1
|
Bonzano E, Barruscotti S, Chiellino S, Montagna B, Bonzano C, Imarisio I, Colombo S, Guerrini F, Saddi J, La Mattina S, Tomasini CF, Spena G, Pedrazzoli P, Lancia A. Current Treatment Paradigms for Advanced Melanoma with Brain Metastases. Int J Mol Sci 2025; 26:3828. [PMID: 40332507 PMCID: PMC12027546 DOI: 10.3390/ijms26083828] [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: 02/15/2025] [Revised: 04/12/2025] [Accepted: 04/17/2025] [Indexed: 05/08/2025] Open
Abstract
The therapeutic management of melanoma brain metastases has undergone a profound revolution during recent decades. Optimal integration of systemic therapies with local treatments seems to represent the strategy to pursue in order to maximize clinical outcomes, stressing the need for real multidisciplinary care in this setting of patients. However, the current approach in the clinics does not necessarily reflect what the current guidelines state, and several pending issues are present, from the ideal therapeutic sequence between stereotactic radiosurgery (SRS) and drug administration to the current role of surgery and whole brain radiotherapy (WBRT), all of which need to be addressed. This narrative review aims to provide practical help for navigating the current controversies, with an eye towards possible future advancements in the field, which could help to obtain a comprehensive molecular characterization of the tumor and a more personalized patient-centered therapeutic approach.
Collapse
Affiliation(s)
- Elisabetta Bonzano
- Department of Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy (S.L.M.)
| | | | - Silvia Chiellino
- Unit of Oncology, Department of Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy (B.M.)
| | - Benedetta Montagna
- Unit of Oncology, Department of Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy (B.M.)
| | - Chiara Bonzano
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, IRCCS Ospedale Policlinico San Martino, University Eye Clinic, 16132 Genoa, Italy
| | - Ilaria Imarisio
- Unit of Oncology, Department of Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy (B.M.)
| | - Sara Colombo
- Department of Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy (S.L.M.)
| | - Francesco Guerrini
- Unit of Neurosurgery, Department of Head & Neck Surgery, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (F.G.)
| | - Jessica Saddi
- Department of Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy (S.L.M.)
| | - Salvatore La Mattina
- Department of Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy (S.L.M.)
| | | | - Giannantonio Spena
- Unit of Neurosurgery, Department of Head & Neck Surgery, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (F.G.)
| | - Paolo Pedrazzoli
- Unit of Oncology, Department of Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy (B.M.)
| | - Andrea Lancia
- Department of Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy (S.L.M.)
| |
Collapse
|
2
|
Aissa T, Aissaoui-Zid D, Moslah W, Khamessi O, Ksiksi R, Oltermann M, Ruck M, Zid MF, Srairi-Abid N. Synthesis, physicochemical and pharmacological characterizations of a tetra-[methylimidazolium] dihydrogen decavanadate, inhibiting the IGR39 human melanoma cells development. J Inorg Biochem 2024; 260:112672. [PMID: 39079338 DOI: 10.1016/j.jinorgbio.2024.112672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/13/2024] [Accepted: 07/23/2024] [Indexed: 09/03/2024]
Abstract
Melanoma is a skin cancer that arises from melanocytes and can spread quickly to the other organs of the body, if not treated early. Generally, melanoma shows an inherent resistance to conventional therapies. In this regard, new potential drugs are being developed as possible treatments for melanoma. In this paper, we report the synthesis of a new decavanadate compound with organic molecules for a potential therapeutic application. The tetra-[methylimidazolium] dihydrogen decavanadate(V) salt (C4H7N2)4[H2V10O28] is characterized by single-crystal X-ray diffraction, by FT-IR, UV-Vis and 51V NMR spectroscopy, as well as by thermal analysis (TGA and DSC). The compound crystallizes in the monoclinic centrosymmetric space group P21/c. Its formula unit consists of one dihydrogen decavanadate anion [H2V10O28]4- and four organic 4-methylimidazolium cations (C4H7N2)+. Important intermolecular interactions are N-H···O and O-H···O hydrogen bonds and π-π stacking interactions between the organic cations, revealed by analysis of the Hirshfeld surface and its two-dimensional fingerprint plots. Interestingly, this compound inhibits the viability of IGR39 cells with IC50 values of 14.65 μM and 4 μM after 24 h and 72 h of treatment, respectively. The analysis of its effect by flow cytometry using an Annexin V-FITC/IP cell labeling, showed that (C4H7N2)4H2V10O28 compound induced IGR39 cell apoptosis and necrosis. Molecular docking studies performed against TNFR1 and GPR40, as putative targets, suggest that the (C4H7N2)4[H2V10O28] compound may act as inhibitor of these proteins, known to be overexpressed in melanoma cells. Therefore, we could consider it as a new potential metallodrug against melanoma.
Collapse
Affiliation(s)
- Taissir Aissa
- University of Tunis El Manar, Faculty of Sciences of Tunis, Laboratory of Materials, Crystal Chemistry and Applied Thermodynamics (LR15ES01), 2092 El Manar II, Tunis, Tunisia
| | - Dorra Aissaoui-Zid
- University of Tunis El Manar, Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, Tunis, Tunisia.
| | - Wassim Moslah
- University of Tunis El Manar, Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, Tunis, Tunisia
| | - Oussema Khamessi
- University of Tunis El Manar, Laboratory of Bioinformatics, Biomathematics and Biostatistics (BIMS), Pasteur Institute of Tunis, Tunis, Tunisia.; Higher Institute of Biotechnology of Sidi Thabet ISBST, University of Manouba, 2020 Ariana,Tunisia
| | - Regaya Ksiksi
- University of Tunis El Manar, Faculty of Sciences of Tunis, Laboratory of Materials, Crystal Chemistry and Applied Thermodynamics (LR15ES01), 2092 El Manar II, Tunis, Tunisia; The Higher Institute of Preparatory Studies in Biology and Geology (ISEP-BG) of Soukra, Carthage University, 49 Avenue "August 13" Choutrana, II-2036 Soukra, Tunisia
| | - Maike Oltermann
- Department of Chemistry and Food Chemistry, Technische Universität Dresden, 01062 Dresden, Germany
| | - Michael Ruck
- Department of Chemistry and Food Chemistry, Technische Universität Dresden, 01062 Dresden, Germany
| | - Mohamed Faouzi Zid
- University of Tunis El Manar, Faculty of Sciences of Tunis, Laboratory of Materials, Crystal Chemistry and Applied Thermodynamics (LR15ES01), 2092 El Manar II, Tunis, Tunisia
| | - Najet Srairi-Abid
- University of Tunis El Manar, Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, Tunis, Tunisia.
| |
Collapse
|
3
|
Rousset P, Nardin C, Maubec E, Heidelberger V, Picard A, Troin L, Gerard E, Kramkimel N, Steff-Naud M, Quéreux G, Gaudy-Marqueste C, Lesage C, Mignard C, Jeudy G, Jouary T, Saint-Jean M, Baroudjian B, Archier E, Mortier L, Lebbe C, Montaudié H. Real-world outcomes of combined lenvatinib and anti-PD-1 in advanced melanoma: the Lenvamel study, a multicenter retrospective study of the French Group of Skin Cancers (Groupe de Cancérologie Cutanée). Oncologist 2024; 29:e1364-e1372. [PMID: 38956747 PMCID: PMC11449033 DOI: 10.1093/oncolo/oyae145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 05/02/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Currently, treatment options for patients with advanced melanoma who experience failed immunotherapy or targeted therapy are lacking. Recent studies suggest the antitumor activity of combined pembrolizumab and lenvatinib in patients with advanced melanoma progressing on immunotherapy. Herein, we report the clinical outcomes of combined lenvatinib and a programmed cell death protein-1 inhibitor (PD-1) in this population. MATERIALS AND METHODS This French multicenter real-world study was conducted between September 2020 and July 2023. The primary endpoint was the objective response rate (ORR) according to the Response Evaluation Criteria in Solid Tumours (version 1.1). Secondary variables were treatment-related adverse events (TRAEs), progression-free survival (PFS), overall survival (OS), and duration of response (DOR). RESULTS Of the 67 patients included (median age, 69 years; median follow-up, 5.0 months), 85% had stage IV-M1c or M1d disease. The overall ORR was 28.4% (95% CI, 18%-41%), including 3 complete (4.5%) and 16 partial (23.9%) responses. Median DOR was 3.1 (interquartile range, 1.3-4.3) months. Median PFS and OS were 3.1 (95% CI, 2.5-3.7) and 9.8 (95% CI, 5.6-13.9) months, respectively. Grades 3-5 TRAEs occurred in 16 (24%) patients; common TRAEs were fatigue (43.3%), nausea/vomiting (26.8%), diarrhea (20.9%), and hypertension (20.9%). No treatment-related deaths occurred. CONCLUSION Our real-world study demonstrates an interesting response rate and acceptable safety profile in a population with poor prognostic factors. Our data support this treatment option for refractory melanoma, as it is not approved by the Food and Drug Administration or European Medicines Agency, and highlight the need for new strategies.
