147
Views
0
CrossRef citations to date
0
Altmetric
Review

Highly Resistant HIV-1 Proteases and Strategies for Their Inhibition

, &
Pages 1023-1038 | Published online: 11 Jun 2015

References

  • Deeks SG , LewinSR, HavlirD V. The end of AIDS: HIV infection as a chronic disease. Lancet382 (9903), 1525–1533 (2013).
  • Passaes CP , Sáez-CiriónA. HIV cure research: advances and prospects. Virology454–455, 340–352 (2014).
  • Clifford DB , AncesBM. HIV-associated neurocognitive disorder. Lancet Infect. Dis.13 (11), 976–986 (2013).
  • Weber IT , WangY-F. HIV protease: role in viral replication, protein-ligand X-ray crystal structures and inhibitor design. In : Aspartic Proteases as Therapeutic Targets. Methods and Principles in Medicinal Chemistry. GhoshAK ( Ed). Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim, Germany, 45, 109–137 (2010).
  • Jacobsen H , YasargilK, WinslowDLet al. Characterization of Human Immunodeficiency Virus type 1 mutants with decreased sensitivity to proteinase inhibitor Ro 31–8959. Virology206 (1), 527–534 (1995).
  • Lloyd SB , KentSJ, WinnallWR. The high cost of fidelity. AIDS Res. Hum. Retroviruses30 (1), 8–16 (2014).
  • Paterson DL , SwindellsS, MohrJet al. Adherence to protease inhibitor therapy and outcomes in patients with HIV infection. Ann. Intern. Med.133 (1), 21–30 (2000).
  • Nolan D , ReissP, MallalS. Adverse effects of antiretroviral therapy for HIV infection: a review of selected topics. Expert Opin. Drug Saf.4 (2), 201–218 (2005).
  • Yasuda JM , MillerC, CurrierJSet al. The correlation between plasma concentrations of protease inhibitors, medication adherence and virological outcome in HIV-infected patients. Antivir. Ther.9 (5), 753–761 (2004).
  • Harrigan PR , HoggRS, DongWWYet al. Predictors of HIV drug-resistance mutations in a large antiretroviral-naive cohort initiating triple antiretroviral therapy. J. Infect. Dis.191 (3), 339–347 (2005).
  • Pham QD , WilsonDP, LawMGet al. Global burden of transmitted HIV drug resistance and HIV-exposure categories. AIDS28 (18), 2751–2762 (2014).
  • Bonura F , TramutoF, VitaleFet al. Transmission of drug-resistant HIV type 1 strains in HAART-naive patients: a 5 year retrospective study in Sicily, Italy. AIDS Res. Hum. Retroviruses26 (9), 961–965 (2010).
  • Tramuto F , BonuraF, MancusoSet al. Detection of a new 3-base pair insertion mutation in the protease gene of Human Immunodeficiency Virus type 1 during highly active antiretroviral therapy (HAART). AIDS Res. Hum. Retroviruses21 (5), 420–423 (2005).
  • Kozísek M , SaskováKG, RezácováPet al. Ninety-nine is not enough: molecular characterization of inhibitor-resistant Human Immunodeficiency Virus type 1 protease mutants with insertions in the flap region. J. Virol.82 (12), 5869–5878 (2008).
  • Pereira-Vaz J , DuqueV, TrindadeLet al. Detection of the protease codon 35 amino acid insertion in sequences from treatment-naïve HIV-1 subtype C infected individuals in the Central Region of Portugal. J. Clin. Virol.46 (2), 169–172 (2009).
  • Shafer RW , SchapiroJM. HIV-1 drug resistance mutations: an updated framework for the second decade of HAART. AIDS Rev.10 (2), 67–84 (2008).
  • Neogi U , RaoSD, BontellIet al. Novel tetra-peptide insertion in Gag-p6 ALIX-binding motif in HIV-1 subtype C associated with protease inhibitor failure in Indian patients. AIDS28 (15), 2319–2322 (2014).
  • Clavel F , MammanoF. Role of Gag in HIV resistance to protease inhibitors. Viruses2 (7), 1411–1426 (2010).
