1
|
Xuan D, Wen W, Tian S, Piao M, Xu D, Liu L. Prognostic value of maximum standard uptake value, metabolic tumor volume, and total lesion glycolysis of 18F-FDG PET/CT in patients with renal carcinoma: A protocol for systematic review and meta analysis. Medicine (Baltimore) 2020; 99:e19988. [PMID: 32443301 PMCID: PMC7254885 DOI: 10.1097/md.0000000000019988] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/18/2020] [Accepted: 03/23/2020] [Indexed: 01/20/2023] Open
Abstract
PURPOSE We present a comprehensive systematic review of the documented literature on parameters derived from F-fluorodeoxyglucose positron emission tomography (F-FDG PET) and meta-analysis of the prognostic value of maximal standard uptake value (SUVmax), metabolic tumor volume (MTV) and total lesional glycolysis (TLG) in patients with renal carcinoma (RCC). PATIENTS AND METHODS Relevant articles in English from PubMed, EMBASE, and the Cochrane Library were retrieved. Pooled hazard ratio (HR) values were used to assess the prognostic value of SUVmax, MTV, and TLG. RESULTS A total of 10 primary studies involving 780 patients with RCC were included. The combined HRs for event-free survival were 1.32 (95% CI 1.10-1.58) for SUVmax, 2.40 (95% CI 1.20-4.79) for MTV, and 3.31 (95% CI 1.68-6.50) for TLG. Pooled HRs for overall survival were 1.264 (95% CI 1.124-1.421) for SUVmax, 3.52 (95% CI 1.451-8.536) for MTV, and 6.33 (95% CI 1.32-30.30) for TLG. Subgroup analysis revealed SUVmax as an independent risk factor for patients with recurrence or metastasis. CONCLUSION The present meta-analysis confirmed that despite the clinical heterogeneity of RCC and adoption of various methods between studies, high SUVmax is a significant prognostic factor, especially in patients with recurrence or metastasis. MTV and TLG were associated with prediction of higher risk of adverse events or death in patients with RCC.
Collapse
Affiliation(s)
- Dongchun Xuan
- Center of Morphological Experiment, Medical College of Yanbian University
- Department of Nuclear Medicine
| | - Weibo Wen
- Center of Morphological Experiment, Medical College of Yanbian University
- Department of Nuclear Medicine
| | | | | | - Dongyuan Xu
- Center of Morphological Experiment, Medical College of Yanbian University
| | - Lan Liu
- Department of Pathology, Yanbian University hospital, Yanji, Jilin Province, China
| |
Collapse
|
2
|
Pankowska V, Malkowski B, Wedrowski M, Wedrowska E, Roszkowski K. FDG PET/CT as a survival prognostic factor in patients with advanced renal cell carcinoma. Clin Exp Med 2018; 19:143-148. [PMID: 30488140 PMCID: PMC6394561 DOI: 10.1007/s10238-018-0539-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 11/22/2018] [Indexed: 12/15/2022]
Abstract
Accurate prediction of the outcome of molecular target-based treatment in advanced renal cell carcinoma (RCC) is an important clinical problem. Positron emission tomography/computed tomography using [18F]-2-fluoro-2-deoxyglucose (FDG PET/CT) is a noninvasive tool for the assessment of glucose accumulation which can be a marker of the biological characteristics of the tumor. In this paper, we assess FDG PET/CT as a survival prognostic marker in patients with advanced RCC. The study included 121 patients treated in the years 2011–2016 with a diagnosis of advanced renal cell carcinoma (stage IV, multifocal metastases in all patients). Assessment using FDG PET/CT was conducted by measuring the maximum standard uptake value (SUVmax) for the marker used (the highest SUV measurement result for each patient in a single examination). SUVmax measurements were compared with various clinical risk factors used as prognostic markers. The median follow-up period was 19 months (ranging from 3 to 61 months). SUVmax measurements in all patients ranged from 1.3 to 30.0 (median 6.9). Higher SUVmax was correlated with poorer prognosis. Multi-way analysis with standard risk factors revealed that SUVmax was an independent factor for overall survival (OS; p < 0.003, hazard ratio 1.312, 95% CI 1.147–1.346). For SUVmax < 7.0, median OS was 32 months. For 7.0 ≤ SUVmax < 12.0, median OS was 12.5 months. For SUVmax ≥ 12.0, median OS was 10 months. The differences were statistically significant. A preliminary SUVmax assessment conducted using FDG PET/CT can provide information useful in the prediction of survival of patients with advanced RCC.
