80 results on '"Busam K"'
Search Results
2. A recurrent germline BAP1 mutation and extension of the BAP1 tumor predisposition spectrum to include basal cell carcinoma
- Author
-
Wadt, K. A.W., Aoude, L. G., Johansson, P., Solinas, A., Pritchard, A., Crainic, O., Andersen, M. T., Kiilgaard, J. F., Heegaard, S., Sunde, L., Federspiel, B., Madore, J., Thompson, J. F., McCarthy, S. W., Goodwin, A., Tsao, H., Jönsson, G., Busam, K., Gupta, R., Trent, J. M., Gerdes, A.-M., Brown, K. M., Scolyer, R. A., and Hayward, N. K.
- Published
- 2015
- Full Text
- View/download PDF
3. Comparison of community pathologists with expert dermatopathologists evaluating Breslow thickness and histopathologic subtype in a large international population-based study of melanoma.
- Author
-
Yardman-Frank, JM, Bronner, B, Rosso, S, From, L, Busam, K, Groben, P, Tucker, P, Cust, A, Armstrong, B, Kricker, A, Marrett, L, Anton-Culver, H, Gruber, S, Gallagher, R, Zanetti, R, Sacchetto, L, Dwyer, T, Venn, A, Orlow, I, Kanetsky, P, Luo, L, Thomas, N, Begg, C, Berwick, M, GEM Study Team, Yardman-Frank, JM, Bronner, B, Rosso, S, From, L, Busam, K, Groben, P, Tucker, P, Cust, A, Armstrong, B, Kricker, A, Marrett, L, Anton-Culver, H, Gruber, S, Gallagher, R, Zanetti, R, Sacchetto, L, Dwyer, T, Venn, A, Orlow, I, Kanetsky, P, Luo, L, Thomas, N, Begg, C, Berwick, M, and GEM Study Team
- Published
- 2021
4. Expert second-opinion pathology review of lymphoma in the era of the World Health Organization classification
- Author
-
Matasar, M. J., Shi, W., Silberstien, J., Lin, O., Busam, K. J., Teruya-Feldstein, J., Filippa, D. A., Zelenetz, A. D., and Noy, A.
- Published
- 2012
- Full Text
- View/download PDF
5. MC1R variants in childhood and adolescent melanoma: a retrospective pooled analysis of a multicentre cohort
- Author
-
Pellegrini, C. Botta, F. Massi, D. Martorelli, C. Facchetti, F. Gandini, S. Maisonneuve, P. Avril, M.-F. Demenais, F. Bressac-de Paillerets, B. Hoiom, V. Cust, A.E. Anton-Culver, H. Gruber, S.B. Gallagher, R.P. Marrett, L. Zanetti, R. Dwyer, T. Thomas, N.E. Begg, C.B. Berwick, M. Puig, S. Potrony, M. Nagore, E. Ghiorzo, P. Menin, C. Manganoni, A.M. Rodolfo, M. Brugnara, S. Passoni, E. Sekulovic, L.K. Baldini, F. Guida, G. Stratigos, A. Ozdemir, F. Ayala, F. Fernandez-de-Misa, R. Quaglino, P. Ribas, G. Romanini, A. Migliano, E. Stanganelli, I. Kanetsky, P.A. Pizzichetta, M.A. García-Borrón, J.C. Nan, H. Landi, M.T. Little, J. Newton-Bishop, J. Sera, F. Fargnoli, M.C. Raimondi, S. Alaibac, M. Ferrari, A. Valeri, B. Sicher, M. Mangiola, D. Nazzaro, G. Tosti, G. Mazzarol, G. Giudice, G. Ribero, S. Astrua, C. Mazzoni, L. Orlow, I. Mujumdar, U. Hummer, A. Busam, K. Roy, P. Canchola, R. Clas, B. Cotignola, J. Monroe, Y. Armstrong, B. Kricker, A. Litchfield, M. Tucker, P. Stephens, N. Switzer, T. Theis, B. From, L. Chowdhury, N. Vanasse, L. Purdue, M. Northrup, D. Rosso, S. Sacerdote, C. Leighton, N. Gildea, M. Bonner, J. Jeter, J. Klotz, J. Wilcox, H. Weiss, H. Millikan, R. Mattingly, D. Player, J. Tse, C.-K. Rebbeck, T. Walker, A. Panossian, S. Setlow, R. Mohrenweiser, H. Autier, P. Han, J. Caini, S. Hofman, A. Kayser, M. Liu, F. Nijsten, T. Uitterlinden, A.G. Kumar, R. Bishop, T. Elliott, F. Lazovich, D. Polsky, D. Hansson, J. Pastorino, L. Gruis, N.A. Bouwes Bavinck, J.N. Aguilera, P. Badenas, C. Carrera, C. Gimenez-Xavier, P. Malvehy, J. Puig-Butille, J.A. Tell-Marti, G. Blizzard, L. Cochrane, J. Branicki, W. Debniak, T. Morling, N. Johansen, P. Mayne, S. Bale, A. Cartmel, B. Ferrucci, L. Pfeiffer, R. Palmieri, G. Kypreou, K. Bowcock, A. Cornelius, L. Council, M.L. Motokawa, T. Anno, S. Helsing, P. Andresen, P.A. Guida, S. Wong, T.H. IMI Study Group GEM Study Group M-SKIP Study Group
- Abstract
Background: Germline variants in the melanocortin 1 receptor gene (MC1R) might increase the risk of childhood and adolescent melanoma, but a clear conclusion is challenging because of the low number of studies and cases. We assessed the association of MC1R variants with childhood and adolescent melanoma in a large study comparing the prevalence of MC1R variants in child or adolescent patients with melanoma to that in adult patients with melanoma and in healthy adult controls. Methods: In this retrospective pooled analysis, we used the M-SKIP Project, the Italian Melanoma Intergroup, and other European groups (with participants from Australia, Canada, France, Greece, Italy, the Netherlands, Serbia, Spain, Sweden, Turkey, and the USA) to assemble an international multicentre cohort. We gathered phenotypic and genetic data from children or adolescents diagnosed with sporadic single-primary cutaneous melanoma at age 20 years or younger, adult patients with sporadic single-primary cutaneous melanoma diagnosed at age 35 years or older, and healthy adult individuals as controls. We calculated odds ratios (ORs) for childhood and adolescent melanoma associated with MC1R variants by multivariable logistic regression. Subgroup analysis was done for children aged 18 or younger and 14 years or younger. Findings: We analysed data from 233 young patients, 932 adult patients, and 932 healthy adult controls. Children and adolescents had higher odds of carrying MC1R r variants than did adult patients (OR 1·54, 95% CI 1·02–2·33), including when analysis was restricted to patients aged 18 years or younger (1·80, 1·06–3·07). All investigated variants, except Arg160Trp, tended, to varying degrees, to have higher frequencies in young patients than in adult patients, with significantly higher frequencies found for Val60Leu (OR 1·60, 95% CI 1·05–2·44; p=0·04) and Asp294His (2·15, 1·05–4·40; p=0·04). Compared with those of healthy controls, young patients with melanoma had significantly higher frequencies of any MC1R variants. Interpretation: Our pooled analysis of MC1R genetic data of young patients with melanoma showed that MC1R r variants were more prevalent in childhood and adolescent melanoma than in adult melanoma, especially in patients aged 18 years or younger. Our findings support the role of MC1R in childhood and adolescent melanoma susceptibility, with a potential clinical relevance for developing early melanoma detection and preventive strategies. Funding: SPD-Pilot/Project-Award-2015; AIRC-MFAG-11831. © 2019 Elsevier Ltd
- Published
- 2019
6. Confocal reflectance imaging of pigmented skin lesions in vivo: initial steps towards a terminology consensus: 885
- Author
-
Benvenuto-Andrade, C, Agero, A L, Dusza, S, Rajadhyaksha, M, Busam, K, Marra, D, Torres, A, Propperova, I, Langley, R, Marghoob, A, Gonzalez, S, and Halpern, A C
- Published
- 2005
7. Melanoma in younger individuals is more likely to contain a BRAF than NRAS mutation: 856
- Author
-
Thomas, N E, Alexander, A, Edmiston, S N, Millikan, R C, Groben, P, Berwick, M, Busam, K, Mattingly, D, Hao, H, Ollila, D, and Conway, K
- Published
- 2005
8. The interaction between vitamin D receptor polymorphisms and sun exposure around time of diagnosis influences melanoma survival.
- Author
-
Orlow, I, Shi, Y, Kanetsky, PA, Thomas, NE, Luo, L, Corrales-Guerrero, S, Cust, AE, Sacchetto, L, Zanetti, R, Rosso, S, Armstrong, BK, Dwyer, T, Venn, A, Gallagher, RP, Gruber, SB, Marrett, LD, Anton-Culver, H, Busam, K, Begg, CB, Berwick, M, GEM Study Group, Orlow, I, Shi, Y, Kanetsky, PA, Thomas, NE, Luo, L, Corrales-Guerrero, S, Cust, AE, Sacchetto, L, Zanetti, R, Rosso, S, Armstrong, BK, Dwyer, T, Venn, A, Gallagher, RP, Gruber, SB, Marrett, LD, Anton-Culver, H, Busam, K, Begg, CB, Berwick, M, and GEM Study Group
- Abstract
Evidence on the relationship between the vitamin D pathway and outcomes in melanoma is growing, although it is not always clear. We investigated the impact of measured levels of sun exposure at diagnosis on associations of vitamin D receptor gene (VDR) polymorphisms and melanoma death in 3336 incident primary melanoma cases. Interactions between six SNPs and a common 3'-end haplotype were significant (p < .05). These SNPs, and a haplotype, had a statistically significant association with survival among subjects exposed to high UVB in multivariable regression models and exerted their effect in the opposite direction among those with low UVB. SNPs rs1544410/BsmI and rs731236/TaqI remained significant after adjustment for multiple testing. These results suggest that the association between VDR and melanoma-specific survival is modified by sun exposure around diagnosis, and require validation in an independent study. Whether the observed effects are dependent or independent of vitamin D activation remains to be determined.
- Published
- 2018
9. Limited variation during circulation of a polyomavirus in the human population involves the COCO-VA toggling site of Middle and Alternative T-antigen(s)
- Author
-
Kazem, S. (Siamaque), Lauber, C. (Chris), van der Meijden, E. (Els), Kooijman, S. (Sander), Kravchenko, A.A. (Alexander A.), Feltkamp, M.C.W. (Mariet C.W.), Gorbalenya, A.E. (Alexander), Browning, J.C. (John C.), Busam, K. (Klaus), Bialasiewicz, S. (Seweryn), Benoit, T. (Taylor), Fleckman, P. (Philip), Hughey, L.C. (Lauren C.), Janssens, R.W.A. (René W.A.), Mechinaud, F. (Francoise), Pope, E. (Elena), Rosenberg, A.S. (Arlene S.), Rácz, E. (Emoke), Sadler, G. (Genevieve), Tabrizi, S.N. (Sepehr N.), Vries, E. (E.) de, Wang, R.C. (Richard C.), Kazem, S. (Siamaque), Lauber, C. (Chris), van der Meijden, E. (Els), Kooijman, S. (Sander), Kravchenko, A.A. (Alexander A.), Feltkamp, M.C.W. (Mariet C.W.), Gorbalenya, A.E. (Alexander), Browning, J.C. (John C.), Busam, K. (Klaus), Bialasiewicz, S. (Seweryn), Benoit, T. (Taylor), Fleckman, P. (Philip), Hughey, L.C. (Lauren C.), Janssens, R.W.A. (René W.A.), Mechinaud, F. (Francoise), Pope, E. (Elena), Rosenberg, A.S. (Arlene S.), Rácz, E. (Emoke), Sadler, G. (Genevieve), Tabrizi, S.N. (Sepehr N.), Vries, E. (E.) de, and Wang, R.C. (Richard C.)
