Cancer genetics Flashcards

1
Q

What are cancer cells?

A

Cancers derived from mutated single cells that have acquired the characteristics of continually dividing in an unrestricted invasive manner into neighboring tissues.
Alterations in the DNA sequence of cancer gene attribute towards the abnormal manner of cancerous cells
All cancers result from changes in the DNA sequence of the genome
Genome within our cells are exposed to mutagens (UV radiation) and accumulates mistakes during replication

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

What is the probability of DNA polymerase incorrectly inserting bases?

A

1 in 10,000 bases

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

How many errors result per generation due to DNA polymerase incorrect base insertion?

A

2.5 x10^-8 per generation

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

How does a specific mutation alter the function of a critical gene, subsequently causing cancer?

A

Provides growth advantage to the cell, over-expression of oncogenes, tumor factor genes and inhibition of tumor suppressor genes, results in increased mitotic rate

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

How is a tumor from an over-proliferating cancer cell?

A

Increased mitotic rate causes tumor formation, causing it to invade surrounding tissues, leading to metastasis - secondary tumours

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

What is a benign tumour?

A

A mass of well-differentiated cells that grows slowly, and is capsulated, lacking the ability to invade neighboring tissue, unable to metastasized.

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

What is a malignant tumour?

A

A malignant tumor is not self-limited (evades apoptosis, causes angiogenesis), cells poorly differentiated; the ability to invade adjacent tissues and metastasis. Biopsy analyzed by a pathologist to determine tumor category

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

How many steps is carcinogenesis divided into?

A

Initiation
Promotion
Progression

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

What is the initiation step of carcinogenesis?

A

Irreversible genetic alteration (Tranversion, transitions or deletions in DNA)

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

What is the promotion stage of carcinogenesis?

A

Expression of the genome, mediated through promoter-receptor interactions

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

What is the progression stage of carcinogenesis?

A

Molecular targets during the stages of carcinogenesis include protooncogenes, cellular oncogenes, and tumor suppressor genes, alterations in both alleles of the latter found exclusively in the stage of progression.

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

How does cancer develop?

A

Begin due to the accumulation of mutation involving oncogenes, tumor suppressor genes, and DNA repair genes.

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

Which gene in colon cancer is a tumor suppressor?

A

APC

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

How does a defect in the tumor suppressor gene, APC lead to cancer?

A

It enables excessive cell proliferation. Proliferating cells tend to acquire mutations involving DNA repair genes other tumor suppressor genes (p53) or growth-related genes (K-ras)

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

What is the histology of cancer cells?

A

Abnormal, non-uniform variations in size and shape. Nucleus has a distorted appearance. Cancer cells have to distinguish histological features visible under the microscope. The nucleus is irregularly large, with cytoplasm abnormalities.

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

What is melanoma?

A

Consumption of epidermis, the spread of melanocytes (melanin-forming cells), nests of melanocytes with variable size and shape. Diagnosis of melanoma accurate with biopsy, and analyzed by a pathologist.

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

What is carcinoma?

A

Cancer arising from cells that cover the external and internal body surfaces. Lung, breast, and colon (epithelial)

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

What is a sarcoma?

A

Cancer arising from cells found in the supporting tissues of the body (bone, cartilage, fat, connective tissue and muscle)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What is lymphoma?

A

Cancers arising from lymph nodes and tissues associated with the immune system

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

What is leukemia?

A

Cancers of immature blood cells that proliferate in the bone marrow, with the tendency to accumulate in large number within the bloodstream

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

How many mutagens are approximately present within tobacco smoke?

A

60 mutagens

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

Which molecules typically cause G-T mutations?

A

Polycyclic aromatic hydrocarbons (PAH), and nicotine derived nitrosamines

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

How are heterocyclic amines and PAH formed?

A

Formed during the Maillard reaction at higher temperatures

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

Why are PAHs and heterocyclic amines cancerous?

A

Express DNA-damaging potential upon bioactivation through metabolism, through metabolism. Aryl-hydrocarbon receptor, a transcription factor activated by ligands (PAH), regulates metabolizing enzymes- enzymes cause carcinogenesis by processing the toxicants to reactive mutagen metabolites

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

What are exogenous factors?

A

induction events through lysogenic viruses can integrate genes into the host genome, alterations to the gene sequence of the genome can result in the expression of oncogenes

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

What are the genetic influences of cancer?

