1
|
Venuta A, Nasso R, Gisonna A, Iuliano R, Montesarchio S, Acampora V, Sepe L, Avagliano A, Arcone R, Arcucci A, Ruocco MR. Celecoxib, a Non-Steroidal Anti-Inflammatory Drug, Exerts a Toxic Effect on Human Melanoma Cells Grown as 2D and 3D Cell Cultures. Life (Basel) 2023; 13:life13041067. [PMID: 37109596 PMCID: PMC10141119 DOI: 10.3390/life13041067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/06/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023] Open
Abstract
Cutaneous melanoma (CM) remains one of the leading causes of tumor mortality due to its high metastatic spread. CM growth is influenced by inflammation regulated by prostaglandins (PGs) whose synthesis is catalyzed by cyclooxygenases (COXs). COX inhibitors, including non-steroidal anti-inflammatory drugs (NSAIDs), can inhibit tumor development and growth. In particular, in vitro experiments have shown that celecoxib, a NSAID, inhibits the growth of some tumor cell lines. However, two-dimensional (2D) cell cultures, used in traditional in vitro anticancer assays, often show poor efficacy due to a lack of an in vivo like cellular environment. Three-dimensional (3D) cell cultures, such as spheroids, are better models because they can mimic the common features displayed by human solid tumors. Hence, in this study, we evaluated the anti-neoplastic potential of celecoxib, in both 2D and 3D cell cultures of A2058 and SAN melanoma cell lines. In particular, celecoxib reduced the cell viability and migratory capability and triggered the apoptosis of melanoma cells grown as 2D cultures. When celecoxib was tested on 3D melanoma cell cultures, the drug exerted an inhibitory effect on cell outgrowth from spheroids and reduced the invasiveness of melanoma cell spheroids into the hydrogel matrix. This work suggests that celecoxib could represent a new potential therapeutic approach in melanoma therapy.
Collapse
Affiliation(s)
- Alessandro Venuta
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Rosarita Nasso
- Department of Movement Sciences and Wellness, University of Naples "Parthenope", 80133 Naples, Italy
| | - Armando Gisonna
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Roberta Iuliano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Sara Montesarchio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Vittoria Acampora
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Leandra Sepe
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Angelica Avagliano
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Rosaria Arcone
- Department of Movement Sciences and Wellness, University of Naples "Parthenope", 80133 Naples, Italy
| | - Alessandro Arcucci
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Maria Rosaria Ruocco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
2
|
The Association of Improved Overall Survival with NSAIDs in Non-Small Cell Lung Cancer Patients Receiving Immune Checkpoint Inhibitors. Clin Lung Cancer 2023; 24:287-294. [PMID: 36804711 DOI: 10.1016/j.cllc.2022.12.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/22/2022] [Accepted: 12/25/2022] [Indexed: 01/22/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) are commonly used in the management of patients with advanced non-small cell lung cancer (NSCLC), but response is suboptimal. Preclinical data suggest ICI efficacy may be enhanced with concomitant nonsteroidal anti-inflammatory (NSAID) medications. PATIENTS AND METHODS In this retrospective study, the Veterans Health Administration Corporate Data Warehouse was queried for patients diagnosed with NSCLC and treated with ICI from 2010 to 2018. Concomitant NSAID use was defined as NSAID dispensation by a VA pharmacy within 90 days of the any ICI infusion. To mitigate immortal time bias, patients who started NSAIDs 60 or more days after ICI initiation were excluded from analysis. Survival was measured from start of ICI. RESULTS We identified 3634 patients with NSCLC receiving ICI; 2336 (64.3%) were exposed to concomitant NSAIDs. On multivariable analysis, NSAIDs were associated with better overall survival (HR = 0.90; 95% CI, 0.83-0.98; P = .010). When stratifying by NSAID type, diclofenac was the only NSAID with significant association with overall survival (HR = 0.75; 95% CI, 0.68-0.83; P < .001). Propensity score matching of the original cohort yielded 1251 patients per cohort balanced in characteristics. NSAIDs remained associated with improved overall survival (HR = 0.85; 95% CI, 0.78-0.92; P < .001). CONCLUSION This study of Veterans with NSCLC treated with ICI demonstrated that concomitant NSAIDs are associated with longer OS. This may indicate that NSAIDs can enhance ICI-induced antitumor immunity and should prospectively validated.
