1
|
Manica D, da Silva GB, Narzetti RA, Dallagnoll P, da Silva AP, Marafon F, Cassol J, de Souza Matias L, Zamoner A, de Oliveira Maciel SFV, Moreno M, Bagatini MD. Curcumin modulates purinergic signaling and inflammatory response in cutaneous metastatic melanoma cells. Purinergic Signal 2025; 21:277-288. [PMID: 38801619 PMCID: PMC12061816 DOI: 10.1007/s11302-024-10023-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Cutaneous melanoma (CM) poses a therapeutic challenge due to its aggressive nature and often limited response to conventional treatments. Exploring novel therapeutic targets is essential, and natural compounds have emerged as potential candidates. This study aimed to elucidate the impact of curcumin, a natural compound known for its anti-inflammatory, antioxidant, and anti-tumor properties, on metastatic melanoma cells, focusing on the purinergic system and immune responses. Human melanoma cell line SK-Mel-28 were exposed to different curcumin concentrations for either 6 or 24 h, after which we assessed components related to the purinergic system and the inflammatory cascade. Using RT-qPCR, we assessed the gene expression of CD39 and CD73 ectonucleotidases, as well as adenosine deaminase (ADA). Curcumin effectively downregulated CD39, CD73, and ADA gene expression. Flow cytometry analysis revealed that curcumin significantly reduced CD39 and CD73 protein expression at specific concentrations. Moreover, the A2A receptor's protein expression decreased across all concentrations. Enzymatic activity assays demonstrated that curcumin modulated CD39, CD73, and ADA activities, with effects dependent on concentration and duration of treatment. Extracellular ATP levels increased after 24 h of curcumin treatment, emphasizing its role in modulating hydrolytic activity. Curcumin also displayed anti-inflammatory properties by reducing NLRP3 gene expression and impacting the levels of key inflammatory cytokines. In conclusion, this study unveils the potential of curcumin as a promising adjuvant in CM treatment. Curcumin modulates the expression and activity of crucial components of the purinergic system and exhibits anti-inflammatory effects, indicating its potential therapeutic role in combating CM. These findings underscore curcumin's promise and warrant further investigation in preclinical and clinical settings for melanoma management.
Collapse
Affiliation(s)
- Daiane Manica
- Department of Biochemistry, Biochemistry Graduate Program, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Gilnei Bruno da Silva
- Multicentric Graduate Program in Biochemistry and Molecular Biology, State University of Santa Catarina, Lages, SC, Brazil
| | - Rafael Antônio Narzetti
- Department of Biochemistry, Biochemistry Graduate Program, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Paula Dallagnoll
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapeco, SC, Brazil
| | - Alana Patrícia da Silva
- Department of Biochemistry, Biochemistry Graduate Program, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Filomena Marafon
- Department of Biochemistry, Biochemistry Graduate Program, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Joana Cassol
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapeco, SC, Brazil
| | - Letícia de Souza Matias
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapeco, SC, Brazil
| | - Ariane Zamoner
- Department of Biochemistry, Biochemistry Graduate Program, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | | | - Marcelo Moreno
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapeco, SC, Brazil.
| | - Margarete Dulce Bagatini
- Department of Biochemistry, Biochemistry Graduate Program, Federal University of Santa Catarina, Florianopolis, SC, Brazil.
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapeco, SC, Brazil.
| |
Collapse
|
2
|
Acar C, Yüksel HÇ, Şahin G, Açar FP, Gunenc D, Karaca B. Prognostic utility of the CALLY index in metastatic melanoma: building a nomogram for Patients on Anti-PD-1 therapy. Clin Transl Oncol 2025:10.1007/s12094-025-03888-z. [PMID: 40091005 DOI: 10.1007/s12094-025-03888-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 02/25/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Despite the success of immune checkpoint inhibitors (ICIs) in metastatic melanoma, many patients fail to derive meaningful benefit, underscoring the urgent need for accessible prognostic biomarkers. The C-reactive protein (CRP)-albumin-lymphocyte (CALLY) index, an immunonutritional index, has shown prognostic value in various cancers. Previous studies indicate that systemic inflammation and nutritional status influence ICI efficacy, suggesting the potential relevance of the CALLY index in metastatic melanoma. This study evaluates the CALLY index's role in metastatic melanoma patients treated with anti-PD-1 therapy. METHODS This retrospective study analysed 92 patients with metastatic melanoma who were treated with anti-PD-1 monotherapy at Ege University's Faculty of Medicine between 2015 and 2023. The CALLY index was calculated using the pre-treatment CRP, albumin and lymphocyte levels. Kaplan-Meier analysis was used to estimate survival outcomes, and univariate and multivariate Cox regression models were employed to identify independent prognostic factors. A predictive nomogram incorporating the CALLY index and other significant variables was then developed. RESULTS The optimal CALLY index cutoff was determined to be 2. Patients with a low CALLY index (≤ 2) had worse median overall survival (OS) and progression-free survival (PFS) when compared with those who had a high CALLY index (> 2) (median OS: 9.6 vs 31.3 months, p < 0.001; median PFS: 3.8 vs 10.6 months, p = 0.001). Multivariate analysis identified the CALLY index, lactate dehydrogenase above the upper limit of normal, Eastern Cooperative Oncology Group score ≥ 2, M1c/M1d staging and acral/mucosal melanoma subtypes to be independent predictors of OS. A nomogram was then constructed based on these factors, yielding a concordance index of 0.705 (95% confidence interval: 0.634-0.776). This model stratified patients into low-, intermediate- and high-risk groups, with the high-risk group showing significantly worse OS than the intermediate- and the low-risk groups (p < 0.001). CONCLUSION The CALLY index is a cost-effective and independent prognostic biomarker that can aid in risk stratification and guide treatment decisions in patients with metastatic melanoma receiving anti-PD-1 therapy.
Collapse
Affiliation(s)
- Caner Acar
- Division of Medical Oncology, Department of Internal Medicine, Ege University Medical Faculty, 35100, Izmir, Turkey.
| | - Haydar Çağatay Yüksel
- Division of Medical Oncology, Department of Internal Medicine, Ege University Medical Faculty, 35100, Izmir, Turkey
| | - Gökhan Şahin
- Division of Medical Oncology, Department of Internal Medicine, Ege University Medical Faculty, 35100, Izmir, Turkey
| | - Fatma Pinar Açar
- Division of Medical Oncology, Department of Internal Medicine, Ege University Medical Faculty, 35100, Izmir, Turkey
| | - Damla Gunenc
- Division of Medical Oncology, Hatay Training and Research Hospital, 3100, Hatay, Turkey
| | - Burçak Karaca
- Division of Medical Oncology, Department of Internal Medicine, Ege University Medical Faculty, 35100, Izmir, Turkey
| |
Collapse
|
3
|
Liu Q, Shaibu Z, Xu A, Yang F, Cao R, Yang F. Predictive value of serum cytokines in patients with non-small-cell lung cancer receiving anti-PD-1 blockade therapy: a meta-analysis. Clin Exp Med 2025; 25:59. [PMID: 39955679 PMCID: PMC11830645 DOI: 10.1007/s10238-025-01587-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
Non-small cell lung cancer (NSCLC) is a leading cause of cancer-related deaths worldwide. Immunotherapy, particularly PD-1 inhibitors, has revolutionized the treatment landscape for NSCLC. However, the predictive biomarkers for PD-1 inhibitor therapy are still limited. Serum cytokines have emerged as potential biomarkers for predicting treatment outcomes. This meta-analysis aims to investigate the predictive value of serum cytokines in PD-1 inhibitor therapy for NSCLC. We conducted a comprehensive literature search in major databases, including PubMed, Google scholar, Embase, and Cochrane database, with a focus on literature published up until October 22, 2024. Studies investigating the association between serum cytokine levels and treatment outcomes in NSCLC patients receiving PD-1 inhibitor therapy were included. The primary outcomes were progression-free survival (PFS) and overall survival (OS). The meta-analysis revealed that elevated IL-6 levels were significantly associated with poorer PFS in NSCLC patients (HR = 2.30, 95% CI [1.39-3.80], P = 0.001). Additionally, high IL-10 expression was related to poorer PFS in NSCLC after therapy (HR = 2.45, 95% CI [1.26-4.76], P = 0.009). In contrast, no significant associations were found between OS and the expression of various cytokines, including IL-4, IL-5, IL-6, IL-8, IL-10, IFN-γ, IL-1β, TNF-α, and IL-12p70. This meta-analysis demonstrates that elevated IL-6 and IL-10 levels are significantly associated with poorer PFS in NSCLC patients receiving PD-1 inhibitor therapy. These findings suggest that serum cytokine levels may serve as predictive biomarkers for treatment outcomes. Further studies are needed to validate these results and explore the underlying mechanisms.
Collapse
Affiliation(s)
- Qian Liu
- Department of Laboratory Medicine, Lianyungang Clinical College Jiangsu University & the Second People'S Hospital of Lianyungang, 41 East Hailian Road, Lianyungang, 222006, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College Xuzhou Medical University & the Second People'S Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China
- Department of Laboratory Medicine, The Second People'S Hospital of Lianyungang Affiliated with Kangda College of Nanjing Medical University, Lianyungang, 222006, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College, Bengbu Medical University & the Second People'S Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China
| | - Zakari Shaibu
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Aiguo Xu
- Department of Oncology, The Second People'S Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China
| | - Fang Yang
- Department of Laboratory Medicine, Lianyungang Clinical College Jiangsu University & the Second People'S Hospital of Lianyungang, 41 East Hailian Road, Lianyungang, 222006, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College Xuzhou Medical University & the Second People'S Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China
- Department of Laboratory Medicine, The Second People'S Hospital of Lianyungang Affiliated with Kangda College of Nanjing Medical University, Lianyungang, 222006, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College, Bengbu Medical University & the Second People'S Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China
| | - Ruoxue Cao
- Department of Laboratory Medicine, Lianyungang Clinical College Jiangsu University & the Second People'S Hospital of Lianyungang, 41 East Hailian Road, Lianyungang, 222006, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College Xuzhou Medical University & the Second People'S Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China
- Department of Laboratory Medicine, The Second People'S Hospital of Lianyungang Affiliated with Kangda College of Nanjing Medical University, Lianyungang, 222006, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College, Bengbu Medical University & the Second People'S Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China
| | - Fumeng Yang
- Department of Laboratory Medicine, Lianyungang Clinical College Jiangsu University & the Second People'S Hospital of Lianyungang, 41 East Hailian Road, Lianyungang, 222006, Jiangsu, China.
- Department of Laboratory Medicine, Lianyungang Clinical College Xuzhou Medical University & the Second People'S Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China.
- Department of Laboratory Medicine, The Second People'S Hospital of Lianyungang Affiliated with Kangda College of Nanjing Medical University, Lianyungang, 222006, Jiangsu, China.
- Department of Laboratory Medicine, Lianyungang Clinical College, Bengbu Medical University & the Second People'S Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China.
| |
Collapse
|
4
|
Al Ageeli E, Abdulhakim JA, Hussein MH, Alnoman MM, Alkhalil SS, Issa PP, Nemr NA, Abdelmaksoud A, Alenizi DA, Fawzy MS, Toraih EA. The HCV-Melanoma Paradox: First Multi-Cohort and Molecular Net-Work Analysis Reveals Lower Incidence but Worse Outcomes-Integrating Clinical, Real-World, and In Silico Data. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1531. [PMID: 39336572 PMCID: PMC11433761 DOI: 10.3390/medicina60091531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/01/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024]
Abstract
Background and Objectives: The relationship between hepatitis C virus (HCV) infection and melanoma remains poorly understood. This study aimed to investigate the association between HCV and melanoma, assess outcomes in patients with both conditions, and explore potential molecular mechanisms connecting the two diseases. Materials and Methods: We conducted a retrospective cohort study of 142 melanoma patients, including 29 with HCV-related cirrhosis, and analyzed their clinical outcomes. For external validation, we used the TriNetX Global Collaborative Network database, comprising 219,960 propensity-matched patients per group. An in silico analysis was performed to identify the molecular pathways linking HCV and melanoma. Results: In the retrospective cohort, HCV-positive melanoma patients showed an increased risk of early relapse (41.4% vs. 18.6%, p = 0.014), recurrence (65.5% vs. 39.8%, p = 0.020), and mortality (65.5% vs. 23.0%, p < 0.001) compared to HCV-negative patients. TriNetX data analysis revealed that HCV-positive patients had a 53% lower risk of developing melanoma (RR = 0.470, 95% CI: 0.443-0.498, p < 0.001). However, HCV-positive melanoma patients had higher all-cause mortality (HR = 1.360, 95% CI: 1.189-1.556, p < 0.001). An in silico analysis identified key molecular players, including IL-6 and CTLA4, in the HCV-melanoma network. Conclusions: While HCV infection may be associated with a lower risk of melanoma development, HCV-positive patients who develop melanoma have poorer outcomes. The identified molecular pathways provide potential targets for future research and therapeutic interventions.