Collapse
Affiliation(s)
- Perrine Rousset
- Dermatology Department, University Hospital of Nice, Nice, France
| | - Charlée Nardin
- Dermatology Department, University Hospital of Besançon, Université de Franche-Comté, Besançon, France
| | - Eve Maubec
- AP-HP, Dermatology Department, Avicenne Hospital, Bobigny, France
| | | | - Alexandra Picard
- Dermatology Department, University Hospital of Nice, Nice, France
| | - Laura Troin
- Dermatology Department, University Hospital of Nice, Nice, France
| | - Emilie Gerard
- Dermatology Department, University Hospital of Bordeaux, Bordeaux, France
| | - Nora Kramkimel
- AP-HP, Dermatology Department, Cochin Hospital, Paris, France
| | - Maud Steff-Naud
- Dermatology Department, CHI Aulnay-Sous-Bois, Aulnay-Sous-Bois, France
| | - Gaëlle Quéreux
- Dermatology Department, University Hospital of Nantes, Nantes, France
| | | | - Candice Lesage
- Dermatology Department, University Hospital of Montpellier, Montpellier, France
| | - Claire Mignard
- Dermatology Department, University Hospital of Rouen, Rouen, France
| | - Géraldine Jeudy
- Dermatology Department, University Hospital of Dijon, Dijon, France
| | - Thomas Jouary
- Dermatology Department, University Hospital of Pau, Pau, France
| | - Mélanie Saint-Jean
- Oncology Department, Institut de Cancérologie de l’Ouest, Saint-Herblain, France
| | - Barouyr Baroudjian
- AP-HP, Oncodermatology Department, Saint-Louis Hospital, Université de Paris, Paris, France
| | - Elodie Archier
- AP-HM, Dermatology Department, Hôpital Saint-Joseph, Marseille, France
| | | | - Céleste Lebbe
- AP-HP, Oncodermatology Department, Saint-Louis Hospital, Université de Paris, Paris, France
| | - Henri Montaudié
- Dermatology Department, University Hospital of Nice, Nice, France
- INSERM U1065, Centre Méditerranéen de Médecine Moléculaire, Université Côte d’Azur, Nice, France
| |
Collapse
|
4
|
Yang P, Zhang J. Indoleamine 2,3-Dioxygenase (IDO) Activity: A Perspective Biomarker for Laboratory Determination in Tumor Immunotherapy. Biomedicines 2023; 11:1988. [PMID: 37509627 PMCID: PMC10377333 DOI: 10.3390/biomedicines11071988] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) is a heme enzyme involved in catalyzing the conversion of tryptophan (Trp) into kynurenine (Kyn) at the first rate-limiting step in the kynurenine pathway of L-tryptophan metabolism. It has been found to be involved in several biological functions such as aging, immune microorganism, neurodegenerative and infectious diseases, and cancer. IDO1 plays an important role in immune tolerance by depleting tryptophan in the tumor microenvironment and inhibiting the proliferation of effector T cells, which makes it an important emerging biomarker for cancer immunotherapy. Therefore, the research and development of IDO1 inhibitors are of great importance for tumor therapy. Of interest, IDO activity assays are of great value in the screening and evaluation of inhibitors. Herein, we mainly review the biological functions of IDO1, immune regulation, key signaling molecules in the response pathway, and the development of IDO1 inhibitors in clinical trials. Furthermore, this review provides a comprehensive overview and, in particular, a discussion of currently available IDO activity assays for use in the evaluation of IDO inhibitors in human blood. We believe that the IDO activity is a promising biomarker for the immune escape and laboratory evaluation of tumor immunotherapy.
Collapse
Affiliation(s)
- Pengbo Yang
- Department of Laboratory Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Junhua Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| |
Collapse
|
5
|
Ou L, Liu S, Wang H, Guo Y, Guan L, Shen L, Luo R, Elder DE, Huang AC, Karakousis G, Miura J, Mitchell T, Schuchter L, Amaravadi R, Flowers A, Mou H, Yi F, Guo W, Ko J, Chen Q, Tian B, Herlyn M, Xu X. Patient-derived melanoma organoid models facilitate the assessment of immunotherapies. EBioMedicine 2023; 92:104614. [PMID: 37229906 PMCID: PMC10277922 DOI: 10.1016/j.ebiom.2023.104614] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND Only a minority of melanoma patients experience durable responses to immunotherapies due to inter- and intra-tumoral heterogeneity in melanoma. As a result, there is a pressing need for suitable preclinical models to investigate resistance mechanisms and enhance treatment efficacy. METHODS Here, we report two different methods for generating melanoma patient-derived organoids (MPDOs), one is embedded in collagen gel, and the other is inlaid in Matrigel. MPDOs in Matrigel are used for assessing the therapeutic effects of anti-PD-1 antibodies (αPD-1), autochthonous tumor infiltrating lymphocytes (TILs), and small molecule compounds. MPDOs in collagen gel are used for evaluating the chemotaxis and migratory capacity of TILs. FINDING The MPDOs in collagen gel and Matrigel have similar morphology and immune cell composition to their parental melanoma tissues. MPDOs show inter- and intra-tumoral heterogeneity and contain diverse immune cells such as CD4+, CD8+ T, Treg, CD14+ monocytic, CD15+, and CD11b+ myeloid cells. The tumor microenvironment (TME) in MPDOs is highly immunosuppressive, and the lymphoid and myeloid lineages express similar levels of PD-1, PD-L1, and CTLA-4 as their parental melanoma tissues. Anti-PD-1 antibodies (αPD-1) reinvigorate CD8+ T cells and induce melanoma cell death in the MPDOs. TILs expanded by IL-2 and αPD-1 show significantly lower expression of TIM-3, better migratory capacity and infiltration of autochthonous MPDOs, and more effective killing of melanoma cells than TILs expanded by IL-2 alone or IL-2 with αCD3. A small molecule screen discovers that Navitoclax increases the cytotoxicity of TIL therapy. INTERPRETATION MPDOs may be used to test immune checkpoint inhibitors and cellular and targeted therapies. FUNDING This work was supported by the NIH grants CA114046, CA261608, CA258113, and the Tara Miller Melanoma Foundation.
Collapse
Affiliation(s)
- Lingling Ou
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Shujing Liu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Huaishan Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yeye Guo
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lei Guan
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Longbin Shen
- The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Ruhui Luo
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - David E Elder
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Alexander C Huang
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Giorgos Karakousis
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - John Miura
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Tara Mitchell
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lynn Schuchter
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ravi Amaravadi
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ahron Flowers
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Haiwei Mou
- The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Fan Yi
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Wei Guo
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jina Ko
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Qing Chen
- The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Bin Tian
- The Wistar Institute, Philadelphia, PA, 19104, USA
| | | | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
6
|
Rousset P, Dalle S, Mortier L, Dereure O, Dalac S, Dutriaux C, Leccia MT, Legoupil D, Brunet-Possenti F, De Quatrebarbes J, Grob JJ, Saiag P, Maubec E, Stoebner PE, Granel-Brocard F, Arnault JP, Allayous C, Oriano B, Lebbe C, Montaudié H. Impact of systemic therapies in metastatic melanoma of unknown primary: A study from MELBASE, a French multicentric prospective cohort. J Am Acad Dermatol 2023; 88:808-815. [PMID: 36543626 DOI: 10.1016/j.jaad.2022.11.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/01/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Clinical outcomes of advanced melanoma of unknown primary (MUP) in the era of novel therapies have been scarcely studied. OBJECTIVE To investigate the efficacy and safety of systemic treatments in patients with advanced MUP compared to patients with stage-matched melanoma of known cutaneous primary (cMKP). METHODS Based on the nationwide MelBase prospective database, this study included advanced melanoma patients treated from March 2013 to June 2021 with first-line immunotherapies, targeted therapies, or chemotherapy. Co-primary outcomes were progression-free survival and overall survival. Secondary outcome was treatment-related toxicities. Multivariate and propensity score analyses were performed. RESULTS Of 1882 patients, 265 (14.1%) had advanced MUP. Patients with advanced MUP displayed more often unfavorable initial prognostic factors than those with cMKP. Progression-free and overall survival did not differ significantly between the groups (P = .73 and P = .93, respectively), as well as treatment-related toxicity rate and severity, regardless of treatment type. LIMITATIONS No record of standard diagnostic criteria of MUP used in the participating centers. CONCLUSIONS Although patients with MUP had less favorable baseline prognostic factors, they benefited from the novel therapies as much as those with cMKP. They should be managed according to similar strategies.
