150 results on '"Redston M"'
Search Results
2. American Society of Colon and Rectal Surgeons 95th Annual Convention Podium and Poster Abstracts June 9–14, 1996 Seattle, Washington
- Author
-
Obrand, D., Gordon, P. H., Rowley, S., Grace, R. H., Rai, S., Moran, M. R., Rai, A. M., Farouk, R., Lee, P. W. R., Edwards, J., Thorne, M., MacDonald, A. W., Duthie, G. S., Monson, J. R. T., Shabahang, M., Brenner, R., Wright, A., Montgomery, E., Trock, B., Buras, R., Schumaker, L., Nolla, J., Buffan, A., Uskokovic, M., Nauta, R., Evans, S., Velázquez, O. C., Zhou, D., Seto, R. W., Choi, J., Jabbar, A., Breen, F., Rombeau, J. L., Casillas, S., Dietz, D. W., Brand, M. I., Vladisavljevic, A., Jones, S. C., Milsom, J. W., Stuntz, M., Wilmoth, G., Ong, J., Stabile, B., Stamos, M. J., Kahn, H., Alexander, A., Rakinic, J., Nagle, D., Fry, R., Simons, A. J., Kerr, R., Toms, C., Groshen, S., Ross, R., Morris, M., Beart, R., Ortega, A., Anthone, G., Lucha, P., Rosen, L., Stasik, J., Olenwine, J., Riether, R., Khubchandani, I., Ogunbivi, O., Birnbaum, E., Fleshman, J., Kodner, I., McLeod, R. S., Geerts, W., Sniderman, K., Greenwood, C., Gregoire, R., Taylor, B., Silverman, R., Atkinson, K., Burnstein, M., Marshall, J., Burul, C., Anderson, D., Ross, T., Wilson, S., Barton, P., Maetani, S., Onodera, H., Morimoto, H., Imamura, M., Hyams, D. M., Mamounas, E., Petrelli, N., Rockette, H., Jones, J., Wolmark, N., Sofo, L., Ratto, C., Valentini, V., Ippoliti, M., Nucera, P., Merico, M., Bellantone, R., Doglietto, G. B., Crucitti, F., Goes, R., Simons, A., Gunderson, L., Grado, G., Streeter, O., Sun, J. H., Decanini-Garza, P., Kim, D. G., Wong, W. D., Rothenberger, D. A., Madoff, R. D., Madlensky, L., Berk, T., Bapat, B., Redston, M., Gallinger, S., Cohen, Z., Winde, G., Schmid, K. W., Brandt, B., Müller, R., Osswald, H., Jang, Y., Steinhagen, R., Heimann, T., Schnitzler, M., Blackstein, M., McLeod, R., Devesa, J. M., Madrid, J. M. Fernandez, Enriquez, J. M., Geerdes, B. P., Heineman, E., Konsten, J., Baeten, C. G. M., Michot, F., Lehur, P. A., Denis, P., Grise, P. H., Leborgne, J., Teniere, P., Buzelin, J. M., Stebbing, J. F., Brading, A. F., Mortensen, N. J. McC, Gunn, J., Gardiner, A., Abdullah, N., Nyam, D. C. N. K., Pemberton, J. H., Ilstrup, D., Lund, J. N., Scholefield, J. H., Stamm, L., Matzel, K. E., Stadelmaier, U., Dünne, A., Hohenberger, W., Sala, C., Garcia-Granero, E., Molina, M. J., Garcia, J. V., Lledo, S., Ternent, C. A., Shashidharan, M., Blatchford, G. J., Christensen, M. A., Thorson, A. G., Sentovich, S. M., Jensen, L. L., Lowry, A. C., Zaheer, S., Reilly, W. T., Tsang, C., Singer, D., Richard, C. S., Stern, H. S., Oliveira, L., Daniel, N., Bernstein, M., DeMarta, D., Weiss, E. G., Nogueras, J. J., Wexner, S. D., Keighley, M. R. B., Korsgen, S., Agachan, F., Kim, D. -S., Goldberg, S. M., Durham, R. M., Pruitt, G., Longo, W. E., Marchesa, P., Oliart, S., Goldblum, J., Fazio, V. W., Rantis, P. C., Daniel, G. L., Vernava, III, A. M., Becker, J. M., Marie, G. St., Ferzoco, S., Franklin, M., Rosenthal, D., Goldstein, E. T., Bass, E. M., DelPino, A., Tan, A., Pearl, R., Orsay, C., Sher, M. E., Sands, L. R., Påhlman, Lars, Hewett, P. J., Thomas, W. M., King, G., Eaton, M., Allendorf, U. D. F., Bessler, M., Whelan, R. L., Trokel, M., Laird, D., Nowygrod, R., Treat, M. R., Vukasin, P., Steele, G., Weston, L., Allendorf, J. D. F., Sellers, G., Joo, J. S., Bruce, C. J., Coller, J. A., Murray, J. J., Schoetz, Jr., D. J., Roberts, P. L., Schoetz, D., Bockler, M., Rosenblatt, M., Malhorta, S., Roberts, P., Murray, J., Coller, J., Rusin, L., Liu, C. D., Newton, T. R., Zinner, M. J., Ashley, S. W., McFadden, D. W., Tusek, D. L., Church, J. M., Strong, S. A., Grass, J., Steinhart, A. H., Greenberg, G. R., Siminovich, K., Blair, J. E., Cruz, C., Prabhakar, L. P., Laramee, C., Nelson, H., Dozois, R. R., Ozuner, G., Hull, T., Fazio, V., Navaro, G., Bauer, J. J., Gorfine, S. R., Gelemt, I. M., Harris, M. T., Kreel, I., Marcello, P. W., Rusin, L. C., Veidenheimer, M. C., Ogunbiyi, O. A., Thibault, C., Sagar, P., Wolff, B. G., Lee, F., Lee, E. C., Pennoyer, W. P., Vignati, P. V., Cohen, J., MacRae, H. M., O'Connor, B., Ton, E., Hain, J. M., Perez-Ramirez, J. J., Spencer, M. P., Gemlo, B. T., Neto, J. A. Reis, Quilici, F. A., Cordeiro, F., Reis, Jr., J. A., Neto, C. I. Reis, Gottesman, L., Tjandra, J., Takano, M., Kuromizu, J., Tsuji, Y., Lee, C. S., Ferrara, A., Levy, J. R., Larach, S. W., Krecker, M., Williamson, P. R., Wong, D. W., Sarmiento, J. M., Burgart, L. J., Frizelle, F. A., Ilstrup, D. M., Salem, R., Smith, L. E., Rooney, P. S., Chapman, M. A. S., Steele, R. J. C., Koren, R., Gal, R., Kyzer, S., Chaimoff, CH., Rodríguez-Bigas, M. A., Mahoney, M. C., Weber, T. K., Petrelli, N. J., Ault, G., Ceron, O., Conti, P., Hadfield, M. B., Turnbull, L. W., Nicholson, A. A., Horsman, A., Shibata, D., Sentovich, S., Hyland, W., Busse, P., Bleday, R., Allendorf, J., Whelan, R., Horvath, K., Treat, M., Wronski, M., Arbit, E., Bilsky, M., Galicich, J. H., Miller, A. S., Lewis, W. G., Williamson, M. E. R., Sagar, P. M., Holdsworth, P. J., Johnston, D., Smith, A. H., Marchetti, F., Thompson-Fawcett, M. W., Warren, B. F., Mortensen, N. J. M., Bouchard, S., Belliveau, P., Trudel, J., Zinsmeister, A. R., Schleck, C. D., McIntyre, P. B., Hanson, R. B., Read, T. E., Dominguez, J. M., Hyman, N. H., Beck, D. E., Dayton, M. T., Stryker, S. J., Wolf, B. G., Young-Fadok, T. M., Meagher, A., Benn, P. L., Takao, Y., Chen, F. C., Wu, J., Milsom, J., Stein, B. L., Vasilevsky, C. A., Hartley, J. E., Cureshi, A., Sellers, G. J., Van, D., Ludwig, K. A., Garcia-Ruiz, A., Espat, N. J., Rao, G. N., Drew, P. J., Pfeifer, J., Park, U. C., Gonzalez, A., Okamoto, T., Konishi, F., Tsukamoto, T., Senba, S., Kashiwagi, H., Kojima, M., Togashi, T., Kanazawa, K., Yoon, W. H., Kang, Y. N., Hong, K. H., Park, H. D., Koo, S. H., Song, K. S., Kim, J. C., Roh, S. A., Park, K. C., Jessup, J. M., Changchien, C. R., Wang, J. Y., Hsu, K. C., Chen, J. S., Tang, R., You, Y. T., Ho, Y. S., Guttman, R., Nelson, R., Sardinha, T. G. S., Gilliland, J., Kroll, M., Lee, E., Wexler, J., Hudzinski, D., Glass, D., Wolff, B. D., King, D. W., Talley, N., Chen, W. S., Lin, W. C., Hsu, H., Wrightson, W. R., Galandiuk, S., LaRocca, R., Myers, S. R., Tada, M., Inoue, H., Tsubaki, M., Endo, M., Sobzcak, S., Welch, J. P., Cohen, J. L., Allen, L. W., Morrow, J. S., Behen, S. L., Smith, K. W., Cali, J. R., Bailey, H. R., Fucini, C., Elbetti, C., Messerini, L., Law, W. L., Butts, D. R., Max, E., Memon, M. A., Devine, J., Feeney, J., Talley, N. J., Stephenson, E. R., Ilahi, O., Koltun, W. A., Spellman, M., Rantis, R. C., Vernava, A. M., Parra, R. O., Breen, E., Hayes, P., Quinn, D., Whitlow, C. B., Opelka, F. G., Gathright, J. B., Golub, R. W., Maccabee, P. J., Combs, A. J., Grose, E. A., Taylor, B. M., Kozell, K., McGannon, E., Krogh, K., Nielsen, J., Djurhuus, J. C., Mosdal, C., Sabroe, S., Laurberg, S., Chen, M. F., Kerner, B. A., Khanduja, K. S., Wise, Jr., W. E., Padmanabhan, A., Meesig, D. M., Yasin, M. T., Aguilar, P. S., Ho, Y. H., Tan, M., Seow-Choen, F., Rustin, R. B., and Harmon, J. M.
