PhotosBiographyFacebookTwitter

From Wikipedia, the free encyclopedia
Bert Vogelstein
Born (1949-06-02) June 2, 1949 (age 74)
Alma mater University of Pennsylvania
Johns Hopkins School of Medicine
Known forp53, Vogelgram, somatic evolution in cancer
SpouseIlene Vogelstein
ChildrenR. Jacob Vogelstein, Joshua T. Vogelstein, and one more, Grandchildren: 5
Awards Breakthrough Prize in Life Sciences (2013) [1]
Warren Triennial Prize (2014) [2]
Scientific career
Fields Oncology, Pathology
Institutions Johns Hopkins School of Medicine
Doctoral students
Website www.hhmi.org/research/investigators/vogelstein_bio.html

Bert Vogelstein (born 1949) is director of the Ludwig Center, Clayton Professor of Oncology and Pathology and a Howard Hughes Medical Institute investigator at The Johns Hopkins Medical School and Sidney Kimmel Comprehensive Cancer Center. [4] A pioneer in the field of cancer genomics, his studies on colorectal cancers revealed that they result from the sequential accumulation of mutations in oncogenes and tumor suppressor genes. These studies now form the paradigm for modern cancer research and provided the basis for the notion of the somatic evolution of cancer.

Research

In the 1980s, Vogelstein developed new experimental approaches to study human tumors. [5] His studies of various stages of colorectal cancers led him to propose a specific model for human tumorigenesis in 1988. In particular, he suggested that "cancer is caused by sequential mutations of specific oncogenes and tumor suppressor genes". [6] [7] [8]

The first tumor suppressor gene validating this hypothesis was that encoding p53. The p53 protein was discovered 10 years earlier by several groups, including that of David Lane and Lionel Crawford, Arnold Levine, and Lloyd Old. But there was no evidence that p53 played a major role in human cancers, and the gene encoding p53 (TP53) was thought to be an oncogene rather than a tumor suppressor gene. In 1989, Vogelstein and his students discovered that TP53 not only played a role in human tumorigenesis, but that it was a common denominator of human tumors, mutated in the majority of them. [9] [10] He then discovered the mechanism through which TP53 suppresses tumorigenesis. Prior to these studies, the only biochemical function attributed to p53 was its binding to heat shock proteins. Vogelstein and his colleagues demonstrated that p53 had a much more specific activity: it bound DNA in a sequence-specific manner. They precisely defined its consensus recognition sequence and showed that virtually all p53 mutations found in tumors resulted in loss of the sequence-specific transcriptional activation properties of p53. [11] [12] They subsequently discovered genes that are directly activated by p53 to control cell birth and cell death. [13] [14] His group's more recent studies examining the entire compendium of human genes have shown that the TP53 gene is more frequently mutated in cancers than any other gene . [15] [16] [12] [17] [18] [19]

In 1991, Vogelstein and long-time colleague Kenneth W. Kinzler, working with Yusuke Nakamura in Japan, discovered another tumor suppressor gene. This gene, called APC, was responsible for Familial Adenomatous Polyposis (FAP), a syndrome associated with the development of numerous small benign tumors, some of which progress to cancer. [20] [21] This gene was independently discovered by Ray White's group at the University of Utah. Vogelstein and Kinzler subsequently showed that non-hereditary (somatic) mutations of APC initiate most cases of colon and rectal cancers. They also showed how APC functions – through binding to beta-catenin and stimulating its degradation. [22] [23]

Vogelstein and Kinzler worked with Albert de la Chapelle and Lauri Aaltonen at the U. Helsinki to identify the genes responsible for Hereditary NonPolyposis Colorectal Cancer ( HNPCC), the other major form of heritable colorectal tumorigenesis. They were the first to localize one of the major causative genes to a specific chromosomal locus through linkage studies. This localization soon led them and other groups to identify repair genes such as MSH2 and MLH1 that are responsible for most cases of this syndrome. [24] [25] [26] [27]

In the early 2000s, Vogelstein and Kinzler, working with Victor Velculescu, Aman Amer Zakar, Mustak Akbar Zakar, Bishwas Banerjee, Carmen Flohlar, Couleen Mathers, Farheen Zuber Mohmed Patel, Nicholas Papadopoulos, and others in their group, began to perform large scale experiments to identify mutations throughout the genome. They were to perform "exomic sequencing", meaning determination of the sequence of every protein-encoding gene in the human genome. The first analyzed tumors included those of the colon, breast, pancreas, and brain. These studies outlined the landscapes of human cancer genomes, later confirmed by massively parallel sequencing of many different tumor types by laboratories throughout the world. [28] In the process of analyzing all the protein-encoding genes within cancers, Vogelstein and his colleagues discovered several novel genes that play important roles in cancer, such as PIK3CA, [29] IDH1, [30] IDH2, [30] ARID1A, [31] ARID2, ATRX, [32] DAXX, [32] MLL2, MLL3, CIC, and RNF43. [33] [34] [35] [36]

Vogelstein pioneered the idea that somatic mutations represent uniquely specific biomarkers for cancer, creating the field now called "liquid biopsies". Working with post-doctoral fellow David Sidransky in the early 1990s, he showed that such somatic mutations were detectable in the stool of colorectal cancer patients and the urine of bladder cancer patients. [37] [38] For this purpose, they developed "Digital PCR" in which DNA molecules are examined one-by-one to determine whether they are normal or mutated. [39] One of the techniques they invented for Digital PCR is called " BEAMing", in which the PCR is carried out on magnetic beads in water-in-oil emulsions. [40] BEAMing is now one of the core technologies used in some next-generation, massively parallel sequencing instruments. More recently, they developed a digital-PCR based technique called SafeSeqS, in which every DNA template molecule is recognized by a unique molecular barcode. SafeSeqS dramatically enhances the ability to identify rare variants among DNA sequences, allowing such variants to be detected when they are present in only 1 in more than 10,000 total DNA molecules. [41] [42] [43] [44] [45]

In mid-2019, Vogelstein started collaborating with the group of Martin Nowak at Harvard University. Together with their groups, they developed mathematical models to explain the evolution of resistance against targeted therapies. [46] They showed that the sequential administration of multiple targeted drugs precludes any chance for cure — even when there are no possible mutations that can confer cross-resistance to both drugs. Thus, simultaneous combination of targeted therapies (as opposed to sequential) is the preferred strategy as there is at least a potential for cure. [47]

Citations

Vogelstein has published nearly 600 scientific papers. Vogelstein's research papers have been cited over 430,000 times. [48]

In 2016 Semantic Scholar AI program included Vogelstein on its list of top ten most influential biomedical researchers. [49]

