T-cell acute lymphoblastic leukemia (ALL) is a rare disease in adults with inferior survival outcomes compared with those seen in pediatric patients. Although potentially curable with ∼50% survival at 5 years, adult patients with relapsed disease have dismal outcomes with <10% of patients surviving long term. This review will discuss the diagnosis and management of adult patients with newly diagnosed T-cell ALL with an emphasis on the immunophenotypic and genetic analyses required to assign prognosis, risk stratify, and guide post-remission therapy. The evidence for the main components of complex T-cell ALL treatment regimens is described. The importance of monitoring minimal residual disease is emphasized, with a discussion of the different methods used. The results of hematopoietic cell transplantation are analyzed, and recommendations made about which patients should be considered for this intervention. The treatment of the adolescent and young adult group is delineated, and the role of using “pediatric-inspired” regimens in older adults considered. We also describe the current data and potential future options for the use of novel therapies, including nelarabine and γ-secretase inhibitors, in adult patients with T-cell ALL.

T-cell lymphoblastic disease is an uncommon disorder in adults. Surveillance, Epidemiology, and End Results Program registry data shows an overall incidence of acute lymphoblastic leukemia (ALL) at 17.3 per million with T-cell ALL comprising ∼25% of all ALL in adults.1  The World Health Organization defines T-cell ALL and T-cell lymphoblastic lymphoma (LBL) as the same disease.2  T-cell ALL is used when there is extensive bone marrow involvement, whereas T-cell LBL is preferred when there is primarily a mass lesion with <20% to 25% blasts in the marrow. Management of T-LBL will not be discussed specifically in this review. T-ALL patients frequently present with a high tumor burden with hyperleukocytosis and large mediastinal masses. Although T-cell ALL is a highly aggressive disease, it is potentially curable in adults with superior 5-year overall survival (OS) rates compared with B-cell ALL (48% vs 41%).1,3,4 

Diagnosis of T-cell ALL requires a combination of morphology, immunophenotyping, and cytogenetic analysis, the results of which have important prognostic implications for patient management.

The immunophenotype of T-cell ALL corresponds to the stage of intrathymic T-cell differentiation and provides the basis for the European Group for the Immunological Characterization of Leukaemias subclassification of T-cell ALL (Table 1).5  Lymphoblasts in T-cell ALL are TdT+ in addition to cytoplasmic CD3+, the only lineage specific marker for lymphoblastic T-cell disease. Other T-cell markers including CD1a, CD2, CD4, CD5, CD7, and CD8 are variably expressed according to the degree of T-cell differentiation. Several study groups have shown a relationship between prognosis and the degree of T-cell differentiation at diagnosis. The Gruppo Italiano Malattie Ematologiche dell’Adulto reported a 91% complete remission (CR) rate in patients with cortical/mature disease as opposed to 56% in those with pre-/pro- T disease.6  The Medical Research Council/Eastern Cooperative Oncology groups performed central immunophenotypic studies of 108 patients uniformly treated on the United Kingdom ALL (UKALL) XII/E2993 trial.3  CD1a-negativity was associated with an increased relapse rate and a lower survival (39% vs 64%; P = .01). Patients with cortical T-cell ALL in the same study had the most favorable prognosis with 5-year OS rates of 62%.4  Others have confirmed these findings,7,8  although Jain et al recently reported no difference in OS between T-cell immunologic subtypes in a total of 111 patients with T-cell ALL/LBL.9 

Table 1.

T-cell CD antigen expression in pro-, pre-, cortical (thymic), and mature T-cell ALL cyCD3 indicates cytoplasmic CD3; sCD3, surface CD3

cyCD3CD7CD5CD2CD1asCD3CD34
Pro-T — — — — ± 
Pre-T — — ± 
Cortical (thymic) ± ± ± — 
Mature ± — — 
cyCD3CD7CD5CD2CD1asCD3CD34
Pro-T — — — — ± 
Pre-T — — ± 
Cortical (thymic) ± ± ± — 
Mature ± — — 

Myeloid antigens are often expressed by T lymphoblasts. The presence of the myeloid antigen CD13 in 51% of cases analyzed was associated with inferior survival (P < .001) in the UKALL 12/E2993 trial.4  Expression of CD33 has also shown to be associated with inferior outcomes.7 

Early T-precursor ALL (ETP)

A novel subgroup of T-cell ALL, ETP-ALL, has recently been described and is characterized by a distinct gene expression profile and immunophenotype.10  ETP cells derive from a subset of immature thymocytes that retain the ability to differentiate into both T-cell and myeloid lineages, suggesting direct derivation from hematopoietic stem cells.11  ETP-ALL blasts are CD8 and CD1a, CD5weak, and express one or more myeloid or stem cell marker.10  The gene expression profile of ETP-ALL has strong similarities to that of normal and myeloid leukemic stem cells and is distinct from non–ETP-ALL. An additional entity of “near” ETP, based on CD5 and myeloid antigen expression, has also been defined.12  ETP-ALL accounts for ∼15% of all T-cell ALL in children10,13,14  and ∼35% in adult T-cell disease,9,15,16  and is associated with high levels of minimal residual disease (MRD) post-induction chemotherapy14  and inferior long-term outcomes.7,9,10,13  However, data from the UKALL 2003 pediatric and young adult ALL trial suggested that these patients have an intermediate-risk outcome when treated on a contemporary protocol.17  Similar results have been reported by the Children’s Oncology Group.14  ETP-ALL has recently been shown to be dependent on aberrant JAK-STAT signaling,18  and data from the Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL) imply that the poor outcome is related to HOXA expression.19 

Cytogenetics

Cytogenetic analysis in B-cell ALL has become an important part of the diagnosis and planning of treatment strategy in both pediatric and adult practice. Although there are an increasing number of molecular abnormalities associated with T-cell ALL, which may represent important targets for future novel therapies, few are currently required for routine diagnosis and treatment planning (Table 2).

Table 2.

The diagnostic work-up of adults with T-cell ALL: essential tests and tests that are currently research based

Essential tests required for routine diagnosis and management of T-cell ALLNon-essential tests currently being investigated within clinical trials in T-cell ALL
Immunophenotyping  
 Determination of pre-T/pro-T, cortical, and mature T-cell phenotype  Determination of ETP subtype 
Oncogenetic analysis  
 Identification of patients with a
complex karyotype 
 NOTCH1/FBXW7 mutations 
  PTEN/NK-RAS mutations 
  JAK/STAT mutations 
  NUP21-ABL1 fusion 
  IL-7 receptor mutations 
Molecular assessment for identification of T-cell receptor rearrangement for MRD monitoring  
Essential tests required for routine diagnosis and management of T-cell ALLNon-essential tests currently being investigated within clinical trials in T-cell ALL
Immunophenotyping  
 Determination of pre-T/pro-T, cortical, and mature T-cell phenotype  Determination of ETP subtype 
Oncogenetic analysis  
 Identification of patients with a
complex karyotype 
 NOTCH1/FBXW7 mutations 
  PTEN/NK-RAS mutations 
  JAK/STAT mutations 
  NUP21-ABL1 fusion 
  IL-7 receptor mutations 
Molecular assessment for identification of T-cell receptor rearrangement for MRD monitoring  

IL-7, interleukin 7.

The genetic lesions underlying T-cell ALL will be discussed in detail elsewhere in this series. Therefore, we will briefly discuss only the oncogenetic abnormalities that are either required for standard practice or are currently being evaluated within treatment algorithms in the context of clinical studies (Tables 2 and 3).

Table 3.