- Published
- 1996
- Full Text
- View/download PDF
3. p53 alterations in oesophageal cancer: association with clinicopathological features, risk factors, and survival
- Author
-
Casson, A G, Tammemagi, M, Eskandarian, S, Redston, M, McLaughlin, J, and Ozcelik, H
- Published
- 1998
4. Microsatellite instability versus immunohistochemistry testing in phenotyping colorectal tumors
- Author
-
Lindor, N.M., Burgart, L.J., Leontovich, O., Goldberg, R.M., Cunningham, J.M., Walsh-Vockley, C., Petersen, G., Redston, M., Young, J., Barker, M., Walsh, M.D., Jass, J., Leggett, B.A., Hopper, J., Bapat, B., Gallinger, S., Selander, T., and Thibodeau, S.N.
- Subjects
Genetic disorders -- Research ,Human chromosome abnormalities -- Research ,Human genetics -- Research ,Immunohistochemistry -- Usage ,Colorectal cancer -- Diagnosis ,Tumors -- Analysis ,Biological sciences - Published
- 2001
5. A diagnostic screening test for tumor microsatellite instability (MSI) using fluorescent multiplex PCR and automated fragment analysis
- Author
-
Rose, M., Selander, T., Redston, M., Gallinger, S., and Bapat, B.
- Subjects
Human genetics -- Research ,Tumors -- Genetic aspects ,Biological sciences - Published
- 2000
6. Genetic, immunohistochemical, and clinical features of medullary carcinoma of the pancreas: A newly described and characterized entity
- Author
-
Wilentz, R. E., Goggins, M., Redston, M., Marcus, V. A., Adsay, N. V., Sohn, T. A., Kadkol, S. S., Yeo, C. J., Choti, M., Zahurak, M., Johnson, K., Tascilar, M., Offerhaus, G. J., Hruban, R. H., Kern, S. E., and Other departments
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,nutritional and metabolic diseases ,neoplasms ,digestive system diseases - Abstract
Medullary carcinomas of the pancreas are a recently described, histologically distinct subset of poorly differentiated adenocarcinomas that may have a unique pathogenesis and clinical course. To further evaluate these neoplasms, we studied genetic, pathological, and clinical features of 13 newly identified medullary carcinomas of the pancreas. Nine (69%) of these had wild-type K-ras genes, and one had microsatellite instability (MSI). This MSI medullary carcinoma, along with three previously reported MSI medullary carcinomas, were examined immunohistochemically for Mlh1 and Msh2 expression, and all four expressed Msh2 but did not express Mlh1. In contrast, all of the medullary carcinomas without MSI expressed both Msh2 and Mlh1. Remarkably, the MSI medullary carcinoma of the pancreas in the present series arose in a patient with a synchronous but histologically distinct cecal carcinoma that also had MSI and did not express Mlh1. The synchronous occurrence of two MSI carcinomas suggests an inherited basis for the development of these carcinomas. Indeed, the medullary phenotype, irrespective of MSI, was highly associated with a family history of cancer in first-degree relatives (P < 0.001). Finally, one medullary carcinoma with lymphoepithelioma-like features contained Epstein-Barr virus-encoded RNA-1 by in situ hybridization. Therefore, because of medullary carcinoma's special genetic, immunohistochemical, and clinical features, recognition of the medullary variant of pancreatic adenocarcinoma is important. Only by classifying medullary carcinoma as special subset of adenocarcinoma can we hope to further elucidate its unique pathogenesis
- Published
- 2000
7. A missense mutation in both hMSH2 and APC in an Ashkenazi Jewish HNPCC kindred: implications for clinical screening
- Author
-
YUAN ZQ, Wong, N., William Foulkes, Alpert, L., Manganaro, F., Andreutti-Zaugg, C., Iggo, R., Anthony, K., Hsieh, E., Redston, M., Pinsky, L., Trifiro, M., Gordon, P., and Lasko, D.
- Subjects
Adaptor Proteins, Signal Transducing ,Adenomatous Polyposis Coli Protein ,Carrier Proteins ,Colorectal Neoplasms, Hereditary Nonpolyposis/genetics ,Cytoskeletal Proteins/genetics ,DNA-Binding Proteins ,Female ,Genetic Predisposition to Disease ,Humans ,Jews ,Male ,Microsatellite Repeats ,MutS Homolog 2 Protein ,Mutation, Missense ,Neoplasm Proteins/genetics ,Nuclear Proteins ,Nucleic Acid Conformation ,Nucleic Acid Hybridization ,Pedigree ,Proto-Oncogene Proteins/genetics ,Risk Factors ,Letters to the Editor - Published
- 1999
8. Allelotype of pancreatic adenocarcinoma
- Author
-
Ab, Seymour, Rh, Hruban, Redston M, Carlos Caldas, Sm, Powell, Kw, Kinzler, Cj, Yeo, and Se, Kern
- Subjects
Pancreatic Neoplasms ,Genotype ,Humans ,Adenocarcinoma ,Alleles ,Gene Deletion - Abstract
Knowledge of the patterns of allelic loss has been useful in identifying the spectrum of the tumor suppressor genes involved in various tumor types. Such analyses in pancreatic carcinoma have been difficult due to the characteristic host desmoplastic reaction to the neoplasm. We have assembled the first allelotype of pancreatic adenocarcinoma, a survey for allelic loss among each chromosomal arm, using seven cryostat-dissected neoplasms. The fractional allelic loss in these seven neoplasms was 0.18, a value similar to that seen previously in colorectal carcinoma. Alleles of chromosome 18q (lost in five of six informative tumors) and of chromosome 17p (lost in four of five informative tumors) were commonly affected. Neither APC mutations (33 neoplasms), allelic shifts of dinucleotide repeats (26 neoplasms), nor immunohistochemical evidence of retinoblastoma protein underexpression (7 neoplasms) were found. Further evaluation of allelic loss in pancreatic cancer would benefit from improved methods for the analysis of lost genetic material which overcome the problems posed by the high admixture of nonneoplastic stromal and inflammatory cells in these tumors.
