BIOL 325 LECTURE 14 MOLECULAR ONCOLOGY STUDY GUIDE

0.0(0)
studied byStudied by 0 people
learnLearn
examPractice Test
spaced repetitionSpaced Repetition
heart puzzleMatch
flashcardsFlashcards
Card Sorting

1/95

flashcard set

Earn XP

Description and Tags

Exam 4

Study Analytics
Name
Mastery
Learn
Test
Matching
Spaced

No study sessions yet.

96 Terms

1
New cards

what is molecular oncology?

interdisciplinary specialty that refers to the investigation of the chemistry of cancer and tumors at the molecular scale

2
New cards

what is the goal of molecular oncology?

  • identify genes that are involved in the development of cancer

    • the protein products may serve as targets for novel cancer treatments or imaging scans

    • this is still a fishing expedition and part of it is scanning known mutants

3
New cards

what are the techniques for molecular oncology? what is there a big area for? why do scientists use a range of techniques?

  • range from genomics, computational biology, tumor imaging, in vitro and in vivo functional models

  • a big area for computational biology but becomes obsolete within a year and requires to be HIGHLY trained for it

  • uses a range of techniques to validate the role of novel candidate genes in the development of cancer

4
New cards

what are driver mutations? what are passenger mutations?

  • driver mutation is directly responsible for the overgrowth of a cell type

  • passenger mutations are usually caused by the driver mutation and they’re not directly implicated in the disease state but might be down the line

    • still important because if a third mutation occurs, the passenger mutation becomes more hazardous for ones health

5
New cards

what is the ultimate goal for molecular oncology?

personalized medicine

  • personalized medicine is truly the only way to cure cancer because no two cancers are identical

  • have to get the information out of one individuals cancer and translate it to the right therapy for that individual

6
New cards

what do clinical labs test for?

tumor markers and genes that are predisposed to cancer

7
New cards

what is oncology? what are malignant tumors? what are the two types of tumors? what is leukemia and lymphoma?

  • the study of tumors (neoplasms)

  • malignant tumors are cancer

  • tumors are solid or hematological

  • leukemia = fluid-based white blood cell cancers

  • lymphoma = WBC cancers that form solid masses (lymph nodes)

8
New cards

what is metastasis?

movement of tumor cells from site of origin

9
New cards

why are proto-oncogene and tumor suppressor genes common targets in cancer diagnosis? name common examples of each (written response)

Proto-oncogenes is a common target in cancer diagnosis as they turn into oncogenes when it mutates. Proto-oncogenes normally promote cell division or cell survival but when a proto-oncogene mutates, it gains a function that causes abnormal cellular division and increased cell survival. Common examples of proto-oncogenes are the RAS gene and HER2. Tumor suppressor genes is also a common target for cancer diagnosis because when tumor suppressor genes are mutated, it loses it function to suppress tumors and allow for uncontrolled cell growth. Common examples of tumor suppressor genes are TP53 and PTEN.

10
New cards

what does tissue specific mean in molecular pathology/analytical testing? what is the problem with it?

  • nucleic acid and protein characteristics of tissue specific markers, such as rearranged IG, or T-cell receptors in leukemia

  • T-cell receptors are a normal part of the tissue system in blood

  • problem = hard to distinguish the tumor or cancer from the normal cell/same markers present in the normal tissue

11
New cards

how are tumor-specific target used in molecular pathology/analytical testing?

tumor-specific targets are more cancer-specific and will only specifically pick up just the cancer because they are not present in normal tissue

12
New cards

understand how LOF, GOF, and chromosomal abnormalities all play roles in cancer (written response question)

Loss-of-function mutations play a role in cancer as the gene, most commonly tumor suppressor genes, loses its normal ability to suppress uncontrolled cell growth. With this, the loss of the function results in the uncontrolled cell growth of cancers and prevents apoptosis in the cancer cells. Gain-of-function mutations play a role in cancer as the proto-oncogene that turns into an oncogene acquires a new mutation that enhances cell division and survivability, which is not normal in a cell. Chromosomal abnormalities play a role in cancer as structural changes like translocations, deletions, and insertions can result in a gain of function or loss of function. Chromosomal number abnormalities like aneuploidy and polysomy can lead to an imbalance in the genes, resulting in enhanced cell division.

13
New cards

how is cancer caused by the nonlethal genetic mutation, proto-oncogene? what are the categories for oncogenes?

  • when proto-oncogenes are mutated into oncogenes (~100 known) where some promote cell division and others prevent apoptosis

  • categories for oncogenes

    • growth factors

    • receptor tyrosine kinase (RTK) which is one of the three common classes of receptors, and is a hot spot for mutations

    • cytoplasmic TKs,

    • Ser/Thr Kinases

    • GTPases

    • transcription factors

14
New cards

how is cancer caused by the nonlethal genetic mutation, tumor suppressor? what are the five broad categories and example them

  • tumor suppressor genes lose its function to slow down cell growth resulting in abnormal cell growth

    • more than 1,000 known tumor suppressor genes and the most common are P53 and PTEN

  • the five broad categories

    • supposed to slow cell growth/cell cycle progression and cell proliferation (2)

    • supposed to fix broken DNA

    • cannot stimulate the cell to die by apoptosis

    • other crucial cellular signaling functions

15
New cards

how is signal transduction and cellular communication involved in cancer/oncogene?