A

Hereditary alterations in genes can result in genetic predisposition to cancer

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

How many original hallmarks of cancer are there?

A

6

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What are the 6 main hallmarks of cancer?

A

1) Sustaining proliferative signaling
2) Evading growth suppressors
3) Resisting cell death
4) Enabling replicative immortality
5) Inducing angiogenesis
6) Activating invasion & metastasis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

How do cancer cells sustain proliferative signaling?

A

1) Self-synthesis of growth factor ligands - autocrine proliferative stimulation.
2)Transmit signals that signal normal cells within the supporting tumor-associated stroma, reciprocate by supplying cancer cells with growth factors.
3)Hyperresponseive to growth factors ligand, increased receptor proteins
Cancer cells re-regulate release of growth-promoting signals, signals are conveyed by growth factors that bind cell-surface (intracellular tyrosine kinase domains)
The latter proceeds to emit signals via branched intracellular signaling pathways that regulate progression through the cell cycle, as well as cell, growth

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

How do cancer cells evade growth suppressors?

A

1) Deactivation of tumor suppresor genes (Limit cell growth and proliferation)
2) Defects in the RB pathway permits cell proliferation. RB transduces growth-inhibitory signals. TP53 inputs from stress and abnormality sensors; the degree of damage to the genome is excessive. TP53 can halt cell–cycle progression. The sensation of irreparable damage can trigger apoptosis. This is circumvented

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

How do cancer cells resist cell death?

A

Loss of TP53 tumour suppressor function eliminates critical damage sensor from apoptosis-inducing circuitry
Tumours may increase expression of antiapoptotic regulators BD-2, BD-X or of survival signals (IGf 1.2)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What is the death ligand?

A

Fas ligand

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What does the FAS ligand activate in terms of apoptosis?

A

Activates latent protease (capsizes 8 & 9) proceeds to initiate the cascade of proteolysis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What are the antiapoptotic regulators?

A

bd-2

bd-x

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

What are the pro-apoptotic factors?

A

Bax, Bim, Puma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

How do cancer cells enable replicative immortality?

A

Norma cell lineages can only pass through a limited number of successive growth and division cycles.
Telomerase adds telomere repeat segments to ends of telomeric DNA, expressed in immortalized cells. Extension of telomeric DNA counters telomere erosion, resistance to senescence and apoptosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

What is the definition of senescence?

A

Irreversible non-proliferative state and crisis (cell death). Beyond the stage of the cell crisis cells exhibit unlimited replicative potential

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What is the role of telomeres?

A

Telomeres are involved in the capability for unlimited proliferation.
Telomeres composed of multiple tendon hexanucleotide repeats shorten progressively in non-immortalized cells (losing ability to protect the ends of chromosomal DNAs). Length correlates with the number of successive cells generations

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

How do cells induce angiogenesis?

A

Tumours require sustenance in the form of nutrients and oxygen, as well as an ability to evacuate metabolic wastes and carbon dioxide. Tumor-associated neovasculature generated by angiogenesis.
Angiogenic switch activated during tumor progression causing vasculature to sprout new vessels to sustain neoplastic growths.
Angiogenic regulators include signaling proteins that bind to stimulatory cell-surface receptors displayed by vascular endothelial growth factors. These growth factors orchestrate new blood vessel growth.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

Which molecule activated cell-cell adhesion? (Begins with E)

A

E-cadherin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What is the role performed by E-cadherins?

A

Forms adheren junction with adjacent epithelial cells, maintaining epithelial cell sheets. Increased expression of E-cadherin behaves as an antagonist of invasion and metastasis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What happens to E-cadherin which causes intravasation of cancer cells?

A

Reduction of its expression through downregulation and mutations inactivation causes reduction in cell adhesion.
Enables for increased transit of cancer cells and intravasation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What are the four new hallmarks of cancer?

A

Deregulating cellular energetics
Avoiding immune destruction
Genome instability and mutation
Tumor promoting inflammation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

Which antigens are released by tumour cells?

A

Tumour antigens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

How are cancer cells eliminated by T cells?

A

Tumor antigens bind to antigen-presenting cels, through antigen presentation, naive T cells via TCRs bind to the presented antigen, and are activated. Activated T cells target and eliminate cancer cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

Which proteins downregulate and suppress T cells, promoting self-tolerance?

