Exam 2 Material Flashcards
what is the major mechanism by which the immune system kills both tumor cells and the cells of tissue transplants ?
CD8+ cells
immune surveillance
immune system usually recognizes and eliminates neoplastic/malignant cells before they start proliferating
why are tumors often able to survive and grow in otherwise immunocompetent individuals?
tumor immunity is often incapable of preventing tumor growth or is easily overwhelmed by rapidly growing tumors
what evidence exists that tumor responses against tumors inhibit tumor growth?
lymphocytic infiltrates around some tumors and enlargement of draining lymph nodes correlate with better prognosis
what evidence exists that tumor recognition shows features of adaptive immunity and is mediated by lymphocytes?
- tumor transplants are rejected by animals (and more rapidly if previously exposed)
- immunity to tumor transplants can be transferred by lymphocyte transfer
what evidence exists that the immune system protects against tumor growth?
immunodeficient people have higher incidence of some tumors
what evidence exists that tumors evade surveillance in part by activating inhibitory receptors on T cells?
theraputic blockade of inhibitory receptors (CTLA-4, PD-1) can lead to tumor remission
5 features of immune response to tumors
- immune system protects against tumor growth
- tumor recognition shows features of adaptive immunity
- tumor recognition is mediated by lymphocytes
- tumor responses against tumors inhibit tumor growth
- tumors evade surveillance in part by activating inhibitory receptors on T cells
what must a tumor do to be recognized by immune system?
must express antigens that are seen as non-self
5 types of tumor antigens may be recognized by the immune system
- mutated self protein that does not contribute to tumorigenesis
- product of oncogene
- product of mutated tumor suppressor gene
- overexpressed/aberrantly expressed self-protein
- oncogenic virus
examples of mutated self proteins that do not contribute to tumorigenesis
various mutant proteins in carcinogen or radiation-induced tumors
examples of products of oncogenes
mutated Ras; Bcr/Abl fusion proteins
examples of overexpressed/aberrantly expressed self-proteins
tyrosinase; gp100; cancer/testis antigens
what are some examples of oncogenic viruses?
HPV E6; cervical carcinoma E7 proteins; EBV-induced lymphoma EBNA proteins
driver mutations
products of mutated or translocated oncogenes or tumor suppressor genes that presumably are involved in the process of malignant transformation
when can structurally normal self-proteins elicit immune responses?
when they are aberrantly expressed
characteristic of majority of tumor antigens that elicit immune responses
endogenously-synthesized cytosolic or nuclear proteins displayed as Class I MHC-associated peptides
how are cytotoxic T cell responses against tumors induced?
recognition of tumor antigens on APCs
from what types of cells can tumors arise?
virtually any nucleated cell (b/c they express MHC I)
3 phases of immune surveillance / tumor growth
- Elimination (immune surveillance)
- Equilibrium (immunoediting)
- Escape
immunoediting
variant tumor cells arise that are more resistant to being killed; over time, a variety of tumor variants develop
Clinical example of equilibrium phase?
woman treated for malignant melanoma 16 yrs before death; both kidneys donated to recipients who developed melanoma 1-2 years later (remember, transplant recipients are immunosuppressed)
3 immunotherapy options for tumors
- passive immunity through transfer of tumor-specific T cells or antibodies
- active T cell immunity enhanced by vaccination with tumor antigen-pulsed dendritic cell
- active immunity enhanced by blocking inhibitory T cell receptors (CTLA-4, PD-1)
immunodiagnostics
- Identification of cell of origin of an undifferentiated tumor
- Monitoring serum levels of cancer markers during treatment
cancer vaccine ideas
- heat shock proteins (clinical trials)
- genetic modifications of tumor cells
- creation of dendritic cell-tumor cell hybrid (hybridoma) – do NOT have to know tumor Ag
- recombinant IFN-alpha (renal cell carcinoma, melanoma)
- IL-2 adoptive cellular therapy (didn’t work)
cross-presentation
dendritic cell activates naive CD8+ T cell specific for antigens of a virus-infected or tumor cell that it has ingested by presenting the antigen to the T cell AND providing costimulation; also called cross-priming
problems with antibody-directed therapy
- tumor heterogeneity (some cells may not express the right antigen)
- low density of tumor antigen expression
- modulation of tumor antigen levels
- antibodies must bind to every tumor cell to kill it (no bystander killing)
ADEPT
antibody-directed enzyme/pro-drug therapy; monoclonal antibody-enzyme conjugate – nontoxic pro-drug given and metabolized to active drug to produce high concentrations of cytotoxic drug localized at tumor site
why do immune responses often fail to check tumor growth?
