Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

Review Article - (2015) Volume 6, Issue 4

The Role of CD4+ T cells in the Development of an Efficacious HIV Vaccine

Haitao Liu1,2, Wei Shen1, Jiayi Shu1 and Xia Jin1*
1Viral Disease and Vaccine Translational Research Unit & Vaccine Centre, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
2School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
*Corresponding Author: Xia Jin, M.D., Ph.D., Professor, Principal Investigator, Viral Disease and Vaccine Translational Research Unit, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China Email:

Abstract

HIV infection has caused serious public health disaster during the past three decades. The rapid discovery of antiretroviral drugs and the implementation of combination antiretroviral therapy in many countries over the past two decades have led to a marked decrease in HIV-associated mortality and morbidity. However, the antiretroviral therapy alone has been unable to eradicate the virus from HIV-infected individuals. To reduce the incidence of HIV infection on a global scale, vaccine is still the most cost-effective method. Due in part to the lack of a comprehensive understanding of immune correlates of protection, an effective HIV vaccine has not yet been developed. Since CD4+ T cells play a central role in orchestrating various arms of immune responses, vaccines that mobilize CD4+ T cells should help to elicit desired immune responses for the prevention of HIV infection. The current knowledge on subsets of CD4+ T cells and their perceived roles in mediation and formation of post-vaccination protective immunity are discussed.

Keywords: CD4+ T cell; Vaccine; HIV

Introduction

Since the first case of Acquired Immunodeficiency Syndrome (AIDS) was reported in 1981, the disease has caused approximately 36 million of deaths globally. AIDS is caused by infection with Human Immunodeficiency Virus (HIV). There are currently an estimated 35 million people living with HIV/AIDS [1,2].

HIV belongs to the Lentivirus genus and Retroviridae family. Its replication is controlled by a reverse transcriptase that lacks proof-reading mechanism, rendering a high mutation rate of approximately 3 × 10-5 per nucleotide per life cycle of HIV replication [3]. In addition, recombination as well as insertion and deletion happen frequently during viral replication and thereby further increased its genetic diversity [4]. As a consequence, HIV is hard to control either by drugs or immune responses.

Although the introduction of combination antiretroviral therapy (ART) has significantly reduced HIV-associated mortality and morbidity, and improved the quality of life in most HIV-infected individuals, the therapy alone cannot cure the disease. Preventative vaccine is considered to be the most cost-effective method to reduce the incident of new HIV infection. To develop an efficacious vaccine against HIV, the crucial role of CD4+ T cells must be better understood. Here, we review the new development on various subsets of CD4+ T cells, and discuss how these information might be employed in the development of vaccines against HIV.

Major Clinical Trials of HIV Vaccine

HIV has a small genome of approximately 10 kb that comprises of 9 genes encoding structural proteins (Gag, Pol, Env) and regulatory proteins (Tat, Rev, Nef, Vpr, Vif, Vpu), respectively. The viral particle is encapsulated by a lipid membrane bilayer in which HIV envelop (Env) glycoprotein spikes are embedded. Through binding to cell surface receptor (CD4) and co-receptor (CCR5 or CXCR4) by the Env protein, HIV infects CD4+ T cells and eventually causes AIDS. These viral structure and virological properties have been exploited to the development of antiviral drugs, and employed in the designing of vaccine targets. Despite vast knowledge has been acquired on HIV virology and immunology, it has been difficult to translate them into the development of an efficacious HIV vaccine.

Table 1 summarized several major HIV vaccine clinical trials conducted in the last two decades; despite capable of inducing measurable immune responses, none of which has yielded satisfactory clinical outcome. Early efforts had been placed on eliciting antibody responses using various forms of Env vaccine, culminating in phase III clinical trials, VAX003 and VAX004, in which recombinant gp120 proteins were tested as vaccine candidates. However, both trials failed to demonstrate protection against HIV acquisition in human volunteers [5,6]. Follow-up mechanistic studies have revealed that the ectodomain of Envprotein is comprised of highly variable, heavily glycosylated core and loop structures surrounding the HIV receptor-binding regions [10]. Since changes occur in N-linked glycosylation frequently [11], antibodies targeting carbohydrates are generally not efficient at preventing HIV infection, with a few exception [12]. Furthermore, high rate of mutation often occurs in the loop region, thus making strain-specific neutralizing antibodies unable to efficiently neutralize the viral mutants. In addition, some Env epitopes induce non-neutralizing antibodies that compromised the induction of neutralization antibodies [13]. Much effort is still being placed on the discovery of broadly neutralizing antibodies and design appropriate immunogens to induce broadly neutralizing antibody responses.

Name Trial sites Immunogen Targeted immune response Efficacy References
VAX004 USA, Canada, Netherlands AIDSVAX B/B gp120 CD4+ T cells, antibodies No efficacy 5
VAX003 Thailand AIDSVAX B/E gp120 CD4+ T cells, antibodies No efficac 6
STEP Study
(HVTN502)
USA, Canada, Latin America MRKAd5 HIV-1 gag/pol/nef B CD4+ T cells, CD8+ T cells No efficacy 7
Phambili trial
(HVTN503)
SouthAfrica MRKAd5 HIV-1 gag/pol/nef B CD4+ T cells, CD8+ T cells No efficacy 8
RV144 Thailand ALVAC-HIV vCP1521 and
AIDSVAX B/E rgp120
CD4+ T cells, CD8+ T cells, antibodies 31.2% efficacy 9

Table 1: Major clinical trials of HIV vaccine.