Collapse
Affiliation(s)
- Essam Al Ageeli
- Department of Basic Medical Sciences, Faculty of Medicine, Jazan University, Jazan 45141, Saudi Arabia;
| | - Jawaher A. Abdulhakim
- Department of Medical Laboratory, College of Applied Medical Sciences, Taibah University, Yanbu 46423, Saudi Arabia;
| | | | - Maryam M. Alnoman
- Department of Biology, Faculty of Science, Taibah University, Yanbu 46423, Saudi Arabia;
| | - Samia S. Alkhalil
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Alquwayiyah 11961, Saudi Arabia;
| | - Peter P. Issa
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA;
| | - Nader A. Nemr
- Endemic and Infectious Diseases Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | - Ahmed Abdelmaksoud
- Department of Internal Medicine, University of California, Riverside, CA 92521, USA;
| | - Dhaifallah A. Alenizi
- Department of Medicine, Faculty of Medicine, Northern Border University, Arar 91431, Saudi Arabia;
| | - Manal S. Fawzy
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar 1321, Saudi Arabia
- Center for Health Research, Northern Border University, Arar 91431, Saudi Arabia
| | - Eman A. Toraih
- Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA
- Genetics Unit, Department of Histology and Cell Biology, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
5
|
Smithy JW, Kalvin HL, Ehrich FD, Shah R, Adamow M, Raber V, Maher CA, Kleman J, McIntyre DAG, Shoushtari AN, Betof Warner A, Callahan MK, Momtaz P, Eton O, Nair S, Wolchok JD, Chapman PB, Berger MF, Panageas KS, Postow MA. Early On-Treatment Assessment of T Cells, Cytokines, and Tumor DNA with Adaptively Dosed Nivolumab + Ipilimumab: Final Results from the Phase 2 ADAPT-IT Study. Clin Cancer Res 2024; 30:3407-3415. [PMID: 38767650 PMCID: PMC11357703 DOI: 10.1158/1078-0432.ccr-23-3643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/12/2024] [Accepted: 05/15/2024] [Indexed: 05/22/2024]
Abstract
PURPOSE The Adaptively Dosed ImmunoTherapy Trial (ADAPT-IT;NCT03122522) investigated adaptive ipilimumab discontinuation in melanoma based on early radiographic assessment. Initial findings indicated similar effectiveness compared with conventional nivolumab-ipilimumab (nivo-ipi). Exploratory biomarker analyses and final clinical results are now reported. PATIENTS AND METHODS Patients with unresectable melanoma received two doses of nivo-ipi. Radiographic assessment at Week 6 informed continuation of ipilimumab before nivolumab maintenance. The primary endpoint was overall response rate at Week 12. Plasma was assayed for circulating tumor DNA and 10 cytokines using a multiplex immunoassay. Flow cytometry of peripheral blood mononuclear cells was performed with an 11-color panel. RESULTS Among the treated patients, expansion of proliferating T-cell populations was observed in responders and nonresponders. Baseline IL6 levels were low in patients achieving an objective radiographic response (median 1.30 vs. 2.86 pg/mL; P = 0.025). High baseline IL6 levels were associated with short progression-free survival [PFS; HR = 1.24, 95% confidence interval (CI), 1.01-1.52; P = 0.041]. At Week 6, patients with response had lower average tumor variant allele fractions than nonresponders (median 0.000 vs. 0.019; P = 0.014). Greater increases in average variant allele fractions from baseline to Week 6 correlated with short PFS (HR = 1.11, 95% CI, 1.01-1.21; P = 0.023). Week 12 overall response rate was 47% (95% CI, 35%-59%) with a median follow-up of 34 months among survivors. Median PFS was 21 months (95% CI, 10-not reached); 76% of responses (95% CI, 64%-91%) persisted at 36 months. CONCLUSIONS Adaptively dosed nivo-ipi responses are durable and resemble historical data for conventional nivo-ipi. Baseline IL6 and circulating tumor DNA changes during treatment warrant further study as biomarkers of nivo-ipi response.
Collapse
Affiliation(s)
- James W Smithy
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Fiona D Ehrich
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ronak Shah
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Matthew Adamow
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Collen A Maher
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jenna Kleman
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Alexander N Shoushtari
- Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
| | - Allison Betof Warner
- Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
| | - Margaret K Callahan
- Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
| | - Parisa Momtaz
- Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
| | - Omar Eton
- Hartford HealthCare Cancer Institute, Hartford, Connecticut
| | - Suresh Nair
- Lehigh Valley Health Network, Allentown, Pennsylvania
| | - Jedd D Wolchok
- Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
| | - Paul B Chapman
- Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
| | | | | | - Michael A Postow
- Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
| |
Collapse
|
6
|
Liu D, Hu L, Shao H. Therapeutic drug monitoring of immune checkpoint inhibitors: based on their pharmacokinetic properties and biomarkers. Cancer Chemother Pharmacol 2023:10.1007/s00280-023-04541-8. [PMID: 37410155 DOI: 10.1007/s00280-023-04541-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 05/03/2023] [Indexed: 07/07/2023]
Abstract
As a new means of oncology treatment, immune checkpoint inhibitors (ICIs) can improve survival rates in patients with resistant or refractory tumors. However, there are obvious inter-individual differences in the unsatisfactory response rate, drug resistance rate and the occurrence of immune-related adverse events (irAE). These questions have sparked interest in researchers looking for a way to screen sensitive populations and predict efficacy and safety. Therapeutic drug monitoring (TDM) is a way to ensure the safety and effectiveness of medication by measuring the concentration of drugs in body fluids and adjusting the medication regimen. It has the potential to be an adjunctive means of predicting the safety and efficacy of ICIs treatment. In this review, the author outlined the pharmacokinetic (PK) characteristics of ICIs in patients. The feasibility and limitations of TDM of ICIs were discussed by summarizing the relationships between the pharmacokinetic parameters and the efficacy, toxicity and biomarkers.
Collapse
Affiliation(s)
- Dongxue Liu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Linlin Hu
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Office of Medication Clinical Institution, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Hua Shao
- Office of Medication Clinical Institution, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
7
|
de la O-Cuevas E, Islas SR, Gallegos-Flores P, E L EI, Tototzintle-Huitle H, Saniger JM. Modulating the interaction of graphenic substrates with human interleukin-6 and its monoclonal antibody: a study by Raman images. RSC Adv 2023; 13:15114-15120. [PMID: 37207094 PMCID: PMC10189315 DOI: 10.1039/d3ra01627g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/03/2023] [Indexed: 05/21/2023] Open
Abstract
Interleukin-6 (IL-6) is a cytokine with wide-ranging biological effects, playing an important role on the immune system and inflammatory responses. Therefore, it is important to develop alternative, highly sensitive and reliable analytical methodologies for the accurate detection of this biomarker in biological fluids. Graphene substrates (GS), such as pristine graphene (G), graphene oxide (GO), and reduced graphene oxide (rGO), have shown great benefits for biosensing and in the development of novel biosensor devices. In this work, we present a proof of concept for the development of a new analytical platform for the specific recognition of human interleukin-6, that is based on the coffee-ring formation of monoclonal antibodies of interleukin-6 (mabIL-6) onto amine functionalized GS. The prepared GS/mabIL-6/IL-6 systems were successfully used to show that IL-6 was specifically and selectively adsorbed onto the area of the mabIL-6 coffee-ring. Raman imaging was confirmed as a versatile tool to investigate different antigen-antibody interactions and their surface distribution. This experimental approach can be used to develop a wide variety of substrates for antigen-antibody interaction allowing the specific detection of an analyte in a complex matrix.
Collapse
Affiliation(s)
- Emmanuel de la O-Cuevas
- Unidad Académica de Ciencias Biológicas, Universidad Autónoma de Zacatecas 98068 Zacatecas Mexico
- Instituto de Ciencias Aplicadas y Tecnología, Universidad Nacional Autónoma de México Circuito Exterior S/N, Cd. Universitaria 04510 Ciudad de México Mexico
- Unidad Académica de Física, Universidad Autónoma de Zacatecas 98068 Zacatecas Mexico
| | - Selene R Islas
- Instituto de Ciencias Aplicadas y Tecnología, Universidad Nacional Autónoma de México Circuito Exterior S/N, Cd. Universitaria 04510 Ciudad de México Mexico
| | - Perla Gallegos-Flores
- Unidad Académica de Ciencias Biológicas, Universidad Autónoma de Zacatecas 98068 Zacatecas Mexico
| | - Esparza-Ibarra E L
- Unidad Académica de Ciencias Biológicas, Universidad Autónoma de Zacatecas 98068 Zacatecas Mexico
| | | | - José M Saniger
- Instituto de Ciencias Aplicadas y Tecnología, Universidad Nacional Autónoma de México Circuito Exterior S/N, Cd. Universitaria 04510 Ciudad de México Mexico
| |
Collapse
|
8
|
Inoue Y, Inui N, Karayama M, Asada K, Fujii M, Matsuura S, Uto T, Hashimoto D, Matsui T, Ikeda M, Yasui H, Hozumi H, Suzuki Y, Furuhashi K, Enomoto N, Fujisawa T, Suda T. Cytokine profiling identifies circulating IL-6 and IL-15 as prognostic stratifiers in patients with non-small cell lung cancer receiving anti-PD-1/PD-L1 blockade therapy. Cancer Immunol Immunother 2023:10.1007/s00262-023-03453-z. [PMID: 37099186 DOI: 10.1007/s00262-023-03453-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 04/16/2023] [Indexed: 04/27/2023]
Abstract
Whether circulating levels of specific cytokines at baseline link with treatment efficacy of immune checkpoint blockade (ICB) therapy in patients with non-small cell lung cancer remains unknown. In this study, serum samples were collected in two independent, prospective, multicenter cohorts before the initiation of ICB. Twenty cytokines were quantified, and cutoff values were determined by receiver operating characteristic analyses to predict non-durable benefit. The associations of each dichotomized cytokine status with survival outcomes were assessed. In the discovery cohort (atezolizumab cohort; N = 81), there were significant differences in progression-free survival (PFS) in accordance with the levels of IL-6 (log-rank test, P = 0.0014), IL-15 (P = 0.00011), MCP-1 (P = 0.013), MIP-1β (P = 0.0035), and PDGF-AB/BB (P = 0.016). Of these, levels of IL-6 and IL-15 were also significantly prognostic in the validation cohort (nivolumab cohort, N = 139) for PFS (log-rank test, P = 0.011 for IL-6 and P = 0.00065 for IL-15) and overall survival (OS; P = 3.3E-6 for IL-6 and P = 0.0022 for IL-15). In the merged cohort, IL-6high and IL-15high were identified as independent unfavorable prognostic factors for PFS and OS. The combined IL-6 and IL-15 status stratified patient survival outcomes into three distinct groups for both PFS and OS. In conclusion, combined assessment of circulating IL-6 and IL-15 levels at baseline provides valuable information to stratify the clinical outcome of patients with non-small cell lung cancer treated with ICB. Further studies are required to decipher the mechanistic basis of this finding.
Collapse
Affiliation(s)
- Yusuke Inoue
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-Ku, Hamamatsu, 431-3192, Japan.
| | - Naoki Inui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-Ku, Hamamatsu, 431-3192, Japan
- Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-Ku, Hamamatsu, 431-3192, Japan
| | - Masato Karayama
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-Ku, Hamamatsu, 431-3192, Japan
- Department of Chemotherapy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-Ku, Hamamatsu, 431-3192, Japan
| | - Kazuhiro Asada
- Department of Respiratory Medicine, Shizuoka General Hospital, 4-27-1 Kita-Ando, Shizuoka, 420-8527, Japan
| | - Masato Fujii
- Department of Respiratory Medicine, Shizuoka City Shizuoka Hospital, 10-93 Otemachi, Shizuoka, 420-8630, Japan
| | - Shun Matsuura
- Department of Respiratory Medicine, Fujieda Municipal General Hospital, 4-1-11 Surugadai, Fujieda, 426-8677, Japan
| | - Tomohiro Uto
- Department of Respiratory Medicine, Iwata City Hospital, 512-3 Ohkubo, Iwata, 438-8550, Japan
| | - Dai Hashimoto
- Department of Pulmonary Medicine, Seirei Hamamatsu General Hospital, 2-12-12 Sumiyoshi, Naka-Ku, Hamamatsu, 430-8558, Japan
| | - Takashi Matsui
- Department of Respiratory Medicine, Seirei Mikatahara General Hospital, 3453 Mikatahara, Kita-Ku, Hamamatsu, 433-8558, Japan
| | - Masaki Ikeda
- Department of Respiratory Medicine, Shizuoka Saiseikai General Hospital, 1-1-1 Oshika, Shizuoka, 422-8527, Japan
| | - Hideki Yasui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-Ku, Hamamatsu, 431-3192, Japan
| | - Hironao Hozumi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-Ku, Hamamatsu, 431-3192, Japan
| | - Yuzo Suzuki
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-Ku, Hamamatsu, 431-3192, Japan
| | - Kazuki Furuhashi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-Ku, Hamamatsu, 431-3192, Japan
| | - Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-Ku, Hamamatsu, 431-3192, Japan
| | - Tomoyuki Fujisawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-Ku, Hamamatsu, 431-3192, Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-Ku, Hamamatsu, 431-3192, Japan
| |
Collapse
|
9
|
Piasek AM, Musolf P, Sobiepanek A. Aptamer-based Advances in Skin Cancer Research. Curr Med Chem 2023; 30:953-973. [PMID: 35400317 DOI: 10.2174/0929867329666220408112735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 01/24/2022] [Accepted: 02/07/2022] [Indexed: 11/22/2022]
Abstract
Cancer diseases have been one of the biggest health threats for the last two decades. Approximately 9% of all diagnosed cancers are skin cancers, including melanoma and non-melanoma. In all cancer cases, early diagnosis is essential to achieve efficient treatment. New solutions and advanced techniques for rapid diagnosis are constantly being sought. Aptamers are single-stranded RNA or DNA synthetic sequences or peptides, which offer novel possibilities to this area of research by specifically binding selected molecules, the so-called cancer biomarkers. Nowadays, they are widely used as diagnostic probes in imaging and targeted therapy. In this review, we have summarized the recently made advances in diagnostics and treatment of skin cancers, which have been achieved by combining aptamers with basic or modern technologies.
Collapse
Affiliation(s)
- Adrianna Maria Piasek
- Laboratory of Biomolecular Interactions Studies, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Paulina Musolf
- Laboratory of Biomolecular Interactions Studies, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Anna Sobiepanek
- Laboratory of Biomolecular Interactions Studies, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| |
Collapse
|
10
|
Bruera S, Suarez-Almazor ME. The effects of glucocorticoids and immunosuppressants on cancer outcomes in checkpoint inhibitor therapy. Front Oncol 2022; 12:928390. [PMID: 36081549 PMCID: PMC9445222 DOI: 10.3389/fonc.2022.928390] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
The emergence of checkpoint inhibitors has created a paradigm shift for the treatment of various malignancies. However, although these therapies are associated with improved survival rates, they also carry the risk of immune-related adverse events (irAEs). Moderate to severe irAEs are typically treated with glucocorticoids, sometimes with the addition of immunosuppressants as steroid-sparing therapy. However, it is unclear how glucocorticoids and immunosuppressants may impact cancer survival and the efficacy of immune checkpoint therapy on cancer. In this narrative review, we discuss the effects of glucocorticoids and immunosuppressants including methotrexate, hydroxychloroquine, azathioprine, mycophenolate mofetil, tumor-necrosis factor (TNF)-inhibitors, interleukin-6 inhibitors, interleukin-1 inhibitors, abatacept, rituximab, and Janus kinase inhibitors (JAKi) on cancer-specific outcomes in the setting of immune checkpoint inhibitor use.
