Page last updated: 2024-10-21

1-anilino-8-naphthalenesulfonate and Disease Exacerbation

1-anilino-8-naphthalenesulfonate has been researched along with Disease Exacerbation in 106 studies

1-anilino-8-naphthalenesulfonate: RN given refers to parent cpd
8-anilinonaphthalene-1-sulfonic acid : A naphthalenesulfonic acid that is naphthalene-1-sulfonic acid substituted by a phenylamino group at position 8.

Research Excerpts

ExcerptRelevanceReference
"The addition of orlistat to a conventional weight loss regimen significantly improved oral glucose tolerance and diminished the rate of progression to the development of impaired glucose tolerance and type 2 diabetes."9.09Effects of weight loss with orlistat on glucose tolerance and progression to type 2 diabetes in obese adults. ( Boldrin, MN; Hauptman, J; Heymsfield, SB; Lucas, CP; Rissanen, A; Segal, KR; Sjöström, L; Wilding, JP, 2000)
"Acute pancreatitis was induced by limitation of pancreatic blood flow by clamping of inferior splenic artery for 30 min, followed by reperfusion."5.31Influence of leptin administration on the course of acute ischemic pancreatitis. ( Bilskl, J; Ceranowicz, P; Dembiński, A; Dembiński, M; Jaworek, J; Konturek, PC; Konturek, SJ; Warzecha, Z, 2002)
"The addition of orlistat to a conventional weight loss regimen significantly improved oral glucose tolerance and diminished the rate of progression to the development of impaired glucose tolerance and type 2 diabetes."5.09Effects of weight loss with orlistat on glucose tolerance and progression to type 2 diabetes in obese adults. ( Boldrin, MN; Hauptman, J; Heymsfield, SB; Lucas, CP; Rissanen, A; Segal, KR; Sjöström, L; Wilding, JP, 2000)
" Single nucleotide polymorphisms (SNPs) near the epidermal growth factor (EGF) (rs4444903), IL28B (rs12979860), and PNPLA3 (rs738409) loci are associated with treatment response, fibrosis, and hepatocellular carcinoma in non-transplant hepatitis C, but allograft population data are limited."3.83Impact of EGF, IL28B, and PNPLA3 polymorphisms on the outcome of allograft hepatitis C: a multicenter study. ( Chung, RT; Corey, KE; Curry, MP; Fuchs, BC; Gao, T; Gogela, NA; Gordon, FD; Johnson, KB; King, LY; Kothari, D; Lee, JH; Lin, MV; Misdraji, J; Mueller, JL; Nephew, LD; Simpson, MA; Tanabe, KK; Wei, L; Zheng, H, 2016)
"Nonalcoholic fatty liver disease (NAFLD) affects around a quarter of the global population, paralleling worldwide increases in obesity and metabolic syndrome."2.66Genetic contributions to NAFLD: leveraging shared genetics to uncover systems biology. ( Eslam, M; George, J, 2020)
"Nonalcoholic fatty liver disease (NAFLD), including nonalcoholic steatohepatitis (NASH), causes hepatic fibrosis, cirrhosis and hepatocellular carcinoma (HCC)."2.66Molecular Mechanisms: Connections between Nonalcoholic Fatty Liver Disease, Steatohepatitis and Hepatocellular Carcinoma. ( Goto, T; Hirotsu, Y; Kanda, T; Masuzaki, R; Moriyama, M; Omata, M, 2020)
"Indeed, acute pancreatitis, especially when presenting in severe forms with hyperstimulation of the pro-inflammatory response, may represent a crucial factor in the progression of COVID-19, entailing both an increase in hospitalization days and in mortality rate."2.66Involvement of the exocrine pancreas during COVID-19 infection and possible pathogenetic hypothesis: a concise review. ( De Biase, D; Fiorino, S; Gallo, C; Hong, W; Maccioni, F; Traversa, G; Zippi, M, 2020)
"Furthermore, NAFLD is believed to be involved in the pathogenesis of common disorders such as type 2 diabetes and cardiovascular disease."2.61Non-alcoholic fatty liver disease: causes, diagnosis, cardiometabolic consequences, and treatment strategies. ( Cusi, K; Häring, HU; Stefan, N, 2019)
"Nonalcoholic fatty liver disease (NAFLD), now the leading cause of liver damage worldwide, is epidemiologically associated with obesity, insulin resistance and type 2 diabetes, and is a potentially progressive condition to advanced liver fibrosis and hepatocellular carcinoma."2.58Genetics of Nonalcoholic Fatty Liver Disease: A 2018 Update. ( Baselli, GA; Valenti, LVC, 2018)
"Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease worldwide."2.58The genetic backgrounds in nonalcoholic fatty liver disease. ( Itoh, Y; Seko, Y; Yamaguchi, K, 2018)
"Both ARLD and NAFLD are multifactorial and refer to a spectrum of disease severity, ranging from steatosis through steatohepatitis to fibrosis and cirrhosis."2.58Genetics of alcoholic liver disease and non-alcoholic steatohepatitis. ( Anstee, QM; Scott, E, 2018)
"Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the United States and represents an increasingly important etiology of hepatocellular carcinoma (HCC) with annual cumulative incidence rates ranging from 2% to 12% in cohorts of NAFLD cirrhosis."2.53Hepatocellular carcinoma in patients with non-alcoholic fatty liver disease. ( Lim, JK; Nguyen, MH; Wong, CR, 2016)
"Nonalcoholic fatty liver disease (NAFLD) encompasses a spectrum that spans simple steatosis, through nonalcoholic steatohepatitis (NASH) to fibrosis and ultimately cirrhosis."2.52The Genetics of Nonalcoholic Fatty Liver Disease: Spotlight on PNPLA3 and TM6SF2. ( Anstee, QM; Day, CP, 2015)
"Nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of the metabolic syndrome and comprises a liver disease spectrum ranging from steatosis to nonalcoholic steatohepatitis (NASH) with risk of progression to liver cirrhosis and hepatocellular carcinoma (HCC)."2.52Challenges and Management of Liver Cirrhosis: Practical Issues in the Therapy of Patients with Cirrhosis due to NAFLD and NASH. ( Halilbasic, E; Hofer, H; Kazemi-Shirazi, L; Kienbacher, C; Munda, P; Rechling, C; Trauner, M; Traussnigg, S, 2015)
"Non-alcoholic fatty liver disease (NAFLD) is the most frequent chronic liver disease in Western countries, ranging from simple steatosis to steatohepatitis, cirrhosis, and hepatocellular cancer."2.50Role of metabolic lipases and lipolytic metabolites in the pathogenesis of NAFLD. ( Claudel, T; Fuchs, CD; Trauner, M, 2014)
"NAFLD is a disease spectrum ranging from simple steatosis, through steatohepatitis to fibrosis and, ultimately, cirrhosis."2.49The genetics of NAFLD. ( Anstee, QM; Day, CP, 2013)
"Nonalcoholic fatty liver disease (NAFLD) in most patients involves only simple hepatic steatosis; however, a minority develop progressive steatohepatitis."2.47Genetic determinants of susceptibility and severity in nonalcoholic fatty liver disease. ( Ballestri, S; Carulli, L; Daly, AK; Day, CP; Loria, P, 2011)
"Thirteen hospitalized dogs with acute pancreatitis diagnosed based on clinical signs, serum cPLI concentrations, and imaging findings were enrolled."1.62Serum concentrations of canine pancreatic lipase immunoreactivity and C-reactive protein for monitoring disease progression in dogs with acute pancreatitis. ( Fosgate, GT; Keany, KM; Perry, SM; Steiner, JM; Stroup, ST, 2021)
"Fibrosis progression in autoimmune hepatitis can be attenuated by immunosuppressive treatment; however, some patients progress despite therapy."1.56The PNPLA3 rs738409 GG genotype is associated with poorer prognosis in 239 patients with autoimmune hepatitis. ( Großhennig, A; Kirstein, MM; Manns, MP; Marhenke, S; Mederacke, I; Mederacke, YS; Metzler, F; Vogel, A, 2020)
"Non-alcoholic fatty liver disease (NAFLD) is a common chronic condition caused by the accumulation of fat in the liver."1.56Validating candidate biomarkers for different stages of non-alcoholic fatty liver disease. ( Al-Otaibi, M; Al-Qarni, R; Al-Saif, F; Alfadda, AA; Alkhalidi, H; Bamehriz, F; Hassanain, M; Iqbal, M, 2020)
"'Severe NAFLD' was defined as the presence of steatohepatitis, NAFLD activity score ≥4 or fibrosis stage ≥2."1.56Liver transcriptomics highlights interleukin-32 as novel NAFLD-related cytokine and candidate biomarker. ( Badiali, S; Baselli, GA; Dongiovanni, P; Fracanzani, AL; Maggioni, M; Mancina, RM; Maurotti, S; Meroni, M; Montalcini, T; Pelusi, S; Pingitore, P; Prati, D; Rametta, R; Romeo, S; Rossi, G; Taliento, AE; Valenti, L, 2020)
"Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in children and adolescents today."1.51Genetic determinants of steatosis and fibrosis progression in paediatric non-alcoholic fatty liver disease. ( Berndt, N; Bläker, H; Bufler, P; Cadenas, C; Golka, K; Hengstler, JG; Henning, S; Holzhütter, HG; Hudert, CA; Jansen, PLM; Loddenkemper, C; Meierhofer, D; Reinders, J; Rudolph, B; Selinski, S; Thielhorn, R; Wiegand, S, 2019)
"Non-alcoholic fatty liver disease (NAFLD) is a common chronic liver illness with a genetically heterogeneous background that can be accompanied by considerable morbidity and attendant health care costs."1.51GWAS and enrichment analyses of non-alcoholic fatty liver disease identify new trait-associated genes and pathways across eMERGE Network. ( Benoit, B; Carey, DJ; Carrell, DS; Carroll, RJ; Cobb, BL; Connolly, JJ; Crosslin, DR; Divanovic, S; Gharavi, AG; Hakonarson, H; Harley, ITW; Harley, JB; Huang, Y; Jarvik, GP; Kullo, IJ; Larson, EB; Li, R; Lingren, T; Mentch, FD; Murphy, S; Namjou, B; Niu, X; Pacheco, JA; Parameswaran, S; Ritchie, MD; Stanaway, IB; Verma, S; Wei, WQ; Williams, MS; Xanthakos, SA, 2019)
"In human breast cancer, high LIPG expression was observed in a limited subset of tumours and was significantly associated with shorter metastasis-free survival in node-negative, untreated patients."1.51LIPG-promoted lipid storage mediates adaptation to oxidative stress in breast cancer. ( Adawy, A; Cadenas, C; Claus, M; Edlund, K; Franckenstein, D; Frank, S; Gianmoena, K; Glotzbach, A; Grgas, K; Hayen, H; Hellwig, B; Hengstler, JG; Käfferlein, HU; Lesjak, MS; Madjar, K; Marchan, R; Mardinoglu, A; Micke, P; Pontén, F; Rahnenführer, J; Schmidt, M; Schriewer, A; Stewart, JD; Thaler, S; Vosbeck, S; Watzl, C; Zhang, C, 2019)
"Identifying NAFLD patients at risk of progression is crucial to orient medical care and resources."1.48Combining Genetic Variants to Improve Risk Prediction for NAFLD and Its Progression to Cirrhosis: A Proof of Concept Study. ( Antonelli-Incalzi, R; Baiocchini, A; Carotti, S; Cecere, R; De Vincentis, A; Del Nonno, F; Dell'Unto, C; Delle Monache, M; Galati, G; Gallo, P; Giannelli, V; Morini, S; Pellicelli, AM; Picardi, A; Rosati, D; Valentini, F; Vespasiani-Gentilucci, U, 2018)
"The ability of lipolysis in breast cancer cells were measured, as well as the expression of the rate-limiting lipase ATGL and fatty acid transporter FABP5."1.48Utilization of adipocyte-derived lipids and enhanced intracellular trafficking of fatty acids contribute to breast cancer progression. ( Li, Y; Ren, G; Sun, J; Tan, J; Tan, Y; Wang, Y; Xiang, T; Xing, L; Yang, D; Zeng, B, 2018)
"Non-alcoholic fatty liver disease (NAFLD) has a prevalence of approximately 30% in western countries, and is emerging as the first cause of liver cirrhosis and hepatocellular carcinoma (HCC)."1.48Promoting genetics in non-alcoholic fatty liver disease: Combined risk score through polymorphisms and clinical variables. ( Antonelli-Incalzi, R; Dell'Unto, C; Gallo, P; Picardi, A; Vespasiani-Gentilucci, U; Volpentesta, M, 2018)
"Advanced liver fibrosis was neither associated with PNPLA3 (p = 0."1.42The Impact of PNPLA3 rs738409 SNP on Liver Fibrosis Progression, Portal Hypertension and Hepatic Steatosis in HIV/HCV Coinfection. ( Aichelburg, MC; Bota, S; Bucsics, T; Ferenci, P; Grabmeier-Pfistershammer, K; Mandorfer, M; Payer, BA; Peck-Radosavljevic, M; Reiberger, T; Scheiner, B; Schwabl, P; Stättermayer, A; Trauner, M, 2015)
"Although most hepatocellular carcinoma (HCC) is related to chronic viral hepatitis or alcoholic liver disease, the incidence of NAFLD-related HCC is increasing."1.40Carriage of the PNPLA3 rs738409 C >G polymorphism confers an increased risk of non-alcoholic fatty liver disease associated hepatocellular carcinoma. ( Anstee, QM; Burt, AD; Daly, AK; Day, CP; Dufour, JF; Leathart, JB; Liu, YL; Patman, GL; Piguet, AC; Reeves, HL, 2014)
"Since hyperglycemia aggravates acute pancreatitis and also activates the receptor for advanced glycation endproducts (RAGE) in other organs, we explored if RAGE is expressed in the pancreas and if its expression is regulated during acute pancreatitis and hyperglycemia."1.39Impact of hyperglycemia and acute pancreatitis on the receptor for advanced glycation endproducts. ( Bürtin, F; Butschkau, A; Genz, B; Kroemer, T; Kuhla, A; Sempert, K; Timm, F; Vollmar, B; Zechner, D, 2013)
"The etiology of NAFLD is multifactorial."1.39PNPLA3, a genetic marker of progressive liver disease, still hiding its metabolic function? ( Burnol, AF; Dubuquoy, C; Moldes, M, 2013)
"Obesity is an emerging risk factor for chronic kidney disease (CKD) in the developed world."1.39Orlistat, an under-recognised cause of progressive renal impairment. ( Coutinho, AK; Glancey, GR, 2013)
"We identify BSSL as a marker for HIV-1 disease progression and emergence of X4 variants."1.38HIV-1 disease progression is associated with bile-salt stimulated lipase (BSSL) gene polymorphism. ( Bakker, M; Kootstra, NA; Paxton, WA; Pollakis, G; Stax, MJ; Tanck, MW; van 't Wout, AB, 2012)
"We defined various liver fibrosis phenotypes on the basis of METAVIR scores, with and without taking the duration of HCV infection into account."1.38Genome-wide association study identifies variants associated with progression of liver fibrosis from HCV infection. ( Abel, L; Argiro, L; Bibert, S; Bochud, PY; Boland, A; Booth, DR; Bourlière, M; Bousquet, L; Bréchot, C; Casanova, JL; Cerny, A; Dufour, JF; George, J; Guergnon, J; Halfon, P; Heim, MH; Hirsch, H; Jacobson, IM; Jouanguy, E; Kutalik, Z; Malinverni, R; Martinetti, G; Moradpour, D; Müllhaupt, B; Munteanu, M; Nalpas, B; Negro, F; Patin, E; Pol, S; Poynard, T; Rice, CM; Semela, D; Stewart, G; Suppiah, V; Talal, AH; Theodorou, I, 2012)
"The severity of acute pancreatitis was assessed 0 h or 1, 2, 3, 5 and 10 days after the last dose of cerulein."1.36Role of hormonal axis, growth hormone - IGF-1, in the therapeutic effect of ghrelin in the course of cerulein-induced acute pancreatitis. ( Ceranowicz, D; Ceranowicz, P; Cieszkowski, J; Dembinski, A; Kato, I; Kusnierz-Cabala, B; Kuwahara, A; Tomaszewska, R; Warzecha, Z, 2010)
" Clinically cardiac adverse events were defined as need for reoperation (n = 88; 46%), reintervention (n = 58; 30%), or angina (n = 89; 46%)."1.35Gene polymorphisms in APOE, NOS3, and LIPC genes may be risk factors for cardiac adverse events after primary CABG. ( Beiras-Fernandez, A; Eifert, S; Lohse, P; Nollert, G; Rasch, A; Reichart, B, 2009)
"To evaluate coronary atherosclerosis, we used CSI (coronary stenosis index) calculated from coronary angiograms."1.33CETP (cholesteryl ester transfer protein) promoter -1337 C>T polymorphism protects against coronary atherosclerosis in Japanese patients with heterozygous familial hypercholesterolaemia. ( Higashikata, T; Inazu, A; Katsuda, S; Kawashiri, MA; Kobayashi, J; Mabuchi, H; Miwa, K; Nohara, A; Takata, M; Yamagishi, M, 2006)
"Chronic pancreatitis is a common cause of exocrine pancreatic insufficiency (EPI) in humans and cats but is rarely recognised in dogs in which pancreatic acinar atrophy (PAA) is reportedly more common."1.32Exocrine pancreatic insufficiency as an end stage of pancreatitis in four dogs. ( Watson, PJ, 2003)
"Acute pancreatitis was induced by limitation of pancreatic blood flow by clamping of inferior splenic artery for 30 min, followed by reperfusion."1.31Influence of leptin administration on the course of acute ischemic pancreatitis. ( Bilskl, J; Ceranowicz, P; Dembiński, A; Dembiński, M; Jaworek, J; Konturek, PC; Konturek, SJ; Warzecha, Z, 2002)
"The following were recorded: disease progression (CDC class), nutritional status (weight Z-score), CD4 lymphocyte count, drug treatment during the previous 12 months, presence of opportunistic infections, clinical evidence of acute pancreatitis (increased serum pancreatic enzymes associated with vomiting, abdominal distention, and intolerance when eating)."1.30Serum pancreatic enzymes in human immunodeficiency virus-infected children. A collaborative study of the Italian Society of Pediatric Gastroenterology and Hepatology. ( Bastoni, K; Bavusotto, A; Boccia, MC; Canani, RB; Carroccio, A; Fontana, M; Guarino, A; Montalto, G; Spagnuolo, MI; Verghi, F; Zuin, G, 1998)

