At diagnosis, approximately 80% of patients with follicular lymphoma (FL) have disseminated disease with one or more extranodal localizations (Ann Arbor stage IV), mostly corresponding to bone marrow (BM) involvement. Conventional chemotherapy is not curative nor does it substantially modify the natural course of the disease. Most patients with disseminated FL ultimately die from the disease, with a median survival time of 8 to 10 years.1 Innovative therapies have to be proposed to these patients. Some form of intensive therapy—for example, high-dose chemotherapy (HDT) or chemoradiotherapy followed by autologous or allogeneic hematopoietic stem cell transplantation—is an option. Recently, the autologous stem cell transplantation (ASCT) procedure has become easier and cheaper, and it has a mortality rate of below 5% and manageable morbidity. It could therefore be considered for patients with FL if it could be shown to improve survival. Allogeneic stem cell transplantation is another innovative approach, adding a graft-versus-lymphoma (GvL) effect. However, the high mortality rate of the procedure limits its indications. Nonmyeloablative allogeneic transplantation could increase the indications by decreasing the transplantation-related mortality (TRM).

Clinical results

Progressing or relapsing patients.

Johnson et al2 have clearly shown that, after each subsequent relapse of FL, the response rate to treatment decreases and the progression-free survival (PFS) time shortens (Table1). This explains why most intensive therapies have been performed in patients with disseminated disease who have either not responded to initial conventional treatment or relapsed after one or more regimens of chemotherapy. The data from several retrospective series of ASCT in progressing/relapsing FL patients have been published. The main reports are summarized in Table2.3-17 They all indicate encouraging results, with a response rate higher than 90%, low (5%-10%) TRM, and long median relapse-free survival (RFS) and overall survival (OS) times. Nevertheless, most patients still relapse after ASCT, and cure has not been demonstrated. Besides, conclusions are difficult to draw on the basis of these reports because of the presence of one or more skewing factors, including different classifications of low-grade lymphomas, patients with and without histological transformation, the selection of patients who were suitable for ASCT, differences in conditioning regimens, the use of BM or peripheral blood stem cells, reinjection of purged or unpurged stem cells, and short follow-up periods.

Disease-free and OS patterns from the largest single center series, which included 153 patients treated with intensive therapy followed by autologous bone marrow transplantation (ABMT) at the Dana Farber Cancer Institute (DFCI, Boston, MA), are shown in Figure1.6 Several groups have compared progression-free survival (PFS) and OS in patients treated with intensive therapy to that in matched historical controls treated with conventional chemotherapy. Patients treated with ASCT had a significantly better rate of freedom from second relapse5,8and, in some series8 but not in others,5better OS. None of these comparisons with historical control groups are case-controlled studies, and they suffer from a major skewing factor in that only those patients who were suitable candidates for intensive therapy, especially with respect to age, performance status, chemosensitivity, marrow infiltration, and response to chemotherapy, were selected.

Fig. 1.

Kaplan-Meier estimates of probability of disease-free survival (A) and overall survival (B) for 153 patients treated with ABMT for progressive/relapsing indolent follicular lymphoma at the DFCI (from Freedman et al6 with the authorization of the author and editor).

Fig. 1.

Kaplan-Meier estimates of probability of disease-free survival (A) and overall survival (B) for 153 patients treated with ABMT for progressive/relapsing indolent follicular lymphoma at the DFCI (from Freedman et al6 with the authorization of the author and editor).

Close modal

Although these results clearly suggest that intensive therapy is probably superior to conventional chemotherapy in relapsing FL patients, only a randomized trial could confirm this hypothesis. Such a trial was launched in Europe for chemotherapy unpurged bone marrow or purged bone marrow (the CUP trial) but was stopped because of poor patient recruitment (89 randomized patients in 4 years). The trial showed a significant improvement in the progression/relapse rate in the ASCT group (44% in the chemotherapy arm as opposed to 32% in the intensive arm, P = .006) without any difference in OS.18 

Several prognostic factors that significantly influenced PFS and/or OS after ASCT have been described: (1) age (with a threshold around 50 years)13; (2) B symptoms at diagnosis and/or on relapse6,19 and/or increased serum lactate dehydrogenase (LDH) levels20; (3) number of previous chemotherapy regimens11,13,21; (4) clinical status at the time of ASCT (patients who have resistant disease are less likely to benefit from ASCT than those with chemosensitive disease)4,11,13,20; (5) presence or absence of histological transformation (several groups have reported on small numbers of patients with histologically transformed FL treated with HDT).11,15,22-27 In the first report of high-risk patients with histologically transformed FL, poor performance status, and bulky or chemoresistant disease, the results were poor with only 1 patient of 10 still surviving one year after HDT.22 More recent series have reported better results (Table3).11,15,23,24,27 The European Bone Marrow Transplantation (EBMT) registry reported on 50 patients treated with HDT for histologically transformed FL.25 Median PFS after HDT was 13 months, and OS was 64% and 51% at 2 and 5 years, respectively. Results of HDT performed after histological transformation were not different from those of 200 matched, untransformed FL and 200 de novo cases of aggressive non-Hodgkin lymphoma (NHL) treated with HDT (P = .67 and .44).25 These results can be considered as better than those reported for patients treated with conventional chemotherapy.28 However, results of HDT in histologically transformed FL are too limited to allow any firm conclusions to be drawn.); (6) histological BM involvement at the time of stem cell harvest6; (7) the bcl-2-JH polymerase chain reaction (PCR) graft status after the purge procedure, if any was performed. The influence of a negative PCR bcl-2-JH rearrangement has been clearly demonstrated by retrospective analysis of patients who were treated with ABMT at the DFCI. PCR detected residual disease at the time of marrow stem cell harvest in 113 of 153 patients. After immunologically purged ABMT, BM PCR analysis showed no detectable disease in 48 patients (42%), whereas 65 (58%) remained PCR positive. Patients whose BM was negative for the bcl-2-JH rearrangement after purged ABMT experienced longer freedom from relapse (FFR) than those whose BM remained positive [6 of 48 relapses versus 49 of 65, and 8-year FFRs of 83% vs 19%, respectively,P < .001].)6; and, finally, (8) residual disease after ASCT. In the DFCI study, continued bcl-2-JH PCR negativity in blood or BM during follow-up was also strongly predictive of prolonged complete remission (CR) after ABMT (5 relapses among the 47 patients with persistently negative samples during follow-up compared to 36 of 39 relapses in patients with positive PCR in all BM samples after ABMT). PCR positivity after purging and in BM samples after transplantation were the only parameters associated with a poorer FFR after ABMT in a univariate analysis (P < .0001).6 Some studies have confirmed the prognostic influence of a negative bcl-2-JH PCR result after ASCT,29-31 but others showed a similar relapse rate whatever the PCR status.32 Real-time quantitative PCR evaluations that provide an accurate and reproducible estimate of the number of gene copies with a low carry-over contamination risk could account for these discrepancies.33 Using this technique as well as limiting dilution assays combined with 2-step PCR, relapses among 15 patients were associated with increasing numbers of circulating t(14;18)-positive cells and continuous CR with stable cell counts.34 For patients without any detectable chromosomal rearrangement, amplification of the third complementary determining region (CDR III) can be useful as a clonal marker for minimal residual disease.35 No relapse occurred in any of the 10 patients in whom HDT eradicated PCR-detectable lymphoma cells with this technique, whereas all 8 patients with PCR-detectable lymphoma cells after ABMT relapsed (P = .0002).35 

In conclusion, intensive therapy with ASCT should be considered for patients younger than 60 to 65 years with chemosensitive relapsing FL, especially if they have poor prognosis factors at relapse.8 36 This conclusion holds true even for patients with histological transformation, which is, per se, an adverse prognostic factor.

ASCT as first-line therapy.

A few Phase II trials of intensive therapy after maximum response to conventional chemotherapy have been reported in FL patients.4,16,37-44 The results are summarized in Table4. In a nonrandomized comparison of 26 patients treated with a high-dose CHOP-like regimen (cyclophosphamide, vincristine, prednisone, and doxorubicin) followed by ABMT, and 14 patients treated with the same regimen without ABMT, there was a significant improvement in DFS in the ABMT group (a DFS of 54% at 36 months in the group treated with ABMT versus 26% in the group treated with conventional chemotherapy) without any difference in OS.38 Horning et al have compared the survival of 37 patients treated with ABMT after a maximum response to conventional chemotherapy and 61 matched historical controls in CR or unconfirmed CR after conventional chemotherapy. With a median follow-up of 6.5 years, the estimated 10-year survival rate was 86% in the transplantation group and 59% in the conventional chemotherapy group (P = .074).39 Recently, Sanz-Rodriguez et al45 has reported on a retrospective comparison of a group of patients initially treated with ASCT and a group of matched controls treated with a conventional treatment (cyclophosphamide, vincristine, and prednisone [CVP] chemotherapy associated with interferon alpha). With a median follow-up of 87 months and 112 months, respectively, there was no difference in PFS (49.5% and 48%, respectively) or OS (76% and 71%).45 In another nonrandomized comparison, when survival was calculated from the date of diagnosis, there was no difference in event-free survival and OS between a group of 33 patients treated with ASCT at the outset and a group of 22 patients treated with ASCT only on relapse.16 

Several large Phase III trials are under way to test the potential benefits of initial ASCT in FL patients with adverse prognostic factors. The German Lymphoma Study Group is comparing conventional treatment (mitoxantrone-chlorambucil-prednisone followed by interferon alpha maintenance) and HDT (4 to 6 cycles of CHOP followed by DexaBEAM chemotherapy, then ASCT with a cyclophosphamide-TBI conditioning regimen). As of May 1999, 288 patients have been randomized. With a very short median follow-up of 17 months, there has been a significant improvement in DFS for patients treated with HDT, with a 32% risk reduction (P = .001) without any difference in OS.46 Two other trials conducted by the GELA group (Groupe d'Etude des Lymphomes de l'Adulte) and the GOELAMS group (Groupe Ouest-Est pour l'Etude des Leucémies Aigues et des Maladies du Sang) are comparing a conventional chemoimmunotherapy program (a CHOP-like regimen concomitantly associated with interferon alpha) and intensive therapy with the transplantation of either unselected (GELA trial) or purged (GOELAMS trial) peripheral stem cells. So far, 400 patients have been included in the GELA trial. The first interim analysis of the GOELAMS trial on 136 patients has been recently reported. The 4-year PFS was 61% in the HDT group and 27% in the conventional group (P = .027), without significant difference in 4-year OS (83% vs 80%).47 

In conclusion, because of the poor outcomes associated with conventional chemotherapy in FL patients with disseminated disease and adverse prognostic factors, intensive therapy with ASCT is a potential improvement and can be considered for patients under 60 years of age.48 Nevertheless, because its efficacy has not yet been proven, this treatment should be considered only in the context of a clinical trial.

