1
|
Bladen JC, Malhotra R, Litwin A. Long-term outcomes of margin-controlled excision for eyelid melanoma. Eye (Lond) 2023; 37:1009-1013. [PMID: 36828958 PMCID: PMC10049999 DOI: 10.1038/s41433-023-02428-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 01/13/2023] [Accepted: 01/25/2023] [Indexed: 02/26/2023] Open
Abstract
OBJECTIVES To provide evidence for long-term outcomes for margin-controlled excision of eyelid melanoma. METHODS Retrospective single-centre observational case series of patients treated for eyelid melanoma between 2007 and 2016, with a minimum of 5-year follow-up. Tumour excision involved rush-paraffin en face horizontal sections and delayed repair (Slow Mohs; SM). RESULTS Twenty-two cases were seen with a survival of 91% (two deaths from nodular and lentigo maligna melanoma) and seven with melanoma in situ (MIS). Invasive melanoma includes eight lentigo maligna melanoma, four nodular, two amelanotic and one desmoplastic. Mean Breslow thickness was 6 mm for invasive (range 0.5-26). Mean excision margin for MIS was 3 mm (range 2-5 mm) and for invasive was 5 mm (range 2-10). Further excisions were performed in nine (41%); two went on to recur. Local recurrence was 36%; six invasive (27%) at a mean of 24 months (range 1.5-5 years) and two for MIS at a mean of 15 months (range 1-1.5 years). Imaging occurred for suspected advanced disease. Sentinel node biopsy was not performed. Advanced melanoma therapy was performed in two cases. No vitamin D testing occurred. CONCLUSIONS Survival rates are in line with 90% overall survival in the UK. Prescriptive excision margins are not applicable in the periocular region and margin-controlled excision with a delayed repair is recommended, but patients need to know further excision may be needed to obtain clearance. Evidence recommending vitamin D therapy needs to be put into clinical practice. In addition, upstaging of MIS occurred advocating excision rather than observation of MIS. More studies are needed to determine the best management of eyelid melanoma.
Collapse
Affiliation(s)
- John C Bladen
- Corneoplastic department, Queen Victoria Hospital, East Grinstead, UK
| | - Raman Malhotra
- Corneoplastic department, Queen Victoria Hospital, East Grinstead, UK
| | - Andre Litwin
- Corneoplastic department, Queen Victoria Hospital, East Grinstead, UK.
| |
Collapse
|
2
|
Skovlund PC, Vind Thaysen H, Schmidt H, Alsner J, Hjollund NH, Lomborg K, Nielsen BK. Effect of patient-reported outcomes as a dialogue-based tool in cancer consultations on patient self-management and health-related quality of life: a clinical, controlled trial. Acta Oncol 2021; 60:1668-1677. [PMID: 34403293 DOI: 10.1080/0284186x.2021.1962972] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 07/27/2021] [Indexed: 11/12/2022]
Abstract
BACKGROUND With increased survival among patients with metastatic melanoma and limited time with health care providers, patients are expected to assume a more active role in managing their treatment and care. Activated patients have the knowledge, skills, and confidence to make effective solutions to self-manage health. The use of patient-reported outcomes (PRO) could have the potential to enhance patient activation. However, PRO-based interventions that facilitate an activation in patients with metastatic melanoma are lacking and warranted. MATERIAL AND METHODS In this prospective non-randomized controlled, clinical trial, patients with metastatic melanoma were assigned to either the intervention (systematic feedback and discussion of PRO during consultation) given at one hospital or the control group (treatment as usual) if they received treatment from two other hospitals in Denmark. The primary outcome was the patient activation measure (PAM), which reflects self-management. Secondary outcomes were health-related quality of life (HRQoL), self-efficacy, and Patient-Physician interaction. Outcomes were measured at baseline, and after 3, 6, and 12 months. The analysis of the effect from baseline to 12 months employed mixed-effects modeling. RESULTS Between 2017 and 2019, patients were allocated to either the intervention group (n = 137) or the control group (n = 142). We found no significant difference in the course of patient activation between the two groups over time. The course of HRQoL was statistically significantly improved by the intervention compared to the control group. Especially, females in the intervention group performed better than males. The other secondary outcomes were not improved by the intervention. CONCLUSION The intervention did not improve knowledge, skills, and confidence for self-management for patients with metastatic melanoma. Neither did it improve coping self-efficacy nor perceived efficacy in Patient-Physician interaction. However, the results suggest that the intervention can have a significant impact on HRQoL and in particular social and emotional well-being among the females.
Collapse
Affiliation(s)
- Pernille Christiansen Skovlund
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital, Aarhus N, Denmark
- Department of Oncology, Aarhus University Hospital, Aarhus N, Denmark
- The Research Centre for Patient Involvement, Aarhus University & the Central Denmark Region, Aarhus N, Denmark
| | - Henriette Vind Thaysen
- The Research Centre for Patient Involvement, Aarhus University & the Central Denmark Region, Aarhus N, Denmark
- Department of Surgery, Aarhus University Hospital, Aarhus N, Denmark
| | - Henrik Schmidt
- Department of Oncology, Aarhus University Hospital, Aarhus N, Denmark
| | - Jan Alsner
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital, Aarhus N, Denmark
| | - Niels Henrik Hjollund
- AmbuFlex - Center for Patient-reported Outcomes, Hospital Unit West Jutland, Herning, Denmark
- Faculty of Health, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus N, Denmark
| | - Kirsten Lomborg
- The Research Centre for Patient Involvement, Aarhus University & the Central Denmark Region, Aarhus N, Denmark
- Faculty of Health, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Berit Kjærside Nielsen
- The Research Centre for Patient Involvement, Aarhus University & the Central Denmark Region, Aarhus N, Denmark
- DEFACTUM, Social & Health Services and Labour Market, Central Denmark Region, Aarhus N, Denmark
| |
Collapse
|
3
|
Zhang Y, Zhou Y, Zhang H, Tian L, Hao J, Yuan Y, Li W, Liu Y. DNA binding and evaluation of anticancer activity in vitro and in vivo of iridium(III) polypyridyl complexes. J Inorg Biochem 2021; 224:111580. [PMID: 34438219 DOI: 10.1016/j.jinorgbio.2021.111580] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/01/2021] [Accepted: 08/11/2021] [Indexed: 01/25/2023]
Abstract
In this report, we synthesized three new iridium(III) complexes: [Ir(piq)2(apip)]PF6 (Ir1, piq = 1-phenylisoquinoline, apip = 2-aminophenyl-1H-imidazo[4,5-f][1,10]phenanthroline), [Ir(piq)2(maip)]PF6 (Ir2, maip = 3-aminophenyl-1H-imidazo[4,5-f][1,10]phenanthroline) and [Ir(piq)2(paip)]PF6 (Ir3, paip = 4-aminophenyl-1H-imidazo[4,5-f][1,10]phenanthroline). The DNA binding was investigated. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method was used to detect the cytotoxic activity of Ir1, Ir2 and Ir3, the complexes show highly active against B16 cells with IC50 values of 0.3 ± 0.2 μM, 3.7 ± 0.2 μM and 4.6 ± 1.1 μM, respectively. Subsequently, cellular uptake suggested that the cytotoxicity of the complexes is attributed to their differences in cellular uptake levels. In addition, complexes Ir1, Ir2 and Ir3 induce cell cycle arrest at the G0/G1 phase and regulate the cell cycle mediators such as cyclin D1, CDK6 (cyclin-dependent kinase 6), CDK4 and p21, leading to the inhibition of B16 cells proliferation. The autophagy was investigated by monodansylcadaverine (MDC) staining. The complexes can promote the change from LC3-I to LC3-II, up-regulate levels of Beclin-1 and down-regulate expression of p62. The complexes induced apoptosis by regulating the expression levels of related indicators such as PARP (poly ADP-ribose polymerase), PI3K (phosphoinositide-3 kinase), AKT (protein kinase B), Caspase, Bcl-2 (B-cell lymphoma-2), Bad (Bcl2 associated death promoter), Bax (Bcl2-associated X) and Cyto C (cytochrome C). Additionally, Ir1 exerted significant antitumor activity in the suppression of malignant melanoma proliferation in vivo. As indicated in the above results, these complexes were highly effective for malignant melanoma treatment through the intrinsic pathway and provided much insight into anticancer drugs for tumor therapy.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yi Zhou
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Huiwen Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Li Tian
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Jing Hao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yuhan Yuan
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Wenlong Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yunjun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China.
| |
Collapse
|
4
|
Zhang Y, Jiang B, Guo C, Liu L, Xu J, Wang Y, Shen C, Zhu J, Shen G, Jiang H, Zhu J, Tao J. Multifunctional Cu xS- and DOX-loaded AuNR@mSiO 2 platform for combined melanoma therapy with inspired antitumor immunity. Biomater Sci 2021; 9:4086-4098. [PMID: 33913979 DOI: 10.1039/d1bm00373a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Combined antitumor therapies based on nanomedicines have shown efficacy in various tumor models in recent years, overcoming the disadvantages of inefficiency and undesired toxicity of traditional therapies. Herein, we present a copper sulfide- and doxorubicin-loaded gold nanorods@mesoporous SiO2 multifunctional nanocomposite (AuNR@mSiO2@DOX-CuxS-PEG) to integrate chemotherapy, the photothermal properties of AuNRs, and the photodynamic properties of CuxS into a single nanoplatform based on hydrophobic interaction and electrostatic attraction. Upon near-infrared light irradiation, the AuNR@mSiO2@DOX-CuxS-PEG nanocomposites exhibit a synergistic therapeutic effect and inhibit the in situ tumor growth and lung metastasis in a melanoma model. This occurs because of the high photothermal conversion efficiency, boosted intracellular reactive oxygen species production, and excellent doxorubicin (DOX) release, as well as an induced tumor-specific immune response. The inspired antitumor immunity was confirmed by elevated infiltration of activated T cells in tumor tissues and improved maturation and activation of dendritic cells in tumor-draining lymph nodes. This study highlights the superior antitumor therapeutic effect elicited by a multifunctional nanoplatform for skin with in situ melanoma and lung metastasis inhibition, indicating its satisfactory clinical application prospects.
