1
|
Comes MC, Fucci L, Mele F, Bove S, Cristofaro C, De Risi I, Fanizzi A, Milella M, Strippoli S, Zito A, Guida M, Massafra R. A deep learning model based on whole slide images to predict disease-free survival in cutaneous melanoma patients. Sci Rep 2022; 12:20366. [PMID: 36437296 PMCID: PMC9701687 DOI: 10.1038/s41598-022-24315-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/14/2022] [Indexed: 11/28/2022] Open
Abstract
The application of deep learning on whole-slide histological images (WSIs) can reveal insights for clinical and basic tumor science investigations. Finding quantitative imaging biomarkers from WSIs directly for the prediction of disease-free survival (DFS) in stage I-III melanoma patients is crucial to optimize patient management. In this study, we designed a deep learning-based model with the aim of learning prognostic biomarkers from WSIs to predict 1-year DFS in cutaneous melanoma patients. First, WSIs referred to a cohort of 43 patients (31 DF cases, 12 non-DF cases) from the Clinical Proteomic Tumor Analysis Consortium Cutaneous Melanoma (CPTAC-CM) public database were firstly annotated by our expert pathologists and then automatically split into crops, which were later employed to train and validate the proposed model using a fivefold cross-validation scheme for 5 rounds. Then, the model was further validated on WSIs related to an independent test, i.e. a validation cohort of 11 melanoma patients (8 DF cases, 3 non-DF cases), whose data were collected from Istituto Tumori 'Giovanni Paolo II' in Bari, Italy. The quantitative imaging biomarkers extracted by the proposed model showed prognostic power, achieving a median AUC value of 69.5% and a median accuracy of 72.7% on the public cohort of patients. These results remained comparable on the validation cohort of patients with an AUC value of 66.7% and an accuracy value of 72.7%, respectively. This work is contributing to the recently undertaken investigation on how treat features extracted from raw WSIs to fulfil prognostic tasks involving melanoma patients. The promising results make this study as a valuable basis for future research investigation on wider cohorts of patients referred to our Institute.
Collapse
Affiliation(s)
- Maria Colomba Comes
- I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Livia Fucci
- I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Fabio Mele
- I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Samantha Bove
- I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Cristian Cristofaro
- I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Ivana De Risi
- I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Annarita Fanizzi
- I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Martina Milella
- I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Sabino Strippoli
- I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Alfredo Zito
- I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Michele Guida
- I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Raffaella Massafra
- I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
| |
Collapse
|
2
|
Duwa R, Jeong JH, Yook S. Immunotherapeutic strategies for the treatment of ovarian cancer: current status and future direction. J IND ENG CHEM 2021. [DOI: 10.1016/j.jiec.2020.11.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
3
|
Dendritic Cells Loaded with Heat Shock-Conditioned Ovarian Epithelial Carcinoma Cell Lysates Elicit T Cell-Dependent Antitumor Immune Responses In Vitro. J Immunol Res 2019; 2019:9631515. [PMID: 31886313 PMCID: PMC6899292 DOI: 10.1155/2019/9631515] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 09/08/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022] Open
Abstract
Ovarian epithelial carcinoma (OEC) is the most frequent ovarian tumor, characterized by a high mortality in advanced stages where conventional therapies are not effective. Based on the role of the immune system in the progression of this disease, immunotherapy using checkpoint blockade has been considered as a therapeutic alternative. Nevertheless, its results do not match up to the positive results in entities like melanoma and other malignancies, suggesting the need to find other therapies to be used alone or in combination. Dendritic cell- (DC-) based vaccines have shown promising results in several types of cancer, such as melanoma, prostate, and lung cancers, due to the essential role played by DCs in the activation of specific T cells, thus using other ways of activating the immune response than immune checkpoint blockade. During the last decade, we have used DC-based vaccines loaded with an allogeneic heat shock-conditioned melanoma cell lysate in the treatment of advanced stage patients in a series of clinical trials. In these studies, 60% of treated patients showed immunological responses which correlated positively with improved survival. Considering the relevance of ovarian cancer and the promising results of our DC-based vaccine, we show here that heat shock-conditioned cell lysates derived from ovarian epithelial carcinoma cell lines have the potential to induce the phenotypic and functional maturation of human DC, which in turn, is able to induce an efficient CD4+ and CD8+ T cell-mediated immune responses against ovarian cancer cell lines in vitro. In summary, OEC heat shock-conditioned cell lysate-loaded DCs may be considered for future combined immunotherapy approaches against ovarian tumors.
