At the intersection between children and older adults, the care of adolescent and young adult (AYA) patients with acute lymphoblastic leukemia (ALL) poses unique challenges and issues beyond those faced by other age groups. Although the survival of AYA patients is inferior to younger children, growing evidence suggests that AYA patients have improved outcomes, with disease-free survival rates of 60% to 70%, when treated with pediatric-based approaches. A holistic approach, incorporating a multidisciplinary team, is a key component of successful treatment of these AYA patients. With the appropriate support and management of toxicities during and following treatment, these regimens are well tolerated in the AYA population. Even with the significant progress that has been made during the last decade, patients with persistence of minimal residual disease (MRD) during intensive therapy still have a poor prognosis. With new insights into disease pathogenesis in AYA ALL and the availability of disease-specific kinase inhibitors and novel targeted antibodies, future studies will focus on individualized therapy to eradicate MRD and result in further improvements in survival. This case-based review will discuss the biology, pharmacology, and psychosocial aspects of AYA patients with ALL, highlighting our current approach to the management of these unique patients.

Acute lymphoblastic leukemia (ALL), a relatively rare malignancy, is one of the few cancers that impacts the entire lifespan, from neonates to the very elderly.1  Although survival now approaches 90% for most children with ALL,2,3  older adolescents and young adults (AYAs) historically have a much poorer prognosis, with an event-free survival (EFS) of only 30% to 45%.4-6  Factors accounting for differences in outcome include heterogeneity in disease biology, host factors (both physiologic and psychosocial), and importantly, the therapeutic approach and experience of the health care teams.7-11  Some authors also suggest that AYAs may have had poorer outcomes, in part, because of low rates of clinical trial enrollment.12  Between 1997 and 2003, fewer than 2% of older adolescents were enrolled in clinical trials, compared with 60% of pediatric patients,13  potentially due to fewer referrals to institutions where clinical trials are offered, limited numbers of clinical trials available for the AYA population, and psychosocial barriers.14 

During the last decade, recognition of the unique characteristics of AYAs with ALL, as well as a new focus on clinical research designed specifically for this population, has led to exciting improvements in treatment outcomes, with EFS now approaching 70% for AYA ALL. The National Cancer Institute has defined the AYA cancer population broadly as being between the ages of 15 to 39 years old.15  Although tremendous heterogeneity in this population clearly exists,16  and the age cutoff of 40 years is somewhat arbitrarily defined, emerging clinical, psychosocial, and biologic features of the disease suggest this may be a distinct population.17,18  This case-based review will focus on the AYA population most commonly treated by adult hematologists-oncologists, ie, patients aged 18 to 39 years old.

A 28-year-old man presents with night sweats, fatigue, palpitations, and abdominal pain. He is found to have a white blood cell count of 23 × 109/L and bulky organomegaly. Bone marrow (BM) biopsy is consistent with precursor B-cell ALL (CD19+, CD20, CD10+,CD22+, CD79a+, CD34+, and TdT+). Fluorescence in situ hybridization (FISH) is negative for MLL rearrangement, BCR/ABL1, ETV5/RUNX1, and trisomies 4, 10, and 17. Cytogenetics shows a normal male karyotype.

What is our approach? If available, we would encourage enrollment onto a clinical trial focused on AYAs with ALL that builds upon an intensive pediatric approach to treatment.

Rationale

Treatment approaches for AYAs with ALL vary considerably, with the choice of regimen predicated on the familiarity and expertise of the treating physician, the availability of a clinical trial, and most importantly, the “door” that the patient enters, namely whether entry is into a pediatric or an adult treatment center. In the United Kingdom, AYA inpatient treatment units already exist and this may facilitate a more uniform treatment approach. However, in the United States, patients younger than 18 years of age are traditionally treated in pediatric departments, whereas AYAs older than 18 years of age are treated by adult hematologists/oncologists and therefore receive “adult” ALL regimens. These “adult” regimens typically consist of intensive use of myelosuppressive agents including daunorubicin, cytarabine, and cyclophosphamide, as well as allogeneic stem cell transplantation (SCT) in first remission.5,19,20  In contrast, pediatric regimens focus on the Berlin-Frankfurt-Munster (BFM) backbone of ALL therapy21 : glucocorticoids, vincristine, asparaginase, early and frequent central nervous system (CNS) prophylaxis, and prolonged maintenance therapy.22-25  Retrospective studies from large North American and European groups suggest that AYAs have superior outcomes when treated with “pediatric” regimens,21-25  and may approach those of younger children, with 5-year EFS of over 70%.26,27  Based on these data, as well as the prospective studies described below, we recommend that AYA patients be treated on a pediatric-based regimen.

Encouraging survival outcomes for AYAs with ALL treated with “pediatric-inspired” regimens have recently been reported from a number of prospective cooperative group clinical trials performed in Europe and the United States.23,28-33  These trials showed an improvement in both EFS and overall survival (OS) compared with historical controls, with >60% EFS and OS in the majority of studies.23,28-33  Both retrospective and prospective studies have included both B- and T-cell ALL, with quite similar EFS and OS.29,33,34  Importantly, although some groups have noted slightly poorer tolerability,29  and certain toxicities (such as hepatotoxicity) may be more common in an older population, overall these regimens have demonstrated feasibility in the AYA population. Several of these trials and regimens are outlined in Table 1 and also reviewed in a recent meta-analysis.35 

