• 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

1/68

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;

68 Cards in this Set

  • Front
  • Back

5 general principles of Organ Transplantation

1. Match compatible blood type & HLA


2. Multi-drug immunosuppressive tx for synergistic effect (allows lower dose/toxicity for each drug w/ higher efficacy)


3. Intensive induction tx, followed by lower-dose maintenance


4. Monitor for rejection, drug toxicity, & infection


5. Reduce/remove drugs that are causing more problems than benefits



Induction therapy typically involves a combination of ..........

immunosuppressive + biologics (monoclonal or polyclonal antilymphocytic Abs)

Induction therapy w/ biologic agents is used to delay the use of nephrotoxic calcineurin inhibitors & to intesify immunosuppresson in high risk pts?




What pts are at high risk of rejection?

repeat transplant pts


broadly presensitized pts


African-American pts


pediatric pts

Biologics may be either polyclonal or monoclonal. Which are safer?

Monoclonal: anti-CD3 mAb


(muromonab-CD3 or OKTC), anti-IL-2R mAbs (daclizumab, basiliximab)




*monoclonals are made from a single source & have more specificity & less variability in efficacy/toxicity

How are Monoclonal Ab's made?

Hybridoma technology:


fuse tumor myeloma cell w/ splenic b cell-->


grow in culture media where only fused cells can grow

_________ is used as induction therapy tx of acute organ transplant rejection




MOA: binds E chain of CD3 on T cell receptor complex-->


internalization of T cell receptor-->


rapid depletion of T cells


&


prevents remaining T cells from recognizing Ag-->


prevents T cell from producing IL-2-->


reduced T cell function

muromonab-CD3




*made using mice

Why are repeated txs w/ muromonab-CD3 (or other mouse monoclonal Abs) CONTRAINDICATED?

repeat use-->


immunization of pt against mouse determinants of Ab-->


neutralizes immunosuppressive effects

Glucocorticoids are administered before injection of muromonab-CD3 to prevent the release of cytokines & reduce first dose rxns.




*also require resuscitation facility




What SEs does muromonab-CD3 toxicity cause?

-"cytokine release syndrome" = fever, chills, N/V, malaise, diarrhea, myalgia--> ARDS, PE, CV collapse (rare, but fatal) (reduced w/ glucocorticoids & successive doses)


-Anaphylaxis


-"Rebound" organ rejection (when stopped)


-Inc infection & cancer



What cytokines are responsible for "cytokine release syndrome" (30 mins after injection)?




(cytokines are released by activated T cells or monocytes)





TNF-alpha


IL-2 & IL-6


IFN-gamma

Why are polyclonal Ab's not as safe?

polyclonals:


lymphocyte immune globulin (ATGAM)


antithymocyte globulin/ ATG (THYMOGLOBULIN)




are made from multiple sources & efficacy & toxicity varies btwn batches, HOWEVER they have a broader target & are more potent

How are polyclonal Ab's made?

repeated injections of human thymocytes (ATG) or lymphocytes (antilymphocyte globulin, ALG) into animals-->


then purifying the serum immunoglobulin fraction

_____________ can be used in combo w/ monoclonal Abs for induction therapy, initial rejection, & steroid resistant rejection




MOA: act on small, long-lived lymphocytes-->


deplete T cells ("thymus dependent" lymphocytes)

ALG (Antilymphocyte Abs)




*obtained from immunization of large animals (horses, etc)

What are the SEs of ALG toxicity?

Pain & redness around the injection site


Anaphylaxis & serum sickness (type 3 HSN)


Histocytic lymphomas


Inc risk of cancer




*depletion of T cells can also impair DTH & cellular immunity

________ is used for induction therapy in acute renal transplant rejection (w/ immunosuppressives) & acute rejection of other organs, & to reduce severity of GVHD after BMT




MOA: deplete circulating lymphocytes by direct cytotoxicity (BOTH complement & cell-mediated) AND block lymphocyte fxn by binding cell surface molecules

ATG (antithymocyte Abs)




*purified gamma globulin obtained from immunization of rabbits

ATG contains various cytotoxic Abs that bind to what targets?

