Genomics Flashcards

1
Q

How does different patterns present?

A

Taking a family history can highlight potential patterns of inheritance. While it can be difficult to be sure about this without a genetic diagnosis, there can be important clues in the family history which may point to a specific pattern of inheritance.

In this talk, we will explore some of the common modes of inheritance, and what clues may suggest these modes of inheritance in a family history.

With autosomal dominant inheritance, only one copy of an altered allele is necessary for the condition to be present
There is a 50% risk to children of affected individual
This risk is independent for each sibling
The gene change does not skip generations
Males and females have same risk of inheriting the gene change
Variable penetrance e.g. the likelihood that disease will manifest if the gene change is present

With autosomal recessive inheritance, two copies of the altered allele are necessary for the condition to be present
If a couple are both carriers, there is a 25% chance of each child they have together being affected by the condition
There is a 50% chance the child will be unaffected but will be a carrier of the condition
There is a 25% chance the child will be unaffected and not a carrier
Typically, both parents of an affected child will be carriers
All children of affected individuals will at least be carriers
Unaffected siblings of an affected individual each have a 2 in 3 chance of being a carrier
Males and females have same risk

X-linked inheritance involves a change in a gene that is on the X chromosome
Males with a gene change on their X chromosome will have the condition
Females with a gene change on one of their two X chromosomes are usually called carriers of the condition
Female carriers are not usually affected, although some females may manifest some symptoms (this is disease specific)
Carriers females have the following reproductive risk with each pregnancy:
25% risk of affected male in each pregnancy
25% chance of a carrier female
25% chance of an unaffected male
25% chance of non-carrier female without the condition
50% risk to sons
50% risk of carrier daughters

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What genes are tested in Germline testing?

A

Tumour Suppressor Genes

Protective role in repairing cells during growth
Germline mutations need a ‘second hit’ in the working copy (Knudson’s hypothesis)
In reality often more complex than this

Oncogenes

Promote growth
Often activate during early/embryonic life
If they acquire ‘gain-of-function’ (i.e. ‘activating’) mutations they are termed ‘proto-oncogenes’…..and increase the chance of cancer
E.g. activating mutations in MEN2 as a cause of Multiple Endocrine Neoplasia type 2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What are examples of tumour suppressor gene and how these can result in inherited cancers?

A

Retinoblastoma
RB1
Cell division, DNA replication, cell death

Li-Fraumeni syndrome (brain tumors, sarcomas, leukemia)
TP53
Cell division, DNA repair, cell death

Familial adenomatous polyposis
APC
Cell division, DNA damage, cell migration, cell adhesion, cell death

Breast and/or ovarian cancer
BRCA1, BRCA2
Repair of double-stranded DNA breaks, cell division, cell death

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

How is germline testing done?

A
How is it done? Need ‘germline’ – generally blood - DNA
Gene by gene – traditional approach e.g. test BRCA1/2 in breast/ovarian family; test CDH1 in diffuse gastric cancer family
Panel approach (often used now) e.g. in family with colorectal cancer/polyps test a panel of 15 relevant genes
‘phenotype agnostic approach’ – large panel to test all known cancer predisposition genes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is array CGH and how does it work?When is it used?

A

Array comparative genome hybridisation (aCGH) allows high resolution chromosome analysis and is done using DNA in solution rather than whole chromosome preparations. In principle, DNA from the patient is broken into tiny fragments which are labelled with a fluorescent dye. DNA from a normal control is treated in the same way but labelled with a different coloured dye. Both are mixed together in equal quantities and allowed to hybridise (stick to their matching sequence).

Each fragment then hybridises to a detector array – essentially, known fragments of DNA sequence fixed to a solid support (e.g. a glass slide). If the patient is missing a fragment, more control DNA will bind to the corresponding detector molecule and will give a ‘normal control’ colour signal when exposed to UV light. If the patient sample and control sample are the same, they will give a fusion colour signal; and if the patient has a duplication of a fragment, more patient DNA will bind to the detector and give a ‘patient’ colour signal.

This is a highly automated process that gives reliable results. It has completely changed the way in which we investigate children with learning disability and is changing the world of prenatal diagnosis.

aCGH will in theory detect trisomy, CNVs, microdeletions and smaller ins/dels but it will not detect single nucleotide changes and cannot, at present, detect balanced translocations.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What are the methods of DNA sequencing?

A

A variety of DNA sequencing technologies has evolved over the past 20 years or so which differ from the original technique (known as Sanger sequencing) and are collectively known as ‘next generation sequencing’ (NGS). The key feature of NGS is the ability to read – and re-read – fragments of DNA in solution, very quickly and reliably. See this paper on DNA sequencing technologies for an overview.

Diagnostic laboratories use a range of specific chemical processes and analytical instruments to sequence DNA and the trend is towards larger ‘reads’ all the way up to whole exome (about 1% of the genome, ‘clinical exome’ testing typically covers around 18,000 genes known to be associated with human disease) and whole genome (6 billion nucleotides per diploid genome). Datasets are enormous and require highly specialised ‘bioinformaticians’ to compare the data with ‘normal’ and to identify variants of likely significance. Analysis is usually restricted to ‘panels’ of genes; indeed, most analysis is now done in silico using ‘virtual panels’. It is also possible to undertake ‘agnostic’ analysis which effectively asks the question “is there anything of interest in this exome/genome?” – this is more complex and more likely to generate variants of uncertain significance (see below).