Optimal procedure

Collection and purging of stem cells.

Autologous peripheral blood stem cell transplantation (APBSCT) has progressively replaced ABMT since retrospective analyses49,50 and at least 3 randomized studies51-53 demonstrated that the time taken for neutrophils or platelet recovery, the duration of the hospital stay, the incidence of infectious complications, and the overall cost are less after APBSCT than after ABMT. Besides, the rate of contamination by lymphoma cells is similar with both harvest procedures,54,55 and posttransplantation PFS and OS rates are similar.50 Finally, immune reconstitution is faster after APBSCT than after ABMT.56 

Leukapheresis for peripheral blood stem cell collection may be performed after either conventional chemotherapy followed by granulocyte colony-stimulating factor (G-CSF) treatment57 or G-CSF treatment alone.58,59 In a nonrandomized comparison between these 2 procedures, there were no differences in transplantation outcome (stem-cell engraftment, risk of relapse).60,61 A combination of stem cell factor (SCF) and G-CSF resulted in the collection of a higher number of stem cells62,63 and has to be more extensively evaluated in patients in whom the harvest is insufficient after chemotherapy plus G-CSF or G-CSF alone.64 65 

Several factors influence the number of stem cells that can be collected, including BM involvement,66 the number of previous chemotherapy regimens,66-71 the time interval between collection and the last cycle of chemotherapy,68,72 previous treatments with radiation and/or cytotoxic drugs (especially the total cumulative dose of alkylating agents66), and fludarabine treatment in some series.73-75 

In general, stem cell collections are poorer and the rate of failure to collect a sufficient number of CD34+ stem cells to perform ASCT is greater in low-grade lymphomas than in aggressive lymphomas. The reasons for this are not understood.68 

Lymphoma cells often contaminate BM69,76,77 and peripheral blood54 stem cell collections and may contribute to relapse after ASCT.78 79 Thus, purging collected stem cells ought to help decrease contamination and lower the risk of relapse. Several procedures have been proposed to purge BM and/or peripheral blood stem cells.

(1) In vitro treatment with cytotoxic agents (4-hydroperoxycyclophosphamide or mafosfamide) has deleterious effects on normal hematopoietic stem cells.19 

(2) In vitro treatment with a cocktail of anti–B-cell monoclonal antibodies (anti-CD20, anti-CD10, and anti-CD19) and fresh rabbit complement,80 or passing the blood81 or BM stem cells82 over a column containing anti–B-cell antibodies bound to immunomagnetic beads. This procedure significantly decreases the number of lymphoma cells in a BM graft as assessed by PCR amplification of the bcl-2-JH rearrangement.83 In the DFCI study, 57 of 114 (50%) patients with PCR-positive BM became negative after purging.80 However, in another study, 18 of 21 (86%) patients remained positive after treatment with a cocktail of 4 monoclonal antibodies.84 

(3) In vitro positive selection of CD34+ hematopoietic stem cells,85,86 which produces a 3- to 4-log reduction in tumor contamination.86,87 Although hematopoietic recovery is not delayed88 after ASCT with CD34-selected stem cells, slow T- and B-cell recovery may increase the risk of opportunistic infections.89,90 Nevertheless, CD34-selected grafts may remain contaminated with lymphoma cells.69 91 

(4) Selection of the CD34+CD19 subpopulation that is less contaminated than the CD34+fraction.91 However, even a CD34+CD19CD20 stem cell population may contain the bcl-2-JH rearrangement.92 93 

(5) Ex vivo expansion of the hematopoietic stem cells using a combination of growth factors such as SCF, interleukin 3, interleukin 6, G-CSF, and Flt-3 ligand that accelerates hematological reconstitution after HDT.94,95 This ex vivo expansion of bcl-2-JH–negative CD34+ progenitor cells does not induce the expansion of lymphoma progenitor cells,93 96 but the data are scarce and need confirmation.

(6) More recently, an in vivo purging method has been proposed using a treatment with rituximab before stem cell collection.97,98The results of the first studies are encouraging. When rituximab is combined with chemotherapy, lymphoma cells became undetectable in peripheral blood stem cell collections in 75% (9 of 12),99 93% (14 of 15),97 or 100% (7 of 7)98 of informative cases, a significantly higher rate than in historical control (26%98 to 40%97). However, a single rituximab infusion 3 to 5 days before stem cell collection is insufficient since 10 of 11 stem cell grafts remained PCR-positive.100 Anti-CD20 monoclonal antibody in vivo purging does not have a deleterious effect on stem cell collection, engraftment time, or transplantation outcome.101 102 A trial conducted by the European Bone Marrow Transplantation group is currently assessing the role of rituximab in ASCT for relapsing FL in a 4-arm randomized trial, rituximab being (1) not given in a control group; (2) given before stem cell collection; (3) given after ASCT; and (4) given before collection and after ASCT.

The clinical benefit of the purge procedure remains to be established. After a reduction of lymphoma cell numbers to below a threshold level of 1 lymphoma cell of 104 to 105 mononucleated cells, the risk of relapse is decreased in some studies5,6,76 but is similar to the relapse rate after unpurged ASCT in other retrospective analyses32,103 or in a comparison of registry data.25 Only a randomized study could confirm the role of purging. One such trial has been initiated but was stopped because of poor patient recruitment (the CUP trial),18 and it is unfortunately unrealistic to undertake another trial comparing ASCT with or without purging of the stem cell graft. In vitro purging is an expensive and cumbersome technique. Treatment before stem cell collection with rituximab or another anti–B-cell antibody such as humanized anti-CD22 (hLL2)104is more promising, given that it is both an anti-lymphoma treatment and an in vivo purging procedure. Confirmatory data are needed.

Conditioning regimens.

Schematically there are 2 types of conditioning regimens for ASCT: (1) those based on chemotherapy alone, such as CBV (cyclophosphamide, carmustine, etoposide),105 BEAM (carmustine, etoposide, cytarabine, melphalan),13 or BEAC (carmustine, etoposide, cytarabine, cyclophosphamide)7; and (2) those using a chemoradiotherapy combination with total body irradiation (TBI).5 6 

The efficacy of very low-dose TBI (1.0 to 2.0 Gy in 0.25 to 0.50 Gy fractions) was demonstrated in low-grade NHLs some 25 years ago.106 Usually, TBI as a conditioning regimen before ASCT is administered in 6 fractions of 2 Gy each (fractionated TBI) rather than as a single dose of 12 Gy, which is more toxic.107TBI is usually combined with high-dose cyclophosphamide5,6and, in some studies, VP16.3 108 

There is no randomized study comparing conditioning regimens in FL. Several retrospective comparisons have been reported, but the results are conflicting. Some report better overall survival with chemotherapy alone,109 whereas the opposite was found in others with better OS when fractionated TBI was used.15 In some studies, no difference was observed.16 110 

Treatment with a murine anti-CD20 monoclonal antibody coupled with a high dose of 131I may be an interesting alternative, given that it allows delivery of a significantly higher dose of radiation to the tumor without too much damage to normal organs. The results of a preliminary phase I-II trial where this high-dose radioimmunotherapy was combined with cyclophosphamide and VP16 were promising. Doses up to 75 Gy could be delivered to affected sites without exceeding doses of 20 Gy to the lungs or other radiosensitive organs.111However, such a conditioning regimen can be given only in a limited number of centers. Other Phase I-II trials using high-dose radioimmunotherapy with humanized anti-CD20 and anti-CD22 MoAbs are in progress.112 

Treatments after ASCT

As previously mentioned, residual disease is often present after ASCT and may contribute to relapse.29,113Several post-ASCT adjuvant treatment modalities have been tested to eradicate residual disease and decrease the relapse rate.114 115 

MoAbs conjugated to toxins such as the anti–B-cell immunotoxin anti–B4-blocked ricin have proven disappointing as an adjunctive therapeutic modality after ABMT.116 Rituximab used as an adjunctive immunotherapy after PBSCT is well tolerated but can induce transient neutropenia100,102 or delayed immune reconstitution, although without any increase in infectious complications.117 However, the ultimate determination of its efficacy will require the results of ongoing studies.

Immune-mediated antitumor therapy after ASCT has been recently reviewed in Blood.111 

(1) Cytokines may be of therapeutic value as their endogenous production is decreased after ASCT,118,119 and their administration soon after ASCT120 can activate cytotoxic T cells directed against lymphoma cell antigens.121-123 A phase II trial of rIL-2 (3 to 6 IU/m2/d × 5 d/wk for 4 weeks) combined with interferon (IFN) alpha (3 × 106U/d × 5 d/wk) given after hematological recovery in relapsing lymphoma patients treated with ABMT has shown a significant improvement in survival when compared to historical controls without post-ABMT immunotherapy (4-year survival of 90% vs 46%) and resulted in a significant decrease in relapse rate (20% vs 46%).124However, the hematological and systemic toxicities of IL-2 restrict its usefulness after ASCT.

(2) Cyclosporine A (CyA), which prevents the thymic clonal deletion of autoreactive T lymphocytes, can increase the incidence of autologous graft-versus-host disease (GVHD),125 a syndrome similar to allogeneic cutaneous GVHD, which is observed in 5% to 10% of patients.126 Encouraging results have been reported in a phase II trial of CyA (in combination with αIFN) given after ABMT for NHL,127 but confirming data are lacking.

(3) An anti-idiotype immune reaction may be induced by infusions of specific idiotypes coupled with a potent adjuvant128-130such as granulocyte-macrophage colony-stimulating factor (GM-CSF),131 the C fragment of tetanus toxin,132 or ex vivo–pulsed dendritic cells that act as antigen-presenting cells133,134 and considerably increase the immune response to idiotype proteins. About 50% of patients develop a cellular and/or an antibody-mediated anti-idiotypic reaction, and in these patients the risk of relapse after ASCT is significantly decreased.130,135,136 Tumor vaccination can be performed either after ASCT with minimal residual disease, or before stem cell harvest, which prevents posttransplantation immunosuppression. Using a syngeneic murine bone marrow transplantation (BMT) model, the tumor-free survival rate was greater after vaccination with irradiated GM-CSF–producing autologous tumor cells in the post-BMT period than in the nontransplantation context.137 Evaluating the potential clinical benefits of these modalities requires further studies.

Complications after ASCT

Relapse.

