The treatment of newly diagnosed acute lymphocytic leukemia (ALL) in adults remains unsatisfactory. Not withstanding the outstanding progress in curing childhood ALL, only approximately one third of adults younger than 60 years can be cured, and the overall published survival curves have not changed significantly during the past 15 years. Recent therapeutic advances in allogeneic transplantation through the conduct of large collaborative studies, better understanding of the relevance of cytogenetics, improved molecular techniques for the detection of minimal residual disease, and clinical research into novel biologic and targeted therapies have all combined to offer potentially a better hope for an improved outcome in this disease. The current approach in 2007 to the management of this disease is presented by way of a discussion of illustrative cases. In this uncommon and difficult disease, well-structured intergroup studies will remain vital for future progress.

In contrast to childhood acute lymphocytic leukemia (ALL), therapy of adults with ALL remains unsatisfactory. The long-term survival rate for adults has not significantly changed during the past 2 decades with a 5-year overall survival of only 30% to 40% for patients younger than 60 years, less than 15% for patients older than 60 years, and less than 5% for patients older than 70 years.1,,,,,,,,,,,,14  At the same time, much information has recently been learned about prognostic factors, cytogenetics, the definition of patients most suitable for an allogeneic transplantation, and the increasing indications for use of alternative donor transplants for high-risk ALL. Good prospective studies for relapsed ALL searching for a better outcome and the treatment of older patients remain an elusive goal. At the same time, recent data suggest that perhaps older teenagers are better treated on traditional pediatric protocols rather than on current adult protocols.

To best illustrate some of these new approaches, several case scenarios are presented, followed by a discussion of the therapeutic implications.

A 33-year-old woman presents with pre-B ALL. Her complete blood cell (CBC) count at presentation shows a white blood cell (WBC) count of 11.0 × 109/L (11 000/μL), the hemoglobin level is 98.0 g/L (9.8 g/dL), and the platelet count is 51.0 × 109/L (51 000/μL). She has a normal karyotype and has an HLA-compatible sibling. What is the most appropriate induction and postremission therapy?

This is a young adult who has Philadelphia chromosome-negative ALL. Such patients are considered “standard risk” (there is no such thing as a low-risk adult ALL). The reasons for this are fairly arbitrary, but, by convention, patients are designated as such by a low WBC count at presentation (<30.0 × 109/L [<30 000/μL] for B-lineage and 100.0 × 109/L [<100 000/μL] for T-lineage ALL) and age younger than 35 years.15,16  A normal karyotype has neither “good” nor “poor” prognostic significance.

Recent studies have reported complete response rates for induction therapy of more than 90%. The large international MRC UKALL XII/ECOG 2993 study reported a complete remission rate of 97% in 533 adults at standard risk.17 

Whether there is a single best induction regimen is not known. Although the regimen used in the MRC/ECOG study showed a high complete remission (CR) rate, an alternative induction therapy using the hyper–cyclophosphamide, vincristine, Adriamycin, and dexamethasone (CVAD) regimen also reported a CR rate in excess of 90%.6 The hyper-CVAD regimen is of particular interest because it does not include l-asparaginase as part of the therapeutic armamentarium. l-Asparaginase has been the mainstay of treatment of childhood ALL for the past 3 decades, since the initial study by the Children's Cancer Study Group reported in 1977.18  Although sequential studies since 1977 have shown an improved outcome with asparaginase, there have been no recent prospective studies that have directly compared the outcome of patients, pediatric or adults, with or without asparaginase. The issue is of particular interest because there are increasing data that emphasize the improved outcome of effective depletion of asparagine compared with patients not achieving effective asparagine depletion,19  making it unlikely that such a study will ever be done. The MRC/ECOG study has proposed several modifications of its previously reported regimen and incorporates the pegylated form of asparaginase in place of the previously used native asparaginase. The polyethylene glycol–conjugated asparaginase was developed to extend the half-life and to lessen the frequency of injections.20  There have been several randomized trials studying the pegylated asparaginase compared with the native Escherichia coli asparaginase, all suggesting an improved efficacy.21,23 

Anthracyclines have been a cardinal feature of induction in ALL.8  However, there is no consensus on the optimal anthracycline, dose, or schedule. Daunorubicin is most frequently used, but idarubicin, doxorubicin, and mitoxantrone have also been given usually in weekly schedules, without convincing data that any agent is better than the other. For young adults a weekly dose of 60 mg of daunorubicin given for 4 weeks has been well tolerated and is the one suggested in Table 1, although alternative weekly and daily schedules are probably equally effective.

Table 1

Induction therapy and early intensification for ALL

DoseRouteSchedule
Phase 1, weeks 1-4    
    Daunorubicin 60 mg/m2 IV Days 1,8,15, and 22 
    Vincristine 1.4 mg/m2 IV Days 1,8,15, and 22 
    Dexamethasone 10 mg/m2 PO Days 1-5, 11-14 
    PEG-asparaginase 2000 U/m2 IV Days 2 and16 
    Methotrexate 12.5 mg IT Day 14 
Phase 2, weeks 5-8*    
    Cyclophosphamide 650 mg/m2 IV Days 1, 15, and 29 
    Cytarabine 75 mg/m2 IV Days 6-9, 13-16, 20-23, and 27-30 
    6-mercaptopurine 6 mg/m2 PO Days 1-30 
    Methotrexate 12.5 mg IT Days 1, 8, 15, and 22 
Intensification/CNS prophylaxis    
    Methotrexate 3 g/m2 IV Days 1 and 8 
    PEG-asparaginase 2000 U/m2 IV Day 2 
DoseRouteSchedule
Phase 1, weeks 1-4    
    Daunorubicin 60 mg/m2 IV Days 1,8,15, and 22 
    Vincristine 1.4 mg/m2 IV Days 1,8,15, and 22 
    Dexamethasone 10 mg/m2 PO Days 1-5, 11-14 
    PEG-asparaginase 2000 U/m2 IV Days 2 and16 
    Methotrexate 12.5 mg IT Day 14 
Phase 2, weeks 5-8*    
    Cyclophosphamide 650 mg/m2 IV Days 1, 15, and 29 
    Cytarabine 75 mg/m2 IV Days 6-9, 13-16, 20-23, and 27-30 
    6-mercaptopurine 6 mg/m2 PO Days 1-30 
    Methotrexate 12.5 mg IT Days 1, 8, 15, and 22 
Intensification/CNS prophylaxis    
    Methotrexate 3 g/m2 IV Days 1 and 8 
    PEG-asparaginase 2000 U/m2 IV Day 2 

IV indicates intravenously; PO, by mouth; and IT, intrathecal.

*Postpone until total WBC count exceeds 3 × 109/L

Although the overall reported induction mortality is approximately 5%, most of those patients died of opportunistic infections, including Aspergillus. The prolonged immunosuppression associated with the 2 phases of induction is thought to contribute to this risk, and the lengthy exposure to corticosteroids is likely to add to this risk. Recent data from the pediatric literature have suggested that the use of dexamethasone, as opposed to prednisone, may improve the outcome. There are also data that suggest that dexamethasone has greater penetration of the central nervous system (CNS) and fewer thromboembolic events than prednisone, and its in vitro antileukemic activity is greater.24  Furthermore, 2 of 3 randomized studies have shown improved survival in children receiving dexamethasone as opposed to prednisone.25,27  For this reason, a truncated use of corticosteroids, in the form of dexamethasone, has been used in the induction regimen, as shown in Table 1.

