Exam 2 week 2 Flashcards

1
Q

Describe components of fibrin and clot formation

**Intrinsic vs extrinsic coagulation system

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Identify and understand the tests appropriate for monitoring the different coagulation pathways

A
  • aPT
  • PT/PTT
  • INR
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Describe the fibrinolytic system, its components and regulators

  • what is lytic state
A

BAckground

  • Fibrinolytic system restricts clot expansion and degrades fibrin during wound healing
    1. Plasmin is a nonspecific protease that degrades fibrin, fibrinogen and other clotting factors
    1. Plasmin action on fibrinogen yields fibrinogen degradation products (FDP) which inhibit further conversion of fibrinogen to fibrin
    1. Plasminogen & Plasmin bind fibrin at a lysine rich binding site
    1. (tissue-plasminogen activator (t-PA) is released from endothelial cells at a site of injury, tPA binds to fibrin via lysine binding sites at the amino terminus and activates bound plasminogen 300x faster than it activates circulating plasminogen

Natural regulation of fibrinolysis

  • a. Damage releases tissue plasminogen activator (t-PA) from endothelium;
  • b. t-PA inactivated by circulating plasminogen activator inhibitor-1 (PAI-1)
  • c. α2-antiplasmin binds covalently to circulating plasmin at a lysine rich binding site causing inactivation;
  • occurs only with circulating plasmin
  • d. LYTIC STATE: circulating plasmin > capacity of α2-antiplasmin
  1. Ideal thrombolytic drug would degrade only desired thrombi and not old fibrin deposits
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Summarize MoA of drugs

Anticoagulants vs antiplatelts vs thrombolytic

A

ANTICOAGULANTS; inhibit fibrin formation (prevent blood clotting)
Drugs; Heparin, LMW-Heparin, Warfarin, Fondaparinux, Argatroban, Dabigatran

ANTIPLATELETS; inhibit platelet aggregation (you already had a heart attack)
Drugs; Aspirin, Dipyridamole, Clopidogrel, Cangelor, Abciximab& Eptifibatide

THROMBOLYTICS/FIBRINOLYTICS; dissolve formed fibrin clots
Drugs; Streptokinase, Alteplase, Anistreplase& Tenecteplase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Anticoagulant Drug - Heparin

  • MoA; what has reversible vs irreversible binding
  • Therapeutic uses; can you use in pregnancy? mode of administration? what happens if you give IM?
  • Clinical test; what do you monitor?
  • Adverse effects
  • Toxicity
  • Antidotes
A
  • *HEPARIN** (Unfractionated contains molecules with MW 15,000-30,000; mean 12 kDa or 40 monosaccharide units; fractionation due to action of endo–D-glucuronidase)
    1. Mechanism of action
  • a. Accelerates (1000 fold) inactivation by Antithrombin III (AT-III) of intrinsic and common pathways including: thrombin (II), IXa, Xa
  • b. Thrombin, IXa, Xa bind irreversibly to Arg-Ser site on AT-III
  • c. Heparin binding to AT-III is reversible; heparin binding site is specific pentasaccharide sequence that contains a 3-O-sulfated glucosamine residue which recognizes AT-III require minimum of 18 monosaccharide units to bind AT-III and thrombin
  1. Therapeutic Use
  • a. Anticoagulant activity In vivo (when injected into body)
  • b. Anticoagulant activity In vitro (when added to blood in test tube)
  • c. Venous thrombosis
  • d. Pulmonary embolism
  • e. Patency of IV cannulas
  • f. Anticoagulant in pregnancy, (discontinue 24 h prior to induction of labor)
  • g. Administer by injection, Large polar molecule, not absorbed orally
  • -Immediate effects if given IV
  • -Onset delayed onset (1-2 hr) with s.c.
  • -Intramuscular Contraindicated (induces painful hematoma)
  1. Other effect Lipid clearing effect heparin activates lipoprotein lipase (cleave TG from VLDL)
  2. Monitoring heparin
  • -Activated partial thromboplastin time (aPTT), monitors common and intrinsic pathway
  • antithrombin III has poor activity against coagulation factor VII (PT time, extrinsic pathway
  1. Adverse effects
  • a. Bleeding
    • 1) Thrombocytopenia Type I (HIT-I) – nonimmune occurs within 2 days of initiating therapy. Mediated by platelet –heparin interaction
    • 2) Type II (HIT-II) – immune mediated, more severe. Heparin therapy needed for 5-10 days. Antibodies form against the heparin-platelet factor 4 complex and bind on platelet surface causing aggregation.
  • b. Osteoporosis if given for more than 6 months

6. Treatment of excess hemorrhage

  • a. Administer plasma or blood containing coagulation factors
  • b. Protamine sulfate: Heparin-protamine complex cannot bind to AT-III so therby disable anticoagulant activity. DO NOT USE in protamine allergy (diabetics, NPH insulin); FISH ALLERGY - DO NOT USE

7. Contraindications

  • -Bleeding disorder
  • -Pre-existing bleeding sites
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Anticoagulant Drug - LOW MOLECULAR WEIGHT HEPARIN (ENOXAPARIN, DALTEPARIN and TINZAPARIN

  • MoA; specific for what factor?
  • Therapeutic uses (can you use in pregnancy?)
  • Contraindication and adverse effects
  • advatages over heparin
A

Description and Mechanism of Action

  • a) Low molecular weight heparins fractionated from heparin MW 1000-10,000 daltons; mean, 4500 daltons, or 15 monosaccharide units
  • b) Higher specificity for Enhanced Antithrombin III inactivation of Xa; >5400 kDa or 18 monosaccharide units required to bind simultaneously AT-III and thrombin
  1. Use
  • a) Prophylaxis and Acute Deep vein thrombosis, Pulmonary embolism, Orthopedic, Abdominal surgery
  • b) Unstable angina or non-Q-wave MI
  • c) Administered sub-cutaneous
  • d) Monitor anti-Xa activity
  • e) Anticoagulant in pregnant women (discontinue 24 h prior to induction of labor)
  1. Contraindications and adverse effects
  • a) Bleeding
  • b) Contraindicated in presence of bleeding disorder or active bleeding site
  1. Current perception of advantages for unfractionated heparin
    • Longer interval between doses (outpatient, once a day dosing)
    • Slight increase or no change in activated partial thromboplastin time
    • Does not require monitoring; could monitor anti-Xa activity
    • Better predictability of response to a given dose
    • Less thrombocytopenia, reduced binding to platelets
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Drug - Synthetic pentasaccharide binds to ATII to accelerate only Factor Xa inactivation

  • MoA
  • Use (pregnancy?)
  • Adverse effect
  • Contraindications
A

FONDAPARINUX

  1. Mechanism of Action
    * -Synthetic pentasaccharide binds to ATII to accelerate only Factor Xa inactivation
  2. Use and administration (mostly SC, IV for acute coronary syndrome)
  • -deep vein thrombosis acute and postoperative (hip or knee replacement)
  • -pulmonary embolism
  • -can be used in pregnancy (discontinue 24 h prior to induction of labor)
  1. Adverse effect BLEEDING
  2. Contraindications
    - active bleeding
    - severe renal impairment (<30 ml/min)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Identify oral anticoagulant

  • Only works in vivo
  • Inhibits hepatic synthesis of biologically active Vitamin K-dependent clotting factors, Protein C and S
    • What are the 4 coagulation factors this drug inhibits
  • Use?
  • how to monitor levels?
  • Adverse reaction and contraindication
    • genetics
  • Treatment of overdose
A

WARFARIN

Therapeutic Use

  • a) Warfarin, drug of choice for oral anticoagulant
  • b) Prophylaxis to prevent Venous thromboembolism and pulmonary embolism
  • c) Used in individuals with Prosthetic Heart Valves
  • d) Arterial thromboembolism prophylaxis in atrial fibrillation
  • e) Delayed therapeutic effect, initial effect occurs in 24 hr and maximal effect takes 5- 7 days
  • f) Warfarin is effective as an anticoagulant only when administered in vivo
  1. Monitoring Warfarin
  • a. INR laboratory standardized Quick one-stage prothrombin time
    • -citrate plasma incubated with tissue thromboplastin & calcium
    • -rate of fibrin generation is dependent on Factors I,II, V, VII and X
    • -Monitor extrinsic pathway with prothrombin time
  • b. Difference in thromboplastin sources and/or lots cause inter and intra laboratory variability
    • FYI INR International Normalized Ratio standardizes reagent thromboplastin to an International Reference Preparation (IRP) of thromboplastin
  • c. Goal for INR is 2.0-3.0 prophylactic for heart valves INR 2.5-3.0