Collapse
Affiliation(s)
- Perrine Rousset
- Dermatology Department, University Hospital of Nice, Nice, France
| | - Stéphane Dalle
- Hospices Civils De Lyon, Cancer Research Center of Lyon, Université Claude Bernard Lyion 1, Immucare, Pierre-Bénite, France
| | - Laurent Mortier
- Dermatology Department, University of Lille, ONCO-THAI INSERM, U1189, Lille, France
| | - Olivier Dereure
- Dermatology Department, University Hospital of Montpellier, Montpellier, France
| | - Sophie Dalac
- Dermatology Department, University Hospital of Dijon, Dijon, France
| | | | | | | | | | | | - Jean-Jacques Grob
- Dermatology Department, Hopital de la Timone, Aix-Marseille University, Marseille, France
| | - Philippe Saiag
- AP-HP, Dermatology, Ambroise Paré Hospital, EA4340, UVSQ University, Paris-Saclay University, Boulogne-Billancourt, France
| | - Eve Maubec
- AP-HP, Dermatology Department, Hôpital Avicenne, Bobigny, France
| | | | | | | | - Clara Allayous
- Université Paris Cite, Dermato-Oncology AP-HP Hôpital Saint Louis, INSERM U976, Paris, France
| | - Bastien Oriano
- AP-HP, Clinical Epidemiology Center, Hôtel-Dieu, Paris, France
| | - Céleste Lebbe
- Université Paris Cite, Dermato-Oncology AP-HP Hôpital Saint Louis, INSERM U976, Paris, France
| | - Henri Montaudié
- Dermatology Department, University Hospital of Nice, Nice, France; INSERM U1065, Centre Méditerranéen de Médecine Moléculaire, Université Côte d'Azur, Nice, France.
| |
Collapse
|
7
|
Prajapat VM, Mahajan S, Paul PG, Aalhate M, Mehandole A, Madan J, Dua K, Chellappan DK, Singh SK, Singh PK. Nanomedicine: A pragmatic approach for tackling melanoma skin cancer. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
8
|
Alia Moosavian S, Hashemi M, Etemad L, Daneshmand S, Salmasi Z. Melanoma-derived exosomes: Versatile extracellular vesicles for diagnosis, metastasis, immune modulation, and treatment of melanoma. Int Immunopharmacol 2022; 113:109320. [DOI: 10.1016/j.intimp.2022.109320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/24/2022] [Accepted: 10/03/2022] [Indexed: 11/05/2022]
|
9
|
Functional Downregulation of PD-L1 and PD-L2 by CpG and non-CpG Oligonucleotides in Melanoma Cells. Cancers (Basel) 2022; 14:cancers14194698. [PMID: 36230620 PMCID: PMC9562717 DOI: 10.3390/cancers14194698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/07/2022] [Accepted: 09/22/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Although metastatic melanoma is still not a curable disease, targeting of immunologically relevant checkpoints represents a turning point in the treatment. Particularly, targeting the interaction between PD-L1 and its referring receptor PD-1 with antibodies has been shown to activate T-cell function abrogating the evasion of tumor cells from immune recognition. Here, we present another approach that interferes with this system by showing that treatment of melanoma cells with oligonucleotides reduces the expression of PD-L1 (and PD-L2) on tumor cells. Specifically, non-CpG-6-PTO, an ODN that forms superstructures known as G-quartets, has been found to inhibit the interferon-γ-induced signaling cascade which fosters PD-L1 expression. These findings suggest a new therapeutic strategy to interfere with one of the most important immune checkpoints. Abstract The clinical application of immune checkpoint inhibitors represents a breakthrough progress in the treatment of metastasized melanoma and other tumor entities. In the present study, it was hypothesized that oligonucleotides (ODNs), known as modulators of the immune response, have an impact on the endogenous expression of checkpoint molecules, namely PD-L1 and PD-L2 (PD-L1/2). IFNγ-stimulated melanoma cells (A375, SK-Mel-28) were treated with different synthetically manufactured oligonucleotides which differed in sequence, length and backbone composition. It was found that a variety of different ODN sequences significantly suppressed PD-L1/2 expression. This effect was dependent on length and phosphorothioate (PTO) backbone. In particular, a sequence containing solely guanines (nCpG-6-PTO) was highly effective in downregulating PD-L1/2 at the protein, mRNA and promoter levels. Mechanistically, we gave evidence that ODNs with G-quartet-forming motifs suppress the interferon signaling axis (JAK/STAT/IRF1). Our findings identify a subset of ODNs as interesting pharmacological compounds that could expand the arsenal of targeted therapies to combat the immunological escape of tumor cells.
Collapse
|
10
|
Zhang Y, Cui Q, Xu M, Liu D, Yao S, Chen M. Current Advances in PD-1/PD-L1 Blockade in Recurrent Epithelial Ovarian Cancer. Front Immunol 2022; 13:901772. [PMID: 35833132 PMCID: PMC9271774 DOI: 10.3389/fimmu.2022.901772] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/30/2022] [Indexed: 12/24/2022] Open
Abstract
Immunotherapies have revolutionized the treatment of a variety of cancers. Epithelial ovarian cancer is the most lethal gynecologic malignancy, and the rate of advanced tumor progression or recurrence is as high as 80%. Current salvage strategies for patients with recurrent ovarian cancer are rarely curative. Recurrent ovarian cancer is a “cold tumor”, predominantly due to a lack of tumor antigens and an immunosuppressive tumor microenvironment. In trials testing programmed death-1 (PD-1)/programmed death ligand 1 (PD-L1) blockade as a monotherapy, the response rate was only 8.0-22.2%. In this review, we illustrate the status of cold tumors in ovarian cancer and summarize the existing clinical trials investigating PD-1/PD-L1 blockade in recurrent ovarian cancer. Increasing numbers of immunotherapy combination trials have been set up to improve the response rate of EOC. The current preclinical and clinical development of immunotherapy combination therapy to convert an immune cold tumor into a hot tumor and their underlying mechanisms are also reviewed. The combination of anti-PD-1/PD-L1 with other immunomodulatory drugs or therapies, such as chemotherapy, antiangiogenic therapies, poly (ADP-ribose) polymerase inhibitors, adoptive cell therapy, and oncolytic therapy, could be beneficial. Further efforts are merited to transfer these results to a broader clinical application.
Collapse
Affiliation(s)
- Yuedi Zhang
- Department of Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qiulin Cui
- Department of Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Manman Xu
- Department of Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Duo Liu
- Department of Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuzhong Yao
- Department of Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Ming Chen, ; Shuzhong Yao,
| | - Ming Chen
- Department of Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Ming Chen, ; Shuzhong Yao,
| |
Collapse
|
11
|
Synergistic antitumor effect of a penicillin derivative combined with thapsigargin in melanoma cells. J Cancer Res Clin Oncol 2022; 148:3361-3373. [PMID: 35751681 DOI: 10.1007/s00432-022-04129-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/08/2022] [Indexed: 10/17/2022]
Abstract
PURPOSE To investigate the effect of TAP7f, a penicillin derivative previously characterized as a potent antitumor agent that promotes ER stress and apoptosis, in combination with thapsigargin, an ER stress inducer, on melanoma cells. METHODS The synergistic antiproliferative effect of TAP7f in combination with thapsigargin was studied in vitro in murine B16-F0 melanoma cells, and in human A375 and SB2 melanoma cells. In vivo assays were performed with C57BL/6J mice challenged with B16-F0 cells. Immunofluorescence and Western blot assays were carried out to characterize the induction of ER stress and apoptosis. Necrotic tumor areas and the potential toxicity of the combined therapy were examined by histological analysis of tissue sections after hematoxylin-eosin staining. RESULTS In vitro, the combination of TAP7f with thapsigargin synergistically inhibited the proliferation of murine B16-F0, and human A375 and SB2 melanoma cells. When non-inhibitory doses of each drug were simultaneously administered to C57BL/6J mice challenged with B16-F0 cells, a 50% reduction in tumor volumes was obtained in the combined group. An apoptotic response characterized by higher expression levels of Baxenhanced PARP-1 cleavage and the presence of active caspase 3 was observed in tumors from the combined treatment. In addition, higher expression levels of GADD153/CHOP and ATF4 were found in tumors of mice treated with both drugs with respect to each drug used alone, indicating the induction of an ER stress response. No signs of tissue toxicity were observed in histological sections of different organs extracted from mice receiving the combination. CONCLUSION The synergistic and effective antitumor action of TAP7f in combination with thapsigargin could be considered as a potential therapeutic strategy for melanoma treatment.