Table 1

AYA treatment regimens

ALL-96 (PETHEMA)29 DFCI Adult ALL 01-17532 C1040334 
Patient population    
 Number of patients 81 92 318 
 Median age at diagnosis (range) 20 years (15-30) 28 years (18-50) 24 years (17-39) 
 Gender Male: 62% Male: 61% Male: 61% 
 Immunophenotype Precursor B- and T-cell ALL Precursor B cell (80%) and precursor T cell (20%) Precursor B cell (76%) and precursor T cell (24%) 
Regimen 
 Induction Vincristine: 2 mg IV (d 1, 8, 15, 22)
Daunorubicin: 30 mg/m2 IV (d 1, 8, 15, 22)
Prednisone: 60 mg/m2 (d 1-27), 30 mg/m2 (d 28-35) PO/IV
Asparaginase: 10 000 U/m2 IV, (d 10-12, 17-19, 24-26)
Cyclophosphamide: 1000 mg/m2 (d 36) 
Vincristine: 2 mg IV (d 1, 8, 15, 22)
Doxorubicin: 30 mg/m2 IV (d 1, 2)
Prednisone: 40 mg/m2 PO (d 1-28)
Methotrexate: 4 g/m2 IV (d 3, 8 to 24 h after doxorubicin)
E colil-asparaginase 25 000 IU/m2 IM (d 5)
Followed by CNS therapy (see below) 
Vincristine: 1.5 mg/m2 - maximum 2 mg IV (d 1, 8, 15, 22)
Daunorubicin: 25 mg/m2 IV (d 1, 8, 15, 22)
Prednisone: 60 mg/m2 IV/PO (d 1-28)
PEG-asparaginase: 2500 IU/m2 IM/IV (d 4)
Extended remission induction (2 wk) administered if failure to achieve morphologic remission on d 29 bone marrow biopsy 
 Consolidation-1/
intensification 
Mercaptopurine: 50 mg/m2 PO (d 1-7) Vincristine: 2 mg (d 1) Vincristine: 1.5 mg/m2 - maximum 2 mg IV (d 15, 22, 43, 50) 
Methotrexate: 3 g/m2 IV (d 1, 28, 56) Methotrexate: 30 m/m2 IV/IM weekly Cyclophosphamide: 1000 mg/m2 (d 1, 29) 
VM-26: 150 mg/m2 IV (d 14, 42) Dexamethasone: 18 mg/m2 PO (d 1-5) Cytarabine: 75 mg/m2 IV/SC (d 1-4, 8-11, 29-32, 36-39) 
Cytarabine: 500 mg/m2 IV (d 14-15, 42, 43) Mercaptopurine: 50 mg/m2 PO (d 1-14) Mercaptopurine: 60 mg/m2 PO (d 1-14, 29-42) 
 E coli asparaginase: 12 500 IU/m2 starting dose PEG-asp: 2500 IU/m2 IM/IV (d 15, 43) 
  Doxorubicin: 30 mg/m2 IV (d 1)   
 Consolidation-2/interim
maintenance 
Dexamethasone: 10 mg/m2 (d 1-14), 5 mg/m2 (d 15-21) PO/IV
Vincristine: 1.5 mg/m2 - maximum 2 mg IV (d 1, 8, 15)
Daunorubicin: 30 mg/m2 IV (d 1, 2, 8, 9)
Cyclophosphamide: 600 mg/m2 (d 1, 15)
Asparaginase: 10 000 U/m2 IV, (d 1-3, 15-17) 
N/A Vincristine: 1.5 mg/m2 - maximum 2 mg IV (d 1, 11, 21, 31, 41)
Methotrexate: 100 m/m2 IV starting, with dose escalation (d 1, 11, 21, 31, 41)
PEG-asp: 2500 IU/m2 IM/IV (d 2, 22) 
 Delayed intensification N/A N/A Vincristine: 1.5 mg/m2 - maximum 2 mg IV (d 1, 8, 15, 43, 50) 
  Dexamethasone: 10 mg/m2 PO/IV (d 1-7, 15-21) 
  Doxorubicin: 25 mg/m2 IV (d 1, 8, 15) 
  PEG-asp: 2500 IU/m2 IM/IV (d 4, 43) 
  Cyclophosphamide: 1000 mg/m2 (d 29) 
  Cytarabine: 75 mg/m2 IV/SC (d 29-32, 36-39) 
  Thioguanine (6-TG): 60 mg/m2 PO (d 29-42) 
 Maintenance/continuation Maintenance-1 (until wk 52)
Methotrexate IM: 20 mg/m2 per wk
Mercaptopurine: PO 50 mg/m2 daily
Reinduction (every 4 wk)
Vincristine: 1.5 mg/m2 - maximum 2 mg IV (d 1)
Prednisone: 60 mg/m2 daily
Asparaginase: 20 000 U/m2 IV (d 1)
Maintenance-2 (wks 53-104)
Methotrexate IM: 20 mg/m2 per wk
Mercaptopurine PO: 50 mg/m2 daily 
Cycles every 3 wk × 74 wk
Vincristine: 2 mg (d 1)
Methotrexate: 30 mg/m2 IV/IM weekly
Dexamethasone: 6 mg/m2 PO (d 1-5)
Mercaptopurine: 50 mg/m2 PO (d 1-14)
Doxorubicin: 30 mg/m2 IV (d 1) 
Duration: Females 2 y, males 3 y (12-wk courses)
Vincristine: 1.5 mg/m2 - maximum 2 mg IV (d 1, 29, 57)
Dexamethasone: 6 mg/m2 PO/IV (d 1-5, 29-33, 57-61)
Mercaptopurine: 75 mg/m2 PO (d 1-84)
Methotrexate: 20 mg/m2 PO weekly (d 8-78) – held on d 29 during 1st 4 courses (when IT methotrexate given) 
 CNS prophylaxis Induction/Consolidation (d 1, 29)
Methotrexate: 15 mg IT
Cytarabine: 30 mg IT
Hydrocortisone: 20 mg IT
Maintenance/reinduction (d 1)
Methotrexate: 15 mg IT
Cytarabine: 30 mg IT
Hydrocortisone: 20 mg IT 
Induction
Cytarabine: 50 mg IT (d 0)
Cytarabine: 40 mg /methotrexate 12 mg/ hydrocortisone 50 mg IT (d 15 and 29)
CNS therapy (3 wk)
Vincristine: 2 mg IV (d 1)
Mercaptopurine: 50 mg/m2 PO × 14 d
Doxorubicin: 30 mg/m2 IV × 1 dose
IT methotrexate/cytarabine twice weekly × 4 doses
Cranial radiation (18-24 Gy)
Intensification (30 wk) and continuation (74 wk)
IT methotrexate/cytarabine/hydrocortisone at start of cycle 
Induction
Methotrexate: 15 mg IT (d 8, 29)
Cytarabine: 70 mg IT (d 1)
Consolidation
Methotrexate: 15 mg IT (d 1, 8, 15, 22)
Interim maintenance
Methotrexate: 15 mg IT (d 1, 31)
Delayed intensification
Methotrexate: 15 mg IT (d 1, 29, 36)
Maintenance
Methotrexate: 15 mg IT (d 1), also given d 29 for first 4 courses 
Outcomes    
 EFS 61% (6-y) 58% (4-y) 66% (2-y) 
 OS 69% (6-y) 67% (4-y) 78% (2-y) 
ALL-96 (PETHEMA)29 DFCI Adult ALL 01-17532 C1040334 
Patient population    
 Number of patients 81 92 318 
 Median age at diagnosis (range) 20 years (15-30) 28 years (18-50) 24 years (17-39) 
 Gender Male: 62% Male: 61% Male: 61% 
 Immunophenotype Precursor B- and T-cell ALL Precursor B cell (80%) and precursor T cell (20%) Precursor B cell (76%) and precursor T cell (24%) 
Regimen 
 Induction Vincristine: 2 mg IV (d 1, 8, 15, 22)
Daunorubicin: 30 mg/m2 IV (d 1, 8, 15, 22)
Prednisone: 60 mg/m2 (d 1-27), 30 mg/m2 (d 28-35) PO/IV
Asparaginase: 10 000 U/m2 IV, (d 10-12, 17-19, 24-26)
Cyclophosphamide: 1000 mg/m2 (d 36) 
Vincristine: 2 mg IV (d 1, 8, 15, 22)
Doxorubicin: 30 mg/m2 IV (d 1, 2)
Prednisone: 40 mg/m2 PO (d 1-28)
Methotrexate: 4 g/m2 IV (d 3, 8 to 24 h after doxorubicin)
E colil-asparaginase 25 000 IU/m2 IM (d 5)
Followed by CNS therapy (see below) 
Vincristine: 1.5 mg/m2 - maximum 2 mg IV (d 1, 8, 15, 22)
Daunorubicin: 25 mg/m2 IV (d 1, 8, 15, 22)
Prednisone: 60 mg/m2 IV/PO (d 1-28)
PEG-asparaginase: 2500 IU/m2 IM/IV (d 4)
Extended remission induction (2 wk) administered if failure to achieve morphologic remission on d 29 bone marrow biopsy 
 Consolidation-1/
intensification 
Mercaptopurine: 50 mg/m2 PO (d 1-7) Vincristine: 2 mg (d 1) Vincristine: 1.5 mg/m2 - maximum 2 mg IV (d 15, 22, 43, 50) 
Methotrexate: 3 g/m2 IV (d 1, 28, 56) Methotrexate: 30 m/m2 IV/IM weekly Cyclophosphamide: 1000 mg/m2 (d 1, 29) 
VM-26: 150 mg/m2 IV (d 14, 42) Dexamethasone: 18 mg/m2 PO (d 1-5) Cytarabine: 75 mg/m2 IV/SC (d 1-4, 8-11, 29-32, 36-39) 
Cytarabine: 500 mg/m2 IV (d 14-15, 42, 43) Mercaptopurine: 50 mg/m2 PO (d 1-14) Mercaptopurine: 60 mg/m2 PO (d 1-14, 29-42) 
 E coli asparaginase: 12 500 IU/m2 starting dose PEG-asp: 2500 IU/m2 IM/IV (d 15, 43) 
  Doxorubicin: 30 mg/m2 IV (d 1)   
 Consolidation-2/interim
maintenance 
Dexamethasone: 10 mg/m2 (d 1-14), 5 mg/m2 (d 15-21) PO/IV
Vincristine: 1.5 mg/m2 - maximum 2 mg IV (d 1, 8, 15)
Daunorubicin: 30 mg/m2 IV (d 1, 2, 8, 9)
Cyclophosphamide: 600 mg/m2 (d 1, 15)
Asparaginase: 10 000 U/m2 IV, (d 1-3, 15-17) 
N/A Vincristine: 1.5 mg/m2 - maximum 2 mg IV (d 1, 11, 21, 31, 41)
Methotrexate: 100 m/m2 IV starting, with dose escalation (d 1, 11, 21, 31, 41)
PEG-asp: 2500 IU/m2 IM/IV (d 2, 22) 
 Delayed intensification N/A N/A Vincristine: 1.5 mg/m2 - maximum 2 mg IV (d 1, 8, 15, 43, 50) 
  Dexamethasone: 10 mg/m2 PO/IV (d 1-7, 15-21) 
  Doxorubicin: 25 mg/m2 IV (d 1, 8, 15) 
  PEG-asp: 2500 IU/m2 IM/IV (d 4, 43) 
  Cyclophosphamide: 1000 mg/m2 (d 29) 
  Cytarabine: 75 mg/m2 IV/SC (d 29-32, 36-39) 
  Thioguanine (6-TG): 60 mg/m2 PO (d 29-42) 
 Maintenance/continuation Maintenance-1 (until wk 52)
Methotrexate IM: 20 mg/m2 per wk
Mercaptopurine: PO 50 mg/m2 daily
Reinduction (every 4 wk)
Vincristine: 1.5 mg/m2 - maximum 2 mg IV (d 1)
Prednisone: 60 mg/m2 daily
Asparaginase: 20 000 U/m2 IV (d 1)
Maintenance-2 (wks 53-104)
Methotrexate IM: 20 mg/m2 per wk
Mercaptopurine PO: 50 mg/m2 daily 
Cycles every 3 wk × 74 wk
Vincristine: 2 mg (d 1)
Methotrexate: 30 mg/m2 IV/IM weekly
Dexamethasone: 6 mg/m2 PO (d 1-5)
Mercaptopurine: 50 mg/m2 PO (d 1-14)
Doxorubicin: 30 mg/m2 IV (d 1) 
Duration: Females 2 y, males 3 y (12-wk courses)
Vincristine: 1.5 mg/m2 - maximum 2 mg IV (d 1, 29, 57)
Dexamethasone: 6 mg/m2 PO/IV (d 1-5, 29-33, 57-61)
Mercaptopurine: 75 mg/m2 PO (d 1-84)
Methotrexate: 20 mg/m2 PO weekly (d 8-78) – held on d 29 during 1st 4 courses (when IT methotrexate given) 
 CNS prophylaxis Induction/Consolidation (d 1, 29)
Methotrexate: 15 mg IT
Cytarabine: 30 mg IT
Hydrocortisone: 20 mg IT
Maintenance/reinduction (d 1)
Methotrexate: 15 mg IT
Cytarabine: 30 mg IT
Hydrocortisone: 20 mg IT 
Induction
Cytarabine: 50 mg IT (d 0)
Cytarabine: 40 mg /methotrexate 12 mg/ hydrocortisone 50 mg IT (d 15 and 29)
CNS therapy (3 wk)
Vincristine: 2 mg IV (d 1)
Mercaptopurine: 50 mg/m2 PO × 14 d
Doxorubicin: 30 mg/m2 IV × 1 dose
IT methotrexate/cytarabine twice weekly × 4 doses
Cranial radiation (18-24 Gy)
Intensification (30 wk) and continuation (74 wk)
IT methotrexate/cytarabine/hydrocortisone at start of cycle 
Induction
Methotrexate: 15 mg IT (d 8, 29)
Cytarabine: 70 mg IT (d 1)
Consolidation
Methotrexate: 15 mg IT (d 1, 8, 15, 22)
Interim maintenance
Methotrexate: 15 mg IT (d 1, 31)
Delayed intensification
Methotrexate: 15 mg IT (d 1, 29, 36)
Maintenance
Methotrexate: 15 mg IT (d 1), also given d 29 for first 4 courses 
Outcomes    
 EFS 61% (6-y) 58% (4-y) 66% (2-y) 
 OS 69% (6-y) 67% (4-y) 78% (2-y) 