It must be emphasized that, although successful induction results are critical for long-term survival in ALL,17  this does not of itself predict for long-term survival because most patients, including those with high-risk ALL, achieve a complete remission. Thus, although it is unlikely that any modifications to the induction regimen will show an improvement in the initial response rate, it is important to continue to study modifications with the view of improving the long-term outcome.

Phase 3 studies are under way to evaluate the role of monoclonal antibody therapy in B-lineage ALL, focusing mostly on either epratuzumab (anti-CD22) or rituximab (anti-CD20).28,29  Such therapy may decrease the level of minimal residual disease (MRD) at the completion of induction therapy. Recent reports suggest an advantage when hyper-CVAD plus rituximab was compared with historical patients with B-ALL treated with hyper-CVAD alone.30  There have also been preliminary reports suggesting a role for rituximab together with hyper-CVAD in patients with CD20+ pre-B ALL.31  Rituximab has already formed part of the standard protocol for older and younger adults with B-precursor ALL in the German Multicenter Study Group for Adult ALL (GMALL) protocols.17  In contrast to the antiproliferative effects of rituximab, epratuzumab appears to function more by an immunomodulatory mechanism,32  and CD22 is expressed on the surface of most pre-B ALL cells. There are mature data in lymphoma, and more preliminary data in ALL, that demonstrate its safety and tolerability as well as significant efficacy, thus making it an attractive agent to study in ALL.33,35  There have also been encouraging data using a combination of rituximab and epratuzumab in lymphomas with reported 60% to 70% response rate and easy tolerability.36,37  Guanine arabinoside, or nelarabine, has been shown to have significant activity in refractory T-cell malignancies; thus, for T-lineage ALL, nelarabine is likely in future studies to be incorporated either into the late phase of induction or the early part of intensification or maintenance therapy, or both.38,39  All the foregoing have much promise and are to be investigated in prospective randomized trials; they cannot yet be considered as standard of care.

Alemtuzumab, a monoclonal antibody directed against CD52, is also an attractive agent to be studied in ALL because of its broad expression on most T- and B-cell ALLs. Limited activity has been reported in advanced ALL,40  and this is likely to be incorporated in future studies for patients with newly diagnosed ALL who are CD52+. Other promising novel agents that are likely to be incorporated into clinical trials in the future include Notch1-activating mutations,41  liposomal vincristine,42  and intrathecal liposomal cytarabine.43 

For postremission therapy the standard of care has evolved during the past few years. Historically, there has been resistance to the use of an allogeneic transplantation for ALL in first complete remission based, in part, on extrapolation from the success in childhood ALL using only chemotherapy,44  as well as retrospective comparisons with registry data.45  This was somewhat surprising because the clinical importance of the graft-versus-leukemia (GvL) effect has been recognized since 1979, and the initial reports were in patients with ALL.46,47  During the past 2 decades, the role of allogeneic transplantation, however, has been recognized in patients at high risk, such as those with Philadelphia chromosome-positive ALL and other high-risk features.16,48,50  Most clinicians have, however, shied away from offering allogeneic transplantations to patients with adult ALL who had an HLA-compatible sibling and were not at high risk.

During the past 14 years the MRC/ECOG study has assigned such therapy to all adult patients, including those with standard-risk ALL, who had an HLA-compatible sibling. The final data were recently reported and showed an unequivocal benefit for HLA-compatible sibling transplantation for patients with standard-risk ALL over other forms of conventional therapy, whether this is consolidation or maintenance therapy or an autologous transplantation.51  Patients with standard-risk ALL (n = 218) who had an HLA-matched donor were compared with 286 patients who did not have a donor with a 5-year overall survival of 63% versus 51%, respectively (P ≤ .05). The patients with the best outcome with standard chemotherapy derived the greatest benefit from an allogeneic transplantation. Such a transplantation should be performed as soon as possible after induction to minimize the transplant-related morbidity and mortality. A total body irradiation (TBI)–based conditioning has been traditionally preferred in ALL, in contrast to acute myelogenous leukemia (AML), and this also provides for added CNS prophylaxis.

The importance of the GvL effect for ALL in first complete remission cannot be overemphasized because the relapse rate is significantly reduced in all patients with ALL. This could have been possibly misunderstood because of the poor data of donor leukocyte infusions in patients with relapsed ALL.52  Extrapolating data from relapsed ALL to patients in first remission has mistakenly led to the assumption that the GvL effect in ALL is not of an overriding significance.

An evaluation of minimal residual disease (MRD) in ALL is assuming increasing clinical relevance and is clearly an important prognostic factor also in adults with ALL, although a decrease in MRD occurs more slowly in adults than in children, and fewer patients reach a negative status.53,,56  Whether such evaluation will result in the ability to better predict for patients who may not require an allogeneic transplantation remains to be proven in prospective studies. Conversely, the presence of detectable MRD by molecular methods after completion of both phases of induction predicts for a poor prognosis even in patients with standard-risk ALL and such patients should proceed to an allogeneic transplantation from an alternative donor.

This patient should be offered a standard induction regimen, similar to the one outlined in Table 1, but without the intensification. She should then receive a myeloablative, TBI-based, allogeneic transplant from an HLA-matched sibling.

An 18-year-old male adolescent presents with common acute lymphoblastic leukemia antigen (CALLA)–positive ALL with a hyperdiploid karyotype. His CBC count at presentation shows a WBC count of 23.0 × 109/L (23 000/μL), the hemoglobin level is 72.0 g/L (7.2 g/dL), and the platelet count is 22.0 × 109/L (22 000/μL). He has an HLA-matched sibling. What is the best induction and postremission therapy?

The issue of treating an adolescent with ALL has come under much scrutiny during the past decade. There are increasing data that suggest that adolescents treated with adult ALL protocols have a worse outcome than similar adolescents treated on pediatric protocols.8,19,57,,,,62  All these comparisons have been retrospective and recognize the inherent difficulty in comparing adolescents who were treated in adult with those treated in pediatric units; for example “older looking” young adults may be referred to an adult ward. Nevertheless, the preponderance of the data suggest that such patients are more likely to benefit from pediatric regimens that, as a general rule, are more intensive, include far more asparaginase, and probably reflect a greater protocol discipline, especially about timeliness of administration and adherence to dosing schedules. Thus, although this particular patient has standard-risk ALL, there are no data that have unequivocally shown the benefit of an allogeneic transplantation for such a patient. Although all standard-risk patients older than 15 years have been included in the MRC/ECOG study,51  a typical adult regimen was used; thus, this remains an open issue. Although children having a hyperdiploid karyotype have been reported to have a more favorable prognosis, this has not been observed in adults.63,65  The data recently reported by the Dana-Farber Cancer Institute in Boston, Massachusetts, for 51 adolescent patients aged 15 to 18 years showed a remarkable event-free survival of 78% in this population. This is clearly better than anything else that has been reported to date, and, if such data are confirmed in an unselected population, there clearly will be no role for an allogeneic transplantation in this age group.66 

This patient should be treated on an institutional pediatric regimen for standard-risk patients with ALL and should not undergo an allogeneic transplantation.

