For acute self-limiting infections a vaccine is successful if it elicits memory at least as good as the natural experience; however, for persistent and chronic infections such as HIV, hepatitis C virus (HCV), human papillomavirus (HPV), and human herpes viruses, this paradigm is not applicable. At best, during persistent virus infection the person must be able to maintain the integrity of the immune system in equilibrium with controlling replicating virus. New vaccine strategies are required that elicit both potent high-avidity CD8+ T-cell effector/memory and central memory responses that can clear the nidus of initial virus-infected cells at mucosal surfaces to prevent mucosal transmission or significantly curtail development of disease. The objective of an HIV-1 T-cell vaccine is to generate functional CD8+ effector memory cells at mucosal portals of virus entry to prevent viral transmission. In addition, long-lived CD8+ and CD4+ central memory cells circulating through secondary lymphoid organs and resident in bone marrow, respectively, are needed to provide a concerted second wave of defense that can contain virus at mucosal surfaces and prevent systemic dissemination. Further understanding of factors which can influence long-lived effector and central memory cell differentiation will significantly contribute to development of effective T-cell vaccines. In this review we will focus on discussing mechanisms involved in T-cell memory and provide promising new approaches toward expanding current vaccine strategies to enhance antiviral memory.

A common defining characteristic of immune memory is that it is both selective and parsimonious. After resolution of primary infection a small somewhat constant fraction of cells remain depending upon initial precursor frequency and the balance between T-cell receptor (TCR) signal strength and prosurvival signals received.1,2  Memory cells are homeostatically maintained in a progrowth state poised to respond rapidly to secondary infection. The fidelity of memory and effectiveness to thwart disease is reflected in the multifunctional character of the recalled response. Memory T cells are heterogenous in terms of phenotype, function, and anatomical locations.3-5  Understanding cytokine and costimulatory signals that influence transcriptional programs regulating T-cell differentiation and memory is key to manipulating vaccine responses. Furthermore, defining different subpopulations of memory CD8+ T cells by differentiation and activation markers representative of transcriptional programs associated with protective recall responses will be key to predicting vaccine efficacy.

To date, strategies for targeted delivery of vaccines and inclusion of cytokines, chemokines, and immunomodulatory molecules for enhancing the magnitude of immune responses and memory have been mostly empirical. For HIV-1 vaccines this has involved the expression of HIV-1 genes, cytokines, and chemokines by in vivo delivery of plasmid DNA and recombinant viral vectors (both replicating and nonreplicating). Recombinant viral vectors that target antigen-presenting cells serve as a means to couple activation of the innate immune system to the induction of adaptive immunity and increase immunogenicity. It is assumed that the use of recombinant viral vectors could induce local innate responses that promote an adaptive immune response to recombinant antigens that might obviate the need for adjuvanting this category of vaccines. It is unclear how antiviral (vector) immunity competes with or skews long-term memory to recombinant antigens and which if any viral vector is capable of inducing the innate immune signature required for coupling adaptive humoral and cellular responses to recombinant antigens that will lead to protective memory responses against HIV-1 infection.

Although central memory cells are considered a renewable source of T effector cells responsible for protection from acute infections and are primed for a rapid effector response, arguably, their ability to exert full effector capacity against the initial infected cell population is delayed sufficiently until the antigen-specific memory population undergoes expansion approximately 3 days after activation.6  In this regard, a number of studies have shown that effector memory cells at mucosal sites can proliferate and exercise a similar role to central memory in protection from disease.7  In addition, a direct relationship between the magnitude and quality of effector CD8+ cytotoxic T lymphocytes (CTL) at mucosal sites during acute simian immunodeficiency virus (SIV) infection was found to correlate with viral load in these animals.8  In a recent study, complete protection from repeated low-dose SIVmac239 intrarectal challenge was achieved in 4 of 12 monkeys immunized with a persistent rhesus cytomegalovirus (RhCMV) expressing SIV Gag, Rev-Tat-Nef, and Env.9  Low-level persistence of the CMV vector was responsible for maintaining a population of multifunctional effector memory SIV-specific CD4+ T cells and activated CD8+ effector T cells (CD28CCR7) in mucosal tissue (as measured in bronchoalveolar lavage) similar to the profile of immune responses directed against the virus during persistent RhCMV infection. Although a direct link between virus-infected cell killing and the specificities of CD8+ CTL involved in the protective response was not determined, this study suggests that differentiated effector or effector memory CD8+ CTL are durable and may play a significant role in extinguishing initial virus replication at local intestinal mucosal effector sites.8 

A recent study has provided evidence of an additional antiviral CD8+ T-cell memory phenotype confined to the site of infection in latently infected HSV-1 trigeminal ganglion.10  A population of tissue-resident CD8+ effector/memory cells (CD69+, CD62LLowCD122Low) were identified that expressed integrins αEβ7 (CD103+) and α1β1 (very late activation antigen-1 [VLA-1]). CD103+ can be induced locally by transforming growth factor-β (TGF-β) and binds E-cadherin on epithelial cells. CD103 is also expressed on interstitial epithelial lymphocytes in the intestine and tumor-infiltrating lymphocytes,11  whereas VLA-1 recognizes collagen and ensures migration and retention of cells within the interstitium after resolution of inflammation. Tissue-resident memory cells can proliferate in response to CD103+ dermal dendritic cells (DCs) and are more effective in local viral protection than CD8+ CTL recruited into skin grafts during reactivation. Expression of VLA-1+ is also found on a subset of human peripheral blood CD4+ memory cells (CD45RO+CD62L+CCR7CD25) associated with delayed-type hypersensitivity (DTH) to recall antigens and in synovial fluid of arthritic patients.12 

Different conclusions regarding the role of central and effector memory in protection from disease may in part be explained by differential transitional states and anatomical location of effector cells associated with different infectious agents in the different studies. The spectrum of effector and memory populations that develop after resolution of infection is a function of the infection potential, anatomical location, and survival niches in which responding cell populations anchor. Moreover, while the original paradigm suggests that the ability of CCR7+ T cells to enter secondary lymph nodes in response to CCL19 and CCL21 provides a reliable marker of CD44+CCR7+CD62L+ central memory cells (Tcm), more recent data suggest that for human CD4+ T-cell memory that proliferative capability and much of the interleukin-2 (IL-2) production is actually derived from CCR7CD4+ T cells. This is consistent with differential regulation of chemokine receptors during activation of CD4+ and CD8+ T cells, in part due to their different functional roles in response to infection and inflammation. Although this relatively simple classification of T-cell memory into Tcm and CD44+CCR7CD62L effector memory cells (Tem) will continue to be useful, it must be complemented by additional markers that reflect origin of priming and activation as well as functional analyses related to proliferative potential and role in protective outcomes. In this review we focus our attention on discussing signal transduction pathways responsible for induction and maintenance of T-cell memory formation and suggest strategies for enhancing vaccine induced memory for effective antiviral responses at mucosal surfaces.

On the basis of chemokine and homing receptors, proliferative capacity, and effector function, CD8+ T-cell memory has been divided into 2 populations after infection, Tcm and Tem. Central memory cells preferentially localize in lymph nodes, secrete IL-2 after antigen stimulation yet exhibit reduced immediate effector function and cytotoxicity.3  In contrast, effector memory cells preferentially localize in nonlymphoid peripheral tissue, have reduced proliferative potential, but exhibit immediate effector functions such as interferon-γ (IFN-γ) production and cytolytic capability. The contribution of Tcm and Tem in protective recall responses has been controversial. Proliferation and expansion of the population of CD62Lhigh memory CD8+ T cells following secondary systemic lymphocytic choriomeningitis virus (LCMV) infection was found to be responsible for virus clearance and resumption of functional memory.13  In contrast, using a dual adoptive transfer model to follow both CD62Lhi and CD62LLo memory cells following intranasal Sendai virus infection, it was found that CD62LLoCD8+ Tem cells proliferated as well as CD62LHighCD8+ Tcm in all organs tested and contributed proportionally to the immediate memory pool.14  However, over time the CD62Lhigh Tcm population evolved under homeostatic maintenance to become the predominant responsive population.

Although Tem are not able to enter lymph nodes under homeostatic conditions and represent a transitional effector phenotype, a recent study showed CXCR3+ Tem cells can migrate to reactive lymph nodes in response to CXCL9 and proinflammatory cytokines where they can undergo antigen-driven expansion and kill antigen-presenting cells.15 

Consistent with this observation, adoptive transfer of a late stage melanoma antigen specific effector CD8+ CTL line in humans (CD28low, CD27low, CD45RA, CD62L, CCR7lowIL-7Rlow) exerted antitumor responses in vivo and evolved into a population of CD28+CD27+CD62L, CCR7lowIL-7R+ effector memory cells.16 

In summary, the relative role of Tcm to Tem in recall responses and protection seems to depend on whether the infection is systemic, sequestered in tissue parenchyma, or mucosal, and the ability of memory T cells to migrate to local reactive lymph nodes and respond to antigen.17-19 

It is thought that the requirements for recalling memory responses are less stringent than those necessary for induction of primary responses. However, recent studies show that memory CD8+ T cells require antigen presentation by lymph node resident CD8α+ DCs and CD70/CD27 costimulation for activation in response to secondary influenza challenge.20  In contrast, naive T cells were immediately responsive to antigen presentation by tissue-resident CD8α DCs in the lung. Although CD8a+ DCs in mice are responsible for priming CD8+ CTL to viruses and intracellular bacteria, a distinct phenotypic counterpart to the mouse CD8α+ conventional DC responsible for cross presentation has not been identified in humans. There is some evidence that a C-type lectin, Clec9A or Clec12A, is a marker of an equivalent human DC subtype.

Compartmentalization between naive precursors and memory cells has 2 important implications for vaccine design. First, targeting antigen to specific DC subtypes provides the opportunity to increase the breadth of T-cell specificities achievable by delivering booster immunizations that target DCs locally at tissue sites of natural infection for de novo induction of new specificities and second, it may be possible to circumvent the immunodominant hierarchy that becomes fixed during boosting and cross-presentation.21,22  One caveat in the interpretation of these results is that although naive CD8+ T cells activated by viral antigens presented by tissue or extrafollicular DCs are capable of migrating into tissue spaces to exert immediate effector function, it is unclear if CD8+ T cells primed in the absence of CD4-proficient help in T cell–rich zones of draining lymph nodes contribute to the long-term memory pool or serve as “kamikaze” cells in response to the immediate viral threat. It is also important to consider how the boosting immunogen and more importantly natural infection dictate the functional outcome of recalled CD8+ CTL memory.23 

In contrast to the simple classification of memory CD8+ T cells based upon peripheral migration, an important new study in mice suggests that the majority of CD4+ T-cell memory is not found in either the Tcm or Tem cell populations but resides in bone marrow.24  This population of “quiescent” CD44hiCD62LCD4+ T cells expressed Ly-6C, similar to memory CD8+ T cells. Furthermore, expression of integrin α2 was required for homing to bone marrow niches where CD4+ memory cells remained adherent to stromal cells or bone marrow DCs which ensure their survival by providing a continuous source of IL-7.25  Upon recall, CD4+ bone marrow–resident memory cells were shown to rapidly mobilize into secondary lymph nodes where they were activated to provide helper function for an anamnestic antibody response. It is interesting to speculate that this niche is reserved for CD4+ memory T-cell maintenance since the size of the peripheral CD4+ memory pool after resolution of infection contracts approximately 50-fold more than the CD8 + T-cell memory population and declines over time as a result of attrition.25  These new findings might require a reevaluation of mechanisms involved in homing and maintenance of long-term CD4+ T-cell memory and have important implications for evaluating CD4+ T helper function induced by vaccines.