Within the NHS, whole exome and whole genome sequencing, followed by focussed analysis of sequence data, will become the standard ‘genetic test’ for most types of inherited disorder. Some unusual types of mutation – triplet repeat expansions, for example – can also be detected using such technologies, with careful bioinformatic analysis. Such approaches will also become standard for tumour DNA analysis to guide cancer therapy. You’ll learn more about that later in your CDM course.

However, Sanger sequencing still has its uses. It is often used – at present, at least – to verify changes detected using NGS, especially when the clinician intends to offer testing to unaffected members of the family.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are the pro’s and cons of genetic testing?

A

Pros: such testing may clarify a genetic diagnosis, the cause of Samantha’s cancer and other diseases risks for Samantha and her family
Cons: a lot to take in at a difficult time, worry about other cancer risks, worry about family
Some patients would genuinely not decide to go ahead, although this is unusual. The usual reasons cited are that the patients have enough on their plate with their current treatment; others don’t wish to ‘open Pandora’s box’.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What are the pro’s and con’s for whole genome sequencing?

A

We are increasingly living in a world in which people want information. A whole genome sequence offers that possibility:

People may have a previously undetected but significant risk of other health problems for which there may be a preventative or therapeutic intervention.

People who carry recessive disease may wish to understand the reproductive implications if they are thinking about having children.

Pharmacogenomic information may help guide current of future drug treatments.

Such comprehensive analysis can reveal unexpected problems and genomic variants of uncertain significance which can complicate an already challenging health problem. This is certainly an area where informed consent is vital. Always remember that germline information generated in one person is likely to have implications for their close relatives.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What are the types of genetic variants?

A

Whole chromosomes

Aneuploidy: too many or too few copies of chromosomes
Common anneuploidies:
Trisomy 21
Trisomy 13
Trisomy 18
Sex chromosome anneuploidies eg: 45 X0 (Turner’s)
Translocation: one chromosome stuck onto another chromosome

There are no missing or extra parts of the chromsome so this is a balanced translocation
This would not be expected to have a phenotypic effect but their offspring may have an unbalanced translocation
Only a karyotype (like this image) will detect a balanced translocation currently

Copy number variant: large chunks of DNA that are either duplicated or deleted
Identified by SNP array currently
The structure of the genome predisposes some chunks of DNA to be deleted or duplicated
extra or missing copies or genes within that chunk of chromosome
dose effect means that this causes a genetic disease
Both deletions and duplications can be pathogenic

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Why do we need to classify gene variants?

A

The Human Genome Project catalysed massive improvements in sequencing genes, resulting in a paradigm shift in genetics

Before we use to look at the phenotype and select genes for sequencing(Sanger sequencing) based on that
 now we use
Genome sequencing
(or large panel of genes, or exome)
Huge numbers of variants to classify

The huge increase in the number of known genetic variants exposed the fact that
Many variants we thought were pathogenic were too common in the general population – they had been incorrectly assumed to be pathogenic
Many variants are extremely rare but we do not have sufficient evidence to say if they cause disease or not
Overall there is more ambiguity about the disease causing status of variants
A robust and structured approach to classifying variants was introduced to improve consistency in interpretation of genetic variants

Single nucleotide variant: a change in a single nucleotide
Synonymous  less likely to be pathogenic
Missense  ?might be pathogenic
Nonsense
Frameshift high likelihood of being pathogenic
Splice site

Other variants-Insertions or deletions of more than one nucleic acid
Can be any number of nucleic acids inserted or deleted
Triplet repeat
Repeat of an amino acid motif at a particular locus
Can be unstable (i.e. get bigger) on transmission:
Premutation  full mutation
Anticipation
Example: Myotonic dystrophy type 1 (DM1)
(CTG)n repeat in a non-coding part of the DMPK gene
Number of repeats corresponds
with phenotype:

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Whats the significance of a genetic diagnosis?

A

What difference does a genetic diagnosis make for a family?
– Diagnostic label is helpful – ending the diagnostic odyssey, psychological impact
– May alter clinical management
• access to specific treatments or screening
• Stop unnecessary screening/treatment/investigations
• May guide withdrawal of care decisions particularly in paediatric/neonates
– Enrolment in clinical trials or disease registries
– Implications for the wider family
• Offer predictive testing to other family members, or counselling about reproductive options
– Reproductive options
• Selection of embryos or fetuses with or without a specific genetic diagnosis as in prenatal testing or preimplantation genetic diagnosis i.e. used as the basis of decisions about making another human being!

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What clues could you use to determine the clinical significance of a genetic variant?

A

Has it been seen before in association with disease?

What effect is it predicted to have on the gene/protein?

Is it the same kind of genetic change that is usually pathogenic in this gene/disease?

Has anyone done experiments modelling this genetic variant?

Does the phenotype match my patient?

How common or rare is it?

Is it present in other family members?

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What are the Criteria for classifying variants?

A

Phenotype
Do variants in this gene cause the same pattern of problems / family history that my patient has?

Population data
Is this variant common or rare, or totally novel?

In silico or computational data
What do computer programs predict will be the effect of the protein?

Family studies
Testing other family members
Reported in a disease database

Functional data
Experiments to model this gene change e.g. in cells or in animal models (mice, zebrafish etc)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What needs to be considered in in Silico?Computational data?

A

What type of variant is it?
Is the variant expected to affect an important part of the protein (‘domain’)?
Does the variant affect an evolutionarily conserved region
How big is the biochemical difference between the substituted amino acids? (only relevant for missense variants)
Computer prediction programs amalgamate all of the above data

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What computer predictive programmes can be used and hoe accurate are they?