Selected current trials in T-cell ALL in adolescent and adult patients

Trial group/trialDescription of studyPopulationPrimary end pointTrial #
UKALL 14 (UK NCRI) Phase 3 nelarabine in induction randomization Adults aged 25-65 y with de novo ALL EFS NCT01085617 
UKALL 2011 (UK NCRI) Phase 3 randomized comparison of dexamethasone and methotrexate schedules Children and young adults up to 25 y with de novo ALL and LBL Steroid-related toxicity; CNS and marrow relapse rate ISRCTN64515327 
MD Anderson Phase 2 nelarabine/HyperCVAD Children and adults CR NCT00501826 
US Intergroup Pediatric-like regimen 21-40 y Survival NCT00558519 
DFCI LY3039478 (notch inhibitor) Relapsed/refractory disease; 2 y and older Overall remission rate NCT02518113 
GRAALL Pediatric-like regimen Adults (median age, 31 y) EFS NCT00327678 
NCI Phase 3, bortezomib randomization 18 y and older EFS NCT02112916 
GIMEMA Phase 2, lineage-targeted methotrexate dosing 18-65 y EFS NCT02067143 
Trial group/trialDescription of studyPopulationPrimary end pointTrial #
UKALL 14 (UK NCRI) Phase 3 nelarabine in induction randomization Adults aged 25-65 y with de novo ALL EFS NCT01085617 
UKALL 2011 (UK NCRI) Phase 3 randomized comparison of dexamethasone and methotrexate schedules Children and young adults up to 25 y with de novo ALL and LBL Steroid-related toxicity; CNS and marrow relapse rate ISRCTN64515327 
MD Anderson Phase 2 nelarabine/HyperCVAD Children and adults CR NCT00501826 
US Intergroup Pediatric-like regimen 21-40 y Survival NCT00558519 
DFCI LY3039478 (notch inhibitor) Relapsed/refractory disease; 2 y and older Overall remission rate NCT02518113 
GRAALL Pediatric-like regimen Adults (median age, 31 y) EFS NCT00327678 
NCI Phase 3, bortezomib randomization 18 y and older EFS NCT02112916 
GIMEMA Phase 2, lineage-targeted methotrexate dosing 18-65 y EFS NCT02067143 

DFCI, Dana-Farber Cancer Institute; GIMEMA, Gruppo Italiano Malattie Ematologiche dell’Adulto; NCI, National Cancer Institute; UK NCRI, United Kingdom National Cancer Research Institute.

Cytogenetic analysis of 303 of 356 patients with T-cell ALL treated on UKALL XII/E2993 was successful in 204 patients (67%) and demonstrated an abnormal karyotype in 72% of patients. The 8% of patients with a complex karyotype (≥5 abnormalities) had significantly inferior outcomes with OS 19% vs 51% at 5 years (P = .006) (Table 2). The 10 patients with 17p abnormalities had 20% survival (confidence interval, 0% to 45%) but this was not statistically different to the remaining T-cell ALL cases.4 

At least 60% of adult patients with T-cell ALL will have a mutation of the NOTCH1/FBXW7 (N/F) pathway at diagnosis.20  The majority of studies that have examined the prognostic impact of N/F pathway mutations have concluded that they are associated with a good response to therapy and improved survival.21-28  However, not all studies report statistically significant results and there is clear heterogeneity in survival rates when comparing similar protocols. It is unclear whether these differences are driven by variable response to therapy, or simply because these mutations are secondary events and few studies have comprehensively factored in the effect of other genetic drivers. One exception is the study by Trinquad et al,29  who elegantly proposed an oncogenetic classifier for 212 adult patients with T-cell ALL based on the presence or absence of mutations in the N/F pathway, as well as the phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and natural killer (NK)-RAS pathways. They described a good-risk group with significantly superior OS and cumulative incidence of relapse (CIR) rates (82% OS and 15% CIR) for patients with mutations in the N/F pathway with no associated mutations in PTEN or NK-RAS. All other patients, including those with mutated N/F genes plus mutations in PTEN or NK-RAS were classified as poor risk with OS 44% and CIR 54%. When combined with MRD assessment post-induction on a pediatric-inspired protocol, investigators identified a good-risk group (MRD <10−4 and good-risk genetic profile) with a 5-year OS of 91%.30  However, it should be noted that an inferior outcome was not reported for T-cell ALL patients with PTEN or RAS pathway mutations treated on UKALL 2003.27  These inconsistent results emphasize the need for a study, which comprehensively assesses all prognostic factors: demographic (age, sex), tumor burden (white cell count [WCC]), early response (MRD), as well as the full spectrum of genetic lesions. The latter must include both primary class defining abnormalities (eg, TAL1, LMO2, TLX1, TLX3, NKX2-1, and MEF2C and those driving HOXA expression), as well as key secondary lesions (eg, mutations and deletions of NOTCH1, FBWX7, NRAS, KRAS, PTEN, TP53, and CDKN2A/B, etc).

Induction therapy

The goal of induction therapy in adult ALL is to achieve a deep remission with low treatment-related toxicity and rapid hematologic recovery to enable prompt progression to further therapy. Although induction regimens vary, most use anthracyclines (daunorubicin or doxorubicin), steroids (dexamethasone or prednisolone), and vincristine plus central nervous system (CNS)-directed prophylaxis with administration of intrathecal (IT) methotrexate. It is usual to include an intensification phase that contains cyclophosphamide and cytarabine.

Effective depletion of extracellular asparagine by administration of the enzyme l-asparaginase has long been an important component of the treatment of pediatric ALL, with T-cell ALL patients benefiting from intensified dosing schedules.31  In recent years, the use of pegylated Escherichia coli-derived (polyethylene glycol-asparaginase [PEG-ASP]) l-asparaginase has been shown to improve outcomes in children,32-36  offering a longer half-life and a lower risk of anti-asparaginase antibody formation. The majority of current adult induction regimens include l-asparaginase, although there is significant variability in how it is delivered. Cyclosphosphamide/vincristine/adriamycin/dexamethasone (HyperCVAD), most commonly used in the United States, contains no asparaginase and may be associated with inferior outcomes in patients with T-cell ALL in some series.37,38  In contrast, the “pediatric-inspired” regimens frequently used in adolescent and young adults (AYAs) contain up to 15 doses of PEG-ASP in the first 30 weeks of therapy.39  PEG-ASP used in combination with a multiagent regimen has been shown to effectively deplete asparagine in adult ALL,35,40  resulting in high CR rates and improved OS outcomes in patients with T-cell ALL, both compared with patients with B-cell ALL and patients treated with native enzyme.35  The German Multicentre Adult ALL (GMALL) study group report improved survival outcomes with intensified PEG-ASP scheduling in a large cohort of patients.41  Similarly, the Dana-Farber Cancer Institute reported excellent results with an intensified PEG-ASP regimen with 75% 3-year OS in T-cell ALL patients.42  Despite interest in intensifying regimens with additional doses of PEG-ASP in adults, there are clear data showing greater toxicities of asparaginase with increasing age, and the optimum dose for adults remains uncertain and may vary with age.43-46  There is a need to investigate the best methods of delivering l-asparaginase in adults with T-cell ALL within a large phase 3 trial.

Patients with T-cell ALL are more likely to have involvement of the CNS at presentation than patients with B-cell ALL (9.6% vs 4.4%; P = .001).47  Patients with CNS disease at diagnosis had inferior 5-year OS outcomes compared with patients without CNS involvement on the UKALL XII/E2993 trial (42% vs 29%) due to an increased risk of both systemic and CNS relapse.47  Pediatric trials have demonstrated improved event-free survival (EFS) outcomes for patients with T-cell ALL when high-dose IV methotrexate (4 doses of 5 g/m2) is added as an intensification phase.48  Although some groups have cited decreased CNS relapses as the primary reason for the improved outcomes,48  others have demonstrated improved systemic disease control with high-dose methotrexate (HDM) as the main benefit.49  Although there are limited data for HDM in the specific treatment of adult T-cell ALL, most ALL trial groups have adopted the use of HDM (doses range from 1-5 g/m2 in adult protocols) in addition to IT chemotherapy within their T-cell ALL protocols (Table 4).4,6,8,9,38,50-52  Combining HDM and PEG-ASP in adults with T-cell ALL is the subject of several ongoing trials (Table 3). The risk of CNS relapse in adult patients on modern regimens who have no evidence of CNS disease at presentation is ∼5%, with or without cranial radiotherapy.47,50,53 

Table 4.