Collapse
Affiliation(s)
- Yamin Zhang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430022, China.
| | - Biling Jiang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430022, China.
| | - Chen Guo
- Key Laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, School of Chemistry and Chemical Engineering, HUST, Wuhan 430074, China.
| | - Liping Liu
- Key Laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, School of Chemistry and Chemical Engineering, HUST, Wuhan 430074, China.
| | - Jian Xu
- Department of Hematology, Union Hospital, Tongji Medical College, HUST, Wuhan 430022, China
| | - Yujue Wang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430022, China.
| | - Chen Shen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430022, China.
| | - Jinjin Zhu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430022, China.
| | - Guanxin Shen
- Department of Immunology, Tongji Medical College, HUST, Wuhan 430022, China
| | - Hao Jiang
- Key Laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, School of Chemistry and Chemical Engineering, HUST, Wuhan 430074, China.
| | - Jintao Zhu
- Key Laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, School of Chemistry and Chemical Engineering, HUST, Wuhan 430074, China.
| | - Juan Tao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430022, China.
| |
Collapse
|
5
|
Close DA, Kirkwood JM, Fecek RJ, Storkus WJ, Johnston PA. Unbiased High-Throughput Drug Combination Pilot Screening Identifies Synergistic Drug Combinations Effective against Patient-Derived and Drug-Resistant Melanoma Cell Lines. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2021; 26:712-729. [PMID: 33208016 PMCID: PMC8128935 DOI: 10.1177/2472555220970917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We describe the development, optimization, and validation of 384-well growth inhibition assays for six patient-derived melanoma cell lines (PDMCLs), three wild type (WT) for BRAF and three with V600E-BRAF mutations. We conducted a pilot drug combination (DC) high-throughput screening (HTS) of 45 pairwise 4×4 DC matrices prepared from 10 drugs in the PDMCL assays: two B-Raf inhibitors (BRAFi), a MEK inhibitor (MEKi), and a methylation agent approved for melanoma; cytotoxic topoisomerase II and DNA methyltransferase chemotherapies; and drugs targeting the base excision DNA repair enzyme APE1 (apurinic/apyrimidinic endonuclease-1/redox effector factor-1), SRC family tyrosine kinases, the heat shock protein 90 (HSP90) molecular chaperone, and histone deacetylases.Pairwise DCs between dasatinib and three drugs approved for melanoma therapy-dabrafenib, vemurafenib, or trametinib-were flagged as synergistic in PDMCLs. Exposure to fixed DC ratios of the SRC inhibitor dasatinib with the BRAFis or MEKis interacted synergistically to increase PDMCL sensitivity to growth inhibition and enhance cytotoxicity independently of PDMCL BRAF status. These DCs synergistically inhibited the growth of mouse melanoma cell lines that either were dabrafenib-sensitive or had acquired resistance to dabrafenib with cross resistance to vemurafenib, trametinib, and dasatinib. Dasatinib DCs with dabrafenib, vemurafenib, or trametinib activated apoptosis and increased cell death in melanoma cells independently of their BRAF status or their drug resistance phenotypes. These preclinical in vitro studies provide a data-driven rationale for the further investigation of DCs between dasatinib and BRAFis or MEKis as candidates for melanoma combination therapies with the potential to improve outcomes and/or prevent or delay the emergence of disease resistance.
Collapse
Affiliation(s)
- David A. Close
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - John M. Kirkwood
- Departments of Medicine, Dermatology, Translational Science, and Melanoma and Skin Cancer Program University of Pittsburgh School of Medicine, Pittsburgh, PA 15232, USA
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Ronald J. Fecek
- Department of Microbiology, Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, PA 15601, USA
| | - Walter J. Storkus
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA 15232, USA
- Departments of Dermatology, Immunology, Bioengineering and Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Paul A. Johnston
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA 15232, USA
| |
Collapse
|
6
|
Meylan P, Pich C, Winkler C, Ginster S, Mury L, Sgandurra M, Dreos R, Frederick DT, Hammond M, Boland GM, Michalik L. Low expression of the PPARγ-regulated gene thioredoxin-interacting protein accompanies human melanoma progression and promotes experimental lung metastases. Sci Rep 2021; 11:7847. [PMID: 33846376 PMCID: PMC8042115 DOI: 10.1038/s41598-021-86329-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/12/2021] [Indexed: 01/10/2023] Open
Abstract
The thioredoxin system plays key roles in regulating cancer cell malignancy. Here we identify the Thioredoxin-interacting protein (TXNIP) as a gene, which expression is regulated by PPARγ in melanoma cells. We show that high TXNIP expression levels associate with benign melanocytic lesions, with tumor regression in patients on MAP kinase targeted therapy, with decreased proliferation in patients’ melanoma biopsies, and with cell cycle arrest in human melanoma cell lines. In contrast, reduced TXNIP expression associates with advanced melanoma and with disease progression in patients. TXNIP depletion in human melanoma cells altered the expression of integrin beta-3 and the localization of the integrin alpha-v/beta-3 dimer at their surface. Moreover, TXNIP depletion affected human melanoma cell motility and improved their capacity to colonize mouse lungs in an in vivo assay. This study establishes TXNIP as a PPARγ-regulated gene in melanoma cells, thereby suggesting a link between these two proteins both involved in the regulation of cancer and of energy metabolism. It also reveals that the decrease in TXNIP expression, which is observed in advanced patient tumors, likely favors lung metastatic seeding of malignant cells.
Collapse
Affiliation(s)
- Patrick Meylan
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015, Lausanne, Switzerland
| | - Christine Pich
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015, Lausanne, Switzerland.,Department of Dermatology and Venereology, University Hospital of Lausanne, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Carine Winkler
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015, Lausanne, Switzerland
| | - Stefanie Ginster
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015, Lausanne, Switzerland
| | - Lionel Mury
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015, Lausanne, Switzerland
| | - Marie Sgandurra
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015, Lausanne, Switzerland.,R&D Philip Morris Products S.A, Neuchâtel, Switzerland
| | - René Dreos
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015, Lausanne, Switzerland
| | - Dennie Tompers Frederick
- Division of Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Marc Hammond
- Division of Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Genevieve Marie Boland
- Division of Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Liliane Michalik
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015, Lausanne, Switzerland.
| |
Collapse
|
7
|
Triphenylphosphonium derivatives disrupt metabolism and inhibit melanoma growth in vivo when delivered via a thermosensitive hydrogel. PLoS One 2020; 15:e0244540. [PMID: 33378390 PMCID: PMC7773266 DOI: 10.1371/journal.pone.0244540] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 12/11/2020] [Indexed: 12/27/2022] Open
Abstract
Despite dramatic improvements in outcomes arising from the introduction of targeted therapies and immunotherapies, metastatic melanoma is a highly resistant form of cancer with 5 year survival rates of <35%. Drug resistance is frequently reported to be associated with changes in oxidative metabolism that lead to malignancy that is non-responsive to current treatments. The current report demonstrates that triphenylphosphonium(TPP)-based lipophilic cations can be utilized to induce cytotoxicity in pre-clinical models of malignant melanoma by disrupting mitochondrial metabolism. In vitro experiments demonstrated that TPP-derivatives modified with aliphatic side chains accumulated in melanoma cell mitochondria; disrupted mitochondrial metabolism; led to increases in steady-state levels of reactive oxygen species; decreased total glutathione; increased the fraction of glutathione disulfide; and caused cell killing by a thiol-dependent process that could be rescued by N-acetylcysteine. Furthermore, TPP-derivative-induced melanoma toxicity was enhanced by glutathione depletion (using buthionine sulfoximine) as well as inhibition of thioredoxin reductase (using auranofin). In addition, there was a structure-activity relationship between the aliphatic side-chain length of TPP-derivatives (5–16 carbons), where longer carbon chains increased melanoma cell metabolic disruption and cell killing. In vivo bio-distribution experiments showed that intratumoral administration of a C14-TPP-derivative (12-carbon aliphatic chain), using a slow-release thermosensitive hydrogel as a delivery vehicle, localized the drug at the melanoma tumor site. There, it was observed to persist and decrease the growth rate of melanoma tumors. These results demonstrate that TPP-derivatives selectively induce thiol-dependent metabolic oxidative stress and cell killing in malignant melanoma and support the hypothesis that a hydrogel-based TPP-derivative delivery system could represent a therapeutic drug-delivery strategy for melanoma.
Collapse
|
8
|
Novel therapies for advanced skin carcinomas. Postepy Dermatol Alergol 2020; 37:660-670. [PMID: 33240003 PMCID: PMC7675086 DOI: 10.5114/ada.2020.100479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 04/02/2019] [Indexed: 12/11/2022] Open
Abstract
Advanced skin carcinomas are a serious therapeutic problem. The statistical analysis shows a continuous increase in the incidence of both melanoma and non-melanoma skin cancers. Traditional therapies are characterized by low effectiveness and patients overall survival is not affected by them. By understanding the molecular pathways that lead to the neoplastic transformation and thanks to the knowledge of the immune system, it is possible to use personalized medicine in novel therapies for advanced skin carcinomas.
Collapse
|
9
|
Zhang Y, Guo C, Liu L, Xu J, Jiang H, Li D, Lan J, Li J, Yang J, Tu Q, Sun X, Alamgir M, Chen X, Shen G, Zhu J, Tao J. ZnO-based multifunctional nanocomposites to inhibit progression and metastasis of melanoma by eliciting antitumor immunity via immunogenic cell death. Am J Cancer Res 2020; 10:11197-11214. [PMID: 33042278 PMCID: PMC7532661 DOI: 10.7150/thno.44920] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 08/27/2020] [Indexed: 12/26/2022] Open
Abstract
Rationale: The development of a highly effective and tumor-specific therapeutic strategy, which can act against the primary tumor and also condition the host immune system to eliminate distant tumors, remains a clinical challenge. Methods: Herein, we demonstrate a facile yet versatile ZnO-capping and Doxorubicin (DOX)-loaded multifunctional nanocomposite (AuNP@mSiO2@DOX-ZnO) that integrates photothermal properties of gold nanoparticles (NPs), pH-responsive properties and preferential selectivity to tumor cells of ZnO QDs and chemotherapeutic agent into a single NP. The photothermal performance, pH-triggered release and preferential phagocytic ability were assessed. The induced anti-tumor immunity was determined by analyzing immune cell profile in tumor in vivo and molecular mechanism were identified by detecting expression of immunogenic cell death (ICD) markers in vitro. Moreover, mice models of unilateral and bilateral subcutaneous melanoma and lung metastasis were established to evaluate the antitumor effects. Results: As an efficient drug carrier, ZnO-capped NPs guarantee a high DOX payload and an in vitro, efficient release of at pH 5.0. In murine melanoma models, the nanocomposite can significantly inhibit tumor growth for a short period upon low-power laser irradiation. Importantly, ZnO NPs not only demonstrate preferential selectivity for melanoma cells but can also induce ICD. Meanwhile, AuNP@mSiO2-based photothermal therapy (PTT) and DOX are directly cytotoxic towards cancer cells and demonstrate an elevated ICD effect. The induced ICD promotes maturation of dendritic cells, further stimulating the infiltration of effector T cells into tumor sites, preventing tumor growth and distant lung metastases. Conclusions: This study highlights the novel mechanism of ZnO-triggered anti-tumor immunity via inducing ICD. Additionally, we shed light on the multifunctionality of nanocomposites in delivering localized skin tumor therapy as well as inhibiting metastatic growth, which holds great promise in clinical applications.
Collapse
|
10
|
Huijberts S, Wang L, de Oliveira RL, Rosing H, Nuijen B, Beijnen J, Bernards R, Schellens J, Wilgenhof S. Vorinostat in patients with resistant BRAFV600E mutated advanced melanoma: a proof of concept study. Future Oncol 2020; 16:619-629. [PMID: 32125175 DOI: 10.2217/fon-2020-0023] [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] [Indexed: 12/21/2022] Open
Abstract
The clinical benefit of treatment with BRAF- and MEK-inhibitors in melanoma is limited due to resistance associated with emerging secondary mutations. Preclinical and clinical studies have shown that short-term treatment with the HDAC inhibitor vorinostat can eliminate cells harboring these secondary mutations causing resistance. This proof of concept study is to determine the efficacy of sequential treatment with vorinostat and BRAFi/MEKi in resistant BRAFV600E mutant melanoma. The primary aim is demonstrating anti-tumor response of progressive lesions according to RECIST 1.1. Secondary end points are to determine that emerging resistant clones with a secondary mutation in the MAPK pathway can be detected in circulating tumor DNA and purged by short-term vorinostat treatment. Exploratory end points include pharmacokinetic, pharmacodynamic and pharmacogenetic analyses (NCT02836548).