With growing knowledge of the antiviral activity of cytotoxic T cell (CTL) in the 1990s, the pendulum of AIDS vaccine development has swung to using various methods to induce HIV-specific CD8+ T cells [14,15]. One of the leading candidates was an adenovirus-based Gag vaccine that induced high magnitude of CD8+ T cell response and protected against SHIV challenged in non-human primates (NHP) [16,17]. Encouraged by the optimistic results in animal models, further clinical trials were performed. Unexpectedly, two large efficacy trials of the CTL-based vaccine neither prevented the acquisition of HIV infection nor reduced viral load or preserved CD4+ T cell counts post infection [7,8], thus revealing discrepancies in the vaccine-induced responses between NHP models and real-life infection in humans. Furthermore, whether CTL-based vaccine is of any importance has been questioned.

However, a subsequent study using DNA vaccine prime and adenovirus vector boost strategy showed that protection against SIV infection in rhesus monkeys can still be achieved. Important, the protected monkeys have high-frequency of CD8+ T cell responses against 11 to 34 epitopes, which is much more than that in the STEP human clinical trial where only and average of 5 epitopes were recognized [18]. This suggests the breadth of CD8+ T cell response maybe important, in addition to the magnitude of the response. Such increased breath, should be important at coping with viral escape from CD8+ T cell control [19]. Therefore, the current consensus is that CD8+ T cell-based vaccines need to elicit responses with great magnitude, improved breadth, and poly-functionality [20-22].

Most intriguingly, in the RV144 phase III clinical trials, a vaccine regimen consists of priming with recombinant canarypox vector designed to stimulate CD8+ T cells, and boosting with recombinant glycoprotein 120 subunit designed to elicit neutralizing antibodies, resulted in a 31.2% vaccine efficacy at the prevention of acquisition of HIV infection [9], despite neither vaccine used alone accomplished any significant effects.

Since CD4+ T cells assist the induction of antibody and CD8+ T cell responses, they should be carefully considered as a component of HIV vaccine. However, CD4+ T cells are the major targets for HIV infection, vaccine-induced CD4+ T cells may acquire an activated phenotypes that make them more susceptible to HIV infection. Indeed, it has been found that vaccine-induced CD4+ T cells contribute to the enhanced SIV replication and accelerated disease progression [23]. In contrast, others have reported that CD4+ T cells are beneficial in the prevention of SIV infection in experimental models [24]. In HIV-infected humans, a strong HIV-specific CD4+ T cell response is associated with a better control of viremia [25]. Further analysis of the RV144 results revealed that HIV-specific CD4+ T cell responses are positively correlated with vaccine efficacy [26]. Other than providing help, there are a small fraction of CD4+ T cells that have cytotoxic capacity and express granzyme A. These cells are expanded during acute HIV infection in some individuals and appear to help to suppress viral replication [27].

Above evidences reinforced the idea that a successful HIV vaccine strategy should aim to induce a CD4+ T cell response, in addition to B cell and CD8+ T cell responses. To harness CD4+ T cells in the development of an efficacious HIV vaccine, we need to first re-evaluate the functional diversity of CD4+ T cell subsets.

The importance of CD4+ T cell subsets in mediation and formation of post-vaccination protective immunity

Upon encountering antigens and adequate co-stimulation signals, naive CD4+ T cells are activated, polarized, and differentiated into distinct subsets including Th1, Th2, Th9, Th17, Th22, Regulatory T cell (Treg), Type I Regulatory cell (Tr1), and Follicular Helper T cell (TFH). Each subset has unique phenotype, cytokine secretion profiles, and transcriptional factors. Based on the stages of differentiation and anatomic locations, they can also be classified into central memory cell (TCM), effector memory cell (TEM), and tissue-resident memory cell (TRM) (Table 2). These T cell subsets either work independently or in concert to modulate other arms of immune responses. Use vaccines to stimulate specific subset of CD4+ T cells, if possible, should help the generation of higher quality antibody and CD8+ T cell responses. Following are a briefly review of the phenotypic and functional properties of each CD4+ T cell subset.

T cell subsets Phenotypic markers Cytokine secretion profile Transcription factors References
Naïve cells CD3+ CD4+CD45RA+ CCR7+ CD62L+ IL-7R+ IL-2 Th-POK 28-30
Th1 CD3+ CD4+CXCR3+IL-12R+ IFNγR+ IFN-γ, IL-2, TNFα,TNFβ T-bet, Eomes 31-36
Th2 CD3+ CD4+ IL-4R+CCR3+ CCR4+CCR8+ CRTh2+ IL-4, IL-5,IL-6,  IL-13 GATA3, STAT5, Gfi-1 32,34-38
Th9 CD3+ CD4+ IL-9 PU.1 12,13
Th17 CD3+ CD4+ IL-23R+ CCR4+ CCR6+ IL-6R+ CD161+ IL-17, IL-17F, IL-21, IL-22, IL-26 RORγt, RORα, RORC2 30,34,36,39-41
Th22 CD3+ CD4+ CCR4+ CCR6+ CCR10+ IL-22 AhR 42-45
Treg CD3+CD4+ CD25+ CTLA4+ CD127- IL-10, TGF-β Foxp3 30,34,36,46-50
Tr1 (type 1 regulatory) CD3+ CD4+ CD25-CD127-,Or CD3+ CD4+ CD49b+ LAG3+ IL-10, TGF-β Not known 30,47,51,52
TFH(follicular helper) CD3+ CD4+CXCR5+ SLAM+ OX40L+ CD40L+ IL-21, IL-4 Bcl6,Ascl2 53-57
TCM (central memory) CD3+ CD4+ CD45RA-IL-7R+ IL-15R+ CD44+ CCR7+ CD62L+ IL-2, IL-21 Bcl6 30, 58, 59
TEM(effector memory) CD3+ CD4+ CD44+ IL-7R+ IL-15R+ CCR7- IFN-γ,IL-4,IL-5,IL-17 Blimp1, T-bet 30, 58, 59
TRM(tissue-resident memory) CD3+ CD4+ CD11a+ CD69+ CD103+ IFN-γ Not Known 59, 60

Table 2: Characteristics of T helper cell subsets.

