• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/493

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

493 Cards in this Set

  • Front
  • Back
screening tests for cystic fibrosis
1. IRT
2. DNA analysis
3. Sweat Test (>60)
Pathophysiology of Cystic Fibrosis
1. Increased mucous viscosity w/ obstruction of lungs, GI tract, Liver, gall bladder, reproductive tract.
2. Respiratory tract colonization/ infection with unique organism
3. increased sodium chloride in serous secretions
Common Treatmnet of Cystic Fibrosis
1. pulmozyme (DNAse)
2. Tobramycin (TOBI)
3. Azithromycin
4. Inhaled hypertonic saline
5. Inhaled Aztreonam (Caystor)
[airway clearance]
[diet/ high protein]
[pancreatic enzyme supplements]
Primary Cause of Down's Syndrome
Non-disjunction in Meiosis I in the maternal line resulting in three copies of chromosome 21. [95% of cases]
Diagnosis of Down's Syndrome
Karyotyping; FISH may be used to get a quick preliminary diagnosis, but it should always be followed by a karyotype to verify.
Prenatal Screening for Down's Syndrome
Serum Measurements [indicative, but not diagnostic]
amniocentesis for karyotyping
Pathology of Down's Syndrome
1. Growth delay
2. Cardiac
a. atrioventricular septal defects[AVSD]
b. ventricular septal defect [vsd]
c. atrial septal defect [asd]
3. gastrointestinal: duodenal atresia or stenosis, imperforate anus
4. myopia and cataracts; protruding tongue; hearing loss
[leukemia 400X more likely]
Most Common Mode of Lysosomal Storage Disease
AR: inability to break down glycoproteins
Biochemical Abnormalities Leading to Lysosomal Storage Disease
1. enzyme deficiency
2. inability to activate lysosomal enzymes
3. abnormality of lysosomal membrane proteins
4. inability to transport enzymes to lysosomes
5. abnormal lysosome structure
Clinical Features of Lysososomal Storage Disease
Abnormal facial features, thickened oral and airway tissues, skeletal abnormalities, enlarged organs, CNS [neurodegeneration, cerebral atrophy, leukodystrophy, mental retardation, seizures]
Therapy of Lysosomal Storage Disease
substrate reduction and enzyme replacement therapy [sometimes chaperone therapy, bone marrow transplantation, liver transplantation, gene therapy]
Treatments for Sickle Cell
1. penecillin until the age of 5
2. hydroxyurea- stimulates the synthesis of HgF to replace HgB
[for crises: heat and hydration NSAID and Narcotic]
Osteogenesis Imperfecta
"Brittle Bone Disease"
impairs triple helix formation of collagen
especially bad if point mutation occurs near the C-terminus
What is necessary to form crosslinks between tropocollagens?
Lysyl oxidase and copper
What is needed to hydroxylate the proline in collagen formation?
prolyl hydroxylase, iron, and vitamin C [ascorbate]
Ehlers-Danlos Syndrome
characterized by stretchy skin; loose joints; poor wound healing; failure of vessel/ organ structure
no cross-linking or no removal of the n-peptide end of tropocollagen [not converted to insoluble form]
Elastin
rubber protein [3d network of crosslinks
made of small nonpolar AA; abundant in proline and lysine
2 chains with 4 crosslinks formed by lysyl oxidase and copper
Elastase
1. degrades elastin for remdeling
2. inhibited by alpha-antitrypsin
Emphysema
mutation in alpha- antitrypsin; elastase degrades elastin in the lungs at too rapid a rate
Keratin
1. repeats of 7 AA
2. has polar and non-polar surfaces
micro to macroscale; alpha helix-protofibril-microfibril-macrofibril of har
3. cystine cross links; insoluble and not stretchy
What happens if trypsin is activated prematurely?
Accute pancreatitis; early activation [cleaving of zymogen in the pancreas, rather than in the intestines] will lead to digestion, by trypsin, of the pancreas
Types of Post-translational Modification
1. folding [aided by chaperones]
2. proteolytic cleavage [cleavage into segments or removal of ends]
3. chemical modification [phosphorylatio, glycosylation, ubiquitination]
Phosphorylation Is very common on which Amino Acid?
Serine; Threonine; Tyrosine
Glycoprotein
n-linked are constructed and then added in the RER
o-linked are built directly on in the Golgi Apparatus
How does the cell recognize which glycoproteins are destined for the lysosome?
the protein is labeled with the mannose-6-phosphate sugar
Proteosomes degrade what?
ubiquitinated intracellular proteins
Types of Enzymes?
1. oxidoreductases
2. transferases
3. hydrolases
4. lyases
5. isomerases
6. ligases
Enzymes may use which cofactors?
1. Metal ions [2/3 of enzymes need metal]
2. coenzyme
3. prosthetic group
Diagnostic lab test for pancreatitis?
blood work to look for elevated levels of pancreatic lipase and anylase
Types of Inhibitors
1. Irreversible; covalent; inactivates the enzyme [ie. Aspirin on cyclooxygenases]
2. reversible
a. competitive; higher Km
b. noncompetiteve; lower Vmax.
Control of Enzyme Activity [FAST]
[S] or allosteric controls
Control of Enzyme activity [ intermediate]
1. covalent modification
2. modular proteins
3. zymogen activation
Control of Enzyme Activity [SLOW]
transcription or translation
tPA
activates plasmin to cut fibrin
removes the clot
given medicinally to limit the size of an infarction
Aspirin
irreversible inhibitor of cyclooxygenases; inhibits clotting [ regenerated by healthy endothelial cells, not platelets]
EACA
E-aminocaproic acid; competetive plasmin inhibitor
slows clot removal [often used in surgery, but keep heparin rady]
What is special about Selenocyseine
a special tRNA for 'stop' codon
Basic Structure of the Nuclear Hormon Receptors
a DNA binding domain [DBD] and a ligand binding domain {LDB}
DNA binding proteins[ zinc fingers]
conformational change upon ligand binding.
Types of dimerization
homo dimerization [same type of protein]
heterodimerization [different type of protein]
G-Protein Coupled Receptors [GPCR]
seven transmembrane domain receptors;
largest family of cell surface receptors;
30% of drugs target GPCR;
cAMP
cyclic AMP;
created by adenyl cyclase;
activates PKA, CREB, BCL-2;
deactivated by phospodiesterase
PIP2
activated by GPCR;
activates IP3 Ca channel;
activates protein kinase C which binds to DAG
Calmodulin
helps Ca do its job
RTK activation
receptor tyrosine Kinase; dimerized and phosphorylated
RTK activates which important protein?
RAS
PTEN
tumor suppressor on the PIP2 pathway
Howis PIP3 made and what does it do?