Collapse
Affiliation(s)
| | - Bogdan Malkowski
- Department of Positron Emission Tomography and Molecular Diagnostic, Nicolaus Copernicus University, Collegium Medicum, Bydgoszcz, Poland
| | - Mateusz Wedrowski
- Department of Positron Emission Tomography and Molecular Diagnostic, Nicolaus Copernicus University, Collegium Medicum, Bydgoszcz, Poland
| | - Ewelina Wedrowska
- Department of Gene Therapy, Faculty of Medicine, Nicolaus Copernicus University, Collegium Medicum, Bydgoszcz, Poland
| | - Krzysztof Roszkowski
- Department of Oncology, Radiotherapy and Gynecologic Oncology, Faculty of Health Sciences, Nicolaus Copernicus University, Collegium Medicum, Bydgoszcz, Poland.
| |
Collapse
|
3
|
Medrano RF, Hunger A, Mendonça SA, Barbuto JAM, Strauss BE. Immunomodulatory and antitumor effects of type I interferons and their application in cancer therapy. Oncotarget 2017; 8:71249-71284. [PMID: 29050360 PMCID: PMC5642635 DOI: 10.18632/oncotarget.19531] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/12/2017] [Indexed: 02/07/2023] Open
Abstract
During the last decades, the pleiotropic antitumor functions exerted by type I interferons (IFNs) have become universally acknowledged, especially their role in mediating interactions between the tumor and the immune system. Indeed, type I IFNs are now appreciated as a critical component of dendritic cell (DC) driven T cell responses to cancer. Here we focus on IFN-α and IFN-β, and their antitumor effects, impact on immune responses and their use as therapeutic agents. IFN-α/β share many properties, including activation of the JAK-STAT signaling pathway and induction of a variety of cellular phenotypes. For example, type I IFNs drive not only the high maturation status of DCs, but also have a direct impact in cytotoxic T lymphocytes, NK cell activation, induction of tumor cell death and inhibition of angiogenesis. A variety of stimuli, including some standard cancer treatments, promote the expression of endogenous IFN-α/β, which then participates as a fundamental component of immunogenic cell death. Systemic treatment with recombinant protein has been used for the treatment of melanoma. The induction of endogenous IFN-α/β has been tested, including stimulation through pattern recognition receptors. Gene therapies involving IFN-α/β have also been described. Thus, harnessing type I IFNs as an effective tool for cancer therapy continues to be studied.
