Abstract
The management of acute lymphoblastic leukemia (ALL) in older patients is challenging. Older patients often have multiple comorbidities and poor performance status, and disease factors associated with poor prognosis are more common in this age group. Patient and disease-related factors should be taken into account to determine whether intensive therapy is appropriate. The use of comorbidity indices and comprehensive geriatric assessment tools can be valuable in this setting. Fit patients should be considered for aggressive therapies including allogeneic hematopoietic stem cell transplantation, whereas low intensity options may be more suitable for the frail. The Philadelphia (Ph) chromosome is present in up to half of the cases of ALL in older patients. The incorporation of TK inhibitors into the treatment plans of older patients with Ph-positive ALL has improved the outcomes significantly. For less fit patients with Ph-positive ALL, the use of TK inhibitors with reduced-intensity chemotherapy or steroids alone results in high rates of remission, but, without further consolidation, relapses are inevitable. Many novel targeted and immunotherapeutic agents are being developed, offering more effective and tolerable treatment options.
Keywords: : acute lymphoblastic leukemia, blinatumomab, elderly, inotuzumab, Philadelphia chromosome, TK inhibitors
Practice points.
The outcomes of older patients with acute lymphoblastic leukemia (ALL) remain poor. Enrollment of these patients on appropriate clinical trials should always be encouraged.
Treatment with intensive chemotherapy is associated with high morbidity and mortality in older patients. Nonetheless, patients able to receive chemotherapy benefit from it.
The use of tyrosine kinase inhibitors (TKIs) has dramatically improved the outcomes of Philadelphia-positive ALL. Achievement of molecular remission should be the goal as it results in improved survival.
There are limited data on the role of allogeneic stem cell transplantation (SCT) in first remission in older patients with ALL, and its use remains controversial. Minimal residual disease assessment may become a useful tool to guide this decision.
For patients with Philadelphia-positive ALL, the use of autologous SCT in first remission might be associated with outcomes comparable to those achieved with allogeneic SCT.
The anti-CD19 bispecific T-cell engager antibody, blinatumomab, and the anti-CD22 antibody–drug conjugate, inotuzumab, are now approved for the treatment of patients with relapsed disease.
Approximately 20–30% of all acute lymphoblastic leukemia (ALL) cases occur in patients older than 55–60 years [1–4]. While the long term outcomes of ALL have improved significantly in the pediatric population and to a lesser extent in young adults, elderly patients still have a very poor prognosis [2,3,5]. Half of the deaths from ALL occur in patients older than 55 years and their 5-year overall survival (OS) rate is at or below 10–20% [1,2,5,6]. The age cutoff to define elderly may vary, but as adopted in most clinical trials, 55–60 years will be used here in this review. The relatively low-age cutoff reflects the substantial challenges in treating this patient population. This review article focuses mainly on studies related to older patients with ALL. If not available, studies that included both younger and older patients were included, with points/outcomes related to older patients highlighted. Randomized clinical trials and studies published in the last 5 years, whenever available, were emphasized.
Older patients with ALL commonly have multiple comorbidities and poor performance status and less often participate in clinical trials or receive intensive therapy [2,3,6]. As discussed later in this review, the inability to deliver optimal therapy to older patients and the higher induction death rate are important factors that contribute to the poor outcomes seen in older patients with ALL. However, even in studies looking at ‘fit’ patients treated with intensive chemotherapy, older age was associated with worse outcomes highlighting different disease biology in this group [2,7]. Factors associated with poor outcomes are more common in elderly patients, and include poor-risk cytogenetics such as the Philadelphia (Ph) chromosome, B-cell immunophenotype and secondary ALL [2].
Approach to treatment of elderly patients with ALL
The first step in treating older patients with ALL is to assess their ability to undergo aggressive therapy. Chronological age alone does not necessarily provide enough information regarding an individual's fitness or frailty, and other important determinants have to be considered. These include the presence of medical comorbidities, cognitive decline, polypharmacy, poor functional status, malnutrition, depression, and lack of social support [8]. Comorbidity burden can be measured by standardized indices such as the Charlson comorbidity index and the hematopoietic cell transplantation comorbidity index. In addition, there are various geriatric assessment tools that incorporate assessment of multiple other domains including cognitive and social function [9]. These have shown to provide valuable information in predicting treatment-related toxicities when applied in older patients with acute myeloid leukemia treated with intensive chemotherapy regimens [10–12]. Patients deemed unfit for intensive chemotherapy can be considered for less intensive approaches or supportive care only. Enrollment on appropriate clinical trials should always be encouraged, especially for this age group, given the paucity of data and the very poor outcomes achieved with current therapies.
Induction
Most ALL induction regimens include vincristine, a corticosteroid, and an anthracycline as a backbone, with other agents such as methotrexate, cytarabine, and cyclophosphamide often added. Asparaginase is also used in many regimens after it demonstrated efficacy in the pediatric population. Pegylated-asparaginase has replaced native Escherichia coli asparaginase as it offers a longer half-life and a lower risk of antiasparaginase antibody formation [13]. Asparaginase is typically given at lower doses in older patients, given its considerable toxicity, especially hepatotoxicity, pancreatitis and thrombosis [14–16].