CD2


CD3, CD4, CD8 (T cells)


CD8, CD 11a, CD18


CD 25, CD44, CD45


HLA class I & class II on T cells

What are the SEs of ATG toxicity?

fever, chills, & hypotension


Serum sickness (type 3 HSN) & glomerulonephritis


Anaphylaxis (rare)


Leukopenia, thrombocytopenia


Inc risk of infection & cancer

What are the 2 types of biologics used for induction therapy?

1. depeleting agents- destroy host immune cells (lymphocyte immune globulin, antithymocyte globulin, & muromonab-CD3 mAb)




2. immune modulators- prevent action of immune cells (muromonab-CD3mAb, anti-IL-2R mAb (bind to alpha chain of IL-2R & block IL-2 from activating T cells)

T/F


muromonab-CD3 mAb is BOTH a depleting agent & an immune modulator

TRUE

What is the difference btwn chimeric & humanized monoclonal Abs?

chimeric = entire variable domains are from mouse, rest of Ab is human




humanized = CDR (complementary determining region) of the variable domains is human, rest of variable domain is mouse


(may be a mix^)




*CDR determines Ag binding specificity for Fab (Fab is formed by the variable domains of the light & heavy chains, coming together at the N-terminal ends)(constant region of heavy chain = Fc portion, determines isotype & fxn (type of Ig))

What is the downfall of chimeric monoclonal Abs?




(humanized have longer half-life & are better)

recipients eventually develop Ab's to the mouse variable domains & neutralize them-->


inhibits therapeutic effects


(eventually become resistant to meds*)

_______ & ________ are IgG1 monoclonal Abs that bind IL-2 receptor alpha chains on activated lymphocytes--->


block IL-2 from binding




*immune modulators (less SE's than depleters*) used in Induction phase (prophylaxis)




Which is humanized & preffered?

Daclizumab (zenapax) & Basiliximab (simulect)




*Daclizumab is humanized & preferred over chimeric basiliximab

What drugs are used in combo for Maintenance therapy?




(Induction phase is predominantly biologics = Abs)

Calcineurin inhibitor + Glucocorticoid + Mycophenolate mofetil (purine metabolism inhibitor)




(usually cut out glucocorticoids in diabetic pts receiving pancreatic transplants)

_____________, is used in maintenance therapy*




MOA: targets downstream pathway of T-cell receptor signalling-->


binds cyclophillin-->


inhibiting calcineurin-->


inhibits NFAT production of IL-2, IL-3, IFN-gamma-->


no T cell proliferation/activation (w/o IL-2)


no B cell or granulocyte activity*

Calcineuron inhibitor: Cyclosporine




(normally, Ag binds to T cell receptor-->


IP3 pathway activation-->


intracellular influx of Ca2+-->


activates calcineurin (phosphatase)-->


dephosphorylates NFAT-->


NFAT enters nucleus & binds DNA-->


IL-2, IL-3, etc production-->


T cell proliferation (IL-2)

__________ can be used for BOTH solid & BMT transplants




*BUT does NOT work on primed T cells (that have already seen Ag), only on naive cells




*causes severe nephrotoxicity!!

Cyclosporine

__________, is also a calcineuron inhibitor that is used in maintenance therapy*, of SOLID organ transplant




* Targets downstream pathway of T-cell receptor signalling by binding to BOTH cyclophilin & FKBP--> more POTENT effects, but also more toxic*


What SE's does it cause?

Calcineurin inhibitor: Tacrolimus




SE's: (2N, 3H)


Nephrotoxicity*


Neurotoxicity


Hyperglycemia


Hypertension


Hyperkalemia

________, is also a calcineurin inhibitor, used in maintenance therapy*, of solid or BMT transplant but it DOES NOT cause nephrotoxicity (less potent)




What SE does it cause?




What is the MOA?