A

PolyPhen and SIFT are commonly used prediction programmes
These are not as robust as we would like, and can be conflicting and even wrong
Use as part of a comprehensive assessment of a variant, not in isolation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What are the benefits and harms of population screening?

A

benefits

Early detection of disease allows for earlier more effective treatment
Identifies at risk individuals so preventive measures can be put in place
Identification of carriers of heritable conditions allows for informed family planning
Increased awareness of own health allowing for lifestyle changes
Control of disease at a population level
Cost-effective

Potential harms

False positive/negative results
Test may be invasive
Insurance implications
Psychological implications

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What is the national breast screening program and when is it done?

A

Most common cancer in the UK
A woman’s lifetime risk – 1 in 8
Around 55,000 people are diagnosed each year (including 400 men)
Just over 80% occur in women over 50 years
National Breast screening programme
Most cases of breast cancer are sporadic
Less than 10% is caused by a germline mutation that increases risk of developing breast cancer

Population screening programme
Offered to women from the age of 50 to 71st birthday every 3 years
May be eligible before the age of 50 if individuals are at a higher risk of developing breast cancer
Women who are 71 or over are still eligible for screening but must request

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What is the national bowel screening program and when is it done?

A

Third most common cancer in the UK
1 in 14 men and 1 in 19 women will get bowel cancer during their lifetime
Mostly sporadic
1 in 4 individuals will have a family history
5-6% inherited e.g. Familial adenomatous polyposis (FAP), Lynch syndrome

Population screening programme
Offered to individuals aged 55 and over
One-off bowel scope screening test offered at age 55 (if available in the individual’s area)
Individuals aged 60-74 offered home testing kit (faecal occult blood test) every 2 years
Individuals 75 or over can request a home testing kit every 2 years

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q
  • Whats the diagnostic criteria for Lynch syndrome?
A

Unaffected carriers

BRCA1

Breast cancer:
Lifetime risks (to 80 yrs): 60-90%
Ovarian cancer:
Lifetime risk 40-60%
Majority of lifetime risk conferred after age of 40
Male breast cancer:
Lifetime risk ~0.1-1%
Prostate cancer:
Lifetime risk similar to population risk ~8%

BRACA2

Breast cancer:
Lifetime risks (to 80 yrs): 45-85%
Ovarian cancer:
Lifetime risk 10-30%
Majority of lifetime risk conferred after age of 50
Male breast cancer:
Lifetime risk ~5-10%
Prostate cancer:
Lifetime risk significantly increased to ~25%

Affected carriers

BRCA1
Breast cancer:
Lifetime risk contralateral breast cancer ~50%
Overall 5 year risk ~10%

BRCA2
Breast cancer:
Lifetime risk contralateral breast cancer ~50%
Overall 5 year risk ~5-10%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

When Should BRACA testing be offered?

A

Individual affected with breast cancer:
In general BRCA1/2 is indicated in a person affected with breast cancer where the Manchester Score is 15 or above
The Manchester Scoring System (MSS) allows the calculation of the probability for the presence of mutations in the BRCA1/2 genes in families suspected of having hereditary breast and ovarian cancer
The MSS, is used alongside the family history:
Scores are added for each cancer
A Manchester score of 15 is equivalent to a 10% threshold for finding a BRCA variant
Individuals with a grade 3 triple negative breast cancer <50 should also be offered BRCA testing
Testing for the 3 Ashkenazi founder mutations offered to women with breast cancer and Ashkenazi ancestry
Full analysis offered when Manchester Scoring is 15 and above
Newer genes such as PALB2 can also be tested
P53, Stk11, CDH1, PTEN can also be considered if appropriate

Individual unaffected with breast or ovarian cancer:
BRCA1/2 testing is indicated in an unaffected person where there is no living affected person available for testing. The Manchester score should be 20 or above
The individual should have a 1st degree relative affected with a relevant cancer
Ideally testing should be offered to an affected individual where there is a greater chance of identifying a mutation
Testing for the 3 Ashkenazi founder mutations offered to women with breast cancer and Ashkenazi ancestry. Full analysis of offered when Manchester Scoring is 20 and above
Testing of other genes e.g. PALB2, P53, Stk11, CDH1, PTEN can also be considered if appropriate

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What is Lynch syndrome?

A

Lynch syndrome, also known as hereditary non-polyposis colorectal cancer (HNPCC), is caused by mutations in one of several DNA mismatch repair (MMR) genes; MLH1, MSH2, MSH6 and PMS2.
Carriers*:
Men – up to 80% risk of colorectal cancer
Women – up to 70% risk of colorectal cancer and 60% endometrial cancer
* Exact risk will depend on the gene

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is predictive testing and when is it done?

A

Predictive testing for known pathogenic variants should be offered to all families where a pathogenic variant has been identified. This is testing in individuals who are not yet affected by the condition.
In some conditions, risk management options are available to individuals who test positive.
For adult-onset conditions, testing is typically offered from the age of 18. Therefore in cancer predisposition syndromes, testing in children is not usually considered as onset is typically in adulthood and surveillance is not normally recommended before the age of 25/30.
Exceptions to this include FAP and TP53 where earlier surveillance may be possible.
Typically predictive testing is offered to first degree relatives in the first instance. If they test positive, then their first degree relatives would then be eligible for predictive testing. If they test positive, their first degree relatives would be eligible for testing, and so on.
It is important that individuals understand the implications of predictive testing. While it is different in some ways to diagnostic testing, it is equally important that patients are fully informed, and are aware of potential medical, psychological, familial and financial implications of genetic testing.