- Published
- 1994
9. Presence of 18q loss of heterozygosity (LOH) and disease-free and overall survival in stage II colon cancer: CALGB Protocol 9581
- Author
-
Bertagnolli, M. M., primary, Niedzwiecki, D., additional, Hall, M., additional, Jewell, S. D., additional, Mayer, R. J., additional, Goldberg, R. M., additional, Colacchio, T. A., additional, Warren, R. S., additional, and Redston, M., additional
- Published
- 2009
- Full Text
- View/download PDF
10. Prediction of response to temozolomide (TMZ)-based therapy by loss of MGMT expression in patients with advanced neuroendocrine tumors (NET)
- Author
-
Kulke, M. H., primary, Frauenhoffer, C. S., additional, Hooshmand, S. M., additional, Ryan, D. P., additional, Enzinger, P. C., additional, Meyerhardt, J. A., additional, Clark, J. W., additional, Hornick, J., additional, Fuchs, C. S., additional, and Redston, M. S., additional
- Published
- 2007
- Full Text
- View/download PDF
11. Microsatellite instability predicts improved response to adjuvant therapy with irinotecan, 5-fluorouracil and leucovorin in stage III colon cancer
- Author
-
Bertagnolli, M. M., primary, Compton, C. C., additional, Niedzwiecki, D., additional, Warren, R. S., additional, Jewell, S., additional, Bailey, G. P., additional, Mayer, R. J., additional, Goldberg, R., additional, Saltz, L., additional, and Redston, M., additional
- Published
- 2006
- Full Text
- View/download PDF
12. Association Between Biallelic and Monoallelic Germline MYH Gene Mutations and Colorectal Cancer Risk
- Author
-
Croitoru, M. E., primary, Cleary, S. P., additional, Di Nicola, N., additional, Manno, M., additional, Selander, T., additional, Aronson, M., additional, Redston, M., additional, Cotterchio, M., additional, Knight, J., additional, Gryfe, R., additional, and Gallinger, S., additional
- Published
- 2004
- Full Text
- View/download PDF
13. Sentinel node staging of resectable colon cancer: Results of CALGB 80001
- Author
-
Bertagnolli, M. M., primary, Redston, M., additional, Miedema, B., additional, Dowell, J., additional, Niedzwiecki, D., additional, Mayer, R., additional, Fleshman, J., additional, Bem, J., additional, and Compton, C., additional
- Published
- 2004
- Full Text
- View/download PDF
14. p53 Missense Mutations in Microdissected High-Grade Ductal Carcinoma In Situ of the Breast
- Author
-
Done, S. J., primary, Eskandarian, S., additional, Bull, S., additional, Redston, M., additional, and Andrulis, I. L., additional
- Published
- 2001
- Full Text
- View/download PDF
15. A missense mutation in both hMSH2 andAPC in an Ashkenazi Jewish HNPCC kindred: implications for clinical screening
- Author
-
YUAN, Z. Q., primary, WONG, N., additional, FOULKES, W. D., additional, ALPERT, L., additional, MANGANARO, F., additional, ANDREUTTI-ZAUGG, C., additional, IGGO, R., additional, ANTHONY, K., additional, HSIEH, E., additional, REDSTON, M., additional, PINSKY, L., additional, TRIFIRO, M., additional, GORDON, P. H., additional, and LASKO, D., additional
- Published
- 1999
- Full Text
- View/download PDF
16. Mismatch Repair Gene Defects Contribute to the Genetic Basis of Double Primary Cancers of the Colorectum and Endometrium
- Author
-
Millar, A. L., primary, Pal, T., additional, Madlensky, L., additional, Sherman, C., additional, Temple, L., additional, Mitri, A., additional, Cheng, H., additional, Marcus, V., additional, Gallinger, S., additional, Redston, M., additional, Bapat, B., additional, and Narod, S., additional
- Published
- 1999
- Full Text
- View/download PDF
17. Hereditary non-polyposis colorectal cancer (HNPCC): Cancer patterns in germline mutation (hMSH2, hMLH1) positive (M+) and mutation negative (M−) families
- Author
-
Temple, LKF, primary, Bapat, B, additional, Madlensky, L, additional, Redston, M, additional, Hiruki, T, additional, Cohen, Z, additional, McLeod, RS, additional, and Gallinger, S, additional
- Published
- 1998
- Full Text
- View/download PDF
18. MSH2 deficient mice are viable and susceptible to lymphoid tumours
- Author
-
Reitmair, A.H., primary, Schmits, R., additional, Ewel, A., additional, Bapat, B., additional, Redston, M., additional, Mitri, A., additional, Waterhouse, P., additional, Mittrücker, H.-W, additional, Wakeham, A., additional, Liu, B., additional, Thomason, A., additional, Griesser, H., additional, Gallinger, S., additional, Ballhausen, W.G., additional, Fishel, R., additional, and Mak, T. W., additional
- Published
- 1995
- Full Text
- View/download PDF
19. Microsatellite instability and loss of heterozygosity at chromosomal location 18q: prospective evaluation of biomarkers for stages II and III colon cancer--a study of CALGB 9581 and 89803.
- Author
-
Bertagnolli MM, Redston M, Compton CC, Niedzwiecki D, Mayer RJ, Goldberg RM, Colacchio TA, Saltz LB, Warren RS, Bertagnolli, Monica M, Redston, Mark, Compton, Carolyn C, Niedzwiecki, Donna, Mayer, Robert J, Goldberg, Richard M, Colacchio, Thomas A, Saltz, Leonard B, and Warren, Robert S
- Published
- 2011
- Full Text
- View/download PDF
20. Microsatellite instability predicts improved response to adjuvant therapy with irinotecan, fluorouracil, and leucovorin in stage III colon cancer: Cancer and Leukemia Group B Protocol 89803.
- Author
-
Bertagnolli MM, Niedzwiecki D, Compton CC, Hahn HP, Hall M, Damas B, Jewell SD, Mayer RJ, Goldberg RM, Saltz LB, Warren RS, Redston M, Bertagnolli, Monica M, Niedzwiecki, Donna, Compton, Carolyn C, Hahn, Hejin P, Hall, Margaret, Damas, Beatrice, Jewell, Scott D, and Mayer, Robert J
- Published
- 2009
- Full Text
- View/download PDF
21. Carcinogenesis in the GI tract: from morphology to genetics and back again.
- Author
-
Redston, Mark and Redston, M
- Published
- 2001
- Full Text
- View/download PDF
22. A missense mutation in both hMSH2 and APC in an Ashkenazi Jewish HNPCC kindred: implications for clinical screening
- Author
-
Redston, M., Yuan, Z.Q., Pinsky, L., Wong, N., Trifiro, M., Foulkes, W.D., Gordon, P.H., Alpert, L., Lasko, D., Manganaro, F., Andreutti-Zaugg, C., Iggo, R., Anthony, K., and Hsieh, E.
- Published
- 1999
23. ALLELOTYPE OF PANCREATIC ADENOCARCINOMA USING XENOGRAFT ENRICHMENT
- Author
-
Stephan Hahn, Seymour, A. B., Hoque, Atms, Schutte, M., Dacosta, L. T., Redston, M. S., Caldas, C., Weinstein, C. L., Fischer, A., Yeo, C. J., Hruban, R. H., and Kern, S. E.
24. Inherited predisposition to pancreatic adenocarcinoma: Role of family history and germ-line p16, BRCA1, and BRCA2 mutations
- Author
-
Geeta Lal, Liu, G., Schmocker, B., Kaurah, P., Ozcelik, H., Narod, S. A., Redston, M., and Gallinger, S.
25. Suppression of intestinal polyps in Msh2-deficient and non-Msh2-deficient multiple intestinal neoplasia mice by a specific cyclooxygenase-2 inhibitor and by a dual cyclooxygenase-1/2 inhibitor
- Author
-
Lal, G., Ash, C., Hay, K., Gallinger, S., Redston, M., Mak, T., Kwong, E., Bruno Hancock, Kargman, S., and Evans, J. F.
26. Mutation profiling of mismatch repair-deficient colorectal cancers using an in silico genome scan to identify coding microsatellites
- Author
-
Park, J., Doron Betel, Gryfe, R., Michalickova, K., Di Nicola, N., Gallinger, S., Hogue, C. W. V., and Redston, M.
27. DETECTION OF K-RAS MUTATIONS IN THE STOOL OF PATIENTS WITH PANCREATIC ADENOCARCINOMA AND PANCREATIC DUCTAL HYPERPLASIA
- Author
-
Caldas, C., Stephan Hahn, Hruban, R. H., Redston, M. S., Yeo, C. J., and Kern, S. E.
28. Erratum: Colorectal carcinomas in mice lacking the catalytic subunit of PI(3)Kγ (Nature (2000) 406 (897-902))
- Author
-
Sasaki, T., Irie-Sasaki, J., Horie, Y., Bachmaier, K., Fata, J. E., Li, M., Suzuki, A., Bouchard, D., Ho, A., Redston, M., Gallinger, S., Khokha, R., Mak, T. W., Hawkins, P. T., len stephens, Scherer, S. W., Tsao, M., and Penninger, J. M.
29. Colorectal carcinomas in mice lacking the catalytic subunit of PI(3)K gamma
- Author
-
Sasaki, T., Irie-Sasaki, J., Horie, Y., Bachmaier, K., Fata, Je, Li, M., Suzuki, A., Bouchard, D., Ho, A., Redston, M., Gallinger, S., Khokha, R., Mak, Tw, Hawkins, Pt, Stephens, L., Stephen W. Scherer, Tsao, M., and Penninger, Jm
30. DNA repair enzyme expression and differential response to temozolomide in a patient with both glioblastoma and metastatic pancreatic neuroendocrine tumor.
- Author
-
Bracht LK, Wen P, Meyerhardt JA, Kulke MH, Hornick JL, Redston M, LaFrankie DC, Black PM, Kesari S, Norden A, Drappatz J, Bracht, Lynn-Kristin, Wen, Patrick, Meyerhardt, Jeffrey A, Kulke, Matthew H, Hornick, Jason L, Redston, Mark, LaFrankie, Debra Conrad, Black, Peter M, and Kesari, Santosh
- Published
- 2008
- Full Text
- View/download PDF
31. Use of RNA Interference to Dissect the Role of Carcinoembryonic Antigen-Related Cell Adhesion Molecule 6 (CEACAM 6) in Pancreatic Adenocarcinoma.
- Author
-
Duxbury, M. S., Matros, E., Redston, M., Ashley, S. W., and Whang, E. E.
- Subjects
ADENOCARCINOMA ,CANCER ,MYC proteins ,TUMOR proteins ,OLIGONUCLEOTIDES ,BIOINFORMATICS ,COMPUTERS in biology ,RNA - Abstract
Transcriptomic analysis identified CEACAM6 as a potential oncoprotein in pancreatic adenocarcinoma. Using retroviral delivery, researchers determined the effects of CEACAM6-specific small interfering RNA (siRNA) on pancreatic adenocarcinoma in vitro and evaluated the siRNA oligonucleotide in vivo. CEACAM6 expression was quantified by realtime RTPCR and Western analysis in PANCl, Capan2, MIAPaCa2 and MIAAR. CEACAM6 was overexpressed by transient transfection. siRNAs, designed using a rational in silico bioinformatics approach, were screened for CEACAM6 gene silencing activity using a novel reverse-transfection siRNA microarray. Cells were infected with replication-deficient retrovirus, engineered to deliver CEACAM6 siRNA (siCEACAM6).
