Anti-Virals

  1. What are Viruses?
    viruses are obligate intracellular parasites, their replication depends on synthetic process of host cells.

    Antivirals:

    • -Block viral entry into or viral exit from the host cells
    • -Inhibit viral replication inside the host cell.
  2. What are the steps of Viral Replication?
    • 1. Adsorption to and penetration into susceptible host cells
    • 2. Uncoating of viral nucleic acid
    • 3. Synthesis of early regulatory proteins
    • 4. Synthesis of RNA or DNA
    • 5. Synthesis of late, stuctural proteins
    • 6. Assembly (maturation) of viral particles
    • 7. Release from the cell
  3. Diagram of steps Antivirals Inhibit:
  4. Drugs used to treat Herpes Simplex Virus (HSV) and Varicella Zoster Virus (VZV)?
    • Acyclovir
    • Valacyclovir
    • Famciclovir
    • Penciclovir
    • Trifluridine
    • Docosanol
  5. Drugs for treament of Cytomegalovirus (CMV)?
    • Ganciclovir
    • Valganciclovir
    • Cidofovir
    • Foscarnet
  6. Drugs for Anti-Hepatities therapy (Hep C and B)?
    • Lamivudine
    • Adefovir
    • Interferon Alfa
    • Pegylated Interferon Alfa
    • Ribavirin
    • Entecacir
    • Telbivudine
    • Tenofivir
    • Emtricitabine
  7. Drugs for Anti-influenza therapy?
    • Admantanes
    • -Amantadine, Rimantadine
    • Neuraminidase Inhibitors
    • -Zanamivir, Oseltamivir
  8. Drugs for AntiRetrovirus (HIV)?
    • Classes:
    • 1. Nucleoside Reverse Transcriptase Inhibitors (NRTIs)
    • 2. Nucleoside Researve Transcriptase Inhibitors
    • 3. NonNucleoside Reserve Transcriptase Inhibitors (NNRTIs)
    • 4. Protease Inhibitors
    • 5. Fusion Inhibitors
    • 6. CCR5 Antagonits
    • 7. Integrase Strand Transfer Inhibitors (InSTIs)
  9. What is Acyclovir?
    -Anti-HSV and anti-VZV drug, only one avaliable for IV use

    -acyclic guanosine derivative- competes with guanosine to bind at catalytic site, leads to DNA chain termination
  10. What is the Mechanism of Action for Acyclovir?
    Requires THREE phosphorylation steps for activation.

    FIRST converted into monophosphate by VIRAL THYMIDINE KINASE

    then, di- and triphosphate in host further phosphorylates to active form

    VIRAL KINASE is required for initial phosphorylation, acyclovir selectively activated and accumulates only in infected cells.

    • Acyclovir TRIphosphate inhibits viral DNA synthesis by:
    • 1. Competitive inhibition with deoxyGTP for viral DNA polymerase- binding to DNA template as trreversible complex
    • 2. Chain Termination following incorporation into viral DNA
  11. What are the resistance to Acyclovir?
    -Through ALTERATIONS (mutations) in Viral Thymidine Kinase or DNA Polymerase
  12. Which agents do NOT require activation by viral thymidine kinase and are ACTIVE against acyclovir-resistant strains?
    foscarnet, cidofivir, trifluridine
  13. What are the major toxicities of Acyclovir?
    nausea, diarrhea, headache
  14. Which agents are use to treat CMV infection?
    Ganciclovir, Cidofivir, Foscarnet
  15. What is Ganciclovir?
    • -Anti-CMV
    • - acyclic guanosine analog
  16. What is the MOA of Ganciclovir?
    -Requires triphosphorylation for activation

    -Initial phophorylation by viral protein kinase phosphotransferase UL97 in CMV-infected cells

    -Ganciclovir triphosphate competitively inhibits DNA ploymerase and causes termination of viral DNA elongation.
  17. What are the mechanisms of Resistance to Ganciclovir?
    -Mutations in UL97- decreased levels of active Ganciclovir triphosphate (these mutations are NOT cross-resistant to cidofivir and foscarnet)

    -Mutations in UL54- results in mutant DNA polymerase
  18. WHat are the major toxicities of Ganciclovir?
    Myelosuppression, neutropenia (more IV, than oral)

    Retinal detachment (CMV retinitis)
  19. What is Cidofivir?

    MOA?