Th1: Through interacting and licensing dendritic cells, T helper 1 (Th1) cells contribute to the induction of cytotoxic T lymphocytes (CTLs) [61], and the maintenance of memory CTLs that can rapidly produce effector cells upon reinfection to eliminate pathogens [62]. In HIV infection, CTLs have been shown to suppress HIV-1 replication in cell cultures and they are associated with better control of viral loads in HIV-1 infected individuals [63-66]. The loss of Th1 cells during HIV-1 infection renders inefficient generation of new CTLs, and failure to prevent disease progression. A vaccine intended to stimulate effective antiviral CTL responses should thus contain a component for inducing sufficient Th1cells.

Th2: T helper 2 (Th2) cells modulate humoral immune responses and promote host defence against extracellular pathogens such as bacteria and parasites [67-70]. By interacting with Th2 cells, B cells undergo activation, proliferation and production of antigen-specific IgG antibodies, or the secretion of IgA antibodies [71]. IgG antibodies are a common form of neutralizing antibody against HIV. Secreted IgA antibodies play a key role in defending HIV-1 infection since the initial HIV infection often occurs at the mucosal surface [72]. Thus, a vaccine geared to stimulate anti-HIV antibodies must also include antigens to activate Th2 cells.

TFH: Follicular T helper cells (Tfh) help the formation and maintenance of germinal centers (GCs) [57]. In GCs, Tfh cells provide survival and proliferative signals to B cells to facilitate B cell proliferation, differentiation into plasma B cells or memory B cells, as well as antibody affinity maturation and class-switching [73]. Furthermore, Tfh cells assist B cells to produce broadly neutralizing antibodies (bNAbs) more swiftly [74]. However, a small fraction of Tfh cells are productively infected by HIV, rendering them defective [75]. Thus, use vaccine to induce Tfh response, and protect them from being destroyed by HIV infection should be beneficial for the elicitation of better quality neutralizing antibody responses against HIV. In fact, elucidating specific phenotypic and functional features of those Tfh cells that help to induce bNAbs, and harnessing these cells in HIV vaccine development had been topics of intense discussion in a recent NIH workshop [76].

Th17: T helper 17 (Th17) cells principally function in the protection against extracellular pathogens [41], through producing cytokines to maintain the integrity of the mucosal barrier and modulate the immune homeostasis at mucosal sites [77]. Mucosal tissues are the first barrier against HIV infection and a major site for HIV replication, thus a robust Th17 cell response contributing positively to the prevention of HIV infection [77]. HIV-specific Th17 cells are induced during early HIV infection [78]. In NHP models, a severe loss of Th17 cells was found in pathogenic SIV infections, whereas no obvious Th17 cell depletion has been shown in nonpathogenic SIV infections that occur in sooty mangabeys and African green monkeys [79,80]. Thus, a vaccine that induces Th17 cells should contribute to the prevention of HIV infection or the control of disease progression post infection.

Treg: Distinguished from other subsets of CD4+ T cells by expressing the master transcription factor Foxp3, regulatory T cells (Treg) suppress immune responses through the secretion of inhibitory cytokines such as IL-10, TGF-β and IL-35 [81,82]. Its role in HIV infection is uncertain currently. On one hand, by suppressing immune activation, Treg helps to reduce the detrimental persistent immune activation causing by HIV infection, and thus preventing disease progression [83]. On the other hand, Treg can inhibit HIV-specific immune responses, and thereby facilitate viral replication under certain circumstances [84,85]. Whether a vaccine should elicit Treg warrants further investigation.

In addition to the above mention CD4+ T cell subset, the potential roles of other subsets such as Th22, Tr1, and TRM in the development of HIV vaccine are unclear. However, the contribution of these other subsets to the overall success of vaccine-induce immune response to HIV should be investigated in future studies.

The Induction of HIV-Specific CD4+ T cell Responses by Vaccination

To design an effective vaccine to stimulate HIV-specific CD4+ T cells, antigens with higher immunogenicity are needed. Studies have demonstrated that HIV Gag can induce strong CD4+ T cell response. In fact, the majority of identified CD4+ T cell epitopes resided in Gag region. In addition to Gag, Nef, Env, Pol and Vpu have also been showed to elicit CD4+ T cell responses [27,86-90]. Furthermore, Env-specific CD4+ T cells are detected in the RV144 phase III clinical trial of HIV vaccines [9]. Thus, multiple epitopes from several HIV proteins should be considered in the design of an effective vaccine [91].

Other than specific HIV protein, the genetic diversity of HIV needs to be addressed in the design of a vaccine against HIV. To solve this problem, mosaic, conserved, central and consensus sequences have all been used in vaccine design in order to cover a wide range of HIV sequences [91-93]. Additionally, by careful selection of flanking sequences, the immunogenicity of polyepitope vaccine may be improved [94]. In future studies, head-to-head comparison among these methods should be performed to discern the pros and cons of each of the method for vaccine design.