PIP2 is converted by p85, p110, and PI3K which activates PTEN
JAK/STAT
Jaks activate stats which dimerize and transcribe
Functions of TGF-B
1. inhibit epithelial growth
2. inhibit immune functions
3. promotion of connective tissue growth
TGF-B pathway
activates SMAD2; which binds to SMAD 4; leads to transcription
What will a mutation in the TGF-B receptor lead to?
colon and gastic cancer
What will mutations in the SMAD4 protein lead to?
pancreatic and colorectal tumors
What will mutations in the SMAD2 protein lead to?
colon cancer
Why is TGF-B bad?
can exacerbate tumor growth by suppression of immune surveilance and by fostering tuor invasion and metastisis
WNT pathway
ligand binds to frizzeled and WNT; bind to axin; allowing B catenin to transcribe
Functions of WNT
cell fate; remodeling, polarity and morphology, adhesion, growth;
especially important in embryogenesis
carcinogenesis
the process by which cells acquire attributes that confer a malignant phenotype
What does telomerase do in respect to cancer?
helps the cell to avoid senescence or natural cell aging; leading to avoidance of apoptosis
The loss of cadherins is a major factor in what hallmark of cancer?
metastasis; cells are no longer bound together and may move freely
What do cancer cells do to harness more energy?
Aerobic glycolysis;
inefficient, but may get energy slightly faster
How do cancer cells escape immunodetection?
turnoff antigen expression; secrete antiproliferative factors;
TGF-B
Microsatellite instability reflects?
decreased DNA repair; NER
miRNA
microRNA
noncoding; downregulate gene expression in development and oncogenesis
Hematoma
coagulated blood tissue
choristoma
ectopic or heterotopic normal tissues [normal in an abnormal location]
granuloma
chronic inflammation
H & E staining
hematoxylin + basophilic; RNA, DNA, cartilage
Eosin - acidophilic; proteins
hamartoma
overgrowth
neoplasms
1. benign tumors
2. malignancies; metastisize
3. hamartomas
Carcinoma
cancer of the epithelium; non-glandular
adenocarcinoma
cancer of the wpithelium; glandular
sarcoma
cancer of connective tissue
leukemia
cancer blood
lymphona
cancer of lymph tissue
high-grade cancer
does not look like tissue of origin [anaplatic]
oncogene
KRAS, HER2
normal gene that, when mutated to active form, contributes toward the neoplastic phenotype
tumor suppressor gene
pRB, TP53
when tumor suppressor gene is absent, cellular events that either express the malignant phenotype proceed at a higher rate
Hallmarks of Cancer
1. resist cell death [apoptosis]
2. sustain proliferatice signaling
3. evade growth suppressors
4. induce angiogenesis
5. enable replicative immortality
6. activate invasion and metastisis
function of BCL-2
limits the signal for apoptosis
HER2
RTK; if mutated, patient may be responsive to herceptin [trastuzumab]
Function of RB
inhibit the growth signal;
antigrowth signal>cell surface receptor>cyclin D> RB> E2F
Master brake for the G1/S phase checkpoint
Prostate Cancer preferentially metastisizes where?
bone marrow>liver>lungs>brain
Pancreatic cancer preferentially metastisizes where?
Liver> lungs
Breast Cancer preferentially metastisizes where?
bone marrow>lungs>liver>brain
Colon Cancer preferentially metastisizes where>
liver> lungs> bone marrow
Key Guardian of the genome?
p53 a tumor suppressor
Chronis Myelogenous Leukemia is characterized by?
anemia and fatigue
philadelphia chromosome
BCR-ABL
What tumor suppressor works on early adenomas?
APC
What tumor suppressor works on intermediate adenomas?
KRAS
What tumor suppressor works on late adenomas?
DCC
What tumor suppressor works on carcinomas?
p53
Familial Adenomatous Polyposis
Carpeting of Polyps in the colon [AD]
usually caused by a loss of APC, a tumor suppressor
Genes expressed from the inactive X chromosome
XIST specifically[ 16 total form the pseudoautosomal region]
3 major types of fibrous proteins
1. collagen
2. elastin
3. keratin
what is the most abundant protein?
collagen
Structure of collagen?
GPx; glycine every 3 AA
triple helix; proline is posttranslationaly modified to hydroxyproline
induces mor H-bonding between fibers in the helix
What is hydroxylated in collagen synthesis?
proline and lysine
What is glycosylated in collagen synthesis?
hydroxylysine
Which way does the triple helix structure of collagen zip up?
c to n-terminus
What happens after the procollagen is secreted from the cell?
1. n and C ends are cleaved
2. tropocollagens stagger [1/4] together
3. cross-linking of lysines by lysyl oxidase
Scurvy
skin lesions; poor wound healing; fragile bones; and bloody gums after brushing
caused by a vitamin C deficiency; lysyl oxidase does not work
Kinases
phosphorylate proteins
phosphatases
enzymes that remove phosphates
What proteins usually undergoes glycosylation?
on extracellular and embrane bound proteins
How does the cell know where the protein go?
the proteins contain a sorting signal recognized by the destination site; the nucleus uses importin alpha and beta
erythrocyte
RBC [red blood cell] contains hemoglobin
Location of Myoglobin
Muscle
Locaton of Hemoglobin
blood
heme
complex of iron and porphyrin
[sheilded from water by Hb and Mb- If water does get in <MHb> methemoglobin]
Hb basic structure
2 alpha and 2 beta subunits each holding a hem in place with his
Does Mb or Hb have a higher affinity for O2
Mb
how can you tell, graphically, what has a higher affinity?
lower Km
Forms of Hb? [allosterism]
raelaxed and taut
What is the effect of lower pH on Hb?
decrease of affinity for O2
What effect does CO2 have on hemoglobin?
decreases its affinity for O2
What does BPG do to Hb?
tightens the bonding; decreases the affinity for O2
necessary to get mor O2 to the body in high altitudes
Does HgF interact with BPG?
very little
Why does fetal hemoglobin take O2 from the maternal blood?
The HgF does not bind well with bpg, increasing its relative affinity for O2 compared to the maternal blood. So as the O2 is released from the maternal blood, the fetal blood will act like myoglobin and pick it up.
Effect of CO on Hb?
binds 200X more tightly than O2, severely decreased affinity for O2;
treat with excess O2
Effect of CN on Hb?
nothing; Bad for mitochondria
how do you treat CN in body?
amyl nitrate
What does amyl nitrate do in blood?
converts Hb to methemoglobin which bind to CN; done to save the mitochondria. Should be careful in dosing, only give enough o convert some of the hemoglobin. Too much would be fatal.
Hb Kansas
ultrarelaxed hemoglobin form: cells will compensate for decreased O2 affinity by cramming 20% more Hb into the RBC
Hb S, Hb D, Hb E, Hb C
forms of sickle cell: decreased solubility and sickling occurs when the cell reaches the deoxy or tight form.