- Abstract
We have recently shown that the trichodysplasia spinulosa-associated polyomavirus (TSPyV) belongs to a large monophyletic group of mammalian polyomaviruses that experienced accelerated codon-constrained Val-Ala (COCO-VA) toggling at a protein site common to both Middle and Alternative T-antigens (MT/ALTO). Here we analyzed thirteen, mostly newly sequenced TSPyV genomes, representing ~40% of reported TS disease cases world-wide. We found two deletions and 30 variable sites (≤0.6%) that included only four sites with non-synonymous substitutions (NSS). One NSS site was under positive selection in the exon shared by Small and Middle T antigens, while three others were segregated in MT/ALTO. Two MT/ALTO sites covaried with five sites elsewhere in the genome and determined separation of twelve TSPyVs into two most populous phylogenetic lineages. The other, most distant TSPyV was distinguished by NSS at the COCO-VA site, observed for the first time during intra-species evolution. Our findings reveal a connection between micro- and macro-evolution of polyomaviruses.
- Published
- 2016
- Full Text
- View/download PDF
10. A recurrent germline BAP1 mutation and extension of the BAP1 tumor predisposition spectrum to include basal cell carcinoma
- Author
-
Wadt, K A W, Aoude, L G, Johansson, P, Solinas, A, Pritchard, A, Crainic, O, Andersen, M T, Kiilgaard, J F, Heegaard, S, Sunde, L, Federspiel, B, Madore, J, Thompson, J F, McCarthy, S W, Goodwin, A, Tsao, H, Jönsson, G, Busam, K, Gupta, R, Trent, J M, Gerdes, A-M, Brown, K M, Scolyer, R A, and Hayward, N K
- Subjects
Male ,Heterozygote ,Tumor Suppressor Proteins ,DNA Mutational Analysis ,Loss of Heterozygosity ,Polymorphism, Single Nucleotide ,Pedigree ,Haplotypes ,Carcinoma, Basal Cell ,Humans ,Female ,Genetic Predisposition to Disease ,Ubiquitin Thiolesterase ,Germ-Line Mutation - Abstract
We report four previously undescribed families with germline BRCA1-associated protein-1 gene (BAP1) mutations and expand the clinical phenotype of this tumor syndrome. The tumor spectrum in these families is predominantly uveal malignant melanoma (UMM), cutaneous malignant melanoma (CMM) and mesothelioma, as previously reported for germline BAP1 mutations. However, mutation carriers from three new families, and one previously reported family, developed basal cell carcinoma (BCC), thus suggesting inclusion of BCC in the phenotypic spectrum of the BAP1 tumor syndrome. This notion is supported by the finding of loss of BAP1 protein expression by immunochemistry in two BCCs from individuals with germline BAP1 mutations and no loss of BAP1 staining in 53 of sporadic BCCs consistent with somatic mutations and loss of heterozygosity of the gene in the BCCs occurring in mutation carriers. Lastly, we identify the first reported recurrent mutation in BAP1 (p.R60X), which occurred in three families from two different continents. In two of the families, the mutation was inherited from a common founder but it arose independently in the third family.
- Published
- 2014
11. A recurrent germline BAP1 mutation and extension of the BAP1 tumor predisposition spectrum to include basal cell carcinoma
- Author
-
Wadt, Karin Anna Wallentin, Aoude, L G, Johansson, P, Solinas, A, Pritchard, A, Crainic, O, Andersen, M.T., Kiilgaard, Jens Folke, Heegaard, S, Sunde, L, Federspiel, Birgitte, Madore, J, Thompson, J F, McCarthy, S W, Goodwin, A, Tsao, H, Jönsson, G, Busam, K, Gupta, R, Trent, J M, Gerdes, A-M, Brown, K M, Scolyer, R A, Hayward, N K, Wadt, Karin Anna Wallentin, Aoude, L G, Johansson, P, Solinas, A, Pritchard, A, Crainic, O, Andersen, M.T., Kiilgaard, Jens Folke, Heegaard, S, Sunde, L, Federspiel, Birgitte, Madore, J, Thompson, J F, McCarthy, S W, Goodwin, A, Tsao, H, Jönsson, G, Busam, K, Gupta, R, Trent, J M, Gerdes, A-M, Brown, K M, Scolyer, R A, and Hayward, N K
- Abstract
We report four previously undescribed families with germline BRCA1-associated protein-1 gene (BAP1) mutations and expand the clinical phenotype of this tumor syndrome. The tumor spectrum in these families is predominantly uveal malignant melanoma (UMM), cutaneous malignant melanoma (CMM) and mesothelioma, as previously reported for germline BAP1 mutations. However, mutation carriers from three new families, and one previously reported family, developed basal cell carcinoma (BCC), thus suggesting inclusion of BCC in the phenotypic spectrum of the BAP1 tumor syndrome. This notion is supported by the finding of loss of BAP1 protein expression by immunochemistry in two BCCs from individuals with germline BAP1 mutations and no loss of BAP1 staining in 53 of sporadic BCCs consistent with somatic mutations and loss of heterozygosity of the gene in the BCCs occurring in mutation carriers. Lastly, we identify the first reported recurrent mutation in BAP1 (p.R60X), which occurred in three families from two different continents. In two of the families, the mutation was inherited from a common founder but it arose independently in the third family.
- Published
- 2015
12. A recurrent germlineBAP1mutation and extension of theBAP1tumor predisposition spectrum to include basal cell carcinoma
- Author
-
Wadt, K.A.W., primary, Aoude, L.G., additional, Johansson, P., additional, Solinas, A., additional, Pritchard, A., additional, Crainic, O., additional, Andersen, M.T., additional, Kiilgaard, J.F., additional, Heegaard, S., additional, Sunde, L., additional, Federspiel, B., additional, Madore, J., additional, Thompson, J.F., additional, McCarthy, S.W., additional, Goodwin, A., additional, Tsao, H., additional, Jönsson, G., additional, Busam, K., additional, Gupta, R., additional, Trent, J.M., additional, Gerdes, A.-M., additional, Brown, K.M., additional, Scolyer, R.A., additional, and Hayward, N.K., additional
- Published
- 2014
- Full Text
- View/download PDF
13. Determinants of BRAF Mutations in Primary Melanomas
- Author
-
Maldonado, J. L., primary, Fridlyand, J., additional, Patel, H., additional, Jain, A. N., additional, Busam, K., additional, Kageshita, T., additional, Ono, T., additional, Albertson, D. G., additional, Pinkel, D., additional, and Bastian, B. C., additional
- Published
- 2003
- Full Text
- View/download PDF
14. Monoclonal antibody MA454 reveals a heterogeneous expression pattern of MAGE-1 antigen in formalin-fixed paraffin embedded lung tumours
- Author
-
Jungbluth, A A, primary, Stockert, E, additional, Chen, Y-T, additional, Kolb, D, additional, Iversen, K, additional, Coplan, K, additional, Williamson, B, additional, Altorki, N, additional, Busam, K J, additional, and Old, L J, additional
- Published
- 2000
- Full Text
- View/download PDF
15. Staphylococcus aureus and derived exotoxins induce nuclear factor kappa B-like activity in murine bone marrow macrophages
- Author
-
Busam, K, primary, Gieringer, C, additional, Freudenberg, M, additional, and Hohmann, H P, additional
- Published
- 1992
- Full Text
- View/download PDF
16. Regulation of transforming growth factor beta 1 gene expression by the product of the retinoblastoma-susceptibility gene.
- Author
-
Kim, S J, primary, Lee, H D, additional, Robbins, P D, additional, Busam, K, additional, Sporn, M B, additional, and Roberts, A B, additional
- Published
- 1991
- Full Text
- View/download PDF
17. Frequent p16-independent inactivation of p14ARF in human melanoma.
- Author
-
Freedberg DE, Rigas SH, Russak J, Gai W, Kaplow M, Osman I, Turner F, Randerson-Moor JA, Houghton A, Busam K, Timothy Bishop D, Bastian BC, Newton-Bishop JA, Polsky D, Freedberg, Daniel E, Rigas, Sushila H, Russak, Julie, Gai, Weiming, Kaplow, Margarita, and Osman, Iman
- Abstract
Background: The tumor suppressors p14(ARF) (ARF) and p16(INK4A) (p16) are encoded by overlapping reading frames at the CDKN2A/INK4A locus on chromosome 9p21. In human melanoma, the accumulated evidence has suggested that the predominant tumor suppressor at 9p21 is p16, not ARF. However, recent observations from melanoma-prone families and murine melanoma models suggest a p16-independent tumor suppressor role for ARF. We analyzed a group of melanoma metastases and cell lines to investigate directly whether somatic alterations to the ARF gene support its role as a p16-independent tumor suppressor in human melanoma, assuming that two alterations (genetic and/or epigenetic) would be required to inactivate a gene.Methods: We examined the p16/ARF locus in 60 melanoma metastases from 58 patients and in 9 human melanoma cell lines using multiplex ligation-dependent probe amplification and multiplex polymerase chain reaction (PCR) to detect deletions, methylation-specific PCR to detect promoter methylation, direct sequencing to detect mutations affecting ARF and p16, and, in a subset of 20 tumors, immunohistochemistry to determine the effect of these alterations on p16 protein expression. All statistical tests were two-sided.Results: We observed two or more alterations to the ARF gene in 26/60 (43%) metastases. The p16 gene sustained two or more alterations in 13/60 (22%) metastases (P = .03). Inactivation of ARF in the presence of wild-type p16 was seen in 18/60 (30%) metastases.Conclusion: Genetic and epigenetic analyses of the human 9p21 locus indicate that modifications of ARF occur independently of p16 inactivation in human melanoma and suggest that ARF is more frequently inactivated than p16. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
18. Matrix Metalloproteinase-9 (MMP-9) polymorphisms in patients with cutaneous malignant melanoma
- Author
-
Busam Klaus, Coit Daniel, Vanderbeek Gretchen, Shah Shivang, Patel Ami, Chuai Shaokun, Ishill Nicole, Mitra Nandita, Reva Boris, Cotignola Javier, Halpern Allan, Houghton Alan, Sander Chris, Berwick Marianne, and Orlow Irene
- Subjects
Internal medicine ,RC31-1245 ,Genetics ,QH426-470 - Abstract
Abstract Background Cutaneous Malignant Melanoma causes over 75% of skin cancer-related deaths, and it is clear that many factors may contribute to the outcome. Matrix Metalloproteinases (MMPs) play an important role in the degradation and remodeling of the extracellular matrix and basement membrane that, in turn, modulate cell division, migration and angiogenesis. Some polymorphisms are known to influence gene expression, protein activity, stability, and interactions, and they were shown to be associated with certain tumor phenotypes and cancer risk. Methods We tested seven polymorphisms within the MMP-9 gene in 1002 patients with melanoma in order to evaluate germline genetic variants and their association with progression and known risk factors of melanoma. The polymorphisms were selected based on previously published reports and their known or potential functional relevance using in-silico methods. Germline DNA was then genotyped using pyrosequencing, melting temperature profiles, heteroduplex analysis, and fragment size analysis. Results We found that reference alleles were present in higher frequency in patients who tend to sunburn, have family history of melanoma, higher melanoma stage, intransit metastasis and desmoplastic melanomas among others. However, after adjustment for age, sex, phenotypic index, moles, and freckles only Q279R, P574R and R668Q had significant associations with intransit metastasis, propensity to tan/sunburn and primary melanoma site. Conclusion This study does not provide strong evidence for further investigation into the role of the MMP-9 SNPs in melanoma progression.