A 48-year-old woman presents with T-lineage ALL. Her WBC count at presentation is 140.0 × 109/L (140 000/μL), the hemoglobin level is 81.0 g/L (8.1 g/dL), and the platelet count is 8.0 × 109/L (8000/μL). Cytogenetic analysis revealed a complex karyotype in 14 of 20 metaphases. She does not have an HLA-matched donor.

This patient clearly has a poor prognosis for several reasons. Her age and elevated WBC count are established poor prognostic features. Until recently, the significance of a complex karyotype in ALL was uncertain. Although this has known poor prognostic significance in AML, and almost intuitively was considered as such in ALL, there had been no data that unequivocally established this. The very recent report of cytogenetics in ALL from the MRC/ECOG study showed for the first time the independently poor prognosis conferred by a complex karyotype.65  In that study there were 41 patients who were negative for the Philadelphia chromosome and had a complex karyotype defined as the presence of 5 or more chromosomal abnormalities. There was a statistically worse overall and event-free survival that was independent from other known prognostic variables.

Historically, most clinical trials have confirmed immunophenotyping as a critical part of the prognostic evaluation of patients with ALL.67  Most trials have reported a superior outcome of T-lineage ALL compared with B-lineage.8,51  However, as with B-lineage disease, this broad classification is likely to be superseded by subtyping for early versus thymic and mature T-ALL.8  Furthermore, Notch1-activating mutations may, in the future, provide further prognostic information for patients with T-ALL.68 

Although the GvL effects for adult ALL in first remission are clearly present in all patients, including those at high risk, this particular patient presents a management dilemma because of the high nonrelapse mortality associated with allogeneic transplantation in this age group. In the MRC/ECOG study the nonrelapse mortality was 29% and 39% at 1 and 2 years, respectively, with most of the deaths related to graft-versus-host disease (GvHD). In fact, the high transplant-related mortality abrogated the reduction in relapse risk for high-risk older patients with a 5-year survival (39%) and event-free survival that were only marginally and nonsignificantly improved at 5 years compared with patients without a donor (36%).51  The difficulties with managing this patient are further compounded in that she did not have an HLA-matched sibling donor.

At the same time, the GvL effect remains the single most potent antileukemic strategy; thus, every effort must be made to offer this patient an allogeneic transplantation, recognizing the need to consider a less toxic transplantation-conditioning regimen.

Unfortunately, there are only scanty data for the use of reduced-intensity transplantation conditioning in ALL. Such an approach takes into consideration all the known hazards, including failure of engraftment and increased likelihood of relapse. Recent preliminary data reported a 1-year overall survival of approximately 70% for high-risk patients with ALL using reduced-intensity conditioning with fludarabine and melphalan.69  The use of alemtuzumab for such patients has also been reported with encouraging results.70,71 

The reported data from Ph-positive ALL showed that for high-risk patients a matched unrelated donor (MUD) is an acceptable alternative for those who do not have a family donor.72  There are also other published data indicating that the risk of a MUD allogeneic transplantation for patients with ALL is not much greater than the risk of a sibling allogeneic transplantation.73,75  Thus, the approach for this patient should be to offer standard induction therapy which is likely to lead to a 90% complete response rate.17  A search for an unrelated donor must be initiated urgently so that this could be offered, without delay, to the patient after successful induction therapy. Another alternative is the use of a haploidentical transplantation. The experience with this is limited by comparison with a MUD transplantation,76  but an update from the group in Perugia, Italy, reported an event-free survival of 46% for the 24 patients with ALL who received a transplant in first remission.77 

This high-risk patient should be offered standard induction followed by an allogeneic transplant from a matched unrelated donor using the best institutional regimen for reduced intensity conditioning. Transplantation-related risks need to be emphasized.

A 53-year-old man presents with Ph-positive ALL. He does not have an HLA-compatible sibling. His CBC count at presentation shows a WBC count of 13.0 × 109/L (13 000/μL), a hemoglobin level of 93.0 g/L (9.3 g/dL), and a platelet count of 23.0 × 109/L (23 000/μL). He received standard induction therapy and imatinib mesylate. At the end of induction and intensification, 3 months from diagnosis, he has no cytogenetic or molecular evidence for BCR-ABL.

Management of patients with Ph-positive ALL has long been recognized as the single worst prognostic predictive factor in ALL. More than 90% of such patients succumb to their disease if an allogeneic transplantation was not performed in first remission. For young adults with Ph-positive ALL an allogeneic transplantation, either from a histocompatible sibling or from a matched unrelated donor, is able to affect a cure in 35% to 50% of patients.

For these reasons, traditional therapy has been to offer some form of an allogeneic transplantation, at almost any cost, to patients with Ph-positive ALL.78,79  Until a year or 2 ago, based on sound trial data, this patient would have been offered a MUD transplantation.67 

The advent of imatinib mesylate for patients with Ph-positive ALL has altered the standard of care.80  Although there has never been a prospective study of patients with ALL comparing the results of an allogeneic transplantation with or without concurrent imatinib mesylate therapy, the preponderance of the data report an important role for imatinib mesylate in inducing a more profound disease response at all ages, making it unethical to conduct any therapeutic protocol in ALL without the use of imatinib mesylate. Important issues remain. At what points should imatinib mesylate be given? Should this be at the start of induction or at some later point? Conversely, is there a role for initiating therapy with imatinib mesylate before chemotherapy? And for those patients undergoing an allogeneic transplantation should imatinib mesylate be given after transplantation and for how long?

One of the most important issues that needs to be emphasized is the fact that since the advent of imatinib mesylate there has never been a study that supports the ongoing imperative for an allogeneic transplantation in Ph-positive ALL.81  Should all adult patients, even those at high risk of transplant-related mortality, be referred for an alternative donor transplantation, when a histocompatible sibling is not available? The transplant-related mortality in these patients, using a conventional transplantation regimen, must be in the order of 40% at 2 years. Moreover, even in the era before imatinib mesylate a small percentage of such patients were cured without a transplantation.

Given that this patient had at 3 months no evidence for MRD and was completely negative for BCR-ABL, there are no compelling data to offer him a matched unrelated donor transplantation, although an allogeneic transplantation would likely still be offered if a sibling donor were available. Much like chronic myeloid leukemia (CML), therapy with imatinib mesylate should be continued indefinitely.

This patient should be continued on the standard maintenance protocol, including imatinib mesylate, which should be continued indefinitely.

A 67-year-old man presents with pre-B ALL. His CBC at presentation shows a WBC count of 53.0 × 109/L (53 000/μL), a hemoglobin level of 82.0 g/L (8.2 g/dL), and a platelet count of 21.0 × 109/L (21 000/μL).

The management of older patients with ALL represents a unique therapeutic challenge. Unfortunately, there have been few prospective studies in this age group, although the median age of ALL in adults is older than 60 years.82  Most older patients are excluded from such trials even if they are eligible. Furthermore, patients who are referred to tertiary care medical centers are a select group and even there, only a few are referred for clinical trials. Thus, the true long-term response rates are likely to be even significantly lower than the published data. Taking all this into account, it is likely that among patients who are aged between 60 and 70 years, there may be 10% who are long-term survivors; and older than 70 years the long-term survival is less than 5%.