Thalassemias
truncation of Hb polypeptide; may have limited function
can sometimes activate HbF to compensate
How do you turn on HbF?
hydroxyurea
What does cyanate do in respect to Sickle Cell?
increases O2 affinity; no deoxy form
Methods of Enzyme Catalysis
concentrating, orienting, straining, dividing
How do enzymes catalyze
decreasing the activation energy by stabilizing the transitional state
Enzyme RXN pathway
E + S= ES= E+P
Assumption of Michaelis-Menten Model of Enzyme Catalysis:
[S] or allosteric controls
Why is an allosteric enzymes plot sigmoidal?
it controls itself based on the [s] and[p]
von Willebrand factor
1. exposed in the collagen of subendothelial cells
2. binds to receptors on platelets
3. aids in the formation of soft blug
Charge of activated platelets?
negative: PS flips to create the - charge on the surface
Which pathway do hemopheliacs use for the coagulation cascade?
extrinsic; they are usually fine except in muscles and joints where tissue factor is not present.
Coumarin
a competitive inhibitor for gamma-carboxyglutaminase/ vitaminK
Heparin
widely used anti coagulant helpd thrombin and antithrombin III get together
Removal or change of single amino acids can do what in proteins?
1. perturb the structure and conformation
2. remove a critical residue
3. alter the interaction of one protein with another
4. destabilize a protein resulting in a shorter biological half-life
Basic structure of an amino acid
alpha carbon flanked by an amino group and a carboxyl group; with an R constituent
What is special about glycine?
has an H side chain; small and packs tightly
What is special about cysteines?
can form a disulfide crosslink
What is special about proline?
has a cyclic side chain that is incompatible with the alpha helix that creates a kink or quick turn
Levels of Protein Structure
1. primary; peptide
2. secondary; alpha helix, beta sheets
3. tertiary; 3d structure of a single protein
4. Quarternary; multiple protein structure
Types of post-translational modifications
1. disulfide crosslinking
2. phosphorylation
3. glycosylation
4. proteolytic processing
5. accessory /co-factor
Prion
diseased protein that has undergone a conformation change; when a prion comes in contact with a non-diseased protein it will induce the same conformational change in the protein.
What is a peptide bond?
the bond between amino acids in a polypeptide chain; water is given off
Intracrine Signaling
signal is produced within a cell and targets cell
Autocrine signaling
signal is excreted and targets the same cell
Juxtacrine signaling
signal is sent via direct connection to target cell
Paracrine signaling
signal over a short distance
Endocrine signaling
signal over a long distance (estradiol)
What kind of ligand can enter the cell?
hydrophobic
Types of nuclear hormone receptors
HSP; heat shock protein
constitutively nuclear
Agonist
bind and activate
Antagonist
bind and repress
Effect of Cholera Toxin on cAMP
prolonged elevation
Effect of Pertussis Toxin on cAMP
increased cAMP levels
Notch Pathway
disposable; only one signal can be used
NF-kB Pathway
activates IkB kinase; usually involved in inflammatory or autoimmune response
HNPCC
Lynch Syndrome; hereditary non-polyposis colon cancer
What mutation predicts decreased responsiveness to monclonal Ab (herceptin) therapy agains EGFR?
KRAS mutations
BRAF
associated with melanomas mutations in the RAS pathway; may respond to BRAF kinase inhibitor
Which neoplasms are always malignant
melanoma, lymphoma, glioma, teratoma
fibro-
connective tissue
leiomyo-
smooth muscle
rhabdomyo
striated or skeletal muscle
lipo-
fat
chondro-
cartilage
osteo-
bone
lymphangio-
lymph fluid
hemangio-
blood
scirrhous
hard
zonula adherens
cadherin adhesion belt at apical end
zonula occludens
tight junctions at apical end
Desmosome
junction in cells anchored with keratin; cadherin in between cells
gap junction
pores made of connexons; known as connexins; allow rapid flow of small particles between adjacent cells
relative sizes of microfilaments
actin< keratin< microtubules
Cilia
doublets of micrtubules at the plasma membrane that aid in movement
centriole
starting point of microtubules; aid in cell division
microvilli
have an actin skeleton about 1/10 the size of cilia
gated transport
used by the nucleus
peroxisome
carries out oxidation reactions
Lysosomes
pH of 5 break down waste
mannose-6-phosphate
ligand for lysosomal pathway
1st cellular pathway
at the end of G1, starts the synthesis of DNA, asks if the environment is favorable?
G2/M checkpoint
Is all Dna replicated? Is environment favorable?
Metaphase to Anaphase Checkpoint
Are all chromosomes attached to the spindle?
Cyclin dependent Kinase
inhibited by p21 , moves cell through checkpoints
Do cancer cells have cilia?
No
Mitogen
Cell growth ligand
p53
will transcribe a protein that inhibits cdk; which inhibits mitosis and cell division
medullary
soft
colloid
muconous or secreting mucous
cystic
having a void space lined with epithelial cells
papillary
nipple-like projection
villous
having a frond-like pattern
cribriform
inside a duct/ having a cookie cutter appearance
hemidesmosomes
on the basal surface incvolve integrins
mRNA has distinctive features
1. 5' cap
2. polyA tail
3. Uracil instead of T
Central Dogma of Genetics
Trascription>Translation>Modification
Stop Codons
1. UAG
2. UGA
3. UAA
Start Codon
AUG
Alternate Splicing
The splicing of different exons in a gene to code for different proteins
What does imprinted mean?
inactivated or silenced
Mitochondria code for how many peptides and where are they utilized?
13 peptides used exclusively in the mitochondria
Do mitochondria have introns in their DNA?
no
What are analogues of purine/pyramidines currenlty being used for?
cancer treatments or antiretroviral; they are introduced into DNA and replace normal nucleotides; this causes a degredation of the DNA
DNA Cloning?
used for copying a small fragment of DNA; used to create DNA libraries.
PCR
Polymerase Chain Reaction; a fst, cheap, easy method to amplify DNA of interest using TAQ; completely in vitro
RT-PCR
reverse-transcriptase PCR; can compare with normal PCR to detect splice site mutations
Southern Blot
DNA gel electrophoresis
Northern Blot
RNA gel electrophoresis
Western Blot
Protein Gel electrophoresis
SNP
single nucleotide polymorphism
Sanger's sequencing
substitution of dideoxysugar base with a standard nucleoside; severs chain at that point; measure and sequence[ run 4 times for each type of nucleotide]
Missense
SNP leading to a different amino acid
Nonsense
SNP leading to a premature stop codon
splice site mutation
SNP leading to the addition or removal of a splice site
Types of long mutation
1. deletion
2. duplication
3. insertion
4. expansion
Frameshift Mutation
a mutation causing a change in the 'reading' frame
Anticipation
growth of a mutation [in expansion related mutation] through geneology
What test is good for large changes in DNA?