    Toxicities?
    -cytosine nucleotide analog

    -phosphorylation to active diphosphate is INDEPENDENT of viral enzymes

    • -INHIBITOR and ALTERNATE substrate for viral DNA polymerase
    • -competitively inhibiting DNA synthesis and becomes incorporated into viral DNA chain

    • Toxicities: dose-dependent nephrotoxicity
    • -decreased intraocular pressure
  20. What is Foscarnet?

    MOA?

    Resistance?
    Toxicities?
    -inorganic pyrophosphate analog

    -inhibits viral DNA polymerase, RNA polymerase, and HIV reverse transcriptase DIRECTLY, without activation by phophorylation

    • -Resistance in HSV and CMV- point mutation in DNA polymerase gene (prolonged or repeated exposure to drug)
    • -Also Mutations in HIV-1 reverse transcriptase gene

    Toxicities: nephrotoxicity, ANemia, CNS Toxicity (headache, hallucinations, seizures)
  21. Importance of AntiRetroViral Therapy
    • -Monotherapy should be AVOIDED
    • -Need Maximal Potency to inhibit viral replication and AVOID premature development of resistance
    • -Combination of drugs (HAART)- highly active antiretroviral therapy--effectivly reduced plasma HIV RNA levels and increase CD4 cells

    -Monitor Viral Load, CD4 cell counts, Drug Resistance Testing
  22. What are NRTIs?
    Nucleoside Reverse Transcriptase Inhibitors

    -binds to DNA polymerase catalytic site of reverse transcriptase enzyme

    • -competitively inhibits DNA polymerase activity
    • -can also be incorporated into viral DNA chain and cause termination
    • -requires activation by phosphorylation to triphosphate (uses human cell kinases)

    • -Resistance by viral mutations
    • -Cross resistance exist amont NRTIs

    -Major Toxicities: Lactic Acidosis, hepatomegaly
  23. How many catalytic sites does Reverse Transcriptase have?
    Reverse Transcriptase enzyme has two active, catalytic sites

    • -RNase H (RNA cleaving) actice site
    • -DNA polymerase (DNA synthesizing) active site

    BOTH sites needed to reverse-transcribe HIV's RNA based genomic material into DNA
  24. What is Zidovudine (Azidothymidine, AZT)?
    • NRTI
    • -deoxythymidine analog (first antiretroviral)

    • -decrease rate of clinical disease progression, prolong HIV infected survival
    • -When administered during pregancy, during labor, AND to neonate, REDUCED rate of VERTICAL (mom to baby) transmission of HIV

    • -Resistance may limit clinical efficacy
    • -high resistance due to three or more mutations

    Toxicities: Myelosuppression (anemia neutropenia)
  25. What are Nucleotide Reserve Transcriptase Inhibitors?
    Example: Tenofovir

    -acyclic nucleoside phosphonate (nucleotide) analog of adenosine

    -Req. activation by phosphorylation to triphosphate (catalyzed by human kinases)

    -Tenofovir competitively inhibits DNA polymerase activity of the HIV reverse transcriptase enzyme, causes chain TERMINATION (once incorporated into viral DNA)

    Toxicities: GI upset, lactic acidosis, hepatomegaly
  26. Is there Cross Resistance between Tenofovir and NRTIs?
    Yes, b/c they share a similar binding site
  27. What is Tenofovir?
    a Nucleotide Reverse Transcriptase Inhibitor
  28. What are NNRTIs?
    • NonNucleoside Reverse Transcriptase Inhibitors
    • -Bind directly to HIV-1 reverse transcriptase
    • -Inhibition of RNA and DNA dependent DNA polymerase activities
  29. Are binding sites of NNRTIs close to NRTIs?
    Binding sites for NNRTIs are NEAR but DIFFERENT from binding sites of NRTIs.

    Binding of the NNRTIs blocks the DNA Polymerase activity by disrupting the DNA polymerase catalytic site of the enzyme

    Unlike NRTIs, NNRTIs DO NOT compete with nucleoside triphosphates and DO NOT req. phosphorylation to be active

    NO cross resistance between NNRTIs and NRTIs or the protease inhibitors
  30. Resistance to NNRTIs?
    Resistance to NNRTI is rapid with monotherapy and associated with viral mutations
  31. What is Nevirapine?
    • Nevirapine is an NNRTI
    • -used in combination with other antiretroviral regimens
    • -Severe/life threatening skin rashes, toxic epidermal necrolysis have occured, discontinue
    • -hepatitis may occur within first 6 weeks of initial therapy, monitor liver function tests

    • -Metabolized by CYP3A4 in liver (substrate)
    • -ALSO is an INDUCER of CYP3A4 metabolism (drug interactions)
  32. What is the purpose of the protease enzyme?
    In later stages of HIV growth cycle, the Gag and Gag-Pol genes are translated into poluproteins, then become immature budding particles.