Prospective clinical trials in adults with T-cell lymphoblastic disease

ReferenceNumber with T-cell diseaseDisease typeTreatment givenCR and OS rates
90 ALL LAL0496 protocol with CNS RT CR-NS, OS-NS 
52 87 ALL ALL 90, 93, and 97 protocols of JALSG; CNS RT for high WCC; alloHCT for almost half CR 75.8% 
OS 35% (5 y) 
744 ALL GMALL 05/93, 06/99, and 07/2003; No alloHCT in CR1 CR 86% 
OS 47% (10 y) 
50 76 ALL GRAALL 2003; CNS RT for all; alloHCT for high-risk patients CR 99% 
EFS 62% (42 mo) 
356 ALL UKALL XII/E2993; alloHCT for those with sibling donor; RCT for the rest chemo vs autograft CNS RT for chemo only arm CR 94% 
OS 48% (5 y) 
38 24 ALL HyperCVAD; alloHCT for high-risk patients (4%) CR 89% 
OS 47% (5 y) 
51 40 ALL/LBL HyperCVAD plus nelarabine CR 91% 
OS 63% (3 y) 
111 ALL/LBL HyperCVAD, HyperCVAD plus nelarabine, or augmented BFM CR 88% 
OS 52% (5 y) 
ReferenceNumber with T-cell diseaseDisease typeTreatment givenCR and OS rates
90 ALL LAL0496 protocol with CNS RT CR-NS, OS-NS 
52 87 ALL ALL 90, 93, and 97 protocols of JALSG; CNS RT for high WCC; alloHCT for almost half CR 75.8% 
OS 35% (5 y) 
744 ALL GMALL 05/93, 06/99, and 07/2003; No alloHCT in CR1 CR 86% 
OS 47% (10 y) 
50 76 ALL GRAALL 2003; CNS RT for all; alloHCT for high-risk patients CR 99% 
EFS 62% (42 mo) 
356 ALL UKALL XII/E2993; alloHCT for those with sibling donor; RCT for the rest chemo vs autograft CNS RT for chemo only arm CR 94% 
OS 48% (5 y) 
38 24 ALL HyperCVAD; alloHCT for high-risk patients (4%) CR 89% 
OS 47% (5 y) 
51 40 ALL/LBL HyperCVAD plus nelarabine CR 91% 
OS 63% (3 y) 
111 ALL/LBL HyperCVAD, HyperCVAD plus nelarabine, or augmented BFM CR 88% 
OS 52% (5 y) 

BFM, Berlin Frankfurt Munster protocol; CNS RT, CNS radiotherapy; JALSG, Japan Adult Leukemia Study Group; NS, not significant; RCT, randomized controlled trial.

There is a paucity of data concerning the best consolidation therapy in adults for T-cell ALL and much current practice is based on pediatric trials. Most regimens use a delayed intensification block of therapy, repeating drugs used in induction, followed by a 2-year period of maintenance with 6-mercaptopurine and methotrexate, pulses of vincristine and steroids, plus additional CNS prophylaxis with IT chemotherapy (Table 4). Although maintenance is not required post-allogeneic hematopoietic cell transplantation (alloHCT), patients receiving a reduced intensity alloHCT (and less CNS-directed therapy) should have HDM prior to transplant and consideration of IT CNS prophylaxis post-HCT in order to minimize their risk of CNS relapse, although the value of this strategy is not proven.54 

Monitoring response to treatment by MRD assessment should be a standard of care in all patients with ALL treated with curative intent. Standardized methods for molecular assessment of MRD identify rearrangements of the immunoglobulin heavy chain gene or T-cell receptor genes by using a large panel of consensus primers spanning the junctional regions of V, (D), and J gene segments. The random insertion and deletion of nucleotides into those segments generates clone-specific rearrangements, which can be sequenced to allow the generation of patient-specific real-time quantitative polymerase chain reaction assays for quantification in ∼90% of patients, with a quantitative range of 10−4. This method has an international standard and is quality-controlled within the European Study Group on MRD. Flow cytometry-based quantification of a “leukemia-associated immunophenotype” is also widely used. This technique has a lower sensitivity (10−3) and is less well standardized due to the wide range of approaches used, although the Euroflow consortium is working toward establishing an international standard.55  Next-generation sequencing can also be used to identify and quantify the same clone-specific re-arrangements and this forms the basis of a commercial Food and Drug Administration-approved test.

Many studies have confirmed early MRD response as a powerful predictor of long-term survival in adult patients with ALL.56-59  GMALL examined MRD at 9 time points in adults receiving GMALL 09/99 ALL therapy and showed that, using their treatment protocols, molecular MRD quantification after 2 courses of therapy (10 to 16 weeks from diagnosis) was the most predictive of outcomes with patients failing to achieve molecular remission (MRD >10−4) post-induction having inferior survival compared with patients who were MRD-negative (MRD <10−4) (42% vs 80%; P < .001).57  Seventy-two of the 196 patients (37%) within this study had T-cell disease.

The GRAALL analyzed the prognostic significance of MRD at 6 weeks from the initiation of induction therapy on the pediatric-inspired GRAALL 2003 and 2005 trials in 163 patients with T-cell ALL (total patients studied = 423). They defined a high-risk group as those with an MRD level ≥10−4 and unfavorable cytogenetics (no N/F mutation and/or N/K-RAS mutation and/or PTEN mutation).30  The 59% of patients in this high-risk group had markedly inferior OS compared with the good-risk group (62% vs 91% at 5 years).

Although it is reasonable to recommend that T-cell ALL patients with persistent MRD should be candidates for an alloHCT in first CR (CR1),60  there are currently no data to show that intervention with novel agents or allogeneic transplantation can overcome the relapse risk and this is currently the focus of many clinical trials (Table 3).

Allogeneic transplantation

alloHCT is the most effective but also the most toxic anti-leukemic therapy for adults with ALL.61  Traditionally, myeloablative alloHCT, with a wide variety of conditioning regimens, has been used in the treatment of ALL. Etoposide and total body irradiation (13.2 Gy in 8 fractions) were used in UKALL XII/E2993. This trial assessed the role of a sibling donor alloHCT by a comparison of outcomes in those with vs those without a sibling donor (Figure 1).4  Two-hundred and fifty-three of 356 T-cell ALL patients were tissue typed; 110 had a sibling donor. OS at 5 years was 46% (38% to 55%) for the no donor group and 61% (51% to 70%) for the donor group (log rank P = .07, χ2 test of difference at 5 years; P = .02); this difference was maintained at 10 years (Figure 1). Having a sibling donor halved the chance of relapse (25% vs 51%; P < .0001) but modestly increased nonrelapse mortality (22% vs 12%; P = .06). The Passweg study showed that acute and/or chronic graft-versus-host disease reduced relapse 2.5-fold (Hazard ratio, 0.4) and that the graft-versus-leukemia effect was similar in patients with T- or B-cell disease.62  These results suggest that myeloablative alloHCT is an effective therapy in adults and can be considered for patients with high-risk T-cell disease (Table 5).4,9,10,17,29,30,44,60,63-65 

Figure 1.

Comparison of OS in patients with T-cell ALL who had a matched sibling donor vs those without a donor within the UKALL XII/E2993 trial.4

Figure 1.

Comparison of OS in patients with T-cell ALL who had a matched sibling donor vs those without a donor within the UKALL XII/E2993 trial.4

Close modal
Table 5.