A

Programmed cell death protein 1 (PD-1) and C279, proteins on cell surface membrane that has a role in regulation in the immune system’s response to the cells of the human body by downregulating the immune system

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What happens during checkpoint pathways in terms of PD-1?

A

PD-1 is activated, negative signals are transmitted to the T-cell, prevents an attack

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

Which ligand is presented on cancer cells which enable them to evade immune attack?

A

PD-L1

49
Q

Which therapy can be used to enable the immune system to recognize and target cancer cells?

A

Nivolumab (Immunotherapy)

50
Q

When is immunotherapy typically used during cancer treatment?

A

Effective treatment to chemotherapy and radiation-resistant cancers (melanomas)

51
Q

What happens during immunotherapy in terms of cancer treatment?

A

Activates a stronger immune response, as well as installing mechanisms for increased recognition and identification of cancer cells.
Checkpoint blockade immunotherapy, inhibits checkpoint pathways, preventing the binding action of PD-1, to PDL-1, thereby activating the T-cell to stimulate tumor degradation.

52
Q

What is a germline mutation

A

A gene change in a body’s reproductive cell, and subsequently incorporated into the DNA of every cell in the body of the offspring either before or during meiosis. Germ cell produce gametes and are exclusive to undergo meiosis as well as mitosis

53
Q

Which cells do germ cells produce?

A

Gametes

54
Q

What are hereditary mutations?

A

Inherited from parents to offspring

55
Q

What are sporadic mutations?

A

Acquired/sporadic mutations are derived during mitosis in cells (non-germline tissues). Somatic mutations are not inherited; alterations in the genome can result in potential cancer
Somatic cells divide by mitosis, mutations in cancer genes accumulate over successive divisions in somatic cells until a sufficient number of accumulative errors can initiate tumor formation
The frequency of genetic events that cause cancer in somatic cells can be influenced by exposure to mutagens. Genetic events are not transmitted to future generations, considering they occur in somatic cells rather than germline variants

56
Q

Which % of cancer are attributed to somatic mutations?

A

90%

57
Q

Which cell line does cancer-predisposing mutations occur in?

A

germline, resulting in inheritance of cancer-causing genes.

58
Q

Which gene leads to an increased risk of breast cancer?

A

BRCA-1 gene

59
Q

Which base transversions are most common in smoking-associated cancers?

A

Guanine to thymine transversions, p53 mutations patterns in lung cancers are distinct between smokers and non-smokers

60
Q

What is the % prevalence of G-T transversions in smoker’ lung cancer?

A

30% in comparison to 12%

61
Q

How does DNA damage caused UV radiation results in signature C>5 or G>A mutations?

A

UV radiation contributes towards the formation of covalent bonds between adjacent pyrimidines (cytosine, and thymine) within DNA molecule. UV-induced mutations

62
Q

Which critical gene when subjected to change can lead to aggressive aberrant cell growth?

A

BRAF, development of malignant melanoma

63
Q

What is a driver mutation?

A

A mutation that provides a selective advantage to a clone in its microenvironment, increasing its survival or reproduction
A minute number of mutations confers this advantage and are currently found in homozygote state
Driver genes are considered to be genes mutated in a greater proportion of cancer samples compared to the expected background mutation rate.

64
Q

What are the two main driver gene mutations?

A

Inactivation of tumour suppressor genes

Oncogenes

65
Q

What is a passenger mutation?

A

Tolerance to multiple mutations by somatic cells (heterozygote state). Passenger mutations have no effect on the viability of a clone however may be associated with clonal expansion because it occurs in the same genome with a driver mutation - hitchhiker

66
Q

Which type of mutation has a tendency to cause clonal expansion?

A

Driver mutations

67
Q

What is the correlation coefficient for the lifetime of developing with how often stem cells in that tissue divide?

A

0.8

68
Q

What is an oncogene?

A

An oncogene is a gene that has the potential to cause cancer. Tumor cells have an elevated proportion of expressed mutated oncogenes

69
Q

What are oncogenes responsible for?

A

Reduction in intracellular survival signals in addition to the expression of proapoptotic factors stimulates cell crisis (apoptosis).
Oncogenes can evade designated apoptosis, resulting in successive proliferation. The predecessor of oncogenes are proto-oncogenes (functional genes involved in cell growth and proliferation, or inhibition of apoptosis).

70
Q

What type of mutation causes oncogene formation?

A

Mutations can upregulate genes for promoting cellular growth (gain of function mutations), and will predispose the cell to cancer and thus termed oncogenes.

