Determinants of the Development and Evolution of HIV-1 Drug Resistance

Total Page:16

File Type:pdf, Size:1020Kb

Determinants of the Development and Evolution of HIV-1 Drug Resistance Determinants of the Development and Evolution of HIV-1 Drug Resistance Axel Fun Determinants of the Development and Evolution of HIV-1 Drug Resistance PhD Thesis, University of Utrecht, the Netherlands Print: Gildeprint drukkerijen, Enschede, the Netherlands ISBN: 978-90-393-5722-4 Coverdesign: Axel Fun and Carlijn van Merode Cover: Flowing waters from nearby hot springs spill across the surface to sculpt magnificent travertine limestone terraces. Mammoth Terraces at Mammoth Hot Springs, Yellowstone National Park, USA. Printing of this thesis was financially supported by the Infection and Immunity Center Utrecht, Merck Sharp & Dohme BV, The Netherlands, Gilead Sciences Netherlands BV and ViiV Healthcare BV. Determinants of the Development and Evolution of HIV-1 Drug Resistance Determinanten van de Ontwikkeling en Evolutie van HIV-1 Medicijn Resistentie (met een samenvatting in het Nederlands) Proefschrift ter verkrijging van de graad van doctor aan de Universiteit Utrecht op gezag van de rector magnificus, prof. dr. G.J. van der Zwaan, ingevolge het besluit van het college voor promoties in het openbaar te verdedigen op donderdag 19 januari 2012 des middags te 2.30 uur door Axel Fun geboren op 7 juni 1981 te Stad Delden Promotor: Prof. dr. E.J.H.J. Wiertz Co-promotoren: Dr. M. Nijhuis Dr. A.M.J. Wensing “Satisfaction of one’s curiosity is one of the greatest sources of happiness in life.” -Linus Pauling- Voor mijn ouders Voor Carlijn Commissie: Prof. dr. R..J. de Boer Prof. dr. M..J.M. Bonten Prof. dr. A.I.M. Hoepelman Prof. dr. F. Miedema Prof. dr. L.P.R. Vandekerckhove Paraninmfen: Evelien Oostdijk Marieke de Regt Contents Chapter 1 General Introduction 9 Partly derived from review articles: Human Immunodeficiency Virus Protease and Gag: Partners in Resistance Submitted for publication Current and Future Integrase Inhibitors: a Systematic Review of literature and Clinical Indications Submitted for publication Chapter 2 In Vitro Selection of High Level Darunavir Resistance in 37 the Absence of Darunavir Resistance Mutations in the Viral Protease In preparation Chapter 3 High Prevalence of Bevirimat Resistance Mutations in 51 Protease Inhibitor-Resistant HIV Isolates AIDS 2010; 24(5):669-673 Chapter 4 HIV-1 Protease Inhibitor Mutations Affect the Development 63 of HIV-1 Resistance to the Maturation Inhibitor Bevirimat Retrovirology 2011; 8:70 Chapter 5 Mutation Q95K Enhances N155H-Mediated Integrase 87 Inhibitor Resistance and Improves Viral Replication Capacity Journal of Antimicrobial Chemotherapy 2010; 65(10):2300-2304 Chapter 6 Impact of the Genetic Background and Population Size 97 on the Evolution of Raltegravir Resistance In preparation Chapter 7 General Discussion 119 Partly derived from review article: Human Immunodeficiency Virus Protease and Gag: Partners in Resistance Submitted for publication Nederlandse samenvatting 137 Publication list 145 Dankwoord/Acknowledgements 147 Curriculum Vitae 151 Chapter 1 General Introduction Partly derived from review articles: Human Immunodeficiency Virus Protease and Gag: Partners in Resistance Axel Fun1, Annemarie MJ Wensing1, Jens Verheyen2 and Monique Nijhuis1 1Department of Virology, Medical Microbiology, University Medical Center Utrecht, The Netherlands; 2Department of Virology, University of Cologne, Cologne, Germany Submitted for publication Current and Future Integrase Inhibitors: a Systematic Review of Literature and Clinical Indications Peter Messiaen1, Annemarie MJ Wensing2, Monique Nijhuis2, Axel Fun2 and Linos Vandekerckhove1 1AIDS Reference Laboratory, Ghent University, Ghent, Belgium; 2Department of Virology, Medical Microbiology, University Medical Center Utrecht, The Netherlands Submitted for publication 10 Chapter 1 The human immunodeficiency virus (HIV) epidemic is still a global health problem with an estimated 33.3 million people infected and 2.6 million newly infected worldwide in 2009[1]. The vast majority is infected with human immunodeficiency virus type 1 (HIV-1). Intensive prevention campaigns and expanding access to treatment resulted in a 19% decline of new infections compared to 1999, the year in which it is thought the epidemic peaked. Despite the global decline of new infections, the incidence in certain risk groups is still rising. For instance, both in the United States and in the Netherlands, the incidence of new HIV diagnosis in young MSM has been steadily rising in recent years[2, 3]. Due to the expanded access to treatment programs, more than 5 million people receive antiretroviral therapy in low- and middle-income countries, which led to a drop in acquired immunodeficiency syndrome (AIDS)-related deaths[1], although early mortality after the initiation of treatment is still a major concern. In the Western countries, the introduction of highly active antiretroviral therapy (HAART) in 1995 resulted in a spectacular improvement of virological suppression rates which led to a dramatic reduction of HIV-1 related morbidity and mortality[4]. Despite these achievements, drug resistance challenges