4. Adverse Rxns and Contraindications

  • a. Hemorrhage
  • b. Adverse rxns more likely to occur if:
    • 1) changes occur in absorption or metabolism of warfarin or Vitamin K
    • 2) alterations occur in synthesis or catabolism of coagulation factors
    • 3) change in fibrin degradation
    • 4) change in platelet function or number
  • c. Genetic predisposition
    • 1) Genetic variants of CYP2C9 and VKORC1 genes use less warfarin
      • -CYP2C9 CYP2C9*2 (30%↓) CYP2C9*3 (90%↓) 10%Caucasian 2% African/Asian
      • -VKORC1 A allele synthesize less VKORC1 carried by 37% Caucasian 14%African
    • 2) FDA approved pharmacogenetic test in 2007
  • d. Contraindicated in patients with bleeding disorder or existing bleeding site
  • e. Contraindicated in pregnant women (Pregnancy Category X)
  • congenital abnormalities occur if fetus exposed first, second or third trimester
  • greater incidence of neonatal and fetal hemorrhage
  1. Treatment of Overdose
  • a. Administer whole blood or plasma
  • b. Administer Vitamin K1– (phytonadione)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Identify drugs that increase vs decrease warfarin response

A

Drugs or Conditions that Increase Warfarin Response

  • Vitamin K deficiency
  • Aspirin (decrease platelet aggregation)
  • Hepatic disease
  • Erythromycin
  • Thyroid hormones
  • Cephalosporins
  • Cimetidine
  • Ketoconazole

Drugs or Conditions that Decrease Warfarin Response

  • Cholestyramine
  • Rifampin
  • Oral Contraceptives (estrogen/progesterone combination only)
  • Excess ingestion of Vitamin K enriched foods
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Identify 2 direct thrombin inhibitors

  • MoA
  • IV vs oral administration
  • side effects and drug interactions
  • which has no P450 interaction
  • which has no antidote
A

DIRECT THROMBIN INHIBITORS (ARGATROBAN and DABIGATRAN ETEXILATE)

  1. Mechanism of Action
  • -Both are Direct thrombin inhibitors, blocks active site of thrombin
  • -Active on free and fibrin bound thrombin (Action independent of ATIII)
  1. ARGATROBAN
  • -IV administration
  • -Treatment and prophylaxis in thrombosis with heparin induced thrombocytopenia
  • -monitor with aPTT
  1. DABIGATRAN
  • -Oral administration
  • -Prodrug converted by esterases to Dabigatran (active)
  • -Converted to 4 Different Glucuronides (active)
  • -NOT A P450 Substrate (so fewer drug interactions)
  • -Substrate for P-glycoprotein transport, drug interactions rifampin induces P-glycoprotein decrease dabigatran blood levels
  • -Venous thrombosis prophylaxis
  • -Prevent stroke with atrial fibrillation
  • -Use with caution in diminished renal excretion (NO ANTIDOTE)
  1. Side Effects and Drug Interactions
  • -Bleeding, no available antidote
  • -P-glycoprotein substrate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Antiplatelet drugs

  • goal of therapy
  • how are they used?
  • examples
A

ANTIPLATELET DRUGS
A. Background

  • The goal of therapy is to diminish platelet function. These drugs are used primarily in Arterial Thrombotic Disease such as:
    • 1) transient ischemic attacks;
    • 2) unstable angina
    • 3) history of Myocardial infarction
  • Examples
    • Aspirin
    • Dipyridamole
    • Clopidogrel
    • cangelor
    • Abciximab
    • Eptifibatide
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Antiplatelet drug -

  • MoA
  • Adverse effects
    *
A

ASPIRIN

Mechanism of Action

  • a. Inhibit platelet aggregation
  • b. Irreversible inhibitor of cyclo-oxygenase (acetylates the enzyme)
  • c. Platelet prostaglandin formation (TXA2) inhibited by 160 mg/day aspirin

Adverse effects

  • a. GI bleeding, pain and peptic ulcer
  • b. Bleeding
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Identify antiplatelet drug

Mechanism of Action
-Inhibits phosphodiesterase (↑ platelet cAMP)

Therapeutic Use

  • Used with other agents, very little benefit if given alone
  • Given with Warfarin for prevention of thromboembolism in patients with prosthetic heart valves
A

DIPYRIDAMOLE

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Identify antiplatelet drug

P2Y12 receotor inhibitor (3)

  • MoA
  • Use
  • Major adverse effects
A

Mechanism of Action

  • a. ADP binding at the purinergic P2Y12 receptor. ADP release stimulates platelet purinergic receptors P2Y1 and P2Y12. P2Y1 activation causes TXA2 release, increase PI hydrolysis and a rise in intracellular Ca. P2Y12 activation inhibits cAMP formation. A full response to ADP requires activity of both P2Y1 and P2Y12 receptors. Inhibition of either P2Y1 or P2Y12 receptors is sufficient to impair ADP action on platelets.
  • b. Clopidogrel prodrug
  • c. Clopidrogrel thiol metabolite is irreversible inhibitor of P2Y12 receptor.
  • d. Cangrelor is a reversible inhibitor, IV administration
  1. Therapeutic Use
    a. Clopidogrel
  • -Reduce risk of thrombotic events in predisposed individuals
  • -Clopidogrel conversion to active mediated by CYP2C19
  • -Normal Platelet function returns when drug is stopped after new platelets are made (7-14 days)

b. Clopidogrel
* -Reduce Thrombotic events following MI, stroke, unstable angina or peripheral arterial disease
c. Cangrelor IV administration

  • -half-life 3-6 minutes, quickly dephosphorylated to inactive metabolite
  • -therapy until oral agent or during surgery
  1. Major adverse effects
  • a) Bleeding due to diminished platelet
  • b) Thrombocytopenia purpura
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

antiplatelet drug

Glycoprotein IIB/IIIA inhibitors

  • MoA
  • Use
  • adverse effects
  • contraindiactaion
A

GLYCOPROTEIN IIB/IIIA INHIBITORS ABCIXIMAB, EPTIFIBATIDE & TIROFIBAN

  • *Platelet Glycoprotein IIb/IIIa Receptor Antagonists
  • Bind to platelet IIb/IIIa receptor to prevent platelet aggregation And crosslinking with fibrinogen**
  • Abciximab Monoclonal antibody Fab fragment and Eptifibatide (peptide)
  • Tirofiban nonpeptide inhibitor
    2. USE
  • Acute coronary syndrome (unstable angina)
  • Prevent acute Adjunct with Angioplasty
  • Administered IV (bolus plus infusion)
    3. Adverse effects
  • Bleeding
  • Increased bruising
    4. Contraindications
  • History of hemorrhagic stroke
  • Active internal bleeding or GI/genitourinary bleeding in past 6 weeks
  • Thrombocytopenia
  • Major surgery or trauma in past 6 weeks

-Cannot use concurrent with warfarin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Fibrinolytic drug (Use, adverse effect, contraindications)

  1. Protein derived from beta-hemolytic streptococci; antigenic
  2. Mechanism of action
    a. Complexes stoichiometrically (1:1) with plasminogen
    b. Conformational change of plasminogen exposes catalytic site for conversion of a second
    plasminogen molecule to plasmin c. Acts on fibrin bound and circulating plasminogen; lytic state may occur
    Lytic state: circulating plasmin > capacity of α2-antiplasmin
A

