as a targeted chemotherapy drug carrier for cancer therapy. NANOTECHNOLOGY 2020; 31:305702. [PMID: 32272454 DOI: 10.1088/1361-6528/ab884d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Melanoma (MM) is a highly aggressive skin cancer with limited treatment options. Although chemotherapy has been using for advanced melanoma treatment, the lack of targetability, the poor biocompatibility and the severe side effects still hamper the wide applications of chemotherapy agents in MM management. Herein, a biocompatible and biodegradable polymeric hyaluronic acid nanoparticle (HANP) encapsulated with Paclitaxel (PTX) was developed for MM targeted therapy. Our results showed that PTX at 37 ± 2.1% (w/w) was able to be loaded into HANP with over 5 d of stability under physiological conditions. In vitro, HANP/PTX presented hyaluronidase-dependent drug release. Compared to free PTX, HANP/PTX demonstrated a 6-75 times higher growth inhibition in five different cancer cells, while only presenting minimum toxicity to normal cells. After intravenous administration at a 10 mg kg-1 equivalent dose of PTX, HANP/PTX significantly ablated MM tumor growth in a mouse model. As confirmed by 18F-fluoro-2-deoxy-D-glucose (FDG) positron emission tomography (PET) imaging, the tumors started to respond to the HANP/PTX as early as 7 d after the initial treatment, which will significantly benefit for personalized treatment. In conclusion, the HANP/PTX nanocomplex demonstrated great promise as a translational nanomedicine for cancer chemotherapy.
Collapse
Affiliation(s)
- Yuting Tang
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
33
|
Karimi H, Leszczyński B, Kołodziej T, Kubicz E, Przybyło M, Stępień E. X-ray microtomography as a new approach for imaging and analysis of tumor spheroids. Micron 2020; 137:102917. [PMID: 32693343 DOI: 10.1016/j.micron.2020.102917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/26/2020] [Accepted: 07/13/2020] [Indexed: 10/23/2022]
Abstract
Three-dimensional (3D) spheroids mimic important properties of tumors and may soon become a reasonable substitute for animal models and human tissue, eliminating numerous problems related to in vivo and ex vivo experiments and pre-clinical drug trials. Currently, various imaging methods including X-ray microtomography (micro-CT), exist but their spatial resolution is limited. Here, we visualized and provided a morphological analysis of spheroid cell cultures using micro-CT and compared it to that of confocal microscopy. An approach is proposed that can potentially open new diagnostic opportunities to determine the morphology of cancer cells cultured in 3D structures instead of using actual tumors. Spheroids were formed from human melanoma cell lines WM266-4 and WM115 seeded at different cell densities using the hanging drop method. Micro-CT analysis of spheroid showed that spheroid size and shape differed depending on the cell line, initial cell number, and duration of culture. The melanoma cell lines used in this study can successfully be cultured as 3D spheroids and used to substitute human and animal models in pre-clinical studies. The micro-CT allows for high-resolution visualization of the spheroids structure.
Collapse
Affiliation(s)
- Hanieh Karimi
- Department of Medical Physics, M. Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland.
| | - Bartosz Leszczyński
- Department of Medical Physics, M. Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland.
| | - Tomasz Kołodziej
- Department of Molecular and Interfacial Biophysics, M. Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland.
| | - Ewelina Kubicz
- Department of Experimental Particle Physics and Applications, M. Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland.
| | - Małgorzata Przybyło
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Kraków, Poland.
| | - Ewa Stępień
- Department of Medical Physics, M. Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
34
|
Steinberg H. Oncology Nurse Coordinators in Clinical Trials - Shaking up the Melanoma Team. Asia Pac J Oncol Nurs 2020; 7:250-254. [PMID: 32642495 PMCID: PMC7325773 DOI: 10.4103/apjon.apjon_19_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/30/2020] [Indexed: 11/17/2022] Open
Abstract
In recent years, melanoma research has undergone a renaissance. The disease that was once viewed, at least in a metastatic setting, as intractable and untreatable is now revealing its molecular “weaknesses.” The year 2011 was a landmark year for melanoma therapy, with the introduction of two new agents – the anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibody ipilimumab and the BRAF (V-raf murine sarcoma viral oncogene homolog B1) inhibitor vemurafenib. These two agents were shown to confer a survival benefit, which was followed by the approval by the Food and Drug Administration (FDA). In 2014, other immune checkpoint inhibitors, such as pembrolizumab and nivolumab, were approved for the treatment of metastatic melanoma. By 2019, the FDA had also approved pembrolizumab as adjuvant therapy. Target therapy and immunotherapy are now the standard of care for melanoma patients. Clinical trials are currently ongoing for new neoadjuvant therapies. Rapidly evolving knowledge will perhaps downgrade melanoma to the level of a chronic, manageable disease from the intractable “black cancer,” it was in the past and a disease that struck fear into the hearts of those who were diagnosed. Changes in immunotherapy treatments were followed by a large volume of clinical trials. This situation has resulted in the need for changes in the roles of existing melanoma multidisciplinary team members, including the clinical trials nurse (CTN). The role of the CTN is not suitable for these new conditions. A new role and tasks need to be established, evolving the CTN into an oncology nurse coordinator (ONC). In this article, we have described the role and responsibilities of an ONC and the changes that have taken place within the multidisciplinary melanoma team.
