ArticleLiterature Review

Cytokine deregulation in cancer. Biomed Pharmacother

Authors:
To read the full-text of this research, you can request a copy directly from the author.

Abstract

Endogenous cytokines are aberrantly produced in many cancers, and serve as autocrine growth factors or indicators in immune response to the tumors. Hence, cytokine deregulation is likely to participate in the development or evolution of the malignant process. Over the last few years, endogenous cytokine levels have been correlated with phenotypic manifestations of cancer and with prognosis. For instance, serum IL-6 levels are elevated in both relapsed and newly-diagnosed Hodgkin's and non-Hodgkin's lymphoma, and these levels correlate with established prognostic features. Furthermore, in diffuse large cell lymphoma, serum IL-6 level is an independent prognostic variable for both complete remission and failure-free survival. Serum IL-10 levels are also elevated in lymphoid malignancies and predict outcome. In some cases, it may be that the balance between endogenous cytokine agonists and antagonists is disrupted. For instance, in chronic myelogenous leukemia, high cellular (leukocyte) levels of IL-1 beta and low levels of IL-1 receptor antagonist (IL-1RA) are seen in advanced disease and correlate with reduced survival. The molecular mechanisms underlying cytokine deregulation are now being investigated, with preliminary data suggesting heterogeneous genetic driving forces, including oncogene aberrations and viral infection.

No full-text available

Request Full-text Paper PDF

To read the full-text of this research,
you can request a copy directly from the author.

... Dysregulated production of cytokines and adhesion molecules has been implicated in the onset and progression of various types of cancer including hematological malignancies [1,2]. Better understanding of tumor microenvironment is essential for development of new treatment approaches [3][4][5]. ...
Article
Full-text available
Aim: To evaluate serum levels of selected cytokine receptors in B-cell precursor acute lymphoblastic leukemia (B-ALL) and their association with acknowledged prognostic factors, relapse-free survival (RFS) and overall survival (OS). Materials and Methods: A total of 42 de novo adult B-ALL patients, 19 BCR/ABL positive, were included in this study. Soluble receptor α for IL-2 (sIL-2Rα), soluble receptor for IL-6 (sIL-6R), soluble receptor for TNF-α type I and II (sTNFR-1, sTNFR-2) and matrix metalloproteinase-9 (MMP-9) were measured by biochip array technology at diagnosis and in complete remission (CR). Results: At diagnosis of B-ALL, we found significantly higher levels of sIL-2Rα, sIL-6R, sTNFR-1, sTNFR-2 and significantly lower levels MMP-9 in comparison with CR (p < 0.001 in all cases). BCR/ABL positive patients had higher levels of sIL-2Rα at diagnosis (r = 0.484; p = 0.014). Serum levels of evaluated cytokines were not associated with achievement of CR after one cycle of induction therapy, RFS or OS. Conclusion: Serum levels of all evaluated cytokines are significantly altered in newly diagnosed B-ALL reflecting activity of the disease. No significant correlations with response to first induction therapy, RFS or OS were found. Further studies with a longer follow-up will be needed.
... As a member of the IL-1 family with multifaceted functions, IL-1RA plays diverse roles in a number of pathological conditions, including cancer. For example, lower levels of IL-1RA were found in several cancer types, such as leukemia, colorectal cancer, and prostate cancer [15][16][17], and it was negatively associated with the development of premalignant oral dysplasia [18]. However, other reports indicate higher levels of IL-1RA in cervical and gastric cancers [19,20], suggesting that IL-1RA may function in a cancer type-specific manner. ...
Article
Full-text available
Background Interleukin-1 receptor antagonist (IL-1RA), a member of the IL-1 family, has diverse roles in cancer development. However, the role of IL-1RA in oral squamous cell carcinoma (OSCC), in particular the underlying mechanisms, remains to be elucidated. Methods Tumor tissues from OSCC patients were assessed for protein expression by immunohistochemistry. Patient survival was evaluated by Kaplan–Meier curve analysis. Impact of differential IL-1RA expression on cultured OSCC cell lines was assessed in vitro by clonogenic survival, tumorsphere formation, soft agar colony formation, and transwell cell migration and invasion assays. Oxygen consumption rate was measured by Seahorse analyzer or multi-mode plate reader. PCR array was applied to screen human cancer stem cell-related genes, proteome array for phosphorylation status of kinases, and Western blot for protein expression in cultured cells. In vivo tumor growth was investigated by orthotopic xenograft in mice, and protein expression in xenograft tumors assessed by immunohistochemistry. Results Clinical analysis revealed that elevated IL-1RA expression in OSCC tumor tissues was associated with increased tumor size and cancer stage, and reduced survival in the patient group receiving adjuvant radiotherapy compared to the patient group without adjuvant radiotherapy. In vitro data supported these observations, showing that overexpression of IL-1RA increased OSCC cell growth, migration/invasion abilities, and resistance to ionizing radiation, whereas knockdown of IL-1RA had largely the opposite effects. Additionally, we identified that EGFR/JNK activation and SOX2 expression were modulated by differential IL-1RA expression downstream of mitochondrial metabolism, with application of mitochondrial complex inhibitors suppressing these pathways. Furthermore, in vivo data revealed that treatment with cisplatin or metformin—a mitochondrial complex inhibitor and conventional therapy for type 2 diabetes—reduced IL-1RA-associated xenograft tumor growth as well as EGFR/JNK activation and SOX2 expression. This inhibitory effect was further augmented by combination treatment with cisplatin and metformin. Conclusions The current study suggests that IL-1RA promoted OSCC malignancy through mitochondrial metabolism-mediated EGFR/JNK activation and SOX2 expression. Inhibition of this mitochondrial metabolic pathway may present a potential therapeutic strategy in OSCC.
... In addition, Nakayama et al. showed that patients with low NRI levels were at higher risk of advanced clinical stage and mortality [37]. The reason for the relationship between low NRI levels and advanced tumors might be due to the various cytokines released by the advanced tumor [38][39][40]. Moreover, several studies have shown that elevated inflammatory factors and poor nutrient absorption are associated with increased catabolism, leading to anorexia and malnutrition [41][42][43]. ...
Article
Full-text available
Background: The nutritional risk index (NRI) is an excellent indicator of nutritional status and a significant prognostic factor in several malignancies, but the relationship between NRI and the prognosis of head and neck soft tissue sarcoma (HNSTS) patients remains unclear. The aim of this study was to investigate the role of NRI in patients with HNSTS. Methods: We retrospectively reviewed patients with HNSTS between 1990 and 2021. In order to determine the optimal cut-off value of NRI, the Maximally selected log-rank statistic was performed. We evaluated the effect of NRI on overall survival (OS) and progression-free survival (PFS) by using the Kaplan-Meier method and Cox regression analysis. Then, OS and PFS nomograms based on NRI were constructed. Results: In total, 436 HNSTS patients were included in this study. The optimal cut-off value of NRI was 99.34. Patients with low-NRI showed significantly worse OS and PFS than patients with high-NRI, respectively (5-year OS rate of 43.0 vs. 70.8%, 5-year PFS rate of 29.0 vs. 45.0%, all p < 0.05). In the multivariate analysis, distant metastasis, deep tumor depth, tumor grade, and NRI were prognostic factors for both PFS and OS, and treatment modality was associated with OS but not PFS. The concordance indexes (C-indexes) of OS and PFS nomograms were 0.794 (95% CI, 0.759-0.829) and 0.663 (95% CI, 0.626-0.700), respectively, which also performed well in the validation set. Conclusions: NRI is an independent predictor of OS and PFS in HNSTS patients. The validated nomograms based on NRI provide useful predictions of OS and PFS for patients with HNSTS.
... To identify potential major regulators along with their subgroup-specific expression behavior, we learned gene regulatory networks associated with the observed expression changes between the three T-PLL subgroups (Fig 6). Most of the 41 revealed genes with increased network connectivity have been reported to be involved in different types of cancer and several of them have been shown to be important in different types of leukemia (Table 1, ALPP [34], AHSP [44], CXCL8 [37], CXCR2 [47], ELANE [53], FFAR2 [40], G0S2 [38], GIMAP2 [55], IL1RN [39], LCN2 [36], MBTD1 [51], PPP1R15A [49]). Thus, it is likely that at least some of these genes may also contribute to T-PLL development and that their subgroup-specific expression behavior may contribute to the globally observed gene expression differences of the T-PLL subgroups. ...
Article
Full-text available
T-cell prolymphocytic leukemia (T-PLL) is a rare blood cancer with poor prognosis. Overexpression of the proto-oncogene TCL1A and missense mutations of the tumor suppressor ATM are putative main drivers of T-PLL development, but so far only little is known about the existence of T-PLL gene expression subtypes. We performed an in-depth computational reanalysis of 68 gene expression profiles of one of the largest currently existing T-PLL patient cohorts. Hierarchical clustering combined with bootstrapping revealed three robust T-PLL gene expression subgroups. Additional comparative analyses revealed similarities and differences of these subgroups at the level of individual genes, signaling and metabolic pathways, and associated gene regulatory networks. Differences were mainly reflected at the transcriptomic level, whereas gene copy number profiles of the three subgroups were much more similar to each other, except for few characteristic differences like duplications of parts of the chromosomes 7, 8, 14, and 22. At the network level, most of the 41 predicted potential major regulators showed subgroup-specific expression levels that differed at least in comparison to one other subgroup. Functional annotations suggest that these regulators contribute to differences between the subgroups by altering processes like immune responses, angiogenesis, cellular respiration, cell proliferation, apoptosis, or migration. Most of these regulators are known from other cancers and several of them have been reported in relation to leukemia (e.g. AHSP, CXCL8, CXCR2, ELANE, FFAR2, G0S2, GIMAP2, IL1RN, LCN2, MBTD1, PPP1R15A). The existence of the three revealed T-PLL subgroups was further validated by a classification of T-PLL patients from two other smaller cohorts. Overall, our study contributes to an improved stratification of T-PLL and the observed subgroup-specific molecular characteristics could help to develop urgently needed targeted treatment strategies.
... Previous studies reported the association of low GNRI with advanced stage in various cancers [19,37,[39][40][41], similar to our findings. Various cytokines are released in advanced cancer [42][43][44], and some studies reported that increased inflammation markers and decreased nutritional marker were associated with increased catabolism, resulting in anorexia and a negative effect on nutritional status [45,46], which may also account for a low GNRI. ...
Article
Full-text available
Abstract Background Several studies investigated the utility of inflammation and nutritional markers in predicting the prognosis in patients with gastric cancer; however, the markers with the best predictive ability remain unclear. This retrospective study aimed to determine inflammation and nutritional markers that predicted prognosis in elderly patients over 75 years of age undergoing curative gastrectomy for gastric cancer. Methods Between January 2005 and December 2015, 497 consecutive elderly gastric cancer patients aged over 75 years underwent curative gastrectomy in 12 institutions. The geriatric nutritional risk index (GNRI), prognostic nutritional index, neutrophil/lymphocyte ratio, platelet/lymphocyte ratio, and C-reactive protein/albumin ratio were examined as prognostic markers for overall survival (OS) and disease-specific survival (DSS) using area under the curve (AUC) using receiver operating characteristic (ROC) curve analysis. Results The GNRI had the highest AUC and predictive value for both OS (0.637, p
... Furthermore, cytokines are associated with the alteration of the µ-opioid receptor level in neural and other immune cells and seem to be associated with peripheral sensitization [10,11]. Notably, cancer cells produce cytokines aberrantly both in hematological diseases and solid tumors [12], thus influencing pain expression and perception. Previous studies focused on adults have found some associations between interleukin-6 (IL-6), interleukin-8 (Il-8), tumor necrosis factor α (TNFα) SNPs, and clinical outcomes in cancer patients. ...
Article
Full-text available
Moderate to severe cancer pain treatment in children is based on the use of weak and strong opioids. Pharmacogenetics play a central role in developing personalized pain therapies, as well as avoiding treatment failure and/or intolerable adverse drug reactions. This observational study aimed to investigate the association between IL-6, IL-8, and TNFα genetic single nucleotide polymorphisms (SNPs) and response to opioid therapy in a cohort of pediatric cancer patients. Pain intensity before treatment (PIt0) significantly differed according to IL-6 rs1800797 SNP, with a higher PI for A/G and G/G individuals (p = 0.017), who required a higher dose of opioids (p = 0.047). Moreover, compared to G/G subjects, heterozygous or homozygous individuals for the A allele of IL-6 rs1800797 SNP had a lower risk of having a PIt0 > 4. Dose24h and Dosetot were both higher in G/G individuals for TNFα rs1800629 (p = 0.010 and p = 0.031, respectively), while risk of having a PIt0 > 4 and a ∆VAS > 2 was higher for G/G subjects for IL-6 rs1800795 SNP compared to carriers of the C allele. No statistically significant association between genotypes and safety outcomes was found. Thus, IL-6 and TNFα SNPs could be potential markers of baseline pain intensity and opioid dose requirements in pediatric cancer patients.
... These cytokines help establish a favorable microenvironment for neoplastic initiation and DNA damage. 7,8 Interleukin-1 (IL-1) is a pro-inflammatory cytokine that exerts a wide range of biological actions, and several case-control studies have shown that IL-1 polymorphisms are significantly associated with different cancers . 9 The IL-1b gene encodes IL-1b, which is one of the most potent pro-inflammatory cytokines that initiates and amplifies both acute and chronic inflammation and is involved in various cellular actions, such as proliferation, differentiation and apoptosis. ...