Patients with T-cell ALL to be considered for alloHCT in first remission

Indication*Reference
Failure to achieve CR after induction 60 
MRD >1 × 10−4 after 2 courses of therapy 30, 60 
Presenting WCC >100 × 109/L 
Complex cytogenetics ≥5 abnormalities 4, 30 
Early T-cell precursor ALL 9, 10, 17 
Unfavorable N/F/PTEN/RAS genetics 29 
Age 40-65 y, especially if delays occur in treatment due to toxicity 64 
Indication*Reference
Failure to achieve CR after induction 60 
MRD >1 × 10−4 after 2 courses of therapy 30, 60 
Presenting WCC >100 × 109/L 
Complex cytogenetics ≥5 abnormalities 4, 30 
Early T-cell precursor ALL 9, 10, 17 
Unfavorable N/F/PTEN/RAS genetics 29 
Age 40-65 y, especially if delays occur in treatment due to toxicity 64 

TRM, treatment-related mortality.

*

Donor quality and the Hematopoietic Cell Transplantation Comorbidity index also affect the decision.65  alloHCT may reduce relapse risk but this should be balanced against the predicted TRM.

MRD negativity may override these unfavorable prognostic factors.

Total body irradiation-based conditioning, although effective in eradicating leukemic cells, causes considerable extramedullary toxicity.61  For this reason, the UK NCRI group has been prospectively studying reduced-intensity conditioning sibling and unrelated donor allografting in older patients (>40 years), in an attempt to reduce TRM but retain the graft-versus-leukemia effect. With 22 months median follow up, 120 patients (15% T cell, median age 51 years, 54% high-risk cytogenetics) had conditioning with fludarabine, melphalan, and alemtuzumab, and experienced 66% survival at 22 months with 18% TRM, suggesting that reduced-intensity conditioning allograft is a valid option for patients >40 years.64  The low TRM was partly due to a low incidence of grade 2-4 graft-versus-host disease but the 20% relapse rate so far may increase. The number of T-cell patients was insufficient for them to be analyzed separately. Patients with MRD >10−4 at alloHCT had a lower EFS (Hazard ratio 2.75; P = .047) compared with those that were MRD-negative ≤10−4). This patient subgroup may require additional pre-transplant therapy in an attempt to achieve MRD-negativity before transplant. TRM will also be affected by the source of stem cells; recent data shows that cord blood produces equivalent survival to unrelated donor HCT.66  There are no mature results of haploidentical HCT in adult ALL; this requires prospective study. Where possible, all patients with T-cell ALL should be treated within a prospective clinical trial. However, a list of potential indications for alloHCT in first CR1 is presented in Table 5.

Autologous stem cell transplantation

UKALL XII/E2993, the only randomized trial to date comparing autograft with standard chemotherapy, showed an 8% survival advantage for 2 years of maintenance chemotherapy over an autograft.61  Ninety-nine of the patients randomized to autograft vs chemotherapy in this study had T-cell ALL. Survival was the same in both arms (Figure 2). Although the study was not powered to analyze the T-cell subset separately, a test of heterogeneity did not suggest patients with T-cell disease should be treated differently to those with B-cell disease (P = .2).4  Further analysis suggested that patients with MRD-negativity (<10−4) prior to autograft have better outcomes than those with persistent MRD, but there is no evidence that autograft is superior to standard chemotherapy in any group of patients with ALL.67 

Figure 2.

Comparison of OS in patients with T-cell ALL treated with autologous stem cell transplantation (auto) or chemotherapy (chemo) within the UKALL XII/E2993 trial.4

Figure 2.

Comparison of OS in patients with T-cell ALL treated with autologous stem cell transplantation (auto) or chemotherapy (chemo) within the UKALL XII/E2993 trial.4

Close modal

In a Russian study, 28 of 77 (36%) patients with T-cell ALL had a BEAM (BCNU, etoposide, cytarabine, and melphalan) autograft in CR1. Although a selected group, 100% of autografted patients survived and none have relapsed with a 27-month median follow up.68 

The incidence of T-cell ALL rises throughout childhood and adolescence (5% of all cases of ALL are <5 years and 27.6% >16 years; P ≤ .0001).69  It is widely accepted that AYA patients with ALL have improved outcomes when treated on pediatric protocols with intensified scheduling of l-asparaginase without the routine use of HCT.50,70-77  Hough et al69  reported OS outcomes of 72% at 5 years for AYA patients aged 16 to 25 years treated on a risk-adapted pediatric protocol where the strongest predictor for survival was immunoglobulin heavy chain gene or T-cell receptor-defined MRD status after induction. Patients with low-risk MRD (<0.01%) had a 93% EFS at 5 years compared with 64% EFS at 5 years for those with high-risk MRD (at least 0.01%). Over 75% of AYA patients with T-cell ALL had high risk or indeterminate MRD post-induction in this study.

Although there is broad agreement that young adults with ALL should be treated on intensive “pediatric-inspired” protocols, there is a lack of clarity on how we define an AYA. In the United Kingdom, anyone up to 25 years of age is considered a young adult, whereas other groups use a cutoff up to 40 years.78  In addition, the definition of what constitutes a “pediatric-inspired” protocol is not clear in the modern era where adult protocols are also based on an MRD risk-based stratification with intensified use of steroids, asparaginase, and other non-myelotoxic agents such as vincristine. Pediatric protocols rarely employ alloHCT in CR1 and yet we know that alloHCT is the most active anti-ALL therapy.61  A subgroup of AYA patients with T-cell ALL are likely to benefit from alloHCT in CR1, but we currently have no risk stratification models for use of alloHCT in frontline therapy in this group. In the United Kingdom, AYA patients with T-cell ALL treated on the NCRI UKALL 2011 trial (ISRCTN64515327) are risk-stratified on MRD alone. Those with very high-risk MRD (>10−1 at day 29 or 5 × 10−3 at week 14) are assigned nelarabine plus multiagent chemotherapy,79  followed by HCT. The current GRAALL study (NCT00327678) stratifies AYA ALL patients for alloHCT based on their oncogenetic classifier.30  The results of these trials are awaited with interest (Table 3). Finally, the toxicity of intensive “pediatric-inspired” protocols with increasing patient age remains a significant cause of morbidity and mortality,69  and needs careful evaluation within the context of large phase 3 studies.

The overall outcome of relapsed ALL in adults is poor with <7% of patients surviving at 5 years.80  There is no agreed standard of care in adults with relapsed T-cell ALL, but standard chemotherapy regimens such as FLAG (fludarabine, cytarabine, and granulocyte colony-stimulating factor) ± idarubicin only result in 30% to 40% response rates with 6 months median OS in responders.81 

Nelarabine

Nelarabine is the only new drug that is licensed specifically for use in relapsed/refractory T-cell ALL/LBL. Initial phase 1/2 pediatric trials reported response rates of 14% to 55% to single-agent nelarabine for patients with relapsed/refractory T-cell lymphoblastic disease, with neurotoxicity being the major toxicity affecting 18% of patients.82,83  Phase 2 trials in adults with relapsed/refractory T-cell ALL/LBL reported similarly encouraging response rates (41% to 46%) to single-agent nelarabine with 28% 1-year OS.84,85  Importantly, 80% of the 36% of patients achieving CR in the Gökbuget study were able to proceed to alloHCT, with 1-year OS for the whole cohort of 24% and 31% for patients reaching HCT. Although 16% of patients experienced some neurologic toxicity, grade 3-4 toxicities were only observed in 7% of patients (n = 9), and most were transient and reversible. The use of nelarabine in combination with other chemotherapeutic agents, particularly cyclophosphamide, has shown promising response rates in children with relapsed/refractory disease.79 

These encouraging results in relapsed disease have created an interest in the use of nelarabine in the initial (upfront) therapy of T-cell ALL. A pilot study from the Children’s Oncology Group added nelarabine to an intensified Berlin Frankfurt Munster protocol for children and young adults, initially reserving this therapy for slow early responders.86  Equivalent outcomes for the slow early responders who received nelarabine (73% with 5-year EFS) and the rapid early responders who did not receive nelarabine (69% with 5-year EFS; P = .64) were reported (Table 5). More recently, Jain et al51  reported the outcomes for 40 adult patients with T-cell ALL/LBL when treated in induction with nelarabine plus HyperCVAD. This single-arm phase 2 study reported a CR rate of 91% with disease-free survival at 3 years of 63% (Table 5). Reported toxicities were acceptable, with all neurologic toxicities being fully reversible (22% grades 2-4).