Collapse
Affiliation(s)
- Sanne Huijberts
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Liqin Wang
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Rodrigo Leite de Oliveira
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Hilde Rosing
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Bastiaan Nuijen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Jos Beijnen
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands.,Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands.,Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Rene Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands.,Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Jan Schellens
- Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Sofie Wilgenhof
- Department of Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| |
Collapse
|
11
|
Fox J, Janda M, Bennett F, Langbecker D. An outreach telephone program for advanced melanoma supportive care: Acceptability and feasibility. Eur J Oncol Nurs 2019; 42:110-115. [PMID: 31493669 DOI: 10.1016/j.ejon.2019.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 08/27/2019] [Accepted: 08/28/2019] [Indexed: 10/26/2022]
Abstract
PURPOSE People with advanced melanoma face an uncertain trajectory as new treatments now have the potential to provide longer-term survival for some. However, the disease course is variable and unpredictable, with many expressing a need for better supportive care. This study aimed to investigate the acceptability and feasibility of extending an existing melanoma-specific self-referral or 'passive' telephone consultation support service to an 'active' outreach call to offer a supportive care program tailored to the needs of the patient. METHOD Participants were enrolled by their oncology nurse into a single group pre-post intervention study. Participants received an outreach telephone call focused on knowledge and skill development. Participants completed questionnaires at baseline and four weeks post-intervention. Post-intervention interviews with patients and involved staff were used to explore acceptability and feasibility of the outreach service call. RESULTS Of 18 participants approached, 15 enrolled and 14 received the intervention. Staff time required for intervention delivery provided evidence for feasibility. Participants perceived the intervention as acceptable, and beneficial. In interviews, having someone with melanoma-specific knowledge to talk with was a key benefit of the outreach call program. Many participants expressed that they would have wished to receive the outreach call at an earlier stage, for example at the time of recurrence of/progression to advanced melanoma. CONCLUSIONS Extending an existing self-referral support service model to use a more 'active' outreach approach is acceptable and feasible. The next step in the evaluation process for this intervention is a randomised controlled trial to determine effectiveness and cost-effectiveness.
Collapse
Affiliation(s)
- Jennifer Fox
- School of Nursing, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Qld, 4075, Australia.
| | - Monika Janda
- Centre for Health Service Research, Faculty of Medicine, Institute of Health and Biomedical Innovation, The University of Queensland, Woolloongabba, Qld, 4102, Australia; School of Public Health and Social Work, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Australia.
| | - Fiona Bennett
- Melanoma Patients Australia, Mater Foundation, 620 Stanley Street, Woolloongabba, Brisbane, 4102, Australia.
| | - Danette Langbecker
- Centre for Online Health - Centre for Health Services Research, The University of Queensland, Ground floor, Building 33, Princess Alexandra Hospital, 199 Ipswich Rd, Woolloongabba, Qld, 4102, Australia.
| |
Collapse
|
12
|
Ailanthone Induces Cell Cycle Arrest and Apoptosis in Melanoma B16 and A375 Cells. Biomolecules 2019; 9:biom9070275. [PMID: 31336757 PMCID: PMC6681521 DOI: 10.3390/biom9070275] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 12/17/2022] Open
Abstract
Malignant melanoma is the most lethal type of skin cancer. Previous studies have shown that ailanthone has potent antitumor activity in a variety of cell lines. However, the anti-tumor effect of ailanthone on malignant melanoma remains unclear. To investigate the anti-tumor mechanisms of ailanthone in human melanoma B16 and mouse melanoma A375 cells, the cell counting kit-8 assay, colony formation assay, DNA content analysis, Hoechst 33258, and Annexin V-FITC/PI staining were used to assess cell proliferation, cell cycle distribution, and cell apoptosis, respectively. Western blotting was performed to evaluate the expression of cell cycle- and apoptosis-related proteins and regulatory molecules. The results showed that ailanthone significantly inhibited melanoma B16 and A375 cell proliferation as well as remarkably induced cell cycle arrest at the G0–G1 phase in B16 cells and the G2–M phase in A375 cells in a dose-dependent manner. Further investigation revealed that ailanthone promoted the expression of p21 and suppressed the expression of cyclin E in B16 cells or cyclin B in A375 cells through the PI3K-Akt signaling pathway. In addition, ailanthone induced B16 and A375 cell apoptosis via a caspase-dependent mechanism. Further studies showed that ailanthone remarkably downregulated Bcl-2 and upregulated Apaf-1 and Bax, and subsequently increased mitochondrial membrane permeabilization and released cytochrome c from the mitochondria in B16 cells and A375 cells. Taken together, ailanthone induces cell cycle arrest via the PI3K-Akt signaling pathway as well as cell apoptosis via the mitochondria-mediated apoptotic signaling pathway. Ailanthone may be potentially utilized as an anti-tumor agent in the management of malignant melanoma.
Collapse
|
13
|
Brouwer NJ, Genders SW, Marinkovic M, van Duinen SG, Jager MJ, Luyten GP. Two Late Recurrences of Conjunctival Melanoma. Ocul Oncol Pathol 2019; 5:262-266. [PMID: 31367588 PMCID: PMC6615343 DOI: 10.1159/000494978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/29/2018] [Indexed: 11/19/2022] Open
Abstract
PURPOSE To report a patient who developed two late recurrences of conjunctival melanoma (CoM), of which one occurred after orbital exenteration. METHODS We describe the case of a patient based on clinical and histopathological examination. RESULTS A 52-year-old patient was treated with local excision and cryotherapy for a CoM with primary acquired melanosis (PAM) near the limbus of the right eye. Twenty-one years later, a recurrence developed in the superior fornix of the same eye in an area with widespread PAM; an orbital exenteration was performed. After another 4 years, a painful nodule developed subcutaneously at the inferior margin of the right orbital socket. Pathology showed a recurrence of CoM with a BRAF V600K mutation, similar to both of the previous lesions (of 25 and 4 years earlier). The nodule was excised without additional therapy. No recurrences or metastases have been observed in the next 2.5 years. The proposed mechanism for the recurrence after surgery could be via dormant tumor cells that have spread prior to the procedure or via residual intraepithelial malignant melanocytes. CONCLUSION Very late recurrences of CoM are rare but may occur. Our case illustrates the need for long-term awareness of doctors and patients, even after extensive surgical procedures such as orbital exenteration.
Collapse
Affiliation(s)
- Niels J. Brouwer
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Stijn W. Genders
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marina Marinkovic
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sjoerd G. van Duinen
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Martine J. Jager
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gré P.M. Luyten
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
14
|
Salmi S, Siiskonen H, Sironen R, Tyynelä-Korhonen K, Hirschovits-Gerz B, Valkonen M, Auvinen P, Pasonen-Seppänen S. The number and localization of CD68+ and CD163+ macrophages in different stages of cutaneous melanoma. Melanoma Res 2019; 29:237-247. [PMID: 30399061 PMCID: PMC6493694 DOI: 10.1097/cmr.0000000000000522] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 09/19/2018] [Indexed: 01/20/2023]
Abstract
The role of tumor-associated macrophages (TAMs) in cutaneous melanoma is controversial. TAMs include immunogenic and immunosuppressive subtypes, and have distinct functions according to their microanatomical localization. Our aim was to investigate TAMs in benign, premalignant, and malignant melanocytic lesions to determine possible associations with tumor progression and clinicopathological characteristics. In total, 184 tissue samples, including benign and dysplastic nevi, in-situ melanomas, superficial (Breslow's depth <1 mm), and deep (Breslow's depth >4 mm) invasive melanomas and lymph node metastases, were analyzed for macrophage content. Samples were stained immunohistochemically for CD68 and CD163, representing all TAMs and M2-macrophages, respectively. Macrophages were counted by hotspot analysis, and assessed semiquantitatively from the tumor cell nests and stromal component of malignant cases. CD68+ and CD163+ TAMs were more abundant in invasive melanomas compared with benign nevi. The proportion of TAMs in the tumor nests was higher in deep melanomas and lymph node metastases compared with superficially invasive melanomas. High amounts of CD68+ macrophages in tumor cell nests were associated with recurrence, whereas low CD163+ macrophage proportion in tumor stroma was associated with recurrence and in primary melanomas also with poor overall survival. TAMs seem to promote tumor progression in cutaneous melanoma. In particular, CD68+ TAMs and their abundance in tumor nests were associated with poor prognostic factors. However, the correlation of low stromal CD163+ TAM proportion with a poor prognosis indicates that the role of TAMs depends on their subtype and microanatomical localization.
Collapse
Affiliation(s)
| | - Hanna Siiskonen
- Department of Dermatology
- Dermatology, Kuopio University Hospital, Kuopio, Finland
| | - Reijo Sironen
- Institute of Clinical Medicine/Clinical Pathology
- Cancer Center of Eastern Finland, University of Eastern Finland
- Departments of Clinical Pathology
| | | | | | | | - Päivi Auvinen
- Cancer Center of Eastern Finland, University of Eastern Finland
- Oncology
| | | |
Collapse
|
15
|
Leeneman B, Franken MG, Coupé VM, Hendriks MP, Kruit W, Plaisier PW, van Ruth S, Verstijnen JA, Wouters MW, Blommestein HM, Uyl – de Groot CA. Stage-specific disease recurrence and survival in localized and regionally advanced cutaneous melanoma. Eur J Surg Oncol 2019; 45:825-831. [DOI: 10.1016/j.ejso.2019.01.225] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 12/20/2018] [Accepted: 01/31/2019] [Indexed: 10/27/2022] Open
|
16
|
Ye Y, Huang S, Wu Y. UNBS5162 and amonafide inhibits tumor progression in human melanoma by the AKT/mTOR pathway. Cancer Manag Res 2019; 11:2339-2348. [PMID: 30962721 PMCID: PMC6434921 DOI: 10.2147/cmar.s177623] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Background Human melanoma is a malignant tumor originated from melanocytes with high invasion, metastasis, and poor prognosis. In this study, the effects of naphthalimides UNBS5162 and amonafide on the properties of proliferation and apoptosis in human melanoma cells were confirmed. Methods Cell proliferation was determined by CCK8 and clone formation assay. Transwell assay was performed to detect the migration and invasion of M14 and A375 cells. Cell apoptosis was estimated using flow cytometry. Results In a drug sensitivity assay, cell viability decreased with increasing concentrations of UNBS5162 or amonafide. Likewise, proliferation of M14 or A375 cells treated with 10 μM UNBS5162 or 8 μM amonafide decreased significantly when compared with negative control (NC) cells, their inhibition effect verified by means of a clone formation assay. After the treatment with UNBS5162 or amonafide, the migration of melanoma cells was inhibited in a dosede-pendent manner. The number of invaded cells treated with UNBS5162 was also significantly reduced when compared with those of the NC cells. The apoptotic cell numbers treated with UNBS5162 or amonafide decreased significantly when compared with the M14 and A375 cells in the NC group. According to Western blot results, phosphorylation of AKT and expressions of mesenchymal marker factors were inhibited in cells treated with UNBS5162 or amonafide. Conclusion These results reveal that UNBS5162 inhibits the cell activity of melanoma cells through the AKT/mTOR signaling pathway, and reverses epithelial–mesenchymal transition conversion in human melanoma cells. This study on UNBS5162 and amonafide in melanomas provides an experimental basis of their uses and potential value on human melanoma treatment.