Collapse
Affiliation(s)
- Sebastian Bruera
- Section of Immunology, Allergy and Rheumatology, Baylor College of Medicine, Houston, TX, United States
| | - Maria E. Suarez-Almazor
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- *Correspondence: Maria E. Suarez-Almazor,
| |
Collapse
|
11
|
Talianová V, Kejík Z, Kaplánek R, Veselá K, Abramenko N, Lacina L, Strnadová K, Dvořánková B, Martásek P, Masařík M, Megová MH, Bušek P, Křížová J, Zdražilová L, Hansíková H, Vlčák E, Filimonenko V, Šedo A, Smetana K, Jakubek M. New-Generation Heterocyclic Bis-Pentamethinium Salts as Potential Cytostatic Drugs with Dual IL-6R and Mitochondria-Targeting Activity. Pharmaceutics 2022; 14:pharmaceutics14081712. [PMID: 36015338 PMCID: PMC9416741 DOI: 10.3390/pharmaceutics14081712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/05/2022] [Accepted: 08/12/2022] [Indexed: 11/22/2022] Open
Abstract
IL-6 signaling is involved in the pathogenesis of a number of serious diseases, including chronic inflammation and cancer. Targeting of IL-6 receptor (IL-6R) by small molecules is therefore an intensively studied strategy in cancer treatment. We describe the design, synthesis, and characteristics of two new bis-pentamethinium salts 5 and 6 (meta and para) bearing indole moieties. Molecular docking studies showed that both compounds have the potential to bind IL-6R (free energy of binding −9.5 and −8.1 kcal/mol). The interaction with IL-6R was confirmed using microscale thermophoresis analyses, which revealed that both compounds had strong affinity for the IL-6R (experimentally determined dissociation constants 26.5 ± 2.5 nM and 304 ± 27.6 nM, respectively). In addition, both compounds were cytotoxic for a broad spectrum of cancer cell lines in micromolar concentrations, most likely due to their accumulation in mitochondria and inhibition of mitochondrial respiration. In summary, the structure motif of bis-pentamethinium salts represents a promising starting point for the design of novel multitargeting compounds with the potential to inhibit IL-6 signaling and simultaneously target mitochondrial metabolism in cancer cells.
Collapse
Affiliation(s)
- Veronika Talianová
- BIOCEV, First Faculty of Medicine, Charles University, CZ-252 42 Vestec, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Ke Karlovu 455/2, CZ-128 08 Prague, Czech Republic
| | - Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, CZ-252 42 Vestec, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Ke Karlovu 455/2, CZ-128 08 Prague, Czech Republic
| | - Robert Kaplánek
- BIOCEV, First Faculty of Medicine, Charles University, CZ-252 42 Vestec, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Ke Karlovu 455/2, CZ-128 08 Prague, Czech Republic
| | - Kateřina Veselá
- BIOCEV, First Faculty of Medicine, Charles University, CZ-252 42 Vestec, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Ke Karlovu 455/2, CZ-128 08 Prague, Czech Republic
| | - Nikita Abramenko
- BIOCEV, First Faculty of Medicine, Charles University, CZ-252 42 Vestec, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Ke Karlovu 455/2, CZ-128 08 Prague, Czech Republic
| | - Lukáš Lacina
- BIOCEV, First Faculty of Medicine, Charles University, CZ-252 42 Vestec, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, CZ-120 00 Prague, Czech Republic
- Department of Dermatovenerology, First Faculty of Medicine, Charles University and General University Hospital, CZ-128 08 Prague, Czech Republic
| | - Karolína Strnadová
- BIOCEV, First Faculty of Medicine, Charles University, CZ-252 42 Vestec, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, CZ-120 00 Prague, Czech Republic
| | - Barbora Dvořánková
- BIOCEV, First Faculty of Medicine, Charles University, CZ-252 42 Vestec, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, CZ-120 00 Prague, Czech Republic
| | - Pavel Martásek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Ke Karlovu 455/2, CZ-128 08 Prague, Czech Republic
| | - Michal Masařík
- BIOCEV, First Faculty of Medicine, Charles University, CZ-252 42 Vestec, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Ke Karlovu 455/2, CZ-128 08 Prague, Czech Republic
- Department of Pathological Physiology and Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Magdalena Houdová Megová
- Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, CZ-120 00 Prague, Czech Republic
| | - Petr Bušek
- Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, CZ-120 00 Prague, Czech Republic
| | - Jana Křížová
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Ke Karlovu 455/2, CZ-128 08 Prague, Czech Republic
| | - Lucie Zdražilová
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Ke Karlovu 455/2, CZ-128 08 Prague, Czech Republic
| | - Hana Hansíková
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Ke Karlovu 455/2, CZ-128 08 Prague, Czech Republic
| | - Erik Vlčák
- Institute of Molecular Genetics, Academy of Sciences, CZ-140 00 Prague, Czech Republic
| | - Vlada Filimonenko
- Institute of Molecular Genetics, Academy of Sciences, CZ-140 00 Prague, Czech Republic
| | - Aleksi Šedo
- Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, CZ-120 00 Prague, Czech Republic
| | - Karel Smetana
- BIOCEV, First Faculty of Medicine, Charles University, CZ-252 42 Vestec, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, CZ-120 00 Prague, Czech Republic
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, CZ-252 42 Vestec, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Ke Karlovu 455/2, CZ-128 08 Prague, Czech Republic
| |
Collapse
|
12
|
Cabaço LC, Tomás A, Pojo M, Barral DC. The Dark Side of Melanin Secretion in Cutaneous Melanoma Aggressiveness. Front Oncol 2022; 12:887366. [PMID: 35619912 PMCID: PMC9128548 DOI: 10.3389/fonc.2022.887366] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/25/2022] [Indexed: 12/11/2022] Open
Abstract
Skin cancers are among the most common cancers worldwide and are increasingly prevalent. Cutaneous melanoma (CM) is characterized by the malignant transformation of melanocytes in the epidermis. Although CM shows lower incidence than other skin cancers, it is the most aggressive and responsible for the vast majority of skin cancer-related deaths. Indeed, 75% of patients present with invasive or metastatic tumors, even after surgical excision. In CM, the photoprotective pigment melanin, which is produced by melanocytes, plays a central role in the pathology of the disease. Melanin absorbs ultraviolet radiation and scavenges reactive oxygen/nitrogen species (ROS/RNS) resulting from the radiation exposure. However, the scavenged ROS/RNS modify melanin and lead to the induction of signature DNA damage in CM cells, namely cyclobutane pyrimidine dimers, which are known to promote CM immortalization and carcinogenesis. Despite triggering the malignant transformation of melanocytes and promoting initial tumor growth, the presence of melanin inside CM cells is described to negatively regulate their invasiveness by increasing cell stiffness and reducing elasticity. Emerging evidence also indicates that melanin secreted from CM cells is required for the immunomodulation of tumor microenvironment. Indeed, melanin transforms dermal fibroblasts in cancer-associated fibroblasts, suppresses the immune system and promotes tumor angiogenesis, thus sustaining CM progression and metastasis. Here, we review the current knowledge on the role of melanin secretion in CM aggressiveness and the molecular machinery involved, as well as the impact in tumor microenvironment and immune responses. A better understanding of this role and the molecular players involved could enable the modulation of melanin secretion to become a therapeutic strategy to impair CM invasion and metastasis and, hence, reduce the burden of CM-associated deaths.
Collapse
Affiliation(s)
- Luís C. Cabaço
- Chronic Diseases Research Center (CEDOC), NOVA Medical School, NMS, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Ana Tomás
- Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E., Lisbon, Portugal
| | - Marta Pojo
- Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E., Lisbon, Portugal
| | - Duarte C. Barral
- Chronic Diseases Research Center (CEDOC), NOVA Medical School, NMS, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
13
|
Tengesdal IW, Dinarello A, Powers NE, Burchill MA, Joosten LAB, Marchetti C, Dinarello CA. Tumor NLRP3-Derived IL-1β Drives the IL-6/STAT3 Axis Resulting in Sustained MDSC-Mediated Immunosuppression. Front Immunol 2021; 12:661323. [PMID: 34531850 PMCID: PMC8438323 DOI: 10.3389/fimmu.2021.661323] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 08/09/2021] [Indexed: 01/05/2023] Open
Abstract
Tumors evade the immune system by inducing inflammation. In melanoma, tumor-derived IL-1β drives inflammation and the expansion of highly immunosuppressive myeloid-derived suppressor cells (MDSCs). Similar in many tumors, melanoma is also linked to the downstream IL-6/STAT3 axis. In this study, we observed that both recombinant and tumor-derived IL-1β specifically induce pSTAT3(Y705), creating a tumor-autoinflammatory loop, which amplifies IL-6 signaling in the human melanoma cell line 1205Lu. To disrupt IL-1β/IL-6/STAT3 axis, we suppressed IL-1β-mediated inflammation by inhibiting the NOD-like receptor protein 3 (NLRP3) using OLT1177, a safe-in-humans specific NLRP3 oral inhibitor. In vivo, using B16F10 melanoma, OLT1177 effectively reduced tumor progression (p< 0.01); in primary tumors, OLT1177 decreased pSTAT3(Y705) by 82% (p<0.01) and II6 expression by 53% (p<0.05). Disruption of tumor-derived NLRP3, either pharmacologically or genetically, reduced STAT3 signaling in bone marrow cells. In PMN-MDSCs isolated from tumor-bearing mice treated with OLT1177, we observed significant reductions in immunosuppressive genes such as Pdcd1l1, Arg1, Il10 and Tgfb1. In conclusion, the data presented here show that the inhibition of NLRP3 reduces IL-1β induction of pSTAT3(Y705) preventing expression of immunosuppressive genes as well as activity in PMN-MDSCs.
Collapse
Affiliation(s)
- Isak W. Tengesdal
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
- Department of Internal Medicine and Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, Netherlands
| | - Alberto Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
- Dipartimento di Biologia, Università degli Studi di Padova, Padova, Italy
| | - Nicholas E. Powers
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Matthew A. Burchill
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Leo A. B. Joosten
- Department of Internal Medicine and Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, Netherlands
| | - Carlo Marchetti
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Charles A. Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
- Department of Internal Medicine and Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
14
|
An HJ, Chon HJ, Kim C. Peripheral Blood-Based Biomarkers for Immune Checkpoint Inhibitors. Int J Mol Sci 2021; 22:9414. [PMID: 34502325 PMCID: PMC8430528 DOI: 10.3390/ijms22179414] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 02/08/2023] Open
Abstract
As cancer immunotherapy using immune checkpoint inhibitors (ICIs) is rapidly evolving in clinical practice, it is necessary to identify biomarkers that will allow the selection of cancer patients who will benefit most or least from ICIs and to longitudinally monitor patients' immune responses during treatment. Various peripheral blood-based immune biomarkers are being identified with recent advances in high-throughput multiplexed analytical technologies. The identification of these biomarkers, which can be easily detected in blood samples using non-invasive and repeatable methods, will contribute to overcoming the limitations of previously used tissue-based biomarkers. Here, we discuss the potential of circulating immune cells, soluble immune and inflammatory molecules, circulating tumor cells and DNA, exosomes, and the blood-based tumor mutational burden, as biomarkers for the prediction of immune responses and clinical benefit from ICI treatment in patients with advanced cancer.
Collapse
Affiliation(s)
- Ho Jung An
- Department of Medical Oncology, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Hong Jae Chon
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam 13496, Korea
| | - Chan Kim
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam 13496, Korea
| |
Collapse
|
15
|
Olszańska J, Pietraszek-Gremplewicz K, Nowak D. Melanoma Progression under Obesity: Focus on Adipokines. Cancers (Basel) 2021; 13:cancers13092281. [PMID: 34068679 PMCID: PMC8126042 DOI: 10.3390/cancers13092281] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/01/2021] [Accepted: 05/05/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Obesity is a rapidly growing public health problem and the reason for numerous diseases in the human body, including cancer. This article reviews the current knowledge of the effect of molecules secreted by adipose tissue-adipokines on melanoma progression. We also discuss the role of these factors as markers of incidence, metastasis, and melanoma patient survival. Understanding the functions of adipokines will lead to knowledge of whether and how obesity promotes melanoma growth. Abstract Obesity is a growing problem in the world and is one of the risk factors of various cancers. Among these cancers is melanoma, which accounts for the majority of skin tumor deaths. Current studies are looking for a correlation between obesity and melanoma. They suspect that a potential cause of its development is connected to the biology of adipokines, active molecules secreted by adipose tissue. Under physiological conditions, adipokines control many processes, including lipid and glucose homeostasis, insulin sensitivity, angiogenesis, and inflammations. However, when there is an increased amount of fat in the body, their secretion is dysregulated. This article reviews the current knowledge of the effect of adipokines on melanoma growth. This work focuses on the molecular pathways by which adipose tissue secreted molecules modify the angiogenesis, migration, invasion, proliferation, and death of melanoma cells. We also discuss the role of these factors as markers of incidence, metastasis, and melanoma patient survival. Understanding the functions of adipokines will lead to knowledge of whether and how obesity promotes melanoma growth. Further studies may contribute to the innovations of therapies and the use of adipokines as predictive and/or prognostic biomarkers.