71
Q

What are acquired mutations?

A

Cancer-causing mutations involving oncogenes are acquired. Mutations activate oncogenes by chromsome rearrangements, gene duplication or mutation

72
Q

Which chimeric gene translocation causes chronic myeloid leukemic?

A

BCR-ABL

73
Q

How are proto-oncogenes activated?

A

Mutation within proto-oncogene or within promoter region results in altered sequence of mRNA, thereby alteration in the protein structure; either causing an increase in enzymatic activity or loss of regulation. Increase in protein concentration can be caused by an increase in protein expression through mis-regulation an increase of protein (mRNA) stability, prolonging its existence and thus activity

74
Q

Which chromosomes are involved in a Philadelphia chromosomal translocation?

A

Chromosomes 9 and 22

75
Q

Which gene encodes for non-receptor tyrosine kinase ABL?

A

ABL1 gene

76
Q

How is the ABL gene activated?

A

In response to growth factors, cytokines, cell adhesion, DNA damage, oxidative stress and other signals

77
Q

What happens upon ABL activation?

A

Cell proliferation or differentiation, survival, or death retraction or migration

78
Q

Which activity is associated with the BCR gene?

A

Serine/threonine kinase activity, and is a guanine nucleotide exchange factor for RHo family GTPase including RHoA

79
Q

What does the product of the BCR-ABL gene lead to?

A

Has elevated tyrosine kinase activity, relocated to the cytoskeleton and phosphorylates multiple cellular substrates.

80
Q

What do RAS proteins control?

A

Control cellular signaling pathways responsible for growth, migration, adhesion, cytoskeletal integrity, survival and differentiation.

81
Q

Which class of proteins are RAS proteins associated with?

A

GTPases

82
Q

How are RAS proteins activated?

A

In response to various extracellular stimuli. Activated receptors transfer the signal to intracellular RAS protein. RAS is a small G-protein upon signal, RAS protein binds to GTP-activation into the signal-emitting mode.

83
Q

What is the main role performed by RAS proteins?

A

Function as binary molecular switches that control intracellular signaling networks. KRAS is a gene that behaves as an activator/deactivator in cell signaling - functionally controls cell proliferation.

84
Q

Which mutation renders the GTPase domain of Ras insensitive to inactivation by GAP?

A

Residue G12 in the ploop and the catalytic residue Q61. Glycine to valine mutation at residue 12 renders the GTPase domain of RAS insensitive

85
Q

What is GAP in terms of GTPase?

A

GTPase activating proteins

86
Q

What are GAPs?

A

Regulatory proteins that bind to activated g-proteins and stimulate their GTPase activity, with the result of terminating the signaling event

87
Q

How is RAS inactivated?

A

By GAP activity

88
Q

What does GTPase convert?

A

GTP to GDP

89
Q

What is the result of GAP insensitivity?

A

Results in increased and continued intracellular signaling & transcription due to secondary messengers within the cell cycles

90
Q

Which residue is responsible for stabilizing the transition state for GTP hydrolysis?

A

Residue 61

91
Q

What is MEN2?

A

Multiple endocrine neoplasia type 2 caused by an inherited mutation in the gene RET- Patients affected by syndrome develop medullary cancer of the thyroid

92
Q

What does the RET proto-oncogene encode for?

A

A receptor tyrosine kinase (Dominant gain of function) concerns uncontrolled proliferation, impaired differentiation and unrestrained survival of the cancer cell

93
Q

What type of tumors arises from an inherited mutation in KIT?

A

Hereditary gastrointestinal stromal tumours

94
Q

What type of cancers arises from inherited mutations in MET?

A

Hereditary papillary renal cancer

95
Q

What type of cancers arises from CDK4 inherited mutations?

A

Malignant melanomas

96
Q

What is a tumor suppressor gene?

A

Is a gene that regulates a cell during cell division and replication. The gene encodes for a protein that controls cell growth, preventing the excessive proliferation

97
Q

What are the results from mutated tumor suppressor genes?

A

Results in a reduction or loss of function, thereby alterations in the base sequence will inhibit mRNA transcription of the functional gene, and the cell regulator proteins cannot be translated.
A combination with other genetic mutations enables the cell to grow aberrantly in an unrestrained manner. mutations attributed to hallmarks of cancer

98
Q

What are the four main functions of tumour suppressor genes?