- Published
- 2004
32. Molecular biology of colorectal cancer
- Author
-
Gryfe, R., Bapat, B., Gallinger, S., Swallow, C., Redston, M., and Couture, J.
- Abstract
Colorectal cancer is a significant cause of morbidity and mortality in Western populations. This cancer develops as a result of the pathologic transformation of normal colonic epithelium to an adenomatous polyp and ultimately an invasive cancer. The multistep progression requires years and possibly decades and is accompanied by a number of recently characterized genetic alterations. Mutations in two classes of genes, tumor-suppressor genes and proto-oncogenes, are thought to impart a proliferative advantage to cells and contribute to development of the malignant phenotype. Inactivating mutations of both copies (alleles) of the adenomatous polyposis coli (APC) gene-a tumor-suppressor gene on chromosome 5q-mark one of the earliest events in colorectal carcinogenesis. Germline mutation of the APC gene and subsequent somatic mutation of the second APC allele cause the inherited familial adenomatous polyposis syndrome. This syndrome is characterized by the presence of hundreds to thousands of colonic adenomatous polyps. If these polyps are left untreated, colorectal cancer develops. Mutation leading to dysregulation of the K-ras protooncogene is also thought to be an early event in colon cancer formation. Conversely, loss of heterozygosity on the long arm of chromosome 18 (18q) occurs later in the sequence of development from adenoma to carcinoma, and this mutation may predict poor prognosis. Loss of the 18q region is thought to contribute to inactivation of the DCC tumor-suppressor gene. More recent evidence suggests that other tumor-suppressor genes-DPC4 and MADR2 of the transforming growth factor @b (TGF-@b) pathway-also may be inactivated by allelic loss on chromosome 18q. In addition, mutation of the tumor-suppressor gene p53 on chromosome 17p appears to be a late phenomenon in colorectal carcinogenesis. This mutation may allow the growing tumor with multiple genetic alterations to evade cell cycle arrest and apoptosis. Neoplastic progression is probably accompanied by additional, undiscovered genetic events, which are indicated by allelic loss on chromosomes 1q, 4p, 6p, 8p, 9q, and 22q in 25% to 50% of colorectal cancers. Recently, a third class of genes, DNA repair genes, has been implicated in tumorigenesis of colorectal cancer. Study findings suggest that DNA mismatch repair deficiency, due to germline mutation of the hMSH2, hMLH1, hPMS1, or hPMS2 genes, contributes to development of hereditary nonpolyposis colorectal cancer. The majority of tumors in patients with this disease and 10% to 15% of sporadic colon cancers display microsatellite instability, also know as the replication error positive (RER+) phenotype. This molecular marker of DNA mismatch repair deficiency may predict improved patient survival. Mismatch repair deficiency is thought to lead to mutation and inactivation of the genes for type II TGF-@b receptor and insulin-like growth-factor II receptor: Individuals from families at high risk for colorectal cancer (hereditary nonpolyposis colorectal cancer or familial adenomatous polyposis) should be offered genetic counseling, predictive molecular testing, and when indicated, endoscopic surveillance at appropriate intervals. Recent studies have examined colorectal carcinogenesis in the light of other genetic processes. Telomerase activity is present in almost all cancers, including colorectal cancer, but rarely in benign lesions such as adenomatous polyps or normal tissues. Furthermore, genetic alterations that allow transformed colorectal epithelial cells to escape cell cycle arrest or apoptosis also have been recognized. In addition, hypomethylation or hypermethylation of DNA sequences may alter gene expression without nucleic acid mutation.
- Published
- 1997
- Full Text
- View/download PDF
33. Celecoxib for the prevention of sporadic colorectal adenomas.
- Author
-
Bertagnolli MM, Eagle CJ, Zauber AG, Redston M, Solomon SD, Kim K, Tang J, Rosenstein RB, Witts J, Corle D, Hess TM, Woloj GM, Boisserie F, Anderson WF, Viner JL, Bagheri D, Burn J, Chung DC, Dewar T, and Foley TR
- Abstract
Background: Studies showing that drugs that inhibit cyclooxygenase-2 (COX-2) reduce the number of colorectal adenomas in animals and patients with familial adenomatous polyposis suggest that COX-2 inhibitors may also prevent sporadic colorectal neoplasia.Methods: We randomly assigned patients who had adenomas removed before study entry to receive placebo (679 patients) or 200 mg (685 patients) or 400 mg (671 patients) of celecoxib twice daily. Randomization was stratified for the use of low-dose aspirin. Follow-up colonoscopies were performed at one and three years after randomization. The occurrence of newly detected colorectal adenomas was compared among the groups with the life-table extension of the Mantel-Haenszel test.Results: Follow-up colonoscopies were completed at year 1 in 89.5 percent of randomized patients, and at year 3 in 75.7 percent. The estimated cumulative incidence of the detection of one or more adenomas by year 3 was 60.7 percent for patients receiving placebo, as compared with 43.2 percent for those receiving 200 mg of celecoxib twice a day (risk ratio, 0.67; 95 percent confidence interval, 0.59 to 0.77; P<0.001) and 37.5 percent for those receiving 400 mg of celecoxib twice a day (risk ratio, 0.55; 95 percent confidence interval, 0.48 to 0.64; P<0.001). Serious adverse events occurred in 18.8 percent of patients in the placebo group, as compared with 20.4 percent of those in the low-dose celecoxib group (risk ratio, 1.1; 95 percent confidence interval, 0.9 to 1.3; P=0.5) and 23.0 percent of those in the high-dose group (risk ratio, 1.2; 95 percent confidence interval, 1.0 to 1.5; P=0.06). As compared with placebo, celecoxib was associated with an increased risk of cardiovascular events (risk ratio for the low dose, 2.6; 95 percent confidence interval, 1.1 to 6.1; and risk ratio for the high dose, 3.4; 95 percent confidence interval, 1.5 to 7.9).Conclusions: These findings indicate that celecoxib is an effective agent for the prevention of colorectal adenomas but, because of potential cardiovascular events, cannot be routinely recommended for this indication. (ClinicalTrials.gov number, NCT00005094 [ClinicalTrials.gov].). [ABSTRACT FROM AUTHOR]- Published
- 2006
34. Tumor microsatellite-instability status as a predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer.
- Author
-
Ribic CM, Sargent DJ, Moore MJ, Thibodeau SN, French AJ, Goldberg RM, Hamilton SR, Laurent-Puig P, Gryfe R, Shepherd LE, Tu D, Redston M, Gallinger S, Ribic, Christine M, Sargent, Daniel J, Moore, Malcolm J, Thibodeau, Stephen N, French, Amy J, Goldberg, Richard M, and Hamilton, Stanley R
- Abstract
Background: Colon cancers with high-frequency microsatellite instability have clinical and pathological features that distinguish them from microsatellite-stable tumors. We investigated the usefulness of microsatellite-instability status as a predictor of the benefit of adjuvant chemotherapy with fluorouracil in stage II and stage III colon cancer.Methods: Tumor specimens were collected from patients with colon cancer who were enrolled in randomized trials of fluorouracil-based adjuvant chemotherapy. Microsatellite instability was assessed with the use of mononucleotide and dinucleotide markers.Results: Of 570 tissue specimens, 95 (16.7 percent) exhibited high-frequency microsatellite instability. Among 287 patients who did not receive adjuvant therapy, those with tumors displaying high-frequency microsatellite instability had a better five-year rate of overall survival than patients with tumors exhibiting microsatellite stability or low-frequency instability (hazard ratio for death, 0.31 [95 percent confidence interval, 0.14 to 0.72]; P=0.004). Among patients who received adjuvant chemotherapy, high-frequency microsatellite instability was not correlated with increased overall survival (hazard ratio for death, 1.07 [95 percent confidence interval, 0.62 to 1.86]; P=0.80). The benefit of treatment differed significantly according to the microsatellite-instability status (P=0.01). Adjuvant chemotherapy improved overall survival among patients with microsatellite-stable tumors or tumors exhibiting low-frequency microsatellite instability, according to a multivariate analysis adjusted for stage and grade (hazard ratio for death, 0.72 [95 percent confidence interval, 0.53 to 0.99]; P=0.04). By contrast, there was no benefit of adjuvant chemotherapy in the group with high-frequency microsatellite instability.Conclusions: Fluorouracil-based adjuvant chemotherapy benefited patients with stage II or stage III colon cancer with microsatellite-stable tumors or tumors exhibiting low-frequency microsatellite instability but not those with tumors exhibiting high-frequency microsatellite instability. [ABSTRACT FROM AUTHOR]- Published
- 2003
35. Common and distinct genomic events in sporadic colorectal cancer and diverse cancer types
- Author
-
Eric S. Martin, Raju Kucherlapati, Alexei Protopopov, Elena Ivanova, Bin Feng, Cameron Brennan, Yonghong Xiao, Giovanni Tonon, Lynda Chin, Gerald Bailey, Ronald A. DePinho, Kate Montgomery, Raktim Sinha, Alec C. Kimmelman, Mark Redston, Martin, E, Tonon, G, Sinha, R, Xiao, Y, Feng, B, Kimmelman, Ac, Protopopov, A, Ivanova, E, Brennan, C, Montgomery, K, Kucherlapati, R, Bailey, G, Redston, M, Chin, L, and Depinho, Ra
- Subjects
Cancer Research ,Colorectal cancer ,Copy number analysis ,Genomics ,Biology ,Genome ,Chromosome instability ,Cell Line, Tumor ,medicine ,Humans ,Genetics ,Chromosome Aberrations ,Models, Genetic ,Gene Expression Profiling ,Microsatellite instability ,Cancer ,Nucleic Acid Hybridization ,Sequence Analysis, DNA ,medicine.disease ,Immunohistochemistry ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,Oncology ,Mutation ,Colorectal Neoplasms ,Genes, Neoplasm - Abstract
Colorectal cancer (CRC) is a major cause of cancer morbidity and mortality, and elucidation of its underlying genetics has advanced diagnostic screening, early detection, and treatment. Because CRC genomes are characterized by numerous non-random chromosomal structural alterations, we sought to delimit regions of recurrent amplifications and deletions in a collection of 42 primary specimens and 37 tumor cell lines derived from chromosomal instability neoplasia and microsatellite instability neoplasia CRC subtypes and to compare the pattern of genomic aberrations in CRC with those in other cancers. Application of oligomer-based array-comparative genome hybridization and custom analytic tools identified 50 minimal common regions (MCRs) of copy number alterations, 28 amplifications, and 22 deletions. Fifteen were highly recurrent and focal (
- Published
- 2007
36. Clinical presentation and outcomes of Helicobacter heilmannii gastritis in children in the New England region of the United States.