Relapses after ASCT often occur in sites of previous disease (78% among 33 patients who relapsed after ABMT for FL).138 In some retrospective analyses, posttransplantion radiation therapy improved event-free survival when compared to historical controls.19 

Relapses after ASCT often have the same pathological aspects as before intensive therapy,138 but patients may relapse either with histological transformation138 or, conversely, after ASCT for transformed FL with a low-grade FL (“down-staging”).24 Relapses after ASCT tend to respond to conventional chemotherapy138 and/or to monoclonal anti-CD 20 antibodies.139 140 

Myelodysplastic syndromes/acute leukemias and other secondary tumors.

Myelodysplastic syndrome and acute leukemia (MDS/AL) are the major long-term complications of ASCT for FL.21,141-145 Their 5-year incidence for NHL following ASCT has been estimated to be 8% to 14%5,144-146 and may be higher in low-grade as opposed to aggressive NHL.147 The prognosis is very poor, with a median survival of 9.4 months.148 

Some factors may increase the risk of MDS/AL, such as advanced age,142,144,149 prior fludarabine therapy,150the total cumulative dose of alkylating agents,141radiation therapy before ASCT,148,149,151 the use of TBI in the conditioning regimens142,144,149 or stem cell priming with high-dose VP-16,151 the use of peripheral blood stem cell (PBSC) rather than BM progenitors,144 or the median number of cells/kg reinfused.148 Cytogenetic BM analysis before ABMT may allow prediction of the risk of MDS/AL.149,152 The incidence of acquired karyotypic instability observed during serial assessment after ASCT may greatly exceed the clinical incidence of MDS/AL (13 instances of karyotypic instability versus 2 cases of MDS/AL in a total of 22 patients).153 

When the follow-up time is prolonged, the pattern of relapses after ASCT appears as continuous, without any plateau. Moreover, ASCT is not feasible when the marrow reserves are inadequate, and it is poorly effective for patients with refractory disease or extensive BM involvement. In such situations, the curative value of allogeneic SCT can now be assessed by analyzing a greater amount of information.

Clinical results

An analysis of 215 patients reported on in 8 different studies,9,10,154-160 including an observational study of 113 patients collected from 50 centers participating in the International Bone Marrow Transplant Registry (IBMTR),155is summarized in Table 5. From these results, it appears that standard allogeneic BMT was offered mainly to patients with advanced, chemotherapy-resistant disease with extensive BM involvement and after extensive prior therapy in most cases. In these conditions, a high TRM (around 40%) is counterbalanced by the very potent antitumoral effect of allogeneic BMT, allowing high lymphoma-free and OS rates (49% to 80%) due to a low recurrence rate (0% to 20%).10,154-157,159 Indeed, if the registry follow-up method is accurate, only 1 relapse occurred among 33 patients monitored for more than 2 years.155 Interestingly, and in contrast to the outcome with many other malignancies, a full response and long-term disease-free survival have been observed even in patients with refractory disease.10,154,158,161 These data indicate that patients with refractory or recurrent low-grade lymphoma have a better chance of cure when they are treated with allogeneic BMT than when they are treated with ASCT. However, in the IBMTR study, a higher survival rate was associated with good pretransplantation performance status, chemotherapy-sensitive disease, use of a TBI-containing conditioning regimen, and age younger than 40 years.155 Better patient selection and earlier transplantation could improve outcomes.159,162 In one study, patients with sensitive disease not heavily treated have a low TRM rate (14% vs 86% in patients with resistant disease).162 In contrast to ASCT, the long-term pattern of which does not reach a plateau, allogeneic SCT has a curative potential as witnessed by the fact that long-term remission (as determined by molecular analysis) was observed in 3 of 7 evaluable patients.163 

Graft-versus-lymphoma effect

The lower relapse rate after allogeneic BMT compared with autologous BMT is likely due to the lack of BM contamination of the graft and to an immune-mediated graft-versus-lymphoma (GvL) effect. Evidence of a GvL effect come from 2 things.

It can come from the response to persistent disease following allogeneic BMT, which is observed at the same time as the development of active acute or chronic GVHD. In an EBMT retrospective study, a lower recurrence rate was found in patients with chronic GVHD compared with those without GVHD (0% vs 35%,P = .02),164 although this relationship is still controversial.155 However, the incidence of GVHD in FL seems to be remarkably high (Table 4) compared with that in leukemia patients, probably because FL recipients and donors tend to be older.156 

Evidence of a GvL effect also comes from induction of remission after cessation of immunosuppression or donor-leukocyte infusion (DLI).156,165 Quantification of the disease activity by real-time PCR with an internal fluorogenic probe for the t(14;18) translocation has been used to investigate the GvL effect of donor leukocytes given for relapsed follicular NHL after allogeneic BMT.166 Cytoreduction after DLI may take several months to occur.154 159 

So, the better outcome after allogeneic BMT in patients with FL compared to that in patients with high-grade subtypes may be explained not only by the more indolent course of the disease and the better condition of patients, but also by the GvL effect.

Optimal procedure

Modalities to reduce the toxicity of the conditioning regimen and the risk of GVHD and to separate the latter from the beneficial GvL effect are required.

Conditioning regimens.

The high response rate after allogeneic or autologous BMT is due at least in part to the intensive conditioning regimen. Several attempts have been made to improve the efficiency and decrease the toxicity of such regimens, but no randomized data are available to assess their respective value. In the IBMTR survey, a higher survival rate was associated with the use of a TBI-containing conditioning regimen. However, only 18% of these patients received non-TBI regimens, and it is possible that unknown factors may have skewed the results.155 An increase in the intensity of the conditioning regimen in 15 patients was associated with an increase in severe acute GVHD despite T-cell depletion of the graft, probably due to supplementary impairment of the recipient's immune status.156 

Source of allogeneic hematopoietic stem cells.

Apheresis products containing G-CSF–mobilized peripheral blood cells can be safely obtained from allogeneic donors. Seven recently reviewed167 randomized trials in various hematological neoplasias have demonstrated that neutrophil and platelet recovery and immune reconstitution were faster with peripheral blood cells than with marrow.168 The risk of acute GVHD is not increased, even though the number of T cells in the blood is 10-fold higher than that in the BM. Moreover, in 2 of these studies, the incidence of chronic GVHD was higher among patients who received peripheral blood cells. This procedure may offer advantages in terms of OS and DFS, especially in patients with high risk of relapse.

GVHD prophylaxis.

The incidence of acute and chronic GVHD following allogeneic BMT can be reduced by several modalities.

(1) T-cell depletion. T-cell depletion, performed in vitro by elutriation or monoclonal antibodies or in vivo by the addition of CAMPATH-1G, may decrease the high TRM rate (4.5% of 22 patients treated with alloBMT for relapsed indolent lymphoma at the DFCI).160,169 Conversely, it may increase graft failure (10%), delay engraftment, mixed chimerism, disease recurrence, and Epstein-Barr virus–associated lymphoproliferative disorder.10,156 Delayed engraftment secondary to T-cell depletion might be overcome by reinfusion of the fraction of small CD34+ cells that coseparate with the small lymphocytes.170 However, decreasing GVHD inhibited the GvL effect. Thus, after T-cell–depleted allogeneic BMT, 4 of 5 patients with clinical CR and informative PCR for the bcl2/IgH translocation remained PCR-positive after transplantation.171 

(2) Delayed administration of small, graded doses of donor lymphocytes172 and the identification and reinjection of specific lymphocyte subtypes.169 For example, CD6+ T-cell–depleted allogeneic BMT resulted in 54% and 59% DFS and OS, respectively, in poor-risk relapsing FL over a median follow-up time of 30 months.169 T-cell–depleted allogeneic BMT followed by donor lymphocyte infusion (DLI) appears to be effective in poor-risk patients with relapsing FL.156Preliminary results of a randomized trial suggest that CD8 depletion of DLI reduces the incidence of GVHD associated with DLI without compromising antitumor activity, conversion to donor hematopoiesis, or immunologic recovery.173 

(3) Transduction of the donor lymphocytes with a suicide gene, such as the herpesvirus thymidine kinase, that confers sensitivity to ganciclovir and allows the effector cells to be eradicated if GVHD occurs.174 

(4) Other immunosuppressive agents such as mycophenolate mofetil, FK506, CAMPATH-1, or monoclonal IL-2 receptor are currently being evaluated.

Nonmyeloablative preparative regimen.

An alternative strategy to reduce TRM and improve the age cutoff is to use a lower dose, nonmyeloablative, preparative regimen designed not to eradicate the malignancy but to provide sufficient immunosuppression to achieve engraftment of an allogeneic graft, thus allowing the development of an immune GvL effect. Results from more than 600 patients with various hematological malignancies have been reported, including 18% with NHL.175-186 

Nonmyeloablative preparative regimens contain fludarabine and 200 cGy TBI,175 cyclophosphamide,161 busulfan with181,182 or without176,185anti–T-lymphocyte globulin, melphalan with184 or without176,183,186 CAMPATH-1H or carmustine.177 Thiotepa-based regimens with fludarabine,180 cyclophosphamide,179 or both178 are also investigated. GVHD prophylaxis consisted in cyclosporine A and methotrexate,176-179tacrolimus,161,187 or mycophenolate mofetil and175,185 or T-cell depletion.186 If residual malignant cells or mixed chimerism are detected, donor lymphocytes are infused to potentiate the GvL effect. Graft rejection occurred in < 5%, whereas the frequency of mixed chimerism ranged from low175 to 40%184 of patients. TRM occurred in between 10% and 40% of cases. Median granulocyte and platelet nadirs are high, 0.3 and 60 G/L, respectively, in one study,175and platelet and red blood cell transfusions are significantly decreased.188 Outpatient allografting has even been performed without subsequent hospitalization.189 However, an increased incidence of early viral infection, especially cytomegalovirus (CMV),190 and late gram-negative infection was reported in nonneutropenic patients.191 GVHD remains the major drawback to this procedure and the main cause of death before day 100. The risk for grade III-IV GVHD in those patients whose median age was older than 50 years remains high (10% to 42%). The addition of in vivo CAMPATH-1H to melphalan and fludarabine significantly reduces TRM and the risk of GVHD with marrow transplants from both related and unrelated donors.184Nonmyeloablative allogenic transplantations are also performed with unrelated donors185,192,193 and in patients who relapsed after autologous BMT.177,178,192,194 195 