Research

Studies (106)

TimeframeStudies, this research(%)All Research%
pre-19900 (0.00)18.7374
1990's5 (4.72)18.2507
2000's15 (14.15)29.6817
2010's71 (66.98)24.3611
2020's15 (14.15)2.80

Authors

AuthorsStudies
Oliveira, CP1
Liu, Q1
Li, L2
Xu, D1
Zhu, J1
Huang, Z1
Yang, J2
Cheng, S1
Gu, Y1
Zheng, L1
Zhang, X2
Shen, H1
Eslam, M1
George, J2
Crisan, D1
Grigorescu, M1
Crisan, N1
Craciun, R1
Lupsor, M1
Radu, C1
Grigorescu, MD1
Suciu, A1
Epure, F1
Avram, L1
Leach, N1
Daijo, K1
Nakahara, T1
Inagaki, Y1
Nanba, M1
Nishida, Y1
Uchikawa, S1
Kodama, K1
Oya, K1
Morio, K1
Fujino, H1
Ono, A1
Murakami, E1
Yamauchi, M1
Kawaoka, T1
Miki, D1
Tsuge, M1
Hiramatsu, A1
Hayes, CN1
Imamura, M1
Aikata, H1
Ochi, H2
Chayama, K2
Baselli, GA2
Dongiovanni, P5
Rametta, R2
Meroni, M1
Pelusi, S1
Maggioni, M2
Badiali, S1
Pingitore, P2
Maurotti, S1
Montalcini, T2
Taliento, AE1
Prati, D1
Rossi, G1
Fracanzani, AL2
Mancina, RM3
Romeo, S3
Valenti, L6
Kanda, T1
Goto, T1
Hirotsu, Y1
Masuzaki, R1
Moriyama, M1
Omata, M1
Mederacke, YS1
Kirstein, MM2
Großhennig, A1
Marhenke, S2
Metzler, F1
Manns, MP1
Vogel, A2
Mederacke, I1
Attané, C1
Muller, C1
Al-Qarni, R1
Iqbal, M1
Al-Otaibi, M1
Al-Saif, F1
Alfadda, AA1
Alkhalidi, H1
Bamehriz, F1
Hassanain, M1
Lim, MA1
Pranata, R1
Jabłońska, J1
Kluska, M1
Lin, XJ1
Liu, R1
Li, C2
Yi, X1
Fu, B1
Walker, MJ1
Xu, XM1
Sun, G1
Lin, CH1
Lan, NSR1
Yeap, BB1
Fegan, PG1
Green, G1
Rankin, JM1
Dwivedi, G1
Habibi, A1
Karami, S1
Varmira, K1
Hadadi, M1
Zhang, M1
Li, Q1
Lan, X1
Li, X1
Zhang, Y2
Wang, Z1
Zheng, J1
Eskandari-Sedighi, G1
Cortez, LM1
Daude, N1
Shmeit, K1
Sim, V1
Westaway, D1
Weigand, S1
O'Connor, M1
Blažek, P1
Kantenwein, V1
Friedrich, L1
Grebmer, C1
Schaarschmidt, C1
von Olshausen, G1
Reents, T1
Deisenhofer, I1
Lennerz, C1
Kolb, C1
Iwata, H1
Sassa, N1
Kato, M2
Murase, Y1
Seko, S1
Kawanishi, H1
Hattori, R1
Gotoh, M1
Tsuzuki, T1
Chen, D1
Zhang, J1
Chen, YP2
Li, Y3
Zhou, M1
Waterhouse, GIN1
Sun, J2
Shi, W1
Ai, S1
Manne, ASN1
Hegde, AR1
Raut, SY1
Rao, RR1
Kulkarni, VI1
Mutalik, S1
Jafari, R1
Hectors, SJ1
Koehne de González, AK1
Spincemaille, P1
Prince, MR1
Brittenham, GM1
Wang, Y2
Banini, BA1
Kumar, DP1
Cazanave, S1
Seneshaw, M1
Mirshahi, F1
Santhekadur, PK1
Wang, L1
Guan, HP1
Oseini, AM1
Alonso, C1
Bedossa, P1
Koduru, SV1
Min, HK1
Sanyal, AJ1
Zippi, M1
Hong, W1
Traversa, G1
Maccioni, F1
De Biase, D1
Gallo, C1
Fiorino, S1
Gao, Y1
Xie, H1
Selen, ES1
Choi, J1
Wolfgang, MJ1
Keany, KM1
Fosgate, GT1
Perry, SM1
Stroup, ST1
Steiner, JM1
Gulley, JL1
Rajan, A1
Spigel, DR1
Iannotti, N1
Chandler, J1
Wong, DJL1
Leach, J1
Edenfield, WJ1
Wang, D1
Grote, HJ1
Heydebreck, AV1
Chin, K1
Cuillerot, JM1
Kelly, K1
Kawamura, Y1
Ikeda, K1
Arase, Y1
Fujiyama, S1
Hosaka, T1
Kobayashi, M2
Saitoh, S1
Sezaki, H1
Akuta, N1
Suzuki, F1
Suzuki, Y1
Kumada, H2
DeWeerdt, S1
Seko, Y1
Yamaguchi, K1
Itoh, Y1
Stickel, F1
Buch, S1
Nischalke, HD1
Weiss, KH1
Gotthardt, D1
Fischer, J1
Rosendahl, J1
Marot, A1
Elamly, M1
Casper, M1
Lammert, F1
McQuillin, A1
Zopf, S1
Spengler, U1
Eyer, F1
von Felden, J1
Wege, H1
Buch, T1
Schafmayer, C1
Braun, F1
Deltenre, P1
Berg, T1
Morgan, MY1
Hampe, J1
Lu, FB1
Hu, ED1
Xu, LM1
Chen, L1
Wu, JL1
Li, H1
Chen, DZ1
Jiménez-Sousa, MÁ1
Gómez-Moreno, AZ1
Pineda-Tenor, D1
Sánchez-Ruano, JJ1
Fernández-Rodríguez, A1
Artaza-Varasa, T1
Gómez-Sanz, A1
Martín-Vicente, M1
Vázquez-Morón, S1
Resino, S1
Scott, E1
Anstee, QM5
Vespasiani-Gentilucci, U2
Dell'Unto, C2
De Vincentis, A1
Baiocchini, A1
Delle Monache, M1
Cecere, R1
Pellicelli, AM1
Giannelli, V1
Carotti, S1
Galati, G1
Gallo, P2
Valentini, F1
Del Nonno, F1
Rosati, D1
Morini, S1
Antonelli-Incalzi, R2
Picardi, A2
Yang, D1
Xing, L1
Tan, Y1
Zeng, B1
Xiang, T1
Tan, J1
Ren, G1
Parajuli, N1
Takahara, S1
Matsumura, N1
Kim, TT1
Ferdaoussi, M1
Migglautsch, AK1
Zechner, R1
Breinbauer, R1
Kershaw, EE1
Dyck, JRB1
Wenzel, TJ1
Klegeris, A1
Stefan, N1
Häring, HU1
Cusi, K1
Garcia, MA1
Rojas, JA1
Millán, SP1
Flórez, AA1
Hu, SH1
Guang, Y1
Wang, WX1
Hudert, CA1
Selinski, S1
Rudolph, B1
Bläker, H1
Loddenkemper, C1
Thielhorn, R1
Berndt, N1
Golka, K1
Cadenas, C2
Reinders, J1
Henning, S1
Bufler, P1
Jansen, PLM1
Holzhütter, HG1
Meierhofer, D1
Hengstler, JG2
Wiegand, S1
Volpentesta, M1
Vosbeck, S1
Edlund, K1
Grgas, K1
Madjar, K1
Hellwig, B1
Adawy, A1
Glotzbach, A1
Stewart, JD1
Lesjak, MS1
Franckenstein, D1
Claus, M1
Hayen, H1
Schriewer, A1
Gianmoena, K1
Thaler, S1
Schmidt, M1
Micke, P1
Pontén, F1
Mardinoglu, A1
Zhang, C1
Käfferlein, HU1
Watzl, C1
Frank, S1
Rahnenführer, J1
Marchan, R1
Valenti, LVC1
Namjou, B1
Lingren, T1
Huang, Y1
Parameswaran, S1
Cobb, BL1
Stanaway, IB1
Connolly, JJ1
Mentch, FD1
Benoit, B1
Niu, X1
Wei, WQ1
Carroll, RJ1
Pacheco, JA1
Harley, ITW1
Divanovic, S1
Carrell, DS1
Larson, EB1
Carey, DJ1
Verma, S1
Ritchie, MD1
Gharavi, AG1
Murphy, S1
Williams, MS1
Crosslin, DR1
Jarvik, GP1
Kullo, IJ1
Hakonarson, H1
Li, R1
Xanthakos, SA1
Harley, JB1
Viganò, M1
Lampertico, P1
Facchetti, F1
Motta, BM1
D'Ambrosio, R2
Romagnoli, S1
Donati, B2
Fargion, S3
Colombo, M2
Calugaru, D1
Dai, Z1
Qi, W1
Lu, J1
Mao, Y1
Yao, Y1
Zhang, T1
Hong, H1
Li, S1
Zhou, T1
Yang, Z1
Yang, X1
Gao, G1
Cai, W1
Coutinho, AK1
Glancey, GR1
Day, CP5
Dunn, W1
O'Neil, M1
Zhao, J1
Wu, CH1
Roberts, B1
Chakraborty, S1
Sherman, C1
Weaver, B1
Taylor, R1
Olson, J1
Olyaee, M1
Gilroy, R1
Schmitt, T1
Wan, YJ1
Weinman, SA1
Zechner, D1
Sempert, K1
Genz, B1
Timm, F1
Bürtin, F1
Kroemer, T1
Butschkau, A1
Kuhla, A1
Vollmar, B1
Myer, JR1
Romach, EH1
Elangbam, CS1
Fares, R1
Lombardi, R1
Zhang, SK1
Cui, NQ1
Zhuo, YZ1
Li, DH1
Liu, JH1
Singal, AG1
Manjunath, H1
Yopp, AC1
Beg, MS1
Marrero, JA1
Gopal, P1
Waljee, AK1
Liu, YL1
Patman, GL1
Leathart, JB1
Piguet, AC1
Burt, AD1
Dufour, JF2
Daly, AK3
Reeves, HL1
Kitamoto, T1
Kitamoto, A1
Yoneda, M1
Hyogo, H1
Mizusawa, S1
Ueno, T1
Nakao, K2
Sekine, A1
Nakajima, A1
Hotta, K1
Mandorfer, M2
Payer, BA2
Schwabl, P2
Steiner, S1
Ferlitsch, A1
Aichelburg, MC2
Stättermayer, AF1
Ferenci, P2
Obermayer-Pietsch, B1
Grabmeier-Pfistershammer, K2
Trauner, M4
Peck-Radosavljevic, M2
Reiberger, T2
De Nicola, S1
Aghemo, A1
Cheroni, C1
Pedrazzini, M1
Marabita, F1
Donnici, L1
De Francesco, R1
Fuchs, CD1
Claudel, T1
Zampino, R1
Coppola, N1
Cirillo, G1
Boemio, A1
Grandone, A1
Stanzione, M1
Capoluongo, N1
Marrone, A1
Macera, M1
Sagnelli, E1
Adinolfi, LE1
del Giudice, EM1
Traussnigg, S1
Kienbacher, C1
Halilbasic, E1
Rechling, C1
Kazemi-Shirazi, L1
Hofer, H1
Munda, P1
Rembeck, K1
Lagging, M1
Tamaki, N1
Kurosaki, M1
Higuchi, M1
Takada, H1
Nakakuki, N1
Yasui, Y1
Suzuki, S1
Tsuchiya, K1
Nakanishi, H1
Itakura, J1
Takahashi, Y1
Ogawa, S1
Tanaka, Y1
Asahina, Y1
Izumi, N1
Spagnuolo, R1
Milano, M1
Brogneri, S1
Morrone, A1
Cosco, C1
Lazzaro, V1
Russo, C1
Ferro, Y1
Pujia, A1
Doldo, P1
Garieri, P1
Piodi, L1
Caprioli, F1
Xiao, W1
Jiang, W1
Shen, J1
Yin, G1
Fan, Y1
Wu, D1
Qiu, L1
Yu, G1
Xing, M1
Hu, G1
Wang, X1
Wan, R1
Scheiner, B1
Bucsics, T1
Bota, S1
Stättermayer, A1
Miyaaki, H1
Ichikawa, T1
Taura, N1
Miuma, S1
Honda, T1
Shibata, H1
Soyama, A1
Hidaka, M1
Takatsuki, M1
Eguchi, S1
Khlaiphuengsin, A1
Kiatbumrung, R1
Payungporn, S1
Pinjaroen, N1
Tangkijvanich, P1
Pieńkowska, J1
Gwoździewicz, K1
Skrobisz-Balandowska, K1
Marek, I1
Kostro, J1
Szurowska, E1
Studniarek, M1
Köpp, J1
Fleßa, S1
Lieb, W1
Markus, MRP1
Teumer, A1
Homuth, G1
Wallaschofski, H1
Marschall, P1
Völzke, H1
Baumeister, SE1
Mueller, JL1
King, LY1
Johnson, KB1
Gao, T1
Nephew, LD1
Kothari, D1
Simpson, MA1
Zheng, H1
Wei, L1
Corey, KE1
Misdraji, J1
Lee, JH1
Lin, MV1
Gogela, NA1
Fuchs, BC1
Tanabe, KK1
Gordon, FD1
Curry, MP1
Chung, RT2
Wong, CR1
Nguyen, MH1
Lim, JK1
Núñez-Torres, R1
Macías, J1
Mancebo, M1
Frías, M1
Dolci, G1
Téllez, F1
Merino, D1
Merchante, N1
Gómez-Mateos, J1
Guaraldi, G1
Rivero-Juárez, A1
Pineda, JA1
Real, LM1
Zhu, Y1
Dong, J1
Wang, P1
Huang, H1
Jin, X1
Zhou, J1
Shi, J1
Gu, G1
Chen, J1
Xu, J1
Song, Y1
Husain, SZ1
Srinath, AI1
Fujii, H1
Fukuda, A1
Tanaka, M1
Kojima, Y1
Ishida, T1
Hirata, K1
Fukagawa, M1
Muller, CA1
McArthur, N1
Belyaev, O1
Burr, W1
Werner, J1
Meiser, A1
Weyhe, D1
Büchler, MW2
Uhl, W2
Higuchi, N1
Enjoji, M1
Eifert, S1
Rasch, A1
Beiras-Fernandez, A1
Nollert, G1
Reichart, B1
Lohse, P1
Algül, H1
Speliotes, EK1
Butler, JL1
Palmer, CD1
Voight, BF1
Hirschhorn, JN1
Rotman, Y1
Koh, C1
Zmuda, JM1
Kleiner, DE1
Liang, TJ1
Koutsari, C1
Lazaridis, KN1
Ceranowicz, D1
Warzecha, Z2
Dembinski, A2
Ceranowicz, P2
Cieszkowski, J1
Kusnierz-Cabala, B1
Tomaszewska, R1
Kuwahara, A1
Kato, I1
Wu, JW1
Wang, SP1
Alvarez, F1
Casavant, S1
Gauthier, N1
Abed, L1
Soni, KG1
Yang, G1
Mitchell, GA1
Ballestri, S1
Carulli, L1
Loria, P1
Trépo, E1
Pradat, P1
Potthoff, A1
Momozawa, Y1
Quertinmont, E1
Gustot, T1
Lemmers, A1
Berthillon, P1
Amininejad, L1
Chevallier, M1
Schlué, J1
Kreipe, H1
Devière, J1
Manns, M1
Trépo, C1
Sninsky, J1
Wedemeyer, H1
Franchimont, D1
Moreno, C1
do O, NT1
Eurich, D1
Trautwein, C1
Neuhaus, P1
Neumann, UP1
Wasmuth, HE1
Schaefer, EA1
Stax, MJ1
Kootstra, NA1
van 't Wout, AB1
Tanck, MW1
Bakker, M1
Pollakis, G1
Paxton, WA1
Maggioni, P1
Piperno, A1
Pelucchi, S1
Mariani, R1
Patin, E1
Kutalik, Z1
Guergnon, J1
Bibert, S1
Nalpas, B1
Jouanguy, E1
Munteanu, M1
Bousquet, L1
Argiro, L1
Halfon, P1
Boland, A1
Müllhaupt, B1
Semela, D1
Heim, MH1
Moradpour, D1
Cerny, A1
Malinverni, R1
Hirsch, H1
Martinetti, G1
Suppiah, V1
Stewart, G1
Booth, DR1
Casanova, JL1
Bréchot, C1
Rice, CM1
Talal, AH1
Jacobson, IM1
Bourlière, M1
Theodorou, I1
Poynard, T1
Negro, F1
Pol, S1
Bochud, PY1
Abel, L1
Miyashita, M1
Ito, T1
Sakaki, M1
Kajiwara, A1
Nozawa, H1
Hiroishi, K1
Imawari, M1
Dubuquoy, C1
Burnol, AF1
Moldes, M1
Jaworek, J1
Konturek, PC1
Dembiński, M1
Bilskl, J1
Konturek, SJ1
Watson, PJ1
Todorova, B1
Kubaszek, A1
Pihlajamäki, J1
Lindström, J1
Eriksson, J1
Valle, TT1
Hämäläinen, H1
Ilanne-Parikka, P1
Keinänen-Kiukaanniemi, S1
Tuomilehto, J1
Uusitupa, M1
Laakso, M1
Whitelock, JM1
Iozzo, RV1
Yan, Q1
Yao, X1
Dai, LC1
Zhang, GL1
Ping, JL1
He, JF1
Han, CF1
Takata, M1
Inazu, A1
Katsuda, S1
Miwa, K1
Kawashiri, MA1
Nohara, A1
Higashikata, T1
Kobayashi, J1
Mabuchi, H1
Yamagishi, M1
Courtney, AE1
O'Rourke, DM1
Maxwell, AP1
MacLaughlin, H1
Macdougall, IC1
Lübbers, H1
Mahlke, R1
Lankisch, PG1
Moerland, M1
Samyn, H1
van Gent, T1
van Haperen, R1
Dallinga-Thie, G1
Grosveld, F1
van Tol, A1
de Crom, R1
Hofbauer, B1
Friess, H1
Weber, A1
Baczako, K1
Kisling, P1
Schilling, M1
Dervenis, C1
Moss, RL1
Musemeche, CA1
Feddersen, RM1
Vallance, BA1
Hewlett, BR1
Snider, DP1
Collins, SM1
Carroccio, A1
Fontana, M1
Spagnuolo, MI1
Zuin, G1
Montalto, G1
Canani, RB1
Verghi, F1
Bavusotto, A1
Bastoni, K1
Boccia, MC1
Guarino, A1
Yuasa, C1
Irimura, K1
Oka, T1
Heymsfield, SB1
Segal, KR1
Hauptman, J1
Lucas, CP1
Boldrin, MN1
Rissanen, A1
Wilding, JP1
Sjöström, L1