Collapse
Affiliation(s)
- Hani Steinberg
- Department of Oncology, Sharett Institute of Oncology, Hadassah Hebrew University Hospital, Kiryat Hadassah, Jerusalem, Israel
| |
Collapse
|
35
|
Kooshkaki O, Derakhshani A, Hosseinkhani N, Torabi M, Safaei S, Brunetti O, Racanelli V, Silvestris N, Baradaran B. Combination of Ipilimumab and Nivolumab in Cancers: From Clinical Practice to Ongoing Clinical Trials. Int J Mol Sci 2020; 21:ijms21124427. [PMID: 32580338 PMCID: PMC7352976 DOI: 10.3390/ijms21124427] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/15/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022] Open
Abstract
Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) are inhibitory checkpoints that are commonly seen on activated T cells and have been offered as promising targets for the treatment of cancers. Immune checkpoint inhibitors (ICIs)targeting PD-1, including pembrolizumab and nivolumab, and those targeting its ligand PD-L1, including avelumab, atezolizumab, and durvalumab, and two drugs targeting CTLA-4, including ipilimumab and tremelimumab have been approved for the treatment of several cancers and many others are under investigating in advanced trial phases. ICIs increased antitumor T cells’ responses and showed a key role in reducing the acquired immune system tolerance which is overexpressed by cancer and tumor microenvironment. However, 50% of patients could not benefit from ICIs monotherapy. To overcome this, a combination of ipilimumab and nivolumab is frequently investigated as an approach to improve oncological outcomes. Despite promising results for the combination of ipilimumab and nivolumab, safety concerns slowed down the development of such strategies. Herein, we review data concerning the clinical activity and the adverse events of ipilimumab and nivolumab combination therapy, assessing ongoing clinical trials to identify clinical outlines that may support combination therapy as an effective treatment. To the best of our knowledge, this paper is one of the first studies to evaluate the efficacy and safety of ipilimumab and nivolumab combination therapy in several cancers.
Collapse
Affiliation(s)
- Omid Kooshkaki
- Student research committee, Birjand University of Medical Sciences, Birjand 9717853577, Iran;
- Department of Immunology, Birjand University of Medical Sciences, Birjand 9717853577, Iran
| | - Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (A.D.); (S.S.)
| | - Negar Hosseinkhani
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran;
| | - Mitra Torabi
- Student research committee, Tabriz University of medical sciences, Tabriz 5165665811, Iran;
| | - Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (A.D.); (S.S.)
| | - Oronzo Brunetti
- Medical Oncology Unit, IRCCS IstitutoTumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
| | - Vito Racanelli
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Nicola Silvestris
- Medical Oncology Unit, IRCCS IstitutoTumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy;
- Correspondence: (N.S.); (B.B.)
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (A.D.); (S.S.)
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran;
- Correspondence: (N.S.); (B.B.)