  • Kozísek M , HenkeS, SaskováKGet al. Mutations in HIV-1 Gag and Pol compensate for the loss of viral fitness caused by a highly mutated protease. Antimicrob. Agents Chemother.56 (8), 4320–4330 (2012).
  • Nijhuis M , van MaarseveenNM, LastereSet al. A novel substrate-based HIV-1 protease inhibitor drug resistance mechanism. PLoS Med.4 (1), e36 (2007).
  • Wensing AM , CalvezV, GünthardHFet al. 2014 Update of the drug resistance mutations in HIV-1. Top. Antivir. Med.22 (3), 642–650 (2014).
  • Hirsch MS , GünthardHF, SchapiroJMet al. Antiretroviral drug resistance testing in adult HIV-1 infection: 2008 recommendations of an International AIDS Society-USA panel. Clin. Infect. Dis.47 (2), 266–285 (2008).
  • Friend J , ParkinN, LieglerTet al. Isolated lopinavir resistance after virological rebound of a ritonavir/lopinavir-based regimen. AIDS18 (14), 1965–1966 (2004).
  • Mo H , KingMS, KingKet al. Selection of resistance in protease inhibitor-experienced, Human Immunodeficiency Virus type 1-infected subjects failing lopinavir- and ritonavir-based therapy: mutation patterns and baseline correlates. J. Virol.79 (6), 3329–3338 (2005).
  • Kagan RM , ShenderovichMD, HeseltinePNR, RamnarayanK. Structural analysis of an HIV-1 protease I47A mutant resistant to the protease inhibitor lopinavir. Protein Sci.14 (7), 1870–1878 (2005).
  • Young TP , ParkinNT, StawiskiEet al. Prevalence, mutation patterns, and effects on protease inhibitor susceptibility of the L76V mutation in HIV-1 protease. Antimicrob. Agents Chemother.54 (11), 4903–4906 (2010).
  • Babrzadeh F , VargheseV, PacoldMet al. Collinearity of protease mutations in HIV-1 samples with high-level protease inhibitor class resistance. J. Antimicrob. Chemother.68 (2), 414–418 (2013).
  • Doherty KM , NakkaP, KingBMet al. A multifaceted analysis of HIV-1 protease multidrug resistance phenotypes. BMC Bioinformatics12 (1), 477 (2011).
  • Varghese V , MitsuyaY, FesselWJet al. Prototypical recombinant multi-protease inhibitor resistant infectious molecular clones of Human Immunodeficiency Virus type-1. Antimicrob. Agents Chemother.57 (9), 4290–4299 (2013).
  • Weber IT , AgniswamyJ. HIV-1 protease: structural perspectives on drug resistance. Viruses1 (3), 1110–1136 (2009).
  • Goldfarb NE , OhanessianM, BiswasSet al. Defective hydrophobic sliding mechanism and active Site expansion in HIV-1 protease drug resistant variant Gly48Thr/Leu89Met: mechanisms for the loss of saquinavir binding potency. Biochemistry54 (2), 422–433 (2014).
  • Hayashi H , TakamuneN, NirasawaTet al. Dimerization of HIV-1 protease occurs through two steps relating to the mechanism of protease dimerization inhibition by DRV. Proc. Natl Acad. Sci. USA111 (33), 12234–12239 (2014).
  • Louis JM , ZhangY, SayerJMet al. The L76V drug resistance mutation decreases the dimer stability and rate of autoprocessing of HIV-1 protease by reducing internal hydrophobic contacts. Biochemistry50 (21), 4786–4795 (2011).
  • Ragland DA , NalivaikaEA, NalamMNLet al. Drug resistance conferred by mutations outside the active site through alterations in the dynamic and structural ensemble of HIV-1 protease. J. Am. Chem. Soc.136 (34), 11956–11963 (2014).
  • Chang YC , XuY, ZhangYet al. Potent antiviral HIV-1 protease inhibitor GRL-02031 adapts to the structures of drug resistant mutants with its P1’-pyrrolidinone ring. J. Med. Chem.55 (7), 3387–3397 (2012).
  • Ohtaka H , SchönA, FreireE. Multidrug resistance to HIV-1 protease inhibition requires cooperative coupling between distal mutations. Biochemistry42 (46), 13659–13666 (2003).
  • Wang Y , LiuZ, BrunzelleJSet al. The higher barrier of darunavir and tipranavir resistance for HIV-1 protease. Biochem. Biophys. Res. Commun.412 (4), 737–742 (2011).
  • Martin P , VickreyJF, ProteasaGet al. “Wide-open” 1.3 A structure of a multidrug-resistant HIV-1 protease as a drug target. Structure13 (12), 1887–1895 (2005).
  • Yedidi RS , ProteasaG, MartinezJLet al. Contribution of the 80s loop of HIV-1 protease to the multidrug-resistance mechanism: crystallographic study of MDR769 HIV-1 protease variants. Acta Crystallogr. D. Biol. Crystallogr.67 (6), 524–532 (2011).
  • Sasková KG , KozísekM, RezácováPet al. Molecular characterization of clinical isolates of Human Immunodeficiency Virus resistant to the protease inhibitor DRV. J. Virol.83 (17), 8810–8818 (2009).
  • Kožíšek M , LepšíkM, Grantz ŠaškováKet al. Thermodynamic and structural analysis of HIVto DRV – analysis of heavily mutated patient-derived HIV-1 proteases. FEBS J.281 (7), 1834–1847 (2014).
  • Louis JM , AnianaA, WeberIT, SayerJM. Inhibition of autoprocessing of natural variants and multidrug resistant mutant precursors of HIV-1 protease by clinical inhibitors. Proc. Natl Acad. Sci. USA108 (22), 9072–9077 (2011).
  • Louis JM , TözsérJ, RocheJet al. Enhanced stability of monomer fold correlates with extreme drug resistance of HIV-1 protease. Biochemistry52 (43), 7678–7688 (2013).
  • Agniswamy J , ShenC-H, WangY-Fet al. Extreme multidrug resistant HIV-1 protease with 20 mutations is resistant to novel protease inhibitors with P1’-pyrrolidinone or P2-tris-tetrahydrofuran. J. Med. Chem.56 (10), 4017–4027 (2013).
  • Agniswamy J , ShenC-H, AnianaAet al. HIV-1 protease with 20 mutations exhibits extreme resistance to clinical inhibitors through coordinated structural rearrangements. Biochemistry51 (13), 2819–2828 (2012).
  • Koh Y , AmanoM, TowataTet al. In vitro selection of highly DRV-resistant and replication-competent HIV-1 variants by using a mixture of clinical HIV-1 isolates resistant to multiple conventional protease inhibitors. J. Virol.84 (22), 11961–11969 (2010).
  • Zhang Y , ChangY-CE, LouisJMet al. Structures of DRV-resistant HIV-1 protease mutant reveal atypical binding of DRV to wide open flaps. ACS Chem. Biol.9 (6), 1351–1358 (2014).
  • De Vera IM , SmithAN, DancelMCet al. Elucidating a relationship between conformational sampling and drug resistance in HIV-1 protease. Biochemistry52 (19), 3278–3288 (2013).
  • Roche J , LouisJM, BaxA. Conformation of inhibitor-free HIV-1 protease derived from NMR spectroscopy in a weakly oriented solution. Chembiochem16 (2), 214–218 (2014).
  • Weber IT , ZhangY, TozserJ. HIV-1 Protease and AIDS therapy. In : Proteases In Biology And Disease, Volume 8. Viral Proteases And Antiviral Protease Inhibitor Therapy. LendeckelU, HooperN ( Eds), Springer, NY, USA, 25–46 (2009).
  • Velthuisen EJ , BaughmanTM, JohnsBAet al. Synthesis and pharmacokinetic profile of highly deuterated brecanavir analogs. Eur. J. Med. Chem.63, 202–212 (2013).
  • Ghosh AK , AndersonDD, WeberIT, MitsuyaH. Enhancing protein backbone binding – a fruitful concept for combating drug-resistant HIV. Angew. Chem. Int. Ed. Engl.51 (8), 1778–1802 (2012).