tumors evolve in the host to evade immune recognition or resist immune effector mechanisms
6 mechanisms of tumor cell resistance to immune recognition and destruction
- tumor stops expressing recognized antigen (antigen loss variants)
- tumor stops expressing class I MHC
- tumor secretes cytokines that suppress immune responses (ex: TGF-β)
- tumor expresses ligands for T cell inhibitory receptors (ex: PD-1)
- tumor induces regulatory T cells, which suppress CD4/CD8 effector
- tumor induces low levels of B7 costimulators on APCs → preferential engagement of inhibitory receptor (CTLA-4) on T cells rather than stimulatory receptor (CD28)
major hallmark of cancer
ability to avoid destruction by the immune system
what activates NK cells?
lack of MHC class I expression
CTLA-4
up-regulated during T cell activation; high affinity for B7; inhibits activated T cells
main strategies/goals for cancer immunotherapy
- provide anti-tumor effectors (antibodies / T cells)
- actively immunize patients against their tumors
- stimulate patients’ own anti-tumor immune responses
problems with chemotherapy / radiation treatment of tumors
damage normal non-tumor tissues; associated with serious toxicities
antibody specific for CD20 used to treat…?
B cell tumors (usually along with chemotherapy); CD20 is not expressed by hematopoietic stem cells, so normal B cells are replenished after antibody treatment is stopped
dendreons provenge vaccine
for advanced prostate cancer; extended life expectancy ~4mos; few side effects; super expensive; company filed for bankruptcy
3 ways antibody treatment could generally help destroy a tumor
- antibodies could bind to tumor antigens and either activate host effector mechanisms, such as phagocytes or the complement system, or deliver toxins to the tumor cells
- antibodies could block growth factor signaling (ex: Her2/Neu in breast cancer)
- antibodies could inhibit angiogenesis
adoptive cellular immunotherapy
T cells isolated from blood or tumor infiltrates of a patient, expanded with growth factors, and injected back into the same patient. The T cells presumably contain tumor-specific CTLs, which find the tumor and destroy it. (Variable results so far.)
example of mutated tumor suppressor gene
mutated p53
PD-1
inhibits T cells (like CTLA-4)
PD-L1/2
inhibitory ligand for PD-1
immune system “backup” for tumor cells that evade CD8+ cells?
NK cells – especially important when tumor cells don’t express MHC I
tumor microenvironment
disrupts CD3 signaling complex (zeta or epsilon chain)
TAM
tumor-associated macrophage; tumor promotes class-switching from M1 to M2, which induces angiogenesis
equilibrium phase of tumor cells
tumor cells that can evade the immune system arise and develop; Treg cells recruited
escape phase of tumor cells
tumor cell has successfully evaded immune system and begins to spread/proliferate
mice without gamma-delta T cells
develop skin cancer
mice without RAG and STAT1
increased incidence of gut and breast tumors
antibodies against CD20
can be used to kill B cell tumors
block CTLA-4 or PD-1
prevent T cell inactivation and hopefully kill tumor cell
what could kill a tumor that does not express MHC Class I?
NK cells
genes that contribute the most to the rejection of grafts
MHC genes
syngeneic grafts
from an identical member of a species
allogeneic grafts
from a different member of a species
xenogeneic grafts
from a different species
which grafts are always rejected by a recipient with a normal immune system?
allografts and xenografts
antigens of allografts that serve as principal targets of rejection?
proteins encoded in MHC
human MHC
HLA complex
MHC Class I corresponds to which HLAs?
HLA-A, -B, and -C
*remember, Class I – CD8!
MHC Class II corresponds to which HLAs?
HLA-DQ, -DP and -DR
*remember, Class II – CD4!
one of the strongest immune responses known?
response to MHC antigens on another individual’s cells
example of immunologic cross-reaction?
T cell recognition of allogeneic MHC molecules in allografts
why do T cells recognize allogeneic molecules?