Collapse
|
3
|
Esmailzadeh A, Shanei A, Attaran N, Hejazi SH, Hemati S. Sonodynamic Therapy Using Dacarbazine-Loaded AuSiO 2 Nanoparticles for Melanoma Treatment: An In-Vitro Study on the B16F10 Murine Melanoma Cell Line. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:1131-1142. [PMID: 35307236 DOI: 10.1016/j.ultrasmedbio.2022.02.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 02/09/2022] [Accepted: 02/13/2022] [Indexed: 06/14/2023]
Abstract
The use of nanoparticles as a sonosensitizer in cancer sonodynamic therapy has been gaining attention because of their great advantages in drug delivery applications. By conjugating chemotherapy agents with nanoparticles, we can develop a drug delivery platform, control drug release and improve the outcome of treatments. The in-vitro study described here evaluates the combination of AuSiO2 nanoparticles and dacarbazine (DTIC@AuSiO2) as a sonosensitizer for sonodynamic therapy of melanoma. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and flow cytometry assays revealed that the viability of B16F10 melanoma cells was significantly inhibited by the increase in apoptosis induction in treatment with DTIC@AuSiO2 nanoparticles under ultrasound exposure compared with treatment with the free DTIC or AuSiO2 nanoparticles. The sonosensitization activity of AuSiO2 nanoparticles and greater uptake of DTIC by tumor cells after loading in DTIC@AuSiO2 nanoparticles inhibited the proliferation of melanoma tumor cells effectively. In conclusion, the DTIC@AuSiO2 nanoparticles established in this study could represent a good drug delivery and sonosensitizer platform for use in melanoma sonodynamic therapy.
Collapse
Affiliation(s)
- Arman Esmailzadeh
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ahmad Shanei
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Neda Attaran
- Department of Medical Nanotechnology, Applied Biophotonics Research Center, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Seyed Hossein Hejazi
- Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Simin Hemati
- Department of Radiation Oncology, School of Medicine, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
4
|
Tudor DV, Bâldea I, Olteanu DE, Fischer-Fodor E, Piroska V, Lupu M, Călinici T, Decea RM, Filip GA. Celecoxib as a Valuable Adjuvant in Cutaneous Melanoma Treated with Trametinib. Int J Mol Sci 2021; 22:4387. [PMID: 33922284 PMCID: PMC8122835 DOI: 10.3390/ijms22094387] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 04/18/2021] [Accepted: 04/20/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Melanoma patients stop responding to targeted therapies mainly due to mitogen activated protein kinase (MAPK) pathway re-activation, phosphoinositide 3 kinase/the mechanistic target of rapamycin (PI3K/mTOR) pathway activation or stromal cell influence. The future of melanoma treatment lies in combinational approaches. To address this, our in vitro study evaluated if lower concentrations of Celecoxib (IC50 in nM range) could still preserve the chemopreventive effect on melanoma cells treated with trametinib. MATERIALS AND METHODS All experiments were conducted on SK-MEL-28 human melanoma cells and BJ human fibroblasts, used as co-culture. Co-culture cells were subjected to a celecoxib and trametinib drug combination for 72 h. We focused on the evaluation of cell death mechanisms, melanogenesis, angiogenesis, inflammation and resistance pathways. RESULTS Low-dose celecoxib significantly enhanced the melanoma response to trametinib. The therapeutic combination reduced nuclear transcription factor (NF)-kB (p < 0.0001) and caspase-8/caspase-3 activation (p < 0.0001), inhibited microphthalmia transcription factor (MITF) and tyrosinase (p < 0.05) expression and strongly down-regulated the phosphatidylinositol-3-kinase/protein kinase B (PI3K/AKT) signaling pathway more significantly than the control or trametinib group (p < 0.0001). CONCLUSION Low concentrations of celecoxib (IC50 in nM range) sufficed to exert antineoplastic capabilities and enhanced the therapeutic response of metastatic melanoma treated with trametinib.