Collapse
|
16
|
Linares J, Marín-Jiménez JA, Badia-Ramentol J, Calon A. Determinants and Functions of CAFs Secretome During Cancer Progression and Therapy. Front Cell Dev Biol 2021; 8:621070. [PMID: 33553157 PMCID: PMC7862334 DOI: 10.3389/fcell.2020.621070] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
Multiple lines of evidence are indicating that cancer development and malignant progression are not exclusively epithelial cancer cell-autonomous processes but may also depend on crosstalk with the surrounding tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs) are abundantly represented in the TME and are continuously interacting with cancer cells. CAFs are regulating key mechanisms during progression to metastasis and response to treatment by enhancing cancer cells survival and aggressiveness. The latest advances in CAFs biology are pointing to CAFs-secreted factors as druggable targets and companion tools for cancer diagnosis and prognosis. Especially, extensive research conducted in the recent years has underscored the potential of several cytokines as actionable biomarkers that are currently evaluated in the clinical setting. In this review, we explore the current understanding of CAFs secretome determinants and functions to discuss their clinical implication in oncology.
Collapse
Affiliation(s)
- Jenniffer Linares
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Juan A. Marín-Jiménez
- Department of Medical Oncology, Catalan Institute of Oncology (ICO) - L'Hospitalet de Llobregat, Barcelona, Spain
| | - Jordi Badia-Ramentol
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Alexandre Calon
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| |
Collapse
|
17
|
Was H, Cichon T, Smolarczyk R, Lackowska B, Mazur-Bialy A, Mazur M, Szade A, Dominik P, Mazan M, Kotlinowski J, Zebzda A, Kusienicka A, Kieda C, Dulak J, Jozkowicz A. Effect of Heme Oxygenase-1 on Melanoma Development in Mice-Role of Tumor-Infiltrating Immune Cells. Antioxidants (Basel) 2020; 9:E1223. [PMID: 33287312 PMCID: PMC7761646 DOI: 10.3390/antiox9121223] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/25/2020] [Accepted: 12/01/2020] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE Heme oxygenase-1 (HO-1) is a cytoprotective, proangiogenic and anti-inflammatory enzyme that is often upregulated in tumors. Overexpression of HO-1 in melanoma cells leads to enhanced tumor growth, augmented angiogenesis and resistance to anticancer treatment. The effect of HO-1 in host cells on tumor development is, however, hardly known. METHODS AND RESULTS To clarify the effect of HO-1 expression in host cells on melanoma progression, C57BL/6xFvB mice of different HO-1 genotypes, HO-1+/+, HO-1+/-, and HO-1-/-, were injected with the syngeneic wild-type murine melanoma B16(F10) cell line. Lack of HO-1 in host cells did not significantly influence the host survival. Nevertheless, in comparison to the wild-type counterparts, the HO-1+/- and HO-1-/- males formed bigger tumors, and more numerous lung nodules; in addition, more of them had liver and spleen micrometastases. Females of all genotypes developed at least 10 times smaller tumors than males. Of importance, the growth of primary and secondary tumors was completely blocked in HO-1+/+ females. This was related to the increased infiltration of leukocytes (mainly lymphocytes T) in primary tumors. CONCLUSIONS Although HO-1 overexpression in melanoma cells can enhance tumor progression in mice, its presence in host cells, including immune cells, can reduce growth and metastasis of melanoma.
Collapse
Affiliation(s)
- Halina Was
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (M.M.); (A.S.); (P.D.); (M.M.); (J.K.); (A.K.); (J.D.); (A.J.)
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, 04-141 Warsaw, Poland;
| | - Tomasz Cichon
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (T.C.); (R.S.)
| | - Ryszard Smolarczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (T.C.); (R.S.)
| | - Bozena Lackowska
- Department of Pathology, Oncology Center, 31-115 Krakow, Poland;
| | - Agnieszka Mazur-Bialy
- Department of Ergonomics and Exercise Physiology, Faculty of Health Science, Jagiellonian University Medical College, 31-126 Krakow, Poland;
| | - Magdalena Mazur
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (M.M.); (A.S.); (P.D.); (M.M.); (J.K.); (A.K.); (J.D.); (A.J.)
| | - Agata Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (M.M.); (A.S.); (P.D.); (M.M.); (J.K.); (A.K.); (J.D.); (A.J.)
| | - Pawel Dominik
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (M.M.); (A.S.); (P.D.); (M.M.); (J.K.); (A.K.); (J.D.); (A.J.)
| | - Milena Mazan
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (M.M.); (A.S.); (P.D.); (M.M.); (J.K.); (A.K.); (J.D.); (A.J.)
| | - Jerzy Kotlinowski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (M.M.); (A.S.); (P.D.); (M.M.); (J.K.); (A.K.); (J.D.); (A.J.)
| | - Anna Zebzda
- Transplantation Centre, Jagiellonian University, 30-663 Krakow, Poland;
| | - Anna Kusienicka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (M.M.); (A.S.); (P.D.); (M.M.); (J.K.); (A.K.); (J.D.); (A.J.)
| | - Claudine Kieda
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, 04-141 Warsaw, Poland;
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (M.M.); (A.S.); (P.D.); (M.M.); (J.K.); (A.K.); (J.D.); (A.J.)
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (M.M.); (A.S.); (P.D.); (M.M.); (J.K.); (A.K.); (J.D.); (A.J.)
| |
Collapse
|
18
|
Kodet O, Kučera J, Strnadová K, Dvořánková B, Štork J, Lacina L, Smetana K. Cutaneous melanoma dissemination is dependent on the malignant cell properties and factors of intercellular crosstalk in the cancer microenvironment (Review). Int J Oncol 2020; 57:619-630. [PMID: 32705148 PMCID: PMC7384852 DOI: 10.3892/ijo.2020.5090] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 06/15/2020] [Indexed: 12/17/2022] Open
Abstract
The incidence of cutaneous malignant melanoma has been steadily increasing worldwide for several decades. This phenomenon seems to follow the trend observed in many types of malignancies caused by multiple significant factors, including ageing. Despite the progress in cutaneous malignant melanoma therapeutic options, the curability of advanced disease after metastasis represents a serious challenge for further research. In this review, we summarise data on the microenvironment of cutaneous malignant melanoma with emphasis on intercellular signalling during the disease progression. Malignant melanocytes with features of neural crest stem cells interact with non‑malignant populations within this microenvironment. We focus on representative bioactive factors regulating this intercellular crosstalk. We describe the possible key factors and signalling cascades responsible for the high complexity of the melanoma microenvironment and its premetastatic niches. Furthermore, we present the concept of melanoma early becoming a systemic disease. This systemic effect is presented as a background for the new horizons in the therapy of cutaneous melanoma.
Collapse
Affiliation(s)
- Ondřej Kodet
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague 2
- Department of Dermatovenereology, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague
- Biotechnology and Biomedicine Center of the Academy of Sciences and Charles University in Vestec (BIOCEV), First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
| | - Jan Kučera
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague 2
- Department of Dermatovenereology, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague
| | - Karolína Strnadová
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague 2
- Biotechnology and Biomedicine Center of the Academy of Sciences and Charles University in Vestec (BIOCEV), First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
| | - Barbora Dvořánková
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague 2
- Biotechnology and Biomedicine Center of the Academy of Sciences and Charles University in Vestec (BIOCEV), First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
| | - Jiří Štork
- Department of Dermatovenereology, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague
| | - Lukáš Lacina
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague 2
- Department of Dermatovenereology, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague
- Biotechnology and Biomedicine Center of the Academy of Sciences and Charles University in Vestec (BIOCEV), First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
| | - Karel Smetana
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague 2
- Biotechnology and Biomedicine Center of the Academy of Sciences and Charles University in Vestec (BIOCEV), First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
| |
Collapse
|
19
|
Yao L, Jia G, Lu L, Bao Y, Ma W. Factors affecting tumor responders and predictive biomarkers of toxicities in cancer patients treated with immune checkpoint inhibitors. Int Immunopharmacol 2020; 85:106628. [PMID: 32474388 DOI: 10.1016/j.intimp.2020.106628] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/24/2020] [Accepted: 05/20/2020] [Indexed: 12/20/2022]
Abstract
Cancer immunotherapy has brought a great revolution in the treatment of advanced human cancer. Immune checkpoint inhibitors (ICIs) that target cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and the programmed cell death protein 1 pathway (PD-1/PD-L1) have been widely administrated in the past years and demonstrated promising in a variety of malignancies. While some patients show benefit from ICIs, others do not respond or even develop resistance to these therapies. Among the responders, the treatments are consequently accompanied with immune-related adverse effects (irAEs), which are diverse in their effected organs, degree of severity and timing. Some of the toxicities are fatal and result in discontinuance of immunotherapy. The toxicity profile from anti-CTLA-4 to anti-PD-1/PD-L1 immunotherapies is distinct from those caused by conventional anticancer therapies, though their presentation may be similar. In order to better help clinicians recognize, monitor and manage irAEs in a growing population of cancer patients who are receiving ICI therapy, this article summarizes the FDA approved ICIs and focuses on (1) existing toxic evidence related to ICIs, (2) occurrence of irAEs, (3) factors influencing tumor responders treated with ICIs, (4) predictive biomarkers of irAEs, and (5) new potential mechanisms of resistance to ICI therapy.
Collapse
Affiliation(s)
- Liqin Yao
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital, Huzhou University School of Medicine, Huzhou, Zhejiang 313000, China
| | - Gang Jia
- Department of Medical Oncology, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, School of Medicine, Center for Biomedical Data Science, Yale Cancer Center, Yale University, 60 College Street, New Haven, CT 06520, USA
| | - Ying Bao
- Department of General Surgery, The First Affiliated Hospital, Huzhou University School of Medicine, Huzhou, Zhejiang 313000, China
| | - Wenxue Ma
- Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
20
|
Vlaykova T, Kurzawski M, Tacheva T, Dimov D, Anastasov A, Vlaykova D, Miteva A, O'donoghue N, Drozdzik M. Effects of the IL6 -174G>C promoter polymorphism and IL-6 serum levels on the progression of cutaneous malignant melanoma. Oncol Lett 2020; 20:1781-1791. [PMID: 32724421 PMCID: PMC7377025 DOI: 10.3892/ol.2020.11740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 03/26/2020] [Indexed: 11/05/2022] Open
Abstract
Cutaneous malignant melanoma (CMM) is one of the most immunogenic types of cancer, with a 6-fold higher rate of spontaneous regression than any other malignancy. In addition to responsiveness to different immunotherapies, the immunogenicity of CMM highlights the important role of the host immune system in the response to CMM. The present study aimed to explore the role of two functional promoter polymorphisms [IL6 -174G>C (rs1800785) and TNFA -308G>A (rs1800629)] in the regulation of the genes encoding the pro-inflammatory cytokines interleukin (IL)-6 and tumor necrosis factor-α, specifically in patients with CMM. A total of 76 patients with CMM and 200 control subjects were genotyped using PCR-restriction fragment length polymorphism. The genotype frequencies for both single nucleotide polymorphisms (SNPs) did not differ significantly between the patients and controls (P=0.358 and P=0.810 for IL6 and TNFA, respectively). However, compared with carriers of C-allele genotypes (CG+CC), patients with the IL6 -174GG genotype exhibited more advanced melanoma (Clark scale ≥3; P=0.037) and shorter survival times, particularly those who worked outdoors (in conditions with increased sunlight exposure; P=0.016). Furthermore, the serum IL-6 levels of patients with CMM were significantly higher than those of the control subjects, which were associated with unfavorable blood and serum characteristics and tumor progression (development of new distant metastases; P=0.004), and with a shorter overall survival time (P=0.042). Using a Cox proportional hazard model, the IL6 -174GG genotype was found to be an independent prognostic factor for reduced survival time (P=0.030), together with sex (being male; P=0.004) and occupations with higher exposure to sunlight (P=0.047). In conclusion, the results of the present study indicated that the promoter polymorphisms IL6 -174G>C and TNFA -308G>A are not predisposing factors for CMM. However, the IL6 -174G>C SNP and IL-6 serum concentrations are likely to influence the progression of the disease, and the GG genotype and higher IL-6 serum levels may indicate shorter survival.
Collapse
Affiliation(s)
- Tatyana Vlaykova
- Department of Chemistry and Biochemistry, Medical Faculty, Trakia University, Stara Zagora 6000, Bulgaria
| | - Mateuzh Kurzawski
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin 70-204, Poland
| | - Tanya Tacheva
- Department of Chemistry and Biochemistry, Medical Faculty, Trakia University, Stara Zagora 6000, Bulgaria
| | - Dimo Dimov
- Department of Chemistry and Biochemistry, Medical Faculty, Trakia University, Stara Zagora 6000, Bulgaria
| | - Asen Anastasov
- Department of Chemistry and Biochemistry, Medical Faculty, Trakia University, Stara Zagora 6000, Bulgaria
| | - Denitsa Vlaykova
- Department of Chemistry and Biochemistry, Medical Faculty, Trakia University, Stara Zagora 6000, Bulgaria
| | - Ani Miteva
- Department of Medical Ethics and Law, Faculty of Public Health, Medical University of Sofia, Sofia 1431, Bulgaria
| | - Niamh O'donoghue
- Department of Chemistry and Biochemistry, Medical Faculty, Trakia University, Stara Zagora 6000, Bulgaria
| | - Marek Drozdzik
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin 70-204, Poland
| |
Collapse
|
21
|
Laino AS, Woods D, Vassallo M, Qian X, Tang H, Wind-Rotolo M, Weber J. Serum interleukin-6 and C-reactive protein are associated with survival in melanoma patients receiving immune checkpoint inhibition. J Immunother Cancer 2020; 8:jitc-2020-000842. [PMID: 32581042 PMCID: PMC7312339 DOI: 10.1136/jitc-2020-000842] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Inflammatory mediators, including acute phase reactants and cytokines, have been reported to be associated with clinical efficacy in patients with melanoma and other cancers receiving immune checkpoint inhibitors (ICI). Analyses of patient sera from three large phase II/III randomized ICI trials, one of which included a chemotherapy arm, were performed to assess whether baseline levels of C-reactive protein (CRP), interleukin-6 (IL-6) or neutrophil/lymphocyte (N/L) ratios were prognostic or predictive. PATIENTS AND METHODS Baseline and on-treatment sera were analyzed by multiplex protein assays from immunotherapy-naïve patients with metastatic melanoma randomized 1:1 on the Checkmate-064 phase II trial of sequential administration of nivolumab followed by ipilimumab or the reverse sequence. Baseline sera, and peripheral blood mononuclear cells using automated cell counting, were analyzed from treatment-naïve patients who were BRAF wild-type and randomly allocated 1:1 to receive nivolumab or dacarbazine on the phase III Checkmate-066 trial, and from treatment-naïve patients allocated 1:1:1 to receive nivolumab, ipilimumab or both ipilimumab and nivolumab on the phase III Checkmate-067 trial. RESULTS Higher baseline levels of IL-6 and the N/L ratio, and to a lesser degree, CRP were associated with shorter survival in patients receiving ICI or chemotherapy. Increased on-treatment levels of IL-6 in patients on the Checkmate-064 study were also associated with shorter survival. IL-6 levels from patients on Checkmate-064, Checkmate-066 and Checkmate-067 were highly correlated with levels of CRP and the N/L ratio. CONCLUSION IL-6, CRP and the N/L ratio are prognostic factors with higher levels associated with shorter overall survival in patients with metastatic melanoma receiving ICI or chemotherapy in large randomized trials. In a multi-variable analysis of the randomized phase III Checkmate-067 study, IL-6 was a significant prognostic factor for survival.