Collapse
Affiliation(s)
- Ruan F.V. Medrano
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of São Paulo/LIM 24, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Aline Hunger
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of São Paulo/LIM 24, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Samir Andrade Mendonça
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of São Paulo/LIM 24, University of São Paulo School of Medicine, São Paulo, Brazil
| | - José Alexandre M. Barbuto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Cell and Molecular Therapy Center, NUCEL-NETCEM, University of São Paulo, São Paulo, Brazil
| | - Bryan E. Strauss
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of São Paulo/LIM 24, University of São Paulo School of Medicine, São Paulo, Brazil
| |
Collapse
|
4
|
Sahu RP, Ocana JA, Harrison KA, Ferracini M, Touloukian CE, Al-Hassani M, Sun L, Loesch M, Murphy RC, Althouse SK, Perkins SM, Speicher PJ, Tyler DS, Konger RL, Travers JB. Chemotherapeutic agents subvert tumor immunity by generating agonists of platelet-activating factor. Cancer Res 2014; 74:7069-78. [PMID: 25304264 DOI: 10.1158/0008-5472.can-14-2043] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Oxidative stress suppresses host immunity by generating oxidized lipid agonists of the platelet-activating factor receptor (PAF-R). Because many classical chemotherapeutic drugs induce reactive oxygen species (ROS), we investigated whether these drugs might subvert host immunity by activating PAF-R. Here, we show that PAF-R agonists are produced in melanoma cells by chemotherapy that is administered in vitro, in vivo, or in human subjects. Structural characterization of the PAF-R agonists induced revealed multiple oxidized glycerophosphocholines that are generated nonenzymatically. In a murine model of melanoma, chemotherapeutic administration could augment tumor growth by a PAF-R-dependent process that could be blocked by treatment with antioxidants or COX-2 inhibitors or by depletion of regulatory T cells. Our findings reveal how PAF-R agonists induced by chemotherapy treatment can promote treatment failure. Furthermore, they offer new insights into how to improve the efficacy of chemotherapy by blocking its heretofore unknown impact on PAF-R activation.
Collapse
Affiliation(s)
- Ravi P Sahu
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana. Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jesus A Ocana
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana. Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Kathleen A Harrison
- Department of Pharmacology, University of Colorado Health Sciences Center, Aurora, Colorado
| | - Matheus Ferracini
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - Mohammed Al-Hassani
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Louis Sun
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Mathew Loesch
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Robert C Murphy
- Department of Pharmacology, University of Colorado Health Sciences Center, Aurora, Colorado
| | - Sandra K Althouse
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Susan M Perkins
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Paul J Speicher
- The Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Douglas S Tyler
- The Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Raymond L Konger
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana. Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jeffrey B Travers
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana. Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana. The Richard L. Roudebush V.A. Medical Center, Indianapolis, Indiana.
| |
Collapse
|
5
|
Herndon TM, Demko SG, Jiang X, He K, Gootenberg JE, Cohen MH, Keegan P, Pazdur R. U.S. Food and Drug Administration Approval: peginterferon-alfa-2b for the adjuvant treatment of patients with melanoma. Oncologist 2012; 17:1323-8. [PMID: 23002124 DOI: 10.1634/theoncologist.2012-0123] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
On March 29, 2011, the U.S. Food and Drug Administration approved peginterferon alfa-2b (PEG-IFN) (Sylatron™; Schering Corporation, Kenilworth, NJ) for the adjuvant treatment of melanoma patients with microscopic or gross nodal involvement following definitive surgical resection including complete lymphadenectomy. The approval was based on a single, open-label, multicenter trial enrolling 1,256 patients. After surgical resection, patients were randomized (1:1) to either PEG-IFN or observation for 5 years. PEG-IFN, 6 μg/kg per week, was administered s.c. for eight doses, followed by 3 μg/kg per week for up to 252 weeks. Stratification factors included microscopic or gross nodal involvement, number of positive nodes, Breslow thickness, ulceration, sex, and study center. Patients were assessed for recurrence by the investigators based on physical examination every 3 months for 2 years and every 6 months thereafter. The relapse-free survival (RFS) interval, the primary efficacy endpoint, was significantly longer in PEG-IFN-treated patients. The median RFS times were 34.8 months and 25.5 months, respectively. There was no statistically significant difference in the overall survival time. The most common (>60%) grade 1-4 adverse reactions were fatigue, increased alanine aminotransferase (ALT) and aspartate aminotransferase (AST), pyrexia, headache, anorexia, myalgia, nausea, chills, and injection site reactions. The most common serious adverse reactions were fatigue, increased ALT and AST, and pyrexia. Thirty-three percent of patients receiving PEG-IFN discontinued treatment as a result of adverse reactions. Five deaths were reported within 30 days of the last treatment dose, two resulting from cardiovascular disease considered as possibly related to treatment.