Novel agents in T-cell ALL

γ-Secretase is required for NOTCH1 signaling through mutant receptors in T-cell ALL and may be an attractive target for therapeutic intervention for the 60% of patients with T- cell ALL with a mutation in the NOTCH-1 signaling pathway.87,88  γ-Secretase inhibitor (GSI) drugs are in early development. A phase 1 study with the GSI BMS-906024 showed activity in 8 of 25 relapsed patients.89  Other studies are now ongoing.90 

The recently reported NUP214-ABL1 fusion gene, reported in 6% of adult T-cell ALL, results in a constitutively activated tyrosine kinase and confers an inferior prognosis.91  Tyrosine kinase inhibitors, including imatinib, have been reported to inhibit the resultant mutant tyrosine kinase in vitro, and offer an interesting and novel therapeutic option in the minority of T-cell ALL patients with this genetic abnormality.92 

Ruxolitinib shows promise in in vitro studies in T-cell ALL blasts and may have activity in the 30% of T-cell ALL patients with increased JAK-STAT signaling.18  Approximately 40% of T-cell ALL expresses CD30, making brentuximab an interesting therapeutic option in these cases for relapsed, refractory disease.93 

CD19 chimeric antigen receptor T-cell (CAR-T) therapy is achieving remarkable results in children and adults with relapsed/refractory pre-B ALL, but this therapeutic modality is not yet a reality for patients with T-cell ALL. There is shared expression of most targetable surface antigens between normal and malignant T cells leading to death of the CAR-T cells (fratricide mechanism) or profound immunodeficiency. However, Mamonkin et al reported targeting CD5 (which is expressed by normal and malignant T cells), and demonstrated activity against T-cell ALL in vivo and in murine xenograft models with only a limited degree of fratricide.94 

About half of adults with T-cell ALL who are intensively treated are currently cured. Further improvements are anticipated with intensification of protocols in younger adults, frontline use of newer agents such as nelarabine, judicious use of HCT, and possibly the use of novel agents and CAR-T cells in poor responders. As always, a better understanding of disease biology will enable the use of specific targeted therapy.

The authors thank Adele Fielding for critically reviewing the manuscript and making a number of important suggestions; Anthony Moorman for several helpful comments on the section on cytogenetics and genetics; and Jenny Bird for manuscript accuracy and consistency.

Contribution: D.I.M. and C.R. wrote, edited, and approved the final version of the manuscript.

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

Correspondence: David I. Marks, University Hospitals Bristol National Health Service Foundation Trust, Adult BMT Unit, Bristol BS2 8ED, United Kingdom; e-mail: david.marks@uhbristol.nhs.uk.