Article
Full-text available
Objective: Oxidative stress caused by the pro-inflammatory cytokine interleukin (IL)-1b has been widely investigated for cancer risk. In this study, we focused on the role of IL-1b rs1143634 polymorphism to reveal its impact on cancer development. Methods: Related studies with fixed inclusion criteria were selected from electronic databases to May 2021. This meta-analysis was performed with odds ratios and 95% confidence intervals. Heterogeneity, publication bias, and sensitivity analyses were also conducted. Trial sequential analysis (TSA) and in-silico gene expression analysis were performed. Results: Forty-four case-control studies involving 18,645 patients with cancer and 22,882 controls were included. We observed a significant association of this single nucleotide polymorphism with overall cancer risk in the codominant model 3 (1.13-fold), recessive model (1.14-fold) and allelic model (1.08-fold). Subgroup analysis revealed that rs1143634 elevated the risk of gastric cancer, breast cancer and multiple myeloma. In addition, Asian and mixed populations and hospital-based controls had a significantly higher risk of cancer development. TSA confirmed our findings. Conclusion: Our meta-analysis revealed that the presence of IL-1b rs1143634 polymorphism increases the risk of cancer development. Among polymorphism carriers, the Asian population has a higher risk than other ethnic populations. This meta-analysis was registered retrospectively at INPLASY (https://inplasy.com/, INPLASY2021100044).
... Besides the immune cells, the levels of particular cytokines in plasma can also reflect the anti-tumor immune state in vivo, and may even be the indicator for predicting the prognosis of cancer patients [25]. IFN-c, a cytokine longestablished as being important in immunoregulation, predominantly plays a significant role in antitumor immunity [26]. ...
Article
Full-text available
Aim To investigate the antitumor efficacy of microwave ablation combined with dendritic cell-derived exosomes (Dex) or dendritic cells (DC) in treating hepatocellular carcinoma using a tumor-bearing mouse model. Methods We used a bilateral tumor-bearing mouse model treated with MWA, MWA + DC (DC-combined group) or MWA + Dex (Dex-combined group). Following tumor ablation on one side, the tumor volume on the contralateral side was monitored. The proportions of CD8⁺ (cytotoxic) T cells and regulatory T (Treg) cells in the spleen were analyzed by flow cytometry, and the number of CD8⁺ T cells and Treg cells in tumor sites was detected by immunohistochemistry. The concentration of interleukin-10 and interferon-γ in plasma was identified using enzyme-linked immunosorbent assay. Results The combination therapy significantly inhibited tumor growth compared with MWA monotherapy. In addition, the tumor immune microenvironment was significantly improved in HCC mice in the combination therapy groups compared to MWA group demonstrated by an increased number of CD8⁺ T cells and a decreased number of Treg cells in tumor sites. A lower proportion of Treg cells were observed in the spleen in the combination therapy groups compared to MWA group. Moreover, the concentration of plasma IFN-γ increased, and the concentration of plasma IL-10 decreased in the combination therapy groups compared to the MWA group. However, there was no statistical difference between the Dex-combined group and the DC-combined group in the comparisons mentioned above. Conclusions Our results provide evidence that MWA combined with Dex can significantly inhibit tumor growth and improve the immune microenvironment compared to MWA alone. Furthermore, the immune-enhancing effect of Dex and DC was equivalent in our combination therapy strategy.
... 10 Not surprisingly, therefore, IL-1Ra and other IL-1 family members play a role in disease progression in solid and hematological malignancies. 5,[12][13][14] In colorectal cancer, for instance, levels of circulating IL-1Ra are elevated and associated with clinicopathological features of disease, including tumor burden and metastasis. 6,7 The data are consistent with our study, which showed median IL-1Ra levels of 947 pg/ml; this level is significantly higher than the 150-300 pg/ml found in healthy people. ...
Article
Full-text available
Bermekimab is a true human monoclonal antibody that targets interleukin-1alpa (IL-1α), an inflammation-mediating alarmin. IL-1 receptor antagonist (IL-1Ra) is a natural molecule that blocks IL-1α activity by occupying the IL-1 receptor. The effect of endogenous IL-1Ra levels on the effectiveness of bermekimab is unknown. We investigated whether pre-treatment levels of circulating IL-1Ra, assessed by an enzyme-linked immunoassay, correlated with achievement of the primary outcome endpoint (effect on lean body mass and symptoms at week 8) in a Phase III study (2:1 randomization) of bermekimab versus placebo (each with best supportive care) in advanced colorectal cancer. Patients who responded to bermekimab in terms of achieving the primary endpoint had lower levels of IL-1Ra than non-responders (N = 204 patients; median = 843 vs. 035 pg/ml, p = 0.0092); no such relationship was observed in the placebo arm (N = 100 patients; 901 vs. 984 pg/ml, p = 0.55). Multivariate analysis corroborated that, in the bermekimab group, patients with lower baseline IL-1Ra levels were more likely to achieve the primary endpoint (odds ratio (OR) 1.7 (95% confidence interval (CI), 1.1 to 2.6), p = 0.017); in contrast, in the placebo arm, pre-treatment plasma IL-1Ra levels were not associated with outcome (OR 1.2 (95% CI 0.6 to 2.5), p = 0.57). The current findings demonstrate that, in a randomized phase III trial, patients with advanced colorectal cancer and lower levels of circulating IL-1Ra are more responsive to treatment with the IL-1α-targeting antibody bermekimab and these observations define a potential biomarker for anti-IL-1α therapy.
... IL-1 has been recently suggested to play a role in the development of breast cancer. Multifunctional cytokines are closely related to the development of inflammatory and immunological responses which play a key role in the pathogenesis of autoimmune and malignant diseases [7,19], making them an important candidate genes in BC. Considering the diverse roles of cytokines in cell growth, proliferation, differentiation and migration in inflammation and cancers, we proposed that polymorphisms in the cytokine genes could affect the risk of BC. ...
Article
Full-text available
Cytokines are known as important regulators of the cancer involved in inflammatory and immunological responses. This fact and plethora of gene polymorphism data prompted us to investigate IL1 gene polymorphisms in breast cancer (BC) patients. Totally, 530 patients with BC and 628 healthy control women were studied. The genetic polymorphisms for IL1 were analyzed by Massarray Sequencing method. Three single nucleotide polymorphisms (SNPs) identified in IL1B, IL1R1 gene are thought to influence breast cancer risk. The results of the association between IL-1B, IL1R1 polymorphisms and breast cancer risk have significant. We found that the variant TT genotype of rs10490571 was associated with a significantly increased breast cancer risk (TT vs. CC: OR = 2.82, 95% CI = 1.12-7.08, P = 0.047 for the codominant model). For rs16944 (AG vs. GG: OR = 0.60, 95% CI = 0.41-0.90, P = 0.034 for the codominant model) and rs1143623 (CG vs. CC: OR = 0.65, 95% CI = 0.45-0.94, P = 0.023 for the codominant model) have significant associations were found in genetic models. In conclusion, the present analysis suggests a correlation of polymorphic markers within the IL-1 gene locus with the risk in developing breast cancer. Taken together with our finding that IL1B, IL1R1 gene three SNP are also associated with the risk for the disease, we suggest that inflammation via innate and adaptive immunity contributes to multifactorial hereditary predisposition to pathogenesis of the breast cancer.
... Cytokines are documented to be closely associated with both inflammation and immune modulation while playing a key role in the development of liver damage in a variety of liver disease such as chronic hepatitis B virus (HBV) infection, alcoholic liver injury, nonalcoholic fatty liver disease, and drug-induced liver injury [5][6][7][8]. It is generally believed that cytokines are deregulated in many kinds of haematological disorders [9,10], while elevation of interleukin-(IL-) 6, IL-10, tumor necrosis factor-(TNF-) α, IL-8, and IL-2 receptor (IL-2R) was demonstrated valuable in the prediction of unfavorable prognosis in lymphoma [11][12][13][14]. ...
Article
Full-text available
Diffuse large B-cell lymphoma (DLBCL) is a heterogeneous group of lymphoma, with different clinical manifestation and prognosis. The International Prognostic Index (IPI), an index designed during the prerituximab era for aggressive lymphoma, showed variable values in the prediction of patient clinical outcomes. The aim of this study was to analyze the prognostic value and causes of pretreatment liver injury in 363 de novo DLBCL patients in our institution. Pretreatment liver impairment, commonly detected in lymphoma patients, showed significant association with poor outcomes and increased serum inflammatory cytokines in DLBCL patients but had no relation to hepatitis B virus replication nor lymphomatous hepatic infiltration. Multivariate analysis revealed that liver dysfunction, advanced Ann Arbor stage, and elevated lactate dehydrogenase (LDH) were independent adverse prognostic factors of both PFS and OS. Accordingly, a new liver-IPI prognostic model was designed by adding liver injury as an important factor in determining IPI score. Based on Kaplan-Meier curves for PFS and OS, the liver-IPI showed better stratification in DLBCL patients than either the IPI or the revised IPI in survival prediction.
... To date, the etiologies of urologic cancer are still largely unknown in spite of extensive studies. However, it has become evident recently that multiple immunomodulatory cytokines are implicated in the process of tumor genesis (Kurzrock 2001;Smyth et al. 2004). Among these cytokines, IL-10 is a multifunctional immunological regulator mainly produced by B cells, T cells and activated monocytes/marcophages. ...
Article
Full-text available
Background Interleukin-10 (IL-10) is a powerful modulator of anti-tumor immune responses. The IL-10 promoter region polymorphisms are known to regulate IL-10 production, and thus are thought to be implicated in tumorigenesis. Recently, the roles of these polymorphisms in urologic cancer have been extensively studied, with conflicting results. Therefore, we conducted the present meta-analysis to better elucidate the correlations between IL-10 polymorphisms and urologic cancer risk. Methods Eligible articles were searched in PubMed, Medline, Embase, Scopus and CNKI up to May 2016. Odds ratios (ORs) and 95% confidence intervals (CIs) were used to detect any potential associations between IL-10 polymorphisms and the risk of urologic cancer. ResultsA total of 22 case–control studies including 8572 patients and 9843 controls were analyzed. The overall meta-analysis results showed that IL-10 −592C>A polymorphism was significantly associated with urologic cancer in CA versus AA (P = 0.04, OR 0.87, 95% CI 0.76–0.99) and AA versus CC+CA (P = 0.03, OR 1.15, 95% CI 1.02–1.31). Subgroup analyses by cancer types suggested there were significant associations between all the three investigated IL-10 polymorphisms and bladder cancer. However, subgroup analyses by ethnicity only detected a weak association between IL-10 −819C>T and Asian population. Conclusions Our findings suggests that IL-10 −592C>A polymorphism may implicate with urologic cancer risk. Besides, promoter region polymorphisms of IL-10 may serve as potential biological markers, especially for bladder cancer. Furthermore, IL-10 −819C>T polymorphism may contribute to urologic cancer susceptibility in Asians while all the three studied variants of IL-10 did not relate to Caucasian urologic cancer predisposition.
... Among these factors, the hypothesis of release and receptor overexpression of pro-and anti-inflammatory cytokines secondary to cancer and chemotherapy has gained the most attention [15]. For instance, a consistent increase in interleukin-6 (IL-6), IL-10 and IL-1b, and consistent decrease in interleukin-1 receptor antagonist (IL-1RA) has been seen in hematological cancer patients [16]. ...
Article
Full-text available
Purpose of review Cancer-related fatigue (CRF) is the most common psychosomatic distress experienced by cancer patients before, during and after chemotherapy. Its impact on functional status and Health Related Quality of Life is a great concern among patients, healthcare professionals and researchers. The primary objective of this systematic review is to determine whether the different chemotherapies affect the association of CRF with individual pro- and anti-inflammatory cytokines. The PRISMA statement guideline has been followed to systematically search and screen article from PubMed and Embase. Recent findings This review has examined 14 studies which included a total of 1312 patients. These studies assayed 20 different kinds of cytokines. The cytokines interleukin-6, interleukin-1RA, TGF-b and sTNF-R2 were associated with CRF in patients receiving anthracycline-based chemotherapy. However, only interleukin-13 was identified in the taxane-based chemotherapy. Similarly, different sets of cytokines were linked with CRF in patients with chemotherapy regimens containing platinum, cyclophosphamides, topotecan or bleomycin. Summary This review has identified that cytokines are differentially linked with CRF according to the various types of chemotherapy regimens
Article
Rubropunctatin, a metabolite isolated from the fungi of the genus Monascus, is a natural lead compound applied for the suppression of tumors with good anti-cancer activity. However, its poor aqueous solubility has limited its further clinical development and utilization. Lecithin and chitosan are excellently biocompatible and biodegradable natural materials, which have been approved by the FDA as drug carrier. Here, we report for the first time the construction of a lecithin/chitosan nanoparticle drug carrier of the Monascus pigment rubropunctatin by electrostatic self-assembly between lecithin and chitosan. The nanoparticles are near-spherical with a size 110-120 nm. They are soluble in water and possess excellent homogenization capacity and dispersibility. Our in vitro drug release assay showed a sustained release of rubropunctatin. CCK-8 assays revealed that lecithin/chitosan nanoparticles loaded with rubropunctatin (RCP-NPs) had significantly enhanced cytotoxicity against mouse mammary cancer 4T1 cells. The flow cytometry results revealed that RCP-NPs significantly boosted cellular uptake and apoptosis. The tumor-bearing mice models we developed indicated that RCP-NPs effectively inhibited tumor growth. Our present findings suggest that lecithin/chitosan nanoparticle drug carriers improve the anti-tumor effect of the Monascus pigment rubropunctatin.