The problem for older adults is 2-fold. First, there are inherent poor prognostic features more common among older adults, putting this age population at high risk. Most important among these is the high incidence of the Philadelphia chromosome, approximately 50%, among older patients with B-lineage ALL.65,83,85  Because approximately 90% of older patients are of B-lineage, it means that almost half of older adults are at high risk.84  The second problem is the associated comorbidities and the inability to tolerate the rigors of protracted therapy. As with AML, a significant proportion of older patients can achieve a complete remission; the problem is maintaining them in such a state. In the few studies that have been designed specifically for patients older than 65 years, initial complete response rates of approximately 50% were obtained with the best results being patients who had low white cell counts, were Ph-negative, or those who had T-lineage. The overall survival, however, was less than 10%.86,87  However, among 519 patients older than 60 years, who received intensive chemotherapy on a prospective trial that was not limited by age, the median remission duration was reported to be 9 months and the median survival was 7 months.88 

It is clear that a design of a regimen for older adults needs to be attenuated from standard therapy for younger patients.89,90  Because achieving a complete remission seems to be a sine qua non for any form of sustained disease-free survival in ALL,51  a serious attempt should be made to get patients into complete remission. A standard regimen for younger adults should be used, but the duration of corticosteroids should be truncated, and full supportive measures with growth factors should be used to minimize the period of neutropenia. The benefit of cytokines in ALL has been unequivocally shown in several well-designed controlled trials.91  To further reduce the toxicity, investigations are ongoing using agents that would allow for maximal drug delivery with lower toxicity such as liposomal vincristine and, possibly, liposomal daunorubicin. Monoclonal antibody therapy assumes even greater importance in this population that is unable to withstand intensive therapy. Clearly imatinib mesylate is the mainstay for patients who are Philadelphia chromosome positive. This tyrosine kinase inhibitor, as well as the new agents dasatinib and nilotinib, are rapidly changing the prognosis for this particular subset of patients. Emerging data suggest that this agent may be offered to older patients in preference to standard chemotherapy because it has considerably less toxicity.10  Other monoclonal antibodies, such as rituximab and epratuzumab, should be incorporated wherever possible among B-lineage patients. Because the percentage of patients who can be cured is low, it is important to administer drugs that preserve the quality of life as far as is possible. However, a palliative approach at diagnosis is not appropriate for those who are reasonably fit because an initial remission can usually be achieved with reasonably acceptable toxicity and may attain a significant period free from disease. Reduced-intensity allogeneic transplantation may in the future affect a cure in some such patients, but there are no data at present to support this.

A suggested regimen for this patient should consist of phase 1 of induction as in Table 1 with the addition of granulocyte-colony stimulating factor (G-CSF). Phase 2 should also be given together with G-CSF, but without the day 29 of cyclophosphamide and the last 4 days of cytarabine (days 27-30). If complete remission is achieved, maintenance therapy should consist only of methotrexate and 6-mercaptopurine, with vincristine and prednisone added every 2 to 3 months. Clearly, if the patient is Ph-positive, imatinib mesylate should be given throughout. If the patient is CD20+, rituximab should also be added every 4 weeks, starting before the first cycle of induction.

A 32-year-old man presents with T-lineage ALL. His CBC at presentation shows a WBC count of 51.0 × 109/L (51 000/μL), hemoglobin level of 111.0 g/L (11.1 g/dL), and a platelet count of 6.0 × 109/L (6000/μL). Cytogenetic analysis showed the t(10;14)(q24;q11.2) karyotype in 7 of 20 metaphases. The patient reported a mild headache, and a routine lumbar puncture revealed the presence of lymphoblasts in his cerebrospinal fluid. He does not have an HLA-matched sibling.

This is a patient whose age, immunophenotype, WBC count at presentation, and karyotype do not place him into the high-risk category. This karyotype is almost uniquely found among patients with T-cell ALL,51  but it has no important prognostic significance. It is clear that such patients require additional therapy to the CNS. However, the issue at hand is whether otherwise standard protocols for ALL are appropriate for such a patient. The overall incidence of CNS involvement at presentation is in the region of 5% to 8%.92,,95  Historically, many such patients were considered to be at inherently high risk of relapse and were excluded from many standard protocols. It was assumed by many that such patients required a far more intensive induction and postremission therapy, even on rapid resolution of findings in the cerebrospinal fluid. During the past few years, 2 large studies reported the results based on protocols that offered unchanged standard of care for patients who presented with CNS disease. The LALA-94 study described 48 patients who had CNS disease at presentation with an outcome that did not differ from the more than 700 remaining patients.96  The MRC/ECOG study reported on 77 patients who had CNS disease at presentation of 1508 patients.97  That study showed an overall survival at 10 years of 34% for patients who did not have CNS disease compared with 29% for patients who did have CNS disease. This improvement is marginally significant (P = .03). However, when tested for heterogeneity, looking at the influence of white cell count at presentation, the differences were not statistically significant, especially for the 25 patients with T-ALL who presented with CNS disease.

Although it is always possible that the incidence of CNS disease at presentation is greater than reported in these main groups, because many clinicians may have selectively excluded such patients from analysis, taken together these data suggest that at the current time there is no evidence that any therapy is known to be better for such patients and such persons should be encouraged to enter standard clinical protocols for ALL. It is clear, however, that such patients need to be given added CNS therapy at diagnosis, and the regimen used by the MRC/ECOG is indicated in Table 2.

Table 2

Management of CNS leukemia present at diagnosis

1. Intrathecal or intraventricular (Ommaya reservoir) methotrexate 12.5 mg three times weekly until clearance of the cerebrospinal fluid. 
2. Lucovorin 10 mg/m2 intravenously or orally every 6 hours for 4 doses, starting 24 hours after the administration of methotrexate. 
3. Cranial irradiation 2400 cGy and 1200 cGy to the spinal cord administered concurrently with phase 2 of induction. 
4. Systemic chemotherapy is not to be delayed while intrathecal or radiation therapy is given. 
1. Intrathecal or intraventricular (Ommaya reservoir) methotrexate 12.5 mg three times weekly until clearance of the cerebrospinal fluid. 
2. Lucovorin 10 mg/m2 intravenously or orally every 6 hours for 4 doses, starting 24 hours after the administration of methotrexate. 
3. Cranial irradiation 2400 cGy and 1200 cGy to the spinal cord administered concurrently with phase 2 of induction. 
4. Systemic chemotherapy is not to be delayed while intrathecal or radiation therapy is given. 

This patient who is at standard risk would have been a candidate for an allogeneic transplantation had a histocompatible sibling been available. In the absence of such a donor there are 3 options, which include standard protracted consolidation maintenance therapy, an autologous transplantation, or a MUD transplantation. Because he is at standard risk and in the absence of data showing the presence of minimal residual disease after induction and intensification, there are no data that would support subjecting him to the increased risks from an unrelated donor transplantation.

