Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jukka Pekka Mecklin is active.

Publication


Featured researches published by Jukka Pekka Mecklin.


Cell | 1993

Mutations of a mutS homolog in hereditary nonpolyposis colorectal cancer

Fredrick S. Leach; Nicholas C. Nicolaides; Nickolas Papadopoulos; Bo Liu; Jin Jen; Ramon Parsons; Päivi Peltomäki; Pertti Sistonen; Lauri A. Aaltonen; Minna Nyström-Lahti; Xin Yuan Guan; Ji Zhang; Paul S. Meltzer; Jing Wei Yu; Fa Ten Kao; David J. Chen; Karen M. Cerosaletti; R. E. Keith Fournier; Sean Todd; Tracey Lewis; Robin J. Leach; Susan L. Naylor; Jean Weissenbach; Jukka Pekka Mecklin; Heikki Järvinen; Gloria M. Petersen; Stanley R. Hamilton; Jane Green; Jeremy R. Jass; Patrice Watson

Recent studies have shown that a locus responsible for hereditary nonpolyposis colorectal cancer (HNPCC) is on chromosome 2p and that tumors developing in these patients contain alterations in microsatellite sequences (RER+ phenotype). We have used chromosome microdissection to obtain highly polymorphic markers from chromosome 2p16. These and other markers were ordered in a panel of somatic cell hybrids and used to define a 0.8 Mb interval containing the HNPCC locus. Candidate genes were then mapped, and one was found to lie within the 0.8 Mb interval. We identified this candidate by virtue of its homology to mutS mismatch repair genes. cDNA clones were obtained and the sequence used to detect germline mutations, including those producing termination codons, in HNPCC kindreds. Somatic as well as germline mutations of the gene were identified in RER+ tumor cells. This mutS homolog is therefore likely to be responsible for HNPCC.


Gastroenterology | 1999

New clinical criteria for hereditary nonpolyposis colorectal cancer (HNPCC, Lynch syndrome) proposed by the International Collaborative Group on HNPCC ☆

Hans F. A. Vasen; Patrice Watson; Jukka Pekka Mecklin; Henry T. Lynch

One of the first successful efforts of the International Collaborative Group on HNPCC (ICG-HNPCC) at its meeting in Amsterdam in 1990 was the establishment of a set of selection criteria for families with hereditary nonpolyposis colorectal cancer (HNPCC, Lynch syndrome) to provide a basis for uniformity in collaborative studies (Table 1).1 Until 1990, such criteria were lacking and the descriptions of the syndrome in the literature varied widely. In the absence of a common nomenclature it was impossible to compare the results of different studies on HNPCC. The wide acceptance of the ICG criteria is reflected by the fact that almost all investigators currently make use of them. However, the criteria have also been criticized. Some investigators feel that the criteria exclude some classic HNPCC families because they do not take into account the extracolonic cancers that are part of the syndrome. There is also concern that the criteria are being used inappropriately for the diagnosis of HNPCC. As criteria for the selection of families for research, they were originally aimed at specificity more than sensitivity. It is now considered that many true HNPCC families would be missed if the criteria are applied to clinical diagnosis, and that families not meeting the criteria might be falsely reassured and excluded from genetic counseling, DNA testing, or surveillance. To resolve these problems, many investigators have developed additional criteria.2–6 At the ninth meeting of the ICG-HNPCC held in Noordwijk, the Netherlands, in 1997, a definition of HNPCC was proposed that was aimed at helping clinicians to identify families. This provided a detailed description of the specific features of HNPCC rather than the rigid criteria required for collaborative studies. In addition, at the ninth meeting, and also at the tenth held in Coimbra, Portugal, in 1998, new selection criteria for collaborative studies were proposed that included the extracolonic cancers associated with HNPCC. Definition of HNPCC


Gastroenterology | 2000

Controlled 15-year trial on screening for colorectal cancer in families with hereditary nonpolyposis colorectal cancer