Article
Leukemia is among the most common types of hematological cancers and the use of herbal medicines to prevent and treat leukemia are under quick development. Among several molecular pathways involved in leukemia pathogenesis and exacerbations, purinergic signaling is revealed as a key component. In the present study, the effects of two doses (5 ug/mL and 10 ug/mL) of Immunity-6™, a phytocomplex composed by beta-glucan, green tea (Camelia sinensis), chamomile (Matricaria chamomilla), and ascorbic acid (vitamin C) was tested in vitro, using chronic myelogenous leukemia cell line (K-562; 5 ×104/mL/well), which were challenged with lipopolysaccharide (LPS; 1 ug/mL) for 24 h. The results demonstrated that both doses of Immunity-6™ inhibited ATP release (p < 0.001) and P2×7 receptor at mRNA levels expression (p < 0.001). Purinergic inhibition by Immunity-6™ was followed by reduced release of proinflammatory cytokines IL-1beta (p < 0.001) and IL-6 (p < 0.001), while only 5 ug/mL of Immunity-6™ reduced the release of TNF-alpha (p < 0.001). Beyond to inhibit the release of pro-inflammatory cytokines, both doses of Immunity-6™ induced the release of anti-inflammatory cytokine IL-10 (p < 0.001), while only the higher dose (10 ug/mL) of Immunity-6™ induced the release of anti-inflammatory IL-1ra (p < 0.05) and klotho (p < 0.001). Thus, Immunity-6™ may be a promising adjuvant in the treatment of leukemia and further clinical trials are guaranteed.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Article
Full-text available
Background: The interleukin-1-receptor antagonist IL1RA (encoded by the IL1RN gene) is a potent competitive antagonist to interleukin-1 (IL1) and thereby is mainly involved in the regulation of inflammation. Previous data indicated a role of IL1RA in muscle-invasive urothelial carcinoma of the bladder (UCB) as well as an IL1-dependent decrease in tissue barrier function, potentially contributing to cancer cell invasion. Objective: Based on these observations, here we investigated the potential roles of IL1RA, IL1A, and IL1B in bladder cancer cell invasion in vitro. Methods: Cell culture, real-time impedance sensing, invasion assays (Boyden chamber, pig bladder model), qPCR, Western blot, ELISA, gene overexpression. Results: We observed a loss of IL1RA expression in invasive, high-grade bladder cancer cell lines T24, UMUC-3, and HT1197 while IL1RA expression was readily detectable in the immortalized UROtsa cells, the non-invasive bladder cancer cell line RT4, and in benign patient urothelium. Thus, we modified the invasive human bladder cancer cell line T24 to ectopically express IL1RA, and measured changes in cell migration/invasion using the xCELLigence Real-Time-Cell-Analysis (RTCA) system and the Boyden chamber assay. The real-time observation data showed a significant decrease of cell migration and invasion in T24 cells overexpressing IL1RA (T24-IL1RA), compared to cells harboring an empty vector (T24-EV). Concurrently, tumor cytokines, e.g., IL1B, attenuated the vascular endothelial barrier, which resulted in a reduction of the Cell Index (CI), an impedance-based dimensionless unit. This reduction could be reverted by the simultaneous incubation with IL1RA. Moreover, we used an ex vivo porcine organ culture system to evaluate cell invasion capacity and showed that T24-IL1RA cells showed significantly less invasive capacity compared to parental T24 cells or T24-EV. Conclusions: Taken together, our results indicate an inverse correlation between IL1RA expression and tumor cell invasive capacity and migration, suggesting that IL1RA plays a role in bladder carcinogenesis, while the exact mechanisms by which IL1RA influences tumor cells migration/invasion remain to be clarified in future studies. Furthermore, we confirmed that real-time impedance sensing and the porcine ex vivo organ culture methods are powerful tools to discover differences in cancer cell migration and invasion.
Article
Background: Acute myeloid leukemia (AML) cells are highly resistant to chemotherapeutic drugs. Cytokines and adhesion molecules may contribute to this resistance and affect treatment outcome. The aim of this study was to evaluate the independence and additional prognostic information of baseline serum levels of selected cytokines and soluble adhesion molecules, included in analyses with standard prognostic indicators. Methods: We used biochip array technology to measure levels of selected cytokines and soluble adhesion molecules in serum samples of 80 newly diagnosed AML patients. The markers of tumour microenvironment were analysed against high risk karyotype, hyperleucocytosis, higher age, lactic dehydrogenase levels and presence of FLT3-ITD and NPM-1 mutation. Results: All evaluated analytes were independent of standard prognostic indicators. Fifteen were associated with overall and eight with progression-free survival in univariate analysis. After correction for multiple testing, we identified soluble interleukin-2 receptor-α as an independent indicator of overall survival. Further, the soluble type I TNF-α receptor was close to statistical significance for both overall and progression-free survival. Conclusions: Baseline levels of soluble interleukin-2 receptor-α predict overall survival in newly diagnosed AML. The TNF-α type I soluble receptor is a candidate prognostic marker in AML and is worth of further investigation.
Article
Recently, the roles of interleukin‐2 (IL‐2), IL‐4, IL‐6 and IL‐8 gene polymorphisms in gastric cancer (GC) have been extensively studied, with conflicting results. Therefore, we conducted the present meta‐analyses to better elucidate the roles of interleukin gene polymorphisms in GC. Eligible articles were searched in PubMed, Medline, Embase, Web of Science and CNKI. Odds ratios (ORs) and 95% confidence intervals (CIs) were used to detect any potential association between interleukin gene polymorphisms and the risk of GC. A total of 63 case‐control studies was finally included in our analyses. Significant associations with the risk of GC were detected for the IL‐6 rs1800796 and IL‐8 rs4073 polymorphisms in overall analyses. Further subgroup analyses based on ethnicities of participants revealed that the IL‐4 rs2243250, IL‐6 rs1800796 and IL‐8 rs4073 polymorphisms were significantly associated with the risk of GC in Asians. Moreover, IL‐8 rs4073 polymorphism was also significantly associated with the risk of GC in Africans. In conclusion, our findings suggested that IL‐4 rs2243250, IL‐6 rs1800796 and IL‐8 rs4073 polymorphisms may serve as genetic biomarkers of GC. This article is protected by copyright. All rights reserved.
Article
The growing resistance against conventional chemotherapy in acute myeloid leukemia (AML) is a noticeable clinical concern. Therefore, many researchers are looking for novel substances to overcome drug resistance in cancer.Staphylococcal enterotoxin B (SEB) is a superantigen (SAg) and a promising compound whichhas lethal effects on malignant cells. Inthis unprecedented study, SEB was used against U937 cells in a co‐culture system in the presence of human bone marrow‐mesenchymal stem cells (hBM‐MSCs). The effects of hBM‐MSCs on the proliferation and survival of U937 cell line with SEB was assessed using MTT assay and AnnexinV/PI flowcytometry, respectively. Moreover, the expression of IL‐6, IL‐10, TGF‐β, and IKKb was evaluated by real‐time PCR technique. Same experiments were also carried out using hBM‐MSCs‐conditioned medium (hBM‐MSCs‐CM).The results showed that SEB reduced the proliferation and survival of U937 cell line, but hBM‐MSCs or hBM‐MSCs‐CM suppressed the effects of SEB. Furthermore, real‐timePCR demonstrated that SEB could decrease the expression of IL‐6, IL‐10, and TGF‐β in hBM‐MSCs (P<0.05), while the production of IKKb was increased in comparison with the control group.These findings help us to have a broader understanding ofthe usage of SEB in the treatment of hematological malignancies, especially if it is targeted against hBM‐MSCs to disrupt their supportive effects on malignant cells. This article is protected by copyright. All rights reserved.
Chapter
Cancer Symptom Science is the first interdisciplinary compilation of research on the mechanisms underlying the expression of cancer-related symptoms. It presents innovations in clinical, animal and in vitro research, research methods in brain imaging, and statistical-descriptive approaches to understanding the mechanistic basis of symptom expression. This volume also provides perspectives from patients, government and industry. By collecting and synthesizing the developing threads of new approaches to understanding cancer-related symptoms, the book promotes a pioneering framework for merging behavioral and biological disciplines to clarify mechanisms of symptom evolution, incorporating new technologies, testing novel agents for symptom control, and improving patient functioning and quality of life both during and after cancer treatment. With an expert editorial team led by Charles S. Cleeland, an internationally-recognized leader in cancer pain assessment and treatment, this is essential reading for surgical, clinical and medical oncologists, academic researchers, and pharmaceutical companies developing new agents to control symptom expression.
Chapter
This chapter proposes a generalized model of formal immune network (FIN) based on self-organizing features of apoptosis (programmed cell death) and autoimmunization both induced by cytokines (messenger proteins). The chapter also describes real-world applications of such FIN to intrusion detection in computer networks and forecast of hydro-physical fields in the ocean. The obtained results demonstrate that FIN outperforms (by training time and accuracy) other approaches of computational intelligence as well as more conventional methods of interpolation.
Article
Full-text available
The introduction of allosteric receptor–receptor interactions in G protein-coupled receptor (GPCR) heteroreceptor complexes of the central nervous system (CNS) gave a new dimension to brain integration and neuropsychopharmacology. The molecular basis of learning and memory was proposed to be based on the reorganization of the homo- and heteroreceptor complexes in the postjunctional membrane of synapses. Long-term memory may be created by the transformation of parts of the heteroreceptor complexes into unique transcription factors which can lead to the formation of specific adapter proteins. The observation of the GPCR heterodimer network (GPCR-HetNet) indicated that the allosteric receptor–receptor interactions dramatically increase GPCR diversity and biased recognition and signaling leading to enhanced specificity in signaling. Dysfunction of the GPCR heteroreceptor complexes can lead to brain disease. The findings of serotonin (5-HT) hetero and isoreceptor complexes in the brain over the last decade give new targets for drug development in major depression. Neuromodulation of neuronal networks in depression via 5-HT, galanin peptides and zinc involve a number of GPCR heteroreceptor complexes in the raphe-hippocampal system: GalR1-5-HT1A, GalR1-5-HT1A-GPR39, GalR1-GalR2, and putative GalR1-GalR2-5-HT1A heteroreceptor complexes. The 5-HT1A receptor protomer remains a receptor enhancing antidepressant actions through its participation in hetero- and homoreceptor complexes listed above in balance with each other. In depression, neuromodulation of neuronal networks in the raphe-hippocampal system and the cortical regions via 5-HT and fibroblast growth factor 2 involves either FGFR1-5-HT1A heteroreceptor complexes or the 5-HT isoreceptor complexes such as 5-HT1A-5-HT7 and 5-HT1A-5-HT2A. Neuromodulation of neuronal networks in cocaine use disorder via dopamine (DA) and adenosine signals involve A2AR-D2R and A2AR-D2R-Sigma1R heteroreceptor complexes in the dorsal and ventral striatum. The excitatory modulation by A2AR agonists of the ventral striato-pallidal GABA anti-reward system via targeting the A2AR-D2R and A2AR-D2R-Sigma1R heteroreceptor complex holds high promise as a new way to treat cocaine use disorders. Neuromodulation of neuronal networks in schizophrenia via DA, adenosine, glutamate, 5-HT and neurotensin peptides and oxytocin, involving A2AR-D2R, D2R-NMDAR, A2AR-D2R-mGluR5, D2R-5-HT2A and D2R-oxytocinR heteroreceptor complexes opens up a new world of D2R protomer targets in the listed heterocomplexes for treatment of positive, negative and cognitive symptoms of schizophrenia.
Article
Full-text available
In the present study we demonstrate that human monocytes activated by lipopolysaccharides (LPS) were able to produce high levels of interleukin 10 (IL-10), previously designated cytokine synthesis inhibitory factor (CSIF), in a dose dependent fashion. IL-10 was detectable 7 h after activation of the monocytes and maximal levels of IL-10 production were observed after 24-48 h. These kinetics indicated that the production of IL-10 by human monocytes was relatively late as compared to the production of IL-1α, IL-1β, IL-6, IL-8, tumor necrosis factor Oi(TNFα), and granulocyte colony-stimulating factor (G-CSF), which were all secreted at high levels 4-8 h after activation. The production of IL-10 by LPS activated monocytes was, similar to that of IL-1α, IL-1β, IL-6, IL-8, TNFα, granulocyte-macrophage colony-stimulating factor (GM-CSF), and G-CSF, inhibited by IL-4. Furthermore we demonstrate here that IL-10, added to monocytes, activated by interferon γ(IFN-γ), LPS, or combinations of LPS and IFN-γ at the onset of the cultures, strongly inhibited the production of IL-1α, IL-1β, IL-6, IL-8, TNFα, GM-CSF, and G-CSF at the transcriptional level. Viral-IL-10, which has similar biological activities on human cells, also inhibited the production of TNFα and GM-CSF by monocytes following LPS activation. Activation of monocytes by LPS in the presence of neutralizing anti-IL-10 monoclonal antibodies resulted in the production of higher amounts of cytokines relative to LPS treatment alone, indicating that endogenously produced IL-10 inhibited the production of IL-1α, IL-1β, IL-6, IL-8, TNFα, GM-CSF, and G-CSF. In addition, IL-10 had autoregulatory effects since it strongly inhibited IL-10 mRNA synthesis in LPS activated monocytes. Furthermore, endogenously produced IL-10 was found to be responsible for the reduction in class II major histocompatibility complex (MHC) expression following activation of monocytes with LPS. Taken together our results indicate that IL-10 has important regulatory effects on immunological and inflammatory responses because of its capacity to downregulate class II MHC expression and to inhibit the production of proinflammatory cytokines by monocytes.