CCR, continuous complete remission; d, day; E coli, Escherichia coli; IM, intramuscular; IT, intrathecal; PO, by mouth; SC, subcutaneous; wk, weeks; y, years.

The largest prospective study to evaluate the feasibility of a pediatric regimen in AYA ALL patients is the US intergroup study, C10403.36  Between November 2007 and December 2012, 318 AYAs between 16 and 39 years of age were treated based on the standard arm of the Children’s Oncology Group (COG) regimen (AALL0232).37  This study demonstrated that toxicities were manageable, with low treatment-related mortality (3%), and that treatment with this pediatric regimen is feasible when administered by an adult hematologist/oncologist to an AYA population up to 40 years of age.36  On this regimen, the 2-year EFS and OS were 66% and 78%, respectively.34  Based on these very encouraging early results, the US cooperative groups are now designing a successor study using the C10403 platform that will attempt to incorporate novel targeted agents to further improve treatment outcomes.

Although the majority of recent studies demonstrate a survival benefit using intensive pediatric regimens for AYA, another recently published comparison study of an “adult” regimen (hyper-CVAD) vs a pediatric regimen (BFM-like) found equivalent EFS (∼70%).20  Because this trial was conducted at an institution with a large, experienced leukemia program, the results may not be widely generalizable but suggest that high-volume referral centers may offer benefits beyond chemotherapeutics. In fact, recent data show that outcomes for AYAs with ALL are significantly improved if treatment is administered at a university or National Cancer Institute-sponsored cancer center with expertise in the complex regimens used to treat ALL.38 

Treating AYA patients with pediatric-based regimens has resulted in impressive increases in EFS from 39% to up to 70%. However, further progress is still needed. Several novel agents that have shown impressive activity for relapsed/refractory disease are being evaluated in the frontline setting and will be included in the next generation of prospective clinical trials for AYAs with ALL. For instance, inotuzumab ozogamicin, a CD22 monoclonal antibody bound to calicheamicin, appears to be safe and active in relapsed/refractory ALL, with response rates of 58% to 82% in recent clinical trials.39,40  Blinatumomab, a bispecific T-cell engaging antibody that directs cytotoxic T cells to CD19-expressing target cells, had a 43% response rate in Ph-negative relapsed/refractory precursor B-cell ALL,41  and recently received US Food and Drug Administration (FDA) approval in relapsed disease. Incorporating these agents into frontline chemotherapy will hopefully help to eradicate minimal residual disease (MRD) and result in further improved survival for AYA patients. Blinatumomab is being added to frontline therapy in an ongoing trial for adults with ALL aged 30 to 70 years (NCT02003222)42  and a National Cancer Institute-approved study in the AYA population (aged 18 to 39 years) will test the addition of inotuzumab ozogamicin to the C10403 backbone.

This patient is started on induction chemotherapy according to the C10403 protocol.

What prevention and monitoring should be performed in this patient during induction therapy?

We recommend administering induction therapy as an inpatient until neutrophil recovery. Early treatment toxicities, including febrile neutropenia, hyperglycemia, and hepatic toxicity occur commonly during induction (typically days 10 to 20) and can be more safely managed with close inpatient observation. We also treat with allopurinol for the first 10 days of induction therapy to prevent hyperuricemia. Following induction therapy, the remainder of treatment can be safely administered as an outpatient.

For antimicrobial prophylaxis, we recommend antiviral (acyclovir) and pneumocystis jiroveci pneumonia prophylaxis (typically trimethoprim-sulfamethoxazole) throughout treatment (including maintenance therapy), keeping in mind that no sulfa drug or non-steroidal anti-inflammatory medications should be given on the days that the patient receives methotrexate. Fungal prophylaxis should include mold coverage during induction therapy. However, broader spectrum azole antifungals cannot be used with vincristine because of the risk of exacerbating vincristine-induced peripheral neuropathy. Therefore, we typically use an echinocandin such as micafungin for prophylaxis during induction and switch to fluconazole for outpatient management during consolidation therapy.

Asparaginase-related toxicities are a challenge when intensive pediatric-inspired regimens are used in the AYA population. The only asparaginase preparation available in North America for frontline ALL therapy is a long-acting pegylated form of asparaginase, or PEG-asp. Little is known about the pharmacokinetics of PEG-asp in AYAs or older adults with ALL.10,11  A dose of 2500 IU/m2 PEG-asp, which was used in both the COG ALL 0232 and the C10403 trials, appears to result in greater hepatic toxicity in the AYA population, particularly during induction chemotherapy36 ; thus, we and others have routinely begun to cap the dose at one vial, or 3750 IU. New methods for safer and more accurate dosing of PEG-asp are available, including a recently FDA-approved assay for measurement of asparaginase activity levels. Pediatric studies have demonstrated that achievement of asparaginase levels of greater than 100U/L for 14 days following a dose of PEG-asp is associated with improved treatment outcomes.43  Our group and others have begun to prospectively test the use of lower initial doses of PEG-asp with subsequent dose adjustment, if necessary, to achieve adequate (but not excessive) asparaginase levels.32  Although we are hopeful that level-based dose adjustment may facilitate the safe use of PEG-asp in AYAs, prospective clinical trials are required to validate this hypothesis. Importantly, asparaginase activity levels allow for detection of “silent” inactivation, which may occur without clinical symptoms in up to one-tenth of patients, as a result of antibody neutralization of asparaginase.43,44  Switching asparaginase preparations when silent inactivation occurred resulted in improved EFS in a pediatric population.43 

Asparaginase-related hypersensitivity reactions occur in as many as 20% of children and adults. Therefore, we routinely premedicate our patients with diphenhydramine, hydrocortisone, and acetaminophen prior to each dose of PEG-asp. In C10403, we found that premedication resulted in a decline in significant hypersensitivity reactions to PEG-asp from 15% to 6%.36  If an anaphylactic reaction occurs, no further PEG-asp is administered and we switch to Erwinia asparaginase at a dose of 25 000 IU/m2. Although some may be concerned about failing to detect antibodies to asparaginase when patients are premedicated (resulting in silent inactivation), previous reports have demonstrated that this is a relatively uncommon event with PEG-asp.45  Furthermore, the FDA-approved assay to measure serum asparaginase levels will obviate this concern. An alternative approach in these patients would be to avoid premedication but, if hypersensitivity occurs, manage the acute toxicities and be prepared to switch to Erwinia asparaginase for subsequent treatment.

Other serious toxicities of asparaginase include asthenia, pancreatitis, thrombosis, and bleeding. For a more detailed discussion regarding the prevention and treatment of asparaginase toxicities in adults, a comprehensive set of recommendations was recently published by an expert panel.46 

This patient completes induction therapy per C10403 protocol without significant complications. BM biopsy shows complete remission (CR) with no detectable MRD by flow cytometry.

When should allogeneic transplant in first CR (CR1) be considered? What role does MRD monitoring play in decisions for treatment?