Collapse
Affiliation(s)
- Diana Valentina Tudor
- Department of Physiology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.V.T.); (I.B.); (M.L.); (R.M.D.); (G.A.F.)
| | - Ioana Bâldea
- Department of Physiology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.V.T.); (I.B.); (M.L.); (R.M.D.); (G.A.F.)
| | - Diana Elena Olteanu
- Department of Physiology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.V.T.); (I.B.); (M.L.); (R.M.D.); (G.A.F.)
| | - Eva Fischer-Fodor
- “Prof. Dr. Ion Chiricuță” Oncology Institute, 400015 Cluj-Napoca, Romania; (E.F.-F.); (V.P.)
| | - Virag Piroska
- “Prof. Dr. Ion Chiricuță” Oncology Institute, 400015 Cluj-Napoca, Romania; (E.F.-F.); (V.P.)
| | - Mihai Lupu
- Department of Physiology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.V.T.); (I.B.); (M.L.); (R.M.D.); (G.A.F.)
| | - Tudor Călinici
- Department of Medical Informatics and Biostatistics, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Roxana Maria Decea
- Department of Physiology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.V.T.); (I.B.); (M.L.); (R.M.D.); (G.A.F.)
| | - Gabriela Adriana Filip
- Department of Physiology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.V.T.); (I.B.); (M.L.); (R.M.D.); (G.A.F.)
| |
Collapse
|
5
|
Sesquiterpene Lactone Deoxyelephantopin Isolated from Elephantopus scaber and Its Derivative DETD-35 Suppress BRAF V600E Mutant Melanoma Lung Metastasis in Mice. Int J Mol Sci 2021; 22:ijms22063226. [PMID: 33810045 PMCID: PMC8004649 DOI: 10.3390/ijms22063226] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/16/2022] Open
Abstract
Melanoma is a highly metastatic disease with an increasing rate of incidence worldwide. It is treatment refractory and has poor clinical prognosis; therefore, the development of new therapeutic agents for metastatic melanoma are urgently required. In this study, we created a lung-seeking A375LM5IF4g/Luc BRAFV600E mutant melanoma cell clone and investigated the bioefficacy of a plant sesquiterpene lactone deoxyelephantopin (DET) and its novel semi-synthetic derivative, DETD-35, in suppressing metastatic A375LM5IF4g/Luc melanoma growth in vitro and in a xenograft mouse model. DET and DETD-35 treatment inhibited A375LM5IF4g/Luc cell proliferation, and induced G2/M cell-cycle arrest and apoptosis. Furthermore, A375LM5IF4g/Luc exhibited clonogenic, metastatic and invasive abilities, and several A375LM5IF4g/Luc metastasis markers, N-cadherin, MMP2, vimentin and integrin α4 were significantly suppressed by treatment with either compound. Interestingly, DET- and DETD-35-induced Reactive Oxygen Species (ROS) generation and glutathione (GSH) depletion were found to be upstream events important for the in vitro activities, because exogenous GSH supplementation blunted DET and DETD-35 effects on A375LM5IF4g/Luc cells. DET and DETD-35 also induced mitochondrial DNA mutation, superoxide production, mitochondrial bioenergetics dysfunction, and mitochondrial protein deregulation. Most importantly, DET and DETD-35 inhibited lung metastasis of A375LM5IF4g/Luc in NOD/SCID mice through inhibiting pulmonary vascular permeability and melanoma cell (Mel-A+) proliferation, angiogenesis (VEGF+, CD31+) and EMT (N-cadherin) in the tumor microenvironment in the lungs. These findings indicate that DET and DETD-35 may be useful in the intervention of lung metastatic BRAFV600E mutant melanoma.
Collapse
|
6
|
Wang DY, McQuade JL, Rai RR, Park JJ, Zhao S, Ye F, Beckermann KE, Rubinstein SM, Johnpulle R, Long GV, Carlino MS, Menzies AM, Davies MA, Johnson DB. The Impact of Nonsteroidal Anti-Inflammatory Drugs, Beta Blockers, and Metformin on the Efficacy of Anti-PD-1 Therapy in Advanced Melanoma. Oncologist 2020; 25:e602-e605. [PMID: 32162820 PMCID: PMC7066699 DOI: 10.1634/theoncologist.2019-0518] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/01/2019] [Indexed: 12/13/2022] Open
Abstract
Anti-programmed cell death protein-1 (anti-PD-1) therapy has greatly improved outcomes of patients with melanoma; however, many fail to respond. Although preclinical studies suggest a potentially synergistic relationship with anti-PD-1 therapy and certain concurrent medications, their clinical role remains unclear. Here, we retrospectively evaluated the use of nonsteroidal anti-inflammatory drugs (NSAIDs) and other drugs in 330 patients with melanoma treated with anti-PD-1 therapy from four academic centers. In the cohort, 37% of patients used NSAIDs including aspirin (acetylsalicylic acid; ASA; 47%), cyclooxygenase (COX)-2 inhibitors (2%), and non-ASA/nonselective COX inhibitor NSAIDs (59%). The objective response rates (ORRs) were similar in patients with NSAID (43.4%) and no NSAID (41.3%) use with no significant difference in overall suvival (OS). There was a trend toward improved progression-free survival (PFS) in patients who took NSAIDs (median PFS: 8.5 vs. 5.2 months; p = .054). Most patients (71.3%) took NSAIDs once daily or as needed. Multivariate analysis did not reveal an association with NSAID use with ORR, PFS, or OS. Concurrent use of metformin or beta blockers did not affect ORR, PFS, or OS. Our study found no conclusive association of concurrent NSAID or other medication use with improved outcomes in patients with melanoma treated with anti-PD-1 therapy. Larger and more systematic analysis is required to confirm these findings.