FL should be a good indication for these nonmyeloablative regimens, as its long natural history gives time to exploit the potentially beneficial GvL effect. So far, few results of nonmyeloablative allogeneic BMT are available in FL161,187,195-200 (Table6). Khouri et al161 first reported on 6 cases of heavily pretreated FL and 9 similar cases of CLL. Eleven of the 15 patients had durable engraftment with 50% to 100% donor cells one month following transplantation, converting to 100% over the next 2 months either spontaneously or after DLI. Hematopoietic recovery was prompt, and 8 of the 11 patients with engraftment achieved CR.161 Maximal responses were slow to develop and gradually occurred over a period of several months to one year. Subsequently, nonmyeloablative allogeneic BMT has been reported in others cases of heavily pretreated, high-risk lymphoma. Evidence for GvL activity have been observed as residual posttransplantation active disease declined with the onset of chronic GVHD161,187,197,198 or after DLI infusion.187,196,197 High doses of rituximab (375 mg/m2 on day −6 and 1000 mg/m2 on days 1, 8, and 15 after transplantation) have been administered to enhance tumor control early after transplantation to allow time for the GvL effect to occur.187 This improves the antitumoral activity of the fludarabine-cyclophosphamide conditioning, as 8 of 8 patients in partial response before transplantation achieved CR, 2 of them concomitant with the withdrawal of tacrolimus and subsequent development of GVHD. At a median follow-up of 21 months, 84% of patients are still in CR despite advance recurrent disease. The low TRM allows to perform nonmyeloablative allogenic PSCT after ABMT conditioned by BEAM therapy.196 The high response rate (9 patients in partial remission after autologous grafting achieved CR after mini-allografting) in this group of heavily pretreated patients is promising. However, acute and chronic GVHD continue to be a problem,195-200 and controlled prospective trials are warranted to assess if nonmyeloablative alloBMT can indeed improve outcomes of patients with FL.

So far, given the encouraging results of allogeneic BMT in patients with advanced and refractory disease, it would be reasonable to consider allogeneic BMT in patients younger than 55 years with disease either refractory or relapsing early after conventional chemotherapy. Nonmyeloablative allogeneic transplantation might be beneficial to patients with FL who would otherwise have been excluded because of age, pre-existing organ dysfunction, or previous treatment.

There is not yet a clear demonstration that HDT followed by ASCT improves the OS of patients when used as first-line treatment. However, the results of several trials will soon allow determination of its role. For patients with chemoresistant disease, allogeneic SCT must be considered if patients have an HLA-identical sibling. Conventional allogeneic SCT is probably the best procedure for patients younger than 50 years. For older patients, nonmyeloablative treatment probably yields a similar cure rate with a lower TRM. For patients who relapse with disseminated disease after a complete or partial response, salvage chemotherapy followed by autologous SCT improves response rate, survival from second relapse, and probably overall survival compared with conventional therapy. Although there is no clear demonstration that stem cell harvest purging decreases the relapse rate and improves survival, in vivo treatment by anti–B monoclonal antibodies such as rituximab are an attractive option, because they have both an antilymphoma and a purging effect on blood and BM. These results must be confirmed by ongoing clinical trials. Despite these improvements, many patients still relapse, and additional therapies must be tested. Posttransplantation immunotherapy is a promising approach.