The observation that memory cell precursors display immediate effector function and the limitation of Tcm and Tem markers to identify early CD8+ T-cell effector and memory cell precursors that develop during the first 4 days and 7 to 10 divisions after initial antigen encounter lead to the assignment of additional markers to define early lineage commitment. Within 48 hours after TCR activation of naive and memory T cells, CD62L is down-regulated and cleaved from the cell surface and IL-7R expression lost. The discovery that the IL-7Rα chain (CD127) was reacquired by a subset of cells that persisted into long-term memory distinguished 2 populations of cells: memory precursor effector cells (MPECs) and short-lived effector cells (SLECs).26,27  The antigen-specific T-cell population that failed to up-regulate IL-7R expressed high levels of killer cell lectin-like receptor G1 (KLRG1) and showed little potential to develop into long-term memory was termed SLECs. KLRG1, a marker of replicative senescence, was recently shown to bind E cadherin expressed on epithelial cells and Langerhans cells (LCs) and inhibit antigen-induced proliferation in both mouse and human studies.28,29  KLRG1 is a good discriminator of antigen experience and is found on a high percentage of CD8+ T cells in persistent infections in humans such as HIV-1 but not hepatitis C virus (HCV).30  For the most part, IL-7R and KLRG expression are inversely related on MPEC (IL-7RhighKLRGlow) and SLECs (IL-7RlowKLRGlow) although this distinction between MPECs and SLECs has not been confirmed in human studies. KLRG1+IL-7Rlo expression in CD8+ effector cells is uniformly associated with Blimp-1 expression in both primary and recall responses to flu consistent with its role in regulating migration and terminal differentiation of effector cells.31,32 

CD4+ T-cell help is necessary for reexpression of IL-7R and IL-6Rα on the MPEC population suggesting an early imprint of CD4+ T-cell help contributing to the survival of activated CD8+ T cells.27,33  It is important to note that IL-7R and KLRG are not sufficient to predict long-term functional memory as CD8+ T cells are uniformly IL-7RhighKLRGlow after DC immunization but few cells evolve into long-term memory.34,35 

A recent study showed that activation markers may more accurately predict long-term memory.36  One month after Sendai virus infection, 3 equally represented antigen-specific populations of memory cells could be identified based upon expression of CD27 and T-cell activation marker CD43 (which also binds E cadherin). Although all 3 subsets displayed similar IFN-γ, tumor necrosis factor-α (TNF-α), and IL-2 multifunctional cytokine responses 1 month after infection and similar granzyme B expression during recall responses, the CD44highCD27highCD43low antigen-specific CD8+ T cells mounted the strongest recall response in lung and airways. After 2 years 90% of long-term memory cells expressed CD27highCD43low. In contrast, peptide/complete Freund adjuvant (CFA)–vaccinated animals displayed the same 3 phenotypes yet the relative proportion of each population was strongly skewed toward the CD27lowCD43high and an additional CD27lowCD43low subset which expressed KLRG1. The progressive loss of poorly proliferating memory cells associated with the activation marker CD43 indicate poor long-term vaccine efficacy relative to protection afforded by natural infection.

These studies indicate that memory CD8+ T cells with the highest proliferative capacity upon recall express CD62Lhigh, CD127high (IL-7R), KLRG1low, CD27high, CD43low, CD122high (IL-2/ IL-15Rβ), and Bcl-2high. Thus, the relative proportion of long-lived antigen-specific peripheral blood CD8+ T cells expressing these markers could be a useful predictor of vaccine efficacy (Figure 1).

Figure 1

Phenotypic markers associated with antigen-induced differentiation of naive CD8+ T cells. Within 48 hours after TCR activation, naive and memory CD8+ T cells up-regulate activation markers including IL-2R, Ki67, PCNA, and respond to signals delivered through JAK/STAT pathways to undergo changes in chemokine and homing receptors. CD62L is down-regulated and cleaved from the cell surface and IL-7Rα (CD127) expression is lost. During the proliferative phase a small subset of cells defined as memory precursor effector cells (MPECs) reacquire the IL-7Rα and have the potential to persist into long-term memory whereas the population that fails to up-regulate this receptor represent short-lived effector cells (SLECs).26,27,37  The antigen-specific T-cell population that fails to up-regulate IL-7R expresses high levels of killer cell lectin-like receptor G1 (KLRG1). Although all antigen-activated CD8+ T cells are thought to express immediate effector function and proliferative capacity, transition to memory is dependent upon multiple sequential signals received by the T cell including the intensity and duration of TCR activation, CD4+-proficient help, costimulation, and cytokines that regulate survival. Transition to memory after natural infection is more dependent on IL-7, whereas T cells that receive weaker signals in the case of soluble protein antigens delivered with adjuvants are equally dependent on IL-7 and IL-15 for survival and transition to memory. Defining phenotypic and functional markers that characterize different transitional phases of CD8+ T cells induced by different viral vectors and other delivery systems will be instrumental in advancing strategies for effective T-cell vaccines for HIV-1, cancer, and other infectious diseases. CD45RA also known as leukocyte common antigen, highly glycosylated protein tyrosine phosphatase regulating TCR, BCR activation and found on naive/resting T cells; CD44, family of cell-surface glycoproteins involved in leukocyte attachment and rolling on endothelial cells and homing to peripheral lymphoid organs by binding mucosal addressin on high endothelial venules. Marker for antigen-experienced cells; CCR7, CC-chemokine G protein–coupled receptor guides cells from peripheral tissue into lymph nodes binding CCL19 (Mip3β) and CCL21(SLC) deposited on HEVs and reticular network; CD62L, L-selectin binds to CD34 and mediates lymphocyte homing through high endothelial venules of peripheral lymphoid tissue and inflamed tissue; CD127, also known as IL-7R; CD122, (IL-2β chain) pairs with γ common chain. Critical component of IL-2 and IL-15–mediated signaling; CD28, constitutive, low-affinity costimulatory signal induces T-cell activation, IL-2 production, and survival; CD27, TNFR superfamily member 7 binds to CD70. Costimulatory signal helps differentiate memory-type CD8+ T cells (CD27+) from effector-type CD8+ T cells (CD27); CD43, leukosialin ligand-receptor complex involved in T-cell activation. Ligand for E-selectin and may regulate T-cell trafficking; CD95, TNF receptor superfamily member 6 also known as Fas. Cell-surface membrane receptor that activates apoptotic pathways when bound by Fas ligand (FasL, CD178); KLRG1, killer cell lectin-like receptor G1, senescence-associated inhibitory receptor binds E cadherin and inhibits AKT phosphorylation; Perforin, is indispensable for granule-mediated cell death by CD8+ CTL; Granzyme B, serine protease–inducing caspase-dependent apoptosis. Performs a key role in the cytotoxic activity mediated by CD8+ CTL (http://www.pathologyoutlines.com/cd100247.html).

Figure 1

Phenotypic markers associated with antigen-induced differentiation of naive CD8+ T cells. Within 48 hours after TCR activation, naive and memory CD8+ T cells up-regulate activation markers including IL-2R, Ki67, PCNA, and respond to signals delivered through JAK/STAT pathways to undergo changes in chemokine and homing receptors. CD62L is down-regulated and cleaved from the cell surface and IL-7Rα (CD127) expression is lost. During the proliferative phase a small subset of cells defined as memory precursor effector cells (MPECs) reacquire the IL-7Rα and have the potential to persist into long-term memory whereas the population that fails to up-regulate this receptor represent short-lived effector cells (SLECs).26,27,37  The antigen-specific T-cell population that fails to up-regulate IL-7R expresses high levels of killer cell lectin-like receptor G1 (KLRG1). Although all antigen-activated CD8+ T cells are thought to express immediate effector function and proliferative capacity, transition to memory is dependent upon multiple sequential signals received by the T cell including the intensity and duration of TCR activation, CD4+-proficient help, costimulation, and cytokines that regulate survival. Transition to memory after natural infection is more dependent on IL-7, whereas T cells that receive weaker signals in the case of soluble protein antigens delivered with adjuvants are equally dependent on IL-7 and IL-15 for survival and transition to memory. Defining phenotypic and functional markers that characterize different transitional phases of CD8+ T cells induced by different viral vectors and other delivery systems will be instrumental in advancing strategies for effective T-cell vaccines for HIV-1, cancer, and other infectious diseases. CD45RA also known as leukocyte common antigen, highly glycosylated protein tyrosine phosphatase regulating TCR, BCR activation and found on naive/resting T cells; CD44, family of cell-surface glycoproteins involved in leukocyte attachment and rolling on endothelial cells and homing to peripheral lymphoid organs by binding mucosal addressin on high endothelial venules. Marker for antigen-experienced cells; CCR7, CC-chemokine G protein–coupled receptor guides cells from peripheral tissue into lymph nodes binding CCL19 (Mip3β) and CCL21(SLC) deposited on HEVs and reticular network; CD62L, L-selectin binds to CD34 and mediates lymphocyte homing through high endothelial venules of peripheral lymphoid tissue and inflamed tissue; CD127, also known as IL-7R; CD122, (IL-2β chain) pairs with γ common chain. Critical component of IL-2 and IL-15–mediated signaling; CD28, constitutive, low-affinity costimulatory signal induces T-cell activation, IL-2 production, and survival; CD27, TNFR superfamily member 7 binds to CD70. Costimulatory signal helps differentiate memory-type CD8+ T cells (CD27+) from effector-type CD8+ T cells (CD27); CD43, leukosialin ligand-receptor complex involved in T-cell activation. Ligand for E-selectin and may regulate T-cell trafficking; CD95, TNF receptor superfamily member 6 also known as Fas. Cell-surface membrane receptor that activates apoptotic pathways when bound by Fas ligand (FasL, CD178); KLRG1, killer cell lectin-like receptor G1, senescence-associated inhibitory receptor binds E cadherin and inhibits AKT phosphorylation; Perforin, is indispensable for granule-mediated cell death by CD8+ CTL; Granzyme B, serine protease–inducing caspase-dependent apoptosis. Performs a key role in the cytotoxic activity mediated by CD8+ CTL (http://www.pathologyoutlines.com/cd100247.html).

Close modal

The factors recognized to affect T-cell memory lineage decisions are precursor frequency, duration of infection, proinflammatory cytokines, expression of activation markers and common γ-chain receptors, and anatomic location. Several models have been proposed to describe the events leading to memory cell formation. The first model proposes that memory cells arise during the contraction phase of the immune response and develop directly from effector cells. Thus, all cells are equipotential and effectors either die or dedifferentiate acquiring memory potential over time as a stochastic function of cytokine deprivation. The linear differentiation model is supported to a certain degree by the observation that activated precursors at some point express effector molecules during transition to memory cells.38  A number of studies have shown that highly activated multiple cytokine-producing CD4+ T cells also transit directly into memory cells during the first 3 to 5 days after antigen encounter suggesting a common developmental program for both CD4+ and CD8+ T cells.39-41 

A second model developed to help explain the phenotypic and functional heterogeneity of effector cells during priming of the immune response is based on the observation that activated CD8+ T cells progress in a stepwise manner gradually losing their memory potential as they differentiate toward a more terminally differentiated phenotype. The decreasing potential model implies that naive cells progress from weakly differentiated to terminally differentiated states as a result of the cumulative signals they receive during infection (eg, TCR-pMHC affinity, antigen dose and duration, cytokine and costimulatory signals). The corollary is that naive T cells entering lymph nodes late during infection (or circumstances in which infection is truncated or inflammation is limited) experience overall weaker signal strength and preferentially contribute to the antigen-specific memory precursor pool.42-44  One caveat of this approach for enhancing memory responses to vaccines is that compromising clonal expansion also severely limits the magnitude of memory cell development and in one study did not influence programming for secondary expansion.2 

A recent study in support of this model identified a population of antigen-specific CD8+ cells 4.5 days (8-10 cell divisions) after LCMV infection that exhibited full effector function, expressed KLRG1int yet were unique in their ability to produce IL-2.45  This KLRG1int population displayed greater potential for homeostatic proliferation and recall proliferation than KLRG1highCD8+ T cells. Importantly, during resolution of infection at day 6, KLRG1int memory precursors were proliferating less and down-regulating granzyme B expression whereas KLRG1high continued to proliferate and express granzyme B. Manipulating the duration of antigen stimulation and inflammation resulted in the largest number of KLRGlowCD62L, CD127high, CD27high IL-2–producing memory cells. Thus, the degree which naive CD8 + T cells are activated by “licensed” DCs, and the extent of clonal expansion before exiting lymph nodes and further encounters with antigen-presenting cells, most likely correlate with the quality and magnitude of the memory cell pool46  (Figure 2). In contrast to the studies that show excessive antigenic stimulation and inflammatory mediators drive CD8+ T-cell terminal differentiation and impairment of memory, evidence suggests that CD4+ T cells require more prolonged antigenic stimulation to drive cellular programs leading to the formation of long-term high-avidity memory.48 

