Abstract
Immune reconstitution following haematopoietic stem cell transplantation (SCT) is an often slow and incomplete process that leads to increased risk of infection and malignant disease. Immunization in SCT is frequently unsuccessful due to the prolonged lymphopenia, especially of CD4 T cells, seen following transplant. The transfusion of T cells, also called ‘adoptive T-cell therapy’, has the potential to enhance anti-tumour and overall immunity, and augment vaccine efficacy in the post-transplant setting. Recent advances in tissue culture, cellular immunology and tumour biology are guiding new approaches to adoptive T-cell therapy. This chapter will discuss the challenges that face the field before adoptive T-cell therapy can be translated into routine clinical practice.
Keywords: T cell, immune reconstitution, lymphopenia, haematopoietic stem cell transplantation, immunotherapy
Stem cell transplantation (SCT) has a well-established role in the treatment of haematological malignancies. Both autologous and allogeneic SCT have demonstrated efficacy as therapy for patients with leukaemia, multiple myeloma and lymphoma. Additionally, the use of SCT is being explored for patients with autoimmune diseases. The post-transplant period is characterized by a prolonged period of immunodeficiency, leading to increased vulnerability to infection.1–3 In one study, the majority of allograft recipients experienced at least one late infection (>50 days to 2 years) after transplant.4 Multivariate analysis showed that infection was the dominant factor associated with non-relapse mortality. In patients with chronic myeloid leukaemia, transplantation with a T-cell-depleted graft has been associated with an increased risk of relapse.5 Several authors have reported a correlation between higher absolute CD4 and CD8 lymphocyte counts and improved disease-free and/or overall survival.6–8 Even after lymphocyte numbers recover, lymphocyte function is often impaired.9–11 These observations reinforce the importance of immune reconstitution in the overall effectiveness of transplantation.
Adoptive immunotherapy is the isolation, ex-vivo activation and infusion of antigen-specific or non-specific lymphocytes. Adoptive cellular therapy can be considered as a strategy aimed at tumour elimination through direct anti-neoplastic effects, or through indirect effects mediated by immunity directed against elements supporting tumour growth such as angiogenesis. Adoptive cellular therapy using autologous or allogeneic cell infusions may also have a role in replacing, repairing or enhancing the immune function damaged as a consequence of cytotoxic therapy. Analysis of the presently available clinical results suggests that, despite some disappointments, there is room for optimism that both adoptive immunotherapy and active immunotherapy (vaccination) may eventually become part of the therapeutic arsenal to prevent or combat cancer in a more efficient way. This chapter will describe the background, rationale, and current clinical use and experimental approach of adoptive cellular therapies to improve immune reconstitution in the setting of SCT for haematological neoplasms.
IMMUNODEFICIENCY FOLLOWING HAEMATOPOIETIC STEM CELL TRANSPLANTATION
In addition to compromising the ability of SCT patients to mount effective anti-tumour immune responses, post-transplant immune suppression clearly increases the risk for serious infections with varicella-zoster virus, cytomegalovirus and Streptococcus pneumoniae.12,13 Early recovery of lymphocytes and lymphocyte function has been linked to improved survival following both auto- and allotransplantation.6,14 In the immediate post-transplant period, lymphocyte restoration is achieved by expansion of mature T cells present in the graft, and not de-novo production from the thymus or bone marrow.15–17 CD4+ T-cell regeneration occurs via a thymus-dependent mechanism, while CD8+ T-cell regeneration occurs via a thymus-independent pathway.10,18 Therefore, after transplant, there is a prolonged deficiency of CD4+ T cells compared with CD8+ T cells, particularly in older patients, secondary to limited thymic regenerative capacity.17,19 While younger patients eventually recover thymic output, the thymic deficiency seen following transplantation may not be fully reversible in older patients.20,21
The CD4+ T-cell deficiency noted after transplant is particularly significant as several studies have demonstrated the importance of these cells in the stimulation of CD8+ T cells and the enhancement of antibody production by B cells. CD8+ T cells that engage antigen in the absence of CD4+ T cells develop normally but do not proliferate well and do not persist, becoming so-called ‘helpless T cells’.22 This phenomenon may be responsible for the poor cytotoxic CD8+ T-cell responses seen in human immunodeficiency virus (HIV),23 and could be operative in transplant patients. In addition to providing a critical stimulus for CD8+ T cells, CD4+ T cells are required for maximal antibody production.24 The importance of CD4+ T cells has been demonstrated in humans where responses to immunization and severity of infection have been correlated with CD4 counts specifically,25,26 and survival correlates with CD4 counts in patients after infection with HIV.25
Humoral immunity recovers more quickly than cellular immunity in the immediate post-transplant period; however, immunoglobulin subset levels are often suppressed such that protective immunity is compromised, and response to vaccination remains a real problem in SCT patients.27 In the setting of allogeneic SCT, immunization of the donor has led to increased titres of Haemophilus influenzae type B and tetanus toxoid antibodies.28,29 Similar findings were noted in patients undergoing autologous SCT who underwent pre-transplant immunizations.30 In this age when so many promising tumour vaccines are in clinical trials, strategies to optimize responses to vaccination in SCT patients have become increasingly important.
ALLOGENEIC T-CELL THERAPY
The first form of human adoptive T-cell therapy was the passenger T cells present in stem cell infusions from bone marrow harvests. In a retrospective analysis, Weiden et al showed that the probability of tumour recurrence was lower for allogeneic transplant recipients compared with those who received syngeneic grafts.31 It is now well established that allogeneic SCT and donor lymphocyte infusions (DLIs) are an effective, but potentially toxic, form of non-specific immunotherapy in chronic myeloid leukaemia,32 although clinical responses for other malignancies have been disappointing.33,34 Early studies showed that the infusion of donor T cells soon after a myeloablative transplant conditioning regimen resulted in the marked augmentation of acute graft-versus-host disease (GvHD).35 In the setting of allogeneic haematopoietic SCT, Kernan et al have shown that the initial dose of infused T cells and the time from initial transplant have a major effect on the incidence and severity of acute GvHD.36 It has only recently been appreciated that the incidence of acute GvHD may be lower following the infusion of donor T cells in the setting of non-myeloablative SCT.37 Studies by Dazzi et al show that in the steady-state setting of relapsed chronic myeloid leukaemia, infusions of resting T cells result in a decreased incidence of GvHD when given by dose fractionation, starting with low doses of donor cells and escalating subsequent doses as required.38 In a non-randomized trial, they compared a bulk, single DLI (average 1.5 × 108 CD3+ cells/kg) with an escalating dose regimen of DLI, where increasing numbers of cells (average total 1.9 × 108 CD3+ cells/kg) were given at 20-week average intervals between infusion. They found that anti-leukaemic effects were preserved but that the incidence of GvHD was much lower using the escalating dose regimen of DLI. The authors have recently reported on a Phase I trial where patients with aggressive haematological malignancies received induction chemotherapy and conventional DLI (median 1.5 × 108 mononuclear cells/kg), followed 12 days later by ex-vivo, CD3/CD28-activated DLI (aDLI). aDLI was dose escalated from 1 × 106 to 1 × 108 CD3+ cells/kg in five levels. Out of 17 evaluable patients, seven developed acute GvHD and four developed chronic GvHD. Eight patients achieved complete remission. This study demonstrated that adoptive transfer of costimulated activated allogeneic T cells is feasible, does not result in excessive GvHD, and may contribute to durable remission in diseases where conventional DLI has been disappointing.39
PRINCIPLES OF T-CELL GROWTH
Adoptive cellular therapy depends on the ability to optimally select or genetically produce cells with the desired antigenic specificity, and then induce cellular proliferation while preserving the effector function, engraftment and homing abilities of the lymphocytes. Unfortunately, many previous clinical trials were carried out with adoptively transferred cells that were propagated in what are now understood to be suboptimal conditions that impair the essential functions of the adoptively transferred cells. Our greater understanding of the process of T-cell activation mediated through cell surface receptors and proteins now indicates that this is a complex multistaged process of recognition, adhesion and stimulation. In vivo, the generation of antigen-specific T cells requires the interaction of dendritic cells (DCs) and naïve T cells in a secondary lymphoid organ, usually a lymph node.40
For over 50 years, immunologists have sought to understand how the balance between immune activation and self tolerance is induced and maintained. Bretscher and Cohn first proposed a two-signal model of B-lymphocyte activation that was later modified by Lafferty and Cunningham for T-cell activation and allograft rejection.41 The essential features of these models were that activation of lymphocytes requires an antigen-specific signal, termed ‘Signal 1’, as well as a second antigen-non-specific event, termed ‘Signal 2’. Moreover, these theories and later modifications proposed that Signal 1 in the absence of Signal 2 led to tolerance or apoptosis. Indeed, in some instances, the binding of tumour antigen to the T-cell receptor in the absence of costimulation not only fails to activate the cell but also leads to functional inactivation.42 It is now appreciated that antigenic stimulation of T cells leads to at least three distinct outcomes: (1) activation, clonal expansion and differentiation to produce cells that secrete distinct subsets of cytokines or to express lytic machinery; (2) induction of an unresponsive state termed ‘anergy’; and (3) induction of apoptosis.43,44
Striving for optimal T-cell function
T cells exist in several distinct stages of differentiation. Naive CD4+ and CD8+ T cells undergo unique developmental programmes after antigen activation, resulting in the generation of effector memory and long-lived central memory T cells (TEM cells and TCM cells, respectively). Three models by which memory CD8+ T cells can be generated have been proposed. In the linear differentiation model, an autonomous antigen-triggered differentiation process consisting of conversion from naïve to effector to TEM cell occurs, followed by the appearance of TCM cells after antigen clearance through a process of dedifferentiation.45 The signal strength model proposes that naïve T cells progress through hierarchical thresholds for proliferation and differentiation as the strength and duration of the interaction with antigen-presenting cells (APCs) is increased.46 T cells receiving the weakest signals do not survive, whereas high-intensity signalling causes the development of terminally differentiated effector T cells that cannot survive into the memory phase. The TCM cells, being the least differentiated of the antigen-stimulated T cells, retain the developmental options of naïve T cells, including their capacity for marked clonal expansion. The stem cell model proposes that the cells within the TCM cell compartment are self-renewing and serve as a source of effector T cells.47 Although the details and mechanisms of differentiation remain to be clarified, it is clear that the various T-cell memory subsets have specialized roles and that not all the subsets would be efficacious in the setting of adoptive T-cell therapy for the treatment of cancer.