| |
Collapse
|
36
|
Steeb T, Wessely A, Drexler K, Salzmann M, Toussaint F, Heinzerling L, Reinholz M, Berking C, Heppt MV. The Quality of Practice Guidelines for Melanoma: A Methodologic Appraisal with the AGREE II and AGREE-REX Instruments. Cancers (Basel) 2020; 12:cancers12061613. [PMID: 32570843 PMCID: PMC7352745 DOI: 10.3390/cancers12061613] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/10/2020] [Accepted: 06/17/2020] [Indexed: 12/21/2022] Open
Abstract
Multiple guidelines on cutaneous melanoma (CM) are available from several consortia and countries. To provide up-to-date guidance in the rapidly changing field of melanoma treatment, guideline developers have to provide regular updates without compromises of quality. We performed a systematic search in guideline databases, Medline and Embase to identify guidelines on CM. The methodological quality of the identified guidelines was independently assessed by five reviewers using the instruments “Appraisal of Guidelines for Research and Evaluation” (AGREE II) and “Recommendation EXcellence” (AGREE-REX). We performed descriptive analysis, explored subgroup differences using the Kruskal–Wallis (H) test and examined the relationship between distinct domains and items of the instruments with Spearman’s correlation. Six guidelines by consortia from Australia, France, Germany, Scotland, Spain and the United States of America were included. The German guideline fulfilled 71%–98% of criteria in AGREE II and 78%–96% for AGREE-REX, obtaining the highest scores. Deficiencies in the domains of “applicability” and “values and preferences” were observed in all guidelines. The German and Spanish guidelines significantly differed from each other in most of the domains. The domains “applicability” and “values and preferences” were identified as methodological weaknesses requiring careful revision and improvement in the future.
Collapse
Affiliation(s)
- Theresa Steeb
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (T.S.); (A.W.); (F.T.); (L.H.); (C.B.)
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nürnberg (CCC ER-EMN), 91054 Erlangen, Germany
| | - Anja Wessely
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (T.S.); (A.W.); (F.T.); (L.H.); (C.B.)
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nürnberg (CCC ER-EMN), 91054 Erlangen, Germany
| | - Konstantin Drexler
- Department of Dermatology, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Martin Salzmann
- Department of Dermatology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Frédéric Toussaint
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (T.S.); (A.W.); (F.T.); (L.H.); (C.B.)
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nürnberg (CCC ER-EMN), 91054 Erlangen, Germany
| | - Lucie Heinzerling
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (T.S.); (A.W.); (F.T.); (L.H.); (C.B.)
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nürnberg (CCC ER-EMN), 91054 Erlangen, Germany
| | - Markus Reinholz
- Department of Dermatology and Allergy, University Hospital, Ludwig-Maximilian University, 80337 Munich, Germany;
| | - Carola Berking
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (T.S.); (A.W.); (F.T.); (L.H.); (C.B.)
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nürnberg (CCC ER-EMN), 91054 Erlangen, Germany
| | - Markus V. Heppt
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (T.S.); (A.W.); (F.T.); (L.H.); (C.B.)
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nürnberg (CCC ER-EMN), 91054 Erlangen, Germany
- Correspondence: ; Tel.: +49-9131-85-35747
| |
Collapse
|
37
|
Yang X, Fu H, Ivanov AA. Online informatics resources to facilitate cancer target and chemical probe discovery. RSC Med Chem 2020; 11:611-624. [PMID: 33479663 PMCID: PMC7429978 DOI: 10.1039/d0md00012d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/30/2020] [Indexed: 12/20/2022] Open
Abstract
The advances in cancer genomics, chemical biology, high-throughput screening technologies, and synthetic medicinal chemistry have tremendously expanded the biological space of cancer targets and chemical space of bioactive small molecules to interrogate oncogenic signaling. To explore and leverage these exponentially growing cancer-associated data, a great number of computational tools, databases, and algorithms have been developed. This review summarizes recent cancer-related web resources that allow researchers working at the interface of chemical, biological, and cancer genomics fields to integrate clinical and genomics data for specific actionable targets and selective chemical compounds to facilitate cancer therapeutic discovery.