Heikki Järvinen; Markku Aarnio; Katja Aktan–Collan‡; Lauri A. Aaltonen; Päivi Peltomäki; Albert de la Chapelle; Jukka Pekka Mecklin

BACKGROUND & AIMSnIdentification of the hereditary nonpolyposis colorectal cancer (HNPCC) syndrome enables prevention of colorectal cancer (CRC) by means of colonoscopy and polypectomies. We evaluated the efficacy of screening in a controlled trial over 15 years.nnnMETHODSnIncidence of CRC and survival were compared in 2 cohorts of at-risk members of 22 families with HNPCC. Colonic screening at 3-year intervals was arranged for 133 subjects; 119 control subjects had no screening. Genetic testing was offered to subjects in whose families the causative mutation was known.nnnRESULTSnCRC developed in 8 screened subjects (6%) compared with 19 control subjects (16 %; P = 0.014). The CRC rate was reduced by 62%. In mutation-positive subjects alone, the CRC rates were 18% in screened subjects and 41% in controls (P = 0.02). The decrease resulted from the removal of adenomas in 13 mutation-positive individuals (30%) and in 6 subjects with unknown mutation status (40%). All CRCs in the study group were local, causing no deaths, compared with 9 deaths caused by CRC in the controls. The overall death rates were 10 vs. 26 subjects in the study and control groups (P = 0.003), 4 vs. 12 in mutation-positive subjects (P = 0.05).nnnCONCLUSIONSnColonoscopic screening at 3-year intervals more than halves the risk of CRC, prevents CRC deaths, and decreases overall mortality by about 65% in HNPCC families.


The Lancet | 2011

Long-term effect of aspirin on cancer risk in carriers of hereditary colorectal cancer: an analysis from the CAPP2 randomised controlled trial

John Burn; Anne-Marie Gerdes; Finlay Macrae; Jukka Pekka Mecklin; Gabriela Moeslein; Sylviane Olschwang; D. Eccles; D. Gareth Evans; Eamonn R. Maher; Lucio Bertario; Marie Luise Bisgaard; Malcolm G. Dunlop; Judy W. C. Ho; Shirley Hodgson; Annika Lindblom; Jan Lubinski; Patrick J. Morrison; Victoria Murday; Raj Ramesar; Lucy Side; Rodney J. Scott; Huw Thomas; Hans F. A. Vasen; Gail Barker; Gillian Crawford; Faye Elliott; Mohammad Movahedi; Kirsi Pylvänäinen; Juul T. Wijnen; Riccardo Fodde

Summary Background Observational studies report reduced colorectal cancer in regular aspirin consumers. Randomised controlled trials have shown reduced risk of adenomas but none have employed prevention of colorectal cancer as a primary endpoint. The CAPP2 trial aimed to investigate the antineoplastic effects of aspirin and a resistant starch in carriers of Lynch syndrome, the major form of hereditary colorectal cancer; we now report long-term follow-up of participants randomly assigned to aspirin or placebo. Methods In the CAPP2 randomised trial, carriers of Lynch syndrome were randomly assigned in a two-by-two factorial design to 600 mg aspirin or aspirin placebo or 30 g resistant starch or starch placebo, for up to 4 years. Randomisation was in blocks of 16 with provision for optional single-agent randomisation and extended postintervention double-blind follow-up; participants and investigators were masked to treatment allocation. The primary endpoint was development of colorectal cancer. Analysis was by intention to treat and per protocol. This trial is registered, ISRCTN59521990. Results 861 participants were randomly assigned to aspirin or aspirin placebo. At a mean follow-up of 55·7 months, 48 participants had developed 53 primary colorectal cancers (18 of 427 randomly assigned to aspirin, 30 of 434 to aspirin placebo). Intention-to-treat analysis of time to first colorectal cancer showed a hazard ratio (HR) of 0·63 (95% CI 0·35–1·13, p=0·12). Poisson regression taking account of multiple primary events gave an incidence rate ratio (IRR) of 0·56 (95% CI 0·32–0·99, p=0·05). For participants completing 2 years of intervention (258 aspirin, 250 aspirin placebo), per-protocol analysis yielded an HR of 0·41 (0·19–0·86, p=0·02) and an IRR of 0·37 (0·18–0·78, p=0·008). No data for adverse events were available postintervention; during the intervention, adverse events did not differ between aspirin and placebo groups. Interpretation 600 mg aspirin per day for a mean of 25 months substantially reduced cancer incidence after 55·7 months in carriers of hereditary colorectal cancer. Further studies are needed to establish the optimum dose and duration of aspirin treatment. Funding European Union; Cancer Research UK; Bayer Corporation; National Starch and Chemical Co; UK Medical Research Council; Newcastle Hospitals trustees; Cancer Council of Victoria Australia; THRIPP South Africa; The Finnish Cancer Foundation; SIAK Switzerland; Bayer Pharma.