At present, naïve T cells are not thought to be useful for adoptive transfer as they are unable to kill tumour cells. The demonstration by Sallusto et al48 that TCM and TEM cell subsets have discrete trafficking and functional properties has the potential to fundamentally alter approaches to adoptive T-cell therapy. In retrospect, previous clinical trials have primarily tested adoptively transferred populations of CD8+ TEM cells.49 This approach was taken because available tissue culture technologies resulted in rapid differentiation of T cells to late-stage effector cells; late-stage TEM cells express CD57 and have poor replicative capacity.50,51 In vitro, TEM cells are superior to TCM cells at tumour cytotoxicity. However, in vivo, TCM cells exhibit superior therapeutic effects when compared with TEM cells on a per cell basis.52 Therefore, in principle, adoptive T-cell transfer strategies are attractive for the ability to generate long-lived populations of central memory T cells capable of immunosurveillance as well as tumour eradication, which is why efforts to test TCM cells in clinical trials are currently of high priority.
Factors determining the optimal CD8+ T cells for adoptive transfer
The cellular basis of immunological memory has been one of the central issues of immunology for more than 50 years. Many studies in mice indicate that true memory with long-lived T cells is only established in the absence of persistent antigen exposure.53,54 This raises a paradox for tumour immunologists regarding whether and how central memory can be established in tumour-bearing patients. Relevant to this issue is that in patients at risk of developing Epstein-Barr virus lymphoma, adoptively transferred gene-marked cytotoxic T lymphocytes (CTLs) persist for years,55 demonstrating that, in principle, it is possible to establish TCM in cancer patients by adoptive transfer. Therefore, to understand approaches to generate persistent immunity in patients where tumour antigens are unlikely to be eliminated completely, it might be more instructive to study the human immune response in patients with chronic persistent viral and parasitic infections.56,57 A major issue of relevance is the differentiation pathway leading to TCM cells. Some studies have indicated that naïve T cells first differentiate into effector T cells, a proportion of which then differentiate into TCM cells.58 In contrast to this linear differentiation model, other studies have suggested that parallel differentiation occurs, with naïve T cells differentiating directly into TCM and effector cells simultaneously through asymmetric division.59,60 Resolution of the above issues is important so that culture systems can be devised to optimally derive populations of TEM and TCM cells.
In humans, adoptive T-cell therapy approaches to date have used peripheral blood, the tumour bed, malignant effusions and draining lymph nodes as the anatomical source from which T cells are derived for adoptive transfer.61–64 Given the recent demonstration that bone marrow is a major reservoir of self-reactive CD8+ T cells,65 it is important to determine if improved anti-tumour effects are observed when bone-marrow-resident T cells are used for adoptive transfer. The bone marrow of breast cancer patients has been shown to contain CD8+ T cells specific for peptide epitopes from the tumour antigens MUC1 and Her-2/neu,66 and adoptive transfer of bone-marrow-derived human CD8+ memory T cells mediates anti-tumour activity in mice bearing tumour xenografts.50 Furthermore, bone marrow from patients with either pancreatic cancer or myeloma has also been shown to be enriched with tumour-reactive CD8+ T cells.67–70
A role for CD4+ T cells in adoptive T-cell transfer
Many studies have shown that the generation and/or maintenance of CD8+ T-cell memory requires CD4+ T-cell help,71 and that tumour-specific immunity is enhanced with CD4+ T-cell help, particularly in the absence of tumour expression of major histocompatibility complex (MHC) class II molecules72 as is commonly observed with aggressive haematological malignancies. Adoptively transferred CD4+ T cells have the potential to augment tumour immunity by several mechanisms that might enhance the survival and function of CD8+ T cells, including the secretion of essential cytokines such as interleukin (IL)-2 and IL-21,73 and the expression of CD40L.74 Besides their intimate involvement in priming tumour-specific CTLs, CD4+ cells participate in additional effector functions. Evidence indicates that other cytokines produced by CD4+ T cells can recruit and activate macrophages and eosinophils that, in turn, mediate anti-tumour effects.75 Clinical adoptive transfer studies also show that the persistence of adoptively transferred cytotoxic CD8+ effector T cells is enhanced with the concomitant administration of IL-276 or CD4+ T cells.77 Recent studies in patients with myeloma show that the adoptive transfer of mixed populations of pathogen-specific CD4+ and CD8+ T cells promoted the establishment of immunity with a robust central memory component.78 However, it is not yet known if this approach enhances the establishment of immunity to self-antigens in cancer patients.
T-cell development is highly dependent on the nature of the cytokine milieu, although species-specific differences exist relating to the primacy of individual cytokines in this process. The common gamma chain (γc) is a shared receptor component for the IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21 receptors. In the mouse, IL-15 is not produced by T cells, whereas human CD4+ memory T cells are reported to constitutively produce and proliferate in response to IL-15,79 suggesting different roles for IL-15 in mouse and human CD4+ memory T cell homeostasis.80 Genetic analysis of patients with a γc deficiency indicates that the human is absolutely dependent on cytokines that signal through γc for T-cell development, as T cells are absent in these individuals, whereas in γc-deficient mice, the spleens of older mice have nearly normal numbers of CD4+ T cells,81 indicating that γc-independent signals can support CD4+ T-cell development in mice but not humans. In mice, adoptively transferred CD8+ T cells cultured in IL-15 show more tumour cytotoxicity than those cultured in IL-2,49 so the use of IL-15 for adoptive T-cell therapy in humans holds substantial promise to augment T-cell numbers and effector functions, although species differences can be expected.
At present, one of the most important issues facing the field is the complexity of CD4+ lineage T cells. Until recently, T cells were separated into two different subsets named Th1 and Th2 cells, based on the pattern of cytokines that they produce when stimulated. Regulatory T cells (Tregs) represent a subset of thymic-derived T cells that exert an immunosuppressive influence and are thought to be essential for mediating tolerance. Tregs express IL-10 and tumour growth factor-β, but may exert inhibitory effects via direct cell contact. Several types of CD4+ Treg cell have now been described in humans,82 and it is probably important to remove Treg cells from adoptively transferred T-cell populations because they suppress anti-tumour immunity.83 Since Treg cells are often enriched in tumour-infiltrating lymphocytes and the peripheral blood in cancer patients,84 it is possible that the outcome of previous adoptive T-cell therapy clinical trials was compromised because the adoptively transferred T-cell populations inadvertently contained Treg cells.