Article
Full-text available
The biological role of interleukin 6 (IL-6) molecules in human B cell tumorigenesis was studied by using an episomal expression vector, pHEBoSV-IL6, to introduce stably the human IL-6 gene into human Epstein Barr virus (EBV)-transformed B lymphoblasts. The gene was present in the IL-6-transfected cells in a high copy number and was efficiently expressed, resulting in the secretion of consistent levels of IL-6 molecules. The constitutive expression of the IL-6 gene led to an altered pattern of growth and to a malignant phenotype, as shown by clonogenicity in to an altered pattern of growth and to a malignant phenotype, as shown by clonogenicity in soft agar cultures and tumorigenicity in nude mice. These data suggest that the combined action of EBV, which exerts an immortalizing function, and of the growth-promoting activity of IL-6 molecules, can give rise to fully transformed B cell tumors in immunodeficient subjects.
Article
Full-text available
Interleukin 10 (IL-10) and viral IL-10 (v-IL-10) strongly reduced antigen-specific proliferation of human T cells and CD4+ T cell clones when monocytes were used as antigen-presenting cells. In contrast, IL-10 and v-IL-10 did not affect the proliferative responses to antigens presented by autologous Epstein-Barr virus-lymphoblastoid cell line (EBV-LCL). Inhibition of antigen-specific T cell responses was associated with downregulation of constitutive, as well as interferon gamma- or IL-4-induced, class II MHC expression on monocytes by IL-10 and v-IL-10, resulting in the reduction in antigen-presenting capacity of these cells. In contrast, IL-10 and v-IL-10 had no effect on class II major histocompatibility complex (MHC) expression on EBV-LCL. The reduced antigen-presenting capacity of monocytes correlated with a decreased capacity to mobilize intracellular Ca2+ in the responder T cell clones. The diminished antigen-presenting capacities of monocytes were not due to inhibitory effects of IL-10 and v-IL-10 on antigen processing, since the proliferative T cell responses to antigenic peptides, which did not require processing, were equally well inhibited. Furthermore, the inhibitory effects of IL-10 and v-IL-10 on antigen-specific proliferative T cell responses could not be neutralized by exogenous IL-2 or IL-4. Although IL-10 and v-IL-10 suppressed IL-1 alpha, IL-1 beta, tumor necrosis factor alpha (TNF-alpha), and IL-6 production by monocytes, it was excluded that these cytokines played a role in antigen-specific T cell proliferation, since normal antigen-specific responses were observed in the presence of neutralizing anti-IL-1, -IL-6, and -TNF-alpha mAbs. Furthermore, addition of saturating concentrations of IL-1 alpha, IL-1 beta, IL-6, and TNF-alpha to the cultures had no effect on the reduced proliferative T cell responses in the presence of IL-10, or v-IL-10. Collectively, our data indicate that IL-10 and v-IL-10 can completely prevent antigen-specific T cell proliferation by inhibition of the antigen-presenting capacity of monocytes through downregulation of class II MHC antigens on monocytes.
Article
Full-text available
Recombinant mouse interleukin 10 (IL-10) was exceedingly potent at suppressing the ability of mouse peritoneal macrophages (m phi) to release tumor necrosis factor alpha (TNF-alpha). The IC50 of IL-10 for the suppression of TNF-alpha release induced by 0.5 microgram/ml lipopolysaccharide was 0.04 +/- 0.03 U/ml, with as little as 1 U/ml suppressing TNF-alpha production by a factor of 21.4 +/- 2.5. At 10 U/ml, IL-10 markedly suppressed m phi release of reactive oxygen intermediates (ROI) (IC50 3.7 +/- 1.8 U/ml), but only weakly inhibited m phi release of reactive nitrogen intermediates (RNI). Since TNF-alpha is a T cell growth and differentiation factor, whereas ROI and RNI are known to inhibit lymphocyte function, it is possible that m phi exposed to low concentrations of IL-10 suppress lymphocytes. m phi deactivated by higher concentrations of IL-10 might be permissive for the growth of microbial pathogens and tumor cells, as TNF-alpha, ROI, and RNI are major antimicrobial and tumoricidal products of m phi. IL-10's effects on m phi overlap with but are distinct from the effects of the two previously described cytokines that suppress the function of mouse m phi, transforming growth factor beta and macrophage deactivation factor. Based on results with neutralizing antibodies, all three m phi suppressor factors appear to act independently.
Article
Full-text available
We have demonstrated the existence of human cytokine synthesis inhibitory factor (CSIF) [interleukin 10 (IL-10)]. cDNA clones encoding human IL-10 (hIL-10) were isolated from a tetanus toxin-specific human T-cell clone. Like mouse IL-10, hIL-10 exhibits strong DNA and amino acid sequence homology to an open reading frame in the Epstein-Barr virus, BCRFI. hIL-10 and the BCRFI product inhibit cytokine synthesis by activated human peripheral blood mononuclear cells and by a mouse Th1 clone. Both hIL-10 and mouse IL-10 sustain the viability of a mouse mast cell line in culture, but BCRFI lacks comparable activity in this assay, suggesting that BCRFI may have conserved only a subset of hIL-10 activities.
Article
Full-text available
The biological role of interleukin 6 (IL-6) molecules in human B cell tumorigenesis was studied by using an episomal expression vector, pHEBoSV-IL6, to introduce stably the human IL-6 gene into human Epstein Barr virus (EBV)-transformed B lymphoblasts. The gene was present in the IL-6-transfected cells in a high copy number and was efficiently expressed, resulting in the secretion of consistent levels of IL-6 molecules. The constitutive expression of the IL-6 gene led to an altered pattern of growth and to a malignant phenotype, as shown by clonogenicity in to an altered pattern of growth and to a malignant phenotype, as shown by clonogenicity in soft agar cultures and tumorigenicity in nude mice. These data suggest that the combined action of EBV, which exerts an immortalizing function, and of the growth-promoting activity of IL-6 molecules, can give rise to fully transformed B cell tumors in immunodeficient subjects.
Article
Full-text available
Article
Full-text available
Interleukin-6 (IL-6) is a growth factor with diverse biologic activity. Originally described as a T-cell product that enhances immunoglobulin (Ig) secretion in antigen-stimulated B cells, it also affects the growth of T cells, plasmacytomas, hybridomas, and hematopoietic stem cells. We report the expression and secretion of IL-6 by two lymphoma cell lines, OCI-LY3 and OCI-LY12. Addition of recombinant IL-6 stimulated their growth, whereas addition of polyclonal anti-recombinant IL-6 (anti-rIL-6) had a marked inhibitory effect on proliferation. These results suggest an autocrine role for IL-6 in the growth of these lymphoma cells in culture.
Article
Full-text available
We report the results of a phase I study of the tolerance and biologic activity of intramuscularly (IM)-administered recombinant interferon-gamma (rIFN-gamma). Forty-four patients with metastatic cancer were given rIFN-gamma at doses ranging from 0.01 to 2.5 mg/m2/d for 42 days. The most common side effects were fever, flulike symptoms, night sweats, and granulocytopenia. The maximum tolerated dose was 0.5 mg/m2/d. Administration of rIFN-gamma resulted in modulation of immune system functions, including induction of major histocompatibility complex-associated antigens on blood leukocytes, an increase in blood surface immunoglobulin-bearing B cell and natural killer (NK) cell number, and NK cell cytotoxicity. Serum lysozyme, determined as an estimate of tissue macrophage activity, also increased. Serum assays for anti-interferon antibodies were negative in all patients. Five of eight evaluable patients with lymphoproliferative disorders showed objective evidence of tumor regression consisting of partial responses (two patients), and minor responses (three patients). These data suggest that further phase II studies of IM-administered rIFN-gamma are indicated.
Article
Full-text available
IL-6, a multifunctional cytokine produced by monocytes, fibroblasts, and endothelial cells, promotes the growth of EBV-immortalized B cells in vitro and renders these cells tumorigenic in athymic mice. In the present study, serum/plasma IL-6 bioactivity was found to be abnormally elevated, albeit transiently, in 17 of 18 solid organ transplant recipients with posttransplant lymphoproliferative disease (PTLD), with a mean maximal level of 196.7 U/ml. This represents a 16.4 increase above the normal mean (11.3 U/ml). In contrast, only 3 of 10 solid organ transplant recipients with uncomplicated courses posttransplant had abnormally elevated serum/plasma IL-6 bioactivity (mean maximal level 41.4 U/ml, P = 0.0007). When transferred to single cell culture, the 11 PTLD tissues produced 640 to 1.25 x 10(6) IL-6 U/ml in the culture supernatant, with a mean maximal level of 35,025 IL-6 U/ml. Cell separation experiments demonstrated that the adherent cells, identified as non-B cells, were the principal source of IL-6 production in vitro by PTLD tissue. Control cultures of inflammatory lymphoid tissue negative for lymphoproliferative disease as well as of PBL from patients with acute EBV-induced infectious mononucleosis consistently produced < 10 IL-6 U/ml. Thus, IL-6 is produced at high levels by PTLD tissues and may play a critical role in the pathogenesis of PTLD.
Article
Full-text available
The in vivo production of interleukin (IL)-10, IL-6, IL-2, and tumor necrosis factor (TNF)-alpha in tumor samples was investigated by immunohistochemistry in 54 non-Hodgkin's lymphomas (NHLs). Respectively, 55, 89, 23, and 29% of tumor samples were found positive for IL-10, IL-6, IL-2, and TNF-alpha expression by immunohistochemistry. Using reverse transcription-PCR, the mRNA of IL-10 and IL-6 were detectable in all samples tested and in 90 and 34% of the samples for TNF-alpha and IL-2, respectively. In 13 patients, fresh tumor tissue was available for B NHL cell purification with Dynabeads. IL-10, IL-6, IL-2, and TNF-alpha were detectable in the supernatant of 38, 100, 0, and 23% of purified tumor cell preparations (PTCPs), respectively. All patients with detectable IL-10 in culture had increased serum IL-10. IL-6 production by tumor cells and serum IL-6 levels were also found to be highly correlated (P < 0.0001). This suggests that tumor cells are a major source of serum IL-1O and IL-6 in these patients. Exogenous IL-10, IL-6, IL-2, and TNF-alpha significantly enhanced the [3H]thymidine uptake in 13 of 13 (100%), 5 of 13 (38%), 9 of 13 (69%), and 2 of 10 (20%) PTCPs costimulated with anti-CD40, respectively. IL-2, IL-6, and TNF-alpha synergized with IL-10 in 54, 23, and 30% of PTCPs. The combination of IL-10, IL-2, and IL-6 induced the maximal level of proliferation in 12 (92%) of 13 PTCPs. CD40 ligand mRNA expression was also detectable in vivo using reverse transcription-PCR in 28 of the 29 (97%) tumor samples tested, including 11 of those tested for [3H]thymidine incorporation. These results show that IL-1O, IL-6, IL-2, and TNF-alpha are produced in NHL tumors and may cooperate in vivo to increase NHL cell proliferation.
Article
Full-text available
The objective of this study was to examine the correlation between serum interleukin-6 (IL-6) and IL-10 levels and outcome in chronic lymphocytic leukemia (CLL). Serum IL-6 and IL-10 levels were measured by enzyme-linked immunoabsorbent assays from 159 and 151 CLL patients, respectively, and from healthy control subjects (n = 55 [IL-6]; n = 37 [IL-10]). Cytokine levels were correlated with clinical features and survival. Serum IL-6 levels were higher in CLL patients (median, 1.45 pg/mL; range, undetectable to 110 pg/mL) than in control subjects (median, undetectable; range, undetectable to 4. 30 pg/mL) (P <.0001). Serum IL-10 levels were higher in CLL patients (median, 5.04 pg/mL; range, undetectable to 74 pg/mL) than in normal volunteers (median, undetectable; range, undetectable to 13.68 pg/mL) (P <.00001). Assays measuring both Epstein-Barr virus-derived and human IL-10 yielded higher values than assays measuring primarily human IL-10 (P <.05). Patients with elevation of serum IL-6 or IL-10 levels, or both, had worse median and 3-year survival (log rank P <.001) and unfavorable characteristics (prior treatment, elevated beta(2)-microglobulin or lactate dehydrogenase, or Rai stage III or IV). Elevated IL-6 and IL-10 levels were independent prognostic factors for survival when analyzed individually or in combination (Cox regression analysis). However, if beta(2)-microglobulin was incorporated into the analysis, it was selected as an independent prognostic feature, and IL-6/IL-10 were no longer selected. In patients with CLL, serum IL-6 and IL-10 (viral and human) levels are elevated and correlate with adverse disease features and short survival. In multivariate analysis, however, beta(2)-microglobulin is the most important prognostic factor.