- Published
- 2007
- Full Text
- View/download PDF
19. Clinical significance of BRAF mutations in metastatic melanoma
- Author
-
Polsky David, Osman Iman, Panageas Katherine S, Chang David Z, Busam Klaus, and Chapman Paul B
- Subjects
Metastatic melanoma ,BRAF mutation ,Clinical significance ,Medicine - Abstract
Abstract Forty to eighty percent of melanoma tumors have activating mutations in BRAF although the clinical importance of these mutations is not clear. We previously reported an analysis of BRAF mutations in metastatic melanoma samples from 68 patients. In this study, we correlated patient baseline characteristics, prognostic factors, and/or clinical outcomes with the presence of BRAF mutations. No significant differences were observed in age, gender, location of primary melanoma, stage at the diagnosis, and depth of primary tumor between patients with and without BRAF mutations. Melanomas harboring BRAF mutations were more likely to metastasize to liver (P = 0.02) and to metastasize to multiple organs (P = 0.048). Neither time to progression to stage IV nor overall survival were associated with BRAF mutations. In conclusion, we observed no significant differences in clinical characteristics or outcomes between melanomas with or without BRAF mutations. Although there was an increased frequency of liver metastasis and tendency to metastasize to multiple organs in tumors with BRAF mutations, there was no detectable effect on survival. Future prospective studies should include analysis of whether BRAF mutations in melanoma tumors correlate with an increased tendency to metastasize to liver or to multiple organs.
- Published
- 2004
- Full Text
- View/download PDF
20. Regulation of the transforming growth factor- 1 and - 3 promoters by transcription factor Sp1
- Author
-
Geiser, A. G., Busam, K. J., Kim, S.-J., and Lafyatis, R.
- Published
- 1993
- Full Text
- View/download PDF
21. MC1R variants in childhood and adolescent melanoma: a retrospective pooled analysis of a multicentre cohort
- Author
-
Pellegrini, Cristina, Botta, Francesca, Massi, Daniela, Martorelli, Claudia, Facchetti, Fabio, Gandini, Sara, Maisonneuve, Patrick, Avril, Marie-Françoise, Demenais, Florence, Bressac-de Paillerets, Brigitte, Hoiom, Veronica, Cust, Anne E, Anton-Culver, Hoda, Gruber, Stephen B, Gallagher, Richard P, Marrett, Loraine, Zanetti, Roberto, Dwyer, Terence, Thomas, Nancy E, Begg, Colin B, Berwick, Marianne, Puig, Susana, Potrony, Miriam, Nagore, Eduardo, Ghiorzo, Paola, Menin, Chiara, Manganoni, Ausilia Maria, Rodolfo, Monica, Brugnara, Sonia, Passoni, Emanuela, Sekulovic, Lidija Kandolf, Baldini, Federica, Guida, Gabriella, Stratigos, Alexandros, Ozdemir, Fezal, Ayala, Fabrizio, Fernandez-de-Misa, Ricardo, Quaglino, Pietro, Ribas, Gloria, Romanini, Antonella, Migliano, Emilia, Stanganelli, Ignazio, Kanetsky, Peter A, Pizzichetta, Maria Antonietta, García-Borrón, Jose Carlos, Nan, Hongmei, Landi, Maria Teresa, Little, Julian, Newton-Bishop, Julia, Sera, Francesco, Fargnoli, Maria Concetta, Raimondi, Sara, Alaibac, Mauro, Ferrari, Andrea, Valeri, Barbara, Sicher, Mariacristina, Mangiola, Daniela, Nazzaro, Gianluca, Tosti, Giulio, Mazzarol, Giovanni, Giudice, Giuseppe, Ribero, Simone, Astrua, Chiara, Mazzoni, Laura, Orlow, Irene, Mujumdar, Urvi, Hummer, Amanda, Busam, Klaus, Roy, Pampa, Canchola, Rebecca, Clas, Brian, Cotignola, Javiar, Monroe, Yvette, Armstrong, Bruce, Kricker, Anne, Litchfield, Melisa, Tucker, Paul, Stephens, Nicola, Switzer, Teresa, Theis, Beth, From, Lynn, Chowdhury, Noori, Vanasse, Louise, Purdue, Mark, Northrup, David, Rosso, Stefano, Sacerdote, Carlotta, Leighton, Nancy, Gildea, Maureen, Bonner, Joe, Jeter, Joanne, Klotz, Judith, Wilcox, Homer, Weiss, Helen, Millikan, Robert, Mattingly, Dianne, Player, Jon, Tse, Chiu-Kit, Rebbeck, Timothy, Walker, Amy, Panossian, Saarene, Setlow, Richard, Mohrenweiser, Harvey, Autier, Philippe, Han, Jiali, Caini, Saverio, Hofman, Albert, Kayser, Manfred, Liu, Fan, Nijsten, Tamar, Uitterlinden, Andre G., Kumar, Rajiv, Bishop, Tim, Elliott, Faye, Lazovich, Deann, Polsky, David, Hansson, Johan, Pastorino, Lorenza, Gruis, Nelleke A., Bouwes Bavinck, Jan Nico, Aguilera, Paula, Badenas, Celia, Carrera, Cristina, Gimenez-Xavier, Pol, Malvehy, Josep, Puig-Butille, Joan Anton, Tell-Marti, Gemma, Blizzard, Leigh, Cochrane, Jennifer, Branicki, Wojciech, Debniak, Tadeusz, Morling, Niels, Johansen, Peter, Mayne, Susan, Bale, Allen, Cartmel, Brenda, Ferrucci, Leah, Pfeiffer, Ruth, Palmieri, Giuseppe, Kypreou, Katerina, Bowcock, Anne, Cornelius, Lynn, Council, M. Laurin, Motokawa, Tomonori, Anno, Sumiko, Helsing, Per, Andresen, Per Arne, Guida, Stefania, Wong, Collaborators (98): Alaibac M, Terence H., Ferrari, A, Valeri, B, Et, Al., Pellegrini, C., Botta, F., Massi, D., Martorelli, C., Facchetti, F., Gandini, S., Maisonneuve, P., Avril, M. -F., Demenais, F., Bressac-de Paillerets, B., Hoiom, V., Cust, A. E., Anton-Culver, H., Gruber, S. B., Gallagher, R. P., Marrett, L., Zanetti, R., Dwyer, T., Thomas, N. E., Begg, C. B., Berwick, M., Puig, S., Potrony, M., Nagore, E., Ghiorzo, P., Menin, C., Manganoni, A. M., Rodolfo, M., Brugnara, S., Passoni, E., Sekulovic, L. K., Baldini, F., Guida, G., Stratigos, A., Ozdemir, F., Ayala, F., Fernandez-de-Misa, R., Quaglino, P., Ribas, G., Romanini, A., Migliano, E., Stanganelli, I., Kanetsky, P. A., Pizzichetta, M. A., Garcia-Borron, J. C., Nan, H., Landi, M. T., Little, J., Newton-Bishop, J., Sera, F., Fargnoli, M. C., Raimondi, S., Alaibac, M., Ferrari, A., Valeri, B., Sicher, M., Mangiola, D., Nazzaro, G., Tosti, G., Mazzarol, G., Giudice, G., Ribero, S., Astrua, C., Mazzoni, L., Orlow, I., Mujumdar, U., Hummer, A., Busam, K., Roy, P., Canchola, R., Clas, B., Cotignola, J., Monroe, Y., Armstrong, B., Kricker, A., Litchfield, M., Tucker, P., Stephens, N., Switzer, T., Theis, B., From, L., Chowdhury, N., Vanasse, L., Purdue, M., Northrup, D., Rosso, S., Sacerdote, C., Leighton, N., Gildea, M., Bonner, J., Jeter, J., Klotz, J., Wilcox, H., Weiss, H., Millikan, R., Mattingly, D., Player, J., Tse, C. -K., Rebbeck, T., Walker, A., Panossian, S., Setlow, R., Mohrenweiser, H., Autier, P., Han, J., Caini, S., Hofman, A., Kayser, M., Liu, F., Nijsten, T., Uitterlinden, A. G., Kumar, R., Bishop, T., Elliott, F., Lazovich, D., Polsky, D., Hansson, J., Pastorino, L., Gruis, N. A., Bouwes Bavinck, J. N., Aguilera, P., Badenas, C., Carrera, C., Gimenez-Xavier, P., Malvehy, J., Puig-Butille, J. A., Tell-Marti, G., Blizzard, L., Cochrane, J., Branicki, W., Debniak, T., Morling, N., Johansen, P., Mayne, S., Bale, A., Cartmel, B., Ferrucci, L., Pfeiffer, R., Palmieri, G., Kypreou, K., Bowcock, A., Cornelius, L., Council, M. L., Motokawa, T., Anno, S., Helsing, P., Andresen, P. A., Guida, S., Wong, T. H., Ege Üniversitesi, Epidemiology, Genetic Identification, and Dermatology
- Subjects
Male ,Skin Neoplasms ,Pediatrics ,Cohort Studies ,0302 clinical medicine ,Odds Ratio ,Developmental and Educational Psychology ,Pediatrics, Perinatology and Child Health ,030212 general & internal medicine ,Child ,Cancer ,Pediatric ,Tumor ,childhood disease ,Middle Aged ,Perinatology and Child Health ,cohort analysis ,Meta-analysis ,Melanocortin ,Cohort ,Female ,MC1R gene ,Receptor, Melanocortin, Type 1 ,Receptor ,Type 1 ,Cohort study ,Adult ,medicine.medical_specialty ,adolescent ,melanoma ,cohort analysi ,Subgroup analysis ,Article ,03 medical and health sciences ,Genetic ,Clinical Research ,030225 pediatrics ,Internal medicine ,Genetics ,Biomarkers, Tumor ,medicine ,Humans ,Genetic Predisposition to Disease ,Polymorphism ,Germ-Line Mutation ,Aged ,Retrospective Studies ,Polymorphism, Genetic ,business.industry ,Prevention ,Case-control study ,Retrospective cohort study ,GEM Study Group ,Odds ratio ,Logistic Models ,M-SKIP Study Group ,Case-Control Studies ,Cutaneous melanoma ,IMI Study Group ,business ,Biomarkers - Abstract
Ferrari, Andrea/0000-0002-4724-0517; Pellegrini, Cristina/0000-0003-2168-8097; Migliano, Emilia/0000-0002-5316-8937; Maisonneuve, Patrick/0000-0002-5309-4704; Guida, Stefania/0000-0002-8221-6694; Pastorino, Lorenza/0000-0002-2575-8331; CARRERA, CRISTINA/0000-0003-1608-8820; Paillerets, Brigitte Bressac-de/0000-0003-0245-8608; Sekulovic, Lidija Kandolf/0000-0002-5221-5068; Caini, Saverio/0000-0002-2262-1102; Potrony, Miriam/0000-0003-2766-0765; Pizzichetta, Maria Antonietta/0000-0002-4201-8490; Little, Julian/0000-0001-5026-5531; Nagore, Eduardo/0000-0003-3433-8707; Polsky, David/0000-0001-9554-5289; Demenais, Florence/0000-0001-8361-0936; Nazzaro, Gianluca/0000-0001-8534-6497; gandini, sara/0000-0002-1348-4548; Cornelius, Lynn A/0000-0002-6329-2819; Palmieri, Giuseppe/0000-0002-4350-2276; Cotignola, Javier/0000-0003-4473-9854; Ghiorzo, Paola/0000-0002-3651-8173; Autier, Philippe/0000-0003-1538-5321; Bishop, Tim/0000-0002-8752-8785; Sera, Francesco/0000-0002-8890-6848; Newton-Bishop, Julia/0000-0001-9147-6802; Litchfield, Melisa/0000-0003-0002-7724, WOS: 000464254100018, PubMed: 30872112, Background Germline variants in the melanocortin 1 receptor gene (MC1R) might increase the risk of childhood and adolescent melanoma, but a clear conclusion is challenging because of the low number of studies and cases. We assessed the association of MC1R variants with childhood and adolescent melanoma in a large study comparing the prevalence of MC1R variants in child or adolescent patients with melanoma to that in adult patients with melanoma and in healthy adult controls. Methods in this retrospective pooled analysis, we used the M-SKIP Project, the Italian Melanoma Intergroup, and other European groups (with participants from Australia, Canada, France, Greece, Italy, the Netherlands, Serbia, Spain, Sweden, Turkey, and the USA) to assemble an international multicentre cohort. We gathered phenotypic and genetic data from children or adolescents diagnosed with sporadic single-primary cutaneous melanoma at age 20 years or younger, adult patients with sporadic single-primary cutaneous melanoma diagnosed at age 35 years or older, and healthy adult individuals as controls. We calculated odds ratios (ORs) for childhood and adolescent melanoma associated