  • cells will sometimes divide and should work where the growth factors interacts with the RTKs and triggers cell division through mitogen

  • if there’s NO growth factor for a long time, then no mitosis should happen, and the cell will go through apoptosis

  • if the RTK is mutated, there is no need for the growth factor anymore because the cell will go through mitosis several times without the stimulation of the growth factor

16
New cards
<p>how does the cell cycle play a role in cancers related to both oncogenes and tumor suppressors? what is done if mutations are detected in the cell cycle?</p>

how does the cell cycle play a role in cancers related to both oncogenes and tumor suppressors? what is done if mutations are detected in the cell cycle?

  • the cell cycle regulates cell division and cell proliferation

  • cells are born in G1 and cannot exit G1 until they pass the checkpoint that sees if it is ok

    • involves cyclin-dependent kinases which only work if phosphorylation is present

  • when a cell is ready to go to the next stage, a phosphorylase and a CDK will interact and allow it to pass the checkpoint

    • cyclin E and CDK2 are common oncogenes and if they’re mutated, they will allow cancerous cells to pass

  • inside of the ring are tumor suppressors and if they’re mutated, they can no longer properly regulate the cell cycle which leads to uncontrolled cell growth

  • if mutations are detected, target treatment may be available or KNOWN mutations indicate high risk so affected individual screens more often

17
New cards

what are the three gene mutations in a gain of function for oncogenes? describe them and give an example

  1. change in protein structure

    • common mutation

    • the proto-oncogene undergoes a point mutation, and within the gene, a protein is produced that does not degrade effectively. As a result, the gene continues to generate fresh proteins, leading to an accumulation within the gene.

    • ex. EGFR M277E, some lung cancers

  2. increase in protein concentration

    • can have gene amplification and during replication, the polymerase puts in more copies of the same gene and each make proteins

    • ex. HER2/Neu breast and prostate

  3. change in gene location

    • translocation, where the gene/your DNA moves to a new location under new controls/promoter

    • ex. t(8:14) which causes Burkitt lymphoma and t(9;22) which creates a Philadelphia chromosome that creates a new fusion protein in CML

18
New cards

what happens if a repair pathway is mutated in cancer?

  • the pathways will lose its function like mismatch repair, ss break and ds break repair

  • every single gene that are involved in the repair pathways are tumor suppressor mutation thats implicated with cancer

    • every target needs a diff therapy which is why cancer therapy is though

19
New cards

what are the three targets for molecular detection of disease? describe them

  • antigens (antigen is on the surface of a T-cell) and mutations

  • disease-specific markers

    • translocations, point mutations, polymorphisms in tumor suppressor or oncogenes

  • viruses

    • EBV, HCV, HPV, HTVL-1

20
New cards

what are the 6 methods for molecular detection of disease?

  1. hybridization, blotting

  2. standard PCR, RT-PCR, electrophoresis

  3. PCR with heteroduplex analysis, SSCP

  4. real-time PCR with gene or patient-specific probes

  5. cancer gene arrays

21
New cards

what is a HER2/Neu oncogene? where is it frequently amplified and what is the percent for it? what are they sensitive to? how can it be detected?

  • common oncogene and falls into the RTK family (intracellular kinase domain)

    • encodes one of family of human epidermal growth-factor receptors (hEFGR2) with tyrosine kinase activity

  • frequently amplified in breast and gastric cells, resulting in increased amounts of cell surface protein

    • 20% of cancers have elevated HER2 expression

  • HER2-expressing tumors are sensitive to HERCEPTIN, a monoclonal antibody

    • herceptin can be used by any cancer with HER2 overexpression

  • DETECTION

    • HER2 protein can be detected by IHC

    • HER2/neu gene amplification is detected by FISH or CISH

      • amplification can slightly change the therapy

22
New cards

what does the intracellular kinase domain do? what is different about them?

  • phosphorylates a signal transduction pathway inside the cell

  • what is different about it = the outside of the cell because different growth factors stimulate all of the different external domains

23
New cards

how does the amplification of HER2 cause overexpression? why? what is often the cause of this?

  • if HER2 is amplified genetically (three copies instead of one), it leads to too much protein

  • normal breast cells already have HER2 protein in the membrane and is there incase an adjacent cell needs to be replaced

    • if there is amplification in the gene or too much protein is being made = will have EXCESS receptors in the same tissue of the cell and will trigger fast cellular division

  • often due to increase copy number of chromosome 17

24
New cards

what is the grading system for HER2 overexpression in IHC? what is the challenge when it comes to IHC?