Collapse
Affiliation(s)
- Andressa S Laino
- Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - David Woods
- Department of Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Melinda Vassallo
- Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | | | - Hao Tang
- Bristol-Myers Squibb, Princeton, New Jersey, USA
| | | | - Jeffrey Weber
- Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| |
Collapse
|
22
|
Zu T, Wen J, Xu L, Li H, Mi J, Li H, Brakebusch C, Fisher DE, Wu X. Up-Regulation of Activating Transcription Factor 3 in Human Fibroblasts Inhibits Melanoma Cell Growth and Migration Through a Paracrine Pathway. Front Oncol 2020; 10:624. [PMID: 32373541 PMCID: PMC7187895 DOI: 10.3389/fonc.2020.00624] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/03/2020] [Indexed: 12/15/2022] Open
Abstract
The treatment of melanoma has remained a difficult challenge. Targeting the tumor stroma has recently attracted attention for developing novel strategies for melanoma therapy. Activating transcription factor 3 (ATF3) plays a crucial role in regulating tumorigenesis and development, but whether the expression of ATF3 in human dermal fibroblasts (HDFs) can affect melanoma development hasn't been studied. Our results show that ATF3 expression is downregulated in stromal cells of human melanoma. HDFs expressing high levels of ATF3 suppressed the growth and migration of melanoma cells in association with downregulation of different cytokines including IL-6 in vitro. In vivo, HDFs with high ATF3 expression reduced tumor formation. Adding recombinant IL-6 to melanoma cells reversed those in vitro and in vivo effects, suggesting that ATF3 expression by HDFs regulates melanoma progression through the IL-6/STAT3 pathway. More importantly, HDFs pretreated with cyclosporine A or phenformin to induce ATF3 expression inhibited melanoma cell growth in vitro and in vivo. In summary, our study reveals that ATF3 suppresses human melanoma growth and that inducing the expression of ATF3 in HDFs can inhibit melanoma growth, a new potential melanoma therapeutic approach.
Collapse
Affiliation(s)
- Tingjian Zu
- Department of Tissue Engineering and Regeneration, School and Hospital of Stomatology, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Jie Wen
- Department of Tissue Engineering and Regeneration, School and Hospital of Stomatology, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Lin Xu
- Department of Tissue Engineering and Regeneration, School and Hospital of Stomatology, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China.,Department of Orthodontics, Liaocheng People's Hospital, Liaocheng, China
| | - Hui Li
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China
| | - Jun Mi
- Department of Tissue Engineering and Regeneration, School and Hospital of Stomatology, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Hui Li
- Department of Hematology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Cord Brakebusch
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - David E Fisher
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Xunwei Wu
- Department of Tissue Engineering and Regeneration, School and Hospital of Stomatology, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China.,Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
23
|
Golan T, Parikh R, Jacob E, Vaknine H, Zemser-Werner V, Hershkovitz D, Malcov H, Leibou S, Reichman H, Sheinboim D, Percik R, Amar S, Brenner R, Greenberger S, Kung A, Khaled M, Levy C. Adipocytes sensitize melanoma cells to environmental TGF-β cues by repressing the expression of miR-211. Sci Signal 2019; 12:12/591/eaav6847. [PMID: 31337739 DOI: 10.1126/scisignal.aav6847] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Transforming growth factor-β (TGF-β) superfamily members are critical signals in tissue homeostasis and pathogenesis. Melanoma grows in the epidermis and invades the dermis before metastasizing. This disease progression is accompanied by increased sensitivity to microenvironmental TGF-β. Here, we found that skin fat cells (adipocytes) promoted metastatic initiation by sensitizing melanoma cells to TGF-β. Analysis of melanoma clinical samples revealed that adipocytes, usually located in the deeper hypodermis layer, were present in the upper dermis layer within proximity to in situ melanoma cells, an observation that correlated with disease aggressiveness. In a coculture system, adipocytes secreted the cytokines IL-6 and TNF-α, which induced a proliferative-to-invasive phenotypic switch in melanoma cells by repressing the expression of the microRNA miR-211. In a xenograft model, miR-211 exhibited a dual role in melanoma progression, promoting cell proliferation while inhibiting metastatic spread. Bioinformatics and molecular analyses indicated that miR-211 directly targeted and repressed the translation of TGFBR1 mRNA, which encodes the type I TGF-β receptor. Hence, through this axis of cytokine-mediated repression of miR-211, adipocytes increased the abundance of the TGF-β receptor in melanoma cells, thereby enhancing cellular responsiveness to TGF-β ligands. The induction of TGF-β signaling, in turn, resulted in a proliferative-to-invasive phenotypic switch in cultured melanoma cells. Pharmacological inhibition of TGF-β prevented these effects. Our findings further reveal a molecular link between fat cells and metastatic progression in melanoma that might be therapeutically targeted in patients.
Collapse
Affiliation(s)
- Tamar Golan
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Roma Parikh
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Etai Jacob
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel.,Department of Structural Biology, Weizmann Institute of Science, Rehovot 7610001, Israel.,Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Hananya Vaknine
- Institute of Pathology, E. Wolfson Medical Center, Holon 58100, Israel
| | | | - Dov Hershkovitz
- Institute of Pathology, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Hagar Malcov
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Stav Leibou
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Hadar Reichman
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Danna Sheinboim
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ruth Percik
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.,Institute of Endocrinology, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Sarah Amar
- Institute of Pathology, E. Wolfson Medical Center, Holon 58100, Israel
| | - Ronen Brenner
- Institute of Pathology, E. Wolfson Medical Center, Holon 58100, Israel
| | | | - Andrew Kung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Mehdi Khaled
- INSERM 1186, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Carmit Levy
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
24
|
Nakamura Y. Biomarkers for Immune Checkpoint Inhibitor-Mediated Tumor Response and Adverse Events. Front Med (Lausanne) 2019; 6:119. [PMID: 31192215 PMCID: PMC6549005 DOI: 10.3389/fmed.2019.00119] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/15/2019] [Indexed: 12/19/2022] Open
Abstract
In the last decade, inhibitors targeting immune checkpoint molecules such as cytotoxic T-lymphocyte antigen 4 (CTLA-4), programmed cell death 1 (PD-1), and programmed cell death-ligand 1 (PD-L1) brought about a major paradigm shift in cancer treatment. These immune checkpoint inhibitors (ICIs) improved the overall survival of a variety of cancer such as malignant melanoma and non-small lung cancer. In addition, numerous clinical trials for additional indication of ICIs including adjuvant and neo-adjuvant therapies are also currently ongoing. Therefore, more and more patients will receive ICIs in the future. However, despite the improved outcome of the cancer treatment by ICIs, the efficacy remains still limited and tumor regression have not been obtained in many cancer patients. In addition, treatment with ICIs is also associated with substantial toxicities, described as immune-related adverse events (irAEs). Therefore, biomarkers to predict tumor response and occurrence of irAEs by the treatment with ICIs are required to avoid overtreatment of ICIs and minimize irAEs development. Whereas, numerous factors have been reported as potential biomarkers for tumor response to ICIs, factors for predicting irAE have been less reported. In this review, we show recent advances in the understanding of biomarkers for tumor response and occurrence of irAEs in cancer patients treated with ICIs.
Collapse
Affiliation(s)
- Yoshiyuki Nakamura
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
25
|
Bridge JA, Lee JC, Daud A, Wells JW, Bluestone JA. Cytokines, Chemokines, and Other Biomarkers of Response for Checkpoint Inhibitor Therapy in Skin Cancer. Front Med (Lausanne) 2018; 5:351. [PMID: 30631766 PMCID: PMC6315146 DOI: 10.3389/fmed.2018.00351] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/29/2018] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy for skin malignancies has ushered in a new era for cancer treatments by demonstrating unprecedented durable responses in the setting of metastatic Melanoma. Consequently, checkpoint inhibitors are now the first-line treatment of metastatic melanoma and widely used as adjuvant therapy for stage III disease. With the observation that higher tumor mutational burden correlates with a better response, checkpoint inhibitors are tested in other skin cancer types of known high tumor mutational burden with promising results and recently became the first-ever FDA-approved treatment for metastatic Merkel cell carcinoma. The emerging new standards-of-care will necessitate more precise biomarkers and predictors for treatment response and immune-related adverse events. Measurable immune-related mediators are currently under investigation as factors that promote or block the response to cancer immunotherapy and may provide insights into the underlying immune response to the tumor. Cytokines and chemokines are such mediators and are crucial for facilitating the recruitment and activation of specific subsets of leukocytes within the microenvironment of skin cancers. The exact mechanisms of how these meditators, both immunological and non-immunological, operate in the tumor microenvironment is an area of active research, so to reliable biomarkers of responses to cancer immunotherapy. Here, we will review and summarize the expanding body of literature for immune-related biomarkers pertaining to Melanoma, Basal cell carcinoma, Squamous cell carcinoma, and Merkel cell carcinoma, highlighting clinically relevant checkpoint inhibitor therapy biomarker advancements.
Collapse
Affiliation(s)
- Jennifer A Bridge
- Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
| | - James C Lee
- Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, United States
| | - Adil Daud
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, United States
| | - James W Wells
- The Faculty of Medicine, The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Jeffrey A Bluestone
- Sean N. Parker Autoimmune Research Laboratory, Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
26
|
Ceylan O, Mishra GK, Yazici M, Qureshi A, Niazi JH, Gurbuz Y. A Hand-Held Point-of-Care Biosensor Device for Detection of Multiple Cancer and Cardiac Disease Biomarkers Using Interdigitated Capacitive Arrays. IEEE TRANSACTIONS ON BIOMEDICAL CIRCUITS AND SYSTEMS 2018; 12:1440-1449. [PMID: 30605085 DOI: 10.1109/tbcas.2018.2870297] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
This paper presents a hand-held point-of-care device that incorporates a lab-on-a-chip module with interdigitated capacitive biosensors for label-free detection of multiple cancer and cardiovascular disease biomarkers. The developed prototype is comprised of a cartridge incorporating capacitive biodetection sensors, a sensitive capacitive readout electronics enclosed in a hand-held unit, and data analysis software calculating the concentration of biomarkers using previously stored reference database. The capacitive biodetection sensors are made of interdigitated circular electrodes, which are preactivated with single (for detecting one biomarker) or multiple specific antibodies (for detecting multiple disease biomarkers). Detection principle of capacitive biosensor is based on measuring the level of capacitance change between interdigitated electrode pairs induced by the change in dielectric constant due to affinity-based electron exchange in between antibodies/antigens and electrodes. The more antibody-antigens binding occurs, the more capacitance change is measured due to the change in dielectric constant of the capacitance media. The device uses preactivated ready-to-use cartridges embedded with capacitive biosensors with shelf-life of three months under optimal conditions, and is capable of onsite diagnosis and can report the result in less than 30 min. The device is verified with real patient blood samples for six different disease biomarkers.
Collapse
|
27
|
Preisner F, Leimer U, Sandmann S, Zoernig I, Germann G, Koellensperger E. Impact of Human Adipose Tissue-Derived Stem Cells on Malignant Melanoma Cells in An In Vitro Co-culture Model. Stem Cell Rev Rep 2018; 14:125-140. [PMID: 29064018 DOI: 10.1007/s12015-017-9772-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
This study focuses on the interactions of human adipose tissue-derived stem cells (ADSCs) and malignant melanoma cells (MMCs) with regard to future cell-based skin therapies. The aim was to identify potential oncological risks as ADSCs could unintentionally be sited within the proximity of the tumor microenvironment of MMCs. An indirect co-culture model was used to analyze interactions between ADSCs and four different established melanoma cell lines (G-361, SK-Mel-5, MeWo and A2058) as well as two low-passage primary melanoma cell cultures (M1 and M2). Doubling time, migration and invasion, angiogenesis, quantitative real-time PCR of 229 tumor-associated genes and multiplex protein assays of 20 chemokines and growth factors and eight matrix metalloproteinases (MMPs) were evaluated. Co-culture with ADSCs significantly increased migration capacity of G-361, SK-Mel-5, A2058, MeWo and M1 and invasion capacity of G-361, SK-Mel-5 and A2058 melanoma cells. Furthermore, conditioned media from all ADSC-MMC-co-cultures induced tube formation in an angiogenesis assay in vitro. Gene expression analysis of ADSCs and MMCs, especially of low-passage melanoma cell cultures, revealed an increased expression of various genes with tumor-promoting activities, such as CXCL12, PTGS2, IL-6, and HGF upon ADSC-MMC-co-culture. In this context, a significant increase (up to 5,145-fold) in the expression of numerous tumor-associated proteins could be observed, e.g. several pro-angiogenic factors, such as VEGF, IL-8, and CCL2, as well as different matrix metalloproteinases, especially MMP-2. In conclusion, the current report clearly demonstrates that a bi-directional crosstalk between ADSCs and melanoma cells can enhance different malignant properties of melanoma cells in vitro.