Collapse
|
12
|
Comito F, Pagani R, Grilli G, Sperandi F, Ardizzoni A, Melotti B. Emerging Novel Therapeutic Approaches for Treatment of Advanced Cutaneous Melanoma. Cancers (Basel) 2022; 14:271. [PMID: 35053435 PMCID: PMC8773625 DOI: 10.3390/cancers14020271] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/29/2021] [Accepted: 01/04/2022] [Indexed: 02/01/2023] Open
Abstract
The prognosis of patients with advanced cutaneous melanoma has radically changed in the past decade. Nevertheless, primary or acquired resistance to systemic treatment occurs in many cases, highlighting the need for novel treatment strategies. This review has the purpose of summarizing the current area of interest for the treatment of metastatic or unresectable advanced cutaneous melanoma, including data from recently completed or ongoing clinical trials. The main fields of investigation include the identification of new immune checkpoint inhibitors (anti-LAG3, GITR agonist and anti-TIGIT), adoptive cell therapy, vaccines, engineered TCR therapy, IL-2 agonists, novel targets for targeted therapy (new MEK or RAF inhibitors, HDAC, IDO, ERK, Axl, ATR and PARP inhibitors), or combination strategies (antiangiogenetic agents plus immune checkpoint inhibitors, intra-tumoral immunotherapy in combination with systemic therapy). In many cases, only preliminary efficacy data from early phase trials are available, which require confirmation in larger patient cohorts. A more in-depth knowledge of the biological effects of the molecules and identifying predictive biomarkers remain crucial for selecting patient populations most likely to benefit from novel emerging treatment strategies.
Collapse
Affiliation(s)
- Francesca Comito
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni, 15-40138 Bologna, Italy; (G.G.); (F.S.); (A.A.); (B.M.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti, 9-40138 Bologna, Italy
| | - Rachele Pagani
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni, 15-40138 Bologna, Italy; (G.G.); (F.S.); (A.A.); (B.M.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti, 9-40138 Bologna, Italy
| | - Giada Grilli
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni, 15-40138 Bologna, Italy; (G.G.); (F.S.); (A.A.); (B.M.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti, 9-40138 Bologna, Italy
| | - Francesca Sperandi
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni, 15-40138 Bologna, Italy; (G.G.); (F.S.); (A.A.); (B.M.)
| | - Andrea Ardizzoni
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni, 15-40138 Bologna, Italy; (G.G.); (F.S.); (A.A.); (B.M.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti, 9-40138 Bologna, Italy
| | - Barbara Melotti
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni, 15-40138 Bologna, Italy; (G.G.); (F.S.); (A.A.); (B.M.)
| |
Collapse
|
13
|
Gómez YM, Gallardo DI, Leão J, Calsavara VF. On a new piecewise regression model with cure rate: Diagnostics and application to medical data. Stat Med 2021; 40:6723-6742. [PMID: 34581460 DOI: 10.1002/sim.9208] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 08/26/2021] [Accepted: 09/11/2021] [Indexed: 12/27/2022]
Abstract
In this article, we discuss an extension of the classical negative binomial cure rate model with piecewise exponential distribution of the time to event for concurrent causes, which enables the modeling of monotonic and non-monotonic hazard functions (ie, the shape of the hazard function is not assumed as in traditional parametric models). This approach produces a flexible cure rate model, depending on the choice of time partition. We discuss local influence on this negative binomial power piecewise exponential model. We report on Monte Carlo simulation studies and application of the model to real melanoma and leukemia datasets.
Collapse
Affiliation(s)
- Yolanda M Gómez
- Facultad de Medicina, Universidad de Atacama, Copiapó, Chile.,Departamento de Matemática, Universidad de Atacama, Copiapó, Chile
| | - Diego I Gallardo
- Departamento de Matemática, Universidad de Atacama, Copiapó, Chile
| | - Jeremias Leão
- Department of Statistics, Federal University of Amazonas, Manaus, Brazil
| | - Vinicius F Calsavara
- Department of Epidemiology and Statistics, A.C. Camargo Cancer Center, São Paulo, Brazil.,Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
14
|
Oxidative Stress-Related Mechanisms in Melanoma and in the Acquired Resistance to Targeted Therapies. Antioxidants (Basel) 2021; 10:antiox10121942. [PMID: 34943045 PMCID: PMC8750393 DOI: 10.3390/antiox10121942] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023] Open
Abstract
Melanoma is a highly aggressive cancer with the poorest prognosis, representing the deadliest form of skin cancer. Activating mutations in BRAF are the most frequent genetic alterations, present in approximately 50% of all melanoma cases. The use of specific inhibitors towards mutant BRAF variants and MEK, a downstream signaling target of BRAF in the MAPK pathway, has significantly improved progression-free and overall survival in advanced melanoma patients carrying BRAF mutations. Nevertheless, despite these improvements, resistance still develops within the first year of therapy in around 50% of patients, which is a significant problem in managing BRAF-mutated advanced melanoma. Understanding these mechanisms is one of the mainstreams of the research on BRAFi/MEKi acquired resistance. Both genetic and epigenetic mechanisms have been described. Moreover, in recent years, oxidative stress has emerged as another major force involved in all the phases of melanoma development, from initiation to progression until the onsets of the metastatic phenotype and chemoresistance, and has thus become a target for therapy. In the present review, we discuss the current knowledge on oxidative stress and its signaling in melanoma, as well as the oxidative stress-related mechanisms in the acquired resistance to targeted therapies.
Collapse
|
15
|
Shi CJ, Xu SM, Han Y, Zhou R, Zhang ZY. Targeting cyclin-dependent kinase 4/6 as a therapeutic approach for mucosal melanoma. Melanoma Res 2021; 31:495-503. [PMID: 34483306 PMCID: PMC8568331 DOI: 10.1097/cmr.0000000000000777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 08/02/2021] [Indexed: 11/26/2022]
Abstract
Mucosal melanoma is a rare but devastating subtype of melanoma which typically has a worse prognosis than other melanoma subtypes. Large-scale next-generation sequencing studies, including our recent research, have also proved that the molecular landscape and potential oncogenic drivers of mucosal melanoma remain distinct from that of cutaneous melanoma. Recently, a number of selective cyclin-dependent kinase 4 (CDK4)/6 inhibitors have been approved for clinical application in breast cancer or entered phase III clinical trial in other solid tumors. Additionally, we have revealed that the dysregulation of cell cycle progression, caused by CDK4 amplification, is a key genetic feature in half of mucosal melanoma and targeting of CDK4 in selected mucosal melanoma patients is a potentially promising direction for precision cancer treatment by using molecular-characterized mucosal melanoma patient-derived-xenograft models. This review summarizes the current literature regarding CDK4/6 dysregulation in mucosal melanoma, preclinical and clinical studies of CDK4/6 inhibitors and potential combinational strategies in treating mucosal melanoma.
Collapse
Affiliation(s)
- Chao-ji Shi
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine
- National Center for Stomatology, National Clinical Research Center for Oral Diseases
| | - Sheng-ming Xu
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine
- National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology
| | - Yong Han
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine
- National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology
| | - Rong Zhou
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine
- National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology
| | - Zhi-yuan Zhang
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine
- National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
16
|
Abstract
Great strides in immunotherapy and targeted therapy have revolutionized the management of previously devastating, advanced melanomas. Although these subfields continue to progress, novel approaches in intratumoral oncolytic therapy, adoptive cell therapy, and vaccine therapies are being developed as adjuncts or alternatives. Cytokines, meanwhile, are seeing a resurgence as a viable option as well. The array of effective agents will, in the next few years, provide options for therapy not only in the adjuvant or unresectable settings but also in the neoadjuvant settings. Perhaps, too, in earlier stage melanomas.