Collapse
Affiliation(s)
- Yingyi Ye
- Department of Dermatology, HwaMei Hospital, University of Chinese Academy of Sciences, Zhejiang, China
| | - Shuhong Huang
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Basic Medical Science, Shandong University, Shandong, China
| | - Yingying Wu
- Department of Oncology, HwaMei Hospital, University of Chinese Academy of Sciences, Zhejiang, China,
| |
Collapse
|
17
|
Pich C, Michalik L. The Janus face of rosiglitazone. Oncotarget 2018; 9:37614-37615. [PMID: 30701018 PMCID: PMC6340879 DOI: 10.18632/oncotarget.26532] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 12/21/2018] [Indexed: 02/06/2023] Open
Affiliation(s)
- Christine Pich
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Liliane Michalik
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
18
|
Ozola A, Ruklisa D, Pjanova D. Association of the 16q24.3 region gene variants rs1805007 and rs4785763 with heightened risk of melanoma in Latvian population. Meta Gene 2018. [DOI: 10.1016/j.mgene.2018.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
19
|
Koppolu V, Rekha Vasigala VK. Checkpoint immunotherapy by nivolumab for treatment of metastatic melanoma. J Cancer Res Ther 2018; 14:1167-1175. [PMID: 30488824 DOI: 10.4103/jcrt.jcrt_1290_16] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Clinical management of metastatic melanoma suffered historically from a lack of effective targeted and immunotherapies due to short-lived clinical responses. Recent advances in our understanding of tumor-immune signaling pathways, discovery of immunosuppressive checkpoints, and subsequent development of antibodies that target these checkpoints reverses the situation to some extent. Two antibodies ipilimumab and nivolumab gained Food and Drug administration approval for the treatment of metastatic melanoma and target two major immunosuppressive checkpoints cytotoxic T lymphocyte antigen and programmed cell death protein 1 (PD-1), respectively. Nivolumab binds to PD-1, prevents PD-1 interaction with ligand Programmed death ligand 1 (PD-L1), and thus releases the T-cell exhaustion events (such as T cell apoptosis, decrease in T cell proliferation, etc.) leading to buildup of potent tumor-specific immune response. Successful Phase I-III results with remarkable antitumor activity and safety led to approval of nivolumab against ipilimumab refractory metastatic melanoma. Nivolumab therapy is exciting in that it not only provides substantial benefit but also provides durable responses. This review focuses on the evolution of immunotherapy leading to nivolumab approval and its potential in treating melanoma either alone or in combination with other therapies.
Collapse
Affiliation(s)
- Veerendra Koppolu
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Veneela Krishna Rekha Vasigala
- Department of General Medicine, Rangaraya Medical College, NTR University of Health Sciences, Vijayawada, Andhra Pradesh, India
| |
Collapse
|
20
|
Pich C, Meylan P, Mastelic-Gavillet B, Nguyen TN, Loyon R, Trang BK, Moser H, Moret C, Goepfert C, Hafner J, Levesque MP, Romero P, Jandus C, Michalik L. Induction of Paracrine Signaling in Metastatic Melanoma Cells by PPARγ Agonist Rosiglitazone Activates Stromal Cells and Enhances Tumor Growth. Cancer Res 2018; 78:6447-6461. [PMID: 30185551 DOI: 10.1158/0008-5472.can-18-0912] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/27/2018] [Accepted: 08/28/2018] [Indexed: 01/10/2023]
Abstract
In addition to improving insulin sensitivity in type 2 diabetes, the thiazolidinedione family of compounds and the pharmacologic activation of their best-characterized target PPARγ have been proposed as a therapeutic option for cancer treatment. In this study, we reveal a new mode of action for the thiazolidinedione rosiglitazone that can contribute to tumorigenesis. Rosiglitazone activated a tumorigenic paracrine communication program in a subset of human melanoma cells that involves the secretion of cytokines, chemokines, and angiogenic factors. This complex blend of paracrine signals activated nonmalignant fibroblasts, endothelial cells, and macrophages in a tumor-friendly way. In agreement with these data, rosiglitazone promoted human melanoma development in xenografts, and tumors exposed to rosiglitazone exhibited enhanced angiogenesis and inflammation. Together, these findings establish an important tumorigenic action of rosiglitazone in a subset of melanoma cells. Although studies conducted on cohorts of diabetic patients report overall benefits of thiazolidinediones in cancer prevention, our data suggest that exposure of established tumors to rosiglitazone may be deleterious.Significance: These findings uncover a novel mechanism by which the thiazolidinedione compound rosiglitazone contributes to tumorigenesis, thus highlighting a potential risk associated with its use in patients with established tumors. Cancer Res; 78(22); 6447-61. ©2018 AACR.
Collapse
Affiliation(s)
- Christine Pich
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Patrick Meylan
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Beatris Mastelic-Gavillet
- Department of Oncology, University of Lausanne, Ludwig Cancer Research Center, Lausanne, Switzerland
| | - Thanh Nhan Nguyen
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Romain Loyon
- Department of Oncology, University of Lausanne, Ludwig Cancer Research Center, Lausanne, Switzerland
| | - Bao Khanh Trang
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Hélène Moser
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Catherine Moret
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Christine Goepfert
- COMPATH, Institute of Animal Pathology, University of Bern, Bern, Switzerland
| | - Jürg Hafner
- Department of Dermatology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Pedro Romero
- Department of Oncology, University of Lausanne, Ludwig Cancer Research Center, Lausanne, Switzerland
| | - Camilla Jandus
- Department of Oncology, University of Lausanne, Ludwig Cancer Research Center, Lausanne, Switzerland
| | - Liliane Michalik
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
21
|
Approaches to decision-making among late-stage melanoma patients: a multifactorial investigation. Support Care Cancer 2018; 27:1059-1070. [PMID: 30136025 PMCID: PMC6373271 DOI: 10.1007/s00520-018-4395-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 08/01/2018] [Indexed: 11/23/2022]
Abstract
Purpose The treatment decisions of melanoma patients are poorly understood. Most research on cancer patient decision-making focuses on limited components of specific treatment decisions. This study aimed to holistically characterize late-stage melanoma patients’ approaches to treatment decision-making in order to advance understanding of patient influences and supports. Methods (1) Exploratory analysis of longitudinal qualitative data to identify themes that characterize patient decision-making. (2) Pattern analysis of decision-making themes using an innovative method for visualizing qualitative data: a hierarchically-clustered heatmap. Participants were 13 advanced melanoma patients at a large academic medical center. Results Exploratory analysis revealed eight themes. Heatmap analysis indicated two broad types of patient decision-makers. “Reliant outsiders” relied on providers for medical information, demonstrated low involvement in decision-making, showed a low or later-in-care interest in clinical trials, and expressed altruistic motives. “Active insiders” accessed substantial medical information and expertise in their networks, consulted with other doctors, showed early and substantial interest in trials, demonstrated high involvement in decision-making, and employed multiple decision-making strategies. Conclusion We identified and characterized two distinct approaches to decision-making among patients with late-stage melanoma. These differences spanned a wide range of factors (e.g., behaviors, resources, motivations). Enhanced understanding of patients as decision-makers and the factors that shape their decision-making may help providers to better support patient understanding, improve patient-provider communication, and support shared decision-making.
Collapse
|
22
|
Patient-reported treatment-related symptom burden for patients with advanced melanoma in Canada. Support Care Cancer 2018; 27:219-227. [PMID: 29934684 PMCID: PMC6281076 DOI: 10.1007/s00520-018-4316-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 06/12/2018] [Indexed: 12/04/2022]
Abstract
Background Little is known on the impact of emerging treatments for advanced melanoma (stages III and IV) on patients’ functioning and well-being. The objective of this study was to describe the patient-reported treatment-related symptom (TRS) burden in advanced melanoma. Method Twenty-nine in-depth, qualitative interviews were conducted among adult patients with advanced melanoma in Canada using a semi-structured interview method. Interviews were transcribed verbatim, and key concepts were identified using a grounded theory analytic approach. Results The 29 patients reported 13 unique treatment journeys involving the following drug therapy categories: cytotoxic chemotherapies, CTLA-4 inhibitors, BRAF or MEK inhibitors, and PD-1 inhibitors. Patients typically underwent multiple treatment episodes over time. Common TRSs included nausea, fatigue, diarrhea or constipation, and skin rashes. Patients described these as impacting their physical functioning, ability to perform activities of daily living, social functioning, and overall quality of life. Conclusion Our findings provide a description of the patient’s experience with treatment for advanced melanoma. Our sample included patients typically diagnosed in mid-life, facing an urgent sequence of medical procedures and a pharmacological treatment journey that was burdensome. There is a need for less toxic and more efficacious treatments earlier in the patient journey to alleviate the impact of advanced melanoma treatment on patients’ health-related quality of life.
Collapse
|
23
|
Abstract
Although microRNAs (miRNAs) have emerged as potent mediators of melanoma development and progression, a precise understanding of their oncogenic role remains unclear. In this study, we analysed formalin-fixed and paraffin-embedded tissues from two separate melanoma cohorts and from a series of benign melanocytic nevi. Using three different quantification methods [array analysis, quantitative PCR (qPCR) and in-situ hybridization (ISH) quantified by digital image analysis], we found considerable miRNA dysregulation in tumours. Using array analysis, samples mainly clustered according to their biological group (benign vs. malignant) and 77 miRNAs differed significantly between nevi and melanoma samples. Increase of miR-21 and miR-142, and decrease of miR-125b, miR-211, miR-101 and miR-513c in the melanomas were verified in both cohorts using qPCR, whereas the decrease of miR-205 observed with array analysis could not be confirmed using qPCR. ISH with digital quantification showed expression of miR-21 and miR-125b in the melanocytic lesions. miR-21 ISH was increased in melanomas, whereas quantification of miR-125b showed uniform ISH expression across nevi and melanomas. Our results support the important involvement of different miRNAs in melanoma biology and may serve as solid basics for further miRNA investigations in melanoma formalin-fixed and paraffin-embedded tissue. In particular, there is increased expression of miR-21 in melanomas compared with benign nevi.
Collapse
|
24
|
Dynamic contrast-enhanced MRI coupled with a subtraction technique is useful for treatment response evaluation of malignant melanoma hepatic metastasis. Oncotarget 2018; 7:38513-38522. [PMID: 27229529 PMCID: PMC5122407 DOI: 10.18632/oncotarget.9567] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 05/08/2016] [Indexed: 01/01/2023] Open
Abstract
PURPOSE To determine whether contrast-enhanced MRI including subtraction sequences can predict the treatment response of melanoma liver metastasis. RESULTS High precontrast T1 signal intensity (SI) of melanoma lesions obscured detection of enhancement after contrast injection. It was impossible to determine whether or not enhancement occurred in the majority of lesions (85.4%, n = 35/41) without including the subtraction technique. Positive enhancement was identified in 14.6% (n = 6/41) of patients without subtraction images, but increased to 68.3% (n = 28/41) by including subtraction images. Follow-up studies determined lesion progression in 34.1% (n = 14/41) of patients. Positive enhancement on the subtraction image (odds ratio = 12.1, P = 0.048) and intermediate high T2 SI (odds ratio = 8.16, P = 0.040) were significantly associated with higher risk of lesion progression. MATERIALS AND METHODS Patients who underwent MRI for melanoma liver metastases between January 2007 and February 2015 were enrolled. The study analyzed 41 liver metastases in 15 patients [11 male and four female; median age 56 years (range 21-81)] for size, lesion enhancement with and without subtraction images, and T2 SI. Follow-up imaging studies were used to determine treatment response. Data were analyzed with generalized estimating equations. CONCLUSIONS MRI including the subtraction technique is useful for determining the treatment response of melanoma liver metastases. Lesion contrast enhancement and intermediate high T2 SI increased the risk of lesion progression.