Nature Genetics | 2009

The common colorectal cancer predisposition SNP rs6983267 at chromosome 8q24 confers potential to enhanced Wnt signaling

Sari Tuupanen; Mikko P. Turunen; Rainer Lehtonen; Outi Hallikas; Sakari Vanharanta; Teemu Kivioja; Mikael Björklund; Gong-Hong Wei; Jian Yan; Iina Niittymäki; Jukka Pekka Mecklin; Heikki Järvinen; Ari Ristimäki; Mariachiara Di-Bernardo; Phil East; Luis Carvajal-Carmona; Richard S. Houlston; Ian Tomlinson; Kimmo Palin; Esko Ukkonen; Auli Karhu; Jussi Taipale; Lauri A. Aaltonen

Homozygosity for the G allele of rs6983267 at 8q24 increases colorectal cancer (CRC) risk ∼1.5 fold. We report here that the risk allele G shows copy number increase during CRC development. Our computer algorithm, Enhancer Element Locator (EEL), identified an enhancer element that contains rs6983267. The element drove expression of a reporter gene in a pattern that is consistent with regulation by the key CRC pathway Wnt. rs6983267 affects a binding site for the Wnt-regulated transcription factor TCF4, with the risk allele G showing stronger binding in vitro and in vivo. Genome-wide ChIP assay revealed the element as the strongest TCF4 binding site within 1 Mb of MYC. An unambiguous correlation between rs6983267 genotype and MYC expression was not detected, and additional work is required to scrutinize all possible targets of the enhancer. Our work provides evidence that the common CRC predisposition associated with 8q24 arises from enhanced responsiveness to Wnt signaling.


American Journal of Human Genetics | 2004

Early-Onset Renal Cell Carcinoma as a Novel Extraparaganglial Component of SDHB-Associated Heritable Paraganglioma

Sakari Vanharanta; Mary Buchta; Sarah R. McWhinney; Sanna K. Virta; Mariola Pęczkowska; Carl Morrison; Rainer Lehtonen; Andrzej Januszewicz; Heikki Järvinen; Matti Juhola; Jukka Pekka Mecklin; Eero Pukkala; Riitta Herva; Maija Kiuru; Nina N. Nupponen; Lauri A. Aaltonen; Hartmut P. H. Neumann; Charis Eng

Hereditary paraganglioma syndrome has recently been shown to be caused by germline heterozygous mutations in three (SDHB, SDHC, and SDHD) of the four genes that encode mitochondrial succinate dehydrogenase. Extraparaganglial component neoplasias have never been previously documented. In a population-based registry of symptomatic presentations of phaeochromocytoma/paraganglioma comprising 352 registrants, among whom 16 unrelated registrants were SDHB mutation positive, one family with germline SDHB mutation c.847-50delTCTC had two members with renal cell carcinoma (RCC), of solid histology, at ages 24 and 26 years. Both also had paraganglioma. A registry of early-onset RCCs revealed a family comprising a son with clear-cell RCC and his mother with a cardiac tumor, both with the germline SDHB R27X mutation. The cardiac tumor proved to be a paraganglioma. All RCCs showed loss of the remaining wild-type allele. Our observations suggest that germline SDHB mutations can predispose to early-onset kidney cancers in addition to paragangliomas and carry implications for medical surveillance.


The New England Journal of Medicine | 2008

Effect of Aspirin or Resistant Starch on Colorectal Neoplasia in the Lynch Syndrome

John Burn; D. Timothy Bishop; Jukka Pekka Mecklin; Finlay Macrae; Gabriela Möslein; Sylviane Olschwang; Marie Luise Bisgaard; Raj Ramesar; Diana Eccles; Eamonn R. Maher; Lucio Bertario; Heikki Järvinen; Annika Lindblom; D. Gareth Evans; Jan Lubinski; Patrick Morrison; Judy W. C. Ho; Hans F. A. Vasen; Lucy Side; Huw Thomas; Rodney J. Scott; Malcolm G. Dunlop; Gail Barker; Faye Elliott; Jeremy R. Jass; Ricardo Fodde; Henry T. Lynch; John C. Mathers

BACKGROUNDnObservational and epidemiologic data indicate that the use of aspirin reduces the risk of colorectal neoplasia; however, the effects of aspirin in the Lynch syndrome (hereditary nonpolyposis colon cancer) are not known. Resistant starch has been associated with an antineoplastic effect on the colon.nnnMETHODSnIn a randomized, placebo-controlled trial, we used a two-by-two design to investigate the effects of aspirin, at a dose of 600 mg per day, and resistant starch (Novelose), at a dose of 30 g per day, in reducing the risk of adenoma and carcinoma among persons with the Lynch syndrome.nnnRESULTSnAmong 1071 persons in 43 centers, 62 were ineligible to participate in the study, 72 did not enter the study, and 191 withdrew from the study. These three categories were equally distributed across the study groups. Over a mean period of 29 months (range, 7 to 74), colonic adenoma or carcinoma developed in 141 participants. Of 693 participants randomly assigned to receive aspirin or placebo, neoplasia developed in 66 participants receiving aspirin (18.9%), as compared with 65 receiving placebo (19.0%) (relative risk, 1.0; 95% confidence interval [CI], 0.7 to 1.4). There were no significant differences between the two groups with respect to the development of advanced neoplasia (7.4% and 9.9%, respectively; P=0.33). Among the 727 participants receiving resistant starch or placebo, neoplasia developed in 67 participants receiving starch (18.7%), as compared with 68 receiving placebo (18.4%) (relative risk, 1.0; 95% CI, 0.7 to 1.4). Advanced adenomas and colorectal cancers were evenly distributed in the two groups. The prevalence of serious adverse events was low, and the events were evenly distributed.nnnCONCLUSIONSnThe use of aspirin, resistant starch, or both for up to 4 years has no effect on the incidence of colorectal adenoma or carcinoma among carriers of the Lynch syndrome. (Current Controlled Trials number, ISRCTN59521990.)


American Journal of Pathology | 2004

Biallelic Inactivation of Fumarate Hydratase (FH) Occurs in Nonsyndromic Uterine Leiomyomas but Is Rare in Other Tumors

Rainer Lehtonen; Maija Kiuru; Sakari Vanharanta; Jari Sjöberg; Leena Maija Aaltonen; Kristiina Aittomäki; Johanna Arola; Ralf Bützow; Charis Eng; Kirsti Husgafvel-Pursiainen; Jorma Isola; Heikki Järvinen; Pasi A. Koivisto; Jukka Pekka Mecklin; Päivi Peltomäki; Reijo Salovaara; Veli Matti Wasenius; Auli Karhu; Virpi Launonen; Nina N. Nupponen; Lauri A. Aaltonen