Collapse
Affiliation(s)
| | | | - Rajat R. Rai
- Melanoma Institute AustraliaSydneyAustralia
- The University of SydneySydneyAustralia
| | - John J. Park
- Royal North Shore and Mater HospitalsSydneyAustralia
| | | | - Fei Ye
- Vanderbilt UniversityNashvilleTenneseeUSA
| | | | | | | | - Georgina V. Long
- Melanoma Institute AustraliaSydneyAustralia
- The University of SydneySydneyAustralia
- Royal North Shore and Mater HospitalsSydneyAustralia
| | - Matteo S. Carlino
- Melanoma Institute AustraliaSydneyAustralia
- The University of SydneySydneyAustralia
- Crown Princess Mary Cancer Centre, Westmead HospitalSydneyAustralia
| | - Alexander M. Menzies
- Melanoma Institute AustraliaSydneyAustralia
- The University of SydneySydneyAustralia
- Royal North Shore and Mater HospitalsSydneyAustralia
| | | | | |
Collapse
|
7
|
Tudor DV, Bâldea I, Lupu M, Kacso T, Kutasi E, Hopârtean A, Stretea R, Gabriela Filip A. COX-2 as a potential biomarker and therapeutic target in melanoma. Cancer Biol Med 2020; 17:20-31. [PMID: 32296574 PMCID: PMC7142851 DOI: 10.20892/j.issn.2095-3941.2019.0339] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/15/2020] [Indexed: 12/15/2022] Open
Abstract
With a constantly increasing incidence, cutaneous melanoma has raised the need for a better understanding of its complex microenvironment that may further guide therapeutic options. Melanoma is a model tumor in immuno-oncology. Inflammation represents an important hallmark of cancer capable of inducing and sustaining tumor development. The inflammatory process also orchestrates the adaptative immunosuppression of tumor cells that helps them to evade immune destruction. Besides its role in proliferation, angiogenesis, and apoptosis, cyclooxygenase-2 (COX-2) is a well-known promoter of immune suppression in melanoma. COX-2 inhibitors are closely involved in this condition. This review attempts to answer two controversial questions: is COX-2 a valuable prognostic factor? Among all COX-2 inhibitors, is celecoxib a suitable adjuvant in melanoma therapy?
Collapse
Affiliation(s)
- Diana Valentina Tudor
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Ioana Bâldea
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Mihai Lupu
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Teodor Kacso
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Eniko Kutasi
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Andreea Hopârtean
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Roland Stretea
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Adriana Gabriela Filip
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| |
Collapse
|
8
|
Brusco I, Li Puma S, Chiepe KB, da Silva Brum E, de David Antoniazzi CT, de Almeida AS, Camponogara C, Silva CR, De Logu F, de Andrade VM, Ferreira J, Geppetti P, Nassini R, Oliveira SM, Trevisan G. Dacarbazine alone or associated with melanoma-bearing cancer pain model induces painful hypersensitivity by TRPA1 activation in mice. Int J Cancer 2019; 146:2797-2809. [PMID: 31456221 DOI: 10.1002/ijc.32648] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 02/06/2023]
Abstract
Antineoplastic therapy has been associated with pain syndrome development characterized by acute and chronic pain. The chemotherapeutic agent dacarbazine, used mainly to treat metastatic melanoma, is reported to cause painful symptoms, compromising patient quality of life. Evidence has proposed that transient receptor potential ankyrin 1 (TRPA1) plays a critical role in chemotherapy-induced pain syndrome. Here, we investigated whether dacarbazine causes painful hypersensitivity in naive or melanoma-bearing mice and the involvement of TRPA1 in these models. Mouse dorsal root ganglion (DRG) neurons and human TRPA1-transfected HEK293 (hTRPA1-HEK293) cells were used to evaluate the TRPA1-mediated calcium response evoked by dacarbazine. Mechanical and cold allodynia were evaluated after acute or repeated dacarbazine administration in naive mice or after inoculation of B16-F10 melanoma cells in C57BL/6 mice. TRPA1 involvement was investigated by using pharmacological and genetic tools (selective antagonist or antisense oligonucleotide treatment and Trpa1 knockout mice). Dacarbazine directly activated TRPA1 in hTRPA1-HEK293 cells and mouse DRG neurons and appears to sensitize TRPA1 indirectly by generating oxidative stress products. Moreover, dacarbazine caused mechanical and cold allodynia in naive but not Trpa1 knockout mice. Also, dacarbazine-induced nociception was reduced by the pharmacological TRPA1 blockade (antagonism), antioxidants, and by ablation of TRPA1 expression. TRPA1 pharmacological blockade also reduced dacarbazine-induced nociception in a tumor-associated pain model. Thus, dacarbazine causes nociception by TRPA1 activation, indicating that this receptor may represent a pharmacological target for treating chemotherapy-induced pain syndrome in cancer patients submitted to antineoplastic treatment with dacarbazine.
