Network


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

Hotspot


Dive into the research topics where Kristen D. Hadfield is active.

Publication


Featured researches published by Kristen D. Hadfield.


Clinical Cancer Research | 2008

Impaired tamoxifen metabolism reduces survival in familial breast cancer patients.

William G. Newman; Kristen D. Hadfield; Ayshe Latif; Stephen A Roberts; Andrew Shenton; Christopher McHague; Fiona Lalloo; Sacha J Howell; D. Gareth Evans

Purpose: Tamoxifen has been the mainstay adjuvant hormonal treatment for breast cancer for many years. Conversion of tamoxifen to its active metabolite, endoxifen, is reduced by low activity of the cytochrome P450 enzyme, CYP2D6. We examined the effect of reduced CYP2D6 activity on the response to tamoxifen in patients with familial early-onset breast cancer. Experimental Design: We conducted a case note review and genotyping for the CYP2D6*3, CYP2D6*4, CYP2D6*5, and CYP2D6*41 alleles in 115 patients (47 BRCA1, 68 BRCA2) with familial breast cancer who had been treated with 20-mg tamoxifen following surgery. Results: Eight (7%) individuals had genotypes consistent with poor metabolizer status, and 4 (3.5%) individuals took CYP2D6 inhibitor drugs concomitant with their tamoxifen and were also considered poor metabolizer. Time to tumor recurrence, disease-free survival, and overall survival were reduced in the patient group with poor metabolizer CYP2D6 activity. However, a significant effect was confined to patients with BRCA2 mutations with a worse overall survival (median survival, 7 versus 28 years; P = 0.008; adjusted hazard ratio, 9.7). Conclusions: Poor metabolizer status for CYP2D6 predicts worse overall survival in patients with familial breast cancer. Therefore, CYP2D6 inhibitor drugs should not be prescribed concomitantly with tamoxifen. Prospective studies should be undertaken to establish the effect of CYP2D6 status on outcome in familial breast cancer patients treated with tamoxifen.


Nature Genetics | 2013

Loss-of-function mutations in SMARCE1 cause an inherited disorder of multiple spinal meningiomas

Miriam J. Smith; James O'Sullivan; Sanjeev Bhaskar; Kristen D. Hadfield; Gemma Poke; John Caird; Saba Sharif; Diana Eccles; David Fitzpatrick; Daniel Rawluk; Daniel du Plessis; William G. Newman; D. Gareth Evans

One-third of all primary central nervous system tumors in adults are meningiomas. Rarely, meningiomas occur at multiple sites, usually occurring in individuals with type 2 neurofibromatosis (NF2). We sequenced the exomes of three unrelated individuals with familial multiple spinal meningiomas without NF2 mutations. We identified two individuals with heterozygous loss-of-function mutations in the SWI/SNF chromatin-remodeling complex subunit gene SMARCE1. Sequencing of SMARCE1 in six further individuals with spinal meningiomas identified two additional heterozygous loss-of-function mutations. Tumors from individuals with SMARCE1 mutations were of clear-cell histological subtype, and all had loss of SMARCE1 protein, consistent with a tumor suppressor mechanism. Our findings identify multiple-spinal-meningioma disease as a new discrete entity and establish a key role for the SWI/SNF complex in the pathogenesis of both meningiomas and tumors with clear-cell histology.


Oncogene | 2010

Rates of loss of heterozygosity and mitotic recombination in NF2 schwannomas, sporadic vestibular schwannomas and schwannomatosis schwannomas

Kristen D. Hadfield; Miriam J. Smith; Jill Urquhart; Andrew Wallace; Naomi L. Bowers; Andrew T. King; Scott A. Rutherford; Dorothy Trump; William G. Newman; D G R Evans