A large prospective randomized international collaborative study (MRC UKALL XII/E2993) demonstrated a significant increase in OS for allogeneic transplant in CR1 when compared with a standard adult ALL regimen (63% vs 52%).19  In contrast, a very recent International Bone Marrow Transplant Registry study of adults 18 to 50 years old found a significant benefit (hazard ratio 3.1; P < .0001) in both disease-free survival (DFS) and OS for patients receiving an intensive pediatric regimen compared with allogeneic transplant in CR1, due to transplant-related mortality.47  Thus, given the risks and complications of transplant, with 20% to 30% nonrelapse transplant mortality in these studies and the high survival (above 70%) and low mortality (3%) rates now being achieved in AYAs with pediatric inspired regimens, we do not routinely recommend allogeneic SCT in CR1. We do, however, routinely perform HLA typing on all patients at diagnosis, but have traditionally reserved transplant for those with high-risk (HR) presenting features, which we consider to be MLL rearrangement48  and hypodiploidy.49,50  More controversial is the negative prognostic significance of early T-cell ALL.51,52  The role of allogeneic transplant in CR1 for a new HR subset, BCR-ABL1–like ALL, remains to be defined and will be discussed briefly in case 2 below.

MRD, measured by flow cytometry or quantitative polymerase chain reaction (qPCR), has emerged as one of the most important prognostic factors in both pediatric and adult ALL and can further inform the decision to transplant in CR1.53-59  Pediatric and adult studies demonstrate that detection of MRD at specified time-points (usually following induction or early consolidation therapy) is associated with high relapse rates and poor survival.55,60-62  For instance, in a study from the German Multicenter Group for Adult ALL, detection of MRD following early consolidation therapy was associated with a continued 5-year CR of only 12%.55  Conversely, the absence of MRD following induction or consolidation therapy using an intensive pediatric regimen has been associated with excellent DFS rates.55  For evaluable patients enrolled on C10403, those with undetectable MRD at end of induction (as detected by a qPCR method) had 93% DFS with a median follow-up of more than 3 years.34  New data incorporating MRD-based prognostic assessments suggest that there is no benefit to allogeneic transplant in CR1 compared with consolidation chemotherapy when MRD levels prior to transplant are low (<10−4 using a qPCR-based assay).57  Thus, we recommend monitoring MRD using a standardized and prospectively validated MRD detection method63,64  and consider referring patients for allogeneic transplant if MRD persists following early consolidation therapy. However, MRD-based referral for transplant requires prospective clinical validation in the AYA patient, and whereas allogeneic transplant performed for AYAs who remain MRD-positive affords some improvement in survival, relapse rates for MRD+ patients are relatively high, ranging from 40% to more than 80%.53,57-59 

Based on the discussion above, we recommend continuing therapy as per protocol (seeTable 1 ). Following intensive postremission consolidation, he continues to maintenance therapy.

During maintenance therapy, we recommend dose escalation of 6-mercaptopurine (6-MP) and methotrexate to maintain adequate myelosuppression, as this has been demonstrated to affect EFS for adolescents with ALL.65  Of note, recent data from the COG suggest that continuous exposure (rather than frequent starting and stopping) is an important determinant of DFS.66  However, practitioners should also be aware that genetic polymorphisms in thiopurine methyltransferase (TPMT) can result in severe hematologic toxicity following treatment with thiopurines, such as 6-MP.67  Because a high proportion of patients have decreased TPMT activity (∼10% with intermediate and 0.3% with low or no activity),67  we now test for genetic polymorphisms in TPMT if patients have prolonged myelosuppression during consolidation therapy or following initiation of maintenance therapy. Other genetic polymorphisms may also contribute to toxicity with 6-MP, such as the recently described NUDT15 variant.68  It is also important that CNS prophylaxis be continued during maintenance therapy to decrease the risk of CNS relapse. Another crucial aspect of long-term maintenance therapy is the close monitoring of drug adherence to the medications that are entirely administered in an outpatient setting (see further discussion below).

Successful treatment of ALL is, unfortunately, associated with the potential for long-term complications that adversely impact the patient’s quality of life. For instance, the intensive use of glucocorticoids in ALL regimens has been associated with significant rates of osteonecrosis, especially in adolescent females69 ; therefore, any joint pain in these patients should be seriously investigated. Neuropathy related to vincristine use is also a common complication from treatment. Neurocognitive dysfunction can occur, although this is less common in modern regimens, which avoid or reduce the dosing of CNS irradiation. Other complications include endocrine and metabolic abnormalities, cardiac toxicity, and secondary malignancies.70  We recommend fertility counseling in all young adults treated for ALL. Comprehensive long-term follow-up guidelines are available through the COG at www.survivorshipguidelines.org and in the National Comprehensive Cancer Network Guidelines for Adolescent and Young Adult Oncology.15 

New data regarding the genetic predisposition to specific treatment toxicities may allow us to further refine our treatment approach to reduce the incidence of these toxicities. For instance, genome-wide analysis has revealed a novel single nucleotide polymorphism in the gene encoding CEP72 that is associated with increased risk of vincristine-induced neuropathy. CEP72 regulates the localization of centrosomal proteins and proper bipolar spindle formation, and knockdown in human ALL cells is associated with augmented vincristine effects.71  Similarly, the risk of glucocorticoid-induced osteonecrosis in children with ALL is associated with single nucleotide polymorphisms in ACP1, which regulates osteoblast differentiation.72  Although further study is needed, one could envision testing for these polymorphisms prior to treatment and adjusting doses based on individual results.

A 28-year-old man is diagnosed with precursor B-cell ALL. PCR and FISH for BCR-ABL1 are negative, and cytogenetics reveals a normal male karyotype. He completes induction therapy on C10403, but day 29 BM biopsy following induction shows significant residual disease (5.5%) by flow cytometry. Chromosomal microarray analysis detects the formation of an EBF1-PDGFRB fusion gene.

The recently described BCR-ABL1–like (or Ph-like) signature is associated with an adverse outcome in both children and young adults with ALL.73  It is characterized by a high frequency of alterations of IKZF1, a gene that encodes the lymphoid transcription factor IKAROS, and carries a gene expression profile similar to that seen in BCR-ABL1+ ALL, but lacks the BCR-ABL1 fusion protein expressed from the t(9;22)(q34.1;q11.2). BCR-ABL1–like ALL is also characterized by overexpression of a number of pathogenetically relevant kinases, some of which may be targeted therapeutically.18  The incidence of the BCR-ABL1–like signature increases with age and is frequent in AYAs (up to 27% in patients aged 20 to 30 years old).73,74  These patients are more likely to have MRD at the end of induction, which, as mentioned above, is an important predictor of DFS.75  These new insights provide further evidence for differences in the biologic basis of ALL as we age, and provide an exciting new rationale for future research to incorporate appropriate kinase inhibitors that may enhance disease response.

At this time, there is not a standardized clinically available assay to identify this genomic signature; however, many groups are working on screening assays to facilitate identification of these cases. A relatively simple assay based on a low-density microarray of several of the highly expressed genes that comprise this signature has already been successfully used to identify patients with the BCR-ABL1–like signature, validated in large pediatric cohorts, and is pending FDA approval.18,76  A variety of other tests, including a panel of FISH probes and/or comparative genomic hybridization are also used to specifically identify recurring fusion genes that result in activated and targetable kinases, including ABL1, ABL2, PDGFRB, JAK1, JAK2, and CRLF2.77,78  Already feasible, these assays are likely to come into common, standardized use within the next year or two to facilitate diagnosis of these cases.

In this particular case, when we noted the gross residual disease at day 28 of induction, we suspected a BCR-ABL1–like signature and performed a comparative genomic hybridization array of the diagnostic sample that identified an EBF1-PDGFRB fusion. During consolidation therapy, as reported by others, we added dasatinib at a dose of 100 mg daily to the standard agents used in consolidation therapy.18,79  The treatment was well tolerated, and 1 month later the disease was in morphologic remission. By the end of consolidation therapy, MRD by flow was undetectable. This patient had an HLA-matched donor and proceeded to allogeneic transplant in CR1. Approximately 180 days posttransplant, he is doing well, has achieved full donor chimerism, and MRD remains undetectable.

It is important to note that the therapeutic approach described above is not yet a standard of care for patients with the BCR-ABL1–like signature. We have a great deal to learn about the feasibility, timing, and impact of incorporating targeted kinase inhibitors (eg, imatinib, dasatinib, ruxolitinib, and others), as well as the role of allogeneic hematopoietic cell transplant for these patients. MRD monitoring in the BCR-ABL1–like cases will also help to refine prognosis and guide treatment choices. Indeed, recently published data demonstrate that MRD measurements can distinguish a subset of relatively good risk vs very poor risk children with BCR-AB1–like ALL.75  Nevertheless, this case illustrates the exciting new therapeutic possibilities that will be studied in future intergroup (and possibly, international) trials that have the very real potential for improving DFS for these HR patients.

A 30-year-old woman presents with fatigue, easy bruising, shortness of breath, and pancytopenia. A BM biopsy is consistent with precursor B-cell ALL. Cytogenetics demonstrates t(9;22)(q34;q11) with a p190 BCR-ABL1 transcript. How should this patient be treated?