Collapse
Affiliation(s)
- Indiara Brusco
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, Brazil
| | - Simone Li Puma
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Kelly Braga Chiepe
- Graduate Program in Health Science, University of the Extreme South of Santa Catarina-Unesc, Criciúma, Brazil
| | - Evelyne da Silva Brum
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, Brazil
| | | | - Amanda Spring de Almeida
- Graduate Program in Physiology and Pharmacology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
| | - Camila Camponogara
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, Brazil
| | - Cássia Regina Silva
- Graduate Program in Genetics and Biochemistry, Biotechnology Institute, Federal University of Uberlandia, Uberlandia, Brazil
| | - Francesco De Logu
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Vanessa Moraes de Andrade
- Graduate Program in Health Science, University of the Extreme South of Santa Catarina-Unesc, Criciúma, Brazil
| | - Juliano Ferreira
- Graduate Program in Pharmacology, Federal University of Santa Catarina, Florianopolis, Brazil
| | | | - Romina Nassini
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Sara Marchesan Oliveira
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, Brazil
| | - Gabriela Trevisan
- Graduate Program in Health Science, University of the Extreme South of Santa Catarina-Unesc, Criciúma, Brazil.,Graduate Program in Physiology and Pharmacology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
| |
Collapse
|
9
|
Gonçalves JP, Potrich FB, Ferreira Dos Santos ML, Costa Gagosian VS, Rodrigues Rossi G, Jacomasso T, Mendes A, Bonciani Nader H, Brochado Winnischofer SM, Trindade ES, Camargo De Oliveira C. In vitro attenuation of classic metastatic melanoma‑related features by highly diluted natural complexes: Molecular and functional analyses. Int J Oncol 2019; 55:721-732. [PMID: 31364728 DOI: 10.3892/ijo.2019.4846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 04/12/2019] [Indexed: 11/05/2022] Open
Abstract
Metastasis is responsible for the majority of deaths among patients with malignant melanoma. Despite recent advances, the majority of current and modern therapies are ineffective and/or financially unfeasible. Thus, in this study, we investigated two low‑cost highly‑diluted natural complexes (HDNCs) that have been shown to be effective against malignant melanoma in a murine model in vivo. The aim of this study was to determine the mechanisms through which these HDNCs directly affect melanoma cells, either alone or in an artificial tumor microenvironment, suppressing the metastatic phenotype, thus explaining previous in vivo effects. For this purpose, HDNC in vitro treatments of B16‑F10 melanoma cells, alone or in co‑culture with Balb/3T3 fibroblasts, were carried out. Molecular biology techniques and standard functional assays were used to assess the changes in molecule expression and in cell behaviors related to the metastatic phenotype. Melanoma progression features were found to be regulated by HDNCs. Molecules related to cell adhesion (N‑cadherin, β1‑integrin and CD44), and migration, extracellular matrix remodeling and angiogenesis were modulated. The cell migratory, invasive and clonogenic capacities were reduced by the HDNCs. No loss of cell proliferation or viability were observed. On the whole, the findings of this study indicate that HDNCs directly reprogram, molecularly and functionally, melanoma cells in vitro, modulating their metastatic phenotype. Such findings are likely to be responsible for the attenuation of tumor growth and lung colonization previously observed in vivo.