  • 0 = negative meaning there is NO staining

  • 1 = negative meaning there is faint staining

  • 2 = weakly positive meaning there is weak to moderate staining of the tumor in >10% of the cells

  • 3 = positive meaning there is strong staining in >10% of the cells

  • heterogeneity is a challenge because there can be minimal staining in one area but strong staining in another on the same tissue

25
New cards

what are the current guidelines for IHC testing for HER2? what are the preferred samples and ideal fixative?

  • labs providing a testing service should carry out a minimum of 250 assays each for year for IHC detection of HER2

  • formalin fixed, paraffin embedded tumor tissue and buffered formalin

    • the use of other alternatives like bouins will prevent testing of FISH

    • other methods of tissue fixation can adversely affect antigen reactivity

26
New cards

how is FISH used to test for breast and gastric cancer? what is it looking for and what does the pink stain indicate?

  • FISH is used to quantitatively measure the level of HER2 gene amplification

  • essentially looking for DNA and uses the DAPI stain to stain the nucleus

  • pink staining = HER2 stain and there’s only 2 HER2 proteins per cell

27
New cards

what role does the EGFR family play in molecular abnormalities in solid tumors? how is HER2 apart of this?

  • growth receptors are often involved in cancer and if theyre constitutively activated, the cells are signaled to divide all the time

    • growth factors are not always activated and will only active when division is needed

  • HER2 is a part of this family and causes all the epithelial cells to divide

    • does NOT have a ligand so it works through heterodimerization with other members of the EGFR family, which leads to uncontrolled cell proliferation

  • more division increases the chance of mutations

  • driver mutations make passenger mutations

28
New cards

what does EGFR oncogene encode for? what are they sensitive to? how are they detected?

  • EGFR oncogene encodes another of the family of EGFR as HER2

    • common target for therapy and diagnosis

    • want to see which one is overexpressed to slow down its cell growth with the correct therapy

  • tumors with activating mutations in EGFR are sensitive to tyrosine kinase inhibitors (TKI)

  • DETECTION

    • EGFR protein = IHC

    • EGFR gene and chromosome abnormalities = FISH

    • EGFR mutations = SSCP, SSP-PCR, or direct sequencing

29
New cards

what is glioblastoma? what was discovered about GBM after using FISH?

  • glioblastoma is a highly aggressive and fast-growing brain tumor that is hard to detect → life expectancy is 2 years after diagnosis

  • used FISH and discovered 24% of glioblastoma showed EGFR amplification but then sequenced it and discovered it was 40-50%

  • also discovered that some therapies are refractory because there are other variants of EGFR

  • EGFR alterations are a prediction of good prognosis and will determine the overall survival of patients with IDH-wild type GBM

30
New cards

what is the RAS gene? what is it involved in? how is it detected?

  • RAS is a proto-oncogene turned into an oncogene after being mutated

    • RAS gene can be amplified

  • RAS signaling is involved in many cellular functions like growth, apoptosis, migration, and differentiation

    • transduction stages uses RAS

  • K-ras gene mutations are detected by SSCP or direct sequencing

31
New cards

where are the K-ras gene mutations implicated in?

  • implicated in various cancers like leukemia, lung, mucinous, colorectal, and pancreatic ductal carcinoma

  • 30% of human cancers are associated with mutations in RAS genes like NRAS, KRAS, HRAS

  • 90% of colon cancers have mutations in RAS

    • if patient has two particular mutations (KRAS) then the medication Cetuximab wont work

  • one mutation in GTP-BD results in permanent activation of RAS and cannot be deactivated

32
New cards

why was RAS considered undruggable until 2022?

  • inhibitors were not working for RAS

  • in 2020 the FDA granted the AMG 510, fast track designation

    • granted means that it was not FDA approved but approved for clinical trials only

  • the AMG 510 binds to KRAS-G12C RAS mutant and slowed cell growth then was approved in 2022

33
New cards

what are the methods to diagnose oncogene activation for gene amplification or chromatin change?

  • FISH/CISH

  • WGS or NextGen

  • qPCR/real-time PCR

  • southern blot

  • PCR breakpoint-specific probes

  • break away probe fish

34
New cards

what are the methods to diagnose oncogene activation for mRNA/protein overabundance?

  • RT-PCR

  • RNA sequencing

  • northern blot

  • in situ staining of protein or RNAs

35
New cards

what might be the most productive method to scan for K-ras mutations?

next-generation sequencing (NGS)

36
New cards

what factors might confound its molecular diagnosis? why?

heterogeneity because if you can miss a mutation

37
New cards

how are oncogenes activated by chromatin modification? what method do they use to detect it? what can occur with overproduction of an oncogenic product?

  • sometimes oncogenes activate by moving around in the genome or become exposed and expressed due to acetylation

  • uses FISH breakaway probes

  • overproduction of an oncogenic product can occur by loss of transcriptional control through chromosomal translocation

38
New cards

what are FISH breakaway probes? what will breakage and no breakage indicate when used?