Collapse
Affiliation(s)
- Fabian Preisner
- ETHIANUM - Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine, Voßstraße 6, 69115, Heidelberg, Germany
| | - Uwe Leimer
- ETHIANUM - Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine, Voßstraße 6, 69115, Heidelberg, Germany
| | - Stefanie Sandmann
- ETHIANUM - Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine, Voßstraße 6, 69115, Heidelberg, Germany
| | - Inka Zoernig
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Im Neuenheimer Feld 460, 60120, Heidelberg, Germany
| | - Guenter Germann
- ETHIANUM - Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine, Voßstraße 6, 69115, Heidelberg, Germany
| | - Eva Koellensperger
- ETHIANUM - Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine, Voßstraße 6, 69115, Heidelberg, Germany.
| |
Collapse
|
28
|
Fujii T, Naing A, Rolfo C, Hajjar J. Biomarkers of response to immune checkpoint blockade in cancer treatment. Crit Rev Oncol Hematol 2018; 130:108-120. [PMID: 30196907 DOI: 10.1016/j.critrevonc.2018.07.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/12/2018] [Accepted: 07/30/2018] [Indexed: 12/18/2022] Open
Abstract
Immune checkpoint inhibitors (ICPis) are emerging as the new corner stone of cancer treatment due to their ability to produce durable responses in patients with various cancers. But, objective responses to ICPis vary among each type of cancer. Further, treatment with ICPis is often associated with risk of developing immune-related adverse event, which are potentially life-threatening if untreated, indicating a need for patient selection. However, given the complexity of the tumor microenvironment and the dynamic interaction between tumor and immune cells, development of robust biomarkers to predict patients who are likely to respond to treatment with ICPis remains a challenge. In this review we present an overview of the immune monitoring strategies that are currently in use to enable appropriate patient selection.
Collapse
Affiliation(s)
- Takeo Fujii
- University of Hawaii Cancer Center, Honolulu, HI, United States
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Christian Rolfo
- Thoracic Medical Oncology, Early Clinical Trials, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center (UMGCCC), Baltimore, MD, United States
| | - Joud Hajjar
- Section of Immunology, Allergy and Rheumatology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, United States.
| |
Collapse
|
29
|
Valpione S, Pasquali S, Campana LG, Piccin L, Mocellin S, Pigozzo J, Chiarion-Sileni V. Sex and interleukin-6 are prognostic factors for autoimmune toxicity following treatment with anti-CTLA4 blockade. J Transl Med 2018; 16:94. [PMID: 29642948 PMCID: PMC5896157 DOI: 10.1186/s12967-018-1467-x] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/30/2018] [Indexed: 11/30/2022] Open
Abstract
Background Ipilimumab is a licensed immunotherapy for metastatic melanoma patients and, in the US, as adjuvant treatment for high risk melanoma radically resected. The use of ipilimumab is associated with a typical but unpredictable pattern of side effects. The purpose of this study was to identify clinical features and blood biomarkers capable of predicting ipilimumab related toxicity. Methods We performed a prospective study aimed at analyzing potential clinical and biological markers associated with immune-related toxicity in patients treated with ipilimumab (3 mg/kg, q3w). We enrolled 140 consecutive melanoma patients treated with ipilimumab for metastatic disease. The following prospectively collected data were utilized: patient characteristics, previous therapies, level of circulating biomarkers associated with tumour burden or immune-inflammation status (lactic dehydrogenase, C-reactive protein, β2-microglobulin, vascular endothelial growth factor, interleukin-2, interleukin-6, S-100, alkaline phosphatase, transaminases) and blood cells subsets (leukocyte and lymphocyte subpopulations). Logistic regression was used for multivariate analysis of data. Results Out of 140 patients, 36 (26%) experienced a severe adverse event, 33 (24%) discontinued treatment for severe toxicity. Among the immune-profile biomarkers analyzed, only interleukin-6 was associated with the risk of toxicity. Female patients had a further increase of immune-related adverse events. Low baseline interleukin-6 serum levels (OR = 2.84, 95% CI 1.34–6.03, P = 0.007) and sex female (OR = 1.5, 95% CI 1.06–2.16 P = 0.022) and were significant and independent risk factors for immune related adverse events. Conclusions Baseline IL6 serum levels and female sex were significantly and independently associated with higher risk of severe toxicity and could be exploited in clinical practice to personalize toxicity surveillance in patients treated with ipilimumab. Electronic supplementary material The online version of this article (10.1186/s12967-018-1467-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sara Valpione
- CRUK Manchester Institute and The Christie NHS Foundation Trust, The University of Manchester, Manchester, M20 4GJ, UK. .,Melanoma and Esophageal Cancer Unit, Istituto Oncologico Veneto-IRCCS, Via Gattamelata 64, 35128, Padua, Italy. .,Department of Surgery, Oncology and Gastroenterology, University of Padova, 64 Gattamelata St, 35128, Padua, Italy.
| | - Sandro Pasquali
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 64 Gattamelata St, 35128, Padua, Italy.,Surgical Oncology, Veneto Oncology Institute, Via Gattamelata 64, 35128, Padua, Italy.,Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, via G Venezian 1, 20133, Milan, Italy
| | - Luca Giovanni Campana
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 64 Gattamelata St, 35128, Padua, Italy.,Surgical Oncology, Veneto Oncology Institute, Via Gattamelata 64, 35128, Padua, Italy
| | - Luisa Piccin
- Melanoma and Esophageal Cancer Unit, Istituto Oncologico Veneto-IRCCS, Via Gattamelata 64, 35128, Padua, Italy.,Department of clinical medicine and surgery, Medical Oncology Unit, University of Naples Federico II, Via S Pansini 5, 80131, Naples, Italy
| | - Simone Mocellin
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 64 Gattamelata St, 35128, Padua, Italy.,Surgical Oncology, Veneto Oncology Institute, Via Gattamelata 64, 35128, Padua, Italy
| | - Jacopo Pigozzo
- Melanoma and Esophageal Cancer Unit, Istituto Oncologico Veneto-IRCCS, Via Gattamelata 64, 35128, Padua, Italy
| | - Vanna Chiarion-Sileni
- Melanoma and Esophageal Cancer Unit, Istituto Oncologico Veneto-IRCCS, Via Gattamelata 64, 35128, Padua, Italy
| |
Collapse
|
30
|
Demonstration of a WNT5A-IL-6 positive feedback loop in melanoma cells: Dual interference of this loop more effectively impairs melanoma cell invasion. Oncotarget 2018; 7:37790-37802. [PMID: 27191257 PMCID: PMC5122349 DOI: 10.18632/oncotarget.9332] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 04/28/2016] [Indexed: 01/07/2023] Open
Abstract
Increased expression and signalling of WNT5A and interleukin-6 (IL-6) have both been shown to promote melanoma progression. Here, we investigated the proposed existence of a WNT5A-IL-6 positive feedback loop that drives melanoma migration and invasion. First, the HOPP algorithm revealed that the invasive phenotype of cultured melanoma cells was significantly correlated with increased expression of WNT5A or IL-6. In three invasive melanoma cell lines, endogenous WNT5A protein expression was related to IL-6 protein secretion. Knockdown with anti-IL-6 siRNAs or treating WM852 melanoma cells with a neutralising anti-IL-6 antibody reduced WNT5A protein expression. Conversely, the silencing of WNT5A expression by WNT5A siRNAs or treating WM852 melanoma cells with Box5 (a WNT5A antagonist) significantly reduced IL-6 secretion. Interestingly, these effects occurred at the protein level but not at the transcriptional levels. Functionally, we demonstrated that combined siRNA knockdown of WNT5A and IL-6 expression or the simultaneous inhibition of WNT5A and IL-6 signalling inhibited melanoma cell invasion more effectively than suppressing each factor individually. Together, our results demonstrate that WNT5A and IL-6 are connected through a positive feedback loop in melanoma cells and that the combined targeting of both molecules could serve as an effective therapeutic means to reduce melanoma metastasis.
Collapse
|
31
|
Lai X, Friedman A. Combination therapy for melanoma with BRAF/MEK inhibitor and immune checkpoint inhibitor: a mathematical model. BMC SYSTEMS BIOLOGY 2017; 11:70. [PMID: 28724377 PMCID: PMC5517842 DOI: 10.1186/s12918-017-0446-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 07/11/2017] [Indexed: 11/24/2022]
Abstract
BACKGROUND The B-raf gene is mutated in up to 66% of human malignant melanomas, and its protein product, BRAF kinase, is a key part of RAS-RAF-MEK-ERK (MAPK) pathway of cancer cell proliferation. BRAF-targeted therapy induces significant responses in the majority of patients, and the combination BRAF/MEK inhibitor enhances clinical efficacy, but the response to BRAF inhibitor and to BRAF/MEK inhibitor is short lived. On the other hand, treatment of melanoma with an immune checkpoint inhibitor, such as anti-PD-1, has lower response rate but the response is much more durable, lasting for years. For this reason, it was suggested that combination of BRAF/MEK and PD-1 inhibitors will significantly improve overall survival time. RESULTS This paper develops a mathematical model to address the question of the correlation between BRAF/MEK inhibitor and PD-1 inhibitor in melanoma therapy. The model includes dendritic and cancer cells, CD 4+ and CD 8+ T cells, MDSC cells, interleukins IL-12, IL-2, IL-6, IL-10 and TGF- β, PD-1 and PD-L1, and the two drugs: BRAF/MEK inhibitor (with concentration γ B ) and PD-1 inhibitor (with concentration γ A ). The model is represented by a system of partial differential equations, and is used to develop an efficacy map for the combined concentrations (γ B ,γ A ). It is shown that the two drugs are positively correlated if γ B and γ A are at low doses, that is, the growth of the tumor volume decreases if either γ B or γ A is increased. On the other hand, the two drugs are antagonistic at some high doses, that is, there are zones of (γ B ,γ A ) where an increase in one of the two drugs will increase the tumor volume growth, rather than decrease it. CONCLUSIONS It will be important to identify, by animal experiments or by early clinical trials, the zones of (γ B ,γ A ) where antagonism occurs, in order to avoid these zones in more advanced clinical trials.
Collapse
Affiliation(s)
- Xiulan Lai
- Institute for Mathematical Sciences, Renmin University of China, Beijing, 100872 People’s Republic of China
| | - Avner Friedman
- Mathematical Bioscience Institute & Department of Mathematics, Ohio State University, Columbus, 43210 OH USA
| |
Collapse
|
32
|
Anticancer and Anti-Inflammatory Properties of Ganoderma lucidum Extract Effects on Melanoma and Triple-Negative Breast Cancer Treatment. Nutrients 2017; 9:nu9030210. [PMID: 28264501 PMCID: PMC5372873 DOI: 10.3390/nu9030210] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/03/2017] [Accepted: 02/16/2017] [Indexed: 11/18/2022] Open
Abstract
Among the most important traditional medicinal fungi, Ganoderma lucidum has been used as a therapeutic agent for the treatment of numerous diseases, including cancer, in Oriental countries. The aim of this study is to investigate the anti-inflammatory, anticancer and anti-metastatic activities of Ganoderma lucidum extracts in melanoma and triple-negative breast cancer cells. Ganoderma lucidum extracts were prepared by using common organic solvents; MDA-MB 231 and B16-F10 cell lines were adopted as cellular models for triple-negative breast cancer and melanoma and characterized for cell viability, wound-healing assay and measurement of cytokines secreted by cancer cells under pro-inflammatory conditions (incubation with lipopolysaccharide, LPS) and pretreatment with Ganoderma lucidum extract at different concentrations. Our study demonstrates, for the first time, how Ganoderma lucidum extracts can significantly inhibit the release of IL-8, IL-6, MMP-2 and MMP-9 in cancer cells under pro-inflammatory condition. Interestingly, Ganoderma lucidum extracts significantly also decrease the viability of both cancer cells in a time- and concentration-dependent manner, with abilities to reduce cell migration over time, which is correlated with a lower release of matrix metalloproteases. Taken together, these results indicate the possible use of Ganoderma lucidum extract for the therapeutic management of melanoma and human triple-negative breast cancer.