Clinical Trials (6)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
Association of Anesthesia Technique With Morbidity and Mortality in Patients With COVID-19 and Surgery for Hip Fracture: a Retrospective Population Cohort Study[NCT05133648]1,000 participants (Anticipated)Observational2023-01-05Active, not recruiting
PAveMenT: Phase Ib Study of Palbociclib and Avelumab in Metastatic AR+ Triple Negative Breast Cancer[NCT04360941]Phase 145 participants (Anticipated)Interventional2020-08-11Recruiting
A Phase I, Open-label, Multiple-ascending Dose Trial to Investigate the Safety, Tolerability, Pharmacokinetics, Biological and Clinical Activity of Avelumab (MSB0010718C) in Subjects With Metastatic or Locally Advanced Solid Tumors and Expansion to Select[NCT01772004]Phase 11,756 participants (Actual)Interventional2013-01-31Completed
The Role of Microbiome Reprogramming on Liver Fat Accumulation[NCT03914495]57 participants (Actual)Interventional2019-05-21Terminated (stopped due to PI carefully considered multiple factors and decided to close study to any further enrollment.)
Comparative Clinical Study to Evaluate the Possible Beneficial Effect of Empagliflozin Versus Pioglitazone on Non-diabetic Patients With Non-Alcoholic Steatohepatitis[NCT05605158]Phase 356 participants (Anticipated)Interventional2022-11-30Not yet recruiting
The Finnish Diabetes Prevention Study: A Follow-up Study on the Effect of a Dietary and Exercise Intervention in the Prevention of Diabetes and Its Vascular Complications[NCT00518167]522 participants (Actual)Interventional1993-11-30Active, not recruiting
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

Dose Escalation Cohort (Excluding Once Weekly Avelumab 10 mg/kg Cohort): Apparent Terminal Half-Life (t1/2) of Avelumab

Apparent terminal half-life was defined as the time required for the serum concentration of drug to decrease 50 percent in the final stage of its elimination. (NCT01772004)
Timeframe: Pre-infusion, at end of 1-hour infusion (Day 1), 0.5, 1, 2, 4, 6,12, 24, 36, 48 hours after end of infusion

InterventionHours (Median)
Dose Escalation Cohort: Avelumab 1.0 mg/kg61.425
Dose Escalation Cohort: Avelumab 3.0 mg/kg89.064
Dose Escalation Cohort: Avelumab 10.0 mg/kg97.440
Dose Escalation Cohort: Avelumab 20.0 mg/kg108.671

Dose Escalation Cohort (Excluding Once Weekly Avelumab 10 mg/kg Cohort): Area Under Serum Concentration-Time Curve From the Time of Dosing to the Time of the Last Observation (AUC0-t) of Avelumab

Area under the serum concentration versus time curve from time zero to the last sampling time t at which the concentration is at or above the lower limit of quantification (LLLQ). AUC(0-t) was calculated according to the mixed log-linear trapezoidal rule. (NCT01772004)
Timeframe: Pre-infusion, at end of 1-hour infusion (Day 1), 0.5, 1, 2, 4, 6,12, 24, 36, 48 hours after end of infusion

InterventionHours*micrograms per milliliter (Mean)
Dose Escalation Cohort: Avelumab 1.0 mg/kg1040
Dose Escalation Cohort: Avelumab 3.0 mg/kg6080
Dose Escalation Cohort: Avelumab 10.0 mg/kg22749.4
Dose Escalation Cohort: Avelumab 20.0 mg/kg45100

Dose Escalation Cohort (Excluding Once Weekly Avelumab 10 mg/kg Cohort): Area Under the Serum Concentration-Time Curve From Time Zero to Infinity (AUC0-infinity) of Avelumab

The AUC(0-inf) was estimated by determining the total area under the curve of the concentration versus time curve extrapolated to infinity. (NCT01772004)
Timeframe: Pre-infusion, at end of 1-hour infusion (Day 1), 0.5, 1, 2, 4, 6,12, 24, 36, 48 hours after end of infusion

InterventionHours*micrograms per milliliter (Mean)
Dose Escalation Cohort: Avelumab 1.0 mg/kg1290
Dose Escalation Cohort: Avelumab 3.0 mg/kg6850
Dose Escalation Cohort: Avelumab 10.0 mg/kg25920.9
Dose Escalation Cohort: Avelumab 20.0 mg/kg46600

Dose Escalation Cohort (Excluding Once Weekly Avelumab 10 mg/kg Cohort): Maximum Observed Serum Concentration (Cmax) of Avelumab

Cmax is the maximum observed serum concentration obtained directly from the concentration versus time curve. (NCT01772004)
Timeframe: Pre-infusion, at end of 1-hour infusion (Day 1), 0.5, 1, 2, 4, 6,12, 24, 36, 48 hours after end of infusion

InterventionMicrograms per milliliter (mcg/mL) (Mean)
Dose Escalation Cohort: Avelumab 1.0 mg/kg18.7
Dose Escalation Cohort: Avelumab 3.0 mg/kg81.9
Dose Escalation Cohort: Avelumab 10.0 mg/kg249.048
Dose Escalation Cohort: Avelumab 20.0 mg/kg489

Dose Escalation Cohort (Excluding Once Weekly Avelumab 10 mg/kg Cohort): Number of Participants Experiencing Dose Limiting Toxicities (DLTs)

DLT: defined using National Cancer Institute Common Toxicity Criteria for Adverse Events Version 4.0, as any one of following: any Grade (Gr) >=3toxicity that is possibly/probably/ definitely related to avelumab, except for any of following: Gr 3 infusion-related reaction resolving within 6 hours and controlled with medical management, Transient Gr 3 flu-like symptoms/fever, which is controlled with medical management, Transient Gr 3 fatigue, local reactions, headache, nausea, emesis that resolves to <= Gr 1, Gr3 diarrhea, Gr 3 skin toxicity, Gr 3 liver function test increase that resolves to <= Gr1 in < 7 days after medical management has been initiated, Single laboratory values out of normal range that were unlikely related to study treatment according to investigator, did not have any clinical correlate, and resolved to <= Gr1 within 7 days with adequate medical management and tumor flare phenomenon defined as local pain, irritation/rash localized at sites of known/suspected tumor. (NCT01772004)
Timeframe: Dose Escalation: Baseline up to Week 3

InterventionParticipants (Count of Participants)
Dose Escalation Cohort: Avelumab 1.0 mg/kg0
Dose Escalation Cohort: Avelumab 3.0 mg/kg0
Dose Escalation Cohort: Avelumab 10.0 mg/kg0
Dose Escalation Cohort: Avelumab 20.0 mg/kg1

Dose Escalation Cohort (Excluding Once Weekly Avelumab 10 mg/kg Cohort): Number of Participants With at Least 1 Positive Anti Drug Antibodies (ADA)

Serum samples were analyzed by a validated electrochemiluminescence immunoassay to detect the presence of antidrug antibodies (ADA). Number of participants with ADA positive results for Avelumab were reported. (NCT01772004)
Timeframe: Dose Escalation: Baseline up to Day 1023

InterventionParticipants (Count of Participants)
Dose Escalation Cohort: Avelumab 1.0 mg/kg0
Dose Escalation Cohort: Avelumab 3.0 mg/kg2
Dose Escalation Cohort: Avelumab 10.0 mg/kg0
Dose Escalation Cohort: Avelumab 20.0 mg/kg0

Dose Escalation Cohort (Excluding Once Weekly Avelumab 10 mg/kg Cohort): Time to Reach Maximum Observed Serum Concentration (Tmax) of Avelumab

Tmax is time to reach maximum observed serum concentration obtained directly from the concentration versus time curve. (NCT01772004)
Timeframe: Pre-infusion, at end of 1-hour infusion (Day 1), 0.5, 1, 2, 4, 6,12, 24, 36, 48 hours after end of infusion

InterventionHours (Median)
Dose Escalation Cohort: Avelumab 1.0 mg/kg1.500
Dose Escalation Cohort: Avelumab 3.0 mg/kg1.500
Dose Escalation Cohort: Avelumab 10.0 mg/kg1.500
Dose Escalation Cohort: Avelumab 20.0 mg/kg1.717

Dose Expansion Cohort: Duration of Response According to Modified Immune-Related Response Criteria (irRC) Per Investigator Assessment

Duration of response according to modified irRC, per investigator assessment was calculated for each participant with a confirmed response (immune-related complete response [irCR] or immune-related partial response [irPR]) as the time from the first observation of response to the first observation of documented disease progression (or death within 12 weeks of the last tumor assessment). irCR: Complete disappearance of all tumor lesions (both index and non-index lesions with no new measurable/unmeasurable lesions). irPR: At least 30% reduction from baseline in the sum of the longest diameter (SLD) of all lesions). Results were calculated based on Kaplan-Meier estimates. (NCT01772004)
Timeframe: Dose Expansion: Baseline up to Day 2023

InterventionMonths (Median)
Primary Expansion Cohort: NSCLC, Post-platinum Doublet21.13
Primary Expansion Cohort: NSCLC, First LineNA
Primary Expansion Cohort: Metastatic Breast Cancer8.31
Primary Expansion Cohort: GC/GEJC Progressed4.14
Primary Expansion Cohort: GC/GEJC Non Progressed22.23
Secondary Expansion Cohort: Adrenocortical CarcinomaNA
Secondary Expansion Cohort: MelanomaNA
Secondary Expansion Cohort: Mesothelioma15.21
Secondary Expansion Cohort: Urothelial CarcinomaNA
Secondary Expansion Cohort: Ovarian CancerNA
Secondary Expansion Cohort: Renal Cell Carcinoma (First Line)10.61
Secondary Expansion Cohort: Renal Cell Carcinoma (Second Line)NA
Efficacy Expansion Cohort: Ovarian CancerNA
Efficacy Expansion Cohort: Urothelial CarcinomaNA
Efficacy Expansion Cohort: GC/GEJC, Third LineNA
Efficacy Expansion Cohort: HNSCCNA

Dose Expansion Cohort: Duration of Response According to Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1 Per Investigator Assessment

Duration of response according to RECIST 1.1, per investigator assessment was calculated for each participant with a confirmed response (complete response [CR] or partial response [PR]) as the time from the first observation of response to the first observation of documented disease progression (or death within 12 weeks of the last tumor assessment). CR: Disappearance of all evidence of target and non-target lesions. PR: At least 30 percent (%) reduction from baseline in the sum of the longest diameter (SLD) of all lesions. Results were calculated based on Kaplan-Meier estimates. (NCT01772004)
Timeframe: Dose Expansion: Baseline up to Day 2023

InterventionMonths (Median)
Primary Expansion Cohort: NSCLC, Post-platinum Doublet17.48
Primary Expansion Cohort: NSCLC, First Line12.02
Primary Expansion Cohort: Metastatic Breast Cancer8.33
Primary Expansion Cohort: GC/GEJC Progressed3.48
Primary Expansion Cohort: GC/GEJC Non Progressed21.42
Secondary Expansion Cohort: Adrenocortical Carcinoma8.41
Secondary Expansion Cohort: MelanomaNA
Secondary Expansion Cohort: Mesothelioma15.21
Secondary Expansion Cohort: Urothelial CarcinomaNA
Secondary Expansion Cohort: Ovarian Cancer10.38
Secondary Expansion Cohort: Renal Cell Carcinoma (First Line)9.94
Secondary Expansion Cohort: Renal Cell Carcinoma (Second Line)NA
Efficacy Expansion Cohort: Ovarian CancerNA
Efficacy Expansion Cohort: Urothelial CarcinomaNA
Efficacy Expansion Cohort: GC/GEJC, Third LineNA
Efficacy Expansion Cohort: HNSCCNA

Dose Expansion Cohort: Immune Related Progression-Free Survival (irPFS) Time According to Modified Immune-Related Response Criteria (irRC)

The irPFS time was defined as the time from first administration of study treatment until first documentation of immune-related progressive disease (irPD) or death when death occurred within 12 weeks of the last tumor assessment or first administration of study treatment (whichever was later). irPD: sum of the longest diameters of target and new measurable lesions increases greater than or equal to [>=] 20%, confirmed by a repeat, consecutive observations at least 4 weeks from the date first documented. The analysis of irPFS will be performed with a Kaplan-Meier method. Data for immune related progression-free survival time has been reported. (NCT01772004)
Timeframe: Dose Expansion: Baseline up to Day 2023

