reviews - Virology Education
Transcription
reviews - Virology Education
A IN TI VIR E AL TH R PY N A & IN CT IO U S DIS SE IN FE S REVIEWS EA Reviews & 3 2011 Pneumonie.nl in Antiviral Therapy INFECTIOUS DISEASES JOURNAL OF ABSTRACTS AND CONFERENCE REPORTS FROM INTERNATIONAL WORKSHOPS ON INFECTIOUS DISEASES & ANTIVIRAL THERAPY Meeting report 6th International Workshop on HIV Transmission Principles of Intervention 14 - 15 July, 2011, Rome, Italy 3rd International Workshop on HIV Pediatrics Dé pneumonieportal voor en door medische professionals 15 - 16 July, 2011, Rome, Italy Het laatste nieuws Scorecards Beeldenbank Video’s Factsheet www.pneumonie.nl pneumonie.nl wordt mogelijk gemaakt door SUPPLEMENT Presentatiemateriaal VACCINATIE Masterclass 2012 DOELGROEP: clinici, waaronder infectiologen, microbiologen, kinderartsen, GGD-artsen, apothekers en arts-assistenten in opleiding voor voorgenoemde specialismen. FORMAT: de masterclass bestaat uit zeven modules die maandelijks op dinsdagavond (18.30 - 21.00 u.) in Utrecht zullen worden georganiseerd. ONDERWERPEN: Vaccinatie en het immuunsysteem, vaccins tegen virussen, bacteriën en protozoa, farmaco-economie, bijwerkingen en acceptatie van vaccins, de ontwikkeling van vaccins inclusief klinische studies, overwegingen om vaccins al dan niet in het Rijksvaccinatieprogramma op te nemen. PROGRAMMA 10 januari 2012 Module I Het begin van vaccinatie, soort vaccins Vaccinatie en het immuunsysteem 7 februari 2012 Module II Adjuvantia en toedieningsvormen Vaccins tegen griep 13 maart 2012 Module III De ontwikkeling van het Rijksvaccinatieprogramma, herd immunity Vaccinatie van kinderen tegen rotavirusinfecties 3 april 2012 Module IV Vaccinatie tegen malaria Ziektelast en kosteneffectiviteit 8 mei 2012 Module V Het meten van werkzaamheid van vaccins, correlaten bescherming Klinische studies: het Capita onderzoek Bijwerkingen en acceptatie van vaccins 5 juni 2012 Module VI Van idee tot product, klinische studies, productie, registratie Vaccinatie tegen S. aureus infecties 19 juni 2012 Module VII Vaccins voor ouderen, immunosenescence Interactieve sessies over de uitbreiding van het vaccinatieprogramma: van evidence based medicine naar maatschappelijk draagvlak ATRIPLA Effectiviteit niet overtroffen in studies 1-12 – de voorkeur van patiënten 13-16* Vertrouwen in de toekomst voor u en uw patiënten. Prof. Dr. Ben van der Zeijst (LUMC) Prof. Dr. Georg Kraal (VU) Dr. Gideon Kersten (RIVM) Dr. Bettie Voordouw (CBG) Prof. Dr. Joost Ruitenberg (Gezondheidsraad) Drs. Patricia Bruijning-Verhagen (UMCU) Prof. Dr. Robert Sauerwein (UMC Radboud) Prof. Dr. Maarten Postma (RUG) Dr. Loek van Alphen (RIVM) Prof. Dr. Marc Bonten (UMCU) Dr. Agnes Kant (Lareb) Prof. Dr. Han van den Bosch (VU) Prof. Dr. Jos van Strijp (UMCU) Prof. Dr. Catharina Mathei (UC Louvain, Belgie) Prof. Dr. Willem van Eden (UMCU) Prof. Dr. Ben van der Zeijst (LUMC) ORGANISEREND COMITÉ Prof. Dr. Marc Bonten Prof. Dr. Han van den Bosch Prof. Dr. Robert Sauerwein Prof. Dr. Ben van der Zeijst Universitair Medisch Centrum Utrecht Vrije Universiteit Amsterdam St. Radboud Universitair Medisch Centrum Nijmegen Leids Universitair Medisch Centrum, Leiden DE VACCINATIE MASTERCLASS WORDT ONDERSTEUND DOOR ACCREDITATIE IS AANGEVRAAGD BIJ DE NIV, NvMM, NvK en NVZA *ATRIPLA is geregistreerd voor volwassen HIV-1-patiënten met virologische suppressie. efavirenz 600mg/emtricitabine 200mg/ tenofovir disoproxil (as fumarate) 245mg Tablets HAART in 1 ©2009 Bristol-Myers Squibb & Gilead Sciences Verkorte productinformatie beschikbaar elders in dit blad. Ad Atripla.indd 1 101223/092 19-01-11 12:12 VOOR MEER INFORMATIE & REGISTRATIE, KIJK OP WWW.VIROLOGY-EDUCATION.COM De masterclass wordt georganiseerd door Virology Education De masterclass wordt financieel mogelijk gemaakt door Pfizer bv Reviews & in Antiviral Therapy INFECTIOUS DISEASES Reviews in Antiviral Therapy & Infectious Diseases is the official journal of abstracts and conference reports from International Workshops on infectious diseases & antiviral therapy. Reviews in Antiviral Therapy & Infectious Diseases publishes peer-reviewed articles relating to viral diseases including HIV, Hepatitis and emerging viruses. Featured topics include clinical management, drug resistance, diagnostic applications, pharmacology, transmission & prevention. Each edition will be dedicated to a specific aspect of viral infection, focusing on the presentations from the latest international meeting on the topic. Reviews in Antiviral Therapy & Infectious Diseases aims at translating the latest key scientific and clinical findings in antiviral therapy into tangible and applicable knowledge to assist readers in routine clinical management. Editors-in-chief: Charles A.B. Boucher, MD, PhD, Erasmus Medical Center Rotterdam, The Netherlands Jonathan M. Schapiro, MD, Sheba Medical Center, Tel Aviv, Israel Publisher: Virology Education, Biltstraat 106, 3572 BJ Utrecht, The Netherlands Phone: +31-30 2307140 Fax: +31-30 2307148 info@virology-education.com; www.virology-education.com ISSN:1872-437X Copyright: All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or transmitted in any form or by any means, electronic, mechanical, photocopying, recording, or otherwise, without prior permission of the publisher. © Virology Education. Reprints:In order to acquire additional reprints of this volume, please contact Virology Education at info@virology-education.com Printed on woodfree coated paper Printed by Labor, Utrecht, The Netherlands (CU-COC-807561) Disclaimer The information in this journal is those of the authors and not necessarily reflects the views of the board or the publisher. No responsibility is assumed by Virology Education for any injury and/or damage to persons or property as result of product liability, negligence or otherwise, or form any use or operation of any methods, products, instructions, or ideas contained in the material herein. All clinical diagnoses and drug regimens/dosages must be independently verified. A qualified healthcare professional should be consulted before using any therapeutic product discussed. Reviews in Antiviral Therapy & Infectious Diseases - volume 1; 2011 - Supplement 1 open een wereld van nieuwe mogelijkheden ISENTRESS, de eerste integraseremmer, nu geregistreerd voor zowel behandelingsnaïeve als eerder behandelde volwassenen met HIV in combinatie met andere antiretrovirale middelen. 0910RTG09NL416J0909 Dus kies vanaf het eerste begin ISENTRESS! ISENTRESS is geïndiceerd in combinatie met andere antiretrovirale geneesmiddelen voor de behandeling van humaan-immunodeficiëntievirus (HIV-1)-infectie bij volwassen patiënten. Raadpleeg de volledige productinformatie (SPC) inclusief dosering, contra-indicaties en waarschuwingen alvorens ISENTRESS voor te schrijven. Zie elders in dit blad voor de verkorte bijsluiter ISENTRESS® en univadis zijn geregistreerde handelsmerken van MERCK & Co., Inc., Whitehouse Station, NJ, USA. Postbus 581, 2003 PC Haarlem, www.msd.nl, www.univadis.nl Nieuw! Viramune met Verlengde afgifte stabiele spiegels met eenmaal daags 1 tablet • het vertrouwde Viramune nu met verlengde afgifte 1 • eenmaaldaagse dosering van 400 mg 1 • goed bijwerkingenprofiel voor de lange termijn3,4,5,6 1. Samenvatting van de productkenmerken 2. Battegay et al. Clin ther. 2011; 33: 1308-1320 3. elzi et al. arch intern med. 2010; 170(1): 57-65 4. reliquet et al. HiV lin tirals 2010; 170: 57-65 5. Vallicello et al. aidS research and human retroviruses. 2011; 27(10) 6. gathe et al. antiviral therapy 2011; 16: 759-69. Voor productinformatie zie elders in dit blad. Sterk op de lange termijn VERKORTE PRODUCTINFORMATIE ISENTRESS 400 mg filmomhulde tabletten. Samenstelling Filmomhulde tablet: 400 mg raltegravir (als kaliumzout). Indicaties ISENTRESS is geïndiceerd in combinatie met andere antiretrovirale geneesmiddelen voor de behandeling van humaan-immunodeficiëntievirus (hiv-1)-infectie bij volwassen patiënten. Deze indicatie is gebaseerd op gegevens over veiligheid en werkzaamheid uit twee dubbelblinde, placebogecontroleerde onderzoeken bij eerder behandelde patiënten en een dubbelblind, met werkzame stof gecontroleerd onderzoek bij niet eerder behandelde patiënten. Klinische ervaring Bewijs voor de effectiviteit van ISENTRESS is gebaseerd op analyses van 96-weeksdata uit twee lopende, gerandomiseerde, dubbelblinde, placebogecontroleerde studies (BENCHMRK 1 en BENCHMRK 2, Protocols 018 en 019) bij eerder met antiretrovirale middelen behandelde, met hiv-1 geïnfecteerde volwassen patiënten, en analyse van 96-weeksdata uit een lopende, gerandomiseerde, dubbelblinde, met werkzame stof gecontroleerde studie (STARTMRK, Protocol 021) bij niet eerder met antiretrovirale middelen behandelde en met hiv-1 geïnfecteerde volwassen patiënten. Contra-indicaties Overgevoeligheid voor het werkzame bestanddeel of voor één van de hulpstoffen. Waarschuwingen en voorzorgen In het algemeen werd in de farmacokinetiek van raltegravir aanzienlijke inter- en intra-individuele variabiliteit waargenomen. Raltegravir heeft een relatief lage genetische barrière voor resistentie. Daarom moet raltegravir waar mogelijk gecombineerd worden met twee andere actieve ARTs om de kans op virologisch falen en het optreden van resistentie te beperken. Bij niet eerder behandelde patiënten zijn de klinische gegevens over gebruik van raltegravir beperkt tot gebruik in combinatie met twee nucleotide-reversetranscriptaseremmers (NRTIs) (emtricitabine en tenofovirdisoproxilfumaraat). De veiligheid en werkzaamheid van ISENTRESS zijn niet vastgesteld bij patiënten met ernstige onderliggende leveraandoeningen. Daarom moet ISENTRESS bij patiënten met een ernstige leverfunctiestoornis met voorzichtigheid worden toegepast. Patiënten met een al eerder bestaande leverfunctiestoornis waaronder chronische hepatitis hebben tijdens antiretrovirale combinatietherapie een verhoogde frequentie van leverfunctiestoornissen gehad en moeten volgens de standaardpraktijk worden gecontroleerd. Er zijn zeer beperkte gegevens over het gebruik van raltegravir bij patiënten die naast hiv ook geïnfecteerd zijn met hepatitis B-virus (HBV) of hepatitis C-virus (HCV). Patiënten met chronische hepatitis B of C die worden behandeld met antiretrovirale combinatietherapie hebben een hogere kans op ernstige en mogelijk fatale leverbijwerkingen. Voorzichtigheid moet worden betracht bij gelijktijdige toediening van ISENTRESS met sterke inductoren van uridinedifosfaat-glucuronosyltransferase (UGT) 1A1 (bijvoorbeeld rifampicine). Rifampicine verlaagt de plasmaconcentraties van raltegravir; de invloed daarvan op de werkzaamheid van raltegravir is onbekend. Maar als gelijktijdige toediening met rifampicine onontkoombaar is, kan verdubbeling van de dosis ISENTRESS worden overwogen. Myopathie en rabdomyolyse zijn gemeld. Gebruik met voorzichtigheid bij patiënten die in het verleden myopathie of rabdomyolyse hebben gehad of anderszins gepredisponeerd zijn. Huiduitslag kwam bij eerder behandelde patiënten die een behandeling met ISENTRESS + darunavir kregen vaker voor dan bij patiënten die ISENTRESS zonder darunavir of darunavir zonder ISENTRESS kregen. ISENTRESS bevat lactose. Patiënten met zeldzame erfelijke stoornissen als galactose-intolerantie, Lapplactasedeficiëntie of glucose-galactosemalabsorptie mogen dit geneesmiddel niet gebruiken. Interacties Uit in vitro-onderzoek blijkt dat raltegravir geen substraat van cytochroom P450 (CYP)-enzymen, is, CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6 of CYP3A niet remt geen inductie van CYP3A4 geeft en het door P-glycoproteïne gereguleerde transport niet remt. Op basis van deze gegevens wordt niet verwacht dat ISENTRESS de farmacokinetiek beïnvloedt van geneesmiddelen die substraten zijn van deze enzymen of P-glycoproteïne. Op basis van in vitro- en in-vivo-onderzoeken wordt raltegravir voornamelijk uitgescheiden door metabolisme via UGT1A1- geïnduceerde glucuronidatieroute. Hoewel uit in-vitro-onderzoek gebleken is dat raltegravir geen remmer is van de UDP-glucuronosyltransferases (UGTs) 1A1 en 2B7, doet één klinisch onderzoek op grond van een waargenomen effect op de glucuronidatie van bilirubine vermoeden dat er in vivo enige remming van UGT1A1 kan voorkomen. Maar de omvang van het effect leidt waarschijnlijk niet tot klinisch belangrijke geneesmiddelinteracties. Bijwerkingen Infecties en parasitaire aandoeningen: soms: genitale herpes†, folliculitis, gastro-enteritis, herpes simplex, infectie met herpesvirus, herpes zoster, influenza, molluscum contagiosum, nasofaryngitis, bovensteluchtweginfectie. Neoplasmata, benigne, maligne en niet-gespecificeerd (inclusief cysten en poliepen): soms: huidpapilloom. Bloed- en lymfestelselaandoeningen: soms: anemie†, anemie wegens ijzergebrek, pijn in lymfeklieren, lymfadenopathie, neutropenie, trombocytopenie‡‡. Immuunsysteemaandoeningen: soms: immuunreconstitutiesyndroom†, geneesmiddelenovergevoeligheid†, overgevoeligheid. Voedings- en stofwisselingsstoornissen: soms: anorexia, cachexie, verminderde eetlust, diabetes mellitus, dyslipidemie, hypercholesterolemie, hyperglykemie, hyperlipidemie, hyperfagie, meer eetlust, polydipsie. Psychische stoornissen: vaak: abnormaal dromen, slapeloosheid, soms: psychische aandoening†, zelfmoordpoging†, angst, verwarring, neerslachtige stemming, depressie, ernstige depressie, slapeloosheid in het midden van de nacht, wisselende stemming, nachtmerrie, paniekaanval, slaapstoornis, suïcidale gedachten‡‡, suïcidaal gedrag (vooral bij patiënten met een voorgeschiedenis van psychische aandoeningen)‡‡. Zenuwstelselaandoeningen: vaak: duizeligheid, hoofdpijn, soms: amnesie, carpaaltunnelsyndroom, cognitieve stoornis, aandachtsstoornis, duizeligheid bij houdingsverandering, dysgeusie, hypersomnie, hypo-esthesie, lethargie, geheugenstoornis, migraine, perifere neuropathie, paresthesie, slaperigheid, spanningshoofdpijn, tremoren. Oogaandoeningen: soms: visusverslechtering. Evenwichtsorgaan- en ooraandoeningen: vaak: vertigo, soms: tinnitus. Hartaandoeningen: soms: palpitaties, sinusbradycardie, ventriculaire extrasystoles. Bloedvataandoeningen: soms: opvliegers, hypertensie. Ademhalingsstelsel-, borstkas- en mediastinumaandoeningen: soms: dysfonie, epistaxis, neusverstopping. Maagdarmstelselaandoeningen: vaak: opgezwollen buik, buikpijn, diarree, winderigheid, misselijkheid, braken, soms: gastritis†, last van de buik, pijn in de bovenbuik, gevoeligheid van de buik, pijn in de anus of het rectum, obstipatie, droge mond, dyspepsie, pijn in de bovenbuik, erosieve duodenitis, oprispingen, gastro-oesofageale reflux, gingivitis, glossitis, odynofagie, acute pancreatitis, maagzweer, rectale bloeding. Lever- en galaandoeningen: soms: hepatitis†, hepatische steatose. Huid- en onderhuidaandoeningen: vaak: huiduitslag‡, soms: acne, alopecia, dermatitis acneiforme, droge huid, erytheem, ingevallen gezicht, hyperhidrose, verkregen lipodystrofie, lipohypertrofie, nachtelijk zweten, prurigo, pruritus, gegeneraliseerde pruritus, maculaire uitslag, maculopapulaire uitslag, uitslag bij pruritus, huidlaesie, urticaria, xeroderma, Stevens Johnson syndroom‡‡. Skeletspierstelsel- en bindweefselaandoeningen: soms: artralgie, artritis, rugpijn, pijn in de flank, musculoskeletale pijn, myalgie, nekpijn, osteopenie, pijn in de extremiteiten, tendinitis, rabdomyolyse‡‡. Nier- en urinewegaandoeningen: soms: nierfalen†, nefritis, nefrolithiase, nycturie, niercyste, nierfunctiestoornis, tubulo-interstitiële nefritis. Voortplantingsstelsel- en borstaandoeningen: soms: erectiestoornis, gynaecomastie, menopauzale verschijnselen. Algemene aandoeningen en toedieningsplaatsstoornissen: vaak: asthenie, vermoeidheid, pyrexie, soms: pijn op de borst, rillingen, oedeem in het gezicht, meer vetweefsel, zich schrikachtig voelen, malaise, perifeer oedeem, pijn. Onderzoeken: vaak: verhoogd alanineaminotransferase, atypische lymfocyten, verhoogd aspartaatamino-transferase, verhoogde triglyceriden in het bloed, verhoogde lipase, soms: verlaagde absolute neutrofielentelling, verhoogde alkalische fosfatase, verlaagd bloedalbumine, verhoogde bloedamylase, verhoogd bloedbilirubine, verhoogd bloedcholesterol, verhoogd bloedcreatinine, verhoogd bloedglucose, verhoogd BUN, verhoogde creatinefosfokinase, nuchter bloedglucose verhoogd, glucose aanwezig in urine, verhoogd HDL, verlaagd LDL, verhoogd LDL, minder trombocyten, positief op rode bloedcellen in urine, grotere tailleomtrek, gewichtstoename, minder witte bloedcellen. Letsels, intoxicaties en verrichtingscomplicaties: soms: onbedoelde overdosis†. † met minstens één ernstige bijwerking ‡ In klinisch onderzoek van eerder behandelde patiënten werd huiduitslag ongeacht causaliteit bij behandelingen met ISENTRESS + darunavir vaker gezien dan bij die met ISENTRESS zonder darunavir of darunavir zonder ISENTRESS. Maar volgens de onderzoeker kwam geneesmiddelgerelateerde huiduitslag ongeveer even vaak voor. De voor blootstelling aangepaste frequenties van huiduitslag (alle oorzaken) waren resp. 10,9, 4,2 en 3,8 per 100 patiëntjaren (PYR) en voor geneesmiddelgerelateerde huiduitslag resp. 2,4, 1,1 en 2,3 per 100 PYR. Deze huiduitslag was licht tot matig-ernstig en leidde niet tot stopzetting van de behandeling. ‡‡ Deze bijwerking is vastgesteld in de periode sinds het geneesmiddel op de markt is, maar werd in gerandomiseerd gecontroleerd klinisch fase III-onderzoek (protocols 018, 019 en 021) niet gemeld als zijnde geneesmiddelgerelateerd. De frequentiecategorie is gedefinieerd als “soms”, in overeenstemming met de richtlijnen voor de samenvatting van de productkenmerken (SmPC) (rev. 2 sept. 2009) op basis van een geschatte bovengrens van het 95 %-betrouwbaarheidsinterval voor 0 voorvallen en het aantal proefpersonen dat in het klinisch fase III-onderzoek met ISENTRESS werd behandeld (n=743). Patiënten die tevens geïnfecteerd zijn met het hepatitis B- en/ of hepatitis C-virus In fase III-onderzoeken mochten eerder behandelde patiënten (N = 114/699 ofwel 16 %; HBV=6 %, HCV=9 %, HBV+HCV=1 %) en niet eerder behandelde patiënten (N = 34/563 of 6 %; HBV=4 %, HCV=2 %, HBV+HCV=0,2 % met gelijktijdige chronische (maar geen acute) infectie met hepatitis B en/of hepatitis C) meedoen op voorwaarde dat de leverfunctietests bij baseline niet hoger waren dan 5 keer de normale bovengrens. In het algemeen was het veiligheidsprofiel van ISENTRESS bij patiënten met een gelijktijdige infectie met hepatitis B-en/of hepatitis C-virus gelijk aan die van patiënten zonder hepatitis B-en/of hepatitis C-virus, hoewel in beide behandelingsgroepen de frequentie van AST- en ALT-afwijkingen in de subgroep met gelijktijdige infectie met hepatitis B en/of hepatitis C iets hoger was. Farmacotherapeutische categorie Antiviraal middel voor systemisch gebruik, Andere antiretrovirale middelen, ATC-code: J05AX08 Afleverstatus UR Verpakking ISENTRESS 400 mg is verkrijgbaar in flesjes met 60 tabletten. Vergoeding en prijzen ISENTRESS 400 mg wordt volledig vergoed. Voor prijzen: zie ZI-index. Raadpleeg de volledige productinformatie (SPC) alvorens ISENTRESS voor te schrijven. 31 augustus 2010. Merck Sharp & Dohme BV Waarderweg 39 2031 BN Haarlem Tel.: 023 - 5153 153 www.msd.nl Verkorte 1b-tekst Viramune® 200 mg tabletten, Viramune® 400 mg tabletten met verlengde afgifte, Viramune® 100 mg tabletten met verlengde afgifte en Viramune® 50 mg/5 ml suspensie voor oraal gebruik Farmacotherapeutische groep: antivirale middelen voor systemisch gebruik , ATC: J05AGO1. Samenstelling: 1 tablet Viramune 200 mg tabletten bevat 200 mg nevirapine-anhydraat (actieve stof). 1 tablet Viramune 400 mg tabletten met verlengde afgifte bevat 400 mg nevirapine-anhydraat. 1 tabletViramune 100 mg tabletten met verlengde afgiftebevat 100 mg nevirapine-anhydraat. Suspensie voor oraal gebruik bevat 10 mg/ml nevirapine (actieve stof). Indicatie: Viramune is geïndiceerd in combinatie met andere antiretrovirale geneesmiddelen voor de behandeling van HIV-1 geïnfecteerde volwassenen en kinderen ongeacht de leeftijd. Dosering: Gewenningsdosering gedurende de eerste 2 weken: eenmaaldaags 200 mg nevirapine. Onderhoudsdosering: 400 mg per dag. Bij starten van Viramune behandeling dient altijd een gewenningsperiode van 2 weken in acht genomen te worden om de kans op huiduitslag te beperken. Deze gewenningsperiode geldt ook indien de behandeling langer dan 1 week onderbroken is. Bij patiënten die tijdens de 14-daagse gewenningsperiode huiduitslag ontwikkelen, mag de dosering niet worden verhoogd voordat de huiduitslag volledig verdwenen is. Alvorens te beginnen met deViramune-therapie, en daarna met regelmatige tussenpozen tijdens de therapie, dient klinisch laboratoriumonderzoek waaronder leverfunctietesten, te worden uitgevoerd. Voor patiënten van 16 jaar en ouder, of met een gewicht van meer dan 50 kg of met een lichaamsoppervlak groter dan 1,25 m2, is de dosering gedurende de eerste 14 dagen 200 mg per dag. Na twee weken wordt de dosering verhoogd naar 400 mg per dag. Voor patiënten jonger dan 16 jaar die minder dan 50 kg wegen of minder dan 1,25 m2 lichaamsoppervlak hebben kan suspensie worden gegeven op basis van of het lichaamsgewicht of het lichaamsoppervlak (zie volledige 1b tekst). De dosering op basis van lichaamsgewicht is voor kinderen tot 8 jaar 4 mg/kg éénmaal daags gedurende twee weken, gevolgd door 7 mg/kg tweemaal daags. De aanbevolen dosering voor patiënten van 8 tot 16 jaar is 4 mg/kg éénmaal daags gedurende twee weken, gevolgd door 4 mg/kg tweemaal daags. De dosering op basis van lichaamsoppervlak (formule van Mosteller) is 150 mg/m2 eenmaal daags gedurende de eerste twee weken gevolgd door 150 mg/m2 tweemaal daags. Contra-indicaties: Overgevoeligheid voor het werkzame bestanddeel nevirapine of voor één van de hulpstoffen. Kruidenpreparaten die sint-janskruid (Hypericum perforatum) bevatten dienen niet tegelijk met Viramune te worden gebruikt. Gelijktijdig gebruik van Viramune met rifampicine of ketoconazol wordt niet aanbevolen. Viramune dient niet opnieuw te worden toegediend aan patiënten bij wie de behandeling definitief moest worden gestaakt vanwege ernstige huiduitslag, huiduitslag gepaard gaande met constitutionele symptomen, overgevoeligheidsreacties, of klinische hepatitis ten gevolge van Viramune gebruik. Viramune dient niet te worden gebruikt bij patiënten met ernstig leverfalen. Indien AST of ALT tijdens de behandeling stijgt tot > 5 keer de bovengrens van de normaalwaarde dient Viramune direct te worden gestaakt. Wanneer AST en ALT genormaliseerd zijn en wanneer de patiënt geen klinische verschijnselen of symptomen heeft gehad van hepatitis, huiduitslag, constitutioneel eczeem of andere verschijnselen die wijzen op een gestoorde orgaanfunctie, dan is het mogelijk Viramune te herintroduceren, per geval beoordeeld, met een startdosering van 200 mg per dag gedurende 14 dagen gevolgd door 400 mg per dag. In deze gevallen is een meer frequente levercontrole vereist. Als de leverfunctie-afwijkingen terugkeren, dient Viramune definitief te worden gestaakt. Waarschuwingen en voorzorgen: De eerste 18 weken van de behandeling met Viramune is een kritische periode, die een nauwkeurige controle van de patiënt vereist om het mogelijk optreden van ernstige en levensbedreigende huidreacties of ernstige hepatitis of leverfalen uit te sluiten. Het hoogste risico op leveraandoeningen en huidreacties bestaat gedurende de eerste 6 weken van de therapie. Bij patiënten met een detecteerbare plasma HIV-1 viral load (≥50 kopieën/ml) zijn vrouwelijk geslacht en hogere uitgangswaarden van CD4+ cellen bij het begin van de therapie risicofactoren voor levercomplicaties. Mannen met meer dan 400 CD4+ cellen/mm3 en vrouwen met meer dan 250 CD4+ cellen/mm3 die een detecteerbare viral load hebben bij aanvang van de behandeling, dienen alleen te starten metViramune behandeling als de voordelen opwegen tegen het risico. Huidreacties: Er zijn ernstige, levensbedreigende en zelfs fatale, huidreacties opgetreden bij patiënten die behandeld werden met Viramune. Hieronder waren gevallen van het Stevens-Johnson syndroom (SJS), toxische epidermale necrolyse (TEN) en overgevoeligheidsreacties gekenmerkt door huiduitslag, constitutionele verschijnselen en verminderd functioneren van inwendige organen. De behandeling met Viramune moet gestaakt worden bij patiënten die ernstige huiduitslag ontwikkelen, of huiduitslag vergezeld van constitutionele symptomen, zoals koorts, blaarvorming, orale laesies, conjunctivitis, oedeem in het gezicht, zwellingen, spier- of gewrichtspijn of algehele onbehaaglijkheid. Vrouw en blijken een hoger risico te hebben op het ontwikkelen van huiduitslag dan mannen, ongeacht het gebruik van Viramune. De patiënt dient er nadrukkelijk op te worden gewezen dat huiduitslag de belangrijkste bijwerking van Viramune is en moet geadviseerd worden huiduitslag onmiddellijk aan de arts te melden. Huiduitslag die samenhangt met het gebruik van Viramune treedt meestal op in de eerste 6 weken na aanvang van de therapie. Daarom dienen patiënten tijdens deze periode zorgvuldig gecontroleerd te worden op het optreden van huiduitslag. Hepatische reacties: Bij patiënten die worden behandeld met Viramune is ernstige en levensbedreigende hepatotoxiciteit, met inbegrip van fatale hepatische necrose, voorgekomen. Er is melding gemaakt van afwijkende leverfunctietests tijdens gebruik van Viramune, ook in de eerste weken van de therapie. Omdat er in enkele gevallen, gedurende de eerste weken na aanvang van de therapie met Viramune, melding is gemaakt van klinische hepatitis dient bepaling van ALT en AST de eerste twee maanden van de behandeling elke twee weken plaats te vinden. In de derde maand dient de controle van de leverfunctie éénmaal, en vervolgens regelmatig plaats te vinden. Overig: Voorzichtigheid is geboden wanneer Viramune wordt voorgeschreven aan zwangere vrouwen. Viramune kan de plasmaconcentraties van hormonale anticonceptiva doen afnemen. De patiënt dient geadviseerd te worden om naast een hormonaal anticonceptivum ook altijd een ander voorbehoedsmiddel te gebruiken. Nevirapine induceert het leverenzym CYP3A en mogelijk 2B6. Middelen die door CYP3A en/of CYP2B6 worden gemetaboliseerd kunnen verlaagd zijn als ze samen met Viramune worden gebruikt. Bij gelijktijdig gebruik van Viramune met fluconazol, warfarine, methadon of claritromycine dient de patiënt nauwkeurig te worden gecontroleerd. Bijwerkingen: De meest frequent gerapporteerde bijwerkingen van Viramune zijn huiduitslag, allergische reacties, hepatitis, afwijkende leverfunctie testen, misselijkheid, braken, diarree, buikpijn, vermoeidheid, koorts, hoofdpijn en spierpijn. De meest voorkomende bijwerking van Viramune is huiduitslag. De huiduitslag bestaat gewoonlijk uit milde tot matig-ernstige, maculopapuleuze, erythemateuze huiduitslag, met of zonder jeuk, op de romp, het gezicht en de armen en benen. Ernstige en levensbedreigende huidreacties, waaronder het Stevens-Johnson syndroom (SJS) en toxische epidermale necrolyse (TEN) zijn opgetreden. Bij patiënten die lever en/of huidreacties ondervinden ten gevolge van Viramune gebruik is rhabdomyolyse waargenomen. Distributie: UR. Vergoeding: Viramune wordt volledig vergoed binnen het GVS. Nadere informatie: de volledige 1b-tekst, productinformatie en publicaties zijn verkrijgbaar bij Boehringer Ingelheim bv, Comeniusstraat 6, 1817 MS Alkmaar, telefoon 0800-2255889. Datum laatste herziening: september 2011. Report 6th Workshop on HIV Transmission Principles of Intervention 14-15 July 2011 in Rome, Italy Report Report on 6th Workshop on HIV Transmission J. Albert1, C. Boucher2, M. Cohen3, W. Heneine4, E. Hunter5, P. Kanki6, B. Mathieson7, G. Scarlatti8, R. Swanstrom9, M. Wainberg10, A. Wensing11 Karolinska Institute, Stockholm, Sweden; 2Erasmus University Rotterdam, The Netherlands; 3University of North Carolina, Chapel Hill, NC, USA; 4Division of HIV/AIDS Prevention, CDC, Atlanta, GA, USA; 5Emory University, Atlanta, GA, USA; 6Harvard School of Public Health, Boston, MA, USA; 7Office of AIDS Research at the National Institutes of Health,Bethesda, MD, USA; 8San Raffaele Scientific Institute in Milan, Italy; 9University of North Carolina, Chapel Hill, NC, USA; 10McGill University, Montreal, Canada; 11University Medical Center Utrecht, The Netherlands 1 Written by Wendy Smith on behalf of the Organizng Committee HIV Transmission – Human Biology Cell biology of HIV sexual transmission Dr Hope (Northwestern University, USA) addressed the topic of the cell biology of sexual transmission, especially the movement of the virus from the lumen of the female genital tract to the target cells, such as T cells and dendritic cells in the sub basal layer of the epithelium1. The process is very rapid: in macaque models, simian immunodeficiency virus (SIV) infections can be established within 30-60 minutes of vaginal challenge. Using virus carrying a photoactivatable GFP fused to Vpr, it has been possible to visualise the virus moving through the vaginal epithelium of explant cultures. Considerable variation was observed in the rate of movement of the viruses from the lumen through the epithelium: most of the viral particles only penetrate a short distance while some are able to penetrate ~50 microns. HIV is able to breach the columnar epithelium of the vagina and access the underlying target cells. Virus often becomes trapped in the mucus that lines the vaginal epithelium and protects the endocervix from infection. When neuraminidase was used to cleave sugars from mucus, HIV was able to penetrate the endocervix more easily. HIV penetrates between the cell junctions of the epithelium (i.e. interstitial diffusion) rather than directly entering the cells. If EDTA is applied to break down the cell junctions, HIV is able to penetrate the epithelium more deeply. Dr Hope’s group is now studying interactions between HIV and tissue obtained from circumcised men in an attempt to understand why circumcision is protective against HIV infection in men. Founder viruses Dr Keane (Murdoch University, Australia) presented a study of an acute heterosexual transmission of HIV-12. A 51 year old, East African woman presented to the clinic five days after non-consensual sexual contact with a man who was known to be HIV infected. p24 antigen was detected in the woman’s blood but the EIA and Western Blot results were negative, indicating early HIV infection prior to seroconversion. The woman had tested HIV negative in 2003 and reported no subsequent risky exposures, apart from the non-consensual sex. The woman started antiretroviral therapy (ART) one year after infection and her viral load dropped to ~50 copies/mL within a few weeks, after having being 70,000 copies/mL in the absence of therapy. Three founder/transmitted Disclosure: Wendy Smith, PhD has disclosed that she does not serve as a consultant for any commercial companies. Dr Smith is a professional medical writer who works for Wordsmiths International Ltd (www.wordsmiths. eu.com) 6 viruses were identified 15 days post-transmission; it is possible that more than one founder virus was transmitted because of the traumatic nature of the sexual contact. There appeared to be transmission of an adapted epitope in the shared recipient and donor HLA C*04:01 restricted epitopes. Phylogenetic analysis demonstrated that the viral strains from the donor and recipient were more closely related to each other than to any strains in the dataset. Limited HIV viral sequence change was observed in patient/ donor HLA-restricted epitopes. Non-selective CTL responses were observed and may have contributed to the rapid rate of disease progression in this case. Compartmentalisation of HIV Paired blood and cervicovaginal lavage (CVL) samples were obtained at Day 0 and Week 16 from 28 women from Malawi and S Africa with chronic HIV infection3. Viral loads were measured and single genome amplification and phylogenetic analysis of env genes were carried out. The Slatkin-Maddison test for compartmentalisation was applied. The CVL viral loads were significantly higher in women Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 6th Workshop on HIV Transmission who had sexually transmitted infections (STI) than in those without STIs, even though the plasma viral loads were similar in both groups. Genetic characterisation of HIV populations in the 10 paired blood and genital tract samples demonstrated that clonal amplification of the virus had occurred in each compartment. A significant amount of genetic difference was observed between paired plasma and CVL viral samples. Compartmentalisation was observed in 60% of subjects and it was significantly associated with higher CD4 cell counts, suggesting that it was associated with the availability of target cells and not due to reduced immunosurveillance in a specific compartment. Longitudinal data suggest that the HIV viral load in the female genital tract is variable over time. Dr Dukhovlinova (University of North Carolina at Chapel Hill, USA) suggested that the presence of a STI could affect the cytokine profile of the female genital tract and facilitate the influx of cells from nearby lymphoid tissue. Further studies are in progress to identify the cytokine profile and tropism of compartmentalized viral strains. Cytokine network in semen of HIV positive men Semen is a complex fluid that contains spermatozoa and a number of soluble factors, including cytokines. The replication of pathogens, such as HIV and herpesviruses, affects cytokine expression as well as is affected by cytokines. Blood and seminal fluid were obtained from 74 HIV-1-infected, ART naïve men and 33 uninfected men: viral loads and cytokine profiles were evaluated4. A relationship was observed between the HIV-1 viral load in the plasma and the semen. No reactivated herpesviruses were detected in samples from HIV negative men, with the exception of low herpesvirus viraemia (CMV, HHV6 and HHV-7) in some semen samples. In HIV-1 infected men, EBV and CMV were detected in the majority of semen samples and their viral loads were significantly higher in the semen than the blood. HSV2, HHV-6, HHV-7 and HHV-8 were detected in HIV-1 infected men’s semen but not in their blood. CMV reactivation was largely compartmentalized in the HIV-1/CMV co-infected men: in 77% of these men, CMV was only found in semen compared to 19% who had detectable CMV levels both in blood and semen. Concurrent EBV and CMV infection was associated with significantly higher levels of HIV-1 shedding in the semen than in HIV-infected men co-infected with only one of these herpesviruses. In HIV-infected men, HIV preferentially upregulated cytokines in semen compared to blood. Based on the results of this study, the investigators (NIH/NICHD, USA) concluded that HIV-1 infection is associated with seminal shedding of CMV and EBV and rearrangement of the cytokine network in semen. These factors may affect the sexual transmissibility of HIV-1. Viral escape – possible mechanism for immune escape Several studies have demonstrated the importance of HIV-1 gp120 variable regions in virus infectivity and immune escape; the majority of these experiments have been carried out on subtype B virus but the most prevalent subtype globally is subtype C5. Virus isolated from recently infected subjects tends to have shorter V1-V2 loops and a lower number of putative N-glycosylation sites (PNGS) than variants isolated during chronic infection. Dr Cenci (Istituto Superiore di Sanita, Italy) reported the results of an analysis of gp120 variable V1-V5 and C3 and C4 constant regions from clade C viruses isolated from patients at different disease stages. During chronic infection, the sequence of amino acids of the V1 and V4 regions and the number of PNGS increased.New PNGS shifted appeared in the C-terminus of all the variable regions with the exception of V3, and in the constant regions near to the CD4 binding site, possibly protecting the virus from the immune response. During both recent and late stage infection, a positive charge correlation was observed between C3 and V5: this could support the hypothesis that an open conformation would be the best fit for binding to cellular receptors. At the contrary, the presence of a negative correlation of positive electric charges between the C3 and V4 regions in the Chronic stage could protect the C3-V4 epitope by immune system. HIV Transmission – Model Systems A number of model systems, such as ex vivo cervical tissue cultures, macaques and humanised mice, have been used to investigate the transmission of retroviruses and potential methods of protecting against infection. Modelling HIV transmission and dissemination SIV infection of rhesus macaques can be used to model HIV transmission and systemic dissemination of the virus, as well as investigating the effectiveness of candidate vaccines and other HIV prevention strategies6. Both single and limited number variant infections can be simulated, using the rectal, vaginal or penile route. SIV env and full length clones of founder viruses have been generated and shown to be functional in in vitro and in vivo assays, thus enabling investigators to analyse the particular characteristics of founder viruses. Transmitted founder clones recapitulate viral load profiles and the pathogenetic features of the parental virus in Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 7 Report naïve rhesus macaques. Longitudinal analyses of putatively single variant infections confirmed that the transmission event involved one viral variant, regardless of the route of infection. A model was established to enable Keele and colleagues (SAIC-Frederick/NIH, USA) to track SIVmac239 infection following intravaginal challenge so that the key events in early replication and systemic dissemination could be dissected6. Synonymous mutations were introduced into a viral isolate to create ten clones that were molecular but isogenic tagged and introduced into female macaques as a nontraumatic vaginal challenge. Real time SGA sequence analysis was used to identify the tagged clones in the blood and tissue of euthanized macaques. There was considerable inter-animal variability in terms of the number of clones that were transmitted; in the majority of infections, one major variant and several minor variants were identified. Infections were multifocal and the virus does not appear to be uniformly distributed. Virus could be found in draining and then distal lymphoid tissues before it was distributed to the plasma, peripheral blood monocyte cells, gastro intestinal tract or spleen. Not all of the variants that were identified at mucosal sites were found in peripheral sites. The initial results were obtained from unsynchronised animals but current experiments are using macaques with synchronised menstrual cycles so that the impact of hormones on SIV transmission can be identified. Productive HIV-1 infection of cervical tissue ex vivo The human cervix is a major route by which women acquire HIV from their sexual partners (Figure 1). Dr Saba (San Raffaele Institute, Italy) has used human cervical tissue explants to study the process of HIV infection ex vivo7. The average age of the donor was 50±1 years old. In this cervical tissue model, X4 HIV-1 was only able to establish a productive infection in a few tissues that were enriched with early differentiated effector memory CD4 T cells whereas R5 HIV-1 infection was supported efficiently. During R5 infection, the CD4 T cells appeared to be HIV-1 gag positive at an earlier stage of infection than the macrophages. R5 HIV-1 replication in the cervical explants was modulated by the phase of the menstrual cycle. There appeared to be a ‘window of vulnerability’ during the secretory stage of the cycle (i.e. when progesterone levels were reaching a peak) when the virus was able to infect the female genital tract tissues more easily, possibly because the release of progesterone caused changes in the mucosa that facilitated infection. Infection was less likely during the proliferative phase of the menstrual cycle and during the menopause. Characterisation of semen leukocytes Dr Bernard-Stoecklin (Comissariat a l’Energie Atomique, France) has investigated whether semen leukocytes act as ‘Trojan horses’ in SIV/HIV mucosal transmission by analysing the infection process in macaques8. In humans or animals with primary infection, the CD4 T cell count in the semen falls rapidly whereas it tends to increase when ART is administered during chronic infection. Lymphocytes in the semen are activated and express higher levels of CD69 (a proliferation marker) and HLADR (a late activation marker) than lymphocytes in the blood. HIV-1 heterosexual transmission in human cervix DCs CD4+ T cells Figure 1. HIV-1 heterosexual transmission in human cervix Source: Dr Saba, San Raffaele Institute, Italy 8 Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement Macrophages 6th Workshop on HIV Transmission They express a number of chemokine receptors and integrins. T cells in the semen are almost all central memory cells, which are the main targets for SIV. They are present in the semen at every stage of the infection process. In summary, seminal leukocytes express markers that may favour HIV/SIV transmission. The impact of semen on HIV infectivity of target cells has been controversial. The effects of semen on CD4 T cell phenotype and susceptibility to HIV infection were investigated by Balandya (Dartmouth Medical School, USA) et al9. They collected semen from HIV negative men and incubated centrifuged seminal plasma from these samples with peripheral blood monocyte cells and X4 or X5 HIV. Levels of HIV infection were assessed using TZM-bl cells and intracellular staining for HIV p24. The results of these experiments indicate that seminal plasma protects CD4 T cells from HIV infection and reduces T cell surface CD4 expression. Seminal plasma also induces CD4 T cell expression of the CCR5 receptor and mediates the preferential transmission of R5 HIV. These data show that semen protects cardinal target cells from HIV infection and contribute to the previously unexplained preferential transmission of R5 HIV. Pre-Exposure Prophylaxis (PrEP) models Humanized mice (RAG-hu), which lack native B, T and NK cells, have been utilised as a model system to investigate the potential of maraviroc and raltegravir to protect against HIV-1 vaginal transmission10. The mice were engrafted with human CD34 cells and multilineage human haematopoiesis was subsequently detected in several organs. HIV-1 was able to infect RAG-hu mice via the vaginal or rectal mucosal tissues. Oral administration of maraviroc or raltegravir for three days prior and post challenge provided 100% protection against HIV-1 infection. CD4 T cell counts declined rapidly in infected, non-treated control animals but the cell counts in the treated mice remained similar to the levels in uninfected control animals. Additionally, maraviroc was formulated as a topical microbicidal gel; it was completely protective against HIV-1 vaginal transmission in RAG-hu mice. The pharmacokinetic and pharmacodynamic profile of a single oral dose of maraviroc (44 mg/kg) in rhesus macaques was studied by Garcia-Lerma and colleagues (CDC, USA)11. Plasma levels of maraviroc peaked two hours after administration but levels in vaginal and rectal secretions peaked at 5 and 24 hours, respectively. Maraviroc was measurable in vaginal and rectal secretions for up to two and seven days, respectively, suggesting that this antiretroviral (ARV) may be useful for PrEP. The pharmacokinetics of maraviroc in macaques were similar to those in human beings who had taken 300 mg maraviroc. A co-receptor occupancy assay was used to analyse maraviroc binding to CCR5. Levels of maraviroc that were found in the rectal tissues 2-3 days after administration were associated with the maximum levels of CCR5 occupancy and protection against infection in vitro. Vaginal absorption of ARVs – the pig tailed macaque model The efficacy of ARVs as microbicides probably depends on how effectively they are absorbed into the vaginal mucosa. During the menstrual cycle, a number of physiological changes occur in the vaginal epithelium that may affect drug absorption (Figure 2)12. Female pig tailed macaques have a bi-phasic menstrual cycle, similar to humans, and so they can be used as a model to examine the effects of menstrual cycle on vaginal drug absorption. During the follicular phase, the vaginal epithelium is thick and keratinised; during the luteal phase, it is thin and more susceptible to HIV infection. A gel containing 5% FTC and 1% tenofovir was applied vaginally once a week for four weeks and then drug levels were measured in the plasma at regular intervals. After a two week washout period, the experiment was repeated but the gel was applied rectally. In the vaginal dosing experiments, there were higher levels of ARV drug absorption during the luteal phase than the follicular phase. When the gel was applied to the rectum, drug absorption was unaffected by the menstrual phase. Maximum levels of drug absorption were observed within 7-10 days of peak progesterone levels and were not drug-dependent. It appears that physiological changes during the menstrual cycle, such as vaginal epithelial thinning, may explain the absorption of ARVs formulated as a microbicide. Prevention of HIV Transmission – Antiviral and Medical Approaches Preventing new HIV infections is an essential component of controlling the global HIV epidemic. Prevention strategies include antiviral approaches, e.g. microbicides, and medical interventions, e.g. use of ARVs to prevent transmission of HIV from mother to child. PrEP, using single or combined ARVs, has been studied as a method of preventing the transmission of HIV from an HIV positive person to his/her sexual partner(s). Preventing sexual transmission of HIV-1 CAPRISA 004 assessed the safety and effectiveness of a 1% tenofovir gel used as a microbicide up to Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 9 Report Follicular Phase Luteal Phase Estradiol Progesterone Day 1 Ovulation 7 Thick, keratinized vaginal lining 14 21 28 Thin, non-keratinized, porous vaginal lining Figure 2. Menstrual cycle phases Source: Dr Heneine, Centers for Disease Control, USA 12 hours before sex and within 12 hours after sex13. The effectiveness of the microbicide at 12 months was 50% and at 30 months it was 39%. The gel also provided 51% protection against herpes virus 2 (HSV-2) infection which infects 50-60% of S African sexually active adults. The results of CAPRISA 004 were hailed as the new hope in HIV prevention, even though the gel did not provide complete protection. Subsequent studies (Figure 3) have shown the following successes: • oral PrEP with tenofovir/FTC prevented HIV infections in men who have sex with men (MSM), with an effectiveness of 44% at the end of the study (iPrEx trial); • ART prevents HIV transmission between sexual partners, with an effectiveness of 96% (HPTN 052); • oral PrEP with daily oral tenofovir or tenofovir/FTC provided 62% and 73% protection in serodiscordant couples, respectively (Partners PrEP); • daily oral tenofovir/FTC provided 63% protection in heterosexual men and women (Botswana TDF2 study). In contrast, the FEM-PrEP study of oral tenofovir/FTC as PrEP in women was halted for futility. The reasons for the study’s outcome of no protection against HIV are being investigated but it is possible that there was poor adherence or inadequate drug levels were present at the possible site of infection. 10 Dr Abdool Karim (University of KwaZulu-Natal, South Africa) provided several additional insights from the CAPRISA 004 trial13. In macaques, genital inflammation has been associated with an increased risk of SIV acquisition. Tenofovir gel use was not associated with elevated levels of genital inflammation compared to placebo. However, genital tract inflammation prior to infection was associated with an increased susceptibility to HIV infection. Achieving detectable tenofovir levels in the female genital tract was associated with a significantly increased level of protection in women without elevated cytokine levels (p=0.05). The effectiveness of the tenofovir gel was not affected by the person’s HSV-2 status. However, one possible hypothesis is that men with high semen viral loads were more likely to transmit HIV than those with low semen viral loads, possibly suggesting that tenofovir gel is less effective if the viral load in the semen is high e.g. in men with acute or early infection. HIV transmission in the vagina appears to be influenced by the host (increased genital inflammation may be associated with increased HIV acquisition); the agent (higher semen viral load may be associated with increased risk of infection); and the environment (higher drug levels in the vagina may reduce the risk of infection). The results of the CAPRISA 004 study have been used to model the effects of widespread use of a 1% tenofovir gel microbicide in S Africa overall and specifically in KwaZulu Natal province14. The Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 6th Workshop on HIV Transmission Study Treatment for prevention Effect size (CI) 96% (73; 99) (Africa, Asia, America’s) PrEP for discordant couples 73% (49; 85) (Partners PrEP) PrEP for heterosexuals 63% (21; 48) (Botswana TDF2) Medical male circumcision 54% (38; 66) (Orange Farm, Rakai, Kisumu) PrEP for MSMs 44% (15; 63) (America’s, Thailand, South Africa) STD treatment 42% (21; 58) (Mwanza) Microbicide 39% (6; 60) (CAPRISA 004 tenofovir gel) HIV Vaccine 31% (1; 51) (Thai RV144) 0% 10 20 30 40 50 60 Efficacy 70 80 90 100% Figure 3. Summary of clinical trial evidence for preventing sexual transmission of HIV in July 2011 Source: Dr. S. Abdool Karim, University of KwaZulu-Natal, Durban, South Africa13. investigators assumed that the presence of HSV2 infection increases the risk of acquiring HIV by 2-3 fold and, in co-infected people, increases HIV infectivity by 30-70% per act. HIV infection increases HSV-2 shedding episodes. In KwaZulu Natal, the number of prevented infections of HIV or HSV-2 would be substantially greater in women than men. The proportion of HSV-2 infections prevented would be approximately independent of the gel’s effectiveness against HIV but it would be affected by coverage (i.e. access to the microbicide). The relationship between HIV effectiveness and coverage on the proportion of HIV infections prevented would be non-linear. On a country-wide scale, the gel would prevent more HIV infections in provinces where the infection rate is high i.e. many more infections would be prevented in KwaZulu Natal than in Northern and Western Cape if the microbicide coverage was assumed to be 90% and the effectiveness was assumed to be 54%. Only 113 women-years of microbicide usage would be required to prevent one HIV infection in women in KwaZulu Natal, while it would require 445 womenyears of usage in Western Cape. Dr Blower (UCLA, USA) concluded by stating that, in the light of financial constraints, a very effective control strategy would be to target the microbicide roll out to provinces with high HIV infection rates. HPTN 052 The results of the HPTN 052 studied were published in the online version of the New England Journal of Medicine on 18 July 2011; Dr Cohen (UNC Chapel Hill, USA) provided an overview of the data to the Workshop15, 16. The aim of the study was to determine if providing ART to HIV infected individuals protected their serodiscordant sexual partners from acquiring the virus; and to evaluate the optimal time to initiate ART in order to reduce morbidity and mortality. A total of 1,763 serodiscordant couples were enrolled. The HIV+ partner had to have a CD4 cell count of 350-550 cells/mm3 and he/she was randomised to receive immediate ART or delayed ART (when the CD4 cell count was <250 cells/mm3). On 28 April 2011, the Data Safety Monitoring Board recommended that the results of the trial be announced as soon as possible. The study was not stopped but modifications to the study design are in progress: all HIV positive participants are now being offered ART. During the trial, 39 infections occurred: 28 were linked and 27 of these occurred in the delayed therapy arm and one in the immediate therapy arm (p<0.001). Seventeen infections occurred in the delayed ART arm when the index partner’s CD4 cell count was >350 cells/mm3. A total of seven unlinked infections occurred: four in the delayed ART arm and three in the immediate ART arm. Four infections are still being analysed for evidence of linkage. The difference between the two arms in terms of morbidity and mortality did not meet the pre-set criterion of >20% but it was significant when no preset value was used (p=0.01): 65 events occurred in the delayed arm and 40 in the immediate arm15. The difference was driven by cases of extrapulmonary TB: 17 in delayed ART arm and three in the immediate Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 11 Report arm (p=0.0013). There were a similar number of deaths in each arm: 13 and 10, respectively. The study will continue for at least one more year to determine the durability of the prevention benefit; any differences between the two arms in terms of prevention benefit; clinical and toxicity events in both arms; and the level of adherence to ART. Dr Cohen explained that this was a proof of concept study, not a public health study: ART was effective in preventing HIV transmission between monogamous couples in this study. Evaluating the effect of ‘Test and Treat’ on HIV transmission ‘Treatment as Prevention’ is the cornerstone of the WHO’s Treatment 2.0 initiative and a number of pilot studies are in progress to evaluate the impact of expanded access to treatment on HIV incidence at the population level. Dr Vasarhelyi (BC Centre for Excellence in HIV/AIDS, Canada) described her group’s efforts to develop a general performance indicator for Seek and Treat strategies using surveillance data17. The indicator was then applied to surveillance data collected 2003-9 (before the STOP HIV/AIDS launch) in British Columbia, a Canadian province, and to simulated data for 2011-17. The outcome of the model was that the proportion of diagnosed HIV infections was approximately constant between 2003 and 2009 in British Columbia. To demonstrate the performance of the indicator in detecting changes in the proportion of diagnosed cases, the model was applied to simulated data for 2011-17. The data were consistent with an annual increase of 1% in the proportion of diagnosed HIV infections since the launch of STOP HIV/AIDS. The development of an indicator for Seek and Test programmes will enable the performance of such programmes to be evaluated at the population level. Activity of griffithsin plus ARVs against subtype C HIV Griffithsin is a mannose-specific lectin that is being investigated as a potential microbicide18. The antiviral potency of griffithsin in combination with tenofovir, maraviroc or enfuvirtide against HIV-1 subtypes B and C, as well as a subtype B X4 virus, was assessed in a range of assays. All of the combinations were synergistic and provided superior antiviral potency against all of the strains studied, compared to griffithsin alone. Antagonism between the therapeutic agents was not observed. Griffithsin plus one or more ARVs has potential as a microbicide; studies are ongoing in the USA to determine the safety and efficacy of this approach. 12 Circumcision The 2009-10 Demographic and Health Survey (DHS) data from Lesotho have been analysed to determine the current prevalence of male circumcision and associated demographic factors for circumcised males; as well as to determine if male circumcision has limited the spread of HIV19. Data on the HIV and circumcision status of 3,074 men was collected: the HIV prevalence was 18% in men aged 15-59 years; and ~56% of men were circumcised. Surprisingly, the HIV prevalence was higher in circumcised men (20%) than uncircumcised men (15%). Male circumcision was more common in rural areas (61%) than urban areas (36%) but HIV prevalence was lower in rural areas compared to urban areas: 17% vs. 20%. The majority of men (79%) lived in rural areas. Only ~8% of circumcisions were carried out using clinical methods, while 92% were carried out with traditional methods, usually at an initiation school. This method does not completely remove the foreskin, and so may be less protective against HIV acquisition than medical circumcision. Approximately half of the men were circumcised after becoming sexually active. The investigators concluded that the use of traditional methods of circumcision, especially after men have become sexually active, might limit the protective effect of male circumcision in Lesotho. Prevention of Mother to Child Transmission of HIV (PMTCT) An HIV infected pregnant woman can transmit HIV to her baby at any point during pregnancy, labour and delivery or breastfeeding. Prevention strategies are therefore needed throughout this long period of time, Dr Scarlatti (San Raffaele Institute, DIBIT, Italy) pointed out20. Administering combinations of ARVs during pregnancy, labour and delivery and avoiding breastfeeding have reduced MTCT rates to almost zero in the developed world. The use of single dose nevirapine changed the perception that it was impossible to prevent MTCT in developing countries. Although its use was associated with the emergence of NNRTI resistance, it not only reduced the number of paediatric HIV infections but also led to PMTCT initiatives in resource limited settings (RLS) using ARV combinations. Since breastfeeding contributes to paediatric health in RLS, a number of ARV-based interventions have been developed to protect a baby from HIV infection during breastfeeding. Only a few viral variants are detected in newborns, regardless of the route of transmission (Figure 4). The transmitted variant is either a major or minor maternal viral variant. Viruses that are transmitted intra partum (during labour and delivery) tend to have shorter variable loops and fewer PNG sites than those that are transmitted during pregnancy. R5 virus is the Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 6th Workshop on HIV Transmission mother Pregnancy Placenta Amniotic fluid Labour Delivery Breast feeding !! a SELECTIVE barrier? Blood Secretions Milk Blood Only FEW virus variants detected in the newborn! child Figure 4. Is MTCT a selective or stochastic process? Source: Dr. G. Scarlatti, San Raffaele Institute, Milan, Italy20 predominant transmitted virus in MTCT: children who are infected with R5broad phenotype are more likely to experience early immunological failure than those who are infected with R5narrow phenotype virus. R5 viruses preferentially cause dendritic cells to extend cellular processes through the intestinal mucosa, thus enabling the R5, but not the X4, viruses to cross the epithelial layer. Dendritic cells that are infected with HIV also act as a local viral reservoir at the mucosal level. Understanding the precise mechanisms by which MTCT occurs will assist in the efforts to prevent children being infected with HIV. The topic of HIV incidence in pregnancy and the first post partum year was addressed by Dr Lu (CDC, USA)21. Routine HIV antibody testing during antenatal care may miss acute infections that are still in the window period (prior to the development of antibodies) or infections that occur during pregnancy. New HIV infections during late pregnancy and breastfeeding are associated with a very high risk of MTCT. In 2010, Dr Lu and colleagues undertook a study of 417 women who had had negative HIV tests during pregnancy and who were approximately one year post partum. The one year HIV incidence in these women was 3.8%. Risk factors for HIV infection were evaluated with a behavioural survey. Although 20% of the women had had more than one sexual partner in the previous two years, this finding was not associated with HIV infection. Women who knew the HIV status of their partner were significantly less likely to acquire HIV than those who did not know. Nearly half (183/417, 43.9%) of the women had been tested together with their partner. Extrapolating these data to the total Batswana population, Dr Lu stated that while the National program found 647 infant infections through the Early Infant Diagnosis program, as many as 1329 infant HIV infections may have occurred in 2010, with 672 (50.6%) due to incident cases of maternal HIV infection that would not have been detected in routine antenatal care. Of the 17 infants born to seroconverting women during the study, three were found to be HIV positive: one baby died before ART was initiated; one died after ART was initiated; and one was alive and taking ART when the study was completed. Dr Lu commented that these data have implications for PMTCT programmes since women continue to be at risk for HIV infection during pregnancy and while breastfeeding, despite a high uptake of antenatal HIV testing and counselling. Women who test HIV negative during pregnancy will breastfeed their babies, breastfeeding babies are at high risk for mother-to-child transmission in the setting of an acute maternal HIV infection. Women are likely to use condoms for protection. However, couples testing and counselling tends to be underused. Many women were not aware of their partners’ HIV status, which was the only significant risk factor found for incident HIV, even in this small cohort. Based on these data, it has been suggested that HIV negative women should be retested when they bring their babies to the clinic for immunization (4 months post partum) or earlier if other risk factors are present. Educational material needs to be modified to explain the risks of HIV infection during pregnancy and breastfeeding. Access to family planning, STI Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 13 Report clinics and couples testing and counselling needs to be improved. Modes of Transmission and HIV Prevention Efforts in Vulnerable Populations Drug users Obtaining precise information on the number of intravenous drug users (IVDU) globally is challenging but there are believed to be approximately three million IVDUs living with HIV worldwide22. HIV transmission occurs by sharing contaminated syringes with other IVDUs as well as unprotected sex with HIV positive partners. Other factors associated with HIV acquisition include unstable housing; and the intensity of use of intravenous cocaine. In certain settings, age, gender and use of crack cocaine have been shown to be risk factors for HIV infection. Preventing HIV transmission amongst IVDUs is complex, mainly because of its illicit nature, and has to take into account a wide range of structural, social and individual factors. Needle and syringe programmes, as well as opioid substitution therapy (OST), have been shown to be effective in reducing HIV transmission amongst IVDUs but political pressures often make it difficult to ensure that prevention programmes provide a complete package of interventions. Data from Australia have shown that full harm reduction programmes are very effective, while programmes that only offer a few harm reduction components are less successful. OST is only useful for people who are dependent on opioids; it is ineffective for cocaine users. Effective OST coverage is poor on a global level, even though good OST programme retain IVDUs in care for longer and OST clients are six times less likely to become HIV positive than IVDUs who do not enter treatment. Increased access to ART by IVDUs in Vancouver has been associated with decreases both in the community plasma viral load and the HIV incidence density. Despite these successes, there is a considerable resource gap – several billion dollars – between the annual amount spent on harm reduction programmes and the estimated total need. Dr Bruneau (University of Montreal, Canada) called for comprehensive healthcare packages for IVDUs. Decreases in the future HIV prevalence and disease burden among IVDUs can only be accomplished by intensifying harm reduction measures among highrisk persons and expanding health care access for those at the greatest risk of disease and progression, including access to HIV prevention technologies such as PrEP. Although IVDU is a major risk factor for HIV transmission, Dr Khayatkhoei (Teheran University, 14 Iran) pointed out that non-IV drug users also participate in high risk behaviour, such as having unprotected or transactional sex23. Dr Khayatkhoei therefore compared the prevalence of HIV in IDU and non-IV drug users in the Imamhossein Hospital Detoxification Center in Tehran, Iran, in a cross sectional study between March 2010 and April 2011. The Center is a referral centre that treats approximately 600 patients per day in the Emergency Department. In Iran, approximately two million people are addicted to drugs; 300,000 of them are IVDUs. In 2006, IVDU was the major route of HIV transmission in Iran. The study population was composed of 200 IVDUs and 200 non-IV DUs. Patients had to have been opioid drug abusers for at least three years. Of the IVDUs, 12.5% (25/200) were HIV positive while this figure was 3.5% (7/200) for non-IV DUs. The risk of acquiring HIV was 3.9 times higher in IVDUs than in non-IV DUs. Dr Khayatkhoei stressed that the HIV prevalence in non-IV DUs is a concern since 82% of all drug abusers are in the reproductive age range and therefore would be expected to be sexually active. She recommended that healthcare workers are sensitised to this issue and that HIV education programmes be developed to decrease behavioural risk factors in both IVDUs and non-IV DUs. Young girls HIV disproportionately affects young girls in subSaharan Africa. Dr Kaggwa (PACE, Uganda) explained that, in Uganda, this part of the epidemic is driven by cross generational sex (CGS): 10% of Ugandan girls aged 15-19 years had had sex with a man who was at least 10 years older in the previous 12 months (Ministry of Health survey 2006). In partnership with Johnson and Johnson, the PACE programme has implemented an anti- CGS project called ‘Go Getters’ in 50 secondary schools in four regions in Uganda. A cross sectional survey was carried out in a representative sample of 2,410 girls aged 15-19 years from 30 secondary schools to assess sexual behaviour and its determinants. Schools were classified as low category (LCS) or high category (HCS), depending on the amount of fees and rates of university entry as a reflection of the girls’ economic circumstances. CGS was more common in LCS than HCS: 9.4% vs. 2.6%. Young girls who had had CGS were more likely to report early sexual debut; have multiple older sexual partners; have low confidence in their ability to reject older men’s advances; and to perceive CGS as socially acceptable. Half of the girls had a boyfriend at the time of the survey and 22% had had sexual intercourse at some point in their life. In previous year, 22% had had sex and 20% had had sex with multiple sexual partners. Girls from HCS were more confident in their ability to reject older Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 6th Workshop on HIV Transmission men’s advances; had social support not to engage in CGS; agreed that women are more at risk of HIV if they have CGS; and had higher self esteem than girls from LCS. Based on these results, HIV prevention programmes should enhance girls’ life skills so that they are more confident in rejecting older men’s sexual advances; campaign against CGS; address the social norms that support CGS; and emphasize the risks associated with CGS. Transmitted Drug Resistance (TDR) Global trends Dr Wensing (University Medical Centre Utrecht, The Netherlands) addressed the issue of transmitted drug resistance (TDR) worldwide24. The SPREAD network is present in 34 European countries. In order to collect information on a representative set of newly diagnosed patients, prospective data are collected annually in 28 SPREAD countries from patients with evidence of recent infection (<1 year) and from patients with an unknown duration of infection. Based on these data, the prevalence of TDR remained stable in Europe between 2002 and 2007: ~8-9% for any class of ARV; ~5-6% for NRTIs; ~3-4% for NNRTIs; and ~2-3% for PIs. Nationwide data are not available for the USA but a number of city or state surveys have demonstrated a similar rate of transmitted drug resistance in various risk groups (~10%). The WHO has developed standardised methods to assess transmitted drug resistance in RLS, especially in recently infected, ART naïve populations. Between 2002 and 2009, 53 surveys had been completed in 22 countries: 83% showed low rates of TDR and 17% had moderate rates of TDR. The overall TDR prevalence was 3.7%. Of the 11 surveys conducted in 2009, five showed moderate levels of TDR, which may have implications for treatment and prevention programmes in these RLS. The majority of TDR is due to single mutations, but resistance profiles differ between treated and untreated individuals24. For example, the M184V and T215Y mutations are far more common in treated individuals than untreated individuals. It has been hypothesised that treated individuals are not the most important source of drug resistant virus, especially as ARV resistant viruses tend to be transmitted less efficiently than susceptible viruses. It is possible that drug resistant viruses evolve in the new host in the absence of drug selective pressure. After conducting a literature survey of cases of TDR, Dr Wensing and her colleagues identified three different evolutionary pathways: 1. Reversion – when the transmitted resistant virus reverted to wild type in the new host. 2. Atypical variants – when the transmitted resistant virus evolved to become an atypical variant(s). 3. Persistence – when the transmitted resistant virus persisted in the new host due to a small difference in the virus’ replicative capacity and/or the emergence of compensatory mutations that became fixed in the population. The clinical implications of TDR were studied retrospectively in the EuroCoord-CHAIN study of >10,000 patients who started ART after 1997 and for whom genotypic and clinical follow up data were available. Although the majority of the patients (90.5%) were infected with fully susceptible virus, approximately 10% were infected with TDR virus. Half of these patients were treated with a regimen that was fully active and half received a regime that the virus was resistant to. Although the level of CD4 cell recovery was similar in all of the patients, individuals who were treated with sub-optimal regimen had a higher risk of virological failure. Patients who received fully active regimens had a similar outcome to patients infected with susceptible virus if they were treated with ritonavir boosted PIs (PI/r) but patients who took NNRTIs were more likely to experience therapy failure. The reasons for this difference are not yet clear but it is possible that the higher genetic barrier to PI/r was a factor. It is also possible that the presence of NNRTI-resistant minority variants affected the clinical outcome. Monitoring TDR is a critical part of ensuring that HIV prevention and treatment programmes are successful. European trends in TDR Prof. Boucher (Erasmus Medical Centre, The Netherlands) described the results of a study by the European Society for Antiviral Resistance to understand the spread of TDR in newly diagnosed European patients, using a pre-defined strategy that ensured representative sampling25. Epidemiological, clinical and virological data were collected prospectively from 2,800 patients between 2002 and 2005 and 1,630 patients between 2006 and 2007. Approximately half of the patients were MSM (48%), 35% were heterosexual and 8% were IVDUs. The majority (79%) were male. Fifty five percent originated in W Europe; 21% from Eastern Europe and Central Asia; 11% from Sub Saharan Africa; and 12% from other continents. The most important mutations that were detected included T215Y, M41L, K103N and L90M. The prevalence of drug resistance was significantly higher in MSM than other groups in relation to any ARV class, NRTIs and NNRTIs but not PIs. Trends towards an increase in resistance to any ARV class and to NRTIs over time were observed in MSM; this change was significant for NNRTIs (+4%, p=0.008). By contrast, there was a trend towards decreased (any ARV class and NRTIs) Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 15 Report or stabilised (PIs and NNRTIs) levels of resistance in heterosexuals. Prof. Boucher concluded that there are still single TAMs and revertants circulating in HIV positive Europeans, especially MSM. Resistance to NNRTIs has increased, mainly in MSM, while the transmission of mutations associated with resistance to PIs has decreased in recent years. TDR in newly diagnosed Japanese patients Viral samples from 408 newly diagnosed (2007-10) patients at Nagoya Medical Center, Japan, were tested for drug resistance using genotypic assays26. Approximately one third (31%) of the patients had seroconverted within the 155 days prior to testing while the remaining patients were classed as ‘long term’ seroconverters. Japanese MSM were diagnosed at a significantly earlier stage in their infection than non-Japanese patients (p<0.01) or those who had acquired HIV via risk factors other than MSM (p<0.05). TDR was observed in similar proportions in both recent and long term seroconverters: resistance to any ARV class was observed in 16% of samples. Resistance to PIs was observed in 7.1% of samples from recent seroconverters compared to 9.6% of long term seroconverters. The figures for NRTIs and NNRTIs, respectively, were 7.9% and 1.6% in the recent seroconverters and 5.3% and 2.1% in the long term seroconverters. There were no statistically significant differences between the two groups in terms of TDR. The mutations, T215X and M46I/L were detected in samples from both recent and long term seroconverters, suggesting that they have stabilised within the viral population and become circulating strains. TDR and clusters The early stage of HIV infection is a window of opportunity for prevention efforts since newly infected individuals are at high risk of transmitting HIV during this period (Figure 5)27. Lack of knowledge about their status means that they are less likely to take protective measures against transmitting HIV. Clusters of infections of both wild type and drug resistant virus have been identified within sexual networks. Over time, the size and number of clusters in a community tend to increase. Clustering can lead to the onward transmission of drug resistant sub-epidemics if these mutations do not reduce the replication capacity of the virus. In some cases, a compensatory mutation arises that reverses the fitness deficits imposed by a specific mutation e.g. a virus carrying E138K plus M184I/V has the same replication capacity as wild type virus. This may have clinical implications for patients who fail therapy with etravirine or other novel NNRTIs. It is essential that compensatory mutations, such as E138K, are monitored in future TDR studies. Mathematical modelling has been used to identify HIV variants that are most likely to generate large transmission clusters and to investigate the effect of more sensitive resistance detection assays on increasing observed persistence times28. The stochastic model of HIV transmission used 10 variants, each with a single point mutation that conferred resistance to NRTIs or NNRTIs. The results of the model suggest that there could be a Viral load & Infectivity Early stage infection: The window of opportunity for prevention 1/5 HIV+ Untested Ag + AbHAART 0 6 mo 2 yrs Acute/recent CLUSTERING >50% 5 yrs Chronic Infection 10-20% Figure 5. Time course of HIV infection Source: Dr. M. Wainberg, McGill University, Montreal Canada27. 16 Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 10 yrs > 20 yrs Chronic treated <<10% 6th Workshop on HIV Transmission hidden epidemic of HIV strains with intermediate to low fitness. The use of assays with increasing sensitivity could substantially increase the detection of intermediate fitness strains. Small differences in fitness result in significant differences in persistence times, cluster sizes and transmission chain lengths. Strains with high fitness costs (>6%), such as K65R, are rarely transmitted and are rarely detected in clusters. Transmissibility of drug resistant variants M184V is one of the most common mutations in patients who fail therapy; it is associated with resistance to NRTIs. A macaque model of infection has been used to determine the impact of M184V on viral transmissibility29. The M184V mutation was introduced into a R5 tropic SHIV genetic background and the transmissibility of the virus was evaluated using a repeat exposure rectal challenge model. The susceptibility of the variant to FTC was similar to that of HIV. At standard doses of virus, the variant was not very transmissible. When the dose was increased four-fold, the variant had similar transmission efficiency to that of wild type virus. The high fitness cost of the M184V mutation resulted in a reduced peak viraemia compared to wild type virus, even when higher doses of the mutated virus were used. Based on the results from this animal model, the presence of mutations that reduce viral fitness may reduce the risk of onward transmission of these mutated viruses. The M41L mutation is associated with resistance to NRTIs; it is a thymidine analogue mutation (TAM). The presence of M41L and at least two other TAMs is associated with resistance to tenofovir but not to FTC. Dr Pingen (University Medical Centre Utrecht, The Netherlands) and colleagues have used genetically engineered and patient-derived viruses to determine the impact of M41L on resistance to tenofovir and FTC30. All viruses that carried M41L as the only resistance-related mutation were fully susceptible to both NRTIs. In vitro selection experiments were carried out in which the viruses were exposed to increasing concentrations of tenofovir, FTC or both drugs. After 5-6 passages, the complete RT gene was sequenced. The results showed that the presence of M41L did not prevent the selection of K65R or select for additional TAMs. M41L did not have a major effect on the development of resistance to tenofovir and/or FTC, suggesting that therapy with these NRTIs could be initiated even in the presence of a M41L mutation. 7 19 - 20 JULY 2 012 DC, USA WASHINGTON MARK - THE - DATE 7th Workshop on HIV Transmission 19 - 20 July 2012, Washington DC, USA Abstracts & Presentations of the 6th Workshop on HIV Transmission are available online at www.virology-education.com Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 17 Report Reference 1. Hope, T. Cell Biology of HIV Sexual Transmission. in 6th International Workshop on HIV Transmission. 2011. Rome, Italy. 2. Keane, N., Limited acute CTL escape among three founder viruses in a case of acute heterosexual transmission of HIV-1 infection. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 3. 3. Dukhovlinova, E., HIV-1 viral populations in the female genital tract can be genetically distinct from virus in the blood. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 4. 4. Introini, A., HIV-1 and reactivated coinfecting Herpes viruses rearrange the cytokine network in semen of infected individuals. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 5. 5. Cenci, A., Characterization of variable regions of the Env protein of HIV-1 subtype C obtained from individuals at different disease stages in Sub-Saharan Africa. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 6-7. 6. Keele, B. Modeling HIV Transmission and Systemic Dissemination in Rhesus Macaques. in 6th International Workshop on HIV Transmission. 2011. Rome, Italy. 7. Saba, E., Productive HIV-1 infection of cervico-vaginal tissue ex vivo. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 9. 8. Bernard-Stoecklin, S., Characterization of semen leukocytes in Macaques infected by SIV. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 10. 9. Balandya, E., Semen protection of CD4+ T cells is less potent against R5 tropic HIV. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 11. 10. Neff, C., Pre-exposure prophylaxis by anti-retrovirals maraviroc and raltegravir protects against HIV-1 vaginal transmission in humanized mice. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 12. 11. Garcia-Lerma, G., Pharmacokinetic and pharmacodynamic profile of maraviroc in rhesus macaques after a single oral dose. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 13. 12. Heneine, W., Substantial changes in vaginal absorption of antiretroviral drugs from gels during the menstrual cycle in Macaques. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 24. 13. Abdool Karim, S. Tenofovir gel and HIV transmission: some insights from the CAPRISA 004 trial. in 6th International Workshop on HIV Transmission. 2011. Rome, Italy. 14. Blower, S., Preventing HIV and herpes infections: Predicting the impact of the CAPRISA tenofovir gel in South Africa. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 25. 15. Cohen, M. Update on HPTN 052. in 6th International Workshop on HIV Transmission. 2011. Rome, Italy. 16. Cohen, M., et al. (2011) Prevention of HIV-1 Infection with Early Antiretroviral Therapy. NEJM. Available from: . 17. Vasarhelyi, K., Evaluating the impact of “Treatment as Prevention” on reducing HIV transmission, using surveillance data. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 26. 18.Schols, D., Synergistic activity of griffithsin in combination with tenofovir, maraviroc and enfuvirtide against HIV-1 subtype C. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 8. 18 19. Coburn, B., Why doesn’t male circumcision work? Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 27. 20.Scarlatti, G. Mother-to-Child transmission of HIV1: Advances and controversies. in 6th International Workshop on HIV Transmission. 2011. Rome, Italy. 21. Lu, L., HIV incidence in women during the first postpartum year and implications for PMTCT programs û Francistown, Botswana, 2010. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 14. 22.Bruneau, J. Preventing HIV transmission among Injection Drug Users: a global perspective. in 6th International Workshop on HIV Transmission. 2011. Rome, Italy. 23. Khayatkhoei, M., Comparison of the prevalence of HIV infection among different types of drug abusers in Imamhossein Detoxification Center, Tehran, Iran. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 22. 24. Wensing, A. Transmission of drug resistant HIV; global trends and implications for first line therapy. in 6th International Workshop on HIV Transmission. 2011. Rome, Italy. 25. Boucher, C., Transmission of HIV resistant to nonnucleoside RT inhibitors is rising in Europe. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 15. 26.Hattori, J., Molecular epidemiology of transmitted drug-resistant HIV among newly diagnosed individuals in Japan. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 16. 27. Wainberg, M., Transmission within clusters of drugresistant variants of HIV-1. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 17. 28. Wagner, B. HIV drug resistance mutations: identifying variants most likely to generate large transmission clusters. in 6th International Workshop on HIV Transmission. 2011. Rome, Italy. 29. Garcia-Lerma, G. Reduced transmissibility of a drugresistant SHIV162P3 isolate containing the M184V mutation in macaques. in Reviews in Antiviral Therapy & Infectious Diseases. 2011. 19. 30. Pingen, M., The frequently transmitted M41L mutation in RT does not affect the in vitro selection of resistance against tenofovir and emtricitabine. Reviews in Antiviral Therapy & Infectious Diseases, 2011. 7: p. 20. Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement In today’s HIV landscape, we expect to keep them there, with durable suppression, well-established tolerability and convenient dosing.1–3 So they can concentrate on dealing with the challenges of life. Voor productinformatie zie elders in deze uitgave. NLRZ-K0003 08/11 687HQ11PM201(3) Verkorte productinformatie REYATAZ® Samenstelling: REYATAZ®, harde capsules, bevatten 150, 200 of 300 mg atazanavir per capsule. Indicaties: REYATAZ capsules, gelijktijdig toegediend met lage doseringen ritonavir, zijn bestemd voor de behandeling van hiv-1 geïnfecteerde volwassenen en pediatrische patiënten in de leeftijd van 6 jaar en ouder in combinatie met andere antiretrovirale middelen. Gebaseerd op de beschikbare virologische en klinische gegevens van volwassen patiënten, is er geen voordeel te verwachten bij volwassen patiënten met stammen die resistent zijn tegen meerdere proteaseremmers (≥ 4 PI mutaties). Er zijn zeer beperkte gegevens beschikbaar van kinderen in de leeftijd van 6 jaar tot jonger dan 18 jaar. De keuze voor REYATAZ bij voorbehandelde volwassen en pediatrische patiënten dient gebaseerd te zijn op individuele virale resistentie tests en de behandelingshistorie van de patiënt. Dosering: volwassen patiënten – De aanbevolen dosering van REYATAZ capsules voor volwassenen is 300 mg eenmaal daags samen met eenmaal daags 100 mg ritonavir en voedsel. pediatrische patienten vanaf 6 jaar – De dosering van REYATAZ capsules bij pediatrische patiënten is gebaseerd op lichaamsgewicht (15-<20 kg: 150 mg, 20-<40 kg: 200 mg, ≥40 kg: 300 mg; samen met 100 mg ritonavir en voedsel). De beschikbare gegevens ondersteunen niet het gebruik bij pediatrische patiënten die minder dan 15 kg wegen. pediatrische patiënten jonger dan 6 jaar: REYATAZ wordt niet aanbevolen. Contra-indicaties: Overgevoeligheid voor atazanavir of voor één van de hulpstoffen; leverinsufficiëntie; gelijktijdig gebruik met PDE-remmer sildenafil voor de behandeling van uitsluitend PAH, rifampicine, St. Jans kruid (Hypericum perforatum) en met substraten van de isovorm CYP3A4 van cytochroom P450 die een smalle therapeutisch breedte hebben (zoals astemizol, terfenadine, cisapride, pimozide, kinidine, bepridil en ergot alkaloïden; met name ergotamine, dihydroergotamine, ergonovine, methylergonovine). Bijzondere waarschuwingen: Patiënten met chronische hepatitis B of C die behandeld worden met een antiretrovirale combinatietherapie hebben een verhoogd risico op ernstige en potentieel fatale leverbijwerkingen. Extra voorzorgsmaatregelen kunnen nodig zijn bij gebruik van REYATAZ bij hemofilie patiënten en bij patiënten met onderliggende leverstoornissen. REYATAZ met ritonavir wordt niet aanbevolen bij patiënten die hemodialyse ondergaan. Speciale voorzichtigheid is nodig bij patiënten met bestaande cardiale geleidingsproblemen (tweedegraads of hoger atrioventriculair of complexe bundeltakblokkade) of risicofactoren (bradycardie, lang congenitaal QT, electrolyt verstoringen), of bij gebruik in combinatie met andere geneesmiddelen die mogelijk het PR- en/of QT-interval verlengen en cardiale controle bij kinderen wordt aanbevolen op geleide van de aanwezigheid van klinische bevindingen. Er zijn meldingen geweest van toegenomen bloeding bij patiënten met type A en B hemofilie. Evaluatie van de fysische kenmerken van de redistributie van vet moet onderdeel uitmaken van klinische beoordeling. Lipiden-stoornissen dienen klinisch passend te worden behandeld. Het ontstaan van diabetes mellitus, hyperglykemie en exacerbatie van bestaande diabetes mellitus zijn gemeld voor patiënten, die proteaseremmers kregen in sommige gevallen ook geassocieerd met keto-acidose. Reversibele verhogingen van indirect (niet-geconjugeerd) bilirubine gerelateerd aan remming van UDP-glucuronosyltransferase (UGT) werden gezien in patiënten die werden behandeld met REYATAZ. Hoewel men aanneemt dat bij de etiologie vele factoren een rol spelen zijn gevallen van osteonecrose vooral gemeld bij patiënten met voortgeschreden hiv-infectie en/of langdurige blootstelling aan CART. Immuunreactiveringssyndroom kan optreden. Alle symptomen van de ontstekingsreactie moeten worden beoordeeld en zo nodig worden behandeld. Nefrolithiasie is gemeld en diverse interacties met geneesmiddelen waaronder: statines, NNRTI’s, sterke CYP3A4 inductoren en/of remmers, PDE-5 remmers voor de behandeling van erectieledisfunctie, antimycotica, gluco- en/of corticosteroïden, salmeterol, maagzuurremmers. Bij gelijktijdig gebruik van een oraal anticonceptivum dient deze tenminste 30 ug ethinylestradiol te bevatten. REYATAZ capsules bevatten lactose. Bijwerkingen: volwassen patiënten – oedeem, palpitaties, hoofdpijn, perifere neuropathie, syncope, amnesie, duizeligheid, slaperigheid, dysgeusie, oculair icterus, dyspneu, braken, diarree, buikpijn, misselijkheid, dyspepsie, pancreatitis, gastritis, opgezette buik, orale aften, flatulentie, droge mond, nierstenen, hematurie, proteïnurie, pollakisurie, pijnlijke nier, huiduitslag, urticaria, alopecia, pruritis, vesiculobulleuze huiduitslag, eczeem, vasodilatatie, spieratrofie, arthralgie, myalgie, myopathie, gewichtsafname, gewichtstoename, anorexie, toegenomen eetlust, hypertensie, lipodystrofie syndroom, moeheid, pijn op de borst, malaise, koorts, asthenie, verstoorde manier van lopen, allergische reacties, geelzucht, hepatitis, hepatosplenomegalie, gynaecomastie, depressie, verwardheid, angst, slapeloosheid, slaapstoornissen, abnormale dromen; pediatrische patiënten – veiligheidsprofiel over het geheel genomen vergelijkbaar met dat gezien bij volwassenen, asymptomatische eerste en tweedegraads atrioventriculaire blokkade. Afleverstatus: UR. Vergoeding en prijzen: volledige vergoeding; voor prijzen zie Z-index. Zie de volledige Samenvatting van Productkenmerken voor aanvullende informatie. Bristol-Myers Squibb BV, Woerden, tel: 0348-574222. Datering: mei 2011 1. Daar ES et al. Ann Intern Med. 2011;154(7):445-456 2. Molina JM and the CASTLE Study Team. J Acquir Immnune Defic Syndr. 2010;53(3):323-332. 3. REYATAZ®/r, SmPC. Available at www.ema.europa.eu. Accessed May 2011. Date of preparation: August 2011 687HQ11PM201(3) NLRZ-A0018 NLRZ_A0018_VPI (210x148mm)_v1b.indd 1 / 200 mg / 245 mg VERKORTE PRODUCTINFORMATIE ATRIPLA 600 mg filmomhulde tabletten 8/30/11 4:39 Als de patiënten een ernstige uitslag ontwikkelen die gepaard gaat met blaren, desquamatie, mucosaletsels of PM koorts, moet de behandeling met Atripla worden stopgezet. Hulpstoffen: Dit geneesmiddel bevat 1 mmol (23,6 mg) natrium per dosis, waarmee rekening moet worden gehouden bij patiënten met een gecontroleerd natriumdieet. Zwangerschap: Atripla dient niet tijdens de zwangerschap te worden gebruikt, tenzij strikt noodzakelijk (d.w.z. er zijn geen andere passende behandelingsopties) Vruchtbare vrouwen: Bij vrouwen die Atripla ontvangen, moet zwangerschap worden voorkomen. Borstvoeding: Vanwege het gevaar van HIVoverdracht en de mogelijkheid van ernstige bijwerkingen bij baby’s die borstvoeding krijgen, dienen moeders te worden geïnstrueerd geen borstvoeding te geven als ze Atripla ontvangen. Bijwerkingen: De meest gerapporteerde, zeer vaak (≥ 1/10) bijwerkingen zijn: duizeligheid, misselijkheid, huiduitslag,abnormale dromen. Vaak (≥ 1/100, <1/10) werden de volgende bijwerkingen waargenomen: anorexie, nachtmerrie, depressie, depressieve stemming, angst, slapeloosheid, stemmingsverandering, slaapstoornis, slaperigheid, hoofdpijn, stupor, lethargie, aandachtsstoornis, vertigo, euforie, opvliegers, diarree, braken, abdominale pijn, flatulentie, opgezette buik, droge mond, nachtelijk zweten, jeuk, hyperpigmentatie van de huid, dermatitis, verhoogde creatininespiegel in het bloed, verminderde eetlust, vermoeidheid, toegenomen energie, koorts. Soms (> 1/1.000, <1/100): herverdeling van de vet, hypertriglyceridemie, gewichtsverlies, verwarring, desorientatie, verandering in de persoonlijkheid, stemmingswisselingen, verminderde libido, amnesie, ataxie, evenwichtsstoornis, onsamenhangende spraak, wazig zien, veranderde visuele diepte-perceptie, dygeusie, acute pancreatitis, orale paresthesie, orale hypo-esthesie, urticaria, droge huid, eczeem, acute hepatitis, myalgie, borstvergroting, zich abnormaal voelen, zich zenuwachtig voelen, rillingen, neutropenie, asthenie, zich dronken voelen, paranoia, psychomotrische agitatie,waanvoorstellingen, agressie, nervositeit. Verpakking: HDPE flacon met een kinderveilige sluiting, met 30 filmomhulde tabletten en een silicagel droogmiddel. Doos met 1 of 3 flacons. Afleverstatus: UR. Vergoeding: volledige vergoeding. Prijs: zie Z-index. Registratiehouder: Gilead Sciences International Limited, Cambridge CB21 6GT, Verenigd Koninkrijk. Datum: 05/2010. Bestudeer de samenvatting van productkenmerken alvorens Atripla voor te schrijven in het bijzonder vanwege dosering, bijwerkingen, waarschuwingen en voorzorgen bij gebruik, en interacties. Neem voor meer inlichtingen contact op met de lokale vertegenwoordiger: Gilead Sciences Netherlands B.V., WTC Tower D, Floor 7, Strawinskylaan 779, 1077 XX Amsterdam. efavirenz 600mg/emtricitabine 200mg/ tenofovir disoproxil (as fumarate) 245mg Tablets Samenstelling: Elke filmomhulde tablet bevat 600 mg efavirenz, 200 mg emtricitabine en 245 mg tenofovirdisoproxil (als fumaraat). Farmaceutische vorm: Filmomhulde tablet. Roze, capsulevormige, filmomhulde tablet, met aan de ene kant “123” gegraveerd en aan de andere kant niets. Farmacotherapeutische groep: Antivirale geneesmiddelen voor de behandeling van HIVinfecties, combinaties, ATCcode: J05AR06. Indicaties: Atripla is een vaste dosiscombinatie efavirenz, emtricitabine en tenofovirdisoproxilfumaraat. Het is geïndiceerd voor de behandeling van infectie met humaan immunodeficiëntievirus1 (HIV1) bij volwassenen met een virussuppressie tot HIV1RNA-concentratie < 50 kopieën/ml onder hun huidige antiretrovirale combinatietherapie gedurende meer dan drie maanden. Er mag bij patiënten geen virologisch falen zijn opgetreden bij eerdere antiretrovirale therapie en het moet bekend zijn dat patiënten voor het begin van hun eerste antiretrovirale behandeling niet geïnfecteerd waren door virusstammen met mutaties die een significante resistentie veroorzaken tegen een van de drie componenten van Atripla. Contra-indicaties: Overgevoeligheid voor de werkzame bestanddelen of voor één van de hulpstoffen. Atripla mag niet worden gebruikt bij patiënten met een ernstige leverfunctiestoornis (CPT graad C). Atripla mag niet gelijktijdig met terfenadine, astemizol, cisapride, midazolam, triazolam, pimozide, bepridil of ergotalkaloïden (bijvoorbeeld ergotamine, dihydro-ergotamine, ergonovine en methylergonovine) worden gebruikt, omdat competitie door efavirenz om cytochroom P450 (CYP) 3A4 kan resulteren in een inhibitie van het metabolisme en potentieel ernstige en/of levensbedreigende bijwerkingen kan uitlokken (bijvoorbeeld hartritmestoornis, langdurige sedatie of respiratoire depressie). Kruidengeneesmiddelen met St.Janskruid (Hypericum perforatum) mogen niet worden gebruikt als tegelijkertijd ook Atripla wordt gebruikt, vanwege het risico van een lagere plasmaconcentratie en een geringer klinisch effect van efavirenz. Efavirenz verlaagt de plasmaconcentratie van voriconazol sterk, terwijl voriconazol de plasmaconcentratie van efavirenz juist sterk verhoogt. Aangezien Atripla een combinatieproduct met vaste doses is, kan de dosering van efavirenz niet worden gewijzigd; daarom mogen voriconazol en Atripla niet gelijktijdig worden toegediend. Waarschuwing en voorzorgen: Als vaste combinatie dient Atripla niet gelijktijdig toegediend te worden met andere geneesmiddelen die een van dezelfde werkzame componenten bevatten: efavirenz, emtricitabine of tenofovirdisoproxilfumaraat. Vanwege overeenkomsten met emtricitabine dient Atripla niet gelijktijdig toegediend te worden met andere cytidineanalogen, zoals lamivudine. Atripla dient niet gelijktijdig toegediend te worden met adefovirdipivoxil. Lactaatacidose: Bij het gebruik van nucleoside-analogen is melding gemaakt van lactaatacidose, gewoonlijk samengaand met hepatische steatose. De behandeling met nucleoside-analogen moet gestopt worden bij het optreden van symptomatische hyperlactatemie en metabole acidose/lactaatacidose, progressieve hepatomegalie of snel stijgende aminotransferasespiegels. Om het risico van lactaatacidosise te minimaliseren bij toediening van nucleoside-analogen moeten de patiënten nauwgezet worden gevolgd. Leverziekte: Atripla is gecontra-indiceerd bij patiënten met een ernstige leverfunctiestoornis. Omdat efavirenz hoofdzakelijk wordt gemetaboliseerd door het cytochroom-P450-systeem (CYP450-systeem), is bij toediening van Atripla aan patiënten met een lichte tot matig-ernstige leverziekte voorzichtigheid geboden. De leverziekte moet periodiek door middel van laboratoriumonderzoek worden gecontroleerd. Patiënten met HIV en gelijktijdige infectie met hepatitisB- of Cvirus (HBV of HCV): Patiënten met chronische hepatitis B of C die een antiretrovirale combinatietherapie ondergaan, lopen een verhoogd risico op ernstige en potentieel fatale leverbijwerkingenStoppen van de behandeling met Atripla bij patiënten met gelijktijdige infectie met HIV en HBV kan gepaard gaan met ernstige acute exacerbaties van hepatitis. Psychische symptomen: Bij patiënten die met efavirenz zijn behandeld, zijn psychische bijwerkingen gemeld. Patiënten met een voorgeschiedenis van psychische stoornissen lijken een groter risico te hebben op deze ernstige psychische bijwerkingen. Convulsies: Bij patiënten die efavirenz gebruikten, zijn convulsies waargenomen, meestal bij een bekende voorgeschiedenis van toevallen. Bij alle patiënten met toevallen in de voorgeschiedenis moet voorzichtigheid worden betracht. Nierfunctiestoornis: Het gebruik van Atripla wordt niet aanbevolen bij patiënten met een matig-ernstige of ernstige nierfunctiestoornis.Het gebruik van Atripla dient te worden vermeden bij gelijktijdig of recent gebruik van een nefrotoxisch geneesmiddel. Indien gelijktijdig gebruik van Atripla en nefrotoxische middelen (bv. aminoglycosiden, amfotericine B, foscarnet, ganciclovir, pentamidine, vancomycine, cidofovir, interleukine2) onvermijdelijk is, moet de nierfunctie wekelijks worden gecontroleerd. Het wordt aangeraden om bij alle patiënten de creatinineklaring te berekenen voordat wordt begonnen met de behandeling met Atripla. De nierfunctie (creatinineklaring en serumfosfaat) wordt gedurende het eerste jaar ook elke vier weken gecontroleerd en daarna elke drie maanden. Aangezien Atripla een combinatieproduct is en het doseringsinterval van de individuele componenten niet kan worden gewijzigd, moet de behandeling met Atripla worden onderbroken bij patiënten bij wie een creatinineklaring van < 50 ml/min of een afname van het serumfosfaatgehalte naar < 1,0 mg/dl (0,32 mmol/l) is bevestigd. Huidreacties: Lichte tot matig-ernstige huiduitslag is gemeld bij gebruik van de individuele componenten van Atripla. HAART in 1 Referenties: 1. Staszewski S, et al. Efavirenz plus zidovudine and lamivudine, efavirenz plus indinavir, and indinavir plus zidovudine and lamivudine in the treatment of HIV-1 infection in adults. N Engl J Med 1999;341:1865–1873. 2. Robbins GK, et al. Comparison of sequential three-drug regimens as initial therapy for HIV-1 infection. N Engl J Med 2003;349:2293–2303. 3. Van Leth F, et al. Comparison of first-line antiretroviral therapy with regimens including nevirapine, efavirenz, or both drugs, plus stavudine and lamivudine: a randomised open-label trial, the 2NN study. Lancet 2004;363:1253–1263. 4. Riddler SA, et al. Class-sparing regimens for initial treatment of HIV-1 infection. N Engl J Med 2008;358:2095–2106. 5. Lennox J, et al. Raltegravir versus efavirenz regimens in treatment-naive HIV-1–infected patients: 96-week efficacy, durability, subgroup, safety, and metabolic analyses. J Acquir Immune Defic Syndr 2010. [Epub ahead of print]. 6. Gotuzzo E, et al. 17th Conference on Retroviruses and Opportunistic Infections. 16–19 February 2010. San Francisco, CA. Abstract K-127. Poster 514. 7. Gallant JE, et al. Tenofovir DF, emtricitabine, and efavirenz vs. zidovudine, lamivudine, and efavirenz for HIV. N Engl J Med 2006;354:251–260. 8. Martinez E, et al. J Acquir Immune Defic Syndr. 2009 Jul 1;51(3):290-7. 9. Sax P, et al. Abacavir–lamivudine versus tenofovir–emtricitabine for initial HIV-1 therapy. N Engl J Med 2009;361:2230–2240. 10. Smith KY, et al. Randomized, double-blind, placebo-matched, multicenter trial of abacavir/lamivudine or tenofovir/ emtricitabine with lopinavir/ritonavir for initial HIV treatment. AIDS 2009;23:1547–1556. 11. Martin A, et al. Simplification of antiretroviral therapy with tenofovir-emtricitabine or abacavir-lamivudine: a randomized, 96-week trial. Clin Infect Dis 2009;49:1591–1601. 12. Post FA, et al. Randomized comparison of renal effects, efficacy, and safety with once-daily abacavir/lamivudine versus tenofovir/emtricitabine, administered with efavirenz, in antiretroviralnaive, HIV-1–infected adults: 48-week results from the ASSERT study. J Acquir Immune Defic Syndr 2010 sept;55(1):49-57. 13. Arribas JR, et al. Tenofovir disoproxil fumarate, emtriciabine, and efavirenz compared with zidovudine/lamivudine and efavirenz in treatment-naive patients. J Acquir Immune Defic Syndr 2008;47:74–78. 14. DeJesus E, et al. Simplification of antiretroviral therapy to a singletablet regimen consisting of efavirenz, emtricitabine, and tenofovir disoproxil fumarate versus unmodified antiretroviral therapy in virologically suppressed HIV-1-infected patients. J Acquir Immune Defic Syndr 2009;51(2):163–174. 15. Hodder SL, et al. Patient-reported outcomes in virologically suppressed, HIV-1-infected subjects after switching to a simpliofied, single-tablet regimen of efavirenz, emtricitabine, and tenofovir DF. AIDS Patient Care STDs 2010;24(2);87–96. 16. Airoldi M, et al. One-pill once-a-day HAART: a simplification strategy that improves adherence and quality of life of HIV-infected subjects. Patient Prefer Adherence 2010;4:1–11. Report 3rd International Workshop on HIV Pediatrics 15 - 16 July 2011 in Rome, Italy Report Progress and Controversies in Pediatric HIV Care Report on the 3rd International Workshop on HIV Pediatrics J. Ananworanich1, C. Boucher2, D. Burger3, E. Capparelli4, M. Cotton5, C. Giaquinto6, D. Mbori-Ngacha7, L. Mofenson8 The Thai Red Cross AIDS Research Center, Thailand, 2Erasmus Medical Center Rotterdam, The Netherlands, University of Nijmegen, The Netherlands, 4University of California San Diego, California, USA, 5Stellenbosch University J8/Tygerberg Children’s Hospital,South Africa, 6University of Padova, Italy, 7UNICEF East and Southern Africa Region, South Africa, 8National Institutes of Health, Bethesda, USA 1 3 Written by Mark Mascolini on behalf of the Organizing Committee Growing up with HIV poses different challenges in resource-rich and poor countries, attendees learned in the opening session of 3rd International Workshop on HIV Pediatrics. But similar challenges in both regions are not hard to find, according to keynote speakers Rohan Hazra1 (Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland) and Philippa Musoke2 (Makerere University, Kampala). Two prospective pediatric cohorts in the United States recorded dramatic drops in mortality from the days before combination antiretroviral therapy (cART) to more recent years,3,4 Hazra noted. The 3553-child PACTG 219/219C cohort recorded a mortality plummet from 7.2 per 100 person-years in 1994 to 0.6 per 100 person-years in 2006.3 In the 364-child PACTS/PACTS-HOPE birth cohort, mortality waned from 18 per 100 person-years in 1986 to 0.8 per 100 person-years in 2004.4 But for youngsters with HIV, decades-long survival with HIV means decades of coping with potential central nervous system disease, metabolic and insulin dysregulation, and bone, liver, renal, and cardiovascular disease. Atherosclerosis does begin in childhood, Hazra stressed. “The use of [cART] has transformed HIV infection in children from a fatal illness to a chronic health condition,” Hazra observed, and, “as with other chronic conditions, long-term treatment, monitoring, and management will be critical, and many challenges will be faced as these children age into adulthood.” At the same time, he argued, survival of perinatally infected children into adulthood in rich countries should encourage “expansion of pediatric treatment programs in low-resource countries, where most HIV-infected children live.” Philippa Musoke spoke for that majority in resource- Disclosure: Mark Mascolini has disclosed that he has not received grants or (research) support from any commercial entity or served as consultant for such a company. 22 limited settings, noting that 400,000 infants become infected with HIV every year, 90% of them in subSaharan Africa.2 While mortality was falling steeply in HIV-positive US children by 2004,4 an African metaanalysis of perinatal prevention trials in breastfeeding infants reported from 1999 through 2002 found that 52.5% of children who became infected had died at 2 years of age and 60% by 2.5 years.5 Delayed diagnosis and treatment of HIV in resourcepoor countries explains much of the high mortality, Musoke said. When women with HIV are not diagnosed during pregnancy, their HIV-positive infants also go undiagnosed. Access to HIV DNA testing of dried blood spots is limited in many lowincome countries, so undiagnosed HIV often persists in newborns. When children are diagnosed, lack of appropriate pediatric formulations—and sometimes perinatal exposure to nevirapine—complicate their treatment. Malnutrition, tuberculosis, and other afflictions complicate the course of children who are diagnosed and begin treatment. Around the world, UNAIDS estimates that 38% of children eligible for cART begin treatment, compared with 43% of adults.6 Across sub-Saharan Africa, only 35% of cART-eligible children get treated, but rates are higher in South Africa (65%) and Botswana (80%). As in high-income countries, children typically respond well to cART in low- and middle-income countries. Musoke cited a 3936-child study of children starting cART before the age of 5 years, 90% of them in Africa and the rest in Asia.7 After a median cART duration of 10.5 months (interquartile range [IQR] 3.7 to 20.6), 6.3% of children in this cohort had died and 10.3% were lost to follow-up. Most deaths, 55%, occurred in the first 6 months of Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 3rd International Workshop on HIV Pediatrics treatment, and only 3.8% of children experienced severe antiretroviral toxicity. rather than waiting until a drop to 250 cells/mm3 lowered the risk of a clinical event 40% (P = 0.01).11 Musoke listed a set of challenges in adolescent HIV care in resource-poor settings, all of which could be cited as challenges in high-income countries: • Knowledge of HIV infection • Linking to (and retaining in) health care • Accepting (and adhering to) therapy • Mental health issues • Complexities of transition to adult care • High risk of HIV transmission to sex partners PREDICT is an open-label trial in Thailand and Cambodia that randomized antiretroviral-naive children from 1 to 12 years old to start cART immediately or to close monitoring every 3 months and to begin cART when the CD4 percent fell below 15%.8 All children were naive to antiretrovirals, had a CD4 percent between 15% and 24% upon enrollment, and had CDC stage A or B disease. Primary endpoints were AIDS-free survival and neurodevelopment as measured by the Berry visualmotor integration score at week 144. PREDICT investigators recruited 300 children (180 Thais and 120 Cambodians) at seven clinical research sites in Thailand and two in Cambodia. Study retention was 97%. Health workers measured CD4 percent every 3 months. All children started the same regimen— nevirapine, zidovudine, and lamivudine Hazra interleaved his data slides with quotes from US children asked for their thoughts about growing up with HIV. “At first, I didn’t think that I was going to grow up with HIV,” one youngster remarked. “I thought I was going to die from HIV.” Now that HIV-positive children around the world are learning that death is not a short-term inevitability, HIV pediatricians everywhere are learning that cross-cultural exchanges like those possible at the International HIV Pediatrics Workshop can afford the deepest insights into a virus that does not pause at Passport Control. Pediatric cART: Controversies and Insights Immediate cART does not prolong survival in 1- to 12-year-olds Contrary to findings in infants under 1 year old and in adults, a 144-week randomized trial in Thailand and Cambodia found that immediate cART for 1- to 12-year-olds with moderately advanced HIV infection did not prolong survival, delay progression to AIDS, or improve neurodevelopment.8 Children in the immediate treatment group did grow faster than those in the delayed treatment group, and they had lower rates of thrombocytopenia and herpes zoster. The findings run counter to results of the South African CHER trial, which found that immediate cART for children under 1 decreased mortality by 76% and HIV progression by 75%.9 As a result of that trial, the World Health Organization (WHO) recommends immediate cART for children up to 2 years old regardless of CD4 count, CD4 percent, or WHO clinical stage.10 For 2- to 5-year-olds, WHO recommends cART for those with a CD4 count at or below 750 cells/mm3 or a CD4 percent at or below 25%, whichever is lower, regardless of WHO clinical stage. For children 5 or older, WHO advises starting cART for all with a CD4 count below 350 cells/mm3, regardless of WHO clinical stage. In adults, the international HPTN 052 trial found that starting cART at a CD4 count between 350 and 550 cells/mm3 Median age was 6.4 years in both treatment arms. Forty-six children (15%) were 1 to 3 years old, 67 (22%) were 4 to 5, and the rest were older than 5 years of age. In the immediate and deferred arms, 52% and 64% were girls, median CD4 percent stood at 19% and 20%, and median viral load measured 4.9 and 4.7 log10 copies/mL. Median weight-for-age Z score and height-for-age Z score were similar in the two groups (-1.3 in both arms for weight and -1.6 to -1.7 for height). After 144 weeks of follow-up, AIDS-free survival was 97.9% in the immediate-cART group and 98.7% in the deferred group, a nonsignificant difference (P = 0.49). Incidence of CDC class C events was 7.6 per 1000 person-years in the immediate group and 4.9 per 1000 person-years in the deferred group (3 events versus 2). The Berry visual-motor integration test score did not differ significantly between the immediate and deferred arms (84.7 and 86.8, P = 0.50) at 144 weeks. The PREDICT investigators called the Berry score a crude measurement of neurodevelopment, and they are further analyzing cognitive function and fine motor test results. Incidence of thrombocytopenia (abnormally low platelets) was lower in the immediate arm (2.5 versus 24.4 per 1000 person-years, hazard ratio [HR] 9.7, P = 0.03), as was incidence of herpes zoster (7.5 versus 31.8 per 1000 person-years, HR 4.2, P = 0.03). All 10 cases of thrombocytopenia in the deferred arm occurred before cART began, and 9 of 13 cases of herpes zoster occurred before cART began. Weightfor-age Z score improved more in the immediate arm (0.20 versus 0.08, P = 0.074), as did height-for-age Z score (0.23 versus 0.00, P = 0.003). Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 23 Report 40 %CD4 after start ART 30 Immediate 20 C D 4% Deferred 10 P=0.74 0 12 24 36 48 60 72 84 96 108 120 132 144 Weeks after commencing ART IMMEDIATE 149 148 147 147 145 143 144 142 142 142 142 142 142 DEFERRED 68 67 60 57 54 51 46 38 27 19 14 8 6 Figure 1. Gains in CD4 percent did not differ between the immediate-cART arm and the delayed arm after treatment began in the randomized PREDICT trial, which found no mortality difference between the immediate and deferred arms after 144 weeks. Source: Thanyawee Puthanakit, HIV-NAT, Bangkok.8 Gains in CD4 percent did not differ significantly between the two study arms after cART began (P = 0.74) (Figure 1). And proportion of children reaching a viral load below 50 copies/mL after treatment began did not differ between the immediate and deferred arms. lower in Thailand and Cambodia than in some other developing countries. Finally, the results should be considered with special caution in 1- to 3-year-old children, who represented only 15% of the study population. Because PREDICT found that HIV-positive children who survive beyond infancy with moderate immune suppression had slow disease progression, the investigators suggested clinicians have the option to monitor such children closely and to start cART when the CD4 percent drops below 15% (although that approach would fall outside WHO guidelines). The PREDICT team proposed that this approach may be useful when children and families are not ready to embark on lifelong therapy and when cART programs are scaling up to cover all children in need. NNRTI or PI first? Clues from four big studies But the investigators cautioned that clinical application of study results should be considered with the following caveats: First, entry criteria stipulated that all participants had survived at least the first year of life without antiretroviral therapy, without an AIDS disease, and without a CD4 percent decline below 15%; and two thirds of study participants had survived the first 5 years of life without AIDS or cART. Thus by definition these children had slowly progressive disease. Second, study participants had their CD4 percent measured every 3 months, an interval that is more frequent than the usual 6 to 12 months in clinical practice in developing countries. Third, morbidity and mortality background rates (for example, form diarrhea and pneumonia) are probably 24 Whether to start cART in infants and children with a regimen based on a nonnucleoside reverse transcriptase inhibitor (NNRTI) or a protease inhibitor (PI) is a pressing question whose answer depends on an array of variables including exposure to singledose nevirapine (sdNVP) or other antiretrovirals for prevention of mother-to-child transmission (PMTCT) and the availability of appropriate, convenient formulations, which typically means generic products in low- and middle-income countries. For children under 3 years old, US Department of Health and Human Services (DHHS) guidelines recommend two nucleos(t)ide reverse transcriptase inhibitors (NRTIs) plus lopinavir/ritonavir, with nevirapine now listed as an alternative (in children not exposed to nevirapine perinatally).12 Because children in both the nevirapine-exposed and unexposed cohorts of IMPAACT P1060 (summarized below) had a better CD4 response and better growth than children in the lopinavir/ritonavir arm, this panel decided that “liquid nevirapine remains an acceptable alternative for infants not exposed to single-dose nevirapine for PMTCT who cannot tolerate lopinavir/ritonavir.” For children 3 to 6 years old, DHHS recommends Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 3rd International Workshop on HIV Pediatrics Time to off study treatment or virologic failure Prop. on TRT with no VF 1.0 0.8 0.6 LPV/r 0.4 NVP 0.2 0.0 Weeks 0 N at risk: NVP 147 LPV/r 140 Week 24 failure rate Week 24: NVP - LPV/r: 21.5% (p<0.001) 24 48 72 96 120 144 168 109 125 68 93 47 54 25 33 20 26 12 17 7 9 Figure 2. Among children not perinatally exposed to nevirapine in IMPAACT P1060 cohort 2, those randomized to treatment with a nevirapine regimen had a 21.5% higher failure/discontinuation rate at 24 weeks than did children randomized to lopinavir/ritonavir. Source: Paul Palumbo, Dartmouth-Hitchcock Medical Center.13 two NRTIs plus efavirenz or lopinavir/ritonavir. And for children 6 years old or older, the panel prefers two NRTIs plus atazanavir/ritonavir, lopinavir/ ritonavir, or efavirenz. WHO guidelines divide first-line treatment choices by age (under 12 months, 12 to 24 months, 24 months to 3 years, and 3 or older) and according to perinatal exposure to nevirapine or other antiretrovirals, recommending regimens based on lopinavir/ritonavir, nevirapine, or efavirenz.10 Study participants were 6 months to 3 years old and eligible for cART by WHO criteria.10 The primary endpoint in both cohorts was (1) virologic failure by week 24, or (2) permanent discontinuation of nevirapine or lopinavir/ritonavir by week 24, or (3) death. The investigators defined virologic failure as a confirmed viral load decline less than 1 log10 copies/ mL (10-fold) between weeks 12 and 24 or a viral load above 400 copies/mL at week 24. The 3rd International Workshop on HIV Pediatrics offered insight into this question in reviews of three randomized trials (IMPAACT P1060, PENPACT 1, and NEVEREST 2) and one large cohort study (EPPICC). Presenting IMPAACT P1060 data, Paul Palumbo (Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire) rated nevirapine a very good pediatric antiretroviral because of its potency, userfriendly formulations, good tolerability, temperature tolerance, and cost-effectiveness.13 A “parade of reports” documenting slowly fading resistance to nevirapine after perinatal exposure began in 2000, Palumbo noted. As these four studies show, the choice between nevirapine (or efavirenz) and a ritonavir-boosted PI is not clear-cut. • IMPAACT P1060. Children in cohort 1 (the nevirapine-exposed group) had (1) an HIV diagnosis by 60 days of age, or (2) strict formula feeding, or (3) an AIDS-defining event by 60 days of age. These children came from four study sites in South Africa and one each in India, Malawi, Tanzania, Uganda, Zambia, and Zimbabwe. Almost all children, 95%, were infected with HIV-1 subtype C, and 20% breastfed.13,14 Cohort 1 included 82 children taking nevirapine and 82 taking lopinavir; both groups had a median age of 0.7 year and a median viral load above 750,000 copies/mL at study entry. Median baseline CD4 percents were 18.9% in the nevirapine group and 19.7% in the lopinavir group. More children taking nevirapine than lopinavir (62% versus 50%) had at least WHO stage 3 disease. Median follow-up stood at 48 weeks (range 0 to 125 weeks). IMPAACT P1060 focused on two cohorts, one exposed to sdNVP and one not exposed.13 The investigators randomized children to nevirapine or lopinavir/ritonavir, both with zidovudine/lamivudine. Among nevirapine-exposed children under 12 months old, virologic failure or discontinuation rates at 24 weeks were 45.3% in the nevirapine group Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 25 Report and 23.3% in the lopinavir groups. Virologic failure/ discontinuation rates at 24 weeks in children 12 months or older were 28.9% in the nevirapine group and 17.5% in the lopinavir group. Among all children without pretreatment nevirapine resistance on a standard assay, 24-week failure rates were 35.8% among those taking nevirapine and 20.3% among those taking lopinavir (P = 0.06). Among the 18 children with a pretreatment nevirapine resistance mutation (Y181C in 15 and K103N in 3), failure rates were 83.3% with nevirapine and 18.2% with lopinavir, a highly significant difference (P = 0.001). The 288 children in cohort 2 (not exposed to nevirapine) came from four sites in South Africa and one each in India, Malawi, Tanzania, Uganda, Zambia, and Zimbabwe. These children had a median age of 1.7 years, and 73% were older than 12 months. Median pretreatment viral load stood at 535,000 copies/mL and median CD4 percent at 15%. Most of the children, 81%, were breastfeeding. Median follow-up was 60 weeks. As in cohort 1, children were randomized to lopinavir/ritonavir or nevirapine plus zidovudine/lamivudine. In cohort 2, virologic failure or off-study rates at week 24 were 40.8% with nevirapine versus 19.3% with lopinavir (P < 0.001) (Figure 2). Failure or off-study rates were significantly higher with nevirapine among children under 12 months old (41.5% versus 19.4%, P = 0.030) and children 12 months old or older (40.6% versus 19.2%, P = 0.001). Rates of discontinuation because of toxicity were higher with nevirapine than with lopinavir for children under 12 months old (7/41 or 17% versus 1/36 or 3%) and for older children (22/106 or 21% versus 5/104 or 5%). Palumbo showed modeling data suggesting that the once-daily lead-in dose of nevirapine may be too low given the high starting viral loads in these children and possible adherence difficulties. Pharmacologist David Burger (University Medical Center, Nijmegen, Netherlands) suggested that the ultimate twice-daily nevirapine dose may also be too low in this population and deserves further evaluation. Palumbo added that some children in cohort 2 had nevirapine resistance mutations at baseline, and IMPAACT investigators are assessing pretreatment samples with more sensitive assays. Standard genotyping detected Y181C in 4 children and K103N in 1 before treatment in cohort 2. • PENPACT 1. Also designated PENTA 9 and PACTG 390, PENPACT 1 was an open-label trial that randomized children to begin antiretroviral therapy with a PI or an NNRTI combination and to switch after virologic failure at a 26 higher versus a lower viral load (above 1000 versus above 30,000 copies/mL).15,16 Of the 263 analyzed study participants, half were from Europe, 27% from the United States, 20% from Brazil or Argentina, and 3% from the Bahamas or Puerto Rico. There were 218 children (83%) in follow-up at the end of the trial in August 2009, and median follow-up stood at 5 years. Study participants had a median age of 6.5 years (range 1 month to 17.8 years), 53% were boys, and 79% were vertically infected. Half were black, one quarter white, and one quarter Hispanic or another race or ethnicity. Median pretreatment CD4 percent stood at 17% (IQR 10% to 25%) and median viral load at 5.1 log10 copies/mL (about 126,000 copies/ mL). Only 15% of children received antiretrovirals for PMTCT, and 4% had one or more major resistance mutations when PENPACT 1 began. At the end of the trial, 70% of children remained on their initial regimen, and the proportion switching antiretroviral classes did not differ between study arms (P = 0.64). Only 7% of children switched to a third-line regimen by the end of follow-up, and proportions did not differ between randomized arms. Among children who changed regimens, the switch point was significantly lower in those randomized to switch at 1000 copies/mL than among those randomized to switch at 30,000 copies/mL (6720 versus 35,712 copies/mL, P < 0.01). Estimated time until 10% of children in the 1000-copy-switch group changed regimens was 54 weeks, compared with 95 weeks until 10% of children in the 30,000-copyswitch arm changed. Change in viral load from baseline to year 4, the primary endpoint, did not differ significantly between the PI group (-3.16 log) and the NNRTI group (-3.31 log) (difference -0.15 log, P = 0.26). Proportions with a viral load below 400 copies/mL at week 204 did not differ between the PI group and the NNRTI group (82% in both) or between the 1000-switch group and 30,000-switch group (83% versus 80%, P = 0.43). Suppression rates with a 50-copy assay were similar in the two randomized comparisons. CD4 percent changes also proved similar in these two comparisons. Adverse event rates did not differ by trial assignment. Among children in whom treatment failed, mutations conferring resistance to PIs were uncommon, and low-level resistance to lopinavir/ritonavir emerged in only 1 child. Among children whose NNRTI regimen failed, NNRTI mutation rates did not differ by viral load switch point, because NNRTI-resistant HIV is selected very early during virologic failure. In the NNRTI arm, Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 3rd International Workshop on HIV Pediatrics NRTI mutations accumulated in approximately 10% more children in the 30,000-switch group than in the 1000-switch group. This accumulation rate was lower than predicted, and NRTI mutations were even less likely to develop in PI recipients. Gareth Tudor-Williams (Imperial College, London) noted that PENPACT 1 is the first randomized trial in the cART era with follow-up long enough to address the consequences of early versus later switching after virologic failure. He suggested the trial raises the question whether nevirapine-exposed infants starting lopinavir/ritonavir should delay switching at virologic failure, because PI resistance rates were low in children in whom a PI failed and—after emergence of M184V— further NRTI mutations were slow to evolve. Tudor-Williams also cautioned that PENPACT 1 results cannot be compared with those of IMPAACT P106013 because of the different study populations and follow-up times. • NEVEREST 2. How to apply results of NEVEREST 2 has perplexed clinicians since these findings were presented at the 1st HIV Pediatrics Workshop and published soon thereafter.17 The study involved nevirapine-exposed South African children who began cART with a PIbased regimen (lopinavir/ritonavir or ritonavir). The 195 children who reached and maintained a viral load below 400 copies/mL for more than 3 months were randomized to keep taking lopinavir/ritonavir or to switch to nevirapine. A significantly higher proportion in the switch arm maintained a viral load below 50 copies/mL to week 52 (56.2% versus 42.4%, P = 0.01). But a significantly higher proportion in the lopinavir/ritonavir maintenance group kept their viral load below 1000 copies/mL (98% versus 80%, P = 0.001). Ashraf Coovadia (Rahima Moosa Mother and Child Hospital, Johannesburg), first author of the published report, updated results through 48 months at the 3rd HIV Pediatrics Workshop.18 Probability of ever reaching a viral load above 50 copies/mL remained lower in the nevirapine switch group at 24 months (51% versus 68%), 36 months (60% versus 69%%), and 48 months (63% versus 75%). The proportion of children with a confirmed viral load above 1000 copies/mL remained stable at 24% in the nevirapineswitch arm through 48 months, while the proportion with a confirmed 1000-copy rebound in the lopinavir/ ritonavir group rose from 5% at 12 months to 11% at 48 months. In the nevirapine-switch group, 59% of all confirmed rebounds above 1000 copies/mL occurred by month 6 and the rest occurred by month 12 (Figure 3). In the lopinavir-maintenance group, only 10% of confirmed rebounds above 1000 copies/mL occurred by month Percent of All “Failures” (confirmed >1000cp/mL) occurring by different time points post randomization 100% 75% 50% 41% By 48m By 12m 50% 40% 59% By 6m 25% 0% 10% LPV/r Stay Half of all failures detected after 12 mos of ART NVP Switch All failures detected within 12 mos of starting ART Figure 3. Among nevirapine-exposed South African children who started a lopinavir/ritonavir regimen, reached and maintained a viral load below 400 copies/mL for more than 3 months, and were randomized to maintain lopinavir/ritonavir or switch to nevirapine in NEVEREST 2, all virologic failures in the nevirapine group occurred within 12 months of the switch, while half of failures in the lopinavir/ritonavir group occurred between months 12 and 48. Source: Ashraf Coovadia, Rahima Moosa Mother and Child Hospital, Johannesburg, South Africa.18 Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 27 Report 6, another 40% occurred by month 12, and the remaining 50% occurred by month 48. Among children with a confirmed viral load above 1000 copies/mL after randomization, the failure rate did not differ between the nevirapine and lopinavir arms in children with no detectable pretreatment mutations. But among those with detectable pretreatment NNRTI mutations, the failure rate was significantly higher in the nevirapine-switch group (above 50%) than in the lopinavir-maintenance group (below 15%) (P = 0.02). Median CD4 percent at 24 weeks was higher in the nevirapine group than the lopinavir group (33.2% versus 30.0%, P < 0.0001), but it is unclear whether this difference is clinically meaningful. CD4 percent declined by at least 10% in significantly more children staying with lopinavir than switching to nevirapine (16.3% versus 3.2%, P = 0.004). Weight for age fell by more than 1 Z score in significantly more children maintaining lopinavir (13.1% versus 4.2% in the nevirapine-switch group, P = 0.03). Grade 2 alanine aminotransferase (ALT) elevations were significantly more frequent in the nevirapine group than the lopinavir group (11.5% versus 2%, P = 0.05), and there was a trend toward a higher proportion of grade 3/4 ALT elevations in the nevirapine arm (5.2% versus 4%, P = 0.08). NEVEREST 2 investigators concluded that switching to an NNRTI regimen after perinatal NNRTI exposure and virologic suppression with lopinavir/ritonavir “can be accomplished safely if adequate virological monitoring is in place.” They observed that failure after a switch to nevirapine occurs within 1 year of the switch, and that viral load monitoring can detect these failures and signal the need to return to lopinavir/ritonavir. The researchers believe more study is needed to discern the clinical significance of low-level viremia and the worse CD4 response in the lopinavir-maintenance group. • EPPICC. Four in 5 European infants starting their first cART regimen after 1996 were still taking their firstline regimen 2 years later, and two thirds had not switched or interrupted antiretrovirals 5 years later, according to results of a 9-cohort analysis.19 By several measures, the best first-line regimen was one NNRTI plus three NRTIs. WHO recommends immediate cART for children up to 2 years old,10 but data on response to early cART are sparse and, as discussed above, IMPAACT 106013 and PENPACT 115 reached different conclusions on the optimal first-line regimen (in highly different 28 study populations). To assess first-line responses in infants, and to determine antiretroviral switch rates and switch predictors, European researchers pooled data from nine cohorts in which children younger than 12 months old started cART between 1996 and 2008.19 European Pregnancy and Paediatric HIV Cohort Collaboration (EPPICC) investigators defined virologic response as a viral load below 400 copies/ mL, CD4 response as absolute change in CD4 Z score from baseline, and switch to second-line therapy as simultaneously changing either (1) three or more drugs for any reason or (2) two drugs because of treatment failure. For purposes of this analysis, treatment interruption meant stopping all antiretrovirals for 14 or more days. The analysis involved 437 infants, 39% in the United Kingdom and Ireland, 23% in Italy, 19% in France, and the rest in other European countries. Median age when cART started was 3.6 months (IQR 2.1 to 5.8), and 31% of infants had an AIDS diagnosis before treatment began. Mothers of 34% of these infants had taken antiretrovirals during pregnancy, and 28% of infants received neonatal antiretroviral prophylaxis. One third of infants were breastfed. Most regimens included either a PI or an NNRTI: • Unboosted PI plus two NRTIs: 38% • Ritonavir-boosted PI plus two NRTIs: 15% • NNRTI plus two NRTIs: 24% • NNRTI plus three NRTIs: 14% • PI plus NNRTI plus NRTI or three NRTIs: 8% The virologic suppression rate rose from 53% in 1996-1999 to 57% in 2000-2003 and to 77% in 20042008. Martina Penazzato (University of Padova, Italy, and CTU-MRC, London), who presented results for EPPICC, suggested that improving virologic response rates reflect better regimen efficacy, more adequate dosing, and “personalized HIV management leading to better caregiver adherence.” Every 10-fold higher pre-cART viral load lowered chances of virologic suppression at 12 months by 33% (adjusted odds ratio [AOR] 0.67, 95% CI 0.50 to 0.89, P = 0.006). Compared with starting an NNRTI plus two NRTIs, starting an NNRTI plus three NRTIs tripled chances of suppression at 12 months (AOR 3.00, 95% CI 1.24 to 7.23, P < 0.001). The study found no evidence that other first-line regimens improved or worsened chances of suppression compared with an NNRTI plus two NRTIs. Among 203 infants with pretreatment and 12-month CD4 counts available, median CD4 change was 520 cells/mm3 (IQR 271 to 1340), median CD4 percent Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 3rd International Workshop on HIV Pediatrics Cumulative probability of switch/planned TI Cumulatieve incidence (%) 60 switch/TI 50 40 36% (31%-41%) 30 switch 20 planned TI 10 0 0 2 4 6 8 10 Number of years from cART initiation Figure 4. Analysis of 437 European infants starting their first antiretroviral combination determined that almost two thirds (64%) were still taking their initial regimen after 5 years of follow-up. Source: Martina Penazzato, University of Padova, Italy, and Clinical Trials Unit, Medical Research Council, London, for the European Pregnancy and Paediatric HIV Cohort Collaboration.19 change 6% (IQR -6% to 16%), and median CD4 Z score change 0.92 (IQR -0.14 to 2.34). Median CD4 Z score rose by 2.29 in infants starting an NNRTI plus three NRTIs, compared with 0.65 in those starting an NNRTI plus two NRTIs and 0.91 in those starting a ritonavir-boosted PI. Infants whose mothers received ART during pregnancy had smaller CD4 Z score gains at 12 months than infants whose mothers did not receive ART. During a median follow-up of 5.9 years (IQR 2.3 to 7.6), 77 of 437 infants (18%) switched to a secondline regimen. Among those who switched, 61% had never reached a viral load below 400 copies/mL on their first regimen. Switch rates were 3.9 per 100 person-years in children starting an NNRTI plus two NRTIs, 2.1 per 100 person-years in children starting an NNRTI plus three NRTIs, and 1.3 per 100 personyears in those starting a boosted PI and two NRTIs; Penazzato cautioned that these results rest on sparse data. Ever attaining a viral load below 400 copies/ mL lowered chances of switching 77% (AOR 0.23, 95% CI 0.15 to 0.37, P < 0.001), whereas a confirmed viral rebound raised chances of switching almost 23 times (AOR 22.8, 95% CI 5.47 to 95.14, P < 0.001). While 28% of children had one or more treatment interruptions, 12% had a planned treatment interruption. Among all children with a treatment interruption, 38% had a viral load below 400 copies/ mL at the first interruption. Cumulative probability of a regimen switch or planned treatment interruption was 36% at 5 years (Figure 4). Thus at the 5-year point 64% of children had taken their initially prescribed regimen consistently. This study is the first to find that an NNRTI plus three NRTIs promote better virologic and immunologic responses than standard three-drug regimens that include an NNRTI or a boosted PI. Penazzato noted that further evaluation of first-line cART options and interruption strategies awaits final analyses of the randomized CHER9 and ARROW20 trials. Viral load at 6 weeks of age predicts mortality up to 1 year Zimbabwean infants with a viral load above the group median at 6 weeks of age had a tripled risk of death during the first 6 months of life, according to results of a retrospective analysis involving children in the years before access to antiretroviral therapy or cotrimoxazole.21,22 Among infants alive at 6 months, a high 6-week viral load doubled the risk of death through 1 year of age. Although viral loads are usually very high in infancy, US research has linked viral load in infants and children with disease progression and death.23-26 Because the value of viral load in predicting HIV disease progression or death in African infants remains unclear, researchers in Zimbabwe undertook this study. Andrew Prendergast (ZVITAMBO Project, Harare) and colleagues analyzed data from the ZVITAMBO vitamin A trial, which took place in Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 29 Report Table 1. Predictors of mortality by 6 and 12 months of age in Zimbabwe Adjusted hazard ratio 95% CI P Infant viral load above vs below median* 3.07 2.16 to 4.38 <0.001 Maternal CD4 count 200-350 vs >350 2.01 1.34 to 3.01 0.001 Maternal CD4 count <200 vs >350 1.70 1.11 to 2.62 0.015 Infant viral load above vs below median* 2.16 2.03 to 4.38 <0.001 Maternal CD4 count 200-350 vs >350 1.69 1.07 to 2.67 0.025 Maternal CD4 count <200 vs >350 1.94 1.19 to 3.16 0.008 Mortality through 6 months of age Mortality through 12 months of age *Median 1.59 million copies/mL. Source: Andrew Prendergast, ZVITAMBO Project, Harare, Zimbabwe.21 Zimbabwe from 1997 through 2001.22 The analysis involved 453 HIV-positive infants with a 6-week plasma sample available, including 151 infected in utero and 302 infected intrapartum. Those two groups did not differ significantly in gender, weight at birth or 6 weeks, hemoglobin at birth or 6 weeks, or rates of vaginal delivery (87.8% and 90.3%) or mixed breastfeeding (58.9% and 56.3%). Mothers of children infected in utero had a significantly higher CD4 count (median 405 versus 355 cells/mm3, P = 0.004) but also a higher viral load (mean 4.52 versus 4.38 log10 copies/mL, P = 0.055). Mortality through 6 months was marginally lower in the intrapartum group than the intrauterine group (P = 0.07) and in intrapartum girls versus intrauterine girls (P = 0.07). Median viral load at 6 weeks stood at 1,589,041 copies/mL (IQR 514,809 to 4,965,854) and did not differ significantly between the intrapartum group and the intrauterine group or between boys and girls. Multivariate analysis determined that a 6-week viral load above the median value tripled the risk of death through 6 months of age and doubled the risk of death through 12 months of age among those alive at 6 months (Table 1). Lower maternal CD4 count also independently predicted infant mortality at 6 and 12 months. The researchers concluded that viral load measured at a single point—at 6 weeks of age—independently predicts mortality in infants who have not begun antiretroviral therapy. They proposed that “rapid suppression of viremia may be important to reduce mortality” in infants, a concept supported by results of the CHER trial.9 30 Viral suppression rates similar with continuous therapy and treatment interruption Two years after a planned treatment interruption (PTI) trial in children ended, viral suppression rates were high and similar in children randomized to the PTI arm and to the continuous therapy arm.27 Nadir CD4 percent above 20% was associated with better CD4 recovery after cART resumed in the PTI arm. The PENTA 11 trial randomized 109 children to continuous cART or CD4-guided PTI if they had (1) a viral load below 50 copies/mL and (2) a CD4 percent at or above 30% for 2- to 6-year-olds or a CD4 percent at or above 25% and a CD4 count at or above 500 cells/mm3 if they were 7 to 15 years old.28 After a median follow-up of 130 weeks, no child in either group had a CDC stage C event, though the PTI group endured more minor clinical events. Alexandra Compagnucci (INSERM SC10, Paris) presented long-term follow-up on 101 of the 109 PENTA 11 participants, 79 from Europe and 22 from Thailand. Fifty children were in the PTI arm and 51 in the continuous-therapy arm. All 101 children completed 1 year of follow-up, 95 completed 2 years, and median follow-up stood at 4.6 years. At the end of PENTA 11, median ages were 11.3 in the PTI group and 12.1 in the continuous group, median CD4 percents 32% and 36%, and proportions with a viral load below 50 copies/mL 60% and 76%. Up to the end of the main trial, children in the PTI arm spent 45.2% of the time off cART, compared with 4.1% in the continuous-therapy arm. During overall follow-up after the end of the main trial, PTI participants spent 10.4% of the time off cART, Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 3rd International Workshop on HIV Pediatrics Table 2. CD4 and viral load outcomes 1 and 2 years after the end of the PENTA 11 treatment interruption trial Mean or proportion CD4 percent CD4 count HIV RNA <50 copies/mL Follow-up year Continuous cART PTI Difference or risk ratio: PTI vs continuous P +1 35.8 32.7 -3.5 0.014 +2 36.0 34.6 -1.6 0.27 +1 925 808 -126 0.048 +2 864 832 -42 0.44 +1 90% 77% 0.85 0.074 +2 86% 82% 0.95 0.57 PTI: Planned treatment interruption. Source: Alexandra Compagnucci, INSERM SC10, Paris.27 compared with 1.3% of time in the control arm. In an analysis excluding follow-up of children on a PTI at the end of the main trial before they restarted cART, proportion of time spent off cART was 4.7% in the PTI group and 1.3% in the continuous-cART group. At the end of long-term follow-up, no child had died and no new CDC stage C event developed. One child had a new CDC stage B event, osteomyelitis, which developed during the main trial in the PTI group. Weight- and height-for-age Z scores did not differ significantly between study arms after 1 and 2 years of long-term follow-up. Two children in each study arm switched antiretrovirals for treatment failure or toxicity after the end of the main trial. Five children in the PTI group and none in the control group switched antiretrovirals for regimen simplification. One year after the main trial ended, CD4 percent and CD4 count were significantly lower in the PTI group than in the continuous-cART group (Table 2). But 2 years after the main trial ended, those differences diminished and were no longer statistically significant. One year after the main trial ended, the proportion of children with a viral load below 50 copies/mL was marginally lower in the PTI group (P = 0.074), but that difference was negligible 2 years after the main trial ended (Table 2, Figure 5). In an adjusted analysis of CD4 recovery based on measurements since children resumed cART after their latest treatment interruption, estimated mean difference in CD4 percent between those with a nadir at or above 20% versus below 20% was 3.7% (95% CI 0.7 to 6.7, P = 0.02). Mean total cholesterol in the 24 months after the end of the main trial did not differ between the PTI group and the control group. Proportion HIV-1 RNA <50 copies/ml from end of main trial Proportion <50 c/ml (%) 100 80 60 CT PTI 40 20 0 0 6 12 18 Months from end of main trial 24 Figure 5. Two years after the end of the PENTA 11 treatment interruption trial,28 proportions of children with a viral load below 50 copies/mL were virtually identical in the planned treatment interruption (PTI) group and the continuous-therapy (CT) group. Source: Alexandra Compagnucci, INSERM SC10, Paris.27 Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 31 Report Table 3. Kaplan-Meier 1-year mortality risk according to CD4 percent, CD4 count, and age Age of child Under 24 months 24 to 35 months 36 months or older CD4% <10 21.2% 4.4% 4.6% CD4% 10 to 14.9 7.4% 2.2% 1.9% CD4% 15 to 19.9 3.0% 1.3% 0.9% CD4% >20 2.2% 1.3% 0.5% CD4 count <100 35.1% (No events) 8.6% CD4 count 100 to 199 8.8% (No events) 2.9% CD4 count 200 to 349 10.2% 2.7% 1.6% CD4 count 350 to 499 10.0% 2.3% 0.9% CD4 count 500 to 749 8.5% 3.6% 0.4% Source: Mary-Ann Davies, University of Cape Town, South Africa, for IeDEA-SA.29 Although antiretroviral treatment interruptions are considered unsafe for adults, viability of this strategy remains an important research topic in children, who face decades of continuous treatment unless interruptions prove feasible. PENTA investigators are pursuing neurocognitive and immunologic substudies in trial participants. CD4 predictors of mortality as guides to switching antiretrovirals When viral load monitoring is not possible, a CD4 percent below 10% may serve as an antiretroviralswitch signal for children 24 to 35 months old, according to results of a 17,000-child analysis in the International Epidemiologic Databases to Evaluate AIDS in Southern Africa (IeDEA-SA).29 A lower threshold might be considered for older children, IeDEA-SA researchers suggested. Clinicians without access to routine viral load monitoring must rely on clinical and CD4 changes to decide when to switch an adult or child from one regimen to another. But because immunologic failure has a low positive predictive value for virologic failure, reliance on CD4 changes may lead to unnecessary switching in a person without virologic failure. Mary-Ann Davies (University of Cape Town, South Africa) and IeDEA-SA colleagues proposed that an alternative approach for clinics without viral load monitoring could be based on CD4 percent and CD4 count thresholds below which mortality begins 32 to increase rapidly in distinct age groups.29 They planned this study to determine 1-year mortality risk according to age and current CD4 percent and count of children taking cART in southern Africa. The analysis focused on 17,173 children under 16 years old beginning a first-line regimen and having at least 180 days of follow-up in IeDEA-SA collaborating cohorts. The investigators considered every time at which CD4 count or percent was measured after the first 4.5 months (135 days) on cART as the start of an observation with time set at 0 and continuing until the first of (1) 365 days, or (2) date of death, or (3) date of transfer out, or (4) date of last visit if the child was lost to follow-up or remained in care at the time of data transfer. Independent variables in Weibull models to predict probability of death were age category (under 24 months, 24 to 35 months, 36 months or older), CD4 percent and its square at the start of the observation, and CD4 count and its square at the start of the observation. As one would expect, 1-year risk of death was higher at lower CD4 percents or counts and younger ages (Table 3). Davies cautioned that these estimates are based on small numbers of children with low CD4 percents or counts in the two younger age groups. She added that censoring due to loss of follow-up may have resulted in an underestimate of mortality risk. In children 24 to 35 months old, the Weibull model Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 3rd International Workshop on HIV Pediatrics determined that mortality risk reached 5% at a CD4 percent of 10%, and started to rise steeply as CD4 percent dropped further. In older children, mortality risk reached 5% at a CD4 percent of 5% and rose more steeply below this value. A CD4 percent of 10% is the 2010 WHO threshold for switching cART in 24- to 59-month-old children.10 WHO guidelines call for switching 24- to 59-month-old children at a CD4 count of 200 cells/mm3, which in IeDEA-SA children this age was associated with a mortality risk of nearly 10%, rising steeply as CD4 count dropped further. In children older than 35 months mortality risk was 5% at a CD4 count of 100 cells/mm3 and rose steeply as CD4 dropped below this. This is the WHO guideline threshold for switching to second-line cART in children 60 months old and older. The IeDEA-SA investigators reached the following conclusions for age-related risk of death based on CD4 percent and count: • The WHO guideline for switching to second-line cART at a CD4 percent of 10% is associated with a 5% risk of dying in the next year for all children 24 months old and older. • For children 36 months old and older, the risk of dying reaches 5% only at a CD4% of 5%. • For children 24 to 35 months old, 1-year mortality risk rapidly approaches 10% at a CD4 count of 200 cells/mm3 and rises steeply as the CD4 count falls further. • For children 36 months old and older, a CD4 count of 100 cells/mm3 is associated with less than a 5% risk of death in the next year. These findings are relevant to the continuing controversy over whether viral load monitoring should be routine for children in resource-poor settings, a question formally debated at the Pediatrics Workshop (see next section). Should viral load monitoring be routine in resource-poor settings? No clinician would dream of caring for HIV-positive children in high-income countries without regular viral load monitoring. US guidelines, for example, say “HIV RNA copy number should be assessed as soon as possible after a child has a positive virologic test for HIV and every 3 to 4 months thereafter; more frequent evaluation may be necessary for children experiencing virologic, immunologic, or clinical deterioration or to confirm an abnormal value.”12 But in countries where viral load monitoring remains too high in cost and too hard to access, the question of routine testing remains unresolved for children and adults. WHO guidelines for infants and children state that “viral load determination is desirable, but not essential, prior to initiating ART” and that “viral load should be assessed to confirm clinical or immunological failure where possible, prior to switching a treatment regimen.”10 Pediatrics Workshop attendees mulled the question of routine HIV RNA monitoring during a debate by two leading international pediatric investigators—Louise Kuhn (Columbia University, New York) arguing that monitoring should be routine for children in resourcelimited settings,30 and Diana Gibb (Medical Research Council, UK) maintaining that it should not.31 Kuhn launched her case for routine viral load testing by citing recent research indicating that annual monitoring could cut the cART failure rate 75% in Thai children,32 that routine testing using the WHO failure threshold of 5000 copies/mL “adds independent predictive value to immunological and clinical assessments for identification of children receiving HAART who are at risk for significant HIVrelated illness,”33 and that regular monitoring may prevent emergence of NNRTI mutations in children whose NNRTI regimen is failing.34 The argument favoring routine viral load monitoring, Kuhn maintained, rests mainly on three pillars: • Plasma HIV RNA is an excellent marker that directly tracks the underlying mechanism driving HIV disease. • Viremia is an early marker allowing for prompt action. • Cost arguments favor routine monitoring over the long term. Viral load assays are accurate and reproducible, Kuhn noted, and they strongly predict negative clinical outcomes in children taking cART. In a study of treated Brazilian children, a viral load above versus below 5000 copies/mL independently raised the risk of a WHO stage 3 or 4 event by more than 80% (adjusted hazard ratio [AHR] 1.81, 95% CI 1.05 to 3.11, P = 0.033).33 Every 0.5-log higher viral load boosted that risk almost 15% (AHR 1.14, 95% CI 1.03 to 1.27, P = 0.016). When an antiretroviral regimen fails, Kuhn added, rebounding viremia is an “early warning signal,” spiking well before CD4 counts wane or HIV-related symptoms emerge. Although Gareth Tudor-Williams emphasized the overall slow emergence of NRTI mutations upon failure of an NNRTI regimen in PENPACT 1 (see above),15,16 Kuhn cited additional data from this trial indicating that three or more NRTI mutations arose significantly more often in children randomized to switch from an NNRTI regimen at 30,000 copies/ Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 33 Report Table 4. Accumulation of NRTI mutations with low versus high viral load switch in PENPACT 116 Switch from NNRTI regimen at: 1000 copies/mL 30,000 copies/mL Number of children 68 64 1 to 2 major NRTI mutations 22% 20% 3 or more major NRTI mutations — 11% 1 to 2 TAMs 5% 8% 3 or more TAMs — 7% P 0.001 0.08 TAMs, thymidine analog mutations. mL rather than 1000 copies/mL (P = 0.001) (Table 4).16 There was also a trend toward more frequent emergence of three or more thymidine analog mutations in the 30,000-copy-switch group (P = 0.08). Although the high initial cost of viral load testing weighs against its use in resource-limited settings, Kuhn argued that long-term costs favor routine monitoring if it delays use of more expensive secondline regimens. The Thai study cited above estimated a cost of $359 for preventing 1 year of virologic failure through optimal viral load monitoring versus $3393 for preventing 1 year of virologic failure and including antiretroviral costs.32 These investigators proposed that optimal monitoring timing in similar settings may be once in the first 6 months of cART then annually. Kuhn maintained that routine viral load testing may also have spinoff benefits, such as providing opportunities for caregiver and child education and for adherence counseling. The question is not whether viral load monitoring should be routine, she concluded, but (1) how frequently testing should be done, (2) what viral load thresholds should be used, (3) what response algorithms will work best, (4) how monitoring systems can be improved, (5) how testing can be made simpler and less expensive, and (6) how more and better pediatric treatment options can be developed. Framing her rebuttal against routine viral load monitoring in children seen in resource-constrained settings, Diana Gibb posed three questions:31 1. What impact does viral load monitoring have on important outcomes? 2. How affordable and feasible is viral load monitoring in low-income settings? 3. What are the competing priorities? 34 The usual answer to the first question, Gibb observed, is to guide decisions on when to start cART and when to switch cART. But a 2003 metaanalysis of 4000 children not receiving cART or zidovudine monotherapy determined that, although CD4 measures and viral load both independently predicted mortality in the short term, CD4 readings were much stronger predictors of disease progression and death.35 Although viral load is also an independent predictor of disease progression before and during cART in adults, Gibb observed, CD4 measures are a much better predictor of immediate risk of death, and several studies found that viral load adds little further prognostic information to CD4 count: The DART trial in Uganda and Zimbabwe enrolled 3321 symptomatic, antiretroviral-naive adults with CD4 counts below 200 cells/mm3 and randomized them to laboratory and clinical monitoring or to clinically driven monitoring alone.36 Gibb, one of the DART investigators, noted that CD4 monitoring every 12 weeks had no impact on disease progression during the first 2 years of cART, and after that it had only a small impact on progression that appeared to be driven by a later switch to clinical monitoring. Although DART did not assess viral load monitoring, she explained, after 6 years of cART about 80% of DART patients had a viral load below 50 copies/mL and about 75% were still taking their first regimen. The unpublished HBAC trial randomized 1200 Ugandan adults to (1) clinical monitoring, (2) clinical plus CD4 monitoring, or (3) clinical plus CD4 plus virologic monitoring. When compared with clinical monitoring alone, CD4 and viral load monitoring had a marginally significant benefit in preventing AIDS or death. But there was no significant difference Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 3rd International Workshop on HIV Pediatrics between the CD4-and-viral load monitoring group and the CD4-and-clinical monitoring group. The PHPT-3 trial randomized 716 Thai adults to a conventional viral load-driven cART switch strategy or to a CD4-driven switch.37 Through 60 months of cART, time to clinical failure (CD4 count below 50 cells/mm3, new AIDS event, or death) was almost identical in the two study arms, and 38% of events occurred within the first 6 months of treatment. In the recently completed PENPACT 1, Gibb reminded attendees, viral suppression rates in children taking cART for 4 or more years did not differ between those who switched when the viral load increased to 1000-copies/mL versus those who waited until the viral load rose to 30,000-copies/mL (around a year later on average), regardless of whether they were taking a PI or an NNRTI.16 Although several groups have described adult African data showing that those switching treatment based on clinical symptoms may have an undetectable viral load (and are thus deemed to have switched “unnecessarily”), a recent analysis of data from the DART study showed this was rare if the CD4 was below 250 cells/mm3.38 Therefore, Gibb argued, use of a 250-CD4 threshold could prevent many “unnecessary” switches to second-line treatment. (Gibb did not mention the 17,000-child IeDEA-SA cohort study summarized in the preceding section of this article, which found that a CD4 percent below 10% may be a useful antiretroviral-switch signal for children 24 to 35 months old.29) To address the question of cost, Gibb cited four model-based cost-effectiveness studies of viral load monitoring, which calculated costs per lifeyear gained ranging from $2526 to $15,25039-42— all estimates above the 3-fold gross domestic product cutoff used to determine cost-effectiveness of interventions in resource-limited countries.43,44 Gibb concluded that “routine viral load monitoring is not necessary and should not be routine for children in resource-limited settings” because: (1) its impact on disease-free survival is very modest, (2) it is not cost-effective, and (3) “its routine use will be a barrier to the goal of maximising ART coverage and actually lead to lives lost.” Evidence from two studies on second-line switching Although HIV Pediatric Workshop attendees weighed data from four studies on first-line cART options for children in countries rich and poor,13-19 for a growing number of children everywhere the question has become which second-line regimen to chose—if there is a choice. Two workshop studies addressed second-line questions: A six-country African study assessed durability of first-line regimens and reasons for switching,45 while an Asian study analyzed efficacy of ritonavir-boosted PI regimens after failure of a nonnucleoside combination.46 IeDEA-West African investigators reported that only small proportions of children who met clinical or immunologic failure criteria in this region switched to a second-line regimen. These researchers collected data from nine clinical centers in six countries— Benin, Burkina Faso, Côte d’Ivoire, Ghana, Mali, and Senegal.45 They included children under 16 years old starting three or more antiretrovirals from 2000 through 2009 and with at least one available measurement of CD4 count, CD4 percent, or WHO clinical stage during the first-line regimen. Routine viral load monitoring is not available throughout most of West Africa. The researchers defined clinical failure as appearance or reappearance of a WHO clinical stage 3 or 4 event after at least 24 weeks of cART in a treatmentadherent child. They defined immunologic failure as reaching or returning to a CD4 percent below 10% for 2- to 5-year-old adherent children or reaching or returning to a CD4 count below 100 cells/mm3 for adherent children 5 or over after the first 24 weeks of therapy. A regimen switch meant (1) changing from three NRTIs to two NRTIs plus a one PI or one NNRTI, or (2) changing from two NRTIs plus one PI to three NRTIs or to two NRTIs plus one NNRTI, or (3) changing from the PI class or the NNRTI class. In 2797 children the overall 24-month probability of switching to a second-line regimen was 23.3% (95% CI 21.5 to 25.4). In the 2450 children (88%) without study-defined clinical or immunologic failure, the 24-month probability of switching was 25.3% (95% CI 23.2 to 27.7). The 227 children (8%) with clinical failure had a 24-month switching probability of only 2.1% (95% CI 0.8 to 5.5). And the 120 children with immunologic failure (4%) had a 24-month switching probability of 26.4% (95% CI 18.8 to 36.4). Median time to switch in all 456 children (16%) who changed to a second-line regimen was 7 months (IQR 3 to 16). A large majority of these children—425 or 93.2%—switched without meeting clinical or immunologic failure criteria. Withdrawal of nelfinavir in 2007 accounted for most of this nonfailure switching. Among switchers, 285 children (62.5%) switched from a first-line nelfinavir regimen. The low switch rates in children who did meet clinical or immunologic failure criteria, the investigators Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 35 Report suggested, indicates “missed opportunities for switching to second-line therapy,” perhaps because key second-line agents such as lopinavir/ritonavir were not available. The IeDEA-West Africa team stressed the importance of advocating for access to monitoring tools and to second-line antiretrovirals for pediatric HIV programs. Researchers from the TREAT Asia Pediatric HIV Observational Database (TApHOD) retrospectively studied 153 children who switched from a first-line NNRTI-based regimen (taken for at least 24 weeks) to a PI-based regimen (taken for at least 24 weeks) up to 20 March 2010.46 All children were younger than 18 years old at the switch and had not taken a PI before the switch. The study group included 80 girls (52%) and 73 boys with a median age of 10 years (IQR 7 to 12), a median CD4 percent of 12.5% (IQR 5.2 to 20.0), a median CD4 count of 237 cells/mm3 (IQR 90 to 466), and a median viral load of 4.6 log10 copies/mL (IQR 3.9 to 5.0). Proportions of children with WHO stage 1, 2, 3, and 4 disease were 7%, 25%, 42%, and 26%. Median first-line duration was 2.6 years (IQR 1.5 to 3.8). Most children (83%) switched to a lopinavir/ ritonavir regimen, while 16% switched to an indinavir/ ritonavir regimen and 1% to atazanavir/ritonavir. Median weight-for-age Z score did not change significantly from the switch point (week 0) (-1.9) to week 48 (-1.9) or week 96 (-1.7). In contrast, median CD4 percent rose from 13.8% at week 0 to 20.3% at week 48 and 22.8% at week 96 (P < 0.001). Proportions of children with viral loads below 400 and 50 copies/mL were 0% and 0% at week 0, 60% and 49% at week 48, and 65% and 62% at week 96 (P < 0.001 for both comparisons). Median total cholesterol rose significantly from 167 mg/dL at week 0 to 187 mg/dL at week 96 (P = 0.002), while median triglycerides climbed from 120 mg/dL at week 0 to 160 mg/dL at week 96 (P = 0.006). Median high-density lipoprotein cholesterol did not change significantly over the study period. Multivariate analysis identified four independent predictors of virologic suppression below 400 copies/mL at 48 weeks: • Longer duration of first-line NNRTI regimen: OR 1.8 per additional year (95% CI 1.2 to 2.9), P = 0.006 • Younger age: OR 0.8 per additional year (95% CI 0.7 to 0.9), P = 0.007 • Higher weight-for-age Z score: OR 1.7 per standard deviation (95% CI 1.1 to 2.7)), P = 0.020 • HIV RNA below 10,000 copies/mL at switch: OR 12.6 (95% CI 1.9 to 81.8), P = 0.049 36 Two variables independently predicted immune recovery, defined as a CD4 percent at or above 25% in children younger than 5 years or a CD4 count at or above 500 cells/mm3 in older children: • Younger age: OR 0.8 per additional year (95%CI 0.7 to 0.9), P < 0.001 • CD4 count at or above 200 cells/mm3 at the switch: OR 7.7 (95% CI 2.8 to 21.5), P = 0.003 The TApHOD researchers concluded that one third of children in this Asian cohort do not reach an undetectable viral load 2 years after switching from an NNRTI regimen to second-line lopinavir/ritonavirbased cART that includes recycled NRTIs. They observed that hyperlipidemia was common after such a switch. PMTCT: Emerging Issues, Persisting Controversies South African PMTCT program cuts early transmission rate below 4% Nine years after South Africa launched a program for prevention of mother-to-child transmission (PMTCT) of HIV, the transmission rate fell below 4% in the first 4 to 8 weeks of life, according to preliminary results of a nationwide population-based analysis.47 South Africa began its PMTCT program in 2002 with single-dose nevirapine and later added zidovudine or combination antiretrovirals for mothers (depending on maternal CD4 count) and extended zidovudine or nevirapine for infants. Estimated national mother-tochild transmission rates in infants up to 2 months old stood at 8.2% in 2008 and 5.8% in 2009. Ameena Goga (Medical Research Council, South Africa) presented results of the new study, which involved caregiver-infant pairs making the 6-week infant immunization visit from June through December 2010. The investigators (Goga, Thu-Ha Dinh and Debra Jackson) identified HIV-exposed infants through caregiver report of maternal HIV positivity or through ELISA analysis of dried blood spots. They tested samples of HIV-exposed infants with qualitative HIV DNA PCR to identify infected infants. The researchers determined that 3003 infants in 9915 caregiver-infant pairs (30.3%) had been exposed to HIV, and 2958 of these 3003 infants (98.5%) had PCR test results. Most mothers of HIV-exposed children (79.8%) were not married or cohabitating. In the 8 days before the study visit, 20% of women practiced exclusive breastfeeding, 18% mixed breastfeeding, and 62% no breastfeeding. Almost two thirds of Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 3rd International Workshop on HIV Pediatrics Table 5. Factors associated with mother-to-child transmission at 6 weeks in South Africa Adjusted odds ratio 95% CI Either maternal ARV or infant ARV prophylaxis 5.2 2.7 to 10.0 <10 weeks of maternal zidovudine plus infant ARV prophylaxis 2.4 1.2 to 5.1 11 to 30 weeks of maternal zidovudine plus infant ARV prophylaxis 1.7 0.9 to 3.5 1.6 1.0 to 2.5 Compared with maternal cART Compared with exclusive breastfeeding or no breast milk Mixed breastfeeding ARV, antiretroviral; cART, combination antiretroviral therapy. Source: Ameena Goga, Medical Research Council, South Africa.47 mothers of HIV-exposed children (62%) said the pregnancy was unplanned. Infant HIV prevalence at 4 to 8 weeks (weighted for sample realization and population live births in 2009) ranged from 0.6% (95% CI 0.3 to 0.9) in Gauteng province to 2.1% (95% CI 1.5 to 2.7) in Mpumalanga province. National infant HIV prevalence stood at 1.1% (95% CI 0.1 to 1.3). Weighted MTCT rate at 4 to 8 weeks ranged from 1.7% (95% CI 0.1 to 4.2) in Northern Cape province to 6% (95% CI 3.8-8.2) in Free State province. The national weighted MTCT rate was 3.5% (95% CI 2.9 to 4.1). Compared with maternal cART, use of either maternal or infant antiretroviral prophylaxis raised the risk of MTCT more than 5 times, and 10 or fewer weeks of maternal zidovudine plus infant antiretroviral prophylaxis more than doubled the risk (Table 5). Compared with maternal cART, there was a borderline association between 11 to 30 weeks of maternal zidovudine plus infant antiretrovirals and higher MTCT risk. Compared with exclusive breastfeeding or no breastfeeding, mixed breastfeeding increased the MTCT risk 60%. Variables that did not affect transmission risk in this analysis were caesarean section, planned versus unplanned pregnancy, or physician versus nonphysician birth attendant. Goga and colleagues cautioned that their analysis is limited by its reliance on primary or community health centers, by exclusion of sick infants needing emergency care and infants older than 8 weeks, by a sample realization below 75% in three provinces, and by potential recall bias. Given those limitations, the investigators concluded that the national South African MTCT rate 4 to 8 weeks after delivery stands at approximately 3.5%. They suggested that rate may be lowered by optimizing antiretroviral use during pregnancy and perinatally and by reducing mixed feeding. High HIV incidence during pregnancy and breastfeeding in Botswana Batswana women who have a negative HIV test during pregnancy remain at high risk for HIV infection later in pregnancy and during breastfeeding, according to results of a 417-woman study.48 Centers for Disease Control and Prevention (CDC) investigators warned that undiagnosed incident HIV infection in pregnant or nursing mothers could result in unanticipated transmission of the virus to infants. HIV testing early in pregnancy is “a critical first step” to reducing HIV transmission, Lydia Lu and CDC colleagues noted. But an early negative test is no guarantee that women will remain free of HIV throughout pregnancy and nursing. In Botswana more than 95% of pregnant women get tested for HIV at a median 22 weeks gestation, and HIV prevalence stood at 32.5% among 43,000 women tested at antenatal clinics in 2009. Early infant testing indicates that 4.2% of HIV-exposed infants in Botswana test positive on their first test. In 2008 CDC-Botswana investigators retested 400 women in a postlabor ward after they had tested negative during pregnancy. HIV incidence in these women was 1.3%. As a result the CDC recommended routine HIV retesting during the third trimester or in the maternity ward for women missed in the third trimester. These researchers planned a Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 37 Report follow-up study for 2010 to determine HIV prevalence in women during the first postpartum year after they tested negative during pregnancy. The 2010 study involved 417 women with a documented negative HIV test during pregnancy who brought a 9- to 15-month-old child to an immunization clinic. Women were tested and counseled according to the standard Botswana algorithm, and they were interviewed about risk behaviors and feeding practices. Eighteen of these 417 women (4.3%) had a positive HIV test, and the investigators estimated 52-week incidence at 3.8%. Because 43,000 women become pregnant in Botswana every year and 32.5% tested positive in antenatal clinics in 2009, the CDC team figured that the remaining 67.5%, or 29,025 women, are assumed to be HIV negative (Figure 6). Based on the 2008 incidence study findings, the researchers estimated that 377 of these 29,025 women (1.3%) become infected during late pregnancy. The 2010 study estimated 1103 (3.8%) become infected during breastfeeding. Next the CDC team estimated the impact of maternal HIV incidence on new infections in infants during 2010. To do so the investigators relied on a 2005 study of Zimbabwean women who became infected with HIV during pregnancy or while breastfeeding.49 That study found that 73% of infants exposed to an incident HIV infection in late pregnancy became infected, and 36% of infants exposed to an incident HIV infection during breastfeeding became infected. If 73% of the 377 women infected during pregnancy in the Botswana study transmitted HIV, 275 infants would be infected. If 36% of the 1103 women infected during breastfeeding in the Botswana study transmitted HIV, 397 infants would be infected. Early infant testing indicates that the transmission rate to HIV-exposed infants in Botswana (where PMTCT is mature and has high uptake) is only 4.2%, or about 587 infants annually. Thus the estimated total number of infected infants is 1259, with 672 of those 1259 (275 + 397, or 53.4%) infected through an undiagnosed maternal HIV infection acquired later in pregnancy or while nursing, and in the absence of PMTCT interventions. The 18 HIV-exposed infants of mothers with a new HIV diagnosis in this study had a median age of 10 months when the infants were tested for HIV. All 18 infants had some exposure to breast milk, and 5 were still breastfeeding at recruitment for the study. Three of 18 infants (17%) had a positive HIV test; because the 5 HIV-exposed infants who were breastfeeding at the time of recruitment did not return for testing, their final HIV status was not determined. In the entire study cohort, 259 infants (62%) were breastfeeding at the time of recruitment. Overall median age at weaning was 7 months. Estimated infections in pregnant and post-partum women, Botswana 2010 43000 pregnant women Women diagnosed before during ANC 13975 (32.5%)* * 2009 ANC Sentinel Surveillance Women assumed to be HIV-negative 29025 (67.5%) Women infected during late pregnancy 377 (1.3%) Women infected while breastfeeding 1103 (3.8%) Figure 6. Based on 2008 and 2010 studies of 417 Batswana women who tested negative for HIV during pregnancy and were retested at the time of delivery (2008) or in the year after delivery (2010), CDC researchers estimated that 1.3% of pregnant women became infected with HIV during late pregnancy and 3.8% became infected while breastfeeding in 2010. Source: Lydia Lu, Centers for Disease Control and Prevention.48 38 Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 3rd International Workshop on HIV Pediatrics Intrapartum perinatal transmission rate Predicted IP transmission rate: With vs. Without sdNVP 0.05 standard treatment standard treatment + sdNVP 0.04 0.03 0.02 0.01 0 3 to 4 –fold reduction 0 10 20 30 40 50 60 Duration of maternal ZDV treatment (days) Figure 7. Bayesian modeling determined that adding single-dose nevirapine (sdNVP) to maternal lopinavir/ ritonavir-based cART would lower intrapartum HIV transmission risk 3- to 4-fold. Source: Marc Lallemant, Chiang Mai University, Chang Mai, Thailand.50 Lu and coworkers concluded that Batswana women assumed to be HIV-negative during pregnancy and the first year after delivery will breastfeed, although some of them may have acquired HIV infection after their initial negative test. The investigators proposed that infants may remain at significant risk for HIV infection in Botswana even when mothers have a negative HIV test during pregnancy. Modeling suggests advantage in adding sdNVP to maternal cART A Bayesian inference model determined that adding single-dose nevirapine (sdNVP) to lopinavir/ritonavirbased cART for pregnant women with fewer than 8 weeks of antiretroviral prophylaxis would lower the risk of perinatal HIV transmission 3- to 4-fold.50 Marc Lallemant (Chiang Mai University, Chang Mai, Thailand) and colleagues based the model on data from three PMTCT trials in Thailand. PHPT-5, a 2008-2010 randomized trial of three PMTCT strategies (clinicaltrials.gov NCT00409591),51 stopped prematurely when Thailand revised its PMTCT guidelines to recommend lopinavir-based cART for all HIV-positive pregnant women as soon as possible after the first trimester. When trial data were analyzed, the overall transmission rate was 2.2%, with no difference between treatment arms. Multivariate analysis determined that shorter zidovudine prophylaxis duration almost doubled the risk of HIV transmission (AOR 1.8 per weekly decrement, 95% CI 1.3 to 2.4), while each log10 higher viral load more than doubled the transmission risk (AOR 2.3, 95% CI 1.1 to 4.8). In planning the Bayesian analysis, Lallemant and coworkers hypothesized that pregnant women with fewer than 8 weeks of prophylaxis did not have enough time to suppress viral replication and that adding maternal and infant sdNVP to maternal lopinavir-based cART would significantly reduce the risk of intrapartum transmission. Using transmission data from 3876 women enrolled in PHPT-1, 2, and 5, the model assumed (1) that maternal prophylaxis duration directly affects viral load at delivery, and (2) that viral load at delivery directly affects risk of intrapartum transmission. Monte Carlo simulations predicted transmission rates with and without sdNVP in relation to duration of maternal and infant prophylaxis. The model estimated an HIV transmission rate of 2.6% (95% probability interval [PI] 0.5% to 9.2%) for the standard of care (lopinavir/ritonavir-based cART during pregnancy and zidovudine for the newborn). Adding sdNVP to the standard of care lowered the estimate to 0.8% (95% PI 0.1% to 2.9%). Maternal sdNVP would decrease the risk of intrapartum transmission approximately 60% (AOR 0.39, 95% CI 0.21 to 0.67). Lallemant and colleagues calculated a 3- to 4-fold lower intrapartum transmission risk with addition of sdNVP (Figure 7). Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 39 Report Higher CD4 count during pregnancy (average + 1 standard deviation [SD] versus average) would lower intrapartum transmission risk more than 35% (AOR 0.63, 95% CI 0.49 to 0.80). Gestational age at delivery (average +/- 1 SD versus average) would increase transmission risk (AOR 1.34, 95% CI 1.07 to 1.64). Trial simulation determined that an ethical and scientifically sound single-arm trial with comparison to historical data and with optimal stopping rules could be designed with fewer than 400 women to test the hypothesis that adding maternal and infant sdNVP to the standard of care lowers the risk of HIV transmission. Impact of formula feeding on HIV-exposed infants in Brazil and South Africa HIV-exposed but uninfected formula-fed South African infants had a higher risk of death if born to mothers with a high viral load or if they had a low birth weight, according to results of a 1000-infant study in South Africa and Brazil.52 Karin Nielsen-Saines (University of California, Los Angeles ) and HPTN 040 colleagues reported that high maternal viral load and lower maternal education were associated with infectious significant adverse events in the overall study group. The analysis involved formula-fed infants enrolled in NICHD HPTN 040, a phase 3 trial conducted between April 2004 and January 2011 at 17 sites in Brazil, South Africa, Argentina, and the United States.53 This randomized open-label trial compared the safety and efficacy of three infant antiretroviral regimens to prevent vertical HIV transmission. Overall HIV transmission and intrapartum transmission were lower in the two- and three-drug arms (singledose zidovudine for 6 weeks plus three doses of nevirapine, and single-dose zidovudine for 6 weeks plus nelfinavir and lamivudine for the first 2 weeks) than in the one-drug arm (single-dose zidovudine for 6 weeks). Primary aims of the new analysis were to assess the frequency of and risk factors for severe morbidity and mortality in a representative subcohort of 1000 HIV-exposed but uninfected formula-fed infants from Brazilian and South African HPTN 040 sites.52 The study included live HIV-exposed infants who were HIV-negative at their 3-month visit and completed 6 months of follow-up. The study pool included HIVexposed infants who died before the 6-month visit and were not determined to be HIV-positive until the time of death. The analysis excluded HIV-positive infants, infants with fewer than 6 months of follow-up, 40 and infants with any evidence of breastfeeding. Most mothers of the 1000 selected infants were of black (48.6%) or mixed black/white race (28.6%), were relatively young (median age 26), had relatively high CD4 counts (median 466 cells/mm3), and had detectable virus at delivery (median viral load 13,580 copies/mL). The study group included 766 infants from Brazil and 234 from South Africa. Their median birth weight was 3012.5 g (6.6 pounds). The overall rate of infectious serious (grade 3 to 5) adverse events was 60 per 100 infant-years, and rates were similar in Brazil (61 per 100 infantyears) and South Africa (59 per 100 infant-years). Gastrointestinal serious adverse events were more frequent in South Africa (24.2 versus 4.7 per 100 infant-years), while congenital serious adverse events (usually congenital syphilis) were more frequent in Brazil (21.8 versus 3.6 per 100 infant years). Almost one quarter of infants (23%) had at least one infectious serious adverse event. Mean weight-for-age Z score (WAZ) at birth was lower in South Africa than Brazil (-0.95 versus -0.49), while mean height-for-age Z score (HAZ) at birth was similar in South Africa (-0.40) and Brazil (-0.54) (Figure 8). Defining malnutrition as a WAZ at or below -2, the investigators determined that 11% of South African infants and 6% of Brazilian infants had malnutrition at 6 months. WAZ and HAZ declined in both the South African and Brazilian cohorts from birth to the month-1 visit, then rebounded to near-normal values through month 6 (Figure 8). At 6 months 15 of 234 South African infants (6.4%) had died, compared with 7 of 766 Brazilian infants (0.9%). Six-month mortality incidence was 64.1 per 1000 infant-years in South Africa and 9.1 per 1000 infant-years in Brazil. In comparison, 12-month overall infant mortality incidence stood at 56 per 1000 infant-years in South Africa and 19 per 1000 infant-years in Brazil according to 2006 WHO data. Multivariate analysis identified four factors that independently affected the risk of infant mortality and three that independently affected the risk of infectious serious adverse events in infants: Risk of infant mortality: • South African versus Brazilian birth: AOR 6.22, 95% CI 2.46 to 15.75, P < 0.0001 • Higher maternal viral load at delivery: AOR 1.71, 95% CI 1.01 to 2.91, P = 0.0476 • WAZ at birth < -2 to > -3: AOR 5.16, 95% CI 1.80 to 14.77, P = 0.0022 • WAZ at birth < -3: AOR 6.18, 95% CI 1.80 to 21.31, P = 0.0039 Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 3rd International Workshop on HIV Pediatrics Weight-for-Age and Heigt-for-Age Z-scores by study visit n=1000 Infants - 0.05 -0.09 Brazil WAZ - 0.15 - 0.25 Mean Z- score - 0.35 - 0.45 - 0.55 -0.34 Brazil HAZ - 0.40 - 0.49 - 0.54 -0.53 South Africa WAZ -0.56 - 0.65 -0.67 - 0.75 -0.86 -0.79 - 0.85 - 0.95 -0.95 - 1.05 -0.67 South Africa HAZ -0.85 -0.95 -1.06 -1.13 - 1.15 - 1.25 Birth Month 1 Month 3 Month 6 Study Visit Figure 8. Figure 8. Mean weight-for-age Z score (WAZ) was lower at birth in HIV-exposed but uninfected South African neonates than in Brazilian neonates in a 1000-infant analysis of HPTN 040. Mean height-for-age Z score (HAZ) was similar in the two groups at birth. Both scores declined in both cohorts through 1 month of age, then rebounded. South African WAZ remained substantially lower than Brazilian WAZ at 6 months (-0.53 versus -0.09). Source: Karin Nielsen-Saines, University of California, Los Angeles.52 Risk of infant infectious serious adverse event: • Maternal education of 8 years (versus more): AOR 3.47, 95% CI 1.49 to 8.09, P = 0.0040 • Maternal education < 8 years (versus more): AOR 3.65, 95% CI 1.71 to 7.80, P = 0.0009 • Maternal viral load at delivery > 1,000,000 copies/ mL: AOR 9.92, 95% CI 1.56 to 63.08, P = 0.0151 Nielsen-Saines and coworkers concluded that mortality of HIV-exposed but uninfected formula-fed infants was lower than the general-population rate in Brazil but slightly higher than the general-population rate in South Africa. South African mortality rates for this group were similar to those of HIV-exposed but uninfected infants in Zimbabwe.54 Text messaging improves infant follow-up in Johannesburg Mobile (cell) phone text messaging to HIV-positive mothers attending a Johannesburg clinic significantly improved infant HIV testing follow-up, according to preliminary results of a randomized study.55 The finding held true for women diagnosed with HIV only at delivery and whose infants thus had a higher risk of HIV infection and mortality. Rahima Moosa Mother and Child Hospital in Johannesburg, South Africa performs approximately 10,000 deliveries annually, about 25% to 30% of them in HIV-positive women. Because testing women for HIV at delivery can be difficult, the Hospital has offered routine postpartum testing since 2008. But follow-up rates are poor, especially in newly diagnosed women, and loss to follow-up at this time breaks the cascade of care, prevention, and monitoring. In an attempt to improve retention in care, KarlGünter Technau (University of the Witwatersrand, Johannesburg) and colleagues planned this study of mobile text messaging to postpartum HIV-positive women. The study called for 10 weeks of texted encouragement and reminders for infant PMTCT medication and appointments. There were three study groups: women who agreed to text messaging randomized 1:1 to receive messages (group A1, n = 160) or not to receive messages (group A2, n = 177), and a third group of women who declined texting (group B, n = 81) Proportions of women whose infants kept an HIV DNA PCR test visit were 90.5% in group A1, 78.2% in group A2, and 63.0% in group B (group A1 versus group B, P = 0.003; group A2 versus group B, P = 0.04). When the investigators considered only the 51 women diagnosed with HIV at delivery, PCR test follow-up rates were 86.7% for group A1, 41.7% for group A2, and 40.9% for group B. For the whole study group, return rates to collect PCR results were 67.3% in group A1, 61.9% in group Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 41 Report Table 6. Impact of cART on vertical HIV transmission at 6 months in Africa Trial: countries Antiretroviral regimen CD4 count Transmission rate Mma Bana: Botswana57 Abacavir, zidovudine/lamivudine or >200 cells/mm3 2.1% Lopinavir/ritonavir, zidovudine/ lamivudine >200 cells/mm3 0.4% Kesho Bora: Burkina Faso, Kenya, South Africa58 Lopinavir/ritonavir, zidovudine/ lamivudine 200 to 500 cells/ mm3 4.9% KiBS: Kenya63 Zidovudine/lamivudine plus nevirapine or nelfinavir 350 to 500 cells/ mm3 3.8% >500 cells/mm3 3.3% Source: François Dabis, Université Bordeaux Segalen, France.61 A2, and 23.5% in group B. For newly diagnosed women, return rates with 67.7% for group A1, 33.3% for group A2, and 4.5% for group B (P = 0.04 for group A1 versus group A2). At the time of the Pediatrics Workshop presentation, PCR results were available for 309 infants, 18 of whom (6%) were HIV-positive. Among 26 women with HIV diagnosed at delivery, 4 (15%) had infants with a positive PCR test. Among 349 infants with vital status data, 8 (2.3%) died before 6 weeks of age. Among 32 women diagnosed with HIV at delivery, 2 (6.3%) had infants who died before 6 weeks. Technau and colleagues concluded that cell text messaging improves follow-up for infant HIV DNA PCR testing and offers additional support for women newly diagnosed with HIV at delivery. (“Sitting alone in the room,” one woman told a staff member, “I felt like I had a friend” when the text message came.) The investigators noted that texting is an inexpensive intervention, costing about $1.20 per woman for 10 weeks of messaging. They cautioned that this is not a “blanket intervention” but requires careful individual counseling to achieve good results. The researchers also noted that women may change cell phones or lose access to them, so a central log-in facility may be needed for optimal results. 42 recommended cART (from 14 weeks’ gestation to 1 week after breastfeeding stops) as PMTCT option B for women judged not to need cART for their own health.59 The alternative PMTCT regimen proposed by WHO, the so-called option A, is antepartum maternal zidovudine, single-dose nevirapine at labor, and in certain cases zidovudine plus lamivudine during labor and delivery and postpartum, plus infant antiretroviral prophylaxis, which varies depending on feeding status. In the United States, where breastfeeding by HIV-positive mothers is not recommended, Department of Health and Human Services guidelines call for at least three antiretrovirals for pregnant women who do not require treatment for their own health, regardless of HIV RNA in plasma.60 Pediatrics Workshop organizers invited two PMTCT experts to debate the proposition that all HIV-positive pregnant women would take cART, with François Dabis (Université Bordeaux Segalen, France) making the case in favor of triple therapy61 and Jeffrey Stringer (University of Alabama at Birmingham, United States, and CIDRZ, Zambia) taking the opposite stance.62 Dabis advanced three arguments for cART as PMTCT: Should all HIV-positive pregnant women receive cART? 1. Maternal universal cART prevents almost all pediatric HIV infections. 2. Maternal universal cART saves children’s lives. 3. Maternal universal cART saves mothers’ lives. Cohort studies47,56 and two randomized trials57,58 found advantages for mothers and neonates when the mother began combination antiretroviral therapy (cART) to prevent vertical HIV transmission (reference47 is reviewed above). Since 2010 WHO has To support his first point, Dabis cited three studies, starting with the Mma Bana trial in Botswana, which found 6-month transmission rates of 0.4% with lopinavir/ritonavir plus zidovudine/lamivudine and Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 3rd International Workshop on HIV Pediatrics 2.1% with abacavir plus zidovudine/lamivudine taken by breastfeeding mothers (Table 6).57 The Kesho Bora trial in Burkina Faso, Kenya, and South Africa recorded a 4.9% transmission rate at 6 months in infants of women taking lopinavir/ritonavir plus zidovudine/lamivudine.58 The single-arm KiBS study in Kenya recorded 6-month transmission rates of 3.3% among women with a CD4 count above 500 cells/mm3 and 3.8% for those with counts between 350 and 500 cells/mm3 while taking zidovudine/ lamivudine plus nevirapine or nelfinavir.63 The infant life-saving potential of cART became clear even earlier, Dabis maintained, in a 2004 pooled analysis of infants born to HIV-positive African mothers in the absence of any maternal antiretroviral therapy.5 This 3468 mother-child pair study found that 35.2% of the 693 HIV-infected children born to these mothers died before 1 year of age and 52.7% died by age 2. Maternal death was the strongest predictor of overall infant death (combining HIVinfected and uninfected children), more than doubling the risk for a child whose mother died compared with those whose mothers survived (AOR 2.27, 95% CI 1.62 to 3.19). Maternal CD4 cell count below 200 cells/mm3 almost doubled the risk of overall infant death compared with infants of mothers with no immunodeficiency (AOR 1.91, 95% CI 1.39 to 2.62). Among the 693 HIV-infected children born to these HIV-positive mothers, maternal death also almost doubled the risk of infant death (AOR 1.84, 95% CI 1.21 to 2.82), as did maternal CD4 count below 200 cells/mm3 (AOR 1.86, 95% CI 1.27 to 2.75). In other words, maternal cART, if available, would have dramatically changed the mortality pattern of all children born to HIV-positive mothers. To make the point that cART promotes survival of HIV-positive mothers, Dabis cited results of a study showing that HIV-positive mothers in Zimbabwe had a 54 times higher risk of death than HIV-negative women in the 24 months after delivery if their CD4 count lay below 200 cells/mm3.64 In contrast, compared with the HIV-negative group, HIV-positive women with 400 to 600 cells/mm3 had a 5.4 times higher risk of death in the 24 months after delivery, much lower than the risk in women with a CD4 count below 200 cells/mm3 but still very high. Dabis also invoked results of the recent HPTN 052 trial to make the points that cART started at a CD4 count between 350 and 550 cells/mm3, rather than below 250 cells/mm3, independently lowers the risk of HIV transmission to cohabitating partners65 and also reduces the risk of tuberculosis and overall severe morbidity and mortality (combined) in the treated person.11 Dabis reported that 15 African countries have picked WHO option A for PMTCT (maternal zidovudine plus infant antiretroviral prophylaxis), while 20 prefer option B (triple therapy from 14 weeks’ gestation to 1 week after breastfeeding stops). Four countries consider either option, while Malawi now recommends cART (efavirenz plus tenofovir/emtricitabine) for life in pregnant women regardless of CD4 count—option B+. Dabis maintained that Malawi’s stance is no longer a theoretical option, because it is simple to implement, minimizes vertical HIV transmission, protects the next pregnancy, improves maternal health and survival, reduces sexual transmission of HIV, reduces the risk of tuberculosis,11 and treats hepatitis B coinfection. He noted, though, that this approach requires careful monitoring of acceptability, feasibility, outcomes, and safety. Stringer began his argument by asking if there is any reason to prefer cART for women who do not meet WHO criteria for triple therapy and have a CD4 count above 350 cells/mm3. To begin addressing this question, he cited the BAN trial in Malawi, which randomized 2369 HIV-positive, breastfeeding mothers with a CD4 count of 250 cells/mm3 or higher and their infants to (1) a maternal antiretroviral regimen, (2) infant nevirapine, or (3) no extended postnatal antiretroviral regimen (the control group).66 Median maternal CD4 counts stood at 429 cells/ mm3 in the cART arm and 440 cells/mm3 in the infant-nevirapine arm. At week 28 HIV transmission rates were significantly lower in the cART arm (2.9%, P = 0.009 versus control) and the infant-nevirapine arm (1.7%, P < 0.001 versus control) than in the control group (7.0%). But the transmission rate did not differ significantly between the two intervention arms (P = 0.10). Does cART for pregnant women offer maternal advantages over WHO option A? Starting cART at a CD4 count above 200 cells/mm3 clearly offers a survival advantage in low-income countries, Stringer noted, citing a Haitian trial that randomized 816 adults with a CD4 count between 200 and 350 cells/ mm3 to begin cART within 2 weeks of enrollment or to wait until their count fell below 200 cells/ mm3.67 Through 36 months of follow-up, early cART significantly improved survival probability by KaplanMeier analysis (P = 0.001). But starting cART sooner also poses some risks, Stringer cautioned, as found in a (yet-unpublished) study of 39,764 women starting cART in Lusaka, Zambia. Through 6 years of follow-up, rates of four “bad events” rose gradually but steadily: missing five pharmacy visits, needing single-antiretroviral substitution, changing to second-line cART, and becoming lost to follow-up. The Kaplan-Meier estimate of at least one “bad event” rose to about Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 43 Report Results: HIV prevalence among those tested 0,4 0,35 HIV prevalence 0,3 0,25 0,2 Female Male 0,15 0,1 0,05 0 12 13 14 15 16 17 18 19 20 21 22 23 24 Age (years) Figure 9. HIV prevalence in adolescents and young adults tested in a Durban outpatient clinic was higher in females starting around age 17. Although prevalence rose in both young women and young men in their early 20s, the rate was significantly higher among young women than men from 18 to 24 years of age (P < 0.01). The arrow indicates the average age of sexual debut in South Africa. Source: Lynn Ramirez-Avila, Children’s Hospital Boston.68 50% after 2 years of follow-up and to about 80% after 6 years. high in the tested group—14% in young men and 22% in young women. The antepartum testing and postpartum prescribing of WHO options A and B are similar in complexity, Stringer argued. Finally, he reminded attendees that antiretroviral access remains limited in many lowincome countries. He maintained that starting cART early in pregnant women could deprive others in greater need of therapy. South Africa has about 15 million people between 15 and 24 years old, and their HIV prevalence stands at 8.7%. The HIV rate rises steeply during the transition from adolescence to adulthood: Prevalence is 6.7% among 15-to-19-year-old girls and 32.7% among 25-to-29-year-old women. Only about one quarter of girls and young women and 15% of boys and young men report being tested for HIV. Stringer offered four conclusions: (1) Women who need cART for their own health should get it. (2) There is no randomized evidence of improved infant or maternal outcomes with universal maternal cART. (3) cART is not simpler or cheaper than well-planned short-term antiretroviral prophylaxis. (4) Starting cART too early in healthcare systems already flush with patients could prevent those in immediate need from gaining access. HIV Testing, Condom Use, Toxicity, and Resistance Low HIV testing rate in adolescents at Durban clinic In a Durban clinic with a well-established HIV testing program, only one third of adolescent boys and half of girls had HIV testing.68 About two thirds of young adults agreed to testing, and HIV prevalence was 44 Lynn Ramirez-Avila (Children’s Hospital Boston) and colleagues at Durban’s McCord Hospital and Massachusetts General Hospital assessed completion of HIV testing by adolescents and young adults who came to this outpatient clinic, which has a routine HIV testing program, and to determine HIV prevalence in those who got tested. Everyone 12 years old or older who comes to the clinic is offered routine opt-out HIV testing, which means a health worker tells them they will be tested unless they decline testing, as sanctioned by South African law. Those who agreed had concurrent fingerprick rapid tests. Testing is included in the clinic fee. The February 2008 to December 2009 study involved 956 adolescents 12 to 17 years old and 2351 young adults 18 to 24 years old. Girls and young women comprised 55% of the adolescent group and 63% of the young adult group. HIV testing rates were Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 3rd International Workshop on HIV Pediatrics significantly higher among girls than boys in the adolescent group (49% versus 33%, P < 0.01), but rates were similar in young women and men (66% and 63%). Ramirez-Avila suggested the possibility that prior HIV testing or known HIV status could explain why some youngsters declined testing. Sixty-two of 389 tested adolescents (16%) had a positive HIV testing, including 42 girls (16%) and 20 boys (16%). Among 1523 tested young adults, 288 (19%) had a positive test, including 213 young women (22%) and 75 young men (14%), a significant difference (P < 0.01). HIV prevalence in both boys and girls was high around 14 years of age, declined in the later teens, and rose again in the early 20s (Figure 9). The researchers had no data to explain the prevalence blip around age 14. Ramirez-Avila and colleagues cautioned that this outpatient clinic may not be representative of public sector clinics because there is a fee for clinic services. Also, reported prevalence involves only youngsters who agreed to testing, so overall prevalence in this age group may be lower. The investigators concluded that HIV testing rates are low but positive test rates are high in this population. They believe these findings point to (1) the need for further studies evaluating individual and clinic-level factors that result in HIV testing uptake by youth and (2) an “urgent need to offer comprehensive and youth-friendly HIV testing to adolescents and young adults in epidemic settings.” Most young people accept opt-out HIV testing in DC emergency room Three quarters of 13- to 24-year-olds accepted rapid opt-out HIV testing in a hospital emergency department in Washington, DC, the city with the highest HIV prevalence in the United States and one with a largely African-American population.69 In 2006 the CDC recommended opt-out HIV screening for all 13- to 64-year-olds seeking medical care, regardless of perceived HIV risk, though the CDC does not suggest the best way to implement this policy.70 Although numerous studies show that opt-out HIV testing is generally well accepted in hospital emergency departments in the United States, adolescents and young adults made up only small parts of these study populations. Washington, DC, the capital of the United States, has an HIV prevalence around 3%,71 about 6 times the national rate. Children’s National Medical Center in Washington launched an opt-out oral-fluid HIV screening program in March 2009 for adolescents and young adults 13 years old or older. Natella Rakhmanina and colleagues planned this study to evaluate implementation of the screening program during its first 20 months and to identify barriers to opt-out screening in a large urban pediatric emergency department. Rakhmanina reported that development of the HIV screening plan involved all relevant staff, including Screening Algorithm Patient Triaged Age ≥ 13? No Patient is given appropratie care Yes Patient and guardian (if present) are informed of the Opt-Out HIV testing No Testing Accepted? Yes Patient and Guardian are informed that the test result will be disclosed to the patient first and then shared with the guardian, unless the patient refuses. Terms of disclosure accepted? Yes No Patient given information about alternate locations of testing and information about testing Patient Tested Figure 10. An HIV screening algorithm for youngsters visiting a Washington, DC, pediatric emergency department considered input from all relevant staff sectors, patients, and parents or guardians. This approach yielded high acceptance rates in the first 20 months of implementation. Source: Natella Rakhmanina, Children’s National Medical Center, Washington, DC.69 Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 45 Report hospital administration, emergency personnel, infectious disease and adolescent health staff, and laboratory specialists. Evaluating attitudes of patients and parents or guardians was helpful in developing the screening algorithm (Figure 10). In the program’s initial stages, nursing staff offered HIV testing with a physician order. Later the hospital shifted to a triage nursing-generated order for HIV testing and added dedicated testing personnel. Through the entire study period, 25,737 adolescents and young adults visited the emergency department, of whom 4566 (17.7%) got tested for HIV. The screening rate rose more than 12-fold from May 2009, when 35 patients agreed to testing, to April 2010, when 428 accepted testing. Proportions of patients approached for HIV screening climbed from 10% in March-August 2009, to 26% in September 2009-February 2010, to 39% in March-August 2010, and to 45% in September-October 2010. In those four study periods, acceptance rates were 78%, 64%, 75%, and 70% and proportions actually tested were 73%, 56%, 70%, and 68%. Age averaged about 16.5 across the four study periods. African Americans comprised large majorities of tested youngsters in every study period, a reflection of the population the hospital serves. Rates of reactive tests in the four study periods were 0.35%, 0%, 0.19%, and 0.26%. Numbers of people who had a confirmed HIV diagnosis and were linked to care were 1, 0, 3, and 0. Primary reasons for not offering HIV testing were lack of time (23.9%), deferral by staff (25.8%), and medical reasons including unstable condition (16.9%). The investigators concluded that routine opt-out rapid oral fluid HIV screening is accepted by most youth attending a pediatric emergency department. Transition from physician- to triage nurse-generated HIV testing orders and addition of dedicated testing staff significantly increased testing rates. Rakhmanina and colleagues believe that having the emergency department staff perform the tests and developing a billing procedure will be necessary to integrate this screening program into the hospital’s standard of care. Video better than counseling in improving Bronx teens’ views on condom use Among sexually active teens and young adults who came to the emergency department at a Bronx medical center, a video series did better than standard counseling in improving scores on intention to use condoms.72 46 Adolescents and young adults account for approximately half of all new HIV infections in the United States every year. Among the 20,000 teens who become infected annually, 75% are members of racial or ethnic minorities. Teens are particularly vulnerable to HIV because (1) half of them begin sexual intercourse before age 16, when they are emotionally and biologically immature, (2) many use alcohol and drugs before having sex, and (3) about two thirds do not use condoms consistently. To promote safer sex behaviors among adolescents attending an urban emergency department, Yvette Calderon (Jacobi Medical Center, Bronx, NY) and colleagues planned this randomized trial comparing the effectiveness of a theory-based HIV educational video with conventional in-person HIV counseling. The study began with qualitative research involving 100 Bronx teens to develop the video series, which provided tailored messaging according to individuals’ initial intentions to use condoms. Those least likely to use condoms viewed a video that demonstrated the negative consequences of not using condoms. Those more likely to use condoms viewed a video that reinforced condom use as a positive behavior and showed other teens’ views on using condoms as a positive behavior. All participants watched a video that demonstrated proper use of male and female condoms and dental dams. Study participants had to be between 15 and 21 years old, sexually active, and clinically stable. All came to the emergency department on Monday through Friday between the hours of 8 AM and 4 PM. (Many sought care for reproductive health problems.) All participants completed pre- and post-intervention tests on three variables hypothesized to reduce unsafe sexual behavior: intention to use condoms, condom outcome expectancy, and condom selfefficacy (belief in one’s ability to use condoms effectively). HIV testing was optional in both study arms. The investigators randomized 102 participants (60.8% female) to the video group and 101 (55.4% female) to the standard-counseling group. Similar proportions in the video and counseling groups were Hispanic (59.8% and 57.4%), and similar proportions were black (34.3% and 42.6%). While 20.6% in the video group were under 18 years old, 25.7% in the counseling group were younger than 18. Two thirds in each group had a prior HIV test. Nearly all study participants reported vaginal sex in the past year (97.1% in the video group and 96.0% in the counseling group). Similar proportions had oral sex in the past year (72.5% video and 62.4% Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 3rd International Workshop on HIV Pediatrics Table 7. Pre- to post-test change in sexual risk variables in Bronx teens Video series mean change Counseling mean change Mean difference (video vs counseling) P Condom use intention score +0.98 –0.04 1.02 0.01 Condom self-efficacy +0.31 +0.05 0.26 0.03 Male condom outcome expectancy +0.19 +0.04 0.15 0.30 Female condom outcome expectancy +0.16 –0.04 0.20 0.06 Source: François Dabis, Université Bordeaux Segalen, France.61 counseling) and anal sex in the past year (26.5% and 21.8%). Mean numbers of sex partners were also similar in the video and counseling groups: 1.41 and 1.42 male partners, and 2.16 and 2.13 female partners. Mean change in the three outcome measures consistently (and often significantly) favored the video group (Table 7). The video’s impact on condom use intention score did not differ by gender or ethnicity. Calderon and colleagues concluded that a youthfriendly video can be a valid way to provide post-test HIV education and prevention messages in an urban emergency department serving a high proportion of minority youth. They noted that longitudinal studies are needed to determine how long such an intervention can affect risk-reduction behavior in this age group. Lipid and body fat disadvantages with PIs vs NNRTIs in African children Compared with South African children who switched to an NNRTI-based regimen, those who stayed with a PI combination had unfavorable lipid profiles and body fat changes, according to results of a 156-child analysis.73 Numerous studies report body fat abnormalities— collectively called lipodystrophy—and dyslipidemia in children taking antiretrovirals in high-income countries. But little work has addressed these abnormalities in sub-Saharan Africa. Antiretroviralassociated lipid changes early in life could have profound long-term consequences for children who will be taking antiretrovirals for decades. This analysis by Stephanie Shiau (Columbia University, New York, NY) and colleagues compared lipid and body fat changes in South African children enrolled in the NEVEREST trial, which randomized nevirapineexposed children who reached an undetectable viral load while taking lopinavir/ritonavir plus stavudine/ lamivudine to continue that regimen or switch to nevirapine with stavudine/lamivudine.17,18 The analysis involved 85 children in the lopinavir group and 71 in the nevirapine group. At the NEVEREST 2 exit visit, age averaged 5.2 years in the lopinavir group and 5.0 in the nevirapine group, and the groups were evenly divided by gender. At that point cART duration averaged 4.2 years in the PI group and 4.1 years in the nevirapine group. Weightand height-for-age Z scores and body mass index Z scores did not differ significantly between the two groups. High proportions in each group had a viral load below 50 copies/mL (84.7% in the PI group and 91.6% in the NNRTI group). The nevirapine group had a higher median CD4 count (1480 versus 1356 cells/ mm3, P = 0.049). Mean total cholesterol was significantly higher in the PI group, and a higher proportion of children taking lopinavir had a total cholesterol level above 200 mg/dL (Table 8). The PI group had lower mean high-density lipoprotein (HDL) cholesterol, higher low-density lipoprotein (LDL) cholesterol, and a higher (worse) total-to-HDL cholesterol ratio. Mean triglycerides were significantly higher in the PI group, and a higher proportion of PI-treated children had a triglyceride level above 150 mg/dL. A sum of skinfold measures was significantly higher in the PI group (43 versus 39 mm, P = 0.029), and percentage of body fat by bioimpedance analysis was significantly higher in children taking lopinavir (17% versus 14%, P = 0.042). The groups did not differ in percentage of upper arm fat, but mean percentage of upper thigh fat was higher in the PI group (22% versus 19%, P = 0.021). A ratio of midthigh skinfold Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 47 Report Table 8. Lipid changes after 4 years of continued lopinavir versus a switch to nevirapine in South African children Lopinavir/ritonavir (n = 85) Nevirapine (n = 71) P Total cholesterol (mg/dL) 171 + 39 161 + 31 0.05 Total cholesterol > 200 mg/dL (%) 16 6 0.03 HDL cholesterol (mg/dL) 51 + 14 59 + 16 0.006 LDL cholesterol (mg/dL) 100 + 34 88 + 27 0.018 Total-to-HDL ratio 3.6 + 1.1 2.9 + 0.9 < 0.001 Triglycerides (mg/dL) 94 + 39 72 + 29 < 0.001 Triglycerides > 150 mg/dL (%) 11 2 0.038 Source: Stephanie Shiau, Columbia University, New York, for the NEVEREST trial.73 sum/skinfold sum was significantly higher in children taking lopinavir (0.24 versus 0.23, P = 0.046). A ratio of trunk-to-thigh fat was significantly lower in the PI group (0.55 versus 0.57, P = 0.03). Shiau and colleagues concluded that, compared with children who switched to the nevirapine regimen, those who continued taking lopinavir/ritonavir had unfavorable cholesterol and triglyceride alterations, greater overall trunk fat, but less trunk fat in relation to thigh fat. They proposed that simple anthropometric measurements, like those used in this study, “appear to be a valuable method to measure body fat in young children on ART.” And they cautioned that unfavorable lipid alterations in children may increase long-term cardiovascular risk and “should be considered in antiretroviral treatment strategies.” High resistance rates in Spanish youngsters transferred to adult care Although Spanish youngsters infected with during childhood had good immunologic virologic control when transferred to adult care, thirds had NRTI-related mutations and almost had PI-related mutations.74 HIV and two half This study involved young people in the Madrid Cohort of HIV-1-Infected Children, an eight-hospital collaboration established in 2000. Of the 534 children enrolled, 198 (37%) are in follow-up, 88 (16%) transferred to adult units, 64 (12%) were lost to follow-up, and 184 (35%) died. Among the 88 cohort members transferred to adult units, 52% were young women, half were infected in 1986-1990, 95.5% were 48 from Europe, 87.5% were white, and age averaged 22.8 years. Miguel de Mulder (Hospital Ramón y Cajal, Madrid) and coworkers genotyped plasma samples from 44 youngsters collected closest to the transfer time. At that point, 64% of the study group had a CD4 percent between 15% and 24%, and 29% had a higher CD4 percent. Viral loads were below 50 copies/mL in 37%, 51 to 499 copies/mL in 22%, and higher in 42%. Most people in this study group, 82.9%, were taking antiretrovirals at the time of transfer. Two thirds of study participants (65%) had at least one resistance mutation, 65% had an NRTI mutation, 49% had a PI mutation, and 33% had an NNRTI mutation. While 41% had mutations conferring resistance to two or more antiretroviral classes, 18% had resistance to three classes. The most frequent NRTI mutations were D67N (50%), M41L (45%), and L210W (32.5%). Only 17.5% had the M184V mutation. The most common PI mutations were L90M (37.5%), M46I (37.5%), and V82A (17.5%). The most common NNRTI mutations were K103N (17.5%) and Y181C (7.5%). The investigators cautioned that this relatively high mutation prevalence “could compromise future longterm ART and clinical management in vertically HIV-1infected patients.” They encouraged multidisciplinary follow-up and psychosocial support for pediatric patients transferring to adult units and new strategies to increase adherence. Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 3rd International Workshop on HIV Pediatrics New data on TB, other Co-infections, HIV in CSF TB therapy raises virologic failure risk in South African children Younger age and tuberculosis cotreatment independently raised the risk of virologic failure of antiretroviral therapy in a retrospective study of 218 South African children.75 TB coinfection is a major complication of HIV infection throughout sub-Saharan Africa. Whereas global TB incidence in 2009 stood at 164 per 100,000 people, incidence measured 340 in Africa, 500 in South Africa, 877 in Cape Town, and 1600 in Khayelitsha, a district outside Cape Town. Elisabetta Walters (Stellenbosch University, Cape Town) and colleagues planned this retrospective study to explore virologic outcomes of cART-treated children also receiving treatment for TB and children on cART only. The analysis included children starting cART between January 2003 and December 2005 and being treated for TB between April 2002 and December 2008 at Cape Town’s Tygerberg Children’s Hospital. The cotreatment group included children treated for TB between April 2002 and December 2008 and receiving cART and anti-TB therapy together for at least 2 weeks. Walters and colleagues defined virologic failure as two consecutive 6-monthly viral loads above 5000 copies/mL. cART was given according to 2003-2005 South African guidelines: KM time to failure curve stratified by TB co - treatment two NRTIs plus ritonavir for infants under 6 months old, two NRTIs plus lopinavir/ritonavir for children from 6 months to 3 years old or weighing less than 10 kg, and two NRTIs plus efavirenz for children older than 3 years. Children taking lopinavir/ritonavir when cotreatment began switched to ritonavir for the duration of cotreatment. Of the 218 children studied, 79 (36%) had TB and 139 did not. Median age was 21 months in the TB group and 36 months in the no-TB group (P = 0.0006), and weight-for-age Z score was significantly lower in children with TB -3.27 versus -1.87, P < 0.0001). A significantly higher proportion in the TB group was taking a PI regimen (72% versus 42%, P = 0.0001). Median baseline CD4 percent was marginally lower in the TB group than in the no-TB group (12% versus 13.5%, P = 0.057), but median baseline viral load was similar in the two groups (5.77 versus 5.67 log10 copies/mL, P = 0.159). Independently of TB cotreatment, virologic suppression 6 and 12 months after cART began was best in the NNRTI group, followed by the lopinavir/ ritonavir group and the ritonavir group. Kaplan-Meier analysis determined that time to virologic failure was significantly faster in children receiving anti-TB therapy and in children under 1 year old (Figure 11). Statistical analysis that adjusted for gender, age, antiTB cotreatment, weight-for-age Z score, and baseline CD4 count and viral load identified younger age and KM time to failure curve stratified by age group Kaplan-Meier survival estimates 1.00 Kaplan-Meier survival estimates stratified by agegroup 0.95 0.90 1.00 No TB 0.85 0.80 3- 5yr 0.90 0.80 0.75 1- 3yr 0.70 0.70 0.65 TB 0.60 P=0.0002 0.60 <1yr 0.50 p=0.0002 Figure 11. Virologic failure of cART by Kaplan-Meier analysis was significantly faster in South African children also being treated for TB than in those without TB and in children younger than 1 year old than in older children Source: Elisabetta Walters, Stellenbosch University, Cape Town.75 Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 49 Report anti-TB cotreatment as independent predictors of virologic failure. Type of antiretroviral therapy could not be assessed as a predictor of failure because of strong colinearity between type of therapy and age (the youngest children were all taking PIs). Walters and coworkers questioned whether age may be a surrogate for other factors contributing to virologic failure in this analysis. They called for further work to identify and implement TB preventive strategies, new antiretroviral strategies for coinfected children, and alternatives to rifampin for children with TB. Most South African infants with TB do not have household TB contact Two thirds of HIV-positive or HIV-exposed infants coinfected with TB did not have an identifiable household contact for their TB, despite intensive tracing, according to results of a 45-infant study in Cape Town, Durban, and Johannesburg, South Africa.76 Avy Violari (Chris Hani Baragwanath Hospital, Soweto, South Africa) and coworkers identified only 3 adult nonhousehold contacts for these children. To determine TB contact status in children, the researchers assessed participants in the P1041 study, a phase 2/3 double-blind trial that randomized HIV-positive and exposed children in South Africa and Botswana to daily isoniazid or matching placebo for 96 weeks.77 All P1041 participants were 3 to 4 months old at recruitment and had Bacille CalmetteGuérin (BCG) vaccination at birth. The study excluded infants with a TB exposure history before enrollment or previous or current treatment for TB. Isoniazid prophylaxis did not improve TB-disease-free survival in HIV-positive children or TB-infection-free survival in HIV-exposed but negative children immunized with BCG vaccine. Violari and colleagues studied all children with definite or probable TB for microbiologically confirmed TB contacts. The analysis involved 22 HIV-positive children and 23 HIV-exposed but uninfected children, with the following numbers and identified contacts from three study sites: • Johannesburg: 32 children, 10 household contacts, 2 nonhousehold contacts • Cape Town: 10 children, 3 household contacts, 1 nonhousehold contact • Durban: 3 children, no contacts identified Overall, the investigators identified contacts for 16 of 45 children (35.5%). Among children with protocol-defined TB, no differences could be found between those with and without contacts in HIV 50 status, isoniazid versus placebo use, infant age at TB diagnosis, maternal age at infant diagnosis, household type (formal versus informal), access to water, more or fewer than 3 people in the household, or presence of an adult over 55 years old in the household. The researchers cautioned, though, that this analysis rests on a small sample size. Violari and colleagues proposed that in countries with a high TB burden, TB exposure of children can occur outside the home more frequently than in the home. They recommended that future studies of childhood TB should undertake aggressive community contact tracing to learn the origin of incident TB. Almost 25% of febrile African children starting cART have bacteremia Almost one quarter of HIV-positive African children enrolled in the ARROW trial in whom fever developed had bacteremia, usually in the first 3 months of antiretroviral therapy.78 Streptococcus pneumoniae was the most frequently isolated pathogen. ARROW is an open-label randomized trial of firstline cART and monitoring strategies at three sites in Uganda and one in Zimbabwe. The 1206 children enrolled have a median age of 6 years (IQR 2 to 9), 51% are girls, and 99.4% were vertically infected with HIV. Median follow-up at the time of this analysis was 156 weeks and maximum follow-up 210 weeks. Victor Musiime (Joint Clinical Research Centre, Kampala) and colleagues assessed ARROW enrollees in whom febrile illness developed, performing blood cultures and sensitivity analyses of isolated pathogens. Most children were receiving cotrimoxazole prophylaxis, most had received Haemophilus influenzae type B vaccination, but none had received pneumococcal vaccination. Culturing 848 samples from 461 children, the investigators found that 123 samples (14.5%) from 105 children (22.8%) were positive. Among the 105 children with positive isolates, median age was 4 years (range 0.5 to 15), and 54 (51%) were girls. Culture positivity rates were 18.8% within 3 months of starting cART, 4.2% between 3 and 11 months, and 1.6% from month 12 onward. The most prevalent pathogens were S pneumoniae (28%), Staphylococcus aureus (9%), other staph species (9%), other strep species (8%), Klebsiella pneumoniae (5%), Salmonella species (5%), and Pseudomonas aeruginosa (5%). Among S pneumoniae isolates, 8 of 22 (36%) were Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 3rd International Workshop on HIV Pediatrics Antibiotic susceptibility patterns of other bacteraemia isolates Number of susceptible isolates (percentage) Name of antibiotic S. aureus Salmonella spp E.coli P.aeruginosa K.pneumoniae Ceftriaxone 4/5(80%) 5/5(100) 2/2(100) 1/2(50) 0/2(0) Cefotaxime 3/4(75%) 2/2(100) 2/2(100) - 0/1 (0) Meropenem/ Imipenem - 2/2(100) 3/3(100) 2/2(100) 2/2(100) Ciprofloxacin 3/3(100%) 3/3(100) - 2/2(100) 1/1(100 ) Amikacin - - 2/2(100) 1/1(100) 1/1(100) Gentamicin 3/5(60) 3/4(75) 1/2(50) 2/3(67) 0 /2(0) Erythromycin 5/8(63) - - - - Chloramphenicol 1/1(100) 0/1(0) - 0/1(0) - Penicillin 1/4(25) 1/4(25) 0/2(0) 0/1 (0) 0 /3(0) Cotrimoxazole 0/8(0) 1/3(33) 0/2(0) 0/1 (0) 0/1(0) Figure 12. Antibiotic susceptibility testing of isolates other than S pneumoniae in 105 of 461 children (22.8%) with positive blood cultures in the ARROW trial showed high rates of resistance to the most frequently prescribed antibiotics. S pneumoniae isolates were also often resistant to common antibiotics (see text). Source: Victor Musiime, Joint Clinical Research Centre, Kampala.78 resistant to oxacillin, 8 of 19 (42%) were resistant to penicillin G, 21 of 22 (95%) were resistant to cotrimoxazole, and 5 of 5 were resistant to gentamicin. All S pneumoniae isolates were susceptible to ceftriaxone, vancomycin, clindamycin, cefotaxime, amoxicillin/clavulanic acid, cefuroxime, and chloramphenicol; 89% were susceptible to erythromycin, 85% to amoxicillin, and 71% to ampicillin. Other bacteremia isolates were usually susceptible to meropenem/imipenem, ciprofloxacin, amikacin, ceftriaxone, and cefotaxime, while most were resistant to chloramphenicol, penicillin, and cotrimoxazole (Figure 12). Musiime and colleagues believe high rates of resistance to commonly used antibiotics suggest that newer agents like ceftriaxone may be more effective for HIV-positive children with possible bacteremia. The high prevalence of S pneumoniae in this pediatric population suggested to the investigators “a need for effective prophylactic antibiotics and/or pneumococcal vaccination.” HBV and HCV rates 5% and 1% in HIVpositive Cambodian children A study of nearly 1000 HIV-positive children in Cambodia determined that approximately 5% have hepatitis B virus (HBV) coinfection and almost 1% have hepatitis C virus (HCV) coinfection.79 Antibody response to HBV vaccination was poor in children not receiving cART, with a low CD4 count, or with a detectable viral load. HBV prevalence is assumed to be high in Cambodia, but rates of HBV and HCV infection in the general population are not well documented. Little is known about HBV or HCV coinfection in HIV-positive children. HBV vaccination, which is essential for HBV-negative people with HIV, became part of Cambodia’s National Immunization Program in 2005. A cross-sectional study involved 974 HIV-positive children seen between April 2009 and March 2010 at the National Pediatric Hospital, which had 1180 HIV-infected children in care in December 2010. Sam Sophan (National Pediatric Hospital, Phnom Penh) and colleagues estimated that 20% of all HIV-positive children in Cambodia are in follow-up at the National Pediatric Hospital. The investigators defined HBV infection as a positive hepatitis B surface antigen (HBsAg) test and HCV infection as a positive antiHCV antibody test. A prospective study involved children eligible for HBV vaccination (negative for HBsAg and hepatitis B surface antibody), who received Engerix-B immunization at months 0, 1, and 6. Children with a severe opportunistic infection or a CD4 percent below 10% were not vaccinated. An anti-HBs antibody level above 100 mIU/mL 1 month after vaccination Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 51 Report The response rate associated with CD4+ status: % 80 70 70 CD4 ≥350 cells/mL or ≥15% (N=759) 67,5 CD4 <350 cells/mL or <15% (N=30) 60 50 40 32,5 30 30 20 10 RR: 2.3 95% CI: 1.3 -3.9 P -value: 0.0001 0 <10 mIU/mL ≥10 mIU/mL Figure 13. Cambodian children with a CD4 percent at or above 15% or a CD4 count at or above 350 cells/ mm3 had more than a doubled chance of responding to HBV vaccination compared with children with lower CD4 measures. Source: Sam Sophan, National Pediatric Hospital, Phnom Penh.79 was considered full protection, a level between 10 and 100 mIU/mL was considered a poor response requiring a single booster, and a level below 10 mIU/ mL was considered failure to respond. The study group had a median age of 8.7 years (IQR 5.7 to 11), and 478 (49%) were girls. Most children (799, 82%) were receiving cART for a median duration of 31 months (IQR 14 to 47), while 175 (18%) had never received antiretrovirals. More than half of these children (61.9%) were born in Phnom Penh. Fortysix children (4.7%) were HBsAg-positive, 122 (12.5%) were anti-HBs-positive, and 9 (0.9%) were anti-HCVpositive. Median age of HBV-coinfected children stood at 9.8 years (range 1.3 to 16.3), compared with a median age of 2.6 years (range 0.4 to 14.5) for HCVcoinfected children. Among 145 children who received HBV vaccination as part of the National Immunization Program after 2004, anti-HBs antibodies developed in only 28 (19.3%). Among 236 children whose caregivers reported that they received three vaccine doses at private clinics, anti-HBs antibodies developed in only 60 (25.4%). Among 789 HIV-positive children eligible for HBV vaccination at the National Pediatric Hospital, median age was 8.6 years (IQR 5.8 to 10.9) and 387 (49%) were girls. In that group, 492 (62.3%) attained full protection after vaccination, 40 (5.1%) had a poor response, and 257 (32.6%) had no response. Children on cART had a median anti-HBs titer of 725 mIU/mL, compared with a titer of 455 mIU/mL in children not on cART. Children receiving cART had a 30% higher chance of attaining a response above 10 mIU/mL (RR 1.3, 95% CI 1.1 to 1.5, P = 0.0001). Children with a CD4 percent at or above 15% or a CD4 count at or above 350 cells/mm3 had more than 52 a doubled chance of responding to vaccination with a titer at or above 10 mIU/mL (RR 2.3, 95% CI 1.3 to 3.9, P = 0.0001) (Figure 13). An HIV RNA load below 400 copies/mL raised the chance of an anti-HBs antibody response 60% (RR 1.6, 95% CI 1.3 to 1.8, P = 0.0001). Sophan and colleagues concluded that a high CD4 count and undetectable viral load are good predictors of the need for HBV vaccination boosting. They proposed that routine HBV vaccination for HIVpositive children without knowledge of CD4 levels or viral load may not yield a good serologic response. The investigators advised deferring HBV vaccination for children until after cART controls HIV. Low CSF HIV load in youth with PML or measles encephalitis Romanian adolescents and young adults had lower HIV RNA in cerebrospinal fluid (CSF) than in plasma if they had progressive multifocal leukoencephalopathy (PML) or subacute measles encephalitis.80 But they had equivalent or higher viral loads in CSF and plasma if they had HIV encephalopathy. Higher HIV RNA in CSF correlates with neurocognitive impairment in adults, but relative viral loads in CSF and plasma have not been closely studied in youngsters, in whom neurologic opportunistic infections are less prevalent than in adults. Many Romanian infants became infected with HIV-1 subtype F from 1987 through 1990 through unsafe injection and transfusion practices. They make up the largest part of a group of young people studied by Luminita Ene (Dr. Victor Babes Hospital, Bucharest) and colleagues to compare CSF and plasma loads in youngsters Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 3rd International Workshop on HIV Pediatrics and in 4 of 8 patients with cryptococcal meningitis, and (2) significantly lower CSF than plasma viral load in patients with PML and subacute measles encephalitis, a difference similar to that in neurologically asymptomatic patients. with and without neurologic complications of HIV. The study also aimed to assess the relevance of CSF load as a marker for central nervous system complications in adolescents and young adults. Of the 132 study participants that had available CSF and plasma samples, 104 (79%) had neurologic complications first noted at an average age of 18.7 years. Median time from HIV diagnosis to neurologic complication was 5.5 years (range 0 to 17 years). Most study participants (116 or 88%) were parenterally infected, 6 were vertically infected, and 10 acquired HIV sexually. Ene and colleagues speculated that HIV RNA may be lower in CSF than plasma in young people with PML or subacute myoclonic measles encephalitis because of possible interactions between HIV and the viruses that causes these diseases. Earlier research found evidence that measles may inhibit HIV replication in plasma.81-83 The investigators suggested that lower viral load in CSF than plasma may be an indirect marker of PML or subacute myoclonic measles encephalitis. In the whole study group, median CSF load was significantly lower than plasma load (2.61 versus 4.23 log10 copies/mL, P < 0.001), though CSF load correlated positively with plasma load (rho 0.68, P < 0.0001), as well as with albumin level (rho 0.22, P = 0.05) and pleocytosis (rho 0.51, P < 0.001). Thirtynine antiretroviral-naive study participants had a significantly higher CSF load than 34 participants on cART but with virologic failure (mean 4.28 versus 2.95 log10 copies/mL, P < 0.001). Among 20 people previously exposed to cART but not on treatment at the time of evaluation, CSF load averaged 4.01 log10 copies/mL, which was significantly lower than in the group with virologic failure but still on cART (P = 0.002). cART may limit EBV load in Ugandan children under 5 Early cART was associated with lower Epstein-Barr virus (EBV) DNA levels in Ugandan children under 5 years old, according to results of a 187-child study. Malaria appeared to promote expansion of EBVpositive B cells in these children. EBV, a herpes virus, infects B lymphocytes and epithelial cells and can cause serious malignancies such as Burkitt’s lymphoma, immunoblastic lymphoma, and nasopharyngeal carcinoma. People infected with HIV run a high risk of EBVrelated diseases, including non-Hodgkin B-cell lymphoma. EBV is endemic in Central Africa, with primary infection occurring during infancy and early childhood. In Africa EBV is strongly associated with Burkitt’s lymphoma, which accounts for 75% of malignancies in children. Thirty-two study participants had HIV encephalopathy, 22 had PML, 8 had cryptococcal meningitis, 6 had cerebral toxoplasmosis, 2 had cytomegalovirus encephalitis, and 2 had TB meningitis. Non-AIDS neurologic conditions were subacute myoclonic measles encephalitis in 13, viral encephalitis in 9, seizures in 4, HIV aseptic meningitis in 2, GuillainBarré syndrome in 1, and facial palsy in 1. Because there have been no studies of EBV viremia in HIV-positive African children, Maria Raffaella Petrara (University of Padova, Padova, Italy) and coworkers planned this analysis of 187 HIV-positive children up to 14 years old in a Kampala cohort. Of the 103 The investigators described two distinct patterns associated with specific neurologic infections (Table 9): (1) higher CSF than plasma HIV RNA in half of the 32 patients with HIV encephalopathy Table 9. HIV RNA in CSF versus plasma in Romanian youth n CSF load* Plasma load* P HIV encephalopathy 32 4.62 4.61 NS PML 21 3.13 4.68 < 0.001 Subacute myoclonic measles encephalitis 13 2.46 3.78 0.006 No neurologic disease 28 2.55 3.23 0.0001 *Mean log10 copies/mL. Source: Luminita Ene, Dr. Victor Babes Hospital, Bucharest.80 Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 53 Report children (55%) still naive to antiretrovirals, 83% had detectable EBV. Among 84 children (45%) receiving cART, 73% had detectable EBV. In the entire cohort, 22% had EBV-1, 18% EBV-2, and 38% both types of EBV. Rates of EBV-1 or EBV-2 or coinfection with both types did not differ substantially between antiretroviral-naive and experienced children. EBV levels were significantly higher in children coinfected with both EBV types than in those infected only with EBV-1 (P = 0.001) or EBV-2 (P = 0.015). But EBV load did not differ significantly between children with EBV-1 and those with EBV-2. Regardless of antiretroviral status, EBV viremia was nonsignificantly higher in children with clinical malaria (P = 0.093), a finding leading Petrara and colleagues to suggest that Plasmodium falciparum malaria may induce B-cell activation and stimulate EBV-infected B cells via toll-like receptor 9 (TLR-9) engagement. Higher EBV viremia in antiretroviral-naive versus treated children under 5 years old prompted the investigators to propose that early cART may restrict EBV replication and expansion of EBV-infected cells during primary EBV infection. EBV viremia did not differ significantly between antiretroviral-treated and naive children. But in children younger than 5 years, EBV load was significantly higher in antiretroviral-naive children (P = 0.017). 4th International Workshop on HIV Pediatrics Washington DC, USA 20 - 21 July 2012 MARK - THE - DATE 4th International Workshop on HIV Pediatrics 20 - 21 July 2012, Washington, DC, USA Abstracts & Presentations of the 3rd International Workshop on HIV Pediatrics are available online at www.virology-education.com 54 Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 3rd International Workshop on HIV Pediatrics Reference 1. Hazra R. Growing up with HIV in resource-rich countries. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Invited lecture. 2. Musoke P. The challenge of growing up HIV infected in resource-limited settings. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Invited lecture. 3. Brady MT, Oleske JM, Williams PL, et al; Pediatric AIDS Clinical Trials Group219/219C Team. Declines in mortality rates and changes in causes of death in HIV1-infected children during the HAART era. J Acquir Immune Defic Syndr. 2010;53:86-94. 4. Kapogiannis B, Soe M, Nesheim S, et al. Mortality trends in the U.S. Perinatal AIDS Collaborative Transmission Study (1986-2004). Clin Infect Dis. 2011; in press. 5. Newell ML, Coovadia H, Cortina-Borja M, Rollins N, Gaillard P, Dabis F; Ghent International AIDS Society (IAS) Working Group on HIV Infection in Women and Children. Mortality of infected and uninfected infants born to HIV-infected mothers in Africa: a pooled analysis. Lancet. 2004;364:1236-1243. Available at http://www.thelancet.com/journals/lancet/article/ PIIS0140-6736%2804%2917140-7/fulltext. Accessed 18 August 2011. 6. UNAIDS. UNAIDS report on the global AIDS epidemic 2010. Geneva: Joint United Nations Programme on HIV/AIDS. 2010. Available at http://www.unaids. org/en/media /unaids/contentassets/documents/ unaidspublication/2010/20101123_globalreport_ en.pdf. Accessed 24 August 2011. 7. Sauvageot D, Schaefer M, Olson D, Pujades-Rodriguez M, O’Brien DP. Antiretroviral therapy outcomes in resource-limited settings for HIV-infected children <5 years of age. Pediatrics. 2010;125:e1039-e1047. 8. Puthanakit T, Vonthanak S, Ananworanich J, et al. Randomized clinical trial of immediate versus deferred antiretroviral therapy initiation in children older than one year with moderate immunodeficiency: the PREDICT study (NCT00234091). 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_01. 9. Violari A, Cotton MF, Gibb DM, Babiker AG, Steyn J, Madhi SA, Jean-Philippe P, McIntyre JA; CHER Study Team. Early antiretroviral therapy and mortality among HIV-infected infants. N Engl J Med. 2008;359:22332244. Available at http://www.nejm.org/doi/ full/10.1056/NEJMoa0800971. Accessed 24 August 2011. 10. World Health Organization. Antiretroviral therapy for HIV infection in infants and children: towards universal access. 2010 revision. Available at http://www.who. int/hiv/pub/paediatric/infants2010/en/. Accessed 8 August 2011. 11. Grinsztejn B, Ribaudo H, Cohen MS, et al. Effects of early versus delayed initiation of antiretroviral therapy (ART) on HIV clinical outcomes: results from the HPTN 052 randomized clinical trial. 6th IAS Conference on HIV Pathogenesis, Treatment and Prevention. 17-20 July 2011. Rome, Italy. Abstract MOAX0105. 12.Panel on Antiretroviral Therapy and Medical Management of HIV-Infected Children. Guidelines for the use of antiretroviral agents in pediatric HIV infection. August 11, 2011; pp 1-268. Available at http:// aidsinfo.nih.gov/ContentFiles/PediatricGuidelines.pdf. Accessed 23 August 2011. 13. Palumbo P. Pediatric HIV treatment option—resource limited settings. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Invited lecture. 14. Palumbo P, Lindsey JC, Hughes MD, et al. Antiretroviral treatment for children with peripartum nevirapine exposure. N Engl J Med. 2010;363:1510-1520. Available at http://www.nejm.org/doi/full/10.1056/ NEJMoa1000931. Accessed 23 August 2011. 15. Tudor-Williams G. What have we learned from the PENPACT 1 trial? 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Invited lecture. 16.PENPACT-1 (PENTA 9/PACTG 390) Study Team, Babiker A, Castro nee Green H, Compagnucci A, et al. First-line antiretroviral therapy with a protease inhibitor versus non-nucleoside reverse transcriptase inhibitor and switch at higher versus low viral load in HIVinfected children: an open-label, randomised phase 2/3 trial. Lancet Infect Dis. 2011;11:273-283. Available at http://www.thelancet.com/journals/laninf/article/ PIIS1473-3099%2810%2970313-3/fulltext. Accessed 8 August 2011. 17. Coovadia A, Abrams EJ, Stehlau R, et al. Reuse of nevirapine in exposed HIV-infected children after protease inhibitor-based viral suppression: a randomized controlled trial. JAMA. 2010;304:10821090. Available at http://jama.ama-assn.org/ content/304/10/1082.long. Accessed 8 August 2011. 18 Coovadia A. Should PI-based therapy be continued indefinitely? 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Invited lecture. 19. Penazzato M, on behalf of the European Pregnancy and Paediatric HIV Cohort Collaboration—EPPICC. Treatment efficacy in European children starting cART in infancy: factors associated with viro-immunological response and 1st-line therapy duration. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_08. 20.Anti-retroviral research for watoto. ARROW trial summary. Available at http://www.arrowtrial.org/trial_ summary.asp. Accessed 9 August 2011. 21. Mutasa K, Prendergast A, Ntozini R, et al. Predictive value of six-week viral load on mortality in HIV-infected Zimbabwean infants. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_07. 22. Malaba LC, Iliff PJ, Nathoo KJ, et al; ZVITAMBO Study Group. Effect of postpartum maternal or neonatal vitamin A supplementation on infant mortality among infants born to HIV-negative mothers in Zimbabwe. Am J Clin Nutr. 2005;81:454-560. Available at http:// www.ajcn.org/content/81/2/454.long. Accessed 8 August 2011. 23. Abrams EJ, Weedon J, Steketee RW, et al. Association of human immunodeficiency virus (HIV) load early in life with disease progression among HIV-infected infants. New York City Perinatal HIV Transmission Collaborative Study Group. J Infect Dis. 1998;178:101108. 24. Shearer WT, Quinn TC, LaRussa P, et al. Viral load and disease progression in infants infected with human immunodeficiency virus type 1. Women and Infants Transmission Study Group. N Engl J Med. 1997;336:1337-1342. Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 55 Report 25.Dickover RE, Dillon M, Leung KM, et al. Early prognostic indicators in primary perinatal human immunodeficiency virus type 1 infection: importance of viral RNA and the timing of transmission on longterm outcome. J Infect Dis. 1998;178:375-387. 26. Mofenson LM, Harris DR, Rich K, et al. Serum HIV1 p24 antibody, HIV-1 RNA copy number and CD4 lymphocyte percentage are independently associated with risk of mortality in HIV-1-infected children. National Institute of Child Health and Human Development Intravenous Immunoglobulin Clinical Trial Study Group. AIDS. 1999;13:31-39. 27. Compagnucci A, Penta SC. Long-term consequences of planned treatment interruptions in HIV-infected children: results from the PENTA 11/TICCH trial. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_10. 28. Paediatric European Network for Treatment of AIDS. Response to planned treatment interruptions in HIV infection varies across childhood. AIDS. 2010;24:231241. 29. Davies M, Bolton C, Eley B, et al. Predicting 1-year mortality using current CD4 percent and count in order to guide switching therapy in children on ART in southern Africa. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_09. 30. Kuhn L. Virologic monitoring should be routine for children in resource-limited settings. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Invited lecture. 31. Gibb D. Viral load monitoring should be routine for children in resource-limited countries: the case against. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Invited lecture. 32. Schneider K, Puthanakit T, Kerr S, et al. Economic evaluation of monitoring virologic responses to antiretroviral therapy in HIV-infected children in resource-limited settings. AIDS. 2011;25:1143-1151. 33. Oliveira R, Krauss M, Essama-Bibi S, et al; NISDI Pediatric Study Group 2010. Viral load predicts new World Health Organization stage 3 and 4 events in HIVinfected children receiving highly active antiretroviral therapy, independent of CD4 T lymphocyte value. Clin Infect Dis. 2010;51:1325-1333. 34. Sutcliffe CG, Moss WJ. ART for children: what to start and when to switch. Lancet Infect Dis. 2011;11:254255. 35.Dunn D; HIV Paediatric Prognostic Markers Collaborative Study Group. Short-term risk of disease progression in HIV-1-infected children receiving no antiretroviral therapy or zidovudine monotherapy: a meta-analysis. Lancet. 2003;362:1605-1611. 36. DART Trial Team, Mugyenyi P, Walker AS, Hakim J, et al. Routine versus clinically driven laboratory monitoring of HIV antiretroviral therapy in Africa (DART): a randomised non-inferiority trial. Lancet. 2010;375:123-131. 37. Jourdain G, Ngo-Giang-Huong N, Le Coeur S, et al. PHPT-3: A randomized clinical trial comparing CD4 vs viral load ART monitoring/switching strategies in Thailand. 18th Conference on Retroviruses and Opportunistic Infections. 27 February-2 March 2011. Boston. Abstract 44. 56 38. Gilks C, Walker S, Munderi P, et al. A single CD4 test with threshold >250 cells/mm3 can markedly reduce switching to second-line ART in African patients managed without CD4 or viral monitoring. 18th Conference on Retroviruses and Opportunistic Infections. 27 February-2 March 2011. Abstract 676. Available at http://www.retroconference.org/2011/ PDFs/676.pdf. Accessed 10 August 2011. 39. Phillips AN, Pillay D, Miners AH, et al. Outcomes from monitoring of patients on antiretroviral therapy in resource-limited settings with viral load, CD4 cell count, or clinical observation alone: a computer simulation model. Lancet. 2008;371:1443-1451. 40. Bendavid E, Young SD, Katzenstein DA, Bayoumi AM, Sanders GD, Owens DK. Cost-effectiveness of HIV monitoring strategies in resource-limited settings: a southern African analysis. Arch Intern Med. 2008;168:1910-1918. 41. Bishai D, Colchero A, Durack DT. The cost effectiveness of antiretroviral treatment strategies in resource-limited settings. AIDS. 2007;21:1333-1340. 42. Kimmel AD, Weinstein MC, Anglaret X, et al; CEPACInternational Investigators. Laboratory monitoring to guide switching antiretroviral therapy in resourcelimited settings: clinical benefits and cost-effectiveness. J Acquir Immune Defic Syndr. 2010;54:258-268. 43. Walensky RP, Ciaranello AL, Park JE, Freedberg KA. Cost-effectiveness of laboratory monitoring in subSaharan Africa: a review of the current literature. Clin Infect Dis. 2010;51:85-92. 44.Walker AS, Gibb DM. Monitoring of highly active antiretroviral therapy in HIV infection. Curr Opin Infect Dis. 2011;24:27-33. 45. Dicko F, Malateste K, Koueta F, et al. Patterns of firstline antiretroviral regimen and switch to second-line in West African children on ART. The IeDEA paediatric West African Database. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract PP_06. 46. Bunupuradah T, Puthanakit T, Fahey P, et al. Secondline highly active antiretroviral therapy in Asian HIVinfected children. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract PP_05. 47. Dinh TH, Goga A, Jackson D, et al. Impact of the national prevention of mother-to-child transmission (PMTCT) program on mother-to-child transmission of HIV (MTCT), South Africa, 2010. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_13. 48. Lu L, Motswere-Chirwa C, Legwaila K, et al. HIV incidence in women during the first postpartum year: implications for PMTCT programs Francistown, Botswana, 2010. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_15. 49. Humphrey J, Marinda E, Moulton L, et al: Breastfeedingassociated HIV transmission among women who seroconvert during late pregnancy and during breastfeeding. XVI International AIDS conference. 14 August 2006. Toronto, Canada. Abstract MOPE0384. 50. Amzal B, Jourdain G, Cressey T, et al. Maternal and infant nevirapine for the prevention of mother-tochild transmission of HIV in women with <8 weeks of prenatal HAART: a Bayesian analysis. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_16. Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 3rd International Workshop on HIV Pediatrics 51. Lallemant M, Jourdain G, Le Coeur S, et al; Perinatal HIV Prevention Trial (Thailand) Investigators. Singledose perinatal nevirapine plus standard zidovudine to prevent mother-to-child transmission of HIV1 in Thailand. N Engl J Med. 2004;351:217-228. Available at http://www.nejm.org/doi/full/10.1056/ NEJMoa033500. Accessed 14 August 2011. 52. Nielsen-Saines K, Watts DH, Joao EC, et al. Infectious morbidity, mortality growth of HIV-exposed, uninfected, formula-fed infants enrolled in NICHD/ HPTN 040/ PACTG 1043. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_11. 53.Nielsen-Saines K, Watts H, Gonçalves Veloso V, et al. Phase III randomized trial of the safety and efficacy of 3 neonatal ARV regimens for prevention of intrapartum HIV-1 transmission: NICHD HPTN 040/ PACTG 1043. 18th Conference on Retroviruses and Opportunistic Infections. 27 February-2 March 2011. Boston. Abstract 124LB. Available at http://www.hptn. org/web%20documents/AnnualMeeting2011/Presen tationJoint/10HPTN040NielsonOK.pdf. Accessed 15 August 2011. 54. Marinda E, Humphrey JH, Iliff PJ, et al. Child mortality according to maternal and infant HIV status in Zimbabwe. Pediatr Infect Dis J. 2007;26:519-526. 55. Technau K, de Tolly K, Sherman G, et al. Mobile text messaging improves PMTCT follow-up in South African public setting. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_12. 56. Liotta G, Mancinelli S, Gennaro E, et al. Is highly active antiretroviral therapy (HAART) in pregnancy protective against maternal mortality? Results from a large DREAM cohort in Malawi and Mozambique. 6th IAS Conference on HIV Pathogenesis, Treatment and Prevention. Rome, Italy. 17-20 July 2011. Abstract TUAB0201. 57. Shapiro RL, Hughes MD, Ogwu A, et al. Antiretroviral regimens in pregnancy and breast-feeding in Botswana. N Engl J Med. 2010;362:2282-2294. Available at http://www.nejm.org/doi/full/10.1056/ NEJMoa0907736. Accessed 18 August 2011. 58. Kesho Bora Study Group, de Vincenzi I. Triple antiretroviral compared with zidovudine and singledose nevirapine prophylaxis during pregnancy and breastfeeding for prevention of mother-to-child transmission of HIV-1 (Kesho Bora study): a randomised controlled trial. Lancet Infect Dis. 2011;11:171-180. Erratum in Lancet Infect Dis. 2011;11:159. 59. World Health Organization. Antiretroviral drugs for treating pregnant women and preventing HIV infection in infants. 2010. Available at http://whqlibdoc.who. int /publications/2010/9789241599818_eng.pdf. Accessed 18 August 2011. 60.Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission. Recommendations for use of antiretroviral drugs in pregnant HIV-1-infected women for maternal health and interventions to reduce perinatal HIV transmission in the United States. May 24, 2010. Available at http://aidsinfo.nih.gov/ContentFiles/PerinatalGL.pdf. Accessed 18 August 2011. 61. Dabis F. HAART should be used for PMTCT in all pregnant women: the pro side. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Invited lecture. 62. Stringer J. PMTCT in 2011: maximizing long-term outcomes through a measured approach. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Invited lecture. 63. Thomas TK, Masaba R, Borkowf CB, et al; for the KiBS study team. Triple-antiretroviral prophylaxis to prevent mother-to-child HIV transmission through breastfeeding—the Kisumu Breastfeeding Study, Kenya: a clinical trial. PLoS Med. 2011;8(3):e1001015. Available at http://www.plosmedicine.org/article/ info%3Adoi%2F10.1371%2Fjournal.pmed.1001015. Accessed 18 August 2011. 64.Hargrove JW, Humphrey JH; ZVITAMBO Study Group. Mortality among HIV-positive postpartum women with high CD4 cell counts in Zimbabwe. AIDS. 2010;24:F11-F14. 65. Cohen MS, Chen YQ, McCauley M, et al; HPTN 052 Study Team. Prevention of HIV-1 infection with early antiretroviral therapy. N Engl J Med. 2011;365:493505. Available at http://www.nejm.org/doi/full/10.1056/ NEJMoa1105243. Accessed 18 August 2011. 66.Chasela CS, Hudgens MG, Jamieson DJ, et al. Maternal or infant antiretroviral drugs to reduce HIV1 transmission. N Engl J Med 2010;362:2271-2281. Available at http://www.nejm.org/doi/full/10.1056/ NEJMoa0911486. Accessed 18 August 2011. 67. Severe P, Juste MA, Ambroise A, et al. Early versus standard antiretroviral therapy for HIV-infected adults in Haiti. N Engl J Med. 2010;363:257-265. Available at http://www.nejm.org/doi/full/10.1056/ NEJMoa0910370. Accessed 18 August 2011. 68. Ramirez-Avila L, Nixon K, Noubary F, et al. Routine HIV testing in adolescents and young adults presenting to an outpatient site in Durban, South Africa. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_05. 69. Rakhmanina N, Johnson B, Souweine K, et al. Screening of adolescents and young adults for HIV in a large urban pediatric emergency department: how to do it right? 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_06. 70. Branson BM, Handsfield HH, Lampe MA, et al. Revised recommendations for HIV testing of adults, adolescents, and pregnant women in health-care settings. MMWR Recomm Rep. 2006;55:1-17. http:// www.cdc.gov/mmwr/preview/mmwrhtml/rr5514a1. htm. Accessed 19 August 2011. 71. Vargas JA, Fears D. At least 3 percent of D.C. residents have HIV or AIDS, city study finds; rate up 22% from 2006. Washington Post. 15 March 2009. Available at http://www.washingtonpost.com/wp-dyn/ content/article/2009/03/14/AR2009031402176.html. Accessed 19 August 2011. 72. Calderon Y, Leu CS, Cowan EA, et al. Project Control. Evaluation of a brief HIV counseling video to improve teenagers risk reduction behavior. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_03. 73. Shiau S, Arpadi S, Strehlau R, et al. Body composition and metabolic abnormalities of perinatally HIV-infected children in South Africa on long-term ARV treatment. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_02. Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement 57 Report 74. Navas A, de Mulder M, Gonzalez-Granados I, et al. High drug resistance prevalence among vertically HIV-infected patients transferred from paediatric care to adult units in Spain. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_04. 75.Walters E, Reichmuth K, Rabie H, Cotton MF, Dramowski A, Marais BJ. Virological outcomes in South African children co-treated with highly-active anti-retroviral therapy (HAART) and anti-tuberculosis therapy. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_20. 76. Nachman S, Zeldow B, Dittmer S, et al. Lack of identification of adult TB contacts in infants with microbiologically confirmed or clinically presumed TB (MCCP TB) in clinical trial P1041. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_21. 77. Madhi SA, Nachman S, Violari A, et al; P1041 Study Team. Primary isoniazid prophylaxis against tuberculosis in HIV-exposed children. N Engl J Med. 2011;365:21-31. Available at http://www.nejm.org/doi/ full/10.1056/NEJMoa1011214. Accessed 21 August 2011. 78. Musiime V, Cook A, Bakeera-Kitaka S, et al. Bacteraemia in HIV-1 infected children on antiretroviral therapy in Uganda and Zimbabwe in the ARROW clinical trial. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_19. 79. Sophan S, Ung V, Huot C, et al. Prevalence of HBV/ HIV and HCV/HIV coinfections and HB vaccination in HIV-infected children at the National Pediatric Hospital, Phnom Penh, Cambodia. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_17. 80. Duiculescu D, Ene L, Tardei G, Achim CL. Divergent trends in HIV RNA levels in the cerebrospinal fluid of children and adolescents with central nervous system complications. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_22. 81. Moss WJ, Ryon JJ, Monze M, Cutts F, Quinn TC, Griffin DE. Suppression of human immunodeficiency virus replication during acute measles. J Infect Dis. 2002;185:1035-1042. 82. Ruel TD, Achan, J, Gasasira AF, et al. Dramatic reductions in HIV RNA among HIV-infected children with acute measles in Uganda. 14th Conference on Retroviruses and Opportunistic Infections. 25-28 February 2007. Los Angeles. Abstract 707. 83. Duiculescu D, Ene L, Ungureanu E, et al. Subacute measles encephalitis: a new AIDS defining disease in HIV-infected young patients. 4th IAS Conference on HIV Pathogenesis, Treatment and Prevention. 22-25 July 2007. Sydney. Poster WEPDB03. 84. Petrara MR, Penazzato M, Massavon W, et al. Dynamics of Epstein-Barr virus in HIV-1-infected children in Uganda. 3rd International Workshop on HIV Pediatrics. 15-16 July 2011. Rome, Italy. Abstract O_18. 58 Reviews in Antiviral Therapy & Infectious Diseases - volume 3; 2011 - Supplement VACCINATIE Masterclass 2012 DOELGROEP: clinici, waaronder infectiologen, microbiologen, kinderartsen, GGD-artsen, apothekers en arts-assistenten in opleiding voor voorgenoemde specialismen. FORMAT: de masterclass bestaat uit zeven modules die maandelijks op dinsdagavond (18.30 - 21.00 u.) in Utrecht zullen worden georganiseerd. ONDERWERPEN: Vaccinatie en het immuunsysteem, vaccins tegen virussen, bacteriën en protozoa, farmaco-economie, bijwerkingen en acceptatie van vaccins, de ontwikkeling van vaccins inclusief klinische studies, overwegingen om vaccins al dan niet in het Rijksvaccinatieprogramma op te nemen. PROGRAMMA 10 januari 2012 Module I Het begin van vaccinatie, soort vaccins Vaccinatie en het immuunsysteem 7 februari 2012 Module II Adjuvantia en toedieningsvormen Vaccins tegen griep 13 maart 2012 Module III De ontwikkeling van het Rijksvaccinatieprogramma, herd immunity Vaccinatie van kinderen tegen rotavirusinfecties 3 april 2012 Module IV Vaccinatie tegen malaria Ziektelast en kosteneffectiviteit 8 mei 2012 Module V Het meten van werkzaamheid van vaccins, correlaten bescherming Klinische studies: het Capita onderzoek Bijwerkingen en acceptatie van vaccins 5 juni 2012 Module VI Van idee tot product, klinische studies, productie, registratie Vaccinatie tegen S. aureus infecties 19 juni 2012 Module VII Vaccins voor ouderen, immunosenescence Interactieve sessies over de uitbreiding van het vaccinatieprogramma: van evidence based medicine naar maatschappelijk draagvlak ATRIPLA Effectiviteit niet overtroffen in studies 1-12 – de voorkeur van patiënten 13-16* Vertrouwen in de toekomst voor u en uw patiënten. Prof. Dr. Ben van der Zeijst (LUMC) Prof. Dr. Georg Kraal (VU) Dr. Gideon Kersten (RIVM) Dr. Bettie Voordouw (CBG) Prof. Dr. Joost Ruitenberg (Gezondheidsraad) Drs. Patricia Bruijning-Verhagen (UMCU) Prof. Dr. Robert Sauerwein (UMC Radboud) Prof. Dr. Maarten Postma (RUG) Dr. Loek van Alphen (RIVM) Prof. Dr. Marc Bonten (UMCU) Dr. Agnes Kant (Lareb) Prof. Dr. Han van den Bosch (VU) Prof. Dr. Jos van Strijp (UMCU) Prof. Dr. Catharina Mathei (UC Louvain, Belgie) Prof. Dr. Willem van Eden (UMCU) Prof. Dr. Ben van der Zeijst (LUMC) ORGANISEREND COMITÉ Prof. Dr. Marc Bonten Prof. Dr. Han van den Bosch Prof. Dr. Robert Sauerwein Prof. Dr. Ben van der Zeijst Universitair Medisch Centrum Utrecht Vrije Universiteit Amsterdam St. Radboud Universitair Medisch Centrum Nijmegen Leids Universitair Medisch Centrum, Leiden DE VACCINATIE MASTERCLASS WORDT ONDERSTEUND DOOR ACCREDITATIE IS AANGEVRAAGD BIJ DE NIV, NvMM, NvK en NVZA *ATRIPLA is geregistreerd voor volwassen HIV-1-patiënten met virologische suppressie. efavirenz 600mg/emtricitabine 200mg/ tenofovir disoproxil (as fumarate) 245mg Tablets HAART in 1 ©2009 Bristol-Myers Squibb & Gilead Sciences Verkorte productinformatie beschikbaar elders in dit blad. Ad Atripla.indd 1 101223/092 19-01-11 12:12 VOOR MEER INFORMATIE & REGISTRATIE, KIJK OP WWW.VIROLOGY-EDUCATION.COM De masterclass wordt georganiseerd door Virology Education De masterclass wordt financieel mogelijk gemaakt door Pfizer bv IN VIR E AL TH R PY A TI A N & IN FE CT IO U S DIS S SE IN REVIEWS EA Reviews & 3 2011 Pneumonie.nl in Antiviral Therapy INFECTIOUS DISEASES JOURNAL OF ABSTRACTS AND CONFERENCE REPORTS FROM INTERNATIONAL WORKSHOPS ON INFECTIOUS DISEASES & ANTIVIRAL THERAPY Meeting report 6th International Workshop on HIV Transmission Principles of Intervention 14 - 15 July, 2011, Rome, Italy 3rd International Workshop on HIV Pediatrics Dé pneumonieportal voor en door medische professionals 15 - 16 July, 2011, Rome, Italy Het laatste nieuws Scorecards Beeldenbank Video’s Factsheet www.pneumonie.nl pneumonie.nl wordt mogelijk gemaakt door SUPPLEMENT Presentatiemateriaal