Once the vaccine antigens are determined, the induction of specific T cell subsets that have biased cytokine secretion profiles may be dependent on the adjuvant of choice. Adjuvants are known to boost the potency, quality, and longevity of antigen specific immune responses through interacting with antigen presenting cells (APCs) [95-97]. Many forms of adjuvants have been examined in clinical and preclinical studies. The most common ones are aluminium salts (Alum), polynucleotides (polyIC, polyICLC)oligonucleotides (CpG), lipopolysaccharide (LPS), lipopeptides (Malp-2, Pam3Cys), and imidazoquinoline (R-8848) [96,98-102]. Results from previous studies have shown that polyIC and polyICLC elicited higher magnitude of CD4+ T cell responses than R-848, CpG, Malp-2, Pam3cys and LPS [103]. Among the squalene-based adjuvant, AS01B is more superior to AS02A and AS02V at eliciting CD4+ T cell responses [104]. These adjuvants should be tested in conjunction with candidate HIV vaccines.

Prospective on the Development of Vaccines Capable of Eliciting HIV-Specific CD4+ T cells

The development of vaccines that activate HIV-specific CD4+ T cell have confronted with many difficulties. First of which is the lack of knowledge on specific phenotype of CD4+ T cells that are immune correlates of protection [105]. Further comparative study between elite controllers and regular progressors should help to identify phenotypic markers of CD4+ T cells in association with the lack of HIV disease progression. Studies have shown that dysregulation of differentiation from central memory CD4+ T cells to effector memory cells is associated with immunological failure in HIV-1 infected individuals [106]. In subjects received candidate HIV-1 vaccine, CD107a+ CD4+ T cells are generated and these cells are relatively more resistant to depletion caused by HIV infection [107]. Thus, vaccines that preserve memory T cells and induce more CD107a+ cells should be beneficial.

Elicitation of specific subsets of CD4+ T cells by vaccination is going to be difficult since we still do not known how to create unique cytokine milieu in vivo. A novel dendritic cell (DC)-targeting strategy may help. In the DC targeting method, an antigen is conjugated with an antibody that recognize specific surface marker on DC surface, and thereby more efficiently bring the antigen to DC for processing and presentation [108], and consequently more efficient priming of T cells. Indeed, antigens target different subsets of DCs appears to elicit differential patterns of immune responses [109]. For instance, antigen targeting at Dectin-1 stimulates more cytokines secretion than targeting CD205 [110]. Further exploration of this vaccination strategy may help to discovery methods for activation specific CD4+ T cell subsets.

Conclusion

An effective HIV vaccine is urgently needed for the prevention of HIV infection and reducing AIDS associated mortality and morbidity. However, there are many obstacles in the development of HIV vaccines. First, high genetic diversity of HIV makes it a challenge to deal with all HIV strains using a single vaccine sequence. Second, even though both humoral and cellular immune responses can be elicited by candidate HIV vaccine or natural HIV infection, the viruses evade immune recognition through generating escape mutations. Third, CD4+ T cells are the major target of HIV, vaccines that activate CD4+ T cells may also generate more target cells that are susceptible to HIV-1 infection. Finally, the immune correlates of protection are not clearly understood. Though a simple principle for designing an efficacious HIV vaccine does not exist, the partial success of RV144 phase III HIV vaccine trial has showed a mixture of vectored vaccine and protein vaccine administer in a prime-and-boost regimen is promising [9]. With a better understanding of the immunology of HIV infection, and more data coming out of human clinical trial of candidate HIV vaccines, an improved version of HIV vaccine will be developed eventually. To carefully select adjuvants in the formulation of vaccines, and to manipulate the activation of different subsets of DCs may lead to directional activation of desirable T helper cell subsets, and thus achieving better vaccine immunogenicity. Whatever that version of vaccine may be, antigens that activate CD4+ T cells surely must be included as a key component.

Acknowledgements

We would like to thank Miss Min Lee for critical reading of the manuscript. This work is supported in part by the National Major Scientific and Technological program Mega Project during the Twelve Five-year Plan (2013ZX10001002002002).