Article
We have recently demonstrated that interleukin (IL)-1 beta levels are elevated in advanced chronic myelogenous leukemia (CML) and that IL-1 inhibitors can suppress CML clonogenic growth. To further assess the clinical implications of increased IL-1 beta expression in CML, we analyzed IL-1 beta and IL-1 receptor antagonist (IL-1RA) levels in leukocyte lysates from a series of CML patients and from normal volunteers. Both IL-1 beta and IL-1RA were measured by enzyme-linked immunosorbent assays (ELISAs), with the lower limits of sensitivity of the assays being 20 pg/mL and 6.5 pg/mL, respectively. The median IL-1 beta level in the 81 CML patients tested was higher (115.8 pg/2.4 x 10(7) cells; range, 0 to 2,000 pg/2.4 x 10(7) cells) than the median level in 25 control samples (10.8 pg/2.4 x 10(7) cells; range, 0 to 95.5 pg/2.4 x 10(7) cells) (P < .01). IL-1 beta was bioactive, as demonstrated with a bioassay based on cytotoxicity to a melanoma cell line (A375). For survival analysis, elevated IL-1 beta levels were defined as those exceeding the mean + 2 SD of normal levels (83 pg/2.4 x 10(7) cells). The survival of the 44 patients with elevated IL-1 beta levels was significantly shorter than that of those who had low IL-1 beta levels (median, 44 v 58 months; P = .049 by Wilcoxon-Gehan method). An association between IL-1 beta and CML prognostic criteria shows that IL-1 beta levels were significantly higher in patients in accelerated/blastic crisis phases of the disease (364.0 pg/2.4 x 10(7) cells) compared with patients in chronic phase (102.0 pg/2.4 x 10(7) cells) (P < .01), and that high IL-1 beta levels correlated with increased blasts in the marrow and peripheral blood (P < .01). In contrast, while IL-1RA levels did not differ between chronic-phase CML patients (median, 471.7 pg/2.4 x 10(5)) and healthy volunteers (median, 454.4 pg/2.4 x 10(5)), patients with accelerated/blast crisis disease had significantly lower levels of IL-1RA (median, 218.7 pg/2.4 x 10(5); P = .03). Finally, although IL-1 beta has been previously shown to increase IL-1RA levels, there was no correlation between IL-1 beta and IL-1RA levels in our CML patients.(ABSTRACT TRUNCATED AT 400 WORDS)
Article
Interleukin-1 (IL-1) has recently been reported to play an important role in acute myelogenous leukemia (AML) blast proliferation. We therefore investigated the effect of soluble IL-1 receptors (sIL-1R) and IL-1 receptor antagonist (IL-1RA) on the growth of AML bone marrow blast progenitors from 25 patients. In the AML blast colony culture assay, sIL-1R and IL-1RA inhibited blast colony-forming cell replication in a dose-dependent fashion, at concentrations ranging from 10 to 500 ng/mL (sIL-1R) and 10 to 1,000 ng/mL (IL-1RA), and their inhibitory effect was partially reversed by IL-1 beta. A similar inhibitory effect was also noted with the use of anti-IL-1 beta neutralizing antibodies. When AML blast progenitors were grown either in the presence of fetal calf serum (FCS) alone or with one of the following: phytohemagglutinin leukocyte-conditioned medium (PHA-LCM), granulocyte-macrophage colony-stimulating factor (GM-CSF), G-CSF, interleukin-3 (IL-3), or stem cell factor (SCF), addition of 100 ng/mL sIL-1R or IL-1RA inhibited blast colony formation by 3% to 96% and 2% to 97%, respectively. In sharp contrast, neither of these IL-1- inhibitory molecules significantly inhibited proliferation of normal marrow hematopoietic progenitors. Lysates of 2 x 10(7) low-density AML marrow cells were tested for intrinsic IL-1 beta content using an enzyme-linked immunoadsorbant assay (ELISA). Samples from five of six patients showed high concentrations (ranging from 501 to 2,041 pg), whereas 2 x 10(7) cells from two normal marrow aspirates yielded 54.6 pg of IL-1 beta. AML blast colony-forming cells from all six patients were inhibited by sIL-1R, IL-1RA, or both. Incubation of nine samples of AML low-density cells with either sIL-1R or IL-1RA reduced GM-CSF concentrations in cell lysates, and supernatants from nine (P less than .01) and six samples (P less than .037), respectively, and G-CSF concentration in lysates from six of nine samples (P less than .03), and in supernatants from five of six samples (P less than .06) when studied by ELISAs. Our data implicate IL-1 in AML blast proliferation and suggest the potential benefits of using IL-1-inhibitory molecules in future therapies for AML.
Article
Philadelphia chromosome1 positive (Ph1) chronic myelogenous leukemia (CML) is characterized by metamorphosis of the chronic phase to blastic crisis. However, cellular events associated with this transition are poorly understood. To examine the possible participation of hematopoietic growth factors in this process, we studied growth factor expression in adherent layers of bone marrows derived from CML Ph1 patients in various stages of the disease. Interleukin-1 beta (IL-1 beta) and IL-6 mRNA were expressed in five of six patients, and granulocyte-macrophage colony-stimulating factor (GM-CSF) in one of six patients with myeloid/undifferentiated blast crisis. In addition, leukemia inhibitory factor (LIF) expression was increased in four of six patients with myeloid/undifferentiated blast crisis phase of the disease. IL-1 beta was also detected in bone marrow adherent layer conditioned medium from two of these patients. These results were in sharp contrast to the lack of detectable levels of uninduced IL-1 beta, IL-6, and GM-CSF mRNA, in samples derived from 4 patients in lymphoid blastic crisis, 3 in accelerated, and 11 in chronic phases of the disease, or from normal controls. The possibility of a paracrine loop formation, whereby the adherent layers representing the bone marrow stroma are induced to express hematopoietic growth factors, was supported by our finding IL-1 beta mRNA expression in the leukemic blast cells in three of four studied patients in blast crisis and IL-1 beta protein production in seven of eight patients studied. Finally, coculturing CML blast crisis cells onto pre-established adherent layers induced the expression of both IL-1 beta and IL-6 genes. From this preliminary study, it appears that abnormal expression of growth factors is a common event with CML Ph1 progression. We hypothesize that IL-1 beta generated by the transformed malignant clone stimulates the marrow stroma to produce various growth factors, and that this process may play a role in disease progression.
Article
In this study, we investigated the role of interleukin-1 beta (IL-1 beta) in the malignant evolution of chronic myelogenous leukemia (CML) and the functional activity of IL-1 inhibitors. Bone marrow (BM) and peripheral blood (PB) low-density cells from 38 CML patients were studied in the colony-forming unit-granulocyte, erythrocyte, monocyte, megakaryocyte colony culture assay. Samples from patients with early stage, interferon-alpha (IFN)-sensitive disease formed hematopoietic colonies in the presence of fetal calf serum (FCS), erythropoietin (Epo), and one of the following: granulocyte-macrophage colony- stimulating factor (10 ng/mL), IL-3 (15 ng/mL), both, or phytohemagglutinin-conditioned medium. The addition of IL-1 beta augmented IFN-sensitive CML colony growth in a dose-dependent manner at concentrations of 10 to 100 U/mL. In sharp contrast, addition of the above growth factors did not augment the colony growth-promoting effect of FCS and Epo in samples from IFN-resistant patients; further, adherent cell fractionation or T-lymphocyte depletion attenuated the “autonomous” colony growth. Lysates of 2.5 x 10(7) low-density cells from each of six IFN-resistant and six IFN-sensitive CML patients and three normal volunteers were tested for intrinsic IL-1 beta content in an enzyme-linked immunosorbent assay and yielded a mean of 610 pg, 54.6 pg, and 49.4 pg of IL-1 beta, respectively (P less than .045). Interestingly, both soluble IL-1 receptors (sIL-1R) and IL-1 receptor antagonist (IL-1RA) at concentrations of 5 to 100 ng/mL (sIL-1R) and 10 to 500 ng/mL (IL-1RA) inhibited CML colony growth in a dose-dependent fashion, with maximal inhibition of 64% and 65%, respectively. A similar effect was noted with the use of anti-IL-1 beta neutralizing antibodies. These data implicate IL-1 beta in CML disease progression and suggest that the inhibitory effects of molecules such as sIL-1R and IL-1RA could conceivably be the basis of a novel therapeutic strategy against this disorder.
Article
The polypeptide cytokine interleukin-1 (IL-1) affects nearly every tissue and organ system. IL-1 is the prototype of the pro-inflammatory cytokines in that it induces the expression of a variety of genes and the synthesis of several proteins that, in turn, induce acute and chronic inflammatory changes. IL-1 is also the prototypic “alarm' cytokine in that it brings about increases in a variety of defense mechanisms, particularly immunologic and hematologic responses. Most studies on the biology of IL-1 have been performed in animals, but human subjects have recently been injected with recombinant IL-1 and the results confirm the two fundamental properties of IL-1 as being both a mediator of disease as well as of host defense. However, in either situation, over or continued production of IL-1 leads to debilitation of normal host functions; therefore, reduction of IL-1 synthesis or its effects becomes a target of therapy in many diseases. In this review, the structure, gene expression, synthesis, and secretion of IL-1 are described. In addition, the two IL-1 surface receptors, possible signal transduction mechanisms, various biologic activities, and production of IL-1 during disease states are discussed. Similarities and differences between IL-1, tumor necrosis factor, and IL-6 are presented. Although various agents for reducing the synthesis and/or for antagonizing the effects of IL-1 have been proposed, the recent cloning of a naturally occurring IL-1 receptor antagonist (IL- 1ra) has opened new experimental and clinical approaches. The ability of this IL-1ra to block the triggering of IL-1 receptors in animals without agonist effects has reduced the severity of diseases such as hemodynamic shock, lethal sepsis, inflammatory bowel disease, experimental arthritis, and the spontaneous proliferation of human leukemic cells.
Article
Abstract Culture of human peripheral blood mononuclear cells (PBMC) with IL-2 stimulates synthesis of cytokines and generation of lymphokine-activated killer (LAK) activity. Both IL-4 and IL-10 [cytokine synthesis inhibitory factor (CSIF)] inhibit IL-2-lnduced synthesis of IFN-γ and tumor necrosis factor (TNF)-α by human PBMC. However, unlike IL-4, IL-10 Inhibits neither IL-2-lnduced proliferation of PBMC and fresh natural killer (NK) cells, nor IL-2-lnduced LAK activity. Moreover, IL-4 inhibits IL-2-lnduced IFN-γ synthesis by purified fresh NK cells, while In contrast the inhibitory effect of IL-10 Is mediated by CD14+ cells (monocytes/macrophages). IL-10 inhibits TNF-a synthesis by monocytes or monocytes plus NK cells, but not by NK cells alone. These results suggest that IL-4 and IL-10 act on NK cells via distinct pathways, and that IL-2-lnduced cytokine synthesis and LAK activity are regulated via different mechanisms.
Article
The objective of this study was to examine the correlation between serum interleukin-6 (IL-6) and IL-10 levels and outcome in chronic lymphocytic leukemia (CLL). Serum IL-6 and IL-10 levels were measured by enzyme-linked immunoabsorbent assays from 159 and 151 CLL patients, respectively, and from healthy control subjects (n = 55 [IL-6]; n = 37 [IL-10]). Cytokine levels were correlated with clinical features and survival. Serum IL-6 levels were higher in CLL patients (median, 1.45 pg/mL; range, undetectable to 110 pg/mL) than in control subjects (median, undetectable; range, undetectable to 4.30 pg/mL) ( P < .0001). Serum IL-10 levels were higher in CLL patients (median, 5.04 pg/mL; range, undetectable to 74 pg/mL) than in normal volunteers (median, undetectable; range, undetectable to 13.68 pg/mL) ( P < .00001). Assays measuring both Epstein-Barr virus-derived and human IL-10 yielded higher values than assays measuring primarily human IL-10 ( P < .05). Patients with elevation of serum IL-6 or IL-10 levels, or both, had worse median and 3-year survival (log rank P < .001) and unfavorable characteristics (prior treatment, elevated β2-microglobulin or lactate dehydrogenase, or Rai stage III or IV). Elevated IL-6 and IL-10 levels were independent prognostic factors for survival when analyzed individually or in combination (Cox regression analysis). However, if β2-microglobulin was incorporated into the analysis, it was selected as an independent prognostic feature, and IL-6/IL-10 were no longer selected. In patients with CLL, serum IL-6 and IL-10 (viral and human) levels are elevated and correlate with adverse disease features and short survival. In multivariate analysis, however, β2-microglobulin is the most important prognostic factor.
