Abstract
Antibodies that naturally develop in some individuals infected with human immunodeficiency virus 1 (HIV-1) and are capable of broadly neutralizing diverse strains of HIV-1 are useful for two applications: they can inform the rational design of vaccine immunogens, and they may be capable of preventing and treating HIV-1 infection when administered passively. A phase IIb study has been initiated with the experimental broadly neutralizing antibody (bnAb) VRC01, which has considerable breadth and potency (also referred to as a phase IIb HVTN 703/HPTN 081 and HVTN 704/HPTN 085 AMP efficacy trials) to evaluate its protective efficacy in individuals at risk of HIV acquisition. bnAbs prevent HIV-1 infection by selectively targeting vulnerable sites on the viral envelope (Env) protein that facilitates the entry of HIV. Although in very early stages, bnAbs capable of neutralizing a broad range of inter- and intraclade HIV-1 isolates have been demonstrated to have potential in treating patients either alone or in combination with antiretroviral drug therapy (cART); however, they are proposed to be advantageous over the latter as far as durability and side effects are concerned. Recent studies have indicated that combination therapy of potent bnAbs along with latency-reversing agents (LRAs) might also target latent reservoirs of HIV and kill them by recruiting effector cells, such as natural killer cells, thus confirming clinical progression. Possession of such qualities makes these new-generation potent bnAbs extremely valuable in effectively complementing the shortcomings of current ART drugs and improving the quality of life of infected individuals.
Keywords: broadly neutralizing antibodies, envelope, HIV-1, treatment
Introduction
Communicable diseases such as acquired immunodeficiency syndrome (AIDS), tuberculosis, malaria and neglected tropical diseases have been the scourge of millions of people in south-east Asia, home to a quarter of the world’s population. Despite our greater understanding of how human immunodeficiency virus 1 (HIV-1) manipulates host machinery in order to favor its own replication, leading to pathogenesis and transmission, a functional cure or protective vaccine remains elusive. Antiretroviral therapy (ART) has indeed significantly brought down the incidence of infection and improved the quality of life of those infected; however, ART possesses limited or no potential to interfere with the viral reservoir, and viral rebound has demonstrated association with treatment interruptions.1,2
HIV-1 continues to be a major global health problem, with more than 35 million individuals (including about 1.8 million children) living with HIV infection in 2016 [as per the United Nations Program of HIV/AIDS (UNAIDS) data: http://www.unaids.org], with a global HIV prevalence of 0.8% among adults in 2015. Although the advent of highly active ART significantly has helped in reducing the incidence and improving the quality of life of HIV-infected individuals, both lack of adherence and poor access to ART are believed to be associated with the continued disease burden. It is estimated that approximately 46% of adults living with HIV had access to ART in 2015 (as per UNAIDS data). Despite its effectiveness, patients infected with HIV need to adhere to ART for life, as it does not eradicate infection. This is because ART drugs primarily target circulating viruses and are largely unable to access those that remain dormant in viral reservoirs as provirus. Hence, although current ART is essential in reduction of disease incidence, it alone is not sufficient in bringing comprehensive reduction in HIV incidence and functional cure. Recent discoveries of a novel class of highly potent and broadly neutralizing HIV-1-specific monoclonal antibodies, by way of single-memory B-cell (obtained from elite neutralizers) cloning technology, have provided new hope in treating patients. Some of these potent broadly neutralizing antibodies (bnAbs) have entered into clinical trials4–8 and few more (either singly or in combinations) are currently being considered to enter into human clinical trials for assessing their efficacy and other clinical evaluations.8 They are currently being investigated for their possible role in compensating the shortcomings of ART drugs. In the present review, we highlight advancements in knowledge of the potential of bnAb-mediated protection and treatment.
What are bnAbs and why are they considered important in disease prevention?
