TY - JOUR AB - Immunotherapy with immune checkpoint inhibitors (ICI) has substantially improved the treatment of advanced renal cell carcinoma (aRCC). Since treatment effects vary among patients, identifying biomarkers to predict response is crucial for optimizing clinical management. Expression of programmed death ligand 1 (PD-L1) is associated with better response to ICI treatment, but is not a valid biomarker. To advance patient stratification for aRCC, we analyzed molecular and clinical data from the JAVELIN Renal 101 (n=726) and IMmotion151 (n=823) trials, both of which compared a combination of PD-L1 inhibitors and anti-angiogenic treatments with sunitinib monotherapy. In JAVELIN Renal 101, we found correlations between genes and progression-free survival (PFS) in PD-L1-positive tumors treated with sunitinib. These associations were independent of variations in the proportion of PD-L1-positive immune cells present in the tumors. Clustering of PD-L1-positive tumors based on PFS-related genes revealed an IMmune CHeckpoint Inhibitor-responsive Phenotype (IMCHIP). In PD-L1-positive tumors with IMCHIP, combined therapy with ICI led to significantly longer PFS relative to sunitinib (13.3 months vs. 4.4 months median PFS), while in PD-L1-positive tumors without IMCHIP, PFS was comparable for both treatments (11.1 months). By de-correlating gene expression measurements from PD-L1 expression, the clustering could be extended to PD-L1-negative tumors resulting in two groups (aRCC1/2) that were independent of PD-L1 status. Stratification based on aRCC1/2 and PD-L1 status allowed treatment effects to be categorized into four groups. A significant difference in PFS between treatments arms was only observed in patients with tumors of type aRCC2/PD-L1-positive, which corresponded to the IMCHIP profile and covered 32.4% of JAVELIN Renal 101. These findings could be validated in the IMmotion151 trial, where IMCHIP represented 20% of the cohort. Compared to other biomarkers currently being investigated, the combination of aRCC1/2 with PD-L1 status exhibited the greatest potential as a predictive biomarker for patient selection, which was confirmed in both cohorts. The prognostic potential of the aRCC1/2 subdivision was evaluated in 476 clear cell RCC from TCGA. Here, aRCC2 was associated with significantly worse overall survival and cancer-specific survival in both univariate and multivariable analyses, accounting for age, sex, and clinicopathological parameters (TNM). In summary, categorization into aRCC1/2 proved to be a new risk stratification of aRCC which, in combination with PD-L1 status, enabled the retrospective identification of responders who benefited from combined therapy with ICI relative to sunitinib monotherapy. The analysis of IMmotion151 trial data utilized data generated by Genentech/Genentech Research and Early Development. AU - Büttner, F.A.* AU - Grünwald, V.* AU - Nößner, E. AU - Tsaur, I.* AU - Bedke, J.* AU - Schwab, M.* AU - Schaeffeler, E.* C1 - 74720 C2 - 57665 SP - 7155 - 7155 TI - Abstract 7155: A PD-L1-independent phenotype uncovers responders to combined therapy with immune checkpoint inhibitors in kidney cancer. JO - Cancer Res. VL - 85 IS - 8_Supplement_1 PY - 2025 SN - 0008-5472 ER - TY - JOUR AB - Lung squamous carcinoma (LUSC) is a lethal cancer with still unknown etiology and limited detection and therapeutic options. One hypothesis links its origin to the transformation of basal epithelium. Notably immunotherapy has changed cancer treatment. We integrated transcriptomics, proteomics, genomics, epigenetics, and single-cell RNA sequencing (scRNAseq) to explore basal cell-dependent signatures as therapy response predictors in LUSC. To identify whether subtypes of LUSC respond differently to immunotherapy, we first used the TCGA-LUSC cohort and performed unsupervised clustering using a set of hub (basal and immune) genes obtained through extensive analysis. We obtained 2 distinct clusters, the "Basalimmune-high" cluster had higher immune scores, immune cell infiltration and a better predictive immunotherapy response. The "Basalimmune-low" showed higher proliferative basal cell score and KRT5/6 and TP63 expression. Interestingly, there was an inverse correlation between KRT5/6 methylation and its RNA expression in "Basalimmune-high", with no difference in TP63 methylation between the groups, when analyzing epigenetic data. The "Basalimmune-high" cluster was associated with poor overall survival, when treated with standard chemotherapy. A prognostic signature of 15 genes was identified. These findings were validated using transcriptomic and proteomic data from other four LUSC cohorts. Using the top upregulated genes between the 2 clusters, we selected and measure their protein serum levels, to identify potential cluster predictors. Changes in serum levels of CXCL5 and CXCL11 in a real-life treatment naïve LUSC cohort (Stage 1-4,n=100) confirmed the clustering and poorer prognosis of the "Basalimmune-high"group (p<0.03). Using MutSig2CV, we found unique mutational landscapes in the two clusters: NOTCH1 in "Basalimmune-high" and ARID1A in "Basalimmune-low". Furthermore, copy number variants showed shared and distinct amplifications (Basalimmune-high: KAT6A; Basalimmune-low: EGFR) and deletions (Basalimmune-high: ROBO1; -low: STK11). GSEA of transcriptomic and proteomic data revealed some biological mechanisms underlying the immune responses of "Basalimmune-high" patients, such as upregulation of signaling pathways for complement, IL2, STAT5, IFN-α/γ, and other inflammatory responses. Given these associations, we performed an intercellular communication analysis on the scRNAseq data to identify novel communications between basal and immune cells that might influence immunotherapy. The pathways identified in silico were validated in vitro in co-cultures of CD8+ T cells, NK cells, and monocytes with primary human bronchial epithelial cells and identified as predictive of basal-immune cell communication. Altogether, this is the first demonstration that a multi-omic integrative approach has successfully identified distinct clusters on LUSC patients and may predict personalized immunotherapy options based on their genetic and molecular profiles. AU - Hains, A.E.* AU - Marques, J.G.* AU - Chetal, K.* AU - Nakatani, T. AU - Ettinger, A. AU - Biagi, C.A.O.d.* AU - Gonzalez‐Sandoval, A.* AU - Pillai, R.* AU - Gatto, A.* AU - Torres-Padilla, M.E. AU - Sadreyev, R.I.* AU - Filbin, M.G.* AU - Rechem, C.V.* C1 - 74716 C2 - 57669 SP - SY23 - 01 TI - Abstract SY23-01: Diffuse midline gliomas: From cell cycle to therapeutic opportunities. JO - Cancer Res. VL - 85 IS - 8_Supplement_2 PY - 2025 SN - 0008-5472 ER - TY - JOUR AU - Papargyriou, A. AU - Reichert, M.* C1 - 74500 C2 - 57497 CY - 615 Chestnut St, 17th Floor, Philadelphia, Pa 19106-4404 Usa SP - 1571-1573 TI - The biology in the pattern: Metastatic organotropism and clinical outcome depend on DNA damage response and immune interactions in pancreatic cancer. JO - Cancer Res. VL - 85 IS - 9 PB - Amer Assoc Cancer Research PY - 2025 SN - 0008-5472 ER - TY - JOUR AB - Cigarette smoke, containing both nicotine and carcinogens, causes lung cancer. However, not all smokers develop lung cancer, highlighting the importance of the interaction between host susceptibility and environmental exposure in tumorigenesis. Here, we aimed to delineate the interaction between metabolizing ability of tobacco carcinogens and smoking intensity in mediating genetic susceptibility to smoking-related lung tumorigenesis. Single-variant and gene-based associations of 43 tobacco carcinogen-metabolizing genes with lung cancer were analyzed using summary statistics and individual-level genetic data, followed by causal inference of Mendelian randomization, mediation analysis, and structural equation modeling. Cigarette smoke-exposed cell models were used to detect gene expression patterns in relation to specific alleles. Data from the International Lung Cancer Consortium (29,266 cases and 56,450 controls) and UK Biobank (2,155 cases and 376,329 controls) indicated that the genetic variant rs56113850 C>T located in intron 4 of CYP2A6 was significantly associated with decreased lung cancer risk among smokers (OR = 0.88, 95% confidence interval = 0.85-0.91, P = 2.18 × 10-16), which might interact (Pinteraction = 0.028) with and partially be mediated (ORindirect = 0.987) by smoking status. Smoking intensity accounted for 82.3% of the effect of CYP2A6 activity on lung cancer risk but entirely mediated the genetic effect of rs56113850. Mechanistically, the rs56113850 T allele rescued the downregulation of CYP2A6 caused by cigarette smoke exposure, potentially through preferential recruitment of transcription factor helicase-like transcription factor. Together, this study provides additional insights into the interplay between host susceptibility and carcinogen exposure in smoking-related lung tumorigenesis. SIGNIFICANCE: The causal pathway connecting CYP2A6 genetic variability and activity, cigarette consumption, and lung cancer susceptibility in smokers highlights the need for behavior modification interventions based on host susceptibility for cancer prevention. AU - Du, M.* AU - Xin, J.* AU - Zheng, R.* AU - Yuan, Q.* AU - Wang, Z.* AU - Liu, H.* AU - Cai, G.* AU - Albanes, D.* AU - Lam, S.* AU - Tardón, A.* AU - Chen, C.* AU - Bojesen, S.E.* AU - Landi, M.T.* AU - Johansson, M.* AU - Risch, A.* AU - Bickeböller, H.* AU - Wichmann, H.-E. AU - Rennert, G.* AU - Arnold, S.* AU - Brennan, P.* AU - Field, J.K.* AU - Shete, S.S.* AU - Le Marchand, L.* AU - Liu, G.* AU - Andrew, A.S.* AU - Kiemeney, L.A.* AU - Zienolddiny, S.* AU - Grankvist, K.* AU - Caporaso, N.E.* AU - Cox, A.* AU - Hong, Y.C.* AU - Yuan, J.M.* AU - Schabath, M.B.* AU - Aldrich, M.C.* AU - Wang, M.* AU - Shen, H.* AU - Chen, F.* AU - Zhang, Z.* AU - Hung, R.J.* AU - Amos, C.I.* AU - Wei, Q.* AU - Lazarus, P.* AU - Christiani, D.C.* C1 - 70065 C2 - 55240 CY - 615 Chestnut St, 17th Floor, Philadelphia, Pa 19106-4404 Usa SP - 616-625 TI - CYP2A6 activity and cigarette consumption interact in smoking-related lung cancer susceptibility. JO - Cancer Res. VL - 84 IS - 4 PB - Amer Assoc Cancer Research PY - 2024 SN - 0008-5472 ER - TY - JOUR AB - Hepatitis B virus (HBV) infections promote liver cancer initiation by inducing inflammation and cellular stress. Despite the primarily indirect effect on oncogenesis, HBV is associated with a recurrent genomic phenotype in HCC, suggesting that it impacts the biology of established HCC. Characterization of the interaction of HBV with host proteins and the mechanistic contributions of HBV to HCC initiation and maintenance could provide insights into HCC biology and uncover therapeutic vulnerabilities. Here, we used affinity purification mass spectrometry to comprehensively map a network of 145 physical interactions between HBV and human proteins in hepatocellular carcinoma (HCC). A subset of the host factors targeted by HBV proteins were preferentially mutated in non-HBV-associated HCC, suggesting that their interaction with HBV influences HCC biology. HBV interacted with proteins involved in mRNA splicing, mitogenic signaling, and DNA repair, with the latter set interacting with the HBV oncoprotein X (HBx). HBx remodeled the PP2A phosphatase complex by excluding striatin regulatory subunits from the PP2A holoenzyme, and the HBx effects on PP2A caused Hippo kinase activation. In parallel, HBx activated mTOR complex 2 (mTORC2), which can prevent YAP degradation. mTORC2-mediated upregulation of YAP was observed in human HCC specimens and mouse HCC models and could be targeted with mTOR kinase inhibitors. Thus, HBV interaction with host proteins rewires HCC signaling rather than directly activating mitogenic pathways, provide an alternative paradigm for the cellular effects of a tumor promoting virus. AU - Turnham, R.E.* AU - Pitea, A.* AU - Jang, G.M.* AU - Xu, Z.* AU - Lim, H.C.* AU - Choi, A.L.* AU - Von Dollen, J.* AU - Levin, R.S.* AU - Webber, J.T.* AU - McCarthy, E.* AU - Hu, J.* AU - Li, X.* AU - Che, L.* AU - Singh, A.* AU - Yoon, A.J.* AU - Chan, G.* AU - Kelley, R.K.* AU - Swaney, D.L.* AU - Zhang, W.* AU - Bandyopadhyay, S.* AU - Theis, F.J. AU - Eckhardt, M.* AU - Chen, X.* AU - Shokat, K.M.* AU - Ideker, T.* AU - Krogan, N.J.* AU - Gordan, J.D.* C1 - 72692 C2 - 56696 TI - HBV remodels PP2A complexes to rewire kinase signaling in hepatocellular carcinoma. JO - Cancer Res. PY - 2024 SN - 0008-5472 ER - TY - JOUR AB - Asymptomatic anthracosis is the accumulation of black carbon particles in adult human lungs. It is a common occurrence, but the pathophysiological significance of anthracosis is debatable. Using in situ high mass resolution matrix-assisted laser desorption/ionization (MALDI) fourier-transform ion cyclotron resonance (FT-ICR) mass spectrometry imaging analysis, we discovered noxious carbon-bound exogenous compounds, such as polycyclic aromatic hydrocarbons (PAHs), tobacco-specific nitrosamines, or aromatic amines, in a series of 330 lung cancer patients in highly variable and unique patterns. The characteristic nature of carbon-bound exogenous compound had a strong association with patient outcome, tumor progression, the tumor immune microenvironment, PD-L1 expression, and DNA damage. Spatial correlation network analyses revealed substantial differences in the metabolome of tumor cells compared to tumor stroma depending on carbon-bound exogenous compounds. Overall, the bioactive pool of exogenous compounds is associated with several changes in lung cancer pathophysiology and correlates with patient outcome. Given the high prevalence of anthracosis in the lungs of adult humans, future work should investigate the role of carbon-bound exogenous compounds in lung carcinogenesis and lung cancer therapy. AU - Kunzke, T. AU - Prade, V.M. AU - Buck, A. AU - Sun, N. AU - Feuchtinger, A. AU - Matzka, M. AU - Fernandez, I.E.* AU - Wuyts, W.A.* AU - Ackermann, M.* AU - Jonigk, D.* AU - Aichler, M. AU - Schmid, R.A.* AU - Eickelberg, O.* AU - Berezowska, S.* AU - Walch, A.K. C1 - 63461 C2 - 51317 CY - 615 Chestnut St, 17th Floor, Philadelphia, Pa 19106-4404 Usa SP - 5862-5875 TI - Patterns of carbon-bound exogenous compounds in lung cancer patients and association with disease pathophysiology. JO - Cancer Res. VL - 81 IS - 23 PB - Amer Assoc Cancer Research PY - 2021 SN - 0008-5472 ER - TY - JOUR AB - Tumor-derived protein tissue inhibitor of metalloproteinases-1 (TIMP1) correlates with poor prognosis in many cancers, including highly lethal pancreatic ductal adenocarcinoma (PDAC). The noncanonical signaling activity of TIMP1 is emerging as one basis for its contribution to cancer progression. However, TIMP1-triggered progression-related biological processes are largely unknown. Formation of neutrophil extracellular traps (NET) in the tumor microenvironment is known to drive progression of PDAC, but factors or molecular mechanisms initiating NET formation in PDAC remain elusive. In this study, gene-set enrichment analysis of a human PDAC proteome dataset revealed that TIMP1 protein expression most prominently correlates with neutrophil activation in patientderived tumor tissues. TIMP1 directly triggered formation of NETs in primary human neutrophils, which was dependent on the interaction of TIMP1 with its receptor CD63 and subsequent ERK signaling. In genetically engineered PDAC-bearing mice, TIMP1 significantly contributed to NET formation in tumors, and abrogation of TIMP1 or NETs prolonged survival. In patient-derived PDAC tumors, NETs predominantly colocalized with areas of elevated TIMP1 expression. Furthermore, TIMP1 plasma levels correlated with DNA-bound myeloperoxidase, a NET marker, in the blood of patients with PDAC. A combination of plasma levels of TIMP1 and NETs with the clinically established marker CA19-9 allowed improved identification of prognostically distinct PDAC patient subgroups. These observations may have a broader impact, because elevated systemic levels of TIMP1 are associated with the progression of a wide range of neutrophil-involved inflammatory diseases. AU - Schoeps, B.* AU - Eckfeld, C.* AU - Prokopchuk, O.* AU - Böttcher, J.* AU - Häußler, D.* AU - Steiger, K.* AU - Demir, I.E.* AU - Knolle, P.* AU - Soehnlein, O.* AU - Jenne, D. AU - Hermann, C.D.* AU - Krüger, A.* C1 - 62473 C2 - 50829 CY - 615 Chestnut St, 17th Floor, Philadelphia, Pa 19106-4404 Usa SP - 3568-3579 TI - Timp1 triggers neutrophil extracellular trap formation in pancreatic cancer. JO - Cancer Res. VL - 81 IS - 13 PB - Amer Assoc Cancer Research PY - 2021 SN - 0008-5472 ER - TY - JOUR AU - Banga, J.* AU - Frappart, L.* AU - Hasenauer, J. AU - Herault, Y.* AU - Jonkers, J.* AU - Koubi, D.* AU - Lange, B.* AU - Lines, G.T.* AU - Plouidou, A.* AU - Rinner, O.* C1 - 59402 C2 - 48788 CY - 615 Chestnut St, 17th Floor, Philadelphia, Pa 19106-4404 Usa SP - 35-35 TI - Predictive modeling, applied to genetically engineered mouse models of breast or lung cancer, provides insights into major oncogenic pathways. JO - Cancer Res. VL - 80 IS - 11 PB - Amer Assoc Cancer Research PY - 2020 SN - 0008-5472 ER - TY - JOUR AB - Observational studies have suggested that physical activity might lower the risk of lung cancer in former and current smokers, but not in never-smokers. Using genetic instruments for self-reported and accelerometer-measured physical activity traits implemented through two-sample Mendelian randomization (MR), we sought to strengthen the evidence for causality. We used 18 genome-wide significant (P < 5 x 10(-8)) single-nucleotide polymorphisms (SNP) for self-reported moderate-to-vigorous physical activity and seven SNP for accelerometer-measured ("average acceleration") physical activity from up to 377,234 UK Biobank participants and evaluated these in relation to risk using 29,266 lung cancer cases (including 11,273 adenocarcinomas, 7,426 squamous cell carcinoma, and 2,664 small-cell carcinoma cases) and 56,450 controls. MR analysis suggested no effect of self-reported physical activity [OR (95% confidence interval (CI)) = 0.67 (0.42-1.05); P = 0.081; Q-value = 0.243] and accelerometer-measured activity [OR (95% CI) = 0.98 (0.93-1.03); P = 0.372; Q-value = 0.562] on lung cancer. There was no evidence for associations of physical activity with histologic types and lung cancer in ever and never smokers. Replication analysis using genetic instruments from a different genome-wide study and sensitivity analysis to address potential pleiotropic effects led to no substantive change in estimates. Collectively, these findings do not support a protective relationship between physical activity and the risk of lung cancer.Significance: A new genetic study provides little evidence that recommending physical activity would help prevent lung cancer. AU - Baumeister, S. AU - Leitzmann, M.F.* AU - Bahls, M.* AU - Meisinger, C. AU - Amos, C.* AU - Hung, R.J.* AU - Teumer, A.* AU - Baurecht, H.* C1 - 60125 C2 - 49244 CY - 615 Chestnut St, 17th Floor, Philadelphia, Pa 19106-4404 Usa SP - 3765-3769 TI - Physical activity does not lower the risk of lung cancer. JO - Cancer Res. VL - 80 IS - 17 PB - Amer Assoc Cancer Research PY - 2020 SN - 0008-5472 ER - TY - JOUR AB - Understanding temporal and spatial hemodynamic heterogeneity as a function of tumor growth or therapy affects the development of novel therapeutic strategies. In this study, we employed eigenspectra multispectral optoacoustic tomography (eMSOT) as a next-generation optoacousticmethod to impart high accuracy in resolving tumor hemodynamics during bevacizumab therapy in two types of breast cancer xenografts (KPL-4 and MDA-MB-468). Patterns of tumor total hemoglobin concentration (THb) and oxygen saturation (sO(2)) were imaged in control and bevacizumab-treated tumors over the course of 58 days (KPL-4) and 16 days (MDA-MB-468), and the evolution of functional vasculature "normalization" was resolved macroscopically. Aninitial sharp drop in tumor sO(2) andTHb content shortly after the initiation of bevacizumab treatment was followed by a recovery in oxygenation levels. Rim-core subregion analysis revealed steep spatial oxygenation gradients in growing tumors that were reduced after bevacizumab treatment. Critically, eMSOT imaging findings were validated directly by histopathologic assessment of hypoxia (pimonidazole) and vascularity (CD31). These data demonstrate how eMSOT brings new abilities for accurate observation of entire tumor responses to challenges at spatial and temporal dimensions not available by other techniques today.Significance: Accurate assessment of hypoxia and vascularization over space and time is critical for understanding tumor development and the role of spatial heterogeneity in tumor aggressiveness, metastasis, and response to treatment. AU - Liapis, E. AU - Klemm, U. AU - Karlas, A. AU - Reber, J. AU - Ntziachristos, V. C1 - 61002 C2 - 49630 CY - 615 Chestnut St, 17th Floor, Philadelphia, Pa 19106-4404 Usa SP - 5291-5304 TI - Resolution of spatial and temporal heterogeneity in bevacizumab-treated breast tumors by eigenspectra multispectral optoacoustic tomography. JO - Cancer Res. VL - 80 IS - 23 PB - Amer Assoc Cancer Research PY - 2020 SN - 0008-5472 ER - TY - JOUR AU - Scheel, C. C1 - 59345 C2 - 48751 CY - 615 Chestnut St, 17th Floor, Philadelphia, Pa 19106-4404 Usa TI - Cell fate plasticity during breast cancer development - where is the translational utility? JO - Cancer Res. VL - 80 IS - 4 PB - Amer Assoc Cancer Research PY - 2020 SN - 0008-5472 ER - TY - JOUR AB - Post-translational modifications are essential for regulating the transcription factor p53 which binds DNA in a highly cooperative manner to control expression of a plethora of tumor suppressive programs. Here we show at the biochemical, cellular, and organismal level that the cooperative nature of DNA binding is reduced by phosphorylation of highly conserved serine residues (human S183/S185, mouse S180) in the DNA binding domain. To explore the role of this inhibitory phosphorylation in vivo, new phosphorylation-deficient p53-S180A knock-in mice were generated. ChIP-seq and RNA-seq studies of S180A knock-in cells demonstrated enhanced DNA binding and increased target gene expression. In vivo, this translated into a tissue-specific vulnerability of the bone marrow that caused depletion of hematopoietic stem cells and impaired proper regeneration of hematopoiesis after DNA damage. Median lifespan was significantly reduced by 20% from 709 days in wild-type to only 568 days in S180A littermates. Importantly, lifespan was reduced by a loss of general fitness and increased susceptibility to age-related diseases, not by increased cancer incidence as often seen in other p53 mutant mouse models. For example, S180A knock-in mice showed markedly reduced spontaneous tumorigenesis and increased resistance to Myc-driven lymphoma and Eml4-Alk-driven lung cancer. Preventing phosphorylation of S183/S185 in human cells boosted p53 activity and allowed tumor cells to be killed more efficiently. Together out data identify p53 DNA binding domain phosphorylation as a druggable mechanism that balances tumorigenesis and aging. AU - Timofeev, O.