APPROXIMATELY 10,000 autologous hematopoietic stem cell (HSC) transplants are performed worldwide each year for malignant diseases.1 Results of randomized trials in recent years suggest that high-dose chemotherapy followed by infusion of autologous HSCs can offer prolonged disease-free survival in hematologic malignancies including non-Hodgkin’s lymphoma in relapse,2 acute myelogenous leukemia,3 and multiple myeloma.4Similarly encouraging results have been seen in the treatment of solid tumors.5 6 

After transplantation, reconstitution of bone marrow (BM) consists of two distinct phenomena, numerical recovery of BM cellular elements on the one hand and functional recovery of cellular interactions on the other.

Although reappearance of neutrophils and platelets is often considered the endpoint of hematologic recovery after intensive chemotherapy and stem cell transplantation, this ignores the second arm of BM recovery, that of immunological reconstitution. In fact, functional recovery of lymphoid and immune effector cells occurs very gradually, and reconstitution of normal humoral and cellular immunity may take a year or more.

Immune reconstitution involves several components of the immune response. These include (1) reappearance of functional B cells, (2) thymic and extra-thymic T-cell development, (3) reconstitution of effector cells including cytotoxic T cells and natural killer (NK) cells, and (4) efficient antigen presentation to reconstitute the pretransplantation immune repertoire. This restoration of immune function is not merely experimental. It may have direct clinical implications: Immediately after the administration of intensive cytotoxic drugs, minimal tumor burden is presumed to be present, providing potentially ideal circumstances to eliminate residual disease altogether by immunotherapeutic means. In this review, several strategies that could lead to enhancement of cellular immune function to take particular advantage of posttransplantation minimal residual disease will be discussed. In addition, the potential to accelerate immune reconstitution and the effect that might have in the therapy of malignant disease will be considered.

Although there are similarities in immune reconstitution after allo-BM transplantation (BMT)7 8 and autologous HSCs, allo-BMT involves graft-versus-host disease (GVHD) and the use of immunosuppressive therapy to control it, both of which interfere in the early developmental stages of immune reconstitution. Autologous HSC transplantation that entails neither GVHD nor immunosuppressive drugs presents more direct insight into the factors involved in immune reconstitution after grafting.

B-Cell Regeneration and Ig VH Gene Expression

Normal B-cell differentiation is accompanied by a set of preprogrammed steps of Ig gene rearrangement and by successive acquisition and loss of differentiation level-specific surface molecules.9 In both its Ig gene rearrangements and in phenotypic expression, B-cell recovery after transplantation appears to recapitulate normal B-cell ontogeny.10 The relative and absolute numbers of circulating cells expressing CD19 and CD20, two markers of mature B cells, are decreased during the first 3 months following transplantation.11,12 Thereafter, the numbers of such cells increase to a plateau at 6 to 9 months. CD23 (the low-affinity receptor for the Fc portion of IgE) and CD38 (the ecto-enzyme of nicotinamide (NAD) glycohydrolase that acts as an adhesion/homing receptor and is involved in intracellular calcium homeostasis) are strongly expressed on the circulating B cells of neonates and B cells from cord blood, whereas in adults, CD23+ CD38+B cells are a minor subset. During the first year postengraftment, the majority of circulating B cells carry the CD23+, CD38+ undifferentiated phenotype.12Furthermore, as in neonates, the percentage of mIgM+ and mIgD+ B cells is high in autologous transplant recipients. Taken together, this suggests that the majority of posttransplantation circulating B cells are poorly differentiated.

Consistent with the notion that autologous BMT (ABMT) ontology follows developmental ontogeny, ABMT recipients have a significant increase in the B-cell precursor marker CD10 in the BM as early as 1 month after grafting which persists for at least 1 year and precedes repopulation of the peripheral blood mature B cells by 1 to 2 months.13 CD10 (also known as CALLA) is a highly conserved neural endopeptidase transiently expressed on early B progenitors before the appearance of heavy μ chain in the cytoplasm and is re-expressed after activation by antigen. Nevertheless, the phenotype of circulating B cells posttransplantation differs from BM B cells because CD10 is expressed on only a negligible fraction of peripheral B cells.10 

In recipients of CD19+ cell-depleted BM, the immunophenotypic features of the resulting BM B-cell precursor populations are similar to those of fetal liver or fetal BM-derived B-cell precursors. As in normal fetal development, the expression of CD10 and CD19 antigens in posttransplantation BM appears to precede expression of other antigens characteristic of normal B-cell ontogeny.14 As might be expected, in patients who received ABMT for B-cell malignancies purged with anti–B-cell monoclonal antibodies (MoAbs), B-cell recovery is further delayed; at 3 months, only 50% of engrafted patients attain normal percentages of CD20+ B cells.15 

Ig gene rearrangements after ABMT, like phenotypic markers, suggest retracing of B-cell ontogeny. Posttransplant Ig gene rearrangements are consistent with wide B-cell polyclonality rather than a narrow oligo or restricted clonality.13 However, Ig VH gene family usage does differ from the normal adult distribution during the first 3 months after both allo-BMT and ABMT.16 Following allo-BMT, the VH repertoire resembles that expressed by the early normal fetal BM with a relative increase in the VH2, VH4, VH5, and VH6 gene families. After ABMT, the same general pattern is observed with a decreased expression of the largest family VH3, offset by a relative increase of the much smaller VH4 and VH5 families.16 This preferential expression of these small VH gene families mimics the pattern seen during normal B-cell ontogeny.17 

By 90 days after BMT, VH3 and VH4 gene usage is indistinguishable from that of normal adults, suggesting that the Ig repertoire may have normalized by then.16 However, even after attaining adult level of VH gene family usage, the rearranged VH genes exhibit much less somatic mutations in BMT recipients than seen in rearrangements of normal adults.18 This may point to a block in antigen-selected affinity maturation of antibody and/or a maturation arrest in B-cell differentiation after Ig V gene rearrangement and may have implications in failures to attain high-specific, high-affinity antibodies in posttransplant vaccinations.

Origin of Posttransplant B Cells

After ABMT or autologous peripheral blood stem cell transplantation (ABSCT), B cells regenerate from several sources: (1) B cells of the transplant recipient which survived the pretransplantation chemotherapeutic intensification treatment; such cells may be seeded in the BM, lymph nodes, or spleen; (2) B cells present in the graft; (3) hematopoietic stem cell progenitors in the transplant that differentiate after grafting in the recipient; and (4) residual recipient stem cells. Because allogeneically transplanted cells can be readily traced, allo-BMT provides direct insight into the origin of B cells in the transplant recipient. Transfer of humoral immunity has been documented from donor to recipient, including immunity to tetanus, varicella, diphteria, influenza virus, cytomegalovirus, hepatitis B virus, and human immunodeficiency virus,19-23 suggesting that functional B cells are passively transferred by transplantation. The corollary of adoptive transfer of immunity is that active immunization of BM donors might serve to reduce the incidence of infection in recipients of allo-BMT and that immunization before intensification might likewise be considered in ABMT and ABSCT recipients. Immunization of ABMT patients before BM harvests with either protein vaccines such as tetanus toxoid or conjugated carbohydrate vaccines such as haemophilus influenzae can enhance early recovery of specific antibody.24 

Transfer of humoral immunity suggests that differentiated antigen-selected B cells in the graft are a significant source of posttransplantation B cells. The ontogeny data presented in the preceding section indicated that stem cells differentiating into Ig expressing B cells represent a significant part of the posttransplantation B-cell population. In addition, findings from B-cell–purged transplants indicate that both recipient and donor stem cells contribute to the posttransplantation B-cell population.15 

B-Cell Function

Deficiencies in humoral responsiveness in HSC recipients is attributed to both decreased T-cell help and to intrinsic B-cell defects.25-27 Serum Ig levels remain low during the first 3 months after ABMT during the same period in which the numbers of circulating B cells is reduced11,28 and B-cell proliferative response to the T-cell–independent antigenStaphylococcus aureus Cowan strain I (SAC) blunted.12 While IgM production in response to pokeweed mitogen and SAC normalize at 3 months, IgG production is suppressed for 12 to 24 months in most patients.12 This delay in Ig production parallels the pattern seen in B-cell ontogeny, and may reflect the failure of posttransplant B cells to receive or respond to T-cell factors involved in isotype switching.

In patients receiving B-cell–purged BM for B-cell hematologic malignancies, responsiveness to the normally proliferative effects of cross-linking anti-Ig antibody is significantly lower than that of normal controls at 3 and 6 months.15 As in engraftment of unpurged marrow, recovery of in vivo B-cell function demonstrates a selective defect with normal serum levels of IgM returning at 6 months, IgG at 12 months, and IgA after 2 years, reflecting a recapitulation of normal B-cell development.15 

BM-derived hematopoietic stem cells in the normal process of differentiation home to the thymus, the major site of T-cell differentiation. However, the thymus is not the only site of T-cell development. T-cell differentiation also occurs through extrathymic pathways in the gut mucosa, in the liver,29 and, at least in the case of murine T-cell development, in the BM as well.30 The contribution of these extrathymic sites may play a role in posttransplant immune reconstitution.

Reconstitution of T-Cell Subsets

Surface markers have proven to be critically useful in characterizing T lymphocytes and their functional subsets. Nevertheless, however useful in our understanding of lymphoid ontogeny, phenotypic subtyping identified to date doubtlessly represents only a part of the overall T-cell functional repertoire. Evidence for the as-yet limited nature of our T-cell characterization is the fact that phenotypic shifts in posttransplantation T cells do not necessary result in immune dysfunction. This must be borne in mind when discussing T-cell phenotypes and their correlation to immune reconstitution after stem cell transplantation.

T-Cell Subsets After ABMT

After ABMT, the relative number of CD3+ cells is significantly decreased compared with those of normal controls during the first month postgrafting, returning to normal levels within 3 months.31 In addition, a decrease in the relative and absolute numbers of CD4+ cells in the peripheral blood is commonly seen and can persist for a year or more.31-33 In contrast, the relative and absolute number of CD8+ cells reconstitutes fairly rapidly resulting in an inverted CD4/CD8 ratio in the months following autologous transplantation.31-33 

The observation of an inverse correlation between the size of the thymus and the level of the CD4+ cells in the peripheral blood after high-dose chemotherapy supports the notion that helper T-cell development depends on residual thymic function.34In addition, thymic epithelium has been shown to play a more important role in helper T-cell development than in differentiation of the suppressor T cell.35 The inverted CD4/CD8 ratio seen after transplantation is consistent with such a schema. Thymic involution starts as early as 1 year of age, continues at a rate of approximately 3% per year until middle age and thereafter decreases to less than 1% per year.36 The fact that CD8+ T-cell reconstitution appears not to be significantly impaired by age-related involution of the thymus suggests that thymic-independent pathways primarily contribute to its regeneration.37 A predominance of CD28 and CD57+ cells among the CD8+ subset has been observed following chemotherapy that may persist up to 9 months.37 The absence of CD28 expression is similar to that seen in extrathymically derived CD8+ cells.38 

Functional subsets of CD4+ and CD8+ T-cell populations have been studied after ABMT and can be distinguished on the basis of the expression of CD45 isoforms (CD45RA, CD45RO). CD45RO+ T cells respond in vitro to recall antigens and correspond to “memory cells” while CD45RA+ T cells correspond to naı̈ve cells recently issued from the thymus. During the first 3 months following ABMT, the number of CD45RA+ lymphocytes is decreased, but returns to normal levels by 1 year posttransplantation.31 39 The CD4+CD45RA+ subset is profoundly reduced and may take up to 2 years to recover. In contrast, the CD8+CD45RA+ population normalizes within the first month after ABMT.

T-Cell Subsets After ABSCT

Because peripheral blood stem cells (PBSC) contain a larger proportion of more differentiated progenitor cells as well as terminally differentiated effector cells than BM, one might suppose that the kinetics of T-cell recovery may be accelerated following ABSCT as compared with ABMT. The fraction of cells designated PBSC in fact does contain large numbers of T cells. CD3+ cells may represent more than 20% of peripheral cells collected after granulocyte colony-stimulating factor (G-CSF) mobilization yielding substantially more T cells (up to 1 log greater) than found in BM.40 41 

Immediately after transplantation of mobilized PBSC, total CD3+ cell levels return to normal, CD4+ cell levels remain below normal, and the number of CD8+ cells increases resulting (depending on the study) in either severely42,43 or slightly44-47 reduced CD4/CD8 ratios. Overall, when CD4/CD8 and CD45RA/CD45RO ratios are examined, recovery of T-cell subsets appears more promptly after ABSCT than after ABMT.43,47 48 As noted however, here as in all similar analyses, precise correlation of T-subset distribution to overall T-cell function is uncertain.

T-Cell Repertoire (TCR)

Shifts in T-cell subsets as defined by TCR V gene expression have been described early after both allo-BMT and ABMT.49-53 It is not clear whether particular T-cell subsets as defined by V gene usage are derived from T-cell precursors (either donor or recipient) or from expansion of donor-derived mature T cells. A significant proportion of patients shows increased usage of TCR γ/δ during the early period post-BMT. Within the TCR γ/δ subpopulation, there is a preferential expression of the Vγ9 and Vδ2 genes as is seen during early fetal life.54 This suggests that recapitulation of T-cell ontogeny may occur early following BMT, analogous to that described for B-cell regeneration.50 51 This early postengraftment predominance of Vγ9+Vδ2+ cells in the periphery may subsequently be further increased by antigen-driven expansion of the newly generated γδ cells. An alternative possibility exists that mature γδ T cells are already present in the BM graft, are expanded after contact with antigen, and do not represent an ontologic recapitulation.

Origin of Posttransplant T Cells and the Role of Thymic and Extrathymic Pathways

Examination of TCR leads to the question of T-cell origin. After ABMT or ABSCT, T cells may reconstitute from at least four different sources: (1) rare recipient T cells that survived the conditioning regimen; these cells might be seeded in the BM, lymph nodes, or spleen; (2) T cells present in the graft; (3) hematopoietic stem cell progenitors of the graft that differentiate in the recipient; and (4) residual recipient stem cells (Fig 1). Studies of TCR alluded to in the preceding section suggest that the quantity of T cells present in the graft might influence the rapidity and the quality of T-cell recovery. However, it remains unclear how much passively transferred T lymphocytes will contribute to sustained cellular immunity.

Fig. 1.

Origin of mature T cells in transplant recipients. Derivation from transplanted stem cells and mature T cells contained in the graft and derivation from surviving host T cells.

Fig. 1.

Origin of mature T cells in transplant recipients. Derivation from transplanted stem cells and mature T cells contained in the graft and derivation from surviving host T cells.

Close modal

In autologous grafting where the source of cells as either transplanted or nontransplanted cannot be readily identified, data from gene marking studies may help to trace the origin of T cells after transplantation. Unfortunately, because gene transfer into BM cells is neither very efficient nor very specific, gene transfer studies may not yet not give a precise quantitative determination of the role of transplanted BM in T-cell recovery. However that may be, following transplantation of autologous BM that had been transduced with a retroviral vector containing a neomycin resistance gene, T cells carrying the gene could be detected by polymerase chain reaction (PCR) as early as 1 month and remain detectable for at least 18 months.55 Nevertheless, although the protocol of retroviral infection in the study cited was intended to transfer the marker gene into stem cells only, one cannot exclude that T cells from the graft had been transduced as well.

Data from allo-BMT are helpful in distinguishing the contribution of grafted BM and T cells from that of postchemotherapy residual stem cells or T cells. In most conditioning regimens used for allo-BMT, recipient hematopoiesis is ablated. However, T cells can survive conditioning regimens.56 Using PCR amplification of minisatellite DNA regions, Roux et al57 were able to determine the recipient or donor origin of T cells after allo-BMT. Their data suggest that in recipients of T-cell–depleted (TCD) BM, within the first year following transplantation, the T-cell compartment has a mixed origin with T cells derived from both transferred donor cells and surviving host T cells. In contrast, when a patient was grafted with an unmanipulated BM, few or no recipient T-cell clones are detected.