- Allogeneic MHC molecules containing peptides derived from allogeneic cells may look like self-MHC molecules with bound foreign peptides
- a single allogeneic graft cell will express thousands of MHC molecules, every one of which may be recognized as foreign by a graft recipient’s T cells
- there is no mechanism for selectively eliminating T cells whose TCRs have a high affinity for allogeneic MHC molecules
how many T cells may react against an allogeneic molecule?
0.1% to 1% of all T cells (compared to 1 in 10^5 or 10^6 T cells that recognize any microbial antigen)
minor histocompatibility antigens
Non-MHC antigens that induce graft rejection (though usually not as strongly); most are allelic forms of normal cellular proteins that happen to differ between donor and recipient; important in blood transfusions and hematopoietic stem cell transplantation
direct allorecognition
T cells in the recipient recognize unprocessed donor allogeneic MHC molecules on graft dendritic cells; stimulates CTLs that attack graft
indirect allorecognition
graft cells (or alloantigens) are ingested by recipient dendritic cells; donor alloantigens are processed and presented by self-MHC molecules on recipient APCs to T cells; alloreactiv CD4+ cells (rather than CTLs) attack graft
CTLs attack graft in what kind of recognition?
direct allorecognition
CD4+ cells attack graft in what kind of recognition?
indirect allorecognition
mixed lymphocyte reaction
in vitro model of T cell recognition of alloantigens – T cells from one individual are cultured with leukocytes of another individual, and T cell responses are assayed; magnitude of response is proportional to the extent of MHC differences btwn these individuals and is a rough predictor of the outcomes of grafts exchanged between these individuals
alloantibodies
also contribute to graft rejection; most are helper T cell–dependent high-affinity antibodies
alloantibody production
recipient B cells recognize donor alloantigens and then process and present peptides derived from these antigens to helper T cells (that may have been previously activated by recipient DCs presenting the same donor alloantigen), thus initiating antibody production. (This is a good example of indirect presentation of alloantigens)
hyperacute rejection
occurs w/i minutes of transplantation; characterized by thrombosis of graft vessels and ischemic necrosis of graft; mediated by antibodies that bind to antigens on the graft vascular endothelium and activate the complement and clotting systems, leading to injury to endothelium and thrombus formation; major barrier to xenotransplantation
acute rejection
occurs within days or weeks after transplantation; due to active immune response of host stimulated by alloantigens in graft; mediated by T cells (CD4+ or CD8+) and antibodies; the principal cause of early graft failure
chronic rejection
occurs over months or years, leading to progressive loss of graft function; may be manifested as graft fibrosis and graft arteriosclerosis; culprits believed to be T cells that react against graft alloantigens and secrete cytokines that stimulate the proliferation and activities of fibroblasts and vascular smooth muscle cells; alloantibodies also contribute
most common complication associated with organ transplant survivability?
chronic rejection
current immunosuppressive therapy designed primarily to prevent what?
acute rejection (activation of alloreactive T cells)
therapeutic monoclonal therapy with an anti-CD25 agent is most similar to which drug therapy?
Rapamycin
principal cause of graft failure?
chronic rejection
mainstay of preventing and treating the rejection of organ transplants?
immunosuppression, primarily of T cell activation and effector functions
cyclosporine / tacrolimus
block T cell cytokine production by inhibiting calcineurin activation of NFAT; very effective in preventing graft/transplant rejection (compared to Azathioprine)
rapamycin
blocks lymphocyte (T and B cell) proliferation by inhibiting mTOR and IL-2 signaling
mTOR
required for T cell responses to cytokine growth factors
major problem with immunosuppressive drugs?
nonspecific immunosuppression – inhibit immune responses to more than just the graft
MHC matching
critical for the success of transplantation of some types of tissues (allogeneic bone marrow grafts) and improves survival of other types of organ grafts (renal allografts), BUT modern immunosuppression is so effective that HLA matching is not considered necessary for many types of organ transplants (heart), especially b/c # of donors is limited and recipients often are too sick to wait for well-matched organs to become available
long-term goal of transplant immunologists?
induce immunological tolerance specifically for the graft alloantigens – will allow graft acceptance without shutting off other immune responses in the host
Xenotransplantation
possible solution for problem of shortage of suitable donor organs
frequent cause of xenotransplant loss?