    Proteases are responsible for cleaving these precursor molecules to produce final sturctural proteins of MATURE virion core.
  33. What are Protease Inhibitors?
    -prevents clevage of Gag-Pol polyprotein by inhibiting the protease enzyme.

    Results in production of IMMATURE noninfectious viral particles.
  34. Resistance of Protease Inhibitors?

    Cross Resistance?
    Specific genotypic mutations confer viral resistance-common

    Monotherapy is contraindicated

    Cross resistance exists, requires minimum of FOUR subsctitutions in gene.
  35. What is a syndrome that is a major toxicity concern with using Protease Inhibitors?
    • -Syndrome of redistribution and accumulation of body fat, Lipodystrophy
    • -increases in triglycerides, LDL levels, glucose intolerance and insulin resistance
    • -abnormalities appear to associate with use of protease inhibitos, cause yet not known
  36. What has been approved by the FDA to treat Lipodystrophy in patients on antiretroviral therapy?
    Tesmorelin, a growth hormone-releasing factor, once daily injection
  37. In Liver, are Protease Inhibitors inducers, inhibitors, or subtrates of CYP3A4 enzyme?
    All protease inhibitors are SUBSTRATES (metabolized by CYP3A4) and INHIBITORS (cause dec. clearance and increased plasma levels of other substraes drugs)

    -SHOULD NOT be administered with other agents that are metabolized by CYP3A4.
  38. What is Ritonavir?
    • Ritonavir is a Protease Inhibitor.
    • -inhibitor of HIV-1 and HIV-2 proteases.

    -Ritonavir is an inhibitor of CYP3A4
  39. What is special about concurrent administration of Ritonavir with other Protease Inhibitors?
    -results in INC plasma levels of these agents (enhanced activity of other protease inhibitor-CYP3A4)

    -Pharmacokinetic interactions, exploited to permit more convenient dosing (patient compliance), improve tolerability, and enhance efficacy.

    EX: Saquinavire and Ritonavir, lopinavir/ritonavir, tipranavir/ritonavir
  40. What can Saquinavir be used in combination with to decrease frequency of dosing, improved antiviral efficacy, and decreased GI side effects of Saquinavir therapy?
    Saquinavir with low (subtherapeutic) dose of Ritonavir.

    The sub therapeutic dose of ritonavir inhibits the CYP3A4-meadiated metabolism of saquinavir, results in increased exposure of saquinavir.

    High saquinavir plasma levels maintain potent viral suppression, and provide pharmacologic barrier to resistance.
  41. What is an example of a Fusion Inhibitor?

    MOA?

    Toxicities?
    Enfuvirtide, peptide, subcutaneously, in combination for HIV-1 therapy

    • -BLOCKS entry of virus into cell
    • -binds to gp41 subunit of viral envelope glycoprotein, preventing conformational changes req. for fusion

    -Resistance can occur- being investigated. NO cross resistance between enfuvirtide and other antiretrovirals

    Toxicities: local injection site reations, hypersensitivity reactions
  42. What are the co-receptors on human cells that HIV may use to enter human CD4 cells?
    • Chemokine Receptor 5 (CCR5)- in early disease
    • Chemokine Receptor 4 (CXCR4) - more advanced disease
  43. What is Maraviroc? What does it target?
    • -new anti-HIV drug, CCR5 Antagonist
    • -used in combination to treat CCR5-tropic HIV-1- detectable disease

    • -targets host CD4 T cells
    • -binds to and BLOCKS CCR5 on CD4 cells
    • -BLOCKS the ENTRY of HIV into CD4 cells, prevents infection

    • -ONLY effective in patients with CCR5-tropic disease
    • -INeffective against HIV that uses both(dual/mixed) or CXCR4 for cell entry.
  44. Patients must undergo what in order to be treated with Maraviroc?
    • -screening test to determine TROPISM of HIV infection.
    • -TROFILE, may fail to detect presence of small CXCR4-tropic virus (thus maravoric would fail)