1.
Dores
GM
,
Devesa
SS
,
Curtis
RE
,
Linet
MS
,
Morton
LM
.
Acute leukemia incidence and patient survival among children and adults in the United States, 2001-2007
.
Blood
.
2012
;
119
(
1
):
34
-
43
.
2.
Swerdlow
SH
,
Campo
E
,
Harris
NL
, et al
.
WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues.
4th ed. Lyon, France: IARC Publications;
2008
.
3.
Rowe
JM
,
Buck
G
,
Burnett
AK
, et al
;
ECOG; MRC/NCRI Adult Leukemia Working Party
.
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
(
12
):
3760
-
3767
.
4.
Marks
DI
,
Paietta
EM
,
Moorman
AV
, et al
.
T-cell acute lymphoblastic leukemia in adults: clinical features, immunophenotype, cytogenetics, and outcome from the large randomized prospective trial (UKALL XII/ECOG 2993)
.
Blood
.
2009
;
114
(
25
):
5136
-
5145
.
5.
Bene
MC
,
Castoldi
G
,
Knapp
W
, et al
;
European Group for the Immunological Characterization of Leukemias (EGIL)
.
Proposals for the immunological classification of acute leukemias
.
Leukemia
.
1995
;
9
(
10
):
1783
-
1786
.
6.
Vitale
A
,
Guarini
A
,
Ariola
C
, et al
.
Adult T-cell acute lymphoblastic leukemia: biologic profile at presentation and correlation with response to induction treatment in patients enrolled in the GIMEMA LAL 0496 protocol
.
Blood
.
2006
;
107
(
2
):
473
-
479
.
7.
Van Vlierberghe
P
,
Ambesi-Impiombato
A
,
De Keersmaecker
K
, et al
.
Prognostic relevance of integrated genetic profiling in adult T-cell acute lymphoblastic leukemia
.
Blood
.
2013
;
122
(
1
):
74
-
82
.
8.
Hoelzer
D
,
Thiel
E
,
Arnold
R
, et al
.
Successful subtype oriented treatment strategies in adult T-All; results of 744 patients treated in three consecutive GMALL studies [abstract]
.
Blood
.
2009
;
114
(
22
).
Abstract 324
.
9.
Jain
N
,
Lamb
AV
,
O’Brien
S
, et al
.
Early T-cell precursor acute lymphoblastic leukemia/lymphoma (ETP-ALL/LBL) in adolescents and adults: a high-risk subtype
.
Blood
.
2016
;
127
(
15
):
1863
-
1869
.
10.
Coustan-Smith
E
,
Mullighan
CG
,
Onciu
M
, et al
.
Early T-cell precursor leukemia: a subtype of very high-risk acute lymphoblastic leukemia identified in two independant cohorts
.
Lancet Oncol
.
2009
;
10
(
2
):
147
-
156
.
11.
Zhang
J
,
Ding
L
,
Holmfeldt
L
, et al
.
The genetic basis of early T-cell precursor acute lymphoblastic leukaemia
.
Nature
.
2012
;
481
(
7380
):
157
-
163
.
12.
Haydu
JK
,
Ferrando
AA
.
Early T-cell precursor acute lymphbastic leukemia (ETP T-ALL)
.
Curr Opin Hematol
.
2013
;
20
(
4
):
9
-
16
.
13.
Ma
M
,
Wang
X
,
Tang
J
, et al
.
Early T-cell precursor leukemia: a subtype of high risk childhood acute lymphoblastic leukemia
.
Front Med
.
2012
;
6
(
4
):
416
-
420
.
14.
Wood
BL
,
Winter
SS
,
Dunsmore
KP
, et al
.
T-lymphoblastic leukemia (T-ALL) shows excellent outcome, lack of significance of the early thymic precursor (ETP) immunophenotype, and validation of the prognostic value of end-induction minimal residual disease (MRD) in Children’s Oncology Group (COG) Study AALL0434 [abstract]
.
Blood
.
2014
;
124
(
21
).
Abstract 1
.
15.
Shah
BD
,
Borate
U
,
Kota
VK
, et al
.
Multi-institution review of adult early T-cell precursor acute lymphoblastic leukemia/lymphoma (ETP-ALL) [abstract]
.
Blood
.
2015
;
126
(
23
).
Abstract 3715
.
16.
Guo
RJ
,
Bahmanyar
M
,
Minden
MD
,
Chang
H
.
CD33, not early precursor T-cell phenotype, is associated with adverse outcome in adult T-cell acute lymphoblastic leukaemia
.
Br J Haematol
.
2016
;
172
(
5
):
823
-
825
.
17.
Patrick
K
,
Wade
R
,
Goulden
N
, et al
.
Outcome for children and young people with early T-cell precursor acute lymphoblastic leukaemia treated on a contemporary protocol, UKALL 2003
.
Br J Haematol
.
2014
;
166
(
3
):
421
-
424
.
18.
Maude
SL
,
Dolai
S
,
Delgado-Martin
C
, et al
.
Efficacy of JAK/STAT pathway inhibition in murine xenograft models of early T-cell precursor (ETP) acute lymphoblastic leukemia
.
Blood
.
2015
;
125
(
11
):
1759
-
1767
.
19.
Bond
J
,
Marchand
T
,
Touzart
A
, et al
.
An early thymic precursor phenotype predicts outcome exclusively in HOXA-overexpressing adult T-cell acute lymphoblastic leukemia: a Group for Research in Adult Acute Lymphoblastic Leukemia study
.
Haematologica
.
2016
;
101
(
6
):
732
-
740
.
20.
Weng
AP
,
Ferrando
AA
,
Lee
W
, et al
.
Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia
.
Science
.
2004
;
306
(
5694
):
269
-
271
.
21.
Clappier
E
,
Collette
S
,
Grardel
N
, et al
;
EORTC-CLG
.
NOTCH1 and FBXW7 mutations have a favorable impact on early response to treatment, but not on outcome, in children with T-cell acute lymphoblastic leukemia (T-ALL) treated on EORTC trials 58881 and 58951
.
Leukemia
.
2010
;
24
(
12
):
2023
-
2031
.
22.
Kox
C
,
Zimmermann
M
,
Stanulla
M
, et al
.
The favorable effect of activating NOTCH1 receptor mutations on long-term outcome in T-ALL patients treated on the ALL-BFM 2000 protocol can be separated from FBXW7 loss of function
.
Leukemia
.
2010
;
24
(
12
):
2005
-
2013
.
23.
Larson Gedman
A
,
Chen
Q
,
Kugel Desmoulin
S
, et al
.
The impact of NOTCH1, FBW7 and PTEN mutations on prognosis and downstream signaling in pediatric T-cell acute lymphoblastic leukemia: a report from the Children’s Oncology Group
.
Leukemia
.
2009
;
23
(
8
):
1417
-
1425
.
24.
Zuurbier
L
,
Homminga
I
,
Calvert
V
, et al
.
NOTCH1 and/or FBXW7 mutations predict for initial good prednisone response but not for improved outcome in pediatric T-cell acute lymphoblastic leukemia patients treated on DCOG or COALL protocols
.
Leukemia
.
2010
;
24
(
12
):
2014
-
2022
.
25.
Asnafi
V
,
Buzyn
A
,
Le Noir
S
, et al
.
NOTCH1/FBXW7 mutation identifies a large subgroup with favorable outcome in adult T-cell acute lymphoblastic leukemia (T-ALL): a Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL) study
.
Blood
.
2009
;
113
(
17
):
3918
-
3924
.
26.
Baldus
CD
,
Thibaut
J
,
Goekbuget
N
, et al
.
Prognostic implications of NOTCH1 and FBXW7 mutations in adult acute T-lymphoblastic leukemia
.
Haematologica
.
2009
;
94
(
10
):
1383
-
1390
.
27.
Jenkinson
S
,
Koo
K
,
Mansour
MR
, et al
.
Impact of NOTCH1/FBXW7 mutations on outcome in pediatric T-cell acute lymphoblastic leukemia patients treated on the MRC UKALL 2003 trial
.
Leukemia
.
2013
;
27
(
1
):
41
-
47
.
28.
Mansour
MR
,
Sulis
ML
,
Duke
V
, et al
.
Prognostic implications of NOTCH1 and FBXW7 mutations in adults with T-cell acute lymphoblastic leukemia treated on the MRC UKALLXII/ECOG E2993 protocol
.
J Clin Oncol
.
2009
;
27
(
26
):
4352
-
4356
.
29.
Trinquand
A
,
Tanguy-Schmidt
A
,
Ben Abdelali
R
, et al
.
Toward a NOTCH1/FBXW7/RAS/PTEN-based oncogenetic risk classification of adult T-cell acute lymphoblastic leukemia: a Group for Research in Adult Acute Lymphoblastic Leukemia study
.
J Clin Oncol
.
2013
;
31
(
34
):
4333
-
4342
.
30.
Beldjord
K
,
Chevret
S
,
Asnafi
V
, et al
;
Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL)
.
Oncogenetics and minimal residual disease are independent outcome predictors in adult patients with acute lymphoblastic leukemia
.
Blood
.
2014
;
123
(
24
):
3739
-
3749
.
31.
Amylon
MD
,
Shuster
J
,
Pullen
J
, et al
.
Intensive high-dose asparaginase consolidation improves survival for pediatric patients with T cell acute lymphoblastic leukemia and advanced stage lymphoblastic lymphoma: a Pediatric Oncology Group study
.
Leukemia
.
1999
;
13
(
3
):
335
-
342
.
32.
Seibel
NL
,
Steinherz
PG
,
Sather
HN
, et al
.
Early postinduction intensification therapy improves survival for children and adolescents with high-risk acute lymphoblastic leukemia: a report from the Children’s Oncology Group
.
Blood
.