Southern Blot; also PCR
Hot' spots for Mutations on DNA
CpG islands; usually by spontaneus deamination of a methylated cytosine
NER
nucleotide excision repair; removes thymine dimers
What causes xeroderma pigmentosum?
lack of NER; only treatment is to avoid UV light
BER
base excision repair; mutations associated with colorectal polyposis
How is a mitochondrial disorder passed on?
by the mother to all of her progeny. Variable expression due to heteroplasmy.
AD
Autosomal Dominant; 50 % chance of passing it on for each child of an affected parent; variable expression due to non-penetrance, and imprinting
AR
Autosomal Recessive; 25% chance for each child and 50 % chance of being a carrier; cariable expression due to consanguinity, population genetics
XLR
X linked recessive; 50 % chance for sons of female carriers; variable expression due to x inactivation
Multifactorial Disease
Mendelian genetics vs. environmental factors
Hardy-weinberg equation
p^2+2pq+q^2=1; q= homozygous recessive
RFLP
restriction fragment length polymorphism; DNA is cut and run to determine lengths and approximate sequence using specific cutting tools
Linkage
mapping of crossing-over events of chromosomes in the tetrad formation; can use markers to determine how frequently thay are recombinant; if they are infrequently seperated, they are said to be linked
1 cM
1 centiMorgan; the genetic distance between two loci where there is only 1% chance of seperation by recombination; approximately 1 Mb
LOD score
Odds of Linkage; >3 evidence of linkage, <-2 evidence against linkage
homozygosity mapping
only useful in AR; determining location by geneology and mapping
Exome sequencing
the use of SNPs to attempt to find missense/ nonsense mutations in distinctive cases
Locus heterogeneity
mutations on different genes causing the same disease
Allelic heterogeneity
mutations on the same chromosome causing the different diseases
G-Banding
Giemse Stain; the standard for staining chromosomes during metaphase or prometaphase in order to identify them.
euploidy
chromosomes are a multiple of 23
aneuploidy
chromosomes are not a multiple of 23
A chromosomal microdeletion is best diagnosed by?
Fish Analysis; flourescent in situ hybridization
Prader Willi Syndrome needs which working copy?
Paternal UBE3A
Angelman Syndrome needs which working copy?
Maternal UBE3A
What are the possible uses of FISH?
1. diagnose chromosomal microdeletion
2. diagnose aneuploidies
3. indentify chromosome translocations [robertsonian also]
CBAVD
congenital bilateral absence of Vas Deferens
deltaF508
most common mutation in cystic fibrosis
Sweat test for Cystic Fibrosis
>60 mmol/L sweat Cl- in two occurrences; used to diagnose 90% of CF
Cystic Fibrosis
1. AR
2. chronic respiratory tract infection
3. pancreatic insufficenccy
4. elevated sweat electrolytes
5. CBAVD- most males infertile
[prevalent in caucasians of european descent]
Huntington's Disease
1. AD
2. chorea movements
3. progressive dementia
4. CAG triplet expansion[34-120 repeats] leading to a gain of function
5. anticipation present
[prevalent in europeans and esp. venezualans.
How does Huntington's disease expand?
through the paternal line
What is ptosis?
Bedroom eyes; can be associated with Oculopharyngeal Muscular Dystrophy
Duchenne's Muscular Dystrophy
1. XLR
2. hypotonia
3. progressive muscle weakness and wasting
4. gower's maneuver
5. mostly affects males
6. out-of-frame mutation
How do you test for Duchenne's MD?
1. PCR
2. Gower's maneuver
3. southern blot
4. northern blot
5. serum creatine kinase [ elevated 50-100 fold]
6. last resort- muscle biopsy
Cause of DMD?
Large deletion on the dystrophin gene causing a frameshift nonsense mutation
What is Becker's Muscular Dystrophy?
similar to Duchenne's, although the deletion resulted in an infram shift. Some dystrophin is still produced. Symptoms are milder
Exon 'hot' spots for mutations on the Dystrophin Gene
1-20 and 45-53
What is the ratio for DMD de novo cases?
0.33333333333333
How do you confirm the clinical diagnosis of DMD?
1. uimmunohistochemical staining
2. western blot
3. PCR/ southern blot
What is Gonadal Mosaicism?
Mutation in the germline cells; mother can have this mutation and not be a 'carrier' for the mutation.
Gentamicin may be used for what?
treatment of Duchenne's MD; forces a read-through of the nonsense mutation;
possibly restore reading frame and decrease severity of symptoms
Fragile X Syndrome
1. XLD
2. CGG expansion through maternal line [>230 repeats][hypermethylated]
3. FMR1 gene
4. prepubertal males; language delays; hyperactivity
FXTAS
prefragile X syndrome; expanded maternally
Sensitivity
frequency of + test when the disease is present
specificity
frequency of - test when the disease is absent
Prenatal Screening Programs
1. maternal serum alpha fetoprotein [MSAFP][identify fetus w/open neural tube defect
2. triple test [identify fetus with down's]
Types of prenatal screens [procedure]
1. amniocentesis [detects neural tube defects][ late in pregnancy]
2. chorion villus sampling [early in pregnancy]
Antisense oligonucleotides
are being used to induce exon skipping to restore open reading frame.
Number of genes in the human genome
approx. 25000
number of genes in mitochondrial DNA
37
Percentage of the human genome actually coding for proteins
1-2%
Pyramidines
CUT
Purines
AG
Acrocentric
centromere is located grossly to one side of the chromosomes [13,14,15,21,22]
Metacentric
centromere is located in the middle of the chormosome
submetacentric
centromere is located away from the middle and the ends of the chromosomes
Reciprocal translocation
parts of chromosomes are 'swapped' equally
robertsonian translocation
part of one chromosome is moved ot another
telomere
The end of the chromosomes; seems to protect the functional portion of DNA; preserved by telomerase
Tandem Repeats
short repeats of codonc or nucleotides used in paternity testing;
euchromatin
active
heterochromatin
inactive
1. always off-constitutive
2. facultative- switched on/off
methylation
process to deactivate segments of DNA; occurs at CpG islands
What is needed to synthesize DNA?
1. primers
2. DNA template
3. DNA polymerase
4. dNTPs
5.Mg+
Charge and handedness of DNA molecule
[-] charge and right-handed in beta-form
Types of DNA transfer
1. transformation
2. conjungation
3. transduction
What is a zygote?
fertilized egg
Ribosome
responsible for mRNA translation for production of amino acids
Okizaki Fragments
additions by short fragments of DNA to the 5' end of the lagging strand during semi0-conservative replication
Where does replication begin?
in an A-T rich region followed by Dna A boxes
Centromere
compacted 'middle' of the chromosome
nucleolus
where ribosomal subunits are assembled
chromatin
relaxed and jumbled chromosomes
what is a chiasma?
a tetrad formation of chromosomes in meiosis
Steps to mitosis?