Recently, a fourth axis to the CD4+ T-cell lineage was recognized, as a new IL-23-driven subset of IL-17-producing CD4+ T cells called ‘Th17’ has been described.85 Th17 cells are pro-inflammatory in some settings, but paradoxically may exert an immunosuppressive effect in the setting of malignancy. An interesting finding is that tumours express IL-23, which may promote the expansion of Th17 cells, resulting in decreased immunosurveillance of tumours by preventing Th1-cell-driven CD8+ T-cell infiltration.86 However, as evidence from several groups indicates that IL-17-producing Th17 cells represent the key effector cells in the induction of autoimmunity, rather than, as once was thought, interferon-γ-producing Th1 cells,87 it remains possible that adoptive transfer of Th17 cells might augment anti-tumour responses. Thus, early in the evolution of tumours, Th17 cells may have a pro-carcinogenic effect, while Th17 cells may have a pharmacological ability to treat established tumours.88
DEVELOPING OPTIMAL CELL CULTURE SYSTEMS
The only form of adoptive cellular therapy routinely employed in the practice of medicine is allogeneic bone marrow or peripheral blood SCT.89 In this setting, the establishment of donor haematopoiesis results in a graft-versus-leukaemia (GvL) effect mediated by donor lymphocytes. The administration of donor leukocyte infusions in the post-transplant period has also been shown to result in the regression of persistent or recurrent disease.90 The adoptive transfer of donor (allogeneic) T cells undergoing ex-vivo activation has promise to augment this effect.39,91 Ex-vivo culture approaches to alter the ratio of effector T cells and Tregs have the potential to decrease the risk of GvHD while preserving anti-tumour effects.92 A clear separation of GvHD and GvL has been demonstrated by the infusion of Tregs in leukaemia-bearing mice.93 However, whether this strategy will work in humans is unknown. Since allogeneic transplantation is associated with significant treatment-related morbidity and mortality, adoptive transfer of tumour-specific autologous T cells has been examined. A central issue for the development of clinical adoptive T-cell therapy strategies has been the development of culture systems in order to produce adequate numbers of effector T cells for autologous therapy.
Two basic approaches are being tested for clinical adoptive T-cell therapy. The first approach has been to isolate and activate in-vitro antigen-specific T cells from peripheral blood or tumour specimens, and then to use repetitive stimulation to clonally expand the antigen-specific T cells in vitro by various approaches. In the second approach, polyclonal ex-vivo activation of the T cells is done based on three assumptions: first, tumour-specific T cells are present in the patient; second, the tumour-specific T cells have been primed in the patient; and third, the in-vivo function of the tumour-specific T cells in the patient is impaired. In the second approach, the cells are activated in a polyclonal fashion by various means in vitro, and are then re-infused into the patient with the expectation that they will respond directly to the tumour or to tumour antigens presented by APCs. The first approach guarantees antigen specificity but is costly and labour intensive, and the second approach is technically more rapid and feasible. In practical terms, only the second approach has been sufficiently robust to support randomized clinical trials.64,78 The rationale for the second approach has been strengthened substantially by the realization that many patients are already primed to their tumour,94–96 and that the major challenge is improving the quality and quantity of the natural immune response.97 However, interest in both approaches has been reinforced by the realization that antigen-independent expansion of the transferred memory T cells can occur in vivo98 under certain conditions.
Strategies to present antigen to T cells ex vivo
The most appropriate methods of ex-vivo T-cell culture mimic the physiological processes whereby DCs generate a constellation of antigen-specific and costimulatory signals in the T cells. DCs are the most efficient APCs for the activation of naïve T cells. However, although useful for therapeutic vaccination, the generation of DCs is laborious and expensive, and yield and potency vary between patients. Therefore, DCs may not be optimal as APCs for large-scale adoptive T-cell therapy trials. In addition, DCs have limited replicative potential; for ex-vivo expansion of autologous T cells, it is desirable to have APCs with extensive replicative potential to facilitate both the scale-up of the process and multiple rounds of T-cell stimulation. As noted previously, since many patients are already primed to their tumours, other less potent forms of APC might suffice to induce T-cell activation. The best results to date have been with the rapid expansion method developed by Riddell and Greenberg, which uses irradiated allogeneic peripheral blood mononuclear cells as APCs (also known as feeder cells) to expand CTLs for adoptive transfer.99 The main limitation of this approach is in scale-up. Schultze et al have shown that CD40-stimulated B cells, which have extensive replicative potential, are an efficient means to propagate antigen-specific T cells.100 Thus, while currently available tissue culture approaches have provided proof of concept for adoptive therapy, a current priority is to develop alternative approaches that can support large-scale trials.
To generate antigen-specific T cells, cell lines and beads can be engineered to create artificial APCs and avoid the need to use autologous APCs for patient-specific cultures.101 General approaches have been to produce artificial APCs, either by coating beads with CD3-specific antibody or peptide–MHC complexes, or by transfecting cells that lack endogenous MHC molecules with MHC molecules and costimulatory molecules. Enhanced polyclonal T-cell activation and proliferation results when cells are stimulated through T-cell receptors and CD28.102 In addition, CD28 stimulation maintains telomere length in human T cells, and this might improve engraftment and persistence of adoptively transferred T cells.103,104 This culture system has been adapted for clinical use, and starting with an initial apheresis product, it is possible to generate the number of mature T cells found in an adult within 2 weeks of ex-vivo culture.105,106
Magnetic beads coated with MHC class I molecules loaded with specific peptide have been used to elicit antigen-specific T-cell propagation.107 Following isolation and expansion, cell populations generated using such beads specifically kill antigen-expressing target cells in vitro, and display anti-viral therapeutic effects in rodents.108 Others have used non-magnetic microspheres coated with complexes of recombinant peptide-loaded MHC molecules to successfully generate CTLs ex vivo from naïve precursors.109 Peptide–MHC tetramers presenting peptides from the melanoma tumour antigens melanoma-associated antigen recognized by T cells 1 (MART1) and glycoprotein 100 (gp100) have also been used to isolate high-avidity tumour-reactive CD8+ T cells from a heterogeneous population by flow cytometry. The tetramer-reactive cells could be cloned and retained their functional activity upon re-stimulation.110,111 Latouche and Sadelain have engineered APCs that could be used to stimulate T cells of any patient expressing a specific human leukocyte antigen (HLA) allele.112 Mouse fibroblasts were transduced retrovirally with a single HLA class I complex along with the human accessory molecules CD80 (also known as B7-1), CD54 (also known as ICAM-1) and CD58 (also known as LFA-3). These artificial APCs consistently elicited and expanded CTLs from patients of the appropriate haploype specific for MART1 and gp100. The authors have also found that artificial APCs that express 4-1BB ligand efficiently expand human central memory CD8+ T cells that have potent cytolytic function,107,113,114 and others have shown that CD83 expression on artificial APCs enhances the generation of CTLs.115
ADOPTIVE CELLULAR THERAPY: DOSE AND SCHEDULING ISSUES
Information on the dose and schedule dependence of adoptively transferred cells is widely scattered in the literature, such that there is no standardized system of administration. There is evidence in non-lymphopenic hosts suggesting that fractionated doses of adoptively transferred T cells are superior to a single infusion of T cells.116 The ideal dose of transferred cells is related to the tumour burden, and the homing and persistence (memory) characteristics of the infused cells.117 Doses of adoptively transferred cells are usually reported as the total number of viable cells administered, or as the total number of viable cells per kg body weight or per square meter body surface area. However, total lymphocyte numbers do not correlate well with body surface area, but rather display a stronger inverse correlation with age. Other variables add to the complexity, particularly the fact that, in the case of T cells or other adoptively transferred cells with high replicative potential, the infused dose may not relate well to the steady-state number of cells. Therefore, dose considerations are more complex than in other areas of transfusion medicine, where, for example, the maximal level of transfused red cells or platelets occurs immediately following infusion. Several recent studies have shown that severe lymphodepletion of the patient prior to cell administration can create optimal conditions to promote anti-tumour effects, and post-transplant adoptive T-cell transfer can take advantage of the homeostatic proliferation that occurs during this time.118
Schedule-dependent efficacy and adverse effects from adoptively transferred cells have been reported. Many studies in rodent tumour models show that the administration of cytotoxic therapy can enhance the effects of adoptively transferred cells through mechanisms that are independent of direct tumour cytoreduction. In studies with cyclophosphamide, enhancement of cellular immunotherapy is due to multiple effects, including: (1) killing of host regulatory lymphocytes that suppress anti-tumour immune responses;119,120 (2) creating ‘space’ in the host so that the adoptively transferred cells can engraft;117 and perhaps (3) enhanced cross-priming of tumour antigens. In contrast, it is probably best, when feasible, to harvest autologous T cells for ex-vivo expansion prior to initiation of chemotherapy. This is because adults have limited regeneration of T cells from the thymus, and therefore, the repertoire remains contracted for long periods of time and, in many cases, never recovers.19,121 Naïve T cells are most sensitive to the effects of cytotoxic chemotherapy, and their numbers are severely depleted in heavily pretreated patients. It is not yet known whether the tumour-specific T cells are derived from primed or naïve T cells in the host, and this likely varies depending on the intrinsic immunogenicity of the tumour. Studies with tetramers binding tumour peptides presented in the context of MHC molecules show that, in some patients, chemotherapy can ablate the tumour-specific T cells that have an effector phenotype, while sparing memory cells.122 The mechanism for this observation is unknown, but the authors speculated that the effector cells were in the active phases of the cell cycle, and were therefore rendered relatively susceptible to the cytotoxic effects of chemotherapy. If these results are confirmed, they would argue that patients should have their repertoire ‘archived’ by apheresis before undergoing chemotherapy.