Collapse
Affiliation(s)
- Thomas M Herndon
- Division of Biological Oncology Products, Food and Drug Administration, 10903 New Hampshire Avenue, Building 22, Room 5222, Silver Spring, Maryland 20993, USA.
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Sahu RP, Turner MJ, DaSilva SC, Rashid BM, Ocana JA, Perkins SM, Konger RL, Touloukian CE, Kaplan MH, Travers JB. The environmental stressor ultraviolet B radiation inhibits murine antitumor immunity through its ability to generate platelet-activating factor agonists. Carcinogenesis 2012; 33:1360-7. [PMID: 22542595 PMCID: PMC3405652 DOI: 10.1093/carcin/bgs152] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Revised: 03/26/2012] [Accepted: 04/14/2012] [Indexed: 11/12/2022] Open
Abstract
Ubiquitous pro-oxidative stressor ultraviolet B radiation (UVB) to human or mouse skin generates platelet-activating factor (PAF) and novel oxidatively modified glycerophosphocholines (Ox-GPCs) with PAF-receptor (PAF-R) agonistic activity. These lipids mediate systemic immunosuppression in a process involving IL-10. The current studies sought to determine the functional significance of UVB-mediated systemic immunosuppression in an established model of murine melanoma. We show that UVB irradiation augments B16F10 tumor growth and is dependent on host, but not melanoma cell; PAF-R-expression as UVB or the PAF-R agonist, carbamoyl PAF (CPAF), both promote B16F10 tumor growth in wild-type (WT) mice, independent of whether B16F10 cells express PAF-Rs, but do not augment tumor growth in Pafr -/- mice. UVB-mediated augmentation of experimental murine tumor growth was inhibited with antioxidants, demonstrating the importance of Ox-GPC PAF-R agonists produced non-enzymatically. Host immune cells are required as CPAF-induced augmentation of tumor growth which is not seen in immunodeficient NOD SCID mice. Finally, depleting antibodies against IL-10 in WT mice or depletion of CD25-positive cells in FoxP3(EGFP) transgenic mice block UVB and/or CPAF-induced tumor growth supporting a requirement for IL-10 and Tregs in this process. These findings indicate that UVB-generated Ox-GPCs with PAF-R agonistic activity enhance experimental murine melanoma tumor growth through targeting host immune cells, most notably Tregs, to mediate systemic immunosuppression.
Collapse
Affiliation(s)
- Ravi P. Sahu
- Department of Dermatology
- Department of Pathology and Laboratory Medicine
| | | | | | | | | | | | - Raymond L. Konger
- Department of Dermatology
- Department of Pathology and Laboratory Medicine
| | | | - Mark H. Kaplan
- Department of Microbiology and Immunology
- H.B. Wells Center for Pediatric Research, Department of Pediatrics
| | - Jeffrey B. Travers
- Department of Dermatology
- H.B. Wells Center for Pediatric Research, Department of Pediatrics
- Department of Pharmacology and Toxicology
- The Richard L. Roudebush V.A. Medical Center, Indiana University School of Medicine, 1044 Walnut St. Rm. 202, Indianapolis, IN 46202, USA
| |
Collapse
|
7
|
Patel JN, Walko CM. Sylatron: a pegylated interferon for use in melanoma. Ann Pharmacother 2012; 46:830-8. [PMID: 22619474 DOI: 10.1345/aph.1q791] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE To review the currently available literature on peginterferon alfa-2b (pegIFN [Sylatron]), including its role in therapy and toxicity for adjuvant treatment of locally advanced melanoma. DATA SOURCES A literature search was performed of PubMed and the American Society of Clinical Oncology abstracts from 1976 to February 2012, using the primary search terms peginterferon alfa-2b, interferon, Sylatron, and melanoma. STUDY SELECTION AND DATA EXTRACTION All available English-language articles and trials that described the pharmacology, pharmacokinetics, pharmacodynamics, clinical activity, or safety profile of pegIFN were reviewed. DATA SYNTHESIS PegIFN was approved in March 2011 for the adjuvant treatment of node-positive melanoma. Interferon (IFN) is commonly used in patients with melanoma who remain at high risk for relapse following surgery; however, the optimal scheduling and dose are not agreed upon. Pegylation of IFN involves conjugation with polyethylene glycol. Following subcutaneous injection of pegIFN, the rate of absorption, renal and cellular clearance, and immunogenicity