Figure 2

Spatial-temporal signals determine the fate of activated CD8+ T cells. The balance between TLR-induced proinflammatory and apoptotic signals via STAT1 pathways in activating DCs and subsequent induction of negative feedback loops that initiate anti-inflammatory signals via the STAT3 axis define the narrow window of DC competency for polarizing CD4+ Th1 responses and cross-presentation of antigen to CD8+ T cells. TCR recognition of pMHC results in rapid down-regulation of CD62L, SIP1, and IL-7R, up-regulation of activation markers, and acquisition of effector function. During the next 3 to 4 days, Th1 CD4+ and CD8+ T cells proliferate in response to autocrine and paracrine IL-2, produce IFN-γ, and up-regulate CTLA-4. CTLA-4 ligation on DCs leads to indoleamine 2,3-dioxygenase (IDO) up-regulation, Foxo3-mediated inhibition of IL-6 production, and a shift from competency to induction of regulatory T cells, anergy, and cell death. This is the cue for activated CD4+ and CD8+ T cells to egress from secondary lymphoid organs. Foxo1 and KLF2 transcription factors, regulated by posttranscriptional modifications, coordinate renewed expression of the IL-7R, CD62L, CCR7, and S1P1, and down-regulate inflammatory chemokine receptors, respectively, on cells destined to become T central memory (Tcm) cells. In contrast, both IL-2 and IL-4 suppress Kruppel-like factor 2 (KLF2) expression and signals transmitted through IL-4R/ STAT6 up-regulate Eomes which induces expression of CXCR3 on CD8+ T cells. Survival after cytokine deprivation is dependent on costimulatory and other survival signals received before egress from lymphoid tissue. Limiting growth factors cause activated T cells to shutdown growth and proliferative programs sustained through TCR/CD28, and IL-2 and the P13K/AKT/mTOR pathway and to up-regulate autophagy pathways during transit into nonactivated lymphoid tissue and tissue niches. Here, T central memory (Tcm) and T effector memory (Tem) cell populations become dependent upon cytokine and tissue-specific interactions for maintenance and homeostasis. CD8+ T cells that receive CD4-proficient help in T cell–rich zones of draining lymph nodes may be more destined for long-term survival. The balance between costimulatory signals that up-regulate antiapoptotic factors, and negative costimulatory molecules up-regulated during the effector phase such as CTLA-4, BTLA-4, and PD1 (during chronic stimulation) that block effector function ultimately determine the population of cells that survive and transit into the memory pool. Expression of CD8+ effector function is regulated by 2 T box–binding transcription factors, Tbet and eomesodermin (Eomes). Although both Tbet and Eomes are essential inducers of CD8+ T-cell IFN-γ, perforin, granzyme B, and cytolytic capability, how these 2 transcription factors are coordinately regulated to control effector function and transition to long-term memory is unclear. During the inductive phase of the immune response, IL-12 and IFN-γ drive the differentiation and expansion of CD4+ Th1 and CD8+ T effector cells. This is reflected in the levels of T-bet expression and secreted osteopontin. During late-stage effector differentiation, inhibitor of DNA binding 2 (Id2), an antagonist of E protein transcription factors is up-regulated by CD8+ T cells and maintained in effector memory cells.47  E proteins are basic helix-loop-helix family of transcriptional activators and repressors, which bind specifically to DNA sequences containing the E-box consensus sequence. E protein homodimers regulate a complex array of genes during T-cell differentiation including expression of CD127 and CD27. Members of helix-loop-helix (HLH) protein family of Id (inhibitor of differentiation) dimerize with bHLH transcription factors and function as negative regulators of differentiation and promote progression of cells into S phase. E-Id2 heterodimer formation leads to diminished E-box–mediated gene expression such as Ctla4 and Bcl2l11 (BimEL) associated with reduced survival of T effector cells and relieves E protein repression of Serpinb9 coding for the serine protease inhibitor (Spi-6) thought to protect cytolytic effector cells from programmed cell death. Thus, Id-2 regulates the size of the Tem (CD62Llow, CD122lowCD127lowCD27high) subset in addition to regulating the survival of effector CD8+ T cells. Understanding transcriptional programs that control proliferation, acquisition of effector function, and survival of CD4+ and CD8+ T cells during the immune response will be key to developing new vaccine strategies.

Figure 2

Spatial-temporal signals determine the fate of activated CD8+ T cells. The balance between TLR-induced proinflammatory and apoptotic signals via STAT1 pathways in activating DCs and subsequent induction of negative feedback loops that initiate anti-inflammatory signals via the STAT3 axis define the narrow window of DC competency for polarizing CD4+ Th1 responses and cross-presentation of antigen to CD8+ T cells. TCR recognition of pMHC results in rapid down-regulation of CD62L, SIP1, and IL-7R, up-regulation of activation markers, and acquisition of effector function. During the next 3 to 4 days, Th1 CD4+ and CD8+ T cells proliferate in response to autocrine and paracrine IL-2, produce IFN-γ, and up-regulate CTLA-4. CTLA-4 ligation on DCs leads to indoleamine 2,3-dioxygenase (IDO) up-regulation, Foxo3-mediated inhibition of IL-6 production, and a shift from competency to induction of regulatory T cells, anergy, and cell death. This is the cue for activated CD4+ and CD8+ T cells to egress from secondary lymphoid organs. Foxo1 and KLF2 transcription factors, regulated by posttranscriptional modifications, coordinate renewed expression of the IL-7R, CD62L, CCR7, and S1P1, and down-regulate inflammatory chemokine receptors, respectively, on cells destined to become T central memory (Tcm) cells. In contrast, both IL-2 and IL-4 suppress Kruppel-like factor 2 (KLF2) expression and signals transmitted through IL-4R/ STAT6 up-regulate Eomes which induces expression of CXCR3 on CD8+ T cells. Survival after cytokine deprivation is dependent on costimulatory and other survival signals received before egress from lymphoid tissue. Limiting growth factors cause activated T cells to shutdown growth and proliferative programs sustained through TCR/CD28, and IL-2 and the P13K/AKT/mTOR pathway and to up-regulate autophagy pathways during transit into nonactivated lymphoid tissue and tissue niches. Here, T central memory (Tcm) and T effector memory (Tem) cell populations become dependent upon cytokine and tissue-specific interactions for maintenance and homeostasis. CD8+ T cells that receive CD4-proficient help in T cell–rich zones of draining lymph nodes may be more destined for long-term survival. The balance between costimulatory signals that up-regulate antiapoptotic factors, and negative costimulatory molecules up-regulated during the effector phase such as CTLA-4, BTLA-4, and PD1 (during chronic stimulation) that block effector function ultimately determine the population of cells that survive and transit into the memory pool. Expression of CD8+ effector function is regulated by 2 T box–binding transcription factors, Tbet and eomesodermin (Eomes). Although both Tbet and Eomes are essential inducers of CD8+ T-cell IFN-γ, perforin, granzyme B, and cytolytic capability, how these 2 transcription factors are coordinately regulated to control effector function and transition to long-term memory is unclear. During the inductive phase of the immune response, IL-12 and IFN-γ drive the differentiation and expansion of CD4+ Th1 and CD8+ T effector cells. This is reflected in the levels of T-bet expression and secreted osteopontin. During late-stage effector differentiation, inhibitor of DNA binding 2 (Id2), an antagonist of E protein transcription factors is up-regulated by CD8+ T cells and maintained in effector memory cells.47  E proteins are basic helix-loop-helix family of transcriptional activators and repressors, which bind specifically to DNA sequences containing the E-box consensus sequence. E protein homodimers regulate a complex array of genes during T-cell differentiation including expression of CD127 and CD27. Members of helix-loop-helix (HLH) protein family of Id (inhibitor of differentiation) dimerize with bHLH transcription factors and function as negative regulators of differentiation and promote progression of cells into S phase. E-Id2 heterodimer formation leads to diminished E-box–mediated gene expression such as Ctla4 and Bcl2l11 (BimEL) associated with reduced survival of T effector cells and relieves E protein repression of Serpinb9 coding for the serine protease inhibitor (Spi-6) thought to protect cytolytic effector cells from programmed cell death. Thus, Id-2 regulates the size of the Tem (CD62Llow, CD122lowCD127lowCD27high) subset in addition to regulating the survival of effector CD8+ T cells. Understanding transcriptional programs that control proliferation, acquisition of effector function, and survival of CD4+ and CD8+ T cells during the immune response will be key to developing new vaccine strategies.

Close modal

A third model proposed to explain the origin of T-cell diversification into memory and effector cells suggests that memory is predetermined by numeric differences in TCR clonotypic precursor frequencies and intraclonal diversification. A number of studies have shown that T-cell clonotypes are represented and persist long-term in both subsets representing the immunodominant hierarchy.49  In addition, an important recent study disputing this model found that memory precursors diverge from a single effector cell through a process of asymmetric division assigning different fates to each daughter cell.50  This finding has lead to the concept that the self-renewal properties of antigen-specific T-cell memory is maintained through asymmetrical mitoses similar to pluripotent stem cells and that a complex interplay of physiologic signals direct the differentiation fates of T cells derived from a limited number of multipotent memory cells. Furthermore, evidence from a growing number of studies suggest that both memory CD4+ and CD8+ T-cell and B-cell lineages share common transcriptional programs with inducible pluripotent stem cells (iPSC) that is evolutionarily conserved between humans and mice.51,52  Integrating knowledge of physiologic signals that regulate cell-cycle controls and signal transduction pathways, epigenetic states, and noncoding microRNAs (miRNAs) is crucial for understanding how cells escape replicative senescence and achieve the balance between self-renewal and cell differentiation.

A self-renewing cell must avoid replicative senescence and divide indefinitely. In addition to the 2 complementary survival signaling pathways: MAPK (Ras/Raf/Mek/Erk) and PI3K/AKT/mTOR activated by growth factors and costimulatory molecules that control the balance between proapoptotic and antiapoptotic factors, cell-cycle controls are integral for maintaining a pluripotential cell. The hallmark of a pluripotential stem cell is an unusual cell-cycle structure lacking a G1-Go restriction point and with a shortened G1 and extended S phase. This is believed to shield cells from the influence of differentiation cues in G1 yet maintain differentiation competency. A recent study found memory CD8+ T cells dwell in G1 phase that is maintained by costimulatory signals.53  Cell-cycle progression is regulated by the balance between cyclins/cyclin-dependent kinases (Cdk) and expression of Cdk inhibitory proteins p21Cip, p27Kip1, and the Ink4/Arf family of tumor suppressors. Antigen activation results in a hyperproliferative state, generation of reactive oxygen species (ROS) and hypoxia inducible factor 1α (HIF1-α) and a DNA-damage response that triggers antiproliferative tumor suppressor pathways leading to p53-mediated apoptosis and p21Cip and p16 INK4a-Rb–mediated senescence. During the contraction period, drivers of the PI3K/AKT pathway which integrate cell proliferation with cell growth, and cMyc, an epigenetic licensing factor, are central to establishment of a metastable multipotent state. Activated AKT phosphorylates the constitutively active serine theronine kinase GSK3β leading to its inactivation. The inactivation of GSK3β/destruction complex is a critical control point in the physiologic regulation of hematopoietic stem cell (HSC) renewal pathways Wnt and Notch. The canonical Wnt pathway leads to inactivation of the GSK3β complex, accumulation of high levels of β-catenin in the cytosol and translocation to the nucleus where it binds Tcf/Lef transcription factors to regulate multiple downstream target genes including cyclin D1, cMyc, and vascular endothelial growth factor (VEGF). Inactivation of GSK3β prevents proteasomal degradation of β-catenin and Smad-1, a cofactor in bone morphogenic protein receptor (Bmpr) signaling. Bmpr and leukemia inhibitory factor receptor (Lifr) signals are essential for stem cell renewal and differentiation and have been shown to represent distinct signaling pathways of self-renewal operative in memory B cells.52 

A seminal study recently showed that activating the Wnt/β-catenin pathway of self-renewal leads to arrested differentiation in activated CD8+ T cells in vitro yet these cells possessed high proliferative capability and function after adoptive transfer and reexposure to antigen in vivo (10-fold > Tcm upon adoptive transfer).54  Stem cell memory (Tscm) could be induced by direct activation of the Wnt pathway by Wnt3a protein or inhibitors of GSK3-β. It is important to note that Wnt-mediated inhibition of differentiation is dependent upon up-regulation of Notch target genes (eg, Hes1, Hey1, and Deltex1) which inhibit Cdk inhibitors mediating the balance between proliferation and differentiation. Further investigation of signaling pathways regulating gene expression by histone modifications and methyl transferases, and noncoding miRNAs will be important in understanding global mechanisms regulating memory cell formation.