Who is eligible for predictive testing

Typically, all first degree relatives of individuals who have a gene change identified in them will be offered predictive testing.
Therefore, if a gene change is identified in Samantha, both of her parents, and her brother and sister would be eligible for predictive testing.
If they test positive, then their first degree relatives would be eligible for testing. For example, if her mum tested positive, her maternal aunt would then be eligible for testing.
Individuals who test positive may then be eligible for risk management options themselves.
Individuals who test negative are usually considered to be at population risk. Their descendants are also ‘off the hook’.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

How does result of genetic tests affect insurance?

A

The ‘Code on Genetic Testing and Insurance’ is an agreement between the Government and the Association of British Insurers (ABI)
Outlines what an insurance company needs to know about testing, and how they should act:
To not require or pressure you into a predictive or diagnostic genetic test
To not ask for or take into account the result of a predictive genetic test if you are applying for insurance (the only exception being if you are applying for life insurance over £500,000 and you have had a predictive genetic test for Huntington’s Disease)
Avoids patients with positive result facing discrimination from insurance companies
However, insurance companies can put two and two together by asking, ‘do you have family history of cancer’, ‘have you had an outpatient’s appointment’ and increase premiums
Advise patients before undergoing genetic testing to consider getting life insurance/mortgage insurance in place, as answering questions with knowledge they had at that time

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What are the factors contributing tio increased breast cancer risk?How can patients risk of developing can classified?

A

Risk factors

Family history
Smoking
Obesity
Alcohol
Lack of exercise
Nulliparity or first pregnancy >30
Early menarche (<12) and/or late menopause (>55)
Current or previous HRT use
Previous radiotherapy
Previous breast cancer
(Prolonged breast feeding is protective)

Risk stratification

can be done based on genotypes and pheotype

The genetics service aims to provide risk assessments for family members and further screening recommendations. An initial family tree is compiled and diagnoses are confirmed via cancer registries or medical records. Genetic testing may also be performed. From this information, we can classify cases into different types of risk categories from very high to low and determine appropriate further management.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What genetic testing is done in breast cancer?

A

Current options
Single mutation (predictive) test – if known mutation in the family
Standard panel test – BRCA1/BRCA2/PALB2 – if fhx breast/ovarian cancer meeting testing criteria of 10% chance of finding a mutation
Bespoke panel test – by adding other relevant genes to above panel

Future options
Large panel testing
Polygenic risk scores

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What are some high pentrance breast cancer genes

A

BRCA1 and BRCA2
PALB2

TP53 Li Fraumeni(Brain tumors,leukemias,sarcomas)
PTEN Cowden(microcephaly,autism,endometrial,kidney,thyroid ca,bengin skin lesions)
CDH1 Hereditary Diffuse Gastric Cancer(lobular breast cancer)
STK11 Peutz Jeghers

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What are other associations of BRACA genes?

A

Only a part of the story of inherited breast cancer
……but in many ways the most important part

The most common single gene cause of breast cancer – implications for screening and prevention; associated with ovarian cancer risk (largely preventable with RR BSO)

Other cancers are associated – more so with BRCA2, and especially male breast, prostate, pancreas

Lifetime breast and ovarian cancer risks higher with BRCA1 compared with BRCA2

28
Q

What are the Management options for families at increased breast cancer risk

A

Check notion for screening and risk reduction

Chemoprohylaxis

Premenopausal women
Tamoxifen 5 years

Post menopausal women
Anastrozole 5 years

29
Q

What are the Pro’s and Cons of risk reduction surgery?

A

Pros
Only effective way of dramatically reducing risk (takes away 90-95% of prior risk)
May be able to be combined with cancer surgery if undergoing treatment for a primary cancer
No screening needed thereafter

Cons
Risks of surgery including reconstruction options – autologous vs implant
Psychological implications
May be an ‘unnecessary’ major procedure - may never have gone on to develop breast cancer
Some residual risk of breast cancer remains

30
Q

What causes Lynch syndrome?

A

LS accounts for approximately 3.3% (1 in 30) of colorectal tumours, and the condition is estimated to lead to over 1,100 colorectal cancers a year in the UK. An estimated 175,000 people in the UK have Lynch syndrome, a large proportion of whom will be unaware that they have the condition. Autosomal dominant

Caused a mutation in one of four
‘mismatch repair’ genes
(MLH1, MSH2, MSH6, PMS2)

Core Lynch cancers are BOWEL
and ENDOMETRIAL

31
Q

How can Lynch syndrome be diagnosed?

A

Clinical (‘Amsterdam’) criteria - 3,2,1 - (3 Lynch tumours, 2 successive generations, 1 diagnosis <50)

Now, NICE recommends tumour testing on ALL COLORECTAL TUMOURS to look for evidence of mismatch repair deficiency

Check notion for pathway

32
Q

What other single gene conditions are associated with increased risk of bowel cancer?

A

Inherited mutations in the APC tumour suppressor gene
Classical FAP: 1000s of colonic adenomas; 95% penetrant by 35; Malignancy risk ~100%
Attenuated FAP: ~10-100 adenomas
Extracolonic features: Gastric fundus polyps, Duodenal polyposis, CHRPE, desmoid tumours, osteomas

Recessive
MUTYH (Recessive mutations cause aFAP like condition)

Dominant
Peutz-Jeghers syndrome (STK11 gene) – perioral hyperpigmentation; GI polyposis, other malignancy risk
Juvenile Polyposis (e.g. SMAD4 gene) – GI polyposis, features of HHT with SMAD4

33
Q

What is Dysmorphology?