  • two fluorescent probes to two targets and are very close to each other and breaks between them

    • can also use three different fluorescent probes to three different targets

  • no breakage = probes will show up next to each other

  • breakage = will have space between them in the nucleus

39
New cards

how do you detect chromosomal aberrations and molecular lesions with FISH, CISH, karyotype, or break-point probes/break away probes? can these molecular tests be used to see disease progression and treatment efficacy? (written response question)

FISH detects chromosomal aberrations and molecular lesions through the use of fluorescent probes to the targeted regions. CISH has a similar premise to FISH but uses chromogenic detection instead. Karyotyping is used to visually inspect the chromosomes by the use of Giemsa staining, which can detect translocations; however, it needs a large translocation to detect it. Karyotyping cannot detect molecular lesions. Break-away probes utilized two to three fluorescent probes to two or three target regions to determine if breakage occurred, indicating chromosomal aberrations and molecular lesions. All these molecular tests can be used to see disease progression and treatment efficacy; however, FISH is recommended for initial diagnosis and PCR for monitoring in hematological tumors.

40
New cards

how was FISH used to see and analyze the t(8;14) translocation? what is the t(8;14) translocation?

  • a green probe was designed to IGH and a red probe was designed to MYC to analyze if the translocation of chromosome 8 to chromosome 14 and vice versa occurred

  • if the two colors were together, it indicated a translocation and a yellow color indicated that a new hybrid chromosome was created

  • the translocation causes the myc oncogene on chromosome 8 to be positioned next to the IG heavy chain on chromosome 14 which causes myc to become constitutively expressed

41
New cards

what is the t(14;18) translocation?

  • reciprocal translocation between the long arms and is common in lymphomas, especially follicular

  • the translocation causes the BCL2 gene from B-cell leukemia and lymphoma leading it to become dysregulated and overexpressed when it comes from chromo 18 to chromo 14

42
New cards

how was the t(14;18) translocation detected by PCR? how is it seen on a gel?

  • PCR will use a downstream primer to the IG region and an upstream primer to the BCL2 gene

    • one primer to be one chromosome and the other primer to a different chromosome

  • in a healthy chromosome, there will be NO PCR product

  • translocation in the major breakpoint region = PCR product further up the gel

  • translocation in the minor cluster region = PCR product further down the gel

  • often need multiple PCR rounds

43
New cards

what are chimeric genes and how might one be identified diagnostically? (written response question)

Chimeric genes are formed when two portions of chromosomes fuse together which are primarily through translocations. Chimeric genes often create a new protein that is not seen in normal tissues due to the fusion. An example of this would be the BCR-ABL gene that is formed through the reciprocal translocation t(9;22). Chimeric genes are identified through FISH for initial diagnosis and through PCR like qPCR and RT-PCR.

44
New cards

what is the philadelphia chromosome? what does the fusion protein activate and result in? why is it a good target?

  • a reciprocal translocation of chromosomes 14 and chromosome 18 which forms a new fusion protein called BCR-ABL

  • 90% of patients with CML have this chromosome

  • BCR-ABL gene activates so many different cellular communication pathways and will lead to increased cell growth, proliferation, and increased gene transcription

    • results in both aberrant activity and subcellular location of the ABL protein TK, leading to cell transformation

  • BCR-ABL is a good target since it is not in normal cells which is the reason why CML has a great cure rate

45
New cards

how are translocations in hematological tumors detected?

  • detected with higher sensitivty using PCR

  • qPCR can be used to quantify the tumor load during patient monitoring

  • FISH is used for initial diagnosis but PCR is used for monitoring

46
New cards

how do you detect chromosomal aberrations and molecular lesions detected with PCR, RT-PCR, qPCR, and SSP-PCR? can these molecular tests be used to see disease progress and treatment efficacy? (written response question)

PCR is used to amplify specific DNA sequences to detect chromosomal aberrations and molecular lesions. RT-PCR turns the specific mRNA sequences into cDNA to detect any translocations, gene fusions, or changes in gene expression. qPCR detects chromosomal aberrations and molecular lesions by quantifying the miRNAs, oncogenes, telomerase, DNA, RNA and more which can also be applied to RT-PCR. SSP-PCR uses sequence specific primers to targeted regions of the molecular lesions and chromosomal aberrations at the 3’ end. These molecular tests can be used to see disease progression and determine the efficacy of treatment.

47
New cards

what do you do in order to get a linear trend in qPCR to ensure that the assay is working? what is it also responsible for?

  • take patients tumor cells and spike it with normal tissue (10x more, 100x more, 1000x more) which creates the linear trend line

  • also responsible for RNA isolation because as you’re adding normal cells to tumor cells, you’re also extracting RNA for RT-qPCR

  • essentially adding cells to cells

48
New cards

in order to get the standard curve for RT-qPCR, what needs to be done?

  • use a standard curve of transcripts of known copy numbers diluted into normal RNA

    • spike the RNA that was isolated from qPCr with normal RNA to create the standard curve

49
New cards

what happens when a tumor suppressor gene gets mutated? what maybe be significant in the development of human cancers? what kind of mutation are they, recessive or dominant?