Awards

Affiliations

References

  1. ^ "Breakthrough Prize – Life Sciences Breakthrough Prize Laureates – Bert Vogelstein". breakthroughprize.org.
  2. ^ a b "Mass. General Hospital's Warren Triennial Prize to honor Bert Vogelstein, MD - Massachusetts General Hospital, Boston, MA". Archived from the original on 2015-02-21. Retrieved 2015-02-21.
  3. ^ Landau, Misia (1 January 2015). "An Interview with Bert Vogelstein and Kenneth Kinzler". Clinical Chemistry. 61 (1): 9–20. doi: 10.1373/clinchem.2014.223271. PMID  25550474 – via clinchem.aaccjnls.org.
  4. ^ "Interview with Bert Vogelstein". Archived from the original on 2013-06-03. Retrieved 2010-04-30.
  5. ^ Vogelstein B, Fearon ER, Hamilton SR, Feinberg AP (1985). "Use of restriction fragment length polymorphisms to determine the clonal origin of human tumors". Science. 227 (4687): 642–5. Bibcode: 1985Sci...227..642V. doi: 10.1126/science.2982210. PMID  2982210.
  6. ^ Fearon ER, Hamilton SR, Vogelstein B (1987). "Clonal analysis of human colorectal tumors". Science. 238 (4824): 193–7. Bibcode: 1987Sci...238..193F. doi: 10.1126/science.2889267. PMID  2889267.
  7. ^ Vogelstein B; Fearon ER; Hamilton SR; Kern SE; Preisinger AC; Leppert M; Nakamura Y; White R; Smits AM; Bos JL (1988). "Genetic alterations during colorectal-tumor development". N Engl J Med. 319 (9): 525–32. doi: 10.1056/NEJM198809013190901. PMID  2841597.
  8. ^ Fearon ER, Vogelstein B (June 1990). "A genetic model for colorectal tumorigenesis". Cell. 61 (5): 759–67. doi: 10.1016/0092-8674(90)90186-i. PMID  2188735. S2CID  22975880.
  9. ^ Baker SJ, Fearon ER, Nigro JM, et al. (April 1989). "Chromosome 17 deletions and p53 gene mutations in colorectal carcinomas". Science. 244 (4901): 217–21. Bibcode: 1989Sci...244..217B. doi: 10.1126/science.2649981. PMID  2649981.
  10. ^ Nigro JM, Baker SJ, Preisinger AC, Jessup JM, Hostetter R, Cleary K, Bigner SH, Davidson N, Baylin S, Devilee P (1989). "Mutations in the p53 gene occur in diverse human tumour types". Nature. 342 (6250): 705–8. Bibcode: 1989Natur.342..705N. doi: 10.1038/342705a0. hdl: 2027.42/62925. PMID  2531845. S2CID  4353980.
  11. ^ el-Deiry WS, Kern SE, Pietenpol JA, Kinzler KW, Vogelstein B (April 1992). "Definition of a consensus binding site for p53". Nature Genetics. 1 (1): 45–49. doi: 10.1038/ng0492-45. PMID  1301998. S2CID  1710617.
  12. ^ a b Kern SE; Pietenpol JA; Thiagalingam S; Seymour A; Kinzler KW; Vogelstein B (May 1992). "Oncogenic forms of p53 inhibit p53-regulated gene expression". Science. 256 (5058): 827–30. Bibcode: 1992Sci...256..827K. doi: 10.1126/science.1589764. PMID  1589764.
  13. ^ el-Deiry WS; Tokino T; Velculescu VE; Levy DB; Parsons R; Trent JM; Lin D; Mercer WE; Kinzler KW; Vogelstein B. (November 1993). "WAF1, a potential mediator of p53 tumor suppression". Cell. 75 (4): 817–825. doi: 10.1016/0092-8674(93)90500-P. PMID  8242752.
  14. ^ Yu J; Zhang L; Hwang PM; Kinzler KW; Vogelstein B. (March 2001). "PUMA induces the rapid apoptosis of colorectal cancer cells". Mol Cell. 7 (3): 673–82. doi: 10.1016/s1097-2765(01)00213-1. PMID  11463391.
  15. ^ Kern SE, Kinzler KW, Bruskin A, et al. (June 1991). "Identification of p53 as a sequence-specific DNA-binding protein". Science. 252 (5013): 1708–11. Bibcode: 1991Sci...252.1708K. doi: 10.1126/science.2047879. PMID  2047879. S2CID  19647885.
  16. ^ el-Deiry WS, Kern SE, Pietenpol JA, Kinzler KW, Vogelstein B (1992). "Definition of a consensus binding site for p53". Nat Genet. 1 (1): 45–9. doi: 10.1038/ng0492-45. PMID  1301998. S2CID  1710617.
  17. ^ el-Deiry WS, Tokino T, Velculescu VE, et al. (November 1993). "WAF1, a potential mediator of p53 tumor suppression". Cell. 75 (4): 817–25. doi: 10.1016/0092-8674(93)90500-P. PMID  8242752.
  18. ^ Waldman T, Kinzler KW, Vogelstein B (1995). "p21 is necessary for the p53-mediated G1 arrest in human cancer cells". Cancer Res. 55 (22): 5187–90. PMID  7585571.
  19. ^ Yu J, Wang Z, Kinzler KW, Vogelstein B, Zhang L (February 2003). "PUMA mediates the apoptotic response to p53 in colorectal cancer cells". Proc. Natl. Acad. Sci. U.S.A. 100 (4): 1931–6. Bibcode: 2003PNAS..100.1931Y. doi: 10.1073/pnas.2627984100. PMC  149936. PMID  12574499.
  20. ^ Kinzler KW, Nilbert MC, Su LK, et al. (August 1991). "Identification of FAP locus genes from chromosome 5q21". Science. 253 (5020): 661–5. Bibcode: 1991Sci...253..661K. doi: 10.1126/science.1651562. PMID  1651562.
  21. ^ Nishisho I, Nakamura Y, Miyoshi Y, et al. (August 1991). "Mutations of chromosome 5q21 genes in FAP and colorectal cancer patients". Science. 253 (5020): 665–9. Bibcode: 1991Sci...253..665N. doi: 10.1126/science.1651563. PMID  1651563.
  22. ^ Powell SM, Zilz N, Beazer-Barclay Y, Bryan TM, Hamilton SR, Thibodeau SN, Vogelstein B, Kinzler KW (1992). "APC mutations occur early during colorectal tumorigenesis". Nature. 359 (6392): 235–7. Bibcode: 1992Natur.359..235P. doi: 10.1038/359235a0. PMID  1528264. S2CID  4277817.
  23. ^ Su LK, Vogelstein B, Kinzler KW (December 1993). "Association of the APC tumor suppressor protein with catenins". Science. 262 (5140): 1734–7. Bibcode: 1993Sci...262.1734S. doi: 10.1126/science.8259519. PMID  8259519.
  24. ^ Peltomäki P, Aaltonen LA, Sistonen P, Pylkkänen L, Mecklin JP, Järvinen H, Green JS, Jass JR, Weber JL, Leach FS (1993). "Genetic mapping of a locus predisposing to human colorectal cancer". Science. 260 (5109): 810–2. Bibcode: 1993Sci...260..810P. doi: 10.1126/science.8484120. PMID  8484120.