Collapse
|
25
|
Cervia D, Assi E, De Palma C, Giovarelli M, Bizzozero L, Pambianco S, Di Renzo I, Zecchini S, Moscheni C, Vantaggiato C, Procacci P, Clementi E, Perrotta C. Essential role for acid sphingomyelinase-inhibited autophagy in melanoma response to cisplatin. Oncotarget 2018; 7:24995-5009. [PMID: 27107419 PMCID: PMC5041885 DOI: 10.18632/oncotarget.8735] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 03/28/2016] [Indexed: 01/03/2023] Open
Abstract
The sphingolipid metabolising enzyme Acid Sphingomyelinase (A-SMase) has been recently shown to inhibit melanoma progression and correlate inversely to tumour grade. In this study we have investigated the role of A-SMase in the chemo-resistance to anticancer treatmentusing mice with melanoma allografts and melanoma cells differing in terms of expression/activity of A-SMase. Since autophagy is emerging as a key mechanism in tumour growth and chemo-resistance, we have also investigated whether an action of A-SMase in autophagy can explain its role. Melanoma sensitivity to chemotherapeutic agent cisplatin in terms of cell viability/apoptosis, tumour growth, and animal survival depended directly on the A-SMase levels in tumoural cells. A-SMase action was due to inhibition of autophagy through activation of Akt/mammalian target of rapamycin (mTOR) pathway. Treatment of melanoma-bearing mice with the autophagy inhibitor chloroquine restored sensitivity to cisplatin of tumours expressing low levels of A-SMase while no additive effects were observed in tumours characterised by sustained A-SMase levels. The fact that A-SMase in melanomas affects mTOR-regulated autophagy and plays a central role in cisplatin efficacy encourages pre-clinical testing on the modulation of A-SMase levels/activity as possible novel anti-neoplastic strategy.
Collapse
Affiliation(s)
- Davide Cervia
- Department for Innovation in Biological, Agro-food and Forest Systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy.,Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milano, Italy
| | - Emma Assi
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy.,Present address: Division of Experimental Oncology, San Raffaele Scientific Institute, Milano, Italy
| | - Clara De Palma
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milano, Italy.,Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, University Hospital "Luigi Sacco", Milano, Italy
| | - Matteo Giovarelli
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milano, Italy
| | - Laura Bizzozero
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy.,Present address: Department of Oncology, Università degli Studi di Torino and Laboratory of Neurovascular Biology, Candiolo Cancer Institute, Candiolo, Italy
| | - Sarah Pambianco
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milano, Italy
| | - Ilaria Di Renzo
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milano, Italy
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milano, Italy
| | - Claudia Moscheni
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milano, Italy
| | | | - Patrizia Procacci
- Department of Biomedical Sciences for Health (SCIBIS), Università degli Studi di Milano, Milano, Italy
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milano, Italy.,Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy.,Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, University Hospital "Luigi Sacco", Milano, Italy
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
26
|
Abstract
PURPOSE To describe the development of uveitis and retinal vasculitis in association with pembrolizumab treatment for metastatic uveal melanoma. METHODS A case report and a brief review of the literature are presented. Information collected and reported include the patient's clinical course, physical examination findings, fluorescein angiogram images, retinal photographs, and her response to treatment. RESULTS A 54-year-old woman was diagnosed with a large choroidal malignant melanoma and had the affected eye enucleated. Pathology confirmed a mixed cell choroidal melanoma, and gene expression profiling was Class 2. Seventeen months after enucleation, the patient was diagnosed with metastatic uveal melanoma to the liver. Disease progression was observed during ipilimumab treatment. Pembrolizumab treatment was initiated, and after four infusions, she presented to clinic complaining of floaters and blurred vision. Examination revealed a nongranulomatous panuveitis characterized by perivascular retinal pigment epithelium pigmentary changes, retinal venous sheathing, 1+ anterior chamber and vitreous cellular reaction, 2+ vitreous haze, and optic disk edema. A dexamethasone sustained-release implant was administered and the uveitis regressed. A relapse in symptoms occurred but quickly subsided with a repeat injection. CONCLUSION Pembrolizumab may induce a uveitic reaction. There is mounting evidence that patients using prembrolizumab should be educated and monitored for signs of uveitis.
Collapse
|
27
|
Brouwer NJ, Marinkovic M, Jochems A, Kapiteijn EW, van Duinen SG, Haeseker BI, Jager MJ, Luyten GPM. Conjunctival Metastasis of a Cutaneous Melanoma. Ocul Oncol Pathol 2017; 4:107-111. [PMID: 30320090 DOI: 10.1159/000479114] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 06/30/2017] [Indexed: 02/03/2023] Open
Abstract
Purpose To report a patient who presented with a conjunctival tumour as a first sign of distant metastasis of cutaneous melanoma. The patient was treated successfully with BRAF/MEK-inhibitors and anti-PD-1 antibodies. Methods Clinical and histopathological examination of the conjunctival lesion. Results A 74-year-old man was referred to our hospital with a pigmented conjunctival tumour, 5 months after having been diagnosed with cutaneous melanoma on his right scapula with loco-regional axillary lymph node metastases. The conjunctival lesion was excised and showed a BRAF V600E mutation. Histopathology showed a melanoma with characteristics suspicious for metastasis, as the lesion did not have a relation with the overlying epithelium. Systemic screening showed multiple distant metastases of the cutaneous melanoma in spleen, liver, and bone. Systemic treatment with the combination of a BRAF-inhibitor (dabrafenib) and MEK-inhibitor (trametinib) was started and followed by a switch to an anti-PD-1 antibody (pembrolizumab). Twenty-two months later, the patient is alive and in good clinical health. Conclusion Conjunctival metastases of cutaneous melanoma may mimic primary conjunctival melanoma. A good medical history and systemic work-up are required to differentiate these diseases. Identification of the proper diagnosis including mutation analysis is crucial, allowing patients to benefit from newly introduced treatment strategies for metastatic cutaneous melanoma.
Collapse
Affiliation(s)
- Niels J Brouwer
- Department of Ophthalmology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Marina Marinkovic
- Department of Ophthalmology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Anouk Jochems
- Department of Medical Oncology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Ellen W Kapiteijn
- Department of Medical Oncology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Sjoerd G van Duinen
- Department of Pathology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Barbara I Haeseker
- Department of Department of Ophthalmology, Alrijne Hospital, Leiden, The Netherlands
| | - Martine J Jager
- Department of Ophthalmology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Gregorius P M Luyten
- Department of Ophthalmology, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
28
|
Cervenka PD, Perez L, Perez DE, Jones B. Melanoma Metastasis to the Mandible—Case Report and Comprehensive Literature Review. J Oral Maxillofac Surg 2017; 75:2025.e1-2025.e12. [DOI: 10.1016/j.joms.2017.05.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/16/2017] [Accepted: 05/16/2017] [Indexed: 10/19/2022]
|
29
|
El Chediak A, Shamseddine A, Bodgi L, Obeid JP, Geara F, Zeidan YH. Optimizing tumor immune response through combination of radiation and immunotherapy. Med Oncol 2017; 34:165. [PMID: 28828581 DOI: 10.1007/s12032-017-1025-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 08/12/2017] [Indexed: 12/13/2022]
Abstract
Radiation therapy and immunotherapy are two highly evolving modalities for the treatment of solid tumors. Immunotherapeutic drugs can either stimulate the immune system via immunogenic pathways or target co-inhibitory checkpoints. An augmented tumor cell recognition by host immune cells can be achieved post-irradiation, as irradiated tissues can release chemical signals which are sensed by the immune system resulting in its activation. Different strategies combining both treatment modalities were tested in order to achieve a better therapeutic response and longer tumor control. Both regimens act synergistically to one another with complimentary mechanisms. In this review, we explore the scientific basis behind such a combination, starting initially with a brief historical overview behind utilizing radiation and immunotherapies for solid tumors, followed by the different types of these two modalities, and the biological concept behind their synergistic effect. We also shed light on the common side effects and toxicities associated with radiation and immunotherapy. Finally, we discuss previous clinical trials tackling this multimodality combination and highlight future ongoing research.
Collapse
Affiliation(s)
- Alissar El Chediak
- Division of Hematology/Oncology, Department of Internal Medicine, Data Management and Clinical Research Unit, Naef K. Basile Cancer Institute- NKBCI American University of Beirut Medical Center, P.O. Box 11-0236, Riad El Solh, Lebanon
| | - Ali Shamseddine
- Division of Hematology/Oncology, Department of Internal Medicine, Data Management and Clinical Research Unit, Naef K. Basile Cancer Institute- NKBCI American University of Beirut Medical Center, P.O. Box 11-0236, Riad El Solh, Lebanon.
| | - Larry Bodgi
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Jean-Pierre Obeid
- Department of Internal Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Fady Geara
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Youssef H Zeidan
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
30
|
Abdel-Rahman O. PD-L1 expression and outcome of advanced melanoma patients treated with anti-PD-1/PD-L1 agents: a meta-analysis. Immunotherapy 2017; 8:1081-9. [PMID: 27485080 DOI: 10.2217/imt-2016-0025] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
AIM This meta-analysis aims at assessing the correlation of PD-L1 levels and response PD-1/PD-L1 inhibitors in advanced malignant melanoma. METHODS Eligible studies included those evaluating PD-1/PD-L1 inhibitors in advanced melanoma and correlating the outcomes to PD-L1 levels. RESULTS After preclusion of ineligible studies, 11 studies were included. For PD-L1 >1% patients versus PD-L1 <1% patients treated with PD-1/PD-L1 targeted agents, the OR of ORR was 2.81 (95% CI: 1.64-4.82; p = 0.0002); while for PD-L1 >5% patients versus PD-L1 <5% patients, it was 2.22 (95% CI: 1.71-2.87; p < 0.00001). CONCLUSION The current analysis indicates the value of PD-L1 positivity in predicting higher response from PD-1/PD-L1 agents. This molecular predictor - together with other predictors - may help further individualize treatment options for metastatic melanoma patients.
Collapse
Affiliation(s)
- Omar Abdel-Rahman
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
31
|
Haass NK, Gabrielli B. Cell cycle-tailored targeting of metastatic melanoma: Challenges and opportunities. Exp Dermatol 2017; 26:649-655. [PMID: 28109167 DOI: 10.1111/exd.13303] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2017] [Indexed: 12/21/2022]
Abstract
The advent of targeted therapies of metastatic melanoma, such as MAPK pathway inhibitors and immune checkpoint antagonists, has turned dermato-oncology from the "bad guy" to the "poster child" in oncology. Current targeted therapies are effective, although here is a clear need to develop combination therapies to delay the onset of resistance. Many antimelanoma drugs impact on the cell cycle but are also dependent on certain cell cycle phases resulting in cell cycle phase-specific drug insensitivity. Here, we raise the question: Have combination trials been abandoned prematurely as ineffective possibly only because drug scheduling was not optimized? Firstly, if both drugs of a combination hit targets in the same melanoma cell, cell cycle-mediated drug insensitivity should be taken into account when planning combination therapies, timing of dosing schedules and choice of drug therapies in solid tumors. Secondly, if the combination is designed to target different tumor cell subpopulations of a heterogeneous tumor, one drug effective in a particular subpopulation should not negatively impact on the other drug targeting another subpopulation. In addition to the role of cell cycle stage and progression on standard chemotherapeutics and targeted drugs, we discuss the utilization of cell cycle checkpoint control defects to enhance chemotherapeutic responses or as targets themselves. We propose that cell cycle-tailored targeting of metastatic melanoma could further improve therapy outcomes and that our real-time cell cycle imaging 3D melanoma spheroid model could be utilized as a tool to measure and design drug scheduling approaches.