Collapse
|
4
|
Lee HM, Okuda KS, González FE, Patel V. Current Perspectives on Nasopharyngeal Carcinoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1164:11-34. [PMID: 31576537 DOI: 10.1007/978-3-030-22254-3_2] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Of the ~129,079 new cases of nasopharyngeal carcinoma (NPC) and 72,987 associated deaths estimated for 2018, the majority will be geographically localized to South East Asia, and likely to show an upward trend annually. It is thought that disparities in dietary habits, lifestyle, and exposures to harmful environmental factors are likely the root cause of NPC incidence rates to differ geographically. Genetic differences due to ethnicity and the Epstein Barr virus (EBV) are likely contributing factors. Pertinently, NPC is associated with poor prognosis which is largely attributed to lack of awareness of the salient symptoms of NPC. These include nose hemorrhage and headaches and coupled with detection and the limited therapeutic options. Treatment options include radiotherapy or chemotherapy or combination of both. Surgical excision is generally the last option considered for advanced and metastatic disease, given the close proximity of nasopharynx to brain stem cell area, major blood vessels, and nerves. To improve outcome of NPC patients, novel cellular and in vivo systems are needed to allow an understanding of the underling molecular events causal for NPC pathogenesis and for identifying novel therapeutic targets and effective therapies. While challenges and gaps in current NPC research are noted, some advances in targeted therapies and immunotherapies targeting EBV NPCs are discussed in this chapter, which may offer improvements in outcome of NPC patients.
Collapse
Affiliation(s)
- Hui Mei Lee
- Cancer Research Malaysia, Subang Jaya, Selangor, Malaysia
| | - Kazuhida Shaun Okuda
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Fermín E González
- Laboratory of Experimental Immunology and Cancer, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Vyomesh Patel
- Cancer Research Malaysia, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
5
|
Szaryńska M, Olejniczak A, Kobiela J, Łaski D, Śledziński Z, Kmieć Z. Cancer stem cells as targets for DC-based immunotherapy of colorectal cancer. Sci Rep 2018; 8:12042. [PMID: 30104575 PMCID: PMC6089981 DOI: 10.1038/s41598-018-30525-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/01/2018] [Indexed: 12/11/2022] Open
Abstract
The therapy of colorectal cancer (CRC) patients is often unsuccessful because of the presence of cancer stem cells (CSCs) resistant to conventional approaches. Dendritic cells (DC)-based protocols are believed to effectively supplement CRC therapy. Our study was aimed to assess how the number and properties of CSCs isolated from tumor tissue of CRC patients will affect the biological characteristics of in vitro modified DCs. Similar procedures were conducted with the using of CRC HCT116 and HT29 cell lines. We found that the detailed configuration of CSC-like markers significantly influenced the maturation and activation of DCs after stimulation with cancer cells lysates or culture supernatants. This basic stimulatory effect was enhanced by LPS that is normally present in CRC CSCs niche. The increased number of CD29+ and CD44+ CSCs presented the opposite impact on treated DCs as showed by many significant correlations. The CD133+ CSCs seemed to impair the functions of DCs. The more CD133+ CSCs in tumor sample the lower number of activated DCs evidenced after stimulation. Moreover, our results showed superiority of the spherical culture model over the adherent one since spherical HCT116 and HT29 cells presented similar influence on DCs properties as CRC patients cancer cells. We concluded that the DCs features may depend directly on the properties of CSCs affected by progression status of tumor.
Collapse
Affiliation(s)
- Magdalena Szaryńska
- Department of Histology, Medical University of Gdansk, 80-210, Gdansk, Poland.