* AU - Koch, L.* AU - Niederau, C.* AU - Tscherne, A.* AU - Schneikert, J.* AU - Klimovich, M.* AU - Elmshäuser, S.* AU - Zeitlinger, M.* AU - Mernberger, M.* AU - Nist, A.* AU - Osterburg, C.* AU - Dötsch, V.* AU - Hrabě de Angelis, M. AU - Stiewe, T.* AU - German Mouse Clinic Consortium (Aguilar-Pimentel, J.A. AU - Schmidt-Weber, C.B. AU - Becker, L. AU - Klopstock, T. AU - Cho, Y.-L. AU - Spielmann, N. AU - Amarie, O.V. AU - Garrett, L. AU - Hölter, S.M. AU - Wurst, W. AU - Calzada-Wack, J. AU - Sanz-Moreno, A. AU - Klein-Rodewald, T. AU - Rathkolb, B. AU - Wolf, E. AU - Östereicher, M.A. AU - Miller, G. AU - Maier, H. AU - Stoeger, C. AU - Leuchtenberger, S. AU - Gailus-Durner, V. AU - Fuchs, H.) C1 - 60042 C2 - 49192 CY - 615 Chestnut St, 17th Floor, Philadelphia, Pa 19106-4404 Usa SP - 5231-5244 TI - Phosphorylation control of p53 DNA binding cooperativity balances tumorigensis and aging. JO - Cancer Res. VL - 80 IS - 23 PB - Amer Assoc Cancer Research PY - 2020 SN - 0008-5472 ER - TY - JOUR AU - Bianco, G.* AU - Montazeri, H.* AU - Quagliata, L.* AU - O'Connor, T. AU - Ehmer, U.* AU - Oellinger, R.* AU - Matter, M.* AU - Gerhard, C.M.* AU - Ng, C.K.Y.* AU - Piscuoglio, S.* AU - Heikenwaelder, M.* AU - Terracciano, L.M.* C1 - 57185 C2 - 47597 CY - 615 Chestnut St, 17th Floor, Philadelphia, Pa 19106-4404 Usa TI - HOXA13 drives hepatocytes proliferation and liver tumorigenesis in mice. JO - Cancer Res. VL - 79 IS - 13 PB - Amer Assoc Cancer Research PY - 2019 SN - 0008-5472 ER - TY - JOUR AB - Mapping tumor heterogeneity and hypoxia within a living intact organism is essential for understanding the processes involved in cancer progression and assessing long-term responses to therapies. Efficient investigations into tumor hypoxia mechanisms have been hindered by the lack of intravital imaging tools capable of multiparametric probing of entire solid tumors with high spatial and temporal resolution. Here, we exploit volumetric multispectral optoacoustic tomography (vMSOT) for accurate, label-free delineation of tumor heterogeneity and dynamic oxygenation behavior. Mice bearing orthotopic MDA-MB-231 breast cancer xenografts were imaged noninvasively during rest and oxygen stress challenge, attaining time-lapse three-dimensional oxygenation maps across entire tumors with 100 mm spatial resolution. Volumetric quantification of the hypoxic fraction rendered values of 3.9% to 21.2%, whereas the oxygen saturation (sO(2)) rate declined at 1.7% to 2.3% permmin all tumors when approaching their core. Three distinct functional areas (the rim, hypoxic, and normoxic cores) were clearly discernible based on spatial sO(2) profiles and responses to oxygen challenge. Notably, although sO(2) readings were responsive to the challenge, deoxyhemoglobin (HbR) trends exhibited little to no variations in all mice. Dynamic analysis further revealed the presence of cyclic hypoxia patterns with a 21% average discrepancy between cyclic fractions assessed via sO(2) (42.2% +/- 17.3%) and HbR fluctuations (63% +/- 14.1%) within the hypoxic core. These findings corroborate the strong potential of vMSOT for advancing preclinical imaging of cancer and informing clinical decisions on therapeutic interventions.Significance: vMSOT provides quantitative measures of volumetric hypoxic fraction and cyclic hypoxia in a label-free and noninvasive manner, providing new readouts to aid tumor staging and treatment decision making. AU - Ron, A. AU - Dean-Ben, X.L. AU - Gottschalk, S. AU - Razansky, D. C1 - 56084 C2 - 46804 CY - 615 Chestnut St, 17th Floor, Philadelphia, Pa 19106-4404 Usa SP - 4767-4775 TI - Volumetric optoacoustic imaging unveils high-resolution patterns of acute and cyclic hypoxia in a murine model of breast cancer. JO - Cancer Res. VL - 79 IS - 18 PB - Amer Assoc Cancer Research PY - 2019 SN - 0008-5472 ER - TY - JOUR AB - Aberrant glutamatergic signaling has been implicated in altered metabolic activity in many cancer types, including malignant melanoma. Previously, we have illustrated the role of metabotropic glutamate receptor 1 (GRM1) in neoplastic transformation of melanocytes in vitro and spontaneous metastatic melanoma in vivo. In this study, we showed that autocrine stimulation constitutively activates the GRM1 receptor and its downstream mitogenic signaling. GRM1-activated (GRM1(+)) melanomas exhibited significantly increased expression of glutaminase (GLS), which catalyzes the first step in the conversion of glutamine to glutamate. In cultured GRM1(+) melanoma cell lines, CB-839, a potent, selective, and orally bioavailable inhibitor of GLS, suppressed cell proliferation, while riluzole, an inhibitor of glutamate release, promoted apoptotic cell death in vitro and in vivo. Combined treatment with CB-839 and riluzole treatment proved to be superior to single-agent treatment, restricting glutamate bioavailability and leading to effective suppression of tumor cell proliferation in vitro and tumor progression in vivo. Hyperactivation of GRM1 in malignant melanoma is an oncogenic driver, which acts independently of canonical melanoma proto-oncogenes, BRAF or NRAS. Overall, these results indicate that expression of GRM1 promotes a metabolic phenotype that supports increased glutamate production and autocrine glutamatergic signaling, which can be pharmacologically targeted by decreasing glutamate bioavailability and the GLS-dependent glutamine to glutamate conversion. AU - Shah, R.* AU - Singh, S.J.* AU - Eddy, K.* AU - Filipp, F.V. AU - Chen, S.* C1 - 55862 C2 - 46622 CY - 615 Chestnut St, 17th Floor, Philadelphia, Pa 19106-4404 Usa SP - 1799-1809 TI - Concurrent targeting of glutaminolysis and metabotropic glutamate receptor 1 (GRM1) reduces glutamate bioavailability in GRM1(+) melanoma. JO - Cancer Res. VL - 79 IS - 8 PB - Amer Assoc Cancer Research PY - 2019 SN - 0008-5472 ER - TY - JOUR AU - Lordick, F.* AU - Haffner, I.* AU - Luber, B.* AU - Maier, D.* AU - Raimundez, E. AU - Hasenauer, J. AU - Kretzschmar, A.K.* AU - von Weikersthal, L.F.* AU - Ahlborn, M.* AU - Knorrenschild, J.R.* AU - Siegler, G.* AU - Rau, B.* AU - Fuxius, S.* AU - Decker, T.* AU - Schierle, K.* AU - Wittekind, C.* C1 - 56371 C2 - 46992 CY - 615 Chestnut St, 17th Floor, Philadelphia, Pa 19106-4404 Usa TI - Heterogeneity of HER2 expression in gastric cancer (GC) leads to high deviation rates between local and central testing and hampers efficacy of anti-HER2 therapy: Survival results from the VARIANZ study. JO - Cancer Res. VL - 78 IS - 13 PB - Amer Assoc Cancer Research PY - 2018 SN - 0008-5472 ER - TY - JOUR AB - Pathogenesis and progression of lung cancer are governed by complex interactions between the environment and host genetic susceptibility, which is further modulated by genetic and epigenetic changes. Autotaxin (ATX, ENPP2) is a secreted glycoprotein that catalyzes the extracellular production of lysophosphatidic acid (LPA), a growth-factor-like phospholipid that is further regulated by phospholipid phosphatases (PLPP). LPA's pleiotropic effects in almost all cell types are mediated through at least six G-protein coupled LPA receptors (LPAR) that exhibit overlapping specificities, widespread distribution, and differential expression profiles. Here we use both preclinical models of lung cancer and clinical samples (from patients and healthy controls) to investigate the expression levels, activity, and biological role of the above components of the ATX/LPA axis in lung cancer. ENPP2 was genetically altered in 8% of patients with lung cancer, whereas increased ATX staining and activity were detected in patient biopsies and sera, respectively. Moreover, PLPP3 expression was consistently downregulated in patients with lung cancer. Comparable observations were made in the two most widely used animal models of lung cancer, the carcinogen urethane-induced and the genetically engineered K-ras(G12D)-driven models, where genetic deletion of Enpp2 or Lpar1 resulted in disease attenuation, thus confirming a procarcinogenic role of LPA signaling in the lung. Expression profiling data analysis suggested that metabolic rewiring may be implicated in the procarcinogenic effects of the ATX/LPA axis in K-ras-(G12D)-driven lung cancer pathogenesis.Significance: These findings establish the role of ATX/LPA in lung carcinogenesis, thus expanding the mechanistic links between pulmonary fibrosis and cancer. AU - Magkrioti, C.* AU - Oikonomou, N.* AU - Kaffe, E.* AU - Mouratis, M.A.* AU - Xylourgidis, N.* AU - Barbayianni, I.* AU - Megadoukas, P.* AU - Harokopos, V.* AU - Valavanis, C.* AU - Chun, J.* AU - Kosma, A.* AU - Stathopoulos, G.T. AU - Bouros, E.* AU - Bouros, D.* AU - Syrigos, K.* AU - Aidinis, V.* C1 - 53946 C2 - 45146 CY - 615 Chestnut St, 17th Floor, Philadelphia, Pa 19106-4404 Usa SP - 3634-3644 TI - The autotaxin - lysophosphatidic acid axis promotes lung carcinogenesis. JO - Cancer Res. VL - 78 IS - 13 PB - Amer Assoc Cancer Research PY - 2018 SN - 0008-5472 ER - TY - JOUR AU - Mueller, S.* AU - Engleitner, T.* AU - Maresch, R.* AU - Zukowska, M.* AU - Lange, S.* AU - Kaltenbacher, T.* AU - Konukiewitz, B.* AU - Oellinger, R.* AU - Zwiebel, M.* AU - Strong, A.* AU - Yen, H.Y.* AU - Banerjee, R.* AU - Louzada, S.* AU - Fu, B.* AU - Seidler, B.* AU - Goetzfried, J.* AU - Schuck, K.* AU - Hassan, Z.* AU - Schoenhuber, N.* AU - Klein, S.* AU - Veltkamp, C.* AU - Friedrich, M.* AU - Rad, L.* AU - Barenboim, M.* AU - Ziegenhain, C.* AU - Hess J. AU - Dovey, O.M.* AU - Eser, S.* AU - Parekh, S.* AU - Constantino-Casas, F.* AU - de la Rosa, J.* AU - Sierra, M.I.* AU - Fraga, M.* AU - Mayerle, J.* AU - Kloeppel, G.* AU - Schmid, R.M.* AU - Cadiñanos, J.* AU - Liu, P.* AU - Vassiliou, G.S.* AU - Weichert, W.* AU - Steiger, K.* AU - Enard, W.* AU - Yang, F.* AU - Unger, K. AU - Schneider, G.* AU - Varela, I.* AU - Bradley, A.* AU - Saur, D.* AU - Rad, R.* C1 - 56321 C2 - 46977 CY - 615 Chestnut St, 17th Floor, Philadelphia, Pa 19106-4404 Usa TI - Evolutionary trajectories and KRAS gene dosage define pancreatic cancer phenotypes. JO - Cancer Res. VL - 78 IS - 13 PB - Amer Assoc Cancer Research PY - 2018 SN - 0008-5472 ER - TY - JOUR AB - Glycolysis and fatty acid synthesis are highly active in cancer cells through cytosolic citrate metabolism, with intracellular citrate primarily derived from either glucose or glutamine via the tricarboxylic acid cycle. We show here that extracellular citrate is supplied to cancer cells through a plasma membrane-specific variant of the mitochondrial citrate transporter (pmCiC). Metabolomic analysis revealed that citrate uptake broadly affected cancer cell metabolism through citrate-dependent metabolic pathways. Treatment with gluconate specifically blocked pmCiC and decreased tumor growth in murine xenografts of human pancreatic cancer. This treatment altered metabolism within tumors, including fatty acid metabolism. High expression of pmCiC was associated with invasion and advanced tumor stage across many human cancers. These findings support the exploration of extracellular citrate transport as a novel potential target for cancer therapy. Significance: Uptake of extracellular citrate through pmCiC can be blocked with gluconate to reduce tumor growth and to alter metabolic characteristics of tumor tissue. AU - Mycielska, M.E.* AU - Dettmer, K.* AU - Rümmele, P.* AU - Schmidt, K.* AU - Prehn, C. AU - Milenkovic, V.M.* AU - Jagla, W.* AU - Madej, M.G.* AU - Lantow, M.* AU - Schladt, M.T.* AU - Cecil, A. AU - Koehl, G.E.* AU - Eggenhofer, E.* AU - Wachsmuth, C.J.* AU - Ganapathy, V.* AU - Schlitt, H.J.* AU - Kunzelmann, K.* AU - Ziegler, C.* AU - Wetzel, C.H.* AU - Gaumann, A.* AU - Lang, S.A.* AU - Adamski, J. AU - Oefner, P.J.* AU - Geissler, E.K.* C1 - 53208 C2 - 44348 SP - 2513-2523 TI - Extracellular citrate affects critical elements of cancer cell metabolism and supports cancer development in vivo. JO - Cancer Res. VL - 78 IS - 10 PY - 2018 SN - 0008-5472 ER - TY - JOUR AU - Schirmer, D.* AU - Storz, I.* AU - Wisskirchen, K.* AU - Feederle, R. AU - Schmidt, O.* AU - Abken, H.* AU - Protzer, U.* AU - Burdach, S.* AU - Richter, G.H.S.* C1 - 56267 C2 - 46906 CY - 615 Chestnut St, 17th Floor, Philadelphia, Pa 19106-4404 Usa TI - GPR64-specific CAR-transgenic T cells selectively kill Ewing sarcoma in vivo. JO - Cancer Res. VL - 78 IS - 19 PB - Amer Assoc Cancer Research PY - 2018 SN - 0008-5472 ER - TY - JOUR AB - Although oncogenic activation of NFkB has been identified in various tumors, the NFkB–activating kinases (inhibitor of NFkB kinases, IKK) responsible for this are elusive. In this study, we determined the role of IKKa and IKKb in KRAS-mutant lung adenocarcinomas induced by the carcinogen urethane and by respiratory epithelial expression of oncogenic KRASG12D. Using NFkB reporter mice and conditional deletions of IKKa and IKKb, we identified two distinct early and late activation phases of NFkB during chemical and genetic lung adenocarcinoma development, which were characterized by nuclear translocation of RelB, IkBb, and IKKa in tumor-initiated cells. IKKa was a cardinal tumor promoter in chemical and genetic KRAS-mutant lung adenocarcinoma, and respiratory epithelial IKKa-deficient mice were markedly protected from the disease. IKKa specifically cooperated with mutant KRAS for tumor induction in a cell-autonomous fashion, providing mutant cells with a survival advantage in vitro and in vivo. IKKa was highly expressed in human lung adenocarcinoma, and a heat shock protein 90 inhibitor that blocks IKK function delivered superior effects against KRAS-mutant lung adenocarcinoma compared with a specific IKKb inhibitor. These results demonstrate an actionable requirement for IKKa in KRAS-mutant lung adenocarcinoma, marking the kinase as a therapeutic target against this disease. Significance: These findings report a novel requirement for IKKa in mutant KRAS lung tumor formation, with potential therapeutic applications. AU - Vreka, M. AU - Lilis, I.* AU - Papageorgopoulou, M. AU - Giotopoulou, G.A. AU - Lianou, M.* AU - Giopanou, I.* AU - Kanellakis, N.I.* AU - Spella, M.* AU - Agalioti, T.* AU - Armenis, V.* AU - Goldmann, T.* AU - Marwitz, S.* AU - Yull, F.E.* AU - Blackwell, T.S.* AU - Pasparakis, M.* AU - Marazioti, A.* AU - Stathopoulos, G.T. C1 - 53606 C2 - 44697 CY - Elsevier House, Brookvale Plaza, East Park Shannon, Co, Clare, 00000, Ireland SP - 2939-2951 TI - IkB kinase a Is required for development and progression of KRAS-mutant lung adenocarcinoma. JO - Cancer Res. VL - 78 IS - 11 PB - Elsevier Ireland Ltd PY - 2018 SN - 0008-5472 ER - TY - JOUR AU - Wierling, C.* AU - Herault, Y.* AU - Jonkers, J.* AU - Ploubidou, A.* AU - Frappart, L.* AU - Hasenauer, J. AU - Banga, J.R.* AU - Rinner, O.* AU - Naumova, V.* AU - Koubi, D.* AU - Lange, B.* C1 - 56322 C2 - 46976 CY - 615 Chestnut St, 17th Floor, Philadelphia, Pa 19106-4404 Usa TI - CanPathPro-development of a platform for predictive pathway modelling using genetically engineered mouse models. JO - Cancer Res. VL - 78 IS - 13 PB - Amer Assoc Cancer Research PY - 2018 SN - 0008-5472 ER - TY - JOUR AU - Zoni, E.* AU - Buck, A. AU - Feuchtinger, A. AU - Spahn, M.* AU - Walch, A.K. AU - Kruithof-de Julio, M.* C1 - 54204 C2 - 45317 CY - 615 Chestnut St, 17th Floor, Philadelphia, Pa 19106-4404 Usa SP - 61-62 TI - Metabolic signature in lethal vs. nonlethal prostate cancer. JO - Cancer Res. VL - 78 IS - 16 PB - Amer Assoc Cancer Research PY - 2018 SN - 0008-5472 ER - TY - JOUR AU - Gires, O. C1 - 50942 C2 - 42750 SP - 1775-1776 TI - EGFR-dependent regulated intramembrane proteolysis of EpCAM - Letter. JO - Cancer Res. VL - 77 IS - 7 PY - 2017 SN - 0008-5472 ER - TY - JOUR AB - In vivo tumor labeling with fluorescent agents may assist endoscopic and surgical guidance for cancer therapy as well as create opportunities to directly observe cancer biology in patients. However, malignant and non-malignant tissues are usually distinguished on fluorescence images by applying empirically determined fluorescence intensity thresholds. Here we report the development of fSTREAM, a set of analytic methods designed to streamline the analysis of surgically excised breast tissues by collecting and statistically processing hybrid multi-scale fluorescence, color, and histology readouts toward precision fluorescence imaging. fSTREAM addresses core questions of how to relate fluorescence intensity to tumor tissue and how to quantitatively assign a normalized threshold that sufficiently differentiates tumor tissue from healthy tissue. Using fSTREAM we assessed human breast tumors stained in vivo with fluorescent bevacizumab at microdose levels Showing that detection of such levels is achievable, we validated fSTREAM for high-resolution mapping of the spatial pattern of labeled antibody and its relation to the underlying cancer pathophysiology and tumor border on a per patient basis. We demonstrated a 98% sensitivity and 79% specificity when using labelled bevacizumab to outline the tumor mass. Overall, our results illustrate a quantitative approach to relate fluorescence signals to malignant tissues and improve the theranostic application of fluorescence molecular imaging. AU - Koch, M. AU - de Jong, J.S.* AU - Glatz, J. AU - Symvoulidis, P. AU - Lamberts, L.E.* AU - Adams, A.L.L.* AU - Kranendonk, M.E.G.* AU - Terwisscha van Scheltinga, A.G.* AU - Aichler, M. AU - Jansen, L.* AU - de Vries, J.* AU - Lub-de, Hoog, M.N.* AU - Schröder, C.P.* AU - Jorritsma-Smit, A.* AU - Linssen, M.D.* AU - de Boer, E.* AU - van der Vegt, B.* AU - Nagengast, W.B.* AU - Elisas,S.G.* AU - Oliveira, S.* AU - Witkamp, A.J.* AU - Mali, W.P.Th.M.* AU - van der Wall, E.* AU - Gracia-Allende, P.B. AU - van Diest, P.J.* AU - de Vries, E.G.* AU - Walch, A.K. AU - van Dam, G.M.* AU - Ntziachristos, V. C1 - 50000 C2 - 41961 CY - Philadelphia SP - 623-631 TI - Threshold analysis and biodistribution of fluorescently labeled bevacizumab in human breast cancer. JO - Cancer Res. VL - 77 IS - 3 PB - Amer Assoc Cancer Research PY - 2017 SN - 0008-5472 ER - TY - JOUR AB - Inherent intermediate-to-low affinity T cell receptors (TCR) that develop during the natural course of immune responses may not allow sufficient activation for tumor elimination, making the majority of T cells suboptimal for adoptive T cell therapy (ATT). TCR affinity enhancement has been implemented to provide stronger T cell activity but carries the risk of creating undesired cross-reactivity leading to potential serious adverse effects in clinical application. We demonstrate here that engineering of low-avidity T cells recognizing a naturally processed and presented tumor-associated antigen with a chimeric PD-1:28 receptor increases effector function to levels seen with high-avidity T cells of identical specificity. Upgrading the function of low-avidity T cells without changing the TCR affinity will allow a large arsenal of low-avidity T cells previously thought to be therapeutically inefficient to be considered for ATT. PD-1:28 engineering re-instated Th1 function in tumor-infiltrating lymphocytes (TILs) that had been functionally disabled in the human renal cell carcinoma (RCC) environment without unleashing undesired Th2 cytokines or IL-10. Involved mechanisms may be correlated to restoration of ERK and AKT signaling pathways. In mouse tumor models of ATT, PD-1:28 engineering enabled low-avidity T cells to proliferate stronger and prevented PD-L1 upregulation and Th2 polarization in the tumor milieu. Engineered T cells combined with checkpoint blockade secreted significantly more IFN-γ compared to T cells without PD-1:28, suggesting a beneficial combination with checkpoint blockade therapy or other therapeutic strategies. Altogether, the supportive effects of PD-1:28 engineering on T cell function makes it an attractive tool for ATT. AU - Schlenker, R. AU - Olguín-Contreras, L.F. AU - Leisegang, M.* AU - Schnappinger, J. AU - Disovic, A. AU - Rühland, S.* AU - Nelson, P.J.* AU - Leonhardt, H.* AU - Harz, H.* AU - Wilde, S.* AU - Schendel, D.J.* AU - Uckert, W.* AU - Willimsky, G.