Collapse
|
33
|
Chen C, Zhang X. IRE1α-XBP1 pathway promotes melanoma progression by regulating IL-6/STAT3 signaling. J Transl Med 2017; 15:42. [PMID: 28222747 PMCID: PMC5320675 DOI: 10.1186/s12967-017-1147-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 02/04/2017] [Indexed: 12/16/2022] Open
Abstract
Background The IRE1α-XBP1 pathway is the most conserved branch of the unfolded protein response pathways, which are activated during endoplasmic reticulum (ER) stress caused by the accumulation of unfolded/misfolded proteins in the ER lumen. The IRE1α-XBP1 pathway plays a critical role in various cancers. However, the role of this pathway in melanoma cell growth remains unclear. Methods Sixty-one pairs of melanoma specimens and corresponding normal tissues from patients were stained with XBP1. Then, XBP1 splicing levels were detected in human tissues and cell lines at the mRNA level. IL-6 expression levels were determined in both melanocytes (HEMn-MP) and melanoma cells (Mel-RMu) overexpressing the spliced form of XBP1 (XBP1s). IL-6 expression was also examined in 4μ8C-treated HEMn-MP and Mel-RMu cells overexpressing IRE1α. Next, we analyzed potential XBP1s binding sites within the IL-6 promoter and conducted ChIP experiments. IL-6/STAT3 signaling was detected by western blotting. Melanoma cell proliferation was examined by CCK8 and BrdU assays. Results The mRNA and protein expression levels of XBP1s were significantly elevated in human melanoma tissues and cell lines compared with normal tissues or melanocytes, thus indicating the activation of the IRE1α-XBP1 branch in melanoma. Ectopic expression of IRE1α or XBP1s robustly enhanced IL-6 expression in HEMn-MP and Mel-RMu cells. Moreover, the inhibition of the RNase activity of IRE1α also abolished the effect of IRE1α in promoting IL-6 expression. Mechanistically, XBP1 binds the IL-6 promoter and activates its expression. Furthermore, secreted IL-6 functions in an autocrine/paracrine manner, activates the intracellular JAK/STAT3 pathway and promotes the proliferation of melanoma cells. Conclusion Our results reveal that the IRE1α-XBP1 pathway regulates Mel-RMu cell proliferation and progression by activating IL-6/STAT3 signaling.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xuejun Zhang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
34
|
Markel G, Imazio M, Koren-Morag N, Galore-Haskel G, Schachter J, Besser M, Cumetti D, Maestroni S, Altman A, Shoenfeld Y, Brucato A, Adler Y. CEACAM1 and MICA as novel serum biomarkers in patients with acute and recurrent pericarditis. Oncotarget 2017; 7:17885-95. [PMID: 26909604 PMCID: PMC4951257 DOI: 10.18632/oncotarget.7530] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 01/23/2016] [Indexed: 11/25/2022] Open
Abstract
Background The immune response plays a significant role in pericarditis, but the mechanisms of disease are poorly defined. Further, efficient monitoring and predictive clinical tools are unavailable. Carcinoembryonic antigen cell adhesion molecule 1 (CEACAM1) is an immune-inhibitory protein, while MHC class I chain related protein A (MICA) and B (MICB) have an immune-stimulating function. Methods and results Serum CEACAM1, MICA and MICB concentrations were measured by ELISA in ∼50 subjects of each group: acute pericarditis (AP), recurrent pericarditis (RP) and lupus (SLE) patients, metastatic melanoma patients as well as healthy donors. Serum CEACAM1 was dramatically elevated in AP and RP patients, but not in SLE patients, and displayed a highly accurate profile in ROC curve analyses. MICA and MICB were elevated in some pericarditis patients. All markers were enhanced in metastatic melanoma patients irrespective of neoplastic pericardial involvement. Etiology-guided analysis of RP patients showed that very low MICA levels were associated with idiopathic RP, while high MICA was associated with autoimmune and post-operative RP. Importantly, MICA was significantly associated with recurrences, independently of other potentially confounding parameters such as age, time of follow up or treatment modality. Conclusions Here we report for the first time on CEACAM1 as a potentially novel biomarker for pericarditis, as well as on MICA as an innovative prognostic marker in these patients. Determination of the roles of these immune factors, as well as their diagnostic and prognostic values should be determined in future prospective studies.
Collapse
Affiliation(s)
- Gal Markel
- Ella Lemelbaum Institute of Melanoma, Sheba Medical Center, Tel Hashomer, Israel.,Talpiot Medical Leadership Program, Sheba Medical Center, Tel Hashomer, Israel.,Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Massimo Imazio
- Cardiology Department, Maria Vittoria Hospital, Torino, Italy
| | - Nira Koren-Morag
- Department of Epidemiology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gilli Galore-Haskel
- Ella Lemelbaum Institute of Melanoma, Sheba Medical Center, Tel Hashomer, Israel
| | - Jacob Schachter
- Ella Lemelbaum Institute of Melanoma, Sheba Medical Center, Tel Hashomer, Israel
| | - Michal Besser
- Ella Lemelbaum Institute of Melanoma, Sheba Medical Center, Tel Hashomer, Israel.,Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | - Arie Altman
- Internal Medicine B, Sheba Medical Center, Tel Hashomer, Israel
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, Israel
| | | | - Yehuda Adler
- Talpiot Medical Leadership Program, Sheba Medical Center, Tel Hashomer, Israel.,Cardiac Rehabilitation Institute, Sheba Medical Center, Tel Hashomer, Israel.,Department of Internal Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
35
|
Lactate, a Neglected Factor for Diabetes and Cancer Interaction. Mediators Inflamm 2016; 2016:6456018. [PMID: 28077918 PMCID: PMC5203906 DOI: 10.1155/2016/6456018] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 11/17/2016] [Accepted: 11/23/2016] [Indexed: 12/23/2022] Open
Abstract
Increasing body of evidence suggests that there exists a connection between diabetes and cancer. Nevertheless, to date, the potential reasons for this association are still poorly understood and currently there is no clinical evidence available to direct the proper management of patients presenting with these two diseases concomitantly. Both cancer and diabetes have been associated with abnormal lactate metabolism and high level of lactate production is the key biological property of these diseases. Conversely, high lactate contribute to a higher insulin resistant status and a more malignant phenotype of cancer cells, promoting diabetes and cancer development and progression. In view of associations between diabetes and cancers, the role of high lactate production in diabetes and cancer interaction should not be neglected. Here, we review the available evidence of lactate's role in different biological characteristics of diabetes and cancer and interactive relationship between them. Understanding the molecular mechanisms behind metabolic remodeling of diabetes- and cancer-related signaling would endow novel preventive and therapeutic approaches for diabetes and cancer treatment.
Collapse
|
36
|
Chiang NY, Peng YM, Juang HH, Chen TC, Pan HL, Chang GW, Lin HH. GPR56/ADGRG1 Activation Promotes Melanoma Cell Migration via NTF Dissociation and CTF-Mediated Gα 12/13/RhoA Signaling. J Invest Dermatol 2016; 137:727-736. [PMID: 27818281 DOI: 10.1016/j.jid.2016.10.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 10/06/2016] [Accepted: 10/17/2016] [Indexed: 02/07/2023]
Abstract
GPR56/ADGRG1 is a versatile adhesion G protein-coupled receptor with diverse biological functions. GPR56 expression is variably detected in human melanoma cell lines and correlates inversely with the metastatic potential of melanoma lesions. GPR56 associates with the tetraspanins CD9 and CD81 on the melanoma cell surface. GPR56 activation by immobilized CG4 monoclonal antibody facilitates N-terminal fragment dissociation in a CD9/CD81-dependent manner specifically inducing IL-6 production, which promotes cell migration and invasion. Interestingly, expression of GPR56-C-terminal fragment alone recapitulates the antibody-induced receptor function, implicating a major role for the C-terminal fragment in GPR56 activation and signaling. Analysis of site-directed mutant receptors attests the importance of the conserved N-terminal residues of the C-terminal fragment for its self-activation. Finally, we show that the GPR56-induced signaling in melanoma cells is mediated by the Gα12/13/RhoA pathway. In summary, the expression and activation of GPR56 may modulate melanoma progression in part by inducing IL-6 production after N-terminal fragment dissociation and C-terminal fragment self-activation.
Collapse
Affiliation(s)
- Nien-Yi Chiang
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan
| | - Yen-Ming Peng
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Horng-Heng Juang
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan; Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Urology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan
| | - Tse-Ching Chen
- Department of Anatomic Pathology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan
| | - Hsiao-Lin Pan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Gin-Wen Chang
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan
| | - Hsi-Hsien Lin
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan; Department of Anatomic Pathology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan; Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
37
|
Tuccitto A, Tazzari M, Beretta V, Rini F, Miranda C, Greco A, Santinami M, Patuzzo R, Vergani B, Villa A, Manenti G, Cleris L, Giardiello D, Alison M, Rivoltini L, Castelli C, Perego M. Immunomodulatory Factors Control the Fate of Melanoma Tumor Initiating Cells. Stem Cells 2016; 34:2449-2460. [PMID: 27301067 DOI: 10.1002/stem.2413] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 04/29/2016] [Indexed: 12/19/2022]
Abstract
Melanoma is a highly heterogeneous tumor for which recent evidence supports a model of dynamic stemness. Melanoma cells might temporally acquire tumor-initiating properties or switch from a status of tumor-initiating cells (TICs) to a more differentiated one depending on the tumor context. However, factors driving these functional changes are still unknown. We focused on the role of cyto/chemokines in shaping TICs isolated directly from tumor specimens of two melanoma patients, namely Me14346S and Me15888S. We analyzed the secretion profile of TICs and of their corresponding melanoma differentiated cells and we tested the ability of cyto/chemokines to influence TIC self-renewal and differentiation. We found that TICs, grown in vitro as melanospheres, had a complex secretory profile as compared to their differentiated counterparts. Some factors, such as CCL-2 and IL-8, also produced by adherent melanoma cells and melanocytes did not influence TIC properties. Conversely, IL-6, released by differentiated cells, reduced TIC self-renewal and induced TIC differentiation while IL-10, produced by Me15888S, strongly promoted TIC self-renewal through paracrine/autocrine actions. Complete neutralization of IL-10 activity by gene silencing and antibody-mediated blocking of the IL-10Rα was required to sensitize Me15888S to IL-6-induced differentiation. For the first time these results show that functional heterogeneity of melanoma could be directly influenced by inflammatory and suppressive soluble factors, with IL-6 favoring TIC differentiation, and IL-10 supporting TIC self-renewal. Thus, understanding the tumor microenvironment (TME) role in modulating melanoma TIC phenotype is fundamental to identifying novel therapeutic targets to achieve long-lasting regression of metastatic melanoma. Stem Cells 2016;34:2449-2460.
Collapse
Affiliation(s)
- Alessandra Tuccitto
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Marcella Tazzari
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Valeria Beretta
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Francesca Rini
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Claudia Miranda
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Molecular Mechanism Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Angela Greco
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Molecular Mechanism Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Mario Santinami
- Melanoma and Sarcoma Unit, Department of Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Roberto Patuzzo
- Melanoma and Sarcoma Unit, Department of Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Barbara Vergani
- Consorzio MIA (Microscopy and Image Analysis), University of Milano-Bicocca, Milano, Italy
| | - Antonello Villa
- Consorzio MIA (Microscopy and Image Analysis), University of Milano-Bicocca, Milano, Italy
| | - Giacomo Manenti
- Department of Predictive and Preventive Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Loredana Cleris
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Daniele Giardiello
- Unit of Clinical Epidemiology and Trial Organization, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Malcolm Alison
- Centre for Tumour Biology, Barts Cancer Institute, Charterhouse Square, London, EC1M 6BQ, United Kingdom
| | - Licia Rivoltini
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Chiara Castelli
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy. .,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.
| | - Michela Perego
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| |
Collapse
|
38
|
Lippitz BE, Harris RA. Cytokine patterns in cancer patients: A review of the correlation between interleukin 6 and prognosis. Oncoimmunology 2016; 5:e1093722. [PMID: 27467926 PMCID: PMC4910721 DOI: 10.1080/2162402x.2015.1093722] [Citation(s) in RCA: 174] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 09/08/2015] [Indexed: 02/09/2023] Open
Abstract
OBJECTIVE In tumor patients, IL-6 appears to be one component of a consistent cancer-associated cytokine network resulting in both a systemic immune stimulation and a microenvironment of cancer-induced immune suppression that ultimately protects the cancer cells. IL-6 has been associated with prognosis in cancer patients, but so far a systemical analysis has not been carried out. METHODS The present meta-analysis studies the relation between IL-6 serum levels and the prognosis of cancer patients in the available clinical literature of 100 articles published between 1993 and 2013 comprising 11,583 patients. RESULTS The IL-6 serum level was described as significantly correlating with survival in 82/101 series comprising 85.6% of patients (9917/11,583) with 23 different cancer types. A total of 64 studies dichotomized patient cohorts according to various cut-off IL-6 serum levels: in 59/64 of these series corresponding to 94.5% of the reported patients (7694/8142) significant correlations between IL-6 serum level and survival were seen. The median survival of cancer patients had been determined above various cut-off levels of serum IL-6 in 24 dichotomized studies (26 cohorts). There was a highly significant inverse correlation between median survival of the cohorts with IL-6 serum level above cut-off (1272 patients) and their corresponding IL-6 cut-off values (Spearman R -0,48 p= < 0.001) following a linear regression when both parameters were log-transformed (p < 0.001). A significant correlation between increasing serum IL-6 and tumor stage or metastases was described in 39/44 studies and 91% of published patients (4221/4636) where clinical parameters had been specified. CONCLUSIONS Closely associated with the patient's clinical condition and independent of the cancer histology, the increased IL-6 serum level uniformly appears to correlate with survival as paraneoplastic condition in later cancer stages independent of the cancer type. Modifications of this paraneoplastic immune reaction may offer new therapeutic options in cancer.
Collapse
Affiliation(s)
- Bodo E Lippitz
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Sjukhuset, Karolinska Institutet , Stockholm, Sweden
| | - Robert A Harris
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Sjukhuset, Karolinska Institutet , Stockholm, Sweden
| |
Collapse
|
39
|
Tocilizumab unmasks a stage-dependent interleukin-6 component in statin-induced apoptosis of metastatic melanoma cells. Melanoma Res 2016; 25:284-94. [PMID: 26020489 PMCID: PMC4492793 DOI: 10.1097/cmr.0000000000000172] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The interleukin (IL)-6 inhibits the growth of early-stage melanoma cells, but not metastatic cells. Metastatic melanoma cells are susceptible to statin-induced apoptosis, but this is not clear for early-stage melanoma cells. This study aimed to investigate the IL-6 susceptibility of melanoma cells from different stages in the presence of simvastatin to overcome loss of growth arrest. ELISA was used to detect secreted IL-6 in human melanoma cells. The effects of IL-6 were measured by western blots for STAT3 and Bcl-2 family proteins. Apoptosis and proliferation were measured by caspase 3 activity, Annexin V staining, cell cycle analysis, and a wound-healing assay. Human metastatic melanoma cells A375 and 518A2 secrete high amounts of IL-6, in contrast to early-stage WM35 cells. Canonical IL-6 signaling is intact in these cells, documented by transient phosphorylation of STAT3. Although WM35 cells are highly resistant to simvastatin-induced apoptosis, coadministration with IL-6 enhanced the susceptibility to undergo apoptosis. This proapoptotic effect of IL-6 might be explained by a downregulation of Bcl-XL, observed only in WM35 cells. Furthermore, the IL-6 receptor blocking antibody tocilizumab was coadministered and unmasked an IL-6-sensitive proportion in the simvastatin-induced caspase 3 activity of metastatic melanoma cells. These results confirm that simvastatin facilitates apoptosis in combination with IL-6. Although endogenous IL-6 secretion is sufficient in metastatic melanoma cells, exogenously added IL-6 is needed for WM35 cells. This effect may explain the failure of simvastatin to reduce melanoma incidence in clinical trials and meta-analyses.