the success of antiretroviral therapy, especially in low and middle income countries where resources and therapeutic options are not as extensively available as in the industrialized countries. With the continually increasing number of people receiving antiretroviral therapy, the incidence of resistance and associated complications are likely to increase in the future. In vivo evolution of HIV The diversity and size of the HIV population within a host is a direct result of a combination of viral and host factors. They include viral replication capacity, the mutation rate of the virus, host cell tropism, the rapid turn-over of viral particles and infected cells, the genetic background of the host (e.g. HLA-type, CCR5Δ32), immune control and target cell availability. HIV has a very high mutation rate which is a feature that is shared by all RNA viruses. The underlying biochemical principal of this high mutation rate is the low fidelity of viral RNA polymerases and reverse transcriptases because these enzymes lack 3’→5’ exonuclease activity, which is a proof-reading mechanism that amends incorrect base-pair insertions. Although the in vivo error rate of HIV-1 reverse transcriptase appears to be lower than predicted from in vitro studies[5-8], the error-rate is still more than a 100,000 times higher than during in vivo DNA transcription in eukaryotic cells, which is approximately one per 10 billion (10-10) basepairs per replication cycle[9, 10]. The estimated in vivo mutation rate of HIV-1 is 3.4x10-5 per basepair per replication cycle[8], which, when multiplied with the size of the HIV-1 genome of about 10 kb, translates to approximately one third of all newly General Introduction 11 generated virus particles containing a change in the viral genome. The high turnover rate of HIV is characterized by rapid clearance of virions and productively infected cells, especially early in infection[11-14]. During acute infection, viral doubling Chapter 1 Chapter time (t2) is estimated to be 10-16 hours[14, 15], which combined with the abundance of permissive target cells can result in extremely high levels of viremia (up to 44 million copies per ml of plasma have been measured)[14]. The doubling time during acute infection is about 3-4 times faster than after treatment interruption during chronic infection (t2 = 1.4- 1.6 days)[16, 17]. The half-life of virions is estimated to be 6-8 hours and that of productively infected cells around 1.5 days. From these data different basic reproductive numbers (R0) have been calculated. Where Little et al[14] calculated that each productively infected cell produces an average of 19 new productively infected cells, Ribeiro et al[15] estimated an R0 of 8.8. The combined high turnover rate and error-prone replication cycle is thought to result in a large and diverse HIV population. However, there is ongoing debate about the exact size and especially the diversity of the population. The concept of population size is critical for understanding the with-in host evolution of HIV-1. The population size ultimately determines if development of HIV-1 drug resistance is largely driven by stochastic or deterministic forces. Considering the large census population within a patient of typically 107-108 HIV-1 infected cells[18], deterministic evolution might seem the major driving force. However, large differences between infected individuals in virological parameters such as viral load set point and rates of emergence and patterns of drug resistance, suggested a more stochastic evolution[19, 20]. In fact, the relevant (effective) population size is always smaller than the census population size. Every productively infected cell produces >1 progeny, therefore all infected cells of a certain generation can not be infected by all productively infected cells of the previous generation, but only by a limited number of infected cells from that generation[21]. Therefore, not every infected cell contributes to the next generation and only the ones that do also contribute to the within host evolution. The number of viral variants that contribute to within host evolution is referred to as the effective population N( e). The mathematical definition of Ne is the size of an idealized population that has the same population genetic properties as the natural population[22] and is an important parameter in models used to study the effect of deterministic and stochastic forces on HIV-1 evolution. To date, many different models have been proposed and large discrepancies exist in the estimates of 2 6 Ne, ranging from 10 to 10 [20-30]. Interestingly, the different estimates span the inverse mutation rate of HIV-1 replication (~10-5), leaving the question whether deterministic or stochastic forces dominate HIV evolution unanswered. 12 Chapter 1 Antiretroviral therapy and drug resistance Zidovudine (AZT) marked the initiation of antiretroviral therapy in 1987, but already soon after its introduction drug resistance was recognized as a serious problem. The subsequent development of other nucleoside reverse-transcriptase inhibitors (NRTIs) revealed that monotherapy would not be able to combat HIV infection very effectively; resistance emerged readily and rapidly.
