Management of patients with antiretroviral treatment failure: guidelines comparison

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

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

Antiretroviral Treatment (ART) of Adult HIV Infection*

Clinical skills building - HIV drug resistance

Clinical support for reduced drug regimens. David A Cooper The University of New South Wales Sydney, Australia

New HIV EACS and Italian Guidelines

Management of NRTI Resistance

Management of Treatment-Experienced Patients: New Agents and Rescue Strategies. Joel E. Gallant, MD, MPH Johns Hopkins University School of Medicine

Somnuek Sungkanuparph, M.D.

Antiretroviral Treatment Strategies: Clinical Case Presentation

Comprehensive Guideline Summary

Introduction to HIV Drug Resistance. Kevin L. Ard, MD, MPH Massachusetts General Hospital Harvard Medical School

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

Updates to the HHS Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents Living with HIV Updated October 17, 2017

Rajesh T. Gandhi, M.D.

Switching ARV Regimens: Managing Toxicity and Improving Tolerability; Switches & Class-Sparing Approaches

DNA Genotyping in HIV Infection

Scottish Medicines Consortium

SA HIV Clinicians Society Adult ART guidelines

Continuing Education for Pharmacy Technicians

Principles of Antiretroviral Therapy

This graph displays the natural history of the HIV disease. During acute infection there is high levels of HIV RNA in plasma, and CD4 s counts

Tunisian recommendations on ART : process and results

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

Second-Line Therapy NORTHWEST AIDS EDUCATION AND TRAINING CENTER

Selected Issues in HIV Clinical Trials

Simplifying HIV Treatment Now and in the Future

Case Study. Dr Sarah Sasson Immunopathology Registrar. HIV, Immunology and Infectious Diseases Department and SydPath, St Vincent's Hospital.

What's new in the WHO ART guidelines How did markets react?

12th European AIDS Conference / EACS ARV Therapies and Therapeutic Strategies A CME Newsletter

HIV Treatment: New and Veteran Drugs Classes

Selected Issues in HIV Clinical Trials

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

PAEDIATRIC HIV INFECTION. Dr Ashendri Pillay Paediatric Infectious Diseases Specialist

Pediatric Antiretroviral Resistance Challenges

When to start: guidelines comparison

Case # 1. Case #1 (cont d)

Anumber of clinical trials have demonstrated

European Guidelines. for the Clinical Management and Treatment of HIV Infected Adults

BHIVA antiretroviral treatment guidelines 2015

Treatment strategies for the developing world

Crafting an ART Regimen for Initiation or Salvage: Are NRTI s Necessary?

Clinical Management of Resistance. AMJ Wensing, MD, PhD

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

2009 Revisions of WHO ART Guidelines. November 2009

HIV 101. Applications of Antiretroviral Therapy

The use of antiretroviral agents during pregnancy in Canada and compliance with North-American guidelines

Paediatric Infectious Diseases Unit, Red Cross War Memorial Children s Hospital & University of Cape Town

What s New. In The 2016 Perinatal HIV Treatment Guidelines? Provided by CDC s Elimination of Perinatal HIV Transmission Stakeholders Group

HIV and the Central Nervous System Impact of Drug Distribution Scott L. Letendre, MD. Professor of Medicine University of California, San Diego

Real Life Experience of Dolutegravir and Lamivudine Dual Therapy As a Switching Regimen in HIVTR Cohort

Pharmacological considerations on the use of ARVs in pregnancy

Persistent low level viraemia on third line ART

Reduced Drug Regimens

Criteria for Oral PrEP

Hepatitis B Case Studies

Resistance Workshop. 3rd European HIV Drug

Virological failure in children. Dr Lee Fairlie

British HIV Association Guidelines for the Management of Hepatitis Viruses in Adults Infected with HIV 2013 Appendix 2

Optimizing the treatment

ART and Prevention: What do we know?

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

Management of ART Failure. EACS Advanced HIV Course 2015 Dr Nicky Mackie

Didactic Series. Archive Genotype Resistance Testing in the Setting of Regimen Switching

Didactic Series. Update: 2012 HIV Treatment Guidelines. Daniel Lee, MD August 30, 2012

European AIDS Clinical Society (EACS) Guidelines for the Clinical Management and Treatment of HIV Infected Adults in Europe

Second and third line paediatric ART strategies

TB/HIV Co-Infection. Tuberculosis and HIV

HIV infection and Primary Care. HIV Care in /30/2013. It s not the AIDS of 85. Stephen Raffanti MD MPH Vanderbilt University School of Medicine

Friday afternoon Programme

Selecting an Initial Antiretroviral Therapy (ART) Regimen

Dr Marta Boffito Chelsea and Westminster Hospital, London

10 : 4. Introduction. R Sajithkumar, Kottayam. Mutations to select agents: Evolution:

Antiretroviral Therapy

Antiretroviral Therapy During Pregnancy and Delivery: 2015 Update

Pediatric HIV Infection and the Medical Management of Pregnant Women infected with HIV. Ernesto Parra, M.D., M.P.H.