References

  1. Rambaut A, Posada D, Crandall KA, Holmes EC (2004) The causes and consequences of HIV evolution. Nat Rev Genet 5: 52-61.
  2. Robertson DL, Hahn BH, Sharp PM (1995) Recombination in AIDS viruses. J MolEvol 40: 249-259.
  3. Flynn NM, Forthal DN, Harro CD, Judson FN, Mayer KH, et al. (2005) Placebo-controlled phase 3 trial of a recombinant glycoprotein 120 vaccine to prevent HIV-1 infection. J Infect Dis 191: 654-665.
  4. Pitisuttithum P, Gilbert P, Gurwith M, Heyward W, Martin M, et al. (2006) Randomized, double-blind, placebo-controlled efficacy trial of a bivalent recombinant glycoprotein 120 HIV-1 vaccine among injection drug users in Bangkok, Thailand. J Infect Dis 194: 1661-1671.
  5. Buchbinder SP, Mehrotra DV, Duerr A, Fitzgerald DW, Mogg R, et al. (2008) Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind, randomised, placebo-controlled, test-of-concept trial. Lancet 372: 1881-1893.
  6. Gray GE, Allen M, Moodie Z, Churchyard G, Bekker LG, et al. (2011) Safety and efficacy of the HVTN 503/Phambili study of a clade-B-based HIV-1 vaccine in South Africa: a double-blind, randomised, placebo-controlled test-of-concept phase 2b study. Lancet Infect Dis 11: 507-515.
  7. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, Kaewkungwal J, Chiu J, et al. (2009) Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N Engl J Med 361: 2209-2220.
  8. Wyatt R, Kwong PD, Desjardins E, Sweet RW, Robinson J, et al. (1998) The antigenic structure of the HIV gp120 envelope glycoprotein. Nature 393: 705-711.
  9. Wei X, Decker JM, Wang S, Hui H, Kappes JC, et al. (2003) Antibody neutralization and escape by HIV-1. Nature 422: 307-312.
  10. Trkola A, Purtscher M, Muster T, Ballaun C, Buchacher A, et al. (1996) Human monoclonal antibody 2G12 defines a distinctive neutralization epitope on the gp120 glycoprotein of human immunodeficiency virus type 1. J Virol 70: 1100-1108.
  11. Moore PL, Crooks ET, Porter L, Zhu P, Cayanan CS, et al. (2006) Nature of nonfunctional envelope proteins on the surface of human immunodeficiency virus type 1. J Virol 80: 2515-2528.
  12. Walker BD, Burton DR (2008) Toward an AIDS vaccine. Science 320: 760-764.
  13. Fauci AS, Johnston MI, Dieffenbach CW, Burton DR, Hammer SM, et al. (2008) HIV vaccine research: the way forward. Science 321: 530-532.
  14. Shiver JW, Fu TM, Chen L, Casimiro DR, Davies ME, et al. (2002) Replication-incompetent adenoviral vaccine vector elicits effective anti-immunodeficiency-virus immunity. Nature 415: 331-335.
  15. Hansen SG, Piatak M Jr, Ventura AB, Hughes CM, Gilbride RM, et al. (2013) Immune clearance of highly pathogenic SIV infection. Nature 502: 100-104.
  16. Wilson NA, Keele BF, Reed JS, Piaskowski SM, MacNair CE, et al. (2009) Vaccine-induced cellular responses control simian immunodeficiency virus replication after heterologous challenge. J Virol 83:6508-6521.
  17. McMichael A (1998) T cell responses and viral escape. Cell 93: 673-676.
  18. Liu J, O'Brien KL, Lynch DM, Simmons NL, La Porte A, et al. (2009) Immune control of an SIV challenge by a T-cell-based vaccine in rhesus monkeys. Nature 457: 87-91.
  19. Hansen SG, Ford JC, Lewis MS, Ventura AB, Hughes CM, et al. (2011) Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature 473: 523-527.
  20. Barouch DH, Stephenson KE, Borducchi EN, Smith K, Stanley K, et al. (2013) Protective efficacy of a global HIV-1 mosaic vaccine against heterologous SHIV challenges in rhesus monkeys. Cell 155: 531-539.
  21. Staprans SI, Barry AP, Silvestri G, Safrit JT, Kozyr N, et al. (2004) Enhanced SIV replication and accelerated progression to AIDS in macaques primed to mount a CD4 T cell response to the SIV envelope protein. Proc Nat AcadSci U S A 101:13026-13031.
  22. Ourmanov I, Brown CR, Moss B, Carroll M, Wyatt L, et al. (2000) Comparative efficacy of recombinant modified vaccinia virus Ankara expressing simian immunodeficiency virus (SIV) Gag-Pol and/or Env in macaques challenged with pathogenic SIV. J Virol 74: 2740-2751.
  23. Rosenberg ES, Billingsley JM, Caliendo AM, Boswell SL, Sax PE, et al. (1997) Vigorous HIV-1-specific CD4+ T cell responses associated with control of viremia. Science 278: 1447-1450.
  24. Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, Tomaras GD, et al. (2012) Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N Engl J Med 366: 1275-1286.
  25. Soghoian DZ, Jessen H, Flanders M, Sierra-Davidson K, Cutler S, et al. (2012) HIV-specific cytolytic CD4 T cell responses during acute HIV infection predict disease outcome. SciTranslMed 4:123ra25.
  26. Yin X, Ladi E, Chan SW, Li O, Killeen N, et al. (2007) CCR7 expression in developing thymocytes is linked to the CD4 versus CD8 lineage decision. J Immunol 179: 7358-7364.
  27. He X, He X, Dave VP, Zhang Y, Hua X, et al. (2005) The zinc finger transcription factor Th-POK regulates CD4 versus CD8 T-cell lineage commitment. Nature 433: 826-833.