1. Prophase
2. Metaphase
3. Anaphase
4. telophase
5. interphase
Steps to Meiosis?
1. Prophase I
2. Metaphase I
3. anaphase I
4. telophase I
5. Prophase II
6. Metaphase II
7. Anaphase II
8. Telophase II
What is the gestational age based on?
age based upon the time from the last normal menstrual period
normal parturition=40 wks
What is fertilization age based on?
age based on the actual time of fertilization
normal parturition is 38 weeks
What weeks in early human development are considered to be the embryonic stage?
weeks 1-8
characterized by the organogenic period
also the time most susceptible to teratogens
What are the characteristics of the Fetal period in early human development?
weeks 9-38
further development of organs and systems
extensive growth
At which point in development does it become possible to survive premature birth and why?
at 24 weeks, the lungs have developed sufficiently; however extensive care will be necessary
What happens to the lungs after birth?
Because only 10-15% of alveoli necessary for survival are present at birth, further development and growth will continue after birth.
What happens in the cardiovascular system after birth?
the closure of fetal chunts and vessels
What happens to the nervous system postnatally?
continued neuronal development
What are the five key processes that must occur for the development of a zygote into a neonate?
1. proliferation
2. growth
3. differentiation
4. pattern formation
5. morphogensis
How is differentiation defined?
the process by which cells or tissues become different from one another.
Also, the progressive acquisition of structural and biochemical specializations leading to unique or highly developed cellular functions
Define totipotent.
able to differentiate into all cell types
Define Pluripotent.
able to differentiate into many cell types
Define Multipotent.
able to differentiate into a restricted group of cells.
Define Unipotent.
determined as to differentiated cell form
Does differential gene activity lead to irreversible turning off of genes?
No
What is the SHH factor important for?
sonic hedgehog
inducing positional growth formations
What is syndactyly?
a malformation due to altered morphogenesis in which two fingers are fused together due to the lack of apoptosis in the cells between the fingers in development
Briefly describe what happens during the first week of development.
1. oocyte released and fertilized
2. rampant cleavage; development of morula
3. compaction and fusion of outer blastomeres; formation of blastocyst
4. hatching of blastocyst from the zona pellucida
5. implantation with syncytiotrophoblasts leading the invasion
In the blastocyst, what is the differentiation potential of the innercell mass?
pluripotential
What are the syncytriotrophoblasts known to secrete?
Human chorionic gonadotropin
metalloproteinases to enviade
What is the most imvsive type of carcinoma?
choriocarcinoma
What does hCG do?
stops the menstrual cycle
What is the percentage of spontaneus abortions?
thought to be at least 60%
What is an ectopic pregnancy?
pregnancy in which the implantation of the blastocyst has occurred in the fillopian tube, not the uterine body
What is meant by placental previa pregnancy?
the blastocyst implanted in the lower part of the uterine body, possibly on the cervix; leading to the placenta being below the fetus during development and possibly birth
What is the differentiatio potential of blastomeres?
totipotential
What are 6 possible indications for the use of IVF?
In Vitro Fertilization
1. blocked fallopian tubes or pelvic adhesions
2. male factor infertility
3. failed 2-4 cycles of ovarian stimulation with intrauterine insemination
4. unexplained infertility
5. vasectomy or tubal ligation
6. preimplantation genetic diagnosis
What occurs during the second week of development?
1. implantation is completed; lies totally within the endometrium
2. formation of primitive uteroplacental circulation
3. formation of bilaminar emryo
4. formation of extraembryonic membranes and cavitied
How does the conceptus hide form the maternal immune system?
1. reduces the expression of major histocompatibility complex (MHC) class I antigen
2. produces locally immunosuppressor molecules
What is the lacunar network?
network of syncytiotrophoblasts that allow maternal blood to leave the maternal vascular system and enter the embryonic system.
What is the bilaminar disk?
one layer of epiblasts on top of one layer of hypoblasts; above is amniotic cavity, below is blastocyst cavity
How is the primary yolk sac formed?
migration/ expansion of hypoblasts around the blastocyst cavity
After the thickening of the conceptus wall, what proliferates around the edges?
extraembryonic mesoderm; which will form the somatic extraembryonic mesoderm, splanchnic extraembryonic mesoderm and the extraembryonic coelom
How is the secondary yolk sac formed?
the splanchnic extraembryonic mesoderm 'pinches' inward. Excess is degraded and absorbed
What is the Chorion comprised of form out to in?
lacunar network [syncytiotrophoblasts and maternal vascular system], cytotrophoblasts, somatic extraembryonic mesoderm
what is the allantois, when is it formed, where is ite?
part of the secondary yolk sac formed caudaly of the caudal end of the bilaminar disk.
What is a hydatidiform Molar Pregnancy?
over development of extraembryonic structures; paternal only genome
what drives trophoblast development?
paternal genome
What drives innercell mass development?
maternal genome
3 characcteristics of an epithelial cell?
1. polarized
2. apical and basal surface
3. sit on top of the extracellular matrix (ECM)
3 characteristics of mesenchymal cells
1. not polarize din ECM
2. surrounded by ECM
3. migrate through the ECM
What layer of the bilaminar disk are all three germ layers formed from?
The epiblast.
What is gastrulation?
the process by which the epiblast differentiates into the ectoderm, mesoderm, and endoderm
What does the ectoderm form?
outler layer, skin, nervous system
What does the mesoderm form?
middle layer, muuscle, heart, skeleton
What does the endoderm form?
inner layer, lining of GI tract
Describe gastrulation.
1. occurs through the primitive streak
2. primitive streak is formed by the thickening of the epiblast.
3. ingression of epiblast through primitive groove forms the mesoderm
4. epiblast that ingresses throught the primitive groove replaces hypoblast to form endoderm
5. begins early in the 3rd week of development as is completed by the end of the 4th week.
Describe neuralation.
1. process by which the neural tube is formed [brain and spinal cord]
2. neural tube formed and closed by the end of the 4th week of development
3. neural crest cells form as neural tube closes
4. neural crest cell form peripharal ganglia and scwann cells of the nervous system.
Another name for neural crest cells.
ectomesenchyme
Neural tube defects are detected how?
ultrasound or measuring alpha-fetoprotein (AFP) in amniotic fluid
Why is folic acid important?
reduces the occurrence of neural tube defect.
Describe meningocele
effect caused by too late degradation of primitive groove; characterized by the meninges protruding into the spine.
What is meninges?
membrane that protects the nervous system.
Describe meningomyelocele?
primitive streak degraded prematurely; causes expose spinal cord out of the spine, but not to the surface; often leads to paralysis beyond that point.
Describe myelochisis.
the premature degradation of the primitive streak resulting in exposure of the spinal cord to the external environment. Often not compatible with life due to infections.