Another source of information is the transfer of antigen-specific cellular immunity from the donor to the recipient. Reports indicate that cellular immunity against varicella zoster virus and tuberculin-purified protein derivative can be effectively transferred from immune marrow donor to recipient.58 59 These findings transposed to autologous transplantation suggest that T cells in the graft can expand.

To study the relative contributions of peripheral lymphoid populations and BM-derived precursors to T-cell regeneration after BMT and to understand the role of the thymic function in the immune recovery,60 Mackall et al61 performed a series of elegant experiments where they studied lethally irradiated thymus-bearing and thymectomized mice that had received congenic lymph node cells as a source of peripheral T-cell progenitors and syngeneic T-cell–depleted BM. They showed that in hosts lacking thymic function, the predominant reconstituting T cells were derived from peripheral T-cell progenitors contained in the transplant which expanded in the thymectomized recipients. In the presence of a functional thymus, the expansion of peripheral T-cell progenitors is downregulated. Furthermore, in thymectomized hosts, the majority of T lymphocytes did not express the high-molecular-weight (HMW) CD45 isoform which indicated normal maturation, while in thymus-bearing animals there was an increase in the level of naive type cells. There is evidence that thymic-independent T-cell regeneration occurs primarily via expansion of peripheral T cells and is Ag driven because significant expansion of CD4+ and CD8+transgenic/TCR-bearing cells appears only in the presence of Ag specific for the TCR.62 In humans, where thymic regenerative capacity is compromised because of age- and chemotherapy-related changes, it appears that a thymic-independent pathway must predominate.60 

The capacity of the thymic and extrathymic pathways to produce CD4+ cells differed in thymectomized and thymus bearing murine hosts.61 Lower numbers of CD4+ cells were derived from TCD BM transplanted into thymectomized recipients than when transplanted in thymus-bearing mice, while the percentage of CD8+ cells did not differ significantly between the two types of recipients. Although the BM is an inefficient source of CD4+ cells in the absence of a functional thymus, the peripheral lymphoid progenitor pathway appears capable of compensating, resulting in adequate numbers of CD4+ cells in transplanted thymectomized mice.

T-Cell Function

T-cell competence after stem cell transplantation can be gauged at three distinct functional levels: cell proliferation, cytokine production, and lytic capacity.

T-Cell Proliferation

Using limiting dilution analysis, the frequency of mitogen-responsive T cells in peripheral blood, including the frequency of cytokine-secreting helper T cells, interleukin-2 (IL-2) responding T cells, and cytotoxic T cells, was found to be low after ABMT.63 

Cayeux et al64 reported that isolated T cells at an early post-ABMT stage (<2 months) had defective responses to normally proliferation-inducing MoAbs such as anti-CD3 or anti-CD2 even in the presence of IL-2 affecting both CD4+ and CD8+subsets. In response to anti-CD3 and IL-2, PB mononuclear cells (PBMC) from ABMT recipients proliferate but to a lesser degree than in normal individuals.65 Sugita et al66 found similar results: anti-CD3 alone or in combination with anti-CD2, anti-CD26, or anti-CD29 could not induce T-cell proliferation within 4 months after ABMT. However, T-cell proliferation induced by anti-CD3 + anti-CD2 and by anti-CD3 + anti-CD26 reaches almost normal levels by 1 year. Just as in normal T cells, coactivation with anti-CD28 MoAb can enhance the response in some transplant patients, particularly in long-term recipients (above 6 months).67 

In a mixed leukocyte reaction (MLR), accessory cells from auto-transplanted recipients were unable to trigger normal levels of proliferation of normal control T cells or to induce them to synthesize IL-2.68 69 In addition, normal allogeneic accessory cells failed to provide the necessary signals to activate transplant-derived T cells or induce them to produce IL-2 and proliferate. This deficiency could be corrected by addition of exogenous IL-2. The fact that recipient T cells do respond to IL-2 indicates that the IL-2 cell receptor is present on their surface and functional. These observations may indicate that during T-cell proliferation cell-to-cell contact does not lead to T-cell activation but can induce IL-2 responsiveness.

T-cell proliferation is also impaired after autologous PBSC, but recovery appears faster than that observed after ABMT.47The depressed immune function might be partly related to the suppressive effects caused by high numbers of monocytes present in growth factor–mobilized PBSC.70 71 Although hematopoietic engraftment after infusion of positively selected CD34+cells appears similar to that observed after unselected PBSCT, no study has compared the immune recovery after infusion of selected CD34+ cells and after PBSC or BM.

Cytolytic Function

Cytomegalovirus (CMV)-specific HLA-restricted CD8+cytotoxic T cells (CTL) have been shown in the majority of patients within the first 3 months after ABMT or APBSCT and are associated with protection from CMV infection.72 In contrast, specific CTL response against Epstein-Barr virus (EBV) are impaired during the first 2 months after autologous stem cell transplantation.73Further investigation of specific CTL activity are required in developing of adjuvant vaccines in patients undergoing transplantation.

Lymphokine-activated killer (LAK)-like activity, that is cells incubated with IL-2 capable of killing NK-resistant target cells, is mediated primarily by CD16+CD3 cells and to a lesser degree by CD16CD3+ cells and appears 4 to 6 weeks after ABMT.74 LAK activity is not observed after conventional chemotherapy not followed by transplantation, but IL-2–responsive LAK precursor cells are rapidly reconstituted after ABMT or ABSCT.75-77 Development of IL-2–mediated cytotoxic activity may prove to be functionally important for eradication of residual malignant cells in vivo. Consistent with such a role is the finding that the cytotoxic activity of PBMC after ABMT against NK-sensitive and NK-resistant target in response to anti-CD3 and IL-2 is actually greater than that of normal controls.67 IL-7 also induces significant LAK activity in ABMT recipients to levels comparable to those obtained with IL-2, suggesting that IL-7 may have an immunotherapeutic role alone or in conjunction with IL-2.78 The potential therapeutic use of cytokines in inducing cytotoxic activity is discussed under NK Cells.

Cytokine Production and T-Cell Responsiveness to Cytokines

In vitro–stimulated PBMC from recipients of ABMT have significant defects in the production of a number of T-cell–derived cytokines important in immune homeostasis, particularly IL-2, in the early posttransplant period.64 79 

The origin of the observed decreased cytokine production (notably IL-2) and of the blunted T-cell proliferation can be ascribed to several causes: subnormal levels of cytokine receptor expression, abnormal accessory function, production of suppressive cytokines, or an intrinsic T-cell defect. Insufficient IL-2 receptor cannot account for the decreased proliferation since stimulation of posttransplant T cells has been shown to induce the expression of the α chain of the IL-2 receptor.66,80,81 Normal accessory cells do not restore normal production of IL-2 and, vice versa, patients’ accessory cells fail to activate normal T cells and induce IL-2 synthesis.68 However, the addition of exogenous IL-2 can at least partially compensate the abnormal proliferative response. Possibly signal transduction required for T-cell–non T-cell interactions are in some way dysfunctional. These may entail the interactions between CD40-CD40L, between CD4-MHC II, or between B7-CD28.

NK cells are defined as large granular lymphocytes capable of mediating major histocompatability complex (MHC)-unrestricted cytolytic reactions against tumor or virally infected cells and not expressing receptors for antigen (ie, neither surface Ig or TCR). In addition, NK cells characteristically have a CD3CD16+CD56+ phenotype. NK cells are among the first cells to recover after transplantation: in contrast to B- and T-cell function, NK activity following both ABMT and ABSCT reaches normal level within 1 month.46,48,80 82-84 

IL-12, a 70-kD heterodimeric cytokine with pleiotropic activities produced by antigen-presenting cells (APC), stimulates the proliferation and cytotoxic activity of NK cells and enhances generation of cytotoxic T cells.85 In addition, it induces transcription and secretion of interferon-γ (IFN-γ) either directly or in synergy with other inducers and promotes commitment of naive T cells to the TH1 pathway resulting in the production of TH1 cytokines including IL-2.85As a result of these effects, IL-12 appears to have strong antitumor and antimetastatic effects as shown in a murine model,86,87with evidence suggesting a similar role in human malignancy.88 SAC stimulated PBMC derived from autologous hematopoietic transplants show no decrease in the production of IL-12 as compared with control PBMC.89 However, this seemingly normal in vitro production of IL-12 does not exclude the possibility that in vivo IL-12 production remains abnormal. T cells play a role in priming APC to produce IL-1290 and posttransplantation T cells may be unable to carry out this function. Transplantation-derived PBMC do not appear to have defective responsiveness to exogeneously provided IL-12 as evidenced by their degree of in vitro proliferation and IL-12–mediated IL-2 production.89 In addition, significant increases in NK and LAK activity are observed with IL-12 alone or in combination with IL-2.91 

The sum of these findings suggests that the recovery of NK cells seen after ABMT and ABSCT is prompt both in quantity and in functional quality. Maturation of NK cells can occur in the absence of a functional thymus in mice and humans.92 93 This may account for the prompt NK recovery. In the early posttransplant period, when specific immunity is still recovering, NK cells may provide an important defense against infections or tumor relapse. Expansion of NK cells with growth factors immediately following transplantation might therefore serve to increase host defenses. Such an immunotherapeutic strategy is discussed below. However, despite our ability to accelerate or potentiate cytotoxic cells, there is no direct evidence showing a correlation between NK activity after transplantation and tumor relapse.

The degree of T-cell incompetence after autologous transplantation is difficult to gauge exactly in vitro. The capacity to mount an effective immune response against foreign antigen better reflects the immune status of transplant recipients than do in vitro tests. Information garnered from allogeneic transplantation indicates that with the passive transfer of allogeneic lymphocytes, transplantation recipients show short-term production of antibodies against viral pathogens.20,94 However, vaccination in the months after transplantation does not always result in sustained protection. This pattern of temporarily effective but nonsustainable protection suggests that B lymphocytes derived from the donor, which generate specific antibody in the short term, are transferred together with the transplant, but that antigen-specific T cells necessary for ongoing sustained protection must be regenerated anew since the transfer of T cells functional against specific infections appears less efficient.95-97 The reduced transfer of antigen-specific T lymphocytes may be caused by an already impaired T-cell–mediated immunity before transplant in patients who have received significant prior chemotherapy.

ABMT patients have an overall increased risk of infections, including CMV, influenza, herpes simplex virus, and varicella zoster virus,98-100 although the incidence of infections and severity are lower in ABMT than among allo-BMT recipients.72,95,101 This disparity may be accountable by the possibility that T-cell transfer from an allogeneic donor is further compromised by GVHD prophylaxis. Lymphocyte proliferative responses to herpes simplex virus and CMV in seropositive patients return more rapidly in ABMT recipients than allo-BMT.72 

Overall, the transfer of antigen-specific T cells is substantially less efficient than the passive transfer of antibody-producing B lymphocytes transfer. This has implications in designing infection-specific vaccines for administration before and after transplantation.

The effectiveness of any cancer immunotherapy in limiting or eliminating malignant cells is a function of the target tumor mass. Even when therapeutically bolstered, immune mechanisms can be rendered ineffective by the presence of overwhelming numbers of target malignant cells. Because high-dose chemotherapy followed by ABMT or ABSCT results in (or is presumed to result) in minimal burden of residual malignant disease it provides a potentially ideal setting for immunotherapy.

Administration of Cytokines

Interleukin-2

It has been 15 years since the first demonstration that IL-2 activates and promotes proliferation of murine and human NK cells in vitro resulting in both a greater degree and wider spectrum of lytic activity and initial preclinical studies had shown that systemic IL-2 had significant antitumor effects.102,103This lead to clinical studies using IL-2 regimens with or without adoptive transfer of in vitro activated LAK cells. After the exhilarating results of the initial clinical trials involving high-dose recombinant IL-2 and LAK cells,104 the true response rate of IL-2 with or without the addition of LAK cells was found to be in the range of 15% to 25% for renal cell carcinoma and melanoma.105 Although a variety of tumors of different histological types are sensitive to IL-2/LAK cell therapy, complete eradication of tumor is rare. Nevertheless, because of the measurable tumor regression seen in a significant minority of patients with metastatic malignancies, a logical extension was to perform similar trials of IL-2 after ABMT for hematologic malignancies. The rationale for using IL-2 is based on the following: (1) preclinical data showing that human hematologic malignant cells can be lysed in vitro by IL-2–activated effector cells106-108; (2) IL-2–responsive cells are present early following transplantation74-76; (3) immunotherapy is likely to be more effective when minimal tumor burden is present; and (4) chemotherapy-resistant cells can be lysed by IL-2–activated NK and T cells. IL-2–based trials have included both preclinical and clinical studies.

Preclinical studies.

Mice inoculated with a leukemic cell line were administered intravenous cyclophosphamide and syngeneic BMT 24 hours later. This was followed by either recombinant (r) IL-2 for 5 days starting on day 1, 7, or 21 or by no further therapy.109Mice not treated with IL-2 relapsed and died within 50 days posttransplantation, whereas mice receiving IL-2 had long-term disease-free survivals. The maximal antileukemic effect was observed in mice receiving IL-2 3 weeks after BMT. The delayed maximal effect is consistent with and likely results from the delay needed to first achieve maximal lymphocyte reconstitution that can then respond to IL-2.

Clinical studies.

Trials of continuous-infusion IL-2 administration after ABMT have been performed primarily in hematologic malignancies,83,110-124although several studies of its use following ABMT for solid tumors have also been performed.83,114 125 

IL-2 administered after ABMT has significant immunomodulatory effects, including an increase in circulating lymphocytes expressing CD3, CD4, CD8, and the NK-associated markers CD16 and CD56 as well as an increase in circulating NK and LAK activity.83,110,111,114,115,117However, in one trial where IL-2 treatment was begun on the day after transplantation for acute lymphoblastic leukemia (ALL), cells with the NK phenotype and circulating NK activity were not enhanced.119 Nevertheless, the majority of IL-2–treated patients showed strong cytotoxicity against an ALL cell line.

One of the primary drawbacks to the use of high-dose IL-2 is its systemic toxicity. Fever, fatigue, diffuse rash, and varying degrees of a capillary leak syndrome with weight gain and hypotension are frequently observed in most of the studies using high doses of IL-2 (between 9 and 24 × 106/IU/m2/d). The major hematopoietic toxicity of IL-2 after ABMT has been thrombocytopenia. Lymphocytosis, neutrophilia, and eosinophilia are commonly reported as well. These effects may be caused by the induction of secretion of other cytokines by IL-2.126 

The conclusion drawn from these clinical trials is that although the administration of IL-2 is complicated by significant side effects in at least 30% of recipients, it remains a viable option for some patients. Most groups administer IL-2 once hematopoietic engrafment has been achieved, generally 2 to 3 months after transplantation while endogenously activated cells and IL-2–responsive immunocompetent precursors cells are present earlier in the posttransplant period. Immune modulation with marked time-dependent increases of NK cells (CD56bright+CD16+CD3) have been achieved with low doses of IL-2 (2 to 4 × 105IU/m2/d) administered for a period of 3 months without significant toxicity.114,122,127 NK and LAK activities were also found to be enhanced by low doses of IL-2.114 

Furthermore, none of the studies including two randomized ones in acute leukemia in adults have shown that the immunomodulatory effects of IL-2 were beneficial in terms of survival.120 124 It is probably unlikely that IL-2 administered alone will be effective, but it may find its place to expand or sustain in vivo NK or cytotoxic T cells injected after transplantation.