Recommended publications
  • The Design, Synthesis and Optimization of Allosteric Hiv-1
    THE DESIGN, SYNTHESIS AND OPTIMIZATION OF ALLOSTERIC HIV-1 INTEGRASE INHIBITORS DISSERTATION Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy in the Graduate School of The Ohio State University By Janet Antwi Graduate Program in Pharmaceutical Sciences The Ohio State University 2017 Dissertation Committee: Professor James R. Fuchs, Advisor Professor Werner Tjarks Professor Karl A. Werbovetz Copyright by Janet Antwi 2017 Abstract In the past quarter century, there has been tremendous progress in the discovery of antiretroviral therapy, making HIV/AIDS a manageable chronic disease. However, the HIV virus is relentless and continues to evolve under drug pressure to escape control and continue infection. The enzyme HIV integrase is responsible for the incorporation of viral double stranded DNA into a host chromosomal DNA and has recently become an attractive target in combating HIV resistance. Raltegravir (RAL), elvitegravir (EVG) and dolutegravir (DTG) are three clinically approved active-site integrase inhibitors. Unfortunately, mutations of the enzyme observed in patients have resulted in resistance to thedrug in the clinic. A new approach to targeting integrase (IN) is the development of allosteric inhibitors that specifically target the protein-protein interaction between IN and its cellular cofactor LEDGF/p75. Recently discovered quinoline-based allosteric integrase inhibitor (ALLINI) B1224436 was the first compound to advance into clinical trials but was discontinued due to poor pharmacokinetic properties including low in vivo clearance. In addition, several reports have revealed the emergence of resistance due to mutation to quinoline based ALLINIs. Applying scaffold hopping approach, several pyridine-based, thiophenes, ii pyrazoles, isoquinolines and other heteroaromatic cores have been studied as ALLINIs.
    [Show full text]
  • Mutational Studies of Novel Screened Molecules Against Wild and Mutated HIV-1 Integrase Using Molecular Docking Studies Pawan Gupta1,2*, Prabha Garg2
    Research Article Mutational studies of novel screened molecules against wild and mutated HIV-1 integrase using molecular docking studies Pawan Gupta1,2*, Prabha Garg2 ABSTRACT Background and Aim: The screened molecules which proposed novel HIV-1 integrase inhibitors were collected from the literature. Mutational studies were performed to check whether these molecules are having good binding affinity against mutated HIV-1 IN or not using molecular docking technique. Materials and Methods: First, homology models of the mutated HIV-1 IN were prepared and subsequently all the models were refined and optimized in MODELLER program. Next, molecular docking studies were performed into the active site of mutated HIV-1 IN models using the proposed inhibitors in AutoDock 4.1 program. The results of these studies were compared with the wild type docking studies. Results: The docking studies were found that some of the screened molecules (ZINC1245110, 131614, 92749, ZINC05181828, and ZINC13147504) followed the same binding patterns (in term of locations, interactions, and binding score) as found with wild type HIV-1 IN. Conclusions: Computationally, the same binding patterns were exhibited by these molecules (ZINC1245110, 131614, 92749, ZINC05181828, and ZINC13147504) against mutated models as wild type. This elucidated that these molecules having susceptibility against the drug-resistant HIV-1 IN. Hence, these molecules may be used as a starting point to design novel inhibitors against mutated HIV-1 IN, which need to be confirmed experimentally. KEY WORDS: Docking, Drug resistance, Homology modeling, HIV-1 integrase, Mutation INTRODUCTION Drug resistance is the inevitable consequence of incomplete suppression of HIV-1 replication. The Human immuno-virus (HIV) causes AIDS.