The reader is referred further to a recent discussion of the models of CD8+ T-cell differentiation and memory.55 

A number of questions regarding the origin of memory precursors and the interpretation of different experimental protocols was resolved in an elegant series of experiments that showed that subset diversification can occur from a single naive precursor during the effector phase.56  Importantly, the pattern of subset diversification was the same when 100 precursors were adoptively transferred and reproduced the diversity seen in the endogenous response. Moreover, by introducing a single naive precursor after infection to reproduce “late comer” cells into the immune response it was shown that a similar pattern of subset diversification synchronized with cells primed earlier with no trend toward preferential development of CD62L+ Tcm populations. In contrast to the pattern of diversification seen by adoptive transfer of a single transgenic T cell followed by infection with Listeria monocytogenes expressing ovalbumin (L.m-Ova), the phenotypic pattern was substantially skewed toward a CD62LlowCD127low effector cell phenotype in animals that were immunized subcutaneously with Ova and CpG oligodeoxynucleotide (ODN). Thus, cumulative signals received by antigen-specific T cells during clonal expansion profoundly shape and synchronize the diversification patterns of effector and memory phenotypes that develop after resolution of infection. Furthermore, cells entering the immune response late during infection or responding to residual antigen depots that persist after resolution of infection will synchronize into similar diversification patterns.

These findings reinforce the importance of phenotypic and functional markers to define heterogenous patterns of multifunctional cells that predict the quality and magnitude of memory T-cell responses elicited by infection or vaccines57  (Figure 1). Extrapolating these findings to defining individual specificities which become representative of the memory cell pool with time is complex. The kinetics of transition to long-term memory will also differ for different infections, vectors, and immunization strategies. Vaccine strategies must be able to deliver the full complement of signals to T cells in a sequential manner to induce clonal expansion of antigen-specific T cells, control the duration of antigen expression and proinflammatory cytokines, and support the expansion and survival of memory precursor cells.

A recent study showed that the T effector memory pool to a single specificity can be expanded by a heterologous prime-boost strategy whereas T central memory is tightly regulated during secondary expansion.58  This study corroborates previous reports that show limiting antigen duration during boosting, including delivery of protein antigens by repeated short interval antigen pulses59  progressively skews the ratio of Tem/Tcm by converting the Tcm population attained after priming toward predominantly Tem cells. The general rule that the size of the Tcm compartment is under exquisite physiologic control begs the question of whether it is possible to significantly expand the antigen-specific Tcm compartment by repeated immunization; and if so, to what extent expansion of one or multiple new specificities compromise preexisting T-cell memory to the detriment of the host.

A balance between T-cell specificities occupying both T effector memory and T central memory pools could conceivably enable better control of virus during acute infection. For HIV vaccines this will require immunization strategies that generate sufficient numbers of high-avidity effector memory CD8+ CTL armed and ready at mucosal sites of infection and CD4+ and CD8+ central memory T cells capable of being rapidly recalled in a second wave of attack to curtail primary infection and prevent dissemination from mucosal compartments.8,60-65 

The production of inflammatory cytokines and in particular the induction of the IFN-γ/IL-12–signaling axis plays a key role in the coordinate regulation of the transcriptional programs that guide CD4+ Th1 and CD8+ T-cell effector and effector/memory differentiation.66  IL-12 levels shift Tbet expression into high gear driving CD4+ Th1 differentiation and cytolytic effector functions of CD8+ T and NK cells. Late in the effector phase as IL-12 levels fall, Tbet expression declines in activated CD8+ T cells while Eomes expression increases. CD8+ T cells induced in the absence of CD4+ T helper cells express high levels of Tbet and fail to develop into effective memory populations.37,67,68  Notably, Tbet represses IL-2 transcription and IL-7R expression. Eomes is expressed mainly in CD8+ T cells and was shown to control IFN-γ and perforin expression in tumor-infiltrating lymphocytes which correlated with positive antitumor responses in colorectal cancer patients.69  Eomes up-regulates IL-2Rβ/IL-15R (CD122) and IL-7R (CD127), and is positively regulated by TGF-β and IL-4R/signal transducer and activator of transcription 6 (STAT6) signals which confer CXCR3 expression on activated CD8+ T cells that can migrate to tissue effector sites. Eomes is negatively regulated by IL-12, CTLA-4, and IL-21.70  Attenuating IFN-γ production by CTLA-4 ligation could prevent cell death of effector/memory cells migrating to tissue effector sites and increase secondary CD8+ CTL responses.71  In summary, temporal regulation of IFN-γ and IL-12 produced by activated DCs during the inductive phase and cytokine and costimulatory signals received during contraction may be critical for determining the population of cells capable of entering the effector/memory cell pool in response to infection or vaccination (Figure 2).

The differential roles of type I and type II IFNs in influencing DC function and CD8+ T-cell responses is complex. Low levels of IFN-α can up-regulate CD40L on DCs and potently enhances CD8+ T-cell expansion and cytotoxicity to cross-presentation of antigen.72,73  However, STAT1 signals driven by high levels of IFN-α and IFN-γ inhibit CD40L induction of IL-12 by DCs and CD8+ T-cell proliferation. Type I IFNAR/STAT1 signals also up-regulate IL-15 production by DCs and have been shown to fuel a feed-forward loop required for CD8+ T-cell survival during the contraction phase and T-cell memory cell formation.41,74,75  It was recently shown that the accelerated secondary responsiveness of CD8+ T cells to antigen is also mediated by DC-produced IL-18 induced through a type I IFNAR-positive regulatory loop.76  Conversely, IFN-α up-regulation of CXCL-9 and CXCL10 on high endothelial venules (HEVs) may contribute to the infiltration CD8+ CXCR3 into lymph nodes (LNs) killing antigen-presenting DCs.

Although the nonredundant role of IL-2 in expansion of effector cells in peripheral tissues and during recall responses is well understood, its role during priming and formation of CD8+ T-cell memory is less clear.77,78  The finding that CD8+ cells generated in the absence of IL-2 signals are strongly impaired in recall responses but not expansion suggest that IL-2 could exert an imprint on early memory progenitor survival programs before reexpression of IL-7R and CD62L.79  Strong TCR/CD28 signals alone are sufficient to initiate G1-S cell-cycle progression. TCR signals up-regulate IL-2 expression while CD28 costimulation up-regulates Aurora B and Survivin downstream of the PI3K pathway. IL-2–PI3K signals activate the survivin–aurora mTOR complex to inactivate p27kip1 allowing Cdk1/2 to stimulate cell-cycle progression.

The transcription factor Blimp-1 is a sequence-specific recruitment factor for chromatin-remodeling enzymes and is well known for its role in silencing plasma cell transcriptional programs and promoting differentiation of B cells into antigen-secreting cells (ASC) and T effector cells.80  Blimp-1 has also been shown to play a pivotal role in T cells by suppressing IL-2 production and limiting proliferation of antigen-activated T cells during contraction of the immune response, increasing IL-10 production, and antigen-induced cell death (AICD).81  Blimp-1 expression is elevated in IL-7Rlow Tem cells and unhelped CD8+ T cells concordant with Tbet, KLRG1high expression.67  Alternately, expression of Bcl-6, a transcriptional repressor and antiapoptotic factor is necessary for generation of antigen-specific CD8+ T-cell memory and may function by suppressing Blimp-1 AICD similar to its reciprocal repression in B cells.82 

When IL-2 and IL-15 were complexed to antibody or soluble membrane IL-15Rα–Fc, respectively, and administered during the contraction phase of the immune response, KLRG1highCD127low effector CD8+ T cells preferentially accumulated whereas IL-7 antibody complexes given during the contraction phase skewed KLRG1lowCD127high memory CD8+83  and CD4+ T cells. Elevated levels of IL-2 were interpreted as sustaining effector responses that might be required in conditions of prolonged inflammation associated with pathogen persistence while IL-15 could prevent activation-induced cell death during the contraction phase. Although IL-15 shares the IL-2Rβ (CD122) and γc (CD132) with IL-2, these 2 cytokines have distinct and often competing roles. IL-2 plays a unique role in activation-induced cell death of self-reactive T cells and maintenance of peripheral CD4+CD25+ regulatory T cells (Tregs). In contrast, IL-15 is important for homeostatic maintenance, proliferation, and survival of NK cells and long-term high-avidity CD8+CD44hiCD122high T-cell memory. We recently showed that the differential effects on T-cell function could be attributed to different signal transduction pathways initiated by the intracellular domains of IL-15Rα and IL-2Rα chains.84  Both IL-15 and IL-15R are coordinately up-regulated on DCs and monocytes by type I and type II IFNs in concert with NFκβ activation triggered by ligation of CD40 or Toll-like receptor 4 (TLR4). Furthermore, IL-15 enhances DC survival. Transpresentation of IL-15 by membrane-bound IL-15Rα on DCs is required for survival and maintenance of CD8+ T-cell memory as well as secondary expansion.85  The receptor undergoes endocytosis upon binding and is recycled back to the cell surface prolonging its expression and availability for transpresentation.

IL-7 is also critical for the survival, homeostatic maintenance, and clonal turnover of antigen-activated memory T cells. TCR avidity, CD28 costimulation, and coreceptor expression have long been known to determine signal strength, up-regulation of CD25 (IL-2Rα), and functional outcome of the T-cell response.48,86  Strong TCR signals up-regulate expression of activation markers including CD5 which inversely correlates with CD8α coreceptor expression and IL-7 responsiveness.87  Scalable signal strengths were shown to regulate the IL-7 responsiveness of CD4+ T cells and influence memory and effector cell fate through increased survival signals transmitted downstream of the PI3K/AKT pathway.88  Costimulatory signals transmitted in parallel through the RAF/MEK/ERK pathway enforce the antiapoptotic program for cell survival of antigen-activated cells.

A recent study showed that administering IL-7 during the contraction phase (days 7-14) but not during the expansion phase is most effective in enhancing the magnitude of the antigen-specific CD8+ memory cell pool after virus infection or DNA immunization.89  In addition to increased survival via STAT5 up-regulation of Bcl-2, IL-7 has also been shown to enhance effector function and resistance to TGF-β–mediated suppression during the contraction phase of the immune response.90  These studies underscore the importance of timing the delivery of IL-7 after T-cell activation and reexpression of IL-7R in promoting T-cell transition to memory.

IL-21 shares the common γ-chain cytokine receptor with IL-2, IL-7, and IL-15 but signals through STAT1 and STAT3 where IL-2 and IL-15 primarily signal through STAT5. In contrast to IL-2 and IL-15, IL-21 limits activated T-cell proliferation. Naive CD4+ T cells stimulated by IL-6, Th17, T follicular helper (Tfh), and NKT cells secrete IL-21 and naive CD8+ T cells and DCs express the IL-21R. The inhibitory effect of IL-21 on T-cell responses is largely due to its potent effect in up-regulating SOCS-1 and SOCS-3 gene expression in DCs and macrophages, and inhibition of TLR2 and TLR4 induced maturation, cytokine production, and IFN-β–mediated signaling.91  IL-21 down-regulates Eomes expression on CD8+ CTL resulting in inhibition of IFN-γ production70  and has been shown to up-regulate perforin production without activation of CD8+ T cells from HIV-infected patients.92  Another study showed that IL-21 repressed CD44, Eomes, and granzyme B expression, and up-regulation of the high-affinity IL-2Rα subunit.93  Interestingly, adoptive transfer of CD8+ T cells primed with antigen in the presence of IL-21 were significantly more effective in undergoing secondary in vivo expansion and mediating tumor regression than either IL-2– or IL-15–primed cells. The enhanced in vivo efficacy of IL-21 versus IL-2– and IL-15–primed CD8+ CTL in tumor rejection was attributed to the ability of IL-21–primed CD8+ cells to express CD62L and maintain a less differentiated state. Primed CD8+ T cells could enter draining lymph nodes and undergo secondary expansion in vivo. IL-2 more than IL-15 is known to down-regulate CD62L on mature antigen-activated CD8+ T cells and this may be reversed by PI3K inhibitors. IL-21 has also been shown to synergize with both IL-7 and IL-15 in boosting CD44hiCD8+ T cells94  and enhanced antitumor functions.95  Thus, IL-21 seems to have a profound effect in inhibiting DC activation and in arresting CD8+ T-cell differentiation yet antigen-activated cells retain the ability to enter antigen-draining lymph nodes and undergo expansion and full effector maturation in vivo.