InterventionMonths (Median)
Primary Expansion Cohort: NSCLC, Post-platinum Doublet4.04
Primary Expansion Cohort: NSCLC, First Line6.93
Primary Expansion Cohort: Metastatic Breast Cancer1.64
Primary Expansion Cohort: GC/GEJC Progressed1.81
Primary Expansion Cohort: GC/GEJC Non Progressed4.14
Secondary Expansion Cohort: Colorectal Cancer2.79
Secondary Expansion Cohort: Castrate-resistant Prostate CancerNA
Secondary Expansion Cohort: Adrenocortical Carcinoma3.81
Secondary Expansion Cohort: Melanoma6.83
Secondary Expansion Cohort: Mesothelioma6.18
Secondary Expansion Cohort: Urothelial Carcinoma4.07
Secondary Expansion Cohort: Ovarian Cancer4.04
Secondary Expansion Cohort: Renal Cell Carcinoma (First Line)8.34
Secondary Expansion Cohort: Renal Cell Carcinoma (Second Line)6.90
Efficacy Expansion Cohort: Ovarian Cancer2.60
Efficacy Expansion Cohort: Urothelial Carcinoma2.46
Efficacy Expansion Cohort: GC/GEJC, Third Line1.35
Efficacy Expansion Cohort: HNSCC2.83

Dose Expansion Cohort: Number of Participants With Atleast 1 Positive Anti Drug Antibodies (ADA) Assay

Serum samples were analyzed by a validated electrochemiluminescence immunoassay to detect the presence of antidrug antibodies (ADA). Number of participants with ADA positive results for Avelumab were reported. (NCT01772004)
Timeframe: Dose Expansion: Baseline up to Day 2023

InterventionParticipants (Count of Participants)
Primary Expansion Cohort: NSCLC, Post-platinum Doublet17
Primary Expansion Cohort: NSCLC, First Line9
Primary Expansion Cohort: Metastatic Breast Cancer17
Primary Expansion Cohort: GC/GEJC Progressed3
Primary Expansion Cohort: GC/GEJC Non Progressed6
Secondary Expansion Cohort: Colorectal Cancer0
Secondary Expansion Cohort: Castrate-resistant Prostate Cancer0
Secondary Expansion Cohort: Adrenocortical Carcinoma4
Secondary Expansion Cohort: Melanoma5
Secondary Expansion Cohort: Mesothelioma3
Secondary Expansion Cohort: Urothelial Carcinoma2
Secondary Expansion Cohort: Ovarian Cancer5
Secondary Expansion Cohort: Renal Cell Carcinoma (First Line)5
Secondary Expansion Cohort: Renal Cell Carcinoma (Second Line)3
Efficacy Expansion Cohort: Ovarian Cancer5
Efficacy Expansion Cohort: Urothelial Carcinoma17
Efficacy Expansion Cohort: GC/GEJC, Third Line6
Efficacy Expansion Cohort: HNSCC5

Dose Expansion Cohort: Overall Survival (OS) Time

Overall survival time was measured as time in months first administration of trial treatment to death. The analysis of OS time was performed with a Kaplan-Meier method. (NCT01772004)
Timeframe: Dose Expansion: Baseline up to Day 2023

InterventionMonths (Median)
Primary Expansion Cohort: NSCLC, Post-platinum Doublet8.57
Primary Expansion Cohort: NSCLC, First Line14.23
Primary Expansion Cohort: Metastatic Breast Cancer8.38
Primary Expansion Cohort: GC/GEJC Progressed6.64
Primary Expansion Cohort: GC/GEJC Non Progressed11.07
Secondary Expansion Cohort: Colorectal Cancer11.20
Secondary Expansion Cohort: Castrate-resistant Prostate Cancer19.32
Secondary Expansion Cohort: Adrenocortical Carcinoma10.55
Secondary Expansion Cohort: Melanoma17.22
Secondary Expansion Cohort: Mesothelioma10.71
Secondary Expansion Cohort: Urothelial Carcinoma13.70
Secondary Expansion Cohort: Ovarian Cancer11.17
Secondary Expansion Cohort: Renal Cell Carcinoma (First Line)NA
Secondary Expansion Cohort: Renal Cell Carcinoma (Second Line)16.85
Efficacy Expansion Cohort: Ovarian Cancer9.13
Efficacy Expansion Cohort: Urothelial Carcinoma6.97
Efficacy Expansion Cohort: GC/GEJC, Third Line3.35
Efficacy Expansion Cohort: HNSCC7.98

Dose Expansion Cohort: Progression-Free Survival (PFS) Time According to Response Evaluation Criteria in Solid Tumors Version (RECIST) 1.1

The PFS time (based on investigator assessments), according to the RECIST 1.1, was defined as the time from first administration of study treatment until first documentation of progressive disease (PD) or death when death occurred within 12 weeks of the last tumor assessment or first administration of study treatment (whichever was later). PD was defined as at least a 20% increase in the sum of longest diameter (SLD), taking as reference the smallest SLD recorded from baseline or the appearance of 1 or more new lesions and unequivocal progression of non-target lesions. The analysis of PFS was performed with a Kaplan-Meier method. (NCT01772004)
Timeframe: Dose Expansion: Baseline up to Day 2023

InterventionMonths (Median)
Primary Expansion Cohort: NSCLC, Post-platinum Doublet2.66
Primary Expansion Cohort: NSCLC, First Line4.04
Primary Expansion Cohort: Metastatic Breast Cancer1.35
Primary Expansion Cohort: GC/GEJC Progressed1.38
Primary Expansion Cohort: GC/GEJC Non Progressed2.76
Secondary Expansion Cohort: Colorectal Cancer1.41
Secondary Expansion Cohort: Castrate-resistant Prostate Cancer5.39
Secondary Expansion Cohort: Adrenocortical Carcinoma2.56
Secondary Expansion Cohort: Melanoma3.06
Secondary Expansion Cohort: Mesothelioma4.11
Secondary Expansion Cohort: Urothelial Carcinoma2.69
Secondary Expansion Cohort: Ovarian Cancer2.60
Secondary Expansion Cohort: Renal Cell Carcinoma (First Line)8.28
Secondary Expansion Cohort: Renal Cell Carcinoma (Second Line)5.55
Efficacy Expansion Cohort: Ovarian Cancer1.45
Efficacy Expansion Cohort: Urothelial Carcinoma1.41
Efficacy Expansion Cohort: GC/GEJC, Third Line1.31
Efficacy Expansion Cohort: HNSCC1.77

Efficacy Expansion Cohorts: Duration of Response According to Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1 as Per Independent Endpoint Review Committee (IERC)

Duration of response according to modified irRC, per investigator assessment was calculated for each participant with a confirmed response (immune-related complete response [irCR] or immune-related partial response [irPR]) as the time from the first observation of response to the first observation of documented disease progression (or death within 12 weeks of the last tumor assessment). irCR: Complete disappearance of all tumor lesions (both index and non-index lesions with no new measurable/unmeasurable lesions). irPR: At least 30% reduction from baseline in the sum of the longest diameter (SLD) of all lesions). Results were calculated based on Kaplan-Meier estimates. (NCT01772004)
Timeframe: Efficacy Expansion: Baseline up to Day 1072

InterventionMonths (Median)
Efficacy Expansion Cohort: Ovarian CancerNA
Efficacy Expansion Cohort: Urothelial CarcinomaNA
Efficacy Expansion Cohort: GC/GEJC, Third LineNA
Efficacy Expansion Cohort: HNSCCNA

Efficacy Expansion Cohorts: Progression-Free Survival (PFS) Time According to Response Evaluation Criteria in Solid Tumors Version (RECIST) 1.1 as Per Independent Endpoint Review Committee (IERC)

The PFS time (based on IERC), according to the RECIST 1.1, was defined as the time from first administration of study treatment until first documentation of progressive disease (PD) or death when death occurred within 12 weeks of the last tumor assessment or first administration of study treatment (whichever was later). PD was defined as at least a 20% increase in the sum of longest diameter (SLD), taking as reference the smallest SLD recorded from baseline or the appearance of 1 or more new lesions and unequivocal progression of non-target lesions. The analysis of PFS was performed with a Kaplan-Meier method. (NCT01772004)
Timeframe: Efficacy Expansion: Baseline up to Day 1072

InterventionMonths (Median)
Efficacy Expansion Cohort: Ovarian Cancer1.87
Efficacy Expansion Cohort: Urothelial Carcinoma1.45
Efficacy Expansion Cohort: GC/GEJC, Third Line1.31
Efficacy Expansion Cohort: HNSCC1.41

Dose Escalation and Expansion Cohorts: Number of Participants With Treatment-Emergent Adverse Events (TEAEs) and TEAEs as Per Severity

Adverse event(AE): any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with use of study drug, whether or not related to study drug. A serious adverse event(SAE) was an AE that resulted in any of following outcomes: death; life threatening; persistent/significant disability/incapacity; initial or prolonged inpatient hospitalization; congenital anomaly/birth defect or was otherwise considered medically important. Treatment-emergent events were events between first dose of study drug that were absent before treatment or that worsened relative to pre-treatment state up to 30 days after last administration.TEAEs included both Serious TEAEs and non-serious TEAEs. Severity of TEAEs were graded using National Cancer Institute-Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 4.0 toxicity grades, as follows: Grade 1= Mild, Grade 2= Moderate, Grade 3= Severe, Grade 4= Life-threatening and Grade 5= Death. (NCT01772004)
Timeframe: Up to Day 2511

,,,,,,,,,,,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
Participants with TEAEsParticipants with TEAEs with Grade 1 severityParticipants with TEAEs with Grade 2 severityParticipants with TEAEs with Grade 3 severityParticipants with TEAEs with Grade 4 severityParticipants with TEAEs with Grade 5 severityParticipants with TEAEs with missing Grade
Dose Escalation Cohort: Avelumab 1.0 mg/kg4002020
Dose Escalation Cohort: Avelumab 10.0 mg/kg15146220
Dose Escalation Cohort: Avelumab 10.0 mg/kg Weekly8233000
Dose Escalation Cohort: Avelumab 20.0 mg/kg212611110
Dose Escalation Cohort: Avelumab 3.0 mg/kg13144040
Efficacy Expansion Cohort: GC/GEJC, Third Line1302305112350
Efficacy Expansion Cohort: HNSCC14914435412251
Efficacy Expansion Cohort: Ovarian Cancer103730456150
Efficacy Expansion Cohort: Urothelial Carcinoma20415468216450
Primary Expansion Cohort: GC/GEJC Non Progressed881419414100
Primary Expansion Cohort: GC/GEJC Progressed6059297100
Primary Expansion Cohort: Metastatic Breast Cancer16122584512240
Primary Expansion Cohort: NSCLC, First Line1563456815250
Primary Expansion Cohort: NSCLC, Post-platinum Doublet18211546619320
Secondary Expansion Cohort: Adrenocortical Carcinoma5011025860
Secondary Expansion Cohort: Castrate-resistant Prostate Cancer17557000
Secondary Expansion Cohort: Colorectal Cancer21296130
Secondary Expansion Cohort: Melanoma5021721550
Secondary Expansion Cohort: Mesothelioma5322123340
Secondary Expansion Cohort: Ovarian Cancer1221152423140
Secondary Expansion Cohort: Renal Cell Carcinoma (First Line)6272520640
Secondary Expansion Cohort: Renal Cell Carcinoma (Second Line)191510120
Secondary Expansion Cohort: Urothelial Carcinoma4431018670

Dose Escalation and Expansion Cohorts: Number of Participants With Treatment-Related Treatment-Emergent Adverse Events (TEAEs) and Treatment-Related TEAEs as Per Severity

"AE was defined as any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of study drug, whether or not related to study drug. Treatment-emergent events were the events between first dose of study drug that were absent before treatment or that worsened relative to pre-treatment state up to 30 days after last administration. TEAEs included both Serious TEAEs and non-serious TEAEs. Treatment related AE was defined as having a Possible or Related relationship to study treatment, as assessed by the Investigator. Severity of Treatment-Related TEAEs were graded using NCI-CTCAE version 4.0 toxicity grades, as follows: Grade 1= Mild, Grade 2= Moderate, Grade 3= Severe, Grade 4= Life-threatening and Grade 5= Death." (NCT01772004)
Timeframe: Baseline up to Day 2511

,,,,,,,,,,,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
Participants with Treatment-Related TEAEsParticipants with Treatment-Related TEAEs with Grade 1 severityParticipants with Treatment-Related TEAEs with Grade 2 severityParticipants with Treatment-Related TEAEs with Grade 3 severityParticipants with Treatment-Related TEAEs with Grade 4 severityParticipants with Treatment-Related TEAEs with Grade 5 severityParticipants with Treatment-Related TEAEs with missing Grade
Dose Escalation Cohort: Avelumab 1.0 mg/kg3111000
Dose Escalation Cohort: Avelumab 10.0 mg/kg14733100
Dose Escalation Cohort: Avelumab 10.0 mg/kg Weekly7241000
Dose Escalation Cohort: Avelumab 20.0 mg/kg17682100
Dose Escalation Cohort: Avelumab 3.0 mg/kg9540000
Efficacy Expansion Cohort: GC/GEJC, Third Line71253311110
Efficacy Expansion Cohort: HNSCC8339349100
Efficacy Expansion Cohort: Ovarian Cancer6530268100
Efficacy Expansion Cohort: Urothelial Carcinoma144507020310
Primary Expansion Cohort: GC/GEJC Non Progressed5727227010
Primary Expansion Cohort: GC/GEJC Progressed281494100
Primary Expansion Cohort: Metastatic Breast Cancer118524613520
Primary Expansion Cohort: NSCLC, First Line109296017300
Primary Expansion Cohort: NSCLC, Post-platinum Doublet146467121800
Secondary Expansion Cohort: Adrenocortical Carcinoma4112218000
Secondary Expansion Cohort: Castrate-resistant Prostate Cancer15861000
Secondary Expansion Cohort: Colorectal Cancer16493000
Secondary Expansion Cohort: Melanoma3915204000
Secondary Expansion Cohort: Mesothelioma438303200
Secondary Expansion Cohort: Ovarian Cancer8646317200
Secondary Expansion Cohort: Renal Cell Carcinoma (First Line)5120233500
Secondary Expansion Cohort: Renal Cell Carcinoma (Second Line)14491000
Secondary Expansion Cohort: Urothelial Carcinoma3211182100

Dose Escalation Cohort (Excluding Once Weekly Avelumab 10 mg/kg Cohort): Number of Participants With Immune Related Best Overall Response (irBOR) According to Modified Immune-Related Response Criteria (irRC)

irBOR defined as best response of any of immune related complete response (irCR), immune related partial response (irPR), immune related stable disease (irSD) and immune related progressive disease (irPD) recorded from baseline until immune related disease progression and determined according to modified irRC per investigator assessment. irCR: Complete disappearance of all tumor lesions (both index and non-index lesions with no new measurable/unmeasurable lesions). irPR: At least 30% reduction from baseline in the sum of the longest diameter (SLD) of all lesions). irSD: SLD of target and new measurable lesions neither irCR, irPR, or irPD. irPD: SLD of target and new measurable lesions increases greater than or equal to [>=] 20%, confirmed by a repeat, consecutive observations at least 4 weeks from the date first documented. Number of participants with immune-related best overall response in each category (irCR, irPR, irSD, irPD) was reported. (NCT01772004)
Timeframe: Dose Escalation: Baseline up to Day 1023

,,,
InterventionParticipants (Count of Participants)
Immune-related Complete ResponseImmune-related Partial ResponseImmune-related Stable DiseaseImmune-related Progressive DiseaseNot Evaluable
Dose Escalation Cohort: Avelumab 1.0 mg/kg00202
Dose Escalation Cohort: Avelumab 10.0 mg/kg01842
Dose Escalation Cohort: Avelumab 20.0 mg/kg011415
Dose Escalation Cohort: Avelumab 3.0 mg/kg00850

Dose Escalation Cohort (Excluding Once Weekly Avelumab 10 mg/kg Cohort): Programmed Death Ligand 1 (PD-L1) Receptor Occupancy

Percentage of PD-L1 receptors occupied by avelumab on human lymphocytes (CD3+ T-cells) was assessed by flow cytometry on peripheral blood mononuclear cell (PBMC) samples. Greater than or equal to [>=] 85 percent [%] of cell viability was required for reliable receptor occupancy assessment. (NCT01772004)
Timeframe: Pre-infusion on Day 1; 48 hours after infusion on Day 3; Pre-infusion on Days 15, 43, and 85