STREPTOKINASE

Use

  • a. Reperfusion of occluded coronaries following acute MI
  • b. Pulmonary emoblism
  • c. Arterial thrombosis
  1. Adverse effects
  • a. High antigenic activity; fever occurs in 33% of patients
  • b. Bleeding
    • -lysis of fibrin at sites of vascular injury
    • -systemic lysis of fibrin, fibrinogen and clotting factors
    • -provide plasma to replenish clotting factors and fibrin
  1. Contraindications
  • a. Surgery or trauma in past 10 days
  • b. Pre-existing bleeding disorder or episode
  • c. Intracranial trauma
  • d. Diastolic blood pressure >110
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Identify fibrinolytic

  1. Streptokinase complexed with human lys-plasminogen. The active catalytic center is blocked by
    an acyl group.
  2. Designed to provide more specific binding to thrombi
  3. Acyl group removed by plasma enzymes
  4. Similar uses and side effects to streptokinase
A

ANTISTREPLASE

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Identify fibrinolytic

MoA

  • a. Activates fibrin bound plasminogen more selectively than circulating plasminogen
  • b. Lytic state is less marked than observed with streptokinase
  1. Therapeutic Use
  • -reperfusion of coronary arteries in acute MI
  • -pulmonary embolism
  • -thrombotic stroke
A

ALTEPLASE Reteplase; TISSUE-TYPE PLASMINOGEN ACTIVATOR (rt -PA)

not as effective

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Identify fibrinolytic

  1. Genetically engineered derivative of alteplase
  2. (compared to alteplase), this drug has longer half-life, greater fibrin specificity than alteplase and slower inactivation by PAI-1
  3. Approved for MI
A

TENECTEPLASE

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Identify fibrolytic INHIBITOR

  1. Inhibitor of Plasminogen activation
  2. Lysine analog, compete for lysine binding site on plasminogen and plasmin
A

**AMINOCAPROIC ACID**

  • Inhibit fibrinolysis
  • inhibit interaction of palsmin and fibrin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

1) A 25 year old pregnant female develops deep vein thrombosis.

  • What agent can cause teratogenicity?
  • What agent would you prescribe?

2) A 63 year old male underwent mitral valve replacement 1 year ago and has been successfully regulated on warfarin for the past 8 months. He develops a sore throat and upper respiratory infection. The patient is placed on a 10 course of erythromycin for his infection.

  • effect of erythromycin on INR
  • Should additional tests be done to monitor the effectiveness of the warfarin?

.3. what induce fever

A

1.

  • Warfarin - cause teratogenicity
  • Heparin should be prescribed in DVT

2.

  • Increase INR ( erythromycin increase warfarin response)
  • Clearance of eythromycin will increase?

3. Streptokinase - cause fever

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q
  1. Describe clinical entities associated with Epstein Barr Virus (EBV) infection
A
  • EBV initiate infections by infecting cells of the respiratory system and regional lymph nodes associated with therespiratory system.
  • EB virus causes a primary infection in the RS and then spread to secondary sites of infection, which leads to the obvious signs and disease symptoms. These viruses infect and are transported by lymphocytes. EBV, for example, infects cells of the B cell lineage. HHV 6,7 and 8 will also be briefly discussed
  • Herpesviridae: Subfamily Gammaherpesvirina
    • α = HSV, VZ; β =CMV; Gamma = EB
    • Double-stranded DNA wound around protein core
    • Icosahedral capsid composed of capsomers
    • Envelope with glycoprotein spikes on its surface
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Properties of EB virus

A
  1. Morphologically same as other herpes viruses, Smaller size DNA genome
  2. Virus replication
    1. Found in lymphoblastoid cell lines,
    • In vivo infects primarily B cells and certain types of epithelial cells in nasopharynx that are CD21 (complement factor C3d) positive cells
    • CD 21 acts as receptor for EBV on B cells
    • Also binds to MHC II
  3. Primary viral antigens found in EBV infected cells; EBNA - DNA binding proteins, VCA - capsid,MA - membrane, EA- early Ags,(EA- R and EA- D)
    • Ags not related to other herpesviruses
    • In total EBV genome can encode over 70 viral proteins
    • Non-structural virus proteins: EB Nuclear Antigens-EBNA 1,2,3A, 3B and 3C; Latent Proteins (LP), Latent Membrane Proteins( LMPs)1 and 2
    • Note: In addition to EBNA (see above) LPs are DNA binding proteins
    • Small viral RNAs (EBER) 1 and 2; detected by in situ hybridization techniques, very senstive approach
  4. Host Range – limited, humans and some non-human primate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Know the three types of EBV infections as it relates to different cells

**which is most common worlwide

A

EB (Epstein-Barr) Virus, 2 subtypes, Type 1 most common world wide

EBV interactions with cells

  1. Permissive cells such B cells and types of epithelia cells support EBV replication, Permissive cells (non-memory resting B cells in tonsils).
    • Immediate early, early and late pahses (gp350/220 are glycoproteins) of cell cycle
  2. Latent infection of memory B cells when competent T cells are present
  3. Immortalization of B cells, enhance proliferation, especially B cells in tissue culture
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Describe in detail the interactions of EBV with cells

A

1) Replication of EBV in permissive cells (non-memory, resting B cells in tonsils):

EBV infection can cause the synthesize up to 70 viral proteins. Immediate early, early and late phases of the cell cycle. (See previous herpesvirus lecture)

Late proteins gp350/220 are glycoproteins that are located in the viral envelop enable attachment of the to specific receptors on the surface of cells (CD21). B Cell entry via fusion, gp42 interacts with MHC class II molecules to promote fusion.

EBV binding to CD 21 initiates the activation and replication of B cells. Expression of certain viral proteins (see full listing above) help maintain the infectious process. Virus replication occurs and progeny virus particles are produced to can be found in high amounts in the infected patient’s saliva. Lytic infection initiates with the immediate early protein, ZEBRA, acting as a transcription factor that furthers the transcription of immediate early genes that then stimulates early gene transcription. Following viral genome synthesis in the nucleus of the infected cell late capsid and envelop proteins are synthesized. Progeny virus is then formed. (The more detailed replication of herpesviruses was covered previously)

2) Latent infections require a different type of EBV and B cell interactions. The lytic infection (in permissive cells) can give rise to latent infections depending on the differentiation state of B cells. Memory B cells are conducive to the formation of latent infections.

In latent infections viral genomes circularize and resemble a plasmid. This form of viral DNA replicates in tandem with cell division via cell DNA polymerase. EBNA-1 is important in maintaining latency

Therefore, only specific genes are expressed from the circular genome.

Viral antigens include EB Nuclear Antigen-1, and latent

proteins (LPs), both EBNA and LPs are DNA binding proteins).

3) B cell immortalization: Latent Membrane proteins 3(LMPs) stimulate the replication of B cells and promote their immortalization.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

Clinical features of EB virus

  • cell type
  • primary symptoms
A

Disease - Infection at early age –no disease, Delayed exposure to virus until adolescence or adulthood causes more severe disease, ie IM

Virus found in saliva, eg. spread by kissing; replication in regional lymph nodes and epithelial cells in the respiratory system

Infected B lymphocytes transmit virus to other lymph nodes and spleen

  • Atypical lymphocytes (Downey Cells) in blood correspond to large T lymphocyte that form in response to EBV infected B cells
  • Some disease symptoms associated with IM result from the immunological attack by T lymphocytesagainst EBV infected B lymphocytes, cytokine production is present
  • Synthesis of a IL-10 analog by EBV during the infection thwarts the TH1 CD4 T cell immunological response

three (3) primary symptoms

  1. lymphadenopathy (enlarged lymph nodes/glands),
  2. splenomegaly (enlarged spleen),and
  3. exudative pharyngitis
  • plus headache, chills, high fever, malaise
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Understand the significance of heterophile antibodies and the use of the “monospot test

Distinguish between EBV and CMV mononucleosis

A

Immune Response: Several responses

  1. Heterophile antibodies - short lived ab - not virus specific; infected B cells differentiate and produce polyclonal antibodies some of which are heterophile and autoantibodies
    1. Heterophile Antibodies agglutinate RBCs of non human origin, horse or sheep RBCs
    2. 60-80% positive sera for heterophile Ab​
  2. Specific antibodies to EB virus proteins
  3. T lymphocyte response to EBV infected B cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Lab diagnosis of EB virus

A

1) Atypical lymphocytes (T lymphocytes= Downey cells), large, basophilic, foamy (vacuolated) cytoplasm
2) Heterophile antibodies (IgM) - “monospot test” - patients serum is treated with GP-RBC to remove nonspecific factors ——> Horse (sheep) RBC——-> Agglutination = + test;

  • some sera from patients are heterophile - negative, must differentiate from heterophile negative CMV mononucleosis,
  • use antibody detection to VCA (IgM),EA, MA, EBNA

3) Best: virus neutralization, immunofluoresence, CF for viral antibodies
4) Time course VCA (IgM) —> VCA (IgG) —-> EA + EBNA Abs,

antibodies peaks few weeks after infection - last for years

antibodies for EBNA in combination with VCA (IgG) indicate a past infection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

EB virus

  • Epidemiology and control
  • Treatment
  • vaccine
A
  1. Epidemiology and control
    1. 90% of adults have antibody
    2. Spread orally - large dose required in saliva “Kissing Disease”, share toothbrush etc
    3. Common in institutions and colleges, asymptomatic primary disease of young children, more severe primary infection for adolescence and adults(college students)
  2. Treatment – symptomatic, limited approval antiviral drugs; Valacyclovir for severe cases + steroids
  3. Vaccine: Experimental subunit vaccine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What 3 conditions can EB virus cause

A

EBV May act as a cofactor rather than the sole cause of malignancies

  1. Infectious mononucleosis
  2. Associtaed with Burkitt’s lymphoma (Africa);
    • ​​Geographical area (africa also where there is malaria)
    • activation of MYC protooncogenes may cause lymphoma
    • Development of lymphoblastoid cell lines that grow in tissue culture - EB viral DNA exist as plasmid in cells from EB patient and imparts increased cell growth
  3. Associated with nasopharyngeal carcinoma (china)

Predilection for B lymphocytes

  • ​​Possibly Burkitts lymphoma and other types of lymphomas,Hodgkins Infectious mononucleosis (heterophile antibody +)nasopharyngeal carcinoma, gastric cancers,oral hairy leukoplakia in AIDS patients, autoimmune disease links
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What is the role of EBV in initiating various cancers?

nasopharyngeal carcinoma vs chronic EBV mononucleosis

A

EBV May act as a cofactor rather than the sole cause of malignancies

1) Nasopharyngeal carcinoma

  1. Nonlymphoid cells, EB genome present in epithelial cells
  2. Males of Chinese extraction Lymphoproliferative disease
  • If T cell deficiency exists in patient, EBV induces B cell leukemia-like or lymphoma may develop. Shows the importance of T cells keeping EBV infected B cell proliferation in check.
    2) Chronic EBV mono - possible association

Fatigue, fever, headaches, mental lapses, numbness and depression, symptoms last months to years

  1. Chronic mononucleosis or chronic viral fatigue syndrome most common, different symptoms per patient, fatigue is common, woman 2X affected compared to men, 1 year average duration
  2. Other possible causative agents include Human herpesvirus 6 or a retrovirus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q
  1. what viruses cause roseola vs karposi sarcoma
A

Roseola is caused by HHV 6 and 7 while Kaposi’s Sarcoma involves HHV 8

1) Human herpes virus 6(HHV6B); not antigenically related to other herpesviruses:

  • Causes Exanthem subitum (roseola) and infect large numbers of children.
  • Infects B and T cell (lymphotropic) and can infect epithelial and endothelia cells as well as neurons.
  • Cause latent infections in T cells and monocytes and can reactivate in transplant patients and immunosuppressed patients.
  • Found at high levels in saliva as a result of replication in the salivary glands

2) HHV 6 and 7 isolates also causes roseola which is characterized by a high fever (103-105 F)and a disseminated rash (trunk and face first and then spreads) follows as the fever resides, rash last 24 to 48 hrs

3) HHV 8 has been associated with Kaposi sarcoma and a rare type of B cell lymphoma. B cells are primarily infected by HHV8 but other cell types can be infected. HHV8 encodes proteins with growth factor characteristics and ability to slow apoptosis of infected and uninfected cells, eg. Interleukin-6 homolog

  • Kaposi sarcoma found at elevated levels in AIDS patients.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

Understand the structural features of human immunodeficiency virus and how its genome RNA differs in the number and types of genes compared to retroviruses discussed previously, ie. Chronic leukemia viruses and acute leukemia/sarcoma viruses.

A

HIV - Virus Structure – basically similar to other retroviruses, especially other Lentiviruses.

  • It is considered to be a complex retrovirus and has genome that possesses genes in addition to gag,pol and env genes, which found in the genomes of the simpler retroviruses. The additional genes are TAT, REV, VIF, NEF and VPU, and VPR, all of which encode accessory proteins that seem to play a regulatory role during HIV infection.

Review: GAG proteins are the core and capsid structural proteins

  • POL proteins are part of a enzymatic complex which includes reverse transcriptase, DNA polymerase, RNAse H, protease and integrase
  • ENV proteins constitute the spikes on the envelop of the virus, gp 160 = gp120 + gp 41
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

Identify accessory genes that encode accessory proteins in HIV

  • transcription transactivator
  • Regulates transport of viral mRNAs from nucleus to cytoplasm
  • key component in enabling a limited HIV infection to progress to full blown HIV replication.
  • blocks cellular protein (APOBEC-3G)
  • enhance release of virus from infeceted cell
  • arrect infected cells in G2 phase - enhance replication?
A

Accessory Genes encode Accessory Proteins

  • TAT = Transcription Transactivator for both viral and cellular control regions of genes, ( Remember from our previous discussion of retroviruses that the LTRs located at each end of the provirus has promoters and enhancers that bind regulatory proteins such as TAT)
  • REV = Regulates transport of viral mRNAs out of the nucleus and into the cytoplasm, also regulates splicing of viral RNAs that leads to the formation of viral mRNAs
  • NEF = Diminishes the expression of CD4 and MHC 1 which changes T cell signaling and is required to achieve a high virus load. NEF is a key component in enabling a limited HIV infection to progress to full blown HIV replication.
  • VIF promotes virus assembly and blocks the action of a cellular protein (APOBEC-3G) that normally acts as an antiviral protein
  • VPU enhances the release of virus from infected cells and inhibits the cell surface expression of CD4 on infected cells
  • VPR arrests infected cells in G2 phase of cell cycle, which is optimal phase for HIV replication and enhances the transport of proviral DNA (provirus) from the cytoplasm into the nucleus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

HIV structure - components and functions

  • fusion protein?
  • what are the major antigens of the protein?
A
  1. Genomic RNA exists in two identical copies in direct association with two proteins; p 9, p 7.
  2. Genome RNA is housed within a cylindrical core composed of p24 - reverse transcriptase is located inside core.
  3. An outer envelope surrounds core. Underlying the envelope is a layer of p 17 protein.
  4. The envelope possesses glycoprotein spikes extending outward from its surface. Each spike is composed of 3 or 4 identical polypeptides, each polypeptide is designated gp 160. (glycoprotein 160,000 mw)
  5. Each gp 160 is usually proteolytically cleaved into two subunits; distal gp 120 (gp 120,000 mw) and proximal gp41.
  6. gp 120 and gp 41 are held together by noncovalent bonds.
  7. gp 160 and gp 120-gp41 are the major antigens of the virus, which change antigenically within a given population or even in a particular individual infected with the virus (see M tropic àT tropic HIV above)
  8. gp 120 important for attachment of virus to receptor on cell surface. gp41 possess amino acid sequence necessary for viral envelope fusion with cell membranes or fusion between an infected cell and uninfected cells. (Remember the HA protein of influenza virus and its role in cell membrane- envelop fusion)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

Know the various stages of HIV replication and the role that accessory genes play in the replication cycle.

A

HIV provirus: Diagram below is not drawn to scale and represents the proviral form derived from the viral genome.

LTR = Long terminal repeat located at each end of the provirus

Note: TAT and REV mRNA are synthesized as a result of multiple splicing events. Segments 1 + 2 + 3 = mRNA; 1,2, and 3 represent different sequences for TAT and REV

Recombination (copy choice) between two copies of genomic

RNA is a source of genetic variation in addition to variations caused by point mutations. Reverse transcriptase can

jump from on 35S RNA to another to yield the final form of the provirus. May be a mechanism to repair breaks in the template genomic RNAs when it is copied into a provirus.

Recombinational changes assume that the two genomic copies within a virus vary in ribonucleotide sequence and are not the same.

37
Q

Understand the process by which HIV binds to specific receptors plus co-receptors at the attachment stage of replication.

A
38
Q

Replication cycle of HIV

A
  1. Cells that are infected by HIV vary depending on the specificity between gp 120 and the receptor/co receptor (chemokine receptors CCR5 and CXCR4)
  2. HIV attaches to cells which possess CD4 molecules on their surfaces, eg. T lymphocytes, macrophages and certain brain cells (microglial cells).
  3. In addition to the CD4 receptor, a second molecule (coreceptor) is involved in the binding and fusion of the virus with the cell. The copreceptor may be one of two types; CCR5coreceptor is present on the surface of certain peripheral T cells/macrophage and HIV strains that bind to these cells is designatedM Tropic HIV.
  4. Other HIV strains bind to cells using CD4 and the CXCR4 coreceptor, which is present on the surface of the majority of T cells (T Tropic HIV)

(some individuals lack CCR5 and are resistant to HIV infection)

As indicated above, the M Tropic (R5) HIV usually is the type that initiates HIV infections. As a result of mutations in the gp120 viral glycoprotein gene, the virus changes to T Tropic(X4) as the HIV infection progresses into the immunosuppressive state.

39
Q

Replication cycle of HIV (cont’d)

What happens following uncoating

  • How can a cancer (kaposi sarcoma) form during HIV infection without an oncogene? what accessory protein is responsible for this?
A
  1. Following uncoating, genomic RNA is reverse transcribed into a provirus which moves to the nucleus where it is integrated into cell DNA.
  2. Once the provirus is integrated into cell DNA it becomes transcriptionally active. A variety of mRNAs are formed that encode the proteins of the virus and the accessory proteins
  3. Viral RNA are spliced once or twice depending on the RNA, mRNAs for REV and TAT are spliced twice (See diagram)
  4. The immature virus buds and to acquire its envelop and the GAG and POL polyproteins located within the immature virus particle are cleaved and a mature virus particle results
  5. Kaposi’s sarcoma and other malignancies are induced during HIV infection. How can a tumor form without an oncogene? TAT seems to play a role as a transactivator and is released from HIV infected cells to induce the sarcoma ( TAT gene was introduced into transgenic mice and the expression of this gene induced a malignancy similar to Kaposi’s sarcoma)

HHV-8 has been implicated at the primary causative agent for Kaposi’s sarcoma (see end of previous lecture on CMV and EBV)

40
Q

Understand the aspects of HIV replication that are targets of drug treatment;

A

attachment, provirus formation, proteolytic cleavage of viral polyproteins, and budding are some of these targets.

41
Q

Understand presumptive and confirmatory diagnostic tests for AIDS. Understand the time course for the development of AIDS and the progression from the initial infection to the disease AIDS

A
  1. HIV infection diagnostics: Detection of HIV specific antigens and antibodie__s specific for HIV proteins; screened in human serum.
  2. CDC/APHL algorithm which includes antigen-antibody combination assay as part of a screening process to detect HIV infections; 1 out of 1,000 to 3000 yield false positive results

A few examples: Abbott Architect HIV Ag/Ab Combo, Bio-Rad GS HIV Combo, Siemens Advia Centaur HIV AG/Ab Combo, etc. Based on immunoassays

  1. If combo test is positive an antibody test can differentiate between HIV-1 and HIV-2, Again an immunoassay, Bio-RAD Geenius HIV 1/2 Assay
  2. If discrepant results are found between initial antigen- antibody screen and the HIV 1/2 Assay then a nucleic acid test, Hologic Aptima HIV-1RNA Qualitative Assay, is employed

Positive RNA test indicates acute HIV infection

  1. Nucleic acid testing is based on Reverse Transcriptase (RT) PCR is used to determine the HIV load in blood. This is reported in RNA copies per ml and is used as an indicator of the extent of HIV replication.
  2. HIV genomes can be sequenced to detect specific mutations in viral genes that encode key enzymes, such as Reverse Transcriptase. The mutations have been shown to impart drug resistance to HIV, such as resistance to reverse trans- scriptase inhibitors.
42
Q
  • What is confirmatory test for HIV?
  • Presumptive test?
A
  • Confirmatory test (picture below); Test for antibodies for presence of HIV viral proteins
    • Detection of HIV specific antigens and antibodie__s specific for HIV proteins; screened in human serum.
  • Presumptive test
    • OraQuick Advance HIV (1/2); fingerstick whole blood, oral fluid; moderate complexity with plasma. Result in 20 minutes. Screens for HIV 1 and HIV 2. Store at room temp.
    • Collect oral fluid specimens by swabbing gums with test device
43
Q
  • Identify major targets of HIV infection
  • Identify cell populations infected with HIV
A
  • •Immune System & CNS
  • •Immune System
    • •Profound immunodeficiency
    • •Affects cell-mediated immunity
  • Cell populations infected
    • CD4+ T cells; few affect in blood but large numbers affected in lymph node. CD4+ T cells are depleted
    • Monocytes/macrophages
    • Dendritic cells
44
Q

In HiV, CD4 cells depleted mostly in lymph nodes and also in blood. How do uninfected cells get killed? (5)

A
  • •Activation-induced apoptosis
  • •Non-cytopathic HIV infection
    • •Activates inflammasome pathway
    • •Cell death due to pyroptosis
    • •Cytokines and other cell contents are released and recruit new cells for possible infection
  • •Destruction of architecture of lymphoid organs
  • •Loss of immature precursors
    • •Thymic progenitor cells are infected
    • •Infection of accessory cells
  • •Syncytia formation
    • Fusion of uninfected & infected
45
Q

In HIV, there is a quantitative defect of CD4+ cells (T cell depletion mostly in lymph node and then in the bloof)

**Identify qualitative defects of CD4+ T cell (can you see in aymptomatic?)

A

Qualitative defect of CD4+ T cells (seen in asyptomatic patients)

  • •Reduction in antigen-induced T cell proliferation
  • •Decrease in TH1 type responses relative to TH2 type
    • •Increased susceptibility to infection by viruses
    • •Marked decrease in cell-mediated immunity
  • •Loss of memory T cells – poor recall function
  • •Decreased helper function for B cell antibody production
  • •All defects can be seen even in asymptomatic HIV-infected individuals
46
Q
  • Latent infection; important feature of HIV infection
  • How are monocytes/macropahes affected in HIV infection
A

Latent infection; Important feature of HIV infection

  • •Latent virus is found in CD4+ T cells and macrophages in lymph nodes
  • •Protected from antiviral therapy
  • •Provides a persistent reservoir of virus

Monocytes/Macrophages

  • •10-50% of macrophages in tissue are infected, many more than are monocytes in blood
  • •Infected macrophages bud few viruses from the cell surface, store large numbers of viral particles, allow replication but are quite resistant to cytopathic effects
  • •Relatively protected from host defenses
  • Serve as reservoirs of infection and virus factories later in infection when CD4+ T cell counts are low
  • •Play an important role in CNS infections – HIV is carried into brain by infected monocytes
  • •Abnormalities in function
    • – Decreased phagocytosis & chemotaxia
    • – Decreased secretion of IL-1
    • – Decreased ability to present antigens to T cells
  • •Certain HIV strains are tropic for macrophages
47
Q

How are dendritic cells (mucosal and follicular) affected in HIV infections

A
  • Mucosal dendritic cells
    • •Initially infected by virus
    • •Transport virus to lymph nodes where virus is transferred to CD4+ T cells
  • •Follicular dendritic cells (in germinal centers)
    • •Reservoirs of infection & trap virions
48
Q

How are B lymphocytes affected in HIV infection

A
  • •Polyclonal activation with germinal center B cell hyperplasia
  • •Bone marrow plasmacytosis
  • •Hypergammaglobulinemias
  • •Increased circulating immune complexes
  • •Inability to mount Ab response to new Ag
  • •Impaired humoral immunity renders patients vulnerable to disseminated infections caused by encapsulated bacteria
49
Q

Identify pathogenesis of CNS involvement with HIV

A

HIV with CNS involvement

  1. Nervous system is a major target of HIV infection
  2. Macrophages and microglia are predominant cell types infected
  3. HIV is carried into brain by infected monocytes but neurons are not infected
  4. Actual mechanisms of damage are not clear but most likely caused indirectly by viral products and other factors, such as cytokines (IL-1, TNF, IL-6)
50
Q

Summarized T cell and non T cell involvement of HIV

A
  • T cell
    • CD4+ T cell quantitative defect/depletion
    • CD4 T cell qualitative defect
  • Non T cell
    • Monocytes/macrophages
    • Dendritic cell (mucosal and follicular)
    • B lymphocytes
51
Q

Summarize natural history of HIV infection

A
  • Primary infection
    • Virus dissemination
    • Acute retroviral syndrome
  • Chronic infection, phase of clinical latency
    • T cell going down and virus going up \
    • oppurtunistic infection (candidiasis)
  • AIDS
52
Q

Natural history of HIV infection - Primary infection

A

Primary infection, virus dissemination and acute retroviral syndrome:

  1. Virus typically enters through mucosal epithelia
  2. Early infection is characterized by infection of memory CD4+ T cells in mucosal lymphoid tissue with death of many infected cells
  3. Next step is dissemination to lymph nodes by infected dendritic cells
  4. In lymph nodes, virus may pass to CD4+ T cells by direct cell-cell contact
  5. Replication in lymph nodes leads to viremia with high numbers of HIV particles in the blood
  6. Infected individual mounts antiviral humoral and cell-mediated immune responses evidenced by seroconversion, usually 3 to 7 weeeks after exposure, which correspond to drop in serum viral titers
  7. Acute retroviral syndrome
  • a. Develops in ~40-90% of patients
  • b. Typically 3-6 weeks after infection; resolves in 2-4 weeks
  • c. Nonspecific symptoms of sore throat, myalgias, fever/sweats, weight loss, fatigue
  • d. Also, rash, cervical adenopathy, diarrhea, vomiting
53
Q

Identify condition

**When does it occur

a. Develops in ~40-90% of HIV patients
b. Typically 3-6 weeks after infection; resolves in 2-4 weeks
c. Nonspecific symptoms of sore throat, myalgias, fever/sweats, weight loss, fatigue
d. Also, rash, cervical adenopathy, diarrhea, vomiting

A

Acute retroviral syndrome

**Primary HIV infection

54
Q

Describe chronic HIV infectin; phase of clinical latency

A
  1. Lymph nodes & spleen are sites of continuous HIV replication & cell destruction
  2. Few or no clinical symptoms
  3. Smoldering destruction of CD4+ T cells in lymphoid tissues
  4. Number of CD4+ T cells in peripheral blood slowly declines
  5. Host defenses start to decrease; viral burden and number of infected CD4+ T cells increases
  6. Patients may be asymptomatic or demonstrate minor opportunistic infections such as candidiasis, herpes zoster, mycobacterial tuberculosis.
  7. Typically lasts 7-10 years, but there are exceptions known as rapid progressors and long-term nonprogressors
55
Q

What is criteria for diagnosis AIDS

presenation?

oppurtunistic infections?

A

The criteria for diagnosis of AIDS have been defined by the CDC

  • a. Certain opportunistic infections and cancers (AIDS-defining opportunistic infections)
  • b. CD4+ T cell count < 200 cells/microliter
  1. Typically present with fatigue, weight loss, and diarrhea
  2. Opportunistic infections account for the majority of deaths in untreated patients with AIDS
  3. Many represent reactivation of a latent infection which was kept in check by a normal immune system
  4. Many of the opportunistic infections have decreased since the introduction of combination antiviral therapy
  5. The most common opportunistic infections include:
  • Pneumocystic jiroveci pneumonia
  • Candidiasis
  • Cytomegalovirus
  • Mycobacteria
  • Cryptococcus
  • JC virus
  • HSV
  • Persistent diarrhea
  • Cancers; Karposi, High grade B cell lymphoma, High risk cervical dysplasia
56
Q

Oppurtunistic infections in AIDS

A
  • a. Pneumocystis jiroveci pneumonia – frequent opportunistic infection; make dx by finding organism in cytologic specimen or biopsy
  • b. Candidiasis – most common fungal infection; commonly infects oral cavity, vagina, esophagus. Invasive infection is uncommon
  • c. Cytomegalovirus – may cause disseminated disease, more commonly affects eye (chorioretinitis, may cause blindness) and gastrointestinal tract (esophagitis, colitis)
  • d. Mycobacteria: Mycobacterium avium-intracellulare disease, disseminated Mycobacterium tuberculosis infection – frequent in AIDS; appearance of M. tuberculosis is similar to non-AIDS, more extensive disease; MAC more unique to AIDS, involves mainly lymph nodes, spleen, liver, marrow, less likely to produce visible granulomas
  • e. Cryptococcus – meningitis is major clinical manifestation
  • f. Toxoplasmosis gondii – encephalitis and cerebral abscess
  • g. JC virus – human papovavirus: progressive multifocal leukoencephalopathy (PML)
57
Q

Oppurtunistic infection in AIDS (cont’d)

A
  • h. Herpes simplex virus – mucocutaneous ulcerations involving the mouth, esophagus,external genitalia and perianal region
  • j. Persistent diarrhea – caused by any number of protozoal and bacterial infections: Cryptosporidium, Isospora belli, microsporidia, Salmonella, Shigella, M. avium-intracellulare
  • k. Cancers – AIDS patients have a high incidence of certain cancers, a common feature is they are all caused by oncogenic DNA viruses
    • 1) Kaposi’s sarcoma is the most common cancer in AIDS patients.
    • a) Normally uncommon vascular neoplasm in the US
    • b) At onset of AIDS epidemic, up to 30% of patients had this
    • c) With use of new AIDS therapy, incidence < 1%
    • d) Can present as purple skin lesions or as disseminated disease in lymph nodes, skin, gastrointestinal tract, or lungs
    • e) Thought to be due to herpes virus 8 (HHV 8) which is sexually transmitted
    • f) Immunosuppression is co-factor
    • 2) High-grade B cell lymphomas
    • a) See development of high-grade B cell lymphomas late in disease course; occurs in peripheral lymph nodes, brain or body cavites
    • b) With lower CD 4 counts, see increased risk
    • c) Associated with Epstein-Barr virus (EBV)
    • 3) High risk of cervical dysplasia in women and anal cancer in men – associated with human papillomavirus (HPV) infection
58
Q

AIDS in CNS

  • how many have clinical invlvemnt?
A

AIDS in the CNS:

  • 40-60% of patients have clinical involvement in CNS,
  • 90% at autopsy.
  • See variety of diseases: aseptic meningitis, peripheral neuropathy, AIDS-dementia complex
59
Q

Effect of antiretroviral drug therapy

A

Effect of antiretroviral drug therapy – Highly active antiretroviral therapy (HAART) or combination antiretroviral therapy

    1. In motivated, compliant patients, HIV replication is reduced below the level of detection and remains there indefinitely
    1. Annual death rate in US reduced from peak of 16-18/100,000 people in 1995-96 to < 4/100,000
    1. Long-term side-effects of antiretroviral therapy
      * a. Lipoatrophy (loss of facial fat
      * b. Excess fat deposition centrally
      * c. Premature cardiovascular, kidney and liver disease (major cause of morbidity in AIDS patients
60
Q

Morphology of HIV/AIDs

  • are there any anatomical changes
  • lesion mean what?
  • lymph nodes?
A
    1. In general, no specific anatomic changes are present in HIV/AIDS
    1. Lesions are characteristic of opportunistic infections or other processes
    1. Lymph nodes:
      * a. Marked follicular hyperplasia seen early in disease and due to polyclonal B cell activation
      * b. Later in disease see follicular involution – follicles are depleted of cells and the organized network of follicular dendritic cells is disrupted
      * c. Leads to release of trapped virus; overall small “burnt-out” lymph nodes; spleen and thymus are also affected in this way.
61
Q

Identify lab tests in HIV infection (5)

A
  1. Rapid HIV test
  2. Antibody studies
  3. CD4+ T cell counts
  4. Level of HIV-1 RNA is important (viral load test)
  5. CDC algorithm for HIV diagnostic testing recommendations (June 2014)
62
Q

Identify lab test for HIV infection

  1. Simultaneously detects HIV-1 p24 antigen and antibodies to both HIV-1 and HIV-2 in human serum, plasma, and venous or fingerstick whole blood specimens
  2. Detection of HIV-1 antigen permits earlier detection of HIV-1 infection than is possible by testing for HIV-1 antibodies alone
  3. Approved by FDA in August 2013
A

Rapid HIV test

  • Look for antigen - antibodies
  • If test come back positive, then identify HIV 1 vs HIV 2
63
Q

lab test in HIV (confirmatory test)

A

Antibody studies:

  1. Antibodies to HIV appear as early as 6 days to 1 month after acute infection
  2. The majority of individuals appear to develop antibodies within 3 months, with rare seroconversion occurring up to 6 months.
64
Q

Lab test of HIV

  • Short term indicator of disease progression

**What is the definition of AIDS with this test

A

CD 4+ T cell counts:

  1. Absolute levels of CD4+ cells are of prognostic value, as disease severity usually increases as CD4 counts decrease.
  2. Also serve as reliable short-term indicator of disease progression.
  3. CDC classifies patient into categories based upon CD4 counts, along with other clinical criteria. Definition of AIDS: CD4+ count of < 200 cells/microliter
65
Q

lab test in HIV

  • Predictor of clinical outcome.
  • Use along with CD4+ T cell count to monitor effectiveness of antiviral therapy
A

Level of HIV-1 RNA is important (viral load test).

  • Viral load increase with HIV (no management)
  • Viral load decrease as HIV is controlled.
66
Q

Identify CDC algorithm for HIV diagnostic testing recommendation

A

CDC algorithm for HIV diagnostic testing recommendations (June 2014)

    1. Diagnosis starts with a fourth-generation test that detects HIV in the blood earlier than antibody tests can; it identifies the viral protein HIV-1 p24 antigen, which appears in the blood before antibodies do
    1. If this test is positive, an immunoassay that differentiates HIV-1 from HIV-2 antibodies should be performed; results from such assays can be obtained faster than they can from the Western blot test
    1. In patients with positive results on the initial antigen test but with negative or indeterminate results on the antibody differentiation assay, HIV-1 nucleic acid testing should be performed to determine whether infection is present
67
Q

HIV - ALLMAN (Review)

Identify medications (what class do all these drugs belong to?)

  • Screen for HLA- B*5701
  • Bone marrow suppression
  • Pigmentation; Nail vs Skin
  • Pancreatitis
  • Peripheral neuropathy (2)
A

HIV Medications review

  • Screen for HLA-B*5701
    • Abacavir (ABC) - NRTI
  • Bone Marrow Suppression
    • Zidovudine (AZT, ZDV) - NRTI
  • Pigmentation
    • Nail = Zidovudine
    • Skin = Emtricitabine -NRTI
  • Pancreatitis
    • Didanosine - NRTI
  • Peripheral Neuropathy
    • Didanosine, Stavudine - NRTI

***All these drugs are NRTI; NRTIs are faulty versions of building blocks that HIV needs to make more copies of itself. When HIV uses an NRTI instead of a normal building block, reproduction of the virus is stalled.

68
Q

HIV meds review

  • Rash (4)
  • Lactic acidosis with hepatic steatosis (2)
  • Do Not Start if CD4 >250 (Woman)/>400 (Men)
  • Lipid abnormalities
A
  • Rash
    • Delvaridine, Efavirenz, Etravirine, Nevirapine (NNRTIs) - RASH RASH RASH
      • Steven Johnson Syndrome (although rare) highest incidence with Nevirapine
      • RASHES frequently occur within the first 2-4 weeks of therapy
        Lactic acidosis with hepatic steatosis
    • Tenofovir
    • All NRTIs; (Abacavir, Didanosine, Emtricitabine, Lamivudine, Stavudine, Zidovudine)
  • Do Not Start if CD4 >250 (Woman)/>400 (Men)
    • Nevirapine
  • Lipid Abnormalities
    • Protease Inhibitors; (Atazanavir, Darunavir, Fosamprenavir, Indinavir, Nelfinavir, Ritonavir, Saquinavir, Tipranavir)
69
Q

HIV meds review

  • Cross sensitvity with sulfa rash (2)
  • hyperbilirubinemia leading to jaundice (2)
  • Paresthesisas (2)
  • Booster antiviral (2)
  • Nephrolithiasis
A
  • Cross sensitivity with Sulfa Rash (good for PI resistant virus)
    • Darunavir, Tipranivir
  • Hyperbilirubinemia leading to jaundice
    • Atazanavir, Indinavir
  • Paresthesias
    • Fosamprenavir, Ritonavir
  • Booster antiviral
    • Ritonavir
    • New booster (no antiviral acivity): cobicistat
  • Nephrolithiasis
    • Indinvavir
70
Q

HIV meds review

  • Avoid in pregnancy
  • Insulin resistance/Diabetes mellitus
A
  • Avoid in Pregnancy
    • Nelfinavir
  • Insulin Resistance/Diabetes Mellitus
    • Protease Inhibitors; (Atazanavir, Darunavir, Fosamprenavir, Indinavir, Nelfinavir, Ritonavir, Saquinavir, Tipranavir)
71
Q

HIV meds review

  • Injection site reaction/irritation
  • Restricted to patients with CCR5 tropic virus
  • Restricted to treatment experienced HIV patients and causes CPK elevation/pyrexia
A
  • Injection site reaction/irritation
    • Enfuvirtide
  • Restricted to patients with CCR5 tropic virus
    • Maraviroc
  • Restricted to treatment experienced HIV patients and causes CPK elevation/pyrexia
    • Raltegravir
72
Q

Identify categories of antiretrovirals

  • CCR5 antagonists
  • Fusion inhibitors
  • NNRTIs (5)
  • NRTIs (7)
  • Integrase Inhibitors (3)
  • Protease Inhibitors (9)
A

Categories of antiretrovirals

  • CCR5 Antagonist
    • Entry inhibitors (Maraviroc)
  • Fusion Inhibitors
    • Fusion inhibitors work by blocking HIV entry into cells (Enfuvirtide)
  • Nonnucleoside Reverse Transcriptase Inhibitors (NNRTIs)
    • NNRTIs bind to and disable reverse transcriptase, a protein that HIV needs to make more copies of itself
    • §Delavirdine (DLV) § Efavirenz (EFV) § Etravirine (ETR) § Nevirapine (NVP) § Rilpivirine (RPV)
  • Nucleoside Reverse Transcriptase Inhibitors (NRTIs)
    • NRTIs are faulty versions of building blocks that HIV needs to make more copies of itself. When HIV uses an NRTI instead of a normal building block, reproduction of the virus is stalled.
    • §Abacavir (ABC) § Didanosine (ddI) § Emtricitabine (FTC) § Lamivudine (3TC) § Stavudine (d4T) § Tenofovir (TDF) § Zidovudine (AZT, ZDV)
  • Integrase Inhibitors (Raltegravir, Elvitegravir, Dolutegravir)
  • Protease Inhibitors (PIs)
    • PIs disable protease, a protein that HIV needs to make more copies of itself
    • §Atazanavir (ATV) § Darunavir (DRV) § Fosamprenavir (FPV) § Indinavir (IDV) § Lopinavir (LPV) § Nelfinavir (NFV) § Ritonavir (RTV) § Saquinavir (SQV), Tipranavir (TPV)
73
Q

Summarize HIV life cycle and how drugs work in the various stages

A

Step 1: Binding HIV attaches itself to the CD4 receptor of the T4 cell. {CCR5 antagonist}

Step 2: The HIV cell fuses to the outside of the cell {Fusion Inhibitor}

Step 3: Reverse Transcription HIV carries 2 strands of RNA which are released after the binding process. The viral enzyme reverse transcriptase allows the RNA to make a DNA copy, so called the “proviral DNA”. {NNRTI, NRTI}

Step 4: Integration HIV DNA is carried into the host’s nucleus, where the viral enzyme integrase hides proviral DNA until the cell wants to make more proteins, thus making more HIV. {Integrase inhibitors}

Step 5: Transcription Viral DNA separate and special enzymes make complementary mRNA.