2008
;
111
(
5
):
2548
-
2555
.
33.
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
(
6
):
1986
-
1994
.
34.
Panetta
JC
,
Gajjar
A
,
Hijiya
N
, et al
.
Comparison of native E. coli and PEG asparaginase pharmacokinetics and pharmacodynamics in pediatric acute lymphoblastic leukemia
.
Clin Pharmacol Ther
.
2009
;
86
(
6
):
651
-
658
.
35.
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
(
10
):
4164
-
4167
.
36.
Kurtzberg
J
,
Asselin
B
,
Bernstein
M
,
Buchanan
GR
,
Pollock
BH
,
Camitta
BM
.
Polyethylene glycol-conjugated L-asparaginase versus native L-asparaginase in combination with standard agents for children with acute lymphoblastic leukemia in second bone marrow relapse: a Children’s Oncology Group Study (POG 8866)
.
J Pediatr Hematol Oncol
.
2011
;
33
(
8
):
610
-
616
.
37.
Kota
VK
,
Hathaway
AR
,
Shah
BD
, et al
.
Poor Outcomes with hyper CVAD induction for T-cell lymphoblastic leukemia/lymphoma [abstract]
.
Blood
.
2015
;
126
(
23
).
Abstract 3762
.
38.
Kozlowski
P
,
Åström
M
,
Ahlberg
L
, et al
;
Swedish Adult ALL Group
.
High relapse rate of T cell acute lymphoblastic leukemia in adults treated with hyper-CVAD chemotherapy in Sweden
.
Eur J Haematol
.
2014
;
92
(
5
):
377
-
381
.
39.
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
(
7
):
813
-
819
.
40.
Rosen
O
,
Müller
HJ
,
Gökbuget
N
, et al
.
Pegylated asparaginase in combination with high-dose methotrexate for consolidation in adult acute lymphoblastic leukaemia in first remission: a pilot study
.
Br J Haematol
.
2003
;
123
(
5
):
836
-
841
.
41.
Göekbuget
N
,
Baumann
A
,
Beck
J
, et al
.
PEG-asparaginase intensification in adult acute lymphoblastic leukemia (ALL): significant improvement of outcome with moderate increase of liver toxicity in the German Multicenter Study Group for Adult ALL (GMALL) Study 07/2003 [abstract]
.
Blood
.
2010
;
116
(
21
).
Abstract 494
.
42.
DeAngelo
DJ
,
Stevenson
K
,
Neuberg
DS
, et al
.
A multicenter phase II study using a dose intensified pegylated-asparaginase pediatric regimen in adults with untreated acute lymphoblastic leukemia: a DFCI ALL Consortium Trial [abstract]
.
Blood
.
2015
;
126
(
23
).
Abstract 80
.
43.
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
(
7
):
2744
-
2750
.
44.
Douer
D
,
Aldoss
I
,
Lunning
MA
, et al
.
Pharmacokinetics-based integration of multiple doses of intravenous pegaspargase in a pediatric regimen for adults with newly diagnosed acute lymphoblastic leukemia
.
J Clin Oncol
.
2014
;
32
(
9
):
905
-
911
.
45.
Rivero
GA
,
Bailey
K
,
Blackford
A
, et al
.
Statistically significant differences in toxicities between L- and PEG asparaginase in adult patients with acute lymphoblastic leukemia [abstract]
.
Blood
.
2010
;
116
(
21
).
Abstract 2121
.
46.
Patel
B
,
Kirkwood
A
,
Dey
A
, et al
.
Pegylated-asparaginase during induction therapy for adult acute lymphoblastic leukaemia: toxicity data from The UKALL14 Trial
.
Leukemia
.
2017
;
31
(
1
):
58
-
64
.
47.
Lazarus
HM
,
Richards
SM
,
Chopra
R
, et al
;
Medical Research Council (MRC)/National Cancer Research Institute (NCRI) Adult Leukaemia Working Party of the United Kingdom and the Eastern Cooperative Oncology Group
.
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
(
2
):
465
-
472
.
48.
Asselin
BL
,
Devidas
M
,
Wang
C
, et al
.
Effectiveness of high-dose methotrexate in T-cell lymphoblastic leukemia and advanced-stage lymphoblastic lymphoma: a randomized study by the Children’s Oncology Group (POG 9404)
.
Blood
.
2011
;
118
(
4
):
874
-
883
.
49.
Clarke
M
,
Gaynon
P
,
Hann
I
, et al
;
Childhood ALL Collaborative Group
.
CNS-directed therapy for childhood acute lymphoblastic leukemia: Childhood ALL Collaborative Group overview of 43 randomized trials
.
J Clin Oncol
.
2003
;
21
(
9
):
1798
-
1809
.
50.
Huguet
F
,
Leguay
T
,
Raffoux
E
, et al
.
Pediatric-inspired therapy in adults with Philadelphia chromosome-negative acute lymphoblastic leukemia: the GRAALL-2003 study [published correction appears in J Clin Oncol. 2009;27(15):2574]
.
J Clin Oncol
.
2009
;
27
(
6
):
911
-
918
.
51.
Jain
P
,
Kantarjian
H
,
Ravandi
F
, et al
.
The combination of hyper-CVAD plus nelarabine as frontline therapy in adult T-cell acute lymphoblastic leukemia and T-lymphoblastic lymphoma: MD Anderson Cancer Center experience
.
Leukemia
.
2014
;
28
(
4
):
973
-
975
.
52.
Yanada
M
,
Jinnai
I
,
Takeuchi
J
, et al
.
Clinical features and outcome of T-lineage acute lymphoblastic leukemia in adults: a low initial white blood cell count, as well as a high count predict decreased survival rates
.
Leuk Res
.
2007
;
31
(
7
):
907
-
914
.
53.
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
(
12
):
2788
-
2801
.
54.
Hamdi
A
,
Mawad
R
,
Bassett
R
, et al
.
Central nervous system relapse in adults with acute lymphoblastic leukemia after allogeneic hematopoietic stem cell transplantation
.
Biol Blood Marrow Transplant
.
2014
;
20
(
11
):
1767
-
1771
.
55.
van Dongen
JJM
,
van der Velden
VHJ
,
Brüggemann
M
,
Orfao
A
.
Minimal residual disease diagnostics in acute lymphoblastic leukemia: need for sensitive, fast, and standardized technologies
.
Blood
.
2015
;
125
(
26
):
3996
-
4009
.
56.
Vidriales
M-B
,
Pérez
JJ
,
López-Berges
MC
, et al
.
Minimal residual disease in adolescent (older than 14 years) and adult acute lymphoblastic leukemias: early immunophenotypic evaluation has high clinical value
.
Blood
.
2003
;
101
(
12
):
4695
-
4700
.
57.
Brüggemann
M
,
Raff
T
,
Flohr
T
, et al
;
German Multicenter Study Group for Adult Acute Lymphoblastic Leukemia
.
Clinical significance of minimal residual disease quantification in adult patients with standard-risk acute lymphoblastic leukemia
.
Blood
.
2006
;
107
(
3
):
1116
-
1123
.
58.
Holowiecki
J
,
Krawczyk-Kulis
M
,
Giebel
S
, et al
.
Status of minimal residual disease after induction predicts outcome in both standard and high-risk Ph-negative adult acute lymphoblastic leukaemia. The Polish Adult Leukemia Group ALL 4-2002 MRD Study
.
Br J Haematol
.
2008
;
142
(
2
):
227
-
237
.
59.
Bassan
R
,
Spinelli
O
,
Oldani
E
, et al
.
Improved risk classification for risk-specific therapy based on the molecular study of minimal residual disease (MRD) in adult acute lymphoblastic leukemia (ALL)
.
Blood
.
2009
;
113
(
18
):
4153
-
4162
.
60.
Gökbuget
N
,
Kneba
M
,
Raff
T
, et al
.
Adult patients with acute lymphoblastic leukemia and molecular failure display a poor prognosis and are candidates for stem cell transplantation and targeted therapies
.
2012
;
120
(
9
):
1868
-
1876
.
61.
Goldstone
AH
,
Richards
SM
,
Lazarus
HM
, et al
.
In adults with standard-risk acute lymphoblastic leukemia, the greatest benefit is achieved from a matched sibling allogeneic transplantation in first complete remission, and an autologous transplantation is less effective than conventional consolidation/maintenance chemotherapy in all patients: final results of the International ALL Trial (MRC UKALL XII/ECOG E2993)
.
Blood
.
2008
;
111
(
4
):
1827
-
1833
.
62.
Passweg
JR
,
Tiberghien
P
,
Cahn
JY
, et al
.
Graft-versus-leukemia effects in T lineage and B lineage acute lymphoblastic leukemia
.
Bone Marrow Transplant
.
1998
;
21
(
2
):
153
-
158
.
63.
Litzow
MR
,
Ferrando
AA
.
How I treat T-cell acute lymphoblastic leukemia in adults
.
Blood
.
2015
;
126
(
7
):
833
-
841
.
64.
Okasha
D
,
Kirkwood
AA
,
Copland
M
, et al
.
Fludarabine, melphalan and alemtuzumab conditioned reduced intensity (RIC) allogeneic hematopoietic cell transplantation for adults aged >40 years with de novo acute lymphoblastic leukemia: a prospective study from the UKALL14 Trial (ISRCTN 66541317) [abstract]