Although CD28 provides the primary costimulatory signal for T-cell activation and proliferation, several members of the TNFR superfamily, such as CD40, CD27, CD30, 4-1BB, OX40, and TNFR2, provide additional signals for induction of CD8+ T-cell effector and memory differentiation. Reciprocal signaling through TNF receptor superfamily (TNFRSF) molecules and ligands expressed on activated DCs and T cells is not only important for influencing the cytokines secreted by DCs but also for shaping CD4+ T-cell lineage commitment and CD8+ T-cell effector function and memory (Figure 2). We previously showed that coimmunization with vaccinia virus expressing a triad of costimulatory molecules (TRICOM), B7-1, ICAM-1, and LFA-3 could induce high-avidity CD8+ CTL responses.96  However, costimulation alone is not sufficient for the development of CD8+ T-cell memory and soluble or membrane-bound ligands have not been able to recapitulate the intimate interactions between DCs, CD4+ Th and CD8+ T cells necessary for imparting long-term memory and secondary expansion to CD8+ T cells.

CD40L continues to be a mainstay in vaccine strategies in combination with TLRs for induction of IL-12 and other TNFSF members on DCs that provide costimulation for augmenting T-cell help and induction of memory CD8+ T-cell responses27,97-100  (Table 1). Although interaction of CD40L, up-regulated on activated CD4+ Th1 cells with CD40 expressed on DCs is considered a primary mechanism for the up-regulation of costimulatory molecules and licensing DCs for CD8+ T-cell priming, a recent study showed that transient CD40L up-regulation on DCs is responsible for CD4-independent priming of CD8+ T cells seen in many virus infections.126 

Table 1

Experimental strategies to optimize vaccine T-cell memory

ApproachCD8 T-cell memory
References
TemTcm
Heterologous prime boost*   58  
    DNA prime viral vector boost ++ ++ 61, 101,103  
    Multiple heterologous recombinant viral vector +++ ++ 104,,107  
    Mucosal heterologous prime boost +++ ++ 61, 108  
    Persistent replicating vectors +++ 9, 109  
    Concurrent heterologous vaccination ++ ++  
Maximize CD4+-proficient help    
    Epitope enhancement, CD40L, agonist CD27 Ab ++ +++ 97, 98, 100, 110  
Cytokines and immunomodulatory molecules    
    IL-12 +++ 111  
    IL-7 ++ +++ 89, 90  
    IL-15 ++ ++ 112,114  
    IL-7 or IL-15 plus IL-21 ++ 94, 95  
    α-galactosylceramide (αGalCer) activation of NKT cell type I +++ ++ 115  
    Synergistic TLR combinations in prime boost +++ 116, 117  
    Dendritic cell targeting combined with synergistic TLR ligands +++ ++  
    Wnt 3a or GSK3β inhibitors  Tscm 54  
Provide effective costimulation    
    TNFSF: 41BBL, OX40L, CD70 ++ ++ 118, 119  
Block negative costimulation    
    Anti–CTLA-4, anti–BTLA-4, anti-PD1 +++ ++ 120, 121  
Push-pull approaches    
    GM-CSF and CD40L, plus IL-13Rα2–Fc +++ +++ 97  
    41BBL plus anti–CTLA-4, +++ +++ 119  
    TLR combinations plus anti–IL-10 +++  
Dendritic cell immunization (Flt3L in vitro expansion)   34  
    TLR 3 plus TLR7/8 activation +++ 116, 122  
    Adenovirus transfection ++ ++ 123  
    miRNA–SOCS-1 inhibition ++  
Repeated immunization at short intervals with multiepitope fusion peptides (Bacillus anthracis lethal factor Lfn) plus protective antigen (PA) ++ ++ 59  
ApproachCD8 T-cell memory
References
TemTcm
Heterologous prime boost*   58  
    DNA prime viral vector boost ++ ++ 61, 101,103  
    Multiple heterologous recombinant viral vector +++ ++ 104,,107  
    Mucosal heterologous prime boost +++ ++ 61, 108  
    Persistent replicating vectors +++ 9, 109  
    Concurrent heterologous vaccination ++ ++  
Maximize CD4+-proficient help    
    Epitope enhancement, CD40L, agonist CD27 Ab ++ +++ 97, 98, 100, 110  
Cytokines and immunomodulatory molecules    
    IL-12 +++ 111  
    IL-7 ++ +++ 89, 90  
    IL-15 ++ ++ 112,114  
    IL-7 or IL-15 plus IL-21 ++ 94, 95  
    α-galactosylceramide (αGalCer) activation of NKT cell type I +++ ++ 115  
    Synergistic TLR combinations in prime boost +++ 116, 117  
    Dendritic cell targeting combined with synergistic TLR ligands +++ ++  
    Wnt 3a or GSK3β inhibitors  Tscm 54  
Provide effective costimulation    
    TNFSF: 41BBL, OX40L, CD70 ++ ++ 118, 119  
Block negative costimulation    
    Anti–CTLA-4, anti–BTLA-4, anti-PD1 +++ ++ 120, 121  
Push-pull approaches    
    GM-CSF and CD40L, plus IL-13Rα2–Fc +++ +++ 97  
    41BBL plus anti–CTLA-4, +++ +++ 119  
    TLR combinations plus anti–IL-10 +++  
Dendritic cell immunization (Flt3L in vitro expansion)   34  
    TLR 3 plus TLR7/8 activation +++ 116, 122  
    Adenovirus transfection ++ ++ 123  
    miRNA–SOCS-1 inhibition ++  
Repeated immunization at short intervals with multiepitope fusion peptides (Bacillus anthracis lethal factor Lfn) plus protective antigen (PA) ++ ++ 59  
*

Although DNA prime and recombinant viral vector boost regimens induce enhanced cell-mediated and humoral immune responses in preclinical models, the results have not yet translated to clinical use. Ongoing efforts to develop new vaccine delivery platforms including DNA electroporation are moving forward toward clinical trial.124 

The cultured ELISPOT assay provides a more sensitive method to enumerate antigen-specific cells not detected in overnight ex vivo assays. This assay may better reveal long-term CD8+ T-cell memory responses achieved by prime boost and highlights differences in measuring human CD4+ and CD8+ T-cell memory.125 

Reciprocal signaling through up-regulated 4-1BBL/4-1BB interactions on activated DCs and antigen-activated CD4+ T cells can suppress IL-10 production and Treg effects thus potentiating Th1 polarization and CD8+ CTL induction.127  The mechanism of 4-1BB ligation in enhancing survival of activated CD8+ T cells was shown to involve activation of TNFR-associated factors TRAF-1 and TRAF-2. TRAF-1 up-regulated prosurvival Bcl-2 family member Bcl-xL and survival in conjunction with ERK-dependent proteasomal degradation of proapoptotic Bcl-2 family member Bim.128  A comparison of the relative role costimulatory molecules CD27, 4-1BB, and OX40 play in antiviral CD8+ T-cell responses showed that both CD27 and 4-1BB were necessary and nonredundant for the induction of CD8+ T-cell responses to influenza infection while OX40 had little effect.129  Collectively, costimulatory signals delivered by all 3 molecules during priming contributed to maximum secondary clonal expansion.

CD70/CD27 costimulation of antigen-activated CD4+ T cells is also a requirement for competent helper function.110,130  CD27 costimulation of activated CD4+ T cells skews the cell population toward a Th1 phenotype and up-regulates expression of a unique membrane marker confined to thymocytes and naive T cells, MS4A4B.110  Interestingly, MS4A4B was also identified as a differentially expressed marker on activated CD8+ T cells receiving CD27-“proficient” CD4+ T-cell help. Moreover, type 1 NKT cells stimulated by α gal-cer have been shown to up-regulate CD70 expression on DCs and effectively prime CD27-dependent CD8+ T-cell responses and effector memory.115,131  Thus, CD27 costimulation during priming appears to exert a strong influence on long-term survival of CD8+ effector memory.118 

The CD28 family members (CD28, CTLA-4, ICOS, PD-1) contain PI3K-binding motifs in their cytoplasmic tail and upon ligation result in strong PI3K/AKT signal transduction in lymphocytes in which the receptor is up-regulated.132  In contrast, TNFRSF members require TRAF adaptor molecules and are dependent upon both TCR signals and CD28 costimulation to deliver their costimulatory signals. CD28 family members CTLA-4, PD-1 and PD-2, T-cell immunoglobulin and mucin domain-containing molecule (TIM-3), and B and T-cell lymphocyte attenuator (BTLA, a member of the immunoglobulin [Ig] superfamily of proteins), are important coinhibitory receptors of TCR activation and effector function.133  Blocking PD-1 and CTLA-4 has been shown to restore T-cell responses in anergic T cells from patients with chronic HCV and HIV infections.120  Blocking the inhibitory receptor BTLA-4 has also been shown to enhance the development of CD8+ T-cell memory.121  A recent study demonstrated the effectiveness of combining anti–4-1BB agonist and anti–CTLA-4 blocking antibodies for induction of effector CD8+ T cell–mediated rejection of an established tumor and long-lasting memory responses. Importantly, the combination ameliorated autoimmune sequelae associated with administration of either antibody alone.119 

Further understanding of the expression kinetics, signal intensity, and dynamic interactions of activating and inhibitory costimulatory signals will better inform vaccine strategies that optimize the magnitude and quality of protective effector/memory and memory T-cell responses while avoiding induction of autoimmune responses.

The emerging view, and one to which we subscribe, of immune memory as an inducible quasi-potent stem cell–like state in which differentiation fates are determined by signaling pathways that regulate transcription factors, epigenetic modifications, and miRNAs is rapidly becoming the new paradigm that links together current research ideas in the fields of stem cell biology and cancer ontogeny. Since the original classification of memory CD8+ cell subsets into Tcm and Teff/mem, additional subsets (eg, memory precursor effector cell [MPEC]), tissue-resident memory, stem cell memory [Tscm]) with distinct phenotypes, anatomical locations, transcriptional programs, and function have been added to the list. Collectively, combination strategies and push-pull approaches that modulate Treg and anergic mechanisms may provide optimum immunization strategies capable of skewing adaptive T-cell responses toward high-avidity effector memory and central memory T-cell populations. New prime boost strategies that enhance the quantity and quality of CD4+ and CD8+ T-cell effector/memory cells in combination with mucosal delivery strategies that provide potent recall responses at mucosal sites of viral infection will greatly advance vaccine strategies against HIV and other intracellular pathogens. In summary, the overall understanding reached is to see the ongoing immune response as a dynamic process of activation and regulation that will require combination strategies and temporal spatial intervention to skew effective T-cell memory formation and augment protective immunity induced by vaccines (see supplemental text for additional information, available on the Blood website; see the Supplemental Materials link at the top of the online article).

The online version of this article contains a data supplement.

We apologize to the authors whose work we were unable to include in our citations due to space limitations.

Contribution: J.D.A. was primary author and illustrator; and I.M.B. was coauthor.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Jeffrey D. Ahlers, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 6700B Rockledge Dr, Rm 5138, Rockville, MD 20817; e-mail: jahlers@niaid.nih.gov; or Igor M. Belyakov, Midwest Research Institute, Frederick, MD 21702; e-mail: igorbelyakov@yahoo.com.