Collapse
Affiliation(s)
- Nikolas K Haass
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, Qld, Australia.,The Centenary Institute, Newtown, NSW, Australia.,Discipline of Dermatology, University of Sydney, Sydney, NSW, Australia
| | - Brian Gabrielli
- Mater Medical Research Institute, Translational Research Institute, The University of Queensland, Brisbane, Qld, Australia
| |
Collapse
|
32
|
Zhao P, Atanackovic D, Dong S, Yagita H, He X, Chen M. An Anti-Programmed Death-1 Antibody (αPD-1) Fusion Protein That Self-Assembles into a Multivalent and Functional αPD-1 Nanoparticle. Mol Pharm 2017; 14:1494-1500. [PMID: 28343398 DOI: 10.1021/acs.molpharmaceut.6b01021] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer immune checkpoint therapy has achieved remarkable clinical successes in various cancers. However, current immune checkpoint inhibitors block the checkpoint of not only the immune cells that are important to cancer therapy but also the immune cells that are irrelevant to the therapy. Such an indiscriminate blockade limits the efficacy and causes the autoimmune toxicity of the therapy. It might be beneficial to use a carrier to target immune checkpoint inhibitors to cancer-reactive immune cells. Here, we explore a method to load the inhibitors into carriers. We used the anti-programmed death-1 antibody (αPD-1) as a model immune checkpoint inhibitor. First, we generated a recombinant single-chain variable fragment (scFv) of αPD-1. Then, we designed and generated a fusion protein consisting of the scFv and an amphiphilic immune-tolerant elastin-like polypeptide (iTEP). Because of the amphiphilic iTEP, the fusion was able to self-assemble into a nanoparticle (NP). The NP was proved to block the PD-1 immune checkpoint in vitro and in vivo. Particularly, the NP exacerbated diabetes development in nonobese diabetic mice as effectively as natural, intact αPD-1. In summary, we successfully expressed αPD-1 as a recombinant protein and linked αPD-1 to a NP, which lays a foundation to develop a delivery system to target αPD-1 to a subpopulation of immune cells.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Pharmaceutics and Pharmaceutical Chemistry, The University of Utah , Salt Lake City, Utah 84112, United States
| | - Djordje Atanackovic
- University of Utah School of Medicine , Salt Lake City, Utah 84112, United States
| | - Shuyun Dong
- Department of Pharmaceutics and Pharmaceutical Chemistry, The University of Utah , Salt Lake City, Utah 84112, United States
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine , Tokyo, Japan
| | - Xiao He
- Department of Pathology, The University of Utah , Salt Lake City, Utah 84112, United States
| | - Mingnan Chen
- Department of Pharmaceutics and Pharmaceutical Chemistry, The University of Utah , Salt Lake City, Utah 84112, United States
| |
Collapse
|
33
|
Abstract
Malignant melanoma of the skin is the most aggressive human cancer given that a primary tumor a few millimeters in diameter frequently has full metastatic competence. In view of that, revealing the genetic background of this potential may also help to better understand tumor dissemination in general. Genomic analyses have established the molecular classification of melanoma based on the most frequent driver oncogenic mutations (BRAF, NRAS, KIT) and have also revealed a long list of rare events, including mutations and amplifications as well as genetic microheterogeneity. At the moment, it is unclear whether any of these rare events have role in the metastasis initiation process since the major drivers do not have such a role. During lymphatic and hematogenous dissemination, the clonal selection process is evidently reflected by differences in oncogenic drivers in the metastases versus the primary tumor. Clonal selection is also evident during lymphatic progression, though the genetic background of this immunoselection is less clear. Genomic analyses of metastases identified further genetic alterations, some of which may correspond to metastasis maintenance genes. The natural genetic progression of melanoma can be modified by targeted (BRAF or MEK inhibitor) or immunotherapies. Some of the rare events in primary tumors may result in primary resistance, while further new genetic lesions develop during the acquired resistance to both targeted and immunotherapies. Only a few genetic lesions of the primary tumor are constant during natural or therapy-modulated progression. EGFR4 and NMDAR2 mutations, MITF and MET amplifications and PTEN loss can be considered as metastasis drivers. Furthermore, BRAF and MITF amplifications as well as PTEN loss are also responsible for resistance to targeted therapies, whereas NRAS mutation is the only founder genetic lesion showing any association with sensitivity to immunotherapies. Unfortunately, there are hardly any data on the possible organ-specific metastatic drivers in melanoma. These observations suggest that clinical management of melanoma patients must rely on the genetic analysis of the metastatic lesions to be able to monitor progression-associated changes and to personalize therapies.
Collapse
|
34
|
Rbah-Vidal L, Vidal A, Billaud EMF, Besse S, Ranchon-Cole I, Mishellany F, Perrot Y, Maigne L, Moins N, Guerquin-Kern JL, Degoul F, Chezal JM, Auzeloux P, Miot-Noirault E. Theranostic Approach for Metastatic Pigmented Melanoma Using ICF15002, a Multimodal Radiotracer for Both PET Imaging and Targeted Radionuclide Therapy. Neoplasia 2016; 19:17-27. [PMID: 27987437 PMCID: PMC5157796 DOI: 10.1016/j.neo.2016.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 11/01/2016] [Indexed: 01/05/2023] Open
Abstract
PURPOSE: This work reports, in melanoma models, the theranostic potential of ICF15002 as a single fluorinated and iodinated melanin-targeting compound. METHODS: Studies were conducted in the murine syngeneic B16BL6 model and in the A375 and SK-MEL-3 human xenografts. ICF15002 was radiolabeled with fluorine-18 for positron emission tomography (PET) imaging and biodistribution, with iodine-125 for metabolism study, and iodine-131 for targeted radionuclide therapy (TRT). TRT efficacy was assessed by tumor volume measurement, with mechanistics and dosimetry parameters being determined in the B16BL6 model. Intracellular localization of ICF15002 was characterized by secondary ion mass spectrometry (SIMS). RESULTS: PET imaging with [18F]ICF15002 evidenced tumoral uptake of 14.33 ± 2.11%ID/g and 4.87 ± 0.93%ID/g in pigmented B16BL6 and SK-MEL-3 models, respectively, at 1 hour post inoculation. No accumulation was observed in the unpigmented A375 melanoma. SIMS demonstrated colocalization of ICF15002 signal with melanin polymers in melanosomes of the B16BL6 tumors. TRT with two doses of 20 MBq [131I]ICF15002 delivered an absorbed dose of 102.3 Gy to B16BL6 tumors, leading to a significant tumor growth inhibition [doubling time (DT) of 2.9 ± 0.5 days in treated vs 1.8 ± 0.3 in controls] and a prolonged median survival (27 days vs 21 in controls). P53S15 phosphorylation and P21 induction were associated with a G2/M blockage, suggesting mitotic catastrophe. In the human SK-MEL-3 model, three doses of 25 MBq led also to a DT increase (26.5 ± 7.8 days vs 11.0 ± 3.8 in controls) and improved median survival (111 days vs 74 in controls). CONCLUSION: Results demonstrate that ICF15002 fulfills suitable properties for bimodal imaging/TRT management of patients with pigmented melanoma.
Collapse
Affiliation(s)
- Latifa Rbah-Vidal
- UMR 990 INSERM/Université d'Auvergne, F-63005 Clermont-Ferrand, France; UMR 892 INSERM/6299 CNRS/Université de Nantes, F-44007 Nantes, France
| | - Aurélien Vidal
- UMR 990 INSERM/Université d'Auvergne, F-63005 Clermont-Ferrand, France; Arronax, CS 10112, F-44817 Saint Herblain Cedex, France
| | | | - Sophie Besse
- UMR 990 INSERM/Université d'Auvergne, F-63005 Clermont-Ferrand, France
| | - Isabelle Ranchon-Cole
- UMR 1107 INSERM/Université d'Auvergne, Equipe Biophysique Neurosensorielle, F-63000 Clermont-Ferrand, France
| | - Florence Mishellany
- Centre Jean Perrin, Laboratoire d'anatomo-pathologie, F-63011 Clermont-Ferrand, France
| | - Yann Perrot
- CNRS/IN2P3/Université Blaise Pascal, Laboratoire de Physique Corpusculaire, F-63000 Clermont-Ferrand, France
| | - Lydia Maigne
- CNRS/IN2P3/Université Blaise Pascal, Laboratoire de Physique Corpusculaire, F-63000 Clermont-Ferrand, France
| | - Nicole Moins
- UMR 990 INSERM/Université d'Auvergne, F-63005 Clermont-Ferrand, France
| | | | - Françoise Degoul
- UMR 990 INSERM/Université d'Auvergne, F-63005 Clermont-Ferrand, France
| | | | - Philippe Auzeloux
- UMR 990 INSERM/Université d'Auvergne, F-63005 Clermont-Ferrand, France
| | | |
Collapse
|
35
|
Dunn J, Watson M, Aitken JF, Hyde MK. Systematic review of psychosocial outcomes for patients with advanced melanoma. Psychooncology 2016; 26:1722-1731. [PMID: 27696578 DOI: 10.1002/pon.4290] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 09/19/2016] [Accepted: 09/29/2016] [Indexed: 11/10/2022]
Abstract
BACKGROUND New advanced melanoma therapies are associated with improved survival; however, quality of survivorship, particularly psychosocial outcomes, for patients overall and those treated with newer therapies is unclear. OBJECTIVE Synthesize qualitative and quantitative evidence about psychosocial outcomes for advanced (stage III/IV) melanoma patients. METHODS Five databases were searched (01/01/1980 to 31/01/2016). Inclusion criteria were as follows: advanced melanoma patients or sub-group analysis; assessed psychosocial outcomes; and English language. RESULTS Fifty-two studies met review criteria (4 qualitative, 48 quantitative). Trials comprise mostly medical not psychosocial interventions, with psychosocial outcomes assessed within broader quality of life measures. Patients receiving chemotherapy or IFN-alpha showed decreased emotional and social function and increased distress. Five trials of newer therapies appeared to show improvements in emotional and social function. Descriptive studies suggest that patients with advanced, versus localized disease, had decreased emotional and social function and increased distress. Contributors to distress were largely unexplored, and no clear framework described coping/adjustment trajectories. Patients with advanced versus localized disease had more supportive care needs, particularly amount, quality, and timing of melanoma-related information, communication with and emotional support from clinicians. Limitations included: lack of theoretical underpinnings guiding study design; inconsistent measurement approaches; small sample sizes; non-representative sampling; and cross-sectional design. CONCLUSIONS Quality trial evidence is needed to clarify the impact of treatment innovations for advanced melanoma on patients' psychosocial well-being. Survivorship research and subsequent translation of that knowledge into programs and services currently lags behind gains in the medical treatment of advanced melanoma, a troubling circumstance that requires immediate and focused attention.