  28. Geginat J, Paroni M, Maglie S, Alfen JS, Kastirr I, et al. (2014) Plasticity of human CD4 T cell subsets. FronImmunol 5: 630.
  29. Thieu VT, Yu Q, Chang HC, Yeh N, Nguyen ET, et al. (2008) Signal transducer and activator of transcription 4 is required for the transcription factor T-bet to promote T helper 1 cell-fate determination. Immunity 29: 679-690.
  30. Szabo SJ, Sullivan BM, Peng SL, Glimcher LH (2003) Molecular mechanisms regulating Th1 immune responses. Annu Rev Immunol 21: 713-758.
  31. Szabo SJ, Kim ST, Costa GL, Zhang X, Fathman CG, et al. (2000) A novel transcription factor, T-bet, directs Th1 lineage commitment. Cell 100:655-669.
  32. Zhu J, Yamane H, Paul WE (2010) Differentiation of effector CD4 T cell populations (*). Annu Rev Immunol 28: 445-489.
  33. Murphy KM, Ouyang W, Farrar JD, Yang J, Ranganath S, et al. (2000) Signaling and transcription in T helper development. Annu Rev Immunol 18: 451-494.
  34. Kanno Y, Vahedi G, Hirahara K, Singleton K, O'Shea JJ (2012) Transcriptional and epigenetic control of T helper cell specification: molecular mechanisms underlying commitment and plasticity. AnnuRev Immunol 30: 707-731.
  35. Ansel KM, Djuretic I, Tanasa B, Rao A (2006) Regulation of Th2 differentiation and Il4 locus accessibility. Annu Rev Immunol 24: 607-656.
  36. Sallusto F, Mackay CR, Lanzavecchia A (2000) The role of chemokine receptors in primary, effector, and memory immune responses. Annu Rev Immunol 18: 593-620.
  37. de Wit J, Souwer Y, van Beelen AJ, de Groot R, Muller FJ, et al. (2011) CD5 costimulation induces stable Th17 development by promoting IL-23R expression and sustained STAT3 activation. Blood 118: 6107-6114.
  38. Alvarez Y, Tuen M, Shen G, Nawaz F, Arthos J, et al. (2013) Preferential HIV infection of CCR6+ Th17 cells is associated with higher levels of virus receptor expression and lack of CCR5 ligands. J Virol 87: 10843-10854.
  39. Korn T, Bettelli E, Oukka M, Kuchroo VK (2009) IL-17 and Th17 Cells. Annu Rev Immunol 27: 485-517.
  40. Fujita H1 (2013) The role of IL-22 and Th22 cells in human skin diseases. J DermatolSci 72: 3-8.
  41. Akdis M, Palomares O, van de Veen W, van Splunter M, Akdis CA (2012) TH17 and TH22 cells: a confusion of antimicrobial response with tissue inflammation versus protection. J Allergy ClinImmunol129:1438-1449; quiz 1450-1451.
  42. Baba N, Rubio M, Kenins L, Regairaz C, Woisetschlager M, et al. (2012) The aryl hydrocarbon receptor (AhR) ligand VAF347 selectively acts on monocytes and naïve CD4(+) Th cells to promote the development of IL-22-secreting Th cells. Hum Immunol 73: 795-800.
  43. Eyerich S, Eyerich K, Pennino D, Carbone T, Nasorri F, et al. (2009) Th22 cells represent a distinct human T cell subset involved in epidermal immunity and remodeling. J ClinInvest 119:3573-3585.
  44. Read S, Malmström V, Powrie F (2000) Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory cells that control intestinal inflammation. J Exp Med 192: 295-302.
  45. Bacchetta R, Gregori S, Roncarolo MG (2005) CD4+ regulatory T cells: mechanisms of induction and effector function. Autoimmun Rev 4: 491-496.
  46. Fontenot JD, Gavin MA, Rudensky AY (2003) Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 4: 330-336.
  47. Hori S, Nomura T, Sakaguchi S (2003) Control of regulatory T cell development by the transcription factor Foxp3. Science 299: 1057-1061.
  48. Vignali DA, Collison LW, Workman CJ (2008) How regulatory T cells work. Nat Rev Immunol 8: 523-532.
  49. Battaglia M, Gregori S, Bacchetta R, Roncarolo MG (2006) Tr1 cells: from discovery to their clinical application. SeminImmunol 18: 120-127.
  50. Levings MK, Roncarolo MG (2000) T-regulatory 1 cells: a novel subset of CD4 T cells with immunoregulatory properties. J Allergy ClinImmunol 106: S109-112.
  51. Yusuf I, Kageyama R, Monticelli L, Johnston RJ, Ditoro D, et al. (2010) Germinal center T follicular helper cell IL-4 production is dependent on signaling lymphocytic activation molecule receptor (CD150). J Immunol185:190-202.
  52. Nurieva RI, Chung Y, Martinez GJ, Yang XO, Tanaka S, et al. (2009) Bcl6 mediates the development of T follicular helper cells. Science 325: 1001-1005.
  53. Yu D, Rao S, Tsai LM, Lee SK, He Y, et al. (2009) The transcriptional repressor Bcl-6 directs T follicular helper cell lineage commitment. Immunity 31: 457-468.
  54. Liu X, Chen X2, Zhong B3, Wang A, Wang X, et al. (2014) Transcription factor achaete-scute homologue 2 initiates follicular T-helper-cell development. Nature 507: 513-518.
  55. Crotty S1 (2011) Follicular helper CD4 T cells (TFH). Annu Rev Immunol 29: 621-663.
  56. Sallusto F, Geginat J, Lanzavecchia A (2004) Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu Rev Immunol 22: 745-763.
  57. Mueller SN, Gebhardt T, Carbone FR, Heath WR (2013) Memory T cell subsets, migration patterns, and tissue residence. Annu Rev Immunol 31: 137-161.
  58. Schenkel JM, Masopust D2 (2014) Tissue-resident memory T cells. Immunity 41: 886-897.