1
Horning
SJ
Follicular lymphoma: have we made any progress?
Ann Oncol.
11
2000
23
27
2
Johnson
PW
Rohatiner
AZ
Whelan
JS
et al
Patterns of survival in patients with recurrent follicular lymphoma: a 20-year study from a single center.
J Clin Oncol.
13
1995
140
147
3
Berglund
A
Enblad
G
Carlson
K
Glimelius
B
Hagberg
H
Long-term follow-up of autologous stem-cell transplantation for follicular and transformed follicular lymphoma.
Eur J Haematol.
65
2000
17
22
4
Voso
MT
Martin
S
Hohaus
S
et al
Prognostic factors for the clinical outcome of patients with follicular lymphoma following high-dose therapy and peripheral blood stem cell transplantation (PBSCT).
Bone Marrow Transplant.
25
2000
957
964
5
Apostolidis
J
Gupta
RK
Grenzelias
D
et al
High-dose therapy with autologous bone marrow support as consolidation of remission in follicular lymphoma: long-term clinical and molecular follow-up.
J Clin Oncol.
18
2000
527
536
6
Freedman
AS
Neuberg
D
Mauch
P
et al
Long-term follow-up of autologous bone marrow transplantation in patients with relapsed follicular lymphoma.
Blood.
94
1999
3325
3333
7
Weaver
CH
Schwartzberg
L
Rhinehart
S
et al
High-dose chemotherapy with BUCY or BEAC and unpurged peripheral blood stem cell infusion in patients with low-grade non-Hodgkin's lymphoma.
Bone Marrow Transplant.
21
1998
383
389
8
Brice
P
Simon
D
Bouabdallah
R
et al
High-dose therapy with autologous stem-cell transplantation (ASCT) after first progression prolonged survival of follicular lymphoma patients included in the prospective GELF 86 protocol.
Ann Oncol.
11
2000
1585
1590
9
Verdonck
LF
Allogeneic versus autologous bone marrow transplantation for refractory and recurrent low-grade non-Hodgkin's lymphoma: updated results of the Utrecht experience.
Leuk Lymphoma.
34
1999
129
136
10
Verdonck
L
Dekker
A
Lokhorst
H
Petersen
E
Nieuwenhuis
H
Allogeneic versus autologous bone marrow transplantation for refractory and recurrent low-grade non-Hodgkin's lymphoma.
Blood.
90
1997
4201
4205
11
Bastion
Y
Brice
P
Haioun
C
et al
Intensive therapy with peripheral blood progenitor cell transplantation in 60 patients with poor-prognosis follicular lymphoma.
Blood.
86
1995
3257
3262
12
Colombat
P
Donadio
D
Fouillard
L
et al
Value of autologous bone marrow transplantation in follicular lymphoma: a France Autogreffe retrospective study of 42 patients.
Bone Marrow Transplant.
13
1994
157
162
13
Bierman
PJ
Vose
JM
Anderson
JR
Bishop
MR
Kessinger
A
Armitage
JO
High-dose therapy with autologous hematopoietic rescue for follicular low-grade non-Hodgkin's lymphoma.
J Clin Oncol.
15
1997
445
450
14
Molina
A
Nademanee
A
O'Donnell
M
et al
Long-term follow-up and analysis of prognostic factors after high-dose therapy and peripheral blood stem cell autografting in 58 patients with a history of low grade follicular lymphoma [abstract].
Blood.
94
1999
171a
15
Cao
TM
Horning
S
Negrin
RS
et al
High-dose therapy and autologous hematopoietic-cell transplantation for follicular lymphoma beyond first remission: the Stanford University experience.
Biol Blood Marrow Transplant.
7
2001
294
301
16
Seyfarth
B
Kuse
R
Sonnen
R
Glass
B
Schmitz
N
Dreger
P
Autologous stem cell transplantation for follicular lymphoma: no benefit for early transplant?
Ann Hematol.
80
2001
398
405
17
Lopez
R
Martino
R
Sureda
A
et al
Autologous stem cell transplantation in advanced follicular lymphoma: a single center experience.
Haematologica.
84
1999
350
355
18
Schouten
H
Kvaloy
S
Sydes
M
Qian
W
Fayers
P
The CUP trial: a randomized study analysing the efficacy of high dose therapy and purging in low-grade non-Hodgkin's lymphoma.
Ann Oncol.
11(suppl 1)
2000
S91
S94
19
Fouillard
L
Laporte
JP
Labopin
M
et al
Autologous stem-cell transplantation for non-Hodgkin's lymphomas: the role of graft purging and radiotherapy posttransplantation—results of a retrospective analysis on 120 patients autografted in a single institution.
J Clin Oncol.
16
1998
2803
2816
20
Cervantes
F
Shu
XO
McGlave
PB
et al
Autologous bone marrow transplantation for non-transformed low-grade non-Hodgkin's lymphoma.
Bone Marrow Transplant.
16
1995
387
392
21
Rohatiner
A
Johnson
P
Price
C
et al
Myeloablative therapy with autologous bone marrow transplantation as consolidation therapy for recurrent follicular lymphoma.
J Clin Oncol.
12
1994
1177
1184
22
Schouten
HC
Bierman
PJ
Vaughan
WP
et al
Autologous bone marrow transplantation in follicular non-Hodgkin's lymphoma before and after histologic transformation.
Blood.
74
1989
2579
2584
23
Friedberg
JW
Neuberg
D
Gribben
JG
et al
Autologous bone marrow transplantation after histologic transformation of indolent B cell malignancies.
Biol Blood Marrow Transplant.
5
1999
262
268
24
Foran
JM
Apostolidis
J
Papamichael
D
et al
High-dose therapy with autologous haematopoietic support in patients with transformed follicular lymphoma: a study of 27 patients from a single centre.
Ann Oncol.
9
1998
865
869
25
Williams
CD
Harrison
CN
Lister
TA
et al
High-dose therapy and autologous stem-cell support for chemosensitive transformed low-grade follicular non-Hodgkin's lymphoma: a case-matched study from the European Bone Marrow Transplant Registry.
J Clin Oncol.
19
2001
727
735
26
Belanger
C
Simon
D
Brice
P
et al
Incidence of histological transformation at first relapse in follicular lymphoma patients included in the prospective GELF 86 protocol [abstract].
Ann Oncol.
10(suppl 3)
1999
216a
27
Chen
CI
Crump
M
Tsang
R
Stewart
AK
Keating
A
Autotransplants for histologically transformed follicular non-Hodgkin's lymphoma.
Br J Haematol.
113
2001
202
208
28
Yuen
AR
Kamel
OW
Halpern
J
Horning
SJ
Long-term survival after histologic transformation of low-grade follicular lymphoma.
J Clin Oncol.
13
1995
1726
1733
29
Moos
M
Schulz
R
Martin
S
Benner
A
Haas
R
The remission status before and the PCR status after high-dose therapy with peripheral blood stem cell support are prognostic factors for relapse-free survival in patients with follicular non-Hodgkin's lymphoma.
Leukemia.
12
1998
1971
1976
30
Gribben
JG
Neuberg
D
Freedman
AS
et al
Detection by polymerase chain reaction of residual cells with the bcl-2 translocation is associated with increased risk of relapse after autologous bone marrow transplantation for B-cell lymphoma.
Blood.
81
1993
3449
3457
31
Corradini
P
Ladetto
M
Astolfi
M
et al
Long-term molecular follow-up in indolent lymphoma patients treated with high-dose sequential chemotherapy and autografting: durable molecular and clinical remission can be achieved only in follicular subtypes [abstract].
Blood.
98
2001
681a
32
Johnson
P
Price
C
Smith
T
et al
Detection of cells bearing the t(14;18) translocation following myeloablative treatment and autologous bone marrow transplantation for follicular lymphoma.
J Clin Oncol.
12
1994
798
805
33
Heid
CA
Stevens
J
Livak
KJ
Williams
PM
Real time quantitative PCR.
Genome Res.
6
1996
986
994
34
Hirt
C
Dolken
G
Quantitative detection of t(14;18)-positive cells in patients with follicular lymphoma before and after autologous bone marrow transplantation.
Bone Marrow Transplant.
25
2000
419
426
35
Zwicky
C
Maddocks
A
Andersen
N
Gribben
J
Eradication of polymerase chain reaction detectable immunoglobulin gene rearrangement in non-Hodgkin's lymphoma is associated with decreased relapse after autologous bone marrow transplantation.
Blood.
88
1996
3314
3322
36
Salles
G
Coiffier
B
Autologous peripheral blood stem cell transplantation for non-Hodgkin's lymphoma.
Baillieres Best Pract Res Clin Haematol.
12
1999
151
169
37
Tarella
C
Caracciolo
D
Corradini
P
et al
Long-term follow-up of advanced-stage low-grade lymphoma patients treated up front with high-dose sequential chemotherapy and autograft.
Leukemia.
14
2000
740
747
38
Morel
P
Laporte
JP
Noel
MP
et al
Autologous bone marrow transplantation as consolidation therapy may prolong remission in newly diagnosed high-risk follicular lymphoma: a pilot study of 34 cases.
Leukemia.
9
1995
576
582
39
Horning
SJ
Negrin
RS
Hoppe
RT
et al
High-dose therapy and autologous bone marrow transplantation for follicular lymphoma in first complete or partial remission: results of a phase II clinical trial.
Blood.
97
2001
404
409
40
Freedman
AS
Gribben
JG
Neuberg
D
et al
High-dose therapy and autologous bone marrow transplantation in patients with follicular lymphoma during first remission.
Blood.
88
1996
2780
2786
41
Bociek
R
Bierman
P
Vose
J
Lynch
J
Morris
M
Armitage
J
High dose therapy with autologous hematopoietic stem cell transplantation for patients with low-grade follicular non-Hodgkin's lymphoma in first complete or partial remission [abstract].
Blood.
94
1999
170a
42
Colombat
P
Cornillet
P
Deconinck
E
et al
Value of autologous stem cell transplantation with purged bone marrow as first-line therapy for follicular lymphoma with high tumor burden: a GOELAMS phase II study.
Bone Marrow Transplant.
26
2000
971
977
43
Gonzalez-Barca
E
Fernandez de Sevilla
A
Domingo-Claros
A
et al
Autologous stem cell transplantation (ASCT) with immunologically purged progenitor cells in patients with advanced stage follicular lymphoma after early partial or complete remission: toxicity, follow-up of minimal residual disease and survival.
Bone Marrow Transplant.
26
2000
1051
1056
44
Ladetto
M
Corradini
P
Sametti
S
et al
The GITMO experience with high-dose chemotherapy and autografting in advanced follicle center lymphoma: a multicenter trial showing good feasibility and frequent achievement of clinical and molecular remission [abstract].
Blood.
98
2001
678a
45
Sanz-Rodriguez
C
Sanchez
A
Rodriguez
F
Gandarillas
M
Conde
E
Arranz
R
Chemotherapy plus interferon alpha-2b vs high-dose therapy for response consolidation in low grade non-Hodgkin's lymphoma: results of a retrospective case control study [abstract].
Blood.
96
2000
791a
46
Hiddemann
W
Unterhalt
M
Wandt
H
et al
Myeloablative radiochemotherapy followed by blood stem cell transplantation significantly prolongs the disease-free interval in patients with low-grade lymphomas as compared to standard maintenance with interferon alpha: results of a prospective randomized comparison by the German Low-Grade Lymphoma Study Group [abstract].
Blood.
96
1999
610a
47
Colombat
P
Foussard
C
Bertrand
P
et al
Value of autologous stem cell transplantation in first line therapy of follicular lymphoma with high tumor burden: first results of the randomized GOELAMS 064 trial [abstract].
Blood.
98
2001
861a
48
Romaguera
JE
Can we identify patients with low grade lymphoma for frontline ABMT?
Leuk Lymphoma.
10
1993
9
15
49
Liberti
G
Pearce
R
Taghipour
G
Majolino
I
Goldstone
A
Comparison of peripheral blood stem-cell and autologous bone marrow transplantation for lymphoma patients: a case-controlled analysis of the EBMT Registry data: Lymphoma Working Party of the EBMT.