hyperacute rejection b/c individuals often contain “natural” antibodies that react with cells from other species and the xenograft cells lack regulatory proteins that can inhibit human complement activation
natural antibodies
mediate hyperacute rejection of xenografts; production does not require prior exposure to the xenoantigens
transfusion
Transplantation of circulating blood cells, platelets, or plasma from one individual to another
major barrier to transfusion
presence of foreign blood group antigens, the prototypes of which are the ABO antigens
ABO antigens
carbohydrates on membrane glycoproteins or glycosphingolipids; contain a core glycan that may have an additional terminal sugar
transfusion reaction
Immunologic reaction against transfused blood products, usually mediated by preformed antibodies in the recipient that bind to donor blood cell antigens, such as ABO blood group antigens or histocompatibility antigens. Transfusion reactions can lead to intravascular lysis of red blood cells and, in severe cases, kidney damage, fever, shock, and disseminated intravascular coagulation.
Rh antigen
Rhesus factor; red cell membrane protein that can be the target of maternal antibodies that may attack a developing fetus when the fetus expresses paternal Rh and the mother lacks the protein
Blood type A
N-acetylgalactosamine terminal sugar; makes anti-B antibodies
Blood type B
galactose terminal sugar; makes anti-A antibodies
Blood type AB
N-acetylgalactosamine AND galactose terminal sugars; no antibodies
Blood type O
no terminal sugars – only core glycan; makes both anti-A and anti-B antibodies
Hematopoietic stem cell transplantation
bone marrow cells or, more often, hematopoietic stem cells mobilized in a donor’s blood are injected into the circulation of a recipient, and the cells home to the marrow; used increasingly to correct hematopoietic defects, to restore bone marrow cells damaged by irradiation and chemotherapy for cancer, and to treat leukemias
Hematopoietic stem cell transplantation problems
- Before transplantation, some of the recipient’s bone marrow has to be destroyed to create space to receive the transplanted stem cells, and this inevitably causes deficiency of blood cells, including immune cells
- The immune system reacts strongly against allogeneic hematopoietic stem cells, so successful transplantation requires careful HLA matching of donor and recipient
graft-vs-host disease
if mature allogeneic T cells are transplanted with the stem cells, these mature T cells can attack the recipient’s tissues and cause a systemic inflammatory reaction characterized by rashes, diarrhea, liver disease, eosinophilia, and enlarged lymph nodes
main factor dictating graft survival?
MHC compatibility
Maternal Tolerance to Fetal Tissues
fetus expresses paternal alloantigens but is not rejected by mother; trophoblast and placenta play key role in tolerance but mechanisms unclear
Graft-vs-Leukemia Effect
mature T cells can kill leukemia cells
mycophenolate mofetil
blocks lymphocyte proliferation by inhibiting guanine nucleotide (DNA) synthesis in lymphocytes
corticosteroids
reduce inflammation; adverse effects include fluid retention, weight gain, diabetes, bone loss, skin thinning
antithyomcyte globulin
binds to and depletes T cells by promoting phagocytosis or complement-mediated lysis; used to treat acute rejection
anti-IL-2 (CD25) receptor antibody
inhibits T cell proliferation by blocking IL-2 binding; may also opsonize and help eliminate activated T cells that express IL-2R
CTLA-4-Ig (belatacept)
inhibits T cell activation by blocking B7 costimulator binding to CD28
anti-CD52 (alemtuzumab)
depletes lymphocytes by complement-mediated lysis
antibody-directed immunotherapy
inject monoclonal antibodies against tumor antigens or against signaling molecules (i.e. T cell inactivators); can combine with a toxin or radioactive molecule to try to directly kill tumor
antibody-directed enzyme/pro-drug therapy (ADEPT)
antibody conjugated with enzyme; antibody-enzyme complex binds target cell; nontoxic pro-drug given, which antibody-enzyme complex cleaves into active form at the tumor site
CSF1/CSF1R Blockade
Reprograms Tumor-Infiltrating Macrophages and Improves Response to T Cell Checkpoint Immunotherapy in Pancreatic Cancer Models
ways to improve efficacy of anti-tumor antibodies
- Humanize antibodies-make mouse Abs more human
- Smaller Abs
- Make bi-functional antibodies or fusion proteins
-Omab
fully mouse antibody
-Ximab
chimeric antibody
-Zumab
humanized antibody
-Umab
fully human antibody
Ipilimumab
human antibody directed against CTLA-4; used for metastatic melanomas; very expensive but very effective (pts survive avg 6-9mos, sometimes longer); tumor cells remain in equilibrium phase
existing cancer vaccine?