The presence of t(9;22)(q34;q11), the Philadelphia chromosome (Ph+), resulting in the BCR-ABL1 fusion gene, increases with age, and occurs in up to 25% to 30% of older adults, although it is less common in younger adults.8  Although Ph+ ALL has historically been associated with poor survival, the addition of tyrosine kinase inhibitors (TKIs) has dramatically improved outcomes for patients of all ages.80,81 

Our current approach for these patients is the addition of a TKI to induction therapy with early CNS-directed therapy. The TKI should be given continuously during induction and all postremission treatment courses. Although imatinib, dasatinib, and nilotinib have all been used effectively and have improved DFS in Ph+ ALL, we typically use dasatinib because of the potential for increased CNS penetration.82  Several different induction regimens added to the TKI have been tested successfully in AYAs, ranging from intensive induction (a BFM-like regimen or hyper-CVAD plus imatinib or dasatinib)80,81,83  to minimal therapy. Data from the Italian Cooperative Group demonstrated that CR rates >90% can be achieved with dasatinib and glucocorticoid therapy alone with no early mortality.84,85  Given the high CR rates and minimal toxicity of the low-intensity regimens, we currently favor enrollment of patients on a clinical trial utilizing this approach to induction therapy (NCT01256398).42  Importantly, we also typically evaluate patients who fail to respond during induction with dasatinib-based therapy for the presence of an abl kinase mutation, because resistant mutations occur more commonly in Ph+ ALL.86 

Induction of remission is typically followed by postremission CNS-directed systemic and intrathecal therapy (continuing the TKI) and allogeneic SCT in CR1 if a donor is available.87  This remains our current recommendation for the AYA patient with Ph+ ALL, based in part, on historical data (prior to TKIs) demonstrating that long-term survival for Ph+ ALL was only possible with allogeneic transplant.88  The power of achieving major or complete molecular remissions with the addition of a TKI to frontline therapy has resulted in prolonged DFS, even in patients who did not undergo allogeneic SCT in CR1.89  Several contemporary trials suggest that excellent DFS can be achieved (70% to 75%) with or without transplant if patients achieve molecular remissions following TKI (dasatinib or nilotinib), plus low-dose chemotherapy and intrathecal prophylaxis.84,90-92  Importantly, these studies still require longer follow up, but suggest that achievement of molecular remissions is a goal of treatment in Ph+ ALL and may obviate the need for allogeneic transplant for achievement of long-term survival. Interestingly, although not specifically designed or powered to evaluate the role of allogeneic transplant, a study by the COG group demonstrated no difference in EFS among 65 pediatric patients (aged 1 to 21 years) with Ph+ ALL who received imatinib plus intensive chemotherapy vs allogeneic BM transplant.83  Thus, the treatment of Ph+ ALL is rapidly evolving and, in addition to the transplant question, future studies may also evaluate the role of newer, more potent TKIs including ponatinib in frontline therapy,93  their impact on achievement of molecular remissions, and the importance and duration of TKI maintenance therapy, including their role following transplant.

A 27-year-old single mother, living apart from her immediate family, has been diagnosed with precursor B-cell ALL and is currently receiving maintenance therapy on the C10403 regimen. She recently lost her job, does not have health insurance, and has not been taking prophylactic medications because of cost. She missed several appointments because of lack of childcare for her 2-year-old daughter and forgot to take her oral methotrexate for the past several weeks. Depressed about her health, she worries about who will help take care of her daughter if her disease relapses.

Nonadherence to treatment regimens and missed appointments are a significant challenge in the AYA population. This is especially true with complicated and prolonged ALL regimens, where the majority of treatment is administered as an outpatient. Clinical trials of AYA patients with leukemia and lymphoma suggest that up to 63% of AYA patients have difficulties adhering to oral treatment regimens.94,95  Factors that affect treatment adherence include emotional factors (such as depression and self-esteem), patient health beliefs, and family environment. Although evidence-based interventions are lacking, several strategies have been suggested to improve treatment adherence, including anticipatory guidance, frequent monitoring of adherence, and interventions such as increasing availability of psychosocial support, modifying communication style, and allowing flexibility in treatment.94  Several groups are now exploring whether medication timing reminders and communication using electronic-based methods (eg, texting and web-based approaches) will facilitate treatment compliance and enhance patient satisfaction. We also encourage patients to keep a treatment diary, so that we can review and address ongoing concerns.

It has been suggested that AYAs face significantly greater challenges accessing health care due to insurance issues, including prescription drug coverage for crucial outpatient medications.96  Young adults with cancer are more likely to present with advanced stage or metastatic disease, be undertreated, and die after a diagnosis of cancer, relative to those who are insured.97  In countries with other health care systems and more comprehensive insurance coverage, this may be less of an issue, and, in the United States, we hope that the implementation of the Affordable Care Act will result in fewer uninsured AYAs and improvement in these statistics.

Perceived social support from family, friends, and health care providers is an important predictor of mental health and symptom distress.98  Support groups can also be a useful resource for adolescent and adult oncology patients and, although there is limited evidence,99  we have found them to be beneficial. A psychologist in our clinic also meets with our patients regularly and helps determine when a referral to psychiatry is warranted. Although the data are inconsistent, some studies have shown that mental health, depression, and anxiety are worse in AYA oncology patients at the time of diagnosis than in the general population.100  This improves as time progresses, but it highlights the importance of good psychological support for these patients.

Given the complexity of ALL treatment and the significant psycho-social and socio-economic challenges, we believe these patients are optimally treated in a supportive outpatient setting with expertise in management of the “whole” patient. At our institution, we have created an AYA cancer clinic that is composed of both pediatric and adult practitioners, as well as the resources and expertise to address the specific issues of a young adult with cancer. Prior to each clinic, we conduct a multidisciplinary meeting with nurses, physicians, pharmacists, social workers, physical therapists, and psychologists to discuss the patient, identify any issues, and determine the best plan of management for the patient. Even though these multidisciplinary teams have already become the standard of care in some countries,101,102  we believe this approach should become the standard for all AYA patients.

The future for AYAs with ALL is bright. Survival rates of 70% or greater are being reported in the recent AYA focused trials, and new targeted therapies individualized to optimize response and minimize toxicity are entering the clinic. Given the tremendous biological heterogeneity of this relatively rare disease and the complexity of the treatment approach, further improvements in survival will be achieved quickly if we can commit to offering these young adults enrollment on novel clinical trials and can provide the multidisciplinary expertise that will facilitate successful treatment outcomes. It “takes a village” to successfully treat ALL; and the AYA with ALL should be able to partake in the best scientific and supportive expertise that we have to offer!

E.C. was supported by grants from the National Institutes of Health/National Institute of General Medical Sciences Clinical Therapeutics grant (T32 GM007019) and the Basic Research Training in Medical Oncology grant (T32 CA009566). W.S. was supported by National Institutes of Health National Cancer Institute grant P30 CA14599-36.

Contribution: E.C. wrote the manuscript, and W.S. wrote the manuscript and supervised the project.

Conflict-of-interest disclosure: W.S. performs consultations for Amgen, Sigma-tau, Gilead, and Jazz Pharma; receives research funding from Sigma-tau; honoraria from the American Board of Internal Medicine (hematology board), the American Society of Hematology, and the National Cancer Institute (Leukemia steering committee). The remaining author declares no competing financial interests.

Correspondence: Wendy Stock, University of Chicago Comprehensive Cancer Center, 5841 S. Maryland Ave, M/C2115, Chicago, IL 60637; e-mail: wstock@medicine.bsd.uchicago.edu.

1
Siegel
 
R
Ma
 
J
Zou
 
Z
Jemal
 
A
Cancer statistics, 2014.
CA Cancer J Clin
2014
, vol. 
64
 
1
(pg. 
9
-
29
)
2
Pulte
 
D
Gondos
 
A
Brenner
 
H
Trends in survival after diagnosis with hematologic malignancy in adolescence or young adulthood in the United States, 1981-2005.
Cancer
2009
, vol. 
115
 
21
(pg. 
4973
-
4979
)
3
Pulte
 
D
Gondos
 
A
Brenner
 
H
Improvement in survival in younger patients with acute lymphoblastic leukemia from the 1980s to the early 21st century.
Blood
2009
, vol. 
113
 
7
(pg. 
1408
-
1411
)
4
Larson
 
RA
Dodge
 
RK
Burns
 
CP
, et al. 
A five-drug remission induction regimen with intensive consolidation for adults with acute lymphoblastic leukemia: cancer and leukemia group B study 8811.
Blood
1995
, vol. 
85
 
8
(pg. 
2025
-
2037
)
5
Stock
 
W
Johnson
 
JL
Stone
 
RM
, et al. 
Dose intensification of daunorubicin and cytarabine during treatment of adult acute lymphoblastic leukemia: results of Cancer and Leukemia Group B Study 19802 [published correction appears in Cancer. 2014;120(14):2222].
Cancer
2013
, vol. 
119
 
1
(pg. 
90
-
98
)
6
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
, vol. 
18
 
3
(pg. 
547
-
561
)
7
Harrison
 
CJ
Moorman
 
AV
Barber
 
KE
, et al. 
Interphase molecular cytogenetic screening for chromosomal abnormalities of prognostic significance in childhood acute lymphoblastic leukaemia: a UK Cancer Cytogenetics Group Study.
Br J Haematol
2005
, vol. 
129
 
4
(pg. 
520
-
530
)
8
Moorman
 
AV
Chilton
 
L
Wilkinson
 
J
Ensor
 
HM
Bown
 
N
Proctor
 
SJ
A population-based cytogenetic study of adults with acute lymphoblastic leukemia.
Blood
2010
, vol. 
115
 
2
(pg. 
206
-
214
)
9
Moorman
 
AV
Harrison
 
CJ
Buck
 
GA
, et al. 
Adult Leukaemia Working Party, Medical Research Council/National Cancer Research Institute
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
, vol. 
109
 
8
(pg. 
3189
-
3197
)
10
Yang
 
L
Panetta
 
JC
Cai
 
X
, et al. 
Asparaginase may influence dexamethasone pharmacokinetics in acute lymphoblastic leukemia.
J Clin Oncol
2008
, vol. 
26
 
12
(pg. 
1932
-
1939
)
11
Veal
 
GJ
Hartford
 
CM
Stewart
 
CF
Clinical pharmacology in the adolescent oncology patient.
J Clin Oncol
2010
, vol. 
28
 
32
(pg. 
4790
-
4799
)
12
Bleyer
 
A
Budd
 
T
Montello
 
M
Adolescents and young adults with cancer: the scope of the problem and criticality of clinical trials.
Cancer
2006
, vol. 
107
 
suppl 7
(pg. 
1645
-
1655
)
13
Fern
 
LA
Whelan
 
JS
Recruitment of adolescents and young adults to cancer clinical trials—international comparisons, barriers, and implications.
Semin Oncol
2010
, vol. 
37
 
2
(pg. 
e1
-
e8
)
14
Tai
 
E
Beaupin
 
L
Bleyer
 
A
Clinical trial enrollment among adolescents with cancer: supplement overview.
Pediatrics
2014
, vol. 
133
 
suppl 3
(pg. 
S85
-
S90
)
15
National Comprehensive Cancer Network
 
Adolescent and Young Adult Oncology Guidelines version 2.2015; September 18, 2014
16
Geiger
 
AM
Castellino
 
SM
Delineating the age ranges used to define adolescents and young adults.
J Clin Oncol
2011
, vol. 
29
 
16
(pg. 
e492
-
e493
)
17
Haïat
 
S
Marjanovic
 
Z
Lapusan
 
S
, et al. 
Outcome of 40 adults aged from 18 to 55 years with acute lymphoblastic leukemia treated with double-delayed intensification pediatric protocol.
Leuk Res
2011
, vol. 
35
 
1
(pg. 
66
-
72
)
18
Roberts
 
KG
Li
 
Y
Payne-Turner
 
D
, et al. 
Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia.
N Engl J Med
2014
, vol. 
371
 
11
(pg. 
1005
-
1015
)
19
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
, vol. 
111
 
4
(pg. 
1827
-
1833
)
20
Rytting
 
ME
Thomas
 
DA
O’Brien
 
SM
, et al. 
Augmented Berlin-Frankfurt-Münster therapy in adolescents and young adults (AYAs) with acute lymphoblastic leukemia (ALL).
Cancer
2014
, vol. 
120
 
23
(pg. 
3660
-
3668
)
21
Schrappe
 
M
Reiter
 
A
Zimmermann
 
M
, et al. 
Long-term results of four consecutive trials in childhood ALL performed by the ALL-BFM study group from 1981 to 1995. Berlin-Frankfurt-Münster.
Leukemia
2000
, vol. 
14
 
12
(pg. 
2205
-
2222
)
22
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
, vol. 
112
 
5
(pg. 
1646
-
1654
)
23
Boissel
 
N
Auclerc
 
MF
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
, vol. 
21
 
5
(pg. 
774
-
780
)
24
de Bont
 
JM
Holt
 
B
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
, vol. 
18
 
12
(pg. 
2032
-
2035
)
25
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
, vol. 
48
 
3
(pg. 
254
-
261
)
26
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
, vol. 
25
 
7
(pg. 
813
-
819
)
27
Nachman
 
JB
Sather
 
HN
Sensel
 
MG
, et al. 
Augmented post-induction therapy for children with high-risk acute lymphoblastic leukemia and a slow response to initial therapy.
N Engl J Med
1998
, vol. 
338
 
23
(pg. 
1663
-
1671
)
28
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
, vol. 
27
 
6
(pg. 
911
-
918
)
29
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
, vol. 
26
 
11
(pg. 
1843
-
1849
)
30
Rijneveld
 
AW
van der Holt
 
B
Daenen
 
SM
, et al. 
Dutch-Belgian HOVON Cooperative group
Intensified chemotherapy inspired by a pediatric regimen combined with allogeneic transplantation in adult patients with acute lymphoblastic leukemia up to the age of 40.
Leukemia
2011
, vol. 
25
 
11
(pg. 
1697
-
1703
)
31
Hocking
 
J
Schwarer
 
AP
Gasiorowski
 
R
, et al. 
Excellent outcomes for adolescents and adults with acute lymphoblastic leukemia and lymphoma without allogeneic stem cell transplant: the FRALLE-93 pediatric protocol.
Leuk Lymphoma
2014
, vol. 
55
 
12
(pg. 
2801
-
2807
)
32
DeAngelo
 
DJ
Stevenson
 
KE
Dahlberg
 
SE
, et al. 
Long-term outcome of a pediatric-inspired regimen used for adults aged 18-50 years with newly diagnosed acute lymphoblastic leukemia.
Leukemia
2015
, vol. 
29
 
3
(pg. 
526
-
534
)
33
Gokbuget
 
N
Beck
 
J
Brandt
 
K
, et al. 
Significant improvement of outcome in adolescents and young adults aged 15-35 years with acute lymphoblastic leukemia 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
, vol. 
122
 
21
 
Abstract 839
34
Stock
 
W
Luger
 
S
Advani
 
A
, et al. 
Favorable outcomes for older adolescents and young adults (AYA) with acute lymphoblastic leukemia (ALL): early results of U.S. Intergroup Trial C10403 [abstract].
Blood (ASH Annual Meeting Abstracts)
2014
, vol. 
124
 
21
 
Abstract 796
35
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
, vol. 
87
 
5
(pg. 
472
-
478
)
36
Advani
 
AS
Sanford
 
B
Luger
 
S
, et al. 
Frontline-treatment of acute lymphoblastic leukemia (ALL) in older adolescents and young adults (AYA) using a pediatric regimen is feasible: toxicity results of the prospective US Intergroup Trial C10403 (Alliance) [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 3903
37
Larsen
 
EC
Salzer
 
WL
Devidas
 
M
, et al. 
Comparison of high-dose methotrexate (HD-MTX) with Capizzi methotrexate plus asparaginase (C-MTX/ASNase) in children and young adults with high-risk acute lymphoblastic leukemia (HR-ALL): a report from the Children’s Oncology Group Study AALL0232 [abstract].
J Clin Oncol
2011
, vol. 
29
 
suppl 18
 
Abstract 3
38
Wolfson
 
JA
Sun
 
C-L
Kang
 
T
Wyatt
 
L
Hurria
 
A
Bhatia
 
S
Impact of care at NCI comprehensive cancer centers (NCICCC) on cancer outcome: results from a population-based study [abstract].
J Clin Oncol
2014
, vol. 
32
 
suppl 5
 
Abstract 6541
39
Kantarjian
 
H
Thomas
 
D
Jorgensen
 
J
, et al. 
Results of inotuzumab ozogamicin, a CD22 monoclonal antibody, in refractory and relapsed acute lymphocytic leukemia.
Cancer
2013
, vol. 
119
 
15
(pg. 
2728
-
2736
)
40
DeAngelo
 
DJ
Stock
 
W
Shustov
 
AR
, et al. 
Weekly inotuzumab ozogamicin (InO) in adult patients with relapsed or refractory CD22-positive acute lymphoblastic leukemia (ALL) [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 3906
41
Topp
 
M
Goekbuget
 
N
Stein
 
A
, et al. 
Confirmatory open-label, single-arm, multicenter phase 2 study of the BiTE antibody blinatumomab in patients (pts) with relapsed/refractory B-precursor acute lymphoblastic leukemia (r/r ALL) [abstract].
J Clin Oncol
2014
, vol. 
32
 
suppl 5
 
Abstract 7005
42
US National Institutes of Health. Available at: www.clinicaltrials.gov. Accessed April 6, 2015
43
Vrooman
 
LM
Stevenson
 
KE
Supko
 
JG
, et al. 
Postinduction dexamethasone and individualized dosing of Escherichia Coli L-asparaginase each improve outcome of children and adolescents with newly diagnosed acute lymphoblastic leukemia: results from a randomized study—Dana-Farber Cancer Institute ALL Consortium Protocol 00-01.
J Clin Oncol
2013
, vol. 
31
 