Collapse
Affiliation(s)
- Jenifer Pendiuk Gonçalves
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Federal University of Paraná, CEP 81530‑980 Curitiba‑PR, Brazil
| | - Francine Bittencourt Potrich
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Federal University of Paraná, CEP 81530‑980 Curitiba‑PR, Brazil
| | - Maria Luiza Ferreira Dos Santos
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Federal University of Paraná, CEP 81530‑980 Curitiba‑PR, Brazil
| | - Viviana Stephanie Costa Gagosian
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Federal University of Paraná, CEP 81530‑980 Curitiba‑PR, Brazil
| | - Gustavo Rodrigues Rossi
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Federal University of Paraná, CEP 81530‑980 Curitiba‑PR, Brazil
| | - Thiago Jacomasso
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Federal University of Paraná, CEP 81530‑980 Curitiba‑PR, Brazil
| | - Aline Mendes
- Biochemistry Department, Federal University of São Paulo, São Paulo ‑ SP 04023‑062, Brazil
| | - Helena Bonciani Nader
- Biochemistry Department, Federal University of São Paulo, São Paulo ‑ SP 04023‑062, Brazil
| | - Sheila Maria Brochado Winnischofer
- Biochemistry and Molecular Biology Department, Section of Biological Sciences, Federal University of Paraná, CEP 81530‑980 Curitiba‑PR, Brazil
| | - Edvaldo S Trindade
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Federal University of Paraná, CEP 81530‑980 Curitiba‑PR, Brazil
| | - Carolina Camargo De Oliveira
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Federal University of Paraná, CEP 81530‑980 Curitiba‑PR, Brazil
| |
Collapse
|
10
|
Ahmed KS, Changling S, Shan X, Mao J, Qiu L, Chen J. Liposome-based codelivery of celecoxib and doxorubicin hydrochloride as a synergistic dual-drug delivery system for enhancing the anticancer effect. J Liposome Res 2019; 30:285-296. [DOI: 10.1080/08982104.2019.1634724] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Kamel S. Ahmed
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Sun Changling
- Department of Otolaryngology–Head and Neck Surgery, Hospital of Jiangnan University, Wuxi, China
| | - Xiaotian Shan
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
| | - Jing Mao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
| | - Lipeng Qiu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
| | - Jinghua Chen
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
| |
Collapse
|
11
|
Buzharevski A, Paskas S, Sárosi MB, Laube M, Lönnecke P, Neumann W, Mijatovic S, Maksimovic-Ivanic D, Pietzsch J, Hey-Hawkins E. Carboranyl Analogues of Celecoxib with Potent Cytostatic Activity against Human Melanoma and Colon Cancer Cell Lines. ChemMedChem 2019; 14:315-321. [PMID: 30602073 DOI: 10.1002/cmdc.201800685] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/14/2018] [Indexed: 12/13/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are the most common way of treating inflammatory disorders. Their widespread use helped reveal their other modes of action as pharmaceuticals, such as a profound effect on various cancers. Celecoxib has proven to be a very prominent member of this group with cytostatic activities. On the other hand, the highly dynamic field of drug design is constantly searching for new ways of modifying known structures to obtain more powerful and less harmful drugs. A very interesting development is the implementation of carboranes in pharmacologically active structures, mostly as phenyl mimetics. Herein we report the synthesis of three carborane-containing derivatives of the COX-2-selective NSAID celecoxib. The new compounds proved to have promising cytostatic potential against various melanoma and colorectal adenocarcinoma cell lines. Inhibited proliferation accompanied by caspase-independent apoptotic cell death was found to be the main cause of decreased cell viability upon treatment with the most efficient celecoxib analogue, 3 b (4-[5-(1,7-dicarba-closo-dodecaboranyl)-3-trifluoromethyl-1H-pyrazol-1-yl]-1-methylsulfonylbenzene).
