1/70
Looks like no tags are added yet.
Name | Mastery | Learn | Test | Matching | Spaced |
|---|
No study sessions yet.
Definition of malignant
cells capable of indefinite growth and metastasis - invasion of distant tissue
Definition of metastasis (1 sentence 1 bullet)
the spread of cancer from original place of formation to another part of the body
occurs when cancerous cells can enter blood vessels and find distant sites & tissue
What causes cancer (3, 2 with subbullets)
chemical substances
physical agents
ionizing radiation
x rays
some infectious agents
HPV, HBV, HCV, EBV
What is a proto-oncogene
genes that promote cell growth and division
What happens when proto-oncogenes are mutated? (1 sentence 1 bullet)
Becomes an oncogene = causes excessive growth via gain of function
ex: KRAS, HER2
What happens when a tumor suppressor gene is mutated? (1 sentence 1 bullet with 2-3 examples)
Causes excessive replication and division via a loss of function
ex: p53, BRCA1/BRCA2
What happens when a DNA Repair gene is mutated?
Allows accumulation of mutations via a loss of function
How many mutations need to occur to have cancer progress?
3
Define cancer immunoediting
our immune system is working every day to remove pre-cancerous cells. the process of trying to get rid of a tumor involves selecting for the toughest, most durable, immune-evasive tumor cells out there
What are the three proposed phases of immunoediting?
elimination = attacking the cells that can be targeted
equilibrium = state of balance between destructive & survival of best cells
escape = most aggressive and least immunogenic cells thrive and spread
How can chronic inflammation cause cancer?
Increase in cellular stress like ROS can lead to genotoxic stress that causes reactive oxygen & nitrogen species to release → increased mutation rate in cells
How can growth factors and cytokines increase cancer?
If they’re in the wrong environment they can induce cell proliferation → chronic cell proliferation
How do pro-angiogenic responses cause cancer?
Increased blood vessel growth allows for greater tumor cell invasion by providing nutrients
What is the function of NK cells in tumor eradication?
target cells with altered, deleted, or ab-labeled MHC I
What is NK cells’ mechanism of action in tumor eradication? (3 bullets)
perforin/granzyme killing
fas/fasL killing
upregulation of stress ligands by target cell
What is the function of macrophages in tumor eradication?
Bind to ab-coated tumor cells
What is the mechanism of action of macrophages in tumor eradication?
Secrete TNF-a which is shown to have strong anti-tumor activity
Name the tumor infiltrating lymphocytes (3)
T cells, NK, NKT cells
How can b cells lead to a downfall in tumor eradication? (2)
ab can bind and block the tumor Ag that the CTL would recognize
Ab can form complexes that block Fc receptors on NK and macrophages limiting their antitumor abilities
What are tumor-associated antigens (TAAs)/neoantigens?
tumors can express unique or inappropriately expressed Ags that may be detected by the immune system
What are the 4 neoantigen groups that T cells can recognize?
viral Ag encoded genes exclusively expressed by tumors
ag encoded by variant forms of normal genes altered by mutations
ag normally expressed only at certain stages of development
ag overexpressed in particular tumors
What are two oncofetal tumor antigens?
Alpha-fetoprotein and carcinoembryonic antigen
How can cancer cells evade cytotoxic T cells? Name 2 ways
Downregulation of MHC Class I expression and mutations/downregulations of second signal molecules
Name the costimulatory marker molecules (2/3)
CD80/86 on APCs
CD28 on T cells
What are the two ways cancer cells can evade NK cells?
downregulate NK activating receptor ligands
expression of mutated or absent Fas/FasL
What are three general ways cancer calls can evade both cytotoxic T cells and NK cells?
upregulation of anti-apoptotic mediators
expression of mutated or absent death receptors
increased expression of co-inhibitory receptors
Name two categories for cancer immunotherapies (that aren’t therapies)
monoclonal antibodies = antibodies that target a specific antigen, varied functions
neoantigen vaccines = stimulate immune response against personalized tumor associated antigens
Name the other two categories for cancer immunotherapies (that are therapies)
Adoptive cell therapy = isolate, expand, and stimulate immune cells ex vivo then transfer back to patient
CAR-T cell therapy = engineer immune cells to recognize specific tumor associated antigens
List the 4 steps of creating monoclonal antibodies
immunize mice with specific Ag to stimulate ab producing plasma cells
fuse plasma cells with human myeloma cells = hybridoma
select and expand for mAb-secreting hybridomas
isolate mAbs from hybridomas
Name the three activating immune mediated killing types
complement dependent cytotoxicity
antibody-dependent cell cytotoxicity
antibody-dependent phagocytosis (opsonization)
Define/explain rituximab (1 sentence 1 bullet)
an anti-CD20 B cell lymphoma mAb that can do all three immune mediated killing types
increases long term survival for B cell lymphoma patients
Explain herceptin’s clinical application
binds to the HER2 receptor, preventing signaling that may enable breast cancer cell growth
Explain the problem and solution when blocking inhibitory immune receptors
ab can target other non-cancerous cells that also express the target Ag, leading to adverse effects
use a pro-body to mask antigen binding site until it reaches tumor site, where specific proteases can degrade it
What does nivolumab bind to?