What is hisrchsprung's disease?
aganglionic megacolon; malformation of neural crest cells migration
What does BMP-4 do?
promotes formation of ventral structures
What does hensen's node do?
secretes factors that block BMP-4 [chordin, noggin]
How does the embryo differentiate between left and right formations?
beating of cilia, to the left from the henson node
Describe what happens during the fourth week of development.
1. organization of mesoderm into groups of tissue [notochord, paraxial mesoderm, intermediate mesoderm, lateral plate mesoderm]

2. segmentation of paraxial mesoderm into somites
3. body folding
What will the cephalic somitomeres go on to form?
the skull
What will the occipital somites go on to form?
skeleton and muscles
How many cephalic somitomers are there?
7
How many somite [occipital + trunk] are there?
42-44
In sclerotome formation [axial skeleton] what do SHH and PAX1 do?
induce formation of sclerotome and cartilage and bone formation respectively
In dermamyotome formation, what do WNT, MYF5, BMP4 nad MYOD do?
1. induce dermamytome formation
2. epaxial musculature
3. hypaxial musculature
What does Hox do?
determines the spatial structure pattern [legs where legs are supposed to go]
What causes holoprosencephaly?
defect in SHH gene possibly from alcohol consumption
What is produced when a stem cell divides?
a stem cell and a cell that must differentiate
What is transdifferentiation?
the conversion of one cell type directly to another
4 types of stem cell research.
1. nuclear transfer
2. iPS cells
3. lineage swithc
4. direct conversion
What doe s the triple screen test for
msAFP, betahCG, uE3
What does the quad screen test
all of the triple test, and dimeric inhibin A
What is pleiotropy?
a birth defect demonstrates pleiotropy when a single underlying causative agent results in abnormalities of more than one organ system in different parts of the embryo or in multiple structures that arise at different times during intrauterine life.
Define syndrome.
a syndrome is generally recognized and defined as a well-characterized constellation of major and minor anomalies that occur together in a predictable fashion presumably due to a single underlying etiology which may be monogenic, chromosomal, mitochondrial, or teratogenic in origin.
What is an association?
an association is a group of anomalies that occur more frequently togetherr than would be expected by chance alone, but that do not have a predictable pattern of recognition and or a suspected unified underlying etiology.
VACTERL
an associatio disorder [anal atresia, cardiac anomalies, other]
Sequence
a sequence is a group of related anomalies that generally stem froma single initial major anomaly that alters the development of other surrounding or related tissues or structures
Field defect
the term field defect is often used to describe raltted malformations in a particular region
Malformation
a malformation signifies that fetal growth and development did not procedd normally due ot underlying genetic, epigenetic, or environmentla factors that altered the development of a particular structure
deformation
a deformation is caused by an abnormal external force on the fetus during in utero development that results in abnormal growth or formation of the fetal structure
Dysplasia
dysplasia occurs where the intrinsic cellular architecture of a tissue is not normally maintained thoughout growth and development.
BMSC
bone marrow stem cell
non seeded technique in bladder
creating a biodegradeable scaffolding for the natural cells to use
Seeding
use of stem cells to regenerate
What is used in recent technology to dice up DNA?
miRNA
What are the number one and number two leading causes of death in the US?
1. heart disease
2. cancer
what are the top three most prevalent types of cancer for men?
1. prostate cancer
2. lung
3. colon
what are the top most prevalent types of cancer in women?
1. breast
2. lung
3. colon
What is the deadliest type of cancer?
apparently lung, also the most preventable
Stages of cancer
stage 1: small, localized, generally resectable
stage 2: larger, less resectable, still fairly localized
stage 3: lymph node involvement, large, diffuse
stage 4: metastasis to one or mor distal sites
At what level does the staging analysis occur?
the whole organ level.
How many grades of cancer are there?
4
What is the first line in cancer therapy?
surgery or radiotherapy
What is the first line in cancer therapy, if surgery and radiotherapy are not safe or effective?
chemotherapy
TPMT
thiopurine methyl transferase
metabolizes thiopurine drugs
When giving thiopurine drugs, what is it important to test for?
for functioning TPMT
What are the criteria for combination chemotherapy?
1. single therapy alone is effective (for a time)
2. should act by different mechanisms of action
3. should have different mechanisms of resistance
Are chemotherapy drugs given simultaneously in combination therapy?
no, the drugs are given individually, but alternating about every 4 weeks
When is cancer considered cured?
when it has been in remission for 10 years
Toxicities associated with chemotherapy.
bone marrow, GI mucosa, oral mucusa, skin [light sensitivity], hair follicle [alopecia], gonads.
Define Amelioration.
to make better
how to ameliorate myelosuppression in chemotherapy
1. Erythrpoietin [EPO][epoetin][epogen]
2. granulocyte colony-stimulating factor [G-CSF][Filgrastim]
3. Granulocyte-macrophage colony-stimulating factor [GM-CSF][Sargramostim/Leukine]
EPO
erythropoeitin [epoeitin][epogen]
fx: stimulates production of erythrocytes
admin: SC or IV
t.5:4-13 h
sfx: hypertension, sweating, thrombosis, myocardial infarction, iron deficiency
TX:do not use in erythroid based cancer
G-CSF
[granulocyte colony-stimulating factor][Filgrastim]
fx: stimulates the production of neutrophiles
t.5: ~3 h, but with PEGylation 15-18 h
sfx: fever, bone pain
TX: do not use in leukemias
GM-CSF
[granulocyte-macrophage colony stimulating factor][Sargramostim/Leukine]
fx: stimulates the production of monocytes and granulocytes]
t.5: ~3h
sfx: fever, rash, bone pain, muscle pain
S-phase specific chemotherapy drugs:
antimetabolites
M-phase specific chemotherapy drug:
Taxanes, Vinca alkaloids
G1-S phase specific chemotherapy:
Topoisomerase II inhibitors
Is it easier to kill dividing cells?
yes
What are cell-cycle nonspecific drugs?
1. alkylating agent
2. platinum analogs
3. anthracycline
4. antitumor antibiotics
5. topoisomerase I inhibitors
What do alkylating agents do?
[cyclophosphamide][nitrogen mustard]
fx: irreversible changes in DNA[cross-linking og guanines]
sfx: mutagenic, teratogenic, carcinogenic
USED: lymphoma, leukemia, prostate, lung, breast and ovarian cancer
What do antimetabolites do?
[s-phase][methotrexate][folic acid analog]
fx:inhibits dihydrofolate reductase, retards DNA/RNA synthesis also inhibits protein synthesis
USED: head, neck, breast and lung carcinoma, sarcoma, testicular and bladder tumors
Mercaptopurine (6-MP)
[thiopurine][antimetabolite][s-phase]
acute leukemia, chronic myelogenous leukemia
Test for mutations of TPMT!