Interleukin-7

As noted, T-cell precursors leaving the BM enter the thymus for terminal differentiation. Data from cytokine-deficient mice (knockout mice) suggest that with the exception of IL-7, cytokines can be removed without significantly affecting intra-thymic development.128,129 Mice lacking the IL-7 gene or a functional IL-7 receptor gene have severe impairment of early lymphocyte expansion.128 129 The severe lymphopenia seen in IL-7–deficient mice is associated with normal distribution of T-cell subsets and response to mitogens, suggesting that IL-7 acts on the expansion and proliferation of T cells rather than on their differentiation and function. These multiple effects of IL-7 on T-cell development have led to preclinical studies in murine BMT.

Preclinical studies.

In a murine model of BMT, IL-7 administration accelerates both T- and B-cell reconstitution by up to 2 to 4 weeks130 and both CD4+ and CD8+ cell counts are found to be expanded. Bolotin et al131 reported that in a syngeneic murine BMT model, injections of IL-7 from day 5 to 18 induced an increase in the cellularity of the thymus by 4 weeks, while the proliferation of early thymic precursor cells was increased nearly eightfold. In contrast, in mice not receiving IL-7, normal numbers of thymocytes appeared only by week 8. Furthermore, in the IL-7–treated mice, distribution of thymic subpopulations approximated those of normal untransplanted mice. Abdul-Hai et al132 reported similar findings of accelerated repopulation of the thymus with IL-7 injected for 10 days immediately after murine transplantation. IL-7 also increased RAG-1 (recombination activator gene) expression in the thymus. Taken together, these data suggest that IL-7 may find a therapeutic role in accelerating T-cell reconstitution after autologous transplantation.

Incubation of HSC With Cytokines and Growth Factors

Preclinical Studies

Incubation of murine BM with IL-2 results in the generation of killer cells with non–MHC-restricted cytotoxicity against tumor cells which appears superior to the cytotoxicity of spleen LAK cells.133-135 Transplantation of IL-2–activated BM (ABM) immediately followed by administration of systemic IL-2 reduces the dissemination of established melanoma and sarcoma in mice more effectively than transplantation with untreated BM with or without systemic IL-2 alone.133,134 This suggests that priming with IL-2 before ABMT induces an antitumor effect capable of eradicating residual malignant disease. When IL-2 therapy is delayed for 1 or 2 weeks after transplantation with ABM, there is a progressive decrease in the cure rate,136 suggesting that ABM cells cannot maintain a prolonged cytotoxicity in vivo. The hematopoietic regenerative capacity of IL-2–activated BM was preserved despite a reduction in viable cell number.

As in the murine studies, in vitro incubation of human BM with IL-2 leads to the generation of cells with cytotoxicity against tumor cell lines.137-141 In vitro activation of BM cells with IL-2 significantly enhances cytotoxicity against chemotherapy-resistant leukemic cells, suggesting that this approach might be useful to eliminate drug-resistant minimal residual disease.142Significantly, PBSC cultured in IL-2 for 24 hours retain adequate potential for hematopoietic reconstitution.143 

NK cells present potentially ideal instruments of immunotherapy. Large scale ex vivo culture of PBSC enriched in monocytes and NK precursors with IL-2 should yield sufficiently high numbers of activated NK cells for clinical use.144 145 It remains to be seen whether optimal regimens of ex vivo and in vivo IL-2 will result in enhanced cytotoxicity against postchemotherapy/transplantation residual malignancy.

Similar studies with granulocyte-macrophage colony-stimulating factor (GM-CSF) indicate that as with IL-2, ex vivo activation of BM with GM-CSF induces cytotoxicity against tumor cells.146 In the case of GM-CSF, however, this is due to the proliferative effect of GM-CSF on macrophages rather than on T cells. When combined with a tumor-specific antibody, GM-CSF induces specific antibody-dependent cytotoxicity (ADCC) against tumor cells.146 

Clinical Studies

Only limited studies of hematopoietic stem cells exposed to IL-2 have been reported so far and these included small numbers of patients with leukemia, breast cancer, and non-Hodgkin’s lymphoma.142 147-149 In the reported studies, BM was maintained with IL-2 for a variety of schedules ranging from 24 hours to 10 days. IL-2–activated BM successfully engrafts patients who had previously received myeloablative chemotherapy. In most studies, additional systemic IL-2 was administered intravenously after transplantation with the expectation that IL-2 would maintain the NK activity. Although in vitro cytotoxicity of IL-2–activated BM against NK-sensitive cell lines was demonstrated, no studies showed any effect against autologous tumor cells. From the preliminary data, it is not yet possible to conclude whether this approach will be successful.

Induction of an Autoaggression Syndrome With Cyclosporine A (CsA)

Preclinical Studies

CsA is a potent immunosuppressive agent that has been used for more than 15 years to prevent GVHD in patients receiving allogeneic BMT. Paradoxically, however, the administration of CsA after syngeneic BMT in rats can lead to the development of an autoimmune phenomenon that is clinically and histologically similar to allogeneic GVHD with the appearance of CD4+ and CD8+ effector cells recognizing MHC class II antigens, including self.150,151Lethally irradiated rats reconstituted with syngeneic BM and treated with CsA for 40 days develop a T-cell–dependent autoimmune syndrome 14 to 28 days after discontinuation of CsA treatment characterized by erythroderma and dermatitis.150 The mechanism for induction of this autoimmune phenomenon remains unclear. It has been suggested that CsA induces modifications in the thymus, including medullary involution, loss of Hassal’s corpuscles, and decreased expression of MHC II antigens in the medulla,152 changes which interfere with intrathymic differentiation of T cells.150,151,153CsA also appears to enhance the development of autoreactive T lymphocytes by blocking their deletion in the thymus.154 The role of the intact thymus in autoaggression is indicated by the finding that syngeneic GVHD cannot be induced in thymectomized animals.153 However, the inhibition of clonal deletion in the thymus and the development of autoreactive T cells in the periphery is insufficient by itself to induce the autoimmune phenomenon. The ablation of the lympho-hematopoietic system with the preparative regimens for transplantation (irradiation or cytotoxic chemotherapy) is also apparently necessary to eliminate peripheral regulatory mechanisms155 156 because infusion of spleen cells from rats with autoreactive disease into normal rats does not transfer the clinical syndrome. In addition, CsA treatment of untransplanted rats does not induce an autoimmune syndrome.

The effector mechanisms of the CsA-induced autoimmune syndrome remain unclear. Because CsA causes a marked decrease in the expression of MHC class II antigens in the thymic medulla,157 developing T cells in the thymus may fail to recognize these MHC determinants as self. Therefore, MHC class II determinants might then turn out to be the principal targets of autoreactivity. The T-cell receptor repertoire of effector T lymphocytes appears restricted, suggesting that only a limited number of class II MHC antigenic determinants may be recognized, and administration of anti–MHC class II antibodies can delay or prevent autoreactivity.158 Recently, the peptide termed CLIP derived from the MHC class II invariant chain which protects the MHC molecules from nonspecific binding of peptides was found to be the target of autoreactive T cells.159 

The autoreactive cells generated by CsA treatment after ABMT have been found to have antitumor effect in vitro.160 Unfortunately, the cytotoxic capacity of these cells is directed against tumor cells expressing the MHC II antigen, limiting the practical application of the therapy. Moreover, CsA induces significant autoimmunity in some, but not all, strains of rats and mice.161 

In an attempt to widen the potential use of CsA, Charak et al,162 using a murine strain that does not develop autoimmune syndrome after CsA therapy, have shown that mice inoculated with non–Ia-bearing tumors (B16 melanoma and C1498 leukemic cells) which received both IL-2 and CsA after BMT had a better survival than mice receiving either IL-2 or CsA alone. However, no mice were cured. The rationale for linking IL-2 and CsA was based on the finding that CsA-generated T cells are highly responsive to IL-2 in vitro.163 The effectiveness of this two-step mechanism is borne out by the finding that the antitumor effect generated in mice receiving IL-2 and CsA could be transferred into secondary tumor-bearing recipients.164 

Clinical Studies

CsA can induce an autoimmune syndrome in patients with lymphoma, acute myeloid leukemia, or breast cancer receiving ABMT.164-170This syndrome is mainly confined to the skin (erythematous maculopapular rash) without clinical evidence of visceral involvement. In one report, the presence of cytotoxic T cells recognizing the patient’s own pretransplant lymphocytes or tumor cell lines that expressed MHC class II determinants could be shown.164 No analysis of cytotoxicity against autologous fresh tumor cells was reported. The reason why clinical signs of autoaggression are located in the skin remains unknown. This could be related to the presence of Langerhans cells in the dermis or a high expression of MHC II in the skin.

Because hematologic malignancies and some solid tumors express MHC class II determinants, they too can presumably serve as the target of a CsA-induced cytotoxic effect. Encouraging results have been reported in a nonrandomized study for the treatment of 40 relapsed or refractory intermediate grade non-Hodgkin’s lymphoma treated with ABMT, CsA, and IFN-α in an attempt to further upregulate the expression of HLA antigens. Thirteen percent of the patients were found to relapse after a median follow-up of 24 months.170 This clinical outcome compares favorably with the trial of ABMT alone in patients with relapsing non-Hodgkin’s lymphoma. However, carefully planned trials comparing APSCT alone versus APBSCT with CsA are still lacking. CsA with or without IFN-γ has been administered after high-dose chemotherapy and ABMT in women with metastatic breast carcinoma, and showed that the combined therapy had an acceptable level of toxicity.166 167 However, no clinical benefits could be shown.

In none of the clinical studies was HLA expression analyzed on primary neoplastic tissue of patients entering the study nor was in vitro evidence of an antitumor effects shown. Overall, it remains to be seen whether there is consistent clinical benefit to the concept of inducing an autoaggressive GVHD-like syndrome in autologous marrow recipients.

Adoptive Transfer of Ex Vivo–Expanded MHC Nonrestricted Effector Cells

As described above, BM or PBSC can be incubated in vitro with cytokines to induce the development of cytotoxic cells. A number of protocols have been devised to select effector cells with cytotoxic activity from PBMC.

Activated NK Cells: Clinical Studies

Initial enthusiasm for the therapeutic use of LAK came from its use in preclinical and clinical studies against lymphoma and leukemia.171,172 In treating minimal residual malignant disease, LAK cells might be expected to be most effective if they are active against chemotherapy-resistant tumor cells which may have survived pretransplantation high-dose chemotherapy regimens. In in vitro studies, LAK cytotoxicity has been demonstrated against tumor cells surviving therapeutic concentrations of chemotherapeutic agents.173 This in vitro data led to pilot clinical trials combining systemic administration of IL-2 followed by apheresis to generate LAK cells after ABMT or ABSCT in lymphoma patients either in relapse or resistant to primary chemotherapy and in acute leukemia with poor prognostic indicators.115 117 

CD3CD56dim peripheral blood cells make up a specific subset of NK cells. These cells are obtained from the post–IL-2 leukapheresis product by adherence to plastic in the presence of IL-2. Such activated NK cells can be expanded in culture for 2 to 3 weeks with IL-2 in the presence of irradiated allogeneic concanavalin A (conA)-preactivated mononuclear cells. The injection of activated NK cells along with IL-2 to support the antitumor activity of NK cells has been used immediately posttransplantation in patients with lymphoma and no major toxicity has been observed with this combination therapy.174 

Such manipulations including those involving apheresis after IL-2, expansion of NK cells, injection of IL-2 to support in vivo activity of NK cells entail many practical difficulties and morbidities limiting their applications to selected patients. In addition, these initial phase I trials have been performed on too small a number of patients to allow any conclusions regarding any potential benefit of these approaches.

Cytokine-Induced Killer Cells

A somewhat different therapeutically useful cytotoxic cell can be obtained by in vitro exposure of PBMC to combinations of IFN-γ, IL-2, and anti-CD3 monoclonal antibody. The resultant effector cell termed cytokine-induced killer cells (CIK) bears a CD3+CD56+ (but CD16) phenotype and demonstrates non-MHC restricted cytotoxicity.175,176 CIK cells have been found to be substantially more cytotoxic in culture than LAK cells against cellular targets and, like LAK cells, they are effective against chemotherapy-resistant cell lines.177 Although ex vivo generation of CIK cells is IL-2 dependent, in vivo use of CIK cells has the advantage over LAK cells of not requiring additional systemic administration of potentially toxic IL-2 to augment their antitumor activity. In direct comparison, CIK cells have been shown to result in greater regression of disseminated human lymphoma in severe combined immunodeficient (SCID) mice than LAK cells.178,179 While a significant minority of CIK-treated lymphoma-bearing SCID mice had long-term survival, none of the LAK-treated mice survived.178 Because of apparent efficient cytotoxicity and limited systemic effects, expansion of CIK cells may find a place in protocols of autologous transplantation.

Cytotoxic Cell Lines

Human cell lines with potent MHC nonrestricted cytotoxicity activity against tumor cells have been reported. A cell line termed TALL-104 bearing the characteristic phenotype of cytotoxic cells has been derived from a human acute T-lymphoblastic leukemia and maintained in continuous culture in the presence of IL-2. TALL-104 cells show cytotoxicity exclusively against tumors across species without deleterious effects on normal tissues.179-181 After lethal irradiation, the cells are no longer leukemogenic when injected into SCID mice, but retain their killer function. In murine models bearing human tumors, administration of the human cytotoxic T-cell line had effective antitumor effects when given at early stage of disease.181 

Additional NK cell lines have been established.182 183Their capacity to lyse cells appears restricted however to certain tumor types, thereby limiting their potential use. Because cell lines can be expanded continuously in culture, they constitute an unlimited source of effector cells. They may ultimately find a therapeutic niche after transplantation when minimal tumor disease is present and when poorly reactive immunity permits the injection of unmatched allogeneic cells.

Adoptive Transfer of Tumor-Specific MHC-Restricted Effector Cells

Adoptive cellular immunotherapy can be described as the transfer of target-specific effector cells to treat malignant disease. Such an approach entails the isolation and expansion of effector CD4+ and CD8+ T cells with specific reactivity for tumor cells from the host or other donor. In addition, adoptive immunotherapy requires that such cells survive in vivo for a sufficient amount of time to eradicate the tumor.184 This approach could be combined with injection of autologous stem cells.

It is currently feasible to grow T cells to large numbers in vitro by stimulating with antigen and by the addition of cytokines such as IL-2.185 As a result, T-cell therapy has been evaluated as a means to restore protective immunity against CMV following allo-BMT. CD8+ cytotoxic T-cell clones specific for CMV isolated from the blood of BM donors and grown in vitro have been infused to the recipients of allogeneic BM transplants to prevent CMV pneumonia.186 Adoptive transfer of antigen-sensitized T cells may in theory be evaluated in the treatment of malignant diseases. However, adoptive transfer of specific immunity is only possible when specific target tumor antigen(s) are identified and antitumor antigen-specific T cells are expanded. For most tumors currently treatable by high-dose chemotherapy and autologous transplantation, tumor-specific antigens are to date not known.

Ideal tumor antigens should be expressed exclusively or at least preferentially by tumors cells. In the emerging concept of tumor-specific cytotoxicity the context of tumor antigen presentation is no less important than the antigen itself. Once processed, the tumor-related antigen is presented on tumor cells as peptides bound to the HLA molecules. For clinical use then, this HLA restriction must also be identified. The antigen must further be shown to induce T-cell cytolytic responses in vitro and in vivo against tumor cells.