  • tumor suppressor gene loses its function to suppress abnormal cell growth and results in uncontrolled cell growth

  • LOF of these genes may be more significant

  • recessive mutation since both alleles that code for a particular protein need to be affected (two hit hypothesis)

  • changes the protein structure

50
New cards

what is TP53/what does it encode? what does it do in the body? what does the mutated form of p53 cause?

  • TP53 has a normal function to assess DNA damage

  • encodes a “caretaker" gene” which is involved in DNA repair, induce apoptosis, transcription control, and regulating the cell cycle

  • binds to a double-stranded break when UV light damages DNA in the genome, which causes cell arrest and gives the cell time to repair the break

  • for the mutated form of p53, the signal transduction cascade cannot activate p53 meaning p53 does not interact with the dsDNA break → hopefully leads to the cells death but in other cases it causes uncontrolled cell growth

51
New cards

how is TP53 specifically connected to cancer? what does its indication mean? how are they detected?

  • 2 million cancer cases each year and about 50% of them are related to the p53 mutation

    • can be dominant negative mutation or LOH

  • TP53 mutations are an indicator of a poor prognosis

  • DETECTION

    • mutated p53 protein is detected by IHC by its persistence

    • TP53 gene mutations are sequenced

    • SSCP is used to look for alternated protein confirmations and silent polymorphisms

52
New cards

what is inside of the p53 gene that was created? what is needed to determine the prognosis of p53 and why?

  • has a ton of subunits that interact with other subunits in different situations and also has a lot of different mutations

  • DNA and protein results often conflict, thus p53 status to determine disease prognosis required 2+ different test types

53
New cards

reason through some broad diagnostic differences when identifying solid tumors vs hematological malignancies (written response question)

solid tumors are abnormal masses of tissue that result from abnormal cell growth due to a mutated tumor suppressor gene. When detecting these, biopsies are required to obtain the tissue samples and analyze them through IHC and FISH. Hematological cancers (INCOMPLETE)

54
New cards

what is looked for in the genes of LOF tumor suppressors?

  • mutations

  • persistent or aberrant proteins

  • copy number variants

    • WES

    • WGS

    • SNP

    • microarray

55
New cards

what is looked for in the mRNA or protein of LOF tumor suppressors?

  • look for presence, may not be produced

  • look for mutation result, such as loss of DNA repair pathway

    • MSI

    • STR analysis

56
New cards

what is von hippel-lindau gene?

  • a tumor sup[pressor and is necessary for normal blood vessel development

  • the normal function is lost by mutation in hemangioblastomas, retinal angioma

  • blood vessel tumors form around the CNS

  • autosomal dominant, 95% penetrance by age 65

57
New cards

what are cancer susceptibility genes and why are they important clinically? (written response question)

Cancer susceptibility genes are mutations in certain genes that MAY increase the risk of some cancers. They are also called cancer predisposing gene mutations because they are usually inherited from one parent. Cancer susceptibility genes are important clinically because knowing its inheritance can help in preventing, detecting, and treating the cancer.

58
New cards

what is BRCA1 and BRCA2? what happens if a women inherits one of the mutations? what does variable expressivity in a patient mean? what are the preventative measures for them?

  • tumor suppressor genes that encode DNA repair proteins that repairs dsDNA breaks during recombination

  • women who inherit a mutation in one of them have a 40% chance of developing breast or ovarian cancer in their lifetime, but it’s truly a range depending on which mutation was inherited.

  • variable expressivity does not mean this person has a 100% chance

  • preventative measures = mascetomy and frequent screening

59
New cards

what are the frequent mutations in BRCA1 and BRCA2? how are they detected?

  • BRCA1 =187delAG and 5382incC

  • BRCA2 = 6174delT

  • DETECTION

    • direct sequencing of both genes but need to know what you’re looking for

    • SSCP and SSP-PCR if you have no hints

    • chromosome breakage tests

60
New cards

how was the the 185delAG of BRCA1 detected by SSP-PCR?

  • used a primer specific to the normal and a red primer specific to the deletion

  • flanks the normal primer to amplify through the region that might have the mutation (120 bp)

    • the red primer will amplify the same primer and is 180 bp on a gel

61
New cards

what is microsatellite instability? why is it linked to DNA repair failure? what is a common way to detect MSI? (written response question)

Microsatellite instability is the production of new alleles from unrepaired replication errors. Microsatellite instability is linked to DNA repair failure because it is a preliminary diagnosis of mismatch repair failure. The mismatch repair system does not properly repair replication errors in microsatellite regions and increases the number of repeats, which leads to microsatellite instability. The common way to detect microsatellite instability is through immunohistochemistry, MSI PCR, and CGE.

62
New cards

what does hereditary nonpolyposis colorectal carcinoma account for? what are the mutations analyzed for and where are the genes often mutated in? what is it associated with and what is looked for?

  • also known as lynch syndrome = accounts for about 5% of colon and endometrial cancer, plus mutations increase the risk of many other cancers

  • associated with mutations in genes encoding components of the mismatch repair system, most frequently MLH1 and MSH2

    • MLH1 is frequently hypermethylated

  • mutations of the genes are analyzed for LOF

  • MLH1 and MSH2 are often mutated in RER

  • looking for microsatellite instability in both genes

    • 85-90% of HNPCC tumors have MSI

63
New cards

what is replication error phenotype (RER)?