HOST LYMPHODEPLETION
The post-transplant setting is an ideal platform for adoptive immunotherapy strategies to capitalize on homeostatic T-cell proliferation,98 in which naive T cells begin to proliferate and differentiate into memory-like T cells when total numbers of naive T cells are reduced below a certain threshold.123,124 Host lymphodepletion may enhance the effectiveness of adoptively transferred T cells.98 Homeostatic T-cell proliferation can result in the induction of auto-immunity,125 providing a clue to improved anti-tumour strategies. T cells can undergo up to seven rounds of cell division after being deprived of contact with APCs.45,126 This homeostatic response of T cells is directed largely to self-antigens.127 This hypothesis has been tested clinically in patients with metastatic melanoma refractory of conventional treatments.62 In this study, a high rate of clinical disease regression was observed in patients who received a lymphodepleting conditioning regimen consisting of cyclophosphamide (60 mg/kg × 2 days) and fludarabine (25 mg/m2 × 5 days) prior to adoptive transfer of T cells.
The authors have completed two Phase I trials in patients transplanted for haematological malignancies using autologous T cells stimulated by coculture with immunomagnetic beads to which anti-CD3 and anti-CD28 monoclonal antibodies had been conjugated. In the first trial, patients with relapsed or refractory non-Hodgkin’s lymphoma were treated with CD34+-selected haematopoietic SCT followed by infusion of up to 1 × 1010 CD3+ autologous T cells at Day 14 after transplant.128 Infusion of autologous costimulated T cells resulted in a rapid, dose-dependent reconstitution of lymphocyte counts. Importantly, the expanded cells were functionally superior to those obtained directly from the patients, as determined by interferon-gamma induction. Complete or partial responses were observed in 10 of the 16 patients infused. ALC recovery was compared with historical controls and found to be superior, but the clinical response was not compared.
In a similar Phase I/II trial, the authors examined the role of pre-transplant immunization and T-cell add-back in autologous transplantation for multiple myeloma.78 All patients received two doses of Prevnar, the 7-valent pneumococcal conjugate vaccine (PCV), beginning 1 month after transplant. Half of the patients received an additional PCV vaccine 2 weeks prior to a steady-state leukapheresis. The harvested T cells were expanded in vitro using the beads described above. Patients received a standard, non-lymphocyte-depleted, autologous SCT after melphalan conditioning, and then received up to 1 × 1010 autologous, expanded CD3+ T cells either 14 days or 100 days after transplantation. As expected, early T-cell recovery was observed in both patient groups that received T-cell add-back on Day 14, while the groups that received add-back on Day 100 were significantly lymphopenic. The authors also found that only those individuals who received PCV-primed T cells early after transplant developed and maintained protective levels of anti-pneumococcal antibodies, as well as PCV-specific CD4 responses. Notably, T-cell responses to antigens not included in the vaccine were also improved in this group. These data demonstrated that combination immunotherapy consisting of a single early post-transplant infusion of antigen-primed, ex-vivo costimulated autologous T cells followed by post-transplant booster immunizations improved the severe immunodeficiency associated with high-dose chemotherapy, and led to clinically relevant immunity in adults within 1 month following transplantation. This pilot study provided a ‘proof-of-concept’ and allowed the authors to extend their studies using activated T cells and cancer vaccines. Based on these early results, a new clinical trial was designed utilizing the ‘prime-boost’ strategy of pre-T-cell harvest vaccination followed by post-transplant booster vaccines. In this Phase I/II trial, myeloma patients are immunized with a multipeptide vaccine composed of HLA-restricted peptides from human telomerase (!hTERT), survivin and cytomegalovirus. Survivin and hTERT are both overexpressed in a number of malignancies, including myeloma.129,130 The vaccinations are followed by T-cell harvest, autologous SCT and infusion of ∼5 × 1010 expanded, autologous T cells at Day +2 after transplantation. The patients go on to receive three additional peptide immunizations. Thus far, T-cell numeric recovery is significantly greater in comparison with the two previous trials. Studies are in progress to determine the mechanism of the enhanced lymphocytosis and response to the peptide vaccine.
SUMMARY
The prolonged immune dysfunction seen after haematopoietic SCT can have devastating consequences, including serious viral and bacterial infection, and tumour relapse secondary to impaired immune surveillance. At the same time, the lymphopenia observed during this period may provide an optimal environment for the adoptive transfer of T cells capable of generating an anti-tumour response and repairing overall immunity. However, although adoptive T-cell therapy of rodent malignancies was first reported in 1955,131 no forms of T-cell therapy have been approved by the US Food and Drug Administration after more than 60 years of research into adoptive immunity for tumours. Still, there is increasing optimism that the scientific barriers preventing clinically effective adoptive immunotherapy have been addressed. Advances in the understanding of T-cell biology and the tumour micro-environment have provided multiple novel adoptive transfer strategies that are now poised for translation into clinical trials. Finally, it is likely that adoptive immunotherapy will not be used alone, but rather in combination with other forms of immunotherapy and chemotherapy to maximize both passive and active immunity.