| | - Agata Olejniczak
- Department of Histology, Medical University of Gdansk, 80-210, Gdansk, Poland
| | - Jarosław Kobiela
- Department of General, Endocrine and Transplant Surgery, Medical University of Gdansk, 80-214, Gdansk, Poland
| | - Dariusz Łaski
- Department of General, Endocrine and Transplant Surgery, Medical University of Gdansk, 80-214, Gdansk, Poland
| | - Zbigniew Śledziński
- Department of General, Endocrine and Transplant Surgery, Medical University of Gdansk, 80-214, Gdansk, Poland
| | - Zbigniew Kmieć
- Department of Histology, Medical University of Gdansk, 80-210, Gdansk, Poland
| |
Collapse
|
6
|
Rojas-Sepúlveda D, Tittarelli A, Gleisner MA, Ávalos I, Pereda C, Gallegos I, González FE, López MN, Butte JM, Roa JC, Fluxá P, Salazar-Onfray F. Tumor lysate-based vaccines: on the road to immunotherapy for gallbladder cancer. Cancer Immunol Immunother 2018; 67:1897-1910. [PMID: 29600445 PMCID: PMC6244977 DOI: 10.1007/s00262-018-2157-5] [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: 09/01/2017] [Accepted: 03/26/2018] [Indexed: 12/14/2022]
Abstract
Immunotherapy based on checkpoint blockers has proven survival benefits in patients with melanoma and other malignancies. Nevertheless, a significant proportion of treated patients remains refractory, suggesting that in combination with active immunizations, such as cancer vaccines, they could be helpful to improve response rates. During the last decade, we have used dendritic cell (DC) based vaccines where DCs loaded with an allogeneic heat-conditioned melanoma cell lysate were tested in a series of clinical trials. In these studies, 60% of stage IV melanoma DC-treated patients showed immunological responses correlating with improved survival. Further studies showed that an essential part of the clinical efficacy was associated with the use of conditioned lysates. Gallbladder cancer (GBC) is a high-incidence malignancy in South America. Here, we evaluated the feasibility of producing effective DCs using heat-conditioned cell lysates derived from gallbladder cancer cell lines (GBCCL). By characterizing nine different GBCCLs and several fresh tumor tissues, we found that they expressed some tumor-associated antigens such as CEA, MUC-1, CA19-9, Erb2, Survivin, and several carcinoembryonic antigens. Moreover, heat-shock treatment of GBCCLs induced calreticulin translocation and release of HMGB1 and ATP, both known to act as danger signals. Monocytes stimulated with combinations of conditioned lysates exhibited a potent increase of DC-maturation markers. Furthermore, conditioned lysate-matured DCs were capable of strongly inducing CD4+ and CD8+ T cell activation, in both allogeneic and autologous cell co-cultures. Finally, in vitro stimulated CD8+ T cells recognize HLA-matched GBCCLs. In summary, GBC cell lysate-loaded DCs may be considered for future immunotherapy approaches.
Collapse
Affiliation(s)
- Daniel Rojas-Sepúlveda
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile.,Faculty of Science, Universidad San Sebastián, Lota 2465, 7510157, Santiago, Chile
| | - Andrés Tittarelli
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - María Alejandra Gleisner
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Ignacio Ávalos
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Cristián Pereda
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Iván Gallegos
- Pathological Anatomy Service, Clinic Hospital, Universidad de Chile, 8380456, Santiago, Chile
| | - Fermín Eduardo González
- Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile.,Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, 8380492, Santiago, Chile
| | - Mercedes Natalia López
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Jean Michel Butte
- Department of Surgery, Fundación Arturo López Pérez, Institute of Oncology, 7500921, Santiago, Chile
| | - Juan Carlos Roa
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, 8330023, Santiago, Chile.,Center for Investigation in Translational Oncology (CITO), Advanced Center for Chronic Diseases (ACCDiS), School of Medicine, Pontificia Universidad Católica de Chile, 8330023, Santiago, Chile
| | - Paula Fluxá
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile. .,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile.
| |
Collapse
|
7
|
Thyagarajan A, Saylae J, Sahu RP. Acetylsalicylic acid inhibits the growth of melanoma tumors via SOX2-dependent-PAF-R-independent signaling pathway. Oncotarget 2017; 8:49959-49972. [PMID: 28636992 PMCID: PMC5564820 DOI: 10.18632/oncotarget.18326] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/06/2017] [Indexed: 12/17/2022] Open
Abstract
Acquired resistance to standard therapies remains a serious challenge, requiring novel therapeutic approaches that incorporate potential factors involved in tumor resistance. As cancers including melanoma express inflammatory cyclooxygenases generating prostaglandins implicated in tumor growth, we investigated mechanism of anti-inflammatory drug, acetylsalicylic acid (ASA) which has been shown to inhibit various tumor types, however, its effects against highly aggressive melanoma model are unclear. Given our reports that an activation of platelet-activating factor-receptor (PAF-R) augments the growth and impede efficacies of therapeutic agents in experimental melanoma, we also sought to determine if PAF-R mediates anti-melanoma activity of ASA. The current studies using stably PAF-R-positive (B16-PAFR) and negative (B16-MSCV) murine melanoma cells and PAF-R-expressing and deficient mice, demonstrate that ASA inhibits the in-vitro and in-vivo growth of highly aggressive B16F10 melanoma via bypassing tumoral or stromal PAF-R signaling. Similar ASA-induced effects in-vitro were seen in human melanoma and nasopharyngeal carcinoma cells positive or negative in PAF-R. Mechanistically, the ASA-induced decrease in cell survival and increase in apoptosis were significantly blocked by prostaglandin F2 alpha (PGF2α) agonists. Importantly, PCR array and qRT-PCR analysis of B16-tumors revealed significant downregulation of sry-related high-mobility-box-2 (SOX2) oncogene by ASA treatment. Interestingly, modulation of SOX2 expression by PGF2α agonists and upregulation by fibroblast growth factor 1 (FGF-1) rescued melanoma cells from ASA-induced decreased survival and increased apoptosis. Moreover, PGF2α-receptor antagonist, AL8810 mimics ASA-induced decreased melanoma cells survival which was significantly blocked by PGF2α and FGF-1. These findings indicate that ASA inhibits the growth of aggressive melanoma via SOX2-dependent-PAF-R-indepedent pathway.