Neutralizing antibodies are specific types of antibodies that have the ability to act on the specific sites on microorganisms such as viruses that aid in establishing an infection. In bacterial infections, antibodies are known for neutralizing toxins, enabling opsonization and facilitating complement-mediated lysis; in viral infections, antibodies interrupt transmission primarily in two ways: by blocking virus entry into the uninfected target cells (by way of disengaging virus–receptor interaction) and via antibody-dependent cell-mediated cytotoxicity (ADCC). While neutralizing antibodies are generally effective in tackling type-specific microbes with limited or no genetic variations, for complex viruses such as HIV-1 and influenza (which display considerable variations in their genotypes), ‘very special’ antibodies capable of efficiently tackling the breadth of these genetic variations are essential. These ‘special antibodies’ are referred to as bnAbs. Since neutralizing antibodies exclusively target the viral surface protein that enables virus entry (the first step in establishing infection), bnAbs are being considered as the ‘blueprint’ for preventive vaccine design, especially for HIV-1 whose genotype varies considerably.9,10 During the course of natural infection, while individuals infected with HIV-1 typically develop type-specific cross-neutralizing antibodies,11 only about 1% of them are found to develop broad and very potent neutralizing antibodies (bnAbs)12 that can neutralize a wide range of genetically diverse HIV-1 subtypes. Although the precise mechanism by which this rare fraction of chronically infected individuals develop bnAbs in the natural course of infection has remained elusive, recent structural, virological and immunological studies have provided strong evidence of how virus–antibody coevolutions in the natural disease course are synergistically associated with sequential development of potent and broad neutralizing antibodies via somatic hypermutations and maturation of antibody genes.13 The extent of suppression of viremia in chronically infected patients by bnAbs is not very clear; nonetheless, because of their ability to directly act on the viral envelope (Env) protein and thus prevent HIV-1 entry, bnAbs hold potential in treating infected patients (through ‘passive’ immunizations) in addition to ART currently given.14 Additionally, since bnAbs can also latch onto the viral Env expressed on the infected cells, they could potentially facilitate fragment crystallizable (Fc)-mediated clearance.15 Although initiation of serum-based immune therapy for treating infectious diseases goes back to the late 1800s,16 as well as the considerable success of monoclonal antibody (mAb)-based cancer therapy in recent times,17 there has been a huge gap in its application in treating infectious diseases.18 In the next section, evidence is highlighted regarding advances made toward understanding the therapeutic potential and nature of applications that bnAbs can offer.
Evidence of bnAb-mediated protection and treatment against HIV-1 and simian immunodeficiency virus (SIV)
Antibodies have been historically used in prophylaxis and treatment of several bacterial and virus-induced infectious diseases,19 and have had success in diverse infections [such as respiratory infection-causing bacteria like Streptococcus pneumoniae, Neisseria meningitidis, Haemophilus influenzae, tetanus, Clostridium botulinum, herpesvirus (cytomagalovirus and varicella zoster virus), enterovirus infection in adults, and vaccinia].19 In fact, the first licensed monoclonal antibody (mAb), palivizumab (licensed in the USA in 1998), was found to prevent infection in infants who were at risk of contracting respiratory syncytial virus infection.20 In early studies with passive immunotherapies, patient sera provided evidence of improved clinical outcomes associated with reduction in plasma viral ribonucleic acid load and delayed appearance of AIDS-defining illnesses.21,22 In contrast to the polyclonal serum samples, due to their homogenous composition, mAbs have higher specific activity than that of serum antibodies and hence provide greater therapeutic efficacy.3 The first-generation bnAbs to HIV-1 such as b12, 2F5, 4E10, 2G12 and KD-247 were used to assess the extent to which they could reduce viremia in infected patients by passive infusion.23–28 Although a transient decrease in plasma viral load and delayed viral rebound was observed post mAb transfusion, frequent virus escape due to emergence of resistance mutations against these mAbs was observed.25,29,30 Isolation of potent bnAbs from elite neutralizers has not only highlighted key vulnerable sites on the viral (HIV-1) Env that the virus uses to evolve in the natural course of infection, but also makes them amenable to effective use in designing improved immunogens via reverse-vaccinology approaches31 and treatment through passive immunization.32,33 Some of these potent bnAbs such as VRC01, 3BNC117 and PGT121 have shown promising results in animal models, as well as in humans, and have shown modest and transient suppression of viremia,14 thus opening a very interesting and important avenue of exploring how bnAbs can be successfully used as therapeutic agents to complement ART in comprehensive treatment of HIV-infected patients.