with MC1R variants by multivariable logistic regression. Subgroup analysis was done for children aged 18 or younger and 14 years or younger. Findings We analysed data from 233 young patients, 932 adult patients, and 932 healthy adult controls. Children and adolescents had higher odds of carrying MC1R r variants than did adult patients (OR 1.54, 95% CI 1.02-2.33), including when analysis was restricted to patients aged 18 years or younger (1.80, 1.06-3.07). All investigated variants, except Arg160Trp, tended, to varying degrees, to have higher frequencies in young patients than in adult patients, with significantly higher frequencies found for Val60Leu (OR 1.60, 95% CI 1.05-2.44; p=0.04) and Asp294His (2.15, 1.05-4.40; p=0.04). Compared with those of healthy controls, young patients with melanoma had significantly higher frequencies of any MC1R variants. Interpretation Our pooled analysis of MC1R genetic data of young patients with melanoma showed that MC1R r variants were more prevalent in childhood and adolescent melanoma than in adult melanoma, especially in patients aged 18 years or younger. Our findings support the role of MC1R in childhood and adolescent melanoma susceptibility, with a potential clinical relevance for developing early melanoma detection and preventive strategies. Copyright (c) 2019 Elsevier Ltd. All rights reserved., [AIRC-MFAG-11831]; NATIONAL CANCER INSTITUTEUnited States Department of Health & Human ServicesNational Institutes of Health (NIH) - USANIH National Cancer Institute (NCI) [P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P01CA206980, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P01CA206980, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P01CA206980, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P01CA206980, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P01CA206980, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P30CA016086, P01CA206980, P30CA016086, P30CA016086] Funding Source: NIH RePORTER, SPD-Pilot/Project-Award-2015; AIRC-MFAG-11831.
- Published
- 2019
22. Improving Accuracy of Somatic Mutation Profiling in Large Epidemiologic Studies: Addressing Cases without Matched Normal Samples.
- Author
-
Arora A, Luo L, Kostrzewa CE, Reiner A, Seshan V, Ernstoff MS, Edmiston SN, Conway K, Gorlov I, Busam K, Orlow I, Hernando E, Thomas NE, Berwick M, Begg CB, and Shen R
- Abstract
Ideally, detection of somatic mutations in a tumor is accomplished using a patient-matched sample of normal cells as the benchmark. In this way somatic mutations can be distinguished from rare germline mutations. In large retrospective studies, archival tissue collection can pose challenges in obtaining samples of normal DNA. In this article we propose a protocol that improves somatic mutation analysis in the absence of a matched normal sample. The method was motivated by the InterMEL study, a large-scale epidemiologic investigation involving multiomic, multi-institutional genomic profiling of 1000 primary melanoma samples. The key insight for accomplishing improved mutation calling is the fact that germline mutations should produce a variant allele frequency (VAF) of around 50%. While a similar VAF of 50% would also be expected for somatic mutations in pure tumor samples, typically the tumor purity is much less than 50%, resulting in a considerably lower VAF. Making use of a technique that can simultaneously estimate both tumor purity and VAF from tumor-only samples we have developed a method for better distinguishing somatic versus germline variants. Based on 137 melanomas from the InterMEL Study with matched normal tissue to provide a gold standard we show that the conventional pipeline using a panel of (unmatched) normal samples has a false positive rate of 15.6% and a false negative rate of 3.5%. Our new technique improves these error rates to 6.4% and 2.1%, respectively.
- Published
- 2024
- Full Text
- View/download PDF
23. Author Correction: Neoadjuvant relatlimab and nivolumab in resectable melanoma.
- Author
-
Amaria RN, Postow M, Burton EM, Tetzlaff MT, Ross MI, Torres-Cabala C, Glitza IC, Duan F, Milton DR, Busam K, Simpson L, McQuade JL, Wong MK, Gershenwald JE, Lee JE, Goepfert RP, Keung EZ, Fisher SB, Betof-Warner A, Shoushtari AN, Callahan M, Coit D, Bartlett EK, Bello D, Momtaz P, Nicholas C, Gu A, Zhang X, Korivi BR, Patnana M, Patel SP, Diab A, Lucci A, Prieto VG, Davies MA, Allison JP, Sharma P, Wargo JA, Ariyan C, and Tawbi HA
- Published
- 2023
- Full Text
- View/download PDF
24. Neoadjuvant relatlimab and nivolumab in resectable melanoma.
- Author
-
Amaria RN, Postow M, Burton EM, Tetzlaff MT, Ross MI, Torres-Cabala C, Glitza IC, Duan F, Milton DR, Busam K, Simpson L, McQuade JL, Wong MK, Gershenwald JE, Lee JE, Goepfert RP, Keung EZ, Fisher SB, Betof-Warner A, Shoushtari AN, Callahan M, Coit D, Bartlett EK, Bello D, Momtaz P, Nicholas C, Gu A, Zhang X, Korivi BR, Patnana M, Patel SP, Diab A, Lucci A, Prieto VG, Davies MA, Allison JP, Sharma P, Wargo JA, Ariyan C, and Tawbi HA
- Subjects
- Humans, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal therapeutic use, Immune Checkpoint Inhibitors adverse effects, Immune Checkpoint Inhibitors therapeutic use, Neoplasm Staging, Macrophages drug effects, Drug Therapy, Combination, Survival Rate, Melanoma drug therapy, Melanoma pathology, Melanoma surgery, Neoadjuvant Therapy adverse effects, Neoadjuvant Therapy methods, Nivolumab adverse effects, Nivolumab therapeutic use
- Abstract
Relatlimab and nivolumab combination immunotherapy improves progression-free survival over nivolumab monotherapy in patients with unresectable advanced melanoma
1 . We investigated this regimen in patients with resectable clinical stage III or oligometastatic stage IV melanoma (NCT02519322). Patients received two neoadjuvant doses (nivolumab 480 mg and relatlimab 160 mg intravenously every 4 weeks) followed by surgery, and then ten doses of adjuvant combination therapy. The primary end point was pathologic complete response (pCR) rate2 . The combination resulted in 57% pCR rate and 70% overall pathologic response rate among 30 patients treated. The radiographic response rate using Response Evaluation Criteria in Solid Tumors 1.1 was 57%. No grade 3-4 immune-related adverse events were observed in the neoadjuvant setting. The 1- and 2-year recurrence-free survival rate was 100% and 92% for patients with any pathologic response, compared to 88% and 55% for patients who did not have a pathologic response (P = 0.005). Increased immune cell infiltration at baseline, and decrease in M2 macrophages during treatment, were associated with pathologic response. Our results indicate that neoadjuvant relatlimab and nivolumab induces a high pCR rate. Safety during neoadjuvant therapy is favourable compared to other combination immunotherapy regimens. These data, in combination with the results of the RELATIVITY-047 trial1 , provide further confirmation of the efficacy and safety of this new immunotherapy regimen., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
25. A diagnostic dilemma: Atypical melanocytic lesions arising in the setting of treatment with the BRAF inhibitor, vemurafenib.
- Author
-
Clark M, Imbriano P, Harwood M, Busam K, and Robinson-Bostom L
- Abstract
Competing Interests: None disclosed.
- Published
- 2022
- Full Text
- View/download PDF
26. Comparison of community pathologists with expert dermatopathologists evaluating Breslow thickness and histopathologic subtype in a large international population-based study of melanoma.
- Author
-
Yardman-Frank JM, Bronner B, Rosso S, From L, Busam K, Groben P, Tucker P, Cust A, Armstrong B, Kricker A, Marrett L, Anton-Culver H, Gruber S, Gallagher R, Zanetti R, Sacchetto L, Dwyer T, Venn A, Orlow I, Kanetsky P, Luo L, Thomas N, Begg C, and Berwick M
- Abstract
Competing Interests: None disclosed.
- Published
- 2021
- Full Text
- View/download PDF
27. Clinical, morphologic, and genomic findings in ROS1 fusion Spitz neoplasms.
- Author
-
Gerami P, Kim D, Compres EV, Zhang B, Khan AU, Sunshine JC, Quan VL, and Busam K
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Female, Humans, Male, Middle Aged, Oncogene Fusion genetics, Oncogene Proteins, Fusion genetics, Nevus, Epithelioid and Spindle Cell genetics, Nevus, Epithelioid and Spindle Cell pathology, Protein-Tyrosine Kinases genetics, Proto-Oncogene Proteins genetics, Skin Neoplasms genetics, Skin Neoplasms pathology
- Abstract
The presence of a characteristic chimeric fusion as the initiating genomic event is one defining feature of Spitz neoplasms. Characterization of specific subtypes of Spitz neoplasms allows for better recognition facilitating diagnosis. Data on clinical outcomes of the specific tumor types may help in predicting behavior. In this study we present the largest series to date on ROS1 fusion Spitz neoplasms. We present the clinical, morphologic, and genomic features of 17 cases. We compared the morphologic features of these 17 cases to a cohort of 99 other non-ROS1 Spitz neoplasms to assess for features that may have high specificity for ROS1 fusions. These tumors consisted of ten Spitz nevi and seven Spitz tumors. None of the cases met criteria for a diagnosis of Spitz melanoma. Morphologically, the ROS1 fusion tumors of this series were characterized by a plaque-like or nodular silhouette, often densely cellular intraepidermal melanocyte proliferation, frequent pagetosis, tendency toward spindle cell cytomorphology, low grade nuclear atypia, and floating nests with occasional transepidermal elimination. However, there was a significant range in microscopic appearances, including two cases with morphologic features of a desmoplastic Spitz nevus. Different binding partners to ROS1 were identified with PWWP2A and TPM3 being the most common. No case had a recurrence or metastasis. Our findings document that most ROS1 fusion Spitz neoplasms have some typical characteristic microscopic features, while a small proportion will have features overlapping with other genomic subtypes of Spitz neoplasms. Preliminary evidence suggests that they tend to be indolent or low grade neoplasms.