Collapse
Affiliation(s)
- Anita Thyagarajan
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH, USA
| | - Jeremiah Saylae
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH, USA
| | - Ravi P. Sahu
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH, USA
| |
Collapse
|
8
|
Pinto R, Strippoli S, De Summa S, Albano A, Azzariti A, Guida G, Popescu O, Lorusso V, Guida M, Tommasi S. MicroRNA expression in BRAF-mutated and wild-type metastatic melanoma and its correlation with response duration to BRAF inhibitors. Expert Opin Ther Targets 2017; 19:1027-35. [PMID: 26156293 DOI: 10.1517/14728222.2015.1065818] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Objective: Currently, the treatment of BRAF V600-mutated metastatic melanoma with BRAF inhibitors gives a response rate of ~ 50% with a progression-free survival of ~ 6 -- 7 months. In order to identify predictive biomarkers capable of stratifying BRAF-mutated patients at high risk of shorter response duration to anti-BRAF therapy, the authors analyzed the expression of 15 microRNAs (miRNAs) targeting crucial genes involved in melanoma biology and drug response.Research design and methods: A total of 15 miRNAs and target gene expression were investigated in 43 patients (30 BRAF-mutated, and 13 BRAF wild-type). Moreover, 20 BRAF-mutated patients treated with vemurafenib were analyzed for miRNA expression in respect to time-to-progression.Results: All miRNAs except miR-192 showed low expression in BRAF-mutated as compared with BRAF wild-type patients. In particular, miR-101, miR-221,miR-21, miR-338-3p and miR-191 resulted in significant downregulation inBRAF-mutated patients. Moreover, high expression of miR-192 and miR-193b* and low expression of miR-132 resulted in significant association with shorter progression.Conclusion: Three miRNAs were significantly associated with clinical outcome in metastatic melanoma patients. An increased understanding of the molecular assessment of BRAF-mutated melanomas could allow development of specific molecular tests able to predict response duration.
Collapse
Affiliation(s)
- Rosamaria Pinto
- 1IRCCS Istituto Tumori "Giovanni Paolo II", Molecular Genetics Laboratory , Bari , Italy +0039 0805555283 ;
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Svedman FC, Pillas D, Taylor A, Kaur M, Linder R, Hansson J. Stage-specific survival and recurrence in patients with cutaneous malignant melanoma in Europe - a systematic review of the literature. Clin Epidemiol 2016; 8:109-22. [PMID: 27307765 PMCID: PMC4887072 DOI: 10.2147/clep.s99021] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Given the increasing incidence in cutaneous malignant melanoma (CMM) and the recent changes in the treatment landscape, it is important to understand stage-specific overall and recurrence-free survival patterns in Europe. Despite publications such as EUROCARE-5, there is limited information on stage-specific survival for CMM in Europe. METHOD We carried out a systematic literature review to provide an up-to-date summary of stage-specific survival and recurrence-free survival patterns in patients with CMM in Europe. Studies were included if they were published in Medline during the past 12 years and included information on stage-specific survival and/or recurrence in CMM. RESULTS Of the 8,749 studies identified, 26 studies were included, representing nine countries. Collectively, the studies covered a population of 152,422 patients and included data from 1978 to 2011. Randomized clinical trials and single-center observational studies comprised the most common study designs, including five large registry-based studies. Stage-specific information for survival and recurrence varied: 5-year overall survival: 95%-100% (stage I), 65%-92.8% (stage II), 41%-71% (stage III), and 9%-28% (stage IV); 5-year relapse-free survival was reported less frequently: 56% (stage II), and 28%-44% (stage III). Studies reporting survival by sentinel node (SN) status reported 5-year overall survival as 80%-95% for negative SN (stage I/II) and 35%-75% for positive SN (stage III) status; recurrence-free survival at 5 years: 76%-90% for negative and 35%-58% for positive SN status. Some studies included comparisons of survival by key patient sociodemographic characteristics, suggesting that these have a substantial influence on survival and recurrence estimates. CONCLUSION The studies identified in this review show large variations in stage-specific overall and recurrence-free survival by study type and by country. Owing to differing study designs and populations, it is difficult to make detailed comparisons. Large population-based studies that include stage-specific survival and recurrence in Europe are therefore important.