Binds to PD-1
What does ipilimumab bind to?
Binds to CTLA-4
What are the 3 steps to making a neoantigen vaccine?
sequence and compare tumor and somatic cell DNA to find mismatches
predicted algorithms determine where these mutations are in proteins and if they’re expressed on MHC/HLA
this ag can be delivered back into the patient via vaccination to help boost the immune response against the tumor
What is the goal of adoptive cell therapy?
select for the strongest anti-tumor lymphocytes
What is the 4 step process to adoptive cell therapy?
excise tumor from patient
culture tumor ex vivo with cytokines that promote growth of lymphocytes
use assay to select lymphocytes with strongest tumor killing capacity
expand and reinfuse strongest lymphocytes into patient
Define chimeric antigen receptor (CAR)
Genetically engineered receptors that involve fusion of tumor targeting antibody with T cell signaling domains
What are the three components of CAR-T cell therapy?
Targeting the antibody, CD3-zeta, and costimulatory domains
What is targeting antibody?
single-chain variable fragment that gives receptor antigen specificity
What is CD3-zeta?
Contains ITAM (immune tyrosine activating motif) for internal signaling
What are the costimulatory domains
CD28, OX40, CD137 = all promote CAR activation
What is the 4 step process for CAR-T cell therapy?
t cells collected from patient blood
genetically engineered recombinant protein is inserted into the t cell
tumor specific t cell is expanded
t cells are placed back into the patient
What can car-t cell therapy result in and what’s the solution?
Can result in cytokine storm
solution: kill switch/suicide gene, CAR-NK Cells
What does histocompatibility determine?
Determines rejection → “histocompatible” grafts share sufficient antigen similarity, decreasing change of rejection
What is the goal of transplantation? (2)
minimize graft rejection
prevent suppressing entire immune response
Who is the donor in an autograft?
Self
Who is the donor in an isograft?
Genetically identical individual (eg identical twin)
Who is the donor in allograft?
Genetically different member of same species (eg parent)
Who is the donor in a xenograft?
Different species
What are the 3 indicators of transplant success?
Blood group antigens matching (AB, Rh), MHC/HLA compatibility, and cross-matching
What is the parent-to-child graft and sibling % compatibility
parent always have 50% compatibility
siblings have 25% chance of MHC compatibility
What is cross-matching and an example of it? (1 sentence 1 bullet)
Blood test that determines compatibility by analyzing circulating antibodies, especially against HLA
ex: luminex bead-based screening assays
What do anti-rejection drugs do?
allow organ transplants between completely mismatched people
Why do certain organs have a higher success rate for transplantations? (3 reasons)
Certain organs could be an immune privileged site, being more resilient and easier to remove, or it being combined with another organ transplant
What is first-set rejection in allograft rejections for animals?
Complete by 12-14 days and memory cells are generated against graft
What is second set rejection in allograft rejections for animals?
Occurs much faster due to memory from previous graft
How do t cells mediate allograft rejections in animals? (2)
CD4+ t cells seem to be more important than CD8+ but both play a significant role
shown through adoptive transfer and t cell neutralizing experiments
What is the timing and mechanism for hyperacute transplant rejections in humans?
Timing: 24-48 hours
Pre-existing antibodies bind to graft cells and activate complement and ADCC and cause inflammation
What is the timing and mechanism for acute transplant rejections in humans?
Timing: 7-10 days
Mediated by t cells in a sensitization stage and effector stage
Describe the sensitization stage (3 steps) for acute transplant rejections in humans
presentation of Ag to T cells
T cells recognize donor MHC (direct presentation) or donor peptides in self MHC (indirect presentation)
T effector and memory cell generation
Describe the effector stage for acute transplant rejections in humans (3 bullets)
t and NK cell killing
antibody production
inflammatory cytokine and chemical production
What is the timing and mechanism for chronic transplant rejections in humans?
Timing: months to years
Humoral and cell-mediated responses, anti-rejection drugs help but aren’t perfect
What is graft vs host disease?
immune cells from donated organ expand and attack recipient
What is the function of irradiation of lymphoid regions? (2 bullets)
wipes out host’s immune cells and donor cells engraft and form a “new” immune system
used in bone marrow transplants, GvHD, and cancer treatment
What is the function of corticosteroids (prednisone, dexamethasone)? (2 bullets)
quiet immune system
given right before and after transplantation
What is the function of calcineurin inhibitors (tacrolimus and cyclosporine)
block calcium-dependent phosphatases that can activate t cells
What are 3 examples of anti-proliferative (anti-miotic) agents?
mycophenolate mofetil
azathioprine
sirolimus
What is the function of anti-thymocyte globulin (ATG) (2 bullets)
ab that bind and neutralize T cells
isolated from animals exposed to human serum
What is the function of belatacept?
soluble CTLA-4 protein that can block co-stimulation and induce T cell anergy
used in kidney transplantations