9
(pg. 
1202
-
1210
)
44
Tong
 
WH
Pieters
 
R
Kaspers
 
GJ
, et al. 
A prospective study on drug monitoring of PEGasparaginase and Erwinia asparaginase and asparaginase antibodies in pediatric acute lymphoblastic leukemia.
Blood
2014
, vol. 
123
 
13
(pg. 
2026
-
2033
)
45
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
, vol. 
109
 
7
(pg. 
2744
-
2750
)
46
Stock
 
W
Douer
 
D
DeAngelo
 
DJ
, et al. 
Prevention and management of asparaginase/pegasparaginase-associated toxicities in adults and older adolescents: recommendations of an expert panel.
Leuk Lymphoma
2011
, vol. 
52
 
12
(pg. 
2237
-
2253
)
47
Seftel
 
MD
Neuberg
 
D
Zhang
 
M-J
, et al. 
Superiority of pediatric chemotherapy over allogeneic hematopoietic cell transplantation for Philadelphia chromosome negative adult ALL in first complete remission: a combined analysis of Dana-Farber ALL Consortium and CIBMTR Cohorts [abstract].
Blood (ASH Annual Meeting Abstracts)
2014
, vol. 
124
 
21
 
Abstract 319
48
Vey
 
N
Thomas
 
X
Picard
 
C
, et al. 
GET-LALA Group the Swiss Group for Clinical Cancer Research (SAKK)
Allogeneic stem cell transplantation improves the outcome of adults with t(1;19)/E2A-PBX1 and t(4;11)/MLL-AF4 positive B-cell acute lymphoblastic leukemia: results of the prospective multicenter LALA-94 study.
Leukemia
2006
, vol. 
20
 
12
(pg. 
2155
-
2161
)
49
Nachman
 
JB
Heerema
 
NA
Sather
 
H
, et al. 
Outcome of treatment in children with hypodiploid acute lymphoblastic leukemia.
Blood
2007
, vol. 
110
 
4
(pg. 
1112
-
1115
)
50
Charrin
 
C
Thomas
 
X
Ffrench
 
M
, et al. 
A report from the LALA-94 and LALA-SA groups on hypodiploidy with 30 to 39 chromosomes and near-triploidy: 2 possible expressions of a sole entity conferring poor prognosis in adult acute lymphoblastic leukemia (ALL).
Blood
2004
, vol. 
104
 
8
(pg. 
2444
-
2451
)
51
Zhang
 
J
Ding
 
L
Holmfeldt
 
L
, et al. 
The genetic basis of early T-cell precursor acute lymphoblastic leukaemia.
Nature
2012
, vol. 
481
 
7380
(pg. 
157
-
163
)
52
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 (ASH Annual Meeting Abstracts)
2014
, vol. 
124
 
21
 
Abstract 1
53
Bassan
 
R
Spinelli
 
O
Oldani
 
E
, et al. 
Different molecular levels of post-induction minimal residual disease may predict hematopoietic stem cell transplantation outcome in adult Philadelphia-negative acute lymphoblastic leukemia.
Blood Cancer J
2014
, vol. 
4
 pg. 
e225
 
54
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
, vol. 
113
 
18
(pg. 
4153
-
4162
)
55
Gökbuget
 
N
Kneba
 
M
Raff
 
T
, et al. 
German Multicenter Study Group for Adult Acute Lymphoblastic Leukemia
Adult patients with acute lymphoblastic leukemia and molecular failure display a poor prognosis and are candidates for stem cell transplantation and targeted therapies.
Blood
2012
, vol. 
120
 
9
(pg. 
1868
-
1876
)
56
Brüggemann
 
M
Raff
 
T
Kneba
 
M
Has MRD monitoring superseded other prognostic factors in adult ALL?
Blood
2012
, vol. 
120
 
23
(pg. 
4470
-
4481
)
57
Dhedin
 
N
Huynh
 
A
Maury
 
S
, et al. 
Allogeneic hematopoietic stem cell transplantation (HSCT) in adults with Philadelphia chromosome (Ph)-negative acute lymphoblastic leukemia (ALL): results from The Group for Research on Adult ALL (GRAALL) [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 552
58
Pulsipher
 
MA
Carlson
 
CS
Mark
 
K
, et al. 
Striking predictive power for relapse and decreased survival associated with detectable minimal residual disease by IGH VDJ deep sequencing of bone marrow pre- and post-allogeneic transplant in children with B-lineage ALL: a subanalysis of the COG ASCT0431/PBMTC ONC051 Study [abstract].
Blood (ASH Annual Meeting Abstracts)
2013
, vol. 
122
 
21
 
Abstract 919
59
Zhang
 
M
Luo
 
Y
Lai
 
X
, et al. 
Minimal residual disease levels at time of CR1 and transplant predict outcome in Philadelphia chromosome-negative adult ALL [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 713
60
Patel
 
B
Rai
 
L
Buck
 
G
, et al. 
Minimal residual disease is a significant predictor of treatment failure in non T-lineage adult acute lymphoblastic leukaemia: final results of the international trial UKALL XII/ECOG2993.
Br J Haematol
2010
, vol. 
148
 
1
(pg. 
80
-
89
)
61
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
, vol. 
142
 
2
(pg. 
227
-
237
)
62
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
, vol. 
107
 
3
(pg. 
1116
-
1123
)
63
Faham
 
M
Zheng
 
J
Moorhead
 
M
, et al. 
Deep-sequencing approach for minimal residual disease detection in acute lymphoblastic leukemia.
Blood
2012
, vol. 
120
 
26
(pg. 
5173
-
5180
)
64
Malnassy
 
G
Carlton
 
V
Moorhead
 
M
Faham
 
M
Stock
 
W
Comparison of next-generation sequencing and ASO-PCR methods for MRD detection in acute lymphoblastic leukemia [abstract].
Haematologica
2013
, vol. 
98
 (pg. 
224
-
225
)
65
Schmiegelow
 
K
Heyman
 
M
Gustafsson
 
G
, et al. 
Nordic Society of Paediatric Haematology and Oncology (NOPHO)
The degree of myelosuppression during maintenance therapy of adolescents with B-lineage intermediate risk acute lymphoblastic leukemia predicts risk of relapse.
Leukemia
2010
, vol. 
24
 
4
(pg. 
715
-
720
)
66
Bhatia
 
S
Landier
 
W
Hageman
 
L
, et al. 
High intra-individual variability in systemic exposure to 6 mercaptopurine (6MP) in children with acute lymphoblastic leukemia (ALL) contributes to ALL relapse: results from a Children’s Oncology Group (COG) Study (AALL03N1) [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 59
67
McLeod
 
HL
Krynetski
 
EY
Relling
 
MV
Evans
 
WE
Genetic polymorphism of thiopurine methyltransferase and its clinical relevance for childhood acute lymphoblastic leukemia.
Leukemia
2000
, vol. 
14
 
4
(pg. 
567
-
572
)
68
Yang
 
JJ
Landier
 
W
Yang
 
W
, et al. 
Inherited NUDT15 variant is a genetic determinant of mercaptopurine intolerance in children with acute lymphoblastic leukemia [published online ahead of print January 26, 2015].
J Clin Oncol
69
te Winkel
 
ML
Pieters
 
R
Hop
 
WC
, et al. 
Prospective study on incidence, risk factors, and long-term outcome of osteonecrosis in pediatric acute lymphoblastic leukemia.
J Clin Oncol
2011
, vol. 
29
 
31
(pg. 
4143
-
4150
)
70
Nathan
 
PC
Wasilewski-Masker
 
K
Janzen
 
LA
Long-term outcomes in survivors of childhood acute lymphoblastic leukemia.
Hematol Oncol Clin North Am
2009
, vol. 
23
 
5
(pg. 
1065
-
1082, vi-vii
)
71
Diouf
 
B
Crews
 
K
Lew
 
G
, et al. 
Genome-wide association analyses identify susceptibility loci for vincristine-induced peripheral neuropathy in children with acute lymphoblastic leukemia [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 618
72
Kawedia
 
JD
Kaste
 
SC
Pei
 
D
, et al. 
Pharmacokinetic, pharmacodynamic, and pharmacogenetic determinants of osteonecrosis in children with acute lymphoblastic leukemia.
Blood
2011
, vol. 
117
 
8
(pg. 
2340
-
2347, quiz 2556
)
73
Mullighan
 
CG
Su
 
X
Zhang
 
J
, et al. 
Children’s Oncology Group
Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia.
N Engl J Med
2009
, vol. 
360
 
5
(pg. 
470
-
480
)
74
Payne-Turner
 
D
Pei
 
D
Becksfort
 
J
, et al. 
Integrated genomic and mutational profiling of adolescent and young adult ALL identifies a high frequency of BCR-ABL1-like ALL with very poor outcome [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 825
75
Roberts
 