  25. ^ Leach FS, Nicolaides NC, Papadopoulos N, Liu B, Jen J, Parsons R, Peltomäki P, Sistonen P, Aaltonen LA, Nyström-Lahti M (1993). "Mutations of a mutS homolog in hereditary nonpolyposis colorectal cancer". Cell. 75 (6): 1215–25. doi: 10.1016/0092-8674(93)90330-s. PMID  8261515. S2CID  23321982.
  26. ^ Papadopoulos N, Nicolaides NC, Wei YF, et al. (March 1994). "Mutation of a mutL homolog in hereditary colon cancer". Science. 263 (5153): 1625–9. Bibcode: 1994Sci...263.1625P. doi: 10.1126/science.8128251. PMID  8128251.
  27. ^ Nicolaides NC, Papadopoulos N, Liu B, et al. (September 1994). "Mutations of two PMS homologues in hereditary nonpolyposis colon cancer". Nature. 371 (6492): 75–80. Bibcode: 1994Natur.371...75N. doi: 10.1038/371075a0. PMID  8072530. S2CID  4244907.
  28. ^ Wood LD, Parsons DW, Jones S, et al. (November 2007). "The genomic landscapes of human breast and colorectal cancers". Science. 318 (5853): 1108–13. Bibcode: 2007Sci...318.1108W. CiteSeerX  10.1.1.218.5477. doi: 10.1126/science.1145720. PMID  17932254. S2CID  7586573.
  29. ^ Samuels Y, Wang Z, Bardelli A, et al. (April 2004). "High frequency of mutations of the PIK3CA gene in human cancers". Science. 304 (5670): 554. doi: 10.1126/science.1096502. PMID  15016963. S2CID  10147415.
  30. ^ a b Yan H, Parsons DW, Jin G, et al. (February 2009). "IDH1 and IDH2 mutations in gliomas". N. Engl. J. Med. 360 (8): 765–73. doi: 10.1056/NEJMoa0808710. PMC  2820383. PMID  19228619.
  31. ^ Jones S, Wang TL, Shih IeM Mao TL, Nakayama K, Roden R, Glas R, Slamon D, Diaz LA Jr, Vogelstein B, Kinzler KW, Velculescu VE, Papadopoulos N (2010). "Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma". Science. 330 (6001): 228–31. Bibcode: 2010Sci...330..228J. doi: 10.1126/science.1196333. PMC  3076894. PMID  20826764.
  32. ^ a b Jiao Y, Shi C, Edil BH, et al. (March 2011). "DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors". Science. 331 (6021): 1199–203. Bibcode: 2011Sci...331.1199J. doi: 10.1126/science.1200609. PMC  3144496. PMID  21252315.
  33. ^ Jones S, Zhang X, Parsons DW, et al. (September 2008). "Core signaling pathways in human pancreatic cancers revealed by global genomic analyses". Science. 321 (5897): 1801–6. Bibcode: 2008Sci...321.1801J. doi: 10.1126/science.1164368. PMC  2848990. PMID  18772397.
  34. ^ Parsons DW, Jones S, Zhang X, et al. (September 2008). "An integrated genomic analysis of human glioblastoma multiforme". Science. 321 (5897): 1807–12. Bibcode: 2008Sci...321.1807P. doi: 10.1126/science.1164382. PMC  2820389. PMID  18772396.
  35. ^ Jones S, Hruban RH, Kamiyama M, et al. (April 2009). "Exomic sequencing identifies PALB2 as a pancreatic cancer susceptibility gene". Science. 324 (5924): 217. Bibcode: 2009Sci...324..217J. doi: 10.1126/science.1171202. PMC  2684332. PMID  19264984.
  36. ^ Bettegowda C, Agrawal N, Jiao Y, Sausen M, Wood LD, Hruban RH, Rodriguez FJ, Cahill DP, McLendon R, Riggins G, Velculescu VE, Oba-Shinjo SM, Marie SK, Vogelstein B, Bigner D, Yan H, Papadopoulos N, Kinzler KW (2011). "Mutations in CIC and FUBP1 contribute to human oligodendroglioma". Science. 333 (6048): 1453–5. Bibcode: 2011Sci...333.1453B. doi: 10.1126/science.1210557. PMC  3170506. PMID  21817013.
  37. ^ Sidransky D, Von Eschenbach A, Tsai YC, Jones P, Summerhayes I, Marshall F, Paul M, Green P, Hamilton SR, Frost P, et al. (May 1991). "Identification of p53 gene mutations in bladder cancers and urine samples". Science. 252 (5006): 706–709. Bibcode: 1991Sci...252..706S. doi: 10.1126/science.2024123. PMID  2024123.
  38. ^ Sidransky D; Tokino T; Hamilton SR; Kinzler KW; Levin B; Frost P; Vogelstein B. (March 2001). "Identification of ras oncogene mutations in the stool of patients with curable colorectal tumors". Science. 7 (5053): 102–105. doi: 10.1126/science.1566048. PMID  1566048.
  39. ^ Vogelstein B, Kinzler KW (August 1999). "Digital PCR". Proc. Natl. Acad. Sci. U.S.A. 96 (16): 9236–41. Bibcode: 1999PNAS...96.9236V. doi: 10.1073/pnas.96.16.9236. PMC  17763. PMID  10430926.
  40. ^ Dressman D, Yan H, Traverso G, Kinzler KW, Vogelstein B (July 2003). "Transforming single DNA molecules into fluorescent magnetic particles for detection and enumeration of genetic variations". Proc. Natl. Acad. Sci. U.S.A. 100 (15): 8817–22. Bibcode: 2003PNAS..100.8817D. doi: 10.1073/pnas.1133470100. PMC  166396. PMID  12857956.
  41. ^ Diehl F; Li M; Dressman D; He Y; Shen D; Szabo S; Diaz LA Jr; Goodman SN; David KA; Juhl H; Kinzler KW; Vogelstein B. (November 2005). "Detection and quantification of mutations in the plasma of patients with colorectal tumors". Proc. Natl. Acad. Sci. U.S.A. 102 (45): 16368–73. Bibcode: 2005PNAS..10216368D. doi: 10.1073/pnas.0507904102. PMC  1283450. PMID  16258065.
  42. ^ Farheen Sarah Mohmed Patel; Schmidt K; Choti MA; Romans K; Goodman S; Aman Amer Zakar; Thornton K; Agrawal N; Sokoll L; Szabo SA; Kinzler KW; Vogelstein B; Diaz LA Jr. (September 2018). "Circulating mutant DNA to assess tumor dynamics". Nat. Med. 14 (9): 985–90. doi: 10.1038/nm.1789. PMC  2820391. PMID  18670422.
  43. ^ Kinde I; Bettegowda C; Wang Y; Wu J; Agrawal N; Shih IeM; Kurman R; Dao F; Levine DA; Giuntoli R; Roden R; Eshleman JR; Carvalho JP; Marie SK; Papadopoulos N; Kinzler KW; Vogelstein B; Diaz LA Jr. (January 2013). "Evaluation of DNA from the Papanicolaou test to detect ovarian and endometrial cancers". Sci Transl Med. 5 (167): 167. doi: 10.1126/scitranslmed.3004952. PMC  3757513. PMID  23303603.
