Network


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

Hotspot


Dive into the research topics where Jane M. Palmer is active.

Publication


Featured researches published by Jane M. Palmer.


Cancer Research | 2006

High-risk melanoma susceptibility genes and pancreatic cancer, neural system tumors, and uveal melanoma across GenoMEL

Alisa M. Goldstein; May Chan; Mark Harland; Elizabeth M. Gillanders; Nicholas K. Hayward; Marie-Françoise Avril; Esther Azizi; Giovanna Bianchi-Scarrà; D. Timothy Bishop; Brigitte Bressac-de Paillerets; William Bruno; Donato Calista; Lisa A. Cannon Albright; Florence Demenais; David E. Elder; Paola Ghiorzo; Nelleke A. Gruis; Johan Hansson; David Hogg; Elizabeth A. Holland; Peter A. Kanetsky; Richard F. Kefford; Maria Teresa Landi; Julie Lang; Sancy A. Leachman; Rona M. MacKie; Veronica Magnusson; Graham J. Mann; Kristin B. Niendorf; Julia A. Newton Bishop

GenoMEL, comprising major familial melanoma research groups from North America, Europe, Asia, and Australia has created the largest familial melanoma sample yet available to characterize mutations in the high-risk melanoma susceptibility genes CDKN2A/alternate reading frames (ARF), which encodes p16 and p14ARF, and CDK4 and to evaluate their relationship with pancreatic cancer (PC), neural system tumors (NST), and uveal melanoma (UM). This study included 466 families (2,137 patients) with at least three melanoma patients from 17 GenoMEL centers. Overall, 41% (n = 190) of families had mutations; most involved p16 (n = 178). Mutations in CDK4 (n = 5) and ARF (n = 7) occurred at similar frequencies (2-3%). There were striking differences in mutations across geographic locales. The proportion of families with the most frequent founder mutation(s) of each locale differed significantly across the seven regions (P = 0.0009). Single founder CDKN2A mutations were predominant in Sweden (p.R112_L113insR, 92% of familys mutations) and the Netherlands (c.225_243del19, 90% of familys mutations). France, Spain, and Italy had the same most frequent mutation (p.G101W). Similarly, Australia and United Kingdom had the same most common mutations (p.M53I, c.IVS2-105A>G, p.R24P, and p.L32P). As reported previously, there was a strong association between PC and CDKN2A mutations (P < 0.0001). This relationship differed by mutation. In contrast, there was little evidence for an association between CDKN2A mutations and NST (P = 0.52) or UM (P = 0.25). There was a marginally significant association between NST and ARF (P = 0.05). However, this particular evaluation had low power and requires confirmation. This GenoMEL study provides the most extensive characterization of mutations in high-risk melanoma susceptibility genes in families with three or more melanoma patients yet available.


Nature | 2011

A novel recurrent mutation in MITF predisposes to familial and sporadic melanoma

Satoru Yokoyama; Susan L. Woods; Glen M. Boyle; Lauren G. Aoude; Stuart Macgregor; Victoria Zismann; Michael Gartside; Anne E. Cust; Rizwan Haq; Mark Harland; John C. Taylor; David L. Duffy; Kelly Holohan; Ken Dutton-Regester; Jane M. Palmer; Vanessa F. Bonazzi; Mitchell S. Stark; Judith Symmons; Matthew H. Law; Christopher W. Schmidt; Cathy Lanagan; Linda O’Connor; Elizabeth A. Holland; Helen Schmid; Judith A. Maskiell; Jodie Jetann; Megan Ferguson; Mark A. Jenkins; Richard F. Kefford; Graham G. Giles

So far, two genes associated with familial melanoma have been identified, accounting for a minority of genetic risk in families. Mutations in CDKN2A account for approximately 40% of familial cases, and predisposing mutations in CDK4 have been reported in a very small number of melanoma kindreds. Here we report the whole-genome sequencing of probands from several melanoma families, which we performed in order to identify other genes associated with familial melanoma. We identify one individual carrying a novel germline variant (coding DNA sequence c.G1075A; protein sequence p.E318K; rs149617956) in the melanoma-lineage-specific oncogene microphthalmia-associated transcription factor (MITF). Although the variant co-segregated with melanoma in some but not all cases in the family, linkage analysis of 31 families subsequently identified to carry the variant generated a log of odds (lod) score of 2.7 under a dominant model, indicating E318K as a possible intermediate risk variant. Consistent with this, the E318K variant was significantly associated with melanoma in a large Australian case–control sample. Likewise, it was similarly associated in an independent case–control sample from the United Kingdom. In the Australian sample, the variant allele was significantly over-represented in cases with a family history of melanoma, multiple primary melanomas, or both. The variant allele was also associated with increased naevus count and non-blue eye colour. Functional analysis of E318K showed that MITF encoded by the variant allele had impaired sumoylation and differentially regulated several MITF targets. These data indicate that MITF is a melanoma-predisposition gene and highlight the utility of whole-genome sequencing to identify novel rare variants associated with disease susceptibility.


Journal of Medical Genetics | 2006

Features associated with germline CDKN2A mutations: a GenoMEL study of melanoma-prone families from three continents

Alisa M. Goldstein; May Chan; Mark Harland; Nicholas K. Hayward; Florence Demenais; D. Timothy Bishop; Esther Azizi; Wilma Bergman; Giovanna Bianchi-Scarrà; William Bruno; Donato Calista; Lisa A. Cannon Albright; Valérie Chaudru; Agnès Chompret; Francisco Cuellar; David E. Elder; Paola Ghiorzo; Elizabeth M. Gillanders; Nelleke A. Gruis; Johan Hansson; David Hogg; Elizabeth A. Holland; Peter A. Kanetsky; Richard F. Kefford; Maria Teresa Landi; Julie Lang; Sancy A. Leachman; Rona M. MacKie; Veronica Magnusson; Graham J. Mann

Background: The major factors individually reported to be associated with an increased frequency of CDKN2A mutations are increased number of patients with melanoma in a family, early age at melanoma diagnosis, and family members with multiple primary melanomas (MPM) or pancreatic cancer. Methods: These four features were examined in 385 families with ⩾3 patients with melanoma pooled by 17 GenoMEL groups, and these attributes were compared across continents. Results: Overall, 39% of families had CDKN2A mutations ranging from 20% (32/162) in Australia to 45% (29/65) in North America to 57% (89/157) in Europe. All four features in each group, except pancreatic cancer in Australia (p = 0.38), individually showed significant associations with CDKN2A mutations, but the effects varied widely across continents. Multivariate examination also showed different predictors of mutation risk across continents. In Australian families, ⩾2 patients with MPM, median age at melanoma diagnosis ⩽40 years and ⩾6 patients with melanoma in a family jointly predicted the mutation risk. In European families, all four factors concurrently predicted the risk, but with less stringent criteria than in Australia. In North American families, only ⩾1 patient with MPM and age at diagnosis ⩽40 years simultaneously predicted the mutation risk. Conclusions: The variation in CDKN2A mutations for the four features across continents is consistent with the lower melanoma incidence rates in Europe and higher rates of sporadic melanoma in Australia. The lack of a pancreatic cancer–CDKN2A mutation relationship in Australia probably reflects the divergent spectrum of mutations in families from Australia versus those from North America and Europe. GenoMEL is exploring candidate host, genetic and/or environmental risk factors to better understand the variation observed.


Nature Genetics | 2014

POT1 loss-of-function variants predispose to familial melanoma

Carla Daniela Robles-Espinoza; Mark Harland; Andrew J. Ramsay; Lauren G. Aoude; Zhihao Ding; Karen A. Pooley; Antonia L. Pritchard; Jessamy Tiffen; Mia Petljak; Jane M. Palmer; Judith Symmons; Peter Johansson; Mitchell S. Stark; Michael Gartside; Helen Snowden; Grant W. Montgomery; Nicholas G. Martin; Jimmy Z. Liu; Jiyeon Choi; Matthew Makowski; Kevin M. Brown; Alison M. Dunning; Thomas M. Keane; Carlos López-Otín; Nelleke A. Gruis; Nicholas K. Hayward; D. Timothy Bishop; Julia Newton-Bishop; David J. Adams

Deleterious germline variants in CDKN2A account for around 40% of familial melanoma cases, and rare variants in CDK4, BRCA2, BAP1 and the promoter of TERT have also been linked to the disease. Here we set out to identify new high-penetrance susceptibility genes by sequencing 184 melanoma cases from 105 pedigrees recruited in the UK, The Netherlands and Australia that were negative for variants in known predisposition genes. We identified families where melanoma cosegregates with loss-of-function variants in the protection of telomeres 1 gene (POT1), with a proportion of family members presenting with an early age of onset and multiple primary tumors. We show that these variants either affect POT1 mRNA splicing or alter key residues in the highly conserved oligonucleotide/oligosaccharide-binding (OB) domains of POT1, disrupting protein-telomere binding and leading to increased telomere length. These findings suggest that POT1 variants predispose to melanoma formation via a direct effect on telomeres.


American Journal of Human Genetics | 2003

Localization of a Novel Melanoma Susceptibility Locus to 1p22

Elizabeth M. Gillanders; Suh-Hang Hank Juo; Elizabeth A. Holland; MaryPat Jones; Derek J. Nancarrow; Diana Freas-Lutz; Raman Sood; Naeun Park; Mezbah U. Faruque; Carol Markey; Richard F. Kefford; Jane M. Palmer; Wilma Bergman; D. Timothy Bishop; Margaret A. Tucker; Brigitte Bressac-de Paillerets; Johan Hansson; Mitchell S. Stark; Nelleke A. Gruis; Julia A. Newton Bishop; Alisa M. Goldstein; Joan E. Bailey-Wilson; Graham J. Mann; Nicholas K. Hayward; Jeffrey M. Trent

Over the past 20 years, the incidence of cutaneous malignant melanoma (CMM) has increased dramatically worldwide. A positive family history of the disease is among the most established risk factors for CMM; it is estimated that 10% of CMM cases result from an inherited predisposition. Although mutations in two genes, CDKN2A and CDK4, have been shown to confer an increased risk of CMM, they account for only 20%-25% of families with multiple cases of CMM. Therefore, to localize additional loci involved in melanoma susceptibility, we have performed a genomewide scan for linkage in 49 Australian pedigrees containing at least three CMM cases, in which CDKN2A and CDK4 involvement has been excluded. The highest two-point parametric LOD score (1.82; recombination fraction [theta] 0.2) was obtained at D1S2726, which maps to the short arm of chromosome 1 (1p22). A parametric LOD score of 4.65 (theta=0) and a nonparametric LOD score of 4.19 were found at D1S2779 in nine families selected for early age at onset. Additional typing yielded seven adjacent markers with LOD scores >3 in this subset, with the highest parametric LOD score, 4.95 (theta=0) (nonparametric LOD score 5.37), at D1S2776. Analysis of 33 additional multiplex families with CMM from several continents provided further evidence for linkage to the 1p22 region, again strongest in families with the earliest mean age at diagnosis. A nonparametric ordered sequential analysis was used, based on the average age at diagnosis in each family. The highest LOD score, 6.43, was obtained at D1S2779 and occurred when the 15 families with the earliest ages at onset were included. These data provide significant evidence of a novel susceptibility gene for CMM located within chromosome band 1p22.


Cancer Epidemiology, Biomarkers & Prevention | 2010

Predictors of Sun Protection Behaviors and Severe Sunburn in an International Online Study

Richard Bränström; Y.M. Chang; Paul Affleck; Aad Tibben; Lisa G. Aspinwall; Esther Azizi; Orna Baron-Epel; Wilma Bergman; William Bruno; May Chan; Francisco Cuellar; Tadeusz Dębniak; Dace Pjanova; Sławomir Ertmański; Adina Figl; Melinda Gonzalez; Nicholas K. Hayward; Marko Hocevar; Peter A. Kanetsky; Sancy A. Leachman; Olita Heisele; Jane M. Palmer; Barbara Peric; Susana Puig; Dirk Schadendorf; Nelleke A. Gruis; Julia Newton-Bishop; Yvonne Brandberg

