• 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

1/168

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;

168 Cards in this Set

  • Front
  • Back
Virion
The virus particle:
Made up of:
1. Viral nucleic acid
2. viral structural proteins

1+2 = nucleocapsid
Properties of viruses
Small, infectious, obligate intracellular parasite

Genome- DNA or RNA

In host cell, genome is replicated and directs synthesis of other virion components by host systems

De novo assembly of of progeny virions
Viral structural proteins
Encoded by viral genome (not host)

Packaged into virion- provides protective coat to nucleic acid

Includes: capsid (protein) and lipoprotein envelope (some viruses: viral protein + cell membrane)
Nonstructural proteins
Encoded by viral genome
Not packaged into virion
Usually enzymes or
transcription factors
Necessary for viral replication in cell

(first segment in ssRNAsense)
Classification
RNA or DNA
Symmetry of capsid
Presence/absence of envelope
Dimensions of virion & caspid

Genome:
RNA: ss: Plus strand
RNA: ss: Minus strand: nonsegmented v. segmented
RNA: ds: segmented

DNA: ss
DNA: ds: linear or ciruclar
Virus Evolution
RNAP lack proofreading fxn
Population = quasispecies

Can evolve by:
Mutation
Recombination (two viruses)
Reassortment (2 viruses w/ segmented genomes)

Ex: human + swine + avian reassortment in host
Growth
One step growth curves for viruses compared to bacteria

Takes longer to replicate, but viral load gets many folds higher than bacteria
Cell surface molecules for virus attachment
and
Binding sites on viruses
Carbohydrates- sialic acid, glycosaminoglycans

Lipids- glycolipids, proteolipids

Proteins- Ig superfamily, complement regulatory proteins, integrins, TNF receptor superfamily

Receptor binding sites on viruses
-Depressions (Picornavirus) OR
-Projections (Rotavirus)
Where RNA and DNA genomes make mRNA...
RNA viruses- CYTOPLASM
(exception: Influenza!)

DNA viruses- NUCLEUS
How to get a lot of information into a small genome
-Overlapping reading frames
-Coding off both strands
-RNA splicing
-Frame-shifting
-RNA editing
How to persist in population
-Do NOT kill host
-Do NOT kill cells in which virus is replicating
-Do NOT get eliminated by immune response
How to have sufficient stability for transmission, but sufficient instability for cell infection
Endocytosis
-enveloped & nonenveloped
-clathrin-coated pits
-Entry into sytoplasm, then fuses w/ endosomes w/ acidiphication
-acidiphication necessary for viral protein conformational changes (like HA of influenza)

Fusion
-Enveloped virus fuses directly w/ cell membrane
-Virus discharged into cytoplasm
Interferons
Viral ds RNA infects cell
Interferon synthesis and liberation
Protein induced blocks replication of unrelated virus

IFN binds to IFN receptors, signal through JAK-STAT pathway to induce antiviral protein genes

IFN control spread of virus prior to appearance of acquired immune response
Antiviral antibodies (tests)
Serological tests- used for dx
-Enzyme immunoassays
-RIA
-W. blot

Biological activity- gives info on fxn of Ab
-Virus neutralization
-Complement fixation
-Hemagglutination inhibition (viruses bound together, can't infect)
Cell-mediated immunity in viral infection
Focus immune response to cells expressing MHC I
Clear infected cells
Recruit other effector cells
Activate macrophages
Provide help for production of antibody by B cells
Human picornavirus
Plus-strand RNA viruses
Small
Rhinoviruses
Enteroviruses (Polioviruses, ECHO viruses, ENteroviruses, Coxsackie)
Hepatitis A virus

RNA replication in cytoplasm

3 surface proteins, 1 interior protein

Receptor binds into "canyon" of picornavirus

Plus strand RNA is translated into a single polyprotein

Virus-encoded proteases process polyprotein into individual proteins

Internal ribosomal entry site (IRES) in 5' nontranslated region of RNA
IRES allows NA to bind directly to ribosome w/o need for 5'7-methyl G cap

Rhinovirus: local URIs (problems in asthma pts)

Enterovirus: cause systemic infection (general viremia-> other organs)
Rash from enterovirus infection

Poliovirus: replication in motor neurons in spinal card
Poliomyelitis: inflammation and death of motor neurons
Virus encoded protease 2A cleaves EIF4G that binds cap-binding protein to assemble initiation complex (shuts off host cell protein synthesis)
Polio
Human picornavirus
Viral Virulence
Under polygenic control
Virulent- causes disease
Attenuated- no or reduced disease
Avirulent- no disease

Virulence genes:
Affect ability of virus to replicate
Defeat host's defense mech
Promote virus spread w/in and among hosts
Gene products that are toxic- cause cell injury directly
Viral Tropism
Virus has to enter cell- susceptibility

Virus has to replicate in cell - permissivity

Neurotropism
Pneumotropism
Enterotropism etc

ALL= PANTROPISM
Viral Receptors
Required for virus entry
Determines host and tissue tropism
Some viruses need co-receptor
Active process

Integrins
Ig-like molecules
GLycosaminoglycans
CHO
Viral Spread
Local replication
Systemic spread- must cross barriers such as basement membrane

Directional release- major determinant of infection pattern

(influenza virus into lumen)
(vesicular stomatitis virus crosses basement membrane)
Viremia
Circulating virus in blood (in circulating WBCs)
DISSEMINATION

Monocytes- Measles, HIV, CMV
Lymphocytes- HIV, Epstein-Barr, Human herpesviruses 6&7
Neutrophils- Influenza virus
Free- Poliovirus, HepB
Viruses vs. Respiratory System
Transmission: hand-shaking, coughing, sneezing

Host Defense: Mucociliary apparatus, Alveolar macrophages, Adaptive immune response

URI: Rhino, Influenza
Lower: Adeno, Influenza
Viruses vs. GI Tract
Transmission: eating, Drinking, poor hygiene

Host Defenses: Stomach pH, digestive enzymes, flow of ingesta, Adaptive immune response

Rotavirus
Reovirus (translocated across M cell)
Measles
Poliovirus
Adeno
Viruses vs. UG Tract
Transmission: Sexual activity, fecal contamination

Host Defenses: Urine flow, thick epithelial layer, acid pH, Adaptive immune response

HIV, HSV (lifelong persistent or latent infections)
HPV- associated w/ cervical cancer
Viruses vs. Eyes
Transmission: COnjunctiva, abrasions, direct inoculation

Host Defenses: Tears, thick epithelia layer, Adaptive immune response

HSV: lifelong persistent or latent
Viruses vs. Skin
Transmission: cuts, abrasions; insect bites; needles

Host Defenses: epidermis, skin oils

Poxviruses
Papillomaviruses
Rabies
Togaviruses- insects
Alphaviruses- insects

Measles virus, chicken pox- come up to skin and replicate
Viruses vs. Nervous System
Transmission: cuts, animal bites; inhalation; cell trafficking

Host Defenses: Blood-brain barrier

Rabies
HSV
HIV
Measles
Alphaviruses

Enter via sensory nerve ending and move retrogradely
Can be latent in DRGs (good example is HSV)
Cytopathic Effects
Cell swelling
Necrosis
Apoptosis
Inclusion bodies
Syncytia/multinucleated giant cells
Cellular hyperplasia/proliferation
Cytopathic Effect: Viral Inclusions
Inclusion Bodies

Viral Inclusions: viral and/or cellular products, present very early in infection, intranuclear and/or intracytoplasmic, Eosinophilic, basophilic, amphophilic
Peripheralization of chromatin

Only aid in diagnosis

Herpesvirus- eosinophiic intranuclear inclusions

CMV- Owl's eye inclusion
Cytopathic Effect: Necrosis
Necrosis
shutdown of protein, nucleic acid synthesis; accumulation of viral components in cell
Ballooning degeneration
Cell death- pyknosis (chromatin clumping); hypereosinophilic cytoplasm
Disruption of tissue architecture
Fetal dev- malformations
Cytopathic Effect: Apoptosis
Promoting Apoptosis: initiation/promotion of cascade
Aids in virus dissemination (lysis)


Inhibiting Apoptosis: permits longterm virus replication, establishes latency
Cytopathic Effects: Syncytia
Viral fusion proteins expressed on surface of cells cause cells to fuse together, producing multinucleated cells

Seen in vivo or in vitro

Purpose: permits virus transmission w/o exposure to host defenses
Cytopathic Effects: Hyperplasia/proliferation
Usually self-limited, transient

May be due to atypical differentiation, accumulation of viral products

May be pre-neoplastic

Ex: Molluscum contagiosum (pox virus)
Virus-induced neoplasia
Often involves integration of viral genomes into host DNA

Ex:
Epstein-Barr virus- NP carcinoma - Burkitts Lymphoma

HPV- cervical carcinoma

HTLV-2- T-cell leukemia
Classic host inflammatory response to viruses
Mononuclear cells:

Lymphocytes
Macrophages
Host susceptibility to viral disease
Genetic determinants:
Diversity of MHC Class I genes
Specific resistance genes

Non-genetic determinants:
Age-related reduced immune competence: infants, elderly
Gender- males, pregnant women
Malnutrition
Corticosteroids, cigarette smoking, stress
Cytopathic Effects: Syncytia
Viral fusion proteins expressed on surface of cells cause cells to fuse together, producing multinucleated cells

Seen in vivo or in vitro

Purpose: permits virus transmission w/o exposure to host defenses
Cytopathic Effects: Hyperplasia/proliferation
Usually self-limited, transient

May be due to atypical differentiation, accumulation of viral products

May be pre-neoplastic

Ex: Molluscum contagiosum (pox virus)
Virus-induced neoplasia
Often involves integration of viral genomes into host DNA

Ex:
Epstein-Barr virus- NP carcinoma - Burkitts Lymphoma

HPV- cervical carcinoma

HTLV-2- T-cell leukemia
Classic host inflammatory response to viruses
Mononuclear cells:

Lymphocytes
Macrophages
Host susceptibility to viral disease
Genetic determinants:
Diversity of MHC Class I genes
Specific resistance genes

Non-genetic determinants:
Age-related reduced immune competence: infants, elderly
Gender- males, pregnant women
Malnutrition
Corticosteroids, cigarette smoking, stress
Key differences between positive and negative strand viruses
Positive Strand RNA Virus:
-First replication step: polymerase synthesis via genomic RNA translation
- strand has non-structural proteins-structural proteins (Polarity)
-Must translate RNAP

Negative Strand RNA Virus
-First replication step is + strand RNA synthesis (mRNA, full length antigenome)
-strand is structural proteins-non-structural proteins (Polarity)
-RNAP is component of incoming virion
Negative strand viruses: Key Features
RNA is not infectious!
Virion contains RNA dependent RNA polymerase
Genomic RNA is packaged in protein (encapsidation)
-Structure = nucleocapsid
Nucleocapsids have helical structure
Virions are enveloped
Entry by virion fusion or cell-cell fusion

Important**
Influenza A,B,C
Parainfluenza 1-4
Mumps
Measles
Respiratory Syncytial
Rabies
Influenza Viruses
3 types A, B, C
A & B antigenically distinct, structurally similar
Influenza A- infects ducks, chx, horses, swine
Influenza virus- avian strains rarely infect humans
Influenza Virion
Structural features:
HA, NA, M2
Genome comprised of segments; 1-3 genes/segment; HA and txn complex on separate segments
Lipid envelope contains viral glycoproteins (HA, NA)
Gene Reassortment
2 different strains infect same cell
Replication of all genome segments
Virus assembly with mixing of segments
End Result: Reassortment Progeny Viruses
Reassortment causes Antigenic SHIFT
For example:

The new virus can replicate in human cells

Humans lack immunity
Influenza virus Hemagglutinin
16 antigenic types
Trimer; globular head on stalk
Multifunctional
-Binds cellular receptor sialic acid
-Binds and agglutinates rbc's
-Mediates fusion of viral envelope w/ cell membrane

HA synthesized in "pro-form"
-Cleavage required for conformational change/activation of fusion function
-Cleavage by tryptase Clara (serine protease secreted by nonciliated Clara cells in bronchial and bronchiolar epithelium found in lumen of respiratory tract)
Target of neutralizing antibodies
-Minor mutations result in antigenic DRIFT

Replacement with gene from alternate hosts results in antigenic SHIFT
Influenza virus entry into host cell
Binding
Virus particle endocytosed
Low pH-induced HA conformational change w/in endosome
-Exposure of hydrophobic aa's in HA (mels w/ membrane)
Fusion of viral envelope w/ endosome membrane
Release of genomes into cytoplasm
No syncytium formation
Influenza virus Neuraminidase
Tetramer
Function: during virus exit, cleaves sialic acid residues on cell surface
Influenza A: 0 recognized subtypes

**If mutant neuraminidase gene, virus cannot cleave and disperse
Influenza nomenclature
Type/location/ # of isolate/ year of first isolation/ HA and NA subtypes
Influenza virus M2 Protein
Tetramer
Spans viral envelope
Function:
Pumps protons into virion at low pH
Loosens protein-protein contact
Facilitates virus uncoating
Target for amantadine
Influenza virus replication
Genome replication occurs in nucleus
-virus lacks mRNA capping and methylating enzymes
-steals caps from host mRNAs
-cap plus 10-13nt are used to prime viral mRNA synthesis
Viral mRNAs are translated on SER and RER
-HA and NA transported thru Golgi to cell membrane

Virus replicates in ciliated columnar epithelial cells in respiratory tract
-causes tracheobronchitis
Lg numbers of virions shed into resp tract- facilitates transmission
Virus-induced apoptosis of infected cells
Infection damages resp tract
-PROTECTIVE MUCUS LAYER DISRUPTED
-Resp epithelium denuded
Influenza Virus Clinical features
Transmission
-Resp
-Seasonal, primarily winter

Symptoms
-Appear 1-4d post infection
-HA, fever, myalgias, nonprod cough, sore throat, no rhinorrhea
-Lasts 3-7 d
-Likely caused by local prod of interferon and IL-1 (immune response)
Influenza Immunity
Innate:
Mucus barrier
Clearance by cilia
Alveolar macrophages

Adaptive
Protection: IgA (mucosal); IgG (serum)
Clearance: IgG + complement; CTL

At risk: Elderly, smokers, chronic pulm disease (emphysema)
*Mucus barrier disruped
Influenza virus complications
Primary virus infection
-Interstitial pneumonitis

Secondary bacterial pneumonia
-Facilitated by damage to innate immune system
-Destruction of ciliated epi cells
-Abnormal macrophage function

Secondary- repsonds to Abx, mortality lower than primary

Primary viral pneumo common in pregnancy-> pregnant women should definitely get vaccinated!
Influenza Immunization
KILLED (inactivated) vaccine (primarily HA, NA)
-Reformulated every year
-Recommended for health care workers, at risk pop
-Provides partial protection

LIVE ATTENUATED INTRANASAL VACCINE (FLU MIST)
Replication restricted to nasopharynx
Cold adapted
Reformulated annually
Approved for use in healthy people 2-65 yrs
Influenza Dx
Direct detection- staining of NP aspirates w/ flu-specific monoclonal antibodies

Culture
Paramyxoviruses
8 medically important viruses
Parainfluenza 1-4
Respiratory Syncytial Virus (Pneumovirus)
Human Metapneumovirus
Measles (Morbillivirus)
Mumps (Rubulavirus)

Characteristics:
Do NOT undergo epidem. important antigenic change
NO natural reservoir- cont. person-person spread
Spread by resp route

Proteins:
H (glycoprotein) mediates receptor binding
F (glycoprotein) mediates fusion

Nonsegmented genome
mRNAs generated by polymerase reinitiation

Glycoproteins- carbs attached in ER, Golgi
Cell attachment protein- H
Fusion protein- F (to be active must be cleaved into F1, F2 by intracellular proteases at neutral pH)

Replication cycle:
Virions fuse at cell membrane
RNAs synthesized in cytoplasm
H/F transported thru ER/Golgi to plasma mem
Nucleocapsids assemble
Budding from cell membrane OR
fusion w/ adjacent cell (fusogenic surface proteins at neutral pH)
Resp Syncytial Virus
Causes outbreaks of resp disease in winter
Direct contact w/ resp secretion
Causes otitis media, bronchitis, bronchiolithis, croup (infection of larynx&trachea), pneumonia
Most severe in young infant
Partial immunity post primary infection

Primary RSV URI can progress in severity
-cough, weezing
-dyspnea, increased RR, hypoxemia

Dx: direct staining of NPAs w/ fluorescent monoclonal antibodies, culture
Vaccine: under dev.
Human Metapneumovirus
Most infections occur in childhood
Serologic evidence of circ for at least 50 yrs
Causes 7-40% peds resp inf.
Dx: RT-PCR
Antibodies for direct detection of viral antigens now available

Parallels RSV: seasonal (winter); initial exposure in childhood
Clinical: range from ild resp symptoms to severe cough, bronchiolitis, pneumonia
Repeated infections occur- prod less severe disease limited to upper resp tract
Parainfluenza Diseases/Prevention
Parainfluenza 1-3
Common cause of URIs
Most common of laryngotracheo-bronchitis croup in young chidren
Most children infected by 5
Dx: culutre
vaccine: not yet available
Measles
Geo: worldwide
Epidemics in vaccinated countries
Endemics in unvaccinated
Routes of transmission
-resp
-aerosol
High attack rate
High mortality rate in infants in developing countries

Systemic replication
-first site- resp epithelium
-second site- local lymph nodes
-dissemination by infected monocytes departing resp LNs (primary viremia)
-Epithelial/endothelial cells infected throughout body release virus into blood (secondary viremia)

Clinical Sx:
Arise during secondary viremia
Prodrome: fever, cough, coyza, conjunctivitis, Koplik spots
Rash arises w/ immune response
(cellular response)
Immune responses
-CD4 help
-CD8 clearance
-IgM, neutralizing IgG
Measles-Induced immune deficiency
-Gen. immunosuppression
Early-lymphopenia
Middle- decreased DTH responses
dec lymphoproliferation
Late- effects on thymus?

Dx:clinical pic, direct stain, culture, serology

Live attenuated vaccine: given to infants 12-15 mo
Mumps
Infection of glandular epithelial cells
-Parotitis, orchitis
-Pancreatitis, ovarian infection infrequent
-Meningitis

Dx: culture (saliva, urine, CSF)
Prev: live attenuated vaccine
Rhabdovirus
Rabies is only important human pathogen

Transmitted in saliva from bite of infected animal
Limited replication in muscle and subepithelial tissue
Uptake by sensory/motor neurons
Retrograde transport to cell body (major site of replication)
Early avoidance of imune response thru transynaptic transmission

Symptoms:
Prodrome: fever, malaise, paresthesias at bite site
Later: anxiety, aggressive behavior, seizures, hypertonia, paralysis

Post-exposure prophylaxis can save!

Dx: clinical (exposure) history, biopsy, PCR

Prev: Inactivated vaccien
-Long incubation period allows for post-exposure use
-Pre-exposre immunization in vocations w/ high risk (vets, wildlife mgrs)
Parvoviruses
small DNA animal viruses
Icosahedral symmetry, nonenveloped
3 protein and linear ss DNA
Only replicate in DIVIDING cells or cells infected w/ helper viruses
Autonomous parvoviruses replicate alone
Dependoviruses require a helper virus
Parvovirus B19
Autonomous parvovirus
Causes childhood rash disease erythema infectiosum (fifth disease), acute recurrent arthritis, aplastic crisis in persons w/ chronic hemolytic anemia, persistent infection w/ chronic anemia in immunocomp indiv, and hydrops fetalis

Replicates preferentially in erythroid precursor cells

Normal child/adult: B19 infection is biphasic- viremic (fever, malaise, myalgias); erythematous RASH on face. Adult often develop arthritis

Acute infection- destruction of erythroid precursors in bone marrow
Transient anemia important in individuals w/ chronic hemolytic anemia due to dzs such as sickle cell, thalassemia, heriditary spherocytosis
Avg. life of circulating erythrocytes is shortened (apoptosis from NSP)
**Aplastic cris
Immunodeficient individuals are unable to clear infection; develop chronic anemia due to RED CELL APLASIA
Fetus- severe anemia-> hydrops fetalis

B19 is ubiquitous, highly contagious
Respiratory secretion, common in school age children & their parents
No vaccien
Bocavirus
Parvovirus
Infects respiratory tract
Adeno-associated virus (AAV)
Dependovirus parvovirus
Causes no recognized disease
Integrates into cellular DNA
Being developed as vector for gene therapy
Papillomaviruses
HpVs
icosahedral
Supercoiled circular dsDNA
Cause of warts, squamous carcinomas
Replication in nucleus of squamous epithelia cellsf
Viral genes expressed in 2 phases:
Early (E) genes encode reg. proteins for replication, txn, transformation
E2, E6, E7 important in disease
Long control region (LCR)- origin of rep and control elements for txn, rep
Late (L) genes encode capsid structural proteins L1, L2
Papillomaviruses cont'd

cutaneous type
Cause warts
Virus enters skin through abrasion
Expression of early genes leads to hyperplasia, formation of wart
Late genes encoding L1 and L2 are expressed and infectious as infected cells in basal layer differentiate into keratinocytes (move to surface)
Papillomaviruses cont'd

mucosal types
Infect genital, oral, respiratory mucosa
Women- proliferating cells at border of squamous and columnar epithelium of cervix infected- flat condyloma develops
Virus clearance (1-2 yrs) delayed in HIV-infected
Persistent infection->cytologic abnormalities on Pap smear

All cervical carcinoma initiated by HPV
HPV-16 and HPV-18 most oncogenic (45, 56)
Oncogenicity- assoc w/ fxn of E6, E7

E7- induces DNA syn in resting cells (bind retinoblastoma tumor suppressor pRB w/ high affinity)
E6 protein activates telomerase, complexes w/ p53 (w/ E6AP, E3 ubiquitin ligase)- targets p53 for ubiquitin-mediated degradation

In malignant cells, viral genome is often integrated to disrupt early gene E2 that controls expression of E6, E7

Papanicolaou smear used as screening device
PCR used to type
Immunization w/ VLPs from L1 capsid protein protects
Recommended for teenage girls
Cervical carcinoma = major cause of death due to malignancy in developing countries
Goals of Antiretroviral therapy
Decrease HIV replication by as much as possible for as long as possible
Nucleoside Analog Reverse Transcriptase Inhibitors (NRTI's)
Approved
**Zidovudine (Azizothymidine, AZT)
Mech: Phosphorylated by cellular enzymes to triphosphate (AZT-TP)
AZTTP is active form!
Inhibits HIV-encoded RT (RNA-dependent DNA polymerase)
Acts as chain terminator
Reduces plasma HIV-1 RNA minimally when used alone

PK: well absorbed
Eliminated by glucuronidation (Phase II conjugation)
Short plasma T1/2 of parent
Longer T1/2 of intracellular AZT-TP (allows infrequent dosing, bid)
Rapid conversion to AZT-MP (which accumulates)
Slow conversion to di- and tri-

Toxicity: Bone marrow suppression, mainly anemia; less commonly granulocytopenia; rare myopathy and lactic acidosis/steatosis

NRTI's toxic because poor selective tox: also inhibits mitochondrial DNA polymerase!!! (toxicity stems from mt malfunction)

Resistance:
Requires 5+ specific amino acid changes
Develops slowly (months to years)
Limited cross-resistance w/ other nucleosides

Tenofovir prescribed more often
Non-Nucleoside Reverse Transcriptase Inhibitors (NNRTI's)
Approved
**Nevirapine
Big difference between nuc and non-nuc is that non-nuc have very different structures (as a group)

Mech: Does not require intracellular activation
Non-competitive inhibitor of RT
Binds to enzyme at site DISTANT from active site and causes conformational change in enzyme rendering it less efficient

Efficacy: Reduces plasma HIV-1 by 2-3 logs; effects are short-lived as monotherapy

Selective Tox: NO effect on human DNA polymerases

PK: Well absorbed
Eliminated by oxidative metab (CYP450 3A4)
Long plasma T1/2, allows infrequent dosing, bid, could be qd)
P450 Enzyme inducer

Toxicity: Rash (common); Steven Johnson Syndrome (rare) (SJS is systemic attack by immune system of epithelium, full body burn); Hepatotoxicity (rare); Drug interactions- P450 enzyme inducer

Resistance: single specific aa change, confers up to 1000-fold resistance to drug; resistance emerges rapidly (days to weeks); Single point mut also confer
Cross resistance to other NNRTI's
Efavirenz
Selective, potent NNRTI
Long half-life
qd
CSF and semen HIV-RNA reduction
Safe, well tolerated
Pregnancy should be avoided

Treatment convenient:
1 pill qd
Coformulation- 3 drugs/pill
HIV Protease Inhibitors PI's
Approved
Saquinavir
Ritonavir

Mech: Does not require intracellular activation; Competitive inhibitor; Transition state peptidomimetic inhibitor (chemical analog of transition state)

*Protease cleaves pre-proteins into mature proteins
Protease inhibitor --I infectious virus (non-infectious immature virus can still persist)

HIV PI's do NOT prevent virion formation and release, but inhibit virion maturation

Selective Tox: Do not inhibit human aspartyl proteases; Reduce plasma HIV-1 RNA by 2-3 logs as monotherapy.
Partially restore CD4 cell count

PK:
Variable bioavailability due to first-pass metab and autoinduction
Highly protein-bound
Eliminated by oxidative metab (CYP450 3A4)
Plasma T1/2 3-5 hours, dosed bid
Potent P450 enzyme inhibitor (3A4>2D6) and hepatic enzyme inducer

Toxicity: GI intolerance n/v, diarrhea;
Hyperlipidemia (common w/ antiretroviral drugs)
Glucose intolerance (rare)

Associtaed w/ fat redistribution (more strongly associated w/ stavudine)
Hepatic transaminits (rare)
Circumorla and extremity parasthesias (common during first few weeks)
Drug Interactions*

Ritonavir enhances plasma concentrations of other protease inhibitors

Resistance:
An initial single aa change confers 3-5 fold resistance = PRIMARY RESISTANCE

SECONDARY aa changes accumulate, conferring ever-increasing resistance (up to 100-fold)
Resistance emerges w/in weeks to months (not in all subjects)
Accumulated mutations confer partial or complete cross-resistance to other PI's
Dose-response curve: higher doses of drugs suppress emergence of resistance genotype.
Integrase Inhibitors
Approved
Raltegravir
Mech: inhibits HIV integrase DNA strand-transfer rxn essential for chromosomal integration
Oral drug, bid
Highly potent anti-HIV effects
Well tolerated, little/no tox
Entry Inhibitors- Enfuvirtide
Approved
Enfuvirtide
Mech: interferes w/ membrane protein helix bundle formation necessary for membrane fusion and entry (analog of piece of HIV envelope protein)
Must be injected bid
Expensive
Reserve for highly treatment-experienced pts
Entry Inhibitor- Maraviroc
Approved
Maraviroc (CCR5 antagonist)
Mech: selectively inhibits HIV entry mediated by CCR5 chemokine co-receptor
Only approved antiretroviral that targets host protein
Oral, bid
Only effective in pts w/ CCR5-trophic HIV (need phenotypic sensitivity test to prescribe)
Treatment-experienced pts
Antiretroviral Usage Today
Combination therapy ONLY
Potent drug combos:
HAART = Highly Active Antiretroviral Therapy
Most popular starting regimens:
Efavirenz + 2 NRTI's
Potent PI + 2 NRTI's

Rational:
Prevention of drug resistance (Minimum of 3 active agents required to prevent emergence of resistance)
No role for synergy

Reduced pill burden:
Co-formulated drugs
Once-daily regimens
Better long-term tolerability
Central role of resistance testing
Influenza A
4 subtypes of flu A have circulated among humans recently:
H1N1, H1N2, H3N2, and novel (swine) H1N1

Flu viruses mutate regularly
Antigenic drift:
-Variations of antigens w/in same H and N class; -Requires annual reformulation of influenza vaccine
Antigenic shift:
-Complete change in H, N or both H and N
-Occurs when bird strain re-assorts w/ human strain
How does Influenza cause a pandemic?
Susceptible population (no immunity)
Transmission from animals to humans
Transmission from human to human
Sustained human to human transmission
(avian flu did not see sustained human-human as seen w/ swine flu)
Influenza
Season: annual epidemic spread; late fall, winter, early spring; most influenza occurs in Jan/Feb

Epidemiology: All ages, highest rates in children, serious illness/death >65, <2, high risk; Annually 36,000 deaths in US (most >65)

Clinical signs:
Abrupt onset of constitutional and resp signs/symptoms: malaise, myalgias, headache, fever, non-productive cough, rhitis, sore throat, otitis
Uncomplicated dz resolves 3-7 days, cough/malaise linger up to 2 weeks

novel influenza A(H1N1) affecting young age group 5-49


Complicated: primarily viral pneumo, exacerbation of underlying med dzs, secondary bac pneumo, co-infection w/ other viral/bac,
**Ask about chest pain, COPD exacerbations, changes in mental status, falls...

Dx: Absence of ILI does not rule out flu; lab dx and high level of suspicion during season necessary!
NP Aspirate; viral culture, rapid dx tests, immunofluorescence, RT-PCR, serology
Influenza Transmission
Transmission:
Person-person
-large particle resp droplets
Close contact
Contaminated surfaces

Incubation 1-4 days
Adults infectious from 1 day prior to symptom onset thru 5 days after
Children shed several days prior to symptom onset for more than 10 days after

Immunocomp can shed for months
Influenza Vaccination
Annual vaccination b/c of antigenic drift

Two types:
Trivalent inactivated vaccine (TIV)
-injected
-grown in eggs
-3 strains
-inactivated, killed
-subunit, subvirion, purified surface antigen
-CANNOT cause influenza (killed)

Live Attenuated Intranasal Vaccine (LAIV)
-Intra-nasal Admin.
-Grown in eggs
-3 strains (same as inact)
-Live, attenuated
-Potential to cause mild sign and symptoms
FluMist
-FDA approved 2-50yrs
-Should NOT be given to pregnant women, immunosuppressed pts


ACIP recs
-Children 6mo-19yrs
-Pregnant women
->50yrs
-People at or who live with/care for people at high risk for complications
-Healthcare personnel

H1N1 vac recs similar:
pregnant, contacts/caregivers of children <6mo, healthcare workers, 6mo-24yrs, at risk of complications from flu

Conditions at risk for Complications:
Chronic Pulm Disease (asthma)
CVD
Renal, hepatic, hema-, metabolic disorders
Immunosuppression
Cognitive/Neuromuscular dysfunction compromising resp fxn
Antiviral Medications
ADJUNCT to vacinate
4 licensed agents in US
Adamantanes
-Adamantadien, Rimantadine
Neurominidase Inhibitors
-Oseltamivir, Zanamivir

Used for: Rx, Prophylaxis

Adamantanes- Seasonal H1N1 susceptible, H3N2 resistant, Pandemic H1N1 resistant

Oseltamivie: Seasonal H1N1 Resistant; Pandemic H1N1 susceptible
Polio
Prior to vaccine:
Infection tended to be early in developing countries and late in industrialized countries.

Caused paralytic poliomyelitis

Jonas Salk- inactivated vaccine
Albert Sabin- live virus vaccine

Inactivated- currently used in US
Live attenuated- generates mucosal immunity; tendency for revertants to wt
When wt poliovirus was eliminated in US, all cases of paralytic polio were due to live virus vaccine

Still endemic in parts of Africa, India
Problems:
-Importation
-Circulation of recombinant viruses
-Prolonged excretion by immunodeficient pts
Togavirus
Enveloped icosahedral virus
Genome = mRNA for nonstructural proteins
Second subgenomic RNA synthesized for translation of structural proteins
*Enveloped plus-strand RNA viruses
Two types:
Rubella virus
Alphavirus
Rubella Virus
Togavirus
Enveloped plus-strand RNA virus
Resp transmission, worldwide distribution
Causes mild rash illness in children and adults
Fetal infection->congenital rubells syndrome w/ mental retardation, heart defects, cataracts...
Congenital infection: Requires maternal exposure, maternal blood invasion, placental infection, entry into baby's blood, fetal infection
Highest in 1st, 3rd trimester
Alphavirus
Togavirus
Enveloped plus-strand RNA virus
Spread by mosquitoes, geographically restricted distribution
Cause encephalitis (EEE, WEE) or rash and arthritis
Encephalitis: mosquito-skin-blood-macrophages, spleen, lymph nodes- brain- encephalitis

Eastern equine encephalitis
Western equine encephalitis
Flaviviruses
Mosquito-born viruses
-Yellow fever virus
-Dengue virus
-Japanes encephalitis virus
-West Nile virus

Tick-borne viruses

Hepatitis C virus

West Nile- causes encephalitis
Coronaviruses
Human Coronaviruses
1. Common cold
2. Severe acute respiratory syndrome (SARS)
Virus properties facilitating infection of GI tract
Resistance to low pH (stomach)
Resistance to detergents (bile)
Resistance to proteases (SI)
Viruses that replicate in GI tract but don't cause GI disease
Enteroviruses (Poliovirus)
Reoviruses
Adenoviruses
Viruses that replicate in GI tract and CAUSE gastroenteritis
**Calicivirus (Norovirus):
+ strand RNA, no envelope

**Rotavirus:
dsRNA, segmented, no envelope
Rotavirus
Rotavirus diarrhea most common cause of severe dehydrating diarrhea in young chidren

Genome: dsRNA, purified segments aren't infectious
Each segment- codes 1 protein
RNA segments form diff viruses; may reassort at high freq during dual infection

IN REOVIRUS FAMILY

Can by typed by Electrophoresis (Electropherotypes, segment sizes)

Capsid structure:
Outer capsid VP4, VP7:
VP7: Viral surface glycoprotein, 30% of virion
VP4: 60 spikes on surface of virus, 1.5% of virion

Trypsin cleavage of VP4 into VP8 and VP5 required for INFECTIVITY
VP5 selectively permeabilizes membranes

Contains own viral RNAP- immediately makes + strand message

Histology: 1-3 days- villus atrophy and blunting, inflammation, mononuclear infiltration of lamina propia, vacuolation of epithelial cells

Rotavirus infects mature absoptive enterocytes in SI
**Enterotoxic nsP4 peptide
Stimuation Cl- secretion by secretory crypt cells
WATERY diarrhea
nsP4 may also stim enteric nervous system-> Diarrhea

Infects younger people
Equal cause of infant diarrhea in developed, developing countries

Dx: antigen-specific enzyme immunoassay

More common in winter

Vaccine:
Two live, oral attenuated vaccines against rotavirus infection FDA approved
-bovine reassortment vaccine
-No clear intussusception risk
Routine component of pediatric vaccine (Not in developing world)
Norovirus
A calicivirus

+ strand RNA, no envelope

Receptor dependent on enzyme encoded by FUT2:
FUT2 encodes enzyme that produces carbohydrate H type 1 on surface of epithelia cells and in mucosal secretions to which NV attaches "secretor" status

Glycosyltransferases, Lewis (FUT3) and A, B enzyme of ABO ystem can modify H type 1 sturcuture at cell surface
Mutations to FUT2 or (FUT3, ABO) can render "nonsecretor" status
Norovirus infection:

O blood group > A or Bj
Secretor status >> nonsecretor

Vomiting when gastric emptying is slowed significantly

No age predilection

Gastroenteritis outbreaks- cruise ships, nursing homes, etc
Acute Gastroenteritis
- 50% of all foodborne outbreaks of gastroenteritis
-Most common etiology

Stool, vomit infectious
Outbreaks common
Many strains, little durable immunity
Dx: no routine tests
Rx: supportive
African Burkitt's Lymphoma
Young boys
Involves maxilla, periorbital region
Occurs in Equatorial Africa (not high altitude, desert)

Herpesvirions seen in tumor cell culture

Latent- no virions being produced
Lytic- virions being produced

Most Burkitt's cells are latently infected
Epstein-Barr Virus
Linked to human cancer- Burkitt's
EBV immortalizes B cells

Immortalized B cells:
Grow indefinitely in tissue culture
Tumorigenic in immunodeficient mice

EBNA1 viral protein in Burkitt's cells and immortalized B cell line
INVISIBLE to CD8+ T cells
(immort B cells- many other proteins!)
EBV latency- in Burkitt's cells; Immortalized B cells- No viral production

Lytic infection- many proteins, viral DNAP, viral thymidine kinase

Latent cells- genome= ds circles; require host DNAP for replication
AND
EBV copy nmbers not afffected by antivirals that inhibit DNAP (acyclovir)
Lytic cells- ds stranded linear molecules; requires viral DNAP for replication
AND
Production of new virions, production of linear DNA blocked by inhibitors of DNAP
Diagnosis of Infectious Mononucleosis
Sore throat
Swollen lymph nodes
Lymphocytosis
Heterophile antibody test~monospot test
EBV Infection and IM Dx
Transmitted in saliva
95% of adults infected
Infection in childhood usually asymptomatic
Infection in adolescence/adulthood assoc w/ syndrome of infectious mono

IM Dx: Monospot
IgM to VCA
IgG to VCA- indicates past

EBV transmission in saliva
Infects B lymphocytes
In lytic infection- spread via infectious virions
In latent infection- spread via cellular proliferation

T and NK response- atypical lymphocytosis characteristic of mononucleosis
Kills man virus infected B cells

After cellular response-
EBV infected resting memory B cell

Virus-infected cell can reactivate and produce virus
Virions infect new cells, which become T cell targets
Immunodeficiency and EBV
Organ Tx Recipients
Immunosuppression w/ cyclosporine, tacrolimus to prevent organ rejection
Develop B cell lymphoma, usually w/ EBV in each tumor cell
If immunosupp stopped- tumor can sometimes regress

Post-Tx: Many EBV B cells, few EBV-specific T cells

Congenital Immunodeficiency
-Severe combined immunodef- often die w/ EBV lymphoma
-X-linked immunodef- often die w/ EBV lymphoma (often boys)

AIDS Lymphoma
-Increased risk of lymphoma
-~50% EBV-associated lymphoma

X-linked agammaglobulinemia
-NO B cells
-NO EBV
-NO B cell lymphoma
**Protected**
EBV and other Tumor Associations
NP carcinomas virtually always EBV-associated, regardless of geo
Tumors are more common in Cantonese, other southern chinese ethnic groups
No evi of immunodeficiency

Hodgkin's Disease
EBV Ag Expression in 30%
-DNA detected; RNA detected; Antigen expresion
-Present at each tumor site, at presentation and relapse
Kaposi's Sarcoma
Children in Africa- involving feet, legs
Old men in Mediterranean
Organ tx recipients
AIDS pts- esp MSM

Most commonly involves skin (GI, lungs)
Neovascular proliferation
Purplish color from red cells in neovasculature

KSHV = HHV8
Detected by PCR in B lymphocytes in seropositive indiv
Does NOT immortalize B lymphocytes in vitro
Carries several genes close mimics of human genes ** IL6

Transmission- early childhood in endemic regions (saliva)
Sexual- MSM

Patho of KS:
KSHV infection required
Immunocompromise
Primary Effusion Lymphoma
B cells floating pleural or peritoneal fluid
Occurs in AIDS pts
Dually infected by EBV, KSHV
Hepatitis
Inflammation of the liver

Non-infectious causes
Alcohol
Acetomeniphen (Tylenol)
Halothane
Isoniazid (INH for TB)
Other meds
Shock, acute obstruction

Infectious Causes
Nonviral: Pneumococcal pneumonia, leptospirosis, sepsis, TB, histoplasmosis, ricketial infections, syphilis

Viral: CMV, EBV, HIV, adenovirus
Hepatitis Virus
Hepatitis Virus
5 viruses
A to E

HAV causes majority of acute viral hep in US

Transmission:
Fecal oral: HAV, HEV
Sexual: HBV, HDV (A,E,C less)
Blood: HBV, HDV, HCV (A,E less)
Perinatal: HBV, HDV (HCV>HAV,HEV)

B,C,D have envelope destroyed by bile- no fecal oral route possible

Symptoms more often w/ A, B
Persistence/Chronic infection w/ C

Healthy liver-Fibrosis- Cirrhosis- Macronodular Cirrhosis- Hepatocellular carcinoma (risk increases w/ persistence of HBV, HCV)

Lab Dx:
Transaminase: ALT, AST (>10-100X normal)
IgM antibody = acute infection, or past
Neutralizing antibodies = recovery
Viral particles = ongoing infection
(protein, nucleic acid)

Vaccine: HAV, HBV
Rx: Acute: none, prevent secondary cases
Chronic: HBV- INFa, laminvudine, adefovir, entecavir, tenofovir
HCV- INFa, ribavirin
HAV
IgM antibodies are markers of acute infection, typically detected for about 6 mo.

Picornaviridae, RNA virus
NO Envelope (bile stable)
Capsid proteins elicit universal neutralizing antibody

Vaccine: YES
HBV
Dx: When products of infectious agent or its nucleic acid detected, it implies ongoing infection (acute or chronic!) and infectivity

S gene and vaccination
Lg, Mid surface antigens
cccDNA and persistence:

Replication- RNA nuclear import- repair-> ccc = completely closed circular DNA (very stable in host nucleus, can integrate and remain as resevoir)
Virion # / Stability, transmission:
VERY transmissable

Genome: very small genome
S gene makes surface antigen
-3 proteins made
-5 specificities, a determinant that elicits neutralizing antibodies
-S gene put into yeast to make first recombinant vaccine
HDV
DEPENDENCY issues- needs HBV
Uses HBV's envelope as its envelope
HCV
HCV Replication
-Rapid turnover
-Error prone replication
*Genomic diversity
*Make mistake at every base, every day, in every person

Mutations- synonymous (silent) or nonsynonymous...
Neutral or Lethal or Nonlethal

Variants can evade adaptive immune system

HCV persists; infectious for decades

Persistence, 80%
Resistant to treatment
HCV vaccine dev
Reservoir
Antiviral drug dev.
HEV
Incubation period avg 40 days
Case-fatality:
PREGNANT WOMEN 15-25%
Illness severity increases w/ age
Vaccine
Any formulation able to elicit antigen specific protective immunological memory.

Classic- vaccine mediated protection based on exposure of an immunogenic agent to a host followed by natural development of immunity
Types of Vaccine formulation
Live Vaccines: Formulations where the antigens are encoded by a genetic material (live, attenuated vaccines) and synthesized IN HOST.
-Infectious agent
-Strong innate inflamm. responses (natural adjuvant)
-Strong induction of all (B&T) adaptive responses and longer memory

"INACTIVE" Vaccines: FOrmulations where the protein or polysaccharide antigens are directly injected into the host (innactivated vaccines, recombinant proteins, purified polysaccharides...)
-Local antigen deposit and dist. to regional lymph nodes
-Weak innate inflammatory response (requires addition of adjuvant)
-Mainly induce antibody responses (weak CD8 cytotoxic responses)
Essential components of vaccine
Appropriate presentation to immune system
(Route, B-cells, CD4 and CD8 T-cells)

Immune stimulatory signals able to start innate immune responses and shape the adaptive effector mechanisms
(Adjuvant)

Antigenic epitope correlated w/ protection: B-cell, linear and conformational: T-cell CD4 and CD8
(Active principle)

Approp. Presentation to imm. sys.

B-cells:
-Polysaccharides: CCD4-Independent antibodies (IgM)
-Proteins: CD4 dependent antibodies (IgG)

T cell:
CD4: protein processed in clas II pathway
CD8: protein expressed intracellular compartment and processed in class I pathway

Adjuvant in clinical use (for inact. and recomb vaccines)
Aluminum compounds
Basic mechanisms of vaccine action
VIRUS
Act. of immune system to produce antigen specific B cell responses (Antibodies)
-Neutralization- block bio. fxn of antigen
-Opsonization- accelerate clearance of antigen

Act of immune system to produce T-cell responses (CD4+ T-helper and CD8+ cytolitic)
-CD4+ cells- cytokine secretion to support B-cell and CD8+ cytolitic cell activation, proliferation, maturation and memory differentiation

Neutralizing antibodies target surface envelope glycoproteins

Cytotoxic T lymphocytes target cytoplasmic non-structural proteins
Protection: BACTERIA
Opsonizing antibodies facilitate phagocytosis; target surface polysaccharides and envelope glycoproteins

Anti-toxin antibodies neutralize toxins by targeting toxins
CD4+ T lymphocytes in HIV infection
CD4+ infection by HIV leads to cell death of infected and uninfected lymphocytes
CD4+ are central to immune response
Natural history of AIDS- loss of CD4+ cells in peripheral blood, lymphoid
CD4+ monocytes are targeted and traffic to tissues->specific tissue macs-> produced virus, cytokines

Normal CD4+ = 1000 cells/ul
AIDS < 200 cells/ul
Retroviruses
Spherical w/ lipid containing envelope (sensitive to detergent)
Dense nucleocapsid core w/ viral RNA genome and Reverse Transcriptase enzyme

Envelope:
Surface glycoprotein (gp120 HIV)
-highly glycosylated
mediates interaction btwn virus and rececptor
-contains epitopes for neutralizing antibodies and cytotoxic lymphocytes (CD8+)

Transmembrane glycoprotein (gp41 HIV)
-Anchors surface glycoprotein
-mediates virus induced cell fusion

Structure:
Nucleocapsid constructed by core protein p24 (HIV) (antibody to p24 used in ELISA assay for HIV infection, quantitative)
-2 copies of RNA genome, each with RT
-Positive strand RNA
-Genome: structural genees, viral proteins, proteins in viral envelope; pol gene- RT, RNase H, protease, integrase

Replication:
Virus Entry
Virus Fusion
Reverse Transcription
Integration of viral DNA
Txn of viral genes
Viral protein expression and virus assembly
Virus budding, Maturation
Virus Entry- HIV
gp120 molecule on virus binds CD4 on cell-> conformational change in protein allows gp120 to interact w/ co-receptor on cells surface- interaction of gp120 causes conform. change in gp41 exposing hydrophobic domain that mediates fusion btwn virus envelop and cell memrane
Virion loses envelope, viral core enters cell

Co-receptors = chemokine
Co-receptor for T-cell tropic HIV = CXCR4 on T-cells and PBMCs

Macrophage-tropic strains co-receptor = CCR5 on PBMCs and mac
Reverse Transcription- HIV
Synthesis of dsDNA from RNA

RTase and tRNA primer at 5' end
Reverse Txn- synthesis of dsDNA (RTase synthesizes - DNA copy
RNase H degrades viral RNA and RTase makes = DNA
ERROR PRONE
Integration of Proviral DNA- HIV
Nucleocapsid w/ dsDNA transported to nucleus
DNA integrated by integrase
Viral DNA replicated w/ cellular DNA

Can remain LATENT
Productive Replication & RNA Transcription
Assembly and Maturation
Cell contains approp. txn factors for virus to express viral mRNA and genomic mRNA

Viral DNA txn-ed into mRNA by cellular RNAPII
5' Capping
AAAA-Tail
Splicing

Processing of Env polyprotein by viral protease in golgi

Immature virion forms at membrane of cell
ONce budded, viral protease cleaves Gag and Gag-Pol-> mature virion (infectious)
HIV Transmission
Genital secretions
Blood highest conc'n

Congenital transmission during gestation , during birth, from breast milk
Clinical Consequences: Acute Infections- HIV
Enlarged painful lymphnodea, fever, rash, muscle ache, meningitis, lymphadenopathy, malaise

Productive replication in lymph nodes--> viremia and decline in CD4+
Acute infection-->elicits cellular & humoral response; virus in blood declines (killing by CD8+, NK, cell-mediated cytotoxicity. Neutralizing antibodies

Virus Load:
Measurement of virus by quantitating virion RNA by RT-PCR
Level of virus (set point) = prognostic indicator of disease progression

During clinically asymptomatic period, virus still present, CD4+ lymphocytes at normal level; virus replication in lymph nodes, spleen, and brain.
FDCs trap virus, present antigens--> cellular recruitment-->hyperplasia
Development of Disease- HIV
Over time, Steady decline in anti-viral immune responses, CD4+ lymphocytes,
INCREASE in viral load in blood

Lymph node destruction (complete in AIDS); Opportunistic infections

Susceptible, activated CD4+ lymphocytes continually recruited to lymphoid tissues, infected, and die
**CD4+ loss throughout course of disease

Problems:
Immune suppression
AIDS
AIDS encephalitis
AIDS dementia
Interstitial pneumonitis
Pathogenesis of AIDS
Depletion of CD4+ lymphocytes--> change in ratio of CD4/CD8
Exhaustion of stem cell pool

Functional CD4+ lymphocyte abnormalities (HIV eliminates CD4 on cells, which is essential for signaling)
HIV downreg. MHC class I expression on cells

Infected monocytes (latent) spread infection to lung, brain, and bone marrow. Activation into macrophages--> virus txn, production

AIDS is catastrophic failure of immune system
Treatment- HIV
Highly active anti-retroviral therapy HAART
Retrovirus Diversity
High rate of mutation
RNA viruses in general higher rate b/c no proofreading fxn
High level of viral diversity in population and in individual
Antigenic variation
HIV Drug Resistance
Selection or pre-existence of drug-resistant virus mutant =
short-lived effects of anti-viral drugs, difficult problem in HIV Rx design
HIV Latency
Long-lived population of latently infected resting CD4+ memory T lymphocytes in peripheral blood containing HIV.
Can reactivate when HAAR Rx stopped
Tissue reservoirs of CD4+ lymphocytes and macrophage.
Clinical Assays to Detect HIV
Evidence of infection- presence of antibodies

Presence of virus- culture or PCR

HIV serology- ELISA
add pts serum to plastic wall containing HIV proteins. If antibodies present, they bind, bound IgG detected w/ enzyme rxn

Positive result confirmed w/ Western Blot assay

Real-Time PCR- highly sensitive; detects virus in plasma and CSF
Can detect as little as 50 copies of HIV RNA in 1ml of plasma or CSF
Measures virus, does NOT depend on immune response to virus to develop antibodies (like ELISA and W. blot)
Why no HIV vaccine?
-Viral vaccines prevent disease caused by virus, not infection
-HIV vaccine should induce "sterilizing immunity" (prevent infection so no latently infected cells)
-Natural infection w/ HIV-1 or HIV-2 does not protective immune responses that can be used to develop vaccine
-HIV-1 exists as genetically diverse population of viruses- require multiple vaccine combos
-HIV-1 infection generates nique quasispecies in each infected individual
Common Features of Herpesviruses
Virus morphology
Viruses are ubiquitous
Infection is often asymptomatic
Common modes of replication
(virus lifecycles have general themes)
-ALL ESTABLISH LATENT INFECTIONS.
(reactivation can produce disease)

Genomes: HUGE! >100kb
Genes: HUGE! >50
-Enzymes
-structural genes
-Non-structural genes
Herpesvirus Infection "Modes"
Productive infection--> release of progeny virions "lytic infection"

Latent Infection:
-No virion production
-Acts as reservoir for recurrent disease
-Recurrent disease occurs as consequence of:
--renewed replication OR
--Cell proliferation (limited to tumor-inducing gamma herpesviruses only)
Lytic Infection v. Latent Infection
Lytic:
Viral envelope formed from NUCLEAR membrane

Gene expression in Lytic:
Immediate early: regulators of viral gene expression
Early: proteins required for genome replication
Late: virion structural proteins

Gene Expression in Latency:
RESTRICTED

Infected cell types:
Lytic: Many >2
Latent: Few; 1 or 2 (lymphocytes and neurons)
Herpesvirus Transmission
Natural modes- skin, mucous membranes
(HSV-1, -2) skin, genital tract
Secretions
-oral
-Respiratory tract (VZV)
Transplacental (CMV)
Iatrogenic Modes
-transfusin
-transplants (CMV)
General Concepts in Herpesvirus Disease
High disease severity
-Primary Infection
-Immune impairment

Low Disease Severity
-Recurrent infection
-Immune competent

Populations w/ severe infections:
-Immunodeficient
-Immunosuppressed
-Fetus/newborns
-Malnourished
-Burn victims
Herpes Simplex Virus Disease
Herpes labialis (lips)
Genital herpes

Primary site- productive infection of epithelial cells
-Retrograde transport-
Secondary site of infection and latent infection: **sensory neuron
-Reactivation, anterograde transport-
Site of recurrent infection: productive infection of epithelia cells

**Orolabial HSV- trigeminal ganglia
**Genital HSV- sacral ganglia

Common features of orolabial and genital herpes recurrence:
Reactivation can be symptomatic (blisters)
OR Asymptomatic
Genital Herpes
Correlates w/ # sexual partners
Women more susceptible
Transmission is commonly UNrecognized
Asymptomatic shedding = COMMON
Primary infections- often asymptomatic

Recurrence: Symptomatic recurrence after yrs of silent infection
Symptoms less severe than primary disease (shorter shedding time, fewer lesions)
Recurrence severity: HSV-2>HSV-1

Factors affecting recurrence:
Time since aquisition
Virus type
Immune status

Transmission:
Unknown- shedding rates have no impact on transmission

Antiviral Efficacy:
Acyclovir
Reduces recurrences
Reduces transmission
Other HSV Presentations
Primary gingivostomatitis (gums, oral cavity, lips)

Herpes Whitlow

HSV Keratitis- #1 cause of infectious blindness in developed world
2 pathogenic mechanisms: autoinoculation; trigeminal nerve opthalmic root infection after ganglion reactivation
Dendritic ulcers
Neonatal Herpes
Transmission through infected birth canal
-most women asymptomatic during labor

Presentation during 1st 1-2 wks of life

Three syndromes:
-skin, eye, mouth (SEM)
----usually not fatal, but recurs
----30% develop neurologic sequelae
-encephalitis
-disseminated

Occurrence of vesicles:
SEM > CNS > Disseminated

Cannot use absence of vesicles to rule out neonatal herpes!
Herpes Encephalitis
Most common acute sporadic encephalitis

Primarily HSV-1

Classic Presentation:
fever and focal neurologic deficits
-temporal lobe involvement
Varicella-zoster Virus (VZV)
Primary infection (Varicella)
-Chicken pox

Sever disease
-Teens/Adults at risk for varicella pneumonia
-Immunocompromised & newborns
----life threatening pneumonia
----encephalitis
----progressive, disseminated varicella

Herpes zoster (shingles)
-reactivation in sensory ganglion
-lesions localized to innervated dermatome
-can disseminate in immunocompromised pts
----- >2dermatomes
------ across midline
Cytomegalovirus
Infection usually asymptomatic
Disease can occur
-Mononucleosis
-Congenital infection
-Recipients of solid-organ, bone marrow Tx, leukemia/lymphoma, AIDS

CMV Mono:
79% EBV, 21% CMV
Frequent manifestation of primary CMV in young adult
Syndrome: fever, lymphadenopathy, lymphocytosis
-exudative pharyngitis freq. absent
Heterophil antiody negative

Congenital CMV:
Disease severity dependent upo maternal serostatus during preg.
-primary maternal infection: severe symptoms in 25% of births
-reactivation during preg.: usually asymptomatic at birth; late onset hearing loss
CMV in Immunocompromised Patients
HIV: reactivation and disease when CD4 < 50
-retinitis
-uncommon w/ HAART

Bone Marrow Transplant
-common presentation; pneumonia after marrow engraftment
-prophylaxis w/ ganciclovir

Solid Organ Transplant
-HUGE problem
-usually manifests as disease in allograft
-Highest risk cases: CMV seronegative recipient receiving organ from seropositive donor
HHV-6 and -7
Roseola infantum (exanthum subitum)- red cheek appearance in very young children
Febrile seizures common w/ roseola
May see infectious mononucleosis, hepatitis, neurologic syndromes
Transplant: fever, hepatitis, SKIN RASHES
Herpesvirus Diagnostics
Viral culture (HSV from all sites except CSF)
Rapid antigen detection: lesions (VZV at JHH)
Detection of nucleic acids: CNS disease, lesions (VZV at JHH)
Antibody detection assay: utility is limited their than for mononucleosis
useful in solid organ tx to id high risk pts
Live-replicating organism (Attenuated virus)
General: Attenuation of the pathogenic properties and preservation of immunogenicity is not always possible

Ex: Varicella Virus Vaccine (VARIVAX)

Formulation, Inoculation:
The virus is unstable
Requires stabilizers for viability

Active Principle (Antigen)
-Adapted in many different cell cultures

Immune stim signals (Adjvant)
-Org. naturally activae innate immune system; no additional adjuvant necessary

Presentation to immune system:
-Viral antigens produced by host and secreted w/ preserved conformational structure
-Antigens access to MHC class I and II processing and presentation
-Elicit good neutralizing antibodies, CD8, CD4 and memory responses
Live-replicating organism (Recombinant/Reassortment)
General: Attenuation of the pathogenic properties by reassortment of bovine and human Rotaviruses genes using reverse genetics

Example: Rotavirus vaccine, oral, pentavalent

Formulation/Inoculation:
Virus is unstable; requires stabilizers

Active principle (Antigen):
5 attenuated Rotavirus viral strains. Reassortants are propagated in Vero cells using cell culture tech.

Immune Stim:
Naturally activate innate immune system; no adjuvant required

Presentation to the immune system:
Viral antigens replicate in SI and secreted w/ preserved conformational structure
Antigens access to MHC class I and II processing and presentation
Elicit good neutralizing antibodies, CD8, CD4, and memory responses
Mucosal IgA responses
Inactive/non-replicating Whole organism
General: Require effective inactivation of pathogenic properties w/ preservation of immunogenicity

Ex: Hepatitis A vaccine (HAV)

Antigen Presentation: Intra-muscular injection

Active principle (Antigen)
HAV is a formalin inactivated whole virus vaccine derived from an attenuated hepatitis A virus (HAV) grown in cell culture in human fibroblasts

Immune Stim: Weak activation of innate immune systems; REQUIRE adjuvant (Aluminum hydroxyphosphate)

MHC Presentation: Antigens access to MHC II processing and presentation
Usually in multimeric antigenic form and induce good antibody responses
Does NOT produced biologically relevant CD8 responses
Elicits GOOD antibody memory response
Subunit- Polysaccharides
General: Require purification of antigen, and inactivation of toxic properties
Does not require epitope mapping

Ex: Miningococcal polysaccharide vaccine

Formulation/Inoculation: subcutaneous; still complex mixture, requires stabilization

Immune Stim: Stimulate innate immune system (TLR receptors)

Active principle: Purified polysaccharide extracted from freeze-dried preparation of Neisseria meningitidis cells and separated from media by purification procedures

Presentation to immune system:
CD4 independent B-cell activation
IgM production
Short-lived antibody memory response
Rapid decline in antibody titer
Subunit- Protein conjugated polysaccharides
General: Require purification of the antigen, and inactivation of toxic properties
Does NOT require epitope mapping

Ex: Conjugated meningococcal polysaccharide vaccine

Formulation/Inoculation: Subcutaneous injection; still complex mixture, and require stabilizers

Immune stim: Stimulate innate immune system (TLR receptors)

Active Principle: Purified polysaccharide extracted from freeze-dried prep of N. meningitidis cells and separated from media by purification processes
Diphtheria protein is purified and inactivated w/ formalin, polysaccharides are covalently linked to toxoid and purified

Presentation to immune system:
Fxnl cooperation btwn strong CD4 responses against diphtheeria toxoid and polysaccharides
CD4 dependent B-cell activation; IgG production
Longer lived antibody memory response
Subunit- Recombinant Protein
General: Protein antigen expression in genetically modified heterologous organism

Ex: Hepatitis B Vaccine

Formulation/Inoculation: IM

Immune Stim: NEEDS adjuvant aluminum hydroxide

Active Principle: Purified surface antigen of virus obtained by culturing genetically engineered Saccaromyces cervisiae cells, which carry surface antigen gene of Hep B virus. ANtigen purified and formulated as suspension of antigen adsorbed on aluminum hydroxide

Presentation to Immune System:
CD4 dependent B-cell act.; IgG production
Longer lived antibody memory response
Vaccine Use
Active Immunity:
Protection produced by person's own immune system
Long lasting

Passive Immunity: (can interfere with vaccination)
-Protection transferred from another person or animal as antibody
-Temp protection, wanes w/ time
-Passive immunity (transplacental IgG antibodies) passsed from mother to child interferees w/ vaccination.
Child vacc scheduled to avoid neutraliztion of vaccine by passive serum
Immunization during Pregnancy
Protects mother & infant
Transplacental antibodies transfer safer and less expensive then Ig therapy

Factors influencing antibody transfer to placenta:
Time between vaccination and delivery
IgG levels and subclass (IgG1=IgG3 >IgG4>IgG2)
IgM, IgA, IgE do NOT cross placenta
Gestational age (33 weeks, IgG maternal = IgG fetal; 40 weeks, IgG fetal>IgG maternal)

Maternal antibodies protect infant
Maternal antibodies neutralize live attenuated vaccines and influence immunization schedule
Immunological Differences in neonates
Immunological immaturity:
immature lymphoid organ arch. (rec immunization 8 weeks post-natal)

CD4+ helper T cel responses:
limited secretion of INF gamma

Innate:
limited T-cell independent responses
Impaired TLR3 and 9

Delayed maturation of dendritic cells
-limited IL-12 responses
Limited activation of CD4+ cells
Delayed and limited induction of germinal centers (low Ab responses)

Influences of maternal antibodies
-Epitope specific
-Does not affect T cell priming

Immunize ASAP and boost as needed
Immunological differences in Elderly
Cognate functions

Thymic regression
-reduced naive T cell population

CD4+ helper T cell responses
-Impaired TRC-MHC signaling

CD8+
Cellular Senescence

Innate
-Deregulation of Macrophage fxn
Impaired signaling and antigen presentation
HBV Antiviral:

Alpha-interferon
Potent inhibitor of HBV
Used to treat chronic active HBV

Mech: Activates Jak/STAT signal transduction pathway-->nuclear translocation of protein complex that interacts w/ genes containing IRES
Genes encode proteins that help cell resist viral infection by interfering w/ penetration, uncoating, RNA txn, translation, virus assembliy, release
**Inhibition of viral protein synthesis

Bioavailability: Poor orally, good IM or subcutaneous

Toxicity: Acute influenza-like symptoms (fever, fatigue, chills, headache, muscle aches), sometimes neuropsych probs, limit tolerance of long-term rx

REsistance: Tolerance to drug develops in most pts. HBV "terminal protein" interferes w/ action of interferons by blocking signal transduction
HBV Antiviral:

Lamivudine (3TC)
Nucleoside analog that inhibits HIV reverse transcriptase AND HBV RT b/c of similarities btwn enzymes

Mech: Cellular enzymes convert drug to triphosphate, which competitively inhibits HBV DNAP causing chain termination

Bioavailability: 80% absorbed orally
Toxicity: Negligible

Resistance: Many resistant strains have mut. in viral DNAP
Discontinuing drug --> rebound HBV viremia to pretreatment levels
14-32% of immunocompetent pts have resistance w/in 1 year, 50% w/i 3 yrs.
Influenza A, B, avian flu Antiviral:

Amantadine & Rimantadine
Mech: Primary effect at early infection; inhibits essential ion channel in virion envelope formed from viral protein M2.
M2 channel- mediates reduction pH in virion, essential for uncoating RNA genome.
Drug binds specific aa lining channel, preventing ion flow
Amantadine dec pH in golgi at late infection--> premature conformational change in viral hemagglutinin protein (HA), which deceases release of infectious particles

Bioavail: orally 50-90% for A; >90% for R

Resistance: Occurs RAPIDLY. 30% of treated people shed resistant virus w/in 5 days.
Drug resistance mutants retain fitness, but remain sensitive to Ribavirin (broad-spectrum, nucleoside analog toxic)
Influenza A, B, avia flu Antiviral

Zanamivir
Oseltamivir (Tamiflu)
Inhibitors of viral Neuraminidase (NA) which cleaves terminal sialic acids from glycoproteins, glycolipids, and proteoglycans
Cleaving sialic acid promotes effective viral spread in resp tract by preventing aggregation/attaching to sialic acid receptors on resp epithelium
Drugs active in prophylaxis and treatment

Mech: Competitive, reversible inhibitors of NA. Active site pocket of viral NA has binding sites for glycerol and carboxylate groups of sialic acid. Zanamivir is analog of sialic acid
Oseltamivir, prodrug of transition state analog of sialic acid

Minimal toxicity

Bio: poor for Zanamivir; given IV or aerosol spray; Good for Oseltamivir, given orally

Resistance: Development of resistance to Z and O is inefficient.
Herpesvirus Antivirals:

Acyclovir (ACV)
Mech: Inhibits DNA synthesis
Initial phosphorylation to ACV-P by viral TK (high affinity for ACV)
ACV-3PP by cellular kinase
ACV-3P competitively inhibits viral DNA polymerase.
Incorporated into DNA causing chain termination

Spectrum of activity: HSV-1, HSV-2, HZV

Bio: ACV 10-30% orally; Valacyclovir- higher oral availability

Tox: well tolerated, some nausea, diarrhea, rash, headache

Resistance: Leading cause of resist= mutation of viral TK, ability to phos. ACV reduced
Resistance from mut. in viral DNAP, decreases incorporation of ACV-3P into DNA
Herpesvirus Antiviral:

Ganciclovir
Analog of guanosine, activity against CMV

Mech: GCV converted to monophosphate by viral kinase; tri-phos by cellular kinase
Potent inhibitor of DNAP; inhibits incorp of GTP into DNA
GCV-3P not chain terminator, but inhibitory effect is due to inhibition of CMV DNA elongation

Bio: 10-20% orally

Tox: Bone marrow progenitor cells inhibited

Resistance: mutations in kinase (UL97 or CMV) making it unable to phos. GCV.
Herpesvirus Antiviral:

Foscarnet
Analog of pyrophosphate that is inhibitory for all herpesviruses and HIV.
Only nonnucleoside replication inhibitor for herpesviruses

Mech: reversibly blocks pyrophosphate binding site on DNAP; inhibits cleavage of pyrophos from dNTPs during DNA chain elong. Selective tox: viral DNAP more susceptible

Bio: 9-17% orally

Tox: Accumulatese in bone, causes kidney toxicity, so used for life-threatening infctions

Resistance: mutations in viral DNAP. Freq.works against ACV and GCV-resistance mutants
Herpesvirus Antiviral:

Fomivirsen
Phosphorothioate-based 21-mer anti-sense oligonucleotide that interferes w/ essential steps in CMV replictaion.

Mech: Anneals w/ "sense" strand of RNA encoding an essential major immediate-early viral protein whose fxn required for virus rep.

Bio: Requires injection into eyes

Toxicity: Produce indicated for local treatment for CMV retinitis in pts w/ AIDS who are intolerant of or have contraindication to other Rxs for CMV retinitis, or who failed other Rxs. Expensive

Resistance: Reported