A

Dysmorphology = Recognition and study of birth defects and syndromes
Malformation: morphological abnormality present at birth and of prenatal origin, it can involve a single organ or a body part and arises because of an abnormal developmental programme.

-> a malformation is a structural birth defect

Syndrome: condition characterised by a set of associated symptoms with a known or assumed single aetiology.

“syndrome” = from the Greek “running together”

34
Q

How can structural defects be calssified?

A

Structural defects can be classified based on their clinical impact, on the pathogenesis (mechanism) or based on recognisable patterns.

Recognising which group they fall into helps us to clarify the need for further investigations and helps us to make a diagnosis

CLINICAL IMPACT
Normal variant
Minor
Major

Normal variants
relatively frequent morphological characteristics with no medical/pathological impact
> 4% of general population
Minor anomalies
structural anomaly that don’t cause significant clinical disease, functional abnormality or cosmetic problems.
No impact on life expectancy or quality of life.
Not frequent (< 4% )
Structural anomaly causing significant clinical, functional or cosmetic problems.
Impact on life expectancy and/or quality of life.

PATHOGENESIS
Malformation
Malformation: morphologic abnormality that arises because of an abnormal developmental process.
It is a primary defect

Deformation
Deformation: distortion by a physical force of an otherwise normal structure.
It is a secondary defect

Dysplasia
Dysplasia: abnormal cellular organisation within a tissue resulting in structural changes/abnormal growth of a tissue.
It is a primary defect

Disruption: destruction of a tissue that was previously normal
It is a secondary defect

RECOGNIZABLE PATTERNS
Syndrome
Syndrome:
A pattern of anomalies known or thought to have the same cause (e.g. Noonan syndrome- PTPN11).

Sequence
Sequence:
morphologic anomalies resulting from a single primary malformation (e.g. Potter sequence; Pierre Robin)

Association
Association:
A pattern of at least 2 anomalies that occur together more often than expected by chance. No common cause identified (e.g. VACTERL).

35
Q

What are the aims of dysmorphology?

A

Diagnosis
Prognosis and follow-up (are any other potential problems?)
Recurrence risk (appropriate counseling for parents and relatives)
Family planning and pregnancy management (PND/PGD?)

36
Q

Why is a diagnosis needed?

A

end of uncertainty
end of unecessary investigations (costly and time consuming for family and healthcare),
better management
prevention of possible complications.

37
Q

What is the significance of PTN11 c.173A>G? and what does it cause?

A

Replaces asparagine with serine at codon 58 of the PTPN11 protein (p.Asn58Ser).
The asparagine residue is moderately conserved
There is a small physicochemical difference between asparagine and serine.
This variant is present in population databases (rs751437780, ExAC 0.003%).
This variant has not been reported in the literature in individuals with PTPN11-related disease.
Algorithms developed to predict the effect of missense changes on protein structure and function do not agree on the potential impact of this missense change (SIFT: “Tolerated”; PolyPhen-2: “Possibly Damaging”; Align-GVGD: “Class C0”).
Variants that disrupt the p.Asn58 amino acid residue in PTPN11 have been observed in affected individuals (PMID: 15956085, 16804314, 19125092, 25914815, 16263833). This suggests that it is a clinically significant residue, and that other variants that disrupt this residue are likely to be causative of disease.
In summary, the available evidence is currently insufficient to determine the role of this variant in disease.
Therefore, it has been classified as a Variant of Uncertain Significance

NOONANS Syndrome
Autosomal dominant genetic condition
Short stature
Distinctive facial features (broad forehead, hypertelorism, downslanting palpebral fissures, a high-arched palate, and low-set, posteriorly rotated ears)
Congenital heart defects (pulmonic stenosis, hypertrophic cardiomyopathy, others)
Other possible features: bleeding tendency (thrombocytopenia), hearing loss, chest deformity (pectus excavatum), scoliosis, kidneys anomalies

38
Q

What are the autosomal recessive diseases and what is their risk of inheritance?

A

CF
Carrier frequency in Northern European population approximately 1 in 25
CFTR gene provides instructions for the production of the Cystic Fibrosis Transmembrane conductance Regulator (CFTR) protein, a cell membrane chloride transporter

Tay sachs
HEXA gene encodes part of the beta-hexosaminidase A enzyme which plays a key role in the breakdown of toxic products in the brain and spinal cord.

Thalasemia

Carrier couples have a 1 in 4 chance of having an affected child

Unaffected children have a 2 in 3 chance of being a carrier

If one parent is a carrier each child has a 1 in 2 chance of being a carrier

Siblings of a carrier have a 1 in 2 chance of being a carrier (presuming no one in the family is affected)

All offspring are obligate carriers if one parent is affected and the other parent isn’t a carrier

If the other parent is a carrier offspring have a 1 in 2 chance of being affected

All offspring are obligate carriers if one parent is affected and the other parent isn’t a carrier

If the other parent is a carrier offspring have a 1 in 2 chance of being affected

39
Q

What is Consanguinity and how is it inherited?

A

A consanguineous relationship is one between individuals who are second cousins or closer
More common in parts of the Middle East, South Asia, certain religious communities and amongst Irish travellers
Prevalence of serious congenital and genetic disorders for children of first cousin parents is approximately double the prevalence in children of unrelated parents (8% compared to 4%)

40
Q

When is Carrier testing offered?and how is risk calculations done?