Calcineurin inhibitor: Sirolimus (Rapamycin)




SE: myelosuppression (thrombocytopenia)




MOA: Targets downstream pathway of IL-2 receptor signalling-->


binds mTOR on FKBP-->


inhibits mTOR (kinase)-->


prevents T cell proliferation



T/F




Although Sirolimus (Rapamycin) is considered a calcineuron inhibitor, it DOES NOT inhibit calcineurin OR inhibit production of IL-2

TRUE

__________, used in maintenance therapy*




MOA: bound to CBG in circulation-->


dissociate & enter cell-->


bind to INTRACELLULAR receptor (GR)-->


induces conformational change-->


causes GB to dissociate from HSP90 & IP-->


enters nucleus w/ GR (as complex)-->


complex binds GRE (on DNA)-->


gene transcription-->


suppress cytokine (IL-1 & IL-6) production

Glucocorticoids (corticosteroids)




*decr cytokine production via neg feedback (low cytokines--> decr endogenous cortisol production)

How do corticosteroids downregulate inflammatory responses (anti-inflammatory effects)?




(*inhibit cellular immunity)

1. inhibition of inflammatory mediators- downregulation of IL-1 & IL-6, T cells inhibited from making IL-2 & proliferating




2. inhibition of inflammatory cell migration- interfere w/ neutrophil & monocyte activity




3. promotion of lymphocyte apoptosis- via increased IkB--> decr NFKB & activation of endonucleases

Glucocorticoid (corticosteroid) uses?




SE: growth retardation in children, hyperglycemia, cataracts, osteopenia, avascular necrosis of bone, inc risk of infection

Uses:


-maintenance therapy for BOTH solid organ transplants & BMT


-autoimmune dz (flares)


-asthma (inhalant w/ beta agonist, avoids systemic SEs)




(try to avoid in diabetics if possible*)

____________is an antimetabolite used for maintenance therapy* post-solid organ transplant & post-BMT, lupus, & RA


MOA:


Inhibits monophosphate dehydrogenase-->


inhibits purine synthesis-->


Inhibits T & B cell production




What is the active metabolite that this drug is hydrolyzed to?

Mycophenolate mofetil




hydrolyzed to mycophenolic acid

_________ is a cytotoxic antimetabolite used for kidney transplants* (renally excreted!)




MOA:


converted to 6-mercaptopurine-->


the to 6-thioinosinic acid-->


inhibits inosinic acid-->


Inhibits purine synthesis-->


inhibits DNA replication & lymphocyte proliferation

Azathioprine




*used in anything involving immunosuppression of kidney*

Azathioprine degradation (via xanthine oxidase & renal excretion is inhibited by _____________




*requires dose lowering of azathioprine

Allopurinol

_________ is an alkylating agent-->


destroys proliferating lymphoid cells




*used in SLE, MS, Wegener's granulomatosis




*may cause hemorrhagic cystitis!

Cyclophosphamide

_______ is the DOC* for RA & GVHD (post-BMT)




MOA:
inhibits pyrimidine synthesis (ONLY ONE!)-->


inhibits DNA synthesis-->


prevents lymphocyte proliferation

Methotrexate

What drugs are used to tx transplant rejection?

high dose PULSATILE glucocorticoids


polyclonal antilymphocyte Abs (ALG)


muromonab-CD3 mAbs




(biologics (polyclonal & monoclonal) are the MOST potent agents!!)

If rejection is d/t high levels of anti-HLA Abs (anti-alloantigen Abs), what can you do to decrease these?

plasmapheresis (remove the Abs)


IVIG (competitive inhibition, IVIG binds Ags)




*can be used in any disorder where pt is creating Abs to something in body (alloantigens or autoantigens (attacked by autoimmune abs)

_________ is the DOC for ITP, Autoimmune hemolytic anemia, & Acute Glomerulonephritis (AGN)

prednisone (glucocorticoid)

Autoreactive tissue disorders (SLE, RA, MS, IBD) are initially tx w/ DMARDs (prednisone, cyclophosphamide, methotrexate, etc), if these fail, what drugs are used?