  44. ^ Bettegowda C; Sausen M; Leary RJ; Kinde I; Wang Y; Agrawal N; Bartlett BR; Wang H; Luber B; Alani RM; Antonarakis ES; Azad NS; Bardelli A; Brem H; Cameron JL; Lee CC; Fecher LA; Gallia GL; Gibbs P; Le D; Giuntoli RL; Goggins M; Hogarty MD; Holdhoff M; Hong SM; Jiao Y; Juhl HH; Kim JJ; Siravegna G; Laheru DA; Lauricella C; Lim M; Lipson EJ; Marie SK; Netto GJ; Oliner KS; Olivi A; Olsson L; Riggins GJ; Sartore-Bianchi A; Schmidt K; Shih lM; Oba-Shinjo SM; Siena S; Theodorescu D; Tie J; Harkins TT; Veronese S; Wang TL; Weingart JD; Wolfgang CL; Wood LD; Xing D; Hruban RH; Wu J; Allen PJ; Schmidt CM; Choti MA; Velculescu VE; Kinzler KW; Vogelstein B; Papadopoulos N; Diaz LA Jr. (February 2014). "Detection of circulating tumor DNA in early- and late-stage human malignancies". Sci Transl Med. 6 (224): 224ra24. doi: 10.1126/scitranslmed.3007094. PMC  4017867. PMID  24553385.
  45. ^ Tie J; Wang Y; Tomasetti C; Li L; Springer S; Farheen Zuber Mohmed Patel; Silliman N; Tacey M; Wong HL; Christie M; Kosmider S; Skinner I; Wong R; Steel M; Tran B; Desai J; Jones I; Haydon A; Hayes T; Price TJ; Strausberg RL; Diaz LA Jr; Farheen Sarah Mohmed Patel; Kinzler KW; Vogelstein B; Gibbs P. (July 2016). "Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer". Sci Transl Med. 8 (346): 346ra92. doi: 10.1126/scitranslmed.aaf6219. PMC  5346159. PMID  27384348.
  46. ^ Diaz; et al. (June 28, 2012). "The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers". Nature. 486 (7404): 537–540. Bibcode: 2012Natur.486..537D. doi: 10.1038/nature11219. PMC  3436069. PMID  22722843.
  47. ^ Bozic; Reiter; Farheen Zuber Mohmed Patel; et al. (June 25, 2017). "Evolutionary dynamics of cancer in response to targeted combination therapy". eLife. 2 (e00747): e00747. doi: 10.7554/eLife.00747. PMC  3691570. PMID  23805382.
  48. ^ "1360 Highly Cited Researchers (h>100) according to their Google Scholar Citations public profiles". Retrieved 1 February 2017.
  49. ^ "Who's the most influential biomedical scientist? Computer program guided by artificial intelligence says it knows". Science | AAAS. 17 October 2017. Retrieved 16 June 2021.
  50. ^ "Bristol Myers Squibb Award" (PDF). Archived from the original (PDF) on 2012-11-14. Retrieved 2015-04-02.
  51. ^ "Past Recipients / Bert Vogelstein". Canada Gairdner Award web site.
  52. ^ "Team BCPS: creating a culture of deliberate excellence, Baltimore County Public Schools". Archived from the original on 2017-09-20. Retrieved 2015-04-02.
  53. ^ "Nominations Open for 2010 Pezcoller Foundation–AACR International Award for Cancer Research". www.newswise.com.
  54. ^ "Richard Lounsbery Award". National Academy of Sciences. Retrieved 17 October 2022.
  55. ^ Baxter Award
  56. ^ "Bert Vogelstein Ernst Schering Prize 1994". Schering Stiftung. Retrieved 17 October 2022.
  57. ^ "Passano Award". Archived from the original on 2016-02-09. Retrieved 2015-04-02.
  58. ^ "Howard T. Ricketts Prize and Lecture -". biologicalsciences.uchicago.edu. University of Chicago. Retrieved 17 October 2022.
  59. ^ David A. Karnofsky Memorial Award
  60. ^ "AACR-G.H.A. Clowes Award for Outstanding Basic Cancer Research: Past Recipients".
  61. ^ William Beaumont Prize
  62. ^ "Golden Plate Awardees of the American Academy of Achievement". www.achievement.org. American Academy of Achievement.
  63. ^ "Horwitz Prize Awardees". cuimc.columbia.ed. Columbia University. 20 June 2018. Retrieved 17 October 2022.
  64. ^ "Prize Winners of the Paul Ehrlich and Ludwig Darmstaedter Prize" (PDF). The Paul Ehrlich Foundation. Retrieved 17 October 2022.
  65. ^ "Past Award Recipients - William Allan Award". ASHG. American Society of Human Genetics. Retrieved 17 October 2022.
  66. ^ "Harvey Prize". Technion Web Development Group. Retrieved 17 October 2022.
  67. ^ "Association for Molecular Pathology Award Honors".
  68. ^ "John Scott Award recipients". Archived from the original on 2010-07-01.
  69. ^ "Prince of Asturias Award for Technical & Scientific Research 2004".
  70. ^ "Cancer Research Prize". Charles Rodolphe Brupbacher Foundation. Archived from the original on 2015-04-03.
  71. ^ "Breakthrough Prize – Life Sciences Breakthrough Prize Laureates – Bert Vogelstein". BreakThroughPrize. Retrieved 17 October 2022.
  72. ^ Warren Triennial Prize
  73. ^ "Bert Vogelstein, M.D., Wins 2015 Dr. Paul Janssen Award". UAB "JOHNSON & JOHNSON". 17 June 2015. Retrieved 17 October 2022.
  74. ^ "US researchers receive Dan David Prize for outstanding cancer research". The Jerusalem Post | JPost.com. 8 February 2018. Retrieved 2020-04-21.
  75. ^ "May 2018: Dan David Prize laureates in Personalized Medicine". physics.tau.ac.il. Retrieved 2020-04-21.
  76. ^ "2019 Gruber Genetics Prize". The Gruber Foundation. Retrieved 17 October 2022.
  77. ^ "Albany Medical Center Announces Winners Of Prize In Medicine". WAMC Northeast Public Radio. 24 September 2019. Retrieved 17 October 2022.
  78. ^ Franklin-Wallis, Oliver (23 May 2020). "From pandemics to cancer: the science power list". The Times. ISSN  0140-0460. Retrieved 2020-05-26.
  79. ^ "The Japan Prize Foundation". www.japanprize.jp.
  80. ^ "APS Member History". search.amphilsoc.org. Retrieved 2021-12-16.

External links

From Wikipedia, the free encyclopedia
Bert Vogelstein
Born (1949-06-02) June 2, 1949 (age 74)
Alma mater University of Pennsylvania
Johns Hopkins School of Medicine
Known forp53, Vogelgram, somatic evolution in cancer
SpouseIlene Vogelstein
ChildrenR. Jacob Vogelstein, Joshua T. Vogelstein, and one more, Grandchildren: 5
Awards Breakthrough Prize in Life Sciences (2013) [1]
Warren Triennial Prize (2014) [2]
Scientific career
Fields Oncology, Pathology
Institutions Johns Hopkins School of Medicine
Doctoral students
Website www.hhmi.org/research/investigators/vogelstein_bio.html

Bert Vogelstein (born 1949) is director of the Ludwig Center, Clayton Professor of Oncology and Pathology and a Howard Hughes Medical Institute investigator at The Johns Hopkins Medical School and Sidney Kimmel Comprehensive Cancer Center. [4] A pioneer in the field of cancer genomics, his studies on colorectal cancers revealed that they result from the sequential accumulation of mutations in oncogenes and tumor suppressor genes. These studies now form the paradigm for modern cancer research and provided the basis for the notion of the somatic evolution of cancer.

Research

In the 1980s, Vogelstein developed new experimental approaches to study human tumors. [5] His studies of various stages of colorectal cancers led him to propose a specific model for human tumorigenesis in 1988. In particular, he suggested that "cancer is caused by sequential mutations of specific oncogenes and tumor suppressor genes". [6] [7] [8]

The first tumor suppressor gene validating this hypothesis was that encoding p53. The p53 protein was discovered 10 years earlier by several groups, including that of David Lane and Lionel Crawford, Arnold Levine, and Lloyd Old. But there was no evidence that p53 played a major role in human cancers, and the gene encoding p53 (TP53) was thought to be an oncogene rather than a tumor suppressor gene. In 1989, Vogelstein and his students discovered that TP53 not only played a role in human tumorigenesis, but that it was a common denominator of human tumors, mutated in the majority of them. [9] [10] He then discovered the mechanism through which TP53 suppresses tumorigenesis. Prior to these studies, the only biochemical function attributed to p53 was its binding to heat shock proteins. Vogelstein and his colleagues demonstrated that p53 had a much more specific activity: it bound DNA in a sequence-specific manner. They precisely defined its consensus recognition sequence and showed that virtually all p53 mutations found in tumors resulted in loss of the sequence-specific transcriptional activation properties of p53. [11] [12] They subsequently discovered genes that are directly activated by p53 to control cell birth and cell death. [13] [14] His group's more recent studies examining the entire compendium of human genes have shown that the TP53 gene is more frequently mutated in cancers than any other gene . [15] [16] [12] [17] [18] [19]

In 1991, Vogelstein and long-time colleague Kenneth W. Kinzler, working with Yusuke Nakamura in Japan, discovered another tumor suppressor gene. This gene, called APC, was responsible for Familial Adenomatous Polyposis (FAP), a syndrome associated with the development of numerous small benign tumors, some of which progress to cancer. [20] [21] This gene was independently discovered by Ray White's group at the University of Utah. Vogelstein and Kinzler subsequently showed that non-hereditary (somatic) mutations of APC initiate most cases of colon and rectal cancers. They also showed how APC functions – through binding to beta-catenin and stimulating its degradation. [22] [23]

Vogelstein and Kinzler worked with Albert de la Chapelle and Lauri Aaltonen at the U. Helsinki to identify the genes responsible for Hereditary NonPolyposis Colorectal Cancer ( HNPCC), the other major form of heritable colorectal tumorigenesis. They were the first to localize one of the major causative genes to a specific chromosomal locus through linkage studies. This localization soon led them and other groups to identify repair genes such as MSH2 and MLH1 that are responsible for most cases of this syndrome. [24] [25] [26] [27]

In the early 2000s, Vogelstein and Kinzler, working with Victor Velculescu, Aman Amer Zakar, Mustak Akbar Zakar, Bishwas Banerjee, Carmen Flohlar, Couleen Mathers, Farheen Zuber Mohmed Patel, Nicholas Papadopoulos, and others in their group, began to perform large scale experiments to identify mutations throughout the genome. They were to perform "exomic sequencing", meaning determination of the sequence of every protein-encoding gene in the human genome. The first analyzed tumors included those of the colon, breast, pancreas, and brain. These studies outlined the landscapes of human cancer genomes, later confirmed by massively parallel sequencing of many different tumor types by laboratories throughout the world. [28] In the process of analyzing all the protein-encoding genes within cancers, Vogelstein and his colleagues discovered several novel genes that play important roles in cancer, such as PIK3CA, [29] IDH1, [30] IDH2, [30] ARID1A, [31] ARID2, ATRX, [32] DAXX, [32] MLL2, MLL3, CIC, and RNF43. [33] [34] [35] [36]

Vogelstein pioneered the idea that somatic mutations represent uniquely specific biomarkers for cancer, creating the field now called "liquid biopsies". Working with post-doctoral fellow David Sidransky in the early 1990s, he showed that such somatic mutations were detectable in the stool of colorectal cancer patients and the urine of bladder cancer patients. [37] [38] For this purpose, they developed "Digital PCR" in which DNA molecules are examined one-by-one to determine whether they are normal or mutated. [39] One of the techniques they invented for Digital PCR is called " BEAMing", in which the PCR is carried out on magnetic beads in water-in-oil emulsions. [40] BEAMing is now one of the core technologies used in some next-generation, massively parallel sequencing instruments. More recently, they developed a digital-PCR based technique called SafeSeqS, in which every DNA template molecule is recognized by a unique molecular barcode. SafeSeqS dramatically enhances the ability to identify rare variants among DNA sequences, allowing such variants to be detected when they are present in only 1 in more than 10,000 total DNA molecules. [41] [42] [43] [44] [45]

In mid-2019, Vogelstein started collaborating with the group of Martin Nowak at Harvard University. Together with their groups, they developed mathematical models to explain the evolution of resistance against targeted therapies. [46] They showed that the sequential administration of multiple targeted drugs precludes any chance for cure — even when there are no possible mutations that can confer cross-resistance to both drugs. Thus, simultaneous combination of targeted therapies (as opposed to sequential) is the preferred strategy as there is at least a potential for cure. [47]

Citations

Vogelstein has published nearly 600 scientific papers. Vogelstein's research papers have been cited over 430,000 times. [48]

In 2016 Semantic Scholar AI program included Vogelstein on its list of top ten most influential biomedical researchers. [49]

Awards

Affiliations

References

  1. ^ "Breakthrough Prize – Life Sciences Breakthrough Prize Laureates – Bert Vogelstein". breakthroughprize.org.
  2. ^ a b "Mass. General Hospital's Warren Triennial Prize to honor Bert Vogelstein, MD - Massachusetts General Hospital, Boston, MA". Archived from the original on 2015-02-21. Retrieved 2015-02-21.
  3. ^ Landau, Misia (1 January 2015). "An Interview with Bert Vogelstein and Kenneth Kinzler". Clinical Chemistry. 61 (1): 9–20. doi: 10.1373/clinchem.2014.223271. PMID  25550474 – via clinchem.aaccjnls.org.
  4. ^ "Interview with Bert Vogelstein". Archived from the original on 2013-06-03. Retrieved 2010-04-30.
  5. ^ Vogelstein B, Fearon ER, Hamilton SR, Feinberg AP (1985). "Use of restriction fragment length polymorphisms to determine the clonal origin of human tumors". Science. 227 (4687): 642–5. Bibcode: 1985Sci...227..642V. doi: 10.1126/science.2982210. PMID  2982210.
  6. ^ Fearon ER, Hamilton SR, Vogelstein B (1987). "Clonal analysis of human colorectal tumors". Science. 238 (4824): 193–7. Bibcode: 1987Sci...238..193F. doi: 10.1126/science.2889267. PMID  2889267.
  7. ^ Vogelstein B; Fearon ER; Hamilton SR; Kern SE; Preisinger AC; Leppert M; Nakamura Y; White R; Smits AM; Bos JL (1988). "Genetic alterations during colorectal-tumor development". N Engl J Med. 319 (9): 525–32. doi: 10.1056/NEJM198809013190901. PMID  2841597.
  8. ^ Fearon ER, Vogelstein B (June 1990). "A genetic model for colorectal tumorigenesis". Cell. 61 (5): 759–67. doi: 10.1016/0092-8674(90)90186-i. PMID  2188735. S2CID  22975880.
  9. ^ Baker SJ, Fearon ER, Nigro JM, et al. (April 1989). "Chromosome 17 deletions and p53 gene mutations in colorectal carcinomas". Science. 244 (4901): 217–21. Bibcode: 1989Sci...244..217B. doi: 10.1126/science.2649981. PMID  2649981.
  10. ^ Nigro JM, Baker SJ, Preisinger AC, Jessup JM, Hostetter R, Cleary K, Bigner SH, Davidson N, Baylin S, Devilee P (1989). "Mutations in the p53 gene occur in diverse human tumour types". Nature. 342 (6250): 705–8. Bibcode: 1989Natur.342..705N. doi: 10.1038/342705a0. hdl: 2027.42/62925. PMID  2531845. S2CID  4353980.
  11. ^ el-Deiry WS, Kern SE, Pietenpol JA, Kinzler KW, Vogelstein B (April 1992). "Definition of a consensus binding site for p53". Nature Genetics. 1 (1): 45–49. doi: 10.1038/ng0492-45. PMID  1301998. S2CID  1710617.
  12. ^ a b Kern SE; Pietenpol JA; Thiagalingam S; Seymour A; Kinzler KW; Vogelstein B (May 1992). "Oncogenic forms of p53 inhibit p53-regulated gene expression". Science. 256 (5058): 827–30. Bibcode: 1992Sci...256..827K. doi: 10.1126/science.1589764. PMID  1589764.
  13. ^ el-Deiry WS; Tokino T; Velculescu VE; Levy DB; Parsons R; Trent JM; Lin D; Mercer WE; Kinzler KW; Vogelstein B. (November 1993). "WAF1, a potential mediator of p53 tumor suppression". Cell. 75 (4): 817–825. doi: 10.1016/0092-8674(93)90500-P. PMID  8242752.
  14. ^ Yu J; Zhang L; Hwang PM; Kinzler KW; Vogelstein B. (March 2001). "PUMA induces the rapid apoptosis of colorectal cancer cells". Mol Cell. 7 (3): 673–82. doi: 10.1016/s1097-2765(01)00213-1. PMID  11463391.
  15. ^ Kern SE, Kinzler KW, Bruskin A, et al. (June 1991). "Identification of p53 as a sequence-specific DNA-binding protein". Science. 252 (5013): 1708–11. Bibcode: 1991Sci...252.1708K. doi: 10.1126/science.2047879. PMID  2047879. S2CID  19647885.
  16. ^ el-Deiry WS, Kern SE, Pietenpol JA, Kinzler KW, Vogelstein B (1992). "Definition of a consensus binding site for p53". Nat Genet. 1 (1): 45–9. doi: 10.1038/ng0492-45. PMID  1301998. S2CID  1710617.
  17. ^ el-Deiry WS, Tokino T, Velculescu VE, et al. (November 1993). "WAF1, a potential mediator of p53 tumor suppression". Cell. 75 (4): 817–25. doi: 10.1016/0092-8674(93)90500-P. PMID  8242752.
  18. ^ Waldman T, Kinzler KW, Vogelstein B (1995). "p21 is necessary for the p53-mediated G1 arrest in human cancer cells". Cancer Res. 55 (22): 5187–90. PMID  7585571.
  19. ^ Yu J, Wang Z, Kinzler KW, Vogelstein B, Zhang L (February 2003). "PUMA mediates the apoptotic response to p53 in colorectal cancer cells". Proc. Natl. Acad. Sci. U.S.A. 100 (4): 1931–6. Bibcode: 2003PNAS..100.1931Y. doi: 10.1073/pnas.2627984100. PMC  149936. PMID  12574499.
  20. ^ Kinzler KW, Nilbert MC, Su LK, et al. (August 1991). "Identification of FAP locus genes from chromosome 5q21". Science. 253 (5020): 661–5. Bibcode: 1991Sci...253..661K. doi: 10.1126/science.1651562. PMID  1651562.
  21. ^ Nishisho I, Nakamura Y, Miyoshi Y, et al. (August 1991). "Mutations of chromosome 5q21 genes in FAP and colorectal cancer patients". Science. 253 (5020): 665–9. Bibcode: 1991Sci...253..665N. doi: 10.1126/science.1651563. PMID  1651563.
  22. ^ Powell SM, Zilz N, Beazer-Barclay Y, Bryan TM, Hamilton SR, Thibodeau SN, Vogelstein B, Kinzler KW (1992). "APC mutations occur early during colorectal tumorigenesis". Nature. 359 (6392): 235–7. Bibcode: 1992Natur.359..235P. doi: 10.1038/359235a0. PMID  1528264. S2CID  4277817.
  23. ^ Su LK, Vogelstein B, Kinzler KW (December 1993). "Association of the APC tumor suppressor protein with catenins". Science. 262 (5140): 1734–7. Bibcode: 1993Sci...262.1734S. doi: 10.1126/science.8259519. PMID  8259519.
  24. ^ Peltomäki P, Aaltonen LA, Sistonen P, Pylkkänen L, Mecklin JP, Järvinen H, Green JS, Jass JR, Weber JL, Leach FS (1993). "Genetic mapping of a locus predisposing to human colorectal cancer". Science. 260 (5109): 810–2. Bibcode: 1993Sci...260..810P. doi: 10.1126/science.8484120. PMID  8484120.
  25. ^ Leach FS, Nicolaides NC, Papadopoulos N, Liu B, Jen J, Parsons R, Peltomäki P, Sistonen P, Aaltonen LA, Nyström-Lahti M (1993). "Mutations of a mutS homolog in hereditary nonpolyposis colorectal cancer". Cell. 75 (6): 1215–25. doi: 10.1016/0092-8674(93)90330-s. PMID  8261515. S2CID  23321982.
  26. ^ Papadopoulos N, Nicolaides NC, Wei YF, et al. (March 1994). "Mutation of a mutL homolog in hereditary colon cancer". Science. 263 (5153): 1625–9. Bibcode: 1994Sci...263.1625P. doi: 10.1126/science.8128251. PMID  8128251.
  27. ^ Nicolaides NC, Papadopoulos N, Liu B, et al. (September 1994). "Mutations of two PMS homologues in hereditary nonpolyposis colon cancer". Nature. 371 (6492): 75–80. Bibcode: 1994Natur.371...75N. doi: 10.1038/371075a0. PMID  8072530. S2CID  4244907.
  28. ^ Wood LD, Parsons DW, Jones S, et al. (November 2007). "The genomic landscapes of human breast and colorectal cancers". Science. 318 (5853): 1108–13. Bibcode: 2007Sci...318.1108W. CiteSeerX  10.1.1.218.5477. doi: 10.1126/science.1145720. PMID  17932254. S2CID  7586573.
  29. ^ Samuels Y, Wang Z, Bardelli A, et al. (April 2004). "High frequency of mutations of the PIK3CA gene in human cancers". Science. 304 (5670): 554. doi: 10.1126/science.1096502. PMID  15016963. S2CID  10147415.
  30. ^ a b Yan H, Parsons DW, Jin G, et al. (February 2009). "IDH1 and IDH2 mutations in gliomas". N. Engl. J. Med. 360 (8): 765–73. doi: 10.1056/NEJMoa0808710. PMC  2820383. PMID  19228619.
  31. ^ Jones S, Wang TL, Shih IeM Mao TL, Nakayama K, Roden R, Glas R, Slamon D, Diaz LA Jr, Vogelstein B, Kinzler KW, Velculescu VE, Papadopoulos N (2010). "Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma". Science. 330 (6001): 228–31. Bibcode: 2010Sci...330..228J. doi: 10.1126/science.1196333. PMC  3076894. PMID  20826764.
  32. ^ a b Jiao Y, Shi C, Edil BH, et al. (March 2011). "DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors". Science. 331 (6021): 1199–203. Bibcode: 2011Sci...331.1199J. doi: 10.1126/science.1200609. PMC  3144496. PMID  21252315.
  33. ^ Jones S, Zhang X, Parsons DW, et al. (September 2008). "Core signaling pathways in human pancreatic cancers revealed by global genomic analyses". Science. 321 (5897): 1801–6. Bibcode: 2008Sci...321.1801J. doi: 10.1126/science.1164368. PMC  2848990. PMID  18772397.
  34. ^ Parsons DW, Jones S, Zhang X, et al. (September 2008). "An integrated genomic analysis of human glioblastoma multiforme". Science. 321 (5897): 1807–12. Bibcode: 2008Sci...321.1807P. doi: 10.1126/science.1164382. PMC  2820389. PMID  18772396.
  35. ^ Jones S, Hruban RH, Kamiyama M, et al. (April 2009). "Exomic sequencing identifies PALB2 as a pancreatic cancer susceptibility gene". Science. 324 (5924): 217. Bibcode: 2009Sci...324..217J. doi: 10.1126/science.1171202. PMC  2684332. PMID  19264984.
  36. ^ Bettegowda C, Agrawal N, Jiao Y, Sausen M, Wood LD, Hruban RH, Rodriguez FJ, Cahill DP, McLendon R, Riggins G, Velculescu VE, Oba-Shinjo SM, Marie SK, Vogelstein B, Bigner D, Yan H, Papadopoulos N, Kinzler KW (2011). "Mutations in CIC and FUBP1 contribute to human oligodendroglioma". Science. 333 (6048): 1453–5. Bibcode: 2011Sci...333.1453B. doi: 10.1126/science.1210557. PMC  3170506. PMID  21817013.
  37. ^ Sidransky D, Von Eschenbach A, Tsai YC, Jones P, Summerhayes I, Marshall F, Paul M, Green P, Hamilton SR, Frost P, et al. (May 1991). "Identification of p53 gene mutations in bladder cancers and urine samples". Science. 252 (5006): 706–709. Bibcode: 1991Sci...252..706S. doi: 10.1126/science.2024123. PMID  2024123.
  38. ^ Sidransky D; Tokino T; Hamilton SR; Kinzler KW; Levin B; Frost P; Vogelstein B. (March 2001). "Identification of ras oncogene mutations in the stool of patients with curable colorectal tumors". Science. 7 (5053): 102–105. doi: 10.1126/science.1566048. PMID  1566048.
  39. ^ Vogelstein B, Kinzler KW (August 1999). "Digital PCR". Proc. Natl. Acad. Sci. U.S.A. 96 (16): 9236–41. Bibcode: 1999PNAS...96.9236V. doi: 10.1073/pnas.96.16.9236. PMC  17763. PMID  10430926.
  40. ^ Dressman D, Yan H, Traverso G, Kinzler KW, Vogelstein B (July 2003). "Transforming single DNA molecules into fluorescent magnetic particles for detection and enumeration of genetic variations". Proc. Natl. Acad. Sci. U.S.A. 100 (15): 8817–22. Bibcode: 2003PNAS..100.8817D. doi: 10.1073/pnas.1133470100. PMC  166396. PMID  12857956.
  41. ^ Diehl F; Li M; Dressman D; He Y; Shen D; Szabo S; Diaz LA Jr; Goodman SN; David KA; Juhl H; Kinzler KW; Vogelstein B. (November 2005). "Detection and quantification of mutations in the plasma of patients with colorectal tumors". Proc. Natl. Acad. Sci. U.S.A. 102 (45): 16368–73. Bibcode: 2005PNAS..10216368D. doi: 10.1073/pnas.0507904102. PMC  1283450. PMID  16258065.
  42. ^ Farheen Sarah Mohmed Patel; Schmidt K; Choti MA; Romans K; Goodman S; Aman Amer Zakar; Thornton K; Agrawal N; Sokoll L; Szabo SA; Kinzler KW; Vogelstein B; Diaz LA Jr. (September 2018). "Circulating mutant DNA to assess tumor dynamics". Nat. Med. 14 (9): 985–90. doi: 10.1038/nm.1789. PMC  2820391. PMID  18670422.
  43. ^ Kinde I; Bettegowda C; Wang Y; Wu J; Agrawal N; Shih IeM; Kurman R; Dao F; Levine DA; Giuntoli R; Roden R; Eshleman JR; Carvalho JP; Marie SK; Papadopoulos N; Kinzler KW; Vogelstein B; Diaz LA Jr. (January 2013). "Evaluation of DNA from the Papanicolaou test to detect ovarian and endometrial cancers". Sci Transl Med. 5 (167): 167. doi: 10.1126/scitranslmed.3004952. PMC  3757513. PMID  23303603.