Collapse
Affiliation(s)
- Sameer Massand
- Division of Plastic Surgery, Penn State University, 500 University Drive, H071, Hershey, PA 17033, USA
| | - Rogerio I Neves
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, 10920 N McKinley Dr, MKC-4, Tampa, FL 33612, USA.
| |
Collapse
|
17
|
Gingrich AA, Kirane AR. Novel Targets in Melanoma: Intralesional and Combination Therapy to Manipulate the Immune Response. Surg Oncol Clin N Am 2021; 29:467-483. [PMID: 32482321 DOI: 10.1016/j.soc.2020.02.012] [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] [Indexed: 11/30/2022]
Abstract
Clinical outcomes for metastatic melanoma have been dramatically altered by recent developments in immunotherapy and targeted strategies, but response to these therapies is not uniform, the majority of patients do not respond, and clinical response can be self-limited. Current directions in melanoma treatment aim to leverage a combination of therapies for tumors refractory to monoimmunotherapy, to include tumor-directed strategies, such as intralesional therapy and inhibitors designed for novel targets, which may augment current systemic agents when used in combination. Here, we summarize new classes of agents and emerging multimodal combination strategies that demonstrate significant promise in future melanoma management.
Collapse
Affiliation(s)
- Alicia A Gingrich
- Department of Surgery, University of California Davis, 4501 X Street, Suite 3010, Sacramento, CA 95817, USA
| | - Amanda R Kirane
- Department of Surgery, University of California Davis, 4501 X Street, Suite 3010, Sacramento, CA 95817, USA.
| |
Collapse
|
18
|
Pisano M, Dettori MA, Fabbri D, Delogu G, Palmieri G, Rozzo C. Anticancer Activity of Two Novel Hydroxylated Biphenyl Compounds toward Malignant Melanoma Cells. Int J Mol Sci 2021; 22:5636. [PMID: 34073232 PMCID: PMC8198844 DOI: 10.3390/ijms22115636] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/15/2022] Open
Abstract
Melanoma, the deadliest form of skin cancer, is still one of the most difficult cancers to treat despite recent advances in targeted and immune therapies. About 50% of advanced melanoma do not benefit of such therapies, and novel treatments are requested. Curcumin and its analogs have shown good anticancer properties and are being considered for use in combination with or sequence to recent therapies to improve patient outcomes. Our group previously published the synthesis and anticancer activity characterization of a novel curcumin-related compound against melanoma and neuroblastoma cells (D6). Here, two hydroxylated biphenyl compounds-namely, compounds 11 and 12-were selected among a small collection of previously screened C2-symmetric hydroxylated biphenyls structurally related to D6 and curcumin, showing the best antitumor potentiality against melanoma cells (IC50 values of 1.7 ± 0.5 μM for 11 and 2.0 ± 0.7 μM for 12) and no toxicity of normal fibroblasts up to 32 µM. Their antiproliferative activity was deeply characterized on five melanoma cell lines by performing dose-response and clonal growth inhibition assays, which revealed long-lasting and irreversible effects for both compounds. Apoptosis induction was ascertained by the annexin V and TUNEL assays, whereas Western blotting showed caspase activation and PARP cleavage. A cell cycle analysis, following cell treatments with either compound 11 or 12, highlighted an arrest in the G2/M transition. Taking all this evidence together, 11 and 12 were shown to be good candidates as lead compounds to develop new anticancer drugs against malignant melanoma.
Collapse
Affiliation(s)
- Marina Pisano
- Institute for Genetic and Biomedical Research (IRGB), National Research Council of Italy (CNR), Traversa la Crucca 3, 07100 Sassari, Italy; (M.P.); (G.P.)
| | - Maria Antonietta Dettori
- Institute of Biomolecular Chemistry (ICB), National Research Council of Italy (CNR), Traversa la Crucca 3, 07100 Sassari, Italy; (M.A.D.); (D.F.); (G.D.)
| | - Davide Fabbri
- Institute of Biomolecular Chemistry (ICB), National Research Council of Italy (CNR), Traversa la Crucca 3, 07100 Sassari, Italy; (M.A.D.); (D.F.); (G.D.)
| | - Giovanna Delogu
- Institute of Biomolecular Chemistry (ICB), National Research Council of Italy (CNR), Traversa la Crucca 3, 07100 Sassari, Italy; (M.A.D.); (D.F.); (G.D.)
| | - Giuseppe Palmieri
- Institute for Genetic and Biomedical Research (IRGB), National Research Council of Italy (CNR), Traversa la Crucca 3, 07100 Sassari, Italy; (M.P.); (G.P.)
| | - Carla Rozzo
- Institute for Genetic and Biomedical Research (IRGB), National Research Council of Italy (CNR), Traversa la Crucca 3, 07100 Sassari, Italy; (M.P.); (G.P.)
| |
Collapse
|
19
|
Borgers JSW, Haanen JBAG. Cellular Therapy and Cytokine Treatments for Melanoma. Hematol Oncol Clin North Am 2021; 35:129-144. [PMID: 33759770 DOI: 10.1016/j.hoc.2020.08.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cancer immunotherapy plays an important role in the treatment of patients with advanced stage melanoma. Recombinant cytokines were the first tested and approved treatments; however, due to disappointing response rates and severe toxicities, their use has significantly decreased. More recently, adoptive cell transfer therapies have shown to be a promising new treatment strategy able to induce complete and durable remissions in patients with melanoma progressive on first-line treatment. This review provides an overview of the cellular therapies (tumor-infiltrating lymphocytes, T-cell receptor T cells, chimeric antigen receptor T cells) and cytokine treatments (interleukin-2 [IL-2], IL-15, IL-7, IL-10, IL-21, interferon alpha, granulocyte-macrophage colony-stimulating factor) for melanoma.
Collapse
Affiliation(s)
- Jessica S W Borgers
- Department of Medical Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - John B A G Haanen
- Department of Medical Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands.
| |
Collapse
|
20
|
Yang D, Chen M, Sun Y, Jin Y, Lu C, Pan X, Quan G, Wu C. Microneedle-mediated transdermal drug delivery for treating diverse skin diseases. Acta Biomater 2021; 121:119-133. [PMID: 33285323 DOI: 10.1016/j.actbio.2020.12.004] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 12/18/2022]
Abstract
Transdermal drug delivery is an attractive route for dermatological disease therapy because it can directly target the lesion site on the skin, reduce adverse reactions associated with systemic administration, and improve patient compliance. However, the stratum corneum, as the main skin barrier, severely limits transdermal drug penetration, with compromised bioavailability. Microneedles (MNs), which are leveraged to markedly improve the penetration of therapeutic agents by piercing the stratum corneum and creating hundreds of reversible microchannels in a minimally invasive manner, have been envisioned as a milestone for effective transdermal drug delivery, especially for superficial disease therapy. Here, the emergence of versatile MNs for the transdermal delivery of various drugs is reviewed, particularly focusing on the application of MNs for the treatment of diverse skin diseases, including superficial tumors, scars, psoriasis, herpes, acne, and alopecia. Additionally, the promises and challenges of the widespread translation of MN-mediated transdermal drug delivery in the dermatology field are summarized.
Collapse
|
21
|
Haugh AM, Salama AKS, Johnson DB. Advanced Melanoma: Resistance Mechanisms to Current Therapies. Hematol Oncol Clin North Am 2021; 35:111-128. [PMID: 33759769 PMCID: PMC7991196 DOI: 10.1016/j.hoc.2020.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Novel therapeutic agents introduced over the past decade, including immune checkpoint inhibitors and targeted therapies, have revolutionized the management of metastatic melanoma and significantly improved patient outcomes. Although robust and durable responses have been noted in some cases, treatment is often limited by innate or acquired resistance to these agents. This article provides an overview of known and suspected mechanisms involved with acquired resistance to BRAF/MEK inhibitors as well as developing insights into innate and acquired resistance to checkpoint inhibitors in patients with melanoma.