Article
With the aim of using interleukin-6 (IL-6)-inducible promoters to express transgenes, we investigated the long-term consequences of high levels of IL-6 in mice. As a first step, we generated transgenic mice constitutively expressing the murine IL-6 at a level sufficient to induce IL-6-responsive genes. These mice were analyzed with respect to the indirect and direct consequences of elevated IL-6 expression over a time period of about 2 years. Although biologically active IL-6 was expressed from the transgene and different alterations could be documented (less immature B cells in bone marrow, expression of IL-6-inducible liver genes), the mice appeared healthy and could easily be used for breeding. Only in mice older than 18 months did we find a high incidence of lymphomas associated with different tissues. These results indicate that the side effects of long-term treatment with IL-6 are relatively moderate, and that IL-6 might be used to mediate the expression of heterologous genes in the context of functional studies.
Article
Interleukin (IL) 6 was measured in the serum of 138 patients with metastatic renal carcinoma before the initiation of IL-2 treatment. IL-6 was detectable in 66 patients with renal cancer (48%) and in only 8 of 70 normal adults (11%). Serum C reactive protein (CRP) and IL-6 levels are correlated, suggesting that IL-6 is involved in CRP increase in these patients. The interval between diagnosis of the primary tumor and metastasis was shorter in patients with a detectable serum IL-6 and/or serum CRP level greater than 50 mg/liter. Serum IL-6 and CRP levels were higher in subgroups of patients previously defined as having a poor life expectancy according to the Eastern Cooperative Oncology Group criteria. Pretreatment concentrations of IL-6 and CRP were higher in patients who experienced progressive disease after IL-2 treatment. Patients with detectable IL-6 had a shorter survival from the beginning of IL-2 treatment than patients without circulating IL-6 (median, 8 versus 16 months). Similarly, the median survival from the beginning of IL-2 therapy of patients with CRP levels greater than 50 mg/liter was 6 months, compared to 16 months in those with CRP levels below this threshold. None of the 21 patients with serum IL-6 concentrations greater than 300 pg/ml achieved response to any of the three IL-2 regimens. This subgroup has a median survival of 5 months after IL-2 treatment and consisted of 15% of the patients in our series. These results indicate that serum IL-6 and CRP levels are adverse prognosis factors in patients with metastatic renal cell carcinoma. Serum IL-6 level could help in the selection or stratification of the patients in future IL-2 trials.
Article
Culture of human peripheral blood mononuclear cells (PBMC) with IL-2 stimulates synthesis of cytokines and generation of lymphokine-activated killer (LAK) activity. Both IL-4 and IL-10 [cytokine synthesis inhibitory factor (CSIF)] inhibit IL-2-induced synthesis of IFN-gamma and tumor necrosis factor (TNF)-alpha by human PBMC. However, unlike IL-4, IL-10 inhibits neither IL-2-induced proliferation of PBMC and fresh natural killer (NK) cells, nor IL-2-induced LAK activity. Moreover, IL-4 inhibits IL-2-induced IFN-gamma synthesis by purified fresh NK cells, while in contrast the inhibitory effect of IL-10 is mediated by CD14+ cells (monocytes/macrophages). IL-10 inhibits TNF-alpha synthesis by monocytes or monocytes plus NK cells, but not by NK cells alone. These results suggest that IL-4 and IL-10 act on NK cells via distinct pathways, and that IL-2-induced cytokine synthesis and LAK activity are regulated via different mechanisms.
Article
Recent in vitro studies indicate that bone marrow mesenchymal elements, residing in close proximity to hematopoietic cell populations, elaborate a network of cytokines that are, at least partially, responsible for modulating the growth and maturation of the latter compartment. Leukemia inhibitory factor (LIF), a molecule with both positive and negative regulatory activities, has been implicated in murine embryogenesis and hematopoiesis. We demonstrate that cultured normal human bone marrow stromal cells constitutively express LIF message. Further, exposure of these cells to other hematopoietic modulators including interleukin 1 alpha (IL-1 alpha), interleukin 1 beta (IL-1 beta), transforming growth factor-beta (TGF-beta), and tumor necrosis factor-alpha (TNF-alpha) (but not interferon-alpha [IFN alpha]) increases the level of LIF RNA. Interestingly, cultured stromal cells derived from three of four patients with chronic myelogenous leukemia showed enhanced LIF expression. These observations suggest that LIF may participate, either alone or through interaction with other cytokines, in the bone marrow microenvironment-mediated influence on both normal and malignant hematopoietic processes.
Article
In this study, we investigated the role of interleukin-1β (IL-1β) in the malignant evolution of chronic myelogenous leukemia (CML) and the functional activity of IL-1 inhibitors. Bone marrow (BM) and peripheral blood (PB) low-density cells from 38 CML patients were studied in the colony-forming unit-granulocyte, erythrocyte, monocyte, megakaryocyte colony culture assay. Samples from patients with early stage, interferon-α (IFN)-sensitive disease formed hematopoietic colonies in the presence of fetal calf serum (FCS), erythropoietin (Epo), and one of the following: granulocyte-macrophage colony-stimulating factor (10 ng/mL), IL-3 (15 ng/mL), both, or phytohemagglutinin-conditioned medium. The addition of IL-1βaugmented IFN-sensitive CML colony growth in a dose-dependent manner at concentrations of 10 to 100 U/mL. In sharp contrast, addition of the above growth factors did not augment the colony growth-promoting effect of FCS and Epo in samples from IFN-resistant patients; further, adherent cell fractionation or T-lymphocyte depletion attenuated the "autonomous" colony growth. Lysates of 2.5 × 107 low-density cells from each of six IFN-resistant and six IFN-sensitive CML patients and three normal volunteers were tested for intrinsic IL-1 βcontent in an enzyme-linked immunosorbent assay and yielded a mean of 610 pg, 54.6 pg, and 49.4 pg of IL-1β, respectively (P < .045). Interestingly, both soluble IL-1 receptors (slL-1R) and IL-1 receptor antagonist (IL-IRA) at concentrations of 5 to 100 ng/mL (sIL-1R) and 10 to 500 ng/mL (IL-1RA) inhibited CML colony growth in a dose-dependent fashion, with maximal inhibition of 64% and 65%, respectively. A similar effect was noted with the use of anti-IL-1β neutralizing antibodies. These data implicate IL-1β in CML disease progression and suggest that the inhibitory effects of molecules such as slL-1R and IL-IRA could conceivably be the basis of a novel therapeutic strategy against this disorder.
Article
Human IL-10 has been reported previously to inhibit the secretion of IFN-gamma in PBMC. In this study, we have found that human IL-10 inhibits T cell proliferation to either mitogen or anti-CD3 mAb in the presence of accessory cells. Inhibited T cell growth by IL-10 was associated with reduced production of IFN-gamma and IL-2. Studies of T cell subset inhibition by human IL-10 showed that CD4+, CD8+, CD45RA high, and CD45RA low cells are all growth inhibited to a similar degree. Dose response experiments demonstrated that IL-10 inhibits secretion of IFN-gamma more readily than T cell proliferation to mitogen. In addition, IL-2 and IL-4 added exogenously to IL-10 suppressed T cell cultures reversed completely the inhibition of T cell proliferation, but had little or no effect on inhibition of IFN-gamma production. Thus, in addition to its previously reported biologic properties, IL-10 inhibits human T cell proliferation and IL-2 production in response to mitogen. Inhibition of IFN-gamma production by IL-10 appears to be independent of the cytokine effect of IL-2 production.
Article
A previous report concluded that a new cytokine, designated IL-10, is a growth cofactor for thymocytes, spleen, and lymph node cells. In this report, we have focused on the effects of IL-10 on CD8+ spleen T cells. We first observed that IL-10 enhances the growth of CD8+ T cells to IL-2. We then investigated the effect of murine rIL-10 on the induction of murine effector CTL from CTL precursors (CTL-p) using both bulk and filler cell-free limiting-dilution cultures. IL-10 alone could not induce Con A-activated FACS-sorted CD8+ T cells either to proliferate or to generate effector CTL. In combination with IL-2, however, IL-10 augmented the cytolytic activity of effector CTL generated from Con A-activated spleen CD8+ T cells in bulk cultures incubated for 5 days. In limiting-dilution cultures (using solid-phase anti-CD3 mAb as stimulus), IL-10, in combination with IL-2, substantially increased the CTL-p frequency and augmented the cytolytic activity per clone expanded from one CD8+ T cell when compared with cells cultured in IL-2 alone. Kinetic studies showed that IL-10 is required at both early and late culture stages for optimal generation of effector CTL. The potentiating effects of IL-10 on CTL function were neutralized by an anti-IL-10 mAb. These results indicate that IL-10 has direct effects on mature T cells, and suggest that IL-10 also functions as a cytotoxic T cell differentiation factor, which promotes a higher number of IL-2-activated CTL-p to proliferate and differentiate into effector CTL. In contrast, IL-10 did not enhance significantly the lymphokine-activated killer cell activity of IL-2-grown CD8+ cytotoxic T cells.
Article
Philadelphia chromosome1 positive (Ph1) chronic myelogenous leukemia (CML) is characterized by metamorphosis of the chronic phase to blastic crisis. However, cellular events associated with this transition are poorly understood. To examine the possible participation of hematopoietic growth factors in this process, we studied growth factor expression in adherent layers of bone marrows derived from CML Ph1 patients in various stages of the disease. Interleukin-1 β (IL-1β) and IL-6 mRNA were expressed in five of six patients, and granulocyte-macrophage colony-stimulating factor (GM-CSF) in one of six patients with myeloid/undifferentiated blast crisis. In addition, leukemia inhibitory factor (LIF) expression was increased in four of six patients with myeloid/undifferentiated blast crisis phase of the disease. IL-1β was also detected in bone marrow adherent layer conditioned medium from two of these patients. These results were in sharp contrast to the lack of detectable levels of uninduced IL-1β, IL-6, and GM-CSF mRNA, in samples derived from 4 patients in lymphoid blastic crisis, 3 in accelerated, and 11 in chronic phases of the disease, or from normal controls. The possibility of a paracrine loop formation, whereby the adherent layers representing the bone marrow stroma are induced to express hematopoietic growth factors, was supported by our finding IL-1β mRNA expression in the leukemic blast cells in three of four studied patients in blast crisis and IL-1β protein production in seven of eight patients studied. Finally, coculturing CML blast crisis cells onto pre-established adherent layers induced the expression of both IL-1β and IL-6 genes. From this preliminary study, it appears that abnormal expression of growth factors is a common event with CML Ph1 progression. We hypothesize that IL-1β generated by the transformed malignant clone stimulates the marrow stroma to produce various growth factors, and that this process may play a role in disease progression.
Article
Increasingly it seems that many cytokines are pleiotropic, and individual molecules may have critical roles in several different organ systems. LIF exemplifies this phenomenon: it influences embryogenesis, bone and lipid metabolism, and hematopoietic and nervous system function. Many of LIF's effects are reminiscent of those of IL-1, TNF, and TGF-beta. Further, even within a single system, LIF can display totally different effects, i.e. induction of differentiation of one leukemic cell line vs. stimulation of proliferation of another. The corollary to these observations is that there appears to be many parallels in developmental systems. For instance, in the case of neuronal "lineage commitment," the events that relate to migration of neural crest cells along various pathways and their ultimate arrest in different locales demonstrate sufficient analogies to hematopoietic lineage commitment phenomena that, in a provocative review, Anderson coined the term "neuropoiesis". This type of analogy becomes even more intriguing when one realizes that some of the same molecules are regulating neuronal and hematopoietic "lineage" proliferation and differentiation. In this respect, several interleukins in addition to LIF are important in neuronal development, and nerve growth factor turns out to also be a hematopoietic regulatory molecule. Similar parallels are enacted in other organ systems as well. The mediation of identical effects by distinct cytokines bound to unique receptors could conceivably be explained by receptor transmodulation or by overlapping signaling sequences. It is nevertheless also unclear how a single cytokine attached to a single receptor can accomplish varied and opposing effects, although divergent intracellular signaling mechanisms could account for some of these phenomena. Yet another enigma relates to how cells from one system can be properly influenced by a pleiotropic molecule such as LIF without significant "cross-effects" on other potentially responsive systems. Cytokine production that is restricted to certain developmental stages, or very localized distribution and spheres of influence within a microenvironment, could be explanatory. The findings of Rathjan and colleagues, i.e. that LIF exists as both a diffusible molecule and as a molecule incorporated into the extracellular matrix, is of special interest in relation to the above questions. Indeed, the distinctions between the roles of diffusible and immobilized signaling molecules could be crucial to the multiplicity of LIF's actions. Diffusible regulatory factors allow communication between spatially separated cells. Cellular responsiveness to such factors is dictated by the presence of appropriate receptors and postreceptor machinery.(ABSTRACT TRUNCATED AT 400 WORDS)
Article
Interleukin-6 is a pleiotropic cytokine with a wide range of effects, including induction of B-cell and cytotoxic T-cell differentiation, and induction of acute phase reactant production by hepatocytes. Interleukin-6 also can act as an autocrine growth factor in malignancy. Various cell types produce interleukin-6, including T and B cells, monocytes, fibroblasts, and some solid tumor cells. In previous work we detected the production of substantial amounts of interleukin-6 by human ovarian cancer cells, including the ovarian cancer cell lines CAOV-3, OVCAR-3, and SKOV-3, and several primary ovarian tumor cultures. In this study we retrospectively examined 90 separate serum specimens for interleukin-6 in 36 patients with epithelial ovarian cancer. The mean serum interleukin-6 concentration of those ovarian cancer patients with macroscopic disease (n = 57) was 0.26 +/- 0.04 U/ml (mean +/- SEM). Healthy adult donors have interleukin-6 serum levels of 0.12 +/- 0.03 U/ml. Sixteen of 21 ovarian cancer patients with macroscopic disease (76%) had elevated (greater than 0.20 U/ml) levels of serum interleukin-6, with levels approaching 1 U/ml in some patients (p less than 0.01). Of those nine patients with bulky tumor (residual greater than 2 cm), eight (89%) had an elevated interleukin-6 level (mean, 0.31 +/- 0.05), while eight of 12 (66%) with minimal residual disease (less than 2 cm) had elevated levels. Only two of 15 (13%) patients who were in clinical remission and who had microscopic disease had elevated values. Of the 36 patients, 22 were CA 125 negative (less than 35 U/ml), and of these, four had elevated interleukin-6 levels. Of the 14 patients with an elevated CA 125 level, 12 (86%) had elevated interleukin-6 levels. In those 16 patients in whom serial levels of interleukin-6 were measured, rising levels were found over a 3 to 4 month interval in nine (56%); this correlated with tumor progression. Furthermore, the subsequent survival of patients was shown to correlate with the level of interleukin-6, such that patients whose levels were elevated greater than 0.20 U/ml interleukin-6 survived a mean of 12.5 months, compared with 27.2 months for patients with normal levels (p less than 0.001). These data support the concept that interleukin-6 may be a useful tumor marker in some patients with epithelial ovarian cancer, as it correlates with the tumor burden, clinical disease status, and survival.