    -Maraviroc treats CCR5-tropic HIV infection ONLY
  45. What are the toxicities of Maraviroc?
    • -hepatotoxicity (boxed warning),
    • -systemic allergic reactions
    • -cardiovascular events- myocardial ischemia/infacrtion, postural hypotension--caution in patients with inc. risk of cardiovascular disease
    • -may INC risk of infections and malignancy
  46. What is Raltegravir?
    HIV-1 integrase strand transfer inhibitors (InSTIs)

    -used in combination therapy for patients with multiple antiretroviral resistance
  47. What is the Mechanism of Action for Raltegravir?
    -inhibits integrase enzyme, preventing viral DNA from integrating with cellular DNA

    -avtive against HIV strains that are resistant to other antiretroviral drugs
  48. What are HIV-1 integrase enzymes?
    enzymes that catalyze important steps in the process of inserting viral DNA into the host cell genome.
  49. Viral resistance to Raltegravir?
    (InSTIs)

    Resistance occurs due to mutations in the integrase enzymes
  50. Major toxicities with Raltegravir?
    Increase in serum creatine kinase, myopathy, and rhabdomyolysis- cause/effect not clear
  51. Can HBV and HCV be cured?
    no, anti-hepatitis therapy is suppressive, not curative.

    -prevalence of HBV/HCV high worldwide.
  52. What is Adefovir?
    • -treats HBV
    • -suppress HBV replication and improve liver histology and fibrosis
    • -HBV DNA appears after cessation of therapy
  53. What is the Mechanism of Action for Adefovir?
    -nucleotide analog of adenosine monophosphate (similar to tenofovir)

    -phosphorylated by cellular kinases to active diphosphate metabolite

    -competitively inhibits HBV DNA polymerase, results in chain termination after incorporation into viral DNA
  54. What are they toxicities associated with Adefovir?
    -dose dependent nephrotoxicity, INC risk in patients with preexisting renal dysfunction/those treated for longer duration

    -Lactic acidosis, hepatomegaly (similar to NRTIs)
  55. What is Interferon Alpha?
    • -treats HBV and HCV
    • -administered SC or IM

    -interferon alphas are cytokines, endogenous protiens, that exert complex antiviral, immunodulatory, and antiproliferative activities invovling synthesis of RNA and proteins.
  56. What is the difference between Pegylated and regular Interferon Alpha?
    Pegylated interferon alpha has a linear/branched polyethylene gycol (PEG) attached by a covalent bond.

    pegylated- have longer half-lives, slower clearance, steadier serum concentrations, less frequent dosing
  57. What is the Mechanism of Action for Interferon Alphas?
    Bind to specific membrance receptors on the cell surface

    • initiates a series of intracellular events:
    • -enzyme induction
    • -suppression of cell proliferation
    • -immunomodulatory activities
    • -inhibition of viral replication

    • -Activate JAK-STAT signal transduction pathway, leads to nuclear translocation of cell protein complex that binds to genes containing IFN-specific responses
    • -Synthesis of over 24 antiviral proteins- works at different stages of viral penetration

    -Modifies immune response to viral infections
  58. What is a major effect of IFN-induced antiviral proteins?
    Inhibition of viral protein synthesis for many viruses.

    A given virus may be inhibitied at several steps, and the perincipal inhibitory effect differs among virus families
  59. How can viruses resist the antiviral effects of IFNs?
    -Blocking production or Activity of selected IFN-inducible antiviral proteins
  60. how does IFNs modify the immune response to viral infections?
    IFN-induced expression of MHC antigens, by enhancing the lytic effects of cytotoxic T-lymphocytes.

    IFNs may mediate some of the systemic symptoms associated with viral infections and contribute totissue damage in certain viral diseases.
  61. What are some commone side effects or toxicities associated with Interferon Alpha therapy?
    Side effects: flu like syndrome within 6 hrs after dosing, resolve upon cont. therapy

    Thrombocytopenia, hypotension, neuropsychiatric side effects.

    Contraindications: phychosis, severe depression, symptomatic heart disease, uncontrolled seizures.