.
Blood
.
2015
;
126
(
23
).
Abstract 733
.
65.
Sorror
ML
,
Storb
RF
,
Sandmaier
BM
, et al
.
Comorbidity-age index: a clinical measure of biologic age before allogeneic hematopoietic cell transplantation
.
J Clin Oncol
.
2014
;
32
(
29
):
3249
-
3256
.
66.
Marks
DI
,
Woo
KA
,
Zhong
X
, et al
.
Unrelated umbilical cord blood transplant for adult acute lymphoblastic leukemia in first and second complete remission: a comparison with allografts from adult unrelated donors
.
Haematologica
.
2014
;
99
(
2
):
322
-
328
.
67.
Mato
AR
,
Luger
SM
.
Autologous stem cell transplant in ALL: who should we be transplanting in first remission?
Bone Marrow Transplant
.
2006
;
37
(
11
):
989
-
995
.
68.
Parovichnikova
E
,
Kuzmina
L
,
Mendeleeva
L
, et al
.
Late intensification with autologous HSCT after non-myeloablative BEAM conditioning has shown to be effective approach in adults with T-cell acute lymphoblastic leukemia treated by non-intensive protocol: results of the RALL study group [abstract]
.
Bone Marrow Transplant
.
2015
;
50
(
suppl 1
):
S57
-
S58
. Abstract 98.
69.
Hough
R
,
Rowntree
C
,
Goulden
N
, et al
.
Efficacy and toxicity of a paediatric protocol in teenagers and young adults with Philadelphia chromosome negative acute lymphoblastic leukaemia: results from UKALL 2003
.
Br J Haematol
.
2016
;
172
(
3
):
439
-
451
.
70.
Ramanujachar
R
,
Richards
S
,
Hann
I
, et al
.
Adolescents with acute lymphoblastic leukaemia: outcome on UK national paediatric (ALL97) and adult (UKALLXII/E2993) trials
.
Pediatr Blood Cancer
.
2007
;
48
(
3
):
254
-
261
.
71.
Boissel
N
,
Auclerc
M-F
,
Lhéritier
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
(
5
):
774
-
780
.
72.
de Bont
JM
,
Holt
Bv
,
Dekker
AW
,
van der Does-van den Berg
A
,
Sonneveld
P
,
Pieters
R
.
Significant difference in outcome for adolescents with acute lymphoblastic leukemia treated on pediatric vs adult protocols in the Netherlands
.
Leukemia
.
2004
;
18
(
12
):
2032
-
2035
.
73.
Hallböök
H
,
Gustafsson
G
,
Smedmyr
B
,
Söderhäll
S
,
Heyman
M
;
Swedish Adult Acute Lymphocytic Leukemia Group; Swedish Childhood Leukemia Group
.
Treatment outcome in young adults and children >10 years of age with acute lymphoblastic leukemia in Sweden: a comparison between a pediatric protocol and an adult protocol
.
Cancer
.
2006
;
107
(
7
):
1551
-
1561
.
74.
Ribera
JM
,
Oriol
A
,
Sanz
MA
, et al
.
Comparison of the results of the treatment of adolescents and young adults with standard-risk acute lymphoblastic leukemia with the Programa Español de Tratamiento en Hematología pediatric-based protocol ALL-96
.
J Clin Oncol
.
2008
;
26
(
11
):
1843
-
1849
.
75.
Stock
W
,
La
M
,
Sanford
B
, et al
;
Children’s Cancer Group; Cancer and Leukemia Group B studies
.
What determines the outcomes for adolescents and young adults with acute lymphoblastic leukemia treated on cooperative group protocols? A comparison of Children’s Cancer Group and Cancer and Leukemia Group B studies
.
Blood
.
2008
;
112
(
5
):
1646
-
1654
.
76.
Usvasalo
A
,
Räty
R
,
Knuutila
S
, et al
.
Acute lymphoblastic leukemia in adolescents and young adults in Finland
.
Haematologica
.
2008
;
93
(
8
):
1161
-
1168
.
77.
Gökbuget
N
,
Beck
J
,
Brandt
K
, et al
.
Significant improvement of outcome in adolescents and young adults (AYAs) aged 15-35 years with acute lymphoblastic leukemia (ALL) with a pediatric derived adult ALL protocol; results of 1529 AYAs in 2 consecutive trials of The German Multicenter Study Group for Adult ALL (GMALL) [abstract]
.
Blood
.
2013
;
122
(
21
).
Abstract 839
.
78.
Ram
R
,
Wolach
O
,
Vidal
L
,
Gafter-Gvili
A
,
Shpilberg
O
,
Raanani
P
.
Adolescents and young adults with acute lymphoblastic leukemia have a better outcome when treated with pediatric-inspired regimens: systematic review and meta-analysis
.
Am J Hematol
.
2012
;
87
(
5
):
472
-
478
.
79.
Commander
LA
,
Seif
AE
,
Insogna
IG
,
Rheingold
SR
.
Salvage therapy with nelarabine, etoposide, and cyclophosphamide in relapsed/refractory paediatric T-cell lymphoblastic leukaemia and lymphoma
.
Br J Haematol
.
2010
;
150
(
3
):
345
-
351
.
80.
Fielding
AK
,
Richards
SM
,
Chopra
R
, et al
;
Medical Research Council of the United Kingdom Adult ALL Working Party; Eastern Cooperative Oncology Group
.
Outcome of 609 adults after relapse of acute lymphoblastic leukemia (ALL); an MRC UKALL12/ECOG 2993 study
.
Blood
.
2007
;
109
(
3
):
944
-
950
.
81.
Specchia
G
,
Pastore
D
,
Carluccio
P
, et al
.
FLAG-IDA in the treatment of refractory/relapsed adult acute lymphoblastic leukemia
.
Ann Hematol
.
2005
;
84
(
12
):
792
-
795
.
82.
Berg
SL
,
Blaney
SM
,
Devidas
M
, et al
;
Children’s Oncology Group
.
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
(
15
):
3376
-
3382
.
83.
Kurtzberg
J
,
Ernst
TJ
,
Keating
MJ
, et al
.
Phase I study of 506U78 administered on a consecutive 5-day schedule in children and adults with refractory hematologic malignancies
.
J Clin Oncol
.
2005
;
23
(
15
):
3396
-
3403
.
84.
DeAngelo
DJ
,
Yu
D
,
Johnson
JL
, et al
.
Nelarabine induces complete remissions in adults with relapsed or refractory T-lineage acute lymphoblastic leukemia or lymphoblastic lymphoma: Cancer and Leukemia Group B study 19801
.
Blood
.
2007
;
109
(
12
):
5136
-
5142
.
85.
Gökbuget
N
,
Basara
N
,
Baurmann
H
, et al
.
High single-drug activity of nelarabine in relapsed T-lymphoblastic leukemia/lymphoma offers curative option with subsequent stem cell transplantation
.
Blood
.
2011
;
118
(
13
):
3504
-
3511
.
86.
Dunsmore
KP
,
Devidas
M
,
Linda
SB
, et al
.
Pilot study of nelarabine in combination with intensive chemotherapy in high-risk T-cell acute lymphoblastic leukemia: a report from the Children’s Oncology Group
.
J Clin Oncol
.
2012
;
30
(
22
):
2753
-
2759
.
87.
Palomero
T
,
Barnes
KC
,
Real
PJ
, et al
.
CUTLL1, a novel human T-cell lymphoma cell line with t(7;9) rearrangement, aberrant NOTCH1 activation and high sensitivity to gamma-secretase inhibitors
.
Leukemia
.
2006
;
20
(
7
):
1279
-
1287
.
88.
Armstrong
F
,
Brunet de la Grange
P
,
Gerby
B
, et al
.
NOTCH is a key regulator of human T-cell acute leukemia initiating cell activity
.
Blood
.
2009
;
113
(
8
):
1730
-
1740
.
89.
Zweidler-McKay
P
,
DeAngelo
DJ
,
Douer
D
, et al
.
The safety and activity of BMS-906024, a gamma secretase inhibitor (GSI) with anti-notch activity, in patients with relapsed T-cell acute lymphoblastic leukemia (T-ALL): initial results of a phase I trial [abstract]
.
Blood
.
2014
;
124
(
21
).
Abstract 968
.
90.
Papayannidis
C
,
DeAngelo
DJ
,
Stock
W
, et al
.
A Phase 1 study of the novel gamma-secretase inhibitor PF-03084014 in patients with T-cell acute lymphoblastic leukemia and T-cell lymphoblastic lymphoma
.
Blood Cancer J
.
2015
;
5
:
e350
.
91.
Graux
C
,
Cools
J
,
Melotte
C
, et al
.
Fusion of NUP214 to ABL1 on amplified episomes in T-cell acute lymphoblastic leukemia
.
Nat Genet
.
2004
;
36
(
10
):
1084
-
1089
.
92.
Quintás-Cardama
A
,
Tong
W
,
Manshouri
T
, et al
.
Activity of tyrosine kinase inhibitors against human NUP214-ABL1-positive T cell malignancies
.
Leukemia
.
2008
;
22
(
6
):
1117
-
1124
.
93.
Zheng
W
,
Medeiros
LJ
,
Young
KH
, et al
.
CD30 expression in acute lymphoblastic leukemia as assessed by flow cytometry analysis
.
Leuk Lymphoma
.
2014
;
55
(
3
):
624
-
627
.
94.
Mamonkin
M
,
Rouce
RH
,
Tashiro
H
,
Brenner
MKA
.
A T-cell-directed chimeric antigen receptor for the selective treatment of T-cell malignancies
.
Blood
.
2015
;
126
(
8
):
983
-
992
.
Sign in via your Institution