Collapse
Affiliation(s)
- Antonio Buzharevski
- Institut für Anorganische Chemie, Universität Leipzig, Johannisallee 29, 04103, Leipzig, Germany
| | - Svetlana Paskas
- Department of Immunology, Institute for Biological Research "Sinisa Stankovic", Belgrade University, Belgrade, Serbia
| | - Menyhárt-Botond Sárosi
- Institut für Anorganische Chemie, Universität Leipzig, Johannisallee 29, 04103, Leipzig, Germany
| | - Markus Laube
- Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Peter Lönnecke
- Institut für Anorganische Chemie, Universität Leipzig, Johannisallee 29, 04103, Leipzig, Germany
| | - Wilma Neumann
- Institut für Anorganische Chemie, Universität Leipzig, Johannisallee 29, 04103, Leipzig, Germany
| | - Sanja Mijatovic
- Department of Immunology, Institute for Biological Research "Sinisa Stankovic", Belgrade University, Belgrade, Serbia
| | - Danijela Maksimovic-Ivanic
- Department of Immunology, Institute for Biological Research "Sinisa Stankovic", Belgrade University, Belgrade, Serbia
| | - Jens Pietzsch
- Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328, Dresden, Germany.,Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstrasse 4, 01062, Dresden, Germany
| | - Evamarie Hey-Hawkins
- Institut für Anorganische Chemie, Universität Leipzig, Johannisallee 29, 04103, Leipzig, Germany
| |
Collapse
|
12
|
Navarro SD, Pessatto LR, Meza A, de Oliveira EJT, Auharek SA, Vilela LC, de Lima DP, de Azevedo RB, Kassuya CAL, Cáceres OIA, da Silva Gomes R, Beatriz A, Oliveira RJ, Martines MAU. Resorcinolic lipid 3-heptyl-3,4,6-trimethoxy-3H-isobenzofuran-1-one is a strategy for melanoma treatment. Life Sci 2018; 209:300-312. [PMID: 30102904 DOI: 10.1016/j.lfs.2018.08.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 08/02/2018] [Accepted: 08/08/2018] [Indexed: 12/12/2022]
Abstract
AIMS Previous studies performed by our research group indicated that cytosporone analogues are capable of prevent or repair DNA damages. This work presents the evaluation of the activity of AMS35AA for metastatic murine melanoma cells (B16F10) in experimental model in vitro and, in pre-clinic assay of metastatic melanoma in vivo, using mice lineage C57BL/6. MAIN METHODS In vitro assays were performed: MTT and comet assay, flow cytometry evaluation, gene expression assay by RT-PCR, qualitative evaluation of cell death using B16F10 cells. In vivo assays: micronucleus and comet assay, splenic phagocytosis, melanoma murine model and histopathological analysis, using mice lineage C57BL/6 (n = 20). KEY FINDINGS In vitro results performed by MTT assay showed that AMS35AA is cytotoxic for B16F10 cells (p < 0.05). Based on comet assay the genotoxicity of the IC50 was determined (95.83 μg/mL) (p < 0.05). These data were corroborated by flow cytometry analysis after the treatment with AMS35AA, which indicates the cellular death by apoptosis (p < 0.05) and increasing of ATR, p53, p21 and GADD45 gene expressions verified using RT-PCR. With respect to in vivo results, it was observed that AMS35AA did not show genotoxic activity. Data of tumor volume ex vivo indicate reduction of tumor for the treated animals with AMS35AA up to 15.84×, which is superior to Dacarbazina (50 mg/Kg, p.c.; i.p.). SIGNIFICANCE In summary, the study showed that AMS35AA reveals relevant results regarding to cytotoxicity of B16F10 murine melanoma cells, inducing death by apoptosis via mitochondrial and/or mediated by DNA damages.
Collapse
Affiliation(s)
- Stephanie Dynczuki Navarro
- Graduate Program in Biotechnology and Biodiversity, Pro Midwest Network, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Lucas Roberto Pessatto
- Research Center in Stem Cells, Cell Therapy and Genetic Toxicology (CeTroGen), "Maria Aparecida Pedrossian" University Hospital, Campo Grande, Mato Grosso do Sul, Brazil; Graduate Program in Genetics and Molecular Biology, General Biology Department, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Alisson Meza
- Graduate Program in Chemistry, Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Edwin José Torres de Oliveira
- Research Center in Stem Cells, Cell Therapy and Genetic Toxicology (CeTroGen), "Maria Aparecida Pedrossian" University Hospital, Campo Grande, Mato Grosso do Sul, Brazil; Graduate Program in Genetics and Molecular Biology, General Biology Department, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Sarah Alves Auharek
- Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Teófilo Otoni, Minas Gerais, Brazil
| | - Lizia Colares Vilela
- Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Teófilo Otoni, Minas Gerais, Brazil
| | - Dênis Pires de Lima
- Graduate Program in Chemistry, Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Ricardo Bentes de Azevedo
- Genetics and Morphology Department, Biosciences Institute, Brasilia University, Brasilia, Federal District, Brazil
| | | | - Osmar Ignacio Ayala Cáceres
- Graduate Program in Chemistry, Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Roberto da Silva Gomes
- Graduate Program in Chemistry, Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil; Synthesis and Molecular Modification Laboratory, Faculty of Exact Sciences and Technology, Federal University of Grande Dourados, Dourados, Mato Grosso do Sul, Brazil
| | - Adilson Beatriz
- Graduate Program in Chemistry, Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Rodrigo Juliano Oliveira
- Research Center in Stem Cells, Cell Therapy and Genetic Toxicology (CeTroGen), "Maria Aparecida Pedrossian" University Hospital, Campo Grande, Mato Grosso do Sul, Brazil; Graduate Program in Genetics and Molecular Biology, General Biology Department, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil; Graduate Program in Health and Development of Midwest Region, Faculty of Medicine "Dr Hélio Mandetta", Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil.