1
Badovinac
 
VP
Haring
 
JS
Harty
 
JT
Initial T cell receptor transgenic cell precursor frequency dictates critical aspects of the CD8(+) T cell response to infection.
Immunity
2007
, vol. 
26
 
6
(pg. 
827
-
841
)
2
Prlic
 
M
Hernandez-Hoyos
 
G
Bevan
 
MJ
Duration of the initial TCR stimulus controls the magnitude but not functionality of the CD8+ T cell response.
J Exp Med
2006
, vol. 
203
 
9
(pg. 
2135
-
2143
)
3
Sallusto
 
F
Lenig
 
D
Förster
 
R
Lipp
 
M
Lanzavecchia
 
A
Two subsets of memory T lymphocytes with distinct homing potentials and effector functions.
Nature
1999
, vol. 
401
 
6754
(pg. 
708
-
712
)
4
Masopust
 
D
Vezys
 
V
Marzo
 
AL
Lefrancois
 
L
Preferential localization of effector memory cells in nonlymphoid tissue.
Science
2001
, vol. 
291
 
5512
(pg. 
2413
-
2417
)
5
Marzo
 
AL
Yagita
 
H
Lefrancois
 
L
Cutting edge: migration to nonlymphoid tissues results in functional conversion of central to effector memory CD8 T cells.
J Immunol
2007
, vol. 
179
 
1
(pg. 
36
-
40
)
6
Whitmire
 
JK
Eam
 
B
Whitton
 
JL
Tentative T cells: memory cells are quick to respond, but slow to divide.
PLoS Pathog
2008
, vol. 
4
 
4
pg. 
e1000041
 
7
Masopust
 
D
Vezys
 
V
Wherry
 
EJ
Barber
 
DL
Ahmed
 
R
Cutting edge: gut microenvironment promotes differentiation of a unique memory CD8 T cell population.
J Immunol
2006
, vol. 
176
 
4
(pg. 
2079
-
2083
)
8
Li
 
Q
Skinner
 
PJ
Ha
 
SJ
, et al. 
Visualizing antigen-specific and infected cells in situ predicts outcomes in early viral infection.
Science
2009
, vol. 
323
 
5922
(pg. 
1726
-
1729
)
9
Hansen
 
SG
Vieville
 
C
Whizin
 
N
, et al. 
Effector memory T cell responses are associated with protection of rhesus monkeys from mucosal simian immunodeficiency virus challenge.
Nat Med
2009
, vol. 
15
 
3
(pg. 
293
-
299
)
10
Gebhardt
 
T
Wakim
 
LM
Eidsmo
 
L
Reading
 
PC
Heath
 
WR
Carbone
 
FR
Memory T cells in nonlymphoid tissue that provide enhanced local immunity during infection with herpes simplex virus.
Nat Immunol
2009
, vol. 
10
 
5
(pg. 
524
-
530
)
11
Le Floc'h
 
A
Jalil
 
A
Vergnon
 
I
, et al. 
Alpha E beta 7 integrin interaction with E-cadherin promotes antitumor CTL activity by triggering lytic granule polarization and exocytosis.
J Exp Med
2007
, vol. 
204
 
3
(pg. 
559
-
570
)
12
Goldstein
 
I
Ben-Horin
 
S
Li
 
J
Bank
 
I
Jiang
 
H
Chess
 
L
Expression of the alpha1beta1 integrin, VLA-1, marks a distinct subset of human CD4+ memory T cells.
J Clin Invest
2003
, vol. 
112
 
9
(pg. 
1444
-
1454
)
13
Wherry
 
EJ
Teichgraber
 
V
Becker
 
TC
, et al. 
Lineage relationship and protective immunity of memory CD8 T cell subsets.
Nat Immunol
2003
, vol. 
4
 
3
(pg. 
225
-
234
)
14
Roberts
 
AD
Ely
 
KH
Woodland
 
DL
Differential contributions of central and effector memory T cells to recall responses.
J Exp Med
2005
, vol. 
202
 
1
(pg. 
123
-
133
)
15
Guarda
 
G
Hons
 
M
Soriano
 
SF
, et al. 
L-selectin-negative CCR7- effector and memory CD8+ T cells enter reactive lymph nodes and kill dendritic cells.
Nat Immunol
2007
, vol. 
8
 
7
(pg. 
743
-
752
)
16
Powell
 
DJ
Dudley
 
ME
Robbins
 
PF
Rosenberg
 
SA
Transition of late-stage effector T cells to CD27+ CD28+ tumor-reactive effector memory T cells in humans after adoptive cell transfer therapy.
Blood
2005
, vol. 
105
 
1
(pg. 
241
-
250
)
17
Bachmann
 
MF
Wolint
 
P
Schwarz
 
K
Oxenius
 
A
Recall proliferation potential of memory CD8+ T cells and antiviral protection.
J Immunol
2005
, vol. 
175
 
7
(pg. 
4677
-
4685
)
18
Stock
 
AT
Jones
 
CM
Heath
 
WR
Carbone
 
FR
Cutting edge: central memory T cells do not show accelerated proliferation or tissue infiltration in response to localized herpes simplex virus-1 infection.
J Immunol
2006
, vol. 
177
 
3
(pg. 
1411
-
1415
)
19
Klonowski
 
KD
Marzo
 
AL
Williams
 
KJ
Lee
 
SJ
Pham
 
QM
Lefrancois
 
L
CD8 T cell recall responses are regulated by the tissue tropism of the memory cell and pathogen.
J Immunol
2006
, vol. 
177
 
10
(pg. 
6738
-
6746
)
20
Belz
 
GT
Bedoui
 
S
Kupresanin
 
F
Carbone
 
FR
Heath
 
WR
Minimal activation of memory CD8+ T cell by tissue-derived dendritic cells favors the stimulation of naive CD8+ T cells.
Nat Immunol
2007
, vol. 
8
 
10
(pg. 
1060
-
1066
)
21
Kastenmuller
 
W
Gasteiger
 
G
Gronau
 
JH
, et al. 
Cross-competition of CD8+ T cells shapes the immunodominance hierarchy during boost vaccination.
J Exp Med
2007
, vol. 
204
 
9
(pg. 
2187
-
2198
)
22
Pavelic
 
V
Matter
 
MS
Mumprecht
 
S
Breyer
 
I
Ochsenbein
 
AF
CTL induction by cross-priming is restricted to immunodominant epitopes.
Eur J Immunol
2009
, vol. 
39
 
3
(pg. 
704
-
716
)
23
Smith
 
MZ
Asher
 
TE
Venturi
 
V
, et al. 
Limited maintenance of vaccine-induced simian immunodeficiency virus-specific CD8 T-cell receptor clonotypes after virus challenge.
J Virol
2008
, vol. 
82
 
15
(pg. 
7357
-
7368
)
24
Tokoyoda
 
K
Zehentmeier
 
S
Hegazy
 
AN
, et al. 
Professional memory CD4+ T lymphocytes preferentially reside and rest in the bone marrow.
Immunity
2009
, vol. 
30
 
5
(pg. 
721
-
730
)
25
Guimond
 
M
Veenstra
 
RG
Grindler
 
DJ
, et al. 
Interleukin 7 signaling in dendritic cells regulates the homeostatic proliferation and niche size of CD4+ T cells.
Nat Immunol
2009
, vol. 
10
 
2
(pg. 
149
-
157
)
26
Kaech
 
SM
Tan
 
JT
Wherry
 
EJ
Konieczny
 
BT
Surh
 
CD
Ahmed
 
R
Selective expression of the interleukin 7 receptor identifies effector CD8 T cells that give rise to long-lived memory cells.
Nat Immunol
2003
, vol. 
4
 
12
(pg. 
1191
-
1198
)
27
Huster
 
KM
Busch
 
V
Schiemann
 
M
, et al. 
Selective expression of IL-7 receptor on memory T cells identifies early CD40L-dependent generation of distinct CD8+ memory T cell subsets.
Proc Natl Acad Sci U S A
2004
, vol. 
101
 
15
(pg. 
5610
-
5615
)
28
Grundemann
 
C
Bauer
 
M
Schweier
 
O
, et al. 
Cutting edge: identification of E-cadherin as a ligand for the murine killer cell lectin-like receptor G1.
J Immunol
2006
, vol. 
176
 
3
(pg. 
1311
-
1315
)
29
Henson
 
SM
Franzese
 
O
Macaulay
 
R
, et al. 
KLRG1 signaling induces defective Akt (ser473) phosphorylation and proliferative dysfunction of highly differentiated CD8+ T cells.
Blood
2009
, vol. 
113
 
26
(pg. 
6619
-
6628
)
30
Bengsch
 
B
Spangenberg
 
HC
Kersting
 
N
, et al. 
Analysis of CD127 and KLRG1 expression on hepatitis C virus-specific CD8+ T cells reveals the existence of different memory T-cell subsets in the peripheral blood and liver.
J Virol
2007
, vol. 
81
 
2
(pg. 
945
-
953
)
31
Kallies
 
A
Xin
 
A
Belz
 
GT
Nutt
 
SL
Blimp-1 transcription factor is required for the differentiation of effector CD8(+) T cells and memory responses.
Immunity
2009
, vol. 
31
 
2
(pg. 
283
-
295
)
32
Rutishauser
 
RL
Martins
 
GA
Kalachikov
 
S
, et al. 
Transcriptional repressor Blimp-1 promotes CD8(+) T cell terminal differentiation and represses the acquisition of central memory T cell properties.
Immunity
2009
, vol. 
31
 
2
(pg. 
296
-
308
)
33
Castellino
 
F
Germain
 
RN
Chemokine-guided CD4+ T cell help enhances generation of IL-6Ralpha high IL-7Ralpha high prememory CD8+ T cells.
J Immunol
2007
, vol. 
178
 
2
(pg. 
778
-
787
)
34
Zammit
 
DJ
Cauley
 
LS
Pham
 
QM
Lefrancois
 
L
Dendritic cells maximize the memory CD8 T cell response to infection.
Immunity
2005
, vol. 
22
 
5
(pg. 
561
-
570
)
35
Badovinac
 
VP
Messingham
 
KA
Jabbari
 
A
Haring
 
JS
Harty
 
JT
Accelerated CD8+ T-cell memory and prime-boost response after dendritic-cell vaccination.
Nat Med
2005
, vol. 
11
 
7
(pg. 
748
-
756
)
36
Hikono
 
H
Kohlmeier
 
JE
Takamura
 
S
Wittmer
 
ST
Roberts
 
AD
Woodland
 
DL
Activation phenotype, rather than central- or effector-memory phenotype, predicts the recall efficacy of memory CD8+ T cells.
J Exp Med
2007
, vol. 
204
 
7
(pg. 
1625
-
1636
)
37
Joshi
 
NS
Cui
 
W
Chandele
 
A
, et al. 
Inflammation directs memory precursor and short-lived effector CD8(+) T cell fates via the graded expression of T-bet transcription factor.
Immunity
2007
, vol. 
27
 
2
(pg. 
281
-
295
)
38
Kaech
 
SM
Hemby
 
S
Kersh
 
E
Ahmed
 
R
Molecular and functional profiling of memory CD8 T cell differentiation.
Cell
2002
, vol. 
111
 
6
(pg. 
837
-
851
)
39
Harrington
 
LE
Janowski
 
KM
Oliver
 
JR
Zajac
 
AJ
Weaver
 
CT
Memory CD4 T cells emerge from effector T-cell progenitors.
Nature
2008
, vol. 
452
 
7185
(pg. 
356
-
360
)
40
Lohning
 
M
Hegazy
 
AN
Pinschewer
 
DD
, et al. 
Long-lived virus-reactive memory T cells generated from purified cytokine-secreting T helper type 1 and type 2 effectors.
J Exp Med
2008
, vol. 
205
 
1
(pg. 
53
-
61
)
41
Bell
 
JJ
Ellis
 
JS
Guloglu
 
FB
, et al. 
Early effector T cells producing significant IFN-gamma develop into memory.
J Immunol
2008
, vol. 
180
 
1
(pg. 
179
-
187
)
42
Badovinac
 
VP
Harty
 
JT
Manipulating the rate of memory CD8+ T cell generation after acute infection.
J Immunol
2007
, vol. 
179
 
1
(pg. 
53
-
63
)
43
D'Souza
 
WN
Hedrick
 
SM
Cutting edge: latecomer CD8 T cells are imprinted with a unique differentiation program.
J Immunol
2006
, vol. 
177
 
2
(pg. 
777
-
781
)
44
Catron
 
DM
Rusch
 
LK
Hataye
 
J
Itano
 
AA
Jenkins
 
MK
CD4+ T cells that enter the draining lymph nodes after antigen injection participate in the primary response and become central-memory cells.
J Exp Med
2006
, vol. 
203
 
4
(pg. 
1045
-
1054
)
45
Sarkar
 
S
Kalia
 
V
Haining
 
WN
Konieczny
 
BT
Subramaniam
 
S
Ahmed
 
R
Functional and genomic profiling of effector CD8 T cell subsets with distinct memory fates.
J Exp Med
2008
, vol. 
205
 
3
(pg. 
625
-
640
)
46
Zehn
 
D
Lee
 
SY
Bevan
 
MJ
Complete but curtailed T-cell response to very low-affinity antigen.
Nature
2009
, vol. 
458
 
7235
(pg. 
211
-
214
)
47
Cannarile
 
MA
Lind
 
NA
Rivera
 
R
, et al. 
Transcriptional regulator Id2 mediates CD8+ T cell immunity.
Nat Immunol
2006
, vol. 
7
 
12
(pg. 
1317
-
1325
)
48
Williams
 
MA
Ravkov
 
EV
Bevan
 
MJ
Rapid culling of the CD4+ T cell repertoire in the transition from effector to memory.
Immunity
2008
, vol. 
28
 
4
(pg. 
533
-
545
)
49
Touvrey
 
C
Derre
 
L
Devevre
 
E
, et al. 
Dominant human CD8 T cell clonotypes persist simultaneously as memory and effector cells in memory phase.
J Immunol
2009
, vol. 
182
 
11
(pg. 
6718
-
6726
)
50
Chang
 
JT
Palanivel
 
VR
Kinjyo
 
I
, et al. 
Asymmetric T lymphocyte division in the initiation of adaptive immune responses.
Science
2007
, vol. 
315
 
