in hi bid ores pro teas a

Upload: angel-martinez

Post on 08-Apr-2018

221 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/6/2019 In Hi Bid Ores Pro Teas A

    1/12

    Vo lu me 3 38 N um be r 18 1281

    Review Article

    Drug Therapy

    A

    L ASTAIR

    J.J. W

    OO D

    , M.D.,

    Editor

    DRUG THERAPY

    HIV-P

    ROTEASE

    I

    NHIBITORS

    C

    HARLES

    F

    LEXNER

    , M.D.

    From the Division of Clinical Pharmacology, Department of Medicineand Department of Pharmacology and Molecular Sciences, Johns HopkinsUniversity School of Medicine, Baltimore. Address reprint requests to Dr.Flexner at Osler 524, Johns Hopkins Hospital, 600 N. Wolfe St., Balti-more, MD 21287-5554.

    1998, Massachusetts Medical Society.

    NHIBITORS of human immunodeficiency virus(HIV)encoded protease, combined with nucle-oside analogues with antiretroviral activity, cause

    profound and sustained suppression of viral replica-tion, reduce morbidity, and prolong life in patients

    with HIV infection.

    1-3

    Recent guidelines recommendthat initial treatment of all HIV-infected patientsinclude the administration of an HIV-protease inhib-itor.

    4

    THE HIV-ENCODED PROTEASE

    The HIV protease, encoded in the 5

    end of the

    pol

    gene, is expressed as part of the gagpol polypro-tein (Fig. 1). This gene encodes a 99-amino-acidprotein. Homodimers of this protein have the aspar-tyl protease activity that is typical of retroviral pro-teases; monomers are enzymatically inactive.

    6

    The

    enzymes targets are amino acid sequences in the gagand gagpol polyproteins, which must be cleaved be-fore nascent viral particles (virions) can mature.

    7-10

    Cleavage of the gag polyprotein produces three largeproteins (p24, p17, and p7) that contribute to thestructure of the virion and to RNA packaging, andthree smaller proteins (p6, p2, and p1) of uncertainfunction.

    11

    Although mammalian cells contain aspar-tyl proteases, none efficiently cleave the gag polypro-tein.

    12,13

    Three of the HIV-cleavage sites are phenyl-alanineproline or tyrosineproline bonds, which areunusual sites of attack for mammalian proteases.

    14

    Proteolytic cleavage of the gag polyprotein resultsin morphologic changes in the virion and condensa-

    tion of the nucleoprotein core. The protease is pack-aged into virions, and the cleavage events it catalyzes

    I

    occur simultaneously with or soon after the budding

    of the virion from the surface of an infected cell.

    15

    Proviral DNA lacking functional protease producesimmature, noninfectious viral particles.

    9

    MECHANISM OF ACTION OFHIV-PROTEASE INHIBITORS

    The four approved HIV-protease inhibitors arebased on amino acid sequences recognized andcleaved in HIV proteins. Indinavir (Crixivan), nelfin-avir (Viracept), ritonavir (Norvir), and saquinavir(Invirase and Fortovase) and the investigational pro-tease inhibitor amprenavir are structurally relatedmolecules (Fig. 2). Most contain a synthetic ana-logue of the phenylalanineproline sequence at po-

    sitions 167 and 168 of the gagpol polyprotein thatis cleaved by the protease.

    14

    These drugs are difficultto synthesize in large quantities because of theircomplex structure.

    HIV-protease inhibitors prevent cleavage of gagand gagpol protein precursors in acutely and chron-ically infected cells, arresting maturation and therebyblocking the infectivity of nascent virions.

    13,16

    Themain antiviral action of HIV-protease inhibitors isthus to prevent subsequent waves of infection; theyhave no effect on cells already harboring integratedproviral DNA. These agents are active against clini-cal isolates of HIV types 1 and 2, with the in vitroconcentration of drug required to reduce viral pro-duction by 50 percent (IC

    50

    ) ranging from 2 to 60nM.

    16-19

    Antiviral activity is correlated with the inhi-bition of enzyme activity, although the drug con-centration required to reduce enzyme activity by 50percent (Ki) is lower than the IC

    50

    , ranging from0.10 to 2.0 nM.

    16-20

    These drugs are inactive orweakly active against human aspartyl proteases, witha Ki of at least 10,000 nM for renin and pepsin.

    16,17

    CLINICAL PHARMACOLOGIC PROPERTIES

    The clinical pharmacokinetic properties of thefour approved protease inhibitors are shown in Table1. Oral bioavailability varies because of differences infirst-pass hepatic metabolism, to which saquinavir isthe most susceptible.

    27

    Food also affects bioavailabil-ity. A high-fat meal increases the bioavailability ofsaquinavir and nelfinavir but reduces that of indina-

    vir. The same high-fat meal increases the bioavail-ability of ritonavir capsules but decreases that of ri-tonavir liquid. The ingestion of indinavir capsules

    with light meals has no effect on the area under theplasma-concentrationtime curve during an averagedosing interval, but variability is large.

    21

    Nelfinavir,ritonavir, and saquinavir should be taken with meals,

    Downloaded from www.nejm.org on September 9, 2006 . Copyright 1998 Massachusetts Medical Society. All rights reserved.

  • 8/6/2019 In Hi Bid Ores Pro Teas A

    2/12

    1282

    Apri l 30, 1998

    The New England Journal of Medicine

    Figure 1.

    Structure and Function of HIV-1 Protease.

    Panel A shows a ribbon diagram of the protease with an inhibitor molecule in the active site (reproduced from Erickson

    5

    with thepermission of the publisher). Panel B shows a horizontal 180-degree rotation of the protease (kindly provided by John W. Erickson,National Cancer Institute, Frederick, Md.). Panel C shows the translational products of the HIV gagpol

    gene and the sites at which

    the gene product is cleaved by the virus-encoded protease. p17 denotes capsid protein, p24 matrix protein, and p7 nucleocapsid;p2, p1, and p6 are small proteins with unknown functions. The arrows denote cleavage events catalyzed by the HIV-specific pro-tease.

    B

    A

    Downloaded from www.nejm.org on September 9, 2006 . Copyright 1998 Massachusetts Medical Society. All rights reserved.

  • 8/6/2019 In Hi Bid Ores Pro Teas A

    3/12

    DRUG THERAPY

    Vo lu me 3 38 N um be r 18

    1283

    and indinavir should be taken after fasting or with alight, low-fat snack.

    All these drugs are metabolized by cytochromeP-450 enzymes, mainly the 3A4 isoform.

    26,28,29

    Theabsorption of the drugs is maximal within fourhours after ingestion. Their elimination half-livesrange from 1.8 to 5 hours. Differences in a drugspharmacokinetic characteristics between patients arelarge, as indicated by a coefficient of variation of 30percent or higher for the mean area under the curve.

    All these drugs except indinavir are at least 98 per-cent protein-bound in plasma. The apparent volumeof distribution ranges from 0.4 to 10.0 liters per kil-ogram of body weight.

    Alpha

    1

    -acid glycoprotein, an inducible plasma pro-tein, reduces the in vitro anti-HIV potency of someprotease inhibitors by a factor of 10 or more.

    30-33

    This effect may be mediated by high-affinity bindingof the drug to the glycoprotein. Since only free drugis available to penetrate cells, plasma concentrationsneed to be high enough to override this effect.

    34

    There are limited data on central nervous systempenetration of these drugs. The cerebrospinal fluidpenetration of ritonavir and saquinavir, defined asthe ratio of the cerebrospinal fluid concentration tothe simultaneous plasma concentration, is less thanor equal to 1 percent.

    23,35

    Indinavir has a higher cer-ebrospinal fluid penetration, ranging from 2.2 to 76percent, perhaps because of decreased plasma pro-tein binding.

    25

    Indinavir, ritonavir, and saquinavir are available ascapsules, and nelfinavir is available as tablets. Sa-quinavir is now also available as soft-gel capsules(Fortovase), which triples oral bioavailability.

    36

    Nel-finavir powder and liquid ritonavir are pediatric for-mulations. Indinavir must be stored in an airtightcontainer with a desiccant.

    21

    Ritonavir is heat-sensi-

    tive and needs to be refrigerated. It also contains al-cohol and is therefore contraindicated in patientstaking disulfiram or metronidazole.

    23

    DRUG INTERACTIONS

    HIV-protease inhibitors can interact with inhibi-tors or inducers of cytochrome P-450 drug-metab-olizing enzymes.