  • slippage in replication common at 1-3 NT repeats

  • the stutters are normally corrected by MMR

  • causes the replication machinery to slip during replication and extends the repeat

64
New cards

what can detect instability and give its results? how are MSI’s analyzed? what does the analysis of the five loci do? what is direct sequencing used for?

  • MSI PCR and CGE can detect instability by comparing PCR amplicons of the microsatellite loci to normal

    • results for PCR in unstable loci = tumor column has more bands compared to the normal column

    • results for CGE in unstable loci = spreads/doesn’t stay in one area

  • analyzed by assessing the stability of at least five MS loci recommended by the NCI

    • dinucleotide or mononucleotide repeating units

  • analysis of five loci determines gene function

    • 2 out of 5 means high instability

  • direct sequencing is used to detect the actual gene mutation

65
New cards

how to detect chromosomal aberrations and molecular lesions with microsatellite instability assay? can this molecular test be used to see disease progression and treatment efficacy? (written response question)

The MSI assay can detect chromosomal aberrations and molecular lesions through their microsatellite instability, which is caused by failure of the MMR system. Specifically, the MSI assay compares the microsatellite instability in tumor tissue and normal tissue. This molecular test can be used to see disease progression and treatment efficacy.

66
New cards

what does it mean when an individual is heterozygous? how is loss of heterogeneity linked in the cancer realm?

  • those who are heterozygous with a germline mutation are predisposed to cancer

  • LOH happens when the other allele (normal) is lost either by deletion or silenced by methylation, and the normal allele is no longer expressed, resulting in the expression of the phenotype of the mutated allele

    • essentially exposes the recessive mutant allele in a hemizygous state

67
New cards

how is loss of heterozygosity detected by STR analysis? what are the results? how do you calculate it?

  • shows the loss of the heterozygous STR

    • compares the colon cancer tissue to adjacent normal tissue

  • uses fluorescent primers to PCR loci associated with cancer and uses CGE or sequencing

    • no LOH = peak of normal and mutant allele is the same

    • LOH = decreased in allele ratio of 20% or more

  • for sequencing, sequence both top and bottom strand and compare how much mutant allele is in normal tissue and tumor tissue

  • calculation = peak normal allele/peak normal allele in tumor divided by peak mt allele/peak mt allele in tumor

68
New cards

what is oncotype Dx? what do the scores indicate for the treatments?

  • a gene expression assay that is commonly used for breast cancer patients

  • score less than 10 for instability = hormone therapy

  • score 11-25 for instability = randomized hormone therapy or hormone + chemotherapy

  • score more than 25 = chemotherapy + hormone therapy

    • bad prognosis and will put you on something that will kill everything

69
New cards

where can tumor markers be found? what are the 7 tumor markers?

  • found in blood, urine, or tissue affected by cancer

  • tumor markers

    1. PSA

    2. CA125

    3. AFP

    4. HCG

    5. carcinoembryonic antigen (CEA)

    6. CA 19-9

70
New cards

what do liquid biopsies look for? who is it more targeted for? what is it done to see?

  • looks for circulating-free DNA (cfDNA) in the plasma or urine OR look for circulating free RNA and circulating tumor cells (CTC)

  • targeted for those who finished their therapy and will go in after 6 months for detection

  • done to see if there is the same MSI/cancerous DNA in the body after therapy/6 months after therapy

71
New cards

how are the circulating tumor cell isolated? what does it end up doing? what does it tend to do? what is FACS?

  • uses antibody capture using antibodies to epithelial cell-specific membrane proteins like epCAM

    • takes the fluid based specimen and sends the cells through a capillary tube

    • there will be a detector with an antibody to the targeted surface proteins of the CTCs

    • as the cells go by one by one, if the receptor on the membrane interacts with the antibody, the machine will suck it up

  • ends up counting how many cancer cells vs normal cells

  • tend to do to see if theres any CTC and if not, you’re in remission

  • FACS = fluorescence activate cell sorting

72
New cards

why do you want to monitor cancer concentration?

  • to see treatment response/to see how well the therapy is working in metastatic disease

  • great way to keep an eye on remission

73
New cards

what are the four PCR variations used in oncology?

  • droplet PCR = type of digital PCR

    • able to quantify how much starting material you have by dividing the number that are positive by the total number of wells that were analyzed

  • TRAP/PCR-CE

    • measures telomerase activity for any cancer

  • allele-specific, non-extendable primer blocker PCR (AS-NEPB-PCR)

  • Δ-PCR

    • uses two forward primers (external and internal) and a reverse primer simultaneously to find translocations

74
New cards

where does traditional PCR measure? how about qPCR? what does droplet PCR do?