    Interferons are ABORTIFACIENT
  62. What is Ribavirin used for?
    combination with IFN alpha to treat HCV infections

    • -guanosine analong, requires intracell. activation
    • -phosphorylated by host enzymes into active triphosphate
  63. What is the Mechanism of Action for Ribavirin?
    -not fully understood

    • 1. Interferes with synthesis of guanosine triphosphate
    • 2. Inhibits capping of viral mRNA
    • 3. Inhibits viral RNA-dependent RNA polumerase
  64. What are the toxicities of Ribavirin?
    dose dependent hemolytic anemia and depression

    contraindicated during pregnancy (teratogenic and mutagenic)
  65. What are the two classes used to treat the flu?
    Admantanes (amantadine, Rimantadine)

    Neuraminidase Inhibitors (Zanamivir, Oseltamivir)
  66. What are Amantadine and Rimantadine?
    anti-influenza therapy

    • Rimantadine is a derivative of Amantadine.
    • -cyclic amines
    • -rimantadine more active than amantadine

    -Resistance of influenza A virus to both agents have INC in recent years
  67. What is the Mechanism of Action for Amantadine and Rimantadine?
    • -inhibit uncoating of viral RNA of influenza A, prevents replication
    • (In some influenza A strains, also inhibit viral assmebly)

    • -Primary target, M2 Protein on viral membrane (highly specific against influenza A)
    • -binds and inhibits M2 Protein, inhibits acid-mediated dissociation of viral ribonucleoprotein complex early in viral replication (inhibits viral uncoating) and alter hemagglutinin processing later in replication- conformational changes- (inhibit viral assembly)
  68. Resistance of Amantadine and rimantadine?
    • Resistance due to mutations, rapidly develops, transmission of resistance virus to household contacts
    • -Both drugs share cross-suscept and cross resistance

    Cross resistance to Zanamivir and Oseltamivir DOES NOT occur.
  69. Toxicities with Amantadine and Rimantadine?
    CNS toxicity- less frequent with Rimantadine

    Serious neurotoxic reactions- with high Amantadine plasma levels (1-5 ug/mL)

    Amantadine is teratogenic
  70. What is different about the Adamantanes and the Neuraminidase Inhibitors?
    Neuraminidase (Zanamivir and Oseltamivir) are active against BOTH influenza A and B

    Adamantanes- only influenza A
  71. Zanamivir vs. Oseltamivir
    both neuraminidase inhibitors

    • zanamivir- oral inhalation, IV
    • Oseltamivir- oral (prodrug), IV

    -both for prophylaxis or treatment of acute uncomplicated influenza infection

    • --Administrator 36-48 hrs after onset of symptoms for 5 days, both agents shortens severity and duration of illness and MAY decrease incidence of respiratory complications
    • (the earlier to start, the more effective in treating influenza)

    -hospitalized patients- both agents reduce duration of hospitalization and risk of death. May also DEC viral shedding and risk of spread to contact (even after 48 hrs after onset)

    • -both are 70-90% effectivie prophylaxis after exposure to influenza A or B
    • -Give within 48 hrs after household exposure or close contact with infected person
  72. Resistance to Zanamivir and Oseltamivir?
    Resistance reported in Influenza A and B strains
  73. Does antiinfluenza therapy reduce the effects of the flu vaccine?
    Antivirals may interfee with the efficacy of the live-attenuated intranasal flu vaccine (Flumist). stop 48 hours before, and atleast 2 weeks after FluMist administration.

    Inactivated flu vaccines are NOT affected by viral neuraminidase inhibitors (Flu shot)
  74. What is the Mechanism of Action for Zanamivir and Oseltamivir?
    -inhibitors of vial neuraminidase enzymes, inhibis viral release/infection

    [Neuraminidase is essential viral glycoprotein for viral release]

    Resistance (oseltamivir) associated with mutation in viral neuraminidase
  75. What are some Toxicities associated with Zanamivir and Oseltamivir?
    Zanamivir- cough, nasal/throat irritations, bronchospasm, NOT for patients with asthma or COPD

    Oseltamivir- headache, nausea, vomiting, take with food to reduce GI effects

    Neuropsychiatric events with Oseltamivir, cause not established
  76. What is Peramivir?
    -FDA 'emergency use authorization' for neuraminidase inhibitor, Peramivir

    -Peramivir, given IV- still investagational drug- inhibits viral neuraminidase

    -FDA authroized use in hospitals for confirmed or suspected pandemic 2009 H1N1 influenza infection who cannot take or are not responding to treatment with Oseltamivir or Zanamivir

    -Many isolates resistant to Oseltamivir, ALSO resistant to Peramivir
  77. How to determine which strains are susceptible to which drugs?
    Susceptibility varies from one strain to another, from one flu season to another.

    strains evolve rapidly, treatment recommendations change during flu seasons

    www.cdc.gov/flu- provides updated info on viral resistance/susceptibility
Author
seyang
ID
69181
Card Set
Anti-Virals
Description
Antiviral Drugs
Updated