Practice points
haematopoietic SCT recipients are at increased risk for infectious complications in the immediate and late post-transplant periods
successful immunization of transplant recipients relies on immune reconstitution of both T and B cells
adoptive T-cell therapy in the immediate post-transplant period enhances quantitative and functional immune recovery, and may allow vaccine introduction and promote tumour immunity
the dose and scheduling of adoptive T-cell therapy in the post-transplant setting has yet to be optimized, so careful monitoring for adverse events is an essential component of this treatment modality
Research agenda
while DLI has proven efficacy for the treatment of chronic myeloid leukaemia, autologous T-cell infusions should still be viewed as experimental, and only offered within the context of a clinical trial
determine optimal timing and doses of adoptively transferred T cells in patients after SCT to augment the effects of host lymphodepletion
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
REFERENCES
- 1.Lum LG. The kinetics of immune reconstitution after human marrow transplantation. Blood. 1987;69:369–380. [PubMed] [Google Scholar]
- 2.Storek J, Gooley T, Witherspoon RP, et al. Infectious morbidity in long-term survivors of allogeneic marrow transplantation is associated with low CD4 T cell counts. Am J Hematol. 1997;54:131–138. doi: 10.1002/(sici)1096-8652(199702)54:2<131::aid-ajh6>3.0.co;2-y. [DOI] [PubMed] [Google Scholar]
- 3.Storek J, Espino G, Dawson MA, et al. Low B-cell and monocyte counts on day 80 are associated with high infection rates between days 100 and 365 after allogeneic marrow transplantation. Blood. 2000;96:3290–3293. [PubMed] [Google Scholar]
- 4.Ochs L, Shu XO, Miller J, et al. Late infections after allogeneic bone marrow transplantations: comparison of incidence in related and unrelated donor transplant recipients. Blood. 1995;86:3979–3986. [PubMed] [Google Scholar]
- 5.Goldman JM, Gale RP, Horowitz MM, et al. Bone marrow transplantation for chronic myelogenous leukemia in chronic phase. Increased risk for relapse associated with T-cell depletion. Ann Intern Med. 1988;108:806–814. doi: 10.7326/0003-4819-108-6-806. [DOI] [PubMed] [Google Scholar]
- 6.*Porrata LF, Gertz MA, Inwards DJ, et al. Early lymphocyte recovery predicts superior survival after autologous hematopoietic stem cell transplantation in multiple myeloma or non-Hodgkin lymphoma. Blood. 2001;98:579–585. doi: 10.1182/blood.v98.3.579. [DOI] [PubMed] [Google Scholar]
- 7.Porrata LF, Gertz MA, Litzow MR, et al. Early lymphocyte recovery predicts superior survival after autologous hematopoietic stem cell transplantation for patients with primary systemic amyloidosis. Clin Cancer Res. 2005;11:1210–1218. [PubMed] [Google Scholar]
- 8.Porrata LF, Ingle JN, Litzow MR, et al. Prolonged survival associated with early lymphocyte recovery after autologous hematopoietic stem cell transplantation for patients with metastatic breast cancer. Bone Marrow Transplant. 2001;28:865–871. doi: 10.1038/sj.bmt.1703236. [DOI] [PubMed] [Google Scholar]
- 9.Nordoy T, Husebekk A, Aaberge IS, et al. Humoral immunity to viral and bacterial antigens in lymphoma patients 4–10 years after high-dose therapy with ABMT. Serological responses to revaccinations according to EBMT guidelines. Bone Marrow Transplant. 2001;28:681–687. doi: 10.1038/sj.bmt.1703228. [DOI] [PubMed] [Google Scholar]
- 10.Mackall CL, Stein D, Fleisher TA, et al. Prolonged CD4 depletion after sequential autologous peripheral blood progenitor cell infusions in children and young adults. Blood. 2000;96:754–762. [PubMed] [Google Scholar]
- 11.Small TN, Papadopoulos EB, Boulad F, et al. Comparison of immune reconstitution after unrelated and related T-cell-depleted bone marrow transplantation: effect of patient age and donor leukocyte infusions. Blood. 1999;93:467–480. [PubMed] [Google Scholar]
- 12.Hoyle C, Goldman JM. Life-threatening infections occurring more than 3 months after BMT. 18 UK Bone Marrow Transplant Teams. Bone Marrow Transplant. 1994;14:247–252. [PubMed] [Google Scholar]
- 13.Ketterer N, Espinouse D, Chomarat M, et al. Infections following peripheral blood progenitor cell transplantation for lymphoproliferative malignancies: etiology and potential risk factors. Am J Med. 1999;106:191–197. doi: 10.1016/s0002-9343(98)00409-4. [DOI] [PubMed] [Google Scholar]
- 14.Pavletic ZS, Joshi SS, Pirruccello SJ, et al. Lymphocyte reconstitution after allogeneic blood stem cell transplantation for hematologic malignancies. Bone Marrow Transplant. 1998;21:33–41. doi: 10.1038/sj.bmt.1701037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Roux E, Helg C, Dumont-Girard F, et al. Analysis of T-cell repopulation after allogeneic bone marrow transplantation: significant differences between recipients of T-cell depleted and unmanipulated grafts. Blood. 1996;87:3984–3992. [PubMed] [Google Scholar]
- 16.Abrahamsen IW, Somme S, Heldal D, et al. Immune reconstitution after allogeneic stem cell transplantation: the impact of stem cell source and graft-versus-host disease. Haematologica. 2005;90:86–93. [PubMed] [Google Scholar]
- 17.Hakim FT, Cepeda R, Kaimei S, et al. Constraints on CD4 recovery postchemotherapy in adults: thymic insufficiency and apoptotic decline of expanded peripheral CD4 cells. Blood. 1997;90:3789–3798. [PubMed] [Google Scholar]
- 18.Mackall CL, Fleisher TA, Brown MR, et al. Distinctions between CD8+ and CD4+ T-cell regenerative pathways result in prolonged T-cell subset imbalance after intensive chemotherapy. Blood. 1997;89:3700–3707. [PubMed] [Google Scholar]
- 19.*Mackall CL, Fleisher TA, Brown MR, et al. Age, thymopoiesis, and CD4+ Tlymphocyte regeneration after intensive chemotherapy. N Engl J Med. 1995;332:143–149. doi: 10.1056/NEJM199501193320303. [DOI] [PubMed] [Google Scholar]
- 20.Storek J, Joseph A, Espino G, et al. Immunity of patients surviving 20 to 30 years after allogeneic or syngeneic bone marrow transplantation. Blood. 2001;98:3505–3512. doi: 10.1182/blood.v98.13.3505. [DOI] [PubMed] [Google Scholar]
- 21.Roux E, Dumont-Girard F, Starobinski M, et al. Recovery of immune reactivity after T-cell-depleted bone marrow transplantation depends on thymic activity. Blood. 2000;96:2299–2303. [PubMed] [Google Scholar]
- 22.*Janssen EM, Droin NM, Lemmens EE, et al. CD4+ T-cell help controls CD8+ T-cell memory via TRAIL-mediated activation-induced cell death. Nature. 2005;434:88–93. doi: 10.1038/nature03337. [DOI] [PubMed] [Google Scholar]
- 23.Lichterfeld M, Kaufmann DE, Yu XG, et al. Loss of HIV-1-specific CD8+ T cell proliferation after acute HIV-1 infection and restoration by vaccine-induced HIV-1-specific CD4+ T cells. J Exp Med. 2004;200:701–712. doi: 10.1084/jem.20041270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Mozdzanowska K, Maiese K, Gerhard W. Th cell-deficient mice control influenza virus infection more effectively than Th- and B cell-deficient mice: evidence for a Th-independent contribution by B cells to virus clearance. J Immunol. 2000;164:2635–2643. doi: 10.4049/jimmunol.164.5.2635. [DOI] [PubMed] [Google Scholar]
- 25.Kroon FP, van Dissel JT, de Jong JC, et al. Antibody response to influenza, tetanus and pneumococcal vaccines in HIV-seropositive individuals in relation to the number of CD4+ lymphocytes. AIDS (London, England) 1994;8:469–476. doi: 10.1097/00002030-199404000-00008. [DOI] [PubMed] [Google Scholar]
- 26.Iorio AM, Francisci D, Camilloni B, et al. Antibody responses and HIV-1 viral load in HIV-1-seropositive subjects immunised with either the MF59-adjuvanted influenza vaccine or a conventional non-adjuvanted subunit vaccine during highly active antiretroviral therapy. Vaccine. 2003;21:3629–3637. doi: 10.1016/s0264-410x(03)00408-0. [DOI] [PubMed] [Google Scholar]
- 27.Avigan D, Pirofski LA, Lazarus HM. Vaccination against infectious disease following hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2001;7:171–183. doi: 10.1053/bbmt.2001.v7.pm11302551. [DOI] [PubMed] [Google Scholar]
- 28.Storek J, Dawson MA, Lim LC, et al. Efficacy of donor vaccination before hematopoietic cell transplantation and recipient vaccination both before and early after transplantation. Bone Marrow Transplant. 2004;33:337–346. doi: 10.1038/sj.bmt.1704336. [DOI] [PubMed] [Google Scholar]
- 29.Molrine DC, Guinan EC, Antin JH, et al. Donor immunization with Haemophilus influenzae type b (HIB)-conjugate vaccine in allogeneic bone marrow transplantation. Blood. 1996;87:3012–3018. [PubMed] [Google Scholar]
- 30.Molrine DC, Guinan EC, Antin JH, et al. Haemophilus influenzae type b (HIB)-conjugate immunization before bone marrow harvest in autologous bone marrow transplantation. Bone Marrow Transplant. 1996;17:1149–1155. [PubMed] [Google Scholar]
- 31.Weiden PL, Flournoy N, Thomas ED, et al. Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts. N Engl J Med. 1979;300:1068–1073. doi: 10.1056/NEJM197905103001902. [DOI] [PubMed] [Google Scholar]
- 32.Dazzi F, Szydlo RM, Cross NC, et al. Durability of responses following donor lymphocyte infusions for patients who relapse after allogeneic stem cell transplantation for chronic myeloid leukemia. Blood. 2000;96:2712–2716. [PubMed] [Google Scholar]
- 33.Collins RH, Jr., Goldstein S, Giralt S, et al. Donor leukocyte infusions in acute lymphocytic leukemia. Bone Marrow Transplant. 2000;26:511–516. doi: 10.1038/sj.bmt.1702555. [DOI] [PubMed] [Google Scholar]
- 34.Salama M, Nevill T, Marcellus D, et al. Donor leukocyte infusions for multiple myeloma. Bone Marrow Transplant. 2000;26:1179–1184. doi: 10.1038/sj.bmt.1702685. [DOI] [PubMed] [Google Scholar]
- 35.Sullivan KM, Storb R, Buckner CD, et al. Graft-versus-host disease as adoptive immunotherapy in patients with advanced hematologic neoplasms. N Engl J Med. 1989;320:828–834. doi: 10.1056/NEJM198903303201303. [DOI] [PubMed] [Google Scholar]
- 36.Kernan NA, Collins NH, Juliano L, et al. Clonable T lymphocytes in T cell-depleted bone marrow transplants correlate with development of graft-v-host disease. Blood. 1986;68:770–773. [PubMed] [Google Scholar]
- 37.Giralt S, Estey E, Albitar M, et al. Engraftment of allogeneic hematopoietic progenitor cells with purine analog-containing chemotherapy: harnessing graft-versus-leukemia without myeloablative therapy. Blood. 1997;89:4531–4536. [PubMed] [Google Scholar]
- 38.Dazzi F, Szydlo RM, Craddock C, et al. Comparison of single-dose and escalating-dose regimens of donor lymphocyte infusion for relapse after allografting for chronic myeloid leukemia. Blood. 2000;95:67–71. [PubMed] [Google Scholar]
- 39.Porter DL, Levine BL, Bunin N, et al. A phase 1 trial of donor lymphocyte infusions expanded and activated ex vivo via CD3/CD28 costimulation. Blood. 2006;107:1325–1331. doi: 10.1182/blood-2005-08-3373. [DOI] [PubMed] [Google Scholar]
- 40.Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245–252. doi: 10.1038/32588. [DOI] [PubMed] [Google Scholar]
- 41.Bretscher P, Cohn M. A theory of self-nonself discrimination. Science (New York, NY) 1970;169:1042–1049. doi: 10.1126/science.169.3950.1042. [DOI] [PubMed] [Google Scholar]
- 42.Chen L, Ashe S, Brady WA, et al. Costimulation of antitumor immunity by the B7 counterreceptor for the T lymphocyte molecules CD28 and CTLA-4. Cell. 1992;71:1093–1102. doi: 10.1016/s0092-8674(05)80059-5. [DOI] [PubMed] [Google Scholar]
- 43.Riley JL, June CH. The CD28 family: a T-cell rheostat for therapeutic control of T-cell activation. Blood. 2005;105:13–21. doi: 10.1182/blood-2004-04-1596. [DOI] [PubMed] [Google Scholar]
- 44.Lenschow DJ, Walunas TL, Bluestone JA. CD28/B7 system of T cell costimulation. Ann Rev Immunol. 1996;14:233–258. doi: 10.1146/annurev.immunol.14.1.233. [DOI] [PubMed] [Google Scholar]
- 45.Kaech SM, Ahmed R. Memory CD8+ T cell differentiation: initial antigen encounter triggers a developmental program in naive cells. Nat Immunol. 2001;2:415–422. doi: 10.1038/87720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Ann Rev Immunol. 2004;22:745–763. doi: 10.1146/annurev.immunol.22.012703.104702. [DOI] [PubMed] [Google Scholar]
- 47.Fearon DT, Manders P, Wagner SD. Arrested differentiation, the self-renewing memory lymphocyte, and vaccination. Science (New York, NY) 2001;293:248–250. doi: 10.1126/science.1062589. [DOI] [PubMed] [Google Scholar]
- 48.*Sallusto F, Lenig D, Forster R, et al. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. 1999;401:708–712. doi: 10.1038/44385. [DOI] [PubMed] [Google Scholar]
- 49.Klebanoff CA, Gattinoni L, Torabi-Parizi P, et al. Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proc Natl Acad Sci USA. 2005;102:9571–9576. doi: 10.1073/pnas.0503726102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Monteiro J, Batliwalla F, Ostrer H, et al. Shortened telomeres in clonally expanded CD28 CD8+ T cells imply a replicative history that is distinct from their CD28+CD8+ counterparts. J Immunol. 1996;156:3587–3590. [PubMed] [Google Scholar]
- 51.Van den Hove LE, Van Gool SW, Vandenberghe P, et al. CD57+/CD28- T cells in untreated hemato-oncological patients are expanded and display a Th1-type cytokine secretion profile, ex vivo cytolytic activity and enhanced tendency to apoptosis. Leukemia. 1998;12:1573–1582. doi: 10.1038/sj.leu.2401146. [DOI] [PubMed] [Google Scholar]
- 52.Berger C, Jensen MC, Lansdorp PM, et al. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J Clin Invest. 2008;118:294–305. doi: 10.1172/JCI32103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Ahmed R, Gray D. Immunological memory and protective immunity: understanding their relation. Science (New York, NY) 1996;272:54–60. doi: 10.1126/science.272.5258.54. [DOI] [PubMed] [Google Scholar]
- 54.Sprent J, Surh CD. T cell memory. Ann Rev Immunol. 2002;20:551–579. doi: 10.1146/annurev.immunol.20.100101.151926. [DOI] [PubMed] [Google Scholar]
- 55.Heslop HE, Ng CY, Li C, et al. Long-term restoration of immunity against Epstein-Barr virus infection by adoptive transfer of gene-modified virus-specific T lymphocytes. Nat Med. 1996;2:551–555. doi: 10.1038/nm0596-551. [DOI] [PubMed] [Google Scholar]
- 56.Champagne P, Ogg GS, King AS, et al. Skewed maturation of memory HIV-specific CD8 T lymphocytes. Nature. 2001;410:106–111. doi: 10.1038/35065118. [DOI] [PubMed] [Google Scholar]
- 57.Appay V, Dunbar PR, Callan M, et al. Memory CD8+ T cells vary in differentiation phenotype in different persistent virus infections. Nat Med. 2002;8:379–385. doi: 10.1038/nm0402-379. [DOI] [PubMed] [Google Scholar]
- 58.Opferman JT, Ober BT, Ashton-Rickardt PG. Linear differentiation of cytotoxic effectors into memory T lymphocytes. Science (New York, NY) 1999;283:1745–1748. doi: 10.1126/science.283.5408.1745. [DOI] [PubMed] [Google Scholar]
- 59.Holmes S, He M, Xu T, et al. Memory T cells have gene expression patterns intermediate between naive and effector. Proc Natl Acad Sci USA. 2005;102:5519–5523. doi: 10.1073/pnas.0501437102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Bouneaud C, Garcia Z, Kourilsky P, et al. Lineage relationships, homeostasis, and recall capacities of central- and effector-memory CD8 T cells in vivo. J Exp Med. 2005;201:579–590. doi: 10.1084/jem.20040876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kono K, Ichihara F, Iizuka H, et al. Expression of signal transducing T-cell receptor zeta molecules after adoptive immunotherapy in patients with gastric and colon cancer. Int J Cancer. 1998;78:301–305. doi: 10.1002/(SICI)1097-0215(19981029)78:3<301::AID-IJC7>3.0.CO;2-Z. [DOI] [PubMed] [Google Scholar]