Biallelic inactivation of the NF2 gene occurs in the majority of schwannomas. This usually involves a combination of a point mutation or multiexon deletion, in conjunction with either a second point mutation or loss of heterozygosity (LOH). We have performed DNA sequence and dosage analysis of the NF2 gene in a panel of 239 schwannoma tumours: 97 neurofibromatosis type 2 (NF2)-related schwannomas, 104 sporadic vestibular schwannomas (VS) and 38 schwannomatosis-related schwannomas. In total, we identified germline NF2 mutations in 86 out of 97 (89%) NF2 patients and a second mutational event in 77 out of 97 (79%). LOH was by far the most common form of second hit. A combination of microsatellite analysis with either conventional comparative genomic hybridization (CGH) or multiplex ligation-dependent probe amplification (MLPA) identified mitotic recombination (MR) as the cause of LOH in 14 out of 72 (19%) total evaluable tumours. Among sporadic VS, at least one NF2 mutation was identified by sequence analysis or MLPA in 65 out of 98 (66%) tumours. LOH occurred in 54 out of 96 (56%) evaluable tumours, but MR only accounted for 5 out of 77 (6%) tested. LOH was present in 28 out of 34 (82%) schwannomatosis-related schwannomas. In all eight patients who had previously tested positive for a germline SMARCB1 mutation, this involved loss of the whole, or part of the long arm, of chromosome 22. In contrast, 5 out of 22 (23%) tumours from patients with no germline SMARCB1 mutation exhibited MR. High-resolution Affymetrix SNP6 genotyping and copy number (CN) analysis (Affymetrix, Santa Clara, CA, USA) were used to determine the chromosomal breakpoint locations in tumours with MR. A range of unique recombination sites, spanning approximately 11.4 Mb, were identified. This study shows that MR is a mechanism of LOH in NF2 and SMARCB1-negative schwannomatosis-related schwannomas, occurring less frequently in sporadic VS. We found no evidence of MR in SMARCB1-positive schwannomatosis, suggesting that susceptibility to MR varies according to the disease context.


Pancreas | 2012

EGFR and KRAS mutational analysis and their correlation to survival in pancreatic and periampullary cancer

Melissa Oliveira-Cunha; Kristen D. Hadfield; Ajith K. Siriwardena; William G. Newman

Objectives Pancreatic and periampullary cancers have a high incidence of activating KRAS mutations. The aim of this study was to determine the incidence of KRAS and EGFR mutations in pancreatic and periampullary cancers and their relationship with survival. Methods One hundred patients undergoing pancreaticoduodenectomy or pancreatic biopsy for cancer were recruited. Samples of formalin-fixed paraffin-embedded or fresh pancreatic tissue were obtained. EGFR was analyzed by DNA sequencing of exons 18 to 21. KRAS was analyzed by pyrosequencing of codons 12, 13, and 61. Results EGFR mutations were found in 2 (2.3%) of 88 assessable cases. One in exon 18 (c.1966C>T, p.Q710X) and 1 in exon 19 (c.2066A>G, p.E734G). A synonymous single-nucleotide polymorphism in exon 20 (c.2361G>A, p.Q787) was identified in 57 (67.8%) of 84 patients studied. Twenty-eight (41.2%) of 68 cases harbored a point mutation in KRAS codon 12 (26 cases) and codon 61 (2 cases). The overall median survival was 308 days (range, 7–2623 days). The presence of KRAS point mutations did not significantly alter median survival time (22.8 vs 28.1 months, P = 0.88). Conclusions EGFR somatic mutations are rare in pancreatobiliary malignancies. KRAS mutations are less common than previous reports and do not correlate with survival.


Journal of Medical Genetics | 2010

SMARCB1 mutations are not a common cause of multiple meningiomas

Kristen D. Hadfield; Miriam J. Smith; Dorothy Trump; William G. Newman; D G R Evans

Background Schwannomas and meningiomas are both part of the tumour spectrum of neurofibromatosis type 2 (NF2) and are associated with somatic loss of chromosome 22. They are also found commonly within the general population, unrelated to NF2. Germline SMARCB1 mutations have recently been identified as a pathogenic cause of a subset of familial schwannomatosis cases, and SMARCB1 is a candidate gene for causation of both schwannomas and meningiomas. Recently, Bacci et al reported a germline SMARCB1 mutation associated with familial schwannomatosis and multiple meningiomas. They concluded that SMARCB1 mutations can predispose to multiple meningiomas. Methods We screened the SMARCB1 gene in a panel of 47 patients with multiple meningioma unrelated to NF2. Results We found no germline mutations. Conclusion We conclude that while meningiomas may be associated with the schwannomatosis phenotype, SMARCB1 is not a major contributor to multiple meningioma disease.