- Published
- 2021
- Full Text
- View/download PDF
28. Primary cutaneous marginal zone lymphoma with leptomeningeal involvement and a durable response to rituximab.
- Author
-
Virmani P, Busam K, Myskowski PL, Yang A, Magro C, and Horwitz S
- Published
- 2017
- Full Text
- View/download PDF
29. Vitamin D receptor polymorphisms and survival in patients with cutaneous melanoma: a population-based study.
- Author
-
Orlow I, Reiner AS, Thomas NE, Roy P, Kanetsky PA, Luo L, Paine S, Armstrong BK, Kricker A, Marrett LD, Rosso S, Zanetti R, Gruber SB, Anton-Culver H, Gallagher RP, Dwyer T, Busam K, Begg CB, and Berwick M
- Subjects
- Australia epidemiology, Canada epidemiology, Female, Genotype, Haplotypes, Humans, Italy epidemiology, Male, Melanoma mortality, Polymorphism, Single Nucleotide, Proportional Hazards Models, Skin Neoplasms mortality, United States epidemiology, Melanoma genetics, Receptors, Calcitriol genetics, Skin Neoplasms genetics
- Abstract
Factors known to affect melanoma survival include age at presentation, sex and tumor characteristics. Polymorphisms also appear to modulate survival following diagnosis. Result from other studies suggest that vitamin D receptor (VDR) polymorphisms (SNPs) impact survival in patients with glioma, renal cell carcinoma, lung, breast, prostate and other cancers; however, a comprehensive study of VDR polymorphisms and melanoma-specific survival is lacking. We aimed to investigate whether VDR genetic variation influences survival in patients with cutaneous melanoma. The analysis involved 3566 incident single and multiple primary melanoma cases enrolled in the international population-based Genes, Environment, and Melanoma Study. Melanoma-specific survival outcomes were calculated for each of 38 VDR SNPs using a competing risk analysis after adjustment for covariates. There were 254 (7.1%) deaths due to melanoma during the median 7.6 years follow-up period. VDR SNPs rs7299460, rs3782905, rs2239182, rs12370156, rs2238140, rs7305032, rs1544410 (BsmI) and rs731236 (TaqI) each had a statistically significant (trend P values < 0.05) association with melanoma-specific survival in multivariate analysis. One functional SNP (rs2239182) remained significant after adjustment for multiple testing using the Monte Carlo method. None of the SNPs associated with survival were significantly associated with Breslow thickness, ulceration or mitosis. These results suggest that the VDR gene may influence survival from melanoma, although the mechanism by which VDR exerts its effect does not seem driven by tumor aggressiveness. Further investigations are needed to confirm our results and to understand the relationship between VDR and survival in the combined context of tumor and host characteristics., (© The Author 2015. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2016
- Full Text
- View/download PDF
30. Dermatopathology quiz: A dome-shaped papule on the cheek.
- Author
-
Elbendary A, Jacobson E, Busam K, and Elston D
- Published
- 2015
- Full Text
- View/download PDF
31. Sun exposure and melanoma survival: a GEM study.
- Author
-
Berwick M, Reiner AS, Paine S, Armstrong BK, Kricker A, Goumas C, Cust AE, Thomas NE, Groben PA, From L, Busam K, Orlow I, Marrett LD, Gallagher RP, Gruber SB, Anton-Culver H, Rosso S, Zanetti R, Kanetsky PA, Dwyer T, Venn A, Lee-Taylor J, and Begg CB
- Subjects
- Female, Humans, Male, Proportional Hazards Models, Sunburn complications, Melanoma mortality, Skin Neoplasms mortality, Sunburn epidemiology, Sunlight adverse effects
- Abstract
Background: We previously reported a significant association between higher UV radiation exposure before diagnosis and greater survival with melanoma in a population-based study in Connecticut. We sought to evaluate the hypothesis that sun exposure before diagnosis was associated with greater survival in a larger, international population-based study with more detailed exposure information., Methods: We conducted a multicenter, international population-based study in four countries-Australia, Italy, Canada, and the United States-with 3,578 cases of melanoma with an average of 7.4 years of follow-up. Measures of sun exposure included sunburn, intermittent exposure, hours of holiday sun exposure, hours of water-related outdoor activities, ambient ultraviolet B (280-320 nm) dose, histologic solar elastosis, and season of diagnosis., Results: Results were not strongly supportive of the earlier hypothesis. Having had any sunburn in 1 year within 10 years of diagnosis was inversely associated with survival; solar elastosis-a measure of lifetime cumulative exposure-was not. In addition, none of the intermittent exposure measures-water-related activities and sunny holidays-were associated with melanoma-specific survival. Estimated ambient UVB dose was not associated with survival., Conclusion: Although there was an apparent protective effect of sunburns within 10 years of diagnosis, there was only weak evidence in this large, international, population-based study of melanoma that sun exposure before diagnosis is associated with greater melanoma-specific survival., Impact: This study adds to the evidence that sun exposure before melanoma diagnosis has little effect on survival with melanoma., (©2014 American Association for Cancer Research.)
- Published
- 2014
- Full Text
- View/download PDF
32. A genome-wide high-resolution array-CGH analysis of cutaneous melanoma and comparison of array-CGH to FISH in diagnostic evaluation.
- Author
-
Wang L, Rao M, Fang Y, Hameed M, Viale A, Busam K, and Jhanwar SC
- Subjects
- Algorithms, Genetic Variation, Genome-Wide Association Study methods, Genome-Wide Association Study standards, Humans, Melanoma genetics, Melanoma pathology, Sensitivity and Specificity, Skin Neoplasms, Melanoma, Cutaneous Malignant, Comparative Genomic Hybridization methods, Comparative Genomic Hybridization standards, In Situ Hybridization, Fluorescence methods, In Situ Hybridization, Fluorescence standards, Melanoma diagnosis
- Abstract
Benign melanocytic nevi and cutaneous melanomas can be difficult to differentiate by means of routine microscopic analysis. Recent evidence has suggested that cytogenomic analysis may be a useful diagnostic method for evaluation of melanocytic proliferations. We investigated the array-based comparative genomic hybridization (aCGH) platform for DNA copy number analysis of formalin-fixed, paraffin-embedded (FFPE) tissues in melanocytic tumors and compared aCGH analysis with fluorescence in situ hybridization (FISH) assays in diagnosis of melanoma. aCGH findings and FISH results were interpreted independently in a blinded fashion. Positive findings were not noted in any benign nevi at aCGH analysis, whereas substantial unbalanced genomic aberrations were revealed in 92% of melanomas. Positive results were obtained in 72% of melanomas via the four-probe FISH assay (RREB1/MYB/CEP6/CCND1). A few additional FISH studies were performed to verify some aCGH findings of focal amplification of oncogenes and homozygous deletion of tumor suppressor genes. The overall concordance in aberrations detected using the two methods was 90%. Most discrepancies were due to a minor abnormal clone identified via FISH that was below analytical sensitivity of the FFPE aCGH test. Our study demonstrated that copy number analysis of FFPE tumor samples via aCGH is a robust and reliable method in diagnosis of melanoma and that aCGH and FISH tests should be used as complementary methods to improve the accuracy of genetic evaluation of melanocytic tumors., (Copyright © 2013 American Society for Investigative Pathology and the Association for Molecular Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
33. Convergent multi-miRNA targeting of ApoE drives LRP1/LRP8-dependent melanoma metastasis and angiogenesis.
- Author
-
Pencheva N, Tran H, Buss C, Huh D, Drobnjak M, Busam K, and Tavazoie SF
- Subjects
- Animals, Cell Line, Tumor, Cells, Cultured, Disease Models, Animal, Endothelial Cells metabolism, Gene Expression Regulation, Neoplastic, HSP40 Heat-Shock Proteins metabolism, Humans, LDL-Receptor Related Proteins metabolism, Low Density Lipoprotein Receptor-Related Protein-1 metabolism, Melanoma drug therapy, Melanoma metabolism, Melanoma pathology, Mice, Mice, Inbred C57BL, Mice, Nude, MicroRNAs antagonists & inhibitors, Neoplasm Metastasis drug therapy, Neoplasm Metastasis pathology, Neovascularization, Pathologic drug therapy, Neovascularization, Pathologic genetics, Oligonucleotides pharmacology, Apolipoproteins E metabolism, Melanoma genetics, MicroRNAs metabolism, Neoplasm Metastasis genetics, Neovascularization, Pathologic metabolism
- Abstract
Through in vivo selection of human cancer cell populations, we uncover a convergent and cooperative miRNA network that drives melanoma metastasis. We identify miR-1908, miR-199a-5p, and miR-199a-3p as endogenous promoters of metastatic invasion, angiogenesis, and colonization in melanoma. These miRNAs convergently target apolipoprotein E (ApoE) and the heat shock factor DNAJA4. Cancer-secreted ApoE suppresses invasion and metastatic endothelial recruitment (MER) by engaging melanoma cell LRP1 and endothelial cell LRP8 receptors, respectively, while DNAJA4 promotes ApoE expression. Expression levels of these miRNAs and ApoE correlate with human metastatic progression outcomes. Treatment of cells with locked nucleic acids (LNAs) targeting these miRNAs inhibits metastasis to multiple organs, and therapeutic delivery of these LNAs strongly suppresses melanoma metastasis. We thus identify miRNAs with dual cell-intrinsic/cell-extrinsic roles in cancer, reveal convergent cooperativity in a metastatic miRNA network, identify ApoE as an anti-angiogenic and metastasis-suppressive factor, and uncover multiple prognostic miRNAs with synergistic combinatorial therapeutic potential in melanoma., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
34. Investigation of the effect of MDM2 SNP309 and TP53 Arg72Pro polymorphisms on the age of onset of cutaneous melanoma.