Ig expressed by B-cell malignancies are unique in that they can be distinguished from those expressed by normal B cells. The idiotype resulting from the combination of the variable regions of Ig heavy and light chain therefore represents the best example of a tumor-specific antigen. A number of hematologic malignancies are associated with mutations or translocations that result in expression of cellular oncogenic or chimeric proteins that play a role in malignant transformations and are present only on malignant cells (Table 1). These antigens presented in MHC molecules might induce a T-cell response which may potentially be expanded for clinical use. Prospective candidates for eliciting such a response are two leukemic-associated neoantigens. Chronic myelogenous leukemia (CML) and acute promyelogenous leukemia (APL) are characterized by recurrent translocations leading to the formation of fusion proteins, p210 bcr-abl and promyelocytic leukemia/retinoic acid receptor-α (PML/RAR) in CML and APL, respectively. In the case of p210, peptides have been derived from the fusion sequences and have been analyzed for consensus anchor residues for binding to a given HLA molecules. However, if a proliferation response can be obtained, a cytolytic response has mainly been performed using as target cell lines or normal PBMC bearing appropriate HLA type and loaded with peptides.207-210 Few data are available using fresh leukemic cells as target of cytolytic T cells. In addition, difficulties in obtaining peptide-specific CTL from PBMC of patients with CML in chronic phase suggest that these T cells may frequently be unresponsive to bcr-abl peptides.211 The demonstration of the presence of the processed peptides on the surface of fresh CML cells needs to be confirmed to validate such an approach. Therefore, search for new tumor antigens in addition to the Ig idiotype are needed.

An identical need for tumor-specific cell-surface antigens exists in the immunotherapy of solid tumors. Tumor-specific antigens and CTL-specific responses to these antigens have been most successfully shown for melanoma.212 Unfortunately, antigens uniquely or preferentially expressed by solid tumors currently treatable by high-dose chemotherapy and autologous stem cell support have not been so extensively described as those for melanoma and are now the object of active investigation.212 Solid-tumor–associated antigens might be oncogenic proteins that are mutated and overexpressed as described for breast carcinoma, mutated tumor suppressor proteins, and oncofetal protein (breast carcinoma), or tissue-specific differentiation antigens such as mucin (breast and ovarian carcinoma)213-216 (Table 1). It remains to be determined exactly whether these molecules may prove to induce an immune response and be biologically active antigens for T-cell therapy. In addition, the frequency of expression of putative tumor-specific antigens on tumor cells and the selection of specific epitopes for targeting T cells will have to be clarified.

Having tumor-specific antigens/peptides in hand does not necessary mean that effective T-cell therapy will follow. A composite picture of tumor antigenicity is still incomplete, but to induce a therapeutically useful T-cell response a number of considerations come into play: (1) the frequency of precursor of CTLs capable of recognizing the HLA-restricted peptides may be very low; (2) the relevant antigenic peptide may be present heterogeneically or not be displayed at the surface of tumor cells because they cannot adequately process the antigen or because a different set of peptides is expressed by tumor cells than the peptides predicted; (3) patients must bear the appropriate HLA for which the epitopes have been identified; (4) many tumors are known to have lost completely or partially their HLA molecules; (5) the concentration of peptides needs to be adjusted to avoid selective expansion of low-avidity CTL which are not tumor-reactive217; (6) finally, tumor may fail to attract and activate tumor-specific T cells at the tumor site.218Therefore, adoptive transfer of effector cells will likely have to be worked out on an antigen-by-antigen basis.

Autologous Antigen-Pulsed Dendritic Cells

Dendritic cells (DC) are powerful specialized antigen-presenting cells that can initiate T-cell–mediated immune responses.219 DC cultured (or pulsed) in vitro with proteins process them into peptides and present them as antigens to naive T cells in such a way that the T cells can recognize and respond to the stimulus. The processed antigen is recognized by T cells in an MHC-restricted fashion. DC have been pulsed with tumor-associated proteins and peptides, thereby generating antitumor cytolytic responses in preclinical models220-226as well in a clinical trial with patients with follicular B-cell lymphoma.227 As noted, however, T-cell–defined epitopes presentable by DC have not been identified for most tumors. Because of the extensive diversity of MHC antigens, designing common peptides for T-cell recognition for different individuals with the same disease may prove difficult. For hematological malignancies having recurrent translocations resulting in common fusion proteins or for solid tumors uniformly overexpressing identifiable oncogenes, synthetic peptides have been designed based on the presence of HLA anchor motifs.211 228-230 Some public and private epitopes have been found to be recognized by CTL in the context of MHC class I restricted molecules (Table 2). However, the affinity of a peptide for its corresponding HLA molecules does not necessarily predict its recognition by T cells and its capacity to elicit a cytotoxic response.

To circumvent this problem and provide DC with presentable tumor-associated peptides, methods have been developed to isolate immunogenic peptides from tumor cells using mild acid elution.235 Transfer of DC cells with tumor peptides isolated by this method has been shown to result in suppression of the growth of weakly immunogenic tumors in a murine model.224However, this individualized therapy with unique tumor peptides might be performed only when a large number of tumor cells are available, as it is the case for leukemia.

DC can be generated in large numbers from CD34+hematopoietic progenitors cells using a combination of cytokines including primarily GM-CSF and tumor necrosis factor-α (TNF-α).236-239 Expansions of up to approximately 1.7 × 106 mature DC have been obtainable from as little as 1 mL of normal adult BM using stem cell factor (SCF), GM-CSF, and TNF-α.237 Combining flt-3 ligand to the above three growth factors, it was estimated that 0.6 × 109/kg DC could be generated starting from peripheral progenitor cells of a single leukapheresis.238 Because of this potential of obtaining DC in large numbers in vitro and because of their importance in processing tumor antigens into specific peptides, DC represent an attractive target for transfer of genes encoding tumor-associated antigens. Recent efforts have shown that expression can be achieved by retroviral transduction of CD34+ progenitor cells while they are made to differentiate into DC by growth factors.240-242 Alternatively, DC can be initially obtained and subsequently transduced with adenovirus vectors in which tumor-associated antigen cDNA have been inserted.243-245These strategies will be helpful to circumvent the difficulties in identifying peptides derived from tumor antigens of each individual patient.

As a result of these initial studies, clinical protocols incorporating DC are envisioned which will include expansion of CD34+populations into DC followed by either pulsing the cells with tumor antigens and antigen-derived peptides epitopes, or introducing genes encoding tumor-associated antigens into them for therapeutic reinjection into patients.

Induction of Antitumor Effect by Combining Autologous HSC Transplantation With Infusions of Allogeneic T Cells

A major antineoplastic advantage of allo-BMT is the possibility of inducing a graft-versus-host effect and graft-versus-leukemia has been mainly described. This same antineoplastic effect could be exploited in autologous transplantation. In this approach, patients would receive autologous stem cells purged of tumor contamination for hematopoietic regeneration following myeloablative conditioning regimens together with lymphocytes from an allogeneic donor. The combination of autologous BM in conjunction with allogeneic lymphocytes and IL-2 has been performed in patients with non-Hodgkin’s lymphoma and leukemia.246 However, the number of patients involved was too low to draw any conclusion regarding its efficacy. This technique may further be rendered even more specific if the allogeneic donors could be previously vaccinated with tumor antigens in an attempt to induce an antitumor response in the recipients. Conceivably, tumor antigens might needed to be altered because tumor antigens may be “self” protein and fail to induce an immune response in healthy individuals. The limitations of this procedure will likely be the difficulty in obtaining an adequate histocompatible donor and control the development of GVHD. The possibility to confer an inducible suicide phenotype to donor T cells by gene transfer of herpes simplex virus thymidine kinase may provide a means of suppressing the development of severe GVHD.247 

Although the regeneration of the CD4 T-cell compartment is generally delayed because of the absence of normal thymic maturation, cellular immunity regains effective levels of function within the first 3 months after transplantation. The use of PBSC in recent years has further contributed to accelerate immune reconstitution in comparison to BM, although the overall advantage of PBSC in this regard remains unclear. Tumor immunotherapy is defined as attempts in vivo to harness the regenerating posttransplant immune responses and apply them against malignant cells. As reviewed here, these attempts fall into one of two categories: induction of nontumor specific cytotoxicity and activation of tumor-directed cellular immunity. Nonspecific methods, including administration of cytokines or incubation of the graft with cytokines, induction of an autoaggression syndrome with cyclosporine, and expansion of NK cells, have not yet been clearly shown to be effective in reducing relapses and improve survival. Furthermore, none of these therapies are without their own side effects. It is unlikely that major progress will be made in the clinical use of cytokines alone, although they may find a therapeutically useful niche enhancing immune effector cells. As for infusion of non–tumor-specific effector cells, administration of activated NK cells or CIK cells requires large-scale cellular expansion, rendering the procedure as yet technically difficult for routine clinical use.

The second category of immunotherapy, tumor-specific modalities, may prove to be most effective when the amount of postchemotherapy residual disease is reduced to a minimum. However, in devising tumor-specific approaches, numerous obstacles have already been recognized that need to be overcome before even hoping for general clinical use. The identification of tumor-associated antigens (even if not strictly unique antigens) inducing immune responses is a prerequisite to the development of adoptive transfer of either effector immune cells or use of APC to activate cytolytic T cells. In addition, many questions remains to be resolved: How to obtain an adequate number of clonal cytotoxic T cells? What is the optimal way to pulse dendritic cells? What is the fate of the generated antitumor immune effector cells once reinjected to the patients? The next 5 years should clarify to what extent specific—and nonspecific—immunotherapy may be able to fill in the shortcoming of conventional chemotherapy and finally lead to more effective long-term control of tumors.

Address reprint requests to Thierry Guillaume, MD, PhD, Laboratory of Experimental Oncology and Hematology, University of Louvain, 54 Avenue Hippocrate, UCL54.71, 1200 Brussels, Belgium.