Journal of Medical Genetics | 2010

Breast cancer susceptibility variants alter risks in familial disease

Ayse Latif; Kristen D. Hadfield; Stephen A Roberts; Andrew Shenton; Fiona Lalloo; Graeme C.M. Black; Anthony Howell; D G R Evans; William G. Newman

Background Recent candidate and genome-wide association studies have identified variants altering susceptibility to breast cancer. Objective To establish the relevance of these variants to breast cancer risk in familial breast cancer cases both with and without BRCA1 or BRCA2 (BRCA1/2) mutations. Methods A cohort of unrelated individuals with breast cancer due to the presence of either BRCA1 (121) or BRCA2 mutations (109) and individuals with familial breast cancer not due to BRCA1/2 mutations (722) were genotyped using Taqman SNP Genotyping Assays. Allele frequencies were compared with an ethnically and gender-matched group (436). Results A synonymous variant (Ser51) in TOX3 (previously TNRC9) was associated with an increased risk of breast cancer (OR=1.82, p<0.001) in BRCA2 mutation carriers. The associations for FGFR2 (OR=1.20, p=0.046), TOX3 (OR=1.5, p<0.001), MAP3K1 (OR=1.26 p=0.03), CASP8 (OR=0.73 p=0.02) and the chromosome 8-associated SNP (OR=1.31, p=0.004) were replicated in individuals without BRCA1/2 mutations. In addition, homozygote carriers of MAP3K1 variants were shown to have a significantly lower Manchester Score (mean 13.8–17.6, p=0.003), whereas individuals carrying one or two copies of the FGFR2 variant had a higher Manchester Score (mean 17.5–17.9, p=0.01). Conclusions This study confirms that susceptibility variants in FGFR2, TOX3 and MAP3K1 and on chromosome 8q are all associated with increased risk of cancer in individuals with a family history of breast cancer, whereas CASP8 is protective in this context. The level of risk is dependent on the strength of the family history and the presence of a BRCA1/2 mutation and contributes to the understanding of the use of these variants in clinical risk prediction.


Proceedings of the National Academy of Sciences of the United States of America | 2015

MiR-204 is responsible for inherited retinal dystrophy associated with ocular coloboma

Ivan Conte; Kristen D. Hadfield; Sara Barbato; Sabrina Carrella; Mariateresa Pizzo; Rajeshwari S. Bhat; Annamaria Carissimo; Marianthi Karali; Louise F. Porter; Jill Urquhart; Sofie Hateley; James O’Sullivan; Forbes D.C. Manson; Stephan C. F. Neuhauss; Sandro Banfi; Graeme C.M. Black

Significance MicroRNAs are key players in the regulation of gene expression. An understanding of human conditions caused by microRNA mutations provides insight into mechanisms of gene regulation and into the interplay between development and maintenance in tissue homeostasis. The eye represents a notable target tissue of genetic diseases. Inherited retinal degenerations and developmental eye disorders are two separate groups that represent leading causes of blindness worldwide. Identifying underlying genetic causes of such conditions is important for diagnosis, counseling, and potential therapy development. We identified a dominant mutation in microRNA-204 as the genetic cause of a unique phenotype of retinal degeneration and coloboma and thus highlight the importance of microRNA-204 as a master regulator of ocular development and normal maintenance. Ocular developmental disorders, including the group classified as microphthalmia, anophthalmia, and coloboma (MAC) and inherited retinal dystrophies, collectively represent leading causes of hereditary blindness. Characterized by extreme genetic and clinical heterogeneity, the separate groups share many common genetic causes, in particular relating to pathways controlling retinal and retinal pigment epithelial maintenance. To understand these shared pathways and delineate the overlap between these groups, we investigated the genetic cause of an autosomal dominantly inherited condition of retinal dystrophy and bilateral coloboma, present in varying degrees in a large, five-generation family. By linkage analysis and exome sequencing, we identified a previously undescribed heterozygous mutation, n.37C > T, in the seed region of microRNA-204 (miR-204), which segregates with the disease in all affected individuals. We demonstrated that this mutation determines significant alterations of miR-204 targeting capabilities via in vitro assays, including transcriptome analysis. In vivo injection, in medaka fish (Oryzias latipes), of the mutated miR-204 caused a phenotype consistent with that observed in the family, including photoreceptor alterations with reduced numbers of both cones and rods as a result of increased apoptosis, thereby confirming the pathogenic effect of the n.37C > T mutation. Finally, knockdown assays in medaka fish demonstrated that miR-204 is necessary for normal photoreceptor function. Overall, these data highlight the importance of miR-204 in the regulation of ocular development and maintenance and provide the first evidence, to our knowledge, of its contribution to eye disease, likely through a gain-of-function mechanism.