* AU - Nößner, E. C1 - 51181 C2 - 43074 SP - 3577-3590 TI - Chimeric PD-1:28 receptor upgrades low-avidity T cells and restores effector function of tumor-infiltrating lymphocytes for adoptive cell therapy. JO - Cancer Res. VL - 77 IS - 13 PY - 2017 SN - 0008-5472 ER - TY - JOUR AB - Identifying genetic variants with pleiotropic associations can uncover common pathways influencing multiple cancers. We took a two-staged approach to conduct genome-wide association studies for lung, ovary, breast, prostate and colorectal cancer from the GAME-ON/GECCO Network (61,851 cases, 61,820 controls) to identify pleiotropic loci. Findings were replicated in independent association studies (55,789 cases, 330,490 controls). We identified a novel pleiotropic association at 1q22 involving breast and lung squamous cell carcinoma, with eQTL analysis showing an association with ADAM15/THBS3 gene expression in lung. We also identified a known breast cancer locus CASP8/ALS2CR12 associated with prostate cancer, a known cancer locus at CDKN2B-AS1 with different variants associated with lung adenocarcinoma and prostate cancer and confirmed the associations of a breast BRCA2 locus with lung and serous ovarian cancer. This is the largest study to date examining pleiotropy across multiple cancer-associated loci, identifying common mechanisms of cancer development and progression. AU - Fehringer, G.* AU - Kraft, P.* AU - Pharoah, P.D.* AU - Eeles, R.A.* AU - Chatterjee, N.* AU - Schumacher, F.R.* AU - Schildkraut, J.M.* AU - Lindström, S.* AU - Brennan, P.* AU - Bickeböller, H.* AU - Houlston, R.S.* AU - Landi, M.T.* AU - Caporaso, N.E.* AU - Risch, A.* AU - Amin Al Olama, A.* AU - Berndt, S.I.* AU - Giovannucci, E.* AU - Grönberg, H.* AU - Kote-Jarai, Z.* AU - Ma, J.* AU - Muir, K.* AU - Stampfer, M.J.* AU - Stevens, V.L.* AU - Wiklund, F.* AU - Willett, W.C.* AU - Goode, E.L. AU - Permuth, J.B.* AU - Risch, H.A.* AU - Reid, B.M.* AU - Bézieau, S.* AU - Brenner, H.* AU - Chan, A.T.* AU - Chang-Claude, J.* AU - Hudson, T.J.* AU - Kocarnik, J.M.* AU - Newcomb, P.A.* AU - Schoen, R.E.* AU - Slattery, M.L.* AU - White, E.S.* AU - Adank, M.A.* AU - Hereditary Breast and Ovarian Cancer Research Group Netherlands (HEBON) (*) AU - Ahsan, H.* AU - Aittomäki, K.* AU - Baglietto, L.* AU - Blomquist, C.H.* AU - Canzian, F.* AU - Czene, K.* AU - Dos Santos Silva, I.* AU - Eliassen, A.H.* AU - Figueroa, J.D.* AU - Flesch-Janys, D.* AU - Fletcher, O.* AU - Garcia-Closas, M.* AU - Gaudet, M.M.* AU - Johnson, N.* AU - Hall, P.* AU - Hazra, A.* AU - Hein, R.* AU - Hofman, A.* AU - Hopper, J.L.* AU - Irwanto, A.* AU - Johansson, M.* AU - Kaaks, R.* AU - Kibriya, M.G.* AU - Lichtner, P. AU - Liu, J.J.* AU - Lund, E.* AU - Makalic, E.* AU - Meindl, A.* AU - Müller-Myhsok, B.* AU - Muranen, T.A.* AU - Nevanlinna, H.* AU - Peeters, P.H.* AU - Peto, J.* AU - Prentice, R.L.* AU - Rahman, N.* AU - Sánchez, M.J.* AU - Schmidt, D.F.* AU - Schmutzler, R.K.* AU - Southey, M.C.* AU - Tamimi, R.M.* AU - Travis, R.C.* AU - Turnbull, C.* AU - Uitterlinden, A.G.* AU - Wang, Z.* AU - Whittemore, A.S.* AU - Yang, X.R.* AU - Zheng, W.* AU - Rafnar, T.* AU - Gudmundsson, J.* AU - Stacey, S.N.* AU - Stefansson, K.* AU - Sulem, P.* AU - Chen, Y.A.* AU - Tyrer, J.P.* AU - Christiani, D.C.* AU - Wei, Y.* AU - Shen, H.* AU - Hu, Z.* AU - Shu, X.O.* AU - Shiraishi, K.* AU - Takahashi, A.* AU - Bossé, Y.* AU - Obeidat, M.* AU - Nickle, D.C.* AU - Timens, W.* AU - Freedman, M.L.* AU - Li, Q.L.* AU - Seminara, D.* AU - Chanock, S.J.* AU - Gong, J.* AU - Peters, U.* AU - Gruber, S.B.* AU - Colorectal Transdisciplinary Study (CORECT) (*) AU - Amos, C.I.* AU - Sellers, T.A.* AU - Easton, D.F.* AU - Hunter, D.J.* AU - Haiman, C.A.* AU - African American Breast Cancer Consortium (AABC) (*) AU - African Ancestry Prostate Cancer Consortium (AAPC) (*) AU - Henderson, B.E.* AU - Hung, R.J.* C1 - 48758 C2 - 41327 CY - Philadelphia SP - 5103-5114 TI - Cross-cancer genome-wide analysis of lung, ovary, breast, prostate and colorectal cancer reveals novel pleiotropic associations. JO - Cancer Res. VL - 76 IS - 17 PB - Amer Assoc Cancer Research PY - 2016 SN - 0008-5472 ER - TY - JOUR AB - Background: Non-small cell lung cancer (NSCLC) is one of the most aggressive tumor entities and first data indicate that microRNAs (miRNAs) are central regulators of NSCLC dissemination. Since each miRNA is able to modulate the expression of several transcripts, they are promising targets for the development of drugs that cause efficient antitumor effects and low resistance. However, the relevant network of miRNA/mRNA driving NSCLC metastasis has not been identified yet. Methods: The differential expression of miRNAs was compared between NSCLC samples from patients with and without lymph node metastasis (N1, N2 and N3 vs. N0) in a cohort of The Cancer Genomic Atlas (TCGA) database (n = 449). The dysregulation of the miRNAs in tumors versus normal lung samples (n = 39) was also analyzed. For validation, fresh-frozen samples from an independent patient cohort (n = 108) were analyzed by qRT-PCR. The role of selected miRNAs in tumor dissemination was assessed by migration and invasion experiments after transfection of respective antagomirs and agomirs in NSCLC cells (time-lapse microscopy). The novel algorithm “miRlastic” was used to identify potential miRNA targets through the integration of miRNA-mRNA expression data by negative multiple linear regression analysis. Moreover, differential methylation of the miRNA genomic locations was studied as a possible mechanism of miRNA dysregulation by analyzing Illumina Infinium 450 k DNA methylation TCGA data (n = 29). Results: By using a stringent selection process, we identified 135 miRNAs differentially induced or reduced in NSCLCs with lymph node metastasis (p≤0.05). Interestingly, 22/135 (16.3%) of the selected miRNAs were located in the chromosomal cluster 14q32.31. Elevated expression of miR-323b, miR-487a and miR-539, which are located in 14q32.31, significantly correlated with poor patient survival. Time-resolved and quantitative analysis of lateral migration illustrated that these miRNAs increased tumor migration without affecting cell viability. Moreover, miRlastic identified several metastasis-related genes as potential downstream targets of these miRNAs. The connection between miRNAs encoded in 14q32.31 and candidate targets was confirmed in NSCLC cell lines (e.g. Pumilio RNA-Binding Family Member-2; PUM2). Lastly, hypomethylation of the 14q32.31 cluster in tumor tissues might explain increased expression of these miRNAs. Conclusions: Our results demonstrate that miRNAs located in the chromosomal cluster 14q32.31 are driving NSCLC dissemination. Therefore, we hypothesize that the coordinated overexpression of these miRNAs is part of a genetic network supporting cancer progression and that they represent promising cancer biomarkers and therapeutic targets. Citation Format: Margarita González-Vallinas, Marco Albrecht, Adriana Pitea, Manuel Rodríguez-Paredes, Damian Stichel, Steffen Sass, Julian Gutekunst, Jennifer Schmitt, Thomas Muley, Michael Meister, Arne Warth, Peter Schirmacher, Fabian J. Theis, Nikola S. Müller, Franziska Matthäus, Kai Breuhahn. Identification of a miRNA/mRNA network driving non-small cell lung cancer (NSCLC) dissemination. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1945. AU - González-Valllinas, M.* AU - Albrecht, M.* AU - Pitea, A. AU - Rodriguez-Paredes, M.* AU - Stichel, D.* AU - Sass, S. AU - Gutekunst, J.* AU - Schmitt, J.* AU - Muley, T.* AU - Meister, M.* AU - Warth, A.* AU - Schirmacher, P.* AU - Theis, F.J. AU - Müller, N.S. AU - Matthäus, F.* AU - Breuhahn, K.* C1 - 50362 C2 - 42166 TI - Abstract 1945: Identification of a miRNA/mRNA network driving non-small cell lung cancer (NSCLC) dissemination. JO - Cancer Res. VL - 76 IS - 14 PY - 2016 SN - 0008-5472 ER - TY - JOUR AB - RAF kinase inhibitor protein (RKIP) is a seminal regulator of intracellular signaling and exhibits both anti-metastatic and anti-tumorigenic properties. Decreased expression of RKIP has been described in several human malignancies, including acute myeloid leukemia (AML). As the mechanisms leading to RKIP loss in AML are still unclear, we aimed to analyze the potential involvement of micro-RNAs (miRNAs) within this study. miRNA microarray and qPCR data of more than 400 AML patient specimens revealed correlation between decreased expression of RKIP and increased expression of miR-23a, a member of the miR-23a/27a/24-2 cluster. In functional experiments, overexpression of miR-23a decreased RKIP mRNA and protein expression, whereas miR-23a inhibition caused the opposite effect. By employing an RKIP 3'UTR luciferase reporter construct with and without mutation or deletion of the putative miR-23a binding site, we could show that RKIP modulation by miR-23a is mediated via direct binding to this region. Importantly, miR-23a overexpression induced a significant increase of proliferation in hematopoietic cells. Simultaneous transfection of an RKIP expression construct lacking the miR-23a binding sites reversed this phenotype, indicating that this effect is truly mediated via downregulation of RKIP. Finally, by analyzing more than 4300 primary patient specimens via database retrieval from The Cancer Genome Atlas (TCGA), we could highlight the importance of the miR-23a/RKIP axis in a broad range of human cancer entities. In conclusion, we have identified miR-23a as a negative regulator of RKIP expression in AML and have provided data that suggest the importance of our observation beyond this tumor entity. AU - Hatzl, S.* AU - Geiger, O.* AU - Kuepper, M.K.* AU - Caraffini, V.* AU - Seime, T.* AU - Furlan, T.* AU - Nussbaumer, E.* AU - Wieser, R.* AU - Pichler, M.* AU - Scheideler, M. AU - Nowek, K.* AU - Lavrencic, M.* AU - Quenhenberger, F.* AU - Wölfler, A.* AU - Troppmair, J.* AU - Sill, H.* AU - Zebisch, A.* C1 - 48631 C2 - 41237 CY - Philadelphia SP - 3644-3654 TI - Increased expression of miR-23a mediates a loss of expression in the RAF kinase inhibitor protein RKIP. JO - Cancer Res. VL - 76 IS - 12 PB - Amer Assoc Cancer Research PY - 2016 SN - 0008-5472 ER - TY - JOUR AB - Sensitive in vivo imaging technologies applicable to the clinical setting are still lacking for adoptive T-cell-based immunotherapies, an important gap to fill if mechanisms of tumor rejection or escape are to be understood. Here, we propose a highly sensitive imaging technology to track human TCR-transgenic T cells in vivo by directly targeting the murinized constant TCR beta domain (TCRmu) with a zirconium-89 ((89)Zr)-labeled anti-TCRmu-F(ab')2 fragment. Binding of the labeled or unlabeled F(ab')2 fragment did not impair functionality of transgenic T cells in vitro and in vivo Using a murine xenograft model of human myeloid sarcoma, we monitored by Immuno-PET imaging human central memory T cells (TCM), which were transgenic for a myeloid peroxidase (MPO)-specific TCR. Diverse T-cell distribution patterns were detected by PET/CT imaging, depending on the tumor size and rejection phase. Results were confirmed by IHC and semiquantitative evaluation of T-cell infiltration within the tumor corresponding to the PET/CT images. Overall, these findings offer a preclinical proof of concept for an imaging approach that is readily tractable for clinical translation. AU - Mall, S.* AU - Yusufi, N.* AU - Wagner, R.* AU - Klar, R.* AU - Bianchi, H.* AU - Steiger, K.* AU - Straub, M.* AU - Audehm, S.* AU - Laitinen, I.* AU - Aichler, M. AU - Peschel, C.* AU - Ziegler, S.* AU - Mustafa, M.* AU - Schwaiger, M.* AU - D'Alessandria, C.* AU - Krackhardt, A.M. C1 - 48964 C2 - 41509 CY - Philadelphia SP - 4113-4123 TI - Immuno-PET imaging of engineered human T cells in tumors. JO - Cancer Res. VL - 76 IS - 14 PB - Amer Assoc Cancer Research PY - 2016 SN - 0008-5472 ER - TY - JOUR AB - Metabolism of nicotine by cytochrome CYP2A6 is a suspected determinant of smoking dose and, consequently, lung cancer risk. We conducted a genome-wide association study (GWAS) of CYP2A6 activity, as measured by the urinary ratio of trans-3'-hydroxycotinine and its glucuronide conjugate over cotinine (total 3HCOT/COT), among 2,239 smokers in the Multiethnic Cohort (MEC) study. We identified 248 CYP2A6 variants associated with CYP2A6 activity (p<5x10-8). CYP2A6 activity was correlated (r=0.32, p<0.0001) with total nicotine equivalents (a measure of nicotine uptake). When we examined the effect of these variants on lung cancer risk in the Transdisciplinary Research in Cancer of the Lung (TRICL) consortium GWAS dataset (13,479 cases, 43,218 controls), we found that the vast majority of these individual effects were directionally consistent and associated with an increased lung cancer risk. 226 of the 248 variants associated with CYP2A6 activity in the MEC were available in TRICL. Of them, 81% had directionally consistent risk estimates and six were globally significantly associated with lung cancer. When conditioning on nine known functional variants and two deletions, the top two SNPs (rs56113850 in MEC and rs35755165 in TRICL) remained significantly associated with CYP2A6 activity in MEC and lung cancer in TRICL. The present data support the hypothesis that a greater CYP2A6 activity causes smokers to smoke more extensively and be exposed to higher levels of carcinogens, resulting in an increased risk for lung cancer. Although the variants identified in these studies may be used as risk prediction markers, the exact causal variants remain to be identified. AU - Patel, Y.M.* AU - Park, S.L.* AU - Han, Y.* AU - Wilkens, L.R.* AU - Bickeböller, H.* AU - Rosenberger, A.* AU - Caporaso, N.* AU - Landi, M.T.* AU - Brüske, I. AU - Risch, A.* AU - Wei, Y.* AU - Christiani, D.C.* AU - Brennan, P.* AU - Houlston, R.S.* AU - Mckay, J.* AU - McLaughlin, J.* AU - Hung, R.J.* AU - Murphy, S.E.* AU - Stram, D.O.* AU - Amos, C.I.* AU - Le Marchand, L.* C1 - 49226 C2 - 33883 CY - Philadelphia SP - 5768-5776 TI - Novel association of genetic markers affecting CYP2A6 activity and lung cancer risk. JO - Cancer Res. VL - 76 IS - 19 PB - Amer Assoc Cancer Research PY - 2016 SN - 0008-5472 ER - TY - JOUR AU - Scheel, C. AU - Linnemann, J.R.* AU - Miura, H. AU - Meixner, L.K. AU - Irmier, M.* AU - Kloos, U.J.* AU - Hirschi, B. AU - Bartsch, H.S.* AU - Sass, S. AU - Beckers, J. AU - Theis, F.J. AU - Gabka, C.* AU - Sotlar, K.* C1 - 48774 C2 - 41404 CY - Philadelphia TI - A force-sensitive organoid assay to quantify regenerative potential of single primary human mammary cells. JO - Cancer Res. VL - 76 PB - Amer Assoc Cancer Research PY - 2016 SN - 0008-5472 ER - TY - JOUR AB - Hepatocellular carcinoma (HCC) represents the second leading cause of cancer-related deaths and is reported to be resistant to chemotherapy caused by tumor-initiating cells. These tumor-initiating cells express stem cell markers. An accumulation of tumor-initiating cells be found in 28-50% of all HCC and is correlated with a poor prognosis. Mechanisms that mediate chemoresistance include drug export, increased metabolism and quiescence. Importantly, the mechanisms that regulate quiescence in tumor-initiating cells have not been analyzed in detail so far. In the present research we have developed a single cell tracking method to follow up the fate of tumor-initiating cells during chemotherapy. Thereby, we were able to demonstrate that mCXCL1 exerts cellular state specific effects regulating the resistance to chemotherapeutics. mCXCL1 is the mouse homolog of the human Interleukin 8, a chemokine which correlates with poor prognosis in HCC patients. We found that mCXCL1 blocks differentiationof premalignant cells and activates quiescence in tumor-initiating cells. This process depends on the activation of the mTORC1 kinase. Blocking of the mTORC1 kinase induces differentiation of tumor-initiating cells and allows their subsequent depletion using the chemotherapeutic drug doxorubicin. Our work deciphers the mCXCL1-mTORC1 pathway as crucial in liver cancer stem cell maintenance and highlights it as a novel target in combination with conventional chemotherapy. AU - Wolf, B.* AU - Krieg, K.* AU - Falk, C.* AU - Breuhahn, K.* AU - Keppeler, H.* AU - Biedermann, T.* AU - Schmid, E.* AU - Warmann, S.* AU - Fuchs, J.* AU - Vetter, S.* AU - Thiele, D.* AU - Nieser, M.* AU - Avci-Adali, M.* AU - Skokowa, Y.* AU - Schöls, L.* AU - Hauser, S.L.* AU - Ringelhan, M. AU - Yevsa, T.* AU - Heikenwälder, M. AU - Kossatz-Boehlert, U.* C1 - 49373 C2 - 41789 CY - Philadelphia SP - 5550-5561 TI - Inducing differentiation of premalignant hepatic cells as a novel therapeutic strategy in hepatocarcinoma. JO - Cancer Res. VL - 76 IS - 18 PB - Amer Assoc Cancer Research PY - 2016 SN - 0008-5472 ER - TY - JOUR AU - Anastasov, N. AU - Höfig, I. AU - Radulovic, V. AU - Richter, S. AU - Lichtenberg, J.* AU - Kelm, J.M.* AU - Thirion, C.* AU - Atkinson, M.J. C1 - 48225 C2 - 41029 CY - Philadelphia TI - Three-dimensional microtissues as phenotypic screening model to identify radiation modifiers for breast cancer. JO - Cancer Res. VL - 75 PB - Amer Assoc Cancer Research PY - 2015 SN - 0008-5472 ER - TY - JOUR AU - Bigalke, I.* AU - Honnashagen, K.* AU - Lundby, M.* AU - Solum, G.* AU - Skoge, L. AU - Inderberg, E.M.S.* AU - Kasten, J. AU - Saboe-Larssen, S.* AU - Schendel, D.J.* AU - Kvalheim, G.* C1 - 48224 C2 - 41030 CY - Philadelphia TI - A new generation of dendritic cells to improve cancer therapy shows prolonged progression-free survival in patients with solid tumors. JO - Cancer Res. VL - 75 PB - Amer Assoc Cancer Research PY - 2015 SN - 0008-5472 ER - TY - JOUR AU - Höfig, I. AU - Rosemann, M. AU - Albrecht, M. AU - Ingawale, Y. AU - Kelm, J.M.* AU - Atkinson, M.J. AU - Thirion, C.* AU - Anastasov, N. C1 - 48223 C2 - 41031 CY - Philadelphia TI - Generation of 3D-microtissues suitable for drug screening with lentivirally GFP-labelled CD44+CD24-breast cancer cells enriched by irradiation. JO - Cancer Res. VL - 75 PB - Amer Assoc Cancer Research PY - 2015 SN - 0008-5472 ER - TY - JOUR AB - Optoacoustic imaging combines the rich contrast of optical methods with the resolution of ultrasound imaging. It can therefore deliver optical visualization of cancer far deeper in tissue than optical microscopy and other conventional optical imaging methods. Technological progress and novel contrast media have resulted in optoacoustic imaging being propagated to basic cancer research and in clinical translation projects. We briefly review recent technological advances, showcase the ability to resolve unique cancer biomarkers based on spectral features at different imaging scales, and highlight the imaging performance achieved in preclinical and clinical imaging applications. AU - Taruttis, A. AU - van Dam, G.M.* AU - Ntziachristos, V. C1 - 44111 C2 - 36744 CY - Philadelphia SP - 1548-1559 TI - Mesoscopic and macroscopic optoacoustic imaging of cancer. JO - Cancer Res. VL - 75 IS - 8 PB - Amer Assoc Cancer Research PY - 2015 SN - 0008-5472 ER - TY - JOUR AU - Toma, M.I.* AU - Wehner, R.* AU - Kloss, A.* AU - Erdmann, K.* AU - Fuessel, S.* AU - Seliger, B.* AU - Brech, D. AU - Nößner, E. AU - Schaekel, K.* AU - Wirth, M.P.* AU - Baretton, G.* AU - Schmitz, M.* C1 - 48226 C2 - 41028 CY - Philadelphia TI - Accumulation of tolerogenic human 6-sulfo LacNAc plus dendritic cells in renal cell carcinoma is associated with poor prognosis. JO - Cancer Res. VL - 75 PB - Amer Assoc Cancer Research PY - 2015 SN - 0008-5472 ER - TY - JOUR AB - Abstract Chromosomal instability (CIN) is associated with poor outcome in epithelial malignancies, including breast carcinomas. Evidence suggests that prognostic signatures in estrogen receptor-positive (ER(+)) breast cancer define tumors with CIN and high proliferative potential. Intriguingly, CIN induction in lower eukaryotic cells and human cells is context dependent, typically resulting in a proliferation disadvantage but conferring a fitness benefit under strong selection pressures. We hypothesized that CIN permits accelerated genomic evolution through the generation of diverse DNA copy-number events that may be selected during disease development. In support of this hypothesis, we found evidence for selection of gene amplification of core regulators of proliferation in CIN-associated cancer genomes. Stable DNA copy-number amplifications of the core regulators TPX2 and UBE2C were associated with expression of a gene module involved in proliferation. The module genes were enriched within prognostic signature gene sets for ER(+) breast cancer, providing a logical connection between CIN and prognostic signature expression. Our results provide a framework to decipher the impact of intratumor heterogeneity on key cancer phenotypes, and they suggest that CIN provides a permissive landscape for selection of copy-number alterations that drive cancer proliferation. AU - Endesfelder, D. AU - Burrell, R.A.* AU - Kanu, N.* AU - McGranahan, N.* AU - Howell, M.* AU - Parker, P.J.* AU - Downward, J.* AU - Swanton, C.* AU - Kschischo, M.* C1 - 32165 C2 - 34975 CY - Philadelphia SP - 4853-4863 TI - Chromosomal instability selects gene copy number variants encoding core regulators of proliferation in ER+ breast cancer. JO - Cancer Res. VL - 74 IS - 17 PB - Amer Assoc Cancer Research PY - 2014 SN - 0008-5472 ER - TY - JOUR AU - Faucz, F.R.* AU - Beuschlei, F.* AU - Fassnacht, M.* AU - Assié, G.* AU - Calebiro, D.* AU - Stratakis, C.A.* AU - Osswald, A.* AU - Ronchi, C.L.* AU - Wieland, T. AU - Sbiera, S.* AU - Schaak, K.* AU - Schmittfull, A.* AU - Schwarzmayr, T. AU - Barreau, O.* AU - Vezzosi, D.* AU - Rizk-Rabbin, M.* AU - Zabel, U.* AU - Szarek, E.* AU - Salpea, P.* AU - Forlino, A.* AU - Vetro, A.* AU - Zuffardi, O.* AU - Kisker, C.* AU - Diener, S. AU - Meitinger, T. AU - Lohse, M.J.* AU - Reincke, M.* AU - Bertherat, J.* AU - Strom, T.M. AU - Allolio, B.