- Author
-
Chang D, Gluchowski NL, Abu Alfa AK, Goldsmith JD, Redston M, and Bonilla S
- Subjects
- Child, Humans, Male, Female, Retrospective Studies, New England, Nausea, Abdominal Pain, Helicobacter heilmannii, Gastritis diagnosis, Gastritis drug therapy, Gastritis microbiology, Helicobacter Infections diagnosis, Helicobacter Infections drug therapy, Helicobacter Infections microbiology, Helicobacter pylori
- Abstract
Objective: To describe the clinical, endoscopic, histologic, and treatment outcomes of Helicobacter heilmannii (H. heilmannii) associated gastritis in children in the New England region of the United States., Methods: Retrospective study of children (1-18 years) with H. heilmannii identified on gastric mucosal biopsies from two pediatric centers over a 21-year period, January 2000-December 2021. Cases were identified by querying pathology databases at each institution. Demographic and clinical data were obtained from the medical record. Endoscopic and histologic findings were extracted from endoscopy and pathology reports, respectively., Results: Thirty-eight children were diagnosed with H. heilmannii-associated gastritis during the study period. The mean age at diagnosis was 10.1 ± 5.3 years, and 25/38 (66%) cases were male. Abdominal pain (32%) and nausea with or without vomiting (26%) were the most common symptoms. Thirty-two children (84%) had endoscopic findings including gastric nodularity (55%) and erythema (26%). All children had histologic signs of chronic gastritis, including those with normal endoscopic exams. Antibiotic regimens used for treating Helicobacter pylori were frequently prescribed. Of the 17 children who underwent a follow-up endoscopy (range 2-68 months), 15 (88%) did not have H. heilmannii identified on gastric biopsies., Conclusion: H. heilmannii was an infrequent but potential cause of epigastric abdominal pain and nausea in our cohort of New England children. While morphologically distinct from H. pylori, the bacteria can result in similar endoscopic and histologic findings of nodularity and chronic gastritis, respectively. The rate of eradication, as assessed by histology following treatment with H. pylori therapies, was below the 90% recommended goal for antimicrobial therapies., (© 2023 European Society for Pediatric Gastroenterology, Hepatology, and Nutrition and North American Society for Pediatric Gastroenterology, Hepatology, and Nutrition.)
- Published
- 2024
- Full Text
- View/download PDF
37. Ang1 and Ang4 differentially affect colitis and carcinogenesis in an AOM-DSS mouse model.
- Author
-
Hu A, Roberts C, Moscalu A, Redston M, and Yoo J
- Subjects
- Animals, Mice, Carcinogenesis genetics, Disease Models, Animal, Interleukin-10 genetics, Interleukin-33, Interleukin-6 genetics, Mice, Inbred C57BL, Tumor Necrosis Factor-alpha genetics, Colitis chemically induced, Colitis complications, Colitis genetics, Colitis-Associated Neoplasms, Ribonuclease, Pancreatic genetics
- Abstract
Introduction: Angiogenin-1 (Ang1) and angiogenin-4 (Ang4) are 14-kDa ribonucleases with potent angiogenic and antimicrobial properties. The role of Ang1 and Ang4 in chronic colitis and colitis-associated cancer has not been previously studied., Methods: Wild-type (WT) and angiogenin-1 knock-out (Ang1-KO) C57BL/6 mice were given azoxymethane, a colon carcinogen, 2 days in advance of three cycles of 3.5% dextran sodium sulfate (DSS). Disease activity index (DAI) was recorded, a colonoscopy was performed after each DSS treatment, and mice were euthanized (colitis, recovery, cancer) with tissue evaluated by histopathology. Ang1, Ang4, TNF-α, Il-1F062, IL-6, IL-10, IL-23, IL-33 mRNA levels were analyzed by RT-PCR., Results: Ang1-KO mice exhibited more severe colitis compared to WT mice during both the acute (P<0.05) and recovery (P<0.05) phases of each DSS cycle. Consistent with these results, colonic TNF-α, IL1-β, IL-6, IL-10, and IL-33 mRNA levels were significantly upregulated in Ang1-KO mice (P<0.05). While Ang4 increased to similar levels in both WT and Ang1-KO mice during colitis and recovery phases, WT mice were distinguished by a significant upregulation of Ang1. Interestingly, despite the reduced colitis, WT mice developed significantly more tumors compared to Ang1-KO mice (P<0.05). 134 tumors formed in WT mice (4.6 tumors/mouse) while only 46 tumors formed (1.5 tumors/mice) in Ang1-KO mice, which were also characterized by a 34-fold decrease in Ang4 compared to WT mice and the complete absence of Ang1., Conclusions: In a mouse model of colitis-associated cancer, Ang1-KO mice develop more severe colitis, but fewer tumors compared to WT mice. Ang1 levels correlate with the severity of colitis and the development of colitis-associated cancer, while Ang4 was upregulated during both colitis and cancer. Ang1 and Ang4 play important regulatory roles in the response to chronic colitis and the development of colitis-associated cancer and may serve as novel therapeutic targets., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2023 Hu et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
38. Prevalence of Mismatch-Repair Deficiency in Rectal Adenocarcinomas.
- Author
-
Papke DJ Jr, Yurgelun MB, Noffsinger AE, Turner KO, Genta RM, and Redston M
- Subjects
- Humans, Neoplastic Syndromes, Hereditary epidemiology, Neoplastic Syndromes, Hereditary genetics, Neoplastic Syndromes, Hereditary pathology, Prevalence, Adenocarcinoma epidemiology, Adenocarcinoma genetics, Adenocarcinoma pathology, Colorectal Neoplasms epidemiology, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, DNA Mismatch Repair genetics
- Published
- 2022
- Full Text
- View/download PDF
39. Giant Filiform Polyposis of the Colon in a 6-Year Old Male With Ulcerative Colitis.
- Author
-
Slack JC, Redston M, Goldsmith JD, Bousvaros B, and Levy Zitomersky N
- Subjects
- Child, Humans, Male, Colitis, Ulcerative complications, Colonic Polyps complications, Colonic Polyps diagnosis, Colonic Polyps surgery, Colorectal Neoplasms
- Abstract
Competing Interests: The authors report no conflicts of interest.
- Published
- 2022
- Full Text
- View/download PDF
40. Abnormal TP53 Predicts Risk of Progression in Patients With Barrett's Esophagus Regardless of a Diagnosis of Dysplasia.
- Author
-
Redston M, Noffsinger A, Kim A, Akarca FG, Rara M, Stapleton D, Nowden L, Lash R, Bass AJ, and Stachler MD
- Subjects
- Adenocarcinoma pathology, Adult, Aged, Aged, 80 and over, Barrett Esophagus pathology, Disease Progression, Esophageal Neoplasms pathology, Female, Humans, Immunohistochemistry, Male, Middle Aged, Prognosis, Risk Assessment, Adenocarcinoma metabolism, Barrett Esophagus metabolism, Esophageal Neoplasms metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Background and Aims: Barrett's esophagus (BE) is the precursor to esophageal adenocarcinoma. A major challenge is identifying the small group with BE who will progress to advanced disease from the many who will not. Assessment of p53 status has promise as a predictive biomarker, but analytic limitations and lack of validation have precluded its use. The aim of this study was to develop a robust criteria for grading abnormal immunohistochemical (IHC) expression of p53 and to test its utility as a biomarker for progression in BE., Methods: Criteria for abnormal IHC of p53 were developed in BE biopsies and validated with sequencing to assess TP53 mutations. The utility of p53 IHC as a biomarker for progression of BE was tested retrospectively in 561 patients with BE with or without known progression. The findings were prospectively validated in a clinical practice setting in 1487 patients with BE., Results: Abnormal p53 IHC highly correlated with TP53 mutation status (90.6% agreement) and was strongly associated with neoplastic progression in the retrospective cohorts, regardless of histologic diagnosis (P < .001). In the retrospective cohort, abnormal p53 was associated with a hazard ratio of 5.03 (95% confidence interval, 3.88-6.5) and a hazard ratio of 5.27 (95% confidence interval, 3.93-7.07) for patients with exclusively nondysplastic disease before progression. In the prospective validation cohort, p53 IHC predicted progression among nondysplastic BE, indefinite for dysplasia, and low-grade dysplasia (P < .001)., Conclusions: p53 IHC identifies patients with BE at higher risk of progression, including in patients without evidence of dysplasia. p53 IHC is inexpensive, easily integrated into routine practice, and should be considered in biopsies from all BE patients without high-grade dysplasia or cancer., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
41. Florid Foreign Body-type Giant Cell Response to Keratin Is Associated With Improved Overall Survival in Patients Receiving Preoperative Therapy for Esophageal Squamous Cell Carcinoma.