  59. Bennett SR, Carbone FR, Karamalis F, Miller JF, Heath WR (1997) Induction of a CD8+ cytotoxic T lymphocyte response by cross-priming requires cognate CD4+ T cell help. J Exp Med 186: 65-70.
  60. Shedlock DJ, Shen H (2003) Requirement for CD4 T cell help in generating functional CD8 T cell memory. Science 300: 337-339.
  61. Koup RA, Safrit JT, Cao Y, Andrews CA, McLeod G, et al. (1994) Temporal association of cellular immune responses with the initial control of viremia in primary human immunodeficiency virus type 1 syndrome. J Virol 68: 4650-4655.
  62. Borrow P, Lewicki H, Hahn BH, Shaw GM, Oldstone MB (1994) Virus-specific CD8+ cytotoxic T-lymphocyte activity associated with control of viremia in primary human immunodeficiency virus type 1 infection. J Virol 68: 6103-6110.
  63. Jin X, Bauer DE, Tuttleton SE, Lewin S, Gettie A, et al. (1999) Dramatic rise in plasma viremia after CD8(+) T cell depletion in simian immunodeficiency virus-infected macaques. J Exp Med 189: 991-998.
  64. Schmitz JE, Kuroda MJ, Santra S, Sasseville VG, Simon MA, et al. (1999) Control of viremia in simian immunodeficiency virus infection by CD8+ lymphocytes. Science 283: 857-860.
  65. Bartlett WC, McCann J, Shepherd DM, Roy M, Noelle RJ (1990) Cognate interactions between helper T cells and B cells. IV. Requirements for the expression of effector phase activity by helper T cells. J Immunol 145: 3956-3962.
  66. Zhu H, Chen Y, Zhou Y, Wang Y, Zheng J, et al. (2012) Cognate Th2-B cell interaction is essential for the autoantibody production in pemphigus vulgaris. J ClinImmunol 32: 114-123.
  67. Cliffe LJ, Grencis RK (2004) The Trichurismuris system: a paradigm of resistance and susceptibility to intestinal nematode infection. AdvParasitol 57: 255-307.
  68. Zaiss DM, Yang L, Shah PR, Kobie JJ, Urban JF, et al. (2006) Amphiregulin, a TH2 cytokine enhancing resistance to nematodes. Science 314: 1746.
  69. Kiyono H, Taguchi T, Aicher WK, Beagley KW, Fujihashi K, et al. (1990) Immunoregulatory confluence: T cells, Fc receptors and cytokines for IgA immune responses. Int Rev Immunol 6: 263-273.
  70. Zhou M, Ruprecht RM2,3 (2014) Are anti-HIV IgAs good guys or bad guys? Retrovirology 11: 109.
  71. Walker LS, Gulbranson-Judge A, Flynn S, Brocker T, Lane PJ (2000) Co-stimulation and selection for T-cell help for germinal centres: the role of CD28 and OX40. Immunol Today 21: 333-337.
  72. Phetsouphanh C, Xu Y, Zaunders J1 (2015) CD4 T Cells Mediate Both Positive and Negative Regulation of the Immune Response to HIV Infection: Complex Role of T Follicular Helper Cells and Regulatory T Cells in Pathogenesis. Front Immunol 5: 681.
  73. Xu Y, Fernandez C, Alcantara S, Bailey M, De Rose R, et al. (2013) Serial study of lymph node cell subsets using fine needle aspiration in pigtail macaques. J Immunol Methods 394: 73-83.
  74. Streeck H, D'Souza MP, Littman DR, Crotty S (2013) Harnessing CD4+ T cell responses in HIV vaccine development. Nat Med 19: 143-149.
  75. Bixler SL, Mattapallil JJ (2013) Loss and dysregulation of Th17 cells during HIV infection. ClinDevImmunol 2013: 852418.
  76. Yue FY, Merchant A, Kovacs CM, Loutfy M, Persad D, et al. (2008) Virus-specific interleukin-17-producing CD4+ T cells are detectable in early human immunodeficiency virus type 1 infection. J Virol 82: 6767-6771.
  77. Cecchinato V, Trindade CJ, Laurence A, Heraud JM, Brenchley JM, et al. (2008) Altered balance between Th17 and Th1 cells at mucosal sites predicts AIDS progression in simian immunodeficiency virus-infected macaques. Mucosal immunology1:279-288.
  78. Brenchley JM, Paiardini M, Knox KS, Asher AI, Cervasi B, et al. (2008) Differential Th17 CD4 T-cell depletion in pathogenic and nonpathogeniclentiviral infections. Blood 112: 2826-2835.
  79. Josefowicz SZ, Lu LF, Rudensky AY (2012) Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol 30: 531-564.
  80. Jenabian MA, Ancuta P, Gilmore N, Routy JP (2012) Regulatory T cells in HIV infection: can immunotherapy regulate the regulator? ClinDevImmunol 2012: 908314.
  81. Hazenberg MD, Otto SA, van Benthem BH, Roos MT, Coutinho RA, et al. (2003) Persistent immune activation in HIV-1 infection is associated with progression to AIDS. AIDS 17: 1881-1888.
  82. Kinter AL, Hennessey M, Bell A, Kern S, Lin Y, et al. (2004) CD25(+)CD4(+) regulatory T cells from the peripheral blood of asymptomatic HIV-infected individuals regulate CD4(+) and CD8(+) HIV-specific T cell immune responses in vitro and are associated with favorable clinical markers of disease status. J Exp Med 200: 331-343.
  83. Kinter A, McNally J, Riggin L, Jackson R, Roby G, et al. (2007) Suppression of HIV-specific T cell activity by lymph node CD25+ regulatory T cells from HIV-infected individuals. ProcNatlAcadSci U S A 104: 3390-3395.