InterventionPercentage of receptors (Mean)
Day 15
Dose Escalation Cohort: Avelumab 10.0 mg/kg93.2

Dose Escalation Cohort (Excluding Once Weekly Avelumab 10 mg/kg Cohort): Programmed Death Ligand 1 (PD-L1) Receptor Occupancy

Percentage of PD-L1 receptors occupied by avelumab on human lymphocytes (CD3+ T-cells) was assessed by flow cytometry on peripheral blood mononuclear cell (PBMC) samples. Greater than or equal to [>=] 85 percent [%] of cell viability was required for reliable receptor occupancy assessment. (NCT01772004)
Timeframe: Pre-infusion on Day 1; 48 hours after infusion on Day 3; Pre-infusion on Days 15, 43, and 85

InterventionPercentage of receptors (Mean)
Day 1Day 3Day 15
Dose Escalation Cohort: Avelumab 20.0 mg/kg0.084.785.0

Dose Escalation Cohort (Excluding Once Weekly Avelumab 10 mg/kg Cohort): Programmed Death Ligand 1 (PD-L1) Receptor Occupancy

Percentage of PD-L1 receptors occupied by avelumab on human lymphocytes (CD3+ T-cells) was assessed by flow cytometry on peripheral blood mononuclear cell (PBMC) samples. Greater than or equal to [>=] 85 percent [%] of cell viability was required for reliable receptor occupancy assessment. (NCT01772004)
Timeframe: Pre-infusion on Day 1; 48 hours after infusion on Day 3; Pre-infusion on Days 15, 43, and 85

InterventionPercentage of receptors (Mean)
Day 1Day 3Day 15Day 43
Dose Escalation Cohort: Avelumab 3.0 mg/kg0.080.190.096.8

Dose Escalation Cohort (Excluding Once Weekly Avelumab 10 mg/kg Cohort): Programmed Death Ligand 1 (PD-L1) Receptor Occupancy

Percentage of PD-L1 receptors occupied by avelumab on human lymphocytes (CD3+ T-cells) was assessed by flow cytometry on peripheral blood mononuclear cell (PBMC) samples. Greater than or equal to [>=] 85 percent [%] of cell viability was required for reliable receptor occupancy assessment. (NCT01772004)
Timeframe: Pre-infusion on Day 1; 48 hours after infusion on Day 3; Pre-infusion on Days 15, 43, and 85

InterventionPercentage of receptors (Mean)
Day 3Day 15Day 43Day 85
Dose Escalation Cohort: Avelumab 1.0 mg/kg92.575.730.319.8

Dose Escalation Cohort: Number of Participants With Best Overall Response (BOR) According to Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1

BOR was determined according to RECIST v1.1 and as per investigator assessment. BOR is defined as the best response of any of complete response (CR), partial response (PR), stable disease (SD) and progressive disease (PD) recorded from date of randomization until disease progression or recurrence (taking the smallest measurement recorded since start of treatment as reference). CR: Disappearance of all evidence of target and non-target lesions. PR: At least 30 percent (%) reduction from baseline in the sum of the longest diameter (SLD) of all lesions. SD =Neither sufficient increase to qualify for PD nor sufficient shrinkage to qualify for PR. PD is defined as at least a 20 % increase in the SLD, taking as reference the smallest SLD recorded from baseline or appearance of 1 or more new lesions and unequivocal progression of non-target lesions. Number of participants with best overall response in each category (CR, PR, SD, PD) was reported. (NCT01772004)
Timeframe: Dose Escalation: Baseline up to Day 2511

,,,,
InterventionParticipants (Count of Participants)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseNot Evaluable
Dose Escalation Cohort: Avelumab 1.0 mg/kg00202
Dose Escalation Cohort: Avelumab 10.0 mg/kg01842
Dose Escalation Cohort: Avelumab 10.0 mg/kg Weekly01412
Dose Escalation Cohort: Avelumab 20.0 mg/kg011433
Dose Escalation Cohort: Avelumab 3.0 mg/kg00850

Dose Expansion Cohort (Secondary Urothelial Carcinoma Cohort): Number of Participants With Confirmed Best Overall Response According to Response Evaluation Criteria in Solid Tumors Version 1.1 as Adjudicated by an Independent Endpoint Review Committee

Confirmed Best Overall Response (BOR) was determined according to Response Evaluation Criteria in Solid Tumors (RECIST) v1.1and as adjudicated by an Independent Endpoint Review Committee (IERC) is defined as best response of any of complete response (CR), partial response (PR), stable disease (SD) and progressive disease (PD) recorded from date of randomization until disease progression/recurrence (taking smallest measurement recorded since start of treatment as reference). CR: Disappearance of all evidence of target/non-target lesions. PR: At least 30% reduction from baseline in sum of longest diameter (SLD) of all lesions. SD: Neither sufficient increase to qualify for PD nor sufficient shrinkage to qualify for PR. PD: at least a 20% increase in SLD, taking as reference smallest SLD recorded from baseline/appearance of 1or more new lesions and unequivocal progression of non-target lesions. Number of participants with BOR in each category (CR, PR, SD, PD) were reported. (NCT01772004)
Timeframe: Secondary Urothelial Carcinoma Dose Expansion: Baseline up to Day 931

InterventionParticipants (Count of Participants)
Complete ResponsePartial ResponseStable DiseaseNon-CR/Non-PDProgressive DiseaseNot Evaluable
Secondary Expansion Cohort: Urothelial Carcinoma61160165

Dose Expansion Cohort: Number of Participants With Best Overall Response (BOR) According to Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1

BOR was determined according to RECIST v1.1 and as per investigator assessment. BOR is defined as the best response of any of complete response (CR), partial response (PR), stable disease (SD) and progressive disease (PD) recorded from date of randomization until disease progression or recurrence (taking the smallest measurement recorded since start of treatment as reference). CR: Disappearance of all evidence of target and non-target lesions. PR: At least 30 percent (%) reduction from baseline in the sum of the longest diameter (SLD) of all lesions. SD = Neither sufficient increase to qualify for PD nor sufficient shrinkage to qualify for PR. PD is defined as at least a 20 % increase in the SLD, taking as reference the smallest SLD recorded from baseline or appearance of 1 or more new lesions and unequivocal progression of non-target lesions. Number of participants with best overall response in each category (CR, PR, SD, PD) was reported. (NCT01772004)
Timeframe: Dose Expansion: Baseline up to Day 2023

,,,,,,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseNot Evaluable
Efficacy Expansion Cohort: GC/GEJC, Third Line17238021
Efficacy Expansion Cohort: HNSCC515506617
Efficacy Expansion Cohort: Ovarian Cancer34374613
Efficacy Expansion Cohort: Urothelial Carcinoma724458735
Primary Expansion Cohort: GC/GEJC Non Progressed24452910
Primary Expansion Cohort: GC/GEJC Progressed0413367
Primary Expansion Cohort: Metastatic Breast Cancer144110715
Primary Expansion Cohort: NSCLC, First Line328684017
Primary Expansion Cohort: NSCLC, Post-platinum Doublet224666824
Secondary Expansion Cohort: Adrenocortical Carcinoma0321224
Secondary Expansion Cohort: Castrate-resistant Prostate Cancer001053
Secondary Expansion Cohort: Colorectal Cancer00993
Secondary Expansion Cohort: Melanoma4716177
Secondary Expansion Cohort: Mesothelioma1426184
Secondary Expansion Cohort: Ovarian Cancer11153519
Secondary Expansion Cohort: Renal Cell Carcinoma (First Line)1938113
Secondary Expansion Cohort: Renal Cell Carcinoma (Second Line)021341
Secondary Expansion Cohort: Urothelial Carcinoma2420144

Dose Expansion Cohort: Number of Participants With Immune Related Best Overall Response (irBOR) According to Modified Immune-Related Response Criteria (irRC)

irBOR defined as best response of any of immune related complete response (irCR), immune related partial response (irPR), immune related stable disease (irSD) and immune related progressive disease (irPD) recorded from baseline until immune related disease progression and determined according to modified irRC per investigator assessment. irCR: Complete disappearance of all tumor lesions (both index and non-index lesions with no new measurable/unmeasurable lesions). irPR: At least 30% reduction from baseline in the sum of the longest diameter (SLD) of all lesions). irSD: SLD of target and new measurable lesions neither irCR, irPR, or irPD. irPD: SLD of target and new measurable lesions increases greater than or equal to [>=] 20%, confirmed by a repeat, consecutive observations at least 4 weeks from the date first documented. Number of participants with immune-related best overall response in each category (irCR, irPR, irSD, irPD) was reported. (NCT01772004)
Timeframe: Dose Expansion: Baseline up to Day 2023

,,,,,,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
Immune-related Complete ResponseImmune-related Partial ResponseImmune-related Stable DiseaseImmune-related Progressive DiseaseNot Evaluable
Efficacy Expansion Cohort: GC/GEJC, Third Line17296233
Efficacy Expansion Cohort: HNSCC615614427
Efficacy Expansion Cohort: Ovarian Cancer34443319
Efficacy Expansion Cohort: Urothelial Carcinoma1024616241
Primary Expansion Cohort: GC/GEJC Non Progressed2453031
Primary Expansion Cohort: GC/GEJC Progressed0520035
Primary Expansion Cohort: Metastatic Breast Cancer16606932
Primary Expansion Cohort: NSCLC, First Line431782122
Primary Expansion Cohort: NSCLC, Post-platinum Doublet226803640
Secondary Expansion Cohort: Adrenocortical Carcinoma0326147
Secondary Expansion Cohort: Castrate-resistant Prostate Cancer003114
Secondary Expansion Cohort: Colorectal Cancer00957
Secondary Expansion Cohort: Melanoma4720119
Secondary Expansion Cohort: Mesothelioma1431107
Secondary Expansion Cohort: Ovarian Cancer115612721
Secondary Expansion Cohort: Renal Cell Carcinoma (First Line)1104065
Secondary Expansion Cohort: Renal Cell Carcinoma (Second Line)021512
Secondary Expansion Cohort: Urothelial Carcinoma2521106

Dose Expansion Cohort: Number of Participants With Positive Programmed Death Receptor-1 Ligand-1 (PD-L1) Biomarker Expression in Tumor Tissue

PD-L1 assessment was performed using immunohistochemistry. PD-L1 expression status was classified as positive or negative based on the following cut-offs: For tumor cells: Participants were considered PD-L1 expression positive (negative): - if at least (less than) 5% of the tumor cells show PD-L1 membrane staining >= 1+, respectively. This was used as the primary cut-off; - if at least (less than) 25% of the tumor cells show PD-L1 membrane staining >=2+, respectively. This was considered as secondary cut-off; - if at least (less than) 1% of the tumor cells show PD-L1 membrane staining >=1+, respectively. This was used as the tertiary cut-off; - if at least (less than) 50% of the tumor cells show PD-L1 membrane staining >=1+, respectively. This was used as the '50% cut-off'; - if at least (less than) 80% of the tumor cells show PD-L1 membrane staining ≥1+, respectively. This was used as the '80% cut-off'. (NCT01772004)
Timeframe: Dose Expansion: Baseline up to Day 2023

,
InterventionParticipants (Count of Participants)
PD-L1 expression status - 1% cutoff
Primary Expansion Cohort: GC/GEJC Progressed20
Primary Expansion Cohort: GC/GEJC Non Progressed26

Dose Expansion Cohort: Number of Participants With Positive Programmed Death Receptor-1 Ligand-1 (PD-L1) Biomarker Expression in Tumor Tissue

PD-L1 assessment was performed using immunohistochemistry. PD-L1 expression status was classified as positive or negative based on the following cut-offs: For tumor cells: Participants were considered PD-L1 expression positive (negative): - if at least (less than) 5% of the tumor cells show PD-L1 membrane staining >= 1+, respectively. This was used as the primary cut-off; - if at least (less than) 25% of the tumor cells show PD-L1 membrane staining >=2+, respectively. This was considered as secondary cut-off; - if at least (less than) 1% of the tumor cells show PD-L1 membrane staining >=1+, respectively. This was used as the tertiary cut-off; - if at least (less than) 50% of the tumor cells show PD-L1 membrane staining >=1+, respectively. This was used as the '50% cut-off'; - if at least (less than) 80% of the tumor cells show PD-L1 membrane staining ≥1+, respectively. This was used as the '80% cut-off'. (NCT01772004)
Timeframe: Dose Expansion: Baseline up to Day 2023

InterventionParticipants (Count of Participants)
PD-L1 expression status - 1% cutoffPD-L1 expression status - 5% cutoffPD-L1 expression status - 25% cutoff
Efficacy Expansion Cohort: GC/GEJC, Third Line39166

Dose Expansion Cohort: Number of Participants With Positive Programmed Death Receptor-1 Ligand-1 (PD-L1) Biomarker Expression in Tumor Tissue

PD-L1 assessment was performed using immunohistochemistry. PD-L1 expression status was classified as positive or negative based on the following cut-offs: For tumor cells: Participants were considered PD-L1 expression positive (negative): - if at least (less than) 5% of the tumor cells show PD-L1 membrane staining >= 1+, respectively. This was used as the primary cut-off; - if at least (less than) 25% of the tumor cells show PD-L1 membrane staining >=2+, respectively. This was considered as secondary cut-off; - if at least (less than) 1% of the tumor cells show PD-L1 membrane staining >=1+, respectively. This was used as the tertiary cut-off; - if at least (less than) 50% of the tumor cells show PD-L1 membrane staining >=1+, respectively. This was used as the '50% cut-off'; - if at least (less than) 80% of the tumor cells show PD-L1 membrane staining ≥1+, respectively. This was used as the '80% cut-off'. (NCT01772004)
Timeframe: Dose Expansion: Baseline up to Day 2023

,
InterventionParticipants (Count of Participants)
PD-L1 expression status - 1% cutoffPD-L1 expression status - 5% cutoffPD-L1 expression status - 25% cutoffPD-L1 expression status - 50% cutoff
Secondary Expansion Cohort: Colorectal Cancer5101
Primary Expansion Cohort: Metastatic Breast Cancer8725313

Dose Expansion Cohort: Number of Participants With Positive Programmed Death Receptor-1 Ligand-1 (PD-L1) Biomarker Expression in Tumor Tissue

PD-L1 assessment was performed using immunohistochemistry. PD-L1 expression status was classified as positive or negative based on the following cut-offs: For tumor cells: Participants were considered PD-L1 expression positive (negative): - if at least (less than) 5% of the tumor cells show PD-L1 membrane staining >= 1+, respectively. This was used as the primary cut-off; - if at least (less than) 25% of the tumor cells show PD-L1 membrane staining >=2+, respectively. This was considered as secondary cut-off; - if at least (less than) 1% of the tumor cells show PD-L1 membrane staining >=1+, respectively. This was used as the tertiary cut-off; - if at least (less than) 50% of the tumor cells show PD-L1 membrane staining >=1+, respectively. This was used as the '50% cut-off'; - if at least (less than) 80% of the tumor cells show PD-L1 membrane staining ≥1+, respectively. This was used as the '80% cut-off'. (NCT01772004)
Timeframe: Dose Expansion: Baseline up to Day 2023

,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
PD-L1 expression status - 1% cutoffPD-L1 expression status - 5% cutoffPD-L1 expression status - 25% cutoffPD-L1 expression status - 50% cutoffPD-L1 expression status - 80% cutoff
Efficacy Expansion Cohort: HNSCC10793485128
Efficacy Expansion Cohort: Ovarian Cancer4523640
Efficacy Expansion Cohort: Urothelial Carcinoma8772253425
Primary Expansion Cohort: NSCLC, First Line8876505338
Primary Expansion Cohort: NSCLC, Post-platinum Doublet12284535441
Secondary Expansion Cohort: Adrenocortical Carcinoma1512352
Secondary Expansion Cohort: Castrate-resistant Prostate Cancer00000
Secondary Expansion Cohort: Melanoma1915782
Secondary Expansion Cohort: Mesothelioma2216852
Secondary Expansion Cohort: Ovarian Cancer7632322
Secondary Expansion Cohort: Renal Cell Carcinoma (First Line)2011210
Secondary Expansion Cohort: Renal Cell Carcinoma (Second Line)41000
Secondary Expansion Cohort: Urothelial Carcinoma1413553

Dose Expansion Phase: Minimum Serum Post-dose (Ctrough) Concentration of Avelumab

