1/175
Looks like no tags are added yet.
Name | Mastery | Learn | Test | Matching | Spaced |
---|
No study sessions yet.
Explain the process of hematopoiesis
Formation of blood cellular components
All cells are derived from HSCs which reside in the medulla of bones
Self-renewing & Pluripotent
What does Self-renewing and Pluripotent mean
Primary lymphoid organ goal
development of immune cells
What are the cells/organs in primary lymphoid
Bone marrow (B cells)
Thymus (T-cells)
What organ gets smaller the more you age?
Thymus
So less T cells
What is the goal for secondary lymphoid organs
Imitates immunological response
What are the organs involved in secondary lymphoid organs
Spleen, MALT, SALT
What is the first line of defense against blood bone pathogens?
Spleen (filters blood)
What two organs of the secondary lymphoid organs are the most highly organized
spleen & lymph nodes
MALT
lines the gut
M cell (microfold cells) lines the gut
Antigen presenting cells → dendritic cell
SALT
Stratum corneum →keratin
Stratum granulosa → have lamellar granules (ceramides) and keratin that form filaments
Langerhans cells
will be presenting antigens
NK cells
innate immune response
causes cell destruction by perforins & granzymes
produces cytokines that activate adaptive immune response
monocytes
involved in tissue repair and remodeling
destroy pathogens; produces cytokines
leave circulation & mature in either macrophages or dendritic cells
Macrophages: phagocytes in tissues
Dendric cells: present antigens to other immune cells to help destroy Dendritic
In neutrophils, are there more segmented or bands
there are more segmented. Remember bands are the babies!
Absolute neutrophil count
WBC x (%segs + %Bands) divided all by 100
Neutrophils
First responders
● Contains granules with lysosomal enzymes for bacterial degradation
● After infections, degradation of neutrophils leads to pus
What immune cell is most abundant (from lowest to highest )
Basophils → neutrophils (bands) → lymphocytes → neutrophils (segmented)
Are mast cells & basophils the same?
NO! They are two different cells
What is the function of basophils/mast cells
inflammation & allergies
What does heparin do
prevent clotsW
What causes inflammation
Histamine, Serotonin, Leukotrines
Eosinophiles
antiviral & anti-parasite
Eosinophil derived neurotoxin damages nerve cells
Major basic protein and peroxidases destroy parasistes
T cell major adaptive immune defense
against intracullar infections
Humoral immunity (antibodies) major adaptive immune defense
against extracullar infection
involves the transprotation of b lymphocytes into plasma cells that produce & secrete antigens
T/F
The complement system is produced in an inactive form
True
They are sequentially activated in three different ways: classical pathway, alternative pathway, lectin pathway
What are the main functions of the complement system?
Cytolysis → C3b
Membrane attack complex (MAC) breaks down & destroys cells
Inflammation → C3a & C5a → bind to mast cells, basophils, platelets to release histamine & chemotactic
opsonization (C3b)
Pathogens coat the surface
What are interferons?
Family of cytokines secreted by host cells to modulate immune respones
Type 1 vs Type 2 interferons
Type 1: INF-a & INF-b which is released by host cells in the presence of pathogens, alerting the other cells via antiviral protiens (AVPs)
Type 2: y-IFN (inflammation, stimulates other immune cells)
What interferon is used to treat multiple sclerosis
INF-b
What interferon is used to treat hairy cell leukemia
INF-a
What cells produce INF-y
Produced by activated T and NK cells
When released, it calls leukocytes to the site of infection, creating inflammation
Autoimmune diseases have been correlated with the upregulation of y-INF
CD4 T helper 1 cell
activates cell mediated immunity pathway
delayed hypersensitivity
CD4 T helper cell 2
drives B cell proliferation
secretes IL4.5.13
CD4 T helper 17 cell
promotes inflammation
secretes IL-17
CD4 regulatory T cells
prevents autoimmunity
controls specific immune response
CD8 Cytotoxic T cells
delayed hypersensitivity
graft (organ) rejection
What are APCs
■ Antigen presenting cells are immune cells that help activate T cells by giving them pathogens. These include B cells, macrophages, and dendritic cells.
■ For a T cell to become fully activated, it needs TWO signals from the APCs
■ If there is only one signal but not two, this leads to anergy. This is a state where T cells become unresponsive even if it sees that antigen again. This is important to prevent autoimmunity.
What two signals are needed from APCs
● Antigen-specific: this is when the T cell receptor on the T cell recognizes and binds to a specific antigen presented by MHC on the APC.
● Co-stimulatory signal
○ This is a safety check. The APC gives an additional singal though other modules, like CD80 binding on the T cell (CD28) to confirm that this is really a threat!