are reduced. As a result of the extended serum half-life, once-weekly administration is feasible, compared with the daily and/or thrice weekly dosing of IFN. When compared with observation alone in patients with resected stage III melanoma, pegIFN demonstrated a significant increase in relapse-free survival, with a marginal impact on overall survival. The most common adverse events were as expected with IFN and included fatigue, increased liver enzymes, pyrexia, headache, anorexia, myalgia, nausea, chills, depression, and injection site reactions. A large Phase 3 study is underway to further assess outcome and toxicity differences between pegIFN weekly and low-dose IFN thrice weekly. CONCLUSIONS PegIFN is a modified version of the previously approved interferon indicated for the adjuvant treatment of melanoma. Although the safety profile remains similar between the pegylated and non-pegylated forms, once-weekly administration is feasible secondary to an extended serum half-life and may have improved convenience for the patient.
Collapse
Affiliation(s)
- Jai N Patel
- Eshelman School of Pharmacy, Division of Pharmacotherapy and Experimental Therapeutics, Institute for Pharmacogenomics and Individualized Therapy, University of North Carolina, Chapel Hill, NC, USA
| | | |
Collapse
|
8
|
Abstract
Achieving optimal patient benefit from biological therapies can be hindered by drug instability, rapid clearance requiring frequent dosing or potential immune reactions. One strategy for addressing these challenges is drug modification through PEGylation, a well established process by which one or more molecules of polyethylene glycol (PEG) are covalently attached to a biological or small-molecule drug, effectively transforming it into a therapy with improved pharmacokinetic and pharmacodynamic properties. Numerous PEGylated therapeutics are currently available, all of which have at least comparable efficacy, safety and tolerability to their unmodified forms. A PEGylated form of interferon-β-1a (PEG-IFNβ-1a) is being developed to address an unmet medical need for safer, more effective and more convenient therapies for multiple sclerosis (MS). Phase I study data suggest that PEG-IFNβ-1a should provide patients with a first-line therapy with a more convenient dosing regimen while maintaining the established efficacy, safety and tolerability of presently available IFNβ-1a. The ongoing global ADVANCE phase III study will determine the clinical efficacy of PEG-IFNβ-1a in patients with relapsing MS.
Collapse
|
9
|
Müller-Quernheim UC, Potthast L, Müller-Quernheim J, Zissel G. Tumor-cell co-culture induced alternative activation of macrophages is modulated by interferons in vitro. J Interferon Cytokine Res 2012; 32:169-77. [PMID: 22280057 DOI: 10.1089/jir.2011.0020] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Tumor-associated macrophages infiltrate tumors and facilitate tumor growth. Here, we analyzed M1 and M2 marker expression in the course of co-culture-driven macrophage differentiation and investigated the influence of interferons (IFNs) on this differentiation. To generate monocyte-derived macrophages (MDMs) 1×10⁶ monocytes of healthy volunteers were cultivated either with 25×10³ adherent A549/mL or in medium containing 50% A549 conditioned medium (CM) for 72 h in the presence or absence of IFN-α, β or γ, respectively. Supernatants were tested for CCL18 (M2 marker) and CXCL10 (M1 marker) by enzyme-linked immunosorbent assay. CCL18 and CXCL10 release by MDM is increased by the presence of A549 cells, but also when cultured in A549 CM. On stimulation with IFN-γ, we observe an increased release of the M1 marker CXCL10 and a decreased release of CCL18. Type I IFNs also increases CXCL10 release. Thus, A549 releases a soluble factor which enhances CCL18 production and M2 polarization, indicating that a localized specific cytokine milieu, as found in the environment of a tumor or in fibrotic lung tissue, favors alternative activation of macrophages. In the presence of IFN-γ, M2 differentiation is attenuated as shown by the decrease of the M2 chemokine CCL18 and by the increase of the M1 chemokine CXCL10. However, CXCL10 levels were also increased by the co-culture, which indicates a simultaneous classical activation (M1) or the formation of a M1/M2 hybrid.