Serum Ctrough concentration of Avelumab is reported. (NCT01772004)
Timeframe: At Day 15, 29, 43, 57, 71, 85, 99, 127 and 169

InterventionMicrogram per milliliter (Mean)
Day 15Day 29Day 43Day 85Day 127
Secondary Expansion Cohort: Castrate-resistant Prostate Cancer28.831.632.337.944.2

Dose Expansion Phase: Minimum Serum Post-dose (Ctrough) Concentration of Avelumab

Serum Ctrough concentration of Avelumab is reported. (NCT01772004)
Timeframe: At Day 15, 29, 43, 57, 71, 85, 99, 127 and 169

InterventionMicrogram per milliliter (Mean)
Day 15Day 29Day 43Day 57Day 71Day 85
Efficacy Expansion Cohort: GC/GEJC, Third Line18.118.619.816.219.619.3

Dose Expansion Phase: Minimum Serum Post-dose (Ctrough) Concentration of Avelumab

Serum Ctrough concentration of Avelumab is reported. (NCT01772004)
Timeframe: At Day 15, 29, 43, 57, 71, 85, 99, 127 and 169

InterventionMicrogram per milliliter (Mean)
Day 15Day 29Day 43Day 85Day 127Day 169
Secondary Expansion Cohort: Colorectal Cancer22.425.523.28.0334.210.5

Dose Expansion Phase: Minimum Serum Post-dose (Ctrough) Concentration of Avelumab

Serum Ctrough concentration of Avelumab is reported. (NCT01772004)
Timeframe: At Day 15, 29, 43, 57, 71, 85, 99, 127 and 169

,,,,
InterventionMicrogram per milliliter (Mean)
Day 15Day 29Day 43Day 57Day 71Day 85Day 169
Efficacy Expansion Cohort: HNSCC20.127.029.030.631.135.632.5
Efficacy Expansion Cohort: Ovarian Cancer22.323.927.728.530.431.228.3
Efficacy Expansion Cohort: Urothelial Carcinoma17.621.325.224.124.228.037.3
Secondary Expansion Cohort: Renal Cell Carcinoma (Second Line)25.127.429.425.527.834.443.6
Secondary Expansion Cohort: Renal Cell Carcinoma (First Line)26.935.735.739.838.439.747.1

Dose Expansion Phase: Minimum Serum Post-dose (Ctrough) Concentration of Avelumab

Serum Ctrough concentration of Avelumab is reported. (NCT01772004)
Timeframe: At Day 15, 29, 43, 57, 71, 85, 99, 127 and 169

,,,,,,,
InterventionMicrogram per milliliter (Mean)
Day 15Day 29Day 43Day 57Day 71Day 85Day 127Day 169
Primary Expansion Cohort: Metastatic Breast Cancer23.726.529.728.929.928.933.131.9
Primary Expansion Cohort: NSCLC, Post-platinum Doublet20.423.524.626.227.927.836.936.6
Secondary Expansion Cohort: Melanoma22.323.829.329.928.835.658.635.3
Secondary Expansion Cohort: Mesothelioma21.720.723.618.221.023.725.928.7
Secondary Expansion Cohort: Ovarian Cancer23.127.728.427.333.439.140.749.3
Primary Expansion Cohort: GC/GEJC (Progressed/Non Progressed)22.525.628.326.528.129.739.131.4
Secondary Expansion Cohort: Adrenocortical Carcinoma17.817.320.622.222.024.928.824.7
Secondary Expansion Cohort: Urothelial Carcinoma21.326.228.129.729.434.735.738.8

Dose Expansion Phase: Minimum Serum Post-dose (Ctrough) Concentration of Avelumab

Serum Ctrough concentration of Avelumab is reported. (NCT01772004)
Timeframe: At Day 15, 29, 43, 57, 71, 85, 99, 127 and 169

InterventionMicrogram per milliliter (Mean)
Day 15Day 29Day 43Day 57Day 71Day 85Day 99Day 169
Primary Expansion Cohort: NSCLC, First Line18.323.523.823.431.027.728.839.1

Dose Expansion Phase: Serum Concentration at End of Infusion (CEOI) of Avelumab

Serum concentration at end of infusion (CEOI) of Avelumab is reported. (NCT01772004)
Timeframe: At Day 1, 15, 29, 43, 85, 127 and 169

InterventionMicrogram per milliliter (Mean)
Day 1Day 43Day 85
Efficacy Expansion Cohort: GC/GEJC, Third Line241240237

Dose Expansion Phase: Serum Concentration at End of Infusion (CEOI) of Avelumab

Serum concentration at end of infusion (CEOI) of Avelumab is reported. (NCT01772004)
Timeframe: At Day 1, 15, 29, 43, 85, 127 and 169

,,,,
InterventionMicrogram per milliliter (Mean)
Day 1Day 43Day 85Day 169
Efficacy Expansion Cohort: HNSCC212249255245
Efficacy Expansion Cohort: Ovarian Cancer247268263233
Efficacy Expansion Cohort: Urothelial Carcinoma224241247313
Primary Expansion Cohort: NSCLC, First Line246250266304
Secondary Expansion Cohort: Renal Cell Carcinoma (Second Line)239255252235

Dose Expansion Phase: Serum Concentration at End of Infusion (CEOI) of Avelumab

Serum concentration at end of infusion (CEOI) of Avelumab is reported. (NCT01772004)
Timeframe: At Day 1, 15, 29, 43, 85, 127 and 169

,
InterventionMicrogram per milliliter (Mean)
Day 1Day 15Day 29Day 43Day 85Day 127Day 169
Secondary Expansion Cohort: Colorectal Cancer272297287306216269272
Secondary Expansion Cohort: Castrate-resistant Prostate Cancer343305291294348339287

Efficacy Expansion Cohort (GC/GEJC, Third Line): Number of Participants With Confirmed Best Overall Response (BOR) as Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1), as Adjudicated by Independent Endpoint Review Committee (IERC)

Confirmed BOR was determined according to RECIST 1.1 and as adjudicated by an Independent Endpoint Review Committee (IERC) and defined as best response of any of complete response (CR), partial response (PR), stable disease (SD) and progressive disease (PD) recorded from date of randomization until disease progression/recurrence (taking smallest measurement recorded since start of treatment as reference). CR: Disappearance of all evidence of target/non-target lesions. PR: At least 30% reduction from baseline in sum of longest diameter (SLD) of all lesions. SD: Neither sufficient increase to qualify for PD nor sufficient shrinkage to qualify for PR. PD: at least a 20% increase in SLD, taking as reference smallest SLD recorded from baseline/appearance of 1 or more new lesions and unequivocal progression of non-target lesions. (NCT01772004)
Timeframe: GC/GEJC, Third Line Efficacy Expansion: Baseline up to Day 871

InterventionParticipants (Count of Participants)
Complete response (CR)Partial response (PR)Stable disease (SD)Progressive disease (PD)Non-CR/Non-PDNon-evaluable
Efficacy Expansion Cohort: GC/GEJC, Third Line252472524

Efficacy Expansion Cohort (HNSCC): Number of Participants With Confirmed Best Overall Response (BOR) as Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1), as Adjudicated by Independent Endpoint Review Committee (IERC)

Confirmed BOR was determined according to RECIST 1.1 and as adjudicated by an Independent Endpoint Review Committee (IERC) and defined as best response of any of complete response (CR), partial response (PR), stable disease (SD) and progressive disease (PD) recorded from date of randomization until disease progression/recurrence (taking smallest measurement recorded since start of treatment as reference). CR: Disappearance of all evidence of target/non-target lesions. PR: At least 30%reduction from baseline in sum of longest diameter (SLD) of all lesions. SD: Neither sufficient increase to qualify for PD nor sufficient shrinkage to qualify for PR. PD: at least a 20% increase in SLD, taking as reference smallest SLD recorded from baseline/appearance of 1or more new lesions and unequivocal progression of non-target lesions. (NCT01772004)
Timeframe: HNSCC Efficacy Expansion: Baseline up to Day 1072

InterventionParticipants (Count of Participants)
Complete response (CR)Partial response (PR)Stable disease (SD)Progressive disease (PD)Non-CR/Non-PDNon-evaluable
Efficacy Expansion Cohort: HNSCC2124667125

Efficacy Expansion Cohort (Ovarian Cancer): Number of Participants With Confirmed Best Overall Response (BOR) as Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1), as Adjudicated by Independent Endpoint Review Committee (IERC)

Confirmed BOR was determined according to RECIST 1.1 and as adjudicated by an Independent Endpoint Review Committee (IERC) and defined as best response of any of complete response (CR), partial response (PR), stable disease (SD) and progressive disease (PD) recorded from date of randomization until disease progression/recurrence (taking smallest measurement recorded since start of treatment as reference). CR: Disappearance of all evidence of target/non-target lesions. PR: At least 30%reduction from baseline in sum of longest diameter (SLD) of all lesions. SD: Neither sufficient increase to qualify for PD nor sufficient shrinkage to qualify for PR. PD: at least a 20% increase in SLD, taking as reference smallest SLD recorded from baseline/appearance of 1or more new lesions and unequivocal progression of non-target lesions. (NCT01772004)
Timeframe: Ovarian Cancer Efficacy Expansion: Baseline up to Day 620

InterventionParticipants (Count of Participants)
Complete response (CR)Partial response (PR)Stable disease (SD)Progressive disease (PD)Non-CR/Non-PDNon-evaluable
Efficacy Expansion Cohort: Ovarian Cancer314139316

Efficacy Expansion Cohort(Urothelial Carcinoma): Number of Participants With Confirmed Best Overall Response (BOR) as Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1), as Adjudicated by Independent Endpoint Review Committee (IERC)

Confirmed BOR was determined according to RECIST 1.1 and as adjudicated by an Independent Endpoint Review Committee (IERC) and defined as best response of any of complete response (CR), partial response (PR), stable disease (SD) and progressive disease (PD) recorded from date of randomization until disease progression/recurrence (taking smallest measurement recorded since start of treatment as reference). CR: Disappearance of all evidence of target/non-target lesions. PR: At least 30% reduction from baseline in sum of longest diameter (SLD) of all lesions. SD: Neither sufficient increase to qualify for PD nor sufficient shrinkage to qualify for PR. PD: at least a 20% increase in SLD, taking as reference smallest SLD recorded from baseline/appearance of 1 or more new lesions and unequivocal progression of non-target lesions. (NCT01772004)
Timeframe: Urothelial Carcinoma Efficacy Expansion: Baseline up to Day 931

InterventionParticipants (Count of Participants)
Complete response (CR)Partial response (PR)Stable disease (SD)Progressive disease (PD)Non-CR/Non-PDNon-evaluable
Efficacy Expansion Cohort: Urothelial Carcinoma6265177137

Primary Expansion Cohorts: Number of Participants With Unconfirmed Response at Week 13 According to Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1

The response criteria evaluation was carried out according to Response Evaluation Criteria in Solid Tumors (RECIST) 1.1. CR and PR did not need to be confirmed by a subsequent tumor assessment due to blinded central assessment. CR: Disappearance of all target lesions since baseline; PR: At least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum of diameters; SD: Neither sufficient increase to qualify for PD nor sufficient shrinkage to qualify for PR and PD: at least a 20% increase in SLD, taking as reference smallest SLD recorded from baseline/appearance of 1or more new lesions and unequivocal progression of non-target lesions. Number of participants with unconfirmed response at week 13 according to response evaluation criteria in solid tumors (RECIST) version 1.1 were reported. (NCT01772004)
Timeframe: Week 13

,,,,
InterventionParticipants (Count of Participants)
Unconfirmed Complete ResponseUnconfirmed Partial ResponseUnconfirmed Stable DiseaseUnconfirmed Progressive DiseaseNon-evaluable
Primary Expansion Cohort: GC/GEJC Non Progressed2544318
Primary Expansion Cohort: GC/GEJC Progressed0611376
Primary Expansion Cohort: Metastatic Breast Cancer173810715
Primary Expansion Cohort: NSCLC, First Line124542849
Primary Expansion Cohort: NSCLC, Post-platinum Doublet228626824

Reviews

23 reviews available for 1-anilino-8-naphthalenesulfonate and Disease Exacerbation

ArticleYear
Genetic contributions to NAFLD: leveraging shared genetics to uncover systems biology.
    Nature reviews. Gastroenterology & hepatology, 2020, Volume: 17, Issue:1

    Topics: 17-Hydroxysteroid Dehydrogenases; Acyltransferases; Adaptor Proteins, Signal Transducing; Carcinoma,

2020
Molecular Mechanisms: Connections between Nonalcoholic Fatty Liver Disease, Steatohepatitis and Hepatocellular Carcinoma.
    International journal of molecular sciences, 2020, Feb-23, Volume: 21, Issue:4

    Topics: 17-Hydroxysteroid Dehydrogenases; Animals; Apoptosis; Carcinoma, Hepatocellular; Diet, High-Fat; Dis

2020
Drilling for Oil: Tumor-Surrounding Adipocytes Fueling Cancer.
    Trends in cancer, 2020, Volume: 6, Issue:7

    Topics: Adipocytes; Disease Progression; Energy Metabolism; Extracellular Vesicles; Fatty Acids, Nonesterifi

2020
Involvement of the exocrine pancreas during COVID-19 infection and possible pathogenetic hypothesis: a concise review.
    Le infezioni in medicina, 2020, Dec-01, Volume: 28, Issue:4

    Topics: COVID-19; Disease Progression; Humans; Hyperamylasemia; Lipase; Pancreas, Exocrine; Pancreatitis; SA

2020
The genetic backgrounds in nonalcoholic fatty liver disease.
    Clinical journal of gastroenterology, 2018, Volume: 11, Issue:2

    Topics: Adaptor Proteins, Signal Transducing; Disease Progression; Genetic Background; Humans; Lipase; Membr

2018
The relationship between obesity and the severity of non-alcoholic fatty liver disease: systematic review and meta-analysis.
    Expert review of gastroenterology & hepatology, 2018, Volume: 12, Issue:5

    Topics: Adult; Aged; Disease Progression; Genetic Predisposition to Disease; Humans; Lipase; Membrane Protei

2018
Genetics of alcoholic liver disease and non-alcoholic steatohepatitis.
    Clinical medicine (London, England), 2018, 04-01, Volume: 18, Issue:Suppl 2

    Topics: Acyltransferases; Disease Progression; Genetic Association Studies; Genetic Predisposition to Diseas

2018
Novel multi-target directed ligand-based strategies for reducing neuroinflammation in Alzheimer's disease.
    Life sciences, 2018, Aug-15, Volume: 207

    Topics: Alzheimer Disease; Amyloid beta-Peptides; Animals; Anti-Inflammatory Agents; Cathepsin B; Dinoprosto

2018
Non-alcoholic fatty liver disease: causes, diagnosis, cardiometabolic consequences, and treatment strategies.
    The lancet. Diabetes & endocrinology, 2019, Volume: 7, Issue:4

    Topics: Antioxidants; Bariatric Surgery; Carcinoma, Hepatocellular; Cardiovascular Diseases; Diabetes Mellit

2019
Non-alcoholic fatty liver disease: causes, diagnosis, cardiometabolic consequences, and treatment strategies.
    The lancet. Diabetes & endocrinology, 2019, Volume: 7, Issue:4

    Topics: Antioxidants; Bariatric Surgery; Carcinoma, Hepatocellular; Cardiovascular Diseases; Diabetes Mellit

2019
Non-alcoholic fatty liver disease: causes, diagnosis, cardiometabolic consequences, and treatment strategies.
    The lancet. Diabetes & endocrinology, 2019, Volume: 7, Issue:4

    Topics: Antioxidants; Bariatric Surgery; Carcinoma, Hepatocellular; Cardiovascular Diseases; Diabetes Mellit

2019
Non-alcoholic fatty liver disease: causes, diagnosis, cardiometabolic consequences, and treatment strategies.
    The lancet. Diabetes & endocrinology, 2019, Volume: 7, Issue:4

    Topics: Antioxidants; Bariatric Surgery; Carcinoma, Hepatocellular; Cardiovascular Diseases; Diabetes Mellit

2019
Genetics of Nonalcoholic Fatty Liver Disease: A 2018 Update.
    Current pharmaceutical design, 2018, Volume: 24, Issue:38

    Topics: Apolipoproteins B; Disease Progression; Gene-Environment Interaction; Humans; Insulin Resistance; Li

2018
The genetics of NAFLD.
    Nature reviews. Gastroenterology & hepatology, 2013, Volume: 10, Issue:11

    Topics: Disease Progression; Epigenomics; Fatty Liver; Genome-Wide Association Study; Humans; Lipase; Membra