5819
(pg. 
1687
-
1691
)
51
Haining
 
WN
Ebert
 
BL
Subrmanian
 
A
, et al. 
Identification of an evolutionarily conserved transcriptional signature of CD8 memory differentiation that is shared by T and B cells.
J Immunol
2008
, vol. 
181
 
3
(pg. 
1859
-
1868
)
52
Tomayko
 
MM
Anderson
 
SM
Brayton
 
CE
, et al. 
Systematic comparison of gene expression between murine memory and naive B cells demonstrates that memory B cells have unique signaling capabilities.
J Immunol
2008
, vol. 
181
 
1
(pg. 
27
-
38
)
53
Allam
 
A
Conze
 
DB
Giardino Torchia
 
ML
, et al. 
The CD8+ memory T-cell state of readiness is actively maintained and reversible.
Blood
2009
, vol. 
114
 
10
(pg. 
2121
-
2130
)
54
Gattinoni
 
L
Zhong
 
XS
Palmer
 
DC
, et al. 
Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells.
Nat Med
2009
, vol. 
15
 
7
(pg. 
808
-
813
)
55
Ahmed
 
R
Bevan
 
MJ
Reiner
 
SL
Fearon
 
DT
The precursors of memory: models and controversies.
Nat Rev Immunol
2009
, vol. 
9
 
9
(pg. 
662
-
668
)
56
Stemberger
 
C
Huster
 
KM
Koffler
 
M
, et al. 
A single naive CD8+ T cell precursor can develop into diverse effector and memory subsets.
Immunity
2007
, vol. 
27
 
6
(pg. 
985
-
997
)
57
Seder
 
RA
Darrah
 
PA
Roederer
 
M
T-cell quality in memory and protection: implications for vaccine design.
Nat Rev Immunol
2008
, vol. 
8
 
4
(pg. 
247
-
258
)
58
Vezys
 
V
Yates
 
A
Casey
 
KA
, et al. 
Memory CD8 T-cell compartment grows in size with immunological experience.
Nature
2009
, vol. 
457
 
7226
(pg. 
196
-
199
)
59
Shaw
 
CA
Starnbach
 
MN
Antigen delivered by anthrax lethal toxin induces the development of memory CD8+ T cells that can be rapidly boosted and display effector functions.
Infect Immun
2008
, vol. 
76
 
3
(pg. 
1214
-
1222
)
60
Belyakov
 
IM
Hel
 
Z
Kelsall
 
B
, et al. 
Mucosal AIDS vaccine reduces disease and viral load in gut reservoir and blood after mucosal infection of macaques.
Nat Med
2001
, vol. 
7
 (pg. 
1320
-
1326
)
61
Belyakov
 
IM
Kuznetsov
 
VA
Kelsall
 
B
, et al. 
Impact of vaccine-induced mucosal high avidity CD8+ CTLs in delay of AIDS-viral dissemination from mucosa.
Blood
2006
, vol. 
107
 
8
(pg. 
3258
-
3264
)
62
Belyakov
 
IM
Ahlers
 
JD
Functional CD8(+) CTLs in mucosal sites and HIV infection: moving forward toward a mucosal AIDS vaccine.
Trends Immunol
2008
, vol. 
29
 
11
(pg. 
574
-
585
)
63
Ahlers
 
JD
Belyakov
 
IM
Strategies for optimizing targeting and delivery of mucosal HIV vaccines.
Eur J Immunol
2009
, vol. 
39
 
10
(pg. 
2657
-
2669
)
64
Belyakov
 
IM
Ahlers
 
JD
What role does the route of immunization play in the generation of protective immunity against mucosal pathogens?
J Immunol
2009
, vol. 
183
 
11
(pg. 
6883
-
6892
)
65
Belyakov
 
IM
Derby
 
MA
Ahlers
 
JD
, et al. 
Mucosal immunization with HIV-1 peptide vaccine induces mucosal and systemic cytotoxic T lymphocytes and protective immunity in mice against intrarectal recombinant HIV-vaccinia challenge.
Proc Natl Acad Sci U S A
1998
, vol. 
95
 (pg. 
1709
-
1714
)
66
Schulz
 
EG
Mariani
 
L
Radbruch
 
A
Hofer
 
T
Sequential polarization and imprinting of type 1 T helper lymphocytes by interferon-gamma and interleukin-12.
Immunity
2009
, vol. 
30
 
5
(pg. 
673
-
683
)
67
Intlekofer
 
AM
Takemoto
 
N
Kao
 
C
, et al. 
Requirement for T-bet in the aberrant differentiation of unhelped memory CD8+ T cells.
J Exp Med
2007
, vol. 
204
 
9
(pg. 
2015
-
2021
)
68
Pearce
 
EL
Shen
 
H
Generation of CD8 T cell memory is regulated by IL-12.
J Immunol
2007
, vol. 
179
 
4
(pg. 
2074
-
2081
)
69
Atreya
 
I
Schimanski
 
CC
Becker
 
C
, et al. 
The T-box transcription factor eomesodermin controls CD8 T cell activity and lymph node metastasis in human colorectal cancer.
Gut
2007
, vol. 
56
 
11
(pg. 
1572
-
1578
)
70
Hegel
 
JK
Knieke
 
K
Kolar
 
P
Reiner
 
SL
Brunner-Weinzierl
 
MC
CD152 (CTLA-4) regulates effector functions of CD8+ T lymphocytes by repressing Eomesodermin.
Eur J Immunol
2009
, vol. 
39
 
3
(pg. 
883
-
893
)
71
Pandiyan
 
P
Hegel
 
JK
Krueger
 
M
Quandt
 
D
Brunner-Weinzierl
 
MC
High IFN-gamma production of individual CD8 T lymphocytes is controlled by CD152 (CTLA-4).
J Immunol
2007
, vol. 
178
 
4
(pg. 
2132
-
2140
)
72
Le Bon
 
A
Durand
 
V
Kamphuis
 
E
, et al. 
Direct stimulation of T cells by type I IFN enhances the CD8+ T cell response during cross-priming.
J Immunol
2006
, vol. 
176
 
8
(pg. 
4682
-
4689
)
73
Quigley
 
M
Huang
 
X
Yang
 
Y
STAT1 signaling in CD8 T cells is required for their clonal expansion and memory formation following viral infection in vivo.
J Immunol
2008
, vol. 
180
 
4
(pg. 
2158
-
2164
)
74
Kolumam
 
GA
Thomas
 
S
Thompson
 
LJ
Sprent
 
J
Murali-Krishna
 
K
Type I interferons act directly on CD8 T cells to allow clonal expansion and memory formation in response to viral infection.
J Exp Med
2005
, vol. 
202
 
5
(pg. 
637
-
650
)
75
Whitmire
 
JK
Eam
 
B
Benning
 
N
Whitton
 
JL
Direct interferon-gamma signaling dramatically enhances CD4+ and CD8+ T cell memory.
J Immunol
2007
, vol. 
179
 
2
(pg. 
1190
-
1197
)
76
Iwai
 
Y
Hemmi
 
H
Mizenina
 
O
Kuroda
 
S
Suda
 
K
Steinman
 
RM
An IFN-gamma-IL-18 signaling loop accelerates memory CD8+ T cell proliferation.
PLoS ONE
2008
, vol. 
3
 
6
pg. 
e2404
 
77
Williams
 
MA
Tyznik
 
AJ
Bevan
 
MJ
Interleukin-2 signals during priming are required for secondary expansion of CD8+ memory T cells.
Nature
2006
, vol. 
441
 
7095
(pg. 
890
-
893
)
78
Dooms
 
H
Wolslegel
 
K
Lin
 
P
Abbas
 
AK
Interleukin-2 enhances CD4+ T cell memory by promoting the generation of IL-7R alpha-expressing cells.
J Exp Med
2007
, vol. 
204
 
3
(pg. 
547
-
557
)
79
Kamimura
 
D
Bevan
 
MJ
Naive CD8+ T cells differentiate into protective memory-like cells after IL-2 anti IL-2 complex treatment in vivo.
J Exp Med
2007
, vol. 
204
 
8
(pg. 
1803
-
1812
)
80
Yu
 
J
Angelin-Duclos
 
C
Greenwood
 
J
Liao
 
J
Calame
 
K
Transcriptional repression by blimp-1 (PRDI-BF1) involves recruitment of histone deacetylase.
Mol Cell Biol
2000
, vol. 
20
 
7
(pg. 
2592
-
2603
)
81
Gong
 
D
Malek
 
TR
Cytokine-dependent Blimp-1 expression in activated T cells inhibits IL-2 production.
J Immunol
2007
, vol. 
178
 
1
(pg. 
242
-
252
)
82
Ichii
 
H
Sakamoto
 
A
Kuroda
 
Y
Tokuhisa
 
T
Bcl6 acts as an amplifier for the generation and proliferative capacity of central memory CD8+ T cells.
J Immunol
2004
, vol. 
173
 
2
(pg. 
883
-
891
)
83
Rubinstein
 
MP
Lind
 
NA
Purton
 
JF
, et al. 
IL-7 and IL-15 differentially regulate CD8+ T cell subsets during contraction of the immune response.
Blood
2008
, vol. 
112
 
9
(pg. 
3704
-
3712
)
84
Wu
 
Z
Xue
 
HH
Bernard
 
J
, et al. 
The IL-15 receptor {alpha} chain cytoplasmic domain is critical for normal IL-15Ralpha function but is not required for trans-presentation.
Blood
2008
, vol. 
112
 
12
(pg. 
4411
-
4419
)
85
Kokaji
 
AI
Hockley
 
DL
Kane
 
KP
IL-15 transpresentation augments CD8+ T cell activation and is required for optimal recall responses by central memory CD8+ T cells.
J Immunol
2008
, vol. 
180
 
7
(pg. 
4391
-
4401
)
86
Hofmann
 
M
Radsak
 
M
Rechtsteiner
 
G
, et al. 
T cell avidity determines the level of CTL activation.
Eur J Immunol
2004
, vol. 
34
 
7
(pg. 
1798
-
1806
)
87
Park
 
JH
Adoro
 
S
Lucas
 
PJ
, et al. 
‘Coreceptor tuning’: cytokine signals transcriptionally tailor CD8 coreceptor expression to the self-specificity of the TCR.
Nat Immunol
2007
, vol. 
8
 
10
(pg. 
1049
-
1059
)
88
Lozza
 
L
Rivino
 
L
Guarda
 
G
, et al. 
The strength of T cell stimulation determines IL-7 responsiveness, secondary expansion, and lineage commitment of primed human CD4+IL-7Rhi T cells.
Eur J Immunol
2008
, vol. 
38
 
1
(pg. 
30
-
39
)
89
Nanjappa
 
SG
Walent
 
JH
Morre
 
M
Suresh
 
M
Effects of IL-7 on memory CD8 T cell homeostasis are influenced by the timing of therapy in mice.
J Clin Invest
2008
, vol. 
118
 
3
(pg. 
1027
-
1039
)
90
Pellegrini
 
M
Calzascia
 
T
Elford
 
AR
, et al. 
Adjuvant IL-7 antagonizes multiple cellular and molecular inhibitory networks to enhance immunotherapies.
Nat Med
2009
, vol. 
15
 
5
(pg. 
528
-
536
)
91
Strengell
 
M
Lehtonen
 
A
Matikainen
 
S
Julkunen
 
I
IL-21 enhances SOCS gene expression and inhibits LPS-induced cytokine production in human monocyte-derived dendritic cells.
J Leukoc Biol
2006
, vol. 
79
 
6
(pg. 
1279
-
1285
)
92
White
 
L
Krishnan
 
S
Strbo
 
N
, et al. 
Differential effects of IL-21 and IL-15 on perforin expression, lysosomal degranulation, and proliferation in CD8 T cells of patients with human immunodeficiency virus-1 (HIV).
Blood
2007
, vol. 
109
 
9
(pg. 
3873
-
3880
)
93
Hinrichs
 
CS
Spolski
 
R
Paulos
 
CM
, et al. 
IL-2 and IL-21 confer opposing differentiation programs to CD8+ T cells for adoptive immunotherapy.
Blood
2008
, vol. 
111
 
11
(pg. 
5326
-
5333
)
94
Zeng
 
R
Spolski
 
R
Finkelstein
 
SE
, et al. 
Synergy of IL-21 and IL-15 in regulating CD8+ T-cell expansion and function.
J Exp Med
2005
, vol. 
201
 (pg. 
139
-
148
)
95
Liu
 
S
Lizee
 
G
Lou
 
Y
, et al. 
IL-21 synergizes with IL-7 to augment expansion and anti-tumor function of cytotoxic T cells.
Int Immunol
2007
, vol. 
19
 