1
Horowitz
MM
New IBMTR/ABMTR slides summarize current use and outcome of allogeneic and autologous transplants.
IBMTR News Lett
2
1995
1
2
Philip
Th
Guglielmi
C
Hagenbeek
A
Somers
R
Van Der Lelie
H
Bron
D
Sonneveld
P
Gisselbrecht
C
Cahn
J-Y
Harousseau
J-L
Coiffier
B
Biron
P
Mandelli
F
Chauvin
F
Autologous bone marrow transplantation as compared with salvage chemotherapy in relapses of chemotherapy-sensitive non-Hodgkin’s lymphoma.
N Engl J Med
333
1995
1540
3
Zittoun
RA
Mandelli
F
Willemze
R
De Witte
T
Labar
B
Resegotti
L
Leoni
F
Damasio
E
Visani
G
Papa
G
Caronia
F
Hayat
M
Stryckmans
P
Rotoli
B
Leoni
P
Peetermans
ME
Dardenne
M
Vegna
ML
Petti
MC
Solbu
G
Suciu
S
for the European Organization for Research and Treatment of Cancer (EORTC) and the Gruppo Italiano Malattie Ematologiche Maligne dell’Adulto (GIMEME) leukemia cooperative groups
Autologous or allogeneic bone marrow transplantation compared with intensive chemotherapy in acute myelogenous leukemia.
N Engl J Med
332
1995
217
4
Attal
M
Harousseau
JL
Stoppa
AM
Sotto
JJ
Fuzibet
JG
Rossi
JF
Casassus
P
Maisonneuve
H
Facon
T
Ifrah
N
Payen
C
Bataille
R
A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma.
N Engl J Med
335
1996
91
5
Humblet
Y
Symann
M
Bosly
A
Delaunois
L
Francis
C
Machiels
J
Beauduin
M
Doyen
C
Weynants
P
Longueville
J
Prignot
J
Late intensification chemotherapy with autologous bone marrow transplantation in selected small cell carcinoma of the lung: A randomized study.
J Clin Oncol
5
1987
1864
6
Bezwoda
WR
Seymour
L
Dansey
RD
High-dose chemotherapy with hematopoietic rescue as primary treatment for metastatic breast cancer: A randomized trial.
J Clin Oncol
13
1995
2483
7
Lum
LG
The kinetics of immune reconstitution after human marrow transplantation.
Blood
69
1987
369
8
Atkinson
K
Reconstruction of the haemopoietic and immune systems after marrow transplantation.
Bone Marrow Transplant
5
1990
209
9
Burrows
PD
Cooper
MD
B cell development and differentiation.
Curr Opin Immunol
9
1997
239
10
Storek
J
Ferrara
S
Ku
N
Giorgi
JV
Champlin
RE
Saxon
A
B cell reconstitution after human bone marrow transplantation: Recapitulation of ontogeny?
Bone Marrow Transplant
12
1993
387
11
Bengtsson
M
Smedmyr
B
Festin
R
Öberg
G
Simonsson
B
Tötterman
TH
B lymphocyte regeneration in marrow and blood after autologous bone marrow transplantation: Increased numbers of B cells carrying activation and progression markers.
Leuk Res
13
1989
791
12
Small
TN
Keever
CA
Weiner-Fedus
S
Heller
G
O’Reilly
RJ
Flomenberg
N
B-cell differentiation following autologous, conventional, or T-cell depleted bone marrow transplantation: A recapitulation of normal B-cell ontogeny.
Blood
76
1990
1647
13
Leitenberg
D
Rappeport
JM
Smith
BR
B-cell precursor bone marrow reconstitution after bone marrow transplantation.
Am J Clin Pathol
102
1994
231
14
Uckun
FM
Haisig
S
Ledbetter
JA
Fidler
P
Myers
DE
Kuebelbeck
V
Weisdorf
D
Gajl-Peczalska
K
Kersey
JH
Ramsay
KC
Developmental hierarchy during early human B cell ontogeny after autologous bone marrow transplantation using autografts depleted of CD19+ B cell precursors by an anti-CD19 pan B cell immunotoxin containing pokeweed antiviral protein.
Blood
79
1992
3369
15
Pedrazzini
A
Freedman
AS
Andersen
J
Heflin
L
Anderson
K
Takvorian
T
Canellos
GP
Whitman
J
Coral
F
Ritz
J
Nadler
LM
Anti-B-cell monoclonal antibody-purged autologous bone marrow transplantation for B-cell non-Hodgkin’s lymphoma: Phenotypic reconstitution and B-cell function.
Blood
74
1989
2203
16
Fumoux
F
Guigou
V
Blaise
D
Maraninchi
D
Fougereau
M
Schiff
C
Reconstitution of human immunoglobulin VH repertoire after bone marrow transplantation mimics B-cell ontogeny.
Blood
81
1993
3153
17
Storek
J
King
L
Ferrara
S
Marcelo
D
Saxon
A
Braun
J
Abundance of a restricted fetal B cell repertoire in marrow transplant recipients.
Bone Marrow Transplant
14
1994
783
18
Suzuki
I
Milner
EC
Glas
AM
Hufnagle
WO
Rao
SP
Pfister
L
Nottenburg
C
Immunoglobulin heavy chain variable region gene usage in bone marrow transplant recipients: Lack of somatic mutation indicates a maturational arrest.
Blood
87
1996
1873
19
Saxon
A
Mitsuyasu
R
Stevens
R
Champlin
RE
Kimata
H
Gale
RP
Designed transfer of specific immune responses with bone marrow transplantation.
J Clin Invest
78
1986
959
20
Wimperis
JZ
Brenner
MK
Prentice
HG
Reittie
JE
Karayannis
P
Griffiths
PD
Hoffbrand
AV
Transfer of a functioning humoral immune system in transplantation of T-lymphocyte-depleted bone marrow.
Lancet
1
1986
339
21
Engelhard
D
Nagler
A
Hardan
I
Morag
A
Aker
M
Baciu
H
Strauss
N
Parag
G
Naparstek
E
Ravid
Z
Or
R
Slavin
S
Zakay-Rones
Z
Antibody response to a two-dose regimen of influenza vaccine in allogeneic T cell-depleted and autologous BMT recipients.
Bone Marrow Transplant
11
1993
1
22
Ilan
Y
Nagler
A
Adler
R
Naparstek
E
Or
R
Slavin
S
Brautbar
C
Shouval
D
Adoptive transfer of immunity to hepatitis B virus after T cell-depleted allogeneic bone marrow transplantation.
Hepatology
18
1993
246
23
Lane
HC
Zunich
KM
Wilson
W
Cefali
F
Easter
M
Kovacs
JA
Masur
H
Leitman
SF
Klein
HG
Steis
RG
Longo
DL
Fauci
AS
Syngeneic bone marrow transplantation and adoptive transfer of peripheral blood lymphocytes combined with zidovudine in human immunodeficiency virus (HIV) infection.
Ann Intern Med
113
1990
512
24
Molrine
DC
Guinan
EC
Antin
JH
Wheeler
C
Parsons
SK
Weinstein
HJ
McGarigle
C
Blanding
P
Phillips
NR
Ciamarra
A
George
S
Ambrosino
DM
Haemophilus influenzae type b (HIB)-conjugate immunization before bone marrow harvest in autologous bone marrow transplantation.
Bone Marrow Transplant
17
1996
1149
25
Witherspoon
RP
Lum
LG
Storb
R
Thomas
ED
In vitro regulation of immunoglobulin synthesis after human marrow transplantation. II. Deficient T and non T-lymphocyte function within 3-4 months of allogeneic, syngeneic, or autologous marrow grafting for hematologic malignancy.
Blood
59
1982
844
26
Kiesel
S
Pezzutto
A
Moldenhauer
G
Haas
R
Körbling
M
Hunstein
W
Dörken
B
B-cell proliferative and differentiative responses after autologous peripheral blood stem cell or bone marrow transplantation.
Blood
72
1988
672
27
Nadler
LM
Takvorian
T
Botnick
L
Bast
RC
Finberg
R
Hellman
S
Canellos
GP
Schlossman
SF
Anti-B1 monoclonal and complement treatment in autologous bone marrow transplantation for relapsed B-cell non-Hodgkin’s lymphoma.
Lancet
2
1984
427
28
Armitage
RJ
Goldstone
AH
Richards
JDM
Cawley
JC
Lymphocyte function after autologous bone marrow transplantation (BMT): A comparison with patients treated with allogeneic BMT and with chemotherapy only.
Br J Haematol
63
1986
637
29
Rocha
B
Guy-Grand
D
Vassalli
P
Extrathymic T cell differentiation.
Curr Opin Immunol
7
1995
235
30
Dejbakhsh-Jones
S
Jerabek
L
Weissman
IL
Strober
S
Extrathymic maturation of αβ T cells from hemopoietic stem cells.
J Immunol
155
1995
3338
31
Sugita
K
Soiffer
RJ
Murray
C
Schlossman
SF
Ritz
J
Morimoto
C
The phenotype and reconstitution of immunoregulatory T cell subsets after T-cell-depleted allogeneic and autologous bone marrow transplantation.
Transplantation
57
1994
1465
32
Olsen
GA
Gockerman
JP
Bast
RC
Borowitz
M
Peters
WP
Altered immunologic reconstitution after standard-dose chemotherapy or high-dose chemotherapy with autologous bone marrow support.
Transplantation
46
1988
57
33
Bengtsson
M
Tötterman
TH
Smedmyr
B
Festin
R
Öberg
G
Simonsson
B
Regeneration of functional and activated NK and T subset cells in the marrow and blood after autologous bone marrow transplantation: A prospective phenotypic study with 2/3-color FACS analysis.
Leukemia
3
1989
68
34
Mackall
CL
Fleisher
TA
Brown
MR
Andrich
MP
Chen
CC
Feuerstein
IM
Horowitz
ME
Magrath
IT
Shad
AT
Steinberg
SM
Wexler
LH
Gress
RE
Age, thymopoiesis, and CD4+ T-lymphocyte regeneration after intensive chemotherapy.
N Engl J Med
332
1995
143
35
Kast
WM
De Waal
LP
Melief
CJM
Thymus dictates major histocompatibility complex (MHC) specificity and immune response gene phenotype of class II MHC-restricted T cells but not of class I MHC restricted T cells.
J Exp Med
160
1984
1752
36
Steinmann
GG
Klaus
B
Müller-Hermelink
H-K
The involution of the ageing human thymic epithelium is independent of puberty. A morphometric study.
Scand J Immunol
22
1985
563
37
Mackall
CL
Fleisher
TA
Brown
MR
Andrich
MP
Chen
CC
Feuerstein
IM
Magrath
IT
Wexler
LH
Dimitrov
DS
Gress
RE
Distinctions between CD8+ and CD4+ T-cell regenerative pathways result in prolonged T-cell subset imbalance after intensive chemotherapy.
Blood
89
1997
3700
38
Azuma
M
Phillips
JH
Lanier
LL
CD28- T lymphocytes. Antigenic and functional properties.
J Immunol
150
1993
1147
39
Storek
J
Witherspoon
RP
Storb
R
T cell reconstitution after bone marrow transplantation into adult patients does not resemble T cell development in early life.
Bone Marrow Transplant
16
1995
413
40
Weaver
CH
Longin
K
Buckner
CD
Bensinger
W
Lymphocyte content in peripheral blood mononuclear cells collected after the administration of recombinant human granulocyte colony-stimulating factor.
Bone Marrow Transplant
13
1994
411
41
Ottinger
HD
Beelen
DW
Scheulen
B
Schaefer
UW
Grosse-Wilde
H
Improved immune reconstitution after allotransplantation of peripheral blood stem cell instead of bone marrow.
Blood
88
1996
2275
42
Hénon
PR
Liang
H
Beck-Wirth
G
Eisenmann
JC
Lepers
M
Wunder
E
Kandel
G
Comparison of hematopoietic and immune recovery after autologous bone marrow or blood stem cell transplants.
Bone Marrow Transplant
9
1992
285
43
Koehne
G
Zeller
W
Stockschlaeder
M
Zander
AR
Phenotype of lymphocyte subsets after autologous peripheral blood stem cell transplantation.
Bone Marrow Transplant
19
1997
149
44
Ho
AD
Maruyama
M
Maghazachi
A
Mason
JR
Glück
S
Corringham
RET
Soluble CD4, soluble CD8, Soluble CD25, lymphopoietic recovery, and endogenous cytokines after high-dose chemotherapy and blood stem cell transplantation.
Blood
84
1994
3550
45
Kiesel
S
Pezzutto
A
Körbling
M
Haas
R
Schulz
R
Hunstein
W
Dörken
B
Autologous peripheral blood stem cell transplantation: Analysis of autografted cells and lymphocyte recovery.
Transplant Proc
21
1989
3084
46
Talmadge
JE
Reed
EC
Kessinger
A
Kuszynski
CA
Perry
GA
Gordy
CL
Mills
KC
Thomas
ML
Pirruccello
SJ
Letheby
BA
Arneson
MA
Jackson
JD
Immunologic attributes of cytokine mobilized peripheral blood stem cells and recovery following transplantation.
Bone Marrow Transplant
17
1996
101
47
Talmadge
JE
Reed
E
Ino
K
Kessinger
A
Kuszynski
C
Heimann
D
Varney
M
Jackson
J
Vose
JM
Bierman
PJ
Rapid immunologic reconstitution following transplantation with mobilized peripheral blood stem cells as compared to bone marrow.
Bone Marrow Transplant
19
1997
161
48
Ashihara
E
Shimazaki
C
Yamagata
N
Hirata
T
Okawa
K
Oku
N
Goto
H
Inaba
T
Fujita
N
Nakagawa
M
Reconstitution of lymphocyte subsets after peripheral blood stem cell transplantation: Two-color flow cytometric analysis.
Bone Marrow Transplant
13
1994
377
49
Vilmer
E
Triebel
F
David
V
Rabian
C
Schumpp
M
Leca
G
Degos
L
Hercend
T
Sigaux
F
Bensussan
A
Prominent expansion of circulating lymphocytes bearing γ T-cell receptors, with preferential expression of variable γ genes after allogeneic bone marrow transplantation.
Blood
72
1988
841
50
van der Harst
D
Brand
A
van Luxemburg-Heijs
SAP
Kooij-Winkelaar
YMC
Zwaan
FE
Koning
F
Selective outgrowth of CD45RO+ Vγ9+/Vδ2+ T-cell receptor γ/δ T cells early after bone marrow transplantation.
Blood
78
1991
1875
51
Villers
D
Milpied
N
Gaschet
J
Davodeau
F
Hallet
M-M
Bonneville
M
Vié
H
Alteration of the T cell repertoire after bone marrow transplantation.
Bone Marrow Transplant
13
1994
19
52
Gorochov
G
Debré
P
Leblond
V
Sadat-Sowti
B
Sigaux
F
Autran
B
Oligoclonal expansion of CD8+CD57+ T cells with restricted T-cell receptor β chain variability after bone marrow transplantation.
Blood
83
1994
587
53
Gaschet
J
Denis
C
Milpied
N
Hallet
M-M
Romagné
F
Necker
A
Vivien
R
David-Ameline
J
Davodeau
F
Bonneville
M
Vié
H
Alterations of T cell repertoire after bone marrow transplantation: Characterization of over-represented subsets.
Bone Marrow Transplant
16
1995
427
54
Krangel
MS
Yssel
H
Brocklehurst
C
Spits
H
A distinct wave of human T cell receptor γ/δ lymphocytes in early life thymus: Evidence for controlled gene rearrangement and cytokine production.
J Exp Med
172
1990
847
55
Brenner
MK
Rill
DR
Holladay
MS
Heslop
HE
Moen
RC
Buschle
M
Krance
RA
Santana
VM
Anderson
FW
Ihle
JN
Gene marking to determine whether autologous marrow infusion restores long-term haematopoiesis in cancer patients.
Lancet
342
1993
1134
56
Butturini
A
Seeger
RC
Gale
RP
Recipient immune-competent T lymphocytes can survive intensive conditioning for bone marrow transplantation.
Blood
68
1986
954
57
Roux
E
Helg
C
Dumont-Girard
F
Chapuis
B
Jeannet
M
Roosnek
E
Analysis of T-cell population after allogeneic bone marrow transplantation: Significant differences between recipients of T-cell depleted and unmanipulated grafts.
Blood
87
1996
3984
58
Kato
S
Yabe
H
Yabe
M
Kimura
M
Ito
M
Tsuchida
F
Tsuji
K
Takahashi
M
Studies on transfer of varicella-zoster-virus specific T cell immunity from bone marrow donor to recipient.
Blood
75
1990
806
59
Rouleau
M
Senik
A
Leroy
E
Vernant
JP
Long-term persistence of transferred PPD-reactive T cells after allogeneic bone marrow transplantation.
Transplantation
55
1993
72
60
Müller-Hermelink
HK
Sale
GE
Borisch
B
Storb
R
Pathology of the thymus after allogeneic bone marrow transplantation in man.
Am J Pathol
129
1987
242
61
Mackall
CL
Granger
L
Sheard
MA
Cepeda
R
Gress
RE
T-cell regeneration after bone marrow transplantation: Differential CD45 isoform expression on thymic-derived versus thymic-independent progeny.
Blood
82
1993
2585
62
Mackall
CL
Bare
CV
Granger
LA
Sharrow
SO
Titus
JA
Gress
RE
Thymic-independent T cell regeneration occurs via antigen-driven expansion of peripheral T cells resulting in a repertoire that is limited in diversity and prone to skewing.
J Immunol
156
1996
4609
63