The issue of offering an autologous transplantation has been studied by several groups. It is theoretically attractive because the possibility of offering a single autologous transplantation with relatively low morbidity and minimal mortality, using current practice, would seem to be an attractive theoretical alternative. Several modestly sized studies have reported that an autologous transplantation was at least comparable with standard chemotherapy. The LALA-87 trial compared 95 patients who underwent an autologous transplantation with 96 patients who were randomly assigned to chemotherapy. The 10-year overall survival was 34% compared with 29% (NS).98  The subsequent LALA-94 study compared 70 patients who received an autologous transplant with 59 patients who received chemotherapy with a 6-year disease-free survival of 21% compared with 6% (NS).96  The ECOG/MRC study prospectively evaluated the role of an autologous transplantation in large numbers and for the first time actually reported that there was no evidence that a single autologous transplantation could replace standard consolidation or maintenance therapy in any risk groups because of an inferior event-free survival of 33% in patients undergoing an autologous transplantation compared with 42% for patients randomly assigned to chemotherapy (P ≤ .05). Thus, such an option cannot be recommended for this patient, although a TBI-based conditioning regimen has a theoretic advantage in this patient with CNS disease at presentation.

This patient with CNS disease at presentation but otherwise with standard-risk features should be treated with standard induction therapy together with treatment of his CNS disease. Once in complete remission, he should receive intensification and postremission therapy. An allogeneic transplantation should be offered if he has a matched sibling. Otherwise he should receive standard maintenance therapy.

A 52-year-old woman with pre-B ALL went into complete remission and was treated with standard intensification or maintenance therapy. She relapsed 2 years from diagnosis while receiving maintenance. Her CBC count shows a WBC count of 1.1 × 109/L (1100/μL), hemoglobin level of 63.0 g/L (6.3 g/dL), and a platelet count of 12.0 × 109/L (12 000/μL). Her bone marrow is completely replaced with lymphoblasts having the same original immunophenotype. She does not have a compatible sibling.

Patients with relapsed ALL have a uniformly poor prognosis. Among 609 relapsed patients, the median survival in the MRC/EGOG study was 24 weeks, and 22% survived for 1 year and only 7% for 5 years.14  The best treatment for patients with relapsed ALL is unknown. Several regimens have been suggested.99,,102  A few facts are clear. The only cure for an adult who has relapsed ALL is an allogeneic transplantation. It is also clear that it is possible to achieve a complete remission with a variety of regimens that range from the administration of vincristine and prednisone alone to a repeat of any of the standard induction regimens or to some of the refractory regimens, such as fludarabine, cytosine arabinoside, and G-CSF (FLAG) or hyper-CVAD, if this was not used in induction. The actual rate of achievement of a CR will depend, much like AML, on the duration of first remission, whether the relapse occurs while on maintenance therapy, and, to some extent, on the degree of the intensity of the prior therapy received and the degree of marrow involvement at relapse. There is also the dilemma of deciding whether to attempt reinduction therapy or to proceed directly to an allogeneic transplantation if a sibling donor is available or if an unrelated donor has been identified. It is clear that the results from an allogeneic transplantation are far superior if performed in remission, and there may be occasions, especially if the relapse occurs after a short remission of say, less than 6 months, when the likelihood of achieving a second remission is so small that the toxicity of reinduction and its effect on a subsequent transplantation may put the balance in favor of an immediate allogeneic transplantation.

The MRC/ECOG also evaluated the efficacy of transplantation after relapse.14  If one selects patients who have survived 3 months from transplantation and had not received prior transplantation in first remission, then patients who undergo an allogeneic transplantation from a sibling donor have an approximately 25% chance of sustained survival, especially if they have achieved a second complete remission. An analysis of 125 such patients reveals a 5-year survival of 25% for 42 patients who underwent a sibling allograft, 16% for those undergoing a MUD transplantation, 15% for those undergoing an autograft, and only 4% for those receiving chemotherapy only.

This particular patient should be reinduced. Although her relapse occurred while on maintenance therapy, this is mitigated by almost 2 years in first remission. A search for an unrelated donor should be instituted as soon as possible, and, if a donor is identified, she should receive a transplant, provided she gets complete or partial response from reinduction. If the disease continues to progress on reinduction therapy, the likelihood of a response from any form of a transplantation is exceedingly low, less than 5%, and at that point consideration should be given for palliative care.

This patient should be reinduced with a regimen such as hyper-CVAD, or any other institutional regimen for relapse. If she achieves a complete or partial response, she should undergo a transplantation from a matched unrelated donor.

We thank Sonia Kamenetsky for assistance in the preparation of the manuscript.

Contribution: J.M.R. and A.H.G. wrote the paper.

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

Correspondence: Jacob M. Rowe, Department of Hematology and Bone Marrow Transplantation, Rambam Medical Center and Technicon, Israel Institute of Technology, Haifa 31096, Israel; e-mail:rowe@jimmy.harvard.edu.