- Author
-
Chen W, Zhao L, Agoston A, White A, Mazzola E, Boyle PJ, Deshpande V, Hornick JL, Bueno R, Bass AJ, Enzinger P, Mamon H, Redston M, and Patil DT
- Subjects
- Adult, Aged, Aged, 80 and over, Chemotherapy, Adjuvant, Esophageal Neoplasms chemistry, Esophageal Neoplasms mortality, Esophageal Neoplasms pathology, Esophageal Squamous Cell Carcinoma mortality, Esophageal Squamous Cell Carcinoma pathology, Female, Humans, Male, Middle Aged, Retrospective Studies, Risk Assessment, Risk Factors, Time Factors, Treatment Outcome, Biomarkers, Tumor analysis, Esophageal Neoplasms therapy, Esophageal Squamous Cell Carcinoma therapy, Esophagectomy adverse effects, Esophagectomy mortality, Gastrectomy adverse effects, Gastrectomy mortality, Granuloma, Foreign-Body pathology, Keratins analysis, Neoadjuvant Therapy adverse effects, Neoadjuvant Therapy mortality
- Abstract
While most resection specimens from patients with neoadjuvantly treated esophageal squamous cell carcinoma show therapy-related changes in the form of inflammation and fibrosis, others harbor a florid foreign body-type giant cell response to keratin debris. The purpose of our study was to perform a detailed clinicopathologic analysis of these histologic types of treatment responses and correlate these findings with patient outcome. Clinical and pathologic parameters from 110 esophagogastrectomies were recorded and analyzed. Two main types of histologic responses were observed: inflammatory-predominant response (59%) and florid foreign body-type giant cell response to keratin (41%). Irrespective of cG, cTNM, and amount of residual cancer, florid foreign body-type giant cell reaction was predominantly noted deep within the esophageal wall, while the inflammatory response was restricted to the mucosa, submucosa, and inner half of muscularis propria. Patients with foreign body-type giant cell response showed significantly better overall survival compared with the inflammatory response group (log-rank test P=0.015). Florid foreign body-type giant cell response was the only factor associated with improved survival in a multivariable analysis for overall survival (hazard ratio=0.5; 95% confidence interval=0.3-1.0; P=0.038), but not in the model for disease-specific survival, whereas ypTNM stage II was the only significant risk factor for disease-specific survival in multivariable analysis (hazard ratio=3.4; 95% confidence interval=1.0-11.2; P=0.047). Our results suggest that in addition to the College of American Pathologists Tumor Regression Score and ypTNM stage, subtype of histologic response to therapy may represent another prognostic marker for neoadjuvantly treated esophageal squamous cell carcinoma., Competing Interests: Conflicts of Interest and Source of Funding: R.B. received grants from NCI, NHLBI, DOD, Roche, Genentech, Verastem, Merck, Siemens Gritstone; Provides Consultations/scientific panel/Scientific Advisory for Regeron, Navigation Sciences, AstraZeneca, Intuitive, Siemens, Novocure, Johnson and Johnson; Holds Equity/Patents for Navigation Sciences. P.E. serves as an Advisor/Board Member for ALX Oncology, Arcus Bioscience, Astellas, AstraZeneca, BMS, Celgene, Daiichi-Sankyo, Five Prime, Lilly, Loxo, Merck, Taiho, Takeda, Zymeworks; is a Consultant/Independent Contractor for Istari, Ono, Legend, Xencor; Received Honorarium Receipt for ALX Oncology, Arcus Bioscience, Astellas, Astra-Zeneca, BMS, Celgene, Daiichi-Sankyo, Five Prime, Istari, Legend, Lilly, Loxo, Merck, Ono, Taiho, Takeda, Xencor, Zymeworks. H.M.: received honoraria from UpToDate as a co-author of several chapters on GI malignancies (none on esophageal cancer); is a member of a Scientific Advisory Committee at Merck, which is related to esophageal cancer. For the remaining authors none were declared., (Copyright © 2021 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
42. Detection of Mutations in Barrett's Esophagus Before Progression to High-Grade Dysplasia or Adenocarcinoma.
- Author
-
Stachler MD, Camarda ND, Deitrick C, Kim A, Agoston AT, Odze RD, Hornick JL, Nag A, Thorner AR, Ducar M, Noffsinger A, Lash RH, Redston M, Carter SL, Davison JM, and Bass AJ
- Subjects
- Adenocarcinoma pathology, Adult, Aged, Aged, 80 and over, Barrett Esophagus pathology, Biopsy, Case-Control Studies, Disease Progression, Esophageal Neoplasms pathology, Esophagoscopy, Female, Humans, Male, Middle Aged, Mutation, Precancerous Conditions pathology, Prognosis, Retrospective Studies, Adenocarcinoma genetics, Barrett Esophagus genetics, Esophageal Neoplasms genetics, Precancerous Conditions genetics, Tumor Suppressor Protein p53 genetics
- Abstract
Background & Aims: Barrett's esophagus (BE) is the greatest risk factor for esophageal adenocarcinoma (EAC), but only a small proportion of patients with BE develop cancer. Biomarkers might be able to identify patients at highest risk of progression. We investigated genomic differences in surveillance biopsies collected from patients whose BE subsequently progressed compared to patients whose disease did not progress., Methods: We performed a retrospective case-control study of 24 patients with BE that progressed to high-grade dysplasia (HGD, n = 14) or EAC (n = 10). The control group (n = 73, called non-progressors) comprised patients with BE and at least 5 years of total endoscopic biopsy surveillance without progression to HGD or EAC. From each patient, we selected a single tissue sample obtained more than 1 year before progression (cases) or more than 2 years before the end of follow-up (controls). Pathogenic mutations, gene copy numbers, and ploidy were compared between samples from progressors and non-progressors., Results: TP53 mutations were detected in 46% of samples from progressors and 5% of non-progressors. In this case-control sample set, TP53 mutations in BE tissues increased the adjusted risk of progression 13.8-fold (95% confidence interval, 3.2-61.0) (P < .001). We did not observe significant differences in ploidy or copy-number profile between groups. We identified 147 pathogenic mutations in 57 distinct genes-the average number of pathogenic mutations was higher in samples from progressors (n = 2.5) than non-progressors (n = 1.2) (P < .001). TP53 and other somatic mutations were recurrently detected in samples with limited copy-number changes (aneuploidy)., Conclusions: In genomic analyses of BE tissues from patients with or without later progression to HGD or EAC, we found significantly higher numbers of TP53 mutations in BE from patients with subsequent progression. These mutations were frequently detected before the onset of dysplasia or substantial changes in copy number., (Copyright © 2018 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
43. A Study of Thymidylate Synthase Expression as a Biomarker for Resectable Colon Cancer: Alliance (Cancer and Leukemia Group B) 9581 and 89803.
- Author
-
Niedzwiecki D, Hasson RM, Lenz HJ, Ye C, Redston M, Ogino S, Fuchs CS, Compton CC, Mayer RJ, Goldberg RM, Colacchio TA, Saltz LB, Warren RS, and Bertagnolli MM
- Subjects
- Aged, Biomarkers, Tumor biosynthesis, Colonic Neoplasms pathology, Colonic Neoplasms surgery, Combined Modality Therapy, Disease-Free Survival, Female, Fluorouracil administration & dosage, Gene Expression Regulation, Neoplastic, Humans, Male, Microsatellite Instability, Middle Aged, Neoplasm Staging, Prognosis, Thymidylate Synthase antagonists & inhibitors, Treatment Outcome, Biomarkers, Tumor genetics, Colonic Neoplasms drug therapy, Colonic Neoplasms genetics, Thymidylate Synthase genetics
- Abstract
Purpose: Tumor levels of thymidylate synthase (TS), a target of 5-fluorouracil (5-FU)-based chemotherapy for colorectal cancer, have been studied as a predictive or prognostic biomarker with mixed results., Patients and Methods: Tumor TS levels were prospectively evaluated in two adjuvant therapy trials for patients with resected stage II or III colon cancer. TS expression was determined by standard immunohistochemistry and by automated quantitative analysis. Tumor mismatch repair deficiency (MMR-D) and BRAF c.1799T > A (p.V600E) mutation status were also examined. Relationships between tumor TS, MMR-D, and BRAF mutation status, overall survival (OS), and disease-free survival (DFS) were investigated in the subset of stage III patients., Results: Patients whose tumors demonstrated high TS expression experienced better treatment outcomes, with DFS hazard ratio (HR) = 0.67, 95% confidence interval (CI) = 0.53, 0.84; and OS HR = 0.68, 95% CI = 0.53, 0.88, for high versus low TS expression, respectively. No significant interaction between TS expression and stage was observed (DFS: interaction HR = 0.94; OS: interaction HR = 0.94). Tumors with high TS expression were more likely to demonstrate MMR-D (22.2% vs. 12.8%; p = .0003). Patients whose tumors demonstrated both high TS and MMR-D had a 7-year DFS of 77%, compared with 58% for those whose tumors had low TS and were non-MMR-D (log-rank p = .0006). Tumor TS expression did not predict benefit of a particular therapeutic regimen., Conclusion: This large prospective analysis showed that high tumor TS levels were associated with improved DFS and OS following adjuvant therapy for colon cancer, although tumor TS expression did not predict benefit of 5-FU-based chemotherapy. The Oncologist 2017;22:107-114Implications for Practice: This study finds that measurement of tumor levels of thymidylate synthase is not helpful in assigning specific adjuvant treatment for colorectal cancer. It also highlights the importance of using prospective analyses within treatment clinical trials as the optimal method of determining biomarker utility., (© AlphaMed Press 2016.)