  84. Walker LE, Vang L, Shen X, Livingston BD, Post P, et al. (2009) Design and preclinical development of a recombinant protein and DNA plasmid mixed format vaccine to deliver HIV-derived T-lymphocyte epitopes. Vaccine 27:7087-7095.
  85. Jin X, Newman MJ, De-Rosa S, Cooper C, Thomas E, et al. (2009) A novel HIV T helper epitope-based vaccine elicits cytokine-secreting HIV-specific CD4+ T cells in a Phase I clinical trial in HIV-uninfected adults. Vaccine 27:7080-7086.
  86. Gauduin MC, Yu Y, Barabasz A, Carville A, Piatak M, et al. (2006) Induction of a virus-specific effector-memory CD4+ T cell response by attenuated SIV infection. J Exp Med 203: 2661-2672.
  87. Zheng N, Fujiwara M, Ueno T, Oka S, Takiguchi M (2009) Strong ability of Nef-specific CD4+ cytotoxic T cells to suppress human immunodeficiency virus type 1 (HIV-1) replication in HIV-1-infected CD4+ T cells and macrophages. Journal of virology. 83:7668-7677.
  88. Sacha JB, Giraldo-Vela JP, Buechler MB, Martins MA, Maness NJ, et al. (2009) Gag- and Nef-specific CD4+ T cells recognize and inhibit SIV replication in infected macrophages early after infection. ProcNatlAcadSci U S A 106: 9791-9796.
  89. Shu J, Fan X2, Ping J2, Jin X, Hao P2 (2014) Designing peptide-based HIV vaccine for Chinese. Biomed Res Int 2014: 272950.
  90. Fonseca SG, Coutinho-Silva A, Fonseca LA, Segurado AC, Moraes SL, et al. (2006) Identification of novel consensus CD4 T-cell epitopes from clade B HIV-1 whole genome that are frequently recognized by HIV-1 infected patients. AIDS (London, England) 20:2263-2273.
  91. Jacobs ES, Persad D, Ran L, Danesh A, Heitman JW, et al. (2014) A CD4+ T cell antagonist epitope down-regulates activating signaling proteins, up-regulates inhibitory signaling proteins and abrogates HIV-specific T cell function. Retrovirology 11: 57.
  92. Reguzova A, Antonets D, Karpenko L, Ilyichev A, Maksyutov R, et al. (2015) Design and evaluation of optimized artificial HIV-1 poly-T cell-epitope immunogens. PLoS One 10: e0116412.
  93. Brunner R, Jensen-Jarolim E, Pali-Schöll I (2010) The ABC of clinical and experimental adjuvants--a brief overview. ImmunolLett 128: 29-35.
  94. Alving CR, Peachman KK, Rao M, Reed SG (2012) Adjuvants for human vaccines. CurrOpinImmunol 24: 310-315.
  95. Leroux-Roels G, Van Belle P2, Vandepapeliere P2, Horsmans Y3, Janssens M2, et al. (2015) Vaccine Adjuvant Systems containing monophosphoryl lipid A and QS-21 induce strong humoral and cellular immune responses against hepatitis B surface antigen which persist for at least 4 years after vaccination. Vaccine 33: 1084-1091.
  96. Coffman RL, Sher A, Seder RA (2010) Vaccine adjuvants: putting innate immunity to work. Immunity 33: 492-503.
  97. Hemmi H, Kaisho T, Takeuchi O, Sato S, Sanjo H, et al. (2002) Small anti-viral compounds activate immune cells via the TLR7 MyD88-dependent signaling pathway. Nat Immunol 3: 196-200.
  98. Barrenschee M, Lex D, Uhlig S (2010) Effects of the TLR2 agonists MALP-2 and Pam3Cys in isolated mouse lungs. PLoS One 5: e13889.
  99. Moyle PM, Toth I (2008) Self-adjuvantinglipopeptide vaccines. Curr Med Chem 15: 506-516.
  100. Reed SG, Orr MT, Fox CB (2013) Key roles of adjuvants in modern vaccines. Nat Med 19: 1597-1608.
  101. Longhi MP, Trumpfheller C, Idoyaga J, Caskey M, Matos I, et al. (2009) Dendritic cells require a systemic type I interferon response to mature and induce CD4+ Th1 immunity with poly IC as adjuvant. J Exp Med 206: 1589-1602.
  102. Leroux-Roels I, Koutsoukos M, Clement F, Steyaert S, Janssens M, et al. (2010) Strong and persistent CD4+ T-cell response in healthy adults immunized with a candidate HIV-1 vaccine containing gp120, Nef and Tat antigens formulated in three Adjuvant Systems. Vaccine 28:7016-7024.
  103. Koup RA, Graham BS, Douek DC (2011) The quest for a T cell-based immune correlate of protection against HIV: a story of trials and errors. Nat Rev Immunol 11: 65-70.
  104. Okoye AA, Picker LJ (2013) CD4(+) T-cell depletion in HIV infection: mechanisms of immunological failure. Immunol Rev 254: 54-64.
  105. Terahara K, Ishii H2, Nomura T2, Takahashi N2, Takeda A2, et al. (2014) Vaccine-induced CD107a+ CD4+ T cells are resistant to depletion following AIDS virus infection. J Virol 88: 14232-14240.
  106. Kastenmüller W, Kastenmüller K2, Kurts C3, Seder RA4 (2014) Dendritic cell-targeted vaccines--hope or hype? Nat Rev Immunol 14: 705-711.
  107. Dudziak D, Kamphorst AO, Heidkamp GF, Buchholz VR, Trumpfheller C, et al. (2007) Differential antigen processing by dendritic cell subsets in vivo. Science 315: 107-111.
  108. Carter RW, Thompson C, Reid DM, Wong SY, Tough DF (2006) Preferential induction of CD4+ T cell responses through in vivo targeting of antigen to dendritic cell-associated C-type lectin-1. J Immunol 177: 2276-2284.
Citation: Liu H, Shen W, Shu J, Jin X (2015) The Role of CD4+ T cells in the Development of an Efficacious HIV Vaccine. J Clin Cell Immunol 6:350.

Copyright: © 2015 Liu H, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top