Article
Study of growth factor RNA levels in the stromal cells derived from the adherent layer of long-term bone marrow culture demonstrated constitutive expression of transforming growth factor beta 1 (TGF-beta 1) and macrophage colony-stimulating factor. These cells did not express granulocyte colony-stimulating factor, granulocyte-monocyte colony-stimulating factor, interleukin (IL) 1 alpha, IL-1 beta, IL-3, and IL-6. However, granulocyte colony-stimulating factor expression could be induced by recombinant human IL-1 beta; while IL-6 could be induced by both IL-1 beta and tumor necrosis factor-alpha. No differences could be detected between adherent layers established from normal and benign phase Ph1 chronic myelogenous leukemia bone marrow. The uninduced expression of TGF-beta 1, a potent hematopoietic cell growth inhibitor, suggests that stromal cells play an inherent role in regulating the proliferation of adjacent bone marrow hematopoietic progenitor cells. However, a defect in stromal TGF-beta 1 production cannot account for the profoundly expanded myeloid compartment in chronic phase chronic myelogenous leukemia. In contrast to the constitutive expression of TGF-beta 1 and macrophage colony-stimulating factor, hematopoietic growth factors are only expressed following a proper stimulation.
Article
In the present report we demonstrate that the IL-6 gene is expressed in anti-Ig-activated and neoplastic B cells. After activation with anti-Ig, splenic B cells rapidly expressed IL-6 mRNA with peak expression occurring at 4 h and declining rapidly thereafter. In an attempt to exclude that the IL-6 mRNA expression was in non-B cells, T cells and monocytes were extensively depleted. In this highly purified B cell population, IL-6 mRNA was retained, whereas the expression of the T cell- and monocyte-restricted CD2 and CD14 genes was nearly undetectable. These results are consistent with the conclusion that activated B cells express IL-6 mRNA. Because we found IL-6 mRNA expression in normal activated B lymphocytes, we examined the expression of IL-6 mRNA in B cell neoplasms. 11 of 25 non-Hodgkins B cell lymphomas and 4 of 4 myelomas and plasma cell leukemias expressed IL-6 mRNA, whereas only 1 of 19 B cell leukemias was positive. To exclude that IL-6 mRNA expression in neoplastic B cells was the result of contaminating non-B cells, T cells and monocytes were extensively depleted from the tumor specimens. In the three IL-6-positive tumor samples depleted of T cells and monocytes, IL-6 mRNA expression was retained in all cases. These observations provide support for the idea that the IL-6 gene is expressed in normal activated and neoplastic B cells.
Article
We report a phase I study of the biological effects, tolerance, and pharmacokinetics of 6- and 24-hour iv infusions of recombinant interferon-gamma (rIFN-gamma) in cancer patients. Twenty-one patients received the 6-hour iv infusion regimen at doses ranging from 0.016 to 0.65 mg/m2/day. Forty-one patients received the 24-hour iv infusion regimen at doses ranging from 0.01 to 0.05 mg/m2/day. Fever and flu-like symptoms were the most common side effects and were seen at all dose levels. The maximum tolerated dose was 0.16 mg/m2 for the 6-hour regimen and 0.01 mg/m2/day for the 24-hour regimen. A dose-dependent granulocytopenia was observed at doses greater than or equal to 0.05 mg/m2/day. A marked increase in beta2 microglobulin occurred by Day 5 of treatment in almost all patients, regardless of the dose level. Consistent serum levels of rIFN-gamma were achieved only at doses of 0.325 mg/m2/day of the 6-hour infusion. The mean serum concentrations at this dose ranged from 18 to 83 units/ml as measured by bioassay (0.64-2.4 ng/ml by enzyme-linked immunoassay). Antibody against rIFN-gamma did not develop in any patient. During the short period of evaluation of this study, one patient with renal cell carcinoma achieved a partial response, and three patients with renal cell (two) and lung carcinoma (one), respectively, achieved minor responses. This study will form the framework for phase II efficacy trials of iv rIFN-gamma.
Article
We report a phase I clinical investigation of 30-minute and four-hour intravenous (IV) infusions of recombinant tumor necrosis factor (rTNF)-alpha. Thirty-nine patients with disseminated cancer received escalating doses of rTNF-alpha for five consecutive days every 2 weeks for a total duration of 8 weeks. Dose escalations followed a modified Fibonacci scale with a minimum of four patients entered at each dose level: 5, 10, 25, 50, 75, 100, 150, 200, and 250 micrograms/m2/d. Toxicities consisted mainly of constitutional symptoms including fever, chills, headache, and fatigue, increasing in severity with dose escalation. No significant differences in dose-limiting toxicities were seen between the two rates of IV infusion. The maximum tolerated dose (MTD) was 200 micrograms/m2 with dose limiting toxicity being constitutional symptoms and hypotension. Hematologic changes included median decrease in both granulocyte and platelet counts of 38% and 41%, respectively (range, 16% to 85%), although clinically significant granulocytopenia and thrombocytopenia were not observed. Hematological parameters returned to baseline within 72 hours after rTNF-alpha was stopped. rTNF-alpha induced changes in lipid metabolism were manifested by median fasting triglyceride elevations above baseline (median, 103 micrograms/dL) of 157% (range, 16% to 389%) after five days of therapy with doses greater than 75 micrograms/m2, associated with a median increase in very-low-density lipoprotein (VLDL) of 80%. Serum rTNF peak levels exceeding 10 ng/mL were observed 30 minutes following rTNF-alpha infusions at MTD dose. Twelve of 34 patients had no change in their evaluable disease for a median duration of 18 weeks (range, 8 to 30 weeks), and 22 patients showed progressive disease. This study forms the framework for phase II trials of IV administered rTNF-alpha.
Article
Interleukin-6 (IL-6) is evaluated as a candidate mediator of morbidity in patients with metastatic adenocarcinoma of the prostate. IL-6 concentration is measured by enzyme-linked immunoadsorbent assay (ELISA) in the ejaculate plasma of healthy men, in primary culture of prostate epithelial cells, in human prostate cancer cell line cultures and SCID mouse xenografts, and in the plasma of 73 men with metastatic adenocarcinoma of the prostate. High levels of IL-6 secretion are found in the normal human ejaculate, in prostate epithelial primary culture, and in three of four anaplastic, androgen-independent human prostate cancer cell lines tested. In contrast, the hormone-responsive and PSA-secreting cell lines and the hormone-independent line PPC-1 do not secrete detectable levels of IL-6 by ELISA: The acquisition of a p53 mutation in LNCaP-GW and PPC-1 is not sufficient to confer the phenotype of high IL-6 secretion. Seventy-three men with well-characterized, advanced, hormone refractory prostate cancer prior to suramin therapy are tested for incidence of abnormal circulating levels of IL-6. Plasma IL-6 levels have a bimodal distribution, with the upper quartile of patients having abnormal levels from 9 to 61 pg/mL. A direct comparison of the high and low serum IL-6 groups show that elevated IL-6 levels are strongly correlated with objective measures of morbidity: decreased hematocrit, hemoglobin, and serum cholesterol, and increased white blood cell count and serum lactate dehydrogenase levels all in the absence of clinical infection. These data show that IL-6 is a prostate exocrine gene product, a candidate mediator of prostate cancer morbidity, and a candidate marker of disease activity for prospective clinical testing.
Article
Interleukin-6 (IL-6) is a potent immunomodulatory cytokine that may have pathogenetic and prognostic significance in a number of disorders. The objective of this study was to examine the correlation between serum IL-6 levels and phenotypic characteristics, as well as outcome of patients with diffuse large-cell lymphoma (DLCL). Using an enzyme-linked immunosorbent assay (ELISA; lower limit of sensitivity, 0.35 pg/mL), we measured IL-6 levels in frozen sera from 33 healthy controls and 58 untreated patients with DLCL who were enrolled onto a single combination chemotherapy protocol. Serum IL-6 levels were correlated with clinical and laboratory features at diagnosis and with failure-free and overall survival. Serum IL-6 levels in the lymphoma patients (median, 4.37 pg/mL; range, < 0.35 to 110 pg/mL) were significantly higher than in the control group (median, < 0.35 pg/mL; range, < 0.35 to 1.87 pg/mL) (P < .0001). Serum IL-6 levels were higher in patients with B symptoms (P = .012), an elevated beta 2-microglobulin level (> or = 3.0 mg/L) (P = .017), and a poor performance status (P = .02). Direct linear correlations with the erythrocyte sedimentation rate (ESR), platelet count, and total WBC count, and an inverse linear correlation with the serum albumin level, were observed (all P < .02). Patients with elevated serum IL-6 levels had inferior failure-free (P = .042) and overall survival (P = .05) compared with those with normal serum IL-6 levels. In patients with DLCL, elevated serum levels of IL-6 at diagnosis are frequent, strongly associated with many adverse disease features, and predictive of a poor failure-free and overall survival.
Article
To investigate the occurrence of non-Hodgkin's lymphoma (NHL) in human immunodeficiency virus (HIV)-infected patients receiving long-term antiretroviral therapy and factors associated with the development of these lymphomas. The charts of 55 patients with advanced HIV infection receiving zidovudine (formerly known as azidothymidine [AZT])-based therapy and 61 patients receiving dideoxyinosine (ddI) were examined for the occurrence of NHL. Stored samples from the AZT-based treatment cohort were examined retrospectively for parameters predictive of the subsequent development of lymphoma. Eight of 55 patients receiving AZT-based therapy developed NHL, yielding an estimated probability of 12% (95% confidence interval [CI], 4.7% to 27.1%) after 24 months, and 29.2% (95% CI, 15.2% to 48.7%) after 36 months. Four of 61 patients receiving ddI developed NHL, yielding a 6.2% (95% CI, 2.1% to 17%) estimated probability after 24 months, and 9.5% (95% CI, 3.6% to 22.8%) after 36 months. The difference between these cohorts was not significant (two-tailed P [P2] = .13). Patients with less than 50 CD4 cells/microL developed NHL at a significantly higher rate (P2 = .0085). This was particularly true for patients who presented with primary CNS lymphoma (PCNSL). For patients receiving AZT-based therapy, pretreatment serum interleukin-6 (IL-6) levels were somewhat higher in those who subsequently developed NHL than in those who did not (P2 = .048). HIV-infected patients with profound immunodeficiency, especially those with less than 50 CD4 cells/microL, are at substantial risk of developing NHL and particularly PCNSL. Additional studies are needed to define the role of other factors such as IL-6 in the pathogenesis of these opportunistic tumors.