The Journal of Clinical Endocrinology and Metabolism | 2010

Novel Corticosteroid-Binding Globulin Variant That Lacks Steroid Binding Activity

Ilias Perogamvros; Caroline Underhill; D E Henley; Kristen D. Hadfield; William G. Newman; David Ray; Stafford L. Lightman; Geoffrey L. Hammond; Peter J Trainer

BACKGROUND Corticosteroid-binding globulin (CBG) is the principal carrier for glucocorticoids in the circulation and a regulator of their bioavailability. Inherited CBG deficiencies are rarely reported, and only three causative mutations in four families have been described. PATIENTS, METHODS, AND RESULTS In a 26-yr-old female with hypotension, fatigue, and undetectable total serum cortisol at presentation, we have identified a novel homozygous c.776g>t transversion in exon 3 of the CBG (SERPINA6) gene. This results in a p.Gly237Val substitution that is predicted to influence the positioning of two β-sheets that constitute part of the CBG steroid-binding site. Two siblings were also homozygous for the variant, whereas her mother and an unaffected sibling were heterozygous. No other symptomatic family members were identified apart from the proband. Individuals homozygous for the variant had serum CBG levels below the reference range when measured by RIA, but CBG was unmeasurable in cortisol-binding capacity assays. In the same individuals, we observed very low baseline and stimulated total serum cortisol levels but normal free serum and salivary cortisol and plasma ACTH. In a study of ultradian cortisol pulsatility, increased pulse frequency was only observed in the proband. CONCLUSION We describe a novel CBG variant that lacks steroid binding activity. All mutant homozygotes have very low total serum cortisol, but normal free serum cortisol levels. The only biochemical feature to distinguish the symptomatic subject was increased cortisol pulsatility, and we suggest that this may influence glucocorticoid signaling and contribute to symptoms previously associated with CBG deficiency.


Pharmacogenomics | 2009

UGT1A1*28 genotype predicts gastrointestinal toxicity in patients treated with intermediate-dose irinotecan

Roberta Ferraldeschi; Laura J Minchell; Stephen A Roberts; Simon Tobi; Kristen D. Hadfield; Fiona Blackhall; Saifee A Mullamitha; Gregory Wilson; Juan W. Valle; Mark P Saunders; William G. Newman

AIMS Variants in UGT1A1 have previously been associated with toxicity from irinotecan chemotherapy. We conducted a pragmatic prospective cohort study to establish the relevance of UGT1A1 variants in the prediction of severe diarrhea and neutropenia in patients with colorectal cancer receiving irinotecan in a routine clinical setting. MATERIALS & METHODS Genotyping of UGT1A1*28 and c.-3156G>A was undertaken in an unselected, prospective cohort of 96 individuals treated with irinotecan at a single major UK oncology centre. Data on cytotoxic drugs received, and toxicity for all irinotecan treatment cycles were collected from case notes. Over 95% (92/96) of patients received an intermediate dose of irinotecan (180 mg/m(2), twice weekly). Irinotecan was given in combination with other cytotoxic drugs in 93/96 subjects and Grade 3 or 4 toxicity occurred in 23% of subjects. RESULTS No association was found between UGT1A1*28 or c.-3156G>A and neutropenia. However, individuals carrying two copies of UGT1A1*28 (p = 0.04; OR: 14; 95% CI: 1.1-185) or c.-3156G>A (p = 0.03) had a significantly increased risk of diarrhea over all cycles. CONCLUSION Our findings indicate that UGT1A1 genotyping is not a good predictor of hematological toxicity in patients treated with intermediate irinotecan doses. However, it may be useful in the identification of patients at risk of severe diarrhea.


Pharmacogenomics | 2012

Pharmacogenetics of aromatase inhibitors

Kristen D. Hadfield; William G. Newman

Aromatase inhibitors (AIs) are an important class of endocrine drugs used in the treatment of early and advanced breast cancer in postmenopausal women. A number of studies have taken candidate approaches to assess the role of variants in genes encoding enzymes important in AI metabolism, notably CYP19A1 (aromatase), in AI response. These studies have shown conflicting, but interesting, results suggesting that CYP19A1 variants may be important in both the efficacy and toxicity of AIs. A recent genome-wide association study has identified a variant, creating an estrogen response element in TCL1A, which is associated with an increased risk of the musculoskeletal side effects associated with AI use. As breast cancer incidence increases, predictive biomarkers of response to AIs will become more important to ensure the most effective use of endocrine treatments.

Collaboration


Dive into the Kristen D. Hadfield's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stephen A Roberts

Manchester Academic Health Science Centre

View shared research outputs
Top Co-Authors

Avatar

Anthony Howell

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ayse Latif

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Roberta Ferraldeschi

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

D. G. Evans

University of Manchester

View shared research outputs
Researchain Logo
Decentralizing Knowledge