Collapse
Affiliation(s)
- Xuan Yang
- Department of Pharmacology and Chemical Biology , Emory University School of Medicine , Emory University , 1510 Clifton Road , Atlanta , GA 30322 , USA . ; ; Tel: +1 404 727 6343
- Emory Chemical Biology Discovery Center , Emory University School of Medicine , Emory University , Atlanta , GA , USA
| | - Haian Fu
- Department of Pharmacology and Chemical Biology , Emory University School of Medicine , Emory University , 1510 Clifton Road , Atlanta , GA 30322 , USA . ; ; Tel: +1 404 727 6343
- Emory Chemical Biology Discovery Center , Emory University School of Medicine , Emory University , Atlanta , GA , USA
- Winship Cancer Institute , Emory University , Atlanta , GA , USA
- Department of Hematology & Medical Oncology , Emory University , Atlanta , GA , USA
| | - Andrey A Ivanov
- Department of Pharmacology and Chemical Biology , Emory University School of Medicine , Emory University , 1510 Clifton Road , Atlanta , GA 30322 , USA . ; ; Tel: +1 404 727 6343
- Emory Chemical Biology Discovery Center , Emory University School of Medicine , Emory University , Atlanta , GA , USA
- Winship Cancer Institute , Emory University , Atlanta , GA , USA
| |
Collapse
|
38
|
A Novel Antibody-Drug Conjugate (ADC) Delivering a DNA Mono-Alkylating Payload to Chondroitin Sulfate Proteoglycan (CSPG4)-Expressing Melanoma. Cancers (Basel) 2020; 12:cancers12041029. [PMID: 32331483 PMCID: PMC7226475 DOI: 10.3390/cancers12041029] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/13/2020] [Accepted: 04/18/2020] [Indexed: 12/18/2022] Open
Abstract
Despite emerging targeted and immunotherapy treatments, no monoclonal antibodies or antibody-drug conjugates (ADCs) directly targeting tumor cells are currently approved for melanoma therapy. The tumor-associated antigen chondroitin sulphate proteoglycan 4 (CSPG4), a neural crest glycoprotein over-expressed on 70% of melanomas, contributes to proliferative signaling pathways, but despite highly tumor-selective expression it has not yet been targeted using ADCs. We developed a novel ADC comprising an anti-CSPG4 antibody linked to a DNA minor groove-binding agent belonging to the novel pyrridinobenzodiazepine (PDD) class. Unlike conventional DNA-interactive pyrrolobenzodiazepine (PBD) dimer payloads that cross-link DNA, PDD-based payloads are mono-alkylating agents but have similar efficacy and substantially enhanced tolerability profiles compared to PBD-based cross-linkers. We investigated the anti-tumor activity and safety of the anti-CSPG4-(PDD) ADC in vitro and in human melanoma xenografts. Anti-CSPG4-(PDD) inhibited CSPG4-expressing melanoma cell growth and colony formation and triggered apoptosis in vitro at low nanomolar to picomolar concentrations without off-target Fab-mediated or Fc-mediated toxicity. Anti-CSPG4-(PDD) restricted xenograft growth in vivo at 2 mg/kg doses. One 5 mg/kg injection triggered tumor regression in the absence of overt toxic effects or of acquired residual tumor cell resistance. This anti-CSPG4-(PDD) can deliver a highly cytotoxic DNA mono-alkylating payload to CSPG4-expressing tumors at doses tolerated in vivo.
Collapse
|
39
|
Wang Y, Zhang D. Tanshinol inhibits growth of malignant melanoma cells via regulating miR-1207-5p/CHPF pathway. Arch Dermatol Res 2019; 312:373-383. [PMID: 31828417 DOI: 10.1007/s00403-019-01992-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 09/22/2019] [Accepted: 10/03/2019] [Indexed: 12/19/2022]
Abstract
Tanshinol possesses anti-tumor activity in melanoma both in vitro and in vivo, and miR-1207-5p is involved in tumor progression in melanoma. However, whether miR-1207-5p can be affected by tanshinol treatment in melanoma is not clear. The expression levels of miR-1207-5p were detected by RT-qPCR. The validation of the direct target of miR-1207-5p was through dual-luciferase reporter assay and western blotting assay. The cell viability rate was determined using MTT assay and colony formation assay. The cell mobility was assessed using Transwell migration/invasion assay. Downregulation of miR-1207-5p was found in melanoma cell lines and tissues and was associated with tumor stages, presence of ulceration, lymph node metastasis, and poor overall survival rate of melanoma patients. Tanshinol treatment and miR-1207-5p overexpression suppressed melanoma cell growth and cell mobility. Chondroitin polymerizing factor (CHPF) is a direct target of miR-1207-5p. Tanshinol exerted anti-tumor activity to melanoma through the regulation of miR-1207-5p/CHPF signaling. Our study highlighted the potential therapeutic application of tanshinol and miR-1207-5p as a supplement to enhance the effect of the traditional cancer treatment methods against melanoma.