Miller
RA
Daley
J
Ghalie
R
Kaizer
H
Clonal analysis of T-cell deficiencies in autotransplant recipients.
Blood
77
1991
1845
64
Cayeux
S
Meuer
S
Pezzutto
A
Körling
M
Haas
R
Schulz
R
Dörken
B
T-cell ontogeny after autologous bone marrow transplantation: Failure to synthesize interleukin-2 (IL-2) and lack of CD2- and CD3-mediated proliferation by both CD4+ and CD8+ cells even in the presence of exogeneous IL-2.
Blood
74
1989
2270
65
Katsanis
E
Anderson
PM
Filipovich
AH
Hasz
DE
Rich
ML
Loeffler
CM
Ochoa
AC
Weisdof
DJ
Proliferation and cytolytic function of anti-CD3 + interleukin-2 stimulated peripheral blood mononuclear cells following bone marrow transplantation.
Blood
78
1991
1286
66
Sugita
K
Nojima
Y
Tachibana
K
Soiffer
RJ
Murray
C
Schlossman
SF
Ritz
J
Morimoto
C
Prolonged impairment of very late activating antigen-mediated T cell proliferation via the CD3 pathway after T cell-depleted allogeneic bone marrow transplantation.
J Clin Invest
94
1994
481
67
Lum
LG
Joshi
ID
Galoforo
SC
Abella
E
Karanes
C
Ratanatharathorn
V
Schultz
KR
Uberti
JP
Sensenbrenner
LL
Ledbetter
JA
June
CH
Coactivation with anti-CD28 monoclonal antibody enhances anti-CD3 monoclonal antibody-induced proliferation and IL-2 synthesis in T cells from autologous bone marrow transplant recipients.
Bone Marrow Transplant
12
1993
565
68
Cayeux
S
Meuer
S
Pezzutto
A
Körbling
M
Haas
R
Schulz
R
Dörken
B
Allogeneic mixed lymphocyte reactions during a second round of ontogeny: Normal accessory cells did not restore defective interleukin-2 (IL-2) synthesis in T cells but induced responsiveness to exogenous IL-2.
Blood
74
1989
2278
69
Welte
K
Keever
CA
Levick
J
Bonilla
MA
Merluzzi
VJ
Mertelsmann
R
Evans
R
O’Reilly
RJ
Interleukin-2 production and response to interleukin-2 by peripheral blood mononuclear cells from patients after bone marrow transplantation: II. Patients receiving soybean lectin-separated and T cell-depleted bone marrow.
Blood
70
1987
1595
70
Mielcarek
M
Martin
PJ
Torok-Storb
B
Suppression of alloantigen-induced T-cell prolidferation by CD14+ cels derived from granulocyte colony-stimulating factor-mobilized peripheral blood mononuclear cells.
Blood
89
1997
1629
71
Ageitos
AG
Ino
K
Ozerol
I
Tarantolo
S
Heimann
DG
Talmadge
JE
Restoration of T and NL cell function in GM-CSF mobilized stem cell products from breast cancer patients by monocyte depletion.
Bone Marrow Transplant
20
1997
117
72
Reusser
P
Attenhofer
R
Hebart
H
Helg
C
Chapuis
B
Einsele
H
Cytomegalovirus-specific T-cell immunity in recipients of autologous peripheral blood stem cell or bone marrow transplants.
Blood
89
1997
3873
73
Nolte
A
Buhmann
R
Straka
C
Emmerich
B
Hallek
M
Assessment and characterization of the cytolytic T lymphocyte response against Epstein-Barr virus in patients with non-Hodgkin’s lymphoma after autologous peripheral blood stem cell transplantation.
Bone Marrow Transplant
21
1998
909
74
Reittie
JE
Gottlieb
D
Heslop
HE
Leger
O
Drexler
HG
Hazlehurst
G
Hoffbrand
AV
Prentice
HG
Brenner
MK
Endogenously generated killer cells circulate after autologous and allogeneic marrow transplantation but not chemotherapy.
Blood
73
1989
1352
75
Higuchi
CM
Thompson
JA
Cox
T
Lindgren
CG
Buckner
CD
Fefer
A
Lymphokine-activated killer function following autologous bone marrow transplantation for refractory hematological malignancies.
Cancer Res
49
1989
5509
76
Neubauer
MA
Benyunes
MC
Thomson
JA
Bensinger
WI
Lindgren
CG
Buckner
CD
Fefer
A
Lymphokine-activated killer (LAK) precursor cell activity is present in infused peripheral blood stem cells and in the blood after autologous peripheral blood stem cell transplantation.
Bone Marrow Transplant
13
1994
311
77
Fegan
C
Thomas
H
Bailey-Wood
R
Coleman
S
Philips
S
Hoy
T
Wittaker
JA
In vitro LAK (lymphokine activated killer) activity following autologous peripheral blood stem cell is significantly greater than that following autologous bone marrow and allogeneic bone marrow transplantation.
Bone Marrow Transplant
16
1995
277
78
Pavletic
Z
Benyunes
MC
Thompson
JA
Lindgren
CG
Massumoto
C
Alderson
MR
Buckner
CD
Fefer
A
Induction by interleukin-7 of lymphokine-activated killer activity in lymphocytes from autologous and syngeneic marrow transplant recipients before and after systemic interleukin-2 therapy.
Exp Hematol
21
1993
1371
79
Guillaume
T
Sekhavat
M
Rubinstein
DB
Hamdan
O
Leblanc
P
Symann
ML
Defective cytokine production following autologous stem cell transplantation for solid tumors and hematologic malignancies regardless of bone marrow or peripheral origin, and lack of evidence for a role for IL-10 in delayed immune reconstitution.
Cancer Res
54
1994
3800
80
Bosly
AE
Staquet
PJ
Doyen
CM
Chatelain
BJ
Humblet
YP
Symann
ML
Recombinant human interleukin-2 restores in vitro T-cell colony formation by peripheral blood mononuclear cells after autologous bone marrow transplantation.
Exp Hematol
15
1987
1048
81
Hanenberg
H
Dilloo
D
Laws
H-J
Zessack
N
Heyll
A
Burdach
S
Time course of interferon-γ production deficiency after autologous and allogeneic stem cell transplantation for malignancies.
Exp Hematol
23
1995
1543
82
Anderson
KC
Ritz
J
Takvorian
T
Coral
F
Daley
H
Gorgone
BC
Freedman
AS
Canellos
GP
Schlossman
SF
Nadler
LM
Hematologic engraftment and immune reconstitution posttransplantation with anti-B1 purged autologous bone marrow.
Blood
69
1987
597
83
Bosly
A
Guillaume
T
Brice
P
Humblet
Y
Staquet
P
Doyen
C
Chatelain
B
Franks
C
Gisselbrecht
C
Symann
M
Effects of escalating doses of recombinant human interleukin-2 (r-IL-2) in correcting functional T-cell defects following autologous bone marrow transplantation for lymphomas and solid tumors.
Exp Hematol
20
1992
962
84
Roberts
MM
To
LB
Gillis
D
Mundy
J
Rawling
C
Ng
K
Juttner
CA
Immune reconstitution following peripheral blood stem cell transplantation, autologous marrow transplantation and allogeneic bone marrow transplantation.
Bone Marrow Transplant
12
1993
469
85
Trinchieri
G
Interleukin-12: A cytokine produced by antigen-presenting cells with immunoregulatory functions in the generation of T-helper type 1 and cytotoxic lymphocytes.
Blood
84
1994
4008
86
Brunda
MJ
Luistro
L
Warrier
RR
Wright
RB
Hubbard
BR
Murphy
M
Wolf
SF
Gately
MK
Antitumor and anti-metastatic activity of interleukin 12 against murine tumors.
J Exp Med
178
1993
1223
87
Nastala
CL
Edington
HD
McKinney
TG
Tahara
H
Nalesnik
MA
Brunda
MJ
Gately
MK
Wolf
SF
Schreiber
RD
Storkus
WJ
Lotze
MT
Recombinant IL-12 administration induces tumor regression in association with IFN-γ production.
J Immunol
153
1994
1697
88
Soiffer
R
Robertson
M
Murray
C
Cochran
K
Ritz
J
Interleukin-12 augments cytolytic activity of peripheral blood lymphocytes from patients with hematological and solid malignancies.
Blood
82
1993
2790
89
Guillaume
T
Kubin
M
Sekhavat
M
Rubinstein
DB
Trinchieri
G
Symann
M
Peripheral blood mononuclear cells derived from patients undergoing autologous hematopoietic stem cell transplantation produce and respond to IL-12.
Bone Marrow Transplant
18
1996
733
90
Shu
U
Kiniwa
M
Wu
CY
Maliszewski
C
Vezzio
N
Hakimi
J
Gately
M
Delespesse
G
Activated T cells induce interleukin-12 production by monocytes via CD40-CD40 ligand interaction.
Eur J Immunol
25
1995
1125
91
Lindgren
CG
Thompson
JA
Robinson
N
Keeler
T
Gold
PJ
Fefer
A
Interleukin-12 induces cytolytic activity in lymphocytes from recipients of autologous and allogeneic stem cell transplants.
Bone Marrow Transplant
19
1997
867
92
Dorshkind
K
Pollack
SB
Bosma
MJ
Phillips
RA
Natural killer cells are present in mice with severe combined immunodeficiency (SCID).
J Immunol
134
1985
3798
93
Hackett
J
Bosma
GC
Bosma
MJ
Bennett
M
Kumar
V
Transplantable progenitors of natural killer cells are distinct from those of T and B lymphocytes.
Proc Natl Acad Sci USA
93
1986
3427
94
Wahren
B
Gahrton
G
Linde
A
Ljungman
P
Lonnqvist
B
Ringden
O
Sundqvist
VA
Transfer and persistence of viral antibody-producing cells in bone marrow transplantation.
J Infect Dis
150
1984
358
95
Gratama
JW
Verdonck
LF
van der Linden
JA
van Heugten
JG
Kreeft
HAJG
D’Amaro
J
Zwaan
FE
de Gast
GC
Cellular immunity to vaccinations and herpes-virus infections after bone marrow transplantation.
Transplantation
41
1986
719
96
Meyers
JD
Flournoy
N
Thomas
ED
Infection with herpes simplex virus and cell-mediated immunity after marrow transplant.
J Infect Dis
142
1980
338
97
Wade
JC
Day
LM
Crowley
JJ
Meyers
JD
Recurrent infection with herpes simplex virus after marrow transplantation: Role of the specific immune response and acyclovir treatment.
J Infect Dis
149
1980
750
98
Reusser
P
Fisher
LD
Buckner
CD
Thomas
ED
Meyers
JD
Cytomegalovirus infection after autologous bone marrow transplantation: Occurrence of cytomegalovirus disease and effect on engrafment.
Blood
75
1990
1888
99
Schuchter
LM
Wingard
JR
Piantadosi
S
Burns
WH
Santos
GW
Saral
R
Herpes zoster infection after autologous bone marrow transplantation.
Blood
74
1989
1424
100
Whimbey
E
Elting
LS
Couch
RB
Lo
W
Williams
L
Champlin
RE
Bodey
GP
Influenza A virus infections among hospitalized adult bone marrow transplant recipients.
Bone Marrow Transplant
13
1994
437
101
Boeckh
M
Gooley
TA
Reusser
P
Buckner
CD
Bowden
RA
Failure of high-dose acyclovir to prevent cytomegalovirus disease after autologous marrow transplantation.
J Infect Dis
172
1995
939
102
Mazumder
A
Rosenberg
SA
Successful immunotherapy of natural killer-resistant established pulmonary melanoma metastases by intravenous adoptive transfer of syngeneic lymphocytes activated in vitro by interleukin-2.
J Exp Med
159
1984
495
103
Rosenberg
SA
Mulé
J
Spiess
P
Reicher
CM
Schwartz
SL
Regression of established pulmonary metastases and subcutaneous tumor mediated by the systemic administration of high-dose recombinant interleukin 2 .
J Exp Med
161
1985
1169
104
Rosenberg
SA
Lotze
MT
Muul
LM
Leitman
S
Chang
AE
Ettinghausen
SE
Matory
YL
Skibber
JM
Shiloni
E
Vetto
JT
Seipp
CA
Simpson
C
Reichert
CM
Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer.
N Engl J Med
313
1985
1485
105
Atkins
MB
Interleukin-2 therapy: A decade of slow but steady progress.
Cancer J Sci Am
2
1996
73
106
Oshimi
K
Oshimi
Y
Akutsu
M
Takei
Y
Saito
H
Okada
M
Mizoguchi
H
Cytotoxicity of interleukin 2-activated lymphocytes for leukemia and lymphoma cells.
Blood
68
1986
938
107
Lotzova
E
Savary
CA
Herberman
RB
Induction of NK cell activity against fresh human leukemia in culture with interleukin-2.
J Immunol
138
1987
2718
108
Fierro
MT
Liao
XS
Lusso
P
Bonferroni
M
Matera
L
Cesano
A
Lista
P
Arione
R
Forni
G
Foa
R
In vitro and in vivo susceptibility of human leukemic cells to lymphokine activated killer activity.
Leukemia
2
1988
50
109
Ackerstein
A
Kedar
E
Slavin
S
Use of recombinant human interleukin-2 in conjunction with syngeneic bone marrow transplantation in mice as a model for control of minimal residual disease in malignant hematologic disorders.
Blood
78
1991
1212
110
Gottlieb
DJ
Prentice
HG
Heslop
HE
Bello-Fernandez
C
Bianchi
AC
Galazka
AR
Brenner
MK
Effect of recombinant interleukin-2 on cytotoxic function following high-dose chemo-radiotherapy for hematological malignancy.
Blood
74
1989
2335
111
Blaise
D
Olive
D
Stoppa
AM
Viens
P
Pourreau
C
Lopez
M
Attal
M
Jasmin
C
Monges
G
Mawas
C
Mannoni
P
Palmer
P
Franks
C
Philip
T
Maraninchi
D
Hematologic and immunologic effects of the systemic administration of recombinant interleukin-2 after autologous bone marrow transplantation.
Blood
76
1990
1092
112
Heslop
H
Duncombe
AS
Reittie
J
Bello-Fernandez
C
Gottlieb
DJ
Prentice
HG
Hoffbrand
AV
Brenner
MK
Interleukin-2 infusion after autologous bone marrow transplantation of chemotherapy enhances hemopoietic regeneration.
Transplant Proc
23
1991
1704
113
Higuchi
C
Thompson
JA
Petersen
FB
Buckner
CD
Fefer
A
Toxicity and immunomodulatory effects of interleukin-2 after autologous bone marrow transplantation for hematological malignancies.
Blood
77
1991
2561
114
Soiffer
R
Murray
C
Cochran
K
Cameron
C
Wang
E
Schow
PW
Daley
JF
Ritz
J
Clinical and immunologic effects of prolonged infusion of low-dose recombinant interleukin-2 after autologous and T-cell-depleted allogeneic bone marrow transplantation.
Blood
79
1992
517
115
Benyunes
MC
Massumoto
C
York
A
Higuchi
CM
Buckner
CD
Thompson
JA
Petersen
FB
Fefer
A
Preliminary report. Interleukin-2 with or without lymphokine-activated killer cells as consolidative immunotherapy after autologous bone marrow transplantation for acute myelogenous leukemia.
Bone Marrow Transplant
12
1993
159
116
Hamon
MD
Prentice
HG
Gottlieb
DJ
Macdonald
ID
Cunningham
JM
Smith
OP
Gilmore
M
Gandhi
L
Collis
C
Immunotherapy with interleukin 2 after ABMT in AML.
Bone Marrow Transplant
11
1993
399
117
Benyunes
MC
Higuchi
C
York
A
Lindgren
C
Thompson
JA
Buckner
CD
Fefer
A
Immunotherapy with interleukin 2 with or without lymphokine-activated killer cells after autologous bone marrow transplantation for malignant lymphoma: A feasibility trial.
Bone Marrow Transplant
16
1995
283
118
Fefer
A
Benyunes
MC
Massumoto
C
Higuchi
C
York
A
Buckner
CD
Thompson
JA
Interleukin-2 therapy after autologous bone marrow transplantation for hematologic malignancies.
Semin Oncol
20
1993
41
119
Weisdorf
DJ
Anderson
PM
Blazar
BR
Uckun
FM
Kersey
JH
Ramsay
NKC
Interleukin 2 immediately after autologous bone marrow transplantation for acute lymphoblastic leukemia: A phase I study.
Transplantation
55
1993
61
120
Attal
M
Blaise
D
Marit
G
Payen
C
Michallet
M
Vernant
JP
Sauvage
C
Troussard
X
Nedellec
G
Pico
J
Huguet
F
Stoppa
AM
Broustet
A
Sotto
JJ
Pris
J
Maraninchi
D
Reiffers J for the BGMT Group
Consolidation treatment of adult acute lymphoblastic leukemia: A prospective, randomized trial comparing allogeneic versus autologous bone marrow transplantation and testing the impact of recombinant interleukin-2 after autologous bone marrow transplantation.
Blood
86
1995
1619
121
Robinson
N
Benyunes
MC
Thomson
JA
York
A
Petersdorf
S
Press
O
Lindgren