KG
Pei
 
D
Campana
 
D
, et al. 
Outcomes of children with BCR-ABL1–like acute lymphoblastic leukemia treated with risk-directed therapy based on the levels of minimal residual disease.
J Clin Oncol
2014
, vol. 
32
 
27
(pg. 
3012
-
3020
)
76
Harvey
 
RC
Kang
 
H
Roberts
 
KG
, et al. 
Development and validation of a highly sensitive and specific gene expression classifier to prospectively screen and identify B-precursor acute lymphoblastic leukemia (ALL) patients with a Philadelphia chromosome-like (“Ph-like” or “BCR-ABL1-like”) signature for therapeutic targeting and clinical intervention [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 826
77
Raca
 
G
Gurbuxani
 
S
Zhang
 
Z
, et al. 
RCSD1-ABL2 fusion resulting from a complex chromosomal rearrangement in high-risk B-cell acute lymphoblastic leukemia.
Leuk Lymphoma
2014
(pg. 
1
-
3
)
78
Chen
 
IM
Harvey
 
RC
Mullighan
 
CG
, et al. 
Outcome modeling with CRLF2, IKZF1, JAK, and minimal residual disease in pediatric acute lymphoblastic leukemia: a Children’s Oncology Group study.
Blood
2012
, vol. 
119
 
15
(pg. 
3512
-
3522
)
79
Weston
 
BW
Hayden
 
MA
Roberts
 
KG
, et al. 
Tyrosine kinase inhibitor therapy induces remission in a patient with refractory EBF1-PDGFRB-positive acute lymphoblastic leukemia.
J Clin Oncol
2013
, vol. 
31
 
25
(pg. 
e413
-
e416
)
80
Thomas
 
DA
Faderl
 
S
Cortes
 
J
, et al. 
Treatment of Philadelphia chromosome-positive acute lymphocytic leukemia with hyper-CVAD and imatinib mesylate.
Blood
2004
, vol. 
103
 
12
(pg. 
4396
-
4407
)
81
Ravandi
 
F
O’Brien
 
S
Thomas
 
D
, et al. 
First report of phase 2 study of dasatinib with hyper-CVAD for the frontline treatment of patients with Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia.
Blood
2010
, vol. 
116
 
12
(pg. 
2070
-
2077
)
82
Porkka
 
K
Koskenvesa
 
P
Lundán
 
T
, et al. 
Dasatinib crosses the blood-brain barrier and is an efficient therapy for central nervous system Philadelphia chromosome-positive leukemia.
Blood
2008
, vol. 
112
 
4
(pg. 
1005
-
1012
)
83
Schultz
 
KR
Bowman
 
WP
Aledo
 
A
, et al. 
Improved early event-free survival with imatinib in Philadelphia chromosome-positive acute lymphoblastic leukemia: a children’s oncology group study.
J Clin Oncol
2009
, vol. 
27
 
31
(pg. 
5175
-
5181
)
84
Chiaretti
 
S
Vitale
 
A
Elia
 
L
, et al. 
First results of the multicenter total therapy gimema LAL 1509 protocol for de novo adult Philadelphia chromosome positive (Ph+) acute lymphoblastic leukemia (ALL) patients [abstract].
Blood
2014
, vol. 
124
 
21
 
Abstract 797
85
Foà
 
R
Vitale
 
A
Vignetti
 
M
, et al. 
GIMEMA Acute Leukemia Working Party
Dasatinib as first-line treatment for adult patients with Philadelphia chromosome-positive acute lymphoblastic leukemia.
Blood
2011
, vol. 
118
 
25
(pg. 
6521
-
6528
)
86
Soverini
 
S
De Benedittis
 
C
Papayannidis
 
C
, et al. 
Drug resistance and BCR-ABL kinase domain mutations in Philadelphia chromosome-positive acute lymphoblastic leukemia from the imatinib to the second-generation tyrosine kinase inhibitor era: the main changes are in the type of mutations, but not in the frequency of mutation involvement.
Cancer
2014
, vol. 
120
 
7
(pg. 
1002
-
1009
)
87
Fielding
 
AK
Rowe
 
JM
Buck
 
G
, et al. 
UKALLXII/ECOG2993: addition of imatinib to a standard treatment regimen enhances long-term outcomes in Philadelphia positive acute lymphoblastic leukemia.
Blood
2014
, vol. 
123
 
6
(pg. 
843
-
850
)
88
Dombret
 
H
Gabert
 
J
Boiron
 
JM
, et al. 
Groupe d’Etude et de Traitement de la Leucémie Aiguë Lymphoblastique de l’Adulte (GET-LALA Group)
Outcome of treatment in adults with Philadelphia chromosome-positive acute lymphoblastic leukemia—results of the prospective multicenter LALA-94 trial.
Blood
2002
, vol. 
100
 
7
(pg. 
2357
-
2366
)
89
Ravandi
 
F
Jorgensen
 
JL
Thomas
 
DA
, et al. 
Detection of MRD may predict the outcome of patients with Philadelphia chromosome-positive ALL treated with tyrosine kinase inhibitors plus chemotherapy.
Blood
2013
, vol. 
122
 
7
(pg. 
1214
-
1221
)
90
Ottmann
 
OG
Pfeifer
 
H
Cayuela
 
J-M
, et al. 
Nilotinib (Tasigna®) and chemotherapy for first-line treatment in elderly patients with de novo Philadelphia chromosome/BCR-ABL1 positive acute lymphoblastic leukemia (ALL): a Trial of the European Working Group for Adult ALL (EWALL-PH-02) [abstract].
Blood (ASH Annual Meeting Abstracts)
2014
, vol. 
124
 
21
 
Abstract 798
91
Kim
 
D-Y
Joo
 
YD
Kim
 
S-D
, et al. 
Nilotinib combined with multi-agent chemotherapy for adult patients with newly diagnosed Philadelphia chromosome-positive acute lymphoblastic leukemia: final results of prospective multicenter phase 2 study [abstract].
Blood (ASH Annual Meeting Abstracts)
2013
, vol. 
122
 
21
 
Abstract 55
92
Yanada
 
M
Matsuo
 
K
Suzuki
 
T
Naoe
 
T
Allogeneic hematopoietic stem cell transplantation as part of postremission therapy improves survival for adult patients with high-risk acute lymphoblastic leukemia: a metaanalysis.
Cancer
2006
, vol. 
106
 
12
(pg. 
2657
-
2663
)
93
Cortes
 
JE
Kim
 
DW
Pinilla-Ibarz
 
J
, et al. 
PACE Investigators
A phase 2 trial of ponatinib in Philadelphia chromosome-positive leukemias.
N Engl J Med
2013
, vol. 
369
 
19
(pg. 
1783
-
1796
)
94
Butow
 
P
Palmer
 
S
Pai
 
A
Goodenough
 
B
Luckett
 
T
King
 
M
Review of adherence-related issues in adolescents and young adults with cancer.
J Clin Oncol
2010
, vol. 
28
 
32
(pg. 
4800
-
4809
)
95
Kondryn
 
HJ
Edmondson
 
CL
Hill
 
J
Eden
 
TO
Treatment non-adherence in teenage and young adult patients with cancer.
Lancet Oncol
2011
, vol. 
12
 
1
(pg. 
100
-
108
)
96
Kantarjian
 
HM
O’Brien
 
S
Insurance policies in the United States may explain part of the outcome differences of adolescents and young adults with acute lymphoblastic leukemia treated on adult versus pediatric regimens.
Blood
2009
, vol. 
113
 
8
(pg. 
1861
-
, author reply 1862
)
97
Aizer
 
AA
Falit
 
B
Mendu
 
ML
, et al. 
Cancer-specific outcomes among young adults without health insurance.
J Clin Oncol
2014
, vol. 
32
 
19
(pg. 
2025
-
2030
)
98
Corey
 
AL
Haase
 
JE
Azzouz
 
F
Monahan
 
PO
Social support and symptom distress in adolescents/young adults with cancer.
J Pediatr Oncol Nurs
2008
, vol. 
25
 
5
(pg. 
275
-
284
)
99
Sansom-Daly
 
UM
Peate
 
M
Wakefield
 
CE
Bryant
 
RA
Cohn
 
RJ
A systematic review of psychological interventions for adolescents and young adults living with chronic illness.
Health Psychol
2012
, vol. 
31
 
3
(pg. 
380
-
393
)
100
Jörngården
 
A
Mattsson
 
E
von Essen
 
L
Health-related quality of life, anxiety and depression among adolescents and young adults with cancer: a prospective longitudinal study.
Eur J Cancer
2007
, vol. 
43
 
13
(pg. 
1952
-
1958
)
101
Haward
 
RA
The Calman-Hine report: a personal retrospective on the UK’s first comprehensive policy on cancer services.
Lancet Oncol
2006
, vol. 
7
 
4
(pg. 
336
-
346
)
102
Fleissig
 
A
Jenkins
 
V
Catt
 
S
Fallowfield
 
L
Multidisciplinary teams in cancer care: are they effective in the UK?
Lancet Oncol
2006
, vol. 
7
 
11
(pg. 
935
-
943
)
Sign in via your Institution