Germline mutations in the fumarate hydratase (FH) gene at 1q43 predispose to dominantly inherited cutaneous and uterine leiomyomas, uterine leiomyosarcoma, and papillary renal cell cancer (HLRCC syndrome). To evaluate the role of FH inactivation in sporadic tumorigenesis, we analyzed a series of 299 malignant tumors representing 10 different malignant tumor types for FH mutations. Additionally, 153 uterine leiomyomas from 46 unselected individuals were subjected to and informative in loss of heterozygosity analysis at the FH locus, and the five (3.3%) tumors displaying loss of heterozygosity were subjected to FH mutation analysis. Although mutation search in the 299 malignant tumors was negative, somatic FH mutations were found in two nonsyndromic leiomyomas; a splice site change IVS4 + 3A>G, leading to deletion of exon four, and a missense mutation Ala196Thr. The occurrence of somatic mutations strongly suggests that FH is a true target of the 1q43 deletions. Although uterine leiomyomas are the most common tumors of women, specific inactivating somatic mutations contributing to the formation of nonsyndromic leiomyomas have not been reported previously. Taking into account the apparent risk of uterine leiomyosarcoma associated with FH germline mutations, the finding raises the possibility that also some nonsyndromic leiomyomas may have a genetic profile that is more prone to malignant degeneration. Our data also indicate that somatic FH mutations appear to be limited to tumor types observed in hereditary leiomyomatosis and renal cell cancer.


Diseases of The Colon & Rectum | 1993

Surveillance in hereditary nonpolyposis colorectal cancer: An international cooperative study of 165 families

Hans F. A. Vasen; Jukka Pekka Mecklin; Patrice Watson; Lucio Bertario; Patrick M. Lynch; Lars Bo Svendsen; Giuseppe Cristofaro; Hansjakob Müller; P. Meera Khan; Henry T. Lynch

During its second meeting at Amsterdam in 1990, the International Collaborative Group on Hereditary Non-Polyposis Colorectal Cancer (ICG-HNPCC) decided to carry out a pilot study on colorectal cancer surveillance in HNPCC. The objectives of the study were to ascertain in each of the participating centers the number of HNPCC families, the recommended screening procedures, the age at diagnosis of colorectal cancer (CRC), and the occurrence of interval cancers. Nine centers in seven countries including Denmark, Finland, Italy, Japan, The Netherlands, Switzerland, and the United States participated. Data were derived from a total of 165 families. With respect to screening, half of the centers advise colonoscopy as the only procedure. The interval between the consecutive examinations varies from one to three years. In the majority of the centers, screening begins at 20 to 25 years. Lifelong screening is recommended by three centers, while the rest advise discontinuation at age 60 to 75 years. The family material included 840 patients with colorectal cancer. The mean age at diagnosis was 45 years, and about 15 percent were diagnosed at age 60 or later. A total of 682 high-risk relatives are being followed. After the follow-up of 1 to 10 years in these families, only six cases of interval cancers were encountered.


Genes, Chromosomes and Cancer | 1999

SMAD genes in juvenile polyposis

Stina Roth; Pertti Sistonen; Reijo Salovaara; Akseli Hemminki; Anu Loukola; Marie Johansson; Egle Avizienyte; Karen A. Cleary; Patrick M. Lynch; Christopher I. Amos; Paula Kristo; Jukka Pekka Mecklin; Ilmo Kellokumpu; Heikki Järvinen; Lauri A. Aaltonen

Juvenile polyposis (JP) is a dominantly inherited condition characterized by the development of multiple hamartomatous tumors, juvenile polyps, in the gastrointestinal tract. The aim of this study was to clarify the role of SMAD4 in JP. DNA from four unrelated JP kindreds and three sporadic JP cases was available for mutation screening. Two truncating defects (one in a familial and one in a sporadic case) and one missense change (in a familial case) that was absent in 55 control samples were detected. To study the possibility that germline mutations in other genes encoding different components of the TGF‐β signaling pathway may be present in these JP patients, mutation analyses of the SMAD2, SMAD3, and SMAD7 genes were also performed. No mutations of these genes were detected in any of the patients. Our results confirm that SMAD4 is a gene predisposing to JP and suggest the existence of further JP loci other than the SMAD2, SMAD3, or SMAD7 genes. Genes Chromosomes Cancer 26:54–61, 1999.

Collaboration


Dive into the Jukka Pekka Mecklin's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Auli Karhu

University of Helsinki

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Finlay Macrae

Royal Melbourne Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge