Network


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

Hotspot


Dive into the research topics where Deborah Packham is active.

Publication


Featured researches published by Deborah Packham.


International Journal of Cancer | 2011

De novo constitutional MLH1 epimutations confer early-onset colorectal cancer in two new sporadic Lynch syndrome cases, with derivation of the epimutation on the paternal allele in one.

Ajay Goel; Thuy–Phuong Nguyen; Hon Chiu Eastwood Leung; Takeshi Nagasaka; Jennifer Rhees; Erin Hotchkiss; Mildred Arnold; Pia Banerji; Minoru Koi; Chau To Kwok; Deborah Packham; Lara Lipton; C. Richard Boland; Robyn L. Ward; Megan P. Hitchins

Lynch syndrome is an autosomal dominant cancer predisposition syndrome classically caused by germline mutations of the mismatch repair genes, MLH1, MSH2, MSH6 and PMS2. Constitutional epimutations of the MLH1 gene, characterized by soma‐wide methylation of a single allele of the promoter and allelic transcriptional silencing, have been identified in a subset of Lynch syndrome cases lacking a sequence mutation in MLH1. We report two individuals with no family history of colorectal cancer who developed that disease at age 18 and 20 years. In both cases, cancer had arisen because of the de novo occurrence of a constitutional MLH1 epimutation and somatic loss‐of‐heterozygosity of the functional allele in the tumors. We show for the first time that the epimutation in one case arose on the paternally inherited allele. Analysis of 13 tumors from seven individuals with constitutional MLH1 epimutations showed eight tumors had lost the second MLH1 allele, two tumors had a novel pathogenic missense mutation and three had retained heterozygosity. Only 1 of 12 tumors demonstrated the BRAF V600E mutation and 3 of 11 tumors harbored a mutation in KRAS. The finding that epimutations can originate on the paternal allele provides important new insights into the mechanism of origin of epimutations. It is clear that the second hit in MLH1 epimutation‐associated tumors typically has a genetic not epigenetic basis. Individuals with mismatch repair–deficient cancers without the BRAF V600E mutation are candidates for germline screening for sequence or methylation changes in MLH1.


Cancer Research | 2007

Epigenetic Inactivation of a Cluster of Genes Flanking MLH1 in Microsatellite-Unstable Colorectal Cancer

Megan P. Hitchins; Andrew Buckle; Kayfong Cheong; Nimita Halani; Su Ku; Chau-To Kwok; Deborah Packham; Catherine M. Suter; Alan P. Meagher; Clare Stirzaker; Susan J. Clark; Nicholas J. Hawkins; Robyn L. Ward

Biallelic promoter methylation and transcriptional silencing of the MLH1 gene occurs in the majority of sporadic colorectal cancers exhibiting microsatellite instability due to defective DNA mismatch repair. Long-range epigenetic silencing of contiguous genes has been found on chromosome 2q14 in colorectal cancer. We hypothesized that epigenetic silencing of MLH1 could occur on a regional scale affecting additional genes within 3p22, rather than as a focal event. We studied the levels of CpG island methylation and expression of multiple contiguous genes across a 4 Mb segment of 3p22 including MLH1 in microsatellite-unstable and -stable cancers, and their paired normal colonic mucosa. We found concordant CpG island hypermethylation, H3-K9 dimethylation and transcriptional silencing of MLH1 and multiple flanking genes spanning up to 2.4 Mb in microsatellite-unstable colorectal cancers. This region was interspersed with unmethylated genes, which were also transcriptionally repressed. Expression of both methylated and unmethylated genes was reactivated by methyltransferase and histone deacetylase inhibitors in a microsatellite-unstable colorectal carcinoma cell line. Two genes at the telomeric end of the region were also hypermethylated in microsatellite-stable cancers, adenomas, and at low levels in normal colonic mucosa from older individuals. Thus, the cluster of genes flanking MLH1 that was specifically methylated in the microsatellite-unstable group of cancers extended across 1.1 Mb. Our results show that coordinate epigenetic silencing extends across a large chromosomal region encompassing MLH1 in microsatellite-unstable colorectal cancers. Simultaneous epigenetic silencing of this cluster of 3p22 genes may contribute to the development or progression of this type of cancer.


Diagnostic Molecular Pathology | 2009

Implementation of novel pyrosequencing assays to screen for common mutations of BRAF and KRAS in a cohort of sporadic colorectal cancers.

Deborah Packham; Robyn L. Ward; Nicholas J. Hawkins; Megan P. Hitchins

Activating mutations of the BRAF and KRAS genes cause constitutive stimulation of an important cell-signaling pathway promoting tumorigenesis, and are increasingly recognized as determinants of response to targeted cancer therapies. The V600E mutation accounts for most of the BRAF mutations in cancer, and KRAS mutations are predominantly encoded by nucleotide substitutions within codons 12 and 13. We designed novel pyrosequencing assays for the detection of the common “hotspot” mutations in these genes, which demonstrated analytical sensitivities of ≤10% in titrations of mutant cell lines. The KRAS pyrosequencing assay has the ability to simultaneously identify all potential nucleotide changes within the mutation cluster at codons 12 and 13, with a sequence output in the sense direction to facilitate results interpretation. These assays were used to determine the mutation status in a prospective series of 1198 sporadic colorectal cancers. The BRAF V600E mutation was detected in 13.2% of the colorectal cancers. The frequency of KRAS mutations in our cohort was 32.4%, with G>A transitions at position 2 of codons 12 and 13 being most prevalent. Both assays proved highly sensitive and specific when applied to clinical specimens, and were applicable to both fresh-frozen and formalin-fixed paraffin-embedded archival tissues. These assays would serve as a suitable platform for large-scale mutation detection in cancer specimens where the facility for pyrosequencing is available.


The Journal of Pathology | 2005

Routine testing for mismatch repair deficiency in sporadic colorectal cancer is justified.

Robyn L. Ward; Jenny Turner; Rachel Williams; Brita Pekarsky; Deborah Packham; Marija Velickovic; Alan P. Meagher; Terence O'Connor; Nicholas J. Hawkins

This study prospectively examines the accuracy of immunohistochemical staining in the identification of mismatch repair defective (MMRD) colorectal cancer in routine clinical practice. The potential impact of this information on decisions regarding adjuvant treatment and germline testing were quantified. A consecutive series of fresh tissue (836 cancers) was obtained from 786 individuals undergoing curative surgery for colorectal cancer at one institution. As part of normal practice, each tumour was screened for the expression of MLH1 and MSH2 by immunohistochemical staining (IHC) and relevant clinicopathological details were documented. Microsatellite instability (MSI) was assessed using standard markers. Overall, 108 (13%) tumours showed loss of staining for either MLH1 (92 tumours) or MSH2 (16 tumours). The positive predictive value of mismatch repair IHC when used alone in the detection of MSI tumours was 88%, and the negative predictive value was 97%. Specificity and positive predictive value were improved by correlation with microsatellite status. Tumour stage (HR 3.5, 95% CI 2.0–6.0), vascular space invasion (HR 1.9, 95% CI 1.2–3.0) and mismatch repair deficiency (HR 0.2, 95% CI 0.05–0.87) were independent prognostic factors in stages II and III disease. Screening by mismatch repair IHC could reasonably have been expected to prevent ineffective treatment in 3.6% of stage II and 7.6% of stage III patients. The frequency of germline mismatch repair mutations was 0.8%, representing six unsuspected hereditary non‐polyposis colorectal cancer (HNPCC) cases. Routine screening of colorectal cancers by mismatch repair IHC identifies individuals at low risk of relapse, and can prevent unnecessary adjuvant treatments in a significant number of individuals. Abnormal immunohistochemistry should be confirmed by microsatellite testing to ensure that false‐positive results do not adversely impact on treatment decisions. Copyright


Biological Procedures Online | 2012

A reinvestigation of somatic hypermethylation at the PTEN CpG island in cancer cell lines

Luke B. Hesson; Deborah Packham; Emily Pontzer; Pauline Funchain; Charis Eng; Robyn L. Ward

BackgroundPTEN is an important tumour suppressor gene that is mutated in Cowden syndrome as well as various sporadic cancers. CpG island hypermethylation is another route to tumour suppressor gene inactivation, however, the literature regarding PTEN hypermethylation in cancer is controversial. Furthermore, investigation of the methylation status of the PTEN CpG island is challenging due to sequence homology with the PTEN pseudogene, PTENP1. PTEN shares a CpG island promoter with another gene known as KLLN. Here we present a thorough reinvestigation of the methylation status of the PTEN CpG island in DNA from colorectal, breast, ovarian, glioma, lung and haematological cancer cell lines.ResultsUsing a range of bisulphite-based PCR assays we investigated 6 regions across the PTEN CpG island. We found that regions 1-4 were not methylated in cancer cell lines (0/36). By allelic bisulphite sequencing and pyrosequencing methylation was detected in regions 5 and 6 in colorectal, breast and haematological cancer cell lines. However, methylation detected in this region was associated with the PTENP1 promoter and not the PTEN CpG island.ConclusionsWe show that methylation of the PTEN CpG island is a rare event in cancer cell lines and that apparent methylation most likely originates from homologous regions of the PTENP1 pseudogene promoter. Future studies should utilize assays that reliably discriminate between PTEN and PTENP1 to avoid data misinterpretation.


JAMA Oncology | 2015

Mosaic Epigenetic Inheritance as a Cause of Early-Onset Colorectal Cancer

Mathew A. Sloane; Andrea C. Nunez; Deborah Packham; Chau-To Kwok; Graeme Suthers; Luke B. Hesson; Robyn L. Ward

IMPORTANCE Constitutional hypermethylation of 1 allele throughout the soma (constitutional epimutation) is an accepted mechanism of cancer predisposition. Understanding the origin and inheritance of epimutations is important for assessing cancer risk in affected families. OBSERVATIONS We report a 29-year-old man with early-onset colorectal cancer who showed a constitutional MLH1 epimutation (approximately 50% of alleles methylated and allele-specific loss of MLH1 expression) that was stable over a 16-year period. The epimutation was inherited without a genetic alteration from his asymptomatic mother. She showed methylation on the same allele but in less than 5% of her somatic cells. CONCLUSIONS AND RELEVANCE These findings indicate that low-level somatic mosaicism for an epimutation in an asymptomatic parent can produce a nonmosaic constitutional epimutation in a child. Asymptomatic low-level methylation in some individuals may be associated with substantial cancer risk to their offspring.


Molecular Cancer Research | 2016

Integrated Genetic, Epigenetic, and Transcriptional Profiling Identifies Molecular Pathways in the Development of Laterally Spreading Tumors

Luke B. Hesson; Benedict Ng; Peter Zarzour; Sameer Srivastava; Chau To Kwok; Deborah Packham; Andrea C. Nunez; Dominik Beck; Regina Ryan; Ashraf Dower; Caroline E. Ford; John E. Pimanda; Mathew A. Sloane; Nicholas J. Hawkins; Michael J. Bourke; Jason Wong; Robyn L. Ward

Laterally spreading tumors (LST) are colorectal adenomas that develop into extremely large lesions with predominantly slow progression to cancer, depending on lesion subtype. Comparing and contrasting the molecular profiles of LSTs and colorectal cancers offers an opportunity to delineate key molecular alterations that drive malignant transformation in the colorectum. In a discovery cohort of 11 LSTs and paired normal mucosa, we performed a comprehensive and unbiased screen of the genome, epigenome, and transcriptome followed by bioinformatics integration of these data and validation in an additional 84 large, benign colorectal lesions. Mutation rates in LSTs were comparable with microsatellite-stable colorectal cancers (2.4 vs. 2.6 mutations per megabase); however, copy number alterations were infrequent (averaging only 1.5 per LST). Frequent genetic, epigenetic, and transcriptional alterations were identified in genes not previously implicated in colorectal neoplasia (ANO5, MED12L, EPB41L4A, RGMB, SLITRK1, SLITRK5, NRXN1, ANK2). Alterations to pathways commonly mutated in colorectal cancers, namely, the p53, PI3K, and TGFβ pathways, were rare. Instead, LST-altered genes converged on axonal guidance, Wnt, and actin cytoskeleton signaling. These integrated omics data identify molecular features associated with noncancerous LSTs and highlight that mutation load, which is relatively high in LSTs, is a poor predictor of invasive potential. Implications: The novel genetic, epigenetic, and transcriptional changes associated with LST development reveal important insights into why some adenomas do not progress to cancer. The finding that LSTs exhibit a mutational load similar to colorectal carcinomas has implications for the validity of molecular biomarkers for assessing cancer risk. Mol Cancer Res; 14(12); 1217–28. ©2016 AACR.


Molecular and Cellular Biology | 2016

Arrested Hematopoiesis and Vascular Relaxation Defects in Mice with a Mutation in Dhfr

Julie A.I. Thoms; Kathy Knezevic; Jia Jenny Liu; Elias N. Glaros; Thuan Thai; Qiao Qiao; Heather Campbell; Deborah Packham; Yizhou Huang; Panagiotis Papathanasiou; Robert Tunningley; Belinda Whittle; Amanda W. S. Yeung; Vashe Chandrakanthan; Luke B. Hesson; Vivien M. Chen; Jason Wong; Louise E. Purton; Robyn L. Ward; Shane R. Thomas; John E. Pimanda

ABSTRACT Dihydrofolate reductase (DHFR) is a critical enzyme in the folate metabolism pathway and also plays a role in regulating nitric oxide (NO) signaling in endothelial cells. Although both coding and noncoding mutations with phenotypic effects have been identified in the human DHFR gene, no mouse model is currently available to study the consequences of perturbing DHFR in vivo. In order to identify genes involved in definitive hematopoiesis, we performed a forward genetic screen and produced a mouse line, here referred to as Orana, with a point mutation in the Dhfr locus leading to a Thr136Ala substitution in the DHFR protein. Homozygote Orana mice initiate definitive hematopoiesis, but expansion of progenitors in the fetal liver is compromised, and the animals die between embryonic day 13.5 (E13.5) and E14.5. Heterozygote Orana mice survive to adulthood but have tissue-specific alterations in folate abundance and distribution, perturbed stress erythropoiesis, and impaired endothelium-dependent relaxation of the aorta consistent with the role of DHFR in regulating NO signaling. Orana mice provide insight into the dual roles of DHFR and are a useful model for investigating the role of environmental and dietary factors in the context of vascular defects caused by altered NO signaling.


bioRxiv | 2018

In search of non-coding driver mutations by deep sequencing of regulatory elements in colorectal cancer

Rebecca C. Poulos; Dilmi Perera; Deborah Packham; Anushi Shah; Caroline Janitz; John E. Pimanda; Nicholas J. Hawkins; Robyn L. Ward; Luke B. Hesson; Jason Wong

Large-scale whole cancer-genome sequencing projects have led to the identification of a handful of cis-regulatory driver mutations in cancer genomes. However, recent studies have demonstrated that very large cancer cohorts will be required in order to identify low frequency non-coding drivers. To further this endeavour, in this study, we performed highdepth sequencing across 95 colorectal cancers and matched normal samples using a unique target capture sequencing (TCS) assay focusing on over 35 megabases of gene regulatory elements. We first assessed coverage and variant detection capability from our TCS data, and compared this with a sample that was additionally whole-genome sequenced (WGS). TCS enabled substantially deeper sequencing and thus we detected 51% more somatic single nucleotide variants (n = 2,457) and 144% more somatic insertions and deletions (n = 39) by TCS than WGS. Variants obtained from TCS data were suitable for somatic mutational signature detection, enabling us to define the signatures associated with germline deleterious variants in MSH6 and MUTYH in samples within our cohort. Finally, we surveyed regulatory mutations to find putative drivers by assessing variant recurrence and function, identifying some regulatory variants that may influence oncogenesis. Our study demonstrates TCS to be a sequencing-efficient alternative to traditional WGS, enabling improved coverage and variant detection when seeking to identify variants at specific loci among larger cohorts. Interestingly, we found no candidate variants that have a clear driver function, suggesting that regulatory drivers may be rare in a colorectal cancer cohort of this size. Author Summary In recent years, some cancer research focus has turned towards the role of somatic mutations in the 98% of the genome that is non-coding. To investigate such mutations, we performed deep sequencing of regulatory regions and a selection of coding genes across 95 colorectal cancer and matched-normal samples. To determine the ability of our targeted deep sequencing methodology to accurately detect variants, we compared our results with those from a sample that was additionally whole-genome sequenced. We found target capture sequencing to enable greater sequencing depth, allowing the detection of 51% and 144% more somatic single nucleotide and insertion/deletion mutations, respectively. Our study here demonstrates target capture sequencing to be a useful approach for researchers seeking to identify variants at specific loci among larger cohorts. Our results also enabled the generation of mutational signatures, implicating deleterious germline single nucleotide variants in coding exons of MSH6 and MUTYH in samples within our cohort. Finally, we surveyed regulatory elements in search of somatic cancer driver mutations. We identified some regulatory variants that may influence oncogenesis, but found no candidate variants with clear driver function. These findings suggest that regulatory driver mutations may be rare in a colorectal cancer cohort of this size.


Clinical Cancer Research | 2018

Disruption of a −35 kb Enhancer Impairs CTCF Binding and MLH1 Expression in Colorectal Cells

Qing Liu; Julie A.I. Thoms; Andrea C. Nunez; Yizhou Huang; Kathy Knezevic; Deborah Packham; Rebecca C. Poulos; Rachel Williams; Dominik Beck; Nicholas J. Hawkins; Robyn L. Ward; Jason Wong; Luke B. Hesson; Mathew A. Sloane; John E. Pimanda

Purpose: MLH1 is a major tumor suppressor gene involved in the pathogenesis of Lynch syndrome and various sporadic cancers. Despite their potential pathogenic importance, genomic regions capable of regulating MLH1 expression over long distances have yet to be identified. Experimental Design: Here, we use chromosome conformation capture (3C) to screen a 650-kb region flanking the MLH1 locus to identify interactions between the MLH1 promoter and distal regions in MLH1-expressing and nonexpressing cells. Putative enhancers were functionally validated using luciferase reporter assays, chromatin immunoprecipitation, and CRISPR-Cas9–mediated deletion of endogenous regions. To evaluate whether germline variants in the enhancer might contribute to impaired MLH1 expression in patients with suspected Lynch syndrome, we also screened germline DNA from a cohort of 74 patients with no known coding mutations or epimutations at the MLH1 promoter. Results: A 1.8-kb DNA fragment, 35 kb upstream of the MLH1 transcription start site enhances MLH1 gene expression in colorectal cells. The enhancer was bound by CTCF and CRISPR-Cas9–mediated deletion of a core binding region impairs endogenous MLH1 expression. A total of 5.4% of suspected Lynch syndrome patients have a rare single-nucleotide variant (G > A; rs143969848; 2.5% in gnomAD European, non-Finnish) within a highly conserved CTCF-binding motif, which disrupts enhancer activity in SW620 colorectal carcinoma cells. Conclusions: A CTCF-bound region within the MLH1-35 enhancer regulates MLH1 expression in colorectal cells and is worthy of scrutiny in future genetic screening strategies for suspected Lynch syndrome associated with loss of MLH1 expression. Clin Cancer Res; 24(18); 4602–11. ©2018 AACR.

Collaboration


Dive into the Deborah Packham's collaboration.

Top Co-Authors

Avatar

Robyn L. Ward

University of Queensland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Luke B. Hesson

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Andrea C. Nunez

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Mathew A. Sloane

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Jason Wong

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

John E. Pimanda

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rachel Williams

St. Vincent's Health System

View shared research outputs
Top Co-Authors

Avatar

Qing Liu

University of New South Wales

View shared research outputs
Researchain Logo
Decentralizing Knowledge