    [Show full text]
  • Fourth Quarter Product Sales of $2.13 Billion, up 11% Year Over Year
    Gilead Sciences Announces Fourth Quarter and Full Year 2011 Financial Results February 2, 2012 4:06 PM ET - Fourth Quarter Product Sales of $2.13 Billion, up 11% Year over Year - - Full Year 2011 Product Sales of $8.10 Billion, up 10% over 2010 - - Full Year 2011 Non-GAAP EPS of $3.86, up 5% over 2010 - - Full Year 2011 Operating Cash Flows of $3.64 Billion - FOSTER CITY, Calif.--(BUSINESS WIRE)--Feb. 2, 2012-- Gilead Sciences, Inc. (Nasdaq:GILD) announced today its results of operations for the fourth quarter and full year 2011. Total revenues for the fourth quarter of 2011 increased 10 percent to $2.20 billion, from $2.00 billion for the fourth quarter of 2010. Net income for the fourth quarter of 2011 was $665.1 million, or $0.87 per diluted share, compared to $629.4 million, or $0.76 per diluted share for the fourth quarter of 2010. Non-GAAP net income for the fourth quarter of 2011, which excludes after-tax acquisition-related, restructuring and stock-based compensation expenses, was $743.1 million, or $0.97 per diluted share, compared to $779.3 million, or $0.95 per diluted share for the fourth quarter of 2010. Full year 2011 total revenues were $8.39 billion, up 5 percent compared to $7.95 billion for 2010. Net income for 2011 was $2.80 billion, or $3.55 per diluted share, compared to $2.90 billion, or $3.32 per diluted share for 2010. Non-GAAP net income for 2011, which excludes after-tax acquisition-related, restructuring and stock-based compensation expenses, was $3.04 billion, or $3.86 per diluted share, compared to $3.21 billion, or $3.69 per diluted share for 2010.
    [Show full text]
  • This Project Has Been Supported with Unrestriced Grants from Abbvie Gilead Sciences HEXAL Janssen-Cilag MSD Viiv Healthcare By
    This project has been supported with unrestriced grants from AbbVie Gilead Sciences HEXAL Janssen-Cilag MSD ViiV Healthcare By Marcus Altfeld, Hamburg/Boston (USA) Achim Barmeyer, Dortmund Georg Behrens, Hannover Dirk Berzow, Hamburg Christoph Boesecke, Bonn Patrick Braun, Aachen Thomas Buhk, Hamburg Rob Camp, Barcelona (Spain/USA) Rika Draenert, Munich Christian Eggers, Linz (Austria) Stefan Esser, Essen Gerd Fätkenheuer, Cologne Gunar Günther, Windhoek (Namibia) Thomas Harrer, Erlangen Christian Herzmann, Borstel Christian Hoffmann, Hamburg Heinz-August Horst, Kiel Martin Hower, Dortmund Christoph Lange, Borstel Thore Lorenzen, Hamburg Tim Niehues, Krefeld Christian Noah, Hamburg Ramona Pauli, Munich Ansgar Rieke, Koblenz Jürgen Kurt Rockstroh, Bonn Thorsten Rosenkranz, Hamburg Bernhard Schaaf, Dortmund Ulrike Sonnenberg-Schwan, Munich Christoph D. Spinner, Munich Thomas Splettstoesser (Figures), Berlin Matthias Stoll, Hannover Hendrik Streeck, Essen/Boston (USA) Jan Thoden, Freiburg Markus Unnewehr, Dortmund Mechthild Vocks-Hauck, Berlin Jan-Christian Wasmuth, Bonn Michael Weigel, Schweinfurt Thomas Weitzel, Santiago (Chile) Eva Wolf, Munich HIV 2015/16 www.hivbook.com Edited by Christian Hoffmann and Jürgen K. Rockstroh Medizin Fokus Verlag IV Christian Hoffmann, M.D., Ph.D. ICH Stadtmitte (Infektionsmedizinisches Centrum Hamburg) Glockengiesserwall 1 20095 Hamburg, Germany Phone: + 49 40 2800 4200 Fax: + 49 40 2800 42020 [email protected] Jürgen K. Rockstroh, M.D., Ph.D. Department of Medicine I University of Bonn Sigmund-Freud-Strasse 25 53105 Bonn, Germany Phone: + 49 228 287 6558 Fax: + 49 228 287 5034 [email protected] HIV Medicine is an ever-changing field. The editors and authors of HIV 2015/16 have made every effort to provide information that is accurate and complete as of the date of publication.
    [Show full text]
  • Interbiotech Ritonavir
    InterBioTech FT-LSI940 Ritonavir Products Description Product Name: Ritonavir Syn: A 84538; ABT 538; Abbott 84538; NSC 693184; RTV; C₃₇H₄₈N₆O₅S₂ Cat N° : LSI940, 10mg LSI941, 50mg LSI942, 100mg LSI943, 500 mg Inquire >=1 g also available as 10 mM solution (1 mL in DMSO) CAS No.: 155213-67-5 MWt: 720.94 Purity: 99.55% (white solid) Melting Point: 175-178°C Solubility: 10 mM in DMSO; < 0.1 mg/mL in H2O Target: CYP3A4 Pathway: Proteaseome Storage:(L) store the product at -20°C (stable for 3 years) or +4°C (stable for 2 years) (M) Product Name: Ritonavir metabolite ( Desthiazolylmethyloxycarbonyl Ritonavir ) Cat N° : XMH680, 5mg XMH681, 10 mg XMH682, 50 mg CAS No.: 176655-55-3 MWt: 579.8 C₃₇H₄₈N₆O₅S₂ Technical and Scientific Information Ritonavir is an inhibitor of HIV protease used to treat HIV infection and AIDS. Biological activity: Ritonavir is an inhibitor of CYP3A4 mediated testosterone 6β-hydroxylation with mean Ki of 19 nM and also inhibits [1] tolbutamide hydroxylation with IC50 of 4.2 μM . Ritonavir is found to be a potent inhibitor of CYP3A-mediated biotransformations (nifedipine oxidation with IC50 of 0.07 mM, 17alpha-ethynylestradiol 2-hydroxylation with IC50 of 2 mM; terfenadine hydroxylation with IC50 of 0.14 mM). Ritonavir is also an inhibitor of the reactions mediated by [2] CYP2D6 (IC50=2.5 mM) and CYP2C9/10 (IC50=8.0 mM) . Ritonavir results in an increase in cell viability in uninfected human PBMC cultures. Ritonavir markedly decreases the susceptibility of PBMCs to apoptosis correlated with lower levels of caspase-1 expression, decreases in annexin V staining, and reduces caspase-3 activity in uninfected human PBMC cultures.
    [Show full text]
  • The Impact of Modern Antiretroviral Therapy on Lipid Metabolism of HIV-1 Infected Patients
    Chapter 6 The Impact of Modern Antiretroviral Therapy on Lipid Metabolism of HIV-1 Infected Patients Joel da Cunha, Luciana Morganti Ferreira Maselli, Sérgio Paulo Bydlowski and Celso Spada Additional information is available at the end of the chapter http://dx.doi.org/10.5772/61061 1. Introduction The highly active antiretroviral therapy (HAART) is the most efficient and safe alternative against HIV-1 infection, to allow the restoration of the immune system, with consequent reduction in mortality rate, increased survival and quality of life of infected patients. Apart from the great benefits of the use of different HAART regimens, laboratory and clinical experience has shown that HAART can induce severe and considerable adverse effects on metabolic complications of lipid metabolism, characterized by signs of dyslipidemia, increased risk of cardiovascular disease and even an increased risk of atherosclerosis. In this context, the class of protease inhibitors has been associated with a higher level of changes of lipid metab‐ olism and an increased risk for cardiovascular disease. In turn, the search for different therapeutic strategies to reverse HAART-associated lipid disorders has led to the use of less metabolically active antiretroviral drugs without compromising antiretroviral efficacy. Thus, the different interactions of antiretroviral drugs are recommended based on their degree of impact on lipid metabolism. Recently, fusion inhibitors, integrase strand transfer inhibitors, entry inhibitors, have been included in the therapeutic arsenal against HIV-1 infection, and are not associated with metabolic disorders, since their mechanisms of action are different from other classes of antiretrovirals. Instead, the use of hypolipidemic drug therapy (statins, fibrates, inhibitors of intestinal cholesterol) becomes necessary when HAART-associated dyslipidemia occurs or persists for a long period and when alterations in diet, exercise and other HAART strategies are ineffective.
    [Show full text]
  • Discovery of Novel Integrase Inhibitors Acting Outside the Active Site Through High-Throughput Screening
    molecules Article Discovery of Novel Integrase Inhibitors Acting outside the Active Site Through High-Throughput Screening 1 2, 2, 1 Cindy Aknin , Elena A. Smith y, Christophe Marchand z, Marie-Line Andreola , Yves Pommier 2 and Mathieu Metifiot 1,* 1 Laboratoire MFP, CNRS UMR5234, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux CEDEX, France; [email protected] (C.A.); [email protected] (M.-L.A.) 2 Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, CCR, NCI, NIH, 37 Convent Drive, Bethesda, MD 20892, USA; [email protected] (E.A.S.); [email protected] (C.M.); [email protected] (Y.P.) * Correspondence: mathieu.metifi[email protected]; Tel.: +33-557571739 Present addresses: Quality Control, Protein Sciences, A Sanofi Company, 1000 Research Parkway, y Meriden, CT 06450, USA. Present addresses: Center for Research Strategy, NCI, NIH, 31 Center Drive, Bethesda, MD 20892, USA. z Received: 20 September 2019; Accepted: 9 October 2019; Published: 12 October 2019 Abstract: Currently, an increasing number of drugs are becoming available to clinics for the treatment of HIV infection. Even if this targeted therapy is highly effective at suppressing viral replication, caregivers are facing growing therapeutic failures in patients, due to resistance with or without treatment adherence concerns. Accordingly, it is important to continue to discover small molecules that have a novel mechanism of inhibition. In this work, HIV integrase inhibitors were selected by high-throughput screening. Chemical structure comparisons enabled the identification of stilbene disulfonic acids as a potential new chemotype. Biochemical characterization of the lead compound stilbenavir (NSC34931) and a few derivatives was performed.
    [Show full text]
  • Hiv Integrase Mechanisms of Resistance to Raltegravir, Elvitegravir, and Dolutegravir Kyla Nicole Ross Wayne State University
    Wayne State University Wayne State University Theses 1-1-2015 Hiv Integrase Mechanisms Of Resistance To Raltegravir, Elvitegravir, And Dolutegravir Kyla Nicole Ross Wayne State University, Follow this and additional works at: https://digitalcommons.wayne.edu/oa_theses Part of the Biochemistry Commons, Bioinformatics Commons, and the Virology Commons Recommended Citation Ross, Kyla Nicole, "Hiv Integrase Mechanisms Of Resistance To Raltegravir, Elvitegravir, And Dolutegravir" (2015). Wayne State University Theses. 458. https://digitalcommons.wayne.edu/oa_theses/458 This Open Access Thesis is brought to you for free and open access by DigitalCommons@WayneState. It has been accepted for inclusion in Wayne State University Theses by an authorized administrator of DigitalCommons@WayneState. HIV INTEGRASE MECHANISMS OF RESISTANCE TO RALTEGRAVIR, ELVITEGRAVIR, AND DOLUTEGRAVIR by KYLA ROSS THESIS Submitted to the Graduate School of Wayne State University, Detroit, Michigan in partial fulfillment of the requirements for the degree of MASTER OF SCIENCE 2015 MAJOR: BIOCHEMISTRY AND MOLECULAR BIOLOGY Approved By: Advisor Date © COPYRIGHT BY KYLA ROSS 2015 All Rights Reserved DEDICATION I dedicate this thesis to my late step brother Carlton Lowry. ii ACKNOWLEDGEMENTS I would like to thank everyone in the Kovari lab for their generous help with this thesis. I thank Ben for helping me with the manuscripts, the research, and the experiments and for being a great colleague and an awesome friend. I thank Brad for answering the most complicated questions, for suggesting alternative approaches, and for being a great friend. I thank Cathy for being there to listen, the awesome conversations and for being a great colleague. I would like to thank Tamaria for giving me this project and offering her professional input.
    [Show full text]
  • The Emerging Role of HIV-1 Integrase in Virion Morphogenesis
    viruses Review Going beyond Integration: The Emerging Role of HIV-1 Integrase in Virion Morphogenesis Jennifer L. Elliott and Sebla B. Kutluay * Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; [email protected] * Correspondence: [email protected] Received: 26 August 2020; Accepted: 7 September 2020; Published: 9 September 2020 Abstract: The HIV-1 integrase enzyme (IN) plays a critical role in the viral life cycle by integrating the reverse-transcribed viral DNA into the host chromosome. This function of IN has been well studied, and the knowledge gained has informed the design of small molecule inhibitors that now form key components of antiretroviral therapy regimens. Recent discoveries unveiled that IN has an under-studied yet equally vital second function in human immunodeficiency virus type 1 (HIV-1) replication. This involves IN binding to the viral RNA genome in virions, which is necessary for proper virion maturation and morphogenesis. Inhibition of IN binding to the viral RNA genome results in mislocalization of the viral genome inside the virus particle, and its premature exposure and degradation in target cells. The roles of IN in integration and virion morphogenesis share a number of common elements, including interaction with viral nucleic acids and assembly of higher-order IN multimers. Herein we describe these two functions of IN within the context of the HIV-1 life cycle, how IN binding to the viral genome is coordinated by the major structural protein, Gag, and discuss the value of targeting the second role of IN in virion morphogenesis. Keywords: HIV-1; integrase; maturation; integrase–RNA interactions; protein–RNA interactions 1.
    [Show full text]
  • Hoffmann Rockstroh |
    Hoffmann| Rockstroh HIV 2015/2016 www.hivbook.com Medizin Fokus Verlag This project has been supported with unrestriced grants from AbbVie Gilead Sciences HEXAL Janssen-Cilag MSD ViiV Healthcare By Marcus Altfeld, Hamburg/Boston (USA) Achim Barmeyer, Dortmund Georg Behrens, Hannover Dirk Berzow, Hamburg Christoph Boesecke, Bonn Patrick Braun, Aachen Thomas Buhk, Hamburg Rob Camp, Barcelona (Spain/USA) Rika Draenert, Munich Christian Eggers, Linz (Austria) Stefan Esser, Essen Gerd Fätkenheuer, Cologne Gunar Günther, Windhoek (Namibia) Thomas Harrer, Erlangen Christian Herzmann, Borstel Christian Hoffmann, Hamburg Heinz-August Horst, Kiel Martin Hower, Dortmund Christoph Lange, Borstel Thore Lorenzen, Hamburg Tim Niehues, Krefeld Christian Noah, Hamburg Ramona Pauli, Munich Ansgar Rieke, Koblenz Jürgen Kurt Rockstroh, Bonn Thorsten Rosenkranz, Hamburg Bernhard Schaaf, Dortmund Ulrike Sonnenberg-Schwan, Munich Christoph D. Spinner, Munich Thomas Splettstoesser (Figures), Berlin Matthias Stoll, Hannover Hendrik Streeck, Essen/Boston (USA) Jan Thoden, Freiburg Markus Unnewehr, Dortmund Mechthild Vocks-Hauck, Berlin Jan-Christian Wasmuth, Bonn Michael Weigel, Schweinfurt Thomas Weitzel, Santiago (Chile) Eva Wolf, Munich HIV 2015/16 www.hivbook.com Edited by Christian Hoffmann and Jürgen K. Rockstroh Medizin Fokus Verlag IV Christian Hoffmann, M.D., Ph.D. ICH Stadtmitte (Infektionsmedizinisches Centrum Hamburg) Glockengiesserwall 1 20095 Hamburg, Germany Phone: + 49 40 2800 4200 Fax: + 49 40 2800 42020 [email protected] Jürgen K. Rockstroh, M.D., Ph.D. Department of Medicine I University of Bonn Sigmund-Freud-Strasse 25 53105 Bonn, Germany Phone: + 49 228 287 6558 Fax: + 49 228 287 5034 [email protected] HIV Medicine is an ever-changing field. The editors and authors of HIV 2015/16 have made every effort to provide information that is accurate and complete as of the date of publication.
    [Show full text]
  • BI 224436 | Medchemexpress
    Inhibitors Product Data Sheet BI 224436 • Agonists Cat. No.: HY-18595 CAS No.: 1155419-89-8 Molecular Formula: C₂₇H₂₆N₂O₄ • Molecular Weight: 442.51 Screening Libraries Target: HIV; HIV Integrase Pathway: Anti-infection; Metabolic Enzyme/Protease Storage: Powder -20°C 3 years 4°C 2 years * The compound is unstable in solutions, freshly prepared is recommended. SOLVENT & SOLUBILITY In Vitro DMSO : ≥ 50 mg/mL (112.99 mM) * "≥" means soluble, but saturation unknown. Mass Solvent 1 mg 5 mg 10 mg Concentration Preparing 1 mM 2.2598 mL 11.2992 mL 22.5984 mL Stock Solutions 5 mM 0.4520 mL 2.2598 mL 4.5197 mL 10 mM 0.2260 mL 1.1299 mL 2.2598 mL Please refer to the solubility information to select the appropriate solvent. In Vivo 1. Add each solvent one by one: 10% DMSO >> 40% PEG300 >> 5% Tween-80 >> 45% saline Solubility: ≥ 2.5 mg/mL (5.65 mM); Clear solution 2. Add each solvent one by one: 10% DMSO >> 90% (20% SBE-β-CD in saline) Solubility: ≥ 2.5 mg/mL (5.65 mM); Clear solution 3. Add each solvent one by one: 10% DMSO >> 90% corn oil Solubility: ≥ 2.5 mg/mL (5.65 mM); Clear solution BIOLOGICAL ACTIVITY Description BI 224436 is a novel HIV-1 noncatalytic site integrase inhibitor with EC50 values of less than 15 nM against different HIV-1 laboratory strains. IC₅₀ & Target EC50: 15 nM (HIV-1)[1] In Vitro BI 224436 has cellular cytotoxicity of more than 90 μM. BI 224436 has a low, 2.1-fold decrease in antiviral potency in the presence of 50% human serum.
    [Show full text]
  • HIV Pipeline 2017: Targets in the HIV Lifecycle Targets in the HIV Lifecycle 1 HIV Attaches to a CD4 Cell
    ISSN 1472-4863 HIV pipeline2017 New drugs in development htb supplement: 2017 Vol 18:(1) HIV pipeline 2017: targets in the HIV lifecycle Targets in the HIV lifecycle 1 HIV attaches to a CD4 cell. 1 HIV Monoclonal Entry inhibitors 2 HIV enters a CD4 cell and gp120 antibodies (mAb) fostemsavir capsid HIV proteins and enzymes combinectin gp41 UB-421 (CD4 receptor) are released into the cell. ibalizumab (CD4 receptor) 3 Reverse transcriptase (RT) CD4 receptor (CCR5 recept.) makes double strand HIV. CCR5 coreceptor PRO-140 4 Integrase enables HIV to join the cell DNA. NRTIs/NtRTIs 2 5 Protease cuts and (nukes) reassembles new HIV. EFdA (MK-8591) 6 Each cell produces GS-9131 CD4 hundreds of new virions. HIV RNA cell NNRTIs (non-nukes) 3 Cell doravirine nucleus Protease inhibitor elsufavirine HIV DNA New viral GS-PS1 rilpivirine LA material 4 5 Capsid inhibitor Integration GS-CA1 INIs (or INSTIs) Maturation Maturation bictegravir and budding inhibitor cabotegravir 6 cabotegravir LA GSK3640254 C O N T E N T S HIVi-base.info i-Base (2017)(www.i-Base.info) New HIV HIV pipeline 2017 2 Introduction to HIV pipeline in 2017 2 Figure 1: HIV pipeline 2017: targets in the HIV lifecycle 3 Recently approved new HIV drugs 3 • Raltegravir HD • Generic dolutegravir • Generic TDF/FTC Submitted applications: completed phase 3 results 4 • Darunavir/cobicistat/FTC/TAF - FDC • Bictegravir/FTC/TAF - FDC • Dolutegravir/rilpivirine - two-drug FDC Table 1: HIV pipeline compounds by development phase 5 Compounds in phase 3 development 6 • Doravirine - NNRTI • Cabotegravir
    [Show full text]