    37

    Table 2 shows the magnitude ofinteractions between HIV-protease inhibitors andother drugs commonly used to treat HIV infection.In many cases, inhibitors of P-450 increase plasmaconcentrations of protease inhibitors. For example,concurrent administration of ketoconazole increasesthe area under the plasma-concentrationtime curveby 62 percent with indinavir,

    21

    by 35 percent withnelfinavir,

    22

    and by 300 percent with saquinavir.

    24

    Because the bioavailability of oral saquinavir is poor,this interaction may be advantageous, increasing theamount of drug reaching the systemic circulation.

    A more important concern is the effect of concur-rent therapy with inducers of P-450, such as rifam-pin and rifabutin, which accelerate the clearance ofprotease inhibitors. The plasma concentration istherefore decreased, and the efficacy of the drugis likely to be reduced, which may result in the de-

    velopment of resistance to the drug. Rifampin re-duces the area under the plasma-concentrationtimecurve by 92 percent with indinavir,

    41

    by 82 percentwith nelfinavir,

    22

    by 35 percent with ritonavir,

    23

    andby 80 percent with saquinavir.

    24

    Rifampin shouldnot be given to patients who require treatment withHIV-protease inhibitors.

    42

    HIV-protease inhibitors alter the pharmacokinet-ics of other drugs by acting as P-450 inhibitors orhepatic-enzyme inducers (Table 2). All these drugsinhibit cytochrome P-450 enzymes. Ritonavir is themost potent,

    28

    indinavir and nelfinavir are less so,

    29,37

    DNA5

    p17 p24 p2 p7 p1 p6 ProteaseReverse

    transcriptaseIntegrase

    3

    RNA

    Precursorpolyproteins

    Functionalproteins

    gag

    gagpol

    gag

    env

    pol

    C

    Downloaded from www.nejm.org on September 9, 2006 . Copyright 1998 Massachusetts Medical Society. All rights reserved.

  • 8/6/2019 In Hi Bid Ores Pro Teas A

    4/12

    1284

    Apri l 30, 1998

    The New England Journal of Medicine

    and saquinavir is the least potent.

    26

    The Ki for theinhibition of terfenadine metabolism is 0.017 m

    Mfor ritonavir

    28

    and 0.7 m

    M for saquinavir,

    26

    a 40-folddifference in potency.

    Concurrent administration of rifabutin with in-dinavir, nelfinavir, or ritonavir increases the area un-der the rifabutin plasma-concentrationtime curveby 204 percent,

    21

    207 percent,

    22

    and 350 percent,

    23

    respectively. For patients taking indinavir or nelfin-avir who require concurrent therapy with rifabutin,the dose of rifabutin should be decreased to 150 mgdaily.

    21,22,42 Concurrent administration of rifabutinwith ritonavir or saquinavir is not recommended.

    23,24

    The combination of ritonavir and rifabutin is associ-ated with an increased incidence of rifabutin-associ-ated toxicity (uveitis).

    43

    Clinicians must be aware ofpotentially toxic drug interactions involving HIV-protease inhibitors and avoid prescribing such drugsas terfenadine, astemizole, cisapride, ergotamines,and potent benzodiazepines such as midazolam andtriazolam to patients receiving an HIV-protease in-hibitor.

    Cytochrome P-450 pharmacokinetic interactionsmay be beneficial when two protease inhibitors aregiven simultaneously. For example, ritonavir inhibitsthe hepatic first-pass metabolism of saquinavir, in-

    Figure 2.

    Structures of Five HIV-Protease Inhibitors with Antiretroviral Activity in Clinical Trials.

    NHtBU denotes amino-tertiary butyl, and Ph phenyl.

    Indinavir

    Nelfinavir

    Ritonavir

    Saquinavir

    Amprenavir

    N

    N

    N N

    N

    N

    N

    N

    N NN

    O

    O

    OO

    OO

    CH3

    O

    O

    O

    H

    H

    H

    H

    H

    S

    S

    S

    S

    OH

    NHtBu

    HO

    OH

    OH

    Ph

    Ph

    Ph

    Ph

    O

    O

    O

    O

    N

    H

    N

    H

    N

    H

    N

    H

    N

    H

    N

    H

    CH3SO3H

    CH3SO3H

    NH2

    NH2

    H2SO4

    O

    N

    H

    N

    H

    OH

    OH

    OH

    O NHtBu

    OO

    Downloaded from www.nejm.org on September 9, 2006 . Copyright 1998 Massachusetts Medical Society. All rights reserved.

  • 8/6/2019 In Hi Bid Ores Pro Teas A

    5/12

    DRUG THERAPY

    Vo lu me 3 38 N um be r 18

    1285

    creasing steady-state plasma concentrations of sa-quinavir by a factor of 20 to 30.

    44

    Nelfinavir increas-es the area under the plasma-concentrationtimecurve of saquinavir by 392 percent and increases thatof indinavir by 51 percent.

    22

    Indinavir increases thearea under the curve of saquinavir by about 500 per-cent.

    38

    Treatment with two protease inhibitors takesadvantage of this pharmacokinetic enhancement andincreases antiviral activity.

    Nelfinavir and ritonavir also reduce the plasmaconcentrations of other drugs, presumably becauseof hepatic enzyme induction (Table 2). Nelfinavirand ritonavir decrease the area under the plasma-concentrationtime curve of ethinyl estradiol by 47percent

    22

    and 40 percent,

    23

    respectively. These twoprotease inhibitors should not be given to womentaking a combination oral contraceptive. Nelfinavirand ritonavir reduce the area under the plasma-con-centrationtime curve of zidovudine by 35 percent

    22

    and 25 percent,

    23

    respectively, presumably becauseof the induction of glucuronyl transferases. Howev-er, the intracellular concentration of zidovudine tri-phosphate (the active drug) is not usually affected bysuch a reduction in the plasma concentration of zi-dovudine,

    45

    and therefore no adjustment of the dose

    of zidovudine is recommended when it is given withnelfinavir or ritonavir. The pharmacokinetics of oth-er nucleoside analogues, which are mainly eliminat-ed by the kidneys, are not affected by protease inhib-itors.

    Ritonavir can induce its own metabolism, a proc-ess known as autoinduction. Steady-state troughplasma concentrations fall by a factor of two to threeduring the first two weeks of therapy in patients giv-en a fixed dose of ritonavir.

    46

    Therefore, the doseshould be higher during the first two weeks of ther-apy. The current recommendations are to start witha dose of 300 mg every 12 hours for three days, fol-lowed by a dose of 400 mg every 12 hours for threedays, then 500 mg every 12 hours for three days, andthen 600 mg every 12 hours, if tolerated.

    23

    Such adose escalation is not recommended for nelfinavir.

    22

    SIDE EFFECTS

    Protease inhibitors have important side effects(Table 3). All approved protease inhibitors have gas-trointestinal side effects. High serum aminotransfer-ase concentrations have been reported in association

    with these drugs, but hepatitis is rare.

    50

    Hyperlipide-mia,

    51

    glucose intolerance, and abnormal fat distri-

    *Data are mean values and ranges in adults without hepatic or renal dysfunction, as reported by Merck (for indinavir),

    21Agouron Pharmaceuticals (fornelfinavir),

    22

    Abbott Laboratories (for ritonavir),

    23

    and Roche Laboratories (for the Invirase formulation of saquinavir).

    24

    C

    max

    denotes maximal concentra-tion during a dosing interval, T

    max

    time to the maximal concentration, T

    1/2

    half-life of the principal elimination (

    b

    ) phase, V

    d

    volume of distribution, CSF

    cerebrospinal fluid, P-450 cytochrome P-450 drug-metabolizing enzymes (with induction denoting a significant increase and inhibition a significantdecrease in the metabolism of other P-450 substrates), and NR not reported.

    Recommended doses and regimens are listed.

    The effect of food is expressed as the change in the area under the plasma-concentrationtime curve after a standard (high-fat) breakfast as comparedwith the area under the curve during fasting.

    Variability is expressed as the coefficient of variation (the standard deviation divided by the mean) for the area under the curve during a dosing interval.

    The values shown are the ratio of the CSF concentration to the simultaneous plasma concentration.

    Lighter meals (toast and coffee or corn flakes with skim milk) have no significant effect on the area under the curve.

    **Data are from Collier et al.

    25

    Data are on file at Abbott Laboratories.

    In vitro studies show that saquinavir can act as a P-450 inhibitor at higher concentrations than those usually achieved with the Invirase formulation.

    26

    T

    ABLE

    1. P

    HARMACOKINETICS

    OF

    A

    PPROVED

    HIV-P

    ROTEASE

    I

    NHIBITORS

    .*

    D

    RUG

    D

    OSE

    A

    PPROXIMATE

    O

    RAL

    B

    IO

    -

    AVAILABILITY

    E

    FFECT

    OF

    F

    OOD

    C

    max

    T

    max

    T

    1

    /

    2

    VARIABILITYPROTEINBINDING Vd

    CSFCONCEN-TRATION

    CLEARANCE(ROUTE) P-450

    INDUC-TION

    INHIBI-TION

    mg % % mg/ml hr hr % % liters/kg % %

    Indinavir 800 every8 hr

    6065 77 7.7 0.8 1.8 2247 6065 NR 2.276** 8890(hepatic)

    No Yes

    Nelfinavir 750 threetimesa day

    78 200 to 300 3.04.0 2.04.0 3.55.0 NR 98 2.07.0 NR 78(hepatic)

    Yes Yes

    Ritonavir 600 twicea day

    6675 7 (liquid);15 (capsules)

    11.2 2.04.0 3.05.0 3036 9899 0.4 1 95(hepatic)

    Yes Yes

    Saquinavir 600 threetimesa day

    4 670 0.2 NR NR 4684 98 10.0 1 97(hepatic)

    No No

    Downloaded from www.nejm.org on September 9, 2006 . Copyright 1998 Massachusetts Medical Society. All rights reserved.

  • 8/6/2019 In Hi Bid Ores Pro Teas A

    6/12

    1286 Apri l 30, 1998

    The New England Journal of Medicine

    bution (buffalo hump) may also occur (Table 3).Hemorrhage has been reported in patients with he-mophilia taking protease inhibitors,21-24 but the roleof the drug is uncertain. Current information on theside effects of protease inhibitors is based on treat-ment of only a few thousand patients per drug. Oth-er toxic effects may be recognized with wider use.

    Each of these drugs has distinct dose-limiting tox-ic effects. Nephrolithiasis is the most important side

    effect of indinavir and can occur within a few daysafter the start of treatment.21 The incidence of flankpain and nephrolithiasis in clinical studies rangesfrom 3 to 15 percent.21,52,53 Indinavir is poorly watersoluble; it is associated with crystalluria, and neph-rolithiasis may result from the precipitation of indin-avir in the renal tubules.53 Patients taking indinavirshould drink at least 1.4 liters (48 oz) of fluid dailyin addition to their normal fluid intake.21 Indinavircauses fewer gastrointestinal problems than the oth-er drugs. Reversible unconjugated hyperbilirubine-mia is frequent in patients taking indinavir but is notusually associated with high serum aminotransferaseconcentrations or overt liver disease.21 Several casesof hemolytic anemia have also been associated withthe use of indinavir.21

    Diarrhea is the dose-limiting side effect of nelfin-avir.22,54,55 It can usually be controlled with antidiar-rheal drugs such as loperamide or fiber supplements.

    Nausea, vomiting, and abdominal pain occur fre-quently with ritonavir, especially during the first few

    weeks of therapy.23 Patients starting treatment shouldbe told to expect these side effects. Ritonavir alsocauses circumoral paresthesias in up to 25 percent of

    *Except as otherwise indicated, data are mean values reported by Merck(for indinavir),21 Agouron Pharmaceuticals (for nelfinavir),22 Abbott Lab-oratories (for ritonavir),23 and Roche Laboratories (for the Invirase formu-lation of saquinavir).24 AUC denotes area under the plasma-concentrationtime curve during an average dosing interval, NR not reported, and NCno statistically significant change.

    The value is reported by McCrea et al.38

    Data are from Cox et al.39

    Data are from Murphy et al.40

    TABLE 2. SELECTED PHARMACOKINETIC DRUG INTERACTIONSINVOLVING APPROVED HIV-PROTEASE INHIBITORS.*

    DRUG INDINAVIR NELFINAVIRRITONA-

    VIR SAQUINAVIR

    % change in AUC

    Effect of other drugs on HIV-protease inhibitors

    HIV-protease inhibitorsIndinavirNelfinavirRitonavirSaquinavir

    51NRNR

    83

    15218

    NR9NR

    500392

    2000

    P-450 inhibitorsKetoconazoleClarithromycinFluconazoleFluoxetine

    622919NR

    35NRNRNR

    NR121219

    300NRNRNR

    P-450 inducersRifabutinRifampin

    3292

    3282

    NR35

    4080

    Nucleoside analoguesDidanosineLamivudine

    StavudineZidovudine

    NRNC

    NC13

    NCNR

    NRNC

    NCNR

    NRNC

    NRNR

    NRNC

    Nonnucleoside reverse-transcriptaseinhibitors

    DelavirdineNevirapine

    7228

    NRNR

    2NC

    52027

    Effect of HIV-protease inhibitors on other drugs

    Anti-infective drugsClarithromycinIsoniazidKetoconazoleRifabutinSulfamethoxazole

    531368

    204NC

    NRNRNR

    207NR

    77NRNR

    350NR

    NRNRNCNRNR

    Nucleoside analoguesDidanosineLamivudineStavudine

    ZalcitabineZidovudine

    NR6

    25

    NR17 to 36

    NR10NC

    NR35

    13NRNR

    NR25

    NRNRNR

    NCNC

    Other drugsDesipramineEthinyl estradiolNorethindroneTheophyllineTrimethoprim

    NR2426NR

    19

    NR4718NRNR

    14540NR

    4320

    NRNRNRNRNR

    *One plus sign indicates toxicity of moderate or severe intensity reported

    in less than 10 percent of treated patients but occurring at least twice asoften as in concurrently treated patients not taking the protease inhibitor.Two plus signs indicate toxicity in at least 10 percent of treated patientsand occurring at least twice as often as in control patients. A minus signindicates toxicity occurring less than twice as often in treated patients as incontrol patients and in less than 3 percent of treated patients. NR denotesnot reported. Data are for the doses listed in Table 1. Data are from Merck(for indinavir),21 Agouron Pharmaceuticals (for nelfinavir),22 Abbott Lab-oratories (for ritonavir),23 and Roche Laboratories (for the Invirase andFortovase formulations of saquinavir).24,36

    Reported by Keruly et al.47 and Dong et al.48

    Reported by Mann et al.49

    TABLE 3. MAJORSIDE EFFECTSOF HIV-PROTEASE INHIBITORS.*

    SIDE EFFECT INDINAVIR NELFINAVIR RITONAVIR SAQUINAVIR

    Nausea

    Vomiting NR

    Diarrhea

    Asthenia or fatigue

    Nephrolithiasis orflank pain

    NR NR NR

    Hyperbilirubinemia NR

    High serum amino-transferase con-centrations

    High serum triglyc-eride concentra-tions

    NR NR NR

    Hyperglycemia

    Fat redistribution

    Paresthesias NR NR

    Downloaded from www.nejm.org on September 9, 2006 . Copyright 1998 Massachusetts Medical Society. All rights reserved.

  • 8/6/2019 In Hi Bid Ores Pro Teas A

    7/12

    DRUG THERAPY

    Vo lu me 3 38 N um be r 18 1287

    patients and, less commonly, paresthesias of thearms and legs.23 The cause is unclear. In most pa-tients, these symptoms resolve during continuedtreatment and are not severe enough to cause dis-continuation of the drug. Ritonavir causes hypertri-glyceridemia in up to 5 percent of patients; serum

    triglyceride concentrations can exceed 1000 mg perdeciliter (11 mmol per liter).23 This side effect hasnot been accompanied by complications such aspancreatitis.

    In its original formulation, saquinavir was associ-ated with occasional diarrhea but few serious sys-temic toxic effects.24 The new formulation, For-tovase, has improved bioavailability; it is much morelikely to cause nausea and diarrhea36 and may havemore frequent systemic toxicity than the older for-mulation, Invirase.

    CLINICAL ANTIVIRAL ACTIVITY

    Monotherapy

    HIV-protease inhibitors rapidly and profoundlyreduce the viral load, as indicated by a decline inplasma HIV RNA concentrations within a few daysafter the start of treatment.56,57 Monotherapy withindinavir, nelfinavir, or ritonavir causes plasma HIVRNA concentrations to be reduced by a factor of100 to 1000 in 4 to 12 weeks.58,59 The rate of de-cline in the viral load suggests that the half-life ofcirculating plasma virus is 6 to 24 hours and thehalf-life of circulating virus-infected cells is 2 to3 days.56,57,60 Reductions in the viral load are paral-leled by mean increases in the CD4 count of 100to 150 cells per cubic millimeter.3,52,58,59

    The magnitude and duration of the reduction inthe viral load with protease-inhibitor monotherapyare directly related to the dose and dosing regimen.In a study of indinavir, doses of less than 2400 mgper day caused only short-lived suppression of the vi-ral load.61 With ritonavir, twice-daily doses of 300,400, 500, or 600 mg resulted in equivalent initial re-ductions in the viral load, but only the 600-mg dosecaused a sustained suppression of the viral load and asustained increase in the CD4 count.58,59 The doseresponse effects of nelfinavir and amprenavir weresimilar.54,62 Because of the poor oral bioavailability ofsaquinavir in the older formulation (Invirase), onlydoses of 3600 to 7200 mg per day, which exceeded

    the approved dose by a factor of 2 to 4, caused re-ductions in the viral load approaching those achievedwith indinavir, nelfinavir, or ritonavir.63

    In an occasional patient, improvements in the vi-ral load and CD4 count may persist for more thanone year with the use of only one protease inhibi-tor.64 Monotherapy is no longer recommended,however, because the duration of the antiviral re-sponse is usually limited and resistance to the drugmay develop.

    Combination Therapy

    Phase 3 clinical trials have evaluated the adminis-tration of protease inhibitors in combination withnucleosides. Current clinical guidelines recommendcombining a protease inhibitor with two nucleosideanalogues (e.g., zidovudine and lamivudine or stav-udine and lamivudine4,65).

    In three large clinical trials, protease inhibitorscombined with nucleoside analogues slowed the pro-gression of disease and improved survival. In a trialinvolving 1090 patients with base-line CD4 countsof less than 100 per cubic millimeter, ritonavir add-ed to nucleoside therapy reduced the combined endpoints of new opportunistic diseases and death by53 percent and reduced the end point of death aloneby 43 percent, as compared with placebo.1,23 Themedian duration of follow-up in this study was sixmonths. Scores for the quality of life declined dur-ing the first four weeks of treatment with ritonavir,

    probably because of side effects, but then improvedsignificantly as compared with base-line values. Thepatients in the placebo group had a gradual declinein the quality of life.66

    Saquinavir in combination with zalcitabine signif-icantly improved survival and slowed the progressionof disease, as compared with saquinavir or zalcita-bine alone, in a randomized, double-blind trial in-

    volving 978 patients treated for a minimum of 16weeks. Combination therapy reduced the combinedclinical end points of disease progression and deathby 40 percent and reduced the single end point ofdeath by 68 percent, as compared with either drugalone.2

    In a randomized, double-blind trial involving1156 patients with base-line CD4 counts of lessthan 200 per cubic millimeter who were followedfor a median of 38 weeks, the combination of indin-avir, zidovudine, and lamivudine reduced the com-bined end points of clinical progression and deathby 50 percent and reduced the end point of deathalone by 57 percent, as compared with the results inpatients treated only with zidovudine and lamivu-dine.3 Studies of nelfinavir and amprenavir with theuse of clinical end points have not been completed.

    Combinations of protease inhibitors and nucleo-side analogues can suppress HIV for long periods oftime. The combination of indinavir with zidovudine

    and lamivudine reduced the plasma viral load to un-detectable concentrations (50 copies per milliliter)in 70 percent of patients after 24 weeks52 and in 60percent after 2 years.67 The combination of nelfin-avir with zidovudine and lamivudine produced sim-ilar results, with the viral load reduced to less than500 copies per milliliter in 80 percent of patients af-ter one year of treatment.68 The response rate wassimilar in patients given ritonavir combined with zi-dovudine and lamivudine.69

    Downloaded from www.nejm.org on September 9, 2006 . Copyright 1998 Massachusetts Medical Society. All rights reserved.

  • 8/6/2019 In Hi Bid Ores Pro Teas A

    8/12

    1288 Apri l 30, 1998

    The New England Journal of Medicine

    With 11 antiretroviral drugs currently approved inthe United States, clinicians and investigators are us-ing a variety of two-, three-, and even four-drugcombinations to treat HIV infection. For example, acombination of two protease inhibitors suppressesthe viral load in patients who have not previously re-

    ceived protease inhibitors. The combination of ri-tonavir (400 mg twice daily) and saquinavir (400mg twice daily) was tolerated by most patients andreduced the plasma viral load to a level of less than500 copies per milliliter for more than 16 weeks.44

    These results are similar to those of studies usingeither drug in combination with nucleoside ana-logues. The effects of combination therapy withritonavir and saquinavir were less impressive in pa-tients in whom prior treatment with a protease in-hibitor had failed. In one small study, only 45 per-cent of patients who did not have responses toindinavir had viral loads of less than 500 copies permilliliter 12 weeks after switching to treatment with

    ritonavir and saquinavir.70 In contrast, all 12 patients who did not have responses to nelfinavir had viralloads of less than 500 copies per milliliter when theyreceived the four-drug regimen of ritonavir, saquin-avir, stavudine, and lamivudine.71

    Combining a protease inhibitor with a nonnucleo-side reverse-transcriptase inhibitor can also profound-ly suppress the viral load. All 12 patients treated withindinavir and the nonnucleoside reverse-transcrip-tase inhibitor nevirapine had viral loads of less than500 copies per milliliter after an average of 24 weeksof treatment.40 Combined treatment with indinavirand the investigational nonnucleoside reverse-trans-criptase inhibitor efavirenz reduced the viral load to

    less than 400 copies per milliliter in 91 percent ofpatients after 60 weeks.72

    RESISTANCE AND TREATMENT FAILURE

    The limited duration of the anti-HIV responsethat occurs in most patients treated with protease-inhibitor monotherapy is associated with the appear-ance of drug-resistant virus.73,74 The major aminoacid mutations associated with clinical resistancehave been mapped.75 A substitution of phenylalaninefor valine at position 82 is the initial mutation asso-ciated with resistance to indinavir and ritonavir, anaspartate-to-asparagine mutation at position 30 re-sults in initial resistance to nelfinavir, and mutationsin glycine at position 48 and leucine at position 90result in initial resistance to saquinavir.75

    In general, initial single amino acid mutations yield only a slight change (by less than a factor of5) in drug sensitivity.18,20,76,77 However, secondarymutations accumulate in the virus and can lead tohigh-level drug resistance. Unlike the initial muta-tions, the secondary mutations in virus from pa-tients with resistance to different protease inhibitorsoverlap.73-75 HIV protease can tolerate a substantial

    amount of mutation; at least one third of its 99 ami-no acids can deviate from the wild-type sequence

    without altering function.73,74 In patients with resist-ance to protease inhibitors, plasma viral loads andCD4 cell counts have returned to pretreatment

    values, indicating that resistant mutants are viru-

    lent.73,74,78,79Prolonged treatment with one protease inhibitor

    can result in the emergence of virus with both pri-mary and secondary resistance mutations. These vi-ruses are resistant not only to the drug being givenbut also to other protease inhibitors that the patienthas never received. Monotherapy with indinavir, forexample, can result in the development of virus thatis resistant to indinavir and to other protease inhib-itors.73 This pattern also occurs with ritonavir mono-therapy74 and could presumably be caused by mono-therapy with any HIV-protease inhibitor. Once apatient has resistant virus, it is retained even aftertreatment stops. In patients with indinavir-resistant

    virus who discontinued monotherapy with indinavirand then received indinavir combined with nucleo-side analogues, an initial small reduction in the viralload was followed by an increase within four weeks.The failure of this treatment was associated with therapid reemergence of indinavir-resistant virus.79

    In some circumstances, the development of virusthat is resistant to protease inhibitors may be a func-tion of plasma drug concentrations. Higher drugdoses, with higher plasma concentrations, were asso-ciated with a more prolonged antiviral response andpresumably a lower propensity for the developmentof resistant virus.54,58,59,61,62 A concentrationresponserelation for indinavir was delineated in a small num-

    ber of patients in whom the maximal reduction inthe viral load occurred with trough plasma concen-trations of more than 0.4 mmol per liter; the reduc-tion was minimal if the trough plasma concentration

    was less than 0.2 mmol per liter.80 In addition, therate of accumulation of mutations causing resistanceto ritonavir was inversely proportional to the troughplasma concentration of the drug during an averagedosing interval.74

    Drug regimens that maintain high plasma concen-trations may be more effective in preventing theemergence of resistant virus than regimens with low-er plasma concentrations. The currently recommend-ed regimens for ritonavir, indinavir, and nelfinavir re-sult in plasma concentrations greater than or equalto the concentration required to reduce virus pro-duction by 90 percent (IC90) throughout an averagedosing interval.27 Although it seems reasonable tomaintain trough concentrations at a level higher thanthe IC90 value, saquinavir in combination with zal-citabine was clinically beneficial with a dosing regi-men that resulted in trough concentrations far be-low the IC90 value.

    2,27 Similarly, nevirapine, whichinduces cytochrome P-450, reduced plasma indina-

    Downloaded from www.nejm.org on September 9, 2006 . Copyright 1998 Massachusetts Medical Society. All rights reserved.

  • 8/6/2019 In Hi Bid Ores Pro Teas A

    9/12

    DRUG THERAPY

    Vo lu me 3 38 N um be r 18 1289

    vir concentrations by 28 percent, on average, but thecombination of these two drugs resulted in a reduc-tion in the viral load of the same magnitude as thatassociated with regimens that did not affect theclearance of indinavir.40

    However, because resistance may be a conse-

    quence of suboptimal plasma drug concentrations,strict adherence to recommended regimens shouldbe encouraged. In a study of compliance in a smallnumber of patients, genotypic resistance was associ-ated with a high plasma viral load or episodes ofpoor compliance.81 Patients who have difficulty tol-erating a high dose of a protease inhibitor may bebetter advised to stop taking all antiretroviral drugsthan to take a lower dose of the protease inhibitor.Physicians should be cautious when prescribing pro-tease inhibitors for patients with a history of poorcompliance.

    Because of the problem of cross-resistance, a switchfrom one protease inhibitor to another should take

    place, if possible, before high-level resistance to theinitial drug occurs (Fig. 3). The sequential additionof single drugs to an ineffective regimen is associated

    with a poor outcome and may result in the sequen-tial selection of drug-resistant virus. For example,

    when indinavir was added after long-term combina-tion therapy with zidovudine and lamivudine, only45 percent of patients had plasma viral loads that

    were less than 500 copies per milliliter67 abouthalf the expected response rate in patients withoutprior antiretroviral therapy who are given the samethree drugs. In patients without responses to initialregimens, salvage therapy should be started at theearliest sign of failure (e.g., a sustained rise in the

    plasma viral load 1.0 log10 copies per milliliter) andshould include two or three drugs the patient hasnever received.4,65

    Combining a potent protease inhibitor with twonucleoside analogues appears to prevent the emer-gence of resistance to either class of drugs.82 Occa-sional lapses in compliance may also be less danger-ous in patients taking combinations of antiretroviraldrugs, because of the ability of one type of drug tosuppress the replication of virus resistant to the oth-er type of drug.

    PATHOPHYSIOLOGIC CONSEQUENCES

    Despite potential problems with resistance, treat-ment with combination antiretroviral regimens con-taining protease inhibitors has a number of beneficialpathophysiologic consequences. Besides increasingthe overall CD4 cell count, combination therapymay increase naive and memory T cells, enhancelymphoproliferative responses, and reduce plasmaconcentrations of harmful cytokines such as tumornecrosis factor.83 Early intervention with combina-tion regimens in a small number of patients with re-cent seroconversion forestalled the loss of HIV-spe-

    cific CD4 lymphocyte responses that characterizesthe natural history of this disease.84

    Unfortunately, protease-inhibitor therapy may notcorrect deficiencies in the T-cell repertoire alreadyinduced by HIV, and lost lymphocyte clones maynot be replaced.85 These observations may explain

    why infections caused by opportunistic pathogenssuch as cytomegalovirus developed in some patientsduring the late stage of the acquired immunodefi-ciency syndrome (AIDS) even though their CD4counts increased from less than 50 to more than 200cells per cubic millimeter while they were receivingtreatment with highly active regimens containingprotease inhibitors.86,87

    Finally, despite the prolonged suppression of HIVviremia in large numbers of patients, no one has yetbeen cured of HIV infection. Patients with unde-

    Figure 3. Changes in HIV Viral Load over Time in Three PatientsTreated with Combination Antiretroviral Regimens Including

    HIV-Protease Inhibitors.Viral load was measured every four weeks. A change of both

    the protease inhibitor and the nucleoside analogues used inthe regimen occurred at the times indicated by the arrows. Thedotted lines indicate the lower limit of detection in an ultrasen-

    sitive polymerase-chain-reaction assay for HIV-1 RNA.

    20

    200,000

    20,000

    2,000

    200

    SuccessfulTherapy

    20

    200,000

    20,000

    2,000

    200

    PlasmaHIVRNA(copies/ml)

    SalvageTherapy

    Changein regimen

    20

    200,000

    20,000

    2,000

    200

    0 4 8 12 16 20 24 28 32 36 40 44 48 52

    Week

    UnsuccessfulTherapy

    Changein regimen

    Downloaded from www.nejm.org on September 9, 2006 . Copyright 1998 Massachusetts Medical Society. All rights reserved.

  • 8/6/2019 In Hi Bid Ores Pro Teas A

    10/12

    1290 Apri l 30, 1998

    The New England Journal of Medicine

    tectable plasma viral loads and greatly reduced num-bers of viral particles in lymph nodes still had detect-able proviral DNA in cells even after 6 to 12 monthsof therapy.88,89 After at least two years of combina-tion therapy consisting of a protease inhibitor andnucleoside analogues, patients with undetectable

    plasma viral loads still had HIV in their lymph nodesand peripheral-blood cells that was capable of repli-cation.90-92

    Eradication of HIV with current combinationregimens may be impossible. Most if not all infectedpatients have a small amount of virus in long-livedcellular reservoirs, and eliminating HIV in even asmall percentage of such patients may require at leastthree to five years of continuous treatment.93,94 Fur-thermore, among patients with undetectable viralloads who are receiving indinavir combined with zi-dovudine and lamivudine, those who stopped takingone or two of the drugs after three to six months oftherapy had relapses up to six times as often as those

    who continued to take all three drugs.95,96

    CONCLUSIONS

    Protease-inhibitor therapy is the most importantrecent advance in the treatment of HIV infection.

    All infected patients with symptomatic disease,CD4 counts of less than 500 cells per cubic milli-meter, or plasma HIV RNA concentrations of morethan 5000 to 10,000 copies per milliliter should re-ceive a protease inhibitor plus two nucleoside ana-logues unless contraindicated.4,64 Although thesecombinations substantially reduce the plasma viralload, increase the CD4 cell count, reduce the pro-gression of disease, and prolong life, they have side

    effects. The emergence of drug-resistant strains ofHIV may limit the long-term effectiveness of treat-ment with protease inhibitors. Monotherapy, sub-therapeutic drug concentrations, and noncompli-ance may promote resistance to this class of drugs.Using protease inhibitors in combination with nu-cleoside analogues or nonnucleoside reverse-trans-criptase inhibitors reduces the development of resist-ance but requires substantial cooperation on thepatients part.

    Protease inhibitors are expensive, with an annualwholesale price ranging from $4,320 to $8,010 perpatient.97 Combination regimens that include pro-tease inhibitors cost about $10,000 per year of lifesaved.97 This figure compares favorably with the costof other prescription drugs given to prevent morbid-ity and mortality in patients with other chronic con-ditions such as hypertension and ischemic heart dis-ease.97

    To maximize the long-term benefit of HIV-pro-tease inhibitors, several issues need to be resolved.

    What are the most appropriate regimens for infants,children, and pregnant women? Can we developsimplified dosing schemes to promote adherence to

    the regimen? Will combinations of two protease in-hibitors be as effective over time as a single proteaseinhibitor combined with reverse-transcriptase inhib-itors? What role should nonnucleoside reverse-trans-criptase inhibitors play in these regimens?

    Important questions about protease inhibitors re-

    main. Much of the information in this article has notbeen published in peer-reviewed journals, and the re-sults of important new studies are reported monthly.The complexity of decisions involving antiretroviraltherapy now rivals that of decisions involving che-motherapy for cancer. Physicians should maintain ahealthy skepticism while awaiting clarification of theoptimal use of protease inhibitors, and wheneverpossible, specialists in the care of patients with AIDSshould be involved in treatment decisions.

    I am indebted to Ms. Laura Rocco for editorial assistance, to Ms. Anne Capriotti for assistance with the figures, and to Ms. Joann

    Nicolette and Dr. Carol Trapnell for assistance with the preparationof the manuscript.

    REFERENCES

    1. Cameron DW, Heath-Chiozzi M, Kravcik S, Mills R, Potthoff A, HenryD. Prolongation of life and prevention of AIDS complications in advancedHIV immunodeficiency with ritonavir: update. In: Volume 1 of Programand abstracts of the 11th International Conference on AIDS, Vancouver,B.C., July 712, 1996:24-5. abstract.2. Salgo MP, Beattie D, Bragman K, Donatacci L, Jones M, MontgomeryL. Saquinavir (Invirase) vs. HIVID (zalcitabine) vs. combination as treat-ment for advanced HIV infection in patients discontinuing/unable to takeRetrovir (zidovudine). In: Volume 1 of Program and abstracts of the 11thInternational Conference on AIDS, Vancouver, B.C., July 712, 1996:24.abstract.3. Hammer SM, Squires KE, Hughes MD, et al. A controlled trial of twonucleoside analogues plus indinavir in persons with human immunodefi-ciency virus infection and CD4 cell counts of 200 per cubic millimeter or

    less. N Engl J Med 1997;337:725-33.4. Carpenter CCJ, Fischl MA, Hammer SM, et al. Antiretroviral therapyfor HIV infection in 1997: updated recommendations of the International

    AIDS Society USA Panel. JAMA 1997;277:1962-9.5. Erickson JW. The not-so-great escape. Nat Struct Biol 1995;2:523-9.[Erratum, Nat Struct Biol 1996;3:103.]6. Pearl LH, Taylor WR. A structural model for the retroviral proteases.Nature 1987;329:351-4.7. Kramer RA, Schaber MD, Skalka AM, Ganguly K, Wong-Staal F, ReddyEP. HTLV-III gagprotein is processed in yeast cells by the virus pol-pro-tease. Science 1986;231:1580-4.8. Graves MC, Lim JJ, Heimer EP, Kramer R A. An 11-kDa form of humanimmunodeficiency virus protease expressed in Escherichia coliis sufficientfor enzymatic activity. Proc Natl Acad Sci U S A 1988;85:2449-53.9. Le Grice SFJ, Mills J, Mous J. Active site mutagenesis of the AIDS v irusprotease and its alleviation by trans complementation. EMBO J 1988;7:2547-53.10. Kohl NE, Emini EA, Schleif WA, et al. Active human immunodeficien-cy virus protease is required for viral infectivity. Proc Natl Acad Sci U S A

    1988;85:4686-90.11. Henderson LE, Bowers MA, Sowder RC II, et al. Gag proteins of thehighly replicative MN strain of human immunodeficiency virus type 1:posttranslational modifications, proteolytic processings, and complete ami-no acid sequences. J Virol 1992;66:1856-65.12. Flexner C, Broyles SS, Earl P, Chakrabarti S, Moss B. Characterizationof human immunodeficiency virus gag/pol gene products expressed by re-combinant vaccinia viruses. Virology 1988;166:339-49.13. Karacostas V, Nagashima K, Gonda MA, Moss B. Human immunode-ficiency virus-like particles produced by a vaccinia virus expression vector.Proc Natl Acad Sci U S A 1989;86:8964-7.14. Debouck C. The HIV-1 protease as a therapeutic target for AIDS.

    AIDS Res Hum Retroviruses 1992;8:153-64.15. Overton HA, McMillan DJ, Gridley SJ, Brenner J, Redshaw S, Mills

    Downloaded from www.nejm.org on September 9, 2006 . Copyright 1998 Massachusetts Medical Society. All rights reserved.

  • 8/6/2019 In Hi Bid Ores Pro Teas A

    11/12

    DRUG THERAPY

    Vo lu me 3 38 N um be r 18 1291

    JS. Effect of two novel inhibitors of the human immunodeficiency virusprotease on the maturation of the HIVgagandgag-polpolyproteins. Vi-rology 1990;179:508-11.16. Roberts NA, Martin JA, Kinchington D, et al. Rational design of pep-tide-based HIV proteinase inhibitors. Science 1990;248:358-61.17. Kempf DJ, Marsh KC, Denissen JF, et al. ABT-538 is a potent inhibitorof human immunodeficiency virus protease and has high oral bioavailabilityin humans. Proc Natl Acad Sci U S A 1995;92:2484-8.

    18. Patick AK, Mo H, Markowitz M, et al. Antiviral and resistance studiesof AG1343, an orally bioavailable inhibitor of human immunodeficiency vi-rus protease. Antimicrob Agents Chemother 1996;40:292-7.19. Vacca JP, Dorsey BD, Schleif WA, et al. L-735,524: an orally bioavail-able human immunodeficiency virus type 1 protease inhibitor. Proc Natl

    Acad Sci U S A 1994;91:4096-100.20. Ho DD, Toyoshima T, Mo H, et al. Characterization of human immu-nodeficiency virus type 1 variants with increased resistance to a C2-symmet-ric protease inhibitor. J Virol 1994;68:2016-20.21. Crixivan (indinavir sulfate) capsules product monograph. West Point,Pa.: Merck, 1997 (package insert).22. Viracept (nelfinavir mesylate) tablets and oral powder product mono-graph. La Jolla, Calif.: Agouron Pharmaceuticals, 1997 (package insert).23. Norvir (ritonavir) capsules product monograph. North Chicago, Ill.:

    Abbott Laboratories, 1997 (package insert).24. Invirase (saquinavir mesylate) capsules product monograph. Nutley,N.J.: Roche Laboratories, 1997 (package insert).25. Collier AC, Marra C, Coombs RW, Zhong L, Stone J, Nguyen B. Cer-ebrospinal fluid indinavir and HIV RNA levels in patients on chronic in-

    dinavir therapy. In: Program and abstracts of the Infectious Diseases Soci-ety of America 35th Annual Meeting, San Francisco, September 1316,1997:75. abstract.26. Fitzsimmons ME, Collins JM. Selective biotransformation of the hu-man immunodeficiency virus protease inhibitor saquinavir by human small-intestinal cytochrome P4503A4: potential contribution to high first-passmetabolism. Drug Metab Dispos 1997;25:256-66.27. Flexner C. Pharmacokinetics and pharmacodynamics of HIV proteaseinhibitors. Infect Med 1996;13:Suppl F:16-23.28. Kumar GN, Rodrigues AD, Buko AM, Denissen JF. CytochromeP450-mediated metabolism of the HIV-1 protease inhibitor ritonavir(ABT-538) in human liver microsomes. J Pharmacol Exp Ther 1996;277:423-31. [Erratum, J Pharmacol Exp Ther 1997;281:1506.]29. Chiba M, Hensleigh M, Nishime JA, Balani SK, Lin JH. Role of cy-tochrome P450 3A4 in human metabolism of MK-639, a potent humanimmunodeficiency virus protease inhibitor. Drug Metab Dispos 1996;24:307-14.30. Bryant M, Getman D, Smidt M, et al. SC-52151, a novel inhibitor ofthe human immunodeficiency virus protease. Antimicrob AgentsChemother 1995;39:2229-34.31. Bilello JA, Bilello PA, Prichard M, Robins T, Drusano GL. Reductionof the in vitro activity of A77003, an inhibitor of human immunodeficien-cy virus protease, by human serum a1 acid glycoprotein. J Infect Dis 1995;171:546-51.32. Lazdins JK, Mestan J, Goutte G, et al. In vitro effect of a1-acid glyco-protein on the anti-human immunodeficiency virus activity of the proteaseinhibitor CGP 61755: a comparative study with other relevant HIV pro-tease inhibitors. J Infect Dis 1997;175:1063-70.33. Kageyama S, Anderson BD, Hoesterey BL, et al. Protein binding ofhuman immunodeficiency virus protease inhibitor KNI-272 and alterationof its in vitro antiretroviral activity in the presence of high concentrationsof proteins. Antimicrob Agents Chemother 1994;38:1107-11.34. Flexner C, Richman DD, Bryant M, et al. Effect of protein bindingon the pharmacodynamics of an HIV protease inhibitor. Antiviral Res1995;26:A282. abstract.35. Moyle GJ, Sadler M, Hawkins D, Buss N. Pharmacokinetics ofsaquinavir at steady state in CSF and plasma: correlation between plasmaand CSF viral load in patients on saquinavir containing regimens. In: Pro-gram and abstracts of the Infectious Diseases Society of America 35th An-nual Meeting, San Francisco, September 1316, 1997:115. abstract.36. Fortovase (saquinavir) soft gelatin capsules product monograph. Nut-ley, N.J.: Roche Laboratories, 1997 (package insert).37. Piscitelli SC, Flexner C, Minor JR, Polis MA, Masur H. Drug interac-tions in patients infected with human immunodeficiency virus. Clin InfectDis 1996;23:685-93.38. McCrea J, Buss N, Stone J, et al. Indinavir-saquinavir single dose phar-macokinetic study. In: Program and abstracts of the Fourth Conference onRetroviruses and Opportunistic Infections, Washington, D.C., January 2226, 1997:177. abstract.39. Cox SR, Ferry JJ, Batts DH, et al. Delavirdine and marketed proteaseinhibitors: pharmacokinetic interaction studies in healthy volunteers. In:Program and abstracts of the Fourth Conference on Retroviruses and Op-

    portunistic Infections, Washington, D.C., January 2226, 1997:133. ab-stract.40. Murphy R, Gagnier P, Lamson M, Dusek A, Ju W, Hsu A. Effect ofnevirapine on pharmacokinetics of indinavir and ritonavir in HIV-1 pa-tients. In: Program and abstracts of the Fourth Conference on Retrovirusesand Opportunistic Infections, Washington, D.C., January 2226, 1997:133. abstract.41. McCrea J, Chodakewitz J, Deutsch P, et al. Effect of rifampin on the

    pharmacokinetics of indinavir in healthy volunteers. Clin Pharmacol Ther1997;61:152. abstract.42. Impact of HIV protease inhibitors on the treatment of HIV-infectedtuberculosis patients with rifampin. MMWR Morb Mortal Wkly Rep 1996;45:921-5.43. Sun E, Heath-Chiozzi M, Cameron DW, et al. Concurrent ritonavirand rifabutin increases risk of rifabutin-associated adverse events. In: Vol-ume 1 of Program and abstracts of the 11th International Conference on

    AIDS, Vancouver, B.C., July 712, 1996:18. abstract.44. Cohen C, Sun E, Cameron W, et al. Ritonavir-saquinavir combinationtreatment in HIV-infected patients. In: Addendum to Program and ab-stracts of the 36th Interscience Conference on Antimicrobial Agents andChemotherapy, New Orleans, September 1518, 1996. Washington, D.C.:

    American Society for Microbiology, 1996:8. abstract.45. Flexner C, Hendrix CW. Pharmacology of antiretroviral agents. In:DeVita VT, Hellman S, Rosenberg SA, eds. AIDS: etiology, diagnosis,treatment, and prevention. 4th ed. Philadelphia: Lippincott-Raven, 1997:479-93.46. Hsu A, Granneman GR, Witt G, et al. Multiple-dose pharmacokinetics

    of ritonavir in human immunodeficiency virus-infected subjects. Antimi-crob Agents Chemother 1997;41:898-905.47. Keruly JC, Chaisson RE, Moore RD. Diabetes and hyperglycemia inpatients receiving protease inhibitors. In: Program and abstracts of theFifth Conference on Retroviruses and Opportunistic Infections, Chicago,February 15, 1998:157. abstract.48. Dong BJ, Gruta C, Legg J, et al. Diabetes and use of protease inhib-itors. In: Program and abstracts of the Fifth Conference on Retrovirusesand Opportunistic Infections, Chicago, February 15, 1998:157. abstract.49. Mann M, Piazza-Hepp T, Koller E, et al. Abnormal fat distribution in

    AIDS patients following protease inhibitor therapy: FDA summary. In:Program and abstracts of the Fifth Conference on Retroviruses and Op-portunistic Infections, Chicago, February 15, 1998:157. abstract.50. Brau N, Leaf HL, Wieczorek RL, Margolis DM. Severe hepatitis inthree AIDS patients treated with indinavir. Lancet 1997;349:924-5.51. Roberts AD, Muesing A, Parenti DM, Hsia J, Wasserman AG, SimonGL. Alterations in serum lipids and lipoproteins with indinavir in HIV-infected patients. In: Program and abstracts of the Infectious Diseases So-ciety of America 35th Annual Meeting, San Francisco, September 1316,1997:114. abstract.52. Gulick RM, Mellors JW, Havlir D, et al. Treatment with indinavir, zi-dovudine, and lamivudine in adults with human immunodeficiency virusinfection and prior antiretroviral therapy. N Engl J Med 1997;337:734-9.53. Kopp JB, Miller KD, Mican JAM, et al. Crystalluria and urinary tractabnormalities associated with indinavir. Ann Intern Med 1997;127:119-25.54. Moyle GL, Youle M, Higgs C, et al. Extended follow-up of the safetyand activity of Agourons HIV protease inhibitor AG1343 (Viracept) in vi-rological responders from the UK phase I/II dose finding study. In: Vol-ume 1 of Program and abstracts of the 11th International Conference on

    AIDS, Vancouver, B.C., July 712, 1996:1:18. abstract.55. Gathe J, Burkhardt B, Hawley P, et al. A randomized phase II studyof Viracept, a novel HIV protease inhibitor, used in combination withstavudine vs. stavudine alone. In: Volume 1 of Program and abstracts ofthe 11th International Conference on AIDS, Vancouver, B.C., July 712,1996:25. abstract.56. Wei X, Ghosh SK, Taylor ME, et al. Viral dynamics in human immu-nodeficiency virus t ype 1 infection. Nature 1995;373:117-22.57. Ho DD, Neumann AU, Perelson AS, Chen W, Leonard JM, MarkowitzM. Rapid turnover of plasma virions and CD4 lymphocytes in HIV-1 in-fection. Nature 1995;373:123-6.58. Danner SA, Carr A, Leonard JM, et al. A short-term study of the safe-ty, pharmacokinetics, and efficacy of ritonavir, an inhibitor of HIV-1 pro-tease. N Engl J Med 1995;333:1528-33.59. Markowitz M, Saag M, Powderly WG, et al. A preliminary study of ri-tonavir, an inhibitor of HIV-1 protease, to treat HIV-1 infection. N EnglJ Med 1995;333:1534-9.60. Perelson AS, Neumann AU, Markowitz M, Leonard JM, Ho DD.HIV-1 dynamics in vivo: virion clearance rate, infected cell life-span, and

    viral generation t ime. Science 1996;271:1582-6.61. Steigbigel RT, Berry P, Mellors J, et al. Efficacy and safety of the HIVprotease inhibitor indinavir sulfate (MK 639) at escalating doses. In: Pro-gram and abstracts of the Third Conference on Retroviruses and Oppor-

    Downloaded from www.nejm.org on September 9, 2006 . Copyright 1998 Massachusetts Medical Society. All rights reserved.

  • 8/6/2019 In Hi Bid Ores Pro Teas A

    12/12

    1292 Apri l 30, 1998

    The New England Journal of Medicine

    tunistic Infections, Washington, D.C., January 28February 1, 1996:80.abstract.62. Schooley RT. Preliminary data on the safety and antiviral efficacy ofthe novel protease inhibitor 141W94 in HIV-infected patients with 150 to400 CD4 cells/mm3. In: Addendum to Program and abstracts of the36th Interscience Conference on Antimicrobial Agents and Chemotherapy,New Orleans, September 1518, 1996. Washington, D.C.: American So-ciety for Microbiology, 1996:8. abstract.

    63. Schapiro JM, Winters MA, Stewart F, et al. The effect of high-dosesaquinavir on viral load and CD4 T-cell counts in HIV-infected patients.Ann Intern Med 1996;124:1039-50.64. Emini EA, Condra JH, Schleif WA, et al. Maintenance of long-term

    virus suppression in patients treated with the HIV-1 protease inhibitorCrixivan (indinavir). In: Volume 1 of Program and abstracts of the 11thInternational Conference on AIDS, Vancouver, B.C., July 712, 1996:18.abstract.65. Department of Health and Human Services Panel on Clinical Practicesfor Treatment of HIV Infection. Guidelines for the use of antiretroviralagents in HIV-infected adults and adolescents. November 5, 1997. or . (Also availablefrom NAPS [document no. 05455, 43 pages], c/o Microfiche Publica-tions, P.O. Box 3513, Grand Central Station, New York, NY 10163-3513.This is not a multiarticle document. Remit in advance [in U.S. funds only]$15 for the first 20 photocopies [$0.50 per page thereafter] or $5 for thefirst microfiche [$1 per microfiche thereafter]. Outside the U.S. and Can-ada add postage of $4.50 for the first 20 pages, $1 for every 10 pages there-after [$1 per microfiche]. There is a $25 invoicing fee for purchase orders.)66.

    Nabulsi AA, Revicki D, Conway D, Maurath C, Mills R, Leonard J.Quality of life consequences of adding ritonavir to current antiviral therapyfor advanced HIV patients. In: Volume 2 of Program and abstracts of the11th International Conference on AIDS, Vancouver, B.C., July 712,1996:31. abstract.67. Gulick R, Mellors J, Havlir D, et al. Indinavir (IDV), zidovudine(ZDV) and lamivudine (3TC): concurrent or sequential therapy in ZDV-experienced patients. In: Program and abstracts of the 37th InterscienceConference on Antimicrobial Agents and Chemotherapy, Toronto, Sep-tember 28October 1, 1997. Washington, D.C.: American Society for Mi-crobiology, 1997:259. abstract.68. Saag M, Gersten M, Chang Y, et al. Long term virological and immu-nological effect of the HIV protease inhibitor Viracept (nelfinavir mesylate)in combination with zidovudine (AZT) and lamivudine (3TC). In: Pro-gram and abstracts of the Infectious Diseases Society of America 35th An-nual Meeting, San Francisco, September 1316, 1997:112. abstract.69. Markowitz M, Cao Y, Hurley A, et al. Triple therapy with AZT, 3TC,and ritonavir in 12 subjects newly infected with HIV-1. In: Supplement toProgram and abstracts of the 11th International Conference on AIDS,

    Vancouver, B.C., July 712, 1996:28. abstract.70. Sampson M, Torres RA, Stein AJ, et al. R itonavir-saquinavir combina-tion treatment in protease inhibitor experienced patients with advancedHIV disease. In: Program and abstracts of the 37th Interscience Confer-ence on Antimicrobial Agents and Chemotherapy, Toronto, September28October 1, 1997. Washington, D.C.: American Society for Microbiol-ogy, 1997:262. abstract.71. Henry K, Kane E, Melroe H, et al. Experience with a ritonavir/saquinavir based regimen for the treatment of HIV-infection in subjectsdeveloping increased viral loads whi le receiving nelfinavir. In: Program andabstracts of the 37th Interscience Conference on Antimicrobial Agents andChemotherapy, Toronto, September 28October 1, 1997. Washington,D.C.: American Society for Microbiology, 1997:282. abstract.72. Kahn J, Mayers D, Riddler S, et al. Durable clinical anti -HIV-1 activity(60 weeks) and tolerability for efavirenz (DMP 266) in combination withindinavir (IDV). In: Program and abstracts of the Fifth Conference on Ret-roviruses and Opportunistic Infections, Chicago, February 15, 1998:208.abstract.73. Condra JH, Schleif WA, Blahy OM, et al. In vivoemergence of HIV-1

    variants resistant to multiple protease inhibitors. Nature 1995;374:569-71.74. Molla M, Korneyeva M, Gao Q, et al. Ordered accumulation of mu-tations in HIV protease confers resistance to ritonavir. Nat Med 1996;2:760-6.75. Schinazi RF, Larder BA, Mellors JW. Mutations in retroviral genes as-sociated with drug resistance. Int Antiviral News 1996;4:95-107.76. el-Farrash MA, Kuroda MJ, Kitazaki T, et al. Generation and charac-terization of a human immunodeficiency virus type 1 (HIV-1) mutant re-sistant to an HIV-1 protease inhibitor. J Virol 1994;68:233-9.77. Otto MJ, Garber S, Winslow DL, et al. In vitroisolation and identifi-cation of human immunodeficiency virus (HIV) variants with reduced sen-sitivity to C-2 symmetrical inhibitors of HIV type 1 protease. Proc Natl

    Acad Sci U S A 1993;90:7543-7.

    78. Schapiro JM, Winters MA, Vierra M, et al. Causes of long-term effi-cacy and/or drug failure in protease inhibitor monotherapy. In: Volume 1of Program and abstracts of the 11th International Conference on AIDS,

    Vancouver, B.C., July 712, 1996:25. abstract.79. Condra J, Schleif WA, Blahy OM, et al. Evidence for the existence oflong lived genetic reservoirs of HIV-1 in infected patients. In: Program andabstracts of the Fourth International Workshop in HIV Drug Resistance,Sardinia, Italy, July 69, 1995:82. abstract.

    80. Stein DS, Fish DG, Bilello JA, Preston SL, Martineau GL, DrusanoGL. A 24-week open-label phase I/II evaluation of the HIV protease in-hibitor MK-639 (indinavir). AIDS 1996;10:485-92.81. Vanhove GF, Schapiro JM, Winters MA, Merigan TC, Blaschke TF. Pa-tient compliance and drug failure in protease inhibitor monotherapy.JAMA 1996;276:1955-6.82. Condra JH, Holder DJ, Schleif WA, et al. Bi-directional inhibition ofHIV-1 drug resistance selection by combination therapy with indinavir andreverse transcriptase inhibitors. In: Supplement to Program and abstractsof the 11th International Conference on AIDS, Vancouver, B.C., July 712, 1996:19. abstract.83. Lederman M, Connick E, Landay A, et al. Partial immune reconstitu-tion after 12 weeks of HAART (AZT, 3TC, ritonavir): preliminary resultsof ACTG 315. In: Program and abstracts of the Fourth Conference on Ret-roviruses and Opportunistic Infections, Washington, D.C., January 2226,1997:208. abstract.84. Rosenberg ES, Billingsley JM, Caliendo AM, et al. Vigorous HIV-1-specific CD4 T cell responses associated with control of viremia. Science1997;278:1447-50.85.

    Connors M, Kovacs JA, Krevat S, et al. HIV infection induces changesin CD4 T-cell phenotype and depletions within the CD4 T-cell reper-toire that are not immediately restored by antiviral or immune-based ther-apies. Nat Med 1997;3:533-40.86. Jacobson MA, Kramer F, Pavan PR, Owens S, Pollard R. Failure ofhighly active antiretroviral therapy (HAART) to prevent CMV retinitis de-spite marked CD4 count increase. In: Program and abstracts of the FourthConference on Retroviruses and Opportunistic Infections, Washington,D.C., January 2226, 1997:129. abstract.87. Gilquin J, Piketty C, Thomas V, Gonzales-Canali G, Kazatchine MD.

    Acute CMV infection in AIDS patients receiving combination therapy in-volving protease inhibitors. In: Program and abstracts of the Fourth Con-ference on Retroviruses and Opportunistic Infections, Washington, D.C.,January 2226, 1997:129. abstract.88. Cavert W, Staskus K, Zupancic M, et al. Quantitative in situ hybridi-zation measurement of HIV-1 RNA clearance kinetics from lymphoid tis-sue cellular compartments during triple-drug therapy. In: Program and ab-stracts of the Fourth Conference on Retroviruses and OpportunisticInfections, Washington, D.C., January 2226, 1997:207. abstract.89. Wong JK, Gunthard HF, Havlir DV, et al. Reduction of HIV-1 inblood and lymph nodes following potent antiretroviral therapy and the vi-rologic correlates of treatment failure. Proc Natl Acad Sci U S A 1997;94:12574-9.90. Finzi D, Hermankova M, Pierson T, et al. Identification of a reservoirfor HIV-1 in patients on highly active antiretroviral therapy. Science 1997;278:1295-300.91. Wong JK, Hezareh M, Gunthard HF, et al. Recovery of replication-competent HIV despite prolonged suppression of plasma vi remia. Science1997;278:1291-5.92. Chun TW, Stuyver L, Mizell SB, et al. Presence of an inducible HIV-1 latent reservoir during highly active antiretroviral therapy. Proc Natl AcadSci U S A 1997;94:13193-7.93. Chun TW, Carruth L, Finzi D, et al. Quantification of latent tissuereservoirs and total body viral load in HIV-1 infection. Nature 1997;387:183-8.94. Perelson AS, Essunger P, Cao Y, et al. Decay characteristics of HIV-1-infected compartments dur ing combination therapy. Nature 1997;387:188-91.95. Havlir DV, Hirsch M, Collier A, et al. Randomized trial of indinavir(IDV) vs. zidovudine (ZDV)/lamivudine (3TC) vs. IDV/ZDV/3TCmaintenance therapy after induction IDV/ADV/3TC therapy. In: Programand abstracts of the Fifth Conference on Retroviruses and OpportunisticInfections, Chicago, February 15, 1998:225. abstract.96. Raffi F, Pialoux G, Brun-Vezinet F, et al. Results of TRILEGE Trial,a comparison of three maintenance regimens for HIV infected adults re-ceiving induction therapy with zidovudine (ZDV), lamivudine (3TC), andindinavir (IDV). In: Program and abstracts of the Fifth Conference on Ret-roviruses and Opportunistic Infections, Chicago, February 15, 1998:225.abstract.97. Moore RD, Bartlett JG. Combination antiretroviral therapy in HIV in-fection. Pharmacoeconomics 1996;10:109-13.

    D l d d f j S t b 9 2006 C i ht 1998 M h tt M di l S i t All i ht d