Azothiopurine
[thiopurine][antimetabolite][s-phase]
Test for Mutations of TPMT!
6-Thioguanine (6-TG)
[thiopurine][antimetabolite][s-phase]
Test for Mutations of TPMT!
Thiopurines:
1. prodrugs requiring activation through the purine salvage pathway
2. act by inducing mutations and by inhibition of de novo purine synthesis
3. toxicity is delayed, since passage through S phase is required
4. also effective as anti-inflammatory and immunosuppressive drugs.
5-FU
[antimetabolite][Fluorouracil]
FX: blocks thymidylate synthase
converted to FdUMP, FdUTP, FUTP
USED: lung, breast, overy, prostate, cervix, bladder, head and neck carcinoma, GI adenocarcinoma
FdUMP
1. inhibits thymidylate synthase by blocking access of dUMP
2. causes dNTP imbalance and increased dUTP levels, which lead to DNA damage
FdUTP
1. incorporated into DNA
2. induces NER, which is futile in the presence of high FdUTP/dTTP levels, this leads eventually to DNA strand breaks and death
FUTP
1. incorporated into RNA
2. inhibits many processes, including processing of pre-mRNA into mature RNA and modification of tRNA
3. profound effects on cell metabolism and viability
Vinka Alkaloids
[M-phase][Vinblastine]
1. isolated from vinca rosea
FX: bind tubulin and prevent mitotic spindle formation
USED: hodgkin's disease, lymphoma, leukemia, kaposi's sarcoma, breast cancer, cervix, lung, ovary, bladder, and testis
Taxoids
[M-phase][Paclitaxel/Taxol]
isolated from yew tree bark
FX: induce polymerization and stabilization of microtubules
USED: metastatic cancer of breast and ovary
Actinomycin D
[antibiotic]
FX: intercalates in DNA and inhibits transcription and DNA synthesis
USED: Wilm's tumor, ewing tumors, osteosarcoma. Neuroblastoma
Doxorubicin
[Adriamycin][anitbiotic]
FX: intercalates in DNA
USED: acute leukemia, sarcoma, hodgkin's disease, neuroblastoma, carcinoma of lung, GI tract, endometrium, ovary,thyroid and breast, wilm's tumor, multiple myeloma
Mitomycin
[antibiotic]
FX: after intracellular activation, reacts with DNA and inhibits its synthesis
USED: carcinoma of head, neck, lung, GI tract, breast, cervix and bladder
Bleomycin
[anitbiotic]
FX: chelates metal ions>produces superoxide and hydroxide free radicals>Dna cleavage, lipid peroxidation
Topotecan
[isolated from happy tree][nonspecific cell cycle][captothecin]
FX: inhibits topoisomerase I, which leads to single-strand breaks in DNA; inhibits replication and transcription
USED: metastatic carcinoma of ovary
Irinotecan
[isolated from happy tree][nonspecific cell cycle][captothecin]
FX: inhibits topoisomerase I, which leads to single-strand breaks in DNA; inhibits replication and transcription
USED: metastatic cancer of colon and rectum
Etoposide
[Epipodophyllotoxins][S-phase]
[isolated from Mayapple plant]
FX: inhibit topoisomerase II, which leads to double strand breaks in DNA; causes errors during DNA replication and eventually apoptosis
Teniposide
[Epipodophyllotoxins][S-phase]
[isolated from Mayapple plant]
FX: inhibit topoisomerase II, which leads to double strand breaks in DNA; causes errors during DNA replication and eventually apoptosis
Cisplatin
FX: intra-interstrand crosslinks in DNA
USED: carcinoma of testes, ovary, head, neck, cervix, endometrium, sarcoma, neuroblastoma
As2O3
USED: acute promyelocytic leukemia
Asparaginase
FX: depletes Asn levels
USED: acute lymphocytic leukemia
Androgens
USED: metastatic breast cancer [may be bad]
Anti-androgens
USED:prostate carcinoma
[Flutamide]
Estrogens
USED: high dose used in postmenopausal breast cancer ; prostate cancer [may be bad]
Anti-Estrogens
[Tamoxifen]
USED: postmenopausal breast cancer, metastatic melanoma [maybe]
Letrozole
[aromatase inhibitor][estrogen pathway]
FX: prevents the conversion of testosterone to estradiol
USED: advanced breast cancer; used in tamoxifen resistance
Fulvestrant
[Estrogen pathway downregulator]
FX: leads to the destruction of estrogen receptor
USED: given to tamoxifen-resistant patients; generally used after letrozole, though some recommend skipping letrozole.
Tretinoin
[retanoic acid receptor][acid form of vit A]
FX: induce the differentiation of undifferentiated cancer cells, targets RAR
USED: acute promyelocytic leukemia [APL]
Q15d, Q3m
SFX:teratogenicity
OTHER: acne vulgaris, keratosis pilaris, cosmetics
OSFX: sensitivity to sunlight, skin irritation
Alitretinoin
[ret acid rec]
FX:target RAR and RXR to induce differentiation of undifferentiated cancer cells
USED: treatment of AIDS-related Kaposi's sarcoma
Other: severe chronic hand eczema
Bexarotene
[retinoic acid receptor]
FX:targets RXR to induce differentiation of undifferentiated cancer cells
USED: treatment of cutaneous T cell lymphoma
Other: lung cancer, breast cancer, kaposi's sarcoma
What type of treatment is used to stop cancer cells from evading growth suppressors?
cyclin-dependent kinase inhibitors
What typ of treatment is used to stop cancer cells from avoiding immune detection?
immune activating anti-CTLA4 mAb
What type of treatment is used to stop cancer cells from enabling replicative immortality?
telomerase inhibitors
What type of treatment is used to stop cancer cells from having tumor promoting inflammation?
selective anti-inflammatory drugs
What type of treatment is used to stop cancer cells from activating invasion and metastisis?
inhibitors of HGF/c Met
What type of treatment is used to stop cancer cells from inducing angiogenesis?
inhibitors of VEGF signaling
What type of treatment is used to stop cancer cells from creating genome instability and mutation?
PARP inhibitors
What type of treatment is used to stop cancer cells from resisting apoptosis?
Proapoptotic BH3 mimetics
What type of treatment is used to stop cancer cells from deregulating cellular energetics?
aerobic glycolysis inhibitors
What type of treatment is used to stop cancer cells from sustaining proliferative signaling?