Significance of baseline viremia on the degree of efficacy of passively infused bnAbs
When passively administered to patients, can bnAbs be more efficacious when circulating viral load is low? Recently, two very potent and broad mAbs that target CD4 binding sites (CD4bs) on the viral Env, 3BNC117 and VRC01, were assessed for their degree of efficacy when administered to HIV-infected patients with low viremia. In a phase II open-label trial, when administered to patients with analytical ART treatment interruption (ATI), 3BNC117 mAb was found to be associated with a significant delay in viral rebound compared with what was found during ART alone.5 Comparable evidence of delayed viral rebound due to virus escape was also found in ATI patients infused with another CD4bs-targeting bnAb, VRC01 (National Institutes of Health [NIH] 15-I-0140 and AIDS Clinical Trials Group [ACTG] A5340 trials);7 however, unlike in the case of the 3BNC117 efficacy trial, profound delay was not observed. Taken together, both these studies highlighted how delayed viral rebound during monotherapy is dependent on low viral load set point in ART-experienced patients who do not harbor pre-existing viruses resistant to the bnAb used for therapy. Nonetheless, combination therapy with a cocktail of bnAbs with distinct specificities on the viral Env holds better promise than monotherapy with regard to minimizing viral escape and rebound.5,7,34
Ability of bnAbs to interfere with latent viral reservoir and clearance
While ART has been shown to significantly improve the quality of life of infected individuals, it largely lacks the ability to directly modulate latently infected cells and hence is, in general, ineffective in curing persistent HIV infection.35,36 Although it has not yet been ascertained whether bnAbs either alone or in combination can significantly limit the size and stability of the latent virus reservoirs, a few studies have provided evidence that bnAbs could possibly act on latently infected cells. For example, bnAbs like VRC01 and PGT121 have been demonstrated in vitro to block HIV-1 replication in activated cells that form the viral reservoir.14 Moreover, while 3BNC117 was found to possess the potential to kill latently infected cells, PGT121 administered passively to nonhuman primates has demonstrated association with significantly depleted proviral DNA.37 Because of their ability to directly bind to viral Env expressed on infected cells, potent bnAbs are thought to be able to latch onto HIV-1 Env expressed on activated cells harboring proviral DNA, which will have dual implications: enable killing of latently infected cells by recruitment of natural killer (NK) cells14 and prevent further forming of latently infected cells by neutralizing HIV-1 released from activated reservoirs. Bruel and colleagues38 recently demonstrated Fc-region-mediated killing of HIV-infected lymphocytes via ADCC activity and engagement of NK cells, mediated by some of the bnAbs that have the ability to limit cell-to-cell HIV-1 transmission.1 While a number of approaches have been proposed toward reactivation of latently infected cells by applying agents that act on cellular transcription factors and host cellular activation signals such as protein kinase-C (PKC), toll-like receptor (TLR) and Mitogen-activated protein kinases (MAPK) agonists, latency-reversing agents (LRAs), and checkpoint blockers, the use of LRAs along with bnAbs for greater virus suppression has gained special interest.39,40 Although no LRA has been reported that can very effectively act on cells latently infected with HIV-1, it has been proposed41,42 that LRAs such as histone deacetylase inhibitors (HDACi) and TLR antagonists (e.g. vorinostat, romidepsin and panobinostat), TLR7 agonists (e.g. GS-9620) and other viral inducers such as I-BET151 and anti-CTLA-4 may work to limit the size and functionality of the latently infected cells when administered with potent bnAbs. Indeed, in experimental humanized mice, in conjunction with different LRAs described above, bnAbs 3BNC117, 10-1074 and PG16 led to beyond-detectable cell-associated proviral load.14,43 Taken together, along with effective LRAs and ART, bnAbs could significantly help to limit the frequency of plasma viral rebound by reducing persistent HIV reservoirs.44,45
bnAbs that block cell-associated HIV-1 transmission act as a ‘double-edged sword’
Typically, the prevention of cellular entry by bnAbs is tested on cell-free viruses; however, cell–cell spread is considered to be a more robust mechanism of HIV-1 spread and transmission.1 Therefore, identifying bnAbs capable of also inhibiting cell–cell transmission would theoretically provide better clinical outcomes during therapy. bnAbs such as those targeting CD4bs (NIH45-46 and 3BNC60) and V3-glycan (10-1074 and PGT121) have been reported possessing properties to neutralize both cell-free and cell-associated viruses1 by acting on the virological synapse that forms when an infected cell contacts an uninfected permissive cell (such as a CD4+ T cell).46,47 bnAbs with the ability to interfere with both cell-free and cell-associated virus transmissions are generally believed to provide better treatment efficacy over the ones that are only effective in neutralizing cell-free viruses. This was perhaps the reason why bnAbs such as 10E8 and 3BC16 that effectively neutralize the majority of cell-free viruses were found to be ineffective in suppressing viremia in vivo,1,34 while dually effective 10-1074 and NIH 45-46 mAbs were effective in modestly suppressing viremia in vivo.34 Although the impact of neutralizing antibodies on the macrophage–T-cell-mediated virological synapse have been documented,48 how neutralizing antibodies act on dendritic cell-mediated cell–cell transmission remains elusive (dendritic cells play a very important role in initial infection and viral transfer).49 Taken together, potent bnAbs capable of preventing cell-free and cell–cell virus transmission are likely to exert better clinical outcomes when used in passive therapy and would be effective in tissues that support HIV infection such as gut-associated lymphoid tissue and lymph nodes.1,50–52
Modification of bnAb arms and implications for improving therapeutic efficacy
The discovery of potent bnAbs has also paved the way for their further modification in order to enhance their utility in vivo for better clinical outcomes when used in therapy. Modifications that prolong half-life, and increase potency, Fc receptor (FcR) binding and polyfunctionality are thought to circumvent the shortcomings of bnAbs when used as whole molecule, and provide durable and long-lasting protection during passive immunotherapy.53,54 A number of studies have demonstrated how modifications of the neonatal FcR (FcRn) can help maintain immunoglobulin (IgG) homeostasis and prolong the serum half-life of IgG in humans.55 Such modifications are also believed to enhance mucosal localization, efficient immune protection, and durability of bnAbs during passive immunotherapy compared with their wild-type counterparts.56 Selective modification of fragment antigen-binding regions (Fabs) of bnAbs has also been demonstrated to considerably enhance potency and breadth of bnAbs. In recent times, bispecific and trispecific bnAbs have gained special attention for their ability to comprehensively arrest virus infection by synchronously latching onto distinct viral and cellular targets that support HIV-1 entry. Very recently, considerable breath and potency of single antibody molecules engineered to possess three distinct specificities to HIV-1 Env have been reported.57 The investigators demonstrated that the combination of three bnAbs with specificities to CD4bs (VRC01, N6), V1V2 (PGDM1400) and membrane proximal external region (MPER) (10E8v4) into one single molecule conferred very potent and broad neutralization in a nonhuman primate model, beyond what each of these individual bnAbs could achieve on their own. In addition, recent reports highlighted the potential of bispecific antibodies with dual specificities to provide considerable potency and neutralization breadth.54,58–60 In one study, the antibodies were engineered such that one arm of the Fab binds to the cellular receptor (e.g. CD4 or CCR5) and the other Fab arm binds to epitopes on viral Env that are targeted by bnAbs. The second group59 constructed a bispecific antibody by putting together the Fabs of two different potent bnAbs with distinct epitope specificities on viral gp120 with a goal to crosslink gp120 domains within a trimer spike. Both the approaches were reported to provide far better virus neutralization activity via their synergistic action over parent antibodies, which suggested the additive effect exerted due to antibody engineering. Antibody engineering has also been suggested as a means to effectively target the latently infected viral reservoir.14 The idea is to engineer antibodies in such a way that they can dually act on the virus as well as the activated cells that harbor HIV-1 in a dormant state. Two classes of such antibody molecules have been investigated: bispecific T-cell engagers and dual-affinity retargeting molecules. Because latently infected cells such as resting CD4+ T cells are not accessed by ART or most antibodies to HIV-1, upon activation, they are believed to express viral Env on the cell surface, enabling bnAbs to latch onto them and facilitate the recruitment of cytotoxic effector cells to kill by ADCC.61 Rational engineering of bnAbs makes them an attractive prospect, by providing additional effector functions beyond virus neutralization and prolonging virus suppression and eradication of viral reservoirs.
Summary
With the advent of single-memory B-cell cloning technology, scientists have been able to discover a number of very potent and broadly neutralizing human monoclonal antibodies from infected individuals. These bnAbs not only are aiding the design of improved immunogens but could also be useful in treating individuals infected with HIV-1. In sharp contrast to the drug regimens that are currently being used as ART, bnAbs have the potential to access latently infected activated T cells in addition to cell-free viruses in the circulation, with help from LRAs. Clinical trials with new-generation promising potent bnAbs are underway to assess their effectiveness in comprehensive treatment, along with their safety, durability and tolerability properties, which should be able to significantly reduce disease burden. Moreover, for effective control of HIV-1 replication, which shows extraordinary diversity in the clinical setting, combination therapy using a cocktail of bnAbs with multiple specificities would concomitantly target distinct and discontinuous epitopes on the viral Env, thereby significantly impeding viral escape and rebound.34,37,62 Finally, while discovery of promising bnAbs to HIV-1 with potential in treating patients are encouraging, there are two areas that need particular attention prior to taking promising bnAbs for clinical development. First, the bnAbs may not necessarily provide comparable breadth and potency toward neutralizing HIV-1 in vivo to that observed under in vitro conditions, as demonstrated recently.63,64 Hence, it will be important to assess the extent (both potency and breadth) to which the promising bnAbs (either as unmodified or engineered forms) neutralize HIV-1 grown in physiologically relevant cells, for example, peripheral blood mononuclear cells (or CD4+ T cells). In addition, it is important to assess the fitness cost of the bnAbs associated with emergence of escape variants of HIV-1.4,64,65 For example, some bnAbs, if given as monotherapy, may select for early escape variants over others that would impact on treatment progress. Understanding such phenomena will help to choose the bnAb combinations that would overcome this problem and provide better treatment outcome. Secondly, it is important to periodically monitor circulating HIV-1 strains across populations and geographical boundaries to assess the breadth, potency and suitability of promising bnAbs.66,67 Typically, viral Envs routinely used to assess the breadth and potency of bnAbs represent viruses which were either isolated in the past or which do not necessarily represent what is currently circulating. This may impact on the bnAb efficacy in clinical trials. In summary, advances made in the discovery and effective antiviral application of bnAbs in HIV-infected patients will provide guidance in developing mAb-based therapies for other emerging and re-emerging pathogens.
Footnotes
Funding: This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
Conflict of interest statement: The authors declare that there is no conflict of interest.
Contributor Information
Rajesh Kumar, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Haryana, India.
Huma Qureshi, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Haryana, India.
Suprit Deshpande, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Haryana, India.
Jayanta Bhattacharya, HIV Vaccine Translational Research Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana-121001, India.
References
- 1. Malbec M, Porrot F, Rua R, et al. Broadly neutralizing antibodies that inhibit HIV-1 cell to cell transmission. J Exp Med 2013; 210: 2813–2821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Cihlar T, Fordyce M. Current status and prospects of HIV treatment. Curr Opin Virol 2016; 18: 50–56. [DOI] [PubMed] [Google Scholar]
- 3. Casadevall A. Antibody-based therapies for emerging infectious diseases. Emerg Infect Dis 1996; 2: 200–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Caskey M, Klein F, Lorenzi JC, et al. Viraemia suppressed in HIV-1-infected humans by broadly neutralizing antibody 3BNC117. Nature 2015; 522: 487–491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Scheid JF, Horwitz JA, Bar-On Y, et al. HIV-1 antibody 3BNC117 suppresses viral rebound in humans during treatment interruption. Nature 2016; 53: 556–560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Ledgerwood JE, Coates EE, Yamshchikov G, et al. Safety, pharmacokinetics and neutralization of the broadly neutralizing HIV-1 human monoclonal antibody VRC01 in healthy adults. Clin Exp Immunol 2015; 182: 289–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Bar KJ, Sneller MC, Harrison LJ, et al. Effect of HIV antibody VRC01 on viral rebound after treatment interruption. N Engl J Med 2016; 375: 2037–2050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Cohen YZ, Caskey M. Broadly neutralizing antibodies for treatment and prevention of HIV-1 infection. Curr Opin HIV AIDS 2018; 13: 366–373. [DOI] [PubMed] [Google Scholar]
- 9. Rusert P, Kouyos RD, Kadelka C, et al. Determinants of HIV-1 broadly neutralizing antibody induction. Nat Med 2016; 22: 1260–1267. [DOI] [PubMed] [Google Scholar]
- 10. Burton DR, Ahmed R, Barouch DH, et al. A blueprint for HIV vaccine discovery. Cell Host Microbe. 2012; 12: 396–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Gray ES, Moody MA, Wibmer CK, et al. Isolation of a monoclonal antibody that targets the alpha-2 helix of gp120 and represents the initial autologous neutralizing-antibody response in an HIV-1 subtype C-infected individual. J Virol 2011; 85: 7719–7729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Simek MD, Rida W, Priddy FH, et al. Human immunodeficiency virus type 1 elite neutralizers: individuals with broad and potent neutralizing activity identified by using a high-throughput neutralization assay together with an analytical selection algorithm. J Virol 2009; 83: 7337–7348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Bonsignori M, Liao HX, Gao F, et al. Antibody-virus co-evolution in HIV infection: paths for HIV vaccine development. Immunol Rev 2017; 275: 145–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Halper-Stromberg A, Nussenzweig MC. Towards HIV-1 remission: potential roles for broadly neutralizing antibodies. J Clin Invest 2016; 126: 415–423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Bhiman JN, Lynch RM. Broadly neutralizing antibodies as treatment: effects on virus and immune system. Curr HIV/AIDS Rep 2017; 14: 54–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Graham BS, Ambrosino DM. History of passive antibody administration for prevention and treatment of infectious diseases. Curr Opin HIV AIDS 2015; 10: 129–134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Scott AM, Wolchok JD, Old LJ. Antibody therapy of cancer. Nat Rev Cancer 2012; 12: 278–287. [DOI] [PubMed] [Google Scholar]
- 18. Saylor C, Dadachova E, Casadevall A. Monoclonal antibody-based therapies for microbial diseases. Vaccine 2009; 27(Suppl. 6): G38–G46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Keller MA, Stiehm ER. Passive immunity in prevention and treatment of infectious diseases. Clin Microbiol Rev 2000; 13: 602–614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Resch B. Product review on the monoclonal antibody palivizumab for prevention of respiratory syncytial virus infection. Hum Vaccin Immunother. 2017; 12: 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Levy J, Youvan T, Lee ML. Passive hyperimmune plasma therapy in the treatment of acquired immunodeficiency syndrome: results of a 12-month multicenter double-blind controlled trial. The Passive Hyperimmune Therapy Study Group. Blood 1994; 84: 2130–2135. [PubMed] [Google Scholar]
- 22. Vittecoq D, Chevret S, Morand-Joubert L, et al. Passive immunotherapy in AIDS: a double-blind randomized study based on transfusions of plasma rich in anti-human immunodeficiency virus 1 antibodies vs. transfusions of seronegative plasma. Proc Natl Acad Sci U S A 1995; 92: 1195–1199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Mehandru S, Vcelar B, Wrin T, et al. Adjunctive passive immunotherapy in human immunodeficiency virus type 1-infected individuals treated with antiviral therapy during acute and early infection. J Virol 2007; 81: 11016–11031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Stiegler G, Armbruster C, Vcelar B, et al. Antiviral activity of the neutralizing antibodies 2F5 and 2G12 in asymptomatic HIV-1-infected humans: a phase I evaluation. AIDS 2002; 16: 2019–2025. [DOI] [PubMed] [Google Scholar]
- 25. Manrique A, Rusert P, Joos B, et al. In vivo and in vitro escape from neutralizing antibodies 2G12, 2F5, and 4E10. J Virol 2007; 81: 8793–8808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Armbruster C, Stiegler GM, Vcelar BA, et al. Passive immunization with the anti-HIV-1 human monoclonal antibody (hMAb) 4E10 and the hMAb combination 4E10/2F5/2G12. J Antimicrob Chemother 2004; 54: 915–920. [DOI] [PubMed] [Google Scholar]
- 27. Armbruster C, Stiegler GM, Vcelar BA, et al. A phase I trial with two human monoclonal antibodies (hMAb 2F5, 2G12) against HIV-1. AIDS 2002; 16: 227–233. [DOI] [PubMed] [Google Scholar]
- 28. Burton DR, Pyati J, Koduri R, et al. Efficient neutralization of primary isolates of HIV-1 by a recombinant human monoclonal antibody. Science 1994; 266: 1024–1027. [DOI] [PubMed] [Google Scholar]
- 29. Trkola A, Kuster H, Rusert P, et al. Delay of HIV-1 rebound after cessation of antiretroviral therapy through passive transfer of human neutralizing antibodies. Nat Med 2005; 11: 615–622. [DOI] [PubMed] [Google Scholar]
- 30. Matsushita S, Yoshimura K, Ramirez KP, et al. Passive transfer of neutralizing mAb KD-247 reduces plasma viral load in patients chronically infected with HIV-1. AIDS 2015; 29: 453–462. [DOI] [PubMed] [Google Scholar]
- 31. Burton DR. What are the most powerful immunogen design vaccine strategies? Reverse vaccinology 2.0 shows great promise. Cold Spring Harb Perspect Biol 2017; 9: a030262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Burton DR, Hangartner L. Broadly neutralizing antibodies to HIV and their role in vaccine design. Annu Rev Immunol 2016; 34: 635–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Sadanand S, Suscovich TJ, Alter G. Broadly neutralizing antibodies against HIV: new insights to inform vaccine design. Annu Rev Med 2016; 67: 185–200. [DOI] [PubMed] [Google Scholar]
- 34. Klein F, Halper-Stromberg A, Horwitz JA, et al. HIV therapy by a combination of broadly neutralizing antibodies in humanized mice. Nature 2012; 492: 118–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Dinoso JB, Kim SY, Wiegand AM, et al. Treatment intensification does not reduce residual HIV-1 viremia in patients on highly active antiretroviral therapy. Proc Natl Acad Sci U S A 2009; 106: 9403–9408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Gandhi RT, Zheng L, Bosch RJ, et al. The effect of raltegravir intensification on low-level residual viremia in HIV-infected patients on antiretroviral therapy: a randomized controlled trial. PLoS Med 2010; 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Barouch DH, Whitney JB, Moldt B, et al. Therapeutic efficacy of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys. Nature 2013; 503: 224–228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Bruel T, Guivel-Benhassine F, Amraoui S, et al. Elimination of HIV-1-infected cells by broadly neutralizing antibodies. Nat Commun 2016; 7: 10844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Perdigao P, Gaj T, Santa-Marta M, et al. Reactivation of latent HIV-1 expression by engineered TALE transcription factors. PLoS One 2016; 11: e0150037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Cary DC, Peterlin BM. Targeting the latent reservoir to achieve functional HIV cure. F1000Res 2016; 5: 1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Barouch DH, Deeks SG. Immunologic strategies for HIV-1 remission and eradication. Science 2014; 345: 169–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Euler Z, Alter G. Exploring the potential of monoclonal antibody therapeutics for HIV-1 eradication. AIDS Res Hum Retroviruses 2015; 31: 13–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Klein F, Nogueira L, Nishimura Y, et al. Enhanced HIV-1 immunotherapy by commonly arising antibodies that target virus escape variants. J Exp Med 2014; 211: 2361–2372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Chun TW, Murray D, Justement JS, et al. Broadly neutralizing antibodies suppress HIV in the persistent viral reservoir. Proc Natl Acad Sci U S A 2014; 111: 13151–13156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Marsden MD, Zack JA. Neutralizing the HIV reservoir. Cell 2014; 158: 971–972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Jolly C, Kashefi K, Hollinshead M, et al. HIV-1 cell to cell transfer across an Env-induced, actin-dependent synapse. J Exp Med 2004; 199: 283–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Jolly C, Sattentau QJ. Retroviral spread by induction of virological synapses. Traffic 2004; 5: 643–650. [DOI] [PubMed] [Google Scholar]
- 48. Duncan CJ, Williams JP, Schiffner T, et al. High-multiplicity HIV-1 infection and neutralizing antibody evasion mediated by the macrophage-T cell virological synapse. J Virol 2014; 88: 2025–2034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Coleman CM, Gelais CS, Wu L. Cellular and viral mechanisms of HIV-1 transmission mediated by dendritic cells. Adv Exp Med Biol 2013; 762: 109–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Li Q, Duan L, Estes JD, et al. Peak SIV replication in resting memory CD4+ T cells depletes gut lamina propria CD4+ T cells. Nature 2005; 434: 1148–1152. [DOI] [PubMed] [Google Scholar]
- 51. Sigal A, Kim JT, Balazs AB, et al. Cell-to-cell spread of HIV permits ongoing replication despite antiretroviral therapy. Nature 2011; 477: 95–98. [DOI] [PubMed] [Google Scholar]
- 52. Abela IA, Berlinger L, Schanz M, et al. Cell-cell transmission enables HIV-1 to evade inhibition by potent CD4bs directed antibodies. PLoS Pathog 2012; 8: e1002634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Ringe R, Bhattacharya J. Preventive and therapeutic applications of neutralizing antibodies to human immunodeficiency virus type 1 (HIV-1). Ther Adv Vaccines 2013; 1: 67–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Montefiori DC. Bispecific antibodies against HIV. Cell 2016; 165: 1563–1564. [DOI] [PubMed] [Google Scholar]
- 55. Robbie GJ, Criste R, Dall’acqua WF, et al. A novel investigational Fc-modified humanized monoclonal antibody, motavizumab-YTE, has an extended half-life in healthy adults. Antimicrob Agents Chemother 2013; 57: 6147–6153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Ko SY, Pegu A, Rudicell RS, et al. Enhanced neonatal Fc receptor function improves protection against primate SHIV infection. Nature 2014; 514: 642–645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Xu L, Pegu A, Rao E, et al. Trispecific broadly neutralizing HIV antibodies mediate potent SHIV protection in macaques. Science 2017; 358: 85–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Huang Y, Yu J, Lanzi A, et al. Engineered bispecific antibodies with exquisite HIV-1-neutralizing activity. Cell 2016; 165: 1621–1631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Bournazos S, Gazumyan A, Seaman MS, et al. Bispecific anti-HIV-1 antibodies with enhanced breadth and potency. Cell 2016; 165: 1609–1620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Asokan M, Rudicell RS, Louder M, et al. Bispecific antibodies targeting different epitopes on the HIV-1 envelope exhibit broad and potent neutralization. J Virol 2015; 89: 12501–12512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Ferrari G, Haynes BF, Koenig S, et al. Envelope-specific antibodies and antibody-derived molecules for treating and curing HIV infection. Nat Rev Drug Discov 2016; 15: 823–834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Shingai M, Nishimura Y, Klein F, et al. Antibody-mediated immunotherapy of macaques chronically infected with SHIV suppresses viraemia. Nature 2013; 503: 277–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Cohen YZ, Lorenzi JCC, Seaman MS, et al. Neutralizing activity of broadly neutralizing anti-HIV-1 antibodies against clade B clinical isolates produced in peripheral blood mononuclear cells. J Virol 2018; 92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Caskey M, Schoofs T, Gruell H, et al. Antibody 10–1074 suppresses viremia in HIV-1-infected individuals. Nat Med 2017; 23: 185–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Lynch RM, Boritz E, Coates EE, et al. Virologic effects of broadly neutralizing antibody VRC01 administration during chronic HIV-1 infection. Sci Transl Med 2015; 7: 319ra206. [DOI] [PubMed] [Google Scholar]
- 66. Hraber P, Korber BT, Lapedes AS, et al. Impact of clade, geography, and age of the epidemic on HIV-1 neutralization by antibodies. J Virol 2014; 88: 12623–12643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Hraber P, Rademeyer C, Williamson C, et al. Panels of HIV-1 subtype C env reference strains for standardized neutralization assessments. J Virol 2017; 91. [DOI] [PMC free article] [PubMed] [Google Scholar]