- Published
- 2017
- Full Text
- View/download PDF
44. Importance of universal mismatch repair protein immunohistochemistry in patients with sebaceous neoplasia as an initial screening tool for Muir-Torre syndrome.
- Author
-
Jessup CJ, Redston M, Tilton E, and Reimann JD
- Subjects
- Adaptor Proteins, Signal Transducing analysis, Adenoma chemistry, Adenoma pathology, Adenosine Triphosphatases analysis, Adult, Aged, Aged, 80 and over, Biopsy, Carcinoma chemistry, Carcinoma pathology, DNA Repair Enzymes analysis, DNA-Binding Proteins analysis, Female, Humans, Male, Middle Aged, Mismatch Repair Endonuclease PMS2, Muir-Torre Syndrome metabolism, Muir-Torre Syndrome pathology, MutL Protein Homolog 1, MutS Homolog 2 Protein analysis, Nuclear Proteins analysis, Predictive Value of Tests, Risk Factors, Adenoma diagnosis, Biomarkers, Tumor analysis, Carcinoma diagnosis, DNA Mismatch Repair, Immunohistochemistry, Muir-Torre Syndrome diagnosis
- Abstract
Muir-Torre syndrome, a Lynch syndrome variant, is characterized by sebaceous neoplasia plus one or more malignancies, typically colon cancer. The significance of DNA mismatch repair (MMR) deficiency detection by immunohistochemistry (IHC) in colorectal carcinomas is well established and is recommended as a screening tool for Lynch syndrome in newly diagnosed colorectal carcinomas. In comparison, literature on IHC application to detect MMR proteins (MLH1, MSH2, MSH6, and PMS2) in sebaceous neoplasia has been less studied and has been derived almost exclusively from tertiary care centers. Herein we describe the largest series to date characterizing MMR deficiency in sebaceous neoplasms, as well as the relative frequencies of each deficiency. Two hundred sixteen consecutive sebaceous neoplasms (216 patients) were analyzed from a community practice setting (133 sebaceous adenomas, 68 sebaceomas, 15 sebaceous carcinomas). One hundred forty-three were MMR deficient (66%), of which 90 were MSH2/MSH6 deficient (63%), 27 MLH1/PMS2 deficient (19%), 22 MSH6 deficient (15%), and 4 PMS2 deficient (3%). MMR deficiency was significantly associated with site, with tumors off of the head and neck more likely to be MMR deficient (specificity 96%). In contrast to prior reports, no significant trend in MMR-deficient versus -nondeficient tumors was seen in age at presentation (median age, 68 versus 66), tumor-infiltrating lymphocytes, or tumor type. Given the low sensitivity of age < 60 years (30%), location off of the head and neck (41%), or presence of tumor-infiltrating lymphocytes (29%) in MMR deficiency detection, IHC screening programs should test all sebaceous neoplasms for MMR deficiency, regardless of their clinicopathological features., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
45. Targeted therapies and predictive markers in epithelial malignancies of the gastrointestinal tract.
- Author
-
McIntire M and Redston M
- Subjects
- Antineoplastic Agents therapeutic use, Drug Resistance, Neoplasm genetics, ErbB Receptors antagonists & inhibitors, ErbB Receptors genetics, Gastrointestinal Neoplasms drug therapy, Humans, Neoplasms, Glandular and Epithelial drug therapy, Biomarkers, Tumor genetics, Gastrointestinal Neoplasms genetics, Molecular Targeted Therapy methods, Neoplasms, Glandular and Epithelial genetics
- Abstract
Context: In recent years, there has been a tremendous amount of interest in the development of targeted therapies for the treatment of human cancers. Increased understanding of the specific molecular pathways and driver mutations critical to cancer cell growth have allowed the development of these advanced therapeutics. Among these, inhibitors of the epidermal growth factor receptor and HER2/neu pathways now play a major role in the management of gastrointestinal cancers in addition to other solid malignancies. In colorectal and gastric cancers, the use of epidermal growth factor receptor inhibitors and HER2/neu inhibitors has increased the available treatment options for patients with advanced disease., Objective: To focus on the current targeted therapies and predictors of response in malignancies of the gastrointestinal tract., Data Sources: Medical literature searchable on PubMed (US National Library of Medicine) as well as older studies revealed by the literature review were used as the source of data., Conclusion: Gene testing of critical elements of the pathways targeted by these agents (such as KRAS mutational analysis in colorectal tumors and HER2/neu testing in gastric cancers) allows the ability to predict which patients will respond to these treatments. As the molecular profiling of tumors and our understanding of cancer genomics and epigenetic alterations continues to grow, it is expected that these personalized targeted therapies will form one of the mainstays of gastrointestinal cancer treatment.
- Published
- 2012
- Full Text
- View/download PDF
46. Immunohistochemical and molecular analysis of tyrosine kinase activity in desmoid tumors.
- Author
-
Cho NL, Carothers AM, Rizvi H, Hasson RM, Redston M, and Bertagnolli MM
- Subjects
- Adult, CSK Tyrosine-Protein Kinase, Cell Transformation, Neoplastic, Female, Humans, Immunohistochemistry, Male, Middle Aged, Phosphorylation, src-Family Kinases, Fibromatosis, Aggressive enzymology, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins c-kit metabolism, beta Catenin metabolism
- Abstract
Background: Optimal surgical and medical therapy for the treatment of desmoid tumors (DT) is still undefined. Partial response to tyrosine kinase inhibitors (TKI) has previously been described. Here, we examined the role of the tyrosine kinases c-Src and c-Kit in driving desmoid tumorigenesis., Methods: Six consecutive DT and matched normal tissues were collected from the operating room. Tissues were embedded in paraffin for immunohistochemical analysis, and protein lysates were prepared for immunoblot and immunoprecipitation., Results: We found increased levels of β-catenin in five of six (83%) DT relative to matched normal tissue by immunoblot analysis. By immunohistochemistry, β-catenin expression was also increased in DT and localized to the nucleus. In contrast, we observed variable levels of total and activated c-Src and c-Kit expression in DT compared with normal tissue. Finally, β-catenin tyrosine phosphorylation (p-Y) among tumors was variably increased, despite the increased amount of total β-catenin in tumors., Conclusions: Our results suggest that c-Src and c-Kit activity in DT is variable, consistent with the heterogeneous nature of this disease. Clinical response to TKI in DT may be via alternative mechanisms unrelated to c-Src or c-Kit activity. Further insight into DT biology will help identify novel drug regimens to limit the morbidity and mortality associated with this disease., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