C
Chauncey
T
Buckner
CD
Bensinger
WI
Appelbaum
FR
Fefer
A
Interleukin-2 after autologous stem cell transplantation for hematologic malignancy: A phase I/II study.
Bone Marrow Transplant
19
1997
435
122
Miller
JS
Tessmer-Tuck
J
Pierson
BA
Weisdorf
D
McGlave
P
Blazar
BR
Katsanis
E
Verfaillie
C
Lebkowsky
J
Radford
J
Jr
Burns
LJ
Low dose subcutaneous interleukin-2 after autologous transplantation generates sustained in vivo natural killer cell activity.
Biol Blood Marrow Transplant
3
1997
34
123
Blaise
D
Attal
M
Pico
JL
Reiffers
J
Stoppa
AM
Bellanger
C
Molina
L
Nedellec
G
Vernant
JP
Legros
M
Gabus
R
Huguet
F
Brandely
M
Hercend
T
Olive
D
Maraninchi
D
The use of a sequential high dose recombinant interleukin 2 regimen after autologous bone marrow transplantation does not improve the disease free survival of patients with acute leukemia transplanted in first complete remission.
Leuk Lymphoma
25
1997
469
124
(abstr, suppl 1)
Blaise
D
Attal
M
Reiffers
J
Stoppa
AM
Huguet
F
Marit
G
Molina
L
Bellanger
C
Pico
JL
Gabus
R
Brandely
M
Hercend
T
Maraninchi
D
Randomized study of interleukin 2 (RIL2) after autologous bone marrow transplantation (BMT) in 130 patients with acute leukemia (AL) in first complete remission (CR1): Results of a multicentric trial with a 5 year follow up.
Blood
90
1997
113a
125
Valteau-Couanet
D
Rubie
H
Meresse
V
Farace
F
Brandely
M
Hartmann
O
Phase I-II study of interleukin-2 after high-dose chemotherapy and autologous bone marrow transplantation in poorly responding neuroblastoma.
Bone Marrow Transplant
16
1995
515
126
Heslop
HE
Duncombe
AS
Reittie
JE
Bello-Fernandez
C
Gottlieb
DJ
Prentice
HG
Mehta
AB
Hoffbrand
AV
Brenner
MK
Interleukin 2 infusion induces haematopoietic growth factors and modifies marrow regeneration after chemotherapy or autologous marrow transplantation.
Br J Haematol
77
1991
237
127
López-Jiménez
Pérez-Oteyza
Muñoz
Parra
C
Villalón
L
Ramos
P
Maldonado
M
Garcı́a-Laraña
Otheo
E
Roldán
E
Garcı́a-Avello
A
Odriozola
J
Subcutaneous versus intravenous low-dose IL-2 therapy after autologous transplantation: Results of a prospective, non-randomized study.
Bone Marrow Transplant
19
1997
429
128
Peschon
JJ
Morrissey
PJ
Grabstein
KH
Ramsdell
FJ
Maraskovsky
E
Gliniak
BC
Park
LS
Ziegler
SF
Williams
DE
Ware
CB
Meyer
JD
Davison
BL
Early lymphocyte expansion is severely impaired in interleukin 7 receptor-deficient mice.
J Exp Med
180
1994
1955
129
von Freeden-Jerry
U
Vierira
P
Lucian
LA
McNeil
T
Burdach
SE
Murray
R
Lymphopenia in interleukin (IL)-7 gene-deleted mice identifies IL-7 as a non redundant cytokine.
J Exp Med
181
1995
1519
130
Boerman
OC
Gregorio
TA
Grzegorzewski
KJ
Faltynek
CR
Kenny
JJ
Wiltrout
RH
Komschlies
KL
Recombinant human IL-7 administration in mice affects colony-forming units-spleen and lymphoid precursors cell localization and accelerates engraftment of bone marrow transplants.
J Leukoc Biol
58
1995
151
131
Bolotin
E
Smogorzewska
M
Smith
S
Widmer
M
Weinberg
K
Enhancement of thymopoiesis after bone marrow transplant by in vivo interleukin-7.
Blood
88
1996
1887
132
Abdul-Hai
A
Reuven
O
Slavin
S
Friedman
G
Weiss
L
Matsa
D
Ben-Yehuda
A
Stimulation of immune reconstitution by interleukin-7 after syngeneic bone marrow transplantation in mice.
Exp Hematol
24
1996
1416
133
Agah
R
Malloy
B
Kerner
M
Mazumder
A
Generation and characterization of IL-2-activated bone marrow cells as a potent graft vs tumor effector in transplantation.
J Immunol
143
1989
3093
134
Agah
R
Malloy
B
Kerner
M
Girgis
E
Bean
P
Twomey
P
Mazumder
A
Potent graft antitumor effect in natural killer-resistant disseminated tumors by transplantation of interleukin-2 activated syngeneic bone marrow in mice.
Cancer Res
49
1989
5959
135
Charak
BS
Brynes
RK
Groshen
S
Chen
S-C
Mazumder
A
Bone marrow transplantation with interleukin-2 activated bone marrow followed by interleukin-2 therapy for acute myeloid leukemia in mice.
Blood
76
1990
2187
136
Charak
BS
Agah
R
Mazumder
A
Granulocyte-macrophage colony-stimulating factor-induced antibody-dependent cellular cytotoxicity in bone marrow macrophages: Application in bone marrow transplantation.
Blood
81
1993
3474
137
Charak
BS
Agah
R
Brynes
RK
Chogyoji
M
Groshen
S
Chen
SC
Mazumder
A
Interleukin-2 (IL-2) and IL-2-activated bone marrow in transplantation: Evaluation from a clinical perspective.
Bone Marrow Transplant
9
1992
479
138
Adler
A
Chervenick
PA
WhitesideTL
Lotzova
E
Herberman
RB
Interleukin 2 induction of lymphokine-activated killer (LAK) activity in the peripheral blood and bone marrow of acute leukemia patients. I. Feasibility of LAK generation in adult patients with active disease and in remission.
Blood
71
1988
706
139
Keever
CA
Pekle
K
Gazzola
MV
Collins
NH
Gillio
A
NK and LAK activities from marrow progenitors. I. The effects of interleukin-2 and interleukin-1.
Cell Immunol
126
1990
211
140
Klingemann
H-G
Deal
H
Reid
D
Eaves
CJ
Design and validation of a clinically applicable culture procedure for the generation of interleukin-2 activated natural killer cells in human bone marrow autografts.
Exp Hematol
21
1993
1263
141
Margolin
KA
Wright
C
Forman
SJ
Autologous bone marrow purging by in situ IL-2 activation of endogenous killer cells.
Leukemia
11
1997
723
142
(abstr, suppl 1)
Meehan
KR
Verma
UN
Frankel
SR
Rajagopal
C
Cahill
R
Arun-Killic
B
Jenson
M
Oquendo
C
Mazumder
A
Immunotherapy with IL-2 and α-IFN after PBSC transplantation for women with breast cancer.
Blood
86
1995
387a
143
Areman
EM
Mazumder
A
Kotula
PL
Verma
UN
Rajagopal
C
Hancock
S
Guevarra
C
Djahanmir
M
Sacher
RA
Meehan
KR
Hematopoietic potential of IL-2-cultured peripheral blood stem cells from breast cancer patients.
Bone Marrow Transplant
18
1996
521
144
Miller
JS
Klingsporn
S
Lund
J
Perry
EH
Verfaillie
C
McGlave
P
Large scale ex vivo expansion and activation of human natural killer cells for autologous therapy.
Bone Marrow Transplant
14
1994
555
145
Miller
JS
Verfaillie
C
McGlave
PB
Expansion and activation of human natural killer cells as therapy for autologous transplantation.
Prog Clin Biol Res
389
1994
39
146
Charak
BS
Brynes
RK
Katsuda
S
Groshen
S
Chen
S-C
Mazumder
A
Induction of graft versus leukemia effect in bone marrow transplantation: Dosage and time schedule dependency of interleukin-2 therapy.
Cancer Res
51
1991
2015
147
Klingemann
H-G
Eaves
CJ
Barnett
MJ
Eaves
AC
Hogge
DE
Nantel
SH
Reece
E
Sheperd
JD
Sutherland
HJ
Phillips
GL
Transplantation of patients with high risk acute myeloid leukemia in first remission with autologous marrow cultured in interleukin-2 followed by interleukin-2 administration.
Bone Marrow Transplant
14
1994
389
148
Beaujean
F
Bernaudin
F
Kuentz
M
Lemerle
S
Cordonnier
C
Le Forestier
C
Reinert
P
Duedari
N
Brandely
M
Vernant
J-P
Successful engraftment after autologous transplantation of 10-day cultured bone marrow activated by interleukin 2 in patients with acute lymphoblastic leukemia.
Bone Marrow Transplant
15
1995
691
149
Margolin
KA
Negrin
RS
Wong
KK
Chatterjee
S
Wright
C
Forman
SJ
Cellular immunotherapy and autologous transplantation for hematologic malignancy.
Immunol Rev
157
1997
231
150
Glazier
AD
Tutschka
PJ
Farmer
ER
Santos
GW
Graft-versus-host disease in cyclosporine A treated rats following syngeneic and autologous bone marrow reconstitution.
J Exp Med
158
1983
1
151
Hess
AD
Horwitz
L
Beshorner
WE
Santos
GW
Development of graft-versus-host disease-like syndrome in cyclosporine-treated rats after syngeneic bone marrow transplantation. I. Development of cytotoxic T-lymphocytes with apparent polyclonal anti-Ia specificity, including autoreactivity.
J Exp Med
161
1985
718
152
Shinozawa
T
Beshorner
WE
Hess
AD
The thymus and prolonged administration of cyclosporine. Irreversible immunopathological changes associated with autologous pseudo-graft-versus-host disease.
Transplantation
50
1990
106
153
Sorokin
R
Kimura
H
Schroder
K
Wilson
DH
Wilson
DB
Cyclosporine-induced autoimmunity. Conditions for expressing disease, requirement for intact thymus, and potency estimates of autoimmune lymphocytes in drug-treated rats.
J Exp Med
164
1986
1615
154
Jenkins
MK
Schwartz
RH
Pardoll
DM
Effects of cyclosporine A on T cell development and clonal deletion.
Science
241
1988
1655
155
Fisher
AC
Beshorner
WE
Hess
AD
Requirements for the induction and adoptive transfer of cyclosporine-induced syngeneic graft-versus-host disease.
J Exp Med
169
1989
1031
156
Fisher
AC
Laulis
MK
Horwitz
L
Beshorner
WE
Hess
AD
Host resistance to cyclosporine induced syngeneic graft-versus-host disease.
J Immunol
143
1989
827
157
(suppl 3)
Beshorner
WE
Suresch
DL
Shinozawa
T
Hess
AD
Influence of irradiation and age on the CsA-induced thymic immunopathology.
Transplant Proc
20
1988
1072
158
Hess
AD
Horwitz
LR
Laulis
MK
Cyclosporin induced syngeneic graft-versus-host disease: Prevention of autoaggression by treatment with monoclonal antibodies to T lymphocyte cell surface determinants and to MHC class II antigens.
Clin Immunol Immunopathol
69
1993
341
159
Hess
AD
Bright
EC
Thoburn
C
Vogelsang
GB
Jones
RJ
Kennedy
MJ
Specificity and antitumor activity of effector T lymphocytes in autologous graft-vs-host disease: Role of the MHC class II invariant chain peptide.
Blood
89
1997
2203
160
Geller
RB
Esa
AH
Beshorner
WE
Frondoza
CG
Santos
GW
Hess
AD
Successful in vitro graft-versus-tumor effect against an Ia-bearing tumor using cyclosporine-induced syngeneic graft-versus-host disease in the rat.
Blood
74
1989
1165
161
Bryson
JS
Jennings
CD
Caywood
BE
Kaplan
AM
Strain specificity in the induction of syngeneic graft-versus-host disease in mice.
Transplantation
51
1991
911
162
Charak
BS
Agah
R
Mazumder
A
Synergism of interleukin-2 and cyclosporine A in induction of a graft-versus-tumor effect without graft-versus-host disease after syngeneic bone marrow transplantation.
Blood
80
1992
179
163
Hooton
JWL
Miller
CL
Helgason
CD
Bleackley
RC
Gillis
S
Paetkau
V
Development of pretoxic T cells in cyclosporine-suppressed mixed lymphocyte reactions.
J Immunol
144
1990
816
164
Jones
RJ
Vogelsang
GB
Hess
AD
Farmer
ER
Mann
RB
Geller
RB
Piantadosi
S
Santos
GW
Induction of graft-versus-host disease after autologous bone marrow transplantation.
Lancet
1
1989
754
165
Yeager
AM
Vogelsang
GB
Jones
RJ
Farmer
ER
Altomonte
V
Hess
AD
Santos
GW
Induction of cutaneous graft-versus-host disease by administration of cyclosporine to patients undergoing autologous bone marrow transplantation for acute myeloid leukemia.
Blood
79
1992
3031
166
Kennedy
MJ
Vogelsang
GB
Beveridge
RA
Farmer
ER
Altomonte
V
Huelskamp
AM
Davidson
NE
Phase I trial of intravenous cyclosporine to induce graft-versus-host disease in women undergoing autologous bone marrow transplantation for breast cancer.
J Clin Oncol
11
1993
478
167
Kennedy
MJ
Vogelsang
GB
Jones
RJ
Farmer
ER
Hess
DA
Altomonte
V
Huelskamp
AM
Davidson
NE
Phase I trial of interferon gamma to potentiate cyclosporine-induced graft-versus-host disease in women undergoing autologous bone marrow transplantation for breast cancer.
J Clin Oncol
12
1994
249
168
Ratanatharathorn
V
Uberti
J
Karanes
C
Lum
LG
Abella
E
Dan
ME
Hussein
M
Sensenbrenner
LL
Phase I study of alpha-interferon augmentation of cyclosporine-induced graft-versus-host disease in recipients of autologous bone marrow transplantation.
Bone Marrow Transplant
13
1994
625
169
Giralt
S
Weber
D
Colome
M
Dimopoulos
M
Mehra
R
Van Besien
K
Gajewski
J
Andersson
B
Khouri
I
Przepiorka
D
von Wolff
B
Delasalle
K
Korbling
M
Seong
D
Alexanian
R
Champlin
R
Phase I trial of cyclosporine-induced autologous graft-versus-host disease in patients with multiple myeloma undergoing high-dose chemotherapy with autologous stem-cell rescue.
J Clin Oncol
15
1997
667
170
Gryn
J
Johnson
E
Goldman
N
Devereux
L
Grana
G
Hageboutros
A
Fernandez
E
Constantinou
C
Harrer
W
Viner
E
Goldberg
J
The treatment of relapsed or refractory intermediate grade non-Hodgkin’s lymphoma with autologous bone marrow transplantation followed by cyclosporine and interferon.
Bone Marrow Transplant
19
1997
221
171
Long
GS
Cramer
DV
Harnaha
JB
Hiserodt
JC
Lymphokine-activated killer (LAK) cell purging of leukemic bone marrow: Range of activity against different hematopoietic neoplasms.
Bone Marrow Transplant
6
1990
169
172
Schlegel
PG
Schmidt-Wolf
G
Schmidt-Wolf
IGH
Kwak
LW
Chao
NJ
Lymphokine-activated killer cells activity against autologous lymphoma cells following bone marrow transplantation.
Cancer Res Ther Control
3
1993
145
173
Gambacorti-Passerini
C
Rivoltini
L
Supino
R
Rodolfo
M
Radrizzani
M
Fossati
G
Parmiani
G
Susceptibility of chemoresistant murine and human tumor cells to lysis by interleukin 2-activated lymphocytes.
Cancer Res
48
1988
2372
174
Lister
J
Rybka
WB
Donnenberg
AD
de Magalhaes-Silverman
M
Pincus
SM
Bloom
EJ
Elder
EM
Ball
ED
Whitheside
TL
Autologous peripheral blood stem cell transplantation and adoptive with activated natural killer cells in the immediate posttransplant period.
Clin Cancer Res
1
1995
607
175
Schmidt-Wolf
IGH
Lefterova
P
Mehta
BA
Fernandez
LP
Huhn
D
Blume
KG
Weissman
IL
Negrin
RS
Phenotypic characterization and identification of effector cells involved in tumor cell recognition of cytokine-induced killer cells.
Exp Hematol
21
1993
1673
176
Lu
PH
Negrin
RS
A novel population of expanded human CD3+CD56+ cells derived from T cells with potent in vivo antitumor activity in mice with severe combined immunodeficiency.
J Immunol
153
1994
1687
177
Schmidt-Wolf
IGH
Lefterova
P
Johnston
V
Shefold
C
Csipai
M
Mehta
B
Tsuruo
T
Huhn
D
Negrin
RS
Sensitivity of multidrug-resistant tumor cell lines to immunologic effector cells.
Cell Immunol
169
1995
85
178
Schmidt-Wolf
IGH
Negrin
RS
Kiem
H
Blume
KG
Weissman
IL
Use of a SCID mouse/human lymphoma model to evaluate cytokine-induced killer cells with potent anti-tumor cell activity.
J Exp Med
174
1991
139
179
Cesano
A
Visonneau
S
Cioé
L
Clark
S
Rovera
G
Santoli
D