A

Initially offered to first degree relatives
Main indication is to find out the chance of having an affected child
Specifically test for the mutation identified in the family
For children testing should be deferred until old enough to consent

Approach depends on:

Prevalence and carrier frequency of the condition
Note carrier frequencies differ significantly between ethnic groups

Does the gene have common mutations which account for a large proportion of cases?
Is there a biochemical test which can identify carrier status?

When it’s not straight forward
De novo mutations
Chromosome abnormalities
Rare or unidentified mutation not picked up on carrier testing
Counsel that carrier testing can reduce the risk of a child being affected but cannot exclude the possibility

41
Q

what is extended carrier testing?

A

Extended carrier testing
Gene panel looking for pathogenic variants in hundreds of genes associated with severe recessive conditions

Based on the cumulative burden of disease associated with individually rare AR disorders
Estimate of AR disease burden: 1.7 in 1000 individuals
Previous research has suggested that each individual is a carrier of a pathogenic mutation in >20 genes associated with AR conditions or traits

Primary objective is to inform couples and enable autonomous family planning

42
Q

What reproductive options are available?

A

Check notion

43
Q

What is Preimplantation genetic diagnosis?What are the advantages and disadvantages?

A

Assisted reproduction with embryo biopsy(5-7 day) to screen embryos for a specific genetic condition prior to implantation

Testing performed following embryo biopsy may be a FISH analysis, SNP array or testing for a known mutation in a single gene disorder

Regulated by the Human Fertilisation and Embryology Authority (HFEA)- list of approved conditions
Common conditions that are reffered to PFD-(dominant-Huntigtons,Myotonic dystrophy,spinal muscular atrophy,recessive-CF,Spinal muscular atrophy. X linked-Fragile X,Duchenne)
Eligibility criteria based on the criteria for NHS IVF treatment
Advantages:
Avoid difficult decisions following testing during a pregnancy
Fertility treatment
Other Considerations:
Success rate (1 in 3 cycles estimated to result in a baby)
Length of treatment(8-18 months)
Travel
Side effects: Ovarian Hyperstimulation syndrome
Multiple pregnancy

44
Q

What are the non-invasive prenatal testing options?

A

Analysis of cell-free fetal DNA (cffDNA) fragments circulating in the maternal bloodstream

Fetal DNA accounts for approximately 10% of cell free DNA in the maternal bloodstream

cffDNA detectable from 4 weeks but reaches levels sufficient for analysis from 10 weeks

NIPT: Non-invasive Prenatal Testing for aneuploidies
NIPD: Non-invasive prenatal Diagnosis

45
Q

What are the Applications of NIPD and NIPT?

A

Determination of fetal sex
X-linked disorders
Disorders with gender specific phenotypes (e.g. Congenital Adrenal Hyperplasia)

Testing for a specific mutation in a single gene disorder
De novo mutation identified in a previous pregnancy or affected child
Known paternal mutation in a dominant disorder
Known paternal mutation in a recessive condition where both parents carry a different mutation

Aneuploidy testing based on DNA molecule counting

46
Q

What are the limitations of NIPT/NIPD?

A

Maternal factors
Some women have very low fetal DNA fractions(DNA process and amplification,nmaternal weight ,gestational age)
Underlying chromosomal abnormality or tumour may affect results

Pregnancy factors
Twin pregnancies
Resolving second gestational sac
Placental mosaicism

47
Q

What factors affect a patient descion regarding reproductive options

A
Family circumstances
Previous experience of the condition
Religious views
Individual beliefs and opinions
Health
Geographical factors
Fertility
Financial factors
Age
48
Q

How could genetic testing improve the management of patients who require warfarin?

Does genetic testing making a clinical difference for warfarin prescription?

A

Pharamacogenic testing can improve patients response to warfarin with longer periods of therapeutic INR however it doesn’t make a clinical difference in the first 4 weeks of treatment

49
Q

What is warfarin?
What are its clinical uses?
What are possible side effects and adverse effects?
How do you start a patient on warfarin therapy?
What problems might there be?
What health and cost implications are there for the individual and the health service?

A

Wafarin is an anticoagulant drug which inhibit vitamin K dependant clotting factors by vitamin K antagonism

Clinical use-

Prophylaxis of embolisation in rheumatic heart disease and atrial fibrillation

Prophylaxis after insertion of prosthetic heart valve

Prophylaxis and treatment of venous thrombosis and pulmonary embolism

Transient ischaemic attacks

Side effects

Haemorrhage

Blue toe syndrome; CNS haemorrhage; diarrhoea; fever; haemothorax; jaundice; pancreatitis; skin necrosis (increased risk in patients with protein C or protein S deficiency); skin reactions

Should not be given in the first trimester of pregnancy.

Frequency not known

Alopecia; nausea; vomiting

Heparin cover given when starting warfarin as it delay to start can result in an initial procoagulant effect

Can cause skin necrosis

50
Q

What is DPYD testing and what is its use?

A

DPYD testing

A very recent example of a pharmacogenomic test to be incorporated into routine clinical practice is testing of the DPYD gene. This is indicated when a clinician is considering prescribing fluorouracil-based chemotherapy. Inherited alterations in DPYD can result in deficiency of the enzyme encoded by this gene, and result in severe toxicity to drugs like 5-fluorouracil. This group of drugs may then be contraindicated in a particular patient, or more commonly a dose reduction can be recommended.

51
Q

What are the advantages and challenges of setting up a new pharmacogenomic test like this which quickly becomes ‘standard of care’ nationally?