A

1) Repression of genes that are essential for continuing of the cell cycle. Lack of genic expression will result in cell cycle disruption a cell division is inhibited
2) Coupling the cell cycle to DNA damage: Damaged DNA sends intracellular survival signals to stimulate DNA repair, senescence or apoptosis. Reparable DNA will enable cell cycle continuation
3) Proteins involved in cell adhesion prevent tumor cells from dispersing block loss of contact inhibition, and inhibits metastasis. Proteins considered as metastasis suppressors
4) DNA repair proteins

99
Q

What is contact inhibition in terms of cancer cells?

A

The process of arresting cell growth when cells come in contact with each other, normal cells stop proliferation, forms monolayer.

100
Q

What are the three main DNA repair proteins?

A

MEN1, HNPCC, & BRCA
An increased mutation rate from decreased DNA repair (ligases) leads to increased inactivation of tumor suppressor and activation of oncogenes

101
Q

What does the two-hit hypothesis of cancer state?

A

The Knudson hypothesis concerns that most genes require two mutations to cause a phenotypic change.
Used to explain the early onset at multiple sites in the body of hereditary retinoblastoma

102
Q

Which case bilateral or unilateral cases are always heritable, in terms of tumour suppressor protein?

A

Bilateral cases

103
Q

Describe the age of onset distribution of bilateral cases

A

Consistent with the presence of a single mutation

104
Q

What is the general inheritance pattern for loss of function mutations that occur in tumor suppressor genes?

A

Recessive

105
Q

What is the first event of the two-hit hypothesis?

A

Inherited germline copy of mutated gene heterozygous insufficient to become cancerous

106
Q

What is the second hit in the two-hit hypothesis?

A

Deleterious mutation to a functional gene within the gene pair occurring somatically -producing cancer

107
Q

What does the two-hit hypothesis state in terms of mutation probability?

A

The hypothesis predicts that the chances for a germline mutation carrier to get a second somatic mutation at any multiple sites in one body cells are much greater than for a non-carrier to get two hits in the same cell

108
Q

What are the main functions of P53?

A

Has a plethora of functions including DNA repair, inducing apoptosis, transcription and regulating the cell cycle. P53 activates transcription of proteins involved in DNA repair

109
Q

What is the role performed by p53 in terms of Mdm2?

A

Regulates the expression of Mdm2 and the genes involved in growth arrest

110
Q

Which syndrome is associated with the inheritance of the p53 gene?

A

Li-Fraumeni Syndrome (LPS) - inheriting one functional copy of the TP53 gene.

111
Q

Why are cancer cells genetically unstable?

A

Stop cell cycling to allow time for DNA repair (p53R2); carry out efficient DNA repair, or undergo apoptosis (Bax, Apadf-1 , PUMA, & NoxA)

112
Q

What is the function of DNA repair genes?

A

Code for proteins that correct mitotic errors and mistakes that may arise during DNA semi-conservative replication prior to cell division

113
Q

What factors is the rate of DNA repair dependent on?

A

Dependent on a plethora of factors, including cell type, cellular age, and the extracellular environment

114
Q

What are the three potential states of cellular dysfunction?

A

1) Irreversible state of dormancy
2) Apoptosis
3) Unregulated cell division and proliferation leading to the cancerous tumor

115
Q

What is the role of the BRCA1 gene?

A

Is phosphorylated by ATM, and CHK2 in response to double-stranded DNA breaks (ionizing radiation potential cause). Phosphorylation results in the activation of molecules
BRCA1 binds to BRCA2 which interacts with RAD51 to form a DNA-repair complex.

116
Q

What are the two categories of cancer viruses?

A

DNA & RNA viruses.

117
Q

How does the lysogenic pathway of viruses lead to cancer?

A

Through the lysogenic pathway, viral DNA is integrated into the host’s genome, viral gene triggers intracellular changes, resulting in cellular proliferation and growth. Integration into host cell chromosomal DNA, stimulate mutations and chromosomal rearrangements that predispose to cancer

Insertion of viral promoter sequence adjacent specific host genes leads to ectopic gene expression resulting in neoplasia
Contain oncogenic strains carrying oncogenes that encode for transforming proteins - influence cell proliferation and differentiation

118
Q

Which virus typically causes cervical cancer?

A
Human papillomavirus (HPV) , causes cancer of cervix and genital warts
99.7% of cervical cancer
119
Q

Which pathogen causes stomach ulcers?

A

H.Pylori