  • Orkin C , DejesusE, KhanlouHet al. Final 192-week efficacy and safety of once-daily DRV/ritonavir compared with lopinavir/ritonavir in HIV-1-infected treatment-naïve patients in the ARTEMIS trial. HIV Med.14 (1), 49–59 (2013).
  • De Meyer S , AzijnH, SurlerauxDet al. TMC114, a novel Human Immunodeficiency Virus type 1 protease inhibitor active against protease inhibitor resistant viruses, including a broad range of clinical isolates. Antimicrob. Agents Chemother.49 (6), 2314–2321 (2005).
  • Brower ET , BachaUM, KawasakiY, FreireE. Inhibition of HIV-2 protease by HIV-1 protease inhibitors in clinical use. Chem. Biol. Drug Des.71 (4), 298–305 (2008).
  • Koh Y , MatsumiS, DasDet al. Potent inhibition of HIV-1 replication by novel non-peptidyl small molecule inhibitors of protease dimerization. J. Biol. Chem.282 (39), 28709–28720 (2007).
  • Ghosh AK , XuCX, RaoKVet al. Probing multidrug resistance/protein-ligand interaction with new oxatricyclic designed ligands in HIV-1 protease inhibitors. ChemMedChem5 (11), 1850–1854 (2010).
  • Amano M , TojoY, Salcedo-GómezPMet al. GRL-0519, a novel oxatricyclic ligand-containing nonpeptidic HIV-1 protease inhibitor (PI), potently suppresses replication of a wide spectrum of multi-PI-resistant HIV-1 variants in vitro. Antimicrob. Agents Chemother.57 (5), 2036–2046 (2013).
  • Zhang H , WangYF, ShenCHet al. Novel P2 tris-tetrahydrofuran group in antiviral compound 1 (GRL-0519) fills the S2 binding pocket of selected mutants of HIV-1 protease. J Med Chem.56 (3), 1074–1083 (2013).
  • Ghosh AK , MartyrCD, SteffeyMet al. Design, synthesis, and X-ray structure of substituted bis-tetrahydrofuran (bis-THF)-derived potent HIV-1 protease inhibitors. ACS Med. Chem. Lett.2 (4), 298–302 (2011).
  • Filler R , SahaR. Fluorine in medicinal chemistry: a century of progress and a 60 year retrospective of selected highlights. Future Med. Chem.1 (5), 777–791 (2009).
  • Salcedo Gómez PM , AmanoM, YashchukSet al. GRL-04810 and GRL-05010, difluoride-containing nonpeptidic HIV-1 protease inhibitors (PIs) that inhibit the replication of multi-PI-resistant HIV-1 in vitro and possess favorable lipophilicity that may allow blood–brain barrier penetration. Antimicrob. Agents Chemother.57 (12), 6110–6121 (2013).
  • Ghosh AK , YashchukS, MizunoAet al. Design of gem-difluoro-bis-tetrahydrofuran as P2 ligand for HIV-1 protease inhibitors to improve brain penetration: synthesis, X-ray studies, and biological evaluation. ChemMedChem10 (1), 107–115 (2015).
  • Qiu X , ZhaoG-D, TangL-Q, LiuZ-P. Design and synthesis of highly potent HIV-1 protease inhibitors with novel isosorbide-derived P2 ligands. Bioorg. Med. Chem. Lett.24 (11), 2465–2468 (2014).
  • Yedidi RS , MaedaK, FyvieWSet al. P2’ benzene carboxylic acid moiety is associated with decrease in cellular uptake: evaluation of novel nonpeptidic HIV-1 protease inhibitors containing P2 bis-tetrahydrofuran moiety. Antimicrob. Agents Chemother.57 (10), 4920–4927 (2013).
  • Yedidi RS , GarimellaH, AokiMet al. A conserved hydrogen-bonding network of P2 bis-tetrahydrofuran-containing HIV-1 protease inhibitors (PIs) with a protease active-site amino acid backbone aids in their activity against PI-resistant HIV. Antimicrob. Agents Chemother.58 (7), 3679–3688 (2014).
  • Cihlar T , HeG-X, LiuXet al. Suppression of HIV-1 protease inhibitor resistance by phosphonate-mediated solvent anchoring. J. Mol. Biol.363 (3), 635–647 (2006).
  • Grantz Šašková K , KozíšekM, StrayKet al. GS-8374, a prototype phosphonate-containing inhibitor of HIV-1 protease, effectively inhibits protease mutants with amino acid insertions. J. Virol.88 (6), 3586–3590 (2014.
  • Blum A , BöttcherJ, DörrSet al. Two solutions for the same problem: multiple binding modes of pyrrolidine-based HIV-1 protease inhibitors. J. Mol. Biol.410 (4), 745–755 (2011).
  • Lexa KW , CarlsonHA. Binding to the open conformation of HIV-1 protease. Proteins79 (7), 2282–2290 (2011).
  • Böttcher J , BlumA, DörrSet al. Targeting the open-flap conformation of HIV-1 protease with pyrrolidine-based inhibitors. Chem. Med. Chem.3 (9), 1337–1344 (2008).
  • Schimer J , CíglerP, VeselýJet al. Structure-Aided Design of Novel Inhibitors of HIV Protease Based on a Benzodiazepine Scaffold. J. Med. Chem.55 (22), 10130–10135 (2012).
  • Perryman AL , ZhangQ, SoutterHHet al. Fragment-based screen against HIV protease. Chem. Biol. Drug Des.75 (3), 257–268 (2010).
  • Mattei S , AndersM, KonvalinkaJet al. Induced maturation of Human Immunodeficiency Virus. J. Virol.88 (23), 13722–13731 (2014).
  • Lee SK , PotempaM, KolliMet al. Context surrounding processing sites is crucial in determining cleavage rate of a subset of processing sites in HIV-1 Gag and Gag-Pro-Pol polyprotein precursors by viral protease. J. Biol. Chem.287 (16), 13279–13290 (2012).
  • Fun A , van MaarseveenNM, PokornáJet al. HIV-1 protease inhibitor mutations affect the development of HIV-1 resistance to the maturation inhibitor bevirimat. Retrovirology8, 70 (2011).
  • Nguyen AT , FeasleyCL, JacksonKWet al. The prototype HIV-1 maturation inhibitor, bevirimat, binds to the CA-SP1 cleavage site in immature Gag particles. Retrovirology8, 101 (2011).
  • Waki K , DurellSR, SoheilianFet al. Structural and functional insights into the HIV-1 maturation inhibitor binding pocket. PLoS Pathog.8 (11), e1002997 (2012).
  • Dang Z , HoP, ZhuLet al. New betulinic acid derivatives for bevirimat-resistant Human Immunodeficiency Virus type-1. J. Med. Chem.56 (5), 2029–2037 (2013).
  • Tang J , JonesSA, JefferyJLet al. Synthesis and biological evaluation of macrocyclized betulin derivatives as a novel class of anti-HIV-1 maturation inhibitors. Open. Med. Chem. J.8, 23–27 (2014).
  • Huang L , ChenC. Understanding HIV-1 protease autoprocessing for novel therapeutic development. Future Med. Chem.5 (11), 1215–1229 (2013).
  • Huang L , LiY, ChenC. Flexible catalytic site conformations implicated in modulation of HIV-1 protease autoprocessing reactions. Retrovirology8, 79 (2011).
  • Davis DA , SouleEE, DavidoffKSet al. Activity of Human Immunodeficiency Virus type 1 protease inhibitors against the initial autocleavage in Gag-Pol polyprotein processing. Antimicrob. Agents Chemother.56 (7), 3620–3628 (2012).
  • Sayer JM , AnianaA, LouisJM. Mechanism of dissociative inhibition of HIV protease and its autoprocessing from a precursor. J. Mol. Biol.422 (2), 230–244 (2012).
  • Tang C , LouisJM, AnianaAet al. Visualizing transient events in amino-terminal autoprocessing of HIV-1 protease. Nature455 (7213), 693–696 (2008).
  • Agniswamy J , SayerJM, WeberIT, LouisJM. Terminal interface conformations modulate dimer stability prior to amino terminal autoprocessing of HIV-1 protease. Biochemistry51 (5), 1041–1050 (2012).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.