1
Taylor
PR
Reid
MM
Bown
N
Hamilton
PJ
Proctor
SJ
Acute lymphoblastic leukemia in patients aged 60 years and over: a population-based study of incidence and outcome.
Blood
1992
80
1813
1817
2
Gokbuget
N
Hoelzer
D
Arnold
R
et al
Treatment of adult ALL according to protocols of the German Multicenter Study Group for Adult ALL (GMALL).
Hematol Oncol Clin North Am
2000
14
1307
1325
3
Linker
C
Damon
L
Ries
C
Navarro
W
Intensified and shortened cyclical chemotherapy for adult acute lymphoblastic leukemia.
J Clin Oncol
2002
20
2464
2471
4
Annino
L
Vegna
ML
Camera
A
et al
Treatment of adult acute lymphoblastic leukemia (ALL): long-term follow-up of the GIMEMA ALL 0288 randomized study.
Blood
2002
99
863
871
5
Takeuchi
J
Kyo
T
Naito
K
et al
Induction therapy by frequent administration of doxorubicin with four other drugs, followed by intensive consolidation and maintenance therapy for adult acute lymphoblastic leukemia: the JALSG-ALL93 study.
Leukemia
2002
16
1259
1266
6
Kantarjian
H
Thomas
D
O'Brien
S
et al
Long-term follow-up results of hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone (Hyper-CVAD), a dose-intensive regimen, in adult acute lymphocytic leukemia.
Cancer
2004
101
2788
2801
7
Larson
RA
The U.S. trials in adult acute lymphoblastic leukemia.
Ann Hematol
2004
83
suppl 1
S127
S128
8
Gokbuget
N
Hoelzer
D
Treatment of adult acute lymphoblastic leukemia.
Hematology Am Soc Hematol Educ Program
2006
133
141
9
Pui
CH
Evans
WE
Treatment of acute lymphoblastic leukemia.
N Engl J Med
2006
354
166
178
10
Ottmann
OG
Wassmann
B
Pfeifer
H
et al
Imatinib compared with chemotherapy as front-line treatment of elderly patients with Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ALL).
Cancer
2007
109
2068
2076
11
Mizuta
S
Kohno
A
Morishita
Y
et al
Long-term follow-up of 14 patients with Philadelphia chromosome-positive acute lymphoblastic leukemia following autologous bone marrow transplantation in first complete remission.
Int J Hematol
2007
85
140
145
12
Vignetti
M
Fazi
P
Cimino
G
et al
Imatinib plus steroids induces complete remissions and prolonged survival in elderly Philadelphia chromosome-positive patients with acute lymphoblastic leukemia without additional chemotherapy: results of the Gruppo Italiano Malattie Ematologiche dell'Adulto (GIMEMA) LAL0201-B protocol.
Blood
2007
109
3676
3678
13
Carpenter
PA
Snyder
DS
Flowers
ME
et al
Prophylactic administration of imatinib after hematopoietic cell transplantation for high-risk Philadelphia chromosome-positive leukemia.
Blood
2007
109
2791
2793
14
Fielding
AK
Richards
SM
Chopra
R
et al
Outcome of 609 adults after relapse of acute lymphoblastic leukemia (ALL); an MRC UKALL12/ECOG 2993 study.
Blood
2007
109
944
950
15
Hoelzer
D
Thiel
E
Loffler
H
et al
Prognostic factors in a multicenter study for treatment of acute lymphoblastic leukemia in adults.
Blood
1988
71
123
131
16
Hunault
M
Harousseau
JL
Delain
M
et al
Better outcome of adult acute lymphoblastic leukemia after early genoidentical allogeneic bone marrow transplantation (BMT) than after late high-dose therapy and autologous BMT: a GOELAMS trial.
Blood
2004
104
3028
3037
17
Rowe
JM
Buck
G
Burnett
AK
et al
Induction therapy for adults with acute lymphoblastic leukemia: results of more than 1500 patients from the international ALL trial: MRC UKALL XII/ECOG E2993.
Blood
2005
106
3760
3767
18
Ortega
JA
Nesbit
ME
Jr
Donaldson
MH
et al
L-Asparaginase, vincristine, and prednisone for induction of first remission in acute lymphocytic leukemia.
Cancer Res
1977
37
535
540
19
Wetzler
M
Sanford
BL
Kurtzberg
J
et al
Effective asparagine depletion with pegylated asparaginase results in improved outcomes in adult acute lymphoblastic leukemia: Cancer and Leukemia Group B Study 9511.
Blood
2007
109
4164
4167
20
Graham
ML
Pegaspargase: a review of clinical studies.
Adv Drug Deliv Rev
2003
55
1293
1302
21
Silverman
LB
Gelber
RD
Dalton
VK
et al
Improved outcome for children with acute lymphoblastic leukemia: results of Dana-Farber Consortium Protocol 91–01.
Blood
2001
97
1211
1218
22
Avramis
VI
Sencer
S
Periclou
AP
et al
A randomized comparison of native Escherichia coli asparaginase and polyethylene glycol conjugated asparaginase for treatment of children with newly diagnosed standard-risk acute lymphoblastic leukemia: a Children's Cancer Group study.
Blood
2002
99
1986
1994
23
Douer
D
Yampolsky
H
Cohen
LJ
et al
Pharmacodynamics and safety of intravenous pegaspargase during remission induction in adults aged 55 years or younger with newly diagnosed acute lymphoblastic leukemia.
Blood
2007
109
2744
2750
24
Nowak-Gottl
U
Ahlke
E
Fleischhack
G
et al
Thromboembolic events in children with acute lymphoblastic leukemia (BFM protocols): prednisone versus dexamethasone administration.
Blood
2003
101
2529
2533
25
Bostrom
BC
Sensel
MR
Sather
HN
et al
Dexamethasone versus prednisone and daily oral versus weekly intravenous mercaptopurine for patients with standard-risk acute lymphoblastic leukemia: a report from the Children's Cancer Group.
Blood
2003
101
3809
3817
26
Mitchell
CD
Richards
SM
Kinsey
SE
et al
Benefit of dexamethasone compared with prednisolone for childhood acute lymphoblastic leukaemia: results of the UK Medical Research Council ALL97 randomized trial.
Br J Haematol
2005
129
734
745
27
Igarashi
S
Manabe
A
Ohara
A
et al
No advantage of dexamethasone over prednisolone for the outcome of standard- and intermediate-risk childhood acute lymphoblastic leukemia in the Tokyo Children's Cancer Study Group L95–14 protocol.
J Clin Oncol
2005
23
6489
6498
28
Gokbuget
N
Hoelzer
D
Rituximab in the treatment of adult ALL.
Ann Hematol
2006
85
117
119
29
Gokbuget
N
Hoelzer
D
Novel antibody-based therapy for acute lymphoblastic leukaemia.
Best Pract Res Clin Haematol
2006
19
701
713
30
Thomas
DA
Faderl
S
O'Brien
S
et al
Chemoimmunotherapy with hyper-CVAD plus rituximab for the treatment of adult Burkitt and Burkitt-type lymphoma or acute lymphoblastic leukemia.
Cancer
2006
106
1569
1580
31
Thomas
DA
Cortes
J
O'Brien
S
et al
Update of the modified hyper-CVAD regimen with or without rituximab in newly diagnosed adult acute lymphocytic leukemia (ALL) [abstract].
Blood
2005
106
521a
Abstract 1831
32
Carnahan
J
Stein
R
Qu
Z
et al
Epratuzumab, a CD22-targeting recombinant humanized antibody with a different mode of action from rituximab.
Mol Immunol
2007
44
1331
1341
33
Leonard
JP
Coleman
M
Ketas
JC
et al
Phase I/II trial of epratuzumab (humanized anti-CD22 antibody) in indolent non-Hodgkin's lymphoma.
J Clin Oncol
2003
21
3051
3059
34
Leonard
JP
Coleman
M
Ketas
JC
et al
Epratuzumab, a humanized anti-CD22 antibody, in aggressive non-Hodgkin's lymphoma: phase I/II clinical trial results.
Clin Cancer Res
2004
10
5327
5334
35
Boronitz
MJ
Carrol
WL
Adamson
PC
et al