Ann Oncol.
5(suppl 2)
1994
151
153
50
Majolino
I
Pearce
R
Taghipour
G
Goldstone
AH
Peripheral-blood stem-cell transplantation versus autologous bone marrow transplantation in Hodgkin's and non-Hodgkin's lymphomas: a new matched-pair analysis of the European Group for Blood and Marrow Transplantation Registry Data: Lymphoma Working Party of the European Group for Blood and Marrow Transplantation.
J Clin Oncol.
15
1997
509
517
51
Schmitz
N
Linch
DC
Dreger
P
et al
Randomised trial of filgrastim-mobilised peripheral blood progenitor cell transplantation versus autologous bone-marrow transplantation in lymphoma patients.
Lancet.
347
1996
353
357
52
Beyer
J
Schwella
N
Zingsem
J
et al
Hematopoietic rescue after high-dose chemotherapy using autologous peripheral-blood progenitor cells or bone marrow: a randomized comparison.
J Clin Oncol.
13
1995
1328
1335
53
Hartmann
O
Le Corroller
AG
Blaise
D
et al
Peripheral blood stem cell and bone marrow transplantation for solid tumors and lymphomas: hematologic recovery and costs: a randomized, controlled trial.
Ann Intern Med.
126
1997
600
607
54
Leonard
BM
Hetu
F
Busque
L
et al
Lymphoma cell burden in progenitor cell grafts measured by competitive polymerase chain reaction: less than one log difference between bone marrow and peripheral blood sources.
Blood.
91
1998
331
339
55
Kanteti
R
Miller
K
McCann
J
et al
Randomized trial of peripheral blood progenitor cell vs bone marrow as hematopoietic support for high-dose chemotherapy in patients with non-Hodgkin's lymphoma and Hodgkin's disease: a clinical and molecular analysis.
Bone Marrow Transplant.
24
1999
473
481
56
Talmadge
JE
Reed
E
Ino
K
et al
Rapid immunologic reconstitution following transplantation with mobilized peripheral blood stem cells as compared to bone marrow.
Bone Marrow Transplant.
19
1997
161
172
57
Pettengell
R
Morgenstern
GR
Woll
PJ
et al
Peripheral blood progenitor cell transplantation in lymphoma and leukemia using a single apheresis.
Blood.
82
1993
3770
3777
58
To
LB
Haylock
DN
Simmons
PJ
Juttner
CA
The biology and clinical uses of blood stem cells.
Blood.
89
1997
2233
2258
59
Weaver
CH
Birch
R
Greco
FA
et al
Mobilization and harvesting of peripheral blood stem cells: randomized evaluations of different doses of filgrastim.
Br J Haematol.
100
1998
338
347
60
Kroger
N
Zeller
W
Fehse
N
et al
Mobilizing peripheral blood stem cells with high-dose G-CSF alone is as effective as with Dexa-BEAM plus G-CSF in lymphoma patients.
Br J Haematol.
102
1998
1101
1106
61
Kessinger
A
Bierman
P
Cowles
M
et al
Mobilized versus non-mobilized peripheral stem cell transplantation after high dose therapy for low grade non-Hodgkin lymphoma.
Can Res Therapy Control.
5
1998
113
119
62
Facon
T
Harousseau
JL
Maloisel
F
et al
Stem cell factor in combination with filgrastim after chemotherapy improves peripheral blood progenitor cell yield and reduces apheresis requirements in multiple myeloma patients: a randomized, controlled trial.
Blood.
94
1999
1218
1225
63
Stiff
PJ
Management strategies for the hard-to-mobilize patient.
Bone Marrow Transplant.
23(suppl 2)
1999
S29
S33
64
Bashford
J
Durrant
S
Schwarer
A
et al
Ancestim (r-metHuSCF) plus filgrastim (r-metHuG-CSF) allows mobilization of peripheral blood stem cells for transplantation in patients failing mobilization with filgrastim alone [abstract].
Blood.
96
2000
178a
65
Azar
N
Vantelon
J
Hasmati
P
et al
Ancestim (r-metHuSCF): mobilization of peripheral blood stem cells (PBSC) in patients with prior unsuccessful mobilization with G-CSF: experience of 67 cases of compassionate use in France [abstract].
Blood.
96
2000
178a
66
Haas
R
Mohle
R
Fruhauf
S
et al
Patient characteristics associated with successful mobilizing and autografting of peripheral blood progenitor cells in malignant lymphoma.
Blood.
83
1994
3787
3794
67
Haas
R
Moos
M
Mohle
R
et al
High-dose therapy with peripheral blood progenitor cell transplantation in low-grade non-Hodgkin's lymphoma.
Bone Marrow Transplant.
17
1996
149
155
68
Perry
A
Watts
M
Peniket
A
Goldstone
A
Linch
D
Progenitor cell yields are frequently poor in patients with histologically indolent lymphomas especially when mobilized within 6 months of previous chemotherapy.
Bone Marrow Transplant.
12
1998
1201
1205
69
McQuaker
I
Haynes
A
Anderson
S
et al
Engraftment and molecular monitoring of CD34+ peripheral-blood stem-cell transplants for follicular lymphoma: a pilot study.
J Clin Oncol.
15
1997
2288
2295
70
Bensinger
W
Appelbaum
F
Rowley
S
et al
Factors that influence collection and engraftment of autologous peripheral-blood stem cells.
J Clin Oncol.
13
1995
2547
2555
71
Vantelon
JM
Koscielny
S
Brault
P
et al
Scoring system for the prediction of successful peripheral blood stem cell (PBSC) collection in non-Hodgkin's lymphoma (NHL): application in clinical practice.
Bone Marrow Transplant.
25
2000
495
499
72
Kotasek
D
Shepherd
KM
Sage
RE
et al
Factors affecting blood stem cell collections following high-dose cyclophosphamide mobilization in lymphoma, myeloma and solid tumors.
Bone Marrow Transplant.
9
1992
11
17
73
O'Donnell
P
Loper
K
Flinn
I
Vogelsang
G
Grever
M
Noga
S
Effect of fludarabine chemotherapy on peripheral blood stem cell transplantation [abstract].
Blood.
92
1998
120a
74
Laszlo
D
Galieni
P
Raspadori
D
et al
Fludarabine-containing regimens may adversely affect peripheral blood stem cell collection in low-grade non Hodgkin lymphoma patients.
Leuk Lymphoma.
37
2000
157
161
75
Micallef
I
Apostolidis
J
Rohatiner
A
et al
Factors which predict unsuccessful mobilization of peripheral blood progenitor cells following G-CSF alone in patients with non-Hodgkin's lymphoma.
Hematology J.
1
2000
367
373
76
Gribben
J
Freedman
A
Woo
S
et al
All advanced stage non-Hodgkin's lymphomas with a polymerase chain reaction amplifiable breakpoint of bcl-2 have residual cells containing the bcl-2 rearrangement at evaluation and after treatment.
Blood.
78
1991
3275
3280
77
Sharp
J
Kessinger
A
Mann
S
et al
Outcome of high-dose therapy and autologous transplantation in non-Hodgkin's lymphoma based on the presence of tumor in the marrow or infused hematopoietic harvest.
J Clin Oncol.
14
1996
214
219
78
Bachier
CR
Giles
RE
Ellerson
D
et al
Hematopoietic retroviral gene marking in patients with follicular non-Hodgkin's lymphoma.
Leuk Lymphoma.
32
1999
279
288
79
Brenner
M
Rill
D
Moen
R
et al
Gene-marking to trace origin of relapse after autologous bone-marrow transplantation.
Lancet.
341
1993
85
86
80
Gribben
J
Freedman
A
Neuberg
D
et al
Immunologic purging of marrow assessed by PCR before autologous bone marrow transplantation for B-cell lymphoma.
N Engl J Med.
325
1991
1525
1533
81
Martin-Henao
GA
Picon
M
Limon
A
et al
Immunomagnetic bone marrow (BM) and peripheral blood progenitor cell (PBPC) purging in follicular lymphoma (FL).
Bone Marrow Transplant.
23
1999
579
587
82
Gribben
J
Saporito
L
Barber
M
et al
Bone marrows of non-Hodgkin's lymphoma patients with a bcl-2 translocation can be purged of polymerase chain reaction-detectable lymphoma cells using monoclonal antibodies and immunomagnetic bead depletion.
Blood.
80
1992
1083
1089
83
Negrin
RS
Pesando
J
Detection of tumor cells in purged bone marrow and peripheral-blood mononuclear cells by polymerase chain reaction amplification of bcl-2 translocations.
J Clin Oncol.
12
1994
1021
1027
84
Pappa
VI
Wilkes
S
Salam
A
Young
BD
Lister
TA
Rohatiner
AZ
Use of the polymerase chain reaction and direct sequencing analysis to detect cells with the t(14;18) in autologous bone marrow from patients with follicular lymphoma, before and after in vitro treatment.
Bone Marrow Transplant.
22
1998
553
558
85
Corradini
P
Astolfi
M
Cherasco
C
et al
Molecular monitoring of minimal residual disease in follicular and mantle cell non-Hodgkin's lymphomas treated with high-dose chemotherapy and peripheral blood progenitor cell autografting.
Blood.
89
1997
724
731
86
Gorin
N
Lopez
M
Laporte
J
et al
Preparation and successful engraftment of purified CD34+ bone marrow progenitor cells in patients with non-Hodgkin's lymphoma.
Blood.
85
1995
1647
1654
87
Voso
M
Hohaus
S
Moos
M
et al
Autografting with CD34+ peripheral blood stem cells: retained engraftment capability and reduced tumour cell content.
Br J Haematol.
104
1999
382
391
88
Marin
G
Dal Cortivo
L
Cayuela
J
et al
Peripheral blood stem cell CD34+ autologous transplant in relapsed follicular lymphoma.
Hematol Cell Ther.
39
1997
33
40
89
Bomberger
C
Singh-Jairam
M
Rodey
G
et al
Lymphoid reconstitution after autologous PBSC transplantation with FACS-sorted CD34+ hematopoietic progenitors.
Blood.
91
1998
2588
2600
90
Divine
M
Boutolleau
D
Delfau-Larue
MH
et al
Poor lymphocyte recovery following CD34-selected autologous peripheral blood stem cell transplantation for non-Hodgkin's lymphoma.
Br J Haematol.
105
1999
349
360
91
Macintyre
E
Belanger
C
Debert
C
et al
Detection of clonal CD34+19+ progenitors in bone marrow of BCL2-IgH-positive follicular lymphoma patients.
Blood.
86
1995
4691
4698
92
Paulus
U
Schmitz
N
Viehmann
K
von Neuhoff
N
Dreger
P
Combined positive/negative selection for highly effective purging of PBPC grafts: towards clinical application in patients with B-CLL.
Bone Marrow Transplant.
20
1997
415
420
93
Yao
M
Fouillard
L
Lemoine
FM
et al
Ex vivo expansion of CD34-positive peripheral blood progenitor cells from patients with non-Hodgkin's lymphoma: no evidence of concomitant expansion of contaminating bcl2/JH-positive lymphoma cells.
Bone Marrow Transplant.
26
2000
497
503
94
Reiffers
J
Cailliot
C
Dazey
B
Attal
M
Caraux
J
Boiron
JM
Abrogation of post-myeloablative chemotherapy neutropenia by ex vivo expanded autologous CD34-positive cells.
Lancet.
354
1999
1092
1093
95
McNiece
I
Briddell
R
Ex vivo expansion of hematopoietic progenitor cells and mature cells.
Exp Hematol.
29
2001
3
11
96
Widmer
L
Pichert
G
Jost
L
Stahel
R
Fate of contaminating t(14; 18)+ lymphoma cells during ex vivo expansion of CD34-selected hematopoietic progenitor cells.
Blood.
88
1996
3166
3175
97
Magni
M
Di Nicola
M
Devizzi
L
et al
Successful in vivo purging of CD34-containing peripheral blood harvests in mantle cell and indolent lymphoma: evidence for a role of both chemotherapy and rituximab infusion.
Blood.
96
2000
864
869
98
Voso
MT
Pantel
G
Weis
M
et al
In vivo depletion of B cells using a combination of high-dose cytosine arabinoside/mitoxantrone and rituximab for autografting in patients with non-Hodgkin's lymphoma.
Br J Haematol.
109
2000
729
735
99
Haioun
C
Delfau-Larue
M
Beaujean
F
et al
Efficiency of in vivo purging with rituximab followed by high-dose therapy (HDT) with autologous peripheral blood stem cell transplantation in B-cell non-Hodgkin's lymphoma: a single institution study [abstract].
Blood.
96
2000
184a
100