Collapse
|
40
|
Pérez-Torres A, Vera-Aguilera J, Sahaza JH, Vera-Aguilera C, Moreno-Aguilera E, Pulido-Camarillo E, Nuñez-Ochoa L, Jeganathan P. Hematological Effects, Serum, and Pulmonary Cytokine Profiles in a Melanoma Mouse Model Treated with GK1. Cancer Biother Radiopharm 2016; 30:247-54. [PMID: 26181852 DOI: 10.1089/cbr.2015.1835] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE In a previous study, we demonstrated the therapeutic efficacy of a subcutaneous injection of GK1 peptide in a melanoma mouse model, effectively increasing the mean survival time by 42.58%, delaying tumor growth, and increasing intratumoral necrosis compared with the control. As a first approach to investigate the anti-melanoma effect of GK1, this study was carried out to determine the hematological effects along with both serum and lung cytokine profiles in a melanoma lung metastatic model. MATERIALS AND METHODS Thirteen C57BL6 female mice were transfected in the lateral tail vein with 2×10(5) B16-F0 melanoma cells. After 7 days, mice were separated in two different groups and treatments were initiated (day 0): The GK1-treated group (seven mice) were injected every 5 days intravenously with GK1 (10 μg) in the lateral tail vein, and the control group (six mice) were injected every 5 days with intravenous saline solution. Blood samples were collected every 5 days from day 0; tumor samples were obtained for cytokine measurements on the day of sacrifice. RESULTS In the peripheral blood, mice treated with GK1 presented a statistically significant decrease in IFN-γ (p<0.05), and lymphocytes tended to be lower compared with the control mice (p=0.06). Lung metastatic analysis demonstrated a significant increase in IFN-γ and IL-12p70 (p<0.05); a significant decrease in IL-17, IL-4, IL-22, IL-23, and IL-12p40 (p<0.05); and a marginal decrease in IL-1β (p=0.07) compared with the control. DISCUSSION Our results suggest that an intratumoral increase of cytokines with antitumor activity along with an intratumoral decrease of cytokines with protumor activity could explain, in part, the anti-melanoma effects of GK1 in a lung metastatic melanoma mouse model. Further studies must be performed to elucidate the precise mechanisms of action for GK1 peptide against melanoma, and their eventual application in humans.
Collapse
Affiliation(s)
- Armando Pérez-Torres
- 1 Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México , México City, México
| | | | - Jorge H Sahaza
- 3 Corporación para Investigaciones Biológicas (CIB) , Unidad de Micología Médica y Experimental, Medellín, Colombia
| | - Carlos Vera-Aguilera
- 1 Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México , México City, México
| | - Eduardo Moreno-Aguilera
- 4 Servicio de Gastrocirugía, Hospital de Especialidades , Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, México DF, México
| | - Evelyn Pulido-Camarillo
- 1 Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México , México City, México
| | - Luis Nuñez-Ochoa
- 5 Departamento de Patología Clínica/Oncología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México , México City, México
| | - Pratheepa Jeganathan
- 6 Department of Mathematics and Statistics, Texas Tech University , Lubbock, Texas
| |
Collapse
|
41
|
Bjoern J, Juul Nitschke N, Zeeberg Iversen T, Schmidt H, Fode K, Svane IM. Immunological correlates of treatment and response in stage IV malignant melanoma patients treated with Ipilimumab. Oncoimmunology 2016; 5:e1100788. [PMID: 27141381 PMCID: PMC4839337 DOI: 10.1080/2162402x.2015.1100788] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 09/15/2015] [Accepted: 09/22/2015] [Indexed: 01/19/2023] Open
Abstract
Introduction: Ipilimumab is effective in the treatment of metastatic malignant melanoma, but few biomarkers reliably predict treatment response. Methods: Patients were treated with Ipilimumab for metastatic malignant melanoma. Blood and serum samples were collected before and during treatment. Mononuclear cells in peripheral blood were subjected to immune phenotypic analyses and cytokine levels were measured in serum samples. Results were correlated with clinical data. Results: A total of 40 patients were included in the analyses. Clinical response were associated with an increase after one series of treatment in absolute lymphocyte count (ALC) (p = 0.008), absolute T cell count (p = 0.02) and the absolute number of activated T cells in peripheral blood (p = 0.003). A high frequency of myeloid derived suppressor cells (MDSC) and a higher level of IL6 were associated with treatment failure, though not significantly. Levels of IL6 in serum above the median showed a tendency to associate with reduced survival by the 4th treatment series. Finally, treatment with Ipilimumab led to a decreased frequency of FOXP3+ regulatory T cells (p = 0.009). Conclusion: Ipilimumab leads to increased ALC, T cell count and T cell activation in malignant melanoma patients responding to treatment. A high baseline frequency of myeloid-derived suppressor cells and high levels of IL6 is associated with a reduced chance of responding to therapy.
Collapse
Affiliation(s)
- Jon Bjoern
- Center for Cancer Immune Therapy, Herlev Hospital, University of Copenhagen, Herlev, Denmark
- Department of Oncology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Nikolaj Juul Nitschke
- Center for Cancer Immune Therapy, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | | | - Henrik Schmidt
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Kirsten Fode
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Inge Marie Svane
- Center for Cancer Immune Therapy, Herlev Hospital, University of Copenhagen, Herlev, Denmark
- Department of Oncology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| |
Collapse
|
42
|
Prasad CP, Mohapatra P, Andersson T. Therapy for BRAFi-Resistant Melanomas: Is WNT5A the Answer? Cancers (Basel) 2015; 7:1900-24. [PMID: 26393652 PMCID: PMC4586801 DOI: 10.3390/cancers7030868] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/09/2015] [Accepted: 09/14/2015] [Indexed: 12/18/2022] Open
Abstract
In recent years, scientists have advocated the use of targeted therapies in the form of drugs that modulate genes and proteins that are directly associated with cancer progression and metastasis. Malignant melanoma is a dreadful cancer type that has been associated with the rapid dissemination of primary tumors to multiple sites, including bone, brain, liver and lungs. The discovery that approximately 40%–50% of malignant melanomas contain a mutation in BRAF at codon 600 gave scientists a new approach to tackle this disease. However, clinical studies on patients have shown that although BRAFi (BRAF inhibitors) trigger early anti-tumor responses, the majority of patients later develop resistance to the therapy. Recent studies have shown that WNT5A plays a key role in enhancing the resistance of melanoma cells to BRAFi. The focus of the current review will be on melanoma development, signaling pathways important to acquired resistance to BRAFi, and why WNT5A inhibitors are attractive candidates to be included in combinatorial therapies for melanoma.
Collapse
Affiliation(s)
- Chandra Prakash Prasad
- Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Centre, Skåne University Hospital, Malmö SE-20502, Sweden.
| | - Purusottam Mohapatra
- Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Centre, Skåne University Hospital, Malmö SE-20502, Sweden.
| | - Tommy Andersson
- Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Centre, Skåne University Hospital, Malmö SE-20502, Sweden.
| |
Collapse
|
43
|
miR-98 suppresses melanoma metastasis through a negative feedback loop with its target gene IL-6. Exp Mol Med 2014; 46:e116. [PMID: 25277211 PMCID: PMC4221693 DOI: 10.1038/emm.2014.63] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 07/31/2014] [Accepted: 08/08/2014] [Indexed: 12/17/2022] Open
Abstract
Dysregulated microRNA (miRNA) expression has a critical role in tumor development and metastasis. However, the mechanism by which miRNAs control melanoma metastasis is unknown. Here, we report reduced miR-98 expression in melanoma tissues with increasing tumor stage as well as metastasis; its expression is also negatively associated with melanoma patient survival. Furthermore, we demonstrate that miR-98 inhibits melanoma cell migration in vitro as well as metastatic tumor size in vivo. We also found that IL-6 is a target gene of miR-98, and IL-6 represses miR-98 levels via the Stat3-NF-κB-lin28B pathway. In an in vivo melanoma model, we demonstrate that miR-98 reduces melanoma metastasis and increases survival in part by reducing IL-6 levels; it also decreases Stat3 and p65 phosphorylation as well as lin28B mRNA levels. These results suggest that miR-98 inhibits melanoma metastasis in part through a novel miR-98-IL-6-negative feedback loop.
Collapse
|
44
|
Interleukin-6 drives melanoma cell motility through p38α-MAPK-dependent up-regulation of WNT5A expression. Mol Oncol 2014; 8:1365-78. [PMID: 24954857 PMCID: PMC5528610 DOI: 10.1016/j.molonc.2014.05.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 05/03/2014] [Accepted: 05/19/2014] [Indexed: 12/24/2022] Open
Abstract
Extensive research has demonstrated a tumor-promoting role of increased WNT5A expression in malignant melanoma. However, very little light has been shed upon how WNT5A expression is up-regulated in melanoma. A potential regulator of WNT5A expression is the pro-inflammatory cytokine Interleukin (IL)-6, which shares the ability of WNT5A to increase melanoma cell invasion. Here, we investigate whether IL-6 can promote melanoma cell motility through an increased expression of WNT5A. We clearly demonstrate that the WNT5A-antagonistic peptide Box5 could inhibit IL-6-induced melanoma cell migration and invasion. Furthermore, IL-6 stimulation of the human melanoma cell lines HTB63 and A375 increased the expression of WNT5A in a dose-dependent manner. To identify the signaling mechanism responsible for this up-regulation, we explored the involvement of the three main signals induced by IL-6; STAT3, Akt and ERK 1/2. Of these, only STAT3 was activated by IL-6 in the melanoma cell lines tested. However, the STAT3 inhibitor S3I-201 failed to inhibit IL-6-induced WNT5A up-regulation in HTB63 and A375 cells. Nor did STAT3 siRNA silencing affect the expression of WNT5A. In search of an alternative signaling mechanism, we detected IL-6-induced activation of p38-MAPK in HTB63 and A375 cells. The p38-MAPK inhibitor SB203580 abolished the IL-6-induced WNT5A up-regulation and blocked IL-6-induced melanoma cell invasion. The latter effect could be rescued by the addition of recombinant WNT5A. Notably, immunoprecipitation analysis revealed that only the p38α-MAPK isoform was activated by IL-6, and subsequent siRNA silencing of p38α-MAPK abolished the IL-6-induced up-regulation of WNT5A. Taken together, we demonstrate a novel link between the two melanoma pro-metastatic agents IL-6 and WNT5A explaining how IL-6 can increase melanoma cell invasion and thus promote the metastatic process. This finding provides a basis for future therapeutic intervention of melanoma progression.
Collapse
|
45
|
Kudchadkar R, Gibney G, Sondak VK. Integrating molecular biomarkers into current clinical management in melanoma. Methods Mol Biol 2014; 1102:27-42. [PMID: 24258972 DOI: 10.1007/978-1-62703-727-3_3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Personalized melanoma medicine has progressed from histopathologic features to serum markers to molecular profiles. Since the identification of activating BRAF mutations and subsequent development of drugs targeting the mutant BRAF protein, oncologists now need to incorporate prognostic and predictive biomarkers into treatment decisions for their melanoma patients. Examples include subgrouping patients by genotype profiles for targeted therapy and the development of serologic, immunohistochemical, and genotype profiles for the selection of patients for immunotherapies. In this chapter, we provide an overview of the current status of BRAF mutation testing, as well as promising serologic and molecular profiles that will impact patient care. As further research helps clarify the roles of these factors, the clinical outcomes of melanoma patients promise to be greatly improved.
Collapse
Affiliation(s)
- Ragini Kudchadkar
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | | | | |
Collapse
|
46
|
Yao X, Huang J, Zhong H, Shen N, Faggioni R, Fung M, Yao Y. Targeting interleukin-6 in inflammatory autoimmune diseases and cancers. Pharmacol Ther 2013; 141:125-39. [PMID: 24076269 DOI: 10.1016/j.pharmthera.2013.09.004] [Citation(s) in RCA: 469] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 09/03/2013] [Indexed: 12/15/2022]
Abstract
Interleukin-6 (IL-6) is a pleiotropic cytokine with significant functions in the regulation of the immune system. As a potent pro-inflammatory cytokine, IL-6 plays a pivotal role in host defense against pathogens and acute stress. However, increased or deregulated expression of IL-6 significantly contributes to the pathogenesis of various human diseases. Numerous preclinical and clinical studies have revealed the pathological roles of the IL-6 pathway in inflammation, autoimmunity, and cancer. Based on the rich body of studies on biological activities of IL-6 and its pathological roles, therapeutic strategies targeting the IL-6 pathway are in development for cancers, inflammatory and autoimmune diseases. Several anti-IL-6/IL-6 receptor monoclonal antibodies developed for targeted therapy have demonstrated promising results in both preclinical studies and clinical trials. Tocilizumab, an anti-IL-6 receptor antibody, is effective in the treatment of various autoimmune and inflammatory conditions notably rheumatoid arthritis. It is the only IL-6 pathway targeting agent approved by the regulatory agencies for clinical use. Siltuximab, an anti-IL-6 antibody, has been shown to have potential benefits treating various human cancers either as a single agent or in combination with other chemotherapy drugs. Several other anti-IL-6-based therapies are also under clinical development for various diseases. IL-6 antagonism has been shown to be a potential therapy for these disorders refractory to conventional drugs. New strategies, such as combination of IL-6 blockade with inhibition of other signaling pathways, may further improve IL-6-targeted immunotherapy of human diseases.
Collapse
Affiliation(s)
- Xin Yao
- MedImmune, LLC, Gaithersburg, MD 20878, USA
| | | | | | - Nan Shen
- Joint Molecular Rheumatology Laboratory of Institute of Health Sciences and Shanghai Renji Hospital, Shanghai, China
| | | | | | - Yihong Yao
- MedImmune, LLC, Gaithersburg, MD 20878, USA.
| |
Collapse
|
47
|
|
48
|
|