Effective targeting of leukemic cells in children with B-precursor acute lymphoblastic leukemia treated with anti-CD22 (Epratuzumab). A Children's Oncology Group (COG) study [abstract].
Blood
2006
108
2585a
Abstract 2585
36
Leonard
JP
Coleman
M
Ketas
JC
et al
Combination antibody therapy with epratuzumab and rituximab in relapsed or refractory non-Hodgkin's lymphoma.
J Clin Oncol
2005
23
5044
5051
37
Strauss
SJ
Morschhauser
F
Rech
J
et al
Multicenter phase II trial of immunotherapy with the humanized anti-CD22 antibody, epratuzumab, in combination with rituximab, in refractory or recurrent non-Hodgkin's lymphoma.
J Clin Oncol
2006
24
3880
3886
38
Berg
SL
Blaney
SM
Devidas
M
et al
Phase II study of nelarabine (compound 506U78) in children and young adults with refractory T-cell malignancies: a report from the Children's Oncology Group.
J Clin Oncol
2005
23
3376
3382
39
Dunsmore
K
Devidas
M
Borowitz
MJ
et al
Nelarabine can be safely incorporated into an intensive, multi-agent chemotherapy regimen for the treatment of T-cell acute lymphocytic leukemia in children: a report from the Children's Oncology Group (AALL00P2 protocol for T-cell leukemia) [abstract].
Blood
2006
108
528a
Abstract 1864
40
Tibes
R
Keating
MJ
Ferrajoli
A
et al
Activity of alemtuzumab in patients with CD52-positive acute leukemia.
Cancer
2006
106
2645
2651
41
Vujovic
S
Henderson
SR
Flanagan
AM
Clements
MO
Inhibition of gamma-secretases alters both proliferation and differentiation of mesenchymal stem cells.
Cell Prolif
2007
40
185
195
42
Thomas
DA
Sarris
AH
Cortes
J
et al
Phase II study of sphingosomal vincristine in patients with recurrent or refractory adult acute lymphocytic leukemia.
Cancer
2006
106
120
127
43
Jabbour
E
O'Brien
S
Kantarjian
H
et al
Neurologic complications associated with intrathecal liposomal cytarabine given prophylactically in combination with high-dose methotrexate and cytarabine to patients with acute lymphocytic leukemia.
Blood
2007
109
3214
3218
44
Jacobs
AD
Gale
RP
Recent advances in the biology and treatment of acute lymphoblastic leukemia in adults.
N Engl J Med
1984
311
1219
1231
45
Zhang
M
Hoelzer
D
Horowitz
M
et al
Long-term follow-up of adults with acute lymphoblastic leukemia in first remission treated with chemotherapy or bone marrow transplantation.
Ann Intern Med
1995
123
428
431
46
Weiden
PL
Flournoy
N
Thomas
ED
et al
Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts.
N Engl J Med
1979
300
1068
1073
47
Sullivan
KM
Weiden
PL
Storb
R
et al
Influence of acute and chronic graft-versus-host disease on relapse and survival after bone marrow transplantation from HLA-identical siblings as treatment of acute and chronic leukemia.
Blood
1989
73
1720
1728
48
Blume
KG
Forman
SJ
Snyder
DS
et al
Allogeneic bone marrow transplantation for acute lymphoblastic leukemia during first complete remission.
Transplantation
1987
43
389
392
49
Sebban
C
Lepage
E
Vernant
JP
et al
Allogeneic bone marrow transplantation in adult acute lymphoblastic leukemia in first complete remission: a comparative study French Group of Therapy of Adult Acute Lymphoblastic Leukemia.
J Clin Oncol
1994
12
2580
2587
50
Attal
M
Blaise
D
Marit
G
et al
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.
BGMT Group Blood
1995
86
1619
1628
51
Rowe
JM
Buck
G
Fielding
A
et al
In adults with standard-risk acute lymphoblastic leukemia (ALL) the greatest benefit is achieved from an allogeneic transplant in first complete remission (CR) and an autologous transplant is less effective than conventional consolidation/maintenance chemotherapy: final results of the International ALL Trial (MRC UKALL XII/ECOG E2993 [abstract].
Blood
2006
108
5a
Abstract 2
52
Collins
RH
Jr
Shpilberg
O
Drobyski
WR
et al
Donor leukocyte infusions in 140 patients with relapsed malignancy after allogeneic bone marrow transplantation.
J Clin Oncol
1997
15
433
444
53
Hoelzer
D
Gokbuget
N
New approaches to acute lymphoblastic leukemia in adults: where do we go.
Semin Oncol
2000
27
540
559
54
Mortuza
FY
Papaioannou
M
Moreira
IM
et al
Minimal residual disease tests provide an independent predictor of clinical outcome in adult acute lymphoblastic leukemia.
J Clin Oncol
2002
20
1094
1104
55
Bruggemann
M
Raff
T
Flohr
T
et al
Clinical significance of minimal residual disease quantification in adult patients with standard-risk acute lymphoblastic leukemia.
Blood
2006
107
1116
1123
56
Szczepanski
T
Why and how to quantify minimal residual disease in acute lymphoblastic leukemia.
Leukemia
2007
21
622
626
57
Stock
W
Sather
H
Dodge
RK
et al
Outcome of adolescents and young adults with ALL: a comparison of Children's Cancer Group (CCG) and Cancer and Leukemia Group B (CALGB) regimens [abstract].
Blood
2000
96
467a
58
Schiffer
CA
Differences in outcome in adolescents with acute lymphoblastic leukemia: a consequence of better regimens? Better doctors? Both?
J Clin Oncol
2003
21
760
761
59
Boissel
N
Auclerc
MF
Lheritier
V
et al
Should adolescents with acute lymphoblastic leukemia be treated as old children or young adults? Comparison of the French FRALLE-93 and LALA-94 trials.
J Clin Oncol
2003
21
774
780
60
de Bont
JM
Holt
B
Dekker
AW
et al
Significant difference in outcome for adolescents with acute lymphoblastic leukemia treated on pediatric vs adult protocols in the Netherlands.
Leukemia
2004
18
2032
2035
61
Testi
AM
Valsecchi
MG
Conter
V
et al
Difference in the outcome of adolescents with acute lymphoblastic leukemia (ALL) enrolled in pediatric (AIEOP) and adult (GIMEMA) protocols [abstract].
Blood
2004
104
539a
Abstract 1954
62
Bleyer
A
The adolescent and young adult gap in cancer care and outcome.
Curr Probl Pediatr Adolesc Health Care
2005
35
182
217
63
Wetzler
M
Dodge
RK
Mrozek
K
et al
Prospective karyotype analysis in adult acute lymphoblastic leukemia: the Cancer and Leukemia Group B experience.
Blood
1999
93
3983
3993
64
Mancini
M
Scappaticci
D
Cimino
G
et al
A comprehensive genetic classification of adult acute lymphoblastic leukemia (ALL): analysis of the GIMEMA 0496 protocol.
Blood
2005
105
3434
3441
65
Moorman
AV
Harrison
CJ
Buck
GA
et al
Karyotype is an independent prognostic factor in adult acute lymphoblastic leukemia (ALL): analysis of cytogenetic data from patients treated on the Medical Research Council (MRC) UKALLXII/Eastern Cooperative Oncology Group (ECOG) 2993 Trial.
Blood
2007
109
3189
3197
66
Barry
E
DeAngelo
DJ
Neuberg
D
et al
Favorable outcome for adolescents with acute lymphoblastic leukemia treated on Dana-Farber Cancer Institute Acute Lymphoblastic Leukemia Consortium Protocols.
J Clin Oncol
2007
25
813
819
67
Boucheix
C
David
B
Sebban
C
et al
Immunophenotype of adult acute lymphoblastic leukemia, clinical, parameters, and outcome: an analysis of a prospective trial including 562 tested patients (LALA87). French Group on Therapy for Adult Acute Lymphoblastic Leukemia.