What cytokine does Treg release
IL-10
Predict the outcome of having a drug that inhibits CD28
T cells will experience hypo response when APC cells present antigens
Predict the outcome of a drug that inhibits CD28
CD40 L of T cell could still interact with CD40 of B cells
What cells are involved in the complement activation
IgM & IgG
igE
allergy & asthma responses mediated by mast cells and basophils.
What is isotope switching
IgM to other Igs.
antibodies recognizing the same antigen but different types.
What must happen for antigen switching
B cell must receive costimulatory signals from CD40 to engage in isotype switching
Different cytokine signals determine the isotype
Activation-induced deaminase (AID) enyzme requires for isotype switching and affinity maturation
Explain isotype switching
○ Occurs in B cells
○ When B cells are first activated by an antigen, they typically produce igM antibodies.
○ Isotype switching is the process of changing the type of isotype of antibody they produce, from IgM to igG or igA, or igE.
Characteristics of an antigen to trigger the immune response
size
chemical composition & heterogeneity
foreignness
uptake & degradability
What are the two self antigens
HLA
MHC
(RBC do not have HLA, instead they have ABO/rh —> determines blood type)
what cells are the antigen presenting cells
dendritic
b cells
macrophages
Self antigen: HLA class 1
● A, B, C, E, F, G
● All nucleated cells & platelets
● Present to CD8+ T cells
Self antigens:
HLA class 2
■ HLA class 2:
● DR, DP, DQ
● Antigen presenting cells: Dendritic, macrophages, and B-cells
● Presented to CD4+ T cells
self- antigen
HLA class 3
● Encodes for TNF, complement, soluble factors
What is tolerance
state of unresponsiveness in which the lymphocytes remain alive, but inactive against a particular antigen
i.e mom doesn’t fight fetus
central tolerance
■ Central tolerance is the immune system quality control check that happens early in the development in B cells (occurs in the bone marrow) and T cell (in the thymus). This makes sures our immune cells do not attack our own body.
■ The B & T cells are engaged in negative selection. This process occurs by putting self-proteins before the cells, and seeing how they react to it. If they react & try to kill it, they are apoptosis.
■ Sometimes, a few T cells react to it just a little bit, jut not enough to be dangerous. These cells become natural T regulatory cells, aka Tregs. They leave the thymus and express FoxP3, a transcription factor that suppress the immune response.
What is peripheral tolerance
■ Peripheral tolerance occurs after the T cells have already left the thymus and are circulating in the body. Their job is to catch & control any self-reactive T cells that escaped that prior screening in the thymus.
■ Occurs though three mechanisms: Anergy (T cells gets turned off) ; Apoptosis (programmed Cell death); become Tregs
How do Tregs work
■ Tregs use inhibitory cytokines like IL-10 & TFG-B & checkpoint molecules like CTLA-4 & PD-1 to send stop signals to immune cells trying to activate
what do tregs need to function
■ Tregs need IL-2 & FoxP3 to function
what triggers the apoptosis pathway
● FasL binding to FasR which triggers apoptosis (this pathway is also used by CD8+ cells)
FasL is also expressed in what cell
NK
what cell is protected against induction of apoptosis
a cell expressing FasL
if FasR is activated, the cell will apoptosis
what happens if a cell does not express PDL-1
co activation of the T cell is predominantly causing immune response
patients will have a higher prevalence of autoimmune disease if they have mutations in
CTL4 & FoxP3
Tumor cells silence the immune system by expressing
PDL1 (anergy)
To treat this: PD-1 & PDL-1 inhibitors activate the immune system
PDL1 is also expressed in
the placenta
it maintains the fetus during pregnancy
testis, retina, and some cancers express _ so they are always protected aginst imune response
FasL
B cells express
CD20
Small molecules vs biologics
Small molecules
targets protein kinases, nucleic acids
JAK inhibitors (tofacinib & methotrexate)
chemically synthesized, low weight, less immunogenic
Biologics
large, compelx molecules from living organisms
IV, SC, costly immunogenic
Monoclonal antibodies like infliximab & fusion proteins (etenercept)
autoantigen
Self antigen on body cells ( HLA & CD markers)
In autoimmune, they are idenitfied as threats
What do we do Pharmacologically for autoimmune diseases
enhance tolerance by boosting inhibitory cytokines (IL-10 & TGF-b)
enhance treg cells by boosting FoxP3, IL-2, PD-1
inhibit immune activation by blocking proinflammatory cytokines (TNR-a, IL-17)
supress T/B cell activation
target protein kinases, cell surface ligand/receptors, nulecic acid pathway
Monoclonal antibodies
● One clone of a B cell that has high specificity and reproducibility. It only recognizes one epitope.