Buckstein
R
Imrie
K
Spaner
D
et al
Autologous stem cell transplants combined with rituximab for relapsed follicular lymphoma achieve prolonged clinical and molecular remissions [abstract].
Blood.
98
2001
680a
101
Buckstein
R
Imrie
K
Spaner
D
et al
Stem cell function and engraftment is not affected by in vivo purging with rituximab for autologous stem cell treatment for patients with low-grade non-Hodgkin's lymphoma.
Semin Oncol.
26
1999
115
122
102
Flinn
IW
O'Donnell
PV
Goodrich
A
et al
Immunotherapy with rituximab during peripheral blood stem cell transplantation for non-Hodgkin's lymphoma.
Biol Blood Marrow Transplant.
6
2000
628
632
103
Blystad
A
Kvaloy
S
Delabie
J
et al
High-dose therapy in high-grade non-Hodgkin's lymphoma patients: comparison between unmanipulated peripheral blood progenitor cells and purged autologous bone marrow as stem cell support [abstract].
Blood.
96
2000
183a
104
Vose
JM
Colcher
D
Gobar
L
et al
Phase I/II trial of multiple dose 131Iodine-MAb LL2 (CD22) in patients with recurrent non-Hodgkin's lymphoma.
Leuk Lymphoma.
38
2000
91
101
105
Wheeler
C
Eickhoff
C
Elias
A
et al
High-dose cyclophosphamide, carmustine, and etoposide with autologous transplantation in Hodgkin's disease: a prognostic model for treatment outcomes.
Biol Blood Marrow Transplant.
3
1997
98
106
106
Johnson
RE
Canellos
GP
Young
RC
Chabner
BA
DeVita
VT
Chemotherapy (cyclophosphamide, vincristine, and prednisone) versus radiotherapy (total body irradiation) for stage III-IV poorly differentiated lymphocytic lymphoma.
Cancer Treat Rep.
62
1978
321
325
107
Girinsky
T
Benhamou
E
Bourhis
JH
et al
Prospective randomized comparison of single-dose versus hyperfractionated total-body irradiation in patients with hematologic malignancies.
J Clin Oncol.
18
2000
981
986
108
Stein
RS
Greer
JP
Goodman
S
et al
Intensified preparative regimens and autologous transplantation in refractory or relapsed intermediate grade non-Hodgkin's lymphoma.
Bone Marrow Transplant.
25
2000
257
262
109
Williams
C
Goldstone
A
Pearce
R
et al
Purging of bone marrow in autologous bone marrow transplantation for non-Hodgkin's lymphoma: a case-matched comparison with unpurged cases by the European Blood and Marrow Transplant Lymphoma Registry.
J Clin Oncol.
14
1996
2454
2564
110
Stockerl-Goldstein
KE
Horning
SJ
Negrin
RS
et al
Influence of preparatory regimen and source of hematopoietic cells on outcome of autotransplantation for non-Hodgkin's lymphoma.
Biol Blood Marrow Transplant.
2
1996
76
85
111
Press
OW
Eary
JF
Gooley
T
et al
A phase I/II trial of iodine-131-tositumomab (anti-CD20), etoposide, cyclophosphamide, and autologous stem cell transplantation for relapsed B-cell lymphomas.
Blood.
96
2000
2934
2942
112
Behr
TM
Wormann
B
Gramatzki
M
et al
Low- versus high-dose radioimmunotherapy with humanized anti-CD22 or chimeric anti-CD20 antibodies in a broad spectrum of B cell-associated malignancies.
Clin Cancer Res.
5
1999
3304s
3314s
113
Hardingham
J
Kotasek
D
Sage
R
et al
Significance of molecular marker-positive cells after autologous peripheral-blood stem-cell transplantation for non-Hodgkin's lymphoma.
J Clin Oncol.
13
1995
1073
1079
114
Guillaume
T
Rubinstein
DB
Symann
M
Immune reconstitution and immunotherapy after autologous hematopoietic stem cell transplantation.
Blood.
92
1998
1471
1490
115
Guillaume
T
Rubinstein
DB
Symann
M
Immunological recovery and tumour-specific immunotherapeutic approaches to post-autologous haematopoietc stem cell transplantation.
Baillières Clin Haematol.
12
1999
293
306
116
Grossbard
ML
Multani
PS
Freedman
AS
et al
A Phase II study of adjuvant therapy with anti–B4-blocked ricin after autologous bone marrow transplantation for patients with relapsed B-cell non-Hodgkin's lymphoma.
Clin Cancer Res.
5
1999
2392
2398
117
Horwitz
S
Breslin
S
Negrin
R
et al
Adjuvant rituximab after autologous peripheral blood stem cell transplant results in delayed immune reconstitution without increase in infectious complications [abstract].
Blood.
96
2000
384a
118
Slavin
S
Nagler
A
Immunotherapy in conjunction with autologous and allogeneic blood or marrow transplantation in lymphoma.
Ann Oncol.
9
1998
S31
S39
119
Slavin
S
Nagler
A
Cytokine-mediated immunotherapy following autologous bone marrow transplantation in lymphoma and evidence of interleukin-2-induced immunomodulation in allogeneic transplants.
Cancer J Sci Am.
3(suppl 1)
1997
S59
S67
120
Neubauer
MA
Benyunes
MC
Thompson
JA
et al
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
316
121
Vey
N
Blaise
D
Tiberghien
P
et al
A pilot study of autologous bone marrow transplantation followed by recombinant interleukin-2 in malignant lymphomas.
Leuk Lymphoma.
21
1996
107
114
122
Klingemann
HG
Grigg
AP
Wilkie-Boyd
K
et al
Treatment with recombinant interferon (alpha-2b) early after bone marrow transplantation in patients at high risk for relapse.
Blood.
78
1991
3306
3311
123
Fierro
MT
Liao
XS
Lusso
P
et al
In vitro and in vivo susceptibility of human leukemic cells to lymphokine activated killer activity.
Leukemia.
2
1988
50
54
124
Nagler
A
Ackerstein
A
Or
R
Naparstek
E
Slavin
S
Immunotherapy with recombinant human interleukin-2 and recombinant interferon-alpha in lymphoma patients postautologous marrow or stem cell transplantation.
Blood.
89
1997
3951
3959
125
Baron
F
Gothot
A
Salmon
JP
et al
Clinical course and predictive factors for cyclosporin-induced autologous graft-versus-host disease after autologous haematopoietic stem cell transplantation.
Br J Haematol.
111
2000
745
753
126
Hess
A
Bright
E
Thoburn
C
Vogelsang
G
Jones
R
Kennedy
M
Specificity of effector T lymphocytes in autologous graft-versus-host disease: role of the major histocompatibility complex class II invariant chain peptide.
Blood.
89
1997
2203
2209
127
Gryn
J
Johnson
E
Goldman
N
et al
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
226
128
Kwak
L
Campbell
M
Czerwinski
D
Hart
S
Miller
R
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
1215
129
Davis
T
Maloney
D
Czerwinski
D
Liles
T
Levy
R
Anti-idiotype antibodies can induce long-term complete remissions in non-Hodgkin's lymphoma without eradicating the malignant clone.
Blood.
92
1998
1184
1190
130
Hsu
FJ
Caspar
CB
Czerwinski
D
et al
Tumor-specific idiotype vaccines in the treatment of patients with B-cell lymphoma—long-term results of a clinical trial.
Blood.
89
1997
3129
3135
131
Bendandi
M
Gocke
CD
Kobrin
CB
et al
Complete molecular remissions induced by patient-specific vaccination plus granulocyte-monocyte colony-stimulating factor against lymphoma.
Nat Med.
5
1999
1171
1177
132
King
CA
Spellerberg
MB
Zhu
D
et al
DNA vaccines with single-chain Fv fused to fragment C of tetanus toxin induce protective immunity against lymphoma and myeloma.
Nat Med.
4
1998
1281
1286
133
Hart
DN
Hill
GR
Dendritic cell immunotherapy for cancer: application to low-grade lymphoma and multiple myeloma.
Immunol Cell Biol.
77
1999
451
459
134
Hsu
FJ
Benike
C
Fagnoni
F
et al
Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells.
Nat Med.
2
1996
52
58
135
Bendandi
M
Anti-idiotype vaccines for human follicular lymphoma.
Leukemia.
14
2000
1333
1339
136
Davis
TA
Hsu
FJ
Caspar
CB
et al
Idiotype vaccination following ABMT can stimulate specific anti-idiotype immune responses in patients with B-cell lymphoma.
Biol Blood Marrow Transplant.
7
2001
517
522
137
Borrello
I
Sotomayor
EM
Rattis
FM
Cooke
SK
Gu
L
Levitsky
HI
Sustaining the graft-versus-tumor effect through posttransplant immunization with granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing tumor vaccines.
Blood.
95
2000
3011
3019
138
Apostolidis
J
Foran
JM
Johnson
PW
et al
Patterns of outcome following recurrence after myeloablative therapy with autologous bone marrow transplantation for follicular lymphoma.
J Clin Oncol.
17
1999
216
221
139
McLaughlin
P
Grillo-Lopez
A
Link
B
et al
Rituximab chimeric anti-CD20 monoclonal antibody therapy for relapsed indolent lymphoma: half of patients respond to a four-dose treatment program.
J Clin Oncol.
16
1998
2825
2833
140
Kaminski
MS
Estes
J
Zasadny
KR
et al
Radioimmunotherapy with iodine (131)I tositumomab for relapsed or refractory B-cell non-Hodgkin lymphoma: updated results and long-term follow-up of the University of Michigan experience.
Blood.
96
2000
1259
1266
141
Pedersen-Bjergaard
J
Andersen
MK
Christiansen
DH
Therapy-related acute myeloid leukemia and myelodysplasia after high-dose chemotherapy and autologous stem cell transplantation.
Blood.
95
2000
3273
3279
142
Stone
RM
Neuberg
D
Soiffer
R
et al
Myelodysplastic syndrome as a late complication following autologous bone marrow transplantation for non-Hodgkin's lymphoma.
J Clin Oncol.
12
1994
2535
2542
143
Bhatia
S
Ramsay
NK
Steinbuch
M
et al
Malignant neoplasms following bone marrow transplantation.
Blood.
87
1996
3633
3639
144
Miller
JS
Arthur
DC
Litz
CE
Neglia
JP
Miller
WJ
Weisdorf
DJ
Myelodysplastic syndrome after autologous bone marrow transplantation: an additional late complication of curative cancer therapy.
Blood.
83
1994
3780
3786
145
Darrington
DL
Vose
JM
Anderson
JR
et al
Incidence and characterization of secondary myelodysplastic syndrome and acute myelogenous leukemia following high-dose chemoradiotherapy and autologous stem-cell transplantation for lymphoid malignancies.
J Clin Oncol.
12
1994
2527
2534
146
Anderson
JR
Vose
J
Kessinger
A
Myelodysplastic syndrome after autologous transplant for lymphoma.
Blood.
84
1994
3988
3989
147
Milligan
DW
Ruiz De Elvira
MC
Kolb
HJ
et al
Secondary leukaemia and myelodysplasia after autografting for lymphoma: results from the EBMT: EBMT Lymphoma and Late Effects Working Parties, European Group for Blood and Marrow Transplantation.
Br J Haematol.
106
1999
1020
1026
148
Friedberg
JW
Neuberg
D
Stone
RM
et al
Outcome in patients with myelodysplastic syndrome after autologous bone marrow transplantation for non-Hodgkin's lymphoma.
J Clin Oncol.
17
1999
3128
3135
149
Traweek
ST
Slovak
ML
Nademanee
AP
Brynes
RK
Niland
JC
Forman
SJ
Clonal karyotypic hematopoietic cell abnormalities occurring after autologous bone marrow transplantation for Hodgkin's disease and non-Hodgkin's lymphoma.
Blood.
84
1994
957
963
150
Micallef
IN
Lillington
DM
Apostolidis
J
et al
Therapy-related myelodysplasia and secondary acute myelogenous leukemia after high-dose therapy with autologous hematopoietic progenitor-cell support for lymphoid malignancies.
J Clin Oncol.
18
2000
947
955
151
Krishnan
A
Bhatia
S
Slovak
ML
et al
Predictors of therapy-related leukemia and myelodysplasia following autologous transplantation for lymphoma: an assessment of risk factors.
Blood.
95
2000
1588
1593
152
Lillington
DM
Micallef
IN
Carpenter
E
et al