- 62.Dudley ME, Wunderlich JR, Robbins PF, et al. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science (New York, NY) 2002;298:850–854. doi: 10.1126/science.1076514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Chang AE, Aruga A, Cameron MJ, et al. Adoptive immunotherapy with vaccine-primed lymph node cells secondarily activated with anti-CD3 and interleukin 2. J Clin Oncol. 1997;15:796–807. doi: 10.1200/JCO.1997.15.2.796. [DOI] [PubMed] [Google Scholar]
- 64.Takayama T, Sekine T, Makuuchi M, et al. Adoptive immunotherapy to lower postsurgical recurrence rates of hepatocellular carcinoma: a randomised trial. Lancet. 2000;356:802–807. doi: 10.1016/S0140-6736(00)02654-4. [DOI] [PubMed] [Google Scholar]
- 65.*Mazo IB, Honczarenko M, Leung H, et al. Bone marrow is a major reservoir and site of recruitment for central memory CD8+ T cells. Immunity. 2005;22:259–270. doi: 10.1016/j.immuni.2005.01.008. [DOI] [PubMed] [Google Scholar]
- 66.Feuerer M, Beckhove P, Bai L, et al. Therapy of human tumors in NOD/SCID mice with patient-derived reactivated memory T cells from bone marrow. Nat Med. 2001;7:452–458. doi: 10.1038/86523. [DOI] [PubMed] [Google Scholar]
- 67.Schmitz-Winnenthal FH, Volk C, Z’Graggen K, et al. High frequencies of functional tumor-reactive T cells in bone marrow and blood of pancreatic cancer patients. Cancer Res. 2005;65:10079–10087. doi: 10.1158/0008-5472.CAN-05-1098. [DOI] [PubMed] [Google Scholar]
- 68.Choi C, Witzens M, Bucur M, et al. Enrichment of functional CD8 memory T cells specific for MUC1 in bone marrow of patients with multiple myeloma. Blood. 2005;105:2132–2134. doi: 10.1182/blood-2004-01-0366. [DOI] [PubMed] [Google Scholar]
- 69.Dhodapkar MV, Krasovsky J, Olson K. T cells from the tumor microenvironment of patients with progressive myeloma can generate strong, tumor-specific cytolytic responses to autologous, tumor-loaded dendritic cells. Proc Natl Acad Sci USA. 2002;99:13009–13013. doi: 10.1073/pnas.202491499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Noonan K, Matsui W, Serafini P, et al. Activated marrow-infiltrating lymphocytes effectively target plasma cells and their clonogenic precursors. Cancer Res. 2005;65:2026–2034. doi: 10.1158/0008-5472.CAN-04-3337. [DOI] [PubMed] [Google Scholar]
- 71.Bevan MJ. Helping the CD8(+) T-cell response. Nat Rev. 2004;4:595–602. doi: 10.1038/nri1413. [DOI] [PubMed] [Google Scholar]
- 72.Ossendorp F, Mengede E, Camps M, et al. Specific T helper cell requirement for optimal induction of cytotoxic T lymphocytes against major histocompatibility complex class II negative tumors. J Exp Med. 1998;187:693–702. doi: 10.1084/jem.187.5.693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Li Y, Bleakley M, Yee C. IL-21 influences the frequency, phenotype, and affinity of the antigen-specific CD8 T cell response. J Immunol. 2005;175:2261–2269. doi: 10.4049/jimmunol.175.4.2261. [DOI] [PubMed] [Google Scholar]
- 74.Schoenberger SP, Toes RE, van der Voort EI, et al. T-cell help for cytotoxic T lymphocytes is mediated by CD40-CD40L interactions. Nature. 1998;393:480–483. doi: 10.1038/31002. [DOI] [PubMed] [Google Scholar]
- 75.Hung K, Hayashi R, Lafond-Walker A, et al. The central role of CD4(+) T cells in the antitumor immune response. J Exp Med. 1998;188:2357–2368. doi: 10.1084/jem.188.12.2357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Yee C, Thompson JA, Byrd D, et al. Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells. Proc Natl Acad Sci USA. 2002;99:16168–16173. doi: 10.1073/pnas.242600099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Einsele H, Roosnek E, Rufer N, et al. Infusion of cytomegalovirus (CMV)-specific T cells for the treatment of CMV infection not responding to antiviral chemotherapy. Blood. 2002;99:3916–3922. doi: 10.1182/blood.v99.11.3916. [DOI] [PubMed] [Google Scholar]
- 78.*Rapoport AP, Stadtmauer EA, Aqui N, et al. Restoration of immunity in lymphopenic individuals with cancer by vaccination and adoptive T-cell transfer. Nat Med. 2005;11:1230–1237. doi: 10.1038/nm1310. [DOI] [PubMed] [Google Scholar]
- 79.Miranda-Carus ME, Benito-Miguel M, Llamas MA, et al. Human T cells constitutively express IL-15 that promotes ex vivo T cell homeostatic proliferation through autocrine/juxtacrine loops. J Immunol. 2005;175:3656–3662. doi: 10.4049/jimmunol.175.6.3656. [DOI] [PubMed] [Google Scholar]
- 80.Prlic M, Lefrancois L, Jameson SC. Multiple choices: regulation of memory CD8 T cell generation and homeostasis by interleukin (IL)-7 and IL 15. J Exp Med. 2002;195:F49–F52. doi: 10.1084/jem.20020767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Nakajima H, Shores EW, Noguchi M, et al. The common cytokine receptor gamma chain plays an essential role in regulating lymphoid homeostasis. J Exp Med. 1997;185:189–195. doi: 10.1084/jem.185.2.189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Jiang H, Chess L. An integrated view of suppressor T cell subsets in immunoregulation. J Clin Invest. 2004;114:1198–1208. doi: 10.1172/JCI23411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.*Shimizu J, Yamazaki S, Sakaguchi S. Induction of tumor immunity by removing CD25+CD4+ T cells: a common basis between tumor immunity and autoimmunity. J Immunol. 1999;163:5211–5218. [PubMed] [Google Scholar]
- 84.Woo EY, Chu CS, Goletz TJ, et al. Regulatory CD4(+)CD25(+) T cells in tumors from patients with early-stage non-small cell lung cancer and late-stage ovarian cancer. Cancer Res. 2001;61:4766–4772. [PubMed] [Google Scholar]
- 85.*Harrington LE, Hatton RD, Mangan PR, et al. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol. 2005;6:1123–1132. doi: 10.1038/ni1254. [DOI] [PubMed] [Google Scholar]
- 86.Langowski JL, Zhang X, Wu L, et al. IL-23 promotes tumour incidence and growth. Nature. 2006;442:461–465. doi: 10.1038/nature04808. [DOI] [PubMed] [Google Scholar]
- 87.McKenzie BS, Kastelein RA, Cua DJ. Understanding the IL-23-IL-17 immune pathway. Trends Immunol. 2006;27:17–23. doi: 10.1016/j.it.2005.10.003. [DOI] [PubMed] [Google Scholar]
- 88.Muranski P, Boni A, Antony PA, et al. Tumor-specific Th17-polarized cells eradicate large established melanoma. Blood. 2008 doi: 10.1182/blood-2007-11-120998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Thomas ED. Bone marrow transplantation: a review. Semin Haematol. 1999;36(Suppl 7):95–103. [PubMed] [Google Scholar]
- 90.Barrett J, Jiang YZ. Allogeneic Immunotherapy for Malignant Diseases. Marcel Dekker, Inc; New York: 2000. [Google Scholar]
- 91.Fowler DH, Odom J, Steinberg SM, et al. Phase I clinical trial of costimulated, IL-4 polarized donor CD4+ T cells as augmentation of allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2006;12:1150–1160. doi: 10.1016/j.bbmt.2006.06.015. [DOI] [PubMed] [Google Scholar]
- 92.June CH, Blazar BR. Clinical application of expanded CD4+25+ cells. Semin Immunol. 2006;18:78–88. doi: 10.1016/j.smim.2006.01.006. [DOI] [PubMed] [Google Scholar]
- 93.Edinger M, Hoffmann P, Ermann J, et al. CD4+CD25+ regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation. Nat Med. 2003;9:1144–1150. doi: 10.1038/nm915. [DOI] [PubMed] [Google Scholar]
- 94.*Zhang L, Conejo-Garcia JR, Katsaros D, et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med. 2003;348:203–213. doi: 10.1056/NEJMoa020177. [DOI] [PubMed] [Google Scholar]
- 95.Germeau C, Ma W, Schiavetti F, et al. High frequency of antitumor T cells in the blood of melanoma patients before and after vaccination with tumor antigens. J Exp Med. 2005;201:241–248. doi: 10.1084/jem.20041379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.*Molldrem JJ, Lee PP, Wang C, et al. Evidence that specific T lymphocytes may participate in the elimination of chronic myelogenous leukemia. Nat Med. 2000;6:1018–1023. doi: 10.1038/79526. [DOI] [PubMed] [Google Scholar]
- 97.Coulie PG, Connerotte T. Human tumor-specific T lymphocytes: does function matter more than number? Curr Opin Immunol. 2005;17:320–325. doi: 10.1016/j.coi.2005.03.002. [DOI] [PubMed] [Google Scholar]