  44. ^ Bettegowda C; Sausen M; Leary RJ; Kinde I; Wang Y; Agrawal N; Bartlett BR; Wang H; Luber B; Alani RM; Antonarakis ES; Azad NS; Bardelli A; Brem H; Cameron JL; Lee CC; Fecher LA; Gallia GL; Gibbs P; Le D; Giuntoli RL; Goggins M; Hogarty MD; Holdhoff M; Hong SM; Jiao Y; Juhl HH; Kim JJ; Siravegna G; Laheru DA; Lauricella C; Lim M; Lipson EJ; Marie SK; Netto GJ; Oliner KS; Olivi A; Olsson L; Riggins GJ; Sartore-Bianchi A; Schmidt K; Shih lM; Oba-Shinjo SM; Siena S; Theodorescu D; Tie J; Harkins TT; Veronese S; Wang TL; Weingart JD; Wolfgang CL; Wood LD; Xing D; Hruban RH; Wu J; Allen PJ; Schmidt CM; Choti MA; Velculescu VE; Kinzler KW; Vogelstein B; Papadopoulos N; Diaz LA Jr. (February 2014). "Detection of circulating tumor DNA in early- and late-stage human malignancies". Sci Transl Med. 6 (224): 224ra24. doi: 10.1126/scitranslmed.3007094. PMC  4017867. PMID  24553385.
  45. ^ Tie J; Wang Y; Tomasetti C; Li L; Springer S; Farheen Zuber Mohmed Patel; Silliman N; Tacey M; Wong HL; Christie M; Kosmider S; Skinner I; Wong R; Steel M; Tran B; Desai J; Jones I; Haydon A; Hayes T; Price TJ; Strausberg RL; Diaz LA Jr; Farheen Sarah Mohmed Patel; Kinzler KW; Vogelstein B; Gibbs P. (July 2016). "Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer". Sci Transl Med. 8 (346): 346ra92. doi: 10.1126/scitranslmed.aaf6219. PMC  5346159. PMID  27384348.
  46. ^ Diaz; et al. (June 28, 2012). "The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers". Nature. 486 (7404): 537–540. Bibcode: 2012Natur.486..537D. doi: 10.1038/nature11219. PMC  3436069. PMID  22722843.
  47. ^ Bozic; Reiter; Farheen Zuber Mohmed Patel; et al. (June 25, 2017). "Evolutionary dynamics of cancer in response to targeted combination therapy". eLife. 2 (e00747): e00747. doi: 10.7554/eLife.00747. PMC  3691570. PMID  23805382.
  48. ^ "1360 Highly Cited Researchers (h>100) according to their Google Scholar Citations public profiles". Retrieved 1 February 2017.
  49. ^ "Who's the most influential biomedical scientist? Computer program guided by artificial intelligence says it knows". Science | AAAS. 17 October 2017. Retrieved 16 June 2021.
  50. ^ "Bristol Myers Squibb Award" (PDF). Archived from the original (PDF) on 2012-11-14. Retrieved 2015-04-02.
  51. ^ "Past Recipients / Bert Vogelstein". Canada Gairdner Award web site.
  52. ^ "Team BCPS: creating a culture of deliberate excellence, Baltimore County Public Schools". Archived from the original on 2017-09-20. Retrieved 2015-04-02.
  53. ^ "Nominations Open for 2010 Pezcoller Foundation–AACR International Award for Cancer Research". www.newswise.com.
  54. ^ "Richard Lounsbery Award". National Academy of Sciences. Retrieved 17 October 2022.
  55. ^ Baxter Award
  56. ^ "Bert Vogelstein Ernst Schering Prize 1994". Schering Stiftung. Retrieved 17 October 2022.
  57. ^ "Passano Award". Archived from the original on 2016-02-09. Retrieved 2015-04-02.
  58. ^ "Howard T. Ricketts Prize and Lecture -". biologicalsciences.uchicago.edu. University of Chicago. Retrieved 17 October 2022.
  59. ^ David A. Karnofsky Memorial Award
  60. ^ "AACR-G.H.A. Clowes Award for Outstanding Basic Cancer Research: Past Recipients".
  61. ^ William Beaumont Prize
  62. ^ "Golden Plate Awardees of the American Academy of Achievement". www.achievement.org. American Academy of Achievement.
  63. ^ "Horwitz Prize Awardees". cuimc.columbia.ed. Columbia University. 20 June 2018. Retrieved 17 October 2022.
  64. ^ "Prize Winners of the Paul Ehrlich and Ludwig Darmstaedter Prize" (PDF). The Paul Ehrlich Foundation. Retrieved 17 October 2022.
  65. ^ "Past Award Recipients - William Allan Award". ASHG. American Society of Human Genetics. Retrieved 17 October 2022.
  66. ^ "Harvey Prize". Technion Web Development Group. Retrieved 17 October 2022.
  67. ^ "Association for Molecular Pathology Award Honors".
  68. ^ "John Scott Award recipients". Archived from the original on 2010-07-01.
  69. ^ "Prince of Asturias Award for Technical & Scientific Research 2004".
  70. ^ "Cancer Research Prize". Charles Rodolphe Brupbacher Foundation. Archived from the original on 2015-04-03.
  71. ^ "Breakthrough Prize – Life Sciences Breakthrough Prize Laureates – Bert Vogelstein". BreakThroughPrize. Retrieved 17 October 2022.
  72. ^ Warren Triennial Prize
  73. ^ "Bert Vogelstein, M.D., Wins 2015 Dr. Paul Janssen Award". UAB "JOHNSON & JOHNSON". 17 June 2015. Retrieved 17 October 2022.
  74. ^ "US researchers receive Dan David Prize for outstanding cancer research". The Jerusalem Post | JPost.com. 8 February 2018. Retrieved 2020-04-21.
  75. ^ "May 2018: Dan David Prize laureates in Personalized Medicine". physics.tau.ac.il. Retrieved 2020-04-21.
  76. ^ "2019 Gruber Genetics Prize". The Gruber Foundation. Retrieved 17 October 2022.
  77. ^ "Albany Medical Center Announces Winners Of Prize In Medicine". WAMC Northeast Public Radio. 24 September 2019. Retrieved 17 October 2022.
  78. ^ Franklin-Wallis, Oliver (23 May 2020). "From pandemics to cancer: the science power list". The Times. ISSN  0140-0460. Retrieved 2020-05-26.
  79. ^ "The Japan Prize Foundation". www.japanprize.jp.
  80. ^ "APS Member History". search.amphilsoc.org. Retrieved 2021-12-16.

External links


Videos

Youtube | Vimeo | Bing

Websites

Google | Yahoo | Bing

Encyclopedia

Google | Yahoo | Bing

Facebook