Collapse
Affiliation(s)
- Yujie Wang
- Yidu Central Hospital of Weifang, No. 4138 Linglongshan South Road, Qingzhou, 262500, Shandong, China
| | - Diancai Zhang
- Yidu Central Hospital of Weifang, No. 4138 Linglongshan South Road, Qingzhou, 262500, Shandong, China.
| |
Collapse
|
40
|
Wang P, Yang W, Shen S, Wu C, Wen L, Cheng Q, Zhang B, Wang X. Differential Diagnosis and Precision Therapy of Two Typical Malignant Cutaneous Tumors Leveraging Their Tumor Microenvironment: A Photomedicine Strategy. ACS NANO 2019; 13:11168-11180. [PMID: 31585038 DOI: 10.1021/acsnano.9b04070] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Elevated hydrogen peroxide (H2O2) in biological tissues is generally recognized to be relevant to the carcinogenesis process that regulates the proliferative activity of cancer cells and the transformation of malignant features. Inspired by this observation, it can be hypothesized that imaging H2O2 in the tumor microenvironment (TME) could help diagnose tumor types and malignancy, and even guide precise therapy. Thus, in this study, a noninvasive photomedicine strategy is demonstrated that leverages the different levels of H2O2 in the TME, and two representative skin cancers, malignant melanoma (MM, clinically higher incidence of metastasis and recurrence) and cutaneous squamous cell carcinoma (cSCC, relatively less dangerous), are differentially diagnosed. The working probe used here is one we previously developed, namely, intelligent H2O2 responsive ABTS-loaded HRP@Gd nanoprobes (iHRANPs). In this study, iHRANPs have advantages over ratiometric imaging due to their bimodal imaging elements, in which the inherent magnetic resonance imaging (MR) mode can be used as the internal imaging reference and the H2O2 responsive photoacoustic (PA) imaging modality can be used for differential diagnosis. Results showed that after intravenous injection of iHRANPs, the tumor signals on both MM and cSCC are obviously enhanced without significant difference under the MR modality. However, under the PA modality, MM and cSCC can be easily distinguished with obvious variations in signal enhancement. Particularly, guided by PA imaging, photothermal therapy (PTT) can be precisely applied on MM, and a strong antitumor effect was achieved owing to the excessive H2O2 in the TME of MM. Furthermore, exogenous H2O2 was injected into cSCC to remedy H2O2 deficiency in the TME of cSCC, and an evident therapeutic efficacy on cSCC can also be realized. This study demonstrated that MM can be differentially diagnosed from cSCC by noninvasive imaging of H2O2 in the TME with iHRANPs; meanwhile, it further enabled imaging-guided precision PTT ablation, even for those unsatisfactory tumor types (cSCC) through exogenously delivering H2O2.
Collapse
Affiliation(s)
- Peiru Wang
- Institute of Photomedicine, Shanghai Skin Disease Hospital , Tongji University School of Medicine , Shanghai 200443 , China
| | - Weitao Yang
- Institute of Photomedicine, Shanghai Skin Disease Hospital , Tongji University School of Medicine , Shanghai 200443 , China
- Tongji University Cancer Center, The Institute for Biomedical Engineering and Nano Science , Tongji University School of Medicine , Shanghai 200092 , China
| | - Shuzhan Shen
- Institute of Photomedicine, Shanghai Skin Disease Hospital , Tongji University School of Medicine , Shanghai 200443 , China
| | - Chao Wu
- School of Physics Science and Engineering , Tongji University , Shanghai 200092 , China
| | - Long Wen
- Institute of Photomedicine, Shanghai Skin Disease Hospital , Tongji University School of Medicine , Shanghai 200443 , China
| | - Qian Cheng
- School of Physics Science and Engineering , Tongji University , Shanghai 200092 , China
| | - Bingbo Zhang
- Institute of Photomedicine, Shanghai Skin Disease Hospital , Tongji University School of Medicine , Shanghai 200443 , China
- Tongji University Cancer Center, The Institute for Biomedical Engineering and Nano Science , Tongji University School of Medicine , Shanghai 200092 , China
| | - Xiuli Wang
- Institute of Photomedicine, Shanghai Skin Disease Hospital , Tongji University School of Medicine , Shanghai 200443 , China
| |
Collapse
|
41
|
Li H, Du H, Zhang G, Wu Y, Qiu P, Liu J, Guo J, Liu X, Sun L, Du B, Tan Y. Curcumin plays a synergistic role in combination with HSV-TK/GCV in inhibiting growth of murine B16 melanoma cells and melanoma xenografts. PeerJ 2019; 7:e7760. [PMID: 31579620 PMCID: PMC6756137 DOI: 10.7717/peerj.7760] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 08/26/2019] [Indexed: 12/24/2022] Open
Abstract