- Author
-
Cotignola J, Chou JF, Roy P, Mitra N, Busam K, Halpern AC, and Orlow I
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Child, Child, Preschool, Confidence Intervals, Female, Genotype, Humans, Kaplan-Meier Estimate, Male, Melanoma pathology, Middle Aged, Odds Ratio, Polymorphism, Single Nucleotide, Proportional Hazards Models, Sex Factors, Skin Neoplasms pathology, Young Adult, Age of Onset, Melanoma genetics, Neoplasm Recurrence, Local genetics, Proto-Oncogene Proteins c-mdm2 genetics, Skin Neoplasms genetics, Tumor Suppressor Protein p53 genetics
- Abstract
Melanoma accounts for the majority of deaths from skin cancer. Women tend to be diagnosed at a younger age and have better survival than men. A tumor-host interaction might be responsible for these gender-specific differences. Recently, a functional single-nucleotide polymorphism in the promoter of the human homolog of mouse double minute 2 (MDM2) gene was characterized: single-nucleotide polymorphism (SNP)309 increases the MDM2 transcription. In melanoma, the effects for SNP309 and the related tumor protein p53 (TP53) Arg72Pro are inconsistent among published reports. This study investigated the association between SNP309 (RefSNP accession ID (rs)2279744) and TP53 codon 72 (rs1042522) polymorphisms, with outcome in a hospital-based cohort of 990 patients with melanoma. We assessed whether these polymorphisms were associated with clinicopathological and phenotypic characteristics and whether these SNPs affect the age of onset of the disease, recurrence, and survival. No significant associations were found between the SNPs and survival. However, women carrying the SNP309 GG genotype were less likely to be diagnosed at a younger age: odds ratio(adjusted<50) 0.52 (0.29-0.92). Our results suggest that women carrying the SNP309 GG genotype might be at lower risk of developing melanoma at a younger age compared with those carrying TG or TT. Further studies are needed to determine whether a nearby functional polymorphism is responsible for this effect in premenopausal women.
- Published
- 2012
- Full Text
- View/download PDF
35. CTLA-4 blockade increases antigen-specific CD8(+) T cells in prevaccinated patients with melanoma: three cases.
- Author
-
Yuan J, Ginsberg B, Page D, Li Y, Rasalan T, Gallardo HF, Xu Y, Adams S, Bhardwaj N, Busam K, Old LJ, Allison JP, Jungbluth A, and Wolchok JD
- Subjects
- Adult, Aged, Antibodies, Monoclonal pharmacology, Antigens, CD metabolism, Antigens, Neoplasm immunology, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes pathology, CTLA-4 Antigen, Cells, Cultured, Cytokines metabolism, Female, Humans, Ipilimumab, Lymphocyte Activation drug effects, Male, Melanoma immunology, Melanoma pathology, Middle Aged, Neoplasm Metastasis, Peptide Fragments immunology, Skin Neoplasms immunology, Skin Neoplasms pathology, Tumor Escape, Antibodies, Monoclonal administration & dosage, CD8-Positive T-Lymphocytes drug effects, Cancer Vaccines, Immunotherapy, Melanoma therapy, Skin Neoplasms therapy
- Abstract
Background: Anti-cytotoxic T-lymphocyte antigen-4 (CTLA-4) antibodies, such as ipilimumab, have generated measurable immune responses to Melan-A, NY-ESO-1, and gp100 antigens in metastatic melanoma. Vaccination against such targets has potential for immunogenicity and may produce an effector-memory T-cell response., Methods: To determine the effect of CTLA-4 blockade on antigen-specific responses following vaccination, in-depth immune monitoring was performed on three ipilimumab-treated patients prevaccinated with gp100 DNA (IMF-24), gp100(209-217) and tyrosinase peptides plus GM-CSF DNA (IMF-32), or NY-ESO-1 protein plus imiquimod (IMF-11); peripheral blood mononuclear cells were analyzed by tetramer and/or intracellular cytokine staining following 10-day culture with HLA-A*0201-restricted gp100(209-217) (ITDQVPFSV), tyrosinase(369-377) (YMDGTMSQV), or 20-mer NY-ESO-1 overlapping peptides, respectively. Tumors from IMF-32 were analyzed by immunohistochemistry to help elucidate mechanism(s) underlying tumor escape., Results: Following vaccination, patients generated weak to no CD4(+) or CD8(+) T-cell response specific to the vaccine antigen but demonstrated increases in effector-memory (CCR7(lo)CD45RA(lo)) tetramer(+)CD8(+) T cells. After ipilimumab induction, patients experienced a robust, although sometimes transient, antigen-specific response for gp100 (IMF-32 and IMF-24) or NY-ESO-1 (IMF-11) and produced polyfunctional intracellular cytokines. Primary and metastatic tumors expressed tyrosinase but not gp100 or class I/II MHC molecules., Conclusion: Vaccination induced a measurable antigen-specific T-cell response that increased following CTLA-4 blockade, potentially "boosting" the vaccine-primed response. Tumor escape may be related to antigen loss or lack of MHC expression necessary for immune activity. These results in a limited number of patients support the need for further research into combining vaccination with ipilimumab and provide insight into mechanisms underlying tumor escape.
- Published
- 2011
- Full Text
- View/download PDF
36. Associations of cumulative sun exposure and phenotypic characteristics with histologic solar elastosis.
- Author
-
Thomas NE, Kricker A, From L, Busam K, Millikan RC, Ritchey ME, Armstrong BK, Lee-Taylor J, Marrett LD, Anton-Culver H, Zanetti R, Rosso S, Gallagher RP, Dwyer T, Goumas C, Kanetsky PA, Begg CB, Orlow I, Wilcox H, Paine S, and Berwick M
- Subjects
- Age Factors, Female, Humans, Male, Melanoma etiology, Melanoma pathology, Middle Aged, Odds Ratio, Phenotype, Skin Neoplasms etiology, Skin Neoplasms pathology, Skin Pigmentation, Elastic Tissue pathology, Elastic Tissue radiation effects, Sunlight adverse effects
- Abstract
Background: Solar elastosis adjacent to melanomas in histologic sections is regarded as an indicator of sun exposure, although the associations of UV exposure and phenotype with solar elastosis are yet to be fully explored., Methods: The study included 2,589 incident primary melanoma patients with assessment of histologic solar elastosis in the population-based Genes, Environment, and Melanoma study. Ambient erythemal UV (UVE) at places of residence and sun exposure hours, including body site-specific exposure, were collected. We examined the association of cumulative site-specific and non-site-specific sun exposure hours and ambient UVE with solar elastosis in multivariable models adjusted for age, sex, center, pigmentary characteristics, nevi, and, where relevant, body site., Results: Solar elastosis was associated most strongly with site-specific UVE [odds ratio (OR) for top exposure quartile, 5.20; 95% confidence interval (95% CI), 3.40-7.96; P for trend <0.001] and also with site-specific sun exposure (OR for top quartile, 5.12; 95% CI, 3.35-7.83; P for trend <0.001). Older age (OR at >70 years, 7.69; 95% CI, 5.14-11.52; P for trend < 0.001) and having more than 10 back nevi (OR, 0.77; 95% CI, 0.61-0.97; P = 0.03) were independently associated with solar elastosis., Conclusion: Solar elastosis had a strong association with higher site-specific UVE dose, older age, and fewer nevi., Impact: Solar elastosis could be a useful biomarker of lifetime site-specific UV. Future research is needed to explore whether age represents more than simple accumulation of sun exposure and to determine why people with more nevi may be less prone to solar elastosis., (©2010 AACR.)
- Published
- 2010
- Full Text
- View/download PDF
37. Lack of evidence for direct involvement of Merkel cell polyomavirus (MCV) in chronic lymphocytic leukemia (CLL).
- Author
-
Tolstov YL, Arora R, Scudiere SC, Busam K, Chaudhary PM, Chang Y, and Moore PS
- Subjects
- Aged, Carcinoma, Merkel Cell pathology, Female, Gene Expression Regulation, Leukemic, Gene Expression Regulation, Viral, Humans, Leukemia, Lymphocytic, Chronic, B-Cell pathology, Leukemia, Lymphocytic, Chronic, B-Cell virology, Male, Neoplasms, Second Primary pathology, Neoplasms, Second Primary virology, Polyomavirus Infections pathology, Tumor Virus Infections pathology, Antigens, Viral, Tumor biosynthesis, Carcinoma, Merkel Cell metabolism, Carcinoma, Merkel Cell virology, Leukemia, Lymphocytic, Chronic, B-Cell metabolism, Neoplasms, Second Primary metabolism, Polyomavirus, Polyomavirus Infections metabolism, Tumor Virus Infections metabolism
- Published
- 2010
- Full Text
- View/download PDF
38. Relationship between germline MC1R variants and BRAF-mutant melanoma in a North Carolina population-based study.
- Author
-
Thomas NE, Kanetsky PA, Edmiston SN, Alexander A, Begg CB, Groben PA, Hao H, Busam K, Ollila DW, Berwick M, and Conway K
- Subjects
- Genetic Variation, Germ-Line Mutation, Humans, Melanoma epidemiology, North Carolina epidemiology, Risk Factors, Skin Neoplasms epidemiology, Melanoma genetics, Proto-Oncogene Proteins B-raf genetics, Receptor, Melanocortin, Type 1 genetics, Skin Neoplasms genetics
- Published
- 2010
- Full Text
- View/download PDF
39. Functional polymorphisms in the promoter regions of MMP2 and MMP3 are not associated with melanoma progression.
- Author
-
Cotignola J, Roy P, Patel A, Ishill N, Shah S, Houghton A, Coit D, Halpern A, Busam K, Berwick M, and Orlow I
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Child, Child, Preschool, Disease Progression, Female, Humans, Male, Melanoma enzymology, Middle Aged, Polymerase Chain Reaction, Matrix Metalloproteinase 2 genetics, Matrix Metalloproteinase 3 genetics, Melanoma genetics, Polymorphism, Genetic, Promoter Regions, Genetic
- Abstract
Background: The matrix metalloproteinases (MMPs) are enzymes that cleave various components of the extracellular matrix (ECM) and basement membranes. MMPs are expressed in melanocytes and their overexpression has been linked to tumor development, progression and metastasis. At the genetic level, the following functional promoter polymorphisms are known to modify the gene transcription: -1306 C/T and -735 C/T in the MMP2 gene, and -1171 5A/6A in the MMP3 gene. Functional polymorphisms in MMP genes' promoter regions may modulate the risk for melanoma progression., Methods: We evaluated MMP2 and MMP3 germline polymorphisms in a group of 1002 melanoma patients using PCR-based methods, including fragment size analysis and melting temperature profiles. Two-sided Chi-Square, Cochran-Armitage tests for trend, Fisher's exact tests, and Kendall's Tau tests were performed to evaluate the associations between genotype and various clinical and epidemiologic factors. Multivariate analyses were conducted using logistic regression, adjusting for known melanoma confounders such as age, sex, phenotypic index, moles, freckles, and race. Survival estimates were computed using the Kaplan-Meier method and differences in survival were assessed using the log rank test., Results: All genotypes were in Hardy-Weinberg equilibrium. After adjustment for age, sex and phenotypic characteristics of melanoma risk, no significant associations were identified with the clinical, pathological, and epidemiological variables studied. The melting profile for MMP2 -735 C/T identified a new change in one sample. A new PCR-amplification followed by direct sequencing confirmed a heterozygote G to A substitution at position -729., Conclusion: This study does not provide strong evidence for further investigation into the role of the MMP2 and MMP3 variants in melanoma progression.
- Published
- 2007
- Full Text
- View/download PDF
40. Expression of the cancer/testis antigen NY-ESO-1 in primary and metastatic malignant melanoma (MM)--correlation with prognostic factors.