Collapse
Affiliation(s)
- Alexandra M Haugh
- Department of Medicine, Vanderbilt University Medical Center, 719 Thompson Lane, Suite 20400, Nashville, TN 37204, USA
| | - April K S Salama
- Department of Medicine, Duke University Medical Center, 20 Duke Medicine Cir, Durham, NC 27710, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Vanderbilt Ingram Cancer Center, 777 PRB, 2220 Pierce Avenue, Nashville, TN 37232, USA.
| |
Collapse
|
22
|
Pilla L, Alberti A, Di Mauro P, Gemelli M, Cogliati V, Cazzaniga ME, Bidoli P, Maccalli C. Molecular and Immune Biomarkers for Cutaneous Melanoma: Current Status and Future Prospects. Cancers (Basel) 2020; 12:E3456. [PMID: 33233603 PMCID: PMC7699774 DOI: 10.3390/cancers12113456] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 12/18/2022] Open
Abstract
Advances in the genomic, molecular and immunological make-up of melanoma allowed the development of novel targeted therapy and of immunotherapy, leading to changes in the paradigm of therapeutic interventions and improvement of patients' overall survival. Nevertheless, the mechanisms regulating either the responsiveness or the resistance of melanoma patients to therapies are still mostly unknown. The development of either the combinations or of the sequential treatment of different agents has been investigated but without a strongly molecularly motivated rationale. The need for robust biomarkers to predict patients' responsiveness to defined therapies and for their stratification is still unmet. Progress in immunological assays and genomic techniques as long as improvement in designing and performing studies monitoring the expression of these markers along with the evolution of the disease allowed to identify candidate biomarkers. However, none of them achieved a definitive role in predicting patients' clinical outcomes. Along this line, the cross-talk of melanoma cells with tumor microenvironment plays an important role in the evolution of the disease and needs to be considered in light of the role of predictive biomarkers. The overview of the relationship between the molecular basis of melanoma and targeted therapies is provided in this review, highlighting the benefit for clinical responses and the limitations. Moreover, the role of different candidate biomarkers is described together with the technical approaches for their identification. The provided evidence shows that progress has been achieved in understanding the molecular basis of melanoma and in designing advanced therapeutic strategies. Nevertheless, the molecular determinants of melanoma and their role as biomarkers predicting patients' responsiveness to therapies warrant further investigation with the vision of developing more effective precision medicine.
Collapse
Affiliation(s)
- Lorenzo Pilla
- Division of Medical Oncology, San Gerardo Hospital, University of Milano-Bicocca School of Medicine, 20900 Monza, Italy; (P.D.M.); (M.G.); (V.C.); (M.E.C.); (P.B.)
| | - Andrea Alberti
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Health Science and Public Health, University of Brescia, ASST Ospedali Civili, 25123 Brescia, Italy;
| | - Pierluigi Di Mauro
- Division of Medical Oncology, San Gerardo Hospital, University of Milano-Bicocca School of Medicine, 20900 Monza, Italy; (P.D.M.); (M.G.); (V.C.); (M.E.C.); (P.B.)
| | - Maria Gemelli
- Division of Medical Oncology, San Gerardo Hospital, University of Milano-Bicocca School of Medicine, 20900 Monza, Italy; (P.D.M.); (M.G.); (V.C.); (M.E.C.); (P.B.)
| | - Viola Cogliati
- Division of Medical Oncology, San Gerardo Hospital, University of Milano-Bicocca School of Medicine, 20900 Monza, Italy; (P.D.M.); (M.G.); (V.C.); (M.E.C.); (P.B.)
| | - Marina Elena Cazzaniga
- Division of Medical Oncology, San Gerardo Hospital, University of Milano-Bicocca School of Medicine, 20900 Monza, Italy; (P.D.M.); (M.G.); (V.C.); (M.E.C.); (P.B.)
| | - Paolo Bidoli
- Division of Medical Oncology, San Gerardo Hospital, University of Milano-Bicocca School of Medicine, 20900 Monza, Italy; (P.D.M.); (M.G.); (V.C.); (M.E.C.); (P.B.)
| | - Cristina Maccalli
- Laboratory of Immune and Biological Therapy, Research Department, Sidra Medicine, Doha 26999, Qatar;
| |
Collapse
|
23
|
Smith HG, Bagwan I, Board RE, Capper S, Coupland SE, Glen J, Lalondrelle S, Mayberry A, Muneer A, Nugent K, Pathiraja P, Payne M, Peach H, Smith J, Westwell S, Wilson E, Rodwell S, Gore M, Turnbull N, Smith MJF. Ano-uro-genital mucosal melanoma UK national guidelines. Eur J Cancer 2020; 135:22-30. [PMID: 32531566 DOI: 10.1016/j.ejca.2020.04.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/22/2020] [Accepted: 04/08/2020] [Indexed: 02/03/2023]
Abstract
Ano-uro-genital (AUG) mucosal melanomas are rare cancers associated with poor outcomes and limited evidence-based management. The United Kingdom AUG mucosal melanoma guideline development group used an evidence-based systematic approach to make recommendations regarding the diagnosis, treatment and surveillance of patients diagnosed with AUG mucosal melanomas. The guidelines were sent for international peer review, and are accredited by The National Institute for Health and Clinical Excellence (NICE). A summary of the key recommendations is presented. The full documents are available on the Melanoma Focus website.
Collapse
Affiliation(s)
| | - Izhar Bagwan
- Royal Surrey County Hospital NHS Foundation Trust, UK
| | - Ruth E Board
- Lancashire Teaching Hospitals NHS Foundation Trust, UK
| | | | | | | | | | | | - Asif Muneer
- NIHR Biomedical Research Centre, University College London Hospitals NHS Foundation Trust and Division of Surgery and Interventional Science University College London, UK
| | - Karen Nugent
- University Hospital Southampton NHS Foundation Trust, UK
| | | | - Miranda Payne
- Oxford University Hospitals NHS Foundation Trust, UK
| | - Howard Peach
- Leeds Teaching Hospitals NHS Foundation Trust, UK
| | | | - Sarah Westwell
- Brighton and Sussex University Hospitals NHS Foundation Trust, UK
| | | | | | | | | | | |
Collapse
|
24
|
Barboza T, Gomes T, da Costa Medeiros P, Ramos IP, Francischetti I, Monteiro RQ, Gutfilen B, de Souza SAL. Development of 131I-ixolaris as a theranostic agent: metastatic melanoma preclinical studies. Clin Exp Metastasis 2020; 37:489-497. [PMID: 32394234 DOI: 10.1007/s10585-020-10036-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 05/04/2020] [Indexed: 12/13/2022]
Abstract
Tissue factor (TF), a blood coagulation protein, plays an important role in tumor growth, invasion, and metastasis. Ixolaris, a tick-derived non-immunogenic molecule that binds to TF, has demonstrated in vivo inhibitory effect on murine models of melanoma, including primary growth and metastasis. This work aimed to: I) develop an efficient and stable labeling technique of ixolaris with Iodine-131(131I); II) compare the biodistribution of 131I and 131I-ixolaris in tumor-free and melanoma-bearing mice; III) evaluate whether 131I-ixolaris could serve as an antimetastatic agent. Ixolaris radioiodination was performed using iodogen, followed by liquid paper chromatography. Labeling stability and anticoagulant activity were measured. Imaging studies were performed after intravenous administration of free 131I or 131I-ixolaris in a murine melanoma model employing the B16-F10 cell line. Animals were divided in three experimental groups: the first experimental group, D0, received a single-dose of 9.25 MBq of 131I-ixolaris at the same day the animals were inoculated with melanoma cells. In the second group, D15, a single-dose of 9.25 MBq of 131I-ixolaris or free 131I was applied into mice on the fifteenth day after the tumor induction. The third group, D1-D15, received two therapeutic doses of 9.25 MBq of 131I-ixolaris or 131I. In vitro studies demonstrated that 131I-ixolaris is stable for up to 24 h and retains its inhibitory activity on blood coagulation. Biodistribution analysis and metastasis assays showed that all treatment regimens with 131I-ixolaris were effective, being the double-treatment (D1/D15) the most effective one. Remarkably, treatment with free 131I showed no anti-metastatic effect. 131I-ixolaris is a promising theranostic agent for metastatic melanoma.