Melanoma is a global concern and accounts for the major mortality of skin cancers. Herpes simplex virus thymidine kinase gene with ganciclovir (HSV-TK/GCV) is a promising gene therapy for melanoma. Despite its low efficiency, it is well known for its bystander effect which is mainly mediated by gap junction. In this study, we found that curcumin reduced B16 melanoma cell viability in both time- and dose-dependent manner. Further study showed that curcumin improved the gap junction intercellular communication (GJIC) function, and upregulated the proteins essential to gap junction, such as connexin 32 and connexin 43, indicating the potential role in enhancing the bystander effect of HSV-TK/GCV. By co-culturing the B16TK cells, which stably expressed TK gene, with wildtype B16 (B16WT) cells, we found that co-treatment of curcumin and GCV synergistically inhibited B16 cell proliferation, but the effect could be eliminated by the gap junction inhibitor AGA. Moreover, curcumin markedly increased apoptosis rate of B16WT cells, suggesting its effect in enhancing the bystander effect of HSV-TK/GCV. In the in-vivo study, we established the xenografted melanoma model in 14 days by injecting mixture of B16TK and B16WT cell in a ratio of 3:7. The result demonstrated that, co-administration of curcumin and GCV significantly inhibited the xenograft growth, as indicated by the smaller size and less weight. The combinational effect was further confirmed as a synergistic effect. In conclusion, the results demonstrated that curcumin could enhance the killing effect and the bystander effect of HSV-TK/GCV in treating melanoma, which might be mediated by improved gap junction. Our data suggested that combination of HSV-TK/GCV with curcumin could be a potential chemosensitization strategy for cancer treatment.
Collapse
Affiliation(s)
- Hong Li
- Department of Biochemistry and Molecular Biology, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haiyan Du
- Department of Biochemistry and Molecular Biology, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guangxian Zhang
- Department of Biochemistry and Molecular Biology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yingya Wu
- Department of Biochemistry and Molecular Biology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pengxiang Qiu
- Department of Biochemistry and Molecular Biology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingjing Liu
- Department of Pathology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jing Guo
- Department of Pathology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xijuan Liu
- Department of Biochemistry and Molecular Biology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingling Sun
- Integrative Cancer Center, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Biaoyan Du
- The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Pathology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuhui Tan
- Department of Biochemistry and Molecular Biology, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
42
|
Eberle J. Countering TRAIL Resistance in Melanoma. Cancers (Basel) 2019; 11:cancers11050656. [PMID: 31083589 PMCID: PMC6562618 DOI: 10.3390/cancers11050656] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 04/27/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022] Open
Abstract
Melanoma of the skin has become a prime example for demonstrating the success of targeted cancer therapy. Nevertheless, high mortality has remained, mainly related to tumor heterogeneity and inducible therapy resistance. But the development of new therapeutic strategies and combinations has raised hope of finally defeating this deadly disease. TNF-related apoptosis-inducing ligand (TRAIL) represents a promising antitumor strategy. The principal sensitivity of melanoma cells for TRAIL was demonstrated in previous studies; however, inducible resistance appeared as a major problem. To address this issue, combination strategies were tested, and survival pathway inhibitors were shown to sensitize melanoma cells for TRAIL-induced apoptosis. Finally, cell cycle inhibition was identified as a common principle of TRAIL sensitization in melanoma cells. Mitochondrial apoptosis pathways, pro- and antiapoptotic Bcl-2 proteins as well as the rheostat consisted of Smac (Second mitochondria-derived activator of caspase) and XIAP (X-linked inhibitor of apoptosis protein) appeared to be of particular importance. Furthermore, the role of reactive oxygen species (ROS) was recognized in this setting. Inducible TRAIL resistance in melanoma can be explained by (i) high levels of antiapoptotic Bcl-2 proteins, (ii) high levels of XIAP, and (iii) suppressed Bax activity. These hurdles have to be overcome to enable the use of TRAIL in melanoma therapy. Several strategies appear as particularly promising, including new TRAIL receptor agonists, Smac and BH3 mimetics, as well as selective kinase inhibitors.
Collapse
Affiliation(s)
- Jürgen Eberle
- Department of Dermatology, Venerology and Allergology, Skin Cancer Center Charité, Charité-Universitätsmedizin Berlin (University Medical Center Charité), 10117 Berlin, Germany.
| |
Collapse
|