Blood
1994
84
1603
1612
68
Chan
SM
Weng
AP
Tibshirani
R
Aster
JC
Utz
PJ
Notch signals positively regulate activity of the mTOR pathway in T-cell acute lymphoblastic leukemia.
Blood
2007
110
278
286
69
Stein
A
O'Donnell
M
Snyder
D
et al
Reduced intensity stem cell transplantation for high-risk acute lymphoblastic leukemia [abstract].
Biol Blood Marrow Transplant
2007
134
suppl 2
134a
70
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
2002
99
1071
1078
71
Delgado
J
Thomson
K
Russell
N
et al
Results of alemtuzumab-based reduced-intensity allogeneic transplantation for chronic lymphocytic leukemia: a British Society of Blood and Marrow Transplantation Study.
Blood
2006
107
1724
1730
72
Goldstone
AH
Chopra
R
Buck
G
et al
The outcome of 267 Philadelphia-positive adults in the International UKALL XII/ECOG E2993 study Final analysis and the role of allogeneic transplant in those under 50 years [abstract].
Blood
2003
104
268a
73
Garderet
L
Labopin
M
Gorin
NC
et al
Patients with acute lymphoblastic leukaemia allografted with a matched unrelated donor may have a lower survival with a peripheral blood stem cell graft compared to bone marrow.
Bone Marrow Transplant
2003
31
23
29
74
Kiehl
MG
Kraut
L
Schwerdtfeger
R
et al
Outcome of allogeneic hematopoietic stem-cell transplantation in adult patients with acute lymphoblastic leukemia: no difference in related compared with unrelated transplant in first complete remission.
J Clin Oncol
2004
22
2816
2825
75
Dahlke
J
Kroger
N
Zabelina
T
et al
Comparable results in patients with acute lymphoblastic leukemia after related and unrelated stem cell transplantation Bone Marrow.
Transplant
2006
37
155
163
76
Marks
DI
Aversa
F
Lazarus
HM
Alternative donor transplants for adult acute lymphoblastic leukaemia: a comparison of the three major options.
Bone Marrow Transplant
2006
38
467
475
77
Aversa
F
Terenzi
A
Tabilio
A
et al
Full haplotype-mismatched hematopoietic stem-cell transplantation: a phase II study in patients with acute leukemia at high risk of relapse.
J Clin Oncol
2005
23
3447
3454
78
Avivi
I
Goldstone
AH
Bone marrow transplant in Ph+ ALL patients.
Bone Marrow Transplant
2003
31
623
632
79
Ottmann
OG
Wassmann
B
Treatment of Philadelphia-chromosome-positive acute lymphoblastic leukemia.
Hematology Am Soc Hematol Educ Program
2005
118
122
80
de Labarthe
A
Rousselot
P
Huguet-Rigal
F
et al
Imatinib combined with induction or consolidation chemotherapy in patients with de novo Philadelphia chromosome-positive acute lymphoblastic leukemia: results of the GRAAPH-2003 study.
Blood
2007
109
1408
1413
81
Yanada
M
Takeuchi
J
Sugiura
I
et al
High complete remission rate and promising outcome by combination of imatinib and chemotherapy for newly diagnosed BCR-ABL-positive acute lymphoblastic leukemia: a phase II study by the Japan Adult Leukemia Study Group.
J Clin Oncol
2006
24
460
466
82
National Cancer Institute, Surveillance, Epidemiology and End Results Program.
2004
Accessed August 12, 2007
83
Secker-Walker
LM
Prentice
HG
Durrant
J
et al
Cytogenetics adds independent prognostic information in adults with acute lymphoblastic leukaemia on MRC trial UKALL XA MRC Adult Leukaemia Working Party.
Br J Haematol
1997
96
601
610
84
Thomas
X
Olteanu
N
Charrin
C
et al
Acute lymphoblastic leukemia in the elderly: the Edouard Herriot Hospital experience.
Am J Hematol
2001
67
73
83
85
Appelbaum
FR
Perry
MC
Impact of age on the biology of acute leukemia.
American Society of Clinical Oncology Educational Book
2005
Alexandria, VA
American Society of Clinical Oncology
528
532
86
Gokbuget
N
de Witt
M
Gerhardt
A
et al
Results of a shortened, dose-reduced treatment protocol in elderly patients with acute lymphoblastic leukemia [abstract].
Blood
2000
96
718a
87
Hoelzer
D
Gokbuget
N
Beck
J
et al
Subtype adjusted therapy improves outcome of elderly patients with acute lymphoblastic leukemia (ALL) [abstract].
Blood
2004
104
2732a
Abstract 2732
88
Pagano
M
Mele
L
Trape
G
et al
Treatment of acute lymphoblastic leukemia in the elderly.
Leuk Lymph
2004
45
117
123
89
Larson
RA
Acute lymphoblastic leukemia: older patients and newer drugs.
Hematology Am Soc Hematol Educ Program
2005
131
136
90
Hoelzer
D
Gokbuget
N
Perry
MC
Treatment of elderly patients with acute lymphoblastic leukemia.
American Society of Clinical Oncology Educational Book
2005
Alexandria, VA
American Society of Clinical Oncology
533
539
91
Frankfurt
O
Tallman
MS
Growth factors in leukemia.
J Natl Compr Canc Netw
2007
5
203
215
92
Fiere
D
Lepage
E
Sebban
C
et al
Adult acute lymphoblastic leukemia: a multicentric randomized trial testing bone marrow transplantation as postremission therapy. The French Group on Therapy for Adult Acute Lymphoblastic Leukemia.
J Clin Oncol
1993
11
1990
2001
93
Cortes
J
O'Brien
SM
Pierce
S
et al
The value of high-dose systemic chemotherapy and intrathecal therapy for central nervous system prophylaxis in different risk groups of adult acute lymphoblastic leukemia.
Blood
1995
86
2091
2097
94
Kantarjian
HM
O'Brien
S
Smith
TL
et al
Results of treatment with hyper-CVAD, a dose-intensive regimen, in adult acute lymphocytic leukemia.
J Clin Oncol
2000
18
547
561
95
Petersdorf
SH
Kopecky
KJ
Head
DR
et al
Comparison of the L10M consolidation regimen to an alternative regimen including escalating methotrexate/L-asparaginase for adult acute lymphoblastic leukemia: a Southwest Oncology Group Study.
Leukemia
2001
15
208
216
96
Thomas
X
Boiron
JM
Huguet
F
et al
Outcome of treatment in adults with acute lymphoblastic leukemia: analysis of the LALA-94 trial.
J Clin Oncol
2004
22
4075
4086
97
Lazarus
HM
Richards
SM
Chopra
R
et al
Central nervous system involvement in adult acute lymphoblastic leukemia at diagnosis: results from the international ALL trial MRC UKALL XII/ECOG E2993.
Blood
2006
108
465
472
98
Thiebaut
A
Vernant
JP
Degos
L
et al
Adult acute lymphocytic leukemia study testing chemotherapy and autologous and allogeneic transplantation. A follow-up report of the French protocol LALA 87.
Hematol Oncol Clin North Am
2000
14
1353
1366
99
Koller
CA
Kantarjian
HM
Thomas
D
et al
The hyper-CVAD regimen improves outcome in relapsed acute lymphoblastic leukemia.
Leukemia
1997
11
2039
2044
100
Giona
F
Annino
L
Rondelli
R
et al
Treatment of adults with acute lymphoblastic leukaemia in first bone marrow relapse: results of the ALL R-87 protocol.
Br J Haematol
1997
97
896
903
101
Montillo
M
Tedeschi
A
Centurioni
R
Leoni
P
Treatment of relapsed adult acute lymphoblastic leukemia with fludarabine and cytosine arabinoside followed by granulocyte colony-stimulating factor (FLAG-GCSF).
Leuk Lymphoma
1997
25
579
583
102
Garcia-Manero
G
Thomas
DA
Salvage therapy for refractory or relapsed acute lymphocytic leukemia.
Hematol Oncol Clin North Am
2001
15
163
205
Sign in via your Institution