* C1 - 45031 C2 - 37150 CY - Philadelphia TI - Constitutive activation of PRKACA in adrenal Cushing's syndrome. JO - Cancer Res. VL - 74 IS - 19 PB - Amer Assoc Cancer Research PY - 2014 SN - 0008-5472 ER - TY - JOUR AB - CD40, a member of the Tumor necrosis factor receptor family, is expressed on all mature B-cells and on most B-cell lymphomas. Recently, we have shown that constitutive activation of CD40-signaling in B-cells induced by a fusion protein consisting of the transmembrane part of the Epstein-Barr viral Latent Membrane Protein 1 (LMP1) and the cytoplasmic part of CD40 (LMP1/CD40) drives B-cell lymphoma development in transgenic mice. Since LMP1/CD40-expressing B-cells showed an upregulation of CD19, we investigated CD19 function in CD40-driven B-cell expansion and lymphomagenesis. Here, we demonstrate that ablation of CD19 in LMP1/CD40 transgenic mice resulted in a severe loss and reduced life span of mature B-cells and completely abrogated development of B-cell lymphoma. CD19 is localized to lipid rafts and constitutively activated by the LMP1/CD40 fusion protein in B-cells. We provide evidence that the improved survival and malignant transformation of LMP1/CD40-expressing B-cells is dependent on activation of the MAPK Erk that is mediated through CD19 in a PI3K-dependent manner. Our data suggest that constitutively active CD40 is dependent on CD19 to transmit survival- and proliferation-signals. Moreover, we detected a similarly functioning prosurvival pathway involving phosphorylated CD19 and PI3K-dependent Erk-phosphorylation in human Diffuse Large B Cell Lymphoma cell lines. Our data provides evidence that CD19 plays an important role in transmitting survival and proliferation signals downstream of CD40 and therefore might be an interesting therapeutic target for the treatment of lymphoma undergoing chronic CD40-signaling. AU - Hojer, C. AU - Frankenberger, S. AU - Strobl, L.J. AU - Feicht, S. AU - Djermanovic, K. AU - Jagdhuber, F. AU - Hömig-Hölzel, C. AU - Ferch, U.* AU - Ruland, J.* AU - Rajewsky, K.* AU - Zimber-Strobl, U. C1 - 31633 C2 - 34621 CY - Philadelphia SP - 4318-4328 TI - B cell expansion and lymphomagenesis induced by chronic CD40 signaling is strictly dependent on CD19. JO - Cancer Res. VL - 74 IS - 16 PB - Amer Assoc Cancer Research PY - 2014 SN - 0008-5472 ER - TY - JOUR AU - Leinhäuser, I. AU - Höfig, I. AU - Anastasov, N. AU - Beuschlein, F.* AU - Mannelli, M.* AU - Atkinson, M.J. AU - Pellegata, N.S. C1 - 45030 C2 - 37151 CY - Philadelphia TI - The Bone morphogenic protein 7 (Bmp7) plays a pro-tumorigenic role in pheochromocytoma. JO - Cancer Res. VL - 74 IS - 19 PB - Amer Assoc Cancer Research PY - 2014 SN - 0008-5472 ER - TY - JOUR AU - Morscher, S.* AU - Driessen, W.H.P.* AU - Burton, N.C.* AU - Sardella, T.* AU - Razansky, D. AU - Ntziachristos, V. C1 - 45028 C2 - 37153 CY - Philadelphia TI - Assessing PK parameters using Dynamic Contrast Enhanced Multispectral Optoacoustic Tomography (DCE-MSOT). JO - Cancer Res. VL - 74 IS - 19 PB - Amer Assoc Cancer Research PY - 2014 SN - 0008-5472 ER - TY - JOUR AU - Morscher, S.* AU - Burton, N.C.* AU - Sardella, T.* AU - Razansky, D. AU - Ntziachristos, V. AU - Driessen, W.H.P.* C1 - 45029 C2 - 37152 CY - Philadelphia TI - Novel approaches for dynamic tumor microenvironment imaging by Multispectral Optoacoustic Tomography (MSOT). JO - Cancer Res. VL - 74 IS - 19 PB - Amer Assoc Cancer Research PY - 2014 SN - 0008-5472 ER - TY - JOUR AB - Although in vivo targeting of tumors using fluorescently-labeled probes has greatly gained in importance over the last few years, most of the clinically applied reagents lack tumor cell specificity. Our novel tumor cell-penetrating peptide-based probe (TPP) recognizes an epitope of Hsp70 that is exclusively present on the cell surface of a broad variety of human and mouse tumors and metastases, but not on normal tissues. Due to the rapid turn-over rate of membrane-Hsp70, fluorescently-labeled TPP is continuously internalized into syngeneic, spontaneous, chemically/genetically induced and xenograft tumors following intravenous administration, thereby enabling site-specific labeling of primary tumors and metastases. In contrast to the commercially available non-peptide small molecule alpha v beta3-integrin antagonist IntegriSense (trademark), TPP exhibits a significantly higher tumor-to-background contrast and stronger tumor-specific signal intensity in all tested tumor models. Moreover, in contrast to IntegriSense (trademark), TPP reliably differentiates between tumor cells and cells of the tumor microenvironment, such as tumor-associated macrophages and fibroblasts which were found to be membrane-Hsp70 negative. Therefore, TPP provides a useful tool for multimodal imaging of tumors and metastases that might help to improve our understanding of tumorigenesis and allow the establishment of improved diagnostic procedures and more accurate therapeutic monitoring. TPP might also be a promising platform for tumor-specific drug delivery and other Hsp70- based targeted therapies. AU - Stangl, S.* AU - Varga, J.* AU - Freysoldt, B.* AU - Trajkovic-Arsic, M.* AU - Siveke, J.T.* AU - Greten, F.R.* AU - Ntziachristos, V. AU - Multhoff, G. C1 - 32523 C2 - 35100 SP - 6903-6912 TI - Selective in vivo imaging of tumors with a tumor cell-specific Hsp70 peptide-based probe. JO - Cancer Res. VL - 74 IS - 23 PY - 2014 SN - 0008-5472 ER - TY - JOUR AU - Anastasov, N. AU - Höfig, I. AU - Lichtenberg, J.* AU - Stroebel, S.* AU - Salomon, M.* AU - Thirion, C.* AU - Kelm, J.* AU - Atkinson, M.J. C1 - 30829 C2 - 33906 CY - Philadelphia TI - Identification of compounds modifying radiation-therapy using a 3D-microtissue technology. JO - Cancer Res. VL - 73 IS - 8 PB - Amer Assoc Cancer Research PY - 2013 SN - 0008-5472 ER - TY - JOUR AU - Driessen, W.H.P.* AU - Burton, N.C.* AU - Sardella, T.* AU - Morscher, S.* AU - Razansky, D. AU - Ntziachristos, V. C1 - 30827 C2 - 33908 CY - Philadelphia TI - Multispectral Optoacoustic Tomography (MSOT) imaging and quantification of apoptosis in vivo. JO - Cancer Res. VL - 73 IS - 8 PB - Amer Assoc Cancer Research PY - 2013 SN - 0008-5472 ER - TY - JOUR AU - Ehrhardt, H. AU - Wachter, F. AU - Grunert, M. AU - Jeremias, I. C1 - 30825 C2 - 33910 CY - Philadelphia TI - TRAIL induces apoptosis preferentially in cell cycle arrested tumor cells, e.g., in tumor cells from children with acute lymphoblastic leukemia upon knockdown of cyclinE. JO - Cancer Res. VL - 73 IS - 8 PB - Amer Assoc Cancer Research PY - 2013 SN - 0008-5472 ER - TY - JOUR AB - Carbonic anhydrase XII (CA XII) is an membrane-tethered cell surface enzyme that is highly expressed on many human tumor cells. CA members in this class of exofacial molecules facilitate tumor metabolism by facilitating CO2 venting and intracellular pH regulation. Accordingly, inhibition of exofacial CAs has been proposed as a general therapeutic strategy to target cancer. The recent characterization of 6A10, the first CA XII-specific inhibitory monoclonal antibody, offered an opportunity to evaluate this strategy with regard to CA XII-mediated catalysis. Using functional assays, we showed that 6A10 inhibited exofacial CA activity in CA XII-expressing cancer cells. 6A10 reduced spheroid growth in vitro, under culture conditions where CA XII was active (i.e. alkaline pH) and where its catalytic activity was likely rate-limiting (i.e. restricted extracellular HCO3- supply). These in vitro results argued that the antibody exerted its growth-retarding effect by acting on the catalytic process, rather than on antigen binding per se. Notably, when administered in a mouse xenograft model of human cancer, 6A10 exerted a significant delay on tumor outgrowth. These results corroborate the notion that exofacial CA is critical for cancer cell physiology, and they establish the immunotherapeutic efficacy of targeting CA XII using an inhibitory antibody. AU - Gondi, G. AU - Mysliwietz, J. AU - Hulikova, A.* AU - Jen, J.P.* AU - Swietach, P.* AU - Kremmer, E. AU - Zeidler, R. C1 - 27725 C2 - 32796 SP - 6494-6503 TI - Antitumor efficacy of a monoclonal antibody that inhibits the activity of cancer-associated carbonic anhydrase XII. JO - Cancer Res. VL - 73 IS - 21 PB - Amer. Assoc. Cancer Research PY - 2013 SN - 0008-5472 ER - TY - JOUR AB - Germ-line mutations of the retinoblastoma gene (RB1) predispose to both sporadic and radiation-induced osteosarcoma, tumors characterized by high levels of genomic instability and activation of ALT (alternative lengthening of telomeres). Mice with haploinsufficiency of the Rb1 gene in the osteoblastic lineage reiterate the radiation-susceptibility to osteosarcoma seen in patients with germ-line RB1 mutations. We demonstrate that the susceptibility is accompanied by an increase in genomic instability resulting from Rb1-dependent telomere erosion. Radiation exposure did not accelerate the rate of telomere loss but amplified the genomic instability resulting from the dysfunctional telomeres. We propose that telomere maintenance is a non-canonical caretaker function of Rb1, and that a deficiency of Rb1 increases susceptibility to radiation-induced carcinogenesis by promoting chromosomal aberrations. AU - González-Vasconcellos, I.M. AU - Anastasov, N. AU - Sanli-Bonazzi, B. AU - Klymenko, O. AU - Atkinson, M.J. AU - Rosemann, M. C1 - 24919 C2 - 31716 SP - 4247-4255 TI - Rb1 haploinsufficiency promotes telomere attrition and radiation-induced genomic instability. JO - Cancer Res. VL - 73 IS - 14 PB - Amer. Assoc. Cancer Research PY - 2013 SN - 0008-5472 ER - TY - JOUR AB - Ewing sarcoma (ES), an osteolytic malignancy that mainly affects children and young adults, is characterized by early metastasis to lung and bone. In this study, we identified the pro-metastatic gene DKK2 as a highly overexpressed gene in ES compared to corresponding normal tissues. Using RNA interference, we demonstrated that DKK2 was critical for malignant cell outgrowth in vitro and in an orthotopic xenograft mouse model in vivo. Analysis of invasion potential in both settings revealed a strong correlation of DKK2 expression to ES invasiveness that may be mediated by the DKK effector matrix metalloproteinase 1 (MMP1). Further, gene expression analyses established the ability of DKK2 to differentially regulate genes such as CXCR4, PTHrP, RUNX2 and TGFβ1, that are associated with homing, invasion and growth of cancer cells in bone tissue as well as genes important for osteolysis, including HIF1α, JAG1, IL6 and VEGF. DKK2 promoted bone infiltration and osteolysis in vivo and further analyses defined DKK2 as a key factor in osteotropic malignancy. Interestingly, in ES cells DKK2 suppression simultaneously increased the potential for neuronal differentiation while decreasing chondrogenic and osteogenic differentiation. Our results provide strong evidence that DKK2 is a key player in ES invasion and osteolysis and also in the differential phenotype of ES cells. AU - Hauer, K.* AU - Calzada-Wack, J. AU - Steiger, K. AU - Grunewald, T.G.* AU - Baumhoer, D.* AU - Plehm, S.* AU - Buch, T.* AU - Prazeres da Costa, O.* AU - Esposito, I. AU - Burdach, S.* AU - Richter, G.H.* C1 - 11707 C2 - 30762 SP - 967-977 TI - DKK2 mediates osteolysis, invasiveness and metastatic spread in Ewing sarcoma. JO - Cancer Res. VL - 73 IS - 2 PB - American Assoc. of Cancer Research PY - 2013 SN - 0008-5472 ER - TY - JOUR AU - Nolte, E.* AU - Szczyrba, J.* AU - Hart, M.* AU - Doell, C.* AU - Wach, S.* AU - Taubert, H.* AU - Keck, B.* AU - Kremmer, E. AU - Stoehr, R.* AU - Hartmann, A.* AU - Wullich, B.* AU - Graesser, F.* C1 - 30828 C2 - 33907 CY - Philadelphia TI - miR-24 influences proliferation of prostate cancer cells in vitro via targeting ZNF217. JO - Cancer Res. VL - 73 IS - 8 PB - Amer Assoc Cancer Research PY - 2013 SN - 0008-5472 ER - TY - JOUR AU - Seibold, P.* AU - Buck, K.* AU - Vrieling, A.* AU - Johnson, T.* AU - Kaaks, R.* AU - Linseisen, J. AU - Heinz, J.* AU - Obi, N.* AU - Flesch-Janys, D.* AU - Chang-Claude, J.* C1 - 30824 C2 - 33911 CY - Philadelphia TI - Enterolactone levels and prognosis after invasive postmenopausal breast cancer: Potential effect modifiers. JO - Cancer Res. VL - 73 IS - 8 PB - Amer Assoc Cancer Research PY - 2013 SN - 0008-5472 ER - TY - JOUR AU - Zhao, Y.* AU - Schwarz, B.* AU - Zhao, L.* AU - Wang, Y.* AU - Tischmacher, A.* AU - Mysliwietz, J. AU - Ellwart, J.W. AU - Bao, Q.* AU - Niess, H.* AU - Modest, D.* AU - Camaj, P.* AU - Jauch, K.* AU - Nelson, P.* AU - Bruns, C.* C1 - 30826 C2 - 33909 CY - Philadelphia TI - Aspirin decreases side population cells by targeting the Wnt pathway in esophageal cancer cells in vitro and enhances the combination chemotherapeutic effect of 5-FU and cisplatin in vivo. JO - Cancer Res. VL - 73 IS - 8 PB - Amer Assoc Cancer Research PY - 2013 SN - 0008-5472 ER - TY - JOUR AB - A major goal of tumor immunotherapy is the induction of long-lasting systemic T-cell immunity. Bispecific antibodies (bsAbs) that lack the immunoglobulin Fc region confer T-cell-mediated killing of tumor cells but do not induce long-term memory. In contrast, trifunctional bsAbs comprise an appropriate Fc region and, therefore, not only recruit T cells but also accessory cells that bear activating Fc gamma receptors (Fc gamma R), providing additional T-cell-activating signals and securing presentation of tumor-derived antigens to T cells. In this study, we show that trifunctional bsAbs induce a polyvalent T-cell response and, therefore, a vaccination effect. Mice were treated with melanoma cells and with a trifunctional bsAb directed against the melanoma target antigen ganglioside GD2 in addition to murine CD3. The trifunctional bsAb activated dendritic cells and induced a systemic immune response that was not replicated by treatment with the F(ab')(2)-counterpart lacking the Fc region. Restimulation of spleen and lymph node cells in vitro yielded T-cell lines that specifically produced interferon-gamma in response to tumor. In addition, trifunctional bsAb-induced T cells recognized various specific peptides derived from melanoma-associated antigens. Moreover, these polyvalent responses proved to be tumor-suppressive and could not be induced by the corresponding bsF(ab')(2)-fragment. Taken together, our findings provide preclinical proof of concept that trifunctional bsAbs can induce tumor-specific T cells with defined antigen specificity. AU - Eissler, N. AU - Ruf, P.* AU - Mysliwietz, J. AU - Lindhofer, H.* AU - Mocikat, R. C1 - 8562 C2 - 30187 SP - 3958-3966 TI - Trifunctional bispecific antibodies induce tumor-specific T cells and elicit a vaccination effect. JO - Cancer Res. VL - 72 IS - 16 PB - Amer. Assoc. Cancer Research PY - 2012 SN - 0008-5472 ER - TY - JOUR AB - NFAT transcription factors control T-cell activation and function. Specifically, the transcription factor NFATc2 affects the regulation of cell differentiation and growth and plays a critical role in the development of colonic inflammation. Here, we used an experimental model of colitis-associated colorectal carcinoma to investigate the contribution of NFATc2 to the promotion of colonic tumors. Compared with wild-type animals that readily presented with multiple colon tumors, NFATc2-deficient mice were protected from tumor development. This observed decrease in colonic tumor progression was associated with reduced endoscopic inflammation, increased apoptosis of lamina propria T lymphocytes, and significantly reduced levels of the critical proin-flammatory cytokines interleukin (IL)-21 and IL-6. Administration of hyper IL-6 abrogated protection from tumor progression in NFATc2-knockout mice and restored tumor incidence to control levels. Taken together, our findings highlight a pivotal role for NFATc2 in the establishment of inflammation-associated colorectal tumors mediated by control of IL-6 expression. AU - Gerlach, K.* AU - Daniel, C. AU - Lehr, H.A.* AU - Nikolaev, A.* AU - Gerlach, T.* AU - Atreya, R.* AU - Rose-John, S.* AU - Neurath, M.F.* AU - Weigmann, B.* C1 - 10449 C2 - 30245 SP - 4340-4350 TI - Transcription factor NFATc2 controls the emergence of colon cancer associated with IL-6-dependent colitis. JO - Cancer Res. VL - 72 IS - 17 PB - Amer. Assoc. Cancer Research PY - 2012 SN - 0008-5472 ER - TY - JOUR AB - The prognosis of relapsed or refractory aggressive lymphoma is poor. The huge variety of currently evolving targeted treatment approaches would benefit from tools for early prediction of response or resistance. We used various ALK-positive anaplastic large cell lymphoma (ALCL) cell lines to evaluate two inhibitors, the HSP90 inhibitor NVP-AUY922, and the mTOR inhibitor everolimus, both of which have shown to interfere with ALK-dependent oncogenic signal transduction. Their therapeutic effect was determined in vitro by MTT assay, [18F]fluorodeoxyglucose- (FDG) and [18F]fluorothymidine- (FLT) uptake, and by biochemical analysis of ALK-induced signalling. Micro FDG- and FLT-PET imaging studies in immunodeficient mice bearing ALCL xenotransplants were performed with the cell lines SUDHL-1 and Karpas299 to assess early treatment response to NVP-AUY922 or everolimus in vivo. SUDHL-1 cells showed sensitivity to both inhibitors in vitro. Importantly, we detected a significant reduction of FLT-uptake in SUDHL-1 bearing animals using both inhibitors compared to baseline as early as 5 days after initiation of targeted therapy. Immunostaining showed a decrease in Ki-67 and an increase in cleaved caspase-3 staining. In contrast, FDG-uptake did not significantly decrease at early time points. Karpas299 xenotransplants, which are resistant to NVP-AUY922 and sensitive to everolimus treatment, showed an increase of mean FLT-uptake on day 2 after administration of NVP-AUY299, but a significant reduction in FLT-uptake upon everolimus treatment. In conclusion, we show that FLT- but not FDG-PET is able to predict response to treatment with specific inhibitors very early in the course of treatment and thus enables early prediction of treatment efficacy. AU - Li, Z.* AU - Graf, N.A.* AU - Herrmann, K.* AU - Jünger, A.* AU - Aichler, M. AU - Feuchtinger, A. AU - Baumgart, A.* AU - Walch, A.K. AU - Peschel, C.* AU - Schwaiger, M.* AU - Buck, A.* AU - Keller, U.* AU - Dechow, T.* C1 - 8450 C2 - 30122 SP - 5014-5024 TI - FLT-PET is superior to FDG-PET for very early response prediction in NPM-ALK-positive lymphoma treated with targeted therapy. JO - Cancer Res. VL - 72 IS - 19 PB - American Assoc. for Cancer Research PY - 2012 SN - 0008-5472 ER - TY - JOUR AB - Traditionally, the goal of nanoparticle-based chemotherapy has been to decrease normal tissue toxicity by improving drug specificity to tumors. The enhanced permeability and retention effect can permit passive accumulation into tumor interstitium. However, suboptimal delivery is achieved with most nanoparticles because of heterogeneities of vascular permeability, which limits nanoparticle penetration. Furthermore, slow drug release limits bioavailability. We developed a fast drug-releasing liposome triggered by local heat that has already shown substantial antitumor efficacy and is in human trials. Here, we show that thermally sensitive liposomes (Dox-TSL) release doxorubicin inside the tumor vasculature. Real-time confocal imaging of doxorubicin delivery to murine tumors in window chambers and histologic analysis of flank tumors illustrates that intravascular drug release increases free drug in the interstitial space. This increases both the time that tumor cells are exposed to maximum drug levels and the drug penetration distance, compared with free drug or traditional pegylated liposomes. These improvements in drug bioavailability establish a new paradigm in drug delivery: rapidly triggered drug release in the tumor bloodstream. Cancer Res; 72(21); 5566-75. (C)2012 AACR. AU - Manzoor, A.A.* AU - Lindner, L.H. AU - Landon, C.D.* AU - Park, J.Y.* AU - Simnick, A.J.* AU - Dreher, M.R.* AU - Das, S.* AU - Hanna, G.* AU - Park, W.* AU - Chilkoti, A.* AU - Koning, G.A.* AU - ten Hagen, T.L.M.* AU - Needham, D.* AU - Dewhirst, M.W.* C1 - 11262 C2 - 30592 SP - 5566-5575 TI - Overcoming limitations in nanoparticle drug delivery: Triggered, intravascular release to improve drug penetration into tumors. JO - Cancer Res. VL - 72 IS - 21 PB - Amer. Assoc. Cancer Research PY - 2012 SN - 0008-5472 ER - TY - JOUR AB - The estrogen receptor-α (ERα) determines the phenotype of breast cancers where it serves as a positive prognostic indicator. ERα is a well-established target for breast cancer therapy, but strategies to target its function remain of interest to address therapeutic resistance and further improve treatment. Recent findings indicate that proteasome inhibition can regulate estrogen-induced transcription, but how ERα function might be regulated was uncertain. In this study, we investigated the transcriptome-wide effects of the proteasome inhibitor bortezomib on estrogen-regulated transcription in MCF7 human breast cancer cells and showed that bortezomib caused a specific global decrease in estrogen-induced gene expression. This effect was specific because gene expression induced by the glucocorticoid receptor was unaffected by bortezomib. Surprisingly, we observed no changes in ERα recruitment or assembly of its transcriptional activation complex on ERα target genes. Instead, we found that proteasome inhibition caused a global decrease in histone H2B monoubiquitination (H2Bub1), leading to transcriptional elongation defects on estrogen target genes and to decreased chromatin dynamics overall. In confirming the functional significance of this link, we showed that RNA interference-mediated knockdown of the H2B ubiquitin ligase RNF40 decreased ERα-induced gene transcription. Surprisingly, RNF40 knockdown also supported estrogen-independent cell proliferation and activation of cell survival signaling pathways. Most importantly, we found that H2Bub1 levels decrease during tumor progression. H2Bub1 was abundant in normal mammary epithelium and benign breast tumors but absent in most malignant and metastatic breast cancers. Taken together, our findings show how ERα activity is blunted by bortezomib treatment as a result of reducing the downstream ubiquitin-dependent function of H2Bub1. In supporting a tumor suppressor role for H2Bub1 in breast cancer, our findings offer a rational basis to pursue H2Bub1-based therapies for future management of breast cancer. AU - Prenzel, T.* AU - Begus-Nahrmann, Y.* AU - Kramer, F.* AU - Hennion, M.* AU - Hsu, C.* AU - Gorsler, T.