A

hings to think about include issues such as:

The strength of evidence
The cost
Consent: who will take consent?
Where do such tests fit in a clinical pathway?
Education: introducing new tests always requires a degree of cultural change and education.

52
Q

What can pharmacogenetics be used for?

A

Tailor individual treatment

  • Drug doses
  • Side effects

When all option are exhausted identify mutation and change treatment

53
Q

What are common features in seen in different trisomy’s?

A

Down syndrome is typically associated with heart defects, most typically atrioventricular septal defects.

Cleft palate is more typical of trisomy 13.

Diabetes mellitus is seen in Down syndrome, but not as frequently as heart defects.

Polydactyly is more typical of trisomy 13

54
Q

How is Myotonic dystrophy inherited?

A

Myotonic dystrophy is an autosomal dominant disorder caused by triplet repeat expansion.

One feature of triplet repeat expansion disorders is genetic anticipation, in which the disease becomes more severe, or starts at any earlier age, with each successive generation.

The risk to Ahmad and Zara’s children is ½ (it is autosomal dominant)

Myotonic dystrophy is an autosomal disease, not X-linked.

Prenatal diagnosis would require triplet repeat expansion testing which is beyond the resolution of comparative genome hybridisation.

Large expansions are a risk if the mother is affected. Large expansions can manifest with fetal akinesia and polyhydramnios. Zara’s pregnancy is at low risk because Ahmad is affected, and a large expansion is unlikely in his children.

55
Q

How can oncogenic disorders inherited?

A

This is a proto-oncogene, like RET. Pathogenic variation can be predicted to result in an oncogene, which will drive cell growth.

No-one with a pathogenic variant in any cancer gene will inevitably develop cancer.

Proto-oncogenes are active in embryonic life and usually dormant in postnatal life.

Oncogenes do not involve the mismatch repair pathway, so microsatellites are unchanged.

Oncogene disorders are usually autosomal dominant.

Oncogenesis involves ‘switching on’ a normal allele by activating mutation.

56
Q

What chromosomal aneuploidies is incompatible with survival to birth?

A

In general, trisomies 13, 18 and 21 may survive to term. Of the sex chromosome aneuploidies, the one most likely to fail early on is 45,Y. 45,X (Turner syndrome) often also fails early on but is more likely to survive to term.

57
Q

What testing is used for

Di George Syndrome

Marfan’s Syndrome

Turner’s Syndrome

Lynch syndrome

Cystic fibrosis

A

Di George syndrome is a chromosome microdeletion syndrome; MSI testing is a somatic test of tumour tissue.

Marfan syndrome is an autosomal dominant disorder caused by pathogenic variation in the FBN1 gene which is beyond the resolution of CGH.

Turner syndrome is diagnosed by chromosome analysis. Whole genome sequencing detects point mutation and small-scale dosage abnormalities.

Lynch syndrome is cause by defective mismatch repair which is detected using MSI analysis.

Cystic fibrosis is caused by pathogenic variation in DNA sequence which is beyond the resolution of CGH.

58
Q

What is the founder effect?

A

Relatively isolated populations in which one of the early founders of the population happened to have a genetic condition

59
Q

What is heterozygote advantage?

A

Increased probability of survival if you are a carrier of a specific disease.
A small advantage over many generations can concentrate some recessive genes in a population.

Genes survive over thousands of generations only if they confer some advantage to the species.
About 1 in 20 caucashianscarry gene for CF.
Being a CF carrier is believed tohavemade it slightly more likely that someone would survive an episode of dysentery (a common problem in mediaeval Europe).

In theneolithicperiod it is believed that a large proportion of the population became iron deficient (diet, threadworm infestation). The increased iron absorption associated with haemochromatosis C282Y carrier status is likely to have offered a survival advantage.

Thalassaemia carriers are believed to survive episodes of malaria, which has been a much greater problem in Asia than Northern Europe!

60
Q

What is expanded carrier screening?

A

Recent developments in low cost, rapid DNA sequencing mean that expanded carrier screening for severe recessive disorders is now possible and, in some countries, routine. Up to 500 different diseases can be covered in a single test.
Such testing is commonplace in the private sector* and may be available as an NHS service in the future.

  • look at the products offered by large American labs such as Invitae, Centogene and Fulgent
61
Q

What are the potential clinical and ethical issues raised by the introduction of expanded carrier screening?

A

Autonomy
The results may provide families with more informationin order forthem to make more informed decisions either before or during a pregnancy, which is a good thing.
Testing doeschallengesthe concept of informedconsent when the carrier testing is for many different conditions

Benificence
Testing potentially spare a child a life with a serious hereditary disease(is that a good thing?)
Or maybe it allowsparents to be better prepared to care for a child with a serious hereditary disease.

Non-maleficence
Easespsychological stress for a couple
Removes the psychological impact on individuals living with aserious hereditary disease
But maybe it creates a potential societal stigma for couples who choose not to have carrier testing and have a child affected by an autosomal recessive condition: could they have done more to prevent it?

Justice
Reduced incidence of recessive conditions associated with high socioeconomic costs (acontroversialeconomic argument!)

62
Q

What other types of cancer are patient with FAP,BCRA2,NF1,CDH1 mutations at risk?

A

Thyroid cancer assoicated in Gardeners syndrome (subtype of FAP). Characterised by polyps in the bowel andextracolonic tumors may includeosteomasof the skull,thyroid cancer, epidermoid cysts,fibromasas well as the occurrence of desmoid tumors in approximately 15% of affected individuals.