Reversal of acute myelogenous leukemia in humanized SCID mice using a novel adoptive transfer approach.
J Clin Invest
94
1994
1076
180
Cesano
A
Visonneau
S
Jeglum
KA
Owen
J
Wilkinson
K
Carner
K
Reese
L
Santoli
D
Phase I clinical trial with a human major histocompatibility complex nonrestricted cytotoxic T-cell line (TALL-104) in dogs with advanced tumors.
Cancer Res
56
1996
3021
181
Visonneau
S
Cesano
A
Torosian
MH
Santoli
D
Cell therapy of a highly invasive human breast carcinoma implanted in immunodeficient (SCID) mice.
Clin Cancer Res
3
1997
1491
182
Gong
J-H
Maki
G
Kingemann
H-G
Characterization of a human cell line (NK-92) with phenotypical and functional characteristics of activated natural killer cells.
Leukemia
8
1994
652
183
Robertson
MJ
Cochran
KJ
Cameron
C
Le
J-M
Tantravahi
R
Ritz
J
Characterization of a cell line, NKL, derived from an aggressive natural killer cell leukemia.
Exp Hematol
24
1996
406
184
Riddell
SR
Greenberg
PD
Cellular adoptive immunotherapy after bone marrow transplantation.
Cancer Treat Res
76
1995
337
185
Riddell
SR
Watanabe
KS
Goodrich
JM
Li
CR
Agha
ME
Greenberg
PD
Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cells clones.
Science
257
1992
238
186
Walter
EA
Greenberg
PD
Gilbert
MJ
Finch
RJ
Watanabe
KS
Thomas
ED
Riddell
SR
Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor.
N Engl J Med
333
1995
1038
187
Kwak
LW
Campbell
MJ
Czerwinski
DK
Hart
S
Miller
RA
Levy
R
Induction of immune responses in patients with B-cell lymphoma against the surface-immunoglobulin idiotype expressed by their tumors.
N Engl J Med
327
1992
1209
188
Nowell
PC
Hungerford
DA
A minute chromosome in human chonic granulocytic leukemia.
Science
132
1960
1497
189
Clark
SS
McLaughlin
J
Crist
WM
Champlin
R
Witte
ON
Unique forms of the abl tyrosine kinase distinguish Ph1-positive CML from Ph1-positive ALL.
Science
235
1987
85
190
Kurzrock
R
Shtalrid
M
Romero
P
Kloetzer
WS
Talpas
M
Trujillo
JM
Blick
M
Beran
M
Gutterman
JU
A novel c-abl protein product in Philadelphia-positive acute lymphoblastic leukaemia.
Nature
325
1987
631
191
de Thé
H
Chomienne
C
Lanotte
M
Degos
L
Dejean
A
The t(15;17) translocation of acute promyelocytic leukemia fuses the retinoic acid receptor alpha gene to a novel transcribed locus.
Nature
347
1990
558
192
Tsujimoto
Y
Finger
LR
Yunis
Nowell
PC
Croce
CM
Cloning of the chromosome breakpoint of neoplastic B cells with the (14;18) chromosome translocation.
Science
226
1984
1097
193
Bos
JL
Verlaan-de Vries
M
van der Eb
AJ
Janssen
JWG
Delwel
R
Löwenberg
B
Colly
LP
Mutations in N-ras predominate in acute myeloid leukemia.
Blood
69
1987
1237
194
Imamura
J
Miyoshi
I
Koeffler
HP
p53 in hematologic malignancies.
Blood
84
1994
2412
195
Dengler
R
Münstermann
U
Al-Batran
S
Hausner
I
Faderl
S
Nerl
C
Emmerich
B
Immunocytochemical and flow cytometric detection of proteinase 3 (myeloblastin) in normal and leukaemic myeloid cells.
Br J Haematol
89
1995
250
196
Sainsbury
JR
Farndon
JR
Needman
GK
Malcolm
AJ
Harris
AL
Epidermal-growth factor receptor status as predictor of early recurrence of and death from breast cancer.
Lancet
1
1987
1398
197
Ro
J
North
SM
Gallick
GE
Hortobagyi
GN
Gutterman
JU
Blick
M
Amplified and overexpressed epidermal growth factor receptor gene in uncultured primary human breast carcinoma.
Cancer Res
48
1988
161
198
Slamon
DJ
Clark
GM
Wong
WJ
Levin
A
Ullrich
A
McGuire
WL
Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene.
Science
235
1987
177
199
Slamon
DJ
Godolphin
W
Jones
LA
Holt
JA
Wong
SG
Keith
DE
Levin
WJ
Stuart
SG
Udoove
J
Ullrich
A
Press
MJ
Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer.
Science
244
1989
707
200
Lemoine
NR
Barnes
DM
Hollywood
DP
Hughes
CM
Smith
P
Dublin
E
Prigent
SA
Gullick
WJ
Hurst
HC
Expression of the ERBB3 gene product in breast cancer.
Br J Cancer
66
1992
1116
201
Theillet
C
Adelaide
J
Louason
G
Bonnet-Dorion
F
Jacquemier
J
Adnane
J
Longy
M
Katsaros
D
Sismondi
P
Gaudray
P
FGFRI and PLAT genes and DNA amplification at 8p12 in breast and ovarian cancers.
Genes Chromosomes Cancer
7
1993
219
202
Saitoh
S
Cunningham
J
de Vries
EMG
McGovern
RM
Schroeder
JJ
Hartmann
A
Blaszyk
H
Wold
LE
Schaid
D
Sommer
SS
Kovach
JS
p53 gene mutations in breast cancers in midwestern US women: Null as well as missense-type mutations are associated with poor prognosis.
Oncogene
9
1994
2869
203
Kovach
JS
Hartmann
A
Blaszyk
H
Cunningham
J
Schaid
D
Sommer
SS
Mutation detection by highly sensitive methods indicates that p53 gene mutations in breast cancer can have important prognostic value.
Proc Natl Acad Sci USA
93
1996
1093
204
Gendler
S
Taylor-Papadimitriou
J
Duhig
T
Rothbard
J
Burchell
J
A highly immunogenic region of a human polymorphic epithelial mucin expressed by carcinomas is made up of tandem repeats.
J Biol Chem
263
1988
12820
205
Siddiqui
J
Abe
M
Hayes
D
Shani
E
Yunis
E
Kufe
D
Isolation and sequencing of a cDNA coding for he human DF3 breast carcinoma-associated antigens.
Proc Natl Acad USA
85
1988
2320
206
Thompson
JA
Grunert
F
Zimmermann
W
Carcinoembryonic antigen gene family: Molecular biology and clinical perspectives.
J Clin Lab Anal
5
1991
344
207
Jung
S
Schluesener
HJ
Human T lymphocytes recognize a peptide of single point-mutated, oncogeneic ras proteins.
J Exp Med
173
1991
273
208
Gambacorti-Passerini
C
Grignani
F
Arienti
F
Pandolfi
PP
Pelicci
PG
Parmiani
G
Human CD4 lymphocytes specifically recognize a peptide representing the fusion region of the hybrid protein pml/RAR alpha present in acute promyelocytic leukemia cells.
Blood
81
1993
1369
209
Bocchia
M
Korontsvit
T
Xu
Q
Mackinnon
S
Yang
SY
Sette
A
Scheinberg
DA
Specific human cellular immunity to bcr-abl oncogene-derived peptides.
Blood
87
1996
3587
210
ten Bosch
GJA
Joosten
AM
Kessler
JH
Melief
CJM
Leeksma
OC
Recognition of BCR-ABL positive leukemic blasts by human CD4+ T cells elicited by primary in vitro immunization with a BCR-ABL breakpoint peptide.
Blood
88
1996
3522
211
Buzyn
A
Ostankovitch
M
Zerbib
A
Kemula
M
Connan
F
Varet
B
Guillet
JG
Choppin
J
Peptides derived from the whole sequence of BCR-ABL bind to several class I molecules allowing specific induction of human cytotoxic T lymphocytes.
Eur J Immunol
27
1997
2066
212
Van Pel
A
van der Bruggen
P
Coulie
PG
Brichard
VG
Lethe
B
van den Eynde
B
Uyttenhove
C
Renauld
JC
Boon
T
Genes coding for tumor antigens recognized by cytolytic T lymphocytes.
Immunol Rev
145
1995
229
213
Barnd
DL
Lan
MS
Metzgar
RS
Finn
OJ
Specific, major histocompatibility complex-unrestricted recognition of tumor-associated mucins by human cytotoxic T cells.
Proc Natl Acad Sci USA
86
1989
7159
214
Ionnanides
CG
Fisk
B
Jerome
KR
Irimura
T
Wharton
JT
Finn
OJ
Cytotoxic T cells from ovarian malignant tumors can recognize polymorphic epithelial mucin core peptide.
J Immunol
151
1993
3963
215
Disis
ML
Smith
JW
Murphy
AE
Chen
W
Cheever
MA
In vitro generation of human cytolytic T-cells specific for peptides derived from the HER-2/neu protooncogene protein.
Cancer Res
54
1994
16
216
Houbiers
JGA
Nijman
HW
van der Burg
SH
Drijfhout
JW
Kenemans
P
van de Velde
CJH
Brand
A
Momburg
F
Kast
WM
Melief
CJM
In vitro induction of human cytotoxic T lymphocyte responses against peptides of mutant and wild-type p53.
Eur J Immunol
23
1993
2072
217
Alexander-Miller
MA
Leggatt
GR
Berzofsky
JA
Selective expansion of high- or low-avidity cytotoxic T lymphocytes and efficacy for adoptive immunotherapy.
Proc Natl Acad Sci USA
93
1996
4102
218
Wick
M
Dubey
P
Koeppen
H
Siegel
CT
Fields
PE
Chen
L
Bluestone
JA
Schreiber
H
Antigenic cancer cells grow progressively in immune hosts without evidence for T cell exhaustion or systemic anergy.
J Exp Med
186
1997
229
219
Steinman
RM
The dendritic cell system and its role in immunogenicity.
Annu Rev Immunol
9
1991
271
220
Paglia
P
Chiodoni
C
Rodolfo
M
Colombo
MP
Murine dendritic cells loaded in vitro with soluble protein prime cytotoxic T lymphocytes against tumor antigen in vivo.
J Exp Med
183
1996
317
221
Inaba
K
Metlay
JP
Crowley
MT
Steinman
RM
Dendritic cells pulsed with protein antigens in vitro can prime antigen-specific, major histocompatibility complex-restricted T cells in situ.
J Exp Med
172
1990
631
222
Flamand
V
Sornasse
T
Thielemans
K
Demanet
C
Bakkus
M
Bazin
H
Tielemans
F
Leo
O
Urbain
J
Moser
M
Murine dendritic cells pulsed in vitro with tumor antigen induce tumor resistance in vivo.
Eur J Immunol
24
1994
605
223
Mayordomo
JI
Zorina
T
Storkus
WJ
Zitvogel
L
Celluzzi
C
Falo
LD
Melief
CJ
Ildstad
ST
Kast
WM
DeLeo
AB
Bone marrow-derived dendritic cells pulsed with synthetic tumor peptides elicit protective and therapeutic antitumor immunity.
Nat Med
1
1995
1297
224
Zitvogel
L
Mayordomo
JI
Tjandrawan
T
DeLeo
AB
Clarke
MR
Lotze
MT
Storkus
WJ
Therapy of murine tumors with tumor peptide-pulsed dendritic cells: Dependence on T cells, B7 costimulation, and T helper cell 1-associated cytokines.
J Exp Med
183
1996
87
225
Celluzi
CM
Mayordomo
JI
Storkus
WJ
Lotze
MT
Falo LD Jr
Peptide-pulsed dendritic cells induce antigen-specific CTL-mediated protective tumor immunity.
J Exp Med
183
1996
283
226
Porgador
A
Snyder
D
Gilboa
E
Induction of antitumor immunity using bone marrow-generated dendritic cells.
J Immunol
156
1996
2918
227
Hsu
FJ
Benike
C
Fagnoni
F
Liles
TM
Czerwinski
D
Taidi
B
Engleman
EG
Levy
R
Vaccinations of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells.
Nat Med
2
1996
52
228
Bocchia
M
Wentworth
PA
Southwood
S
Sidney
J
McGraw
K
Scheinberg
DA
Sette
A
Specific binding of leukemia oncogene fusion protein peptides to HLA class I molecules.
Blood
85
1995
2680
229
Fisk
B
Blevins
TL
Taylor Wharton
J
Ioannides
CG
Identification of an immunodominant peptide of HER-2/neu protooncogene recognized by ovarian tumor-specific cytotoxic T lymphocyte lines.
J Exp Med
181
1995
2109
230
Buzyn
A
Ostankovitch
M
Zerbib
A
Kemula
M
Connan
F
Varet
B
Guillet
J-G
Choppin
J
Peptides derived from the whole sequence of BCR-ABL bind to several class I molecules allowing specific induction of human cytotoxic T lymphocytes.
Eur J Immunol
27
1997
2066
231
Molldrem
JJ
Clave
E
Jiang
YZ
Mavroudis
D
Raptis
A
Hensel
N
Agarwala
V
Barrett
AJ
Cytotoxic T lymphocytes specific for a nonpolymorphic proteinase 3 peptide preferentially inhibit chronic myeloid leukemia colony-forming units.
Blood
90
1997
2529
232
Peoples
GE
Goedegebuure
PS
Smith
R
Linehan
DC
Yoshino
I
Eberlein
TJ
Breast and ovarian cancer-specific cytotoxic T lymphocytes recognize the same HER2/neu-derived peptide.
Proc Natl Acad USA
92
1995
432
233
Linehan
DC
Goedegebuure
PS
Peoples
GE
Rogers
SO
Eberlein
TJ
Tumor-specific and HLA-A2-restricted cytolysis by tumor-associated lymphocytes in human metastatic breast cancer.
J Immunol
155
1995
4486
234
Doménech
N
Henderson
RA
Finn
OJ
Identification of an HLA-A11-restricted epitope from the tandem repeat domain of the epithelial tumor antigen mucin.
J Immunol
155
1995
4766
235
Storkus
WJ
She HJ
III
Salter
RD
Lotze
MT
Identification of T-cell epitopes: Rapid isolation of class I-presented peptides from viable cells by mild acid elution.
J Immunother
14
1993
94
236
Bernhardt
H
Disis
ML
Heimfeld
S
Hand
S
Gralow
JR
Cheever
MA
Generation of immunostimulatory dendritic cells from human CD34+ hematopoietic progenitor cells of the bone marrow and peripheral blood.
Cancer Res
55
1995
1099
237
Szabolcs
P
Moore
MAS
Young
JW
Expansion of immunostimulatory dendritic cells among the myeloid progeny of human CD34+ bone marrow precursors cultured with c-kit ligand, granulocyte-macrophage colony-stimulating factor, and TNF-α.
J Immunol
154
1995
5851
238
Siena
S
Di NicolaM
Bregni
M
Mortarini
R
Anichini
A
Lombardi
L
Ravagnani
F
Parmiani
G
Gianni
AM
Massive ex vivo generation of functional dendritic cells from mobilized CD34+ blood progenitors for anticancer therapy.
Exp Hematol
23
1995
1463
239
Fisch
P
Kohler
G
Garbe
A
Herbst
B
Wider
D
Kohler
H
Schaefer
H
Mertelsmann
R
Brugger
W
Kanz
L
Generation of antigen-presenting cells for soluble protein antigens ex vivo from peripheral blood CD34+ hematopoietic progenitor cells in cancer patients.
Eur J Immunol
26
1996
595
240
Henderson
RA
Nimgaonkar
MT
Watkins
SC
Robbins
PD
Ball
ED
Finn
OJ
Human dendritic cells genetically engineered to express high levels of the human epithelial tumor antigen mucin (MUC-1).
Cancer Res
56
1996
3763
241
Reeves
ME
Royal
RE
Lam
JS
Rosenberg
SA
Hwu
P
Retroviral transduction of human dendritic cells with a tumor-associated antigen gene.
Cancer Res
56
1996
5672
242
Aicher
A
Westermann
J
Cayeux
S
Willimsky
G
Daemen
K
Blankenstein
T
Uckert
W
Dörken
B
Pezzutto
A
Successful retroviral mediated transduction of a reporter gene in human dendritic cells: Feasibility of therapy with gene-modified antigen presenting cells.
Exp Hematol
25
1997
39
243
Ribas
A
Butterfield
LH
McBride
WH
Jilani
SM
Bui
LA
Vollmer
CM
Lau
R
Dissette
VB
Hu
B
Chen
AY
Glaspy
JA
Economou
JS
Genetic immunization for the melanoma antigen MART-1/Melan-A using recombinant adenovirus-transduced murine dendritic cells.
Cancer Res
57
1997
2865
244
Wan
Y
Bramson
J
Carter
R
Graham
F
Gauldie
J
Dendritic cells transduced with an adenoviral vector encoding a model tumor-associated antigen for tumor vaccination.
Human Gene Ther
8
1997
1355
245
Arthur
JF
Butterfield
LH
Roth
MD
Bui
LA
Kiertscher
SM
Lau
R
Dubinett
S
Glaspy
J
McBride
WH
Economou
JS
A comparison of gene transfer methods in human dendritic cells.
Cancer Gen Ther
4
1997
17
246
(abstr, suppl 1)
Slavin
S
Naparstek
E
Nagler
A
Ackerstein
A
Drakos
P
Kapelushnik
Y
Brautbar
C
Or
R
Cell mediated immunotherapy (CMI) for the treatment of malignant hematological diseases in conjunction with autologous bone marrow tranplantation (ABMT).
Blood
82
1993
292a
247
Bonini
C
Ferrari
G
Verzeletti
PS
Zappone
E
Ruggieri
L
Ponzoni
M
Rossini
S
Mavilio
F
Traversari
C
Bordignon
C
HSV-tk gene transfer into donor lymphocytes for control of allogeneic graft-versus-leukemia.
Science
276
1997
1719
Sign in via your Institution