  • traditional PCR measures at the plateau/measures how much amplicon is made at the end of the PCR

  • qPCR measures at the exponential phase

  • droplet PCR (dPCR) counts individual molecules for absolute quantification

75
New cards

how do you better quantify the sample for dPCR? what is the process for dPCR?

emulsify into a little droplet to better quantify how much sample and how much mutated target a patient has

  1. isolate DNA from tissue biopsy and elute it in a certain amount of sample

    • best way is to take 1 microliter of the diluted sample and put it into each well

  2. after putting the sample into 1 microliter wells, can perform amplification of the wells

    • PCR amplify the wells separately and analyze each separately

  3. after amplification = will end up with a 0 or 1 score

    • 0 = no template

    • 1 = template

76
New cards

what are the 5 benefits of dPCR? explain them

  • good for quantification

    • shows exactly how many copies you have and how many cells you started with and how many are mutated

  • high tolerance to inhibitors

    • during isolation sometimes there are inhibitors so you can just dilute to 1 microliter and still perform PCR

  • superior precision

    • detects very small fold changes

  • increased sensitivity

    • detect rare mutations and low abundance targets

  • high reproducibility

    • elimate amplification efficiency bias

77
New cards

what are the 5 applications for dPCR?

  1. CNV

  2. rare mutation detection

  3. NGS library quantification

  4. viral load detection

  5. gene expression analysis

78
New cards

what are the 6 viruses that can lead to cancer? explain them

  1. EBV = linked to hodgkins lymphoma and detected by qPCR

  2. HBV = test for surface antigen and core antibody

    • treatment of virus occurs in all cancer patients if HBV+ to prevent HBV liver destruction

  3. HCV = screens for antibodies, curing virus improves cancer outcomes

    • linked to liver cancer and other liver problems

  4. HTLV = tests for T-cell lymphotropic viral antibodies

    • most have no symptoms, some will develop adult T-cell leukemia-lymphoma

  5. HHV8 (herpes 8, karposi SV) = uses qPCR

    • antiviral drugs improve cancer treatment

  6. Merkel cell polyomavirus (MCPV) = uses qRT-PCR to test and is associated with poor outcomes of skin (head, limb, and trunk) cancers

79
New cards

what is the relation of HPV in cervical cancers? how does a Pap smear detect cervical cancers? what can tests do now for cervical cancers? what do vaccines do for HPV?

  • HPV has been implicated in 99.7% of cervical cancer cases worldwide

  • Pap smears detect abnormal cervical cells

  • tests can look at different strains of HPV

    • one particular strain can put an individual at very high risk for cervical cancer and will immediately start a preventative measure when detected

    • there’s also at home screening that can test for diff strains too

  • vaccine that prevents HPV associated with cancers

    • vaccines can be used after virus acquisition to prevent cancer

80
New cards

what is hematopoesis?

  • in your bone marrow, there’s a stem cell called multipotential hematopoietic stem cell aka a hemocytoblast

  • hemocytoblast can become a myeloid progenitor and become platelets, RBCs, or WBCs

  • hemocytoblast can also become lymphoid progenitor and become the B and T lymphocytes

81
New cards

what is needed to respond to mean pathogens? what mediates antigen recognition? what rearranges similar manner?

  • need to make a ton of T-cell receptors to the B-cells which is how you respond to a lot of mean pathogens

  • two classes of receptor proteins that mediate antigen recognition are antibodies and T-cell receptors

  • antibody genes (IG heavy, IG light chain, VDJ regions) and T-cell receptor genes (alpha, gamma, beta, delta) rearrange in a similar manner

    • recombination is normal in these cells

82
New cards
<p>what do we have in our chromosomes related to this picture? what happens when a person is exposed to something different?</p>

what do we have in our chromosomes related to this picture? what happens when a person is exposed to something different?

  • have variable regions, variable diversity joining regions, and constant regions

  • when a person is exposed to something different → chromosomes splice things together to make different combinations of the antibodies

    • by the time you get down to the mRNA, you need one of the VDJ regions

  • compound the diversity of the chromosome with how many different alleles exist in different populations

    • relates to antibody diversity

83
New cards

what do you have to screen for in hematological disease? what happens in most hematological diseases?

  • have to screen person for their genetic recombinations

  • what happens:

    • the VDJ regions go through somatic hypermutatin (SMH) which is normal and occurs after the B-cell encounters the antigen

    • all the different cell populations you’re supposed to have, all the diff T-cell receptors, and all the different antibodies lose their polyclonality turning into a monoclonal population

84
New cards

what do you test for in hematological diseases? how are they detected?

  • tests for clonality with blood tests

    • normal lymphocyte populations are polyclonal with respect to IG (heavy and light) and TCR genes → all 3 may need analyzed

    • leukemia or lymphoma are monoclonal

      • depends on the clone but this is how to test/diagnose for leukemia or lymphoma

  • clonality is detected by protein or NA or Sblots

85
New cards

how are monoclonal populations detected? how are RENs used for them? what are the results?