2013
PNPLA3 I148M polymorphism and progressive liver disease.
    World journal of gastroenterology, 2013, Nov-07, Volume: 19, Issue:41

    Topics: Carcinoma, Hepatocellular; Cholangitis, Sclerosing; Disease Progression; Fatty Liver; Fatty Liver, A

2013
The effect of PNPLA3 on fibrosis progression and development of hepatocellular carcinoma: a meta-analysis.
    The American journal of gastroenterology, 2014, Volume: 109, Issue:3

    Topics: Carcinoma, Hepatocellular; Disease Progression; Genetic Predisposition to Disease; Humans; Lipase; L

2014
Role of metabolic lipases and lipolytic metabolites in the pathogenesis of NAFLD.
    Trends in endocrinology and metabolism: TEM, 2014, Volume: 25, Issue:11

    Topics: Animals; Disease Progression; Humans; Lipase; Lipolysis; Liver; Non-alcoholic Fatty Liver Disease; R

2014
Challenges and Management of Liver Cirrhosis: Practical Issues in the Therapy of Patients with Cirrhosis due to NAFLD and NASH.
    Digestive diseases (Basel, Switzerland), 2015, Volume: 33, Issue:4

    Topics: Anticholesteremic Agents; Antioxidants; Bariatric Surgery; Carcinoma, Hepatocellular; Disease Progre

2015
Impact of IL28B, ITPA and PNPLA3 genetic variants on therapeutic outcome and progression of hepatitis C virus infection.
    Pharmacogenomics, 2015, Volume: 16, Issue:10

    Topics: Disease Progression; Genetic Variation; Hepacivirus; Hepatitis C; Humans; Interferons; Interleukins;

2015
The Genetics of Nonalcoholic Fatty Liver Disease: Spotlight on PNPLA3 and TM6SF2.
    Seminars in liver disease, 2015, Volume: 35, Issue:3

    Topics: Animals; Disease Progression; Genetic Association Studies; Genetic Markers; Genetic Predisposition t

2015
Genetics of Alcoholic Liver Disease.
    Seminars in liver disease, 2015, Volume: 35, Issue:4

    Topics: Alcohol Drinking; Alcoholism; Carcinoma, Hepatocellular; Disease Progression; Ethanol; Fatty Liver,

2015
Hepatocellular carcinoma in patients with non-alcoholic fatty liver disease.
    World journal of gastroenterology, 2016, Oct-07, Volume: 22, Issue:37

    Topics: Adiponectin; Carcinoma, Hepatocellular; Disease Progression; Humans; Immune System; Inflammation; Li

2016
What's unique about acute pancreatitis in children: risk factors, diagnosis and management.
    Nature reviews. Gastroenterology & hepatology, 2017, Volume: 14, Issue:6

    Topics: Abdominal Pain; Acute Disease; Amylases; Antioxidants; Carrier Proteins; Child; Cholangiopancreatogr

2017
Genetic determinants of susceptibility and severity in nonalcoholic fatty liver disease.
    Expert review of gastroenterology & hepatology, 2011, Volume: 5, Issue:2

    Topics: Animals; Cytokines; Disease Progression; Fatty Liver; Female; Genetic Predisposition to Disease; Gen

2011
The impact of human gene polymorphisms on HCV infection and disease outcome.
    Seminars in liver disease, 2011, Volume: 31, Issue:4

    Topics: Antiviral Agents; Disease Progression; Genetic Predisposition to Disease; Genotype; Hepacivirus; Hep

2011
Heparan sulfate: a complex polymer charged with biological activity.
    Chemical reviews, 2005, Volume: 105, Issue:7

    Topics: Chromosome Aberrations; Cytokines; Disease Progression; Extracellular Matrix; Fibroblast Growth Fact

2005

Trials

3 trials available for 1-anilino-8-naphthalenesulfonate and Disease Exacerbation

ArticleYear
Avelumab for patients with previously treated metastatic or recurrent non-small-cell lung cancer (JAVELIN Solid Tumor): dose-expansion cohort of a multicentre, open-label, phase 1b trial.
    The Lancet. Oncology, 2017, Volume: 18, Issue:5

    Topics: Aged; Antibodies, Monoclonal; Antibodies, Monoclonal, Humanized; Antineoplastic Agents; Carcinoma, N

2017
Avelumab for patients with previously treated metastatic or recurrent non-small-cell lung cancer (JAVELIN Solid Tumor): dose-expansion cohort of a multicentre, open-label, phase 1b trial.
    The Lancet. Oncology, 2017, Volume: 18, Issue:5

    Topics: Aged; Antibodies, Monoclonal; Antibodies, Monoclonal, Humanized; Antineoplastic Agents; Carcinoma, N

2017
Avelumab for patients with previously treated metastatic or recurrent non-small-cell lung cancer (JAVELIN Solid Tumor): dose-expansion cohort of a multicentre, open-label, phase 1b trial.
    The Lancet. Oncology, 2017, Volume: 18, Issue:5

    Topics: Aged; Antibodies, Monoclonal; Antibodies, Monoclonal, Humanized; Antineoplastic Agents; Carcinoma, N

2017
Avelumab for patients with previously treated metastatic or recurrent non-small-cell lung cancer (JAVELIN Solid Tumor): dose-expansion cohort of a multicentre, open-label, phase 1b trial.
    The Lancet. Oncology, 2017, Volume: 18, Issue:5

    Topics: Aged; Antibodies, Monoclonal; Antibodies, Monoclonal, Humanized; Antineoplastic Agents; Carcinoma, N

2017
The G-250A promoter polymorphism of the hepatic lipase gene predicts the conversion from impaired glucose tolerance to type 2 diabetes mellitus: the Finnish Diabetes Prevention Study.
    The Journal of clinical endocrinology and metabolism, 2004, Volume: 89, Issue:5

    Topics: Alleles; Diabetes Mellitus, Type 2; Disease Progression; Exercise; Female; Genetic Predisposition to

2004
Effects of weight loss with orlistat on glucose tolerance and progression to type 2 diabetes in obese adults.
    Archives of internal medicine, 2000, May-08, Volume: 160, Issue:9

    Topics: Adult; Anti-Obesity Agents; Blood Glucose; Diabetes Mellitus, Type 2; Disease Progression; Double-Bl

2000

Other Studies

80 other studies available for 1-anilino-8-naphthalenesulfonate and Disease Exacerbation

ArticleYear
Should PNPLA3 polymorphism be performed in clinical practice in patients with NAFLD to predict the risk of disease progression?
    Hepatology (Baltimore, Md.), 2022, Volume: 76, Issue:5

    Topics: Disease Progression; Genetic Predisposition to Disease; Genotype; Humans; Lipase; Non-alcoholic Fatt

2022
Identification of novel immune-related targets mediating disease progression in acute pancreatitis.
    Frontiers in cellular and infection microbiology, 2022, Volume: 12

    Topics: Acute Disease; Amylases; Animals; Anti-Inflammatory Agents; Ceruletide; Disease Models, Animal; Dise

2022
Association between PNPLA3[G]/I148M variant, steatosis and fibrosis stage in hepatitis C virus - genetic matters.
    Journal of physiology and pharmacology : an official journal of the Polish Physiological Society, 2019, Volume: 70, Issue:4

    Topics: Antiviral Agents; Disease Progression; Fatty Liver; Female; Hepatitis C, Chronic; Humans; Interferon

2019
Risk factors for histological progression of non-alcoholic steatohepatitis analyzed from repeated biopsy cases.
    Journal of gastroenterology and hepatology, 2020, Volume: 35, Issue:8

    Topics: Adult; Aged; Aged, 80 and over; Alanine Transaminase; Alleles; Biomarkers; Biopsy; Disease Progressi

2020
Liver transcriptomics highlights interleukin-32 as novel NAFLD-related cytokine and candidate biomarker.
    Gut, 2020, Volume: 69, Issue:10

    Topics: Adult; Biomarkers; Disease Progression; Drug Discovery; Female; Gene Expression Profiling; Genetic P

2020
The PNPLA3 rs738409 GG genotype is associated with poorer prognosis in 239 patients with autoimmune hepatitis.
    Alimentary pharmacology & therapeutics, 2020, Volume: 51, Issue:11

    Topics: Adolescent; Adult; Aged; Child; Child, Preschool; Disease Progression; Female; Follow-Up Studies; Ge

2020
Validating candidate biomarkers for different stages of non-alcoholic fatty liver disease.
    Medicine, 2020, Sep-04, Volume: 99, Issue:36

    Topics: Adult; Biomarkers; Case-Control Studies; Disease Progression; Enzyme-Linked Immunosorbent Assay; Fem

2020
    Journal of clinical orthopaedics and trauma, 2021, Volume: 12, Issue:1

    Topics: Acute Coronary Syndrome; Adolescent; Adsorption; Adult; Aged; Animals; Aspergillus; Aspergillus oryz

2021
Identification of a Metabolic, Transcriptomic, and Molecular Signature of Patatin-Like Phospholipase Domain Containing 3-Mediated Acceleration of Steatohepatitis.
    Hepatology (Baltimore, Md.), 2021, Volume: 73, Issue:4

    Topics: Animals; Diet, High-Fat; Diet, Western; Disease Models, Animal; Disease Progression; Gene Expression

2021
Characterization of the Relationship Between the Expression of Aspartate β-Hydroxylase and the Pathological Characteristics of Breast Cancer.
    Medical science monitor : international medical journal of experimental and clinical research, 2020, Dec-31, Volume: 26

    Topics: Acyltransferases; Adult; Aged; Atlases as Topic; Breast Neoplasms; Calcium-Binding Proteins; Carcino

2020
Discordant hepatic fatty acid oxidation and triglyceride hydrolysis leads to liver disease.
    JCI insight, 2021, 01-25, Volume: 6, Issue:2

    Topics: Animals; Carnitine O-Palmitoyltransferase; Disease Progression; Fatty Acids; Female; Hydrolysis; Lip

2021
Serum concentrations of canine pancreatic lipase immunoreactivity and C-reactive protein for monitoring disease progression in dogs with acute pancreatitis.
    Journal of veterinary internal medicine, 2021, Volume: 35, Issue:5

    Topics: Acute Disease; Animals; C-Reactive Protein; Disease Progression; Dog Diseases; Dogs; Lipase; Pancrea

2021
New Discriminant Method for Identifying the Aggressive Disease Phenotype of Non-alcoholic Fatty Liver Disease.
    Internal medicine (Tokyo, Japan), 2017, Volume: 56, Issue:12

    Topics: Adult; Age Factors; Aged; Biopsy; Body Mass Index; Disease Progression; Female; Fibrosis; Genotype;

2017
Disease progression: Divergent paths.
    Nature, 2017, 11-23, Volume: 551, Issue:7681

    Topics: Age Factors; Body Mass Index; Disease Progression; Epigenesis, Genetic; Genetic Predisposition to Di

2017
Genetic variants in PNPLA3 and TM6SF2 predispose to the development of hepatocellular carcinoma in individuals with alcohol-related cirrhosis.
    The American journal of gastroenterology, 2018, Volume: 113, Issue:10

    Topics: Acyltransferases; Aged; Carcinoma, Hepatocellular; Case-Control Studies; Disease Progression; Europe

2018
PNPLA3 rs738409 polymorphism is associated with liver fibrosis progression in patients with chronic hepatitis C: A repeated measures study.
    Journal of clinical virology : the official publication of the Pan American Society for Clinical Virology, 2018, Volume: 103

    Topics: Adult; Disease Progression; Disease Susceptibility; Elasticity Imaging Techniques; Female; Genetic A

2018
Combining Genetic Variants to Improve Risk Prediction for NAFLD and Its Progression to Cirrhosis: A Proof of Concept Study.
    Canadian journal of gastroenterology & hepatology, 2018, Volume: 2018

    Topics: Adult; Aged; Case-Control Studies; Disease Progression; Gene Frequency; Genetic Loci; Genotype; Huma

2018
Utilization of adipocyte-derived lipids and enhanced intracellular trafficking of fatty acids contribute to breast cancer progression.
    Cell communication and signaling : CCS, 2018, 06-18, Volume: 16, Issue:1

    Topics: 3T3-L1 Cells; Adipocytes; Animals; Biological Transport; Breast Neoplasms; Cell Communication; Disea

2018
Atglistatin ameliorates functional decline in heart failure via adipocyte-specific inhibition of adipose triglyceride lipase.
    American journal of physiology. Heart and circulatory physiology, 2018, 10-01, Volume: 315, Issue:4

    Topics: Adipocytes; Animals; Disease Models, Animal; Disease Progression; Enzyme Inhibitors; Heart Failure;

2018
Neutral lipid storage disease with myopathy and dropped head syndrome. Report of a new variant susceptible of treatment with late diagnosis.
    Journal of clinical neuroscience : official journal of the Neurosurgical Society of Australasia, 2018, Volume: 58

    Topics: Delayed Diagnosis; Disease Progression; Female; Humans; Lipase; Lipid Metabolism, Inborn Errors; Mid

2018
Protective Effects of Calcitonin Gene-Related Peptide-Mediated p38 Mitogen-Activated Protein Kinase Pathway on Severe Acute Pancreatitis in Rats.
    Digestive diseases and sciences, 2019, Volume: 64, Issue:2

    Topics: Acute Disease; Amylases; Animals; Calcitonin Gene-Related Peptide; Cytokines; Disease Progression; E

2019
Genetic determinants of steatosis and fibrosis progression in paediatric non-alcoholic fatty liver disease.
    Liver international : official journal of the International Association for the Study of the Liver, 2019, Volume: 39, Issue:3

    Topics: Adaptor Proteins, Signal Transducing; Adolescent; Age Factors; Case-Control Studies; Child; Disease

2019
Promoting genetics in non-alcoholic fatty liver disease: Combined risk score through polymorphisms and clinical variables.
    World journal of gastroenterology, 2018, Nov-21, Volume: 24, Issue:43

    Topics: Disease Progression; Genetic Predisposition to Disease; Humans; Lipase; Liver; Membrane Proteins; No

2018
LIPG-promoted lipid storage mediates adaptation to oxidative stress in breast cancer.
    International journal of cancer, 2019, 08-15, Volume: 145, Issue:4

    Topics: Breast Neoplasms; Cell Line, Tumor; Disease Progression; Disease-Free Survival; Female; Humans; Lipa

2019
GWAS and enrichment analyses of non-alcoholic fatty liver disease identify new trait-associated genes and pathways across eMERGE Network.
    BMC medicine, 2019, 07-17, Volume: 17, Issue:1

    Topics: Adult; Aged; Body Mass Index; Case-Control Studies; Community Networks; Disease Progression; Electro

2019
Patatin-like phospholipase domain-containing 3 I148M affects liver steatosis in patients with chronic hepatitis B.
    Hepatology (Baltimore, Md.), 2013, Volume: 58, Issue:4

    Topics: Adult; Alcoholism; Cohort Studies; Disease Progression; Fatty Liver; Female; Genetic Predisposition

2013
[Genotyping in progressive age-related macular degeneration].
    Oftalmologia (Bucharest, Romania : 1990), 2012, Volume: 56, Issue:4

    Topics: Biomarkers; Bruch Membrane; Cholesterol, HDL; Complement Factor H; Complement Inactivating Agents; D

2012
Dual regulation of adipose triglyceride lipase by pigment epithelium-derived factor: a novel mechanistic insight into progressive obesity.
    Molecular and cellular endocrinology, 2013, Sep-05, Volume: 377, Issue:1-2

    Topics: 3T3-L1 Cells; Adipocytes; Adipose Tissue; Animals; Body Weight; Diet, High-Fat; Disease Progression;

2013
Orlistat, an under-recognised cause of progressive renal impairment.
    Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association, 2013, Volume: 28 Suppl 4

    Topics: Acute Kidney Injury; Aged; Anti-Obesity Agents; Diabetes Mellitus, Type 2; Disease Progression; Esse

2013
Donor PNPLA3 rs738409 genotype affects fibrosis progression in liver transplantation for hepatitis C.
    Hepatology (Baltimore, Md.), 2014, Volume: 59, Issue:2

    Topics: Biopsy; Cohort Studies; Disease Progression; Female; Follow-Up Studies; Genotype; Hepatitis C; Human

2014
Impact of hyperglycemia and acute pancreatitis on the receptor for advanced glycation endproducts.
    International journal of clinical and experimental pathology, 2013, Volume: 6, Issue:10

    Topics: Animals; Diabetes Mellitus, Experimental; Disease Progression; Hyperglycemia; Lipase; MAP Kinase Sig