Collapse
|
10
|
Krauze MT, Tarhini A, Gogas H, Kirkwood JM. Prognostic significance of autoimmunity during treatment of melanoma with interferon. Semin Immunopathol 2011; 33:385-91. [PMID: 21279809 PMCID: PMC8635120 DOI: 10.1007/s00281-011-0247-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 01/11/2011] [Indexed: 12/19/2022]
Abstract
Since the pivotal cooperative group trials in the 1980's-90's,, high-dose interferon (HDI) has been the standard of adjuvant therapy. Despite multiple other trials evaluating potential new therapies in melanoma, HDI remains the only FDA-approved therapy for stage IIB and III melanoma. Initial reports from the more recent phase III international trials of modifications of the original HDI regimen linked the appearance of autoimmunity with improved outcomes of disease. Trials of high-dose interleukin-2, many years earlier, reported anecdotal observations that were consistent with the hypothesis that autoimmunity and clinical benefit of immunotherapies of melanoma are linked with one another. The only prospectively conducted study examining the appearance of clinical and laboratory evidence of autoimmunity during HDI therapy was published by Gogas and colleagues, demonstrating statistically significant impact on relapse-free survival and overall survival. Retrospectively conducted studies of different intermediate dosage regimens of interferon (IFN) have not fully confirmed the linkage of serological evidence of autoimmunity and improved survival outcomes. With the emergence of new immunotherapies in treatment of melanoma, this review highlights the importance of autoimmunity for future applications in melanoma and reviews significant differences of past studies evaluating the appearance of autoimmunity during IFN therapy in high-risk melanoma.
Collapse
Affiliation(s)
- Michal T Krauze
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | | | | | | |
Collapse
|
11
|
Abstract
Interferon alpha (IFNα) is widely used for treatment of melanoma and certain other malignancies. This cytokine as well as the related IFNβ exerts potent anti-tumorigenic effects; however, their efficacy in patients is often suboptimal. Here, we report that inflammatory signaling impedes the effects of IFNα/β. Melanoma cells can secrete pro-inflammatory cytokines that inhibit cellular responses to IFNα/β via activating the ligand-independent pathway for the phosphorylation and subsequent ubiquitination and accelerated degradation of the IFNAR1 chain of type I IFN receptor. Catalytic activity of the p38 protein kinase was required for IFNAR1 downregulation and inhibition of IFNα/β signaling induced by proinflammatory cytokines such as interleukin 1 (IL-1). Activation of p38 kinase inversely correlated with protein levels of IFNAR1 in clinical melanoma specimens. Inhibition of p38 kinase augmented the inhibitory effects of IFNα/β on cell viability and growth in vitro and in vivo. The roles of inflammation and p38 protein kinase in regulating cellular responses to IFNα/β in normal and tumor cells are discussed.
Collapse
|
12
|
Current world literature. Curr Opin Oncol 2011; 23:227-34. [PMID: 21307677 DOI: 10.1097/cco.0b013e328344b687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
13
|
Current World Literature. Curr Opin Support Palliat Care 2010; 4:293-304. [DOI: 10.1097/spc.0b013e328340e983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|