Central Nervous System Penetration of ARVs: Does it Matter?

La terapia dell infezione da HIV: passato, presente e futuro.

Resistance Analyses of Integrase Strand Transfer Inhibitors within Phase 3 Clinical Trials of Treatment-Naive Patients

HIV Treatment: State of the Art 2013

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

ID Week 2016: HIV Update

WHAT S NEW IN THE 2015 PERINATAL HIV GUIDELINES?

Virological suppression and PIs. Diego Ripamonti Malattie Infettive - Bergamo

HIV Drugs and the HIV Lifecycle

HIV replication and selection of resistance: basic principles

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

Antiviral Therapy 2013; 18: (doi: /IMP2329)

Kiat Ruxrungtham. Professor of Medicine Chulalongkorn University, and HIV-NAT, Thai Red Cross AIDS Research Centre

Differentiating emtricitabine (FTC) from lamivudine (3TC): what a fine-tuning of antiretroviral therapy might entail

Clinical Commissioning Policy: Use of cobicistat (Tybost ) as a booster in treatment of HIV positive adults and adolescents

Frailty and age are independently associated with patterns of HIV antiretroviral use in a clinical setting. Giovanni Guaraldi

ANTIRETROVIRAL THERAPY

Dr Alan Winston. Imperial College Healthcare NHS Trust London. 7-8 October 2010, Queen Elizabeth II Conference Centre, London.

How to best manage HIV patient?

James Nuttall Paediatric Infectious Diseases Unit Red Cross Children s Hospital

Pediatric HIV Update NORTHWEST AIDS EDUCATION AND TRAINING CENTER

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

Update on Antiretroviral Treatment for HIV Infection 2008

Third Agent Advantages Disadvantages. Component Tenofovir/emtricitabine (TDF/FTC) 300/200 mg (coformulated with EFV as Atripla) 1 tab once daily

Transcription:

The editorial staff Management of patients with antiretroviral treatment failure: guidelines comparison A change of therapy should be considered for patients if they experience sustained rebound in viral load levels, having either had previously undetectable levels or never achieved undetectable levels on their current treatment regimen after 24 36 weeks. The likelihood of achieving an undetectable viral load on changing therapy is predicted by the number of active drugs in the new regimen, plus factors influencing tolerability and adherence. The decision to change therapy should be guided by the availability of a treatment option that is likely to have the potency to suppress viral load to undetectable levels (<50 copies/ml) and which the patient is likely to be able to adhere to and tolerate. On BHIVA 2008 Guidelines opinion, the choice of a new regimen should be guided by the results of current and previous resistance testing, treatment history and the ability of the patient to adhere to and tolerate individual drugs. Resistance testing is important to identify which drugs will possibly be of most benefit, i.e. active. A drug is defined as active where it is likely to have significant antiviral activity in vivo based on antiretroviral treatment and virological failure histories and the results of all current and previous resistance testing. There are comparative data assessing which ritona vir boosted PI regimen is more effective in PI-experienced patients with or without detectable PI mutations at baseline and further data will be available from ongoing trials. Similar virological efficacy at 48 weeks has been demonstrated for lopinavir/r and atazanavir/r in patients who have previously failed at least two regimens including at least one containing a PI. Gastrointestinal side effects and hyperlipidaemia were more common with lopinavir/r. Hyperbilirubinaemia and, in a small number of patients, clinical jaundice were the most common side effects with atazanavir/r. In treatment-experienced patients, of whom 69% were PI- and 76% NNRTI-experienced and all of whom were lopinavir/r naıve, treatment with darunavir/r twice daily had superior virological efficacy at 48 weeks compared with lopinavir/r. However, in secondary analyses, similar virological efficacy was seen in patients who had no detectable primary PI mutations at baseline and in those who had two or more active agents in the optimized background therapy. In patients who had previously experienced treatment failure on one or two PIs, non inferiority of fosamprenavir/r compared with lopinavir/r using the primary endpoint of time-averaged change in viral load from baseline could not be established. However, similar proportions of patients achieved viral load <50 copies/ml at 48 weeks with lopinavir/r and twice-daily fosamprenavir/r, but not with once-daily fosamprenavir/r. Once-daily fosamprenavir/r is not recommended in patients with previous PI failure. In EACS 2009 Guidelines, the definition of virological failure is a confirmed plasma HIV RNA > 50 copies/ ml 6 months after starting therapy (initiation or modification) in patients that remain on ART. General measures are: Evaluate adherence, compliance, tolerability, drugdrug interactions, drug-food interactions, psychosocial issues Perform resistance testing on failing therapy (usually reliable with plasma HIV RNA levels >500-1000 copies/ml) and obtain historical resistance testing for archived mutations Consider TDM Review antiretroviral history Identify treatment options, active, potentially active drugs/combination If plasma HIV RNA > 50 and <500-1000 copies/ml Check for adherence Check plasma HIV RNA 1 to 2 months later Improve boosted PI s PK (if applicable) 299

300 If plasma HIV RNA confirmed > 500/1000 copies/ml, change regimen as soon as possible: what to change will depend on the resistance testing results: No resistance mutations found: re-check for adherence, perform TDM Resistance mutations found: switch to a suppressive regimen based on drug history; multidisciplinary experts discussion advised Goal of new regimen: plasma HIV RNA < 400 c/ ml after 3 months, plasma HIV RNA < 50 c/ml after 6 months In case of resistance mutations demonstrated, general recommendations are: Use 2 or preferably 3 active drugs in the new regimen (including active drugs from previously used classes) Any regimen should use at least 1 fully active PI/r (e.g. darunavir/r) plus 1 drug from a class not used previously e.g. fusion, integrase or CCR inhibitor (if tropism test shows R5 virus only), or 1 NNRTI (e.g. etravirine), assessed by genotypic testing Defer change if < 2 active drugs available, based on resistance data, except in patients with low CD4 count (<100/mm3) or with high risk of clinical deterioration for whom the goal is the preservation of immune function through partial reduction of plasma HIV RNA (> 1 log reduction) by recycling. If limited options, consider experimental and new mechanistic drugs, favouring clinical trials (but avoid functional monotherapy) Treatment interruption is not recommended Optimisation of new regimen is: If demonstrated NRTI multiple resistance, avoid NRTI but Consider continuation of 3TC or FTC even if documented resistance mutation (M184V/I) Select 1 active ritonavir-boosted PI. If at all possible avoid double boosted PIs Etravirine potentially active in selected NNRTImutation profiles Always check for drug-drug-interactions, and when necessary perform TDM of drugs of new regimen if available If many options are available, criteria of preferred choice include: simplicity of the regimen, toxicity risks evaluation, drug-drug-interactions, future salvage therapy. On 2009 DHHS Guidelines, recommendations are: In treatment-experienced patients with suppressed viremia, assess adherence frequently and simplify the regimen when feasible. Change individual antiretroviral drugs to reduce or man-

age toxicity, as needed. Evaluation of antiretroviral treatment failure in a patient should include an assessment of the severity of HIV disease of the patient; the antiretroviral treatment history, including the duration, drugs used, antiretroviral potency and response, adherence history, and drug intolerance/toxicity; the use of concomitant medica- 301

302 tions with consideration of adverse drug interactions with antiretrovirals; HIV RNA and CD4 T-cell count trends over time; and the results of prior drug resistance testing. Optimal virologic response to treatment is maximal virologic suppression (e.g., HIV RNA level <400 copies/ml after 24 weeks, <50 copies/ml after 48 weeks). Persistent low-level viremia (e.g., HIV RNA 50 200 copies/ml) does not necessarily indicate virologic failure or a reason to change treatment. Drug resistance testing should be obtained (AI) while the patient is taking the failing antiretroviral regimen (or, if not possible, within 4 weeks of treatment discontinuation). The goal of treatment for patients with prior drug exposure and drug resistance is to re-establish maximal virologic suppression, with HIV RNA suppressed to below the limit of detection of a sensitive assay (e.g., <50 copies/ml) (AI). The patient s treatment history and the past and current resistance test results should be used to identify fully active agents to design a new regimen (AII). A fully active agent is one that is likely to have antiretroviral activity on the basis of the patient s treatment history, susceptibility on drug resistance testing, and mechanistic class. Assessing and managing a patient who has antiretroviral experience, who exhibits drug resistance, and who is experiencing treatment failure is complex and expert advice is critical and should be sought. Adding at least two (preferably three) fully active agents to an optimized background antiretroviral regimen can provide significant antiretroviral activity (AII). Immunologic failure can be defined as a failure to achieve and maintain an adequate CD4 response despite virologic suppression. For immunologic failure, current medications, untreated coinfection, and serious medical conditions should be assessed. There is no consensus for when and how to treat immunologic failure. The immunomodulator interleukin-2 has not demonstrated clinical

benefits in randomized trials and is not recommended (AI). For some highly treatment experienced patients, maximal virologic suppression is not possible. In this case, antiretroviral therapy should be continued with regimens designed to minimize toxicity, preserve CD4 cell counts, and avoid clinical progression. In this scenario, expert advice is essential and should be sought. In WHO 2009 guidelines, recommendations are: 1. Where available, use viral load (VL) to confirm treatment failure. 2. Where routinely available, use VL every 6 months to detect viral replication. 3. A persistent VL above 5 000 copies/ml confirms treatment failure. 4. When VL is not available, use immunological criteria to confirm clinical failure. In developing these recommendations, the panel was concerned by the limitations of clinical and immunological monitoring for diagnosing treatment failure, and placed high value on avoiding premature or unnecessary switching to expensive second line ART. They also valued the need to optimize the use of virological monitoring and ensure adherence. Based on the pooled analysis of the size effects from two randomized trials (HBAC and DART trials), clinical monitoring alone (compared to combined immunological and clinical monitoring or to combined virological, immunological, and clinical monitoring) resulted in increases in mortality, disease progression and unnecessary switches, but no difference in serious adverse events. However, in one of these trials (HBAC trial), combined immunological and clinical monitoring was compared to combined virological, immunological, and clinical monitoring, and resulted in no difference in mortality, disease progression, unnecessary switches or virological treatment failures. 1. A boosted protease inhibitor (PI/r) plus two nucleoside analogues (NRTIs) are recommended for second-line ART. 2. ATV/r and LPV/r are the preferred boosted PI s for secondline ART. 3. Simplification of second NRTI options is recommended. If d4t or AZT has been used in first-line, use TDF + 3TC or FTC as the NRTI backbone in secondline. If TDF has been used in first-line, use AZT + 3TC as the NRTI backbone in second-line. In developing these recommendations, the panel placed high value on using simpler second-line regimens and the availability of heat-stable, fixed-dose combinations. On the question of whether PI monotherapy could be used as secondline ART, there is moderate quality of evidence from nine RCTs and individual study reports showing less virological suppression and higher rates of viral rebound for PI monotherapy compared to standard triple ART regimens. The panel concluded that an NRTI backbone should be maintained. On the question of which bpi to use in second-line therapy, there is moderate quality of evidence that ATV/r is non-inferior to LPV/r (in combination with TDF and an optimized second NRTI) in treatment experienced patients. Non-serious adverse events varied by boosted PI with no significant difference in serious adverse events. On the question of which NRTI backbone to use in second-line, few studies of relevance were identified. The recommended NRTI sequencing is based on likely resist- 303

304 ance mutations and potential for retained antiviral activity. If AZT+3TC are used in first-line with sensitive monitoring and early switching, the NRTIs with remaining activity are TDF and ddi, (both very likely) and ABC (likely). In the scenario of insensitive monitoring and late switching, TDF and ddi activity are less likely, with activity of ABC unlikely. If TDF+3TC are use in first-line, with early switching, the NRTIs with remaining activity are AZT and d4t (very likely), ddi, ABC (possible). In the scenario of late switching, activity of AZT and d4t is very likely, with activity of ddi and ABC unlikely. Retained activity of 3TC is likely in the early switching scenario and less likely in the case of late switching. AZT+3TC, TDF+FTC and TDF+3TC are available as fixed-dose combinations. 1. National programmes should develop policies for third-line therapy that consider funding, sustainability and the provision of equitable access to ART. 2. Third-line regimens should include new drugs likely to have anti HIV activity such as integrase inhibitors and second generation NNRTIs and PIs. 3. Patients on a failing second-line regimen with no new ARV options, should continue with a tolerated regimen. The panel were concerned by unpublished cohort reports of high mortality among patients failing second-line therapy, but placed high value on balancing the need to develop policies for third-line therapy whilst maintaining increased access to first-line therapy. It was recognized that many counties have financial constraints that might limit the adoption of third-line regimens. From a targeted literature review of relevant studies, the evidence is limited with few studies of newer agents in resource-limited settings. Data from RCTs predominantly in developed countries are available for boosted darunavir, etravirine and raltegravir. Taken together, these data support the efficacy of these agents in highly ART experienced patients. There was no uncertainty among the panel concerning the need for third-line regimens. However, there was uncertainty about how making third-line regimens available would impact the provision of first-line ART. There was also uncertainty about what third-line drugs should be provided and many studies are ongoing. On 2010 IAS opinion, the virologic goal of treatment for first and multiple-regimen failure is to achieve a plasma HIV-1 RNA level below the limit of detection of the most sensitive assays available. With the availability of new drugs and regimens, this goal now is achievable, even in most patients with multi regimen failure. Reasons for viral rebound after complete suppression, such as poor adherence, drug-drug interactions, concurrent infections, and recent vaccinations, should be considered before the regimen is changed. Testing for an isolated detectable viral load should be repeated to exclude measurement error or self-resolving low level viremia. Stage of HIV disease, nadir and current CD4 cell count, comorbidities, treatment history, current and previous drug resistance tests, and concomitant medications with potential for interactions should be considered when the new regimen is designed. Ideally 3, but at least 2, fully active drugs should be included and drugs from new classes should be considered. The toxicities of stavudine, didanosine, and to a lesser extent zidovudine make their use problematic, and they should be used only when options are limited. Once failure has been confirmed, an NNRTIcontaining regimen should be discontinued as soon as possible to minimize the selection of additional mutations. Initial NNRTI failures traditionally have been treated with 2 active NRTIs plus a PI/r, but raltegravir, maraviroc, and etravirine now provide additional options. According to potency and high genetic barrier, the inclusion of a PI/r should be considered whenever possible, but when not possible, an agent from a new class should be considered. Treatment experienced patients receiving etravirine and darunavir/r plus an optimized background regimen had better virologic responses than those receiving placebo plus background regimen, with comparable tolerability at 48 weeks. Resistance to the PI/r component does not always emerge when regimen failure is detected, allowing the same drug or another in the PI class to be used in the next regimen. For early failures, strategic sequencing of PIs should be considered. If some degree of PI resistance exists, darunavir/r is likely to be preferred over lopinavir/r or tipranavir/r because of its superior tolerability and toxicity profile, as well as problematic drug interactions associated with tipranavir/r. If not previously used, an NNRTI may be included, provided that potential drug interactions are considered. Whenever possible, a new antiretroviral regimen should contain at least 2 fully active drugs. In a setting of a multidrug (including PI and NNRTI) resistance, 3 active drugs, including new classes of agents (integrase strand transfer inhibitors or entry inhibitors), should be used. Individuals with multidrug-resistant virus usually benefit from a PI/r with activity against resistant strains, such as darunavir/r or tipranavir/r. Etravirine can be paired with darunavir/r (but not tipranavir/r) and may be of value, depending on the number of NNRTI mutations present. Enfuvirtide may be an option if no other new class can be used, despite the inconvenience of subcutaneous injection and injection site reactions. Dualboosted PIs are not recommended. Lamivudine or emtricitabine is sometimes included to maintain the M184V mutation and decrease viral fitness, but there is no new evidence to support this approach. Another theoretically beneficial strategy is to use zidovudine to prevent the emergence of the K65R mutation in the presence of thymidine analogue mutations when using tenofovir in patients in whom nrti-containing regimens are failing. However, no clinical benefit has been shown for this approach. Maintenance of regimen potency is the objective when switching ART regimens. Virologic failure of an initial regimen (confirmed measurable viremia) should be identified and treated as early as possible with at least 2 fully active drugs (AIa) to avoid the accumulation of resistance mutations. For NNRTI failures, the new combination usually should include a PI/r or an agent from a new class (AIa) if a PI/r is not possible. Etravirine may be a useful component of a new regimen for NNRTI failure but must be supported by a potent combination including a PI/r (AIa).

305

306 Depending on the resistance profile and options available, inclusion of agents from new drug classes (raltegravir or maraviroc) should be considered (BIIb). Monotherapy with a PI/r should be avoided unless other drugs cannot be considered for reasons of toxicity/tolerability (AIa). Design of a new regimen should consider previous drug exposure, previous resistance profile, drug interactions, and history of intolerance/toxicity (CIII). Treatment interruptions should be avoided, except in the context of controlled clinical trials (AIa). Elective treatment interruptions should consider the different half-lives of the regimen components, with stopping the drugs in a staggered manner when an NNRTI is a component (CIII).