2013
Species- and dose-specific pancreatic responses and progression in single- and repeat-dose studies with GI181771X: a novel cholecystokinin 1 receptor agonist in mice, rats, and monkeys.
    Toxicologic pathology, 2014, Volume: 42, Issue:1

    Topics: Amylases; Animals; Benzodiazepines; Disease Models, Animal; Disease Progression; Dose-Response Relat

2014
Modified Xiaochaihu Decoction () prevents the progression of chronic pancreatitis in rats possibly by inhibiting transforming growth factor-β1/Sma- and mad-related proteins signaling pathway.
    Chinese journal of integrative medicine, 2013, Volume: 19, Issue:12

    Topics: Amylases; Animals; Base Sequence; Blood Glucose; Body Weight; Chronic Disease; Disease Progression;

2013
Carriage of the PNPLA3 rs738409 C >G polymorphism confers an increased risk of non-alcoholic fatty liver disease associated hepatocellular carcinoma.
    Journal of hepatology, 2014, Volume: 61, Issue:1

    Topics: Adult; Aged; Carcinoma, Hepatocellular; Case-Control Studies; Cohort Studies; Disease Progression; F

2014
Targeted next-generation sequencing and fine linkage disequilibrium mapping reveals association of PNPLA3 and PARVB with the severity of nonalcoholic fatty liver disease.
    Journal of human genetics, 2014, Volume: 59, Issue:5

    Topics: Actinin; Adult; Aged; Biopsy; Case-Control Studies; Chromosome Mapping; Disease Progression; Female;

2014
Revisiting liver disease progression in HIV/HCV-coinfected patients: the influence of vitamin D, insulin resistance, immune status, IL28B and PNPLA3.
    Liver international : official journal of the International Association for the Study of the Liver, 2015, Volume: 35, Issue:3

    Topics: Adult; Coinfection; Disease Progression; Female; Genotype; Hepatitis C; Hepatitis C, Chronic; HIV; H

2015
Interaction between PNPLA3 I148M variant and age at infection in determining fibrosis progression in chronic hepatitis C.
    PloS one, 2014, Volume: 9, Issue:8

    Topics: Adolescent; Adult; Age Factors; Aged; Disease Progression; Fatty Liver; Gene Frequency; Genetic Pred

2014
Patatin-Like Phospholipase Domain-Containing 3 I148M Variant Is Associated with Liver Steatosis and Fat Distribution in Chronic Hepatitis B.
    Digestive diseases and sciences, 2015, Volume: 60, Issue:10

    Topics: Abdominal Fat; Adult; Aged; Anthropometry; Body Mass Index; Chi-Square Distribution; Cohort Studies;

2015
Genetic Polymorphisms of IL28B and PNPLA3 Are Predictive for HCV Related Rapid Fibrosis Progression and Identify Patients Who Require Urgent Antiviral Treatment with New Regimens.
    PloS one, 2015, Volume: 10, Issue:9

    Topics: Aged; Antiviral Agents; Disease Progression; Fatty Liver; Female; Genome-Wide Association Study; Gen

2015
PNPLA3 148M Carriers with Inflammatory Bowel Diseases Have Higher Susceptibility to Hepatic Steatosis and Higher Liver Enzymes.
    Inflammatory bowel diseases, 2016, Volume: 22, Issue:1

    Topics: Alanine Transaminase; Alleles; Aspartate Aminotransferases; Biomarkers; Cohort Studies; Disease Prog

2016
Retinoic Acid Ameliorates Pancreatic Fibrosis and Inhibits the Activation of Pancreatic Stellate Cells in Mice with Experimental Chronic Pancreatitis via Suppressing the Wnt/β-Catenin Signaling Pathway.
    PloS one, 2015, Volume: 10, Issue:11

    Topics: Actins; Active Transport, Cell Nucleus; Animals; Apoptosis; Axin Protein; Cells, Cultured; Ceruletid

2015
The Impact of PNPLA3 rs738409 SNP on Liver Fibrosis Progression, Portal Hypertension and Hepatic Steatosis in HIV/HCV Coinfection.
    PloS one, 2015, Volume: 10, Issue:11

    Topics: Adult; Age Factors; Alleles; Biopsy; Cohort Studies; Coinfection; Cross-Sectional Studies; Disease P

2015
Impact of Donor and Recipient Single Nucleotide Polymorphisms in Living Liver Donor Transplantation for Hepatitis C.
    Transplantation proceedings, 2015, Volume: 47, Issue:10

    Topics: Cohort Studies; Disease Progression; Female; Genotype; Hepacivirus; Hepatitis C; Humans; Lipase; Liv

2015
Association of PNPLA3 Polymorphism with Hepatocellular Carcinoma Development and Prognosis in Viral and Non-Viral Chronic Liver Diseases.
    Asian Pacific journal of cancer prevention : APJCP, 2015, Volume: 16, Issue:18

    Topics: Aged; Carcinoma, Hepatocellular; Case-Control Studies; Disease Progression; Fatty Liver, Alcoholic;

2015
Perfusion-CT--Can We Predict Acute Pancreatitis Outcome within the First 24 Hours from the Onset of Symptoms?
    PloS one, 2016, Volume: 11, Issue:1

    Topics: Adult; Aged; Aged, 80 and over; Amylases; Disease Progression; Early Diagnosis; Female; Humans; Lipa

2016
Association of PNPLA3 rs738409 and TM6SF2 rs58542926 with health services utilization in a population-based study.
    BMC health services research, 2016, Feb-03, Volume: 16

    Topics: Adult; Aged; Disease Progression; Female; Genotype; Germany; Health Services; Humans; Lipase; Male;

2016
Impact of EGF, IL28B, and PNPLA3 polymorphisms on the outcome of allograft hepatitis C: a multicenter study.
    Clinical transplantation, 2016, Volume: 30, Issue:4

    Topics: Adult; Allografts; Antiviral Agents; Carcinoma, Hepatocellular; Cohort Studies; Disease Progression;

2016
[Relationship between changes of increased amylase or lipase levels and pancreas injury in critically ill children].
    Zhonghua er ke za zhi = Chinese journal of pediatrics, 2016, Volume: 54, Issue:9

    Topics: Amylases; Blood Urea Nitrogen; Calcitonin; Child; Child, Preschool; Creatinine; Critical Illness; Di

2016
The PNPLA3 Genetic Variant rs738409 Influences the Progression to Cirrhosis in HIV/Hepatitis C Virus Coinfected Patients.
    PloS one, 2016, Volume: 11, Issue:12

    Topics: Adult; Alleles; Coinfection; Cross-Sectional Studies; Disease Progression; Elasticity Imaging Techni

2016
Serum Lipase as Clinical Laboratory Index for Chronic Renal Failure Diagnosis.
    Clinical laboratory, 2016, Jul-01, Volume: 62, Issue:7

    Topics: Adult; Aged; Aged, 80 and over; Amylases; Biomarkers; Case-Control Studies; Clinical Enzyme Tests; C

2016
Putative role of endothelial lipase in dialysis patients with hypoalbuminemia and inflammation.
    American journal of nephrology, 2008, Volume: 28, Issue:6

    Topics: Aged; Atherosclerosis; Cholesterol, HDL; Disease Progression; Female; Humans; Hypoalbuminemia; Infla

2008
The effect of synacthen on acute necrotizing pancreatitis in rats.
    Pancreas, 2008, Volume: 37, Issue:3

    Topics: Adrenal Insufficiency; Amylases; Animals; Cosyntropin; Cytokines; Disease Models, Animal; Disease Pr

2008
Reduced hepatic expression of adipose tissue triglyceride lipase and CGI-58 may contribute to the development of non-alcoholic fatty liver disease in patients with insulin resistance.
    Scandinavian journal of gastroenterology, 2008, Volume: 43, Issue:8

    Topics: 1-Acylglycerol-3-Phosphate O-Acyltransferase; Adipose Tissue; Biomarkers; Disease Progression; Fatty

2008
Gene polymorphisms in APOE, NOS3, and LIPC genes may be risk factors for cardiac adverse events after primary CABG.
    Journal of cardiothoracic surgery, 2009, Aug-19, Volume: 4

    Topics: Apolipoproteins E; Cohort Studies; Coronary Artery Bypass; Disease Progression; Female; Genetic Pred

2009
[Management of chronic pancreatitis].
    MMW Fortschritte der Medizin, 2010, Mar-25, Volume: 152, Issue:12

    Topics: Amylases; Analgesics; Cholangiopancreatography, Endoscopic Retrograde; Combined Modality Therapy; Di

2010
PNPLA3 variants specifically confer increased risk for histologic nonalcoholic fatty liver disease but not metabolic disease.
    Hepatology (Baltimore, Md.), 2010, Volume: 52, Issue:3

    Topics: Adult; Alleles; Biopsy; Case-Control Studies; Disease Progression; Fatty Liver; Female; Genetic Pred

2010
The association of genetic variability in patatin-like phospholipase domain-containing protein 3 (PNPLA3) with histological severity of nonalcoholic fatty liver disease.
    Hepatology (Baltimore, Md.), 2010, Volume: 52, Issue:3

    Topics: Adolescent; Adult; Aged; Alleles; Biopsy; Case-Control Studies; Child; Chromosomes, Human, Pair 10;

2010
Emerging genes associated with the progression of nonalcoholic fatty liver disease.
    Hepatology (Baltimore, Md.), 2010, Volume: 52, Issue:3

    Topics: Alleles; Biopsy; Disease Progression; Fatty Liver; Genetic Predisposition to Disease; Humans; Lipase

2010
Role of hormonal axis, growth hormone - IGF-1, in the therapeutic effect of ghrelin in the course of cerulein-induced acute pancreatitis.
    Journal of physiology and pharmacology : an official journal of the Polish Physiological Society, 2010, Volume: 61, Issue:5

    Topics: Amylases; Animals; Ceruletide; Disease Progression; Ghrelin; Growth Hormone; Hypophysectomy; Insulin

2010
Deficiency of liver adipose triglyceride lipase in mice causes progressive hepatic steatosis.
    Hepatology (Baltimore, Md.), 2011, Volume: 54, Issue:1

    Topics: Alanine Transaminase; Animals; Cytoplasm; Disease Models, Animal; Disease Progression; Energy Metabo

2011
Impact of patatin-like phospholipase-3 (rs738409 C>G) polymorphism on fibrosis progression and steatosis in chronic hepatitis C.
    Hepatology (Baltimore, Md.), 2011, Volume: 54, Issue:1

    Topics: Adult; Aged; Antiviral Agents; Belgium; Cross-Sectional Studies; Disease Progression; Fatty Liver; F

2011
The common I148 M variant of PNPLA3 does not predict fibrosis progression after liver transplantation for hepatitis C.
    Hepatology (Baltimore, Md.), 2011, Volume: 54, Issue:4

    Topics: Cohort Studies; Disease Progression; End Stage Liver Disease; Female; Genetic Markers; Graft Rejecti

2011
HIV-1 disease progression is associated with bile-salt stimulated lipase (BSSL) gene polymorphism.
    PloS one, 2012, Volume: 7, Issue:3

    Topics: Alleles; CD4 Lymphocyte Count; Disease Progression; Genotype; HIV Infections; HIV Seropositivity; HI

2012
Patatin-like phospholipase domain containing-3 gene I148M polymorphism, steatosis, and liver damage in hereditary hemochromatosis.
    World journal of gastroenterology, 2012, Jun-14, Volume: 18, Issue:22

    Topics: Adult; Biomarkers; Biopsy; Chi-Square Distribution; Disease Progression; Fatty Liver; Female; Gene F

2012
Genome-wide association study identifies variants associated with progression of liver fibrosis from HCV infection.
    Gastroenterology, 2012, Volume: 143, Issue:5

    Topics: Adult; Apoptosis; c-Mer Tyrosine Kinase; Disease Progression; Eye Proteins; Female; Genome-Wide Asso

2012
Genetic polymorphism in cyclooxygenase-2 promoter affects hepatic inflammation and fibrosis in patients with chronic hepatitis C.
    Journal of viral hepatitis, 2012, Volume: 19, Issue:9

    Topics: Asian People; Cyclooxygenase 2; Disease Progression; Female; Genetic Predisposition to Disease; Hepa

2012
PNPLA3, a genetic marker of progressive liver disease, still hiding its metabolic function?
    Clinics and research in hepatology and gastroenterology, 2013, Volume: 37, Issue:1

    Topics: Disease Progression; Fatty Liver; Genetic Markers; Humans; Lipase; Membrane Proteins; Non-alcoholic

2013
Influence of leptin administration on the course of acute ischemic pancreatitis.
    Journal of physiology and pharmacology : an official journal of the Polish Physiological Society, 2002, Volume: 53, Issue:4 Pt 2

    Topics: Amylases; Animals; Disease Progression; DNA; Interleukin-1; Interleukin-10; Ischemia; Leptin; Lipase

2002
Exocrine pancreatic insufficiency as an end stage of pancreatitis in four dogs.
    The Journal of small animal practice, 2003, Volume: 44, Issue:7

    Topics: Amylases; Animals; Case-Control Studies; Chronic Disease; Disease Progression; Dog Diseases; Dogs; E

2003
Effect of early administration of exogenous basic fibroblast growth factor on acute edematous pancreatitis in rats.
    World journal of gastroenterology, 2006, May-21, Volume: 12, Issue:19

    Topics: Amylases; Animals; Ceruletide; Disease Progression; DNA; Edema; Fibroblast Growth Factor 2; Immunohi

2006
CETP (cholesteryl ester transfer protein) promoter -1337 C>T polymorphism protects against coronary atherosclerosis in Japanese patients with heterozygous familial hypercholesterolaemia.
    Clinical science (London, England : 1979), 2006, Volume: 111, Issue:5

    Topics: Adult; Aged; Aged, 80 and over; Cholesterol Ester Transfer Proteins; Coronary Artery Disease; Diseas

2006
Rapidly progressive renal failure associated with successful pharmacotherapy for obesity.
    Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association, 2007, Volume: 22, Issue:2

    Topics: Acute Kidney Injury; Anti-Obesity Agents; Biopsy; Disease Progression; Female; Follow-Up Studies; Gl

2007
Rapidly progressive renal failure associated with successful pharmacotherapy for obesity.
    Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association, 2007, Volume: 22, Issue:8

    Topics: Acute Kidney Injury; Anti-Obesity Agents; Biopsy; Cohort Studies; Disease Progression; Follow-Up Stu

2007
[Acute pancreatitis: what is really important in diagnosis and therapy].
    Medizinische Klinik (Munich, Germany : 1983), 2007, Sep-15, Volume: 102, Issue:9

    Topics: Aged; Amylases; Critical Care; Disease Progression; Fatal Outcome; Female; Humans; Lipase; Pancreas;

2007
Acute elevation of plasma PLTP activity strongly increases pre-existing atherosclerosis.
    Arteriosclerosis, thrombosis, and vascular biology, 2008, Volume: 28, Issue:7

    Topics: Animals; Apolipoproteins B; Atherosclerosis; Cholesterol, Dietary; Collagen; Disease Models, Animal;

2008
Hyperlipaemia intensifies the course of acute oedematous and acute necrotising pancreatitis in the rat.
    Gut, 1996, Volume: 38, Issue:5

    Topics: Acute Disease; Amylases; Animals; Cholesterol; Disease Models, Animal; Disease Progression; Edema; H

1996
Progressive pan-colonic fibrosis secondary to oral administration of pancreatic enzymes.
    Pediatric surgery international, 1998, Volume: 13, Issue:2-3

    Topics: Child; Colon; Constriction, Pathologic; Cystic Fibrosis; Disease Progression; Fibrosis; Humans; Inte

1998
T cell-mediated exocrine pancreatic damage in major histocompatibility complex class II-deficient mice.
    Gastroenterology, 1998, Volume: 115, Issue:4

    Topics: Amylases; Animals; Blood Glucose; Digestive System; Disease Progression; Histocompatibility Antigens

1998
Serum pancreatic enzymes in human immunodeficiency virus-infected children. A collaborative study of the Italian Society of Pediatric Gastroenterology and Hepatology.
    Scandinavian journal of gastroenterology, 1998, Volume: 33, Issue:9

    Topics: Adolescent; AIDS-Related Opportunistic Infections; Amylases; CD4-Positive T-Lymphocytes; Child; Chil

1998
Ethanol administration delays recovery from acute pancreatitis induced by exocrine hyperstimulation.
    Journal of pharmacological and toxicological methods, 1998, Volume: 39, Issue:4

    Topics: Acute Disease; Amylases; Animals; Calcium; Central Nervous System Depressants; Ceruletide; Disease P

1998