biologics:


Anti-TNF Abs (infliximab, etanercept, adalimumab)


Abatacept (recombinant fusion protein)

Methotrexate is the DOC for RA, if sxs remain, what is the next step?

add Anti-TNF Abs: Infliximab, etanercept, adalimumab



_________, a chimeric IgG1 monoclonal Ab-->


Anti-TNF-alpha




-used for RA & Crohn's disease*




SE: develop ANA (already present in RA) & normal monoclonal Ab SE's


*CI in CHF pts

*Infliximiab (remicade)

______, dimeric fusion protein w/ human IgG1-->


Anti- TNF-alpha & TNF-beta




-used for RA, polyarticular juvenile RA*, & psoriatic arthritis*




SE: develop anti-dsDNA Abs (seen in SLE), ANA

Etanercept (enebrel)

_____, human IgG1 monoclonal Ab-->


Anti-TNF-alpha




-used in RA (only)

Adalimumab (humira)

________ may also be used in pts w/ RA refractive to methotrexate (DMARDs) & is cheaper than Anti TNF-Abs w/ less global SEs




*CI w/ Anti TNF-Abs & w/ Anakinra (IL-IR antagonist, also used for (adult) RA not responding to DMARDs)

Abatacept

Abatacept is a recombinant fusion protein composed of chimeric CTLA-4 + human IgG Fc




MOA?

CTLA-4 on T cells binds to CD80/CD86/B7 on APCs (Abs)-->


prevents costimulatory CD28 on T cells from binding to B7 (TCR recognizes autoimmune Ag presented by MHC on APC, but also requires costimulatory binding to be activated)-->


prevents T cell activation-->


T cell anergy-->


decreases the amount of autoreactive T cells


& CTLA-4 binding to B7 sends inhibitory signals to active T cells-->


down-regulating activity

________ is a humanized IgG1 monoclonal Ab that binds to IgE fc region-->


IgE antagonist




*used to tx asthma

Omalizumab




(per Krishna)

Multiple Sclerosis (MS) is an inflammatory disease of CNS white matter. Auto-Abs to _____ & _____ cause (triad):


mononuclear cell infiltration


demyelination


scarring (gliosis)




*predominantly in females (20-35)


*Type 4 HSN**

Auto-Abs (activated T cells) to


myelin basic protein (MBP) &


myelin oligodendrocyte glycoprotein (MOG)




*causes demyelination, sx depend on area affected

How is relapsing-remitting MS (85% of cases) tx?




(reduces relapses by 33%)




what is the MOA?

IFN-Beta (1a or Ib)


(or w/ Natalizumab, per Krishna)




MOA:


interferes w/ costimulatory molecules (B7/CD28 & CD40/CD40L)-->


reduces T cell activation




deviates response toward TH2-->


dampens TH1 response




interferes w/ T cell adhesion to endothelium-->


prevents migration across BB

When relapsing-remitting pts who have been taking IFN-beta begin to show neurological deterioration, it becomes secondary progressive MS.


How is this tx?




(reduces relapses by 67% (better than IFN-Beta)




MOA?

Mitoxantrone




MOA:


intercalates DNA-->


inhibits DNA synthesis

Immunostimulatory drugs are used to tx?




(immunizations, cytokines, levamisole, thalidomide, BCG)

infection


Immunodeficiency


cancer

_____________ is used post-surgery for Dukes class C colorectal cancer & to tx Hodgkin's disease




*stimulates immune response by activating macrophages




what SE's does it cause?

Levamisole (ergamisol)




(originally anti-parasitic)




SE: agranulocytosis

__________ is used in Multiple Myeloma, Erythema nodosum leprosum (leprosy), & skin manifestations of SLE




*Has mixed effects--> inhibits TNF-alpha & neutrophils & inc IL-10 (immunosuppressive), but also Enhances Cell Mediated Immunity




what SE's does it cause?