Detection of chromosome abnormalities pre-high-dose treatment in patients developing therapy-related myelodysplasia and secondary acute myelogenous leukemia after treatment for non-Hodgkin's lymphoma.
J Clin Oncol.
19
2001
2472
2481
153
Horning
S
Cherry
A
Bangs
D
Negrin
R
Blume
K
Serial assessment of marrow cytogenetics after autologous transplantation of follicular lymphoma [abstract].
Blood.
96
2000
406a
154
van Besien
K
Khouri
I
Giralt
S
et al
Allogeneic bone marrow transplantation for refractory and recurrent low-grade lymphoma: the case for aggressive management.
J Clin Oncol.
13
1995
1096
1102
155
van Besien
K
Sobocinski
K
Rowlings
P
et al
Allogeneic bone marrow transplantation for low-grade lymphoma.
Blood.
92
1998
1832
1836
156
Mandigers
C
Raemaekers
J
Schattenberg
A
et al
Allogeneic bone marrow transplantation with T-cell-depleted marrow grafts for patients with poor-risk relapsed low-grade non-Hodgkin's lymphoma.
Br J Haematol.
100
1998
198
206
157
Stein
R
Greer
J
Goodman
S
Kallianpur
A
Ahmed
M
Wolff
S
High-dose therapy with autologous or allogeneic transplantation as salvage therapy for small cleaved cell lymphoma of follicular center cell origin.
Bone Marrow Transplant.
23
1999
227
233
158
Toze
CL
Shepherd
JD
Connors
JM
et al
Allogeneic bone marrow transplantation for low-grade lymphoma and chronic lymphocytic leukemia.
Bone Marrow Transplant.
25
2000
605
612
159
Forrest
D
Matheson
K
Couban
S
Nevill
T
Fernandez
L
High-dose therapy and allogeneic hematopoietic stem cell transplantation for progressive follicular lymphoma [abstract].
Blood.
98
2001
408a
160
Cull
GM
Haynes
AP
Byrne
JL
et al
Preliminary experience of allogeneic stem cell transplantation for lymphoproliferative disorders using BEAM-CAMPATH conditioning: an effective regimen with low procedure-related toxicity.
Br J Haematol.
108
2000
754
760
161
Khouri
I
Keating
M
Korbling
M
et al
Transplant-lite: induction of graft-versus-malignancy using fludarabine-based nonablative chemotherapy and allogeneic blood progenitor-cell transplantation as treatment for lymphoid malignancies.
J Clin Oncol.
16
1998
2817
2824
162
Berdeja
JG
Jones
RJ
Zahurak
ML
et al
Allogeneic bone marrow transplantation in patients with sensitive low-grade lymphoma or mantle cell lymphoma.
Biol Blood Marrow Transplant.
7
2001
561
567
163
Mitterbauer
M
Neumeister
P
Kalhs
P
et al
Long-term clinical and molecular remission after allogeneic stem cell transplantation (SCT) in patients with poor prognosis non-Hodgkin's lymphoma.
Leukemia.
15
2001
635
641
164
Chopra
R
Goldstone
AH
Pearce
R
et al
Autologous versus allogeneic bone marrow transplantation for non-Hodgkin's lymphoma: a case-controlled analysis of the European Bone Marrow Transplant Group Registry data.
J Clin Oncol.
10
1992
1690
1695
165
van Besien
K
de Lima
M
Giralt
S
et al
Management of lymphoma recurrence after allogeneic transplantation: the relevance of graft-versus-lymphoma effect.
Bone Marrow Transplant.
19
1997
977
982
166
Mandigers
C
Meijerink
J
Raemaekers
J
Schattenberg
A
Mensink
E
Graft-versus-lymphoma effect of donor leucocyte infusion shown by real-time quantitative PCR analysis of t(14;18).
Lancet.
352
1998
1522
1523
167
Korbling
M
Anderlini
P
Peripheral blood stem cell versus bone marrow allotransplantation: does the source of hematopoietic stem cells matter?
Blood.
98
2001
2900
2908
168
Sharp
JG
Kessinger
A
Lynch
JC
Pavletic
ZS
Joshi
SS
Blood stem cell transplantation: factors influencing cellular immunological reconstitution.
J Hematother Stem Cell Res.
9
2000
971
981
169
Soiffer
RJ
Freedman
AS
Neuberg
D
et al
CD6+ T cell-depleted allogeneic bone marrow transplantation for non-Hodgkin's lymphoma.
Bone Marrow Transplant.
21
1998
1177
1181
170
Noga
S
Seber
A
Davis
J
et al
CD34 augmentation improves allogeneic T cell-depleted bone marrow engraftment.
J Hematother.
7
1998
151
157
171
Juckett
M
Rowlings
P
Hessner
M
et al
T cell-depleted allogeneic bone marrow transplantation for high-risk non-Hodgkin's lymphoma: clinical and molecular follow-up.
Bone Marrow Transplant.
21
1998
893
899
172
Mackinnon
S
Papadopoulos
E
Carabasi
M
et al
Adoptive immunotherapy evaluating escalating doses of donor leukocytes for relapse of chronic myeloid leukemia after bone marrow transplantation: separation of graft-versus-leukemia responses from graft-versus-host disease.
Blood.
86
1995
1261
1268
173
Soiffer
R
Alyea
E
Canning
C
et al
A randomized trial of CD8+ T cell depletion to prevent graft-vs-host disease associated with donor lymphocyte infusions.
Blood.
98
2001
3554
3561
174
Bonini
C
Ferrari
G
Verzeletti
S
et al
HSV-TK gene transfer into donor lymphocytes for control of allogeneic graft-versus-leukemia.
Science.
276
1997
1719
1724
175
Sandmaier
B
Maloney
D
Gooley
T
et al
Nonmyeloablative haematopoietic stem cell for HLA-matched related donors for patients with hematologic malignancies: clinical results of a TBI-based conditioning regimen [abstract].
Blood.
98
2001
742a
176
Martino
R
Caballero
MD
Canals
C
et al
Allogeneic peripheral blood stem cell transplantation with reduced-intensity conditioning: results of a prospective multicentre study.
Br J Haematol.
115
2001
653
659
177
Wasch
R
Reisser
S
Hahn
J
et al
Rapid achievement of complete donor chimerism and low regimen-related toxicity after reduced conditioning with fludarabine, carmustine, melphalan and allogeneic transplantation.
Bone Marrow Transplant.
26
2000
243
250
178
Corradini
P
Tarella
C
Olivieri
A
et al
Reduced-intensity conditioning followed by allografting of hematopoietic cells can produce clinical and molecular remissions in patients with poor-risk hematologic malignancies.
Blood.
99
2002
75
82
179
Raiola
AM
Van Lint
MT
Lamparelli
T
et al
Reduced intensity thiotepa-cyclophosphamide conditioning for allogeneic haemopoietic stem cell transplants (HSCT) in patients up to 60 years of age.
Br J Haematol.
109
2000
716
721
180
Alessandrino
EP
Bernasconi
P
Colombo
AA
et al
Thiotepa and fludarabine (TT-FLUDA) as conditioning regimen in poor candidates for conventional allogeneic hemopoietic stem cell transplant.
Ann Hematol.
80
2001
521
524
181
Slavin
S
Nagler
A
Naparstek
E
et al
Nonmyeloablative stem cell transplantation and cell therapy as an alternative to conventional bone marrow transplantation with lethal cytoreduction for the treatment of malignant and nonmalignant hematologic diseases.
Blood.
91
1998
756
763
182
Slavin
S
Or
R
Prighozina
T
et al
Immunotherapy of hematologic malignancies and metastatic solid tumors in experimental animals and man.
Bone Marrow Transplant.
25(suppl 2)
2000
S54
S57
183
Giralt
S
Thall
PF
Khouri
I
et al
Melphalan and purine analog-containing preparative regimens: reduced-intensity conditioning for patients with hematologic malignancies undergoing allogeneic progenitor cell transplantation.
Blood.
97
2001
631
637
184
Kottaridis
PD
Milligan
DW
Chopra
R
et al
In vivo CAMPATH-1H prevents graft-versus-host disease following nonmyeloablative stem cell transplantation.
Blood.
96
2000
2419
2425
185
Bornhauser
M
Thiede
C
Platzbecker
U
et al
Dose-reduced conditioning and allogeneic hematopoietic stem cell transplantation from unrelated donors in 42 patients.
Clin Cancer Res.
7
2001
2254
2262
186
Craddock
C
Bardy
P
Kreiter
S
et al
Short Report: engraftment of T-cell-depleted allogeneic haematopoietic stem cells using a reduced intensity conditioning regimen.
Br J Haematol.
111
2000
797
800
187
Khouri
IF
Saliba
RM
Giralt
SA
et al
Nonablative allogeneic hematopoietic transplantation as adoptive immunotherapy for indolent lymphoma: low incidence of toxicity, acute graft-versus-host disease, and treatment-related mortality.
Blood.
98
2001
3595
3599
188
Weissinger
F
Sandmaier
BM
Maloney
DG
Bensinger
WI
Gooley
T
Storb
R
Decreased transfusion requirements for patients receiving nonmyeloablative compared with conventional peripheral blood stem cell transplants from HLA-identical siblings.
Blood.
98
2001
3584
3588
189
Carella
AM
Champlin
R
Slavin
S
McSweeney
P
Storb
R
Mini-allografts: ongoing trials in humans.
Bone Marrow Transplant.
25
2000
345
350
190
Martino
R
Caballero
MD
Canals
C
et al
Reduced-intensity conditioning reduces the risk of severe infections after allogeneic peripheral blood stem cell transplantation.
Bone Marrow Transplant.
28
2001
341
347
191
Mohty
M
Faucher
C
Vey
N
et al
High rate of secondary viral and bacterial infections in patients undergoing allogeneic bone marrow mini-transplantation.
Bone Marrow Transplant.
26
2000
251
255
192
Chakraverty
R
Peggs
K
Chopra
R
et al
Limiting transplantation-related mortality following unrelated donor stem cell transplantation by using a nonmyeloablative conditioning regimen.
Blood.
99
2002
1071
1078
193
Maris
M
Niederwieser
D
Sandmaier
B
et al
Nonmyeloablative hematopoietic stem cell transplants using 10/10 HLA antigen matched unrelated donors for patients with advanced hematologic malignancies ineligible for conventional HSCT [abstract].
Blood.
98
2001
858a
194
Nagler
A
Or
R
Naparstek
E
Varadi
G
Slavin
S
Second allogeneic stem cell transplantation using nonmyeloablative conditioning for patients who relapsed or developed secondary malignancies following autologous transplantation.
Exp Hematol.
28
2000
1096
1104
195
Mohty
M
Fegueux
N
Exbrayat
C
et al
Reduced intensity conditioning: enhanced graft-versus-tumor effect following dose-reduced conditioning and allogeneic transplantation for refractory lymphoid malignancies after high-dose therapy.
Bone Marrow Transplant.
28
2001
335
339
196
Carella
AM
Cavaliere
M
Lerma
E
et al
Autografting followed by nonmyeloablative immunosuppressive chemotherapy and allogeneic peripheral-blood hematopoietic stem-cell transplantation as treatment of resistant Hodgkin's disease and non-Hodgkin's lymphoma.
J Clin Oncol.
18
2000
3918
3924
197
Nagler
A
Slavin
S
Varadi
G
Naparstek
E
Samuel
S
Or
R
Allogeneic peripheral blood stem cell transplantation using a fludarabine-based low intensity conditioning regimen for malignant lymphoma.
Bone Marrow Transplant.
25
2000
1021
1028
198
Dreger
P
Glass
B
Seyfarth
B
et al
Reduced-intensity allogeneic stem cell transplantation as salvage treatment for patients with indolent lymphoma or CLL after failure of autologous SCT.
Bone Marrow Transplant.
26
2000
1361
1363
199
Sykes
M
Preffer
F
McAfee
S
et al
Mixed lymphohaemopoietic chimerism and graft-versus-lymphoma effects after non-myeloablative therapy and HLA-mismatched bone-marrow transplantation.
Lancet.
353
1999
1755
1759
200
Hou
J
Fowler
D
Wilson
W
et al
Potent graft-versus-lymphoma effect after nonmyeloablative stem cell transplant in refractory non-Hodgkin's lymphoma: role of rapid complete donor chimerism [abstract].
Blood.
98
2001
404a

Author notes

Philippe Solal-Celigny, Centre Jean Bernard, 9 rue Beauverger 72 000 Le Mans, France; e-mail: p.solal-celigny@noos.fr.

Sign in via your Institution