Collapse
Affiliation(s)
- Thiago Barboza
- Departamento de Radiologia, Faculdade de Medicina, Laboratório de Marcação de Células E Moléculas, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Tainá Gomes
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Priscylla da Costa Medeiros
- Departamento de Radiologia, Faculdade de Medicina, Laboratório de Marcação de Células E Moléculas, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Isalira Peroba Ramos
- Centro Nacional de Bioimagem E Biologia Estrutural, Bloco M, Unidade 2, Centro de Ciências da Saúde, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Ivo Francischetti
- Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Bethesda, USA
| | - Robson Q Monteiro
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Bianca Gutfilen
- Departamento de Radiologia, Faculdade de Medicina, Laboratório de Marcação de Células E Moléculas, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Sergio Augusto Lopes de Souza
- Departamento de Radiologia, Faculdade de Medicina, Laboratório de Marcação de Células E Moléculas, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brasil. .,Centro Nacional de Bioimagem E Biologia Estrutural, Bloco M, Unidade 2, Centro de Ciências da Saúde, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brasil.
| |
Collapse
|
25
|
Louie KS, Banks V, Scholz F, Richter H, Öhrling K, Mohr P, Haferkamp S. Real-world use of talimogene laherparepvec in Germany: a retrospective observational study using a prescription database. Future Oncol 2020; 16:317-328. [PMID: 32050787 DOI: 10.2217/fon-2019-0838] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 01/24/2020] [Indexed: 12/23/2022] Open
Abstract
Aim: There is a growing body of data on real-world use of talimogene laherparepvec (T-VEC). We aimed to characterize real-world T-VEC use using a nationally representative German prescription database covering 60% of prescriptions reimbursed. Patients & methods: A retrospective analysis was conducted using the German IMS® LRx prescription database, analyzing patients aged ≥18 years with an initial T-VEC prescription at 106 plaque-forming units (PFU)/ml and ≥1 subsequent prescription at 108 PFU/ml. Median time on T-VEC treatment, patient characteristics and patterns of T-VEC use were described. Results: Of 127 patients prescribed T-VEC, 72 patients (57%) met study criteria. About two-thirds of these patients initiated T-VEC in 2017. Median age at T-VEC initiation was 74 years (range: 44 to 91). Most prescriptions (88%) were dispensed from hospitals. At study end, 26 (36%) patients remained on T-VEC; 46 (64%) had ended treatment. Median duration of T-VEC treatment for all patients was 18.7 weeks (95% CI: 15.3-26.9) and was longer among those who initiated treatment in 2017 versus 2016 (26.7 vs 15.6 weeks, respectively). Median volume administered for the first 106 PFU/ml and second 108 PFU/ml was 4 ml; the volume decreased for subsequent administrations (2 ml by the eighth administration and 1 ml by the 16th administration). Conclusion: This real-world prescription database study showed that patients who initiated treatment in 2017 had a treatment duration in clinical practice that corresponded with the European Summary of Product Characteristics guideline of continuing T-VEC for ≥6 months. Additional long-term data linking drug use with clinical outcomes are needed.
Collapse
Affiliation(s)
- Karly S Louie
- Amgen Ltd, 1 Uxbridge Business Park, Uxbridge, UB8 1DH, UK
| | - Victoria Banks
- VLB Contractors Ltd, 17 Marling Way, Gravesend, DA12 4DW, UK
| | - Florian Scholz
- IQVIA, Real World Insights, Unterschweinstiege 2-14, 60549, Frankfurt, Germany
| | - Hartmut Richter
- IQVIA, Real World Insights, Unterschweinstiege 2-14, 60549, Frankfurt, Germany
| | | | - Peter Mohr
- Elbe-Klinikum, Am Krankenhaus 1, 21614, Buxtehude, Germany
| | - Sebastian Haferkamp
- University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| |
Collapse
|
26
|
Guo Y, Yang L, Lei S, Tan W, Long J. NEDD4 Negatively Regulates GITR via Ubiquitination in Immune Microenvironment of Melanoma. Onco Targets Ther 2019; 12:10629-10637. [PMID: 31824170 PMCID: PMC6900405 DOI: 10.2147/ott.s212317] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 11/06/2019] [Indexed: 11/23/2022] Open
Abstract
Introduction Melanoma is a common skin cancer that is usually associated with poor clinical outcomes. Recently, the immune checkpoint GITR has been identified as a promising target for immunotherapy of melanoma. In this study, we aimed to investigate the post-translational regulation mechanism of GITR in melanoma. Methods Western blotting was used to evaluate the protein expression of NEDD4, GITR and Foxp3. Real-time PCR (RT-PCR) was performed to determine expression levels of NEDD4, GITR, Foxp3 and IL-2. Cell viability was detected by MTT assay. The ubiquitination of GITR was evaluated by immunoprecipitation. NEDD4 expression data and melanoma survival data were obtained from The Cancer Genome Atlas (TCGA) and cBioPortal databases. Results We demonstrate that E3 ligase NEDD4 binds to GITR and mediates ubiquitination and degradation of GITR. Overexpression of NEDD4 inhibits anti-tumor immunity mediated by T cells against melanoma cells. We also found that the expression of NEDD4 is increased in metastatic melanoma. High NEDD4 expression level is correlated with the poor prognosis of melanoma patients. Discussion In summary, our findings demonstrated that E3 ligase NEDD4 mediates ubiquitination and degradation of GITR and suppresses T-cell-mediated-killings on melanoma cells. Our work highlighted the E3 ligase NEDD4 as a novel prognosis biomarker and therapeutic target for melanoma.
Collapse
Affiliation(s)
- Yu Guo
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
| | - Lichang Yang
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
| | - Shaorong Lei
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
| | - Wuyuan Tan
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
| | - Jianhong Long
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
| |
Collapse
|
27
|
Calsavara VF, Milani EA, Bertolli E, Tomazella V. Long-term frailty modeling using a non-proportional hazards model: Application with a melanoma dataset. Stat Methods Med Res 2019; 29:2100-2118. [DOI: 10.1177/0962280219883905] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The semiparametric Cox regression model is often fitted in the modeling of survival data. One of its main advantages is the ease of interpretation, as long as the hazards rates for two individuals do not vary over time. In practice the proportionality assumption of the hazards may not be true in some situations. In addition, in several survival data is common a proportion of units not susceptible to the event of interest, even if, accompanied by a sufficiently large time, which is so-called immune, “cured,” or not susceptible to the event of interest. In this context, several cure rate models are available to deal with in the long term. Here, we consider the generalized time-dependent logistic (GTDL) model with a power variance function (PVF) frailty term introduced in the hazard function to control for unobservable heterogeneity in patient populations. It allows for non-proportional hazards, as well as survival data with long-term survivors. Parameter estimation was performed using the maximum likelihood method, and Monte Carlo simulation was conducted to evaluate the performance of the models. Its practice relevance is illustrated in a real medical dataset from a population-based study of incident cases of melanoma diagnosed in the state of São Paulo, Brazil.
Collapse
Affiliation(s)
- Vinicius F Calsavara
- Department of Epidemiology and Statistics, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Eder A Milani
- Institute of Mathematics and Statistics, Federal University of Goiás, Goiânia, Brazil
| | - Eduardo Bertolli
- Skin Cancer Department, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Vera Tomazella
- Department of Statistics, Federal University of São Carlos, São Carlos, Brazil
| |
Collapse
|
28
|
Pisano M, Arru C, Serra M, Galleri G, Sanna D, Garribba E, Palmieri G, Rozzo C. Antiproliferative activity of vanadium compounds: effects on the major malignant melanoma molecular pathways. Metallomics 2019; 11:1687-1699. [PMID: 31490510 DOI: 10.1039/c9mt00174c] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Malignant melanoma (MM) is the most fatal skin cancer, whose incidence has critically increased in the last decades. Recent molecular therapies are giving excellent results in the remission of melanoma but often they induce drug resistance in patients limiting their therapeutic efficacy. The search for new compounds able to overcome drug resistance is therefore essential. Vanadium has recently been cited for its anticancer properties against several tumors, but only a few data regard its effect against MM. In a previous work we demonstrated the anticancer activity of four different vanadium species towards MM cell lines. The inorganic anion vanadate(v) (VN) and the oxidovanadium(iv) complex [VO(dhp)2] (VS2), where dhp is 1,2-dimethyl-3-hydroxy-4(1H)-pyridinonate, showed IC50 values of 4.7 and 2.6 μM, respectively, against the A375 MM cell line, causing apoptosis and cell cycle arrest. Here we demonstrate the involvement of Reactive Oxygen Species (ROS) production in the pro-apoptotic effect of these two V species and evaluate the activation of different cell cycle regulators, to investigate the molecular mechanisms involved in their antitumor activity. We establish that VN and VS2 treatments reduce the phosphorylation of extracellular-signal regulated kinase (ERK) by about 80%, causing the deactivation of the mitogen activated protein kinase (MAPK) pathway in A375 cells. VN and VS2 also induce dephosphorylation of the retinoblastoma protein (Rb) (VN 100% and VS2 90%), together with a pronounced increase of cyclin-dependent kinase inhibitor 1 p21 (p21Cip1) protein expression up to 1800%. Taken together, our results confirm the antitumor properties of vanadium against melanoma cells, highlighting its ability to induce apoptosis through generation of ROS and cell cycle arrest by counteracting MAPK pathway activation and strongly inducing p21Cip1 expression and Rb hypo-phosphorylation.