Collapse
Affiliation(s)
- Jeff Dunn
- Cancer Council Queensland, Brisbane, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia.,School of Social Science, The University of Queensland, St Lucia, Queensland, Australia.,Institute for Resilient Regions, University of Southern Queensland, Toowoomba, Australia
| | - Maggie Watson
- Pastoral and Psychological Care, Royal Marsden Hospital, Sutton, Surrey, UK
| | - Joanne F Aitken
- Cancer Council Queensland, Brisbane, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia.,Institute for Resilient Regions, University of Southern Queensland, Toowoomba, Australia.,School of Public Health and Social Work, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Melissa K Hyde
- Cancer Council Queensland, Brisbane, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
36
|
van Amerongen RA, Retèl VP, Coupé VM, Nederlof PM, Vogel MJ, van Harten WH. Next-generation sequencing in NSCLC and melanoma patients: a cost and budget impact analysis. Ecancermedicalscience 2016; 10:684. [PMID: 27899957 PMCID: PMC5102690 DOI: 10.3332/ecancer.2016.684] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Indexed: 02/06/2023] Open
Abstract
Next-generation sequencing (NGS) has reached the molecular diagnostic laboratories. Although the NGS technology aims to improve the effectiveness of therapies by selecting the most promising therapy, concerns are that NGS testing is expensive and that the ‘benefits’ are not yet in relation to these costs. In this study, we give an estimation of the costs and an institutional and national budget impact of various types of NGS tests in non-small-cell lung cancer (NSCLC) and melanoma patients within The Netherlands. First, an activity-based costing (ABC) analysis has been conducted on the costs of two examples of NGS panels (small- and medium-targeted gene panel (TGP)) based on data of The Netherlands Cancer Institute (NKI). Second, we performed a budget impact analysis (BIA) to estimate the current (2015) and future (2020) budget impact of NGS on molecular diagnostics for NSCLC and melanoma patients in The Netherlands. Literature, expert opinions, and a data set of patients within the NKI (n = 172) have been included in the BIA. Based on our analysis, we expect that the NGS test cost concerns will be limited. In the current situation, NGS can indeed result in higher diagnostic test costs, which is mainly related to required additional tests besides the small TGP. However, in the future, we expect that the use of whole-genome sequencing (WGS) will increase, for which it is expected that additional tests can be (partly) avoided. Although the current clinical benefits are expected to be limited, the research potentials of NGS are already an important advantage.
Collapse
Affiliation(s)
- Rosa A van Amerongen
- Department of Epidemiology and Biostatistics, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Valesca P Retèl
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, PO Box 90203, 1006 BE Amsterdam, The Netherlands; School of Governance and Management, University of Twente, MB-HTSR, PO Box 217, 7500 AE Enschede, The Netherlands
| | - Veerle Mh Coupé
- Department of Epidemiology and Biostatistics, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Petra M Nederlof
- Department of Molecular Diagnostics, Pathology, The Netherlands Cancer Institute, PO Box 90203, 1006 BE Amsterdam, The Netherlands
| | - Maartje J Vogel
- Department of Molecular Diagnostics, Pathology, The Netherlands Cancer Institute, PO Box 90203, 1006 BE Amsterdam, The Netherlands
| | - Wim H van Harten
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, PO Box 90203, 1006 BE Amsterdam, The Netherlands; School of Governance and Management, University of Twente, MB-HTSR, PO Box 217, 7500 AE Enschede, The Netherlands
| |
Collapse
|
37
|
Dany M, Nganga R, Chidiac A, Hanna E, Matar S, Elston D. Advances in immunotherapy for melanoma management. Hum Vaccin Immunother 2016; 12:2501-2511. [PMID: 27454404 PMCID: PMC5085014 DOI: 10.1080/21645515.2016.1190889] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/26/2016] [Accepted: 05/13/2016] [Indexed: 12/31/2022] Open
Abstract
Melanoma remains a leading cause of death among young adults. Evidence that melanoma tumor cells are highly immunogenic and a better understanding of T-cell immune checkpoints have changed the therapeutic approach to advanced melanoma. Instead of targeting the tumor directly, immunotherapy targets and activates the immune response using checkpoint inhibitors, monoclonal antibodies, vaccines, and adoptive T cell therapy. This review focuses on the immune signaling and biological mechanisms of action of recent immune-based melanoma therapies as well as their clinical benefits.
Collapse
Affiliation(s)
- Mohammed Dany
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Rose Nganga
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Alissar Chidiac
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Edith Hanna
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Sara Matar
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Dirk Elston
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
38
|
Catalani E, Proietti Serafini F, Zecchini S, Picchietti S, Fausto AM, Marcantoni E, Buonanno F, Ortenzi C, Perrotta C, Cervia D. Natural products from aquatic eukaryotic microorganisms for cancer therapy: Perspectives on anti-tumour properties of ciliate bioactive molecules. Pharmacol Res 2016; 113:409-420. [PMID: 27650755 DOI: 10.1016/j.phrs.2016.09.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/12/2016] [Accepted: 09/16/2016] [Indexed: 11/27/2022]
Abstract
Several modern drugs, including those for cancer therapy, have been isolated from natural sources, are based on natural products and its derivatives, or mime natural products. Some of them are in clinical use, others in clinical trials. The success of natural products in drug discovery is related to their biochemical characteristics and to the technologic methods used to study their feature. Natural compounds may acts as chemo-preventive agents and as factors that increase therapeutic efficacy of existing drugs, thus overcoming cancer cell drug resistance that is the main factor determining the failure in conventional chemotherapy. Water environment, because of its physical and chemical conditions, shows an extraordinary collection of natural biological substances with an extensive structural and functional diversity. The isolation of bioactive molecules has been reported from a great variety of aquatic organisms; however, the therapeutic application of molecules from eukaryotic microorganisms remains inadequately investigated and underexploited on a systematic basis. Herein we describe the biological activities in mammalian cells of selected substances isolated from ciliates, free-living protozoa common almost everywhere there is water, focusing on their anti-tumour actions and their possible therapeutic activity. In particular, we unveil the cellular and molecular machine mediating the effects of cell type-specific signalling protein pheromone Er-1 and secondary metabolites, i.e. euplotin C and climacostol, in cancer cells. To support the feasibility of climacostol-based approaches, we also present novel findings and report additional mechanisms of action using both in vitro and in vivo models of mouse melanomas, with the scope of highlighting new frontiers that can be explored also in a therapeutic perspective. The high skeletal chemical difference of ciliate compounds, their sustainability and availability, also through the use of new organic synthesis/modifications processes, and the results obtained so far in biological studies provide a rationale to consider some of them a potential resource for the design of new anti-cancer drugs.
Collapse
Affiliation(s)
- Elisabetta Catalani
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - Francesca Proietti Serafini
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - Silvia Zecchini
- Unit of Clinical Pharmacology, University Hospital "Luigi Sacco"-ASST Fatebenefratelli Sacco, Milano, Italy
| | - Simona Picchietti
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - Anna Maria Fausto
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - Enrico Marcantoni
- School of Sciences and Technologies, Section of Chemistry, Università degli Studi di Camerino, Italy
| | - Federico Buonanno
- Laboratory of Protistology and Biology Education, Department of Education, Cultural Heritage and Tourism, Università degli Studi di Macerata, Italy
| | - Claudio Ortenzi
- Laboratory of Protistology and Biology Education, Department of Education, Cultural Heritage and Tourism, Università degli Studi di Macerata, Italy
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Italy.
| | - Davide Cervia
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy; Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Italy.
| |
Collapse
|
39
|
Abdel-Rahman O. Combination or single-agent ipilimumab as immunotherapy of advanced melanoma: a critical review. Melanoma Manag 2016; 3:231-243. [PMID: 30190892 DOI: 10.2217/mmt-2016-0011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Accepted: 06/16/2016] [Indexed: 01/08/2023] Open
Abstract
Aim A pooled analysis of the efficacy and toxicity of combination immunotherapy versus single-agent ipilimumab in the management of advanced melanoma has been conducted. Methodology Eligible studies included randomized controlled studies evaluating ipilimumab-based doublet immunotherapy versus ipilimumab monotherapy for the management of unresectable melanoma. Results Nivolumab/ipilimumab combination strategy is associated with a significant improvement in objective response rate (odds ratio: 7.38; 95% CI: 3.71-14.67; p < 0.00001) and progression-free survival (0.42; 95% CI: 0.34-0.52; p < 0.00001) as well as a higher relative risk for high-grade elevated alanine aminotransferase (5.58; 95% CI: 2.28-13.67; p = 0.0002). Conclusion This analysis demonstrated that nivolumab/ipilimumab combination is associated with a higher objective response rate and progression-free survival in the management of advanced melanoma.
Collapse
Affiliation(s)
- Omar Abdel-Rahman
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Lotfy Elsayed Street, Cairo 11665, Egypt
| |
Collapse
|
40
|
Schwartz H, Blacher E, Amer M, Livneh N, Abramovitz L, Klein A, Ben-Shushan D, Soffer S, Blazquez R, Barrantes-Freer A, Müller M, Müller-Decker K, Stein R, Tsarfaty G, Satchi-Fainaro R, Umansky V, Pukrop T, Erez N. Incipient Melanoma Brain Metastases Instigate Astrogliosis and Neuroinflammation. Cancer Res 2016; 76:4359-71. [PMID: 27261506 DOI: 10.1158/0008-5472.can-16-0485] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/02/2016] [Indexed: 11/16/2022]
Abstract
Malignant melanoma is the deadliest of skin cancers. Melanoma frequently metastasizes to the brain, resulting in dismal survival. Nevertheless, mechanisms that govern early metastatic growth and the interactions of disseminated metastatic cells with the brain microenvironment are largely unknown. To study the hallmarks of brain metastatic niche formation, we established a transplantable model of spontaneous melanoma brain metastasis in immunocompetent mice and developed molecular tools for quantitative detection of brain micrometastases. Here we demonstrate that micrometastases are associated with instigation of astrogliosis, neuroinflammation, and hyperpermeability of the blood-brain barrier. Furthermore, we show a functional role for astrocytes in facilitating initial growth of melanoma cells. Our findings suggest that astrogliosis, physiologically instigated as a brain tissue damage response, is hijacked by tumor cells to support metastatic growth. Studying spontaneous melanoma brain metastasis in a clinically relevant setting is the key to developing therapeutic approaches that may prevent brain metastatic relapse. Cancer Res; 76(15); 4359-71. ©2016 AACR.
Collapse
Affiliation(s)
- Hila Schwartz
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eran Blacher
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Malak Amer
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nir Livneh
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lilach Abramovitz
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Anat Klein
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel. Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Dikla Ben-Shushan
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shelly Soffer
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Raquel Blazquez
- Department of Internal Medicine III, Hematology and Medical Oncology, University Hospital Regensburg, Regensburg, Germany
| | | | - Meike Müller
- Tumor Models Unit, German Cancer Research Center, Heidelberg, Germany
| | | | - Reuven Stein
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Galia Tsarfaty
- Department of Diagnostic Imaging, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Tobias Pukrop
- Department of Internal Medicine III, Hematology and Medical Oncology, University Hospital Regensburg, Regensburg, Germany. Department of Hematology/Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Neta Erez
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
41
|
Climacostol reduces tumour progression in a mouse model of melanoma via the p53-dependent intrinsic apoptotic programme. Sci Rep 2016; 6:27281. [PMID: 27271364 PMCID: PMC4895139 DOI: 10.1038/srep27281] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 05/17/2016] [Indexed: 12/12/2022] Open
Abstract
Climacostol, a compound produced by the ciliated protozoan Climacostomum virens, displayed cytotoxic properties in vitro. This study demonstrates that it has anti-tumour potential. Climacostol caused a reduction of viability/proliferation of B16-F10 mouse melanoma cells, a rapidly occurring DNA damage, and induced the intrinsic apoptotic pathway characterised by the dissipation of the mitochondrial membrane potential, the translocation of Bax to the mitochondria, the release of Cytochrome c from the mitochondria, and the activation of Caspase 9-dependent cleavage of Caspase 3. The apoptotic mechanism of climacostol was found to rely on the up-regulation of p53 and its targets Noxa and Puma. In vivo analysis of B16-F10 allografts revealed a persistent inhibition of tumour growth rate when melanomas were treated with intra-tumoural injections of climacostol. In addition, it significantly improved the survival of transplanted mice, decreased tumour weight, induced a remarkable reduction of viable cells inside the tumour, activated apoptosis and up-regulated the p53 signalling network. Importantly, climacostol toxicity was more selective against tumour than non-tumour cells. The anti-tumour properties of climacostol and the molecular events associated with its action indicate that it is a powerful agent that may be considered for the design of pro-apoptotic drugs for melanoma therapy.