47. Five-year efficacy and safety analysis of the Adenoma Prevention with Celecoxib Trial.
- Author
-
Bertagnolli MM, Eagle CJ, Zauber AG, Redston M, Breazna A, Kim K, Tang J, Rosenstein RB, Umar A, Bagheri D, Collins NT, Burn J, Chung DC, Dewar T, Foley TR, Hoffman N, Macrae F, Pruitt RE, Saltzman JR, Salzberg B, Sylwestrowicz T, and Hawk ET
- Subjects
- Adult, Aged, Aged, 80 and over, Celecoxib, Colonoscopy, Cyclooxygenase 2 Inhibitors administration & dosage, Dose-Response Relationship, Drug, Female, Follow-Up Studies, Humans, Male, Middle Aged, Neoplasm Recurrence, Local prevention & control, Pyrazoles administration & dosage, Sulfonamides administration & dosage, Adenoma prevention & control, Cardiovascular Diseases chemically induced, Colorectal Neoplasms prevention & control, Cyclooxygenase 2 Inhibitors adverse effects, Pyrazoles adverse effects, Sulfonamides adverse effects
- Abstract
The Adenoma Prevention with Celecoxib Trial examined the efficacy and safety of the cyclooxygenase (Cox)-2 inhibitor, celecoxib, for sporadic colorectal adenoma prevention in patients at high risk for colorectal cancer. The trial randomized 2,035 subjects to receive either placebo, celecoxib 200 mg twice daily, or celecoxib 400 mg twice daily. The primary study safety and efficacy analyses involved 3 years of treatment. The results showed significant antitumor effect but also indicated increased cardiovascular adverse events in patients treated with celecoxib compared with placebo. A total of 933 patients participated in an extension of the Adenoma Prevention with Celecoxib Trial, with a planned total treatment and surveillance duration of 5 years. Study medication was stopped early, resulting in a median treatment duration of 3.1 years for those with a year 5 colonoscopy. Patients treated on the placebo arm had a cumulative adenoma incidence of 68.4% over 5 years of observation. This figure was 59.0% (P < 0.0001) for those receiving low-dose celecoxib, and 60.1% (P < 0.0001) for those receiving high-dose celecoxib. The cumulative incidence of advanced adenomas over 5 years was 21.3% of those taking placebo, 12.5% (P < 0.0001) of those taking low dose celecoxib and 15.8% (P < 0.0001) of those taking high-dose celecoxib. Investigator reported treatment emergent adverse events were similar across all treatment groups for categories including renal and hypertensive events and gastrointestinal ulceration and hemorrhage events. For a category composed of cardiovascular and thrombotic events, the risk relative to placebo was 1.6 (95% confidence interval, 1.0, 2.5) for those using 200 mg twice daily celecoxib and 1.9 (95% confidence interval, 1.2, 3.1) for those using 400 mg twice daily celecoxib. Secondary analysis showed an interaction between a baseline history of atherosclerotic heart disease and study drug use with respect to cardiovascular and thrombotic adverse events (P = 0.004). These results confirm the inhibitory effect of celecoxib on colorectal adenoma formation, and provide additional safety data indicating an elevated risk for cardiovascular and thrombotic adverse events, particularly for patients with preexisting atherosclerotic heart disease.
- Published
- 2009
- Full Text
- View/download PDF
48. p27Kip1 in stage III colon cancer: implications for outcome following adjuvant chemotherapy in cancer and leukemia group B protocol 89803.
- Author
-
Bertagnolli MM, Warren RS, Niedzwiecki D, Mueller E, Compton CC, Redston M, Hall M, Hahn HP, Jewell SD, Mayer RJ, Goldberg RM, Saltz LB, and Loda M
- Subjects
- Adult, Aged, Aged, 80 and over, Chemotherapy, Adjuvant, Colonic Neoplasms chemistry, Colonic Neoplasms mortality, Colonic Neoplasms pathology, Cyclin-Dependent Kinase Inhibitor p27, DNA Mismatch Repair, Female, Humans, Intracellular Signaling Peptides and Proteins physiology, Male, Microsatellite Instability, Middle Aged, Neoplasm Staging, Prospective Studies, Treatment Outcome, Colonic Neoplasms drug therapy, Intracellular Signaling Peptides and Proteins analysis
- Abstract
Background: In retrospective studies, loss of p27(Kip1) (p27), a cyclin-dependent kinase inhibitor, has been associated with poor prognosis following colorectal cancer treatment. In a prospective study, we validated this relationship in patients enrolled on a trial of adjuvant chemotherapy for stage III colon cancer., Methods: Cancer and Leukemia Group B protocol 89803 randomized 1,264 stage III colon cancer patients to receive weekly bolus 5-fluorouracil/leucovorin or weekly bolus irinotecan, 5-fluorouracil, and leucovorin (IFL). The primary endpoint was overall survival (OS); disease-free survival was a secondary endpoint. Expression of p27 and DNA mismatch repair proteins were determined by immunohistochemistry in primary tumor and normal tissue from paraffin blocks. Data were analyzed using log-rank test., Results: Of 601 tumors analyzed, 207 (34.4%) showed p27 loss, 377 (62.8%) retained p27, and 17 (2.8%) were indeterminate. Patients with p27-negative tumors showed reduced OS [5-year OS 66%: 95% confidence interval (95% CI), 0.59-0.72 versus 75%: 95% CI, 0.70-0.79; log-rank P = 0.021]. This relationship was not influenced by treatment arm. Combination of p27 status with mismatch repair status, however, identified a small subset of patients that may benefit from IFL (n = 36; 5-year disease-free survival 81%: 95% CI, 0.64-0.98 versus 47%: 95% CI, 0.21-0.72; log-rank P = 0.042; 5-year OS 81%: 95% CI, 0.64-0.98 versus 60%: 95% CI, 0.35-0.85; log-rank P = 0.128)., Conclusions: Loss of p27 is associated with reduced survival in stage III colon cancer but by itself does not indicate a significant difference in outcome between patients treated IFL or 5-fluorouracil/leucovorin.
- Published
- 2009
- Full Text
- View/download PDF
49. Genomic alterations link Rho family of GTPases to the highly invasive phenotype of pancreas cancer.
- Author
-
Kimmelman AC, Hezel AF, Aguirre AJ, Zheng H, Paik JH, Ying H, Chu GC, Zhang JX, Sahin E, Yeo G, Ponugoti A, Nabioullin R, Deroo S, Yang S, Wang X, McGrath JP, Protopopova M, Ivanova E, Zhang J, Feng B, Tsao MS, Redston M, Protopopov A, Xiao Y, Futreal PA, Hahn WC, Klimstra DS, Chin L, and DePinho RA
- Subjects
- Animals, Carcinoma, Pancreatic Ductal pathology, Cell Line, Transformed, Cell Movement physiology, Gene Expression Regulation, Neoplastic, Genomics, Humans, Mice, Mice, Nude, Neoplasm Invasiveness, Pancreatic Ducts cytology, Pancreatic Neoplasms pathology, Phenotype, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-akt genetics, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction physiology, p21-Activated Kinases metabolism, rho GTP-Binding Proteins metabolism, Carcinoma, Pancreatic Ductal genetics, Pancreatic Ducts physiology, Pancreatic Neoplasms genetics, Protein Serine-Threonine Kinases genetics, p21-Activated Kinases genetics, rho GTP-Binding Proteins genetics
- Abstract
Pancreas ductal adenocarcinoma (PDAC) is a highly lethal cancer that typically presents as advanced, unresectable disease. This invasive tendency, coupled with intrinsic resistance to standard therapies and genome instability, are major contributors to poor long-term survival. The genetic elements governing the invasive propensity of PDAC have not been well elucidated. Here, in the course of validating resident genes in highly recurrent and focal amplifications in PDAC, we have identified Rio Kinase 3 (RIOK3) as an amplified gene that alters cytoskeletal architecture as well as promotes pancreatic ductal cell migration and invasion. We determined that RIOK3 promotes its invasive activities through activation of the small G protein, Rac. This genomic and functional link to Rac signaling prompted a genome wide survey of other components of the Rho family network, revealing p21 Activated Kinase 4 (PAK4) as another amplified gene in PDAC tumors and cell lines. Like RIOK3, PAK4 promotes pancreas ductal cell motility and invasion. Together, the genomic and functional profiles establish the Rho family GTP-binding proteins as integral to the hallmark invasive nature of this lethal disease.
- Published
- 2008
- Full Text
- View/download PDF
50. Missense polymorphisms in the adenomatous polyposis coli gene and colorectal cancer risk.
- Author
-
Cleary SP, Kim H, Croitoru ME, Redston M, Knight JA, Gallinger S, and Gryfe R
- Subjects
- Adenomatous Polyposis Coli epidemiology, Adult, Aged, Alleles, Analysis of Variance, Case-Control Studies, Colorectal Neoplasms epidemiology, DNA Mutational Analysis, Female, Humans, Incidence, Male, Middle Aged, Ontario epidemiology, Open Reading Frames, Polymerase Chain Reaction, Risk Factors, Statistics, Nonparametric, Surveys and Questionnaires, Adenomatous Polyposis Coli genetics, Colorectal Neoplasms genetics, Genes, APC, Mutation, Missense, Polymorphism, Genetic
- Abstract
Purpose: Whereas truncating germline mutations of the adenomatous polyposis coli (APC) gene give rise to familial adenomatous polyposis, missense polymorphisms of APC may confer a weaker risk for colorectal cancer., Methods: We sequenced the entire open reading frame of the APC gene and tested for two common MYH mutations in a population-based series of patients with colorectal cancer and 5 to 99 adenomas. Missense adenomatous polyposis coli alterations identified in this colorectal cancer multiple-polyp population were analyzed in a population-based series of patients with colorectal cancer and healthy control subjects., Results: Germline APC or mutY human homologue (MYH) alterations were identified in 16 of 39 colorectal cancer-multiple polyp patients. Four missense APC gene alterations (S130G, E1317Q, D1822V, G2502S) were observed in 13 individuals and 3 additional patients carried presumed pathogenic (APC Y94X, biallelic MYH Y165C and heterozygous MYH G382D) mutations. When independently assessed in 971 patients with colorectal cancer and 954 healthy control subjects, none of the identified missense APC alterations conferred a significantly increased risk for colorectal cancer, odds ratio (95 percent confidence intervals): S130G = 3.1 (0.29-32.25), E1317Q = 1.08 (0.59-2.74), G2502S = 1 (0.65-1.63), D1822V (heterozygous) = 0.79 (0.64-0.98), D1822V (homozygous) = 0.82 (0.63-1.27)., Conclusions: Germline missense APC alterations observed in 33 percent of patients with multiple colorectal neoplasms seemed to play a limited role in colorectal cancer risk when independently assessed by a population-based, case-control analysis.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.