EGFR inhibitors
Imatinib
[Gleevec]
FX:tyrosine kinase inhibitor [Abl, cKit, PDGFR]
USED: chronic myeloid leukemia, acute lymphocytic leukemia, gastrointestinal stromal tumors
Trastuzumab
[herceptin][stop proliferative signaling]
FX: humanized monoclonal anti-HER2 antibody used in mutated HER2 patients
T.5: 6 d
Q7d IV with chemotherapy
Q21d IV after surgery/ chemotherapy
SFX: cardiac dysfunction
Lapatinib
[kinase inhibitor]
FX: HER2 mutation
t.5: 24h
q1d
sfx: nausea, vomiting, heartburn, fatigue
Erlotinib
[Tarceva][stop proliferative signaling]
FX:EGFR inhibitor
USED: advanced NSCLC; 3 month life extension
SFX: rash, diarrhea, fatigue
Gefitinib
[Iressa][stop proliferative signaling]
FX: EGFR inhibitor
USED: advanced NSCLC
SFX: acne, diarrhea, nausea, vomiting
Cetuximab
[Erbitux][stop proliferative signaling]
FX: prevents EGF binding
USED: squamous cell carcinoma and colon cancer
TEST: check for mutation on KRAS
Bevacizumab
[Avastin][angiogenesis inhibitor]
FX: prevents VEGF from binding to its receptor
USED: metastatic breast cancer
OTHER: age-related macular degeneration
SFX: risk of bleeding, bowel proliferation
Sunitinib
[Sutent][angiogenesis inhibitor]
FX: inhibitor of VEGFRs, PDGFRs, and cKit
USED: RCC and imatinib-resistant GIST
Sorafenib
[Nexavar][angiogenesis inhibitor]
FX: inhibitor of VEGFR2-3, PDGFR, cKit, Raf-1, B-Raf
USED: advanced renal cell carcinoma and unresectable hepatocellular carcinoma
Vemurafenib/zelboraf
[Zelboraf][kinase inhibitor]
FX:inhibits V600E B-Raf, not wild type
USED: late stage melanoma; 6 months life ext.; regression for 2-18 m
Bortezomib/velcade
[Velcade][proteasome inhibitor]
FX: inhibits active site of 26 S proteasome
USED: multiple myeloma
What preserves methylation?
DNMT1
What may be unique about a cancer epigenome?
1. genome-wide hypomethylation
2. site-specific CpG island promoter hypermethylation
What is one possible bad reason for hypermethylation?
the silencing of tumor suppressors
Azacytidine
[Vidaza][DNA demethylating Agent]
FX: inhibits DNA methyltransferase after incorporation into DNA; reactivation of tumor suppressor genes
USED: myelodysplastic syndrome
5-Aza-2'-deoxyxytidine
[decitabine][DNA demethylating agent]
FX: similar to azacytidine
USED: myelodysplastic syndrome
What is HDAC?
histone deacetylases
Vorinostat/ SAHA
[Zolinza][HDAC]
FX: reactivates tumor suppressor
USED: cutaneous T-cell lymphoma
Romidepsin
[Istodax][HDAC]
FX: reactivates tumor suppressor
USED: cutaneous t-Cell lymphoma
To improve results when treating epigenetically, what would you use synergistically?
1. HDAC
2. demethylating agents
Define immunotherapy.
treatment of disease by inducing, enhancing or suppressing an immune response.
What are the three reasons to use immunotherapy in cancer treatment?
1. immune cells kill tumor cells
2. immune cells affect cancer growth and metastisis
3. adjunct treatment to ameliorate SFX of chemotherapy
IL-2
[immunotherapy]
FX:antigen binding to T cell receptor leads to secretion of IL-2
T.5:30-90 min
Admin: IV
USED: metastatic renal carcinoma and malignant melanoma
SFX: especially capillary leak [due to activated T cells damaging the endothelium]
IFN-alpha2a
[immunotherapy]
FX: induced by viral infection and produced by leukocytes
t.5: 2.5 h
Admin: SC or IM
USED: renal cell carcinoma
SFX: myelosuppression and flulike symptoms
IFN-gamma
[immunotherapy]
FX: induced by viral infections and produced by T-lymphocytes and NK cells
Admin: SC and IV
T.5: 30 min
USED: renal cell carcinoma
SFX: flulike symptoms, GI upset
Levamisole
[immunotherapy]
FX: non-specific immunostimulatory properties
USED: colon cancer, melanoma, head and neck cancer
SFX: suppress the production of leukocytes, inflammatory destruction of blood vessels
WITHDRAWN FROM US; common in COCAINE
Briefly describe adoptive immunotherapy for cancer.
Take out T cells, select and stimulate IFN-gamma producers, suppress immune system, put selected T cells back
CAR
chimeric antigen receptor [artificial T cell receptor]
Provenge
[sipuleucel-T]
FX: autologous cellular immunotherapy 'cancer vaccine'
3 courses Q 14d ($90000)
USED: metastatic, castration-resistant prostate cancer
SFX: fever, fatigue, chills, nausea, joint and headache
Ipilimumab
[immunotherapy]
FX: blocks CTLA-4
USED: late stage melanoma; possible proatate and lung cancer
SFX: potentially fatal immunological adverse effects, GI, Fever
Denileukin Diftitox
[Ontak][immunotoxin]
FX:IL-2 conjugated to diptheria toxin
USED: cutaneous T cell Lymphoma
Adcetris
[immunotoxin]
FX: antibody conjugated to poison
USED: refractory Hodgkin's lymphoma and systemis anaplastic large cell lymphoma
On what mutation was the first succesful gene therapy done on?
X-linked IL2RG mutation
ONYX-015
oncolytic virus involving p53
Measles
leads to spontaneous regression of leukemias and lymphomas
What are the common pleiotropic manifestations of neurofibromatosis 1?
1. 6 café-au-lait spots over 15mm
2. 1 plexiform neurofibroma or 2 other neurofibromas
3. axillary or inguinal freckling
4. distinstinctive osseous lesion
5. optic glioma
6. 2 lisch nodules
7. affect 1st degree relative
8. mutation in NF1
How many pleiotropic manifestations are required before you can clinically diagnose neurofibromatosis 1?
2
Genetics of neurofibromatosis
Autosomal Dominant; 50 % os cases de novo
Diagnosis of Neurofibromatosis 2
1. bilateral vestibular schwannomas, or
2. a first degree relative with NF2, plus either
a. unilateral vestibular schwannoma under age 30y
b. Two of the following: neurofibroma, meningioma, gliomma, schwannoma, juvenile posterior subscapsular lenticular opacity 'cataract'
3. unilateral vestibular schwannoma and any two: neurofibroma, meingioma, glioma, schwannoma, cataract
4. multiple meningiomas and
a. inilateral vestibular schwannoma
b. any two of: neurofibroma, glioma, schwannoma, cataract
Legius Syndrome
SPRED1
café au lait spots
axillary freckling
nodules are lipomas, not neurofibromas
mild disorder
What did Osler believe?
The body is a machine and there is one root cause proximate; look for similarity between cases
What did Garrod believe?
Disease is caused by incongruence between the body and environment, disease can come from many causes, near or far, and every case is different.