Collapse
|
42
|
Whiteman DC, Green AC, Olsen CM. The Growing Burden of Invasive Melanoma: Projections of Incidence Rates and Numbers of New Cases in Six Susceptible Populations through 2031. J Invest Dermatol 2016; 136:1161-1171. [PMID: 26902923 DOI: 10.1016/j.jid.2016.01.035] [Citation(s) in RCA: 417] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 01/13/2016] [Accepted: 01/28/2016] [Indexed: 10/22/2022]
Abstract
New melanoma therapies are being developed rapidly, complementing prevention and detection strategies for disease control. Estimating the future burden of melanoma is necessary for deciding how best to deploy limited resources to achieve effective melanoma control. Using three decades of cancer registry data (1982-2011) from six populations with moderate to high melanoma incidence (US whites and the populations of the United Kingdom, Sweden, Norway, Australia, New Zealand), we applied age-period-cohort models to describe current trends and project future incidence rates and numbers of melanomas out to 2031. Between 1982 and 2011, melanoma rates in US whites, and the populations of the United Kingdom, Sweden, and Norway increased at more than 3% annually and are projected to continue rising until at least 2022. Melanoma incidence in Australia has been declining since 2005 (-0.7% per year), and melanoma incidence in New Zealand is increasing but is projected to decline soon. The numbers of new melanoma cases will rise in all six populations because of aging populations and high age-specific rates in the elderly. In US whites, annual new cases will rise from around 70,000 in 2007-2011 to 116,000 in 2026-2031, with 79% of the increase attributable to rising age-specific rates and 21% to population growth and aging. The continued increases in case numbers in all six populations through 2031 will increase the challenges of melanoma control.
Collapse
Affiliation(s)
- David C Whiteman
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia; The University of Queensland, School of Public Health, Herston Road, Herston, QLD 4006, Australia.
| | - Adele C Green
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia; The University of Queensland, School of Public Health, Herston Road, Herston, QLD 4006, Australia; Cancer Research UK Manchester Institute and Institute of Inflammation and Repair, University of Manchester, Manchester, UK
| | - Catherine M Olsen
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia; The University of Queensland, School of Public Health, Herston Road, Herston, QLD 4006, Australia
| |
Collapse
|
43
|
Middleton MR, Dalle S, Claveau J, Mut P, Hallmeyer S, Plantin P, Highley M, Kotapati S, Le TK, Brokaw J, Abernethy AP. Real-world treatment practice in patients with advanced melanoma in the era before ipilimumab: results from the IMAGE study. Cancer Med 2016; 5:1436-43. [PMID: 27118102 PMCID: PMC4944869 DOI: 10.1002/cam4.717] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 03/04/2016] [Accepted: 03/07/2016] [Indexed: 11/21/2022] Open
Abstract
The therapeutic landscape for advanced melanoma has recently been transformed by several novel agents (immune checkpoint inhibitors and molecular‐targeted agents). The prospective, multi‐site, observational study IMAGE (ipilimumab: management of advanced melanoma in real practice) included a retrospective cohort to describe real‐world treatment prior to approval of the immune checkpoint inhibitor ipilimumab. This retrospective cohort of patients, who started second‐line/subsequent treatment (index therapy) for advanced melanoma within 3 years before ipilimumab approval, was selected randomly by chart review. Collected data included treatment history, patient outcomes, and healthcare resource utilization. All patients had ≥1 year of follow‐up data. This analysis included 177 patients from Europe (69%) and North America (31%). The most common index therapies (used alone or in combination) were fotemustine (23%), dacarbazine (21%), temozolomide (14%), and platinum‐based chemotherapy (14%). Most patients (89%) discontinued index treatment during the study period; the most common reason was disease progression (59%). Among patients with tumor assessment (153/177; 86%), 2% had complete response, 5% had partial response, and 12% had stable disease on last tumor assessment. At 1‐year study follow‐up, median progression‐free survival was 2.6 months (95% confidence interval [CI], 2.1–2.9) and median overall survival was 8.8 months (95% CI, 6.5–9.7). During follow‐up, 95% of the patients had healthcare visits for advanced melanoma, 74% of whom were hospitalized or admitted to a hospice facility. These results provide insights into patient care with advanced melanoma in the era before ipilimumab and may serve as a benchmark for new agents in future real‐world studies.
Collapse
Affiliation(s)
- Mark R Middleton
- National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom
| | | | - Joel Claveau
- Centre Hospitalier Universitaire de Québec, Quebec City, Canada
| | - Pilar Mut
- Hospital Son Llatzer, Illes Balears, Spain
| | | | | | - Martin Highley
- Plymouth Oncology Centre, Derriford Hospital, Plymouth, United Kingdom
| | | | | | | | | |
Collapse
|
44
|
Feng JH, Nakagawa-Goto K, Lee KH, Shyur LF. A Novel Plant Sesquiterpene Lactone Derivative, DETD-35, Suppresses BRAFV600E Mutant Melanoma Growth and Overcomes Acquired Vemurafenib Resistance in Mice. Mol Cancer Ther 2016; 15:1163-76. [PMID: 27048951 DOI: 10.1158/1535-7163.mct-15-0973] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/30/2016] [Indexed: 12/22/2022]
Abstract
Acquired resistance to vemurafenib develops through reactivation of RAF/MEK/ERK signaling or bypass mechanisms. Recent combination therapies such as a MEK inhibitor combined with vemurafenib show improvement in major clinical end points, but the percentage of patients with adverse toxic events is higher than with vemurafenib monotherapy and most patients ultimately relapse. Therefore, there is an urgent need to develop new antimelanoma drugs and/or adjuvant agents for vemurafenib therapy. In this study, we created a novel semiorganically modified derivative, DETD-35, from deoxyelephantopin (DET), a plant sesquiterpene lactone demonstrated as an anti-inflammatory and anti-mammary tumor agent. Our results show that DETD-35 inhibited proliferation of a panel of melanoma cell lines, including acquired vemurafenib resistance A375 cells (A375-R) established in this study, with superior activities to DET and no cytotoxicity to normal melanocytes. DETD-35 suppressed tumor growth and reduced tumor mass as effectively as vemurafenib in A375 xenograft study. Furthermore, DETD-35 also reduced tumor growth in both acquired (A375-R) and intrinsic (A2058) vemurafenib resistance xenograft models, where vemurafenib showed no antitumor activity. Notably, the combination of DETD-35 and vemurafenib exhibited the most significant effects in both in vitro and in vivo xenograft studies due to synergism of the compound and the drug. Mechanistic studies suggested that DETD-35 overcame acquired vemurafenib resistance at least in part through deregulating MEK-ERK, Akt, and STAT3 signaling pathways and promoting apoptosis of cancer cells. Overall, our results suggest that DETD-35 may be useful as a therapeutic or adjuvant agent against BRAF(V600E) mutant and acquired vemurafenib resistance melanoma. Mol Cancer Ther; 15(6); 1163-76. ©2016 AACR.
Collapse
Affiliation(s)
- Jia-Hua Feng
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan. Institute of Plant Biology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Kyoko Nakagawa-Goto
- College of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan. Natural Products Research Laboratories, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina
| | - Kuo-Hsiung Lee
- Natural Products Research Laboratories, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina
| | - Lie-Fen Shyur
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan. PhD Program in Translational Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
45
|
Voss RK, Woods TN, Cromwell KD, Nelson KC, Cormier JN. Improving outcomes in patients with melanoma: strategies to ensure an early diagnosis. PATIENT-RELATED OUTCOME MEASURES 2015; 6:229-42. [PMID: 26609248 PMCID: PMC4644158 DOI: 10.2147/prom.s69351] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Patients with thin, low-risk melanomas have an excellent long-term prognosis and higher quality of life than those who are diagnosed at later stages. From an economic standpoint, treatment of early stage melanoma consumes a fraction of the health care resources needed to treat advanced disease. Consequently, early diagnosis of melanoma is in the best interest of patients, payers, and health care systems. This review describes strategies to ensure that patients receive an early diagnosis through interventions ranging from better utilization of primary care clinics, to in vivo diagnostic technologies, to new "apps" available in the market. Strategies for screening those at high risk due to age, male sex, skin type, nevi, genetic mutations, or family history are discussed. Despite progress in identifying those at high risk for melanoma, there remains a lack of general consensus worldwide for best screening practices. Strategies to ensure early diagnosis of recurrent disease in those with a prior melanoma diagnosis are also reviewed. Variations in recurrence surveillance practices by type of provider and country are featured, with evidence demonstrating that various imaging studies, including ultrasound, computed tomography, positron emission tomography, and magnetic resonance imaging, provide only minimal gains in life expectancy, even for those with more advanced (stage III) disease. Because the majority of melanomas are attributable to ultraviolet radiation in the form of sunlight, primary prevention strategies, including sunscreen use and behavioral interventions, are reviewed. Recent international government regulation of tanning beds is described, as well as issues surrounding the continued use artificial ultraviolet sources among youth. Health care stakeholder strategies to minimize UV exposure are summarized. The recommendations encompass both specific behaviors and broad intervention targets (eg, individuals, social spheres, organizations, celebrities, governments).
Collapse
Affiliation(s)
- Rachel K Voss
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tessa N Woods
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kate D Cromwell
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kelly C Nelson
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Janice N Cormier
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
46
|
Doublet BRAF/MEK inhibition versus single-agent BRAF inhibition in the management of BRAF-mutant advanced melanoma, biological rationale and meta-analysis of published data. Clin Transl Oncol 2015; 18:848-58. [DOI: 10.1007/s12094-015-1438-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 10/19/2015] [Indexed: 02/03/2023]
|
47
|
Stavraka C, Blagden S. The La-Related Proteins, a Family with Connections to Cancer. Biomolecules 2015; 5:2701-22. [PMID: 26501340 PMCID: PMC4693254 DOI: 10.3390/biom5042701] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/21/2015] [Accepted: 10/07/2015] [Indexed: 01/09/2023] Open
Abstract
The evolutionarily-conserved La-related protein (LARP) family currently comprises Genuine La, LARP1, LARP1b, LARP4, LARP4b, LARP6 and LARP7. Emerging evidence suggests each LARP has a distinct role in transcription and/or mRNA translation that is attributable to subtle sequence variations within their La modules and specific C-terminal domains. As emerging research uncovers the function of each LARP, it is evident that La, LARP1, LARP6, LARP7 and possibly LARP4a and 4b are dysregulated in cancer. Of these, LARP1 is the first to be demonstrated to drive oncogenesis. Here, we review the role of each LARP and the evidence linking it to malignancy. We discuss a future strategy of targeting members of this protein family as cancer therapy.
Collapse
Affiliation(s)
- Chara Stavraka
- Ovarian Cancer Research Centre, Institute for Reproductive and Developmental Biology, Imperial College, Du Cane Road, London W12 0HS, UK.
| | - Sarah Blagden
- Ovarian Cancer Research Centre, Institute for Reproductive and Developmental Biology, Imperial College, Du Cane Road, London W12 0HS, UK.
- Department of Oncology, University of Oxford, Churchill Hospital, Old Road, Oxford OX3 7LE, UK.
| |
Collapse
|