Collapse
Affiliation(s)
- Marina Pisano
- Istituto di Chimica Biomolecolare (ICB), Consiglio Nazionale delle Ricerche (CNR), Traversa La Crucca 3, 07100 Sassari, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Oliveira Pinho J, Matias M, Gaspar MM. Emergent Nanotechnological Strategies for Systemic Chemotherapy against Melanoma. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E1455. [PMID: 31614947 PMCID: PMC6836019 DOI: 10.3390/nano9101455] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/04/2019] [Accepted: 10/10/2019] [Indexed: 12/24/2022]
Abstract
Melanoma is an aggressive form of skin cancer, being one of the deadliest cancers in the world. The current treatment options involve surgery, radiotherapy, targeted therapy, immunotherapy and the use of chemotherapeutic agents. Although the last approach is the most used, the high toxicity and the lack of efficacy in advanced stages of the disease have demanded the search for novel bioactive molecules and/or efficient drug delivery systems. The current review aims to discuss the most recent advances on the elucidation of potential targets for melanoma treatment, such as aquaporin-3 and tyrosinase. In addition, the role of nanotechnology as a valuable strategy to effectively deliver selective drugs is emphasized, either incorporating/encapsulating synthetic molecules or natural-derived compounds in lipid-based nanosystems such as liposomes. Nanoformulated compounds have been explored for their improved anticancer activity against melanoma and promising results have been obtained. Indeed, they displayed improved physicochemical properties and higher accumulation in tumoral tissues, which potentiated the efficacy of the compounds in pre-clinical experiments. Overall, these experiments opened new doors for the discovery and development of more effective drug formulations for melanoma treatment.
Collapse
Affiliation(s)
- Jacinta Oliveira Pinho
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Mariana Matias
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Maria Manuela Gaspar
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| |
Collapse
|
30
|
Incorvaia L, Badalamenti G, Rinaldi G, Iovanna JL, Olive D, Swayden M, Terruso L, Vincenzi B, Fulfaro F, Bazan V, Russo A, Fanale D. Can the plasma PD-1 levels predict the presence and efficiency of tumor-infiltrating lymphocytes in patients with metastatic melanoma? Ther Adv Med Oncol 2019; 11:1758835919848872. [PMID: 31205506 PMCID: PMC6535916 DOI: 10.1177/1758835919848872] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/13/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The immune response in melanoma patients is locally affected by presence of tumor-infiltrating lymphocytes (TILs), generally divided into brisk, nonbrisk, and absent. Several studies have shown that a greater presence of TILs, especially brisk, in primary melanoma is associated with a better prognosis and higher survival rate. PATIENTS AND METHODS We investigated by enzyme-linked immunosorbent assay (ELISA) the correlation between PD-1 levels in plasma and the presence/absence of TILs in 28 patients with metastatic melanoma. RESULTS Low plasma PD-1 levels were correlated with brisk TILs in primary melanoma, whereas intermediate values correlated with the nonbrisk TILs, and high PD-1 levels with absent TILs. Although the low number of samples did not allow us to obtain a statistically significant correlation between the plasma PD-1 levels and the patients' overall survival depending on the absence/presence of TILs, the median survival of patients having brisk type TILs was 5 months higher than that of patients with absent and nonbrisk TILs. CONCLUSIONS This work highlights the ability of measuring the plasma PD-1 levels in order to predict the prognosis of patients with untreated metastatic melanoma without a BRAF mutation at the time of diagnosis.
Collapse
Affiliation(s)
- Lorena Incorvaia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Giuseppe Badalamenti
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Gaetana Rinaldi
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Juan Lucio Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Daniel Olive
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Mirna Swayden
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Lidia Terruso
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Bruno Vincenzi
- Medical Oncology Department, University Campus Bio-Medico, Rome, Italy
| | - Fabio Fulfaro
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Viviana Bazan
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Via del Vespro 129, 90127 Palermo, Italy
| | - Daniele Fanale
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| |
Collapse
|
31
|
Porcu M, De Silva P, Solinas C, Battaglia A, Schena M, Scartozzi M, Bron D, Suri JS, Willard-Gallo K, Sangiolo D, Saba L. Immunotherapy Associated Pulmonary Toxicity: Biology Behind Clinical and Radiological Features. Cancers (Basel) 2019; 11:cancers11030305. [PMID: 30841554 PMCID: PMC6468855 DOI: 10.3390/cancers11030305] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/17/2019] [Accepted: 02/26/2019] [Indexed: 12/22/2022] Open
Abstract
The broader use of immune checkpoint blockade in clinical routine challenges clinicians in the diagnosis and management of side effects which are caused by inflammation generated by the activation of the immune response. Nearly all organs can be affected by immune-related toxicities. However, the most frequently reported are: fatigue, rash, pruritus, diarrhea, nausea/vomiting, arthralgia, decreased appetite and abdominal pain. Although these adverse events are usually mild, reversible and not frequent, an early diagnosis is crucial. Immune-related pulmonary toxicity was most frequently observed in trials of lung cancer and of melanoma patients treated with the combination of the anti-cytotoxic T lymphocyte antigen (CTLA)-4 and the anti-programmed cell death-1 (PD-1) antibodies. The most frequent immune-related adverse event in the lung is represented by pneumonitis due to the development of infiltrates in the interstitium and in the alveoli. Clinical symptoms and radiological patterns are the key elements to be considered for an early diagnosis, rendering the differential diagnosis crucial. Diagnosis of immune-related pneumonitis may imply the temporary or definitive suspension of immunotherapy, along with the start of immuno-suppressive treatments. The aim of this work is to summarize the biological bases, clinical and radiological findings of lung toxicity under immune checkpoint blockade, underlining the importance of multidisciplinary teams for an optimal early diagnosis of this side effect, with the aim to reach an improved patient care.
Collapse
Affiliation(s)
- Michele Porcu
- Department of Radiology, University Hospital of Cagliari, 09042 Monserrato (Cagliari), Italy.
| | - Pushpamali De Silva
- Molecular Immunology Unit, Institut Jules Bordet, Universitè Libre de Bruxelles (ULB), 1000 Brussels, Belgium.
- Clinical and Experimental Hematology, Institute Jules Bordet, Universitè Libre de Bruxelles (ULB), 1000 Brussels, Belgium.
| | - Cinzia Solinas
- Molecular Immunology Unit, Institut Jules Bordet, Universitè Libre de Bruxelles (ULB), 1000 Brussels, Belgium.
- Department of Medical Oncology and Hematology, Regional Hospital of Aosta, 11100 Aosta, Italy.
| | - Angelo Battaglia
- Department of Medical Oncology and Hematology, Regional Hospital of Aosta, 11100 Aosta, Italy.
| | - Marina Schena
- Department of Medical Oncology and Hematology, Regional Hospital of Aosta, 11100 Aosta, Italy.
| | - Mario Scartozzi
- Department of Medical Oncology, University Hospital of Cagliari, 09042 Monserrato (Cagliari), Italy.
| | - Dominique Bron
- Clinical and Experimental Hematology, Institute Jules Bordet, Universitè Libre de Bruxelles (ULB), 1000 Brussels, Belgium.
| | - Jasjit S Suri
- Lung Diagnostic Division, Global Biomedical Technologies, Inc., Roseville, CA 95661, USA.
- AtheroPoint™ LLC, Roseville, CA 95661, USA.
| | - Karen Willard-Gallo
- Molecular Immunology Unit, Institut Jules Bordet, Universitè Libre de Bruxelles (ULB), 1000 Brussels, Belgium.
| | - Dario Sangiolo
- Department of Oncology, University of Torino, 10043 Orbassano (Torino), Italy.
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute FPO-IRCCS, 10060 Candiolo (Torino), Italy.
| | - Luca Saba
- Department of Radiology, University Hospital of Cagliari, 09042 Monserrato (Cagliari), Italy.
| |
Collapse
|