● Produced by hybridoma method
● Used for targeted therapy and precision tests
Polyclonal antibodies
● Many clones of B cells that recognize multiple types of epitopes. They have lower specificity, and their reproducibility varies by batch.
● They are produced by animal immunization
● Used for general detection & broad screening
what do biologics target
○ Receptors: TNF & IL receptor
○ Ligands: TNF-a, IL-17
○ Cells: B cells, T cells
○ Signaling pathways: JAK-STAT, NF-B
Biologics vs biosimilars
Biologics = original and fully tested complex drug made from living cells.
Biosimilars = close copies; not generic drugs but similar versions requiring comparability studies. Only some biosimilar drugs achieve interchangeability
what biologics are a higher risk for hypersensitivity
monoclonal antibodies, especially chimeric and murine derived
Compare the PK of small molecules with biologics
○ Small molecules
■ Oral, rapid absorption
■ Disturbed widely, often though passive diffusion
■ Eliminated via renal or hepatic metabolism (CYP450)
■ Low immunogenicity
■ Short half life
○ Biologics
■ Absorption through SC/IV, slow( lymphatic uptake)
■ Distribution is limited by size, receptor mediated
■ Eliminated via proteolysis by hepatocytes, antigens
■ Long half life (up to 3 weeks due to FcRN recycling)
■ High immunogenicity
what are the accreditation organizations for specialty pharmacy
URAC, ACHC, JCAHO, CPPA, NABP, and NCQA
Autoimmune conditions
often caused by a dysregulation of inflammatory cytokines (cytokines are proteins that communicate between other cells)
Proinflammatory cytokines activate immune cells & release more cytokines, starting the inflammatory cascade
Centralized vs decentralized specialty pharmacist
Centralized works from a call center
Decentralized works in a clinic
What is insurance formularies Tiers 1-4
Insurance formularies are a list of drugs covered by a drug plan
■ Tier 1 = typical, generic drugs that are automatically approved
■ Tier 2 = brand name drugs or more expensive generics
■ Tier 3 = more expensive brand name drugs or non preferred
■ Tier 4 = specialty drugs, new approved pharmaceutical drugs that payer want to discourage because of cost
What is step therapy
Trying less expensive drugs before stepping up to pricy drugs
what two things ensure that medications are prescribed appropriately
prior authorization & step therpay
T/F
Biologics are large weight
True
How are biologics adminstrered
parenterally
Can biologics have biosimilar products?
■ if considered to have no clinical different in safety, purity, and potency to the reference product
Can small molecules have generic products?
■ products if considered bioequivalent to the innovator product
Why would you use monoclonal antibodies (mAbs)
■ Type of biologic that can be used for more specific targeting and more tolerable safety profile
● End in mab (i.e: dupilumab)
■ The MOA can be direct cell toxicity, immune-mediated cell toxicity, vascular disruption, and the modulation of the immune system
How are biosimilars, which differ from batch to batch, show their slight variants?
they show variations in protein mixes by adding 4 random letter suffixes to the name
denosumab-bbs
why would biosimilars go though additional testing?
■ testing to be considered interchangeable to the reference product (allows for pharmacist substitution pending specific state laws)
What determines the categorization or workflow for a specific medication
Medication risk assessment (MRA) or proactive risk assessment (PRA
What percent of drugs are specialty drugs
75% of 7000 new drugs
They take up 50% of the drug spending
What are the leading areas of drugs
Immunology & oncology
Type 1: hypersensitivity reaction
Immediate
IgE mediated
(triggered by an allergen binding to IgE on mast cells
Mast cell degradation releases histamine
Type 2: hypersensitivity reaction
Antibodies IgM or IgG bind to antigens, activating the complement system & NK & macrophages to destroy cells
Type 3: hypersensitivity reaction
Immune complexes (antigens + IgG & IgM deposit in tissues, triggering inflammation & tissue damage
Type 4: hypersensitivity reaction
Antigen is processed & presented to T cells
CD8+ T cells & Th1 cells release cytokines
Pseduoallergy hypersensitivity reaction
Not antibody mediated; no IgE involved
direct activation of complement & mast cells
How do WBC and immune system get affected by glucorticoids
○ Glucocorticoids decrease circulating lymphocytes, monocytes, eosinophils, basophils, and macrophages, but they increase neutrophils (via bone marrow release and inhi ition of diapedesis)
What are the pro-inflammatory cytokines
IL-1,2,3,4,5,6,8,TNF
What is the MOA for antihistamines
blocks H1 histamine receptors, but do NOT block histamine release