HPV-16; does not eliminate established tumors but can prevent infection
generating specific anti-tumor T cells
Adoptive T cell immunotherapy; viral vector used to recombine T cell alpha and beta chain genes with specific vector DNA; viral particles infect other T cells and cause them to express these genes; T cells become specific for tumor and attack it
Chimeric Antigen Receptor (CAR)–Modified T Cells
anti-CD19/CD137 (costimulatory receptor) / CD3zeta (signal transduction) CAR–modified T cells re-infused into a patient expanded 1000 fold; chimeric T cells still detectable 6 months after infusion; regression of axillary lymphadenopathy occurred within 1 month after infusion and was sustained; B cell lymphopenia is complication but can be managed
3 stages of tumor development
- elimination / immune surveillance
- equilibrium / immunoediting
- escape
Adoptive T cell immunotherapy
infuses the patient’s modified TCR (CAR) lymphocytes using CD3 antibody and IL-2; efficacy due to robust clonal expansion of infused cells, resulting in destruction of tumor and development of anti-tumor memory cell
most promising immunotherapy to combat already established tumors?
Generation of anti-tumor T cells
“Second Set” Graft
introducing 2nd graft from same donor to same recipient; can show that exposure to 1st graft created immunological memory
graft rejection requires what type of lymphocytes?
T cells – depletion or inactivation of T cells leads to reduced graft rejection
graft rejection is mediated by what cells?
lymphocytes – the ability to reject a graft rapidly can be transferred to a naive individual through lymphocytes from a sensitized individual
graft rejection shows which 2 cardinal features of adaptive immunity?
memory and specificity – prior exposure to donor MHC molecules leads to accelerated graft rejection
how to mitigate cytotoxic effects of steroids?
lower doses of steroids + other immunosuppressive drugs
Anti-metabolites
originally used to treat cancer but now used in post-transplantation therapy; includes Azathioprine, Cyclophosphamide, Mycophenolate
Azathioprine
purine analog that interferes with DNA synthesis; cytotoxic to T & B cells
Cyclophosphamide
alkylating agent; used in chemical weapons (nitrogen mustard)
Immunosuppressors that interfere with T cell signaling
Cyclosporin/Tacrolimus, Rapamycin
why does matching HLA between donor and recipient not prevent organ rejection?
multiple minor histocompatibility loci differences
what kinds of transplants are most likely to cause graft-vs-host disease?
Hematopoietic stem cell transplants
Type I Hypersensitivity
immediate; Th2, IgE, mast cells, eosinophils; allergy/atopy
Hypersensitivity
excessive or aberrant immune response causing injury or pathology to tissues of the body; can be:
1) dysregulated / uncontrolled response to foreign antigen causing damage
2) aberrant response against self – “autoimmunity “
allergy
Disorder caused by an immediate hypersensitivity reaction, often referring to the type of antigen that elicits the disease, such as food allergy, bee sting allergy, and penicillin allergy. All these conditions are the result of antigen-induced TH2 generation and IgE production, and mast cell or basophil activation.
Hypersensitivity reaction classification
classified on the basis of the principal immunologic mechanism that is responsible for tissue injury and disease
atopy
Propensity of an individual to produce IgE antibodies in response to various environmental antigens and to develop strong immediate hypersensitivity (allergic) responses. People who have allergies to environmental antigens, such as pollen or house dust, are said to be atopic.
development of immediate hypersensitivity reaction
- Th2 cells activated, produce IL-4 and IL-13
- B cells stimulated to produce IgE
- IgE binds to FceRI on mast cells
- multiple receptors cross-linked
- mast cell degranulation
sensitization
“first exposure” to an allergen
2 phases of immediate hypersensitivity reaction
1) immediate phase
2) late phase
immediate phase of immediate hypersensitivity reactions
develops within minutes of exposure; characterized by the release of preformed granules from the mast cell, mainly proteases and vasoactive amines (histamine), which promote vasodilation and smooth mm contraction. Over a slightly longer period of time, prostaglandins and leukotrienes also produce vasodilation and smooth mm contraction
histamine
released by mast cells in immediate hypersensitivity reactions; promotes vasodilation, increase in vascular permeability, and smooth mm contraction; does NOT play a role in bronchial constriction / asthma
late phase of immediate hypersensitivity reactions
develops 2-24 hours after exposure; activation of cytokine genes such as TNF that promote the recruitment of neutrophils and eosinophils that liberate proteases; primarily responsible for the tissue damage seen with repeat exposures
major mediator of a type I hypersensitivity reaction
mast cell
Mast cell degranulation
activated by crosslinking of the FcεRI leading to release of the preformed mediators; responsible for type I hypersensitivity
How an antigen or allergen affects the body and the extent of the response it stimulates depends on?