- Author
-
Velazquez EF, Jungbluth AA, Yancovitz M, Gnjatic S, Adams S, O'Neill D, Zavilevich K, Albukh T, Christos P, Mazumdar M, Pavlick A, Polsky D, Shapiro R, Berman R, Spira J, Busam K, Osman I, and Bhardwaj N
- Subjects
- Female, Humans, Male, Melanoma diagnosis, Middle Aged, Neoplasm Metastasis, Neoplasm Staging, Prognosis, Recurrence, Survival Analysis, Antigens, Neoplasm metabolism, Melanoma metabolism, Melanoma pathology, Membrane Proteins metabolism
- Abstract
Cancer/testis (CT) antigens are potential targets for cancer immunotherapy, with NY-ESO-1 being among the most immunogenic. In several clinical trials in malignant melanoma (MM) patients, NY-ESO-1 protein/peptides showed clear evidence of inducing specific immunity. However, little is known about NY-ESO-1 expression in primary and metastatic MM and its relationship to disease progression. We analyzed NY-ESO-1 expression immunohistochemically in a series of primary and metastatic MMs and its relation to prognostic parameters and survival. We studied 61 primary and 63 metastatic MM specimens (from 61 and 56 patients, respectively). The prevalence of NY-ESO-1 expression was significantly higher in metastatic versus primary tumors [18/56 (32%) versus 8/61 (13%), P = 0.015]. There was a significant association between initial stage at presentation and NY-ESO-1 expression [stage I (3.45%), stage II (9.52%) and stage III (45.45%), P = 0.0014]. Primary MMs expressing NY-ESO-1 were significantly thicker than NY-ESO-1 negative cases (median thickness 4.7 mm versus 1.53 mm respectively, P = 0.03). No significant difference was seen in overall survival. In conclusion, NY-ESO-1 is more frequently expressed in metastatic than in primary MM and its expression is associated with thicker primary lesions and a higher frequency of metastatic disease, indicative of a worse prognosis. Our study suggests that patients with metastatic MM who express NY-ESO-1 may benefit from NY-ESO-1-based immunotherapy.
- Published
- 2007
41. CDKN2A germline mutations in individuals with cutaneous malignant melanoma.
- Author
-
Orlow I, Begg CB, Cotignola J, Roy P, Hummer AJ, Clas BA, Mujumdar U, Canchola R, Armstrong BK, Kricker A, Marrett LD, Millikan RC, Gruber SB, Anton-Culver H, Zanetti R, Gallagher RP, Dwyer T, Rebbeck TR, Kanetsky PA, Wilcox H, Busam K, From L, and Berwick M
- Subjects
- Cyclin-Dependent Kinase Inhibitor p16 physiology, DNA, Neoplasm genetics, Exons genetics, Genetic Predisposition to Disease, Genetic Testing, Humans, Melanoma physiopathology, Risk Factors, Skin Neoplasms physiopathology, Cyclin-Dependent Kinase Inhibitor p16 genetics, Germ-Line Mutation genetics, Melanoma genetics, Skin Neoplasms genetics
- Abstract
Cyclin-dependent kinase inhibitor type 2A (CDKN2A) has been identified as a major melanoma susceptibility gene based on the presence of germline mutations in high-risk melanoma families. In this study, we sought to identify and characterize the spectrum of CDKN2A mutations affecting p16 inhibitor of cyclin-dependent kinase type 4 (INK4a) in individuals with melanoma using a population-based study design. DNA samples from 1189 individuals with incident multiple primary melanoma (MPM) and 2424 with incident single primary melanoma unselected for family history of melanoma were available for screening of CDKN2A (p16INK4a) mutations. Variants were classified for functional impact based on intragenic position, existing functional data, sequence, and structural analysis. The impact of individual mutations and functional groupings was assessed by comparing frequencies in cases of MPM versus cases with a single first primary melanoma, and by comparing the reported incidence rates in first-degree relatives. Our results show that mutations occur infrequently in these high-risk groups, and that they occur mainly in exons 1alpha and 2. Rare coding variants with putative functional impact are observed to increase substantially the risk of melanoma. With the exception of the variant in position -34 of CDKN2A of known functional consequence, the remaining rare variants in the non-coding region have no apparent impact on risk.
- Published
- 2007
- Full Text
- View/download PDF
42. Number of nevi and early-life ambient UV exposure are associated with BRAF-mutant melanoma.
- Author
-
Thomas NE, Edmiston SN, Alexander A, Millikan RC, Groben PA, Hao H, Tolbert D, Berwick M, Busam K, Begg CB, Mattingly D, Ollila DW, Tse CK, Hummer A, Lee-Taylor J, and Conway K
- Subjects
- Confidence Intervals, DNA Mutational Analysis, DNA, Neoplasm analysis, DNA, Neoplasm chemistry, DNA, Neoplasm genetics, Female, Genes, ras genetics, Humans, Male, Melanoma genetics, Middle Aged, Mutation, North Carolina epidemiology, Odds Ratio, Phenotype, Polymorphism, Single-Stranded Conformational, Risk Factors, Sequence Analysis, DNA, Skin Neoplasms genetics, Melanoma epidemiology, Nevus pathology, Proto-Oncogene Proteins B-raf genetics, Skin Neoplasms epidemiology, Ultraviolet Rays adverse effects
- Abstract
Malignant melanomas often contain BRAF or NRAS mutations, but the relationship of these mutations to ambient UV exposure in combination with phenotypic characteristics is unknown. In a population-based case series from North Carolina, 214 first primary invasive melanoma patients in the year 2000 were interviewed regarding their risk factors. Ambient solar UV exposures were estimated using residential histories and a satellite-based model. Cases were grouped on the basis of BRAF and NRAS somatic mutations, determined using single-strand conformation polymorphism analysis and radiolabeled DNA sequencing, and the risk profiles of these groups were compared. Mutually exclusive BRAF-mutant and NRAS-mutant cases occurred at frequencies of 43.0% and 13.6% with mean ages at diagnosis of 47.3 and 62.1 years, respectively. Tumors from patients with >14 back nevi were more likely to harbor either a BRAF mutation [age-adjusted odds ratio (OR), 3.2; 95% confidence interval (95% CI), 1.4-7.0] or an NRAS mutation (age-adjusted OR, 1.7; 95% CI, 0.6-4.8) compared with patients with 0 to 4 back nevi. However, BRAF-mutant and NRAS-mutant tumors were distinctive in that BRAF-mutant tumors were characteristic of patients with high early-life ambient UV exposure (adjusted OR, 2.6; 95% CI, 1.2-5.3). When ambient UV irradiance was analyzed by decadal age, high exposure at ages 0 to 20 years was associated with BRAF-mutant cases, whereas high exposure at ages 50 and 60 years was characteristic of NRAS-mutant cases. Our results suggest that although nevus propensity is important for the occurrence of both BRAF and NRAS-mutant melanomas, ambient UV irradiance influences risk differently based on the age of exposure. The association of BRAF mutations with early-life UV exposure provides evidence in support of childhood sun protection for melanoma prevention.
- Published
- 2007
- Full Text
- View/download PDF
43. Ambient UV, personal sun exposure and risk of multiple primary melanomas.
- Author
-
Kricker A, Armstrong BK, Goumas C, Litchfield M, Begg CB, Hummer AJ, Marrett LD, Theis B, Millikan RC, Thomas N, Culver HA, Gallagher RP, Dwyer T, Rebbeck TR, Kanetsky PA, Busam K, From L, Mujumdar U, Zanetti R, and Berwick M
- Subjects
- Adolescent, Adult, Child, Environmental Exposure, Female, Humans, Male, Middle Aged, Occupational Exposure, Odds Ratio, Radiation Dosage, Risk Factors, Risk Reduction Behavior, Melanoma etiology, Neoplasms, Multiple Primary etiology, Neoplasms, Radiation-Induced, Skin Neoplasms etiology, Sunlight adverse effects, Ultraviolet Rays adverse effects
- Abstract
Objective: Sun exposure is the main cause of melanoma in populations of European origin. No previous study has examined the effect of sun exposure on risk of multiple primary melanomas compared with people who have one melanoma., Methods: We identified and enrolled 2,023 people with a first primary melanoma (controls) and 1,125 with multiple primary melanomas (cases) in seven centers in four countries, recorded their residential history to assign ambient UV and interviewed them about their sun exposure., Results: Risk of multiple primary melanomas increased significantly (P<0.05) to OR=2.10 for the highest exposure quarter of ambient UV irradiance at birth and 10 years of age, to OR=1.38 for lifetime recreational sun exposure, to OR=1.85 for beach and waterside activities, to OR=1.57 for vacations in a sunnier climate, to OR=1.50 for sunburns. Occupational sun exposure did not increase risk (OR=1.03 for highest exposure). Recreational exposure at any age increased risk and appeared to add to risk from ambient UV in early life., Conclusions: People who have had a melanoma can expect to reduce their risk of a further melanoma by reducing recreational sun exposure whatever their age. The same is probably true for a person who has never had a melanoma.
- Published
- 2007
- Full Text
- View/download PDF
44. Matrix Metalloproteinase-9 (MMP-9) polymorphisms in patients with cutaneous malignant melanoma.
- Author
-
Cotignola J, Reva B, Mitra N, Ishill N, Chuai S, Patel A, Shah S, Vanderbeek G, Coit D, Busam K, Halpern A, Houghton A, Sander C, Berwick M, and Orlow I
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Child, Child, Preschool, Female, Genotype, Humans, Male, Melanoma enzymology, Middle Aged, Polymerase Chain Reaction, Promoter Regions, Genetic, Skin Neoplasms enzymology, Matrix Metalloproteinase 9 genetics, Melanoma genetics, Polymorphism, Single Nucleotide, Skin Neoplasms genetics
- Abstract
Background: Cutaneous Malignant Melanoma causes over 75% of skin cancer-related deaths, and it is clear that many factors may contribute to the outcome. Matrix Metalloproteinases (MMPs) play an important role in the degradation and remodeling of the extracellular matrix and basement membrane that, in turn, modulate cell division, migration and angiogenesis. Some polymorphisms are known to influence gene expression, protein activity, stability, and interactions, and they were shown to be associated with certain tumor phenotypes and cancer risk., Methods: We tested seven polymorphisms within the MMP-9 gene in 1002 patients with melanoma in order to evaluate germline genetic variants and their association with progression and known risk factors of melanoma. The polymorphisms were selected based on previously published reports and their known or potential functional relevance using in-silico methods. Germline DNA was then genotyped using pyrosequencing, melting temperature profiles, heteroduplex analysis, and fragment size analysis., Results: We found that reference alleles were present in higher frequency in patients who tend to sunburn, have family history of melanoma, higher melanoma stage, intransit metastasis and desmoplastic melanomas among others. However, after adjustment for age, sex, phenotypic index, moles, and freckles only Q279R, P574R and R668Q had significant associations with intransit metastasis, propensity to tan/sunburn and primary melanoma site., Conclusion: This study does not provide strong evidence for further investigation into the role of the MMP-9 SNPs in melanoma progression.