Step 6: Translation mRNA is processed & corresponding strand of HIV proteins are made.

Step 7: Viral Assembly and Maturation Long strands of viral proteins are cut up by protease into smaller proteins that serve as enzymes or elements of new HIV. Once new HIV is assembled, they bud off and create a new virus. {Protease Inhibitors}

74
Q

Primary goals of HIV therapy

A
  • Reduce HIV related morbidity and prolong survival
  • Improve quality of life
  • Restore and preserve immunologic function
  • Maximally and durably suppress HIV viral load
  • Prevent vertical HIV transmission
75
Q

CD4 vs viral load in HIV

A

Uncontrolled HIV; increased viral load, decreased CD4 count

Controlled HIV; decreased viral load, increased CD4 count

  • CD4 T Cell Count
    • Major clinical indicator of immunocompetence in HIV infections
    • Usually the single most important decision to initiate therapy
    • Strongest predictor of subsequent disease progression and survival
    • Obtain baseline repeated every 3-6 months to re-assess
  • Viral Load Testing
    • Obtain plasma viral load at initiation repeated in 2-8 weeks after treatment initiation or alterations in therapy.
    • Once prescribed stable antiviral regimen, check every 3-4 months
76
Q

Opportunistic Infections in Patients with HIV
Landmark CD4 Counts

A
  • CD4 Count <200
    • Pneumocystis carinii pneumonia (PCP)
    • Aka Pnemocystis jirovecii
    • Px: Bactrim DS daily or 3 times weekly
  • CD4 Count <100
    • Toxoplasmosis
    • Toxoplasmosis Px: Positive serology àBactrim DS daily
  • CD4 Count <50
    • MAI/MAC & CMV (routine px not as clear for CMV)
    • MAC Px: Azithromycin 1200mg/week
    • CMV Px: routine px not as clear: Ganciclovir
77
Q

Combination products of HIV meds

  • Atripla
  • COmplera
  • Stribild
  • Combivir
  • Epzicom
  • Trizivir
  • Truvada
A
  • Atripla® = efavirenz + emtricitabine + tenofovir
    • *(1 tab daily: preferably QHS & empty stomach)
  • Complera® = rilpivirine + emtricitabine + tenofovir
    • *(1 tab daily: watch if high viral load >100K viral load)
  • Stribild = elvitegravir + cobicstat + emtricitabine + tenofovir
    • *(1 tab daily: treatment naïve ptns) “Quad Pill”
  • Combivir® = lamivudine + zidovudine (1 tab BID)
  • Epzicom® = abacavir + lamivudine (1 tab daily)
  • Trizivir® = abacarvir + lamivudine + zidovudine (1 tab BID)
  • Truvada® = emtricitabine + tenofovir (1 tab daily)
78
Q

Idnetify HIV med

  • Booster , always used in combo, no antiviral activity itself
  • Potent inhibitor of cytochrome P450 3A (CYP3A)
    • Acts as a pharmacoenhancing or “boosting” agent for antiviral drugs used in the treatment of HIV infection
    • Pharmacokinetic enhancer of atazanavir or darunavir
  • Utilized with Elvitegravir
  • Part of the “Quad Pill” approved Aug 2012
  • 150mg daily to be used with Atazanavir or darunavir approved in Sept 2014
A

Odd Ball: Co bicistat (Tybost)

79
Q

Identfiy HIV med

  • Mechanism of Action
    • Binds to the CCR5 receptor of the CD4 T-cell
  • Indication/Special Characteristics
    • RESTRICTED to HIV infected patients infected with CCR5 tropic virus
  • Adverse Effects
    • Abdominal pain, Rash
    • Upper resp tract infections/cough
    • Dizziness/ orthostatic hypotension
    • Musculoskeletal symptoms
A

CCR5 Antagonist (Entry Inhibitor)

Maraviroc (Selzentry)

80
Q

Identify NRTI

Slightly different mechanism allowing continued efficacy again HIV strains resistant to other Reverse Transcriptase Inhibitors

ADR

  • Lactic acidosis with hepatic steatosis (fatty liver)
  • Decreased bone density
  • Acute exacerbation of hepatitis if co-inf with Hep B
A

Tenofovir

2 types

TAF and TDF

81
Q

HIV class medicatin

nMOA: Inhibit HIV-1 protease

nHIV-1 protease is responsible for cleaving large viral polypeptides into smaller functional virons

nAll PI cleared via Hepatic oxidative metabolism à drug-drug interactions

nInhibitors of 3A4 – all to a varying degree

nRitonavir “booster” allowing lower doses and pill burden for several antivirals

nFood effects all over the board

nFrom Increased of 70-570%

nTo Decrease of 70%

A

Protease Inhibitors (PI)

  • All PIs are associated with Endocrine type side effects:
    • nHyperlipidemia
    • nFat maldistribution
    • nInsulin Resistance/Diabetes mellitus
    • nOsteonecrosis
  • Ritonavir – the booster of all boosters!!!
    • nAll regimens booster with ritonavir are associated with increased LDL, TG and HDL (esp TG)
    • nAllow lower dose or less pill burden
82
Q

HIV med in HIV infected women

A

§Efavirenz

  • §Teratogenic in nonhuman primates
  • §Risk of neural tube defects occurs during the first 5-6 weeks of pregnancy, and pregnancy usually is not recognized before 4-6 weeks of pregnancy
  • §Do pregnancy test before starting EFV (women of childbearing potential)
  • §Counsel about potential risk to fetus and desirability of avoiding pregnancy while on EFV
  • §Use alternative ARV agent in women who are trying to conceive or who are not using effective contraception, if feasible
83
Q

Post exposure prophylaxis

A

PEP

  • Raltegravir (Isentress) 400 mg BID
  • PLUS Truvada Daily
    • Truvada = tenofovir 300mg & emtricitabine 200mg)

Ideally start within 2 hours of exposure (best animal data shows w/in 72hrs)

Do not delay starting PEP for any reason

Continue to take for 28 days (best animal data, not exact science)

Medicolegal issues: offer PEP, proper baseline and follow up assessments

84
Q
A
85
Q

HIV case discussion

  • false positive from what test
  • age group to test
  • most common combo drug
  • primary infection
A
  • Don’t rely on viral load from finger stick – lots of false positive
  • Who to test? from age 13 – 75 years old (atleast once). More often if sexually active or higher risk.
  • Juluca; most common combo drug (2 drugs)
  • Acute retroviral syndrome; primary infection from HIV. 2-4 weeks after exposure. – flu, LAD, pharyngitis, rash, myalgiaarthralgia, thrombocytopenia, leukopenia, diarrhea, HA, transaminitis, HSM, thrush, neuropathy, encephalopathy, night sweats
86
Q

HIV case discussion

  • HIV in pregnancy; scrrened when? transmission how?
  • IRIS? what organ is affected
A

HIV in pregnancy; must be screened at beginning of pregnancy and tested at end of pregnancy

  • Transmission in; pregnancy, labor and delivery and breastfeeding.
  • With suppression; risk is <2%

IRIS; immune reconstitution inflammatory syndrome

  • Start ART and then elicit previously absent host inflammatory response
  • Classic; TB, Cryptococcus (wait 4 weeks before starting ART)
87
Q

Hospital case

43 y.o female admitted with left knee bursa abscess

  • WBC 0.2. Hgb 8, Plt 14 - pancytopenia (bone marrow failure)
  • drug user. shared needles with dead brother from OD
  • Allergy; bactrim
  • PMH; anxiety
  • Patient is on cefazolin for MSSA
A
  • Patient has untreated HIV; why she as bone marrow depletion (pancytopenia)
88
Q

Hospital case

38 y.o male with fever, rash, diarrhea, LAD, knee abscess, axillary abcess

  • Syphilis
  • Groin lymphadenpathy
  • Emphetemine positive
A

Syphilis and HIV