where antigen contacts immune system
systemic anaphylaxis
- Type I hypersensitivity
- drugs, venom, food, serum
- intravenous entry
- edema, increased vascular permeability, laryngeal edema, circulatory collapse, death
acute urticaria (wheal-and-flare)
- Type I hypersensitivity
- animal hair, insect bites, allergy testing
- entry through skin or systemic
- local increase in vascular permeability & blood flow, edema
seasonal allergies
- Type I hypersensitivity
- pollens, dust mite feces
- entry through contact with conjunctiva of eye, nasal mucosa
- edema of conjunctiva and nasal mucosa, sneezing
asthma
- Type I hypersensitivity
- dander, pollen, dust mite feces
- inhalation leading to contact with mucosal lining of lower airways
- bronchial constriction, increased mucus production, airway inflammation
food allergy
- Type I hypersensitivity
- nuts, shellfish, milk, eggs, soy, wheat, etc
- oral entry
- vomiting, diarrhea, pruritis, urticaria, anaphylaxis
dendritic cells secrete what to promote Th2 differentiation?
IL-4, IL-5, IL-9, IL-13
Th2 cells secrete what to perform effector functions?
IL-4 (germinal center rxn), IL-5 (eosinophils), IL-13 (mucus production)
how could you inhibit allergic rxn?
- inhibit the CD-40 receptor or ligand necessary for B cell activation
- inhibit IL-4 or IL-13 which promote class switching to IgE
- inhibit transcription factor STAT6, which promotes differentiation of Th2 cells
unique features of mast cells that make them ideally suited to mediate allergic rxns
- location in epithelia – can recruit pathogen-specific lymphocytes and nonspecific effectors to sites where pathogens most often enter body
- can promote mm contraction through lipid mediators
mast cells secrete what?
- enzymes (tryptase, chymase)
- toxic mediators (histamine, heparin)
- cytokines (IL-4, IL-13 [Th2]; IL-3, IL-5, GM-CSF [eosinophils]; TNF-alpha [inflammation])
- chemokines (CCL3 – monocytes, macrophages, neutrophils)
- lipid mediators (prostaglandins, leukotrienes [smooth mm contraction, vascular permeability, bronchoconstriction]; platelet-activating factor [attract leukocytes, activate neutrophils/eosinophils])
eosinophils
extremely cytotoxic granules great for fighting parasites, but can also cause tissue damage; multiple levels of regulation; after initial exposure to cytokines, threshold for degranulation drops, leading to development of allergic rxns
eosinophil regulation
- bone marrow produces very few
- require eotaxins (2nd signal) to activate & allow entry into tissues
- FcεRI not expressed constitutively, but upregulated when eosinophil activated
chronic airway inflammation
can be caused by chronic Th2 activation / response
Airway remodeling
hypertrophy of smooth muscle cells leads to thickened airway walls –> fibrosis from chronic asthma
clinical pathological triad of chronic bronchial asthma
1) Intermittent airway obstruction
2) Chronic bronchial inflammation w/eosinophils
3) Bronchial muscle hyper-reactivity to bronchoconstrictors (cold air, exercise, viral infections, pollutants)
treatment of anaphylaxis
epinephrine – vascular smooth mm contraction, increased cardiac output, bronchodilation, inhibition of mast cell degranulation
treatment of chronic bronchial asthma
corticosteroids, leukotriene antagonists, phosphodiesterase inhibitors – reduce inflammation, relax bronchial smooth mm
allergic disease treatment
1) desensitization – inject w/small doses of allergen over time to induce tolerance
2) anti-IgE antibody
3) antihistamines
4) cromolyn – inhibit mast cell degranulation
role of histamine in airway constriction?
none – no antihistamines for asthma treatment!