10
(pg. 
1213
-
1221
)
96
Oh
 
S
Hodge
 
JW
Ahlers
 
JD
Burke
 
DS
Schlom
 
J
Berzofsky
 
JA
Selective induction of high avidity CTL by altering the balance of signals from antigen presenting cells.
J Immunol
2003
, vol. 
170
 (pg. 
2523
-
2530
)
97
Ahlers
 
JD
Belyakov
 
IM
Terabe
 
M
, et al. 
A push-pull approach to maximize vaccine efficacy: abrogating suppression with an IL-13 inhibitor while augmenting help with GM-CSF and CD40L.
Proc Natl Acad Sci U S A
2002
, vol. 
99
 
20
(pg. 
13020
-
13025
)
98
Liu
 
J
Yu
 
Q
Stone
 
GW
, et al. 
CD40L expressed from the canarypox vector, ALVAC, can boost immunogenicity of HIV-1 canarypox vaccine in mice and enhance the in vitro expansion of viral specific CD8+ T cell memory responses from HIV-1-infected and HIV-1-uninfected individuals.
Vaccine
2008
, vol. 
26
 
32
(pg. 
4062
-
4072
)
99
Ahonen
 
CL
Doxsee
 
CL
McGurran
 
SM
, et al. 
Combined TLR and CD40 triggering induces potent CD8+ T cell expansion with variable dependence on type I IFN.
J Exp Med
2004
, vol. 
199
 
6
(pg. 
775
-
784
)
100
Sanchez
 
PJ
McWilliams
 
JA
Haluszczak
 
C
Yagita
 
H
Kedl
 
RM
Combined TLR/CD40 stimulation mediates potent cellular immunity by regulating dendritic cell expression of CD70 in vivo.
J Immunol
2007
, vol. 
178
 
3
(pg. 
1564
-
1572
)
101
Wilson
 
NA
Keele
 
BF
Reed
 
JS
, et al. 
Vaccine-induced cellular responses control simian immunodeficiency virus replication after heterologous challenge.
J Virol
2009
, vol. 
83
 
13
(pg. 
6508
-
6521
)
102
Goonetilleke
 
N
Moore
 
S
Dally
 
L
, et al. 
Induction of multifunctional human immunodeficiency virus type 1 (HIV-1)-specific T cells capable of proliferation in healthy subjects by using a prime-boost regimen of DNA- and modified vaccinia virus Ankara-vectored vaccines expressing HIV-1 Gag coupled to CD8+ T-cell epitopes.
J Virol
2006
, vol. 
80
 
10
(pg. 
4717
-
4728
)
103
Harari
 
A
Bart
 
PA
Stohr
 
W
, et al. 
An HIV-1 clade C DNA prime, NYVAC boost vaccine regimen induces reliable, polyfunctional, and long-lasting T cell responses.
J Exp Med
2008
, vol. 
205
 
1
(pg. 
63
-
77
)
104
Liu
 
J
O'Brien
 
KL
Lynch
 
DM
, et al. 
Immune control of an SIV challenge by a T-cell-based vaccine in rhesus monkeys.
Nature
2009
, vol. 
457
 
7225
(pg. 
87
-
91
)
105
Belyakov
 
IM
Ahlers
 
JD
Nabel
 
GJ
Moss
 
B
Berzofsky
 
JA
Generation of functionally active HIV-1 specific CD8(+) CTL in intestinal mucosa following mucosal, systemic or mixed prime-boost immunization.
Virology
2008
, vol. 
381
 (pg. 
106
-
115
)
106
Corbett
 
M
Bogers
 
WM
Heeney
 
JL
, et al. 
Aerosol immunization with NYVAC and MVA vectored vaccines is safe, simple, and immunogenic.
Proc Natl Acad Sci U S A
2008
, vol. 
105
 
6
(pg. 
2046
-
2051
)
107
Xu
 
R
Nasar
 
F
Megati
 
S
, et al. 
Prime-boost vaccination with recombinant mumps virus and recombinant vesicular stomatitis virus vectors elicits an enhanced human immunodeficiency virus type 1 Gag-specific cellular immune response in rhesus macaques.
J Virol
2009
, vol. 
83
 
19
(pg. 
9813
-
9823
)
108
Belyakov
 
IM
Isakov
 
D
Zhu
 
Q
Dzutsev
 
A
Berzofsky
 
JA
A novel functional CTL avidity/activity compartmentalization to the site of mucosal immunization contributes to protection of macaques against simian/human immunodeficiency viral depletion of mucosal CD4+ T cells.
J Immunol
2007
, vol. 
178
 (pg. 
7211
-
7221
)
109
Verreck
 
FA
Vervenne
 
RA
Kondova
 
I
, et al. 
MVA. 85A boosting of BCG and an attenuated, phoP deficient M. tuberculosis vaccine both show protective efficacy against tuberculosis in rhesus macaques.
PLoS ONE
2009
, vol. 
4
 
4
pg. 
e5264
 
110
Xiao
 
Y
Peperzak
 
V
Keller
 
AM
Borst
 
J
CD27 instructs CD4+ T cells to provide help for the memory CD8+ T cell response after protein immunization.
J Immunol
2008
, vol. 
181
 
2
(pg. 
1071
-
1082
)
111
Halwani
 
R
Boyer
 
JD
Yassine-Diab
 
B
, et al. 
Therapeutic vaccination with simian immunodeficiency virus (SIV)-DNA + IL-12 or IL-15 induces distinct CD8 memory subsets in SIV-infected macaques.
J Immunol
2008
, vol. 
180
 
12
(pg. 
7969
-
7979
)
112
Villinger
 
F
Miller
 
R
Mori
 
K
, et al. 
IL-15 is superior to IL-2 in the generation of long-lived antigen specific memory CD4 and CD8 T cells in rhesus macaques.
Vaccine
2004
, vol. 
22
 
25–26
(pg. 
3510
-
3521
)
113
Kutzler
 
MA
Robinson
 
TM
Chattergoon
 
MA
, et al. 
Coimmunization with an optimized IL-15 plasmid results in enhanced function and longevity of CD8 T cells that are partially independent of CD4 T cell help.
J Immunol
2005
, vol. 
175
 
1
(pg. 
112
-
123
)
114
Kraynyak
 
KA
Kutzler
 
MA
Cisper
 
NJ
, et al. 
Plasmid-encoded interleukin-15 receptor alpha enhances specific immune responses induced by a DNA vaccine in vivo.
Hum Gene Ther
2009
, vol. 
20
 
10
(pg. 
1143
-
1156
)
115
Guillonneau
 
C
Mintern
 
JD
Hubert
 
FX
, et al. 
Combined NKT cell activation and influenza virus vaccination boosts memory CTL generation and protective immunity.
Proc Natl Acad Sci U S A
2009
, vol. 
106
 
9
(pg. 
3330
-
3335
)
116
Grossmann
 
C
Tenbusch
 
M
Nchinda
 
G
, et al. 
Enhancement of the priming efficacy of DNA vaccines encoding dendritic cell-targeted antigens by synergistic Toll-like receptor ligands.
BMC Immunol
2009
, vol. 
10
 pg. 
43
 
117
Scarlett
 
UK
Cubillos-Ruiz
 
JR
Nesbeth
 
YC
, et al. 
In situ stimulation of CD40 and Toll-like receptor 3 transforms ovarian cancer-infiltrating dendritic cells from immunosuppressive to immunostimulatory cells.
Cancer Res
2009
, vol. 
69
 
18
(pg. 
7329
-
7337
)
118
Dolfi
 
DV
Boesteanu
 
AC
Petrovas
 
C
Xia
 
D
Butz
 
EA
Katsikis
 
PD
Late signals from CD27 prevent Fas-dependent apoptosis of primary CD8+ T cells.
J Immunol
2008
, vol. 
180
 
5
(pg. 
2912
-
2921
)
119
Kocak
 
E
Lute
 
K
Chang
 
X
, et al. 
Combination therapy with anti-CTL antigen-4 and anti-4-1BB antibodies enhances cancer immunity and reduces autoimmunity.
Cancer Res
2006
, vol. 
66
 
14
(pg. 
7276
-
7284
)
120
Ha
 
SJ
West
 
EE
Araki
 
K
Smith
 
KA
Ahmed
 
R
Manipulating both the inhibitory and stimulatory immune system towards the success of therapeutic vaccination against chronic viral infections.
Immunol Rev
2008
, vol. 
223
 (pg. 
317
-
333
)
121
Krieg
 
C
Boyman
 
O
Fu
 
YX
Kaye
 
J
B and T lymphocyte attenuator regulates CD8+ T cell-intrinsic homeostasis and memory cell generation.
Nat Immunol
2007
, vol. 
8
 
2
(pg. 
162
-
171
)
122
Boullart
 
AC
Aarntzen
 
EH
Verdijk
 
P
, et al. 
Maturation of monocyte-derived dendritic cells with Toll-like receptor 3 and 7/8 ligands combined with prostaglandin E(2) results in high interleukin-12 production and cell migration.
Cancer Immunol Immunother
2008
, vol. 
57
 
11
(pg. 
1589
-
1597
)
123
Ahlers
 
JD
Belyakov
 
IM
Strategies for recruiting and targeting dendritic cells for optimizing HIV vaccines.
Trends Mol Med
2009
, vol. 
15
 
6
(pg. 
263
-
274
)
124
Mooij
 
P
Balla-Jhagjhoorsingh
 
SS
Beenhakker
 
N
, et al. 
Comparison of human and rhesus macaque T-cell responses elicited by boosting with NYVAC encoding human immunodeficiency virus type 1 clade C immunogens.
J Virol
2009
, vol. 
83
 
11
(pg. 
5881
-
5889
)
125
Todryk
 
SM
Pathan
 
AA
Keating
 
S
, et al. 
The relationship between human effector and memory T cells measured by ex vivo and cultured ELISPOT following recent and distal priming.
Immunology
2009
, vol. 
128
 
1
(pg. 
83
-
91
)
126
Johnson
 
S
Zhan
 
Y
Sutherland
 
RM
, et al. 
Selected Toll-like receptor ligands and viruses promote helper-independent cytotoxic T cell priming by upregulating CD40L on dendritic cells.
Immunity
2009
, vol. 
30
 
2
(pg. 
218
-
227
)
127
Choi
 
BK
Bae
 
JS
Choi
 
EM
, et al. 
4-1BB-dependent inhibition of immunosuppression by activated CD4+CD25+ T cells.
J Leukoc Biol
2004
, vol. 
75
 
5
(pg. 
785
-
791
)
128
Sabbagh
 
L
Pulle
 
G
Liu
 
Y
Tsitsikov
 
EN
Watts
 
TH
ERK-dependent Bim modulation downstream of the 4-1BB-TRAF1 signaling axis is a critical mediator of CD8 T cell survival in vivo.
J Immunol
2008
, vol. 
180
 
12
(pg. 
8093
-
8101
)
129
Hendriks
 
J
Xiao
 
Y
Rossen
 
JW
, et al. 
During viral infection of the respiratory tract, CD27, 4-1BB, and OX40 collectively determine formation of CD8+ memory T cells and their capacity for secondary expansion.
J Immunol
2005
, vol. 
175
 
3
(pg. 
1665
-
1676
)
130
Keller
 
AM
Groothuis
 
TA
Veraar
 
EA
, et al. 
Costimulatory ligand CD70 is delivered to the immunological synapse by shared intracellular trafficking with MHC class II molecules.
Proc Natl Acad Sci U S A
2007
, vol. 
104
 
14
(pg. 
5989
-
5994
)
131
Taraban
 
VY
Rowley
 
TF
Tough
 
DF
Al-Shamkhani
 
A
Requirement for CD70 in CD4+ Th cell-dependent and innate receptor-mediated CD8+ T cell priming.
J Immunol
2006
, vol. 
177
 
5
(pg. 
2969
-
2975
)
132
Fos
 
C
Salles
 
A
Lang
 
V
, et al. 
ICOS ligation recruits the p50alpha PI3K regulatory subunit to the immunological synapse.
J Immunol
2008
, vol. 
181
 
3
(pg. 
1969
-
1977
)
133
Jones
 
RB
Ndhlovu
 
LC
Barbour
 
JD
, et al. 
Tim-3 expression defines a novel population of dysfunctional T cells with highly elevated frequencies in progressive HIV-1 infection.
J Exp Med
2008
, vol. 
205
 
12
(pg. 
2763
-
2779
)
Sign in via your Institution