* AU - Hintermair, C. AU - Eick, D. AU - Kremmer, E. AU - Simons, M.* AU - Beissbarth, T.* AU - Johnsen, S.A.* C1 - 6589 C2 - 28951 SP - 5739-5753 TI - Estrogen-dependent gene transcription in human breast cancer cells relies upon proteasome-dependent monoubiquitination of histone H2B. JO - Cancer Res. VL - 71 IS - 17 PB - AACR PY - 2011 SN - 0008-5472 ER - TY - JOUR AB - Primary mediastinal B-cell lymphoma (PMBL) and classical Hodgkin lymphoma (cHL) share a frequent constitutive activation of JAK (Janus kinase)/STAT signaling pathway. Because of complex, nonlinear relations within the pathway, key dynamic properties remained to be identified to predict possible strategies for intervention. We report the development of dynamic pathway models based on quantitative data collected on signaling components of JAK/STAT pathway in two lymphoma-derived cell lines, MedB-1 and L1236, representative of PMBL and cHL, respectively. We show that the amounts of STAT5 and STAT6 are higher whereas those of SHP1 are lower in the two lymphoma cell lines than in normal B cells. Distinctively, L1236 cells harbor more JAK2 and less SHP1 molecules per cell than MedB-1 or control cells. In both lymphoma cell lines, we observe interleukin-13 (IL13)-induced activation of IL4 receptor α, JAK2, and STAT5, but not of STAT6. Genome-wide, 11 early and 16 sustained genes are upregulated by IL13 in both lymphoma cell lines. Specifically, the known STAT-inducible negative regulators CISH and SOCS3 are upregulated within 2 hours in MedB-1 but not in L1236 cells. On the basis of this detailed quantitative information, we established two mathematical models, MedB-1 and L1236 model, able to describe the respective experimental data. Most of the model parameters are identifiable and therefore the models are predictive. Sensitivity analysis of the model identifies six possible therapeutic targets able to reduce gene expression levels in L1236 cells and three in MedB-1. We experimentally confirm reduction in target gene expression in response to inhibition of STAT5 phosphorylation, thereby validating one of the predicted targets. Cancer Res; 71(3); 693-704. ©2010 AACR. AU - Raia, V.* AU - Schilling, M.* AU - Böhm, M.* AU - Hahn, B.* AU - Kowarsch, A. AU - Raue, A.* AU - Sticht, C.* AU - Bohl, S.* AU - Saile, M.* AU - Möller, P.* AU - Gretz, N.* AU - Timmer, J.* AU - Theis, F.J. AU - Lehmann, W.D.* AU - Lichtner, P.* AU - Klingmüller, U.* C1 - 2483 C2 - 28098 SP - 693-704 TI - Dynamic mathematical modeling of IL13-induced signaling in Hodgkin and primary mediastinal B-Cell lymphoma allows prediction of therapeutic targets. JO - Cancer Res. VL - 71 IS - 3 PB - American Assoc. for Cancer Research PY - 2011 SN - 0008-5472 ER - TY - JOUR AB - A-Raf kinase can inhibit apoptosis by binding to the proapoptotic mammalian sterile 20-like kinase (MST2). This function relies on expression of hnRNP H, which ensures the correct splicing of a-raf mRNA needed to produce full-length A-Raf protein. Here, we showed that expression of hnRNP H and production of full-length A-Raf is positively controlled by c-Myc. Low c-Myc reduces hnRNP H expression and switches a-raf splicing to produce A-Raf(short), a truncated protein. Importantly, A-Raf(short) fails to regulate MST2 but retains the Ras-binding domain such that it functions as a dominant negative mutant suppressing Ras activation and transformation. Human colon and head and neck cancers exhibit high hnRNP H and high c-Myc levels resulting in enhanced A-Raf expression and reduced expression of A-Raf(short). Conversely, in normal cells and tissues in which c-Myc and hnRNP H are low, A-Raf(short) suppresses extracellular signal regulated kinase activation such that it may act as a safeguard against oncogenic transformation. Our findings offered a new paradigm to understand how c-Myc coordinates diverse cell functions by directly affecting alternate splicing of key signaling components. AU - Rauch, J.* AU - Moran-Jones, K.* AU - Albrecht, V.* AU - Schwarzl, T.* AU - Hunter, K.* AU - Gires, O. AU - Kolch, W. C1 - 6390 C2 - 28600 SP - 4664-4674 TI - c-Myc regulates RNA splicing of the A-raf kinase and its activation of the ERK pathway. JO - Cancer Res. VL - 71 IS - 3 PB - Amer Assoc Cancer Research PY - 2011 SN - 0008-5472 ER - TY - JOUR AB - Tumor cells generate substantial amounts of reactive oxygen species (ROS), engendering the need to maintain high levels of antioxidants such as thioredoxin (Trx)- and glutathione (GSH)-dependent enzymes. Exacerbating oxidative stress by specifically inhibiting these types of ROS-scavenging enzymes has emerged as a promising chemotherapeutic strategy to kill tumor cells. However, potential redundancies among the various antioxidant systems may constrain this simple approach. Trx1 and thioredoxin reductase 1 (Txnrd1) are up-regulated in numerous cancers, and Txnrd1 has been reported to be indispensable for tumorigenesis. However, we report here that genetic ablation of Txnrd1 has no apparent effect on tumor cell behavior based on similar proliferative, clonogenic, and tumorigenic potential. This finding reflects widespread redundancies between the Trx- and GSH-dependent systems based on evidence of a bypass to Txnrd1 deficiency by compensatory upregulation of GSH-metabolizing enzymes. Because the survival and growth of Txnrd1-deficient tumors were strictly dependent on a functional GSH system, Txnrd1(-/-) tumors were highly susceptible to experimental GSH depletion in vitro and in vivo. Thus, our findings establish for the first time that a concomitant inhibition of the two major antioxidant systems is highly effective in killing tumor, highlighting a promising strategy to combat cancer. AU - Mandal, P.K. AU - Schneider, M.* AU - Kölle, P.* AU - Kuhlencordt, P.* AU - Förster, H. AU - Beck, H.* AU - Bornkamm, G.W. AU - Conrad, M. C1 - 5689 C2 - 27971 CY - Philadelphia SP - 9505-9514 TI - Loss of thioredoxin reductase 1 renders tumors highly susceptible to pharmacologic glutathione deprivation. JO - Cancer Res. VL - 70 IS - 22 PB - Amer. Assoc. Cancer Research PY - 2010 SN - 0008-5472 ER - TY - JOUR AB - A-Raf belongs to the family of oncogenic Raf kinases that are involved in mitogenic signaling by activating the mitogen-activated protein (MAP)/extracellular signal-regulated kinase (ERK) kinase (MEK)-ERK pathway. Low kinase activity of A-Raf toward MEK suggested that A-Raf might have alternative functions. Here, we show that A-Raf prevents cancer cell apoptosis contingent on the expression of the heterogeneous nuclear ribonucleoprotein H (hnRNP H) splice factor, which is required for the correct transcription and expression of a-raf. Apoptosis was prevented by A-Raf through sequestration and inactivation of the proapoptotic MST2 kinase. Small interfering RNA-mediated knockdown of hnRNP H or A-Raf resulted in MST2-dependent apoptosis. In contrast, enforced expression of either hnRNP H or A-Raf partially counteracted apoptosis induced by etoposide. In vivo expression studies of colon specimens corroborated the overexpression of hnRNP H in malignant tissues and its correlation with A-Raf levels. Our findings define a novel mechanism that is usurped in tumor cells to escape naturally imposed apoptotic signals. AU - Rauch, J.* AU - O'Neill, E.* AU - Mack, B.* AU - Matthias, C.* AU - Münz, M. AU - Kolch, W.* AU - Gires, O. C1 - 5852 C2 - 27690 SP - 1679-1688 TI - Heterogeneous nuclear ribonucleoprotein H blocks MST2-mediated apoptosis in cancer cells by regulating A-Raf transcription. JO - Cancer Res. VL - 70 IS - 4 PB - American Assoc. for Cancer Research PY - 2010 SN - 0008-5472 ER - TY - JOUR AB - Carcinogenesis is the result of mutations and subsequent clonal expansions of mutated, selectively advantageous cells. To investigate the relative contributions of mutation versus cell selection in tumorigenesis, we compared two mathematical models of carcinogenesis in two different cancer types: lung and colon. One approach is based on a population genetics model, the Wright-Fisher process, whereas the other approach is the two-stage clonal expansion model. We compared the dynamics of tumorigenesis predicted by the two models in terms of the time period until the first malignant cell appears, which will subsequently form a tumor. The mean waiting time to cancer has been calculated approximately for the evolutionary colon cancer model. Here, we derive new analytic approximations to the median waiting time for the two-stage lung cancer model and for a multistage approximation to the Wright-Fisher process. Both equations show that the waiting time to cancer is dominated by the selective advantage per mutation and the net clonal expansion rate, respectively, whereas the mutation rate has less effect. Our comparisons support the idea that the main driving force in lung and colon carcinogenesis is Darwinian cell selection. AU - Schöllnberger, H. AU - Beerenwinkel, N.* AU - Hoogenveen, R.* AU - Vineis, P.* C1 - 5601 C2 - 27515 SP - 6797-6803 TI - Cell selection as driving force in lung and colon carcinogenesis. JO - Cancer Res. VL - 70 IS - 17 PB - American Assoc. for Cancer Research PY - 2010 SN - 0008-5472 ER - TY - JOUR AU - Torsvik, A.* AU - Røsland, G.V.* AU - Svendsen, A.* AU - Molven, A.* AU - Immervoll, H.* AU - McCormack, E.* AU - Lønning, P.E.* AU - Primon, M.* AU - Sobala, E.* AU - Tonn, J.C.* AU - Goldbrunner, R.* AU - Schichor, C.* AU - Mysliwietz, J. AU - Lah, T.T.* AU - Motaln, H.* AU - Knappskog, S.* AU - Bjerkvig, R.* C1 - 5311 C2 - 27585 SP - 6393-6396 TI - Spontaneous malignant transformation of human mesenchymal stem cells reflects cross-contamination: Putting the research field on track - letter. JO - Cancer Res. VL - 70 IS - 15 PB - American Assocation Cancer Research PY - 2010 SN - 0008-5472 ER - TY - JOUR AB - Initially discovered as a dominant antigen on colon carcinomas, the epithelial cell adhesion molecule (EpCAM) was considered a mere cell adhesion molecule and reliable surface-binding site for therapeutic antibodies. Recent findings can better explain the relevance of EpCAM's high-level expression on human cancers and cancer propagating cells, and its negative prognostic potential for survival of patients with certain cancers. EpCAM has oncogenic potential and is activated by release of its intracellular domain, which can signal into the cell nucleus by engagement of elements of the wnt pathway. AU - Münz, M.* AU - Baeuerle, P.A.* AU - Gires, O. C1 - 544 C2 - 27144 SP - 5627-5629 TI - The emerging role of EpCAM in cancer and stem cell signaling. JO - Cancer Res. VL - 69 IS - 14 PB - Amer Assoc Cancer Research PY - 2009 SN - 0008-5472 ER - TY - JOUR AB - Anaplastic thyroid carcinoma (ATC) is one of the most aggressive and chemoresistant cancers. The serine/threonine kinase Polo-like kinase 1 (PLK1), a key regulator of multiple steps during mitotic progression, is highly expressed in ATC. Here, we used the BI 2536 PLK1 inhibitor on ATC and nontransformed thyroid follicular cell tines. Our data show that ATC cells are addicted to high levels of PLK1 activity for proliferation, survival, anchorage-independent growth, and tumorigenicity. On treatment with nanomolar doses of BI 2536, ATC cells progressed normally through S phase but died thereafter, directly from mitotic arrest. Immunofluorescence microscopy, immunoblot, and flow cytometry analysis showed that, on PLK1 blockade, ATC cells arrested in prometaphase with a 4N DNA content. Treated ATC cells accumulated phosphohistone H3 and displayed characteristic mitotic (Polo) spindle aberrations. Nontransformed thyroid cells were 3.2- to 18.4-fold less susceptible to BI 2536-induced cell cycle effects compared with ATC cells. These findings identify PLK1 as a promising target for the molecular therapy of ATC. AU - Nappi, T.C.* AU - Salerno, P.* AU - Zitzelsberger, H. AU - Carlomagno, F.* AU - Salvatore, G.* AU - Santoro, M.* C1 - 1515 C2 - 26963 SP - 1916-1923 TI - Identification of Polo-like kinase 1 as a potential therapeutic target in anaplastic thyroid carcinoma. JO - Cancer Res. VL - 69 IS - 5 PB - Amer Assoc Cancer Research PY - 2009 SN - 0008-5472 ER - TY - JOUR AB - Human mesenchymal stem cells (hMSC) aid in tissue maintenance and repair by differentiating into specialized cell types. Due to this ability, hMSC are currently being evaluated for cell-based therapies of tissue injury and degenerative diseases. However, extensive expansion ex vivo is a prerequisite to obtain the cell numbers required for human cell-based therapy protocols. Recent studies indicate that hMSC may contribute to cancer development and progression either by acting as cancer-initiating cells or through interactions with stromal elements. If spontaneous transformation ex vivo occurs, this may jeopardize the use of hMSC as therapeutic tools. Whereas murine MSC readily undergo spontaneous transformation, there are conflicting reports about spontaneous transformation of hMSC. We have addressed this controversy in a two-center study by growing bone marrow-derived hMSC in long-term cultures (5-106 weeks). We report for the first time spontaneous malignant transformation to occur in 45.8% (11 of 24) of these cultures. In comparison with hMSC, the transformed mesenchymal cells (TMC) showed a significantly increased proliferation rate and altered morphology and phenotype. In contrast to hMSC, TMC grew well in soft agar assays and were unable to undergo complete differentiation. Importantly, TMC were highly tumorigenic, causing multiple fast-growing lung deposits when injected into immunodeficient mice. We conclude that spontaneous malignant transformation may represent a biohazard in long-term ex vivo expansion of hMSC. On the other hand, this spontaneous transformation process may represent a unique model for studying molecular pathways initiating malignant transformation of hMSC. AU - Røsland, G.V.* AU - Svendsen, A.* AU - Torsvik, A.* AU - Sobala, E.* AU - McCormack, E.* AU - Immervoll, H.* AU - Mysliwietz, J. AU - Tonn, J.C.* AU - Goldbrunner, R.* AU - Lonning, P.E.* AU - Bjerkvig, R.* AU - Schichor, C.* C1 - 978 C2 - 26607 SP - 5331-5339 TI - Long-term cultures of bone marrow-derived human mesenchymal stem cells frequently undergo spontaneous malignant transformation. JO - Cancer Res. VL - 69 IS - 13 PB - Amer Assoc Cancer Research PY - 2009 SN - 0008-5472 ER - TY - JOUR AB - Mutations of the tumor suppressor E-cadherin and overexpression of the receptor tyrosine kinase epidermal growth factor receptor (EGFR) are among the most frequent genetic alterations associated with diffuse-type gastric carcinoma. Accumulating evidence suggests a functional relationship between E-cadherin and EGFR that regulates both proteins. We report that somatic mutation of E-cadherin is associated with increased activation of EGFR followed by enhanced recruitment of the downstream acting signaling components growth factor receptor binding protein 2 and Shc, and activation of Ras. Reduced complex formation of mutant E-cadherin - with an in frame deletion of exon 8 in the extracellular domain resulting in reduced adhesion and increased motility - with EGFR was observed compared with wild-type E-cadherin. We conclude that reduced binding of mutant E-cadherin to EGFR in a multicomponent complex or reduced stability of the complex may enhance EGFR surface motility, thereby facilitating EGFR dimerization and activation. Furthermore, reduced surface localization due to enhanced internalization of mutant E-cadherin compared with the wild-type protein was observed. The internalization of EGFR was decreased in response to epidermal growth factor stimulation in cells expressing mutant E-cadherin, suggesting that mutation of E-cadherin also influences the endocytosis of EGFR. Moreover, we show increased activation of EGFR in gastric carcinoma samples with mutant E-cadherin lacking exons 8 or 9. In summary, we describe activation of EGFR by mutant E-cadherin as a novel mechanism in tumor cells that explains the enhanced motility of tumor cells in the presence of an extracellular mutation of E-cadherin. AU - Bremm, A.* AU - Walch, A.K. AU - Fuchs, M.* AU - Mages, J.* AU - Duyster, J.* AU - Keller, G.* AU - Hermannstädter, C. AU - Becker, K.F.* AU - Rauser, S. AU - Langer, R.* AU - von, Weyhern, C.H.* AU - Höfler, H. AU - Luber, B.* C1 - 3389 C2 - 25174 SP - 707-714 TI - Enhanced activation of epidermal growth factor receptor caused by tumor-derived E-cadherin mutations. JO - Cancer Res. VL - 68 IS - 3 PB - AACR PY - 2008 SN - 0008-5472 ER - TY - JOUR AB - Regulation of chromatin is an important aspect of controlling promoter activity and gene expression. Posttranslational modifications of core histones allow proteins associated with gene transcription to access chromatin. Closely associated with promoters of actively transcribed genes, trimethylation of histone H3 at lysine 4 (H3K4me3) is a core histone mark set by several protein complexes. Some of these protein complexes contain the trithorax protein ASH2 combined with the MLL oncoproteins. We identified human ASH2 in a complex with the oncoprotein MYC. This finding, together with the observation that hASH2 interacts with MLL, led us to test whether hASH2 itself is involved in transformation. We observed that hASH2 cooperates with Ha-RAS to transform primary rat embryo fibroblasts (REF). Furthermore, transformation of REFs by MYC and Ha-RAS required the presence of rAsh2. In an animal model, the hASH2/Ha-RAS-transformed REFs formed rapidly growing tumors characteristic of fibrosarcomas that, compared with tumors derived from MYC/Ha-RAS transformed cells, were poorly differentiated. This finding suggests that ASH2 functions as an oncoprotein. Although hASH2 expression at the mRNA level was generally not deregulated, hASH2 protein expression was increased in most human tumors and tumor cell lines. In addition, knockdown of hASH2 inhibited tumor cell proliferation. Taken together, these observations define hASH2 as a novel oncoprotein. AU - Luscher-Firzlaff, J.* AU - Gawlista, I.* AU - Vervoorts, J.* AU - Kapelle, K.* AU - Braunschweig, T.* AU - Walsemann, G.* AU - Rodgarkia-Schamberger, C.* AU - Schuchlautz, H.* AU - Dreschers, S.* AU - Kremmer, E. AU - Lilischkis, R.* AU - Cerni, C.* AU - Wellmann, A.* AU - Lüscher, B.* C1 - 1435 C2 - 25698 SP - 749-758 TI - The human trithorax protein hASH2 functions as an oncoprotein. JO - Cancer Res. VL - 68 IS - 3 PB - AACR PY - 2008 SN - 0008-5472 ER - TY - JOUR AB - Formaldehyde is an aliphatic monoaldehyde and is a highly reactive environmental human carcinogen. Whereas humans are continuously exposed to exogenous formaldehyde, this reactive aldehyde is a naturally occurring biological compound that is present in human plasma at concentrations ranging from 13 to 97 micromol/L. It has been well documented that DNA-protein crosslinks (DPC) likely play an important role with regard to the genotoxicity and carcinogenicity of formaldehyde. However, little is known about which DNA damage response pathways are essential for cells to counteract formaldehyde. In the present study, we first assessed the DNA damage response to plasma levels of formaldehyde using chicken DT40 cells with targeted mutations in various DNA repair genes. Here, we show that the hypersensitivity to formaldehyde is detected in DT40 mutants deficient in the BRCA/FANC pathway, homologous recombination, or translesion DNA synthesis. In addition, FANCD2-deficient DT40 cells are hypersensitive to acetaldehyde, but not to acrolein, crotonaldehyde, glyoxal, and methylglyoxal. Human cells deficient in FANCC and FANCG are also hypersensitive to plasma levels of formaldehyde. These results indicate that the BRCA/FANC pathway is essential to counteract DPCs caused by aliphatic monoaldehydes. Based on the results obtained in the present study, we are currently proposing that endogenous formaldehyde might have an effect on highly proliferating cells, such as bone marrow cells, as well as an etiology of cancer in Fanconi anemia patients. AU - Ridpath, J.R.* AU - Nakamura, A.* AU - Tano, K.* AU - Luke, A.M.* AU - Sonoda, E.* AU - Arakawa, H. AU - Buerstedde, J.M. AU - Gillespie, D.A.* AU - Sale, J.E.* AU - Yamazoe, M.* AU - Bishop, D.K.* AU - Takata, M.* AU - Takeda, S.* AU - Watanabe, M.* AU - Swenberg, J.A.* AU - Nakamura, J.* C1 - 3056 C2 - 24992 SP - 11117-11122 TI - Cells deficient in the FANC/BRCA pathway are hypersensitive to plasma levels of formaldehyde. JO - Cancer Res. VL - 67 IS - 23 PB - AACR PY - 2007 SN - 0008-5472 ER - TY - JOUR AB - Histone deacetylases (HDAC) reverse the acetylation of histone and nonhistone proteins and thereby modulate chromatin structure and function of nonhistone proteins. Many tumor cell lines and experimental tumors respond to HDAC inhibition. To assess the role of an individual HDAC isoenzyme in physiology and tumor development, HDAC2-mutant mice were generated from a gene trap embryonic stem cell clone. These mice express a catalytically inactive fusion protein of the NH(2)-terminal part of HDAC2 and beta-galactosidase, which fails to integrate into corepressor complexes with mSin3B. They are the first class 1 HDAC mutant mice that are viable although they are approximately 25% smaller than their littermates. Cell number and thickness of intestinal mucosa are reduced. Mutant embryonic fibroblasts fail to respond to insulin-like growth factor I (IGF) by the IGF-I-induced increase in cell number observed in wild-type cells. These data suggest a novel link between HDACs and IGF-I-dependent responses. Crossing of HDAC2-mutant with tumor-prone APC(min) mice revealed tumor rates that are lower in HDAC2-deficient mice by 10% to 100% depending on segment of the gut and sex of the mice. These mice provide evidence that the key functions of HDAC2, although not essential for survival of the organism, play a rate-limiting role for tumor development in vivo. AU - Zimmermann, S. AU - Kiefer, F. AU - Prudenziati, M.