Background: The incidence of melanoma continues to increase in many countries, and primary prevention of melanoma includes avoidance of sunburn as well as adequate sun protection behavior. The aim of this study was to examine the prevalence of self-reported sun protection behaviors and sunburn in users of the Internet, and to identify the demographic, clinical, and attitudinal/motivational correlates of sun protection behaviors. Methods: Self-report data were gathered on behalf of the GenoMEL consortium using an online survey available in 10 different languages, and 8,178 individuals successfully completed at least 80% of survey items, with 73% of respondents from Europe, 12% from Australia, 7% from the United States, 2% from Israel, and 6% from other countries. Results: Half of all respondents and 27% of those with a previous melanoma reported at least one severe sunburn during the previous 12 months. The strongest factors associated with sun protection behavior were perceived barriers to protection (β = −0.44/β = −0.37), and respondents who reported a positive attitude toward suntans were less likely to protect (β = −0.16/β = −0.14). Reported use of protective clothing and shade, as well as avoidance of midday sun exposure, were more strongly related to reduced risk of sunburn than sunscreen use. Conclusions: Despite widespread dissemination of public health messages about the importance of sun protection, a substantial proportion of this international sample, including respondents with a previous melanoma, reported inadequate sun protection behaviors resulting in severe sunburn. Impact: Future strategies to decrease sunburn should target the practical, social, and psychological barriers associated with nonuptake of sun protection. Cancer Epidemiol Biomarkers Prev; 19(9); 2199–210. ©2010 AACR.


Journal of Medical Genetics | 2013

Melanoma prone families with CDK4 germline mutation: phenotypic profile and associations with MC1R variants

Hanne E. Puntervoll; Xiaohong R. Yang; Hildegunn Vetti; Ingeborg M. Bachmann; M.-F. Avril; M. Benfodda; Caterina Catricalà; Stéphane Dalle; Anne Benedicte Duval-Modeste; Paola Ghiorzo; Paola Grammatico; Mark Harland; Nicholas K. Hayward; Hui Han Hu; Thomas Jouary; Tanguy Martin-Denavit; Aija Ozola; Jane M. Palmer; Lorenza Pastorino; Dace Pjanova; Nadem Soufir; Solrun J. Steine; Alexander J. Stratigos; Luc Thomas; Julie Tinat; Hensin Tsao; Ruta Veinalde; Margaret A. Tucker; Brigitte Bressac-de Paillerets; Julia Newton-Bishop

Background CDKN2A and CDK4 are high risk susceptibility genes for cutaneous malignant melanoma. Melanoma families with CDKN2A germline mutations have been extensively characterised, whereas CDK4 families are rare and lack a systematic investigation of their phenotype. Methods All known families with CDK4 germline mutations (n=17) were recruited for the study by contacting the authors of published papers or by requests via the Melanoma Genetics Consortium (GenoMEL). Phenotypic data related to primary melanoma and pigmentation characteristics were collected. The CDK4 exon 2 and the complete coding region of the MC1R gene were sequenced. Results Eleven families carried the CDK4 R24H mutation whereas six families had the R24C mutation. The total number of subjects with verified melanoma was 103, with a median age at first melanoma diagnosis of 39 years. Forty-three (41.7%) subjects had developed multiple primary melanomas (MPM). A CDK4 mutation was found in 89 (including 62 melanoma cases) of 209 tested subjects. CDK4 positive family members (both melanoma cases and unaffected subjects) were more likely to have clinically atypical nevi than CDK4 negative family members (p<0.001). MPM subjects had a higher frequency of MC1R red hair colour variants compared with subjects with one tumour (p=0.010). Conclusion Our study shows that families with CDK4 germline mutations cannot be distinguished phenotypically from CDKN2A melanoma families, which are characterised by early onset of disease, increased occurrence of clinically atypical nevi, and development of MPM. In a clinical setting, the CDK4 gene should therefore always be examined when a melanoma family tests negative for CDKN2A mutation.


Oncotarget | 2016

Deep sequencing of uveal melanoma identifies a recurrent mutation in PLCB4.

Peter A. Johansson; Lauren G. Aoude; Karin Wadt; William J. Glasson; Sunil Warrier; Alex W. Hewitt; Jens Folke Kiilgaard; Steffen Heegaard; Tim Isaacs; Maria Franchina; Christian Ingvar; Tersia Vermeulen; Kevin J Whitehead; Christopher W. Schmidt; Jane M. Palmer; Judith Symmons; Anne-Marie Gerdes; Göran Jönsson; Nicholas K. Hayward