Collapse
Affiliation(s)
- Fernanda Costa Svedman
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | | | - Aliki Taylor
- Centre for Observational Research, Amgen Ltd, Uxbridge, UK
| | - Moninder Kaur
- Centre for Observational Research, Amgen Ltd, Uxbridge, UK
| | | | - Johan Hansson
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
10
|
Para-Phenylenediamine Induces Apoptotic Death of Melanoma Cells and Reduces Melanoma Tumour Growth in Mice. Biochem Res Int 2016; 2016:3137010. [PMID: 27293892 PMCID: PMC4886052 DOI: 10.1155/2016/3137010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 03/28/2016] [Accepted: 04/04/2016] [Indexed: 01/08/2023] Open
Abstract
Melanoma is one of the most aggressive forms of cancer, usually resistant to standard chemotherapeutics. Despite a huge number of clinical trials, any success to find a chemotherapeutic agent that can effectively destroy melanoma is yet to be achieved. Para-phenylenediamine (p-PD) in the hair dyes is reported to purely serve as an external dyeing agent. Very little is known about whether p-PD has any effect on the melanin producing cells. We have demonstrated p-PD mediated apoptotic death of both human and mouse melanoma cells in vitro. Mouse melanoma tumour growth was also arrested by the apoptotic activity of intraperitoneal administration of p-PD with almost no side effects. This apoptosis is shown to occur primarily via loss of mitochondrial membrane potential (MMP), generation of reactive oxygen species (ROS), and caspase 8 activation. p-PD mediated apoptosis was also confirmed by the increase in sub-G0/G1 cell number. Thus, our experimental observation suggests that p-PD can be a potential less expensive candidate to be developed as a chemotherapeutic agent for melanoma.
Collapse
|
11
|
González FE, Gleisner A, Falcón-Beas F, Osorio F, López MN, Salazar-Onfray F. Tumor cell lysates as immunogenic sources for cancer vaccine design. Hum Vaccin Immunother 2015; 10:3261-9. [PMID: 25625929 DOI: 10.4161/21645515.2014.982996] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Autologous dendritic cells (DCs) loaded with tumor-associated antigens (TAAs) are a promising immunological tool for cancer therapy. These stimulate the antitumor response and immunological memory generation. Nevertheless, many patients remain refractory to DC approaches. Antigen (Ag) delivery to DCs is relevant to vaccine success, and antigen peptides, tumor-associated proteins, tumor cells, autologous tumor lysates, and tumor-derived mRNA have been tested as Ag sources. Recently, DCs loaded with allogeneic tumor cell lysates were used to induce a potent immunological response. This strategy provides a reproducible pool of almost all potential Ags suitable for patient use, independent of MHC haplotypes or autologous tumor tissue availability. However, optimizing autologous tumor cell lysate preparation is crucial to enhancing efficacy. This review considers the role of cancer cell-derived lysates as a relevant source of antigens and as an activating factor for ex vivo therapeutic DCs capable of responding to neoplastic cells. These promising therapies are associated with the prolonged survival of advanced cancer patients.
Collapse
Key Words
- AM, Cytokine-activated monocytes
- Ags, Antigens
- CDAMs, Cell death-associated molecules
- CRT, Calreticulin
- CTLs, Cytotoxic T lymphocytes
- DAMPs
- DAMPs, Damage-associated molecular patterns
- DCs, Dendritic cells
- DTH, Delayed-type IV hypersensitivity
- GM-CSF, Granulocyte and macrophage colony stimulating factor
- HMGB1, High-mobility group box 1 protein
- HSPs, Heat shock proteins
- ICD, Immunogenic cell death
- MAAs, Melanoma-associated antigens
- MHC, Major histocompatibility complex
- MM, Malignant melanoma
- NKT, Natural killer T cell
- PAMPs, Pathogen-associated molecular patterns
- PBMCs, Peripheral blood mononuclear cells
- PCCL, Prostate cancer cell lysate
- PD1, Programmed cell death protein 1
- PRRs, Pattern recognition receptors
- PSA, Prostate specific antigen
- RAGE, Receptor for advanced glycation endproducts
- SNPs, Single nucleotide polymorphisms
- TAAs, Tumor-associated antigens
- TAPCells, Tumor antigen presenting cells
- TCRs, T cell receptors
- TLRs, Toll-like receptors
- TNF, Tumor necrosis factor
- TRIMEL, Allogeneic melanoma cell lysate
- TRIPRO, Allogeneic prostate cell lysate
- Toll-like receptors
- Tregs, Regulatory T lymphocytes
- cancer immunotherapy
- dendritic cells
- immunogenic cell death
Collapse
Affiliation(s)
- Fermín E González
- a Millennium Institute on Immunology and Immunotherapy; Institute of Biomedical Sciences; Faculty of Medicine ; University of Chile ; Santiago , Chile
| | | | | | | | | | | |
Collapse
|
12
|
Porcelli L, Guida G, Quatrale AE, Cocco T, Sidella L, Maida I, Iacobazzi RM, Ferretta A, Stolfa DA, Strippoli S, Guida S, Tommasi S, Guida M, Azzariti A. Aurora kinase B inhibition reduces the proliferation of metastatic melanoma cells and enhances the response to chemotherapy. J Transl Med 2015; 13:26. [PMID: 25623468 PMCID: PMC4314759 DOI: 10.1186/s12967-015-0385-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 01/08/2015] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The poor response to chemotherapy and the brief response to vemurafenib in metastatic melanoma patients, make the identification of new therapeutic approaches an urgent need. Interestingly the increased expression and activity of the Aurora kinase B during melanoma progression suggests it as a promising therapeutic target. METHODS The efficacy of the Aurora B kinase inhibitor barasertib-HQPA was evaluated in BRAF mutated cells, sensitive and made resistant to vemurafenib after chronic exposure to the drug, and in BRAF wild type cells. The drug effectiveness has been evaluated as cell growth inhibition, cell cycle progression and cell migration. In addition, cellular effectors of drug resistance and response were investigated. RESULTS The characterization of the effectors responsible for the resistance to vemurafenib evidenced the increased expression of MITF or the activation of Erk1/2 and p-38 kinases in the newly established cell lines with a phenotype resistant to vemurafenib. The sensitivity of cells to barasertib-HQPA was irrespective of BRAF mutational status. Barasertib-HQPA induced the mitotic catastrophe, ultimately causing apoptosis and necrosis of cells, inhibited cell migration and strongly affected the glycolytic metabolism of cells inducing the release of lactate. In association i) with vemurafenib the gain in effectiveness was found only in BRAF(V600K) cells while ii) with nab-paclitaxel, the combination was more effective than each drug alone in all cells. CONCLUSIONS These findings suggest barasertib as a new therapeutic agent and as enhancer of chemotherapy in metastatic melanoma treatment.
Collapse
Affiliation(s)
- Letizia Porcelli
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Gabriella Guida
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Anna E Quatrale
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Tiziana Cocco
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Letizia Sidella
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Immacolata Maida
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Rosa M Iacobazzi
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Anna Ferretta
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Diana A Stolfa
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Sabino Strippoli
- Medical Oncology Department, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Stefania Guida
- Unit of Dermatology and Venereology, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Stefania Tommasi
- Molecular Genetics Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Michele Guida
- Medical Oncology Department, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Amalia Azzariti
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| |
Collapse
|
13
|
Garrisi VM, Strippoli S, De Summa S, Pinto R, Perrone A, Guida G, Azzariti A, Guida M, Stefania T. Proteomic profile and in silico analysis in metastatic melanoma with and without BRAF mutation. PLoS One 2014; 9:e112025. [PMID: 25437182 PMCID: PMC4249853 DOI: 10.1371/journal.pone.0112025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 10/11/2014] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Selective inhibitors of BRAF, vemurafenib and dabrafenib are the standard of care for metastatic melanoma patients with BRAF V600, while chemotherapy continued to be widely used in BRAF wild type patients. MATERIALS AND METHODS In order to discover novel candidate biomarkers predictive to treatment, serum of 39 metastatic melanoma vemurafenib (n = 19) or chemotherapy (n = 20) treated patients at baseline, at disease control and at progression, were analyzed using SELDI-TOF technology. In silico analysis was used to identify more significant peaks. RESULTS In patients with different BRAF status, we found 5 peptides significantly deregulated, with the down-regulation of the m/z 9176 peak strongly associated with BRAF mutation. At baseline as predictive biomarkers we identified 2 peptides - m/z 6411, 4075 - as significantly up-regulated in responders to chemotherapy and 4 peaks - m/z 5900, 12544, 49124 and 11724 - significantly up-regulated in longer vs shorter responders to vemurafenib. After response, 3 peptides (m/z 4658, 18639, and 9307) resulted significantly down regulated while 3 peptides m/z 9292, 7765 and 9176 appeared up-regulated respectively in chemotherapy and vemurafenib responder patients. In vemurafenib treated patients, 16 peaks appeared deregulated at progression compared to baseline time. In silico analysis identified proteins involved in invasiveness (SLAIN1) and resistance (ABCC12) as well as in the pathway of detoxification (NQO1) and apoptosis (RBM10, TOX3, MTEFD1, TSPO2). Proteins associated with the modulation of neuronal plasticity (RIN1) and regulatory activity factors of gene transcription (KLF17, ZBTB44) were also highlighted. CONCLUSION Our exploratory study highlighted some factors that deserve to be further investigated in order to provide a framework for improving melanoma treatment management through the development of biomarkers which could act as the strongest surrogates of the key biological events in stage IV melanoma.
Collapse
Affiliation(s)
- Vito Michele Garrisi
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Sabino Strippoli
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Simona De Summa
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Rosamaria Pinto
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Antonella Perrone
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Gabriella Guida
- Dept. of Basic Medical Sciences, Faculty of Medicine and Surgery, School of Medicine, University of Bari, Bari, Italy
| | - Amalia Azzariti
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Michele Guida
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Tommasi Stefania
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
- * E-mail:
| |
Collapse
|
14
|
Sahu RP, Ocana JA, Harrison KA, Ferracini M, Touloukian CE, Al-Hassani M, Sun L, Loesch M, Murphy RC, Althouse SK, Perkins SM, Speicher PJ, Tyler DS, Konger RL, Travers JB. Chemotherapeutic agents subvert tumor immunity by generating agonists of platelet-activating factor. Cancer Res 2014; 74:7069-78. [PMID: 25304264 DOI: 10.1158/0008-5472.can-14-2043] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Oxidative stress suppresses host immunity by generating oxidized lipid agonists of the platelet-activating factor receptor (PAF-R). Because many classical chemotherapeutic drugs induce reactive oxygen species (ROS), we investigated whether these drugs might subvert host immunity by activating PAF-R. Here, we show that PAF-R agonists are produced in melanoma cells by chemotherapy that is administered in vitro, in vivo, or in human subjects. Structural characterization of the PAF-R agonists induced revealed multiple oxidized glycerophosphocholines that are generated nonenzymatically. In a murine model of melanoma, chemotherapeutic administration could augment tumor growth by a PAF-R-dependent process that could be blocked by treatment with antioxidants or COX-2 inhibitors or by depletion of regulatory T cells. Our findings reveal how PAF-R agonists induced by chemotherapy treatment can promote treatment failure. Furthermore, they offer new insights into how to improve the efficacy of chemotherapy by blocking its heretofore unknown impact on PAF-R activation.
Collapse
Affiliation(s)
- Ravi P Sahu
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana. Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jesus A Ocana
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana. Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Kathleen A Harrison
- Department of Pharmacology, University of Colorado Health Sciences Center, Aurora, Colorado
| | - Matheus Ferracini
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - Mohammed Al-Hassani
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Louis Sun
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Mathew Loesch
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Robert C Murphy
- Department of Pharmacology, University of Colorado Health Sciences Center, Aurora, Colorado
| | - Sandra K Althouse
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Susan M Perkins
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Paul J Speicher
- The Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Douglas S Tyler
- The Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Raymond L Konger
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana. Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jeffrey B Travers
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana. Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana. The Richard L. Roudebush V.A. Medical Center, Indianapolis, Indiana.
| |
Collapse
|
15
|
Vemurafenib in the French temporary authorization for use metastatic melanoma cohort: a single-centre trial. Melanoma Res 2014; 24:75-82. [PMID: 24241686 DOI: 10.1097/cmr.0000000000000034] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Vemurafenib, a selective BRAF inhibitor, has recently shown an improved overall survival (OS) in metastatic melanoma with V600E mutation in phase 2 and 3 trials. Patients with BRAF V600E metastatic melanoma received vemurafenib orally, in the French temporary authorization for use program from April 2011 to April 2012. We analysed the clinical benefit and safety of vemurafenib. Secondary analyses included the impact of brain metastases on median OS and progression-free survival (PFS). Fifty patients were enrolled, of whom 20% had stage IIIC and 80% stage IV disease. The majority were men (58%), with a median age of 58 years (51-69). Forty-three patients had an Eastern Cooperative Oncology Group performance status of 0 or 1 (86%). Twenty patients had brain metastases (40%). Overall response rate was 53%. Complete response was achieved in five patients (10%), partial response in 21 patients (43%) and stable disease in seven patients (14%). Median OS was 7.5 months (95% confidence interval 5.6-12.7) and PFS was 3.6 months (95% confidence interval 2.9-5.9). Patients with brain metastasis had a response rate of 50% (nine partial response, one complete response), and median OS and PFS were, respectively, 4.3 and 3.1 months. Common adverse events were fatigue, arthralgia and cutaneous side effects. Sixteen per cent developed squamous cell carcinoma. Grade 3/4 was observed in 11 patients (22%). Six per cent required temporary discontinuation and/or dose reduction because of toxic effects. This study confirms the considerable clinical benefit of vemurafenib for patients with brain metastasis, with manageable toxicity.
Collapse
|
16
|
Asundi J, Lacap JA, Clark S, Nannini M, Roth L, Polakis P. MAPK Pathway Inhibition Enhances the Efficacy of an Anti-Endothelin B Receptor Drug Conjugate by Inducing Target Expression in Melanoma. Mol Cancer Ther 2014; 13:1599-610. [DOI: 10.1158/1535-7163.mct-13-0446] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
17
|
The pan-Aurora kinase inhibitor, PHA-739358, induces apoptosis and inhibits migration in melanoma cell lines. Melanoma Res 2014; 23:102-13. [PMID: 23344158 DOI: 10.1097/cmr.0b013e32835df5e4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Treatment of metastatic melanoma has long been a challenge because of its resistance to traditional chemotherapeutics, leading to the search for alternative strategies. Aurora kinases are key mitotic regulators that are frequently overexpressed in various cancers including melanoma, making them ideal targets for drug development. Several Aurora kinase inhibitors have been developed and tested preclinically and clinically. PHA-739358 is currently one of the most advanced clinical compounds being tested in phase II clinical trials; however, its antitumor effect has not been tested in melanoma. In this study, the antiproliferative and anti-invasive effects of PHA-739358 were investigated in melanoma cell lines. The results demonstrated that PHA-739358 produces a time-dependent and dose-dependent inhibition of cell proliferation, induction of apoptosis, and inhibition of cell migration. Downregulation of matrix metalloproteinase-2 by the inhibition of NFκB-signaling pathway may contribute to PHA-739358-induced inhibition of migration. Furthermore, PHA-739358 enhanced temozolomide and Plx4032-induced apoptosis. This study suggests that Aurora kinase inhibitors may provide a new strategy for the treatment of advanced melanoma.
Collapse
|
18
|
Wu X, Marmarelis ME, Hodi FS. Activity of the heat shock protein 90 inhibitor ganetespib in melanoma. PLoS One 2013; 8:e56134. [PMID: 23418523 PMCID: PMC3572008 DOI: 10.1371/journal.pone.0056134] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 01/05/2013] [Indexed: 01/09/2023] Open
Abstract
Heat shock protein 90 (HSP90) is involved in the regulation of diverse biological processes such as cell signaling, proliferation and survival, and has been recently recognized as a potential target for cancer therapy. Ganetespib is a potent ATP competitive inhibitor of HSP90. Ganetespib downregulated the expression of multiple signal transducing molecules including EGFR, IGF-1R, c-Met, Akt, B-RAF and C-RAF, resulting in pronounced decrease in phosphorylation of Akt and Erk1/2 in a panel of five cutaneous melanoma cell lines including those harboring B-RAF and N-RAS mutations. Ganetespib exhibited potent antiproliferative activity on all five of these cell lines, with IC50 values between 37.5 and 84 nM. Importantly, Ganetespib is active on B-RAF mutated melanoma cells that have acquired resistance to B-RAF inhibition. Ganetespib induced apoptosis and cell cycle arrest at G1 and/or G2/M phase. Ganetespib induced cell cycle arrest was accompanied by altered expression of cyclin-dependent kinase inhibitor (CDKI) p21(Cip1) and p27(Kip1), cyclins B1, D1 and E, and/or cyclin-dependent kinases 1, 2 and 4. HSP90 is functionally important for melanoma cells and HSP90 inhibitors such as ganetespib could potentially be effective therapeutics for melanoma with various genetic mutations and acquired resistance to B-RAF inhibition.
Collapse
Affiliation(s)
- Xinqi Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
- Melanoma Disease Center, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts, United States of America
| | | | - F. Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
- Melanoma Disease Center, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts, United States of America
| |
Collapse
|
19
|
Hickerson RP, Gonzalez-Gonzalez E, Vlassov AV, Li M, Lara MF, Contag CH, Kaspar RL. Intravital fluorescence imaging of small interfering RNA-mediated gene repression in a dual reporter melanoma xenograft model. Nucleic Acid Ther 2012; 22:438-43. [PMID: 23098239 DOI: 10.1089/nat.2012.0364] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Development of RNA interference (RNAi)-based therapeutics has been hampered by the lack of effective and efficient means of delivery. Reliable model systems for screening and optimizing delivery of RNAi-based agents in vivo are crucial for preclinical research aimed at advancing nucleic acid-based therapies. We describe here a dual fluorescent reporter xenograft melanoma model prepared by intradermal injection of human A375 melanoma cells expressing tandem tomato fluorescent protein (tdTFP) containing a small interfering RNA (siRNA) target site as well as enhanced green fluorescent protein (EGFP), which is used as a normalization control. Intratumoral injection of a siRNA specific to the incorporated siRNA target site, complexed with a cationic lipid that has been optimized for in vivo delivery, resulted in 65%±11% knockdown of tdTFP relative to EGFP quantified by in vivo imaging and 68%±10% by reverse transcription-quantitative polymerase chain reaction. No effect was observed with nonspecific control siRNA treatment. This model provides a platform on which siRNA delivery technologies can be screened and optimized in vivo.
Collapse
|