Article
In the current study, we used a monoclonal antibody-based enzyme-linked immunosorbent assay and bioassay to assess leukemia inhibitory factor (LIF) protein levels, activity, and function in supernatants of 59 adherent layers derived from acute and chronic myelogenous leukemia, myelodysplastic syndrome, and hairy cell leukemia patients and from normal controls. We demonstrate that biologically active LIF protein is constitutively produced and secreted by cultured bone marrow stromal cells from all of the studied subjects. Furthermore, various cytokines can alter endogenous LIF protein levels. Twenty-four h of exposure to recombinant human (rh) interleukin (IL) 4 (100 units/ml) significantly decreased LIF protein levels in adherent layer conditioned media [median base line level, 2.6 ng/ml; range, 1.6-8.0 ng/ml; median post rhIL-4 exposure levels, 1.9 ng/ml; range, 0.9-5.8 ng/ml (n = 7; P = 0.022)]. In contrast, rhIL-1 beta and rh tumor necrosis factor alpha consistently increased LIF protein levels. In the samples exposed to 50 units/ml rhIL-1 beta, median base line LIF level was 2.6 ng/ml; median post-LIF level was 9.0 ng/ml (n = 8; P = 0.014). In the two samples exposed to rh tumor necrosis factor alpha (200 units/ml), LIF levels increased from baseline levels of 2.6 and 2.7 ng/ml to postexposure levels of 7.7 and 12.2 ng/ml, respectively. Finally, the presence of LIF may be relevant to both normal and malignant hematopoietic processes as evidenced by: (a) LIF protein levels in adherent layer conditioned media were significantly elevated in samples from patients with a spectrum of hematological neoplasms [acute myelogenous leukemia: median level, 3.0 ng/ml (range, 1.6-11.0 ng/ml); myelodysplastic syndrome: median level, 4.5 ng/ml (range 1.4-15.5 ng/ml); hairy cell leukemia; median level, 3.5 ng/ml (range 2.2-10.3 ng/ml); chronic myelogenous leukemia-chronic phase: median level, 4.35 ng/ml (range 0.3-19.0 ng/ml); and chronic myelogenous leukemia-blast crisis: median level, 6.25 ng/ml (range 0.7-20.3 ng/ml)] as compared to samples from normal individuals (median level, 2.0 ng/ml; range, 0.7-4.6 ng/ml; P < 0.05); and (b) in normal controls, in vitro abrogation of endogenous LIF bioactivity by neutralizing antibody decreased the number of committed granulocyte-macrophage hemopoietic progenitors.
Article
Several cytokines including gamma-interferon, tumor necrosis factor alpha, interleukin 1 beta (IL-1 beta), and interleukin 6 (IL-6) are pyrogenic and can inhibit lipogenic processes. Because patients with lymphoma often suffer from fever, weight loss, and night sweats (B symptoms), the etiology of which is unknown, the authors investigated serum levels of these cytokines in normal volunteers and in patients with Hodgkin's and non-Hodgkin's lymphoma. Sixty serum samples from patients with Hodgkin's disease (28 patients) or non-Hodgkin's lymphoma (32 patients), as well as 20 samples from normal volunteers, were collected. The majority of patients had advanced (Stage III or IV) or relapsed disease. The assay for gamma-interferon was a specific and sensitive radioimmunoassay (lower limit of detection = 0.1 unit/ml); the assays for tumor necrosis factor alpha, IL-1 beta, and IL-6 were enzyme-linked immunoassays with lower limits of sensitivity of 10 pg/ml, 20 pg/ml, and 22 pg/ml, respectively. There were no statistically significant differences in gamma-interferon, tumor necrosis factor alpha, or IL-1 beta levels between lymphoma patients and normal subjects. In contrast, 20 of 57 patients (35%) with lymphoma as compared with 0 of 19 normal volunteers (0%) had detectable serum IL-6 levels (P < 0.005, chi 2 test). Interestingly, 17 of 29 lymphoma patients with B symptoms (59%) as opposed to 3 of 28 lymphoma patients without B symptoms (11%) had detectable serum IL-6 levels (P < 0.001, chi 2 test); the median IL-6 level was 28.9 pg/ml (B symptoms present) versus undetectable (no B symptoms) (P < 0.005, Mann-Whitney U test). Analyzing Hodgkin's and non-Hodgkin's lymphoma groups separately revealed similar results. IL-6 levels showed no significant correlation with time from diagnosis, beta 2-microglobulin, or lactate dehydrogenase levels. However, analysis by the method of Kaplan and Meir demonstrated that the median survival of Hodgkin's disease patients with detectable IL-6 levels (> or = 22 pg/ml) was 10 mo, whereas the median survival has not been reached at a median follow-up time of 37.5 mo in those patients with lower values (Wilcoxon P value = 0.0012). There were too few patients in each subset of non-Hodgkin's lymphoma to determine the correlation between IL-6 and survival but, considered as a single group, a statistically significant correlation was not found.(ABSTRACT TRUNCATED AT 400 WORDS)
Article
Interleukin-6 (IL-6) is a potent immunomodulatory cytokine that may have pathogenetic significance in several malignancies. In addition, high IL-6 levels have been associated with a poor prognosis in multiple myeloma, nonHodgkin's lymphoma, ovarian cancer, and renal cancer, as well in advanced Hodgkin's lymphoma. In this study, we analyzed IL-6 levels in newly diagnosed Hodgkin's disease and determined clinical correlates of elevated levels. Using a sensitive enzyme-linked immunosorbent assay (lower limit of sensitivity = 0.35 pg/mL) we measured IL-6 levels in sera from 33 healthy controls and 65 untreated patients with Hodgkin's disease. Interleukin-6 levels in the Hodgkin's patients (median 2.7 pg/mL; range < 0.35 to 38.4 pg/mL) were significantly higher than in the controls (median < 0.35 pg/mL; range < 0.35 to 1.87 pg/mL; P < 0.0001). Interleukin-6 levels were also higher in males (P = 0.03) and in patients with bulky disease (P = 0.026) or advanced Ann Arbor stage (P = 0.017). In addition, serum levels of IL-6 also showed direct linear correlations with the erythrocyte sedimentation rate (r = 0.64, P = 0.0007), platelet count (r = 0.53, P < 0.0001), leukocyte count (r = 0.36, P = 0.003), and beta (2)-microglobulin level (r = 0.4, P = 0.0012); and an inverse linear correlation with serum albumin level (r = -0.43, P = 0.0003). In the 10 patients tested who had elevated serum IL-6 levels pretherapy and who achieved complete remission, serum IL-6 values decreased at the time of remission to the range found in healthy controls. Our observations suggest that, in patients with Hodgkin's disease, serum levels of IL-6 are frequently elevated at diagnosis, normalize during remission, and are associated with specific disease characteristics including several adverse prognostic features.
Article
Interleukin-6 has important lymphoid bioregulatory effects, and serum levels of interleukin-6 are often elevated in patients with lymphoma. To determine the relation between serum levels of interleukin-6 before treatment and outcome in patients with diffuse large-cell lymphoma. Retrospective cohort analysis with multivariate analysis. Tertiary referral center. 118 untreated patients with diffuse large-cell lymphoma who were enrolled in frontline chemotherapy protocols and 45 healthy controls. Serum levels of interleukin-6 were measured by using a sensitive enzyme-linked immunosorbent assay. Levels below the upper limit of the range for controls were considered normal. Outcomes were complete response, failure-free survival, and overall survival. Serum levels of interleukin-6 were higher in patients with lymphoma (median, 4.6 pg/ml [range, undetectable to 224 pg/mL]) than in controls (median, undetectable [range, undetectable to 4.3 pg/mL]) (P = 0.009). The complete response rate was 95% for persons with normal interleukin-6 levels and 55% for persons with high interleukin-6 levels (P = 0.001). Patients with high interleukin-6 levels had inferior failure-free and overall survival rates (P < 0.001 for both comparisons). The actuarial 4-year failure-free and overall survival rates were 72% and 85%, respectively, for persons with normal interleukin-6 levels and 37% and 46%, respectively, for persons with high interleukin-6 levels. In multivariate analysis, interleukin-6 was selected as the most significant predictor of complete response and failure-free survival. Failure-free and overall survival of patients stratified according to International Prognostic Index score could be further stratified by interleukin-6 level (P < or = 0.03 for all comparisons). In patients with diffuse large-cell lymphoma, serum interleukin-6 levels are an independent prognostic factor for complete response and failure-free survival.
Article
Interleukin-10 (IL-10) is a pleiotropic cytokine produced by type 2 helper cells (Th2), as well as by monocytes and macrophages, and normal and neoplastic B lymphocytes. It is highly homologous to an open reading frame of Epstein-Barr virus (EBV) called BCRF1, and EBV infection of B-cells up-regulates IL-10. IL-10 production has strong immunosuppressive effects via inhibition of Th1 type cytokines, including interferon gamma and interleukin-2. On B-cells, IL-10 has a potent stimulating effect, inducing proliferation and differentiation. Interestingly, in cell lines derived from B-cell lymphomas, IL-10 production has been found to be up-regulated, and it serves as an autocrine growth factor. In patients with non-Hodgkin's lymphoma (NHL), serum IL-10 levels are significantly increased when compared to normal individuals and NHL patients in remission. The prognostic significance of these increased levels vary according to the assay used. Both human IL-10 and viral IL-10 are increased, and when specific assays for human IL-10 are used, there seems to be no prognostic significance, whereas when the assay cross-reacts with viral IL-10, high levels correlate with poor prognosis. These results suggest that viral IL-10 might have some pathogenic role in NHL.
Article
The purpose of this review is to provide an overview of the literature linking Ras signaling pathways and leukemia and to discuss the biologic and potential therapeutic implications of these observations. A search of MEDLINE from 1966 to October 1998 was performed. A wealth of data has been published on the role of Ras pathways in cancer. To be biologically active, Ras must move from the cytoplasm to the plasma membrane. Importantly, a posttranslational modification--addition of a farnesyl group to the Ras C-terminal cysteine--is a requisite for membrane localization of Ras. Farnesylation of Ras is catalyzed by an enzyme that is designated farnesyltransferase. Recently, several compounds have been developed that can inhibit farnesylation. Preclinical studies indicate that these molecules can suppress transformation and tumor growth in vitro and in animal models, with little toxicity to normal cells. An increasing body of data suggests that disruption of Ras signaling pathways, either directly through mutations or indirectly through other genetic aberrations, is important in the pathogenesis of a wide variety of cancers. Molecules such as farnesyl transferase inhibitors that interfere with the function of Ras may be exploitable in leukemia (as well as in solid tumors) as novel antitumor agents.
Article
Interleukin (IL)-1beta is constitutively expressed in many leukemias and operates as an autocrine growth factor. To study the cellular basis for this aberrant production, we analyzed two cell lines, B1 (acute lymphoblastic leukemia) and W1 (juvenile chronic myelogenous leukemia), which express high levels of IL-1beta and have mutations in the K-RAS and N-RAS genes, respectively. Electromobility shift assays demonstrated transcription factor binding at multiple IL-1beta promoter elements [nuclear factor (NF)-IL6/CREB, NFB1, NFkappaB, and NF-IL6], consistent with the activation of an upstream signaling pathway. To determine whether activated Ras was involved, two structurally distinct classes of farnesyltransferase (FTase) inhibitors (the monoterpenes and a peptidomimetic) and an adenoviral vector expressing antisense targeted to K-RAS were used to specifically interfere with Ras function and/or expression. Treatment with the FTase inhibitors resulted in a concentration-dependent decrease in both NF-IL6/CREB binding to the IL-1beta promoter and IL-1beta protein levels, without a significant change in total cellular protein levels. Furthermore, exposure of the B1 cells to antisense against K-RAS resulted in an approximately 50% reduction in both p21Ras and IL-1beta protein levels. Growth suppression was observed after FTase inhibitor or antisense exposure, an effect that was partially reversible by the addition of recombinant IL-1beta to the cultures. Our observations suggest that mutated RAS genes may mediate autocrine IL-1beta production in some leukemias by stimulating signal transduction pathways that activate the IL-1beta promoter.
Interleukin 6 expression in normal and neo-plastic B cells Autocrine interleukin-1beta production in leukemia: evidence for the involvement of mutated RAS
  • Freeman Gj Freedman As
  • Sn
  • J Segil Jm
Freeman GJ, Freedman AS, Rabinowe SN, Segil JM, Horow-itz J, Rosen K, et al. Interleukin 6 expression in normal and neo-plastic B cells. J Clin Invest 1989 ; 83 : 1512-8. 27 Beaupre DM, Talpaz M, Marini III, Frank C, Cristiano RJ, Roth J, Estrov Z, et al. Autocrine interleukin-1beta production in leukemia: evidence for the involvement of mutated RAS. Cancer Res 1999 ; 59 : 2871-980.
Differential effects of interleukin-4 and -10 and interleukin-2 induced interferon-γ synthesis and lymphokine-activated killer activity Il-10 inhibits cytokine synthesis by human monocytes: an auto-regulatory role of Il-10 produced by monocytes
  • Hsu
  • Moore Kw R Dh
  • Bennett B C Figdar
Hsu DH, Moore KW, Spits H. Differential effects of interleukin-4 and -10 and interleukin-2 induced interferon-γ synthesis and lymphokine-activated killer activity. Int Immunol 1992 ; 4 : 563-9. 37 De Waal-Malefyt R, Abrams J, Bennett B, Figdar C, de Vries J. Il-10 inhibits cytokine synthesis by human monocytes: an auto-regulatory role of Il-10 produced by monocytes. J Exp Med 1991 ; 174 : 1209-20.
Interleukin-10: a novel cytotoxic T cell differentiation factor RAS and leukemia: from basic mecha-nisms to gene-directed therapy Cytokine deregulation in cancer
  • Chen W-F Zlotnik
Chen W-F, Zlotnik A. Interleukin-10: a novel cytotoxic T cell differentiation factor. J Immunol 1991 ; 147 : 5288-534. 41 Beaupre DM. Kurzrock R. RAS and leukemia: from basic mecha-nisms to gene-directed therapy. J Clin Oncol 1999 ; 17 : 1071-9. Cytokine deregulation in cancer
Interleukin-6: an overview
  • Van
  • Sj
Van SJ. Interleukin-6: an overview. Annu Rev Immunol 1990 ; 8 : 253-7.