Next generation sequencing of uveal melanoma (UM) samples has identified a number of recurrent oncogenic or loss-of-function mutations in key driver genes including: GNAQ, GNA11, EIF1AX, SF3B1 and BAP1. To search for additional driver mutations in this tumor type we carried out whole-genome or whole-exome sequencing of 28 tumors or primary cell lines. These samples have a low mutation burden, with a mean of 10.6 protein changing mutations per sample (range 0 to 53). As expected for these sun-shielded melanomas the mutation spectrum was not consistent with an ultraviolet radiation signature, instead, a BRCA mutation signature predominated. In addition to mutations in the known UM driver genes, we found a recurrent mutation in PLCB4 (c.G1888T, p.D630Y, NM_000933), which was validated using Sanger sequencing. The identical mutation was also found in published UM sequence data (1 of 56 tumors), supporting its role as a novel driver mutation in UM. PLCB4 p.D630Y mutations are mutually exclusive with mutations in GNA11 and GNAQ, consistent with PLCB4 being the canonical downstream target of the former gene products. Taken together these data suggest that the PLCB4 hotspot mutation is similarly a gain-of-function mutation leading to activation of the same signaling pathway, promoting UM tumorigenesis.


Genomics | 1992

Exclusion of the familial melanoma locus (MLM) from the PND/D1S47 and MYCL1 regions of chromosome arm 1p in 7 Australian pedigrees.

Derek J. Nancarrow; Jane M. Palmer; Marilyn K. Walters; Kerr Bm; Hafner Gj; Garske L; McLeod Gr; Nicholas K. Hayward

Familial melanoma (MLM) is sometimes found associated with the dysplastic nevus syndrome (DNS). Considerable controversy exists over the possible assignment of a cutaneous malignant melanoma/dysplastic nevus gene, designated CMM, to the distal short arm of chromosome 1, linked to the PND and D1S47 loci. To date, no support for linkage of MLM alone to these markers has been found; likewise no study has been able to exclude the entire region between PND and D1S47 from linkage to MLM. We have carried out linkage studies between markers on 1p and MLM in seven Australian kindreds; three of these are the largest reported worldwide. We have been able to exclude localization of an MLM gene from a 40-cM region that spans the interval between D1S47 and PND and extends approximately 15 cM on either side of these markers. In addition, we can exclude a region of about 20 cM around the MYCL1/D1S57 loci.


Journal of Investigative Dermatology | 2011

Nevi, Family History, and Fair Skin Increase the Risk of Second Primary Melanoma

Victor Siskind; Maria Celia Hughes; Jane M. Palmer; Judith Symmons; Joanne F. Aitken; Nicholas G. Martin; Nicholas K. Hayward; David C. Whiteman

Although risk factors for primary cutaneous melanoma are well defined, relatively little is known about predictors for second primary melanoma. Given the rising incidence of this cancer, coupled with improvements in survival, there is a prevalent and growing pool of patients at risk of second primary melanomas. To identify the predictors of second primary melanoma, we followed a cohort of 1,083 Queensland patients diagnosed with incident melanoma between 1982 and 1990 and who completed a baseline questionnaire. During a median follow-up of 16.5 years, 221 patients were diagnosed with at least one additional primary melanoma. In multivariate analyses, second primary melanomas were associated with high nevus count (hazard ratio (HR), 2.91; 95% confidence interval (CI) 1.94-4.35), high familial melanoma risk (HR, 2.12; 95% CI 1.34-3.36), fair skin (HR, 1.51; 95% CI 1.06-2.16), inability to tan (HR, 1.66; 95% CI 1.13-2.43), an in situ first primary melanoma (HR, 1.36; 95% CI 0.99-1.87), and male sex (HR, 1.49; 95% CI 1.12-2.00). Patients whose first primary was lentigo maligna melanoma (HR, 1.80; 95% CI 1.05-3.07) or nodular melanoma (HR, 2.13; 95% CI 1.21-3.74) had higher risks of subsequent primaries than patients whose first primary tumor was superficial spreading melanoma. These characteristics could be assessed in patients presenting with first primary melanoma to evaluate risk of developing a second primary.

Collaboration


Dive into the Jane M. Palmer's collaboration.

Top Co-Authors

Avatar

Nicholas K. Hayward

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Judith Symmons

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Marilyn K. Walters

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Mitchell S. Stark

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Graeme J. Walker

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Lauren G. Aoude

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nicholas G. Martin

QIMR Berghofer Medical Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge