Strategies of Antiretroviral Therapy in Adults

Similar documents
0.14 ( 0.053%) UNAIDS 10% (94) ( ) (73-94/6 ) 8,920

Terapia antirretroviral inicial y de rescate: Utilidad actual y futura de nuevos medicamentos

2 nd Line Treatment and Resistance. Dr Rohit Talwani & Dr Dave Riedel 12 th June 2012

National AIDS Treatment Advocacy Project

Quick Reference Guide to Antiretrovirals. Guide to Antiretroviral Agents

C h a p t e r 5 5 HIV Therapy Where are We Now?

MEDICAL COVERAGE GUIDELINES ORIGINAL EFFECTIVE DATE: 03/07/18 SECTION: DRUGS LAST REVIEW DATE: 02/19/19 LAST CRITERIA REVISION DATE: ARCHIVE DATE:

HIV MEDICATIONS AT A GLANCE. Atripla 600/200/300 mg tablet tablet daily. Complera 200/25/300 mg tablet tablet daily

HIV medications HIV medication and schedule plan

Principles of Antiretroviral Therapy

Anumber of clinical trials have demonstrated

Medscape's Antiretroviral Pocket Guide for the Treatment of HIV Infection

The Annotated Bibliography of the UCSF HIV Solid Organ Transplantation Project. ARV Dosing in End Stage Renal Disease

HIV - Life cycle. HIV Life Cyle

PAEDIATRIC HIV INFECTION. Dr Ashendri Pillay Paediatric Infectious Diseases Specialist

Antiretroviral therapy for patients with HIV disease

THE HIV LIFE CYCLE. Understanding How Antiretroviral Medications Work

AIDS 101: From Abstinence to Zidovudine

Management of NRTI Resistance

Continuing Education for Pharmacy Technicians

TOXICITY, TOLERABILITY, AND ADHERENCE TO THERAPY

Guidelines for the Use of Antiretroviral Agents in HIV-1-Infected Adults and Adolescents

ABC/3TC/ZDV ABC PBO/3TC/ZDV

Somnuek Sungkanuparph, M.D.

HIV 101: Fundamentals of HIV Infection

0% 0% 0% Parasite. 2. RNA-virus. RNA-virus

HIV Treatment Update. Awewura Kwara, MD, MPH&TM Associate Professor of Medicine and Infectious Diseases Brown University

Malaysian Consensus Guidelines on Antiretroviral Therapy Cheng Joo Thye Hospital Raja Permaisuri Bainun Ipoh

National AIDS Treatment Advocacy Project

Perspective Resistance and Replication Capacity Assays: Clinical Utility and Interpretation

HIV Update Objectives. Epidemiology. Epidemiology, Transmission and Natural History. Transmission Risk by Exposure. Transmission 9/29/2014

HIV epidemiology since HIV in the United States. HIV Transmission

Treatment strategies for the developing world

Special Contribution Questions to and Answers from the International AIDS Society USA Resistance Testing Guidelines Panel

Recommendations for Use of Antiretroviral Drugs in Pregnant HIV-1-Infected Women for Maternal Health and

POST-EXPOSURE PROPHYLAXIS, PRE-EXPOSURE PROPHYLAXIS, & TREATMENT OF HIV

MedChem 401~ Retroviridae. Retroviridae

Management of patients with antiretroviral treatment failure: guidelines comparison

Although the public s awareness of infection with the

Hydroxyurea with ddi or ddi/d4t: a novel approach to HIV therapy

ANTIRETROVIRAL therapy directed at the reverse

HIV THERAPY STRATEGIES FOR THIRD LINE. issues to consider when faced with few drug options

Antiviral Chemotherapy

HIV Drugs and the HIV Lifecycle

Combination of protease inhibitors for the treatment of HIV-1-infected patients: a review of pharmacokinetics and clinical experience

Concise Review for Primary-Care Physicians

Nobel /03/28. HIV virus and infected CD4+ T cells

Structured Treatment Interruption in HIV Positive Patients. Leah Jackson, BScPhm Pharmacy Resident HIV Rotation January 23, 2007

MDR HIV and Total Therapeutic Failure. Douglas G. Fish, MD Albany Medical College Albany, New York Cali, Colombia March 30, 2007

Distribution and Effectiveness of Antiretrovirals in the Central Nervous System

ANTIRETROVIRAL (ART) DRUG INFORMATION FOR HEALTH CARE PROFESSIONAL

The advent of protease inhibitors (PIs) as PROCEEDINGS CLINICAL EXPECTATIONS OF EFFICACY: PROTEASE INHIBITOR POTENCY * Benjamin Young, MD, PhD

Understanding Viruses CHAPTER 38. Antiviral Agents. Understanding Viruses (cont'd) Viral Infections (cont'd) Viral Infections.

Comprehensive Guideline Summary

When to start: guidelines comparison

Many highly active antiretroviral therapy PROCEEDINGS

Updated Recommendations of the International AIDS Society USA Panel JAMA. 2000;283:

Northwest AIDS Education and Training Center Educating health care professionals to provide quality HIV care

Treatment of HIV-1 in Adults and Adolescents: Part 2

Terapia antirretroviral I: Casos especiales


Supplementary information

Simplifying HIV Treatment Now and in the Future

The legally binding text is the original French version TRANSPARENCY COMMITTEE OPINION. 14 December 2011

Treatment of respiratory virus infection Influenza A & B Respiratory Syncytial Virus (RSV)

Supplementary Figure 1. Gating strategy and quantification of integrated HIV DNA in sorted CD4 + T-cell subsets.

AMERICAN ACADEMY OF PEDIATRICS. Guidance for the Clinician in Rendering Pediatric Care. Peter L. Havens, MD, and the Committee on Pediatric AIDS

TB/HIV Co-Infection. Tuberculosis and HIV

CCC ARV Dosing Recommendations for HIV-exposed infants Updated

TB Intensive Tyler, Texas December 2-4, Tuberculosis and HIV Co-Infection. Lisa Y. Armitige, MD, PhD. December 4, 2008.

Supplemental Digital Content 1. Combination antiretroviral therapy regimens utilized in each study

HIV replication and selection of resistance: basic principles

WOMENS INTERAGENCY HIV STUDY ANTIRETROVIRAL DOSAGE FORM SECTION A. GENERAL INFORMATION

Decision Analysis Example

The New England Journal of Medicine

Objectives. HIV in the Trenches HIV Update for the Primary Care Provider, An Overview The HIV Continuum of Care.

HIV 101: Overview of the Physiologic Impact of HIV and Its Diagnosis Part 2: Immunologic Impact of HIV and its Effects on the Body

The US Food and Drug Administration has

Virologic and CD4 Cell Response to Zidovudine or Zidovudine and Lamivudine Following Didanosine Treatment of Human Immunodeficiency Virus Infection

NOTICE TO PHYSICIANS. Division of AIDS (DAIDS), National Institute of Allergy and Infectious Diseases, National Institutes of Health

Reverse transcriptase and protease inhibitor resistant mutations in art treatment naïve and treated hiv-1 infected children in India A Short Review

VIKING STUDIES Efficacy and safety of dolutegravir in treatment-experienced subjects

A. K. PATICK,* T. J. BORITZKI, AND L. A. BLOOM Agouron Pharmaceuticals, Inc., San Diego, California 92121

Scottish Medicines Consortium

Sysmex Educational Enhancement and Development No

The New England Journal of Medicine NELFINAVIR, EFAVIRENZ, OR BOTH AFTER THE FAILURE OF NUCLEOSIDE TREATMENT OF HIV INFECTION

Treatment experience in South Africa. Dr Ian Sanne Clinical HIV Research Unit University of the Witwatersrand

The prevalence of antiretroviral drug resistance in the United States

Perspective Antiretroviral Therapy: Select Lessons From Recent Studies

Natural history of HIV Infection

Antiretroviral Dosing in Renal Impairment

Antiretroviral Therapy During Pregnancy and Delivery: 2015 Update

A Genetic Test to Screen for Abacavir Hypersensitivity Reactions

CONCISE COMMUNICATION

Selecting an Initial Antiretroviral Therapy (ART) Regimen

The legally binding text is the original French version TRANSPARENCY COMMITTEE OPINION. 9 May 2012

The Future of HIV: Advances in Drugs and Research. Shauna Gunaratne December 17, 2018

PHARMACOKINETICS OF ANTIRETROVIRAL AND ANTI-HCV AGENTS

TORONTO GENERAL HOSPITAL HIV AMBULATORY CARE ROTATION

NATAPNATIONAL. HIV 102: Care & Treatment AIDS TREATMENT ADVOCACY PROJECT. Origins of HIV. Table of Contents. National AIDS Treatment Advocacy Project

Transcription:

Strategies of Antiretroviral Therapy in Adults JOHN A. BARTLETT Duke University Medical Center, Durham, North Carolina, USA Key Words. HIV Treatment strategies AIDS Antiretroviral treatment HIV therapy ABSTRACT Recent progress in antiretroviral treatment has led to dramatic improvements in HIV-related morbidity and mortality. These improvements have been fostered by advances in our understanding of HIV-related pathogenesis, the use of plasma HIV RNA levels to monitor patients, and the availability of 11 licensed antiretroviral drugs, including the potent protease inhibitors. Numerous drug INTRODUCTION Tremendous popular interest is focused on the development of new treatments for HIV-infected persons, and the pendulum of public understanding has swung from euphoria over the availability of antiretroviral combinations which provide complete suppression of plasma viremia to despair over treatment failure in patients who have exhausted all available options. An objective summary of progress in the last three years clearly identifies dramatic treatment successes, tempered by the realization that HIV disease is a chronic infection for which we have no immediately curative therapy. Many reports now describe marked decreases in AIDS-related complications, hospitalizations, and deaths [1, 2]. Within the Adult Infectious Diseases Clinic at Duke University Medical Center, the number of persons dying from AIDS has declined from 144 in 1995 to a projected total of 30 in 1997. Three major advances underlie these improvements in clinical outcomes: a better understanding of HIV-related pathogenesis, the ability to directly and sensitively measure HIV activity with plasma HIV RNA levels, and the availability of an increasing number of new therapeutic agents with potent antiretroviral activity, differing mechanisms of action, and nonoverlapping patterns of resistance. Given the marked expansion in the tools to monitor and treat HIVinfected persons, we must now develop the optimal strategies to guide clinical practice. In many respects, the evolution of HIV medicine is similar to oncology 20 years ago. The paradigms of oncologic medicine may offer important models for the development of new HIV treatment strategies. combinations, especially those containing three agents, can suppress plasma HIV RNA levels below the lower limit of detection in the majority of treated patients. However, the limitations of this therapeutic response patient compliance, drug resistance, and a residual burden of chronically infected cells which are refractory to treatment should be familiar to the oncologist. The Oncologist 1998;3:111-118 RELEVANT PATHOPHYSIOLOGY Acute HIV Infection After the acquisition of HIV, most commonly through sexual contact, the initially infected cells appear to be macrophages within the genital tract. Macrophage-tropic HIV strains, also possessing the in vitro culture characteristic of not inducing syncytia, are most commonly transmitted [3]. Animal studies suggest that a regional lymphadenitis soon follows [4]. From this initial localized infection HIV disseminates hematogenously, and the virus may then localize on follicular dendritic cells throughout lymphoid tissue [5, 6]. From the surface of follicular dendritic cells, HIV can then infect CD4 lymphocytes trafficking through the germinal centers of lymphoid tissue. This process creates a large reservoir of chronically HIV-infected cells within lymph nodes, the entire reticuloendothelial system, lymphoid tissue in the gut, and microglial cells in the central nervous system [5, 6]. During acute HIV infection, high-grade viremia can be documented by high titers of culturable plasma virus or high levels of plasma HIV RNA [7, 8]. Resolution of this high-grade viremia may correspond to the development of an anti-hiv cytotoxic T-lymphocyte response [9]. Most diagnostic tests for HIV infection measure anti- HIV antibodies. The serologic response against HIV usually takes 4-12 weeks to develop, and therefore conventional diagnostic tests are not useful during acute HIV infection. More useful tests during this period, such as p24 antigen levels Correspondence: John A. Bartlett, M.D., Duke University Medical Center, Box 3238, Durham, North Carolina 27710, USA; Telephone: 919-681-6060; Fax: 919-681-8474; e-mail: jab5@acpub.duke.edu Accepted for publication January 23, 1998. AlphaMed Press 1083-7159/98/$5.00/0 The Oncologist 1998;3:111-118

112 Advances in HIV Therapy (a core-associated protein) or plasma HIV RNA levels, may directly measure virus. Preliminary evidence suggests that the measurement of plasma HIV RNA levels may be most sensitive [10], but significant impediments to its widespread use include availability, timeliness, and cost. Much attention is currently focused on the treatment of acute HIV infection. The use of potent combination regimens during this period could limit the dissemination of virus to lymphoid tissue and create a smaller reservoir of chronically infected cells, thereby limiting the potential for viral diversity and resistance [11]. Established HIV Infection Established HIV infection infers that the dissemination of HIV has occurred and the reservoir of chronically infected cells has been created. In patients with established infection, virus within plasma and peripheral blood mononuclear cells exists in a dynamic state of turnover [12, 13]. It has been observed that 10 10 virions are produced every day within an HIV-infected person. Within this background of rapid virus turnover, it is easy to appreciate the potential for the rapid emergence of drug-resistant HIV strains. The mechanisms of CD4 + lymphocyte depletion are not yet fully elucidated, but it appears that chronic viral replication contributes both directly and indirectly to CD4 + lymphocyte decline. Prognostic Markers Within the past several years, the prognostic value of plasma HIV RNA measurements has become more clearly appreciated [14-16]. Interestingly, plasma HIV RNA levels may reach a temporary steady state following the resolution of acute HIV infection [14]. This level has been described as the viral set point and clearly portends the risk of clinical progression to AIDS or death over the next 10 years. Measurement of plasma HIV RNA levels throughout the course of HIV disease, not just at the set point, also offers important prognostic information concerning the risk of clinical progression to AIDS and death [15, 16] and the risk of vertical transmission [17]. Therapeutic interventions which decrease plasma HIV RNA levels are associated with an improved clinical prognosis [18-22]. Recognition of the prognostic value of plasma HIV RNA levels has led to the incorporation of their measurement in routine clinical practice, usually on a quarterly schedule [23]. Plasma HIV RNA levels may display considerable variability, and changes up to twofold may not be considered biologically important [24]. Immunizations and intercurrent illnesses have been demonstrated to transiently increase plasma HIV RNA levels [25], and measurement following these events may be misleading. The methods for measuring plasma HIV RNA include RT-PCR, bdna, and NASBA. All offer relatively consistent measurements, although they differ in their lower limit of detection. Clinical trial results now suggest that the nadir of plasma HIV RNA levels on treatment may be very important in determining the durability of an antiretroviral regimen [26]. This observation carries a profound implication for clinical practice, and strongly suggests that the goal of treatment for all patients should be the complete suppression of plasma HIV RNA below the lower limit of detection. In addition, great efforts are under way to improve the sensitivity of plasma HIV RNA measurements to even lower levels, (i.e., ultra-sensitive assays which measure down to 20 copies/ml). Absolute CD4 + cell counts also offer important prognostic information for HIV-infected persons and complement the measurement of plasma HIV RNA levels. Absolute CD4 + cell counts remain important predictors of complicating opportunistic infections. Many clinicians now measure absolute CD4 + cell counts every six months unless a patient is near a decision point for the initiation of prophylaxis for an opportunistic infection. AVAILABLE AGENTS Nucleoside Reverse Transcriptase Inhibitors (NRTIs) General Observations The NRTIs are the most extensively evaluated class of antiretroviral drugs. NRTIs require intracellular triphosphorylation to become activated. When used alone, NRTIs typically offer 0.5-1.0 log 10 decreases in plasma HIV RNA levels, and when two NRTIs are combined, the decreases are usually 1.0-1.5 log 10 copies/ml. They may be combined with non-nucleoside reverse transcriptase inhibitors (NNRTIs) and the protease inhibitors (PIs). Specific NRTIs are shown in Table 1. Combinations NRTIs may be combined with one definite exception: zidovudine and stavudine. These two drugs have exhibited both in vitro and in vivo antagonism [27]. Zalcitabine should probably not be combined with stavudine or didanosine due to overlapping toxicities. Stavudine may be combined with didanosine cautiously, but patients should be followed closely for peripheral neuropathy and pancreatitis [28]. NNRTIs General Observations NNRTIs are potent inhibitors of HIV in vitro and in vivo, but their use can be complicated by the rapid development of resistance through a single mutation. As a

Bartlett 113 Table 1. Specific NRTIs Dose Toxicities Unique characteristics Approved: Zidovudine 200 mg tid or Macrocytic anemia, neutropenia, Central nervous system penetration; decreases (ZDV, AZT, Retrovir) 300 mg bid myopathy, nausea, fatigue, vertical transmission; 3TC resistance may headaches reverse ZDV resistance Didanosine (DDI, Videx) 200 mg bid Peripheral neuropathy, pancreatitis, May be used as monotherapy; formulated nausea, bloating, diarrhea with buffer; must not be given with food Zalcitabine (DDC, HIVID) 0.75 mg tid Peripheral neuropathy, aphthous ulcers, rash Stavudine (D4T, Zerit) 40 mg bid or Peripheral neuropathy, pancreatitis, Central nervous system penetration 30 mg bid < 60 kg transaminitis, agitation Lamivudine (3TC, Epivir) 150 mg bid Pancreatitis, peripheral neuropathy, 3TC resistance may reverse ZDV resistance; nausea active against hepatitis B; 3TC resistance may cross over to DDI and DDC Unapproved: Abacavir (GW 1592, Ziagen) 300 mg bid Nausea, headaches Central nervous system penetration; potent antiretroviral effect in previously untreated patients Table 2. Specific NNRTIs Dose Toxicities Unique characteristics Approved: Nevirapine (Viramune) 200 mg qd 2 weeks, Rash, nausea, Central nervous system penetration; rapid then 200 mg bid transaminitis onset of activity; drug interactions Delaviridine (Rescriptor) 400 mg tid Rash Unapproved: Etavirenz (DMP-266, Sustiva) 600 mg qd Nausea, dizziness Rapid onset of potent activities; once daily dosing; relatively delayed resistance? result, they must be combined with other antiretroviral drugs. A new NNRTI, DMP-266, may require the acquisition of at least two mutations for resistance, perhaps enhancing its usefulness. NNRTIs require no intracellular phosphorylation and distribute well throughout the body, including the CNS and across the placenta. NNRTIs are commonly metabolized through the cytochrome p450 system and therefore may interact with PIs. NNRTIs may share a common toxicity a macular erythematous rash which is usually self-limited. Specific NNRTIs are shown in Table 2. Combinations NNRTI combinations cannot be recommended because of potential cross-resistance. NNRTIs should always be combined with other antiretroviral agents, either NRTIs or PIs. When combined with PIs, pharmacokinetic interactions may occur which require PI dosage adjustment. Protease Inhibitors General Observations PIs represent the most potent class of antiretroviral agents. They require no intracellular activation and enter the CNS in relatively low concentrations due to plasma protein binding. Careful attention to dosing schedule is crucial because low trough levels of the drug are clearly associated with the appearance of resistance mutations. In the future, therapeutic drug monitoring may become important for use with PIs. PIs should not be used as monotherapy because of potential resistance. PIs may interact with many other drugs, including NNRTIs, because of their extensive cytochrome p450 metabolism. Specific PIs are shown in Table 3. Combinations PIs may be combined based upon drug interactions and resistance patterns. Attractive combinations may include

114 Advances in HIV Therapy Table 3. Specific PIs Dose Toxicities Unique characteristics Approved: Saquinavir (Fortovase) 1200 mg tid Nausea New formulation has improved bioavailability Ritonavir (Norvir) Initial 300 mg bid Nausea, diarrhea, perioral paresthesias, Increase to full dose over first one to two weeks; Full 600 mg bid triglyceride elevations, transaminitis potential drug interactions; must be refrigerated; may be taken with food Indinavir (Crixivan) 800 mg q8h Nausea, bloating, hyperbilirubinemia, Cannot be taken with food; must drink at least nephrolithiasis 1.5 l/day to prevent stones Nelfinavir (Viracept) 750 mg tid Diarrhea, nausea Should be taken with food Unapproved: Amprenavir (GW 141; VX-478) ritonavir and saquinavir (20-30 increase in saquinavir levels) [29], nelfinavir and saquinavir (2-3 increase in saquinavir levels) [30], ritonavir and indinavir (under evaluation), and ritonavir and nelfinavir (under evaluation). The combination of indinavir and saquinavir has demonstrated in vitro antagonism. PIs may be combined with NRTIs, but may require dosage adjustments when combined with NNRTIs. TREATMENT STRATEGIES Nausea General Principles Acknowledging the current availability of 11 antiretroviral agents and anticipating the FDA approval of several new agents within the next year, it is critical to identify the guiding principles for antiretroviral therapy. These principles should include the following considerations: The Perceived Benefit Must be Greater Than the Toxicities, Imposition, and Cost Clear decreases in the risk of clinical progression to AIDS or death have been demonstrated for patients with late-stage HIV disease (absolute CD4 + cells < 200/mm 3 ), receiving triple combination therapy including a protease inhibitor [31, 32]. For patients with an intermediate stage of HIV disease, (absolute CD4 + cells < 500/mm 3 ), combination therapy with two NRTIs offers improved clinical outcomes over ZDV monotherapy [33, 34]. For patients with early HIV disease (absolute CD4 + cells > 500/mm 3 ), there is no clinical trial yet completed which has evaluated clinical outcomes in the recipients of combination therapy. Therefore, it may be reasonable to delay therapy in patients at low risk of clinical progression (low plasma HIV RNA, high absolute CD4 + cells), with close monitoring [35]. Compliance is Crucial! Antiretroviral drug resistance is most likely to occur in regimens which incompletely suppress virus replication. Lower trough plasma levels of protease inhibitors are associated with the appearance of mutations conferring protease inhibitor resistance [36]. Taken together, these observations underscore the need for patient adherence to antiretroviral regimens. Patients who develop resistant virus may also transmit these mutant strains to their sexual partners [37]. Careful patient selection and education before initiating treatment are crucial strategies for therapeutic success. All Regimens Should Include at Least Two Drugs Most currently prescribed antiretroviral regimens include two NRTIs, to which a third drug such as a protease inhibitor or NNRTI is added. Two NRTIs commonly suppress plasma HIV RNA levels by 1.0-1.5 log 10 copies/ml in previously untreated patients [38]. Therefore, unless pretreatment plasma HIV RNA levels are less than 5,000-10,000 copies/ml, it is unlikely that a two-nrti combination can suppress levels below the lower limit of detection. In most patients, a threedrug combination will be required to achieve full suppression, and it is possible that in patients with very high baseline plasma HIV RNA levels, four drugs may be required to reach full suppression. Relative Potency is Greatest for the Protease Inhibitors PIs can suppress plasma HIV RNA levels by 1.5-2.0 log 10 copies/ml [39, 40], greater than the decrease observed with currently available NRTIs or NNRTIs. Therefore, PIs should be considered the backbone of the most active antiretroviral combinations. In the future, NRTIs such as abacavir and NNRTIs such as efavirenz may offer similar suppression.

Bartlett 115 All Regimens are Likely to have a Finite Period of Effectiveness Current evidence suggests that the potent suppression of plasma HIV RNA levels is accompanied by suppression of HIV RNA within lymphoid tissue, although HIV DNA in chronically infected cells does not appear to be decreasing [41, 42]. These observations suggest that antiretroviral therapy achieves suppression only, without any reduction in the chronic viral reservoir. Antiretroviral combinations can suppress plasma HIV RNA levels below the lower limit of detection in up to 90% of subjects for 48 weeks [43], but the longer-term durability of this response remains uncertain. It seems unlikely that this level of suppression can persist indefinitely. Old Drugs May Not be Recyclable The available evidence suggests that resistant strains appear in the plasma and lymphoid tissue almost simultaneously [44]. When a given antiretroviral drug is discontinued, the predominant plasma isolates no longer display drug resistance. However, soon after the reintroduction of the same antiretroviral drug, these resistant strains rapidly reappear within the plasma. Taken together, these observations suggest that lymphoid tissue may serve as an important reservoir for drug-resistant strains and that drugs from a previously failed regimen are unlikely to be effective when reintroduced. Drug Interactions are Important The PIs and NNRTIs are extensively metabolized through the cytochrome p450 system. As a result, the clinician must be aware of potential drug interactions involving these agents. Monitor the Success of Treatment Plasma HIV RNA levels decline rapidly following the initiation of antiretroviral therapy with a t1/2 <12 hours. If patients require early positive reinforcement for their commitment to antiretroviral adherence, measuring plasma HIV RNA levels at two to three weeks may be helpful. Preliminary evidence suggests that a plasma HIV RNA level <1,000 copies/ml at week 12 correlates with a level below the lower limit of detection at week 24 [26]. If patients have not achieved this landmark by week 12, questions should be raised about compliance and should lead to the consideration of treatment intensification with the addition of one to two new antiretroviral drugs. Once on a stable regimen, most clinicians follow plasma HIV RNA levels every three months. Immune Reconstitution? When plasma HIV RNA levels fall below the lower limit of detection, absolute CD4 + cells usually increase by 100-150/mm 3 [43]. However, the functional status of these cells remains uncertain [45, 46]. Treated patients have a lower risk of opportunistic infections and death [31, 32] and refractory complications of progressive HIV disease such as molluscum contagiosum may resolve [47], suggesting that some degree of meaningful immunologic reconstitution is achieved. However, most clinicians are reluctant to discontinue opportunistic infection prophylaxis, especially for Pneumocystis carinii pneumonia, until clinical trials results clarify these issues. Initiating Treatment Timing The decision to initiate therapy should be guided by three factors: Plasma HIV RNA levels Absolute CD4 + cell counts Patient acceptance Currently, there are no clinical trial results to guide consideration of the proper plasma HIV RNA level at which to begin treatment. Therefore, recommendations must be made by extrapolating from cohort studies following plasma HIV RNA levels. Most clinicians would strongly encourage treatment at a plasma HIV RNA level >20,000 copies/ml, would encourage treatment with plasma HIV RNA levels = 10,000-20,000 copies/ml, and would carefully consider treatment with plasma HIV RNA levels <10,000 copies/ml. The International AIDS Society USA Panel recently recommended therapy for all patients with plasma HIV RNA levels >5,000-10,000 copies/ml. Similarly, many clinicians would strongly encourage treatment with absolute CD4 + cells <200/mm 3, would encourage treatment with cells 200-500/mm 3, and would carefully consider treatment with cells >500/mm 3. All recommendations to initiate treatment must involve an informed, committed patient. Poor compliance will inevitably lead to a shortened duration of activity and viral resistance. Initial Combinations The goal of all antiretroviral therapy should be suppression of plasma HIV RNA levels below the limit of detection. The optimal timing to achieve this goal remains to be determined, but should be reached by 24 weeks. The optimal initial regimen also remains to be determined. Here are some options: Two NRTIs: Extensive experience, well tolerated but offer only 1 log 10 suppression of plasma HIV RNA levels at one year. Two NRTIs + NNRTI: Limited experience, well tolerated, with suppression of plasma HIV RNA levels below detectable in 50%-60% at one year; spares the PIs for later.

116 Advances in HIV Therapy Two NRTIs + PI: Moderate experience, well tolerated, extremely potent, with suppression of plasma HIV RNA levels below detectable in 80%-90% at one year. Two PIs: Limited experience, appears to be well tolerated and extremely potent, with suppression of plasma HIV RNA levels below detectable in 80%-90% at six months; uncertain role as initial therapy. Salvage Regimens Given the apparent inability of antiretroviral treatment to significantly diminish the reservoir of chronically infected cells and the likelihood of the eventual emergence of resistant strains, all regimens of suppressive antiretroviral treatment will ultimately fail. Therefore, it is critically important to recognize failing regimens quickly and reinstitute effective treatment with a salvage intervention. Timing Most clinicians follow plasma HIV RNA levels every three months in treated patients. Currently, there is no absolute consensus on the proper plasma HIV RNA threshold at which to change treatment. It is likely that the kinetics of the increase in plasma HIV RNA levels may vary in individual patients depending upon the degree of antiretroviral resistance, viral fitness, and other presently unknown factors. However, many clinicians will agree on changing therapy when plasma HIV RNA levels are >5,000-10,000 copies/ml. In the future, it is very likely that decisions to change treatment may be guided by resistance assays. New Combinations The goal of salvage regimens is also to suppress plasma HIV RNA levels to below the limit of detection. This goal appears to be best achieved by including at least two new drugs without cross-resistance to previously used drugs. Some examples include: NRTI a + NRTI b NRTI c + PI NRTI a + NRTI b + NNRTI NRTI c + PI NRTI a + NRTI b + PI a NRTI c + PI b NRTI c + PI b PI b + PI c + NNRTI CRITICAL ISSUES FOR THE FUTURE Despite the tremendous treatment advances in the past three years, numerous clinical problems need solutions. A partial listing of these problems includes: The need to optimize the performance of available regimens with considerations for the sequencing of combinations, enhancing compliance, and development of clinically useful resistance assays. The continuous need for new salvage regimens through new drug development. The need to diminish the reservoir of chronically infected cells by understanding their natural turnover and identifying methods to accelerate the turnover rate. The need to assess immunologic reconstitution through the use of antiretroviral drugs alone and to consider methods for enhancing this reconstitution. The need to define the potential benefits and optimal treatment strategies for important clinical circumstances such as acute HIV infection and HIV-infected pregnant women. SUMMARY In 1997, HIV-treating clinicians and their patients enjoy far more treatment opportunities than in years past. However, the limitations of these treatments patient compliance, drug resistance, and a residual population of chronically infected cells should be familiar to the oncologist. Perhaps the therapeutic paradigms of clinical oncology will provide a fast track to further improvements in antiretroviral strategies. REFERENCES 1 Chiasson M, Berenson L, Li W et al. Declining AIDS mortality in New York City. Presented at Fourth Conference on Retroviruses and Opportunistic Infections. Washington, DC, 1997; abstract 376. 2 Update: trends in AIDS incidence, deaths, and prevalence United States, 1996. MMWR 1997;46:165-173. 3 Roos MTL, Lange J, de Goede R et al. Viral phenotype and immune response in primary human immunodeficiency virus type 1 infection. J Infect Dis 1992;165:427-432. 4 Musey L, Hughes J, Shacker T et al. Cytotoxic T-cell responses, viral load and disease progression in early human immunodeficiency virus type 1 infection. N Engl J Med 1997;337:1267-1274. 5 Pantaleo G, Graziosi C, Butini L et al. Lymphoid organs function as major reservoirs for human immunodeficiency virus. Proc Natl Acad Sci USA 1991;88:9838-9842. 6 Pantaleo G, Graziosi C, Demarest JF et al. HIV infection is active and progressive in lymphoid tissue during the clinically latent stage of disease. Nature 1993;362:355-358. 7 Daar ES, Mougdil T, Meyer RD et al. Transient high levels of viremia in patients with primary human immunodeficiency virus type 1 infection. N Engl J Med 1991;324:961-964. 8 Clark SJ, Saag MS, Decker WD et al. High titers of cytopathic virus in plasma of patients with symptomatic primary HIV-1 infection. N Engl J Med 1991;324:954-960.

Bartlett 117 9 Spira A, Marx P, Patterson B et al. Cellular targets of infection and route of viral dissemination after an intravaginal inoculation of simian immunodeficiency virus into rhesus macaques. J Exp Med 1996;183:215-225. 10 Shacker T, Collier AC, Hughes J et al. Clinical and epidemiologic features of primary HIV infection. Ann Intern Med 1996;125:257-264. 11 Markowitz M, Cao Y, Hurley A et al. Triple therapy with AZT, 3TC and ritonavir in 12 subjects newly infected with HIV-1. Presented at XIth International Conference on AIDS. Vancouver, B.C., Canada, 1996; Abstract TH.B.933. 12 Wei X, Ghosh SK, Taylor ME et al. Viral dynamics in human immunodeficiency virus type 1 infection. Nature 1995;373:117-122. 13 Ho DD, Neumann AU, Perelson AS et al. Rapid turnover of plasma virions and CD4 lymphocytes in HIV-1 infection. Nature 1995;373:123-126. 14 Mellors JW, Kingsley LA, Rinaldo CR et al. Quantitation of HIV-1 RNA in plasma predicts outcome after seroconversion. Ann Intern Med 1995;122:573-579. 15 Mellors JW, Rinaldo CR, Gupta P et al. Prognosis of HIV-1 infection predicted by the quantity of virus in plasma. Science 1996;272:1167-1170. 16 Mellors JW, Munoz A, Giorgi J et al. Plasma viral load and CD4 + lymphocytes as prognostic markers of HIV-1 infection. Ann Intern Med 1997;126:946-954. 17 Cao Y, Krogstad P, Korber B et al. Maternal HIV-1 viral load and vertical transmission of infection: the Ariel Project for the prevention of HIV transmission from mother to infant. Nat Med 1997;3:549-552. 18 O Brien WA, Hartigan PM, Martin D et al. Changes in plasma HIV-1 RNA and CD4 + lymphocyte counts and the risk of progression to AIDS. N Engl J Med 1996;334:426-431. 19 Phillips AN, Eron J, Bartlett JA et al. HIV-1 RNA levels and the development of clinical disease. AIDS 1996;10:859-865. 20 Katzenstein D, Hammer S, Hughes M et al. The relation of virologic and clinical outcomes after nucleoside therapy in HIVinfected adults with 200 to 500 CD4 cells per cubic millimeter. N Engl J Med 1996;335:1091-1098. 21 Hughes MD, Johnson VA, Hirsch M et al. Monitoring plasma HIV-1 RNA levels in addition to CD4 + lymphocyte count improves assessment of antiretroviral therapeutic response. Ann Intern Med 1997;126:929-938. 22 O Brien WA, Hartigan PM, Daar ES et al. for the VA Cooperative Study Group on AIDS. Changes in plasma HIV RNA levels and CD4 + lymphocyte counts predict both response to antiretroviral therapy and therapeutic failure. Ann Intern Med 1997;126:939-945. 23 Saag M, Holodniy M, Kuritzkes DR et al. HIV viral load markers in clinical practice. Nat Med 1996;2:625-629. 24 Paxton WB, Coombs RW, McElrath MJ et al. Longitudinal analysis of quantitative virologic measures in human immunodeficiency virus-infected subjects with 400 CD4 lymphocytes: implications for applying measurements to individual patients. J Infect Dis 1997;195:247-254. 25 O Brien WA, Grovit-Ferbas K, Namazi A et al. Human immunodeficiency virus-type 1 replication can be increased in peripheral blood of seropositive patients after influenza immunization. Blood 1995;86:1082-1089. 26 Kempf D, Rode R, Xu Y et al. The durability of response to protease inhibitor therapy is predicted by viral load. Abstract 62 presented at International Workshop on HIV Drug Resistance, Treatment Strategies and Eradication. June 1997. 27 Merrill DP, Moonis M, Ting-Chao C et al. Lamivudine or stavudine in two and three drug combinations against human immunodeficiency virus type 1 or replication in vitro. J Infect Dis 1996;173:355-364. 28 Pollard R, Peterson D, Hardy D et al. Stavudine (D4T) and didanosine (ddi) combination therapy in HIV-infected subjects: antiviral effect and safety in an ongoing pilot randomized double-blinded trial. Presented at XIth International Conference on AIDS. Vancouver, B.C., Canada, 1996; Abstract ThB. 293. 29 Granneman G, Hsu A, Sun E et al. Pharmacokinetics/pharmacodynamics of ritonavir-saquinavir combination therapy. Presented at Fourth Conference on Retroviruses and Opportunistic Infections. Washington, DC, February 1997; abstract 609. 30 Kravcik S, Sahai J, Karr B et al. Nelfinavir mesylate (NFV) increases Saquinavir soft gel capsule (SQV - SGC) exposure in HIV+ patients. Presented at fourth Conference on Retroviruses and Opportunistic Infections. Washington, DC 1997; abstract 371. 31 Cameron W, Heath-Chiozzi M, Kravcik S et al. Prolongation of life and prevention of AIDS in advanced HIV immunodeficiency with ritonavir. Presented at the Third National Conference on Retroviruses and Opportunistic Infections. Washington, DC: February 1, 1996, abstract LB6a. 32 Hammer S, Squires K, Hughes M et al. A controlled trial of two nucleoside analogues plus indinavir in persons with human immunodeficiency virus infection and CD4 cell counts of 200 per cubic millimeter or less. N Engl J Med 1997;337:725-733. 33 Delta Coordinating Committee. Delta: a randomised double-blind controlled trial comparing combinations of zidovudine plus didanosine or zalcitabine with zidovudine alone in HIV-infected individuals. Lancet 1996;348:283-291. 34 Hammer S, Katzenstein D, Hughes M et al. A trial comparing nucleoside monotherapy with combination therapy in HIVinfected adults with CD4 cell counts from 200 to 500 per cubic millimeter. New Engl J Med 1996;335:1081-1090. 35 Carpenter C, Fischl M, Hammer S et al. Antiretroviral therapy for HIV infection in 1997: updated recommendations of the International AIDS Society-USA Panel. JAMA 1997;227:1962-1969. 36 Molla A, Kornevera M, Gao Q et al. Ordered accumulation of mutations in HIV-protease confers resistance to ritonavir. Nat Med 1996;2:760-766. 37 Erice A, Mayers D, Strike D et al. Brief report: primary infection with zidovudine-resistant human immunodeficiency virus type 1. New Engl J Med 1993;328:1163-1165. 38 Eron JJ, Benoit SL, Jemsek J et al. The safety and efficacy of lamivudine, zidovudine or both in the treatment of HIV-positive

118 Advances in HIV Therapy patients with CD4 + cell counts of 200-500 cells per cubic millimeter. N Engl J Med 1995;333:1662-1669. 39 Danner SA, Carr A, Leonard JM et al. A short-term study of the safety, pharmacokinetics, and efficacy of ritonavir, an inhibitor of HIV-1 protease. N Engl J Med 1995;333:1528-1533. 40 Markowitz M, Saag M, Powderly WG et al. A preliminary study of ritonavir, an inhibitor of HIV-1 protease, to treat HIV-1 infection. N Engl J Med 1995;333:1534-1539. 41 Chun T, Carruth L, Finzi D et al. Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature 1997;387:183-187. 42 Cavert W, Notermans D, Staskus K et al. Kinetics of response in lymphoid tissues to antiretroviral therapy of HIV-1 infection. Science 1997;276:960-964. 43 Gulick RM, Mellors J, Havlir D et al. Treatment with indinavir, zidovudine and lamivudine in adults with human immunodeficiency virus infection on prior antiretroviral therapy. N Engl J Med 1997;337:734-739. 44 Schapiro J, Winters M, Vierra M et al. Causes of long-term efficacy and/or drug failure in protease (PR) inhibitor monotherapy. Presented at XIth International Conference on AIDS. Vancouver, B.C., Canada, 1996; abstract Mo.B414. 45 Connors M, Kovacs J, Krevat S et al. HIV infection induces changes in CD4 + T-cell phenotype and depletions within the CD4 + T-cell repertoire that are not immediately restored by antiviral or immune-based therapies. Nat Med 1997;5:533-540. 46 Autran B, Carcelain G, Li TS et al. Positive effects of combined antiretroviral therapy on CD4 + T-cell homeostasis and function in advanced HIV disease. Science 1997;277:112-116. 47 Hicks C, Myers S, Giner J. Resolution of intractable molluscum contagiosum in a human immunodeficiency virus-infected patient after institution of antiretroviral therapy with ritonavir. Clin Infect Dis 1997;24:1023-1025.