  • monoclonal populations are detected by rearranged bands unique to the tumor cell population

  • three different RENs (EcoR1, BamH1, and Hind3) are specific to the constant regions

    • if there is recombination in the VDJ regions, one of their chromosomes has recombined and made a different sequence (not good) and the REN’s will cut at these different sequences at the specific REN site

    • essentially uses three different REN digests to show different banding patterns in the patient to show their chromosome has undergone rearrangement

  • EcoR1 and Hind3 results

    • one band in germline

    • two bands in rearranged

  • BamH1 results

    • one band in germline

    • three bands in rearranged

86
New cards

how was next gen sequencing used to test for clonality?

  • used paraffin fixed tissue and sequenced and compared the number of diff DNA seqeunces

    • there is a bunch of genetic info in all the cells in a healthy individual

  • polyclonal cell population → should be genetically different

  • monoclonal cell population → one particular population is overexpressed which shows patient has lymphoma or leukemia

87
New cards
<p>how was southern blot used to detect monoclonal lymphocyte populations? what are the advantages and disadvantages of sblot?</p>

how was southern blot used to detect monoclonal lymphocyte populations? what are the advantages and disadvantages of sblot?

  • determined the B cell clonality in patient blood

  • looked at the IG chain gene with three different REN digests

    • negative control different RENs should give 3 bands in total (one band for each digest)

    • in patient = there is a single additional (rearranged) band to indicate a monoclonal population

  • advantages = cheap and sensitive

  • disadvantage = uses radiation

88
New cards

how was capillary gel electrophoresis used to detect t-cell clonality? what are the results? is it expensive?

  • detects small sequence changes because DNA must be single stranded (denaturing)

    • can also use for STRs because it can detect a three NT difference (extra STR)

  • in a healthy control = it should look like a hill (genetically different)

    • y axis = abundance of the amplified fluorescent TCR-gamma (500-1500)

  • in a patient with monoclonal population = does not look like a hill and has multiple peaks and one peak that is overexpressed which indicates a monoclonal population

  • not an expensive assay and doesn’t jeopardize the health of the technician

89
New cards

what are the three common translocations in hematological tumors?

  • t(8;14)

  • t(9;22)

  • t(14;18)

90
New cards

what is wrong with chromosome 14?

  • IGH is on chromosome 14 and is a common translocation and is put next to a proto-oncogene to turn it onto an oncogene

    • now that oncogene is constitutively expressed

  • proto-oncogenes are under the control of the IGH gene, leading to uncontrolled cell growth

  • the translocations with chromosome 14 are commonly linked to lymphomas

91
New cards

how was the translocation (9;22) detected by RT-PCR?

  • designed an upstream primer to the BCR gene and a downstream primer to the ABL gene

    • turned mRNA into cDNA then PCR amplified the cDNA

    • designed a primer after ABL so it can pick up all possible translocations

  • looking for the expression of the fusion protein of the BCR-ABL gene

    • easier to pick up than mRNA

  • in the gel it shows all the different translocations that can occur

92
New cards

what is the cause of a majority of childhood cancers?

  • caused by fusions

  • 80% of childhood cancers are leukemias or lymphomas and of those, more than 30% (as high as 75%) are caused by chromosomal translocations

93
New cards

how do you detect chromosomal aberrations and molecular lesions with RFLP and southern blot? can these molecular tests be used to see disease progression and treatment efficacy? (written response question)

Both RFLP and Southern blot can be used to detect chromosomal aberrations and molecular lesions through the use of restriction enzyme digests to analyze the different DNA fragments. Particularly, RFLP and Southern blotting are used for clonality testing and gene rearrangements in cancer patients. RFLP and Southern blot can be used to see disease progression and treatment efficacy by monitoring the changes in the patient’s DNA patterns.

94
New cards

how do you detect chromosomal aberrations and molecular lesions with capillary and gel electrophoresis? can these molecular tests be used to see disease progression and treatment efficacy? (written response question)

Capillary gel electrophoresis separates DNA fragments that can reveal chromosomal abnormalities like loss of heterozygosity and molecular lesions like microsatellite instability by analyzing the peaks. Gel electrophoresis can also be used to detect chromosomal aberrations and molecular lesions because it is used to resolve PCR products, which can detect deletions in chromosomes and mutations in genes. Capillary and gel electrophoresis can be used to monitor disease progression and treatment efficacy by observing the changes in the patients.

95
New cards

how do you detect chromosomal aberrations and molecular lesions with sequencing and microarrays? can these molecular tests be used to see disease progression and treatment efficacy? (written response questions)

Sequencing can be used to detect chromosomal aberrations and molecular lesions by analyzing the DNA sequence for mutations or any structural changes. Microarrays can detect chromosomal aberrations and molecular lesions by scanning the expression of multiple genes at the same time. These tests can be used to see disease progression and treatment efficacy.

96
New cards

what are the three genetic aberrations that cause hematological cancers? they are also common diagnostic and treatment targets (written response question)

One genetic aberration that cause hematological cancer are gene mutations. Gene mutations are often scanned in diagnostics to determine the mutation in the individual which can determine the treatment for the individual. Another genetic aberration is chromosome structural abnormalities like translocations, deletions, and insertions. Lastly, another genetic aberration that can cause hematological cancers is chromosome number abnormalities like aneuploidy and polysomy.