Thalidomide (thalomid)




(originally sleep aid)




SE: teratogenesis, thrombosis (in multiple myeloma pts)

Multiple myeloma pts taking thalidomide can be given _________ if they develop thrombosis

Warfarin




(Gemcitabine is also an alt. tx for multiple myeloma & lymphoma per Krishna)

__________ is used as an adjuvant in bladder cancer




*activates macrophages-->


enhances cell-mediated immunity

BCG (Bacille Calmette-Guerin)




*viable strain of bovine Tb (mycobacterium bovis)

Without _____________ there is no immune system.




What limits their use?

cytokines (IFN, CSF, IL)




SEVERE systemic SEs--> fever, flu-like sx, anorexia, fatigue, malaise (may be enough to kill pt)

(G-/GM-)CSF & IL-3 push hematopoetic cells towards ________ lineage




What is G-/ GM-CSF used for?

myeloid lineage




GM-CSF used in pre-term neonates, neutropenic pt, & cancer pts receiving chemo

_______ pushes hematopoetic cells towards lymphoid lineage

IL-7

_______ is used to tx hairy cell leukemia, malignant melanoma, kaposi's sarcoma, & Hep B & C

IFN-alpha




(IFN-beta used in MS*)

_____ is used to tx chronic granulomatous disease

IFN-gamma

_____ is used to tx METASTATIC renal cell carcinoma




*non-metastatic tx w/ surgery*

IL-2

___________ immunization involves administration of pre-formed abs from vaccinated individual (artificial) or from mother to fetus (natural)


&


conveys immediate, short lived (humoral immunity) protection against Ag,


BUT, does NOT lead to development of cell mediated immunity

Passive Immunization (IVIG & Hyperimmune globulins)




(**Active immunizations (vaccinations) DO induce host immune response & lead to formation of cell-mediated immunity against future infection)

When are IVIGs indicated?


(passive immunity, made from pooled plasma, largely IgG, non-specific)

XLA, HyperIgM immunodeficiency


risk of exposure to Rabies or Hep B (not-confirmed)


Tetanus infection (or botulism, diptheria)

When are Hyperimmune globulins indicated?


(passive immunity, made from high titers of Ag specific Ig's)

Hep B infection


Rabies (1 shot + 5 doses of vaccine)


Tetanus infection


Hemolytic Disease of Newborn


VZV, CMV, RSV

when is Rho (D) immune globulin indicated?


(hyperimmune globulin)

to prevent hemolytic disease of the newborn


give mother @ 28 wks & immediately after delivery (prevent development of anti-Rh IgG)




If Rh (-) mother has hx of pregnancy w/ Rh (+) fetus (Rh + dad)-->


Will develop Anti-Rh IgG-->


Anti-Rh IgG crosses placenta & attacks second Rh (+) child--->


hemolytic disease of newborn

________ is indicated in metastatic breast cancer




*binds to tumor expressing human epidermal growth factor receptor (HER-2/neu)-->


blocks ligand binding & down-regulates receptor

*Trastuzumab (herceptin)

_______ is indicated in refractory cases of non-Hodgkin's lymphoma




*murine-human monoclonal IgG1 (human Fc) Ab-->


binds to CD20 on B cells-->


Ab-dependent cellular cytotoxicity-->


apoptosis of malignant lymphoma cells




(doesn't interfere w/ healthy B cell signalling, bc only CD19 & CD21 are involved)

*Rituximab (rituxan)




(can also be used in RA to wipe out B cells, per Krishna)

________ is indicated for prophylactic tx of RSV in exposed neonates




*monoclonal Ab-->


binds fusion (f) protein of RSV-->


neutralizes infectivity

*Palivizumab (synagis)

________ is indicated post cardiac intervention (angioplasty, stent, etc) to prevent complications d/t clot formation




*Fab monoclonal Ab-->


binds to GPIIb/IIIa receptors on activated platelets (glycoprotein 2b/3a antagonist)-->


prevents binding of adhesion molecules to receptor-->


inhibits platelet aggregation

*Abciximab (reopro)