* AU - Spiller, C. AU - Hansen, J. AU - Floß, T. AU - Wurst, W. AU - Minucci, S.* AU - Göttlicher, M. C1 - 2214 C2 - 24859 SP - 9047-9054 TI - Reduced body size and decreased intestinal tumor rates in HDAC2-mutant mice. JO - Cancer Res. VL - 67 IS - 19 PB - AACR PY - 2007 SN - 0008-5472 ER - TY - JOUR AB - Allogeneic cell therapy as a means to break immunotolerance to solid tumors is increasingly used for cancer treatment. To investigate cellular alloimmune responses in a human tumor model, primary cultures were established from renal cell carcinoma (RCC) tissues of 56 patients. In three patients with stable RCC line and human leukocyte antigen (HLA)-identical sibling donor available, allogeneic and autologous RCC reactivities were compared using mixed lymphocyte/tumor cell cultures (MLTC). Responding lymphocytes were exclusively CD8(+) T cells, whereas CD4(+) T cells or natural killer cells were never observed. Sibling MLTC populations showed higher proliferative and cytolytic antitumor responses compared with their autologous counterparts. The allo-MLTC responders originated from the CD8(+) CD62L(high)(+) peripheral blood subpopulation containing naive precursor and central memory T cells. Limiting dilution cloning failed to establish CTL clones from autologous MLTCs or tumor-infiltrating lymphocytes. In contrast, a broad panel of RCC-reactive CTL clones was expanded from each allogeneic MLTC. These sibling CTL clones either recognized exclusively the original RCC tumor line or cross-reacted with nonmalignant kidney cells of patient origin. A minority of CTL clones also recognized patient-derived hematopoietic cells or other allogeneic tumor targets. The MHC-restricting alleles for RCC-reactive sibling CTL clones included HLA-A2, HLA-A3, HLA-A11, HLA-A24, and HLA-B7. In one sibling donor-RCC pair, strongly proliferative CD3(+)CD16(+)CD57(+) CTL clones with non-HLA-restricted antitumor reactivity were established. Our results show superior tumor-reactive CD8 responses of matched allogeneic compared with autologous T cells. These data encourage the generation of antitumor T-cell products from HLA-identical siblings and their potential use in adoptive immunotherapy of metastatic RCC patients. AU - Kausche, S.* AU - Wehler, T.* AU - Schnürer, E.* AU - Lennerz, V.* AU - Brenner, W.* AU - Melchior, S.* AU - Gröne, M.* AU - Nonn, M.* AU - Strand, S.* AU - Meyer, R.* AU - Ranieri, E.* AU - Huber, C.* AU - Falk, C.S. AU - Herr, W.* C1 - 2566 C2 - 24830 SP - 11447-11454 TI - Superior antitumor in vitro responses of allogeneic matched sibling compared with autologous patient CD8plus T cells. JO - Cancer Res. VL - 66 IS - 23 PB - AACR PY - 2006 SN - 0008-5472 ER - TY - JOUR AB - Cross-linking agents that induce DNA interstrand cross-links (ICL) are widely used in anticancer chemotherapy. Yeast genetic studies show that nucleotide excision repair (NER), Rad6/Rad18-dependent postreplication repair, homologous recombination, and cell cycle checkpoint pathway are involved in ICL repair. To study the contribution of DNA damage response pathways in tolerance to cross-linking agents in vertebrates, we made a panel of gene-disrupted clones from chicken DT40 cells, each defective in a particular DNA repair or checkpoint pathway, and measured the sensitivities to cross-linking agents, including cis-diamminedichloroplatinum (II) (cisplatin), mitomycin C, and melphalan. We found that cells harboring defects in translesion DNA synthesis (TLS), Fanconi anemia complementation groups (FANC), or homologous recombination displayed marked hypersensitivity to all the cross-linking agents, whereas NER seemed to play only a minor role. This effect of replication-dependent repair pathways is distinctively different from the situation in yeast, where NER seems to play a major role in dealing with ICL. Cells deficient in Rev3, the catalytic subunit of TLS polymerase Polζ, showed the highest sensitivity to cisplatin followed by fanc-c. Furthermore, epistasis analysis revealed that these two mutants work in the same pathway. Our genetic comprehensive study reveals a critical role for DNA repair pathways that release DNA replication block at ICLs in cellular tolerance to cross-linking agents and could be directly exploited in designing an effective chemotherapy. AU - Nojima, K.* AU - Arakawa, H. AU - Buerstedde, J.-M. C1 - 4589 C2 - 23208 SP - 11704-11711 TI - Multiple repair pathways mediate tolerance to chemotherapeutic cross-linking agents in vertebrate cells. JO - Cancer Res. VL - 65 IS - 24 PY - 2005 SN - 0008-5472 ER - TY - JOUR AB - A phase I clinical trial with granulocyte-macrophage colony-stimulating factor tumor cell vaccines in patients with metastatic renal cell carcinoma (RCC) showed immune cell infiltration at vaccine sites and delayed-type hypersensitivity (DTH) responses to autologous tumor cells indicative of T-cell immunity. To further characterize RCC T-cell responses and identify relevant RCC-associated antigens, we did a detailed analysis of CD8+ T-cell responses in two vaccinated RCC patients who generated the greatest magnitude of DTH response and also displayed a strong clinical response to vaccination (>90% reduction in metastatic tumor volume). Three separate CD8+ T-cell lines (and subsequent derived clones) derived from patient 24 recognized distinct RCC-associated antigens. One recognized a shared HLA-A*0201-restricted antigen expressed by both renal cancer cells and normal kidney cells. This recognition pattern correlated with a positive DTH test to normal kidney cells despite no evidence of impairment of renal function by the patient's remaining kidney after vaccination. A second line recognized a shared HLA-C7-restricted antigen that was IFN-γ inducible. A third line recognized a unique HLA-A*0101-restricted RCC antigen derived from a mutated KIAA1440 gene specific to the tumor. In addition, two independent CTL lines and three clones were also generated from patient 26 and they recognized autologous tumor cells restricted through HLA-A*0205, HLA-A/B/C, and HLA-B/C. These results show that paracrine granulocyte-macrophage colony-stimulating factor tumor vaccines may generate a diverse repertoire of tumor-reactive CD8+ T-cell responses and emphasize the importance of polyvalency in the design of cancer immunotherapies. AU - Zhou, X.* AU - Jun, D.Y.* AU - Thomas, A.M.* AU - Huang, X.* AU - Huang, L.-Q.* AU - Mautner, J. AU - Mo, W.* AU - Robbins, P.F.* AU - Pardoll, D.M.* AU - Jaffee, E.M.* C1 - 2534 C2 - 23442 SP - 1079-1088 TI - Diverse CD8+ T-cell responses to renal cell carcinoma antigens in patients treated with an autologous granulocyte-macrophage colony-stimulating factor gene-transduced renal tumor cell vaccine. JO - Cancer Res. VL - 65 IS - 3 PY - 2005 SN - 0008-5472 ER - TY - JOUR AB - Mutator phenotypes, a common and largely unexplained attribute of human cancer, might be better understood in mouse tumors containing reporter genes for accurate mutation enumeration and analysis. Previous work on peritoneal plasmacytomas (PCTs) in mice suggested that PCTs have a mutator phenotype caused by Myc-deregulating chromosomal translocations and/or phagocyte-induced mutagenesis due to chronic inflammation. To investigate this hypothesis, we generated PCTs that harbored the transgenic shuttle vector, pUR288, with a lacZ reporter gene for the assessment of mutations in vivo. PCTs exhibited a 5.5 times higher mutant frequency in lacZ (40.3 +/- 5.1 x 10(-5)) than in normal B cells (7.36 +/- 0.77 x 10(-5)), demonstrating that the tumors exhibit the phenotype of increased mutability. Studies on lacZ mutant frequency in serially transplanted PCTs and phagocyte-induced lacZ mutations in B cells in vitro indicated that mutant levels in tumors are not determined by exogenous damage inflicted by inflammatory cells. In vitro studies with a newly developed transgenic model of inducible Myc expression (Tet-off/MYC) showed that deregulated Myc sensitizes B cells to chemically induced mutations, but does not cause, on its own, mutations in lacZ. These findings suggested that the hypermutability of PCT is governed mainly by intrinsic features of tumor cells, not by deregulated Myc or chronic inflammation. AU - Felix, K.* AU - Polack, A. AU - Pretsch, W. AU - Jackson, S.H.* AU - Feigenbaum, L.* AU - Bornkamm, G.W. AU - Janz, S.* C1 - 1757 C2 - 21686 SP - 530-537 TI - Moderate Hypermutability of a Transgenic lacZ Reporter Gene in Myc-Dependent Inflammation-Induced Plasma Cell Tumors in Mice. JO - Cancer Res. VL - 64 IS - 2 PB - AACR PY - 2004 SN - 0008-5472 ER - TY - JOUR AB - The role of the micronutrient, selenium, in human cancers associated with chronic inflammations and persistent infections is poorly understood. Peritoneal plasmacytomas (PCTs) in strain BALB/c (C), the premier experimental model of inflammation-dependent plasma cell transformation in mice, may afford an opportunity to gain additional insights into the significance of selenium in neoplastic development. Here, we report that selenium-depleted C mice (n = 32) maintained on a torula-based low-selenium diet (5-8 mug of selenium/kg) were totally refractory to pristane induction of PCT. In contrast, 11 of 26 (42.3%) control mice maintained on a selenium adequate torula diet (300 mug of selenium/kg) and 15 of 40 (37.5%) control mice fed standard Purina chow (440 mug of selenium/kg) developed PCT by 275 days postpristane. Abrogation of PCT was caused in part by the striking inhibition of the formation of the inflammatory tissue in which PCT develop (pristane granuloma). This was associated with the reduced responsiveness of selenium-deficient inflammatory cells (monocytes and neutrophils) to chemoattractants, such as thioredoxin and chemokines. Selenium-deficient C mice exhibited little evidence of disturbed redox homeostasis and increased mutant frequency of a transgenic lacZ reporter gene in vivo. These findings implicate selenium, via the selenoproteins, in the promotion of inflammation-induced PCT and suggest that small drug inhibitors of selenoproteins might be useful for preventing human cancers linked with chronic inflammations and persistent infections. AU - Felix, K.* AU - Gerstmeier, S.* AU - Kyriakopoulos, A.* AU - Dong, H.-F. AU - Eckhaus, M.* AU - Behne, D.* AU - Bornkamm, G.W. AU - Janz, S.* C1 - 3482 C2 - 21760 SP - 2910-2917 TI - Selenium deficiency abrogates inflammation-dependent plasma cell tumors in mice. JO - Cancer Res. VL - 64 IS - 8 PY - 2004 SN - 0008-5472 ER - TY - JOUR AB - Age is the largest single risk factor for the development of cancer in mammals. Age-associated chromosomal changes, such as aneuploidy and telomere erosion, may be vitally involved in the initial steps of tumorigenesis. However, changes in gene expression specific for increased aneuploidy with age have not yet been characterized. Here, we address these questions by using a panel of fibroblast cell lines and lymphocyte cultures from young and old age groups. Oligonucleotide microarrays were used to characterize the expression of 14,500 genes. We measured telomere length and analyzed chromosome copy number changes and structural rearrangements by multicolor interphase fluorescence in situ hybridization and 7-fluorochrome multiplex fluorescence in situ hybridization, and we tried to show a relationship between gene expression patterns and chromosomal changes. These analyses revealed a number of genes involved in both the cell cycle and proliferation that are differently expressed in aged cells. More importantly, our data show an association between age-related aneuploidy and the gene expression level of genes involved in centromere and kinetochore function and in the microtubule and spindle assembly apparatus. To verify that some of these genes may also be involved in tumorigenesis, we compared the expression of these genes in chromosomally stable microsatellite instability and chromosomally unstable chromosomal instability colorectal tumor cell lines. Three genes (Notch2, H2AFY2, and CDC5L) showed similar expression differences between microsatellite instability and chromosomal instability cell lines as observed between the young and old cell cultures suggesting that they may play a role in tumorigenesis. AU - Geigl, J.B.* AU - Langer, S.* AU - Barwisch, S.* AU - Pfleghaar, K.* AU - Lederer, G.* AU - Speicher, M.R. C1 - 2358 C2 - 22266 SP - 8550-8557 TI - Analysis of gene expression patterns and chromosomal associated with aging. JO - Cancer Res. VL - 64 IS - 23 PY - 2004 SN - 0008-5472 ER - TY - JOUR AB - Because of its amplification and/or overexpression in many human tumors, the HER-2/neu proto-oncogene represents an attractive target for T-cell-mediated vaccination strategies. However, overexpression of oncogenes is often associated with defective expression of components of the MHC class I antigen-processing machinery (APM), thereby resulting in an immune escape phenotype of oncogene-transformed cells. To determine whether HER-2/neu influences the MHC class I antigen-processing pathway, the expression pattern of different APM components was examined in murine in vitro models of constitutive and tetracycline-controlled HER-2/neu expression. In comparison with HER-2/neu− control cells, HER-2/neu+ fibroblasts exhibit reduced levels of MHC class I surface antigens that were associated with impaired expression and/or function of the peptide transporter associated with antigen processing, the proteasome subunits low molecular weight protein 2 and low molecular weight protein 10, the proteasome activators PA28α and PA28β, and tapasin. These APM abnormalities resulted in reduced sensitivity to lysis by CTLs. The HER-2/neu-mediated immune escape phenotype could be corrected by IFN-γ treatment. The clinical relevance of this finding was supported by an inverse correlation between HER-2/neu and the peptide transporter associated with antigen-processing protein expression as determined by immunhistochemical analysis of a series of HER-2/neu− and HER-2/neu+ breast cancer specimens. Thus, a functional link between deficient APM component expression and HER-2/neu overexpression is proposed that might influence the design of HER-2/neu-targeted T-cell-based immunotherapeutic strategies. AU - Hermann, F.* AU - Lehr, H.-A.* AU - Drexler, I. AU - Sutter, G. AU - Hengstler, J.* AU - Wollscheid, U.* AU - Seliger, B.* C1 - 1338 C2 - 22324 SP - 215-220 TI - HER-2/neu-mediated regulation of components of the MHC class I antigen-processing pathway. JO - Cancer Res. VL - 64 IS - 1 PY - 2004 SN - 0008-5472 ER - TY - JOUR AB - Loss-of-function mutations in Patched (Ptch1) are implicated in constitutive activation of the Sonic hedgehog pathway in human basal cell carcinomas (BCCs), and inherited Ptch1 mutations underlie basal cell nevus syndrome in which a typical feature is multiple BCC occurring with greater incidence in portals of radiotherapy. Mice in which one copy of Ptch1 is inactivated show increased susceptibility to spontaneous tumor development and hypersensitivity to radiation-induced tumorigenesis, providing an ideal in vivo model to study the typical pathologies associated with basal cell nevus syndrome. We therefore examined BCC development in control and irradiated Ptch1neo67/+ mice. We show that unirradiated mice develop putative BCC precursor lesions, i.e., basaloid hyperproliferation areas arising from both follicular and interfollicular epithelium, and that these lesions progress to nodular and infiltrative BCCs only in irradiated mice. Data of BCC incidence, multiplicity, and latency support the notion of epidermal hyperproliferations, nodular and infiltrative BCC-like tumors representing different stages of tumor development. This is additionally supported by the pattern of p53 protein expression observed in BCC subtypes and by the finding of retention of the normal remaining Ptch1 allele in all nodular, circumscribed BCCs analyzed compared with its constant loss in infiltrative BCCs. Our data suggest chronological tumor progression from basaloid hyperproliferations to nodular and then infiltrative BCC occurring in a stepwise fashion through the accumulation of sequential genetic alterations. AU - Mancuso, M.* AU - Pazzaglia, S.* AU - Tanori, M.* AU - Hahn, H.* AU - Merola, P.* AU - Rebessi, S.* AU - Atkinson, M.J. AU - di Majo, V.* AU - Covelli, V.* AU - Saran, A.* C1 - 2698 C2 - 22237 SP - 934-941 TI - Basal cell carcinoma and its development: Insights from radiation-induced tumors in Ptch1-deficient mice. JO - Cancer Res. VL - 64 IS - 3 PY - 2004 SN - 0008-5472 ER - TY - JOUR AB - We described previously a basal cell carcinoma (BCC) and medulloblastoma (MB) phenotype for CD1Ptch1neo67/+ mice exposed to ionizing radiation. Ptch1 heterozygous mice mimic the predisposition to BCC and MB development of patients affected by nevoid BCC syndrome that inherit a mutant Patched (Ptch1) allele. To examine the impact of genetic background on development of BCCs and other tumors we used two outbred mouse lines characterized by extremely high, carcinogenesis-susceptible (Car-S), and low, carcinogenesis-resistant (Car-R), susceptibility to skin carcinogenesis. Crosses between Ptch1neo67/+ mice and Car-S (F1S) or Car-R mice (F1R) were exposed to ionizing radiation. F1SPtch1neo67/+ mice were highly susceptible to radiation-induced BCCs, whereas F1RPtch1neo67/+ mice were completely resistant, indicating that tumor penetrance can be modulated by genetic background. Development of microscopic and macroscopic BCC lesions was influenced by Car-S and Car-R genotypes, suggesting a genetic-background effect on both initiation and progression of BCC. Susceptibility was additionally increased in N2 backcross mice (Car-S x F1SPtch1neo67/+), showing a contribution from recessive-acting Car-S modifiers. The modifying effects of Car-S-derived susceptibility alleles were tissue specific. In fact, despite higher susceptibility to BCC induction, Car-S-derived lines had lower MB incidence compared with CD1Ptch1neo67/+ mice. BCC-associated somatic events were not influenced by genetic background, as shown by similar rate of wild-type Ptch1 loss in BCCs from F1SPtch1neo67/+ (93%) and CD1Ptch1neo67/+ mice (100%). Finally, microsatellite analysis of BCCs showed Ptch1 loss through interstitial deletion. These results are relevant to humans, in which BCC is the commonest malignancy, because this model system may be used to study genes modifying BCC development. AU - Pazzaglia, S.* AU - Mancuso, M.* AU - Tanori, M.* AU - Atkinson, M.J. AU - Merola, P.* AU - Rebessi, S.* AU - di Majo, V.* AU - Covelli, V.* AU - Hahn, H.* AU - Saran, A.* C1 - 2197 C2 - 21850 SP - 3798-3806 TI - Modulation of patched-associated susceptibility to radiation induced tumorigenesis by genetic background. JO - Cancer Res. VL - 64 IS - 11 PY - 2004 SN - 0008-5472 ER - TY - JOUR AB - The nonclassical HLA-G molecule exhibits a limited tissue distribution and exerts multiple immune regulatory functions. Recent studies indicate that HLA-G expression plays a key role in the induction of immune tolerance and may represent a novel immune escape mechanism of tumor cells. Despite a high frequency of tumor-infiltrating T lymphocytes in renal cell carcinoma (RCC) lesions, outgrowth of tumor cells occurs that might be attributable to abrogation-efficient antitumor responses. To delineate the potential role of HLA-G in RCC immunology, the HLA-G expression pattern and its functional consequences on immune responses were analyzed in cell lines and lesions derived from primary RCC lesions. A heterogeneous constitutive and IFN-gamma-inducible HLA-G mRNA and protein expression was found in 12.5% of RCC cell lines but not in autologous normal kidney cells. Western blot analysis of 37 primary RCC lesions revealed HLA-G protein expression in 27% of RCC lesions. Functional studies performed with alloreactive natural and lymphokine-activated killer cells as well as antigen-specific CD8(+) T-cell populations demonstrated that HLA-G expression inhibits lysis of RCC cells by these different immune effector cells, whereas HLA-G(-) normal kidney cells were recognized. Furthermore, the HLA-G-mediated counteraction of immune response could be restored by antibody blocking experiments. Thus, aberrant HLA-G expression is found at a relatively high frequency in RCC and might participate in evasion of these tumor cells from immunosurveillance. AU - Bukur, J.* AU - Rebmann, V.* AU - Grosse-Wilde, H.* AU - Luboldt, H.* AU - Ruebben, H.* AU - Drexler, I.* AU - Sutter, G. AU - Huber, C.* AU - Seliger, B.* C1 - 23863 C2 - 36382 SP - 4107-4111 TI - Functional role of human leukocyte antigen-G up-regulation in renal cell carcinoma. JO - Cancer Res. VL - 63 IS - 14 PB - Amer. Assoc. Cancer Research PY - 2003 SN - 0008-5472 ER - TY - JOUR AB - The EBV latent membrane protein 1 (LMP1) is an integral membrane protein that acts like a constitutively activated receptor. LMP1 interacts with members of the tumor necrosis factor receptor-associated factor family, as well as with tumor necrosis factor receptor-associated death domain, resulting in induction of nuclear factor-κB, the p38 mitogen-activated protein kinase pathway, and the c-Jun NH2-terminal kinase activator protein 1-signaling cascade. The binding of Janus kinase 3 results in activation of signal transducers and activators of transcription. The domain structure of LMP1 has been mapped extensively, but the quantitative contribution of distinct LMP1 domains to the efficiency of B-cell proliferation by EBV has not been determined. On the basis of the maxi-EBV system, which allows us to introduce and study mutations in the context of the complete EBV genome, a panel of 10 EBV mutants with alterations in the LMP1 gene locus was established. The mutant EBVs were tested for their efficiency to induce and maintain proliferation of clonal B-cell lines in vitro. Surprisingly and with reduced frequency, EBV mutants which deleted LMP1’s COOH terminus, transmembrane domains, or the entire open reading frame were able to generate proliferating B-cell clones that were dependent on the presence of human fibroblast feeder cells. A B-cell clone carrying the LMP1-null mutant EBV genome was also analyzed for oncogenicity in severe combined immunodeficiency mice. Our results demonstrate that LMP1 is critical but not mandatory for the generation of proliferating B cells in vitro. LMP1 functions greatly contribute to EBV’s transformation potential and appear essential for its oncogenicity in severe combined immunodeficiency mice. AU - Dirmeier, U. AU - Neuhierl, B. AU - Kilger, E. AU - Reisbach, G. AU - Sandberg, M.L.* AU - Hammerschmidt, W. C1 - 22335 C2 - 21192 SP - 2982-2989 TI - Latent Membrane Protein 1 is Critical for Efficient Growth Transformation of Human B Cells by Epstein-Barr Virus. JO - Cancer Res. VL - 63 IS - 11 PY - 2003 SN - 0008-5472 ER - TY - JOUR AB - The antitumor activity of IFN-alpha is well established. However, the role of the plasmacytoid dendritic cell (PDC), the major producer of IFN-alpha upon viral infection, in tumor biology is unknown. We sought to study the presence and function of PDC in a human solid tumor. Here, we demonstrate that PDCs infiltrate tumor tissue of patients with head and neck squamous cell carcinoma (HNSCC). Functional activity of PDC was examined by using CpG motif containing oligonucleotides, a defined microbial stimulus for PDCs (recognized via toll-like receptor 9). We found that HNSCC diminished the ability of PDC to produce IFN-alpha in response to CpG motif containing oligonucleotide. Tumor-induced down-regulation of toll-like receptor 9 was identified as one mechanism likely contributing to impaired PDC function within the tumor environment. In tumor-draining lymph nodes, suppression of CpG-induced IFN-alpha production was less pronounced than in single-cell suspensions of primary tumor tissue. In these lymph nodes, CpG-induced IFN-alpha production was associated with increased levels of interferon-induced protein 10 and IFN-gamma and activation of CD4 and CD8 T cells. These results show for the first time the presence of PDCs in human solid tumor tissue and that tumors suppress the capacity of PDCs to produce IFN-alpha. PDCs, which in the absence of appropriate stimulation are reported to promote regulatory CD8 T cells, may contribute to an impaired T-cell-mediated immune response in HNSCC. AU - Hartmann, E.* AU - Wollenberg, B.* AU - Rothenfusser, S.* AU - Wagner, M.* AU - Mack, B.* AU - Giese, Th.* AU - Gires, O. AU - Endres, S.* AU - Hartmann, G.* C1 - 10311 C2 - 21731 SP - 6478-6487 TI - Identification and functional analysis of tumor-infiltrating plasmacytoid dendritic cells in head and neck cancer. JO - Cancer Res. VL - 63 IS - 19 PY - 2003 SN - 0008-5472 ER - TY - JOUR AB -   Genetic instability appears to be required for a normal colorectal epithelial cell to evolve into a cancerous one. Bloom syndrome patients have a strong predisposition to cancer that affects a variety of tissues. The mechanism of disease is attributed to genomic instability, but many questions about the nature of this instability have not yet been answered. To investigate these issues, we used gene-targeting techniques to disrupt the BLM gene in karyotypically stable colorectal cancer epithelial cells. BLM knockout cells showed an increased tendency of sister chromatids to exchange DNA strands and were substantially more likely to undergo homologous recombination at chromosomal loci than parental cells. Surprisingly, BLM-deficient colorectal cancer epithelial cells did not display gross chromosomal rearrangements nor a change in the rates of chromosome gains and losses. However, the enhanced homologous recombination was associated with losses of heterozygosity. These observations define a type of genetic instability that has significant implications for the evolution of cancer. AU - Traverso, G.* AU - Bettegowda, C.* AU - Kraus, J. AU - Speicher, M.R. AU - Kinzler, K.W.* AU - Vogelstein, B.* AU - Lengauer, C.* C1 - 10310 C2 - 21585 SP - 8578-8581 TI - Hyper-recombination and genetic instability in BLM-deficient epithelial cells. JO - Cancer Res. VL - 63 IS - 24 PY - 2003 SN - 0008-5472 ER - TY - JOUR AB - Expression analysis of genes encoding components of the phosphotyrosine signaling system by cDNA array hybridization revealed elevated levels of FGFR4 transcripts in several mammary carcinoma cell lines. In the FGFR4 gene transcript from MDA-MB-453 mammary carcinoma cells, a G to A conversion was discovered that results in the substitution of glycine by arginine at position 388 in the transmembrane domain of the receptor. The Arg388 allele was also found in cell lines derived from a variety of other tumor types as well as in the germ-line of cancer patients and healthy individuals. Analysis of three geographically separated groups indicated that it occurs in approximately 50% of the human population. Investigation of the clinical data of 84 breast cancer patients revealed that homo- or heterozygous carriers of the Arg388 allele had a significantly reduced disease-free survival time (P = 0.01) within a median follow-up of 62 months. Moreover, the FGFR4 Arg388 allele was associated with early lymph node metastasis and advanced tumor-node-metastasis (TNM) stage in 82 colon cancer patients. Consistent with this finding, MDA-MB-231 mammary tumor cells expressing FGFR4 Arg388 exhibited increased motility relative to cells expressing the FGFR4 Gly388 isotype. Our results support the conclusion that the FGFR4 Arg388 allele represents a determinant that is innocuous in healthy individuals but predisposes cancer patients for significantly accelerated disease progression. AU - Bange, J.* AU - Prechtl, D.* AU - Cheburkin, Y.* AU - Specht, K. AU - Harbeck, N.* AU - Schmitt, M.* AU - Knyazeva, T.* AU - Müller, S.* AU - Gärtner, S.* AU - Sures, I.* AU - Wang, H.* AU - Imyanitov, E.* AU - Häring, H.-U.* AU - Knayzev, P.* AU - Iacobelli, S.* AU - Höfler, H. AU - Ullrich, A.* C1 - 21960 C2 - 20482 SP - 840-847 TI - Cancer Progression and Tumor Cell Motility Are Associated with the FGFR4 Arg388 Allele. JO - Cancer Res. VL - 62 IS - 3 PY - 2002 SN - 0008-5472 ER - TY - JOUR AB - Lymphokine activated killer (LAK) cells represent mixtures of natural killer (NK) and non-MHC-restricted CTLs that have the capacity to lyse a variety of tumor cells and MHC class I-negative target cells. Although it is clear that NK cells are negatively regulated by interactions with MHC class Ia or class Ib molecules, the regulation of LAK-derived T cells has not been clarified to date. In the studies presented here, we demonstrate that IFN-  treatment of tumor cells can induce their resistance to LAK- derived T cells in a manner similar to that seen for NK cells. The IFN-  -mediated suppression of LAK activity correlates with increased MHC class I expression by the tumor cells, and the inhibition of LAK- mediated cytotoxicity can be reversed in the presence of class I-specific antibody. Furthermore, the expression of MHC class Ia or class Ib mol- ecules in class I-negative cell lines can reduce their susceptibility to LAK-mediated cytotoxicity. This principle of negative regulation by MHC class I molecules applies to LAK-derived T cells generated from periph- eral blood lymphocytes of renal cell carcinoma patients and healthy, control donors. Although LAK-derived T cells can be inhibited in their lytic activity through interactions with MHC class Ia and class Ib mole- cules, they do not express the known inhibitory receptors specific for these ligands that are found on NK cells. Apparently, LAK-derived T cells are negatively regulated by as yet undefined inhibitory receptors. AU - Falk, C.S. AU - Nößner, E. AU - Weiss, E.H.* AU - Schendel, D.J. C1 - 10313 C2 - 20296 SP - 480-487 TI - Retaliation against Tumor Cells Showing Aberrant HLA Expression Using Lymphokine Activated Killer-derived T Cells1. JO - Cancer Res. VL - 62 PB - AACR PY - 2002 SN - 0008-5472 ER - TY - JOUR AB - The relative cytotoxicity of a diphtheria toxin (DT) human interleukin 3(IL3) fusion protein (DT388IL3) was tested against primitive normal (n = 3)and acute myeloid leukemia (AML) progenitors (n = 7). After 24-h culture with 50 ng/ml DT388IL3, the mean percentages of kill of AML colony-forming cells (CFCs), long-term culture-initiating cells (LTC-ICs), and suspension culture-ICs (SC-ICs) were 82% (range, 47–100), 56% (range, 28–91), and 74% (range, 43–87), respectively, with most surviving progenitors being cytogenetically normal. Engraftment of DT388IL-3-treated AML cells in nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice followed for 16 weeks was eradicated for two of these samples. In contrast, with normal bone marrow, mean percentages of CFC kill of 49 and 64% were seen with 50 or 250 ng/ml DT388IL3, respectively, whereas no significant kills were observed in the LTC-IC and SC-IC assays. The NOD/SCID mouse repopulating cell (RC) frequency in normal BM cells was also not reduced by DT388IL3 treatment. In subsequent experiments, NOD/SCID mice that received AML blasts i.v. followed in 24 h by 0.045 μg/g DT388IL3 daily i.p. × 5 showed mean percentages of reduction in AML engraftment of 83% (range, 14–100) and 57% (range, 0–98) after 4 and 12 weeks, respectively (n = 6). No evidence of leukemia was detected with two of six AML samples 12 weeks after one 5-day course of DT388IL3. Repeating the DT388IL3 treatment every 4 weeks enhanced its effectiveness against two additional samples. Thus, DT388IL3 kills primitive leukemic progenitors from a proportion of AML patients but shows no significant toxicity against equivalent normal cells. AU - Feuring-Buske, M. AU - Frankel, A.E.* AU - Alexander, R.L.* AU - Gerhard, B.* AU - Hogge, D.E.* C1 - 22231 C2 - 20955 SP - 1730-1736 TI - A Diphtheria Toxin-Interleukin 3 Fusion Protein is Cytotoxic to Primitive Acute Myeloid Leukemia Progenitors but Spares Normal Progenitors. JO - Cancer Res. VL - 62 IS - 6 PY - 2002 SN - 0008-5472 ER - TY - JOUR AB - We describe a novel hereditary cancer syndrome in the rat that is transmitted by a recessive gene mutation. Animals exhibiting the mutant phenotype develop multiple neuroendocrine malignancies within the first year of life. The endocrine neoplasia is characterized by bilateral adrenal pheochromocytoma, multiple extra-adrenal pheochromocytoma, bilateral medullary thyroid cell neoplasia, bilateral parathyroid hyperplasia, and pituitary adenoma. The appearance of neoplastic disease is preceded by the development of bilateral juvenile cataracts. Although the spectrum of affected tissues is reminiscent of human forms of multiple endocrine neoplasia (MEN), no germ-line mutations were detected in the Ret or Menin genes that are responsible for the dominantly inherited MEN syndromes in humans. Segregation studies in F1 and F2 crosses yielded frequencies of affected animals entirely consistent with a recessive autosomal mode of inheritance. The lack of the phenotype in F1 animals effectively excludes a germ-line tumor suppressor gene mutation as the causal event. The absence of mutation of known MEN genes and the unique constellation of affected tissues, plus the recessive mode of inheritance, lead us to conclude that the mutation of an as yet unknown gene is responsible for this syndrome of inherited neuroendocrine cancer. AU - Fritz, A. AU - Walch, A.K. AU - Piotrowska, K. AU - Rosemann, M. AU - Schäffer, E.H. AU - Weber, K.* AU - Timper, A. AU - Wildner, G.* AU - Graw, J. AU - Höfler, H. AU - Atkinson, M.J. C1 - 10312 C2 - 20412 SP - 3048-3051 TI - Recessive Transmission of a Multiple Endocrine Neoplasia Syndrome in the Rat. JO - Cancer Res. VL - 62 IS - 11 PB - AACR PY - 2002 SN - 0008-5472 ER - TY - JOUR AB - Cyclin D1 is downstream of erbB2 and is required for erbB2 transformation. Here we report thatcyclin D1 functions are essential, rate limiting for erbB2 transformation, and reciprocally increase erbB2 levels. This interaction depends on three cyclin D1 activities: cyclin-dependent kinase 4-dependent kinase activity, titration of p27, and an intrinsic transcriptional activity of cyclin D1. Drugs active against erbB2 and cyclin D1 (Herceptin and flavopiridol) were synergistically cytotoxic against erbB2-positive breast cancer cell lines. Addition of flavopiridol to Herceptin synergistically lowered erbB2 levels in these cells. Our data suggest the potential use of combinations of cyclin-dependent kinase inhibitors and Herceptin in breast cancer. AU - Nahta, R.* AU - Iglehart, J.D.* AU - Kempkes, B. AU - Schmidt, E.V.* C1 - 21969 C2 - 20493 SP - 2267-2271 TI - Rate-limiting Effects of Cyclin D1 in Transformation by ErbB2 Predicts Synergy between Herceptin and Flavopiridol. JO - Cancer Res. VL - 62 IS - 8 PY - 2002 SN - 0008-5472 ER - TY - JOUR AB - Various evidence suggests that adoptive transfer of polyclonal, tumor-specific, IFN-γ-producing CD4+ T cells [T helper type 1 (Th1) cells] should be highly efficient for tumor immune therapy. However, this approach could not be tested because very few MHC class II-restricted tumor peptides have been defined. Here we show that stimulation of freshly isolated T helper cells with syngeneic tumor cells and antigen-presenting cells in the presence of immunostimulatory CpG DNA allows the generation of large numbers of strongly polarized, tumor-specific Th1 cells within 3 weeks of culture, even when T helper cells were derived from tumor-bearing mice. A single injection of 0.5 × 106 A20-specific Th1 cells even eradicated disseminated A20 lymphomas and provided lifelong protection without inducing autoimmune disease. The therapy was largely independent of CD8+ cells but required IFN-γ and CD40-CD40L interactions, suggesting that tumor-specific Th1 cells eradicate established tumors by activating proinflammatory macrophages. AU - Egeter, O.* AU - Mocikat, R. AU - Ghoreschi, K.* AU - Dieckmann, A. AU - Röcken, M.* C1 - 21357 C2 - 19473 SP - 1515-1520 TI - Eradication of Disseminated Lymphomas with CpG-DNA Activated T Helper Type Cells from Nontransgenic Mice. JO - Cancer Res. VL - 60 IS - 6 PY - 2000 SN - 0008-5472 ER - TY - JOUR AB - Evaluation of 20 cases of radiation-induced childhood papillary thyroid carcinoma using fluorescence in situ hybridization demonstrated the presence of clonal translocations affecting the RET locus. Semiquantitative reverse transcription-PCR indicated overexpression of the RET tyrosine kinase (TK) domain in four cases. In two cases, the RET rearrangements PTC6 and PTC7 were identified and assigned to balanced translocations t(7;10)(q32;q11.2) and t(1;10)(p13;q11.2), respectively. In one case with a balanced translocation t(10;14)(q11.2;q22.1), 5' rapid amplification of cDNA ends revealed a novel type of RET oncogenic activation (PTC8), arising from a fusion of the 5' part of the kinectin (KTN1) gene to the TK domain of the RET gene. The presence of coiled-coil domains in the resulting ktn1/ret fusion protein suggests ligand-independent dimerization and thus constitutive activation of the ret TK domain. AU - Salassidis, K. AU - Bruch, J. AU - Zitzelsberger, H. AU - Lengfelder, E.* AU - Kellerer, A.M.* AU - Bauchinger, M. C1 - 23356 C2 - 31123 SP - 2786-2679 TI - Translocation t(10;14)(q11.2:q22.1) fusing the kinectin to the RET gene creates a novel rearranged form (PTC8) of the RET proto-oncogene in radiation-induced childhood papillary thyroid carcinoma. JO - Cancer Res. VL - 60 IS - 11 PB - American Assoc. of Cancer Research PY - 2000 SN - 0008-5472 ER - TY - JOUR AB - Risk assessment of dioxins is currently based on induction of liver tumors in rats. The toxicity of dioxins is characterized by large sensitivity differences among animal species and even strains of the same species, which complicates the risk assessment. The significance of these differences in dioxin-induced carcinogenicity is not known. We therefore studied the liver tumor-promoting activity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in the sensitive Long-Evans (L-E) and the resistant Han/Wistar (H/W) rats differing >1000-fold in their sensitivity to the acute lethality of TCDD. Female rats were partially hepatectomized, initiated with nitrosodiethylamine, and treated with TCDD for 20 weeks. Altered hepatic foci (AHF) were stereologically quantitated using glutathione S-transferase P as a marker. AHF were significantly (P < 0.001) and dose dependently increased in L-E rats at 10 and 100 ng/kg/day, but in H/W rats only at 1000 ng/kg/day and above, indicating a remarkable (∼100-fold) sensitivity difference between L-E and H/W rats. The same sensitivity difference but 10-fold less foci were observed between nonhepatectomized/noninitiated L-E and H/W rats. Induction of AHF was related to hepatotoxicity but not to cytochrome P4501A1 activity in the liver. Liver TCDD concentrations were similar in both strains. H/W rats are exceptionally resistant to induction of AHF by TCDD, and the resistance is associated with an altered transactivation domain of the aryl hydrocarbon receptor. Genetic differences may account for significant interindividual/intraspecies sensitivity differences in dioxin-induced carcinogenesis. Understanding the role of transactivation domain of the aryl hydrocarbon receptor in carcinogenesis is therefore likely to improve dioxin risk assessment. AU - Viluksela, M.* AU - Bager, Y.* AU - Tuomisto, J.T.* AU - Scheu, G.* AU - Unkila, M.* AU - Pohjanvirta, R.* AU - Flodström, S.* AU - Kosma, V.M.* AU - Mäki-Paakkanen, J.* AU - Vartiainen, T.* AU - Klimm, C. AU - Schramm, K.-W. AU - Wärngard, L.* AU - Tuomisto, J.* C1 - 21660 C2 - 19827 SP - 6911-6920 TI - Liver Tumor-promoting Activity of 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD-sensitive and TCDD- resistant Rat Strains1. JO - Cancer Res. VL - 60 IS - 24 PY - 2000 SN - 0008-5472 ER - TY - JOUR AB - Metabolic activation of the K-region trans-8,9-diol of the highly carcinogenic hexacyclic aromatic hydrocarbon dibenzo[a,l]pyrene (DB[a,l]P) by human cytochrome P-450 (P450) 1A1 and 1B1 was investigated in Chinese hamster V79 cell lines expressing human P450 1A1 or 1B1. P450 1A1 and 1B1 are the major P450s involved in metabolic activation of polycyclic aromatic hydrocarbons in human cells. The major DNA adducts formed by metabolism of DB[a,l]P in cultures expressing P450 1A1 or 1B1 resulted mainly from the fjord region (-)-anti-DB[a,l]P-11,12-diol 13,14-epoxide [(-)-anti-DB[a,l]PDE] and, to a lesser extent, (+)-syn-DB[a,l]PDE. In V79 cells expressing human P450 1A1, high amounts of as yet unidentified highly polar DNA adducts are formed in addition to the DNA adducts derived from DB[a,l]PDEs. Human P450 1A1 has been found to metabolize DB[a,l]P on its K-region to the trans-8,9-diol, and it has been proposed that the DNA binding of the parent compound in P450 1A1-expressing tissues may be partially mediated by activation of the K-region trans-8,9-diol to form bis-diol epoxides. V79 cells expressing human P450 1A1 or 1B1 formed only low amounts of DNA adducts after treatment with high doses of the K-region trans-8,9-diol. None of the adducts formed were identical to the main adducts formed in the same cell lines by metabolic activation of DB[a,l]P or (-)-DB[a,l]P-trans-11,12-diol. These results demonstrate that the K-region trans-8,9-diol does not significantly contribute to the genotoxicity of the very potent carcinogen DB[a,l]P in human cells or tissues expressing P450 1A1 or 1B1. AU - Luch, A.* AU - Kishiyama, S.* AU - Seidel, A.* AU - Doehmer, J.* AU - Greim, H. AU - Baird, W.M.* C1 - 23984 C2 - 31430 SP - 4603-4609 TI - The K-region trans-8,9-diol does not significantly contribute as an intermediate in the metabolic activation of dibenzo[a,l]pyrene to DNA-binding metabolites by human cytochrome P450 1A1 or 1B1. JO - Cancer Res. VL - 59 IS - 18 PB - Amer. Assoc. Cancer Research PY - 1999 SN - 0008-5472 ER - TY - JOUR AB - A major goal of tumor immunotherapy is the induction of a systemic immune response against tumor antigens such as the tumor-specific immunoglobulin idiotype (Id) expressed by lymphomas of the B-cell lineage. We describe an approach based on specific redirection of the tumor Id toward professional antigen-presenting cells (APCs), thereby overcoming the inefficient presentation on the parental transformed B cell. Lymphoma cells are fused to a xenogeneic hybridoma cell line that secretes an antibody against a surface molecule on APCs. Due to preferential assembly between heavy and light chains of antibodies of different species-origin, the resulting "trioma" cells produce at high yield a bispecific antibody containing the lymphoma Id and the APC-binding arm, which redirects the Id to APCs. Processing and presentation of the Id will lead to T-cell activation. An absolute requirement for inducing a complete tumor protection was the immunization with antibody-secreting trioma cells as a cell-based vaccine instead of the soluble bispecific antibody. Tumor immunity was specific and long-lasting. Both CD4+ and CD8+ T cells were necessary for inducing tumor immunity. AU - Mocikat, R. AU - Selmayr, M. AU - Thierfelder, S. AU - Lindhofer, H. C1 - 58138 C2 - 0 SP - 2346-9 TI - Trioma-based vaccination against B-cell lymphoma confers long-lasting tumor immunity. JO - Cancer Res. VL - 57 IS - 12 PY - 1997 SN - 0008-5472 ER - TY - JOUR AB - EBV is a human tumor virus that is associated with different types of tumors. A unique feature of EBV is its capability to infect and immortalize human B cells both in vivo and in vitro. In cell culture, this progress is termed immortalization and infected B cells grow out to permanent, so-called lymphoblastoid cell lines. During our experiments, we observed that B lymphocytes derived from adenoids are infected efficiently by EBV and proliferate much more rapidly than any other known type of B cell. High concentrations of adhesion molecules and of CD21, the EBV receptor, present on these cells may account for this phenomenon. Adenoid B cells may therefore represent a particular subpopulation of preactivated B lymphocytes that can greatly simplify and enhance the production of lymphoblastoid cell lines for, e.g., antigen-presenting cells for gene therapeutic approaches and similar applications. AU - Zeidler, R. AU - Meissner, P.* AU - Eissner, G. AU - Lazis, S. AU - Hammerschmidt, W. C1 - 27620 C2 - 32770 SP - 5610-5614 TI - Rapid proliferation of B cells from adenoids in response to Epstein-Barr virus infection. JO - Cancer Res. VL - 56 IS - 24 PY - 1996 SN - 0008-5472 ER - TY - JOUR AB - Both retroviral infections as well as human tumors may cause immunosuppression. One of the factors involved in immunosuppression in patients with squamous cell carcinoma of the head and neck (SCC-HN) is a protein related in the retroviral protein p15E. A conserved, 17-amino acid sequence represents the immunosuppressive epitope of retroviral p15E. In order to study the relationship between SCC-HN associated immunosuppression and retroviral p15E, we produced three new monoclonal antibodies (MAbs: ER- IS1, ER-IS2, and ER-185) directed against the immunosuppressive synthetic CKS-17 peptide. These MAbs react with the immunosuppressive peptide (in enzyme-linked immunosorbent assay), with human tumor cell lines (in FACScan analysis), with retroviral p15E (on Western blot), and with cryostat sections of SCC-HN tumor tissue. In addition, the MAbs neutralize the immunosuppressive low molecular weight factors present in sera of patients with SCC-HN. These results show that retroviral p15E and the immunosuppressive factors associated with SCC-HN share a conserved immunosuppressive epitope and that MAbs against this epitope can be used for detection and centralization of the tumor-associated immunosuppressive protein(s). AU - Lang, M.* AU - Oostendorp, R.A.J. AU - Simons, P.J.* AU - Boersma, W.J.A.* AU - Knegt, P.P.M.* AU - Van Ewijk, W.* C1 - 33540 C2 - 38515 SP - 1831-1836 TI - New monoclonal antibodies against the putative immunosuppressive site of retroviral p15E. JO - Cancer Res. VL - 54 IS - 7 PY - 1994 SN - 0008-5472 ER - TY - JOUR AB - Surgical biopsies from ten head and neck squamous cell carcinomas were labeled in vitro with bromodeoxyuridine. In histological sections, bromodeoxyuridine-positive nuclei and β-fibroblast growth factor (β-FGF) were stained using immunohistochemistry. In clearly discernible clusters of tumor cells, the cytoplasm shows strong positive β-FGF staining, whereas other tumor regions are completely β-FGF negative. Within positively stained areas, the tumor cell bromodeoxyuridine labeling index is higher in comparison to β-FGF-negative areas by a factor of 5 ± 0.8. This is reflected in a positive correlation of the tumor cell labeling index and the relative extent of β-FGF-positive tumor areas. Viable tumor areas bordering on necrosis, which are known to be hypoxic, are β-FGF negative. The average tumor endothelial cell labeling index was 1.8 ± 0.6%, as compared to 0.16% in adjacent normal mucosa. Since endothelial cell pulse labeling indices are too low for a further quantitative analysis, the relationship of β-FGF expression and endothelial cell turnover was studied in more detail in two fairly well-differentiated murine squamous cell carcinoma lines (AT 84 and AT 478). Labeling indices were higher and endothelial cell doubling times were significantly shorter in β-FGF-positive as compared to β-FGF-negative tumor areas (AT 84, 9.3 h versus 25.4 h; AT 478/25, 6.8 h versus 16 h). Thus, the discrete expression of β-FGF is associated with regional differences in endothelial cell kinetics. In two generations of the tumor line AT 478, characterized by different volume doubling times of 18 days (AT 478/25) and 36 days (AT 478/4), β-FGF-positive areas represent 75.5 ± 6% and 19.7 ± 7% of the viable tumor tissue, respectively. This indicates a correlation between β-FGF production of tumor cells and growth rate. AU - Schultz-Hector, S. AU - Haghayegh, S. C1 - 40313 C2 - 13060 SP - 1444-1449 TI - β-fibroblast growth factor expression in human and murine squamous cell carcinomas and its relationship to regional endothelial cell proliferation. JO - Cancer Res. VL - 53 IS - 6 PY - 1993 SN - 0008-5472 ER - TY - JOUR AB - To analyze the region upstream of c-myc, a number of novel probes were established. These were generated by chromosomal walking starting from the breakpoint of the chromosomal translocation of the B-cell line 380 and by cloning the breakpoint of the translocation of the Burkitt lymphoma cell line IARC/BL72. Using the newly isolated probes a detailed physical map of 500 kilobases of the region upstream of c-myc was established applying pulsed-field gel electrophoresis. The chromosomal breakpoint of IARC/BL72 cells was mapped to a site 55 kilobases 5′ of c-myc. A region 20 kilobases in length and containing the breakpoints of 380, EW36, P3HR-1, and Daudi cells was identified 170-190 kilobases upstream of c-myc. In addition the HPV18 integration site in HeLa cells was located between 340 and 500 kilobases 5′ of c-myc. The probes were used to define the c-myc amplification units in Colo320-HSR and HL60 cells as well as in four cases of small cell lung cancer. Evidence is provided that the amplicon of HL60 cells is discontinuously organized. AU - Joos, S. AU - Haluska, F.G.* AU - Falk, M.H. AU - Henglein, B.* AU - Hameister, H.* AU - Croce, C.M.* AU - Bornkamm, G.W. C1 - 40490 C2 - 38884 SP - 6547-6552 TI - Mapping chromosomal breakpoints of Burkitt's t(8;14) translocations far upstream of c-myc. JO - Cancer Res. VL - 52 IS - 23 PY - 1992 SN - 0008-5472 ER - TY - JOUR AU - Jenke, H.-S. AU - Michel, G. AU - Hornhardt, S. C1 - 18958 C2 - 11347 TI - Polychlorinated Biphenyls (PCB) Interfere with Cellular Oncogene-Expression in Rat Liver at the Transcriptional Level. JO - Cancer Res. PY - 1990 SN - 0008-5472 ER - TY - JOUR AB - Endogenous retroviruses and retroviral elements represent a substantial component of vertebrate genomes. They are inherited as stable Mendelian genes and may be activated spontaneously or by physical or chemical agents. In the human genome various retroviral elements have been detected by their relationship with mammalian endogenous and exogenous retroviruses. The structure of these elements resembles either full-length or truncated proviruses. The biological function of human retrovirus-related sequences is still unknown, but like other transposable elements, they may have contributed in shaping the eukaryotic genome. Furthermore, they exhibit a number of features giving them a potential for involvement in carcinogenesis. Expression of endogenous retroviral elements has been detected in various human tissues and cell lines and in some cases appears to be associated with human neoplasias. AU - Leib-Mösch, C. AU - Brack-Werner, R. AU - Werner, T. AU - Bachmann, M.* AU - Faff, O.* AU - Erfle, V.F. AU - Hehlmann., R.* C1 - 41157 C2 - 34287 SP - 5636s-5642s TI - Endogenous retroviral elements in human DNA. JO - Cancer Res. VL - 50 PY - 1990 SN - 0008-5472 ER - TY - JOUR AU - Petrides, P.E. AU - Bock, S. AU - Bovens, J. AU - Hofmann, R. AU - Jakse, G. C1 - 18235 C2 - 11448 TI - Modulation of pro-EGF, pro TGF-alpha and EGF-receptor Gene Expression in Human Renal Carcinoma. JO - Cancer Res. PY - 1990 SN - 0008-5472 ER - TY - JOUR AB - We have analyzed the expression of the genes for the precursors of epidermal growth factor (pro-EGF) and transforming growth factor α (proTGF-α) as well as for the EGF receptor in tissue specimens of a large number of adult patients with renal cell carcinoma. Since normal kidney tissue was available from the same patients we could directly compare the expression of these genes in tumors with that in adjacent normal renal tissue. Our experiments reveal underexpression of the proEGF gene in all tumors analyzed (21 of 21) and overexpression of the genes for proTGF-α (33 of 33 analyzed) and EGF receptor (22 of 23 analyzed) in tumor samples, when compared with normal kidney tissue. The expression of the proTGF-α gene appeared to depend on grade and differentiation of the tumor, since well differentiated tumors (grade 1) expressed more proTGF-α mRNA than the adjacent normal tissue but significantly less than poorly differentiated tumors (grade 2 or 3), which are the most aggressive ones. In none of these tissue specimens did we find, by Southern analysis, amplification of the proTGF-α or EGF receptor gene. Therefore, overexpression of these genes must be due to another effect, perhaps an alteration of their mRNA turnover. Although the EGF receptor gene (c-ereB1) is overexpressed in nearly all carcinomas analyzed, there was no linear coexpression with the proTGF-α gene. In contrast, transcription of the proEGF gene was completely turned off in tumor tissue. Although we have found by restriction fragment length polymorphism analysis, in one of three tumor samples, evidence for a somatic mutation within the proEGF gene, we do not know yet, due to the limited number of Southern analyses, whether this somatic mutation is causally involved in the decrease of proEGF mRNA expression and, hence, is representative of renal cell carcinoma. To our knowledge, this is the first observation on primary tumor tissue in humans that upon malignant transformation the gene for a polypeptide growth factor gene is underexpressed. AU - Petrides, P.E. AU - Bock, S.* AU - Bovens, J.* AU - Hofmann, R.* AU - Jakse, G.* C1 - 42050 C2 - 34282 SP - 3934-3939 TI - Modulation of pro-epidermal growth factor, pro-transforming growth factor α and epidermal growth factor receptor gene expression in human renal carcinomas. JO - Cancer Res. VL - 50 IS - 13 PY - 1990 SN - 0008-5472 ER - TY - JOUR AB - Fifty-one radiation-induced murine osteosarcomas were investigated for alterations in c-myc gene structure and c-myc expression. Amplification of c-myc was found in 30% of BALB/c tumors and 13% of NMRI tumors. A region of common proviral integration, Mlvi-1, localized on the same region on chromosome 15, was amplified concomitantly. Multiple copies of both loci were localized on double minutes. Three of the tumors with c-myc amplification also showed rearrangements of the c-myc gene region. One of these rearrangements included the 5′ and 3′-flanking sequences and the noncoding part of the third exon. Repetitive sequences were found in the 5′ region of the c-myc gene, and the 3′ flanking region was substituted by sequences normally present in a more distant part of chromosome 15. Increased levels of c-myc transcripts of apparently normal size were found in tumors carrying amplified c-myc sequences. Abnormally high expression of c-myc in some tumors was correlated with an early stage of osteogenic differentiation, suggesting the involvement of the c-myc gene in the control of the osteogenic differentiation of transformed cells. AU - Sturm, S.A. AU - Strauß, P.G. AU - Adolph, S.* AU - Hameister, H.* AU - Erfle, V.F. C1 - 42134 C2 - 40264 SP - 4146-4153 TI - Amplification and rearrangement of c-myc in radiation-induced murine osteosarcomas. JO - Cancer Res. VL - 50 IS - 13 PY - 1990 SN - 0008-5472 ER - TY - JOUR AB - We recently found that exposure of cells to different aminothiols promotes cystine uptake and leads to an increase of cellular glutathione by new biosynthesis (Issels et al., Biochem. Pharmacol. 37: 881-888, 1988). Therefore, we further investigated whether the known radioprotective and chemoprotective aminothiol derivative S-2-(3-aminopropylamino)ethylphosphorothioic acid (WR-2721) or its dephosphorylated form (WR-1065) will lead to similar effects. In order to convert WR-2721 to the free thiol compound (WR-1065) in vitro, the medium also contained 20 U/ml alkaline phosphatase (AP). For uptake studies a modified McCoy's 5A medium supplemented with 0.1 mM [35S]cystine was used. In Chinese hamster ovary (CHO) and Chinese hamster ovarian carcinoma (OvCa) cells, WR-2721 exposure alone did not increase the cystine uptake relative to that of control (untreated) cells, while WR-2721 + AP enhanced the uptake of cystine more than twofold in both cell lines. The increase of cystine uptake was dependent on the time of exposure (0-60 min) and the concentrations of WR-2721 (0-8 mM) + AP. Half-maximal uptake of cystine was observed at concentrations of 0.69 and 0.57 mM WR-2721 in CHO and OvCa cells, respectively. Determination of both reduced (GSH) and oxidized (GSSG) cellular glutathione levels after the exposure (0-300 min) to WR-2721 + AP in CHO cells showed a depletion of GSH to less than 10% of the pretreatment value and a 4-fold reduction of the GSH/GSSG ratio. In contrast, in OvCa cells the amount of total glutathione rather increased with no significant change of the GSH/GSSG ratio by the exposure to WR-2721 + AP. Further analysis using high-performance liquid chromatography of cell extracts revealed that the relative amount of incorporated [35S]cystine into glutathione was increased similarly in both cell lines. The data show that precursor availability and new biosynthesis of glutathione is enhanced by the exposure to WR-2721 + AP in vitro despite the differential modulation of the cellular glutathione status in the two cell lines. These findings may have important implications for the use of aminothiols like WR-2721 in various cells and tissues in regard of their response to chemotherapeutic agents, ionizing radiation and/or hyperthermia. AU - Issels, R.D. AU - Nagele, A. C1 - 42456 C2 - 36486 SP - 2082-2086 TI - Promotion of cystine uptake, increase of glutathione biosynthesis, and modulation of glutathione status by S-2-(3-aminopropylamino)ethyl phosphorothioic acid (WR-2721) in Chinese hamster cells. JO - Cancer Res. VL - 49 IS - 8 PY - 1989 SN - 0008-5472 ER - TY - JOUR AB - When human tumor xenotransplants into nude mice are used as experimental models, it is important to know whether their microvascular anatomy is rather host or tumor specific. Therefore a morphometric comparison of the vascular network in human squamous cell carcinomas and their xenotransplants was carried out. Biopsies were taken from surgical specimens of three squamous cell carcinomas of the oral cavity. Part of the material was processed for histology and the rest was cut into 1-mm3 cubes and transplanted s.c. into the lateral thorax of athymic nude mice [NCr/Sed(nu/nu)]. The microvascular architecture of original tumors and of three first, one second, and one late generation xenografts was compared. Capillaries were identified in original human tumors by anti-factor VIII staining and in xenografts with antilaminin staining. The median distances between interphase tumor cells and blood vessels were determined and were found to be much longer in original human tumors than in xenografts, ranging from 81 microns to 99 microns and 53 microns to 65 microns, respectively. However, the characteristic qualitative histology of tumors appeared to be preserved in xenotransplants. Analysis of the topographic distribution of mitotic figures revealed that in both original tumors and xenotransplants proliferation of tumor cells was concentrated around blood vessels. Again, vascular distances in original tumors were significantly longer than in xenotransplants. In addition, xenotransplants into nude mice from a long passaged cell line from a human pharyngeal squamous cell carcinoma, FaDu, was investigated. FaDu showed a rare-fication of the capillary network with increasing tumor volume, but a constant median distance of mitotic figures from blood vessels. In conclusion, the pattern of spatial distribution of proliferating tumor cells as well as differentiation characteristics appear to be retained in xenograft tumors, but the density of the vascular system is host specific. This has to be taken into account when physiological parameters of blood supply are studied in xenotransplanted tumors. AU - Lauk, S. AU - Zietman, A. AU - Skates, S. AU - Fabian, R. AU - Suit, H.D. C1 - 17513 C2 - 10442 SP - 4557-4561 TI - Comparative Morphometric Study of Tumor Vasculature in Human Squamous Cell Carcinomas and Their Xenotransplants in Athymic Nude Mice. JO - Cancer Res. VL - 49 IS - 16 PY - 1989 SN - 0008-5472 ER - TY - JOUR AB - The clonogenic assay is widely considered to be the most valid test for predicting tumor cell sensitivity to cytostatic drugs. In this study it was compared with early growth curves of human leukemic cell lines (HL-60, K562, Reh) after treatment with different types of cytostatic drugs (Adriamycin,cis-diamminedichloroplatinum(II):,1-β-D-arabinofuranosyl- cytosine, and 5-fluorouracil) for 1 and 24 h. Following drug treatment two parallel cultures were started: a soft agar culture for the clonogenic assay; and a liquid suspension culture for vital cell counting by measuring esterase activity with fluorescein diacetate at different time points. The latter was recorded using flow cytometry during the following 3 days in 12-h intervals. For each drug concentration a survival factor was calculated from the growth curve between 24 and 72 h. This survival factor takes into account both the y intercept of the extrapolated growth curve and the slope of the growth curve. The dose-response curves resulting from either the survival factors or the clonogenic assay were always nearly identical. The results demonstrate that in established cell lines flow cytometric determination of vital cell increase rates provides a convenient alternative to the clonogenic assay for drug testing. AU - Ellwart, J.W. AU - Kremer, J.P. AU - Dörmér, P.G. C1 - 42123 C2 - 36135 SP - 5722-5725 TI - Drug testing in established cell lines by flow cytometric vitality measurements versus clonogenic assay. JO - Cancer Res. VL - 48 IS - 20 PY - 1988 SN - 0008-5472 ER - TY - JOUR AU - Issels, R.D. AU - Nagele, A. AU - Wilmanns, W. C1 - 17275 C2 - 10332 TI - Promotion of Cystine Uptake, Increase of Glutathione Biosynt hesis, and Modulation of Glutathione Status by S-2-(3-aminopylamino)-ethyl phosphorothioic acid (WR-2721) in Chinese Hamster Cells. JO - Cancer Res. PY - 1988 SN - 0008-5472 ER - TY - JOUR AU - Schmidt, J. AU - Strauß, P.G. AU - Schön, A. AU - Luz, A. AU - Murray, A.B. AU - Melchiori, A. AU - Aresu, O. AU - Erfle, V. C1 - 17382 C2 - 9926 TI - Characterization of Tumor Cell Heterogenity in Clonal Sublines from a Spontaneous Murine Osteosarcoma. JO - Cancer Res. PY - 1988 SN - 0008-5472 ER - TY - JOUR AB - Biopterin accumulation had been demonstrated as the result of normal and, especially, of malignant hemopoietic cell proliferation . Among 13 major intermediates of pterin metabolism and two lumazines, xanthopterin (but not dihydroxyanthopterin) was found to inhibit cell proliferation (half-maximum inhibition at 1.8 x 10 -5 M) during concanavalin A-induced lymphocyte activation in pre-stimulated lymphocytes and in a lymphoid cel line grown in continuous culture (LS-2). LS-2 cells exposed to maximum inhibitor concentrations largely maintained the initial thymidine incorporation rate for about 40 hr but failed to enter logarithmic growth. Isoxanthopterin inhibition was found only in serum-free medium, since it is trapped by the α-acid glycoprotein present in the serum. The reduced biopterin derivatives, sepiapterin, dihydrobiopterin, and tetrahydrobiopterin, are costimulators during concanavalin A-induced lymphocyte activation. Their costimulatory effect follows an optimum curve and peaks at 1.5 to 3 x 10 -5 M. It is highest at the suboptimal and supraoptimal concanavalin A concentration. The D-erythro isomer dihydroneopterin was inactive. The results indicate that the anabolic-reduced biopterin derivatives are not simply lymphocytic products, but, in combination with the catabolites xanthopterin and isoxanthopterin, they also participate in the regulation of lymphocyte activation. Hence, they fulfill the criteria for lymphokines. AU - Ziegler, I. AU - Hamm, U. AU - Berndt, J. C1 - 40909 C2 - 38315 SP - 5356-5359 TI - Participation of pterins in the control of lymphocyte stimulation and lymphoblast proliferation. JO - Cancer Res. VL - 43 IS - 11 PY - 1983 SN - 0008-5472 ER - TY - JOUR AB - A glycoprotein was selectively enriched in the supernatant (Fraction b) obtained by alcohol and trichloroacetic acid fractionation of digitonin extracts from blood of patients with neoplastic diseases and of control subjects. Subsequent chromatography with concanavalin A:Sepharose separated a concanavalin A-reactive fraction from a concanavalin A-nonreactive one. In sodium dodecyl sulfate gel electrophoresis, the fractions from both malignant origin as well as control subjects appeared as single bands showing the same mobility. They were identical with the band obtained from commercial α1-acid glycoprotein. In Fraction b of malignant origin, greatly increased amounts of the α1-acid glycoprotein from malignant cases (AGP(M)) were found as compared to α1-acid glycoprotein from controls (AGP(C)). Furthermore, AGP(C) had a higher glycine content than did AGP(M). The electrofocusing pattern of AGP(M) showed additional bands between pH 3.7 and 4.4, whereas AGP(C) and commercial α1-acid glycoprotein focused between pH 3.2 and 3.8. In contrast to AGP(C) and to a commercial α1-acid glycoprotein, AGP(M) is characterized by a chromophoric group with maximal absorbance at 400 nm. It could be detached by treatment with 6 M guanidine hydrochloride, thus indicating a noncovalent binding. The spectral data of the separated chromophore at pH 0.5 agreed with that of a 6,7-substituted pteridine. After detachment with reducing agents, a pteridine in its 7k8-dihydro form was indicated by spectral analysis. AU - Ziegler, I. AU - Maier, K.C. AU - Fink, M.K. C1 - 41417 C2 - 38662 SP - 1567-1573 TI - Pteridine-binding α1-acid glycoprotein from blood of patients with neoplastic diseases. JO - Cancer Res. VL - 42 IS - 4 PY - 1982 SN - 0008-5472 ER -