BRCA2- prostate cancer, breast, ovarian, pancreatic,NF1- NEUROFIBROMATOSIS, autosomal domiant disorder causing tumours in the nervous system
CDH1- gastric cancer

63
Q

What direct-to consumer genetic testing?

A

Genetic testing tends to be requested by a doctor or genetic counsellor following a consent process during which the consumer has the opportunity to discuss the pros and cons of testing.
This generally applies to both somatic genetic testing (which can inform cancer care, as we have seen) and germline testing (which is focussed on rare Mendelian disease).
Sample collection, DNA extraction, the technical process of DNA analysis and the scientific process of reporting are all undertaken by quality assured diagnostic laboratories.

Direct-to-consumer genetic testing is different in a number of ways:
Tests are marketed via a range of web- and print-based media, including social media platforms; they can even be bought over-the-counter

DNA sample are typically collected using saliva kits and sent directly to the testing laboratory by the consumer.
Results are uploaded to a secure website or provided in a written report format.

64
Q

What do direct-to-consumer genetic tests offer?

A

Disease risk prediction

Several companies market tests which claim to estimate the consumer’s risk of developing a number of ‘common’ conditions such as:

Dementia / Alzheimer’s disease
Inflammatory bowel disease
Type II diabetes
Coeliac disease
Autoimmune disease
Parkinson’s disease

Many of these common disorders are multifactorial.

Direct-to-consumer genetic testing can only provide information about genetic variation and may overlook important factors such as diet, smoking and family history.

Such factors would be considered by a doctor or genetic counsellor – but are not usually addressed by consumers of direct-to-consumer genetic tests.

Carrier status

Some companies are able to test for carrier status for some common autosomal recessive conditions such as cystic fibrosis, sick cell disease and many different rare disorders.

One popular ancestry DNA site offers testing of 44 different rare recessive disorders:

Mendelian disease diagnosis

Many companies offer diagnosis for Mendelian diseases, but this is usually only undertaken following genetic counselling.

Lifestyle testing

Such tests are marketed with the aim of providing information about a range of lifestyle factors and often link to the targeted marketing of lifestyle products or services, for example:
Fitness and exercise capacity
Nutrition, metabolism and weight
Sleep preferences
Skin care (
Taste – even which wine you should prefer

Ancestry

Some very popular services provide data on ancestral origins and can even link you to other consumers who share DNA sequence with you.

Such products are based on statistical prediction programmes which compare a large number of genetic markers with ‘standards’ from different areas of the world. As such they are based on current population distributions rather than reference DNA samples extracted from ancient tissues like bone or teeth.

My Neanderthal genotype

Segments of our H.sapiens genome are believed to have been introduced through interbreeding with H.neanderthalis. The same is true for the more recent, but also archaic, Denisovans.

65
Q

What are the issues with privacy in direct to consumer testing?

A

Privacy

Genetic data are personal and private. Consumers need to be reassured that their data will be stored safely and to know whether their it will be used for research or commercial purposes (marketing).

They should understand issues like:
What does the company do with the consumer’s DNA sample once the analysis is complete? Will it be stored, shared, sold, or destroyed?

Who owns the consumer’s genetic data? Is it the company? Do have a right to decide how the data are used?

Can the consumer delete their data?

What are the company’s data security measures? How are the data stored? Can the data be accessed for unauthorised use?

Does the company use your information for internal research or other ‘secondary purposes’?

Does the company share the consumer’s data with, or sell it to, pharmaceutical or biotechnology companies, academic institutions, or nonprofit organizations? If so, is it identifiable? Is the consumer informed? Can the consumer opt out?

Does the company update consumers if it changes its privacy policy?

What would happen to the consumer’s data if the company goes out of business or is sold?

There are concerns about the possibility that DNA data may be accessed by lawful disclosure, as part of the investigation of a crime (for example).

Many companies now provide explicit information about this. Data which is passed on to third party interpretation services (see next section) could also in theory be accessed by law enforcement agencies and may be more liable to being accessed unlawfully.

66
Q

What is reanaylsis?

A

Some direct-to-consumer genetic testing companies – particularly ancestry DNA companies – will allow you to download your raw data, although most urge caution in the way in which these data are used. Not all of the 600,000 or so SNPs will have been used in the original analysis.

Most people would never be able to analyse this data on their own. A number of third party services have developed which off re-analysis and re-intepretation of your raw data, often for a fee (look here for some examples). These third party services offer more information about disease risks or lifestyle traits, for example.
These services are not risk-free, however:
The original DNA analysis often includes false positive findings, which are carried through to re-analysis.
In one recent study, 40% of reported findings were not confirmed in a clinical laboratory.
Once the consumer has downloaded their raw data file, it is no longer protected by the original service’s privacy policy.
Third-party re-interpretation services are usually unregulated.

67
Q

What are the Pros and Cons of Direct to Consumer genetic testing?

A

Pros
Sample collection is easy and usually doesn’t require a blood sample.
The range of tests can be attractive to the consumer and is often much broader than they could access through a clinical service.
Results are generally returned quickly, direct to the consumer.
The consumer can feel more in control of their genetic testing.
Consumers are often given the opportunity to add their data to company databases to be used in research and product development

Cons
The cost is borne by the consumer.
The technology may not detect all variants in any given gene. There is a risk of false positives and false negatives.
Results may give false reassurance (e.g. BRCA testing).
Evidence linking a specific genetic variant with a disease may not be scientifically robust. Health risk testing is not predictive.
Ancestry testing may give misleading information.
This are largely unregulated services.
Genetic privacy might not be guaranteed.