| | - Marco Antonio Utrera Martines
- Graduate Program in Biotechnology and Biodiversity, Pro Midwest Network, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil; Graduate Program in Chemistry, Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil.
| |
Collapse
|
13
|
Zhou P, Qin J, Li Y, Li G, Wang Y, Zhang N, Chen P, Li C. Combination therapy of PKCζ and COX-2 inhibitors synergistically suppress melanoma metastasis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:115. [PMID: 28865485 PMCID: PMC5581453 DOI: 10.1186/s13046-017-0585-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/18/2017] [Indexed: 01/22/2023]
Abstract
Background Metastatic malignant melanoma is one of the most aggressive malignancies and its treatment remains challenging. Recent studies demonstrate that the melanoma metastasis has correlations with the heightened activations of protein kinase C ζ (PKCζ) and cyclooxygenase-2 (COX-2) signaling pathways. Targeted inhibitions for PKCζ and COX-2 have been considered as the promising strategies for the treatment of melanoma metastasis. Thus, the PKCζ inhibitor J-4 and COX-2 inhibitor Celecoxib were combined to treat melanoma metastasis in this study. Methods The Transwell assay, Wound-healing assay and Adhesion assay were used to evaluate the inhibition of combined therapy of J-4 and Celecoxib on melanoma cells invasion, migration and adhesion in vitro, respectively. The impaired actin polymerization was observed by confocal microscope and inactivated signal pathways about PKCζ and COX-2 were confirmed by the Western blotting assay. The B16-F10/C57BL mouse melanoma model was used to test the inhibition of combined therapy of J-4 and Celecoxib on melanoma metastasis in vivo. Results The in vitro results showed that the combination of J-4 and Celecoxib exerted synergistic inhibitory effects on the migration, invasion and adhesion of melanoma B16-F10 and A375 cells with combination index less than 1. The actin polymerization and phosphorylation of Cofilin required in cell migration were severely impaired, which is due to the inactivation of PKCζ related signal pathways and the decrease of COX-2. The combined inhibition of PKCζ and COX-2 induced Mesenchymal-Epithelial Transition (MET) in melanoma cells with the expression of E-Cadherin increasing and Vimentin decreasing. The secretion of MMP-2/MMP-9 also significantly decreased after the combination treatment. In C57BL/6 mice intravenously injected with B16-F10 cells (5 × 104 cells/mouse), co-treatment of J-4 and Celecoxib also severely suppressed melanoma lung metastasis. The body weight monitoring and HE staining results indicated the low toxicity of the combination therapy. Conclusions This study demonstrates that the combination therapy of PKCζ and COX-2 inhibitors can significantly inhibit melanoma metastasis in vitro and in vivo, which will be an efficient strategy for treatment of melanoma metastasis in clinics.
Collapse
Affiliation(s)
- Ping Zhou
- Department of Thoracic Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, International Medical School, School of Pharmacy, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, People's Republic of China
| | - Jiaqi Qin
- Department of Thoracic Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, International Medical School, School of Pharmacy, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, People's Republic of China
| | - Yuan Li
- Department of Thoracic Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, International Medical School, School of Pharmacy, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, People's Republic of China
| | - Guoxia Li
- Department of Thoracic Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, International Medical School, School of Pharmacy, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, People's Republic of China
| | - Yinsong Wang
- Department of Thoracic Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, International Medical School, School of Pharmacy, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, People's Republic of China
| | - Ning Zhang
- Department of Thoracic Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, International Medical School, School of Pharmacy, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, People's Republic of China
| | - Peng Chen
- Department of Thoracic Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, International Medical School, School of Pharmacy, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, People's Republic of China.
| | - Chunyu Li
- Department of Thoracic Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, International Medical School, School of Pharmacy, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, People's Republic of China.
| |
Collapse
|