- 98.Dummer W, Niethammer AG, Baccala R, et al. T cell homeostatic proliferation elicits effective antitumor autoimmunity. J Clin Invest. 2002;110:185–192. doi: 10.1172/JCI15175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Riddell SR, Greenberg PD. Rapid Expansion Method (“REM”) for In Vitro Propagation of T Lymphocytes. US Patent. 1998
- 100.Schultze JL, Michalak S, Seamon MJ, et al. CD40-activated human B cells: an alternative source of highly efficient antigen presenting cells to generate autologous antigen-specific T cells for adoptive immunotherapy. J Clin Invest. 1997;100:2757–2765. doi: 10.1172/JCI119822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Kim JV, Latouche JB, Riviere I, et al. The ABCs of artificial antigen presentation. Nat Biotechnol. 2004;22:403–410. doi: 10.1038/nbt955. [DOI] [PubMed] [Google Scholar]
- 102.Levine BL, Bernstein WB, Connors M, et al. Effects of CD28 costimulation on long-term proliferation of CD4+ T cells in the absence of exogenous feeder cells. J Immunol. 1997;159:5921–5930. [PubMed] [Google Scholar]
- 103.Weng N, Levine BL, June CH, et al. Regulation of telomerase RNA template expression in human T lymphocyte development and activation. J Immunol. 1997;158:3215–3220. [PubMed] [Google Scholar]
- 104.Zhou J, Shen X, Huang J, et al. Telomere length of transferred lymphocytes correlates with in vivo persistence and tumor regression in melanoma patients receiving cell transfer therapy. J Immunol. 2005;175:7046–7052. doi: 10.4049/jimmunol.175.10.7046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Kalamasz D, Long SA, Taniguchi R, et al. Optimization of human T-cell expansion ex vivo using magnetic beads conjugated with anti-CD3 and Anti-CD28 antibodies. J Immunother. 2004;27:405–418. doi: 10.1097/00002371-200409000-00010. [DOI] [PubMed] [Google Scholar]
- 106.Levine BL, Cotte J, Small CC, et al. Large-scale production of CD4+ T cells from HIV-1-infected donors after CD3/CD28 costimulation. J Hematother. 1998;7:437–448. doi: 10.1089/scd.1.1998.7.437. [DOI] [PubMed] [Google Scholar]
- 107.Oelke M, Maus MV, Didiano D, et al. Ex vivo induction and expansion of antigen-specific cytotoxic T cells by HLA-Ig-coated artificial antigen-presenting cells. Nat Med. 2003;9:619–624. doi: 10.1038/nm869. [DOI] [PubMed] [Google Scholar]
- 108.Luxembourg AT, Borrow P, Teyton L, et al. Biomagnetic isolation of antigen-specific CD8+ T cells usable in immunotherapy. Nat Biotechnol. 1998;16:281–285. doi: 10.1038/nbt0398-281. [DOI] [PubMed] [Google Scholar]
- 109.Lone YC, Motta I, Mottez E, et al. In vitro induction of specific cytotoxic T lymphocytes using recombinant single-chain MHC class I/peptide complexes. J Immunother. 1998;21:283–294. doi: 10.1097/00002371-199807000-00006. [DOI] [PubMed] [Google Scholar]
- 110.Dunbar PR, Chen JL, Chao D, et al. Cutting edge: rapid cloning of tumor-specific CTL suitable for adoptive immunotherapy of melanoma. J Immunol. 1999;162:6959–6962. [PubMed] [Google Scholar]
- 111.Yee C, Savage PA, Lee PP, et al. Isolation of high avidity melanoma-reactive CTL from heterogeneous populations using peptide-MHC tetramers. J Immunol. 1999;162:2227–2234. [PubMed] [Google Scholar]
- 112.Latouche JB, Sadelain M. Induction of human cytotoxic T lymphocytes by artificial antigen-presenting cells. Nat Biotechnol. 2000;18:405–409. doi: 10.1038/74455. [DOI] [PubMed] [Google Scholar]
- 113.Maus MV, Kovacs B, Kwok WW, et al. Extensive replicative capacity of human central memory T cells. J Immunol. 2004;172:6675–6683. doi: 10.4049/jimmunol.172.11.6675. [DOI] [PubMed] [Google Scholar]
- 114.Suhoski MM, Golovina TN, Aqui N, et al. Engineering artificial antigen-presenting cells to express a diverse array of costimulatory molecules. Mol Ther. 2007;15:981–988. doi: 10.1038/mt.sj.6300134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Hirano N, Butler MO, Xia Z, et al. Engagement of CD83 ligand induces prolonged expansion of CD8+ T cells and preferential enrichment for antigen specificity. Blood. 2006;107:1528–1536. doi: 10.1182/blood-2005-05-2073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Kircher MF, Allport JR, Graves EE, et al. In vivo high resolution three-dimensional imaging of antigen-specific cytotoxic T-lymphocyte trafficking to tumors. Cancer Res. 2003;63:6838–6846. [PubMed] [Google Scholar]
- 117.Freitas AA, Rocha B. Peripheral T cell survival. Curr Opin Immunol. 1999;11:152–156. doi: 10.1016/s0952-7915(99)80026-0. [DOI] [PubMed] [Google Scholar]
- 118.*Williams KM, Hakim FT, Gress RE. T cell immune reconstitution following lymphodepletion. Semin Immunol. 2007;19:318–330. doi: 10.1016/j.smim.2007.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Melief CJ. Tumor eradication by adoptive transfer of cytotoxic T lymphocytes. Adv Cancer Res. 1992;58:143–175. doi: 10.1016/s0065-230x(08)60294-8. [DOI] [PubMed] [Google Scholar]
- 120.North RJ. Models of adoptive T-cell-mediated regression of established tumors. Contemp Topics Immunobiol. 1984;13:243–257. doi: 10.1007/978-1-4757-1445-6_12. [DOI] [PubMed] [Google Scholar]
- 121.Mackall CL, Gress RE. Pathways of T-cell regeneration in mice and humans: implications for bone marrow transplantation and immunotherapy. Immunol Rev. 1997;157:61–72. doi: 10.1111/j.1600-065x.1997.tb00974.x. [DOI] [PubMed] [Google Scholar]
- 122.Lee PP, Yee C, Savage PA, et al. Characterization of circulating T cells specific for tumor-associated antigens in melanoma patients. Nat Med. 1999;5:677–685. doi: 10.1038/9525. [DOI] [PubMed] [Google Scholar]
- 123.Goldrath AW, Bevan MJ. Low-affinity ligands for the TCR drive proliferation of mature CD8+ T cells in lymphopenic hosts. Immunity. 1999;11:183–190. doi: 10.1016/s1074-7613(00)80093-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Surh CD, Sprent J. Homeostatic T cell proliferation: how far can T cells be activated to self-ligands? J Exp Med. 2000;192:F9–F14. doi: 10.1084/jem.192.4.f9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.King C, Ilic A, Koelsch K, et al. Homeostatic expansion of T cells during immune insufficiency generates autoimmunity. Cell. 2004;117:265–277. doi: 10.1016/s0092-8674(04)00335-6. [DOI] [PubMed] [Google Scholar]
- 126.van Stipdonk MJ, Lemmens EE, Schoenberger SP. Naive CTLs require a single brief period of antigenic stimulation for clonal expansion and differentiation. Nat Immunol. 2001;2:423–429. doi: 10.1038/87730. [DOI] [PubMed] [Google Scholar]
- 127.Ernst B, Lee DS, Chang JM, et al. The peptide ligands mediating positive selection in the thymus control T cell survival and homeostatic proliferation in the periphery. Immunity. 1999;11:173–181. doi: 10.1016/s1074-7613(00)80092-8. [DOI] [PubMed] [Google Scholar]
- 128.Laport GG, Levine BL, Stadtmauer EA, et al. Adoptive transfer of costimulated T cells induces lymphocytosis in patients with relapsed/refractory non-Hodgkin lymphoma following CD34+-selected hematopoietic cell transplantation. Blood. 2003;102:2004–2013. doi: 10.1182/blood-2003-01-0095. [DOI] [PubMed] [Google Scholar]
- 129.Schmidt SM, Schag K, Muller MR, et al. Survivin is a shared tumor-associated antigen expressed in a broad variety of malignancies and recognized by specific cytotoxic T cells. Blood. 2003;102:571–576. doi: 10.1182/blood-2002-08-2554. [DOI] [PubMed] [Google Scholar]
- 130.Vonderheide RH, Hahn WC, Schultze JL, et al. The telomerase catalytic subunit is a widely expressed tumor-associated antigen recognized by cytotoxic T lymphocytes. Immunity. 1999;10:673–679. doi: 10.1016/s1074-7613(00)80066-7. [DOI] [PubMed] [Google Scholar]
- 131.Mitchison NA. Studies on the immunological response to foreign tumor transplants in the mouse. I. The role of lymph node cells in conferring immunity by adoptive transfer. J Exp Med. 1955;102:157–177. doi: 10.1084/jem.102.2.157. [DOI] [PMC free article] [PubMed] [Google Scholar]