- Published
- 2007
- Full Text
- View/download PDF
45. Population-based study of natural variation in the melanocortin-1 receptor gene and melanoma.
- Author
-
Kanetsky PA, Rebbeck TR, Hummer AJ, Panossian S, Armstrong BK, Kricker A, Marrett LD, Millikan RC, Gruber SB, Culver HA, Zanetti R, Gallagher RP, Dwyer T, Busam K, From L, Mujumdar U, Wilcox H, Begg CB, and Berwick M
- Subjects
- Adult, Aged, Female, Genetic Variation, Humans, Male, Middle Aged, Sequence Analysis, DNA, Melanoma genetics, Receptor, Melanocortin, Type 1 genetics
- Abstract
Natural variation in the coding region of the melanocortin-1 receptor (MC1R) gene is associated with constitutive pigmentation phenotypes and development of melanoma and nonmelanoma skin cancers. We investigated the effect of MC1R variants on melanoma using a large, international population-based study design with complete determination of all MC1R coding region variants. Direct sequencing was completed for 2,202 subjects with a single primary melanoma (controls) and 1,099 subjects with second or higher-order primary melanomas (cases) from Australia, the United States, Canada, and Italy. We observed 85 different MC1R variants, 10 of which occurred at a frequency >1%. Compared with controls, cases were more likely to carry two previously identified red hair ("R") variants [D84E, R151C, R160W, and D294H; odds ratio (OR), 1.6; 95% confidence interval (95% CI), 1.1-2.2]. This effect was similar among individuals carrying one R variant and one r variant (defined as any non-R MC1R variant; OR, 1.6; 95% CI, 1.3-2.2) and among those carrying only one R variant (OR, 1.5; 95% CI, 1.1-1.9). There was no statistically significant association among those carrying only one or two r variants. Effects were similar across geographic regions and categories of pigmentation characteristics or number of moles. Our results confirm that MC1R is a low-penetrance susceptibility locus for melanoma, show that pigmentation characteristics may not modify the relationship of MC1R variants and melanoma risk, and suggest that associations may be smaller than previously reported in part due to the study design.
- Published
- 2006
- Full Text
- View/download PDF
46. The prevalence of CDKN2A germ-line mutations and relative risk for cutaneous malignant melanoma: an international population-based study.
- Author
-
Berwick M, Orlow I, Hummer AJ, Armstrong BK, Kricker A, Marrett LD, Millikan RC, Gruber SB, Anton-Culver H, Zanetti R, Gallagher RP, Dwyer T, Rebbeck TR, Kanetsky PA, Busam K, From L, Mujumdar U, Wilcox H, and Begg CB
- Subjects
- Aged, Australia epidemiology, Canada epidemiology, Case-Control Studies, Chromatography, High Pressure Liquid, Exons genetics, Female, Humans, International Agencies, Introns genetics, Italy epidemiology, Male, Melanoma epidemiology, Middle Aged, Neoplasms, Multiple Primary epidemiology, Polymerase Chain Reaction, Polymorphism, Genetic, Skin Neoplasms epidemiology, United States epidemiology, Cyclin-Dependent Kinase Inhibitor p16 genetics, Germ-Line Mutation, Melanoma genetics, Neoplasms, Multiple Primary genetics, Skin Neoplasms genetics
- Abstract
Germ-line mutations of CDKN2A have been identified as strong risk factors for melanoma in studies of multiple-case families. However, an assessment of their relative risk for melanoma in the general population has been difficult because they occur infrequently. We addressed this issue using a novel population-based case-control study design in which "cases" have incident second- or higher-order melanomas [multiple primary melanoma (MPM)] and "controls" have incident first primary melanoma [single primary melanoma (SPM)]. Participants were ascertained from nine geographic regions in Australia, Canada, Italy, and United States. In the 1,189 MPM cases and 2,424 SPM controls who were eligible and available for analysis, the relative risk of a subsequent melanoma among patients with functional mutations who have an existing diagnosis of melanoma, after adjustments for age, sex, center, and known phenotypic risk factors, is estimated to be 4.3 (95% confidence interval, 2.3-7.7). The odds ratio varied significantly depending on the type of mutation involved. The results suggest that the relative risk of mutation carriers in the population may be lower than currently believed and that different mutations on the CDKN2A gene may confer substantially different risks of melanoma.
- Published
- 2006
- Full Text
- View/download PDF
47. A design for cancer case-control studies using only incident cases: experience with the GEM study of melanoma.
- Author
-
Begg CB, Hummer AJ, Mujumdar U, Armstrong BK, Kricker A, Marrett LD, Millikan RC, Gruber SB, Culver HA, Zanetti R, Gallagher RP, Dwyer T, Rebbeck TR, Busam K, From L, and Berwick M
- Subjects
- Adult, Age Distribution, Aged, Case-Control Studies, Eye Color, Family Health, Female, Hair Color, Humans, Incidence, Male, Melanoma pathology, Middle Aged, Nevus epidemiology, Nevus pathology, Phenotype, Reproducibility of Results, Risk Factors, Sex Distribution, Skin Neoplasms pathology, Melanoma epidemiology, Neoplasms, Second Primary epidemiology, Skin Neoplasms epidemiology
- Abstract
Background: The population-based case-control study is not suited to the evaluation of rare genetic (or environmental) factors. The use of a novel case-control design in which cases have second primaries and controls are cancer survivors has been proposed for this purpose., Methods: We report results from an international study of melanoma that involved population-based ascertainment of incident cases of second or subsequent primary melanoma as the 'case' group and incident cases of first primary melanoma as the 'control' group. We evaluate the validity of the study design by comparing the results obtained for phenotypic factors that have been shown consistently to be associated with melanoma in previous conventional studies with the results from a conventional case-control study conducted in Connecticut and from literature reviews., Results: All but one of the known risk factors for melanoma were shown to be significantly associated with melanoma in our study, though the individual odds ratios appear to be somewhat attenuated relative to the magnitudes typically observed in the literature., Conclusions: Patients with a second or subsequent primary cancer of a single type represent a potentially valuable and under-utilized resource for the study of cancer aetiology.
- Published
- 2006
- Full Text
- View/download PDF
48. Polymorphisms in nucleotide excision repair genes and risk of multiple primary melanoma: the Genes Environment and Melanoma Study.
- Author
-
Millikan RC, Hummer A, Begg C, Player J, de Cotret AR, Winkel S, Mohrenweiser H, Thomas N, Armstrong B, Kricker A, Marrett LD, Gruber SB, Culver HA, Zanetti R, Gallagher RP, Dwyer T, Rebbeck TR, Busam K, From L, Mujumdar U, and Berwick M
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Case-Control Studies, Child, Female, Genotype, Humans, Male, Melanoma etiology, Middle Aged, Odds Ratio, Risk Factors, Skin Neoplasms etiology, DNA Repair, Melanoma genetics, Polymorphism, Genetic, Skin Neoplasms genetics, Xeroderma Pigmentosum Group D Protein genetics
- Abstract
Polymorphisms in six genes involved in nucleotide excision repair of DNA were examined in a large population-based case-control study of melanoma. Genotyping was conducted for 2485 patients with a single primary melanoma (controls) and 1238 patients with second or higher order primary melanomas (cases). Patients were ascertained from nine geographic regions in Australia, Canada, Italy and the United States. Positive associations were observed for XPD 312 Asn/Asn versus Asp/Asp [odds ratio (OR) = 1.5, 95% confidence interval (CI) 1.2-1.9] and XPD 751 Gln/Gln versus Lys/Lys (OR = 1.4, 95% CI 1.1-1.7) genotypes and melanoma. The combined XPD Asn (A) 312 + Gln (C) 751 haplotype was significantly more frequent in cases (32%) compared with controls (29%) (P = 0.003) and risk of melanoma increased significantly with one and two copies of the haplotype (ORs 1.2, 95% CI 1.0-1.4, and 1.6, 95% CI 1.2-2.0, trend P = 0.002). No significant associations were observed for HR23B codon 249, XPG codon 1104, XPC codon 939, XPF codon 415, XPF nt 2063, ERCC6 codon 1213 or ERCC6 codon 1230. ORs for XPD and XPC genotypes were stronger for melanoma diagnosed at an early age, but tests for interaction were not statistically significant. The results provide further evidence for a role of XPD in the etiology of melanoma.
- Published
- 2006
- Full Text
- View/download PDF
49. Lifetime risk of melanoma in CDKN2A mutation carriers in a population-based sample.
- Author
-
Begg CB, Orlow I, Hummer AJ, Armstrong BK, Kricker A, Marrett LD, Millikan RC, Gruber SB, Anton-Culver H, Zanetti R, Gallagher RP, Dwyer T, Rebbeck TR, Mitra N, Busam K, From L, and Berwick M
- Subjects
- Adult, Age Distribution, Aged, Australia epidemiology, Canada epidemiology, Case-Control Studies, Female, Genetic Predisposition to Disease, Genotype, Humans, Incidence, Italy epidemiology, Logistic Models, Male, Melanoma mortality, Middle Aged, Penetrance, Risk Assessment, Skin Neoplasms mortality, Survival Rate, United States epidemiology, Genes, p16, Germ-Line Mutation, Melanoma epidemiology, Melanoma genetics, Skin Neoplasms epidemiology, Skin Neoplasms genetics
- Abstract
Background: Germline mutations in the CDKN2A gene have been linked to melanoma incidence in many families with multiple cases of the disease. Previous studies of multiple-case families have indicated that the lifetime risk (i.e., penetrance) of melanoma in CDKN2A mutation carriers is very high, ranging from 58% in Europe to 91% in Australia by age 80 years. In this study, we examined lifetime melanoma risk among CDKN2A mutation carriers using carriers who were identified in a population-based study of melanoma., Methods: Probands for the study were incident case patients with either first or subsequent melanoma who were identified in nine geographic regions in Australia, Canada, the United States, and Italy. A total of 3626 probands (53% participation rate) with adequate DNA for analysis were recruited and genotyped for CDKN2A mutations. From the 3550 probands whose DNA could be amplified by polymerase chain reaction of CDKN2A exons 1alpha, 2, and 3 and surrounding regions, 65 mutation carriers were identified. Melanoma histories in first-degree relatives of these probands were used to calculate the lifetime risk in CDKN2A mutation carriers using the kin-cohort method., Results: The risk of melanoma in CDKN2A mutation carriers was approximately 14% (95% CI = 8% to 22%) by age 50 years, 24% (95% CI = 15% to 34%) by age 70 years, and 28% (95% CI = 18% to 40%) by age 80 years. Eighteen probands had three or more first-degree relatives with melanoma, but only one was a carrier of a CDKN2A mutation., Conclusions: CDKN2A mutation carriers in the general population have a much lower risk of melanoma than that suggested by estimates obtained from multiple-case families. The preponderance of familial clustering of melanoma occurs in families without identifiable mutations in CDKN2A.
- Published
- 2005
- Full Text
- View/download PDF
50. Clinical significance of BRAF mutations in metastatic melanoma.
- Author
-
Chang DZ, Panageas KS, Osman I, Polsky D, Busam K, and Chapman PB
- Abstract
Forty to eighty percent of melanoma tumors have activating mutations in BRAF although the clinical importance of these mutations is not clear. We previously reported an analysis of BRAF mutations in metastatic melanoma samples from 68 patients. In this study, we correlated patient baseline characteristics, prognostic factors, and/or clinical outcomes with the presence of BRAF mutations. No significant differences were observed in age, gender, location of primary melanoma, stage at the diagnosis, and depth of primary tumor between patients with and without BRAF mutations. Melanomas harboring BRAF mutations were more likely to metastasize to liver (P = 0.02) and to metastasize to multiple organs (P = 0.048). Neither time to progression to stage IV nor overall survival were associated with BRAF mutations. In conclusion, we observed no significant differences in clinical characteristics or outcomes between melanomas with or without BRAF mutations. Although there was an increased frequency of liver metastasis and tendency to metastasize to multiple organs in tumors with BRAF mutations, there was no detectable effect on survival. Future prospective studies should include analysis of whether BRAF mutations in melanoma tumors correlate with an increased tendency to metastasize to liver or to multiple organs.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.