Data have demonstrated that elderly patients may derive a benefit from intensive chemotherapy, although less than half of them are able to receive it [2,3,6]. In an analysis of 100 patients with ALL between the ages of 55 and 65 years treated with intensive chemotherapy, drug-dose reductions, omissions or delays occurred in approximately 50% of patients [14]. Intensive multiagent chemotherapy regimens in older populations achieve complete response (CR) rates of around 50–70%, with treatment-related mortality rates approaching 20–40% (Table 1) [1,2,6,17,18].
Table 1. . Selected studies of induction chemotherapy regimens in older patients with acute lymphoblastic leukemia.
Year† | Group/study | Age | No. of patients | CR rate % | Induction/early death rate % | OS rate % | ||
---|---|---|---|---|---|---|---|---|
1 year | 3 years | 5 years | ||||||
2000 | MD Anderson [69] | >60 | 44 | 79 | 16 | 17 | ||
2001 | SWOG 8419 [70] | 50–84 | 85 | 41 | 37 | <10 | ||
2002 | GIMEMA 0208 [71] | 50–60 | 121 | 68 | – | 15 | ||
2005 | CALGB [72] | >60 | 129 | 57 | – | 12 | ||
2007 | PETHEMA ALL96‡ [17] | 56–67 | 33 | 58 | 36 | 39 | ||
2008 | SWOG 9400 [73] | 50–65 | 43 | 63 | – | 23 | ||
2008 | EWALL [74] | 56–73 | 40 | 85 | 0 | 61 | ||
2012 | MRC UKALL XII/ECOG2993 [14] | 55–64 | 100 | 70 | 18 | 21 | ||
2012 | GMALL‡ [18] | 55–85 | 268 | 76 | 14 | 23 | ||
2016 | Multicenter [39] | 51–72 | 30 | 67 | 3 | 63 | ||
2016 | PETHEMA, ALLOLD07‡ [75] | 56–79 | 54 | 74 | 13 | 50 |
†Year of study publication.
‡Included patients with Ph-negative ALL only.
ALL: Acute lymphoblastic leukemia; CR: Complete response; OS: Overall survival; Ph: Philadelphia.
The GMALL group conducted the largest prospective trial in 268 older patients with newly-diagnosed Ph-negative ALL, using a dose-reduced pediatric-based (Berlin–Frankfurt–Munster) induction regimen [18]. The median age was 68 years, with 51% of patients aged between 66 and 75 years and 10% above 75 years. The induction regimen was divided into two phases (idarubicin, vincristine, and dexamethasone in Phase I, and cyclophosphamide and cytarabine in Phase II), followed by six alternating consolidation courses of intermediate-dose methotrexate and asparaginase with high-dose cytarabine for 1 year, and maintenance therapy up to 2 years. Eight treatments with rituximab were added for patients with CD20+ disease. The overall CR and early death rates were 76 and 14%, respectively (the CR and early death rates for the different age groups were: 84 and 7% in patients aged 55–65 years, 74 and 14% in patients aged 66–75 years, and 52 and 37% in patients older than 75 years). The 5-year OS rate was 23%. In a large single-center retrospective study, MD Anderson reported their experience with 122 older (≥60 years) patients with ALL treated with eight alternating courses of hyper-CVAD (cyclophosphamide, vincristine, doxorubicin and dexamethasone) with high doses of methotrexate and cytarabine followed by maintenance with 6-mercaptopurine, vincristine, methotrexate, and prednisone (POMP), and compared their outcomes with 34 older patients who received less intensive regimens and 409 younger patients treated with hyper-CVAD [19]. The CR rate was 84% for older patients treated with hyper-CVAD, 59% for older patients treated with less intensive regimens, and 92% for younger patients treated with hyper-CVAD, with treatment related mortality rates of 10, 12 and 2%, respectively. Infections were the major cause of death. In patients treated with hyper-CVAD, death in CR occurred in 34% of older patients and 7% of younger patients. The 5-year OS rates were 20, 9, and 48%, respectively [19].
Given the pronounced myelosuppression from both ALL and disease-directed therapy, particularly in older patients, supportive therapy with blood product transfusions, granulocyte colony-stimulating factor (CSF) and prophylactic antibiotics is a crucial component of ALL induction therapies. The use of granulocyte-CSF is associated with a shorter duration of neutropenia, a lower rate of neutropenic fever and a higher CR rate, but with no significant effect on survival [20].
CNS prophylaxis
As the CNS is a sanctuary site for ALL, prophylaxis with intrathecal chemotherapy given at intervals throughout induction and postremission therapy is an essential part of ALL treatment protocols. Methotrexate, cytarabine and steroids are the most commonly used intrathecal agents for CNS prophylaxis. In contrast to other TK inhibitors (TKIs), dasatinib crosses the blood–brain barrier; however, it results in low CSF levels, and prophylaxis with intrathecal chemotherapy is still recommended [21–23].
Ph-positive ALL
Approximately half of the elderly patients with ALL have the Ph chromosome, making it the most common cytogenetic abnormality in this age group [24,25]. While the Ph chromosome has historically been associated with worse outcomes, this has improved with the introduction of the TKIs [25,26]. For example, a Swedish population-based study of 155 older patients with ALL showed no evidence of worse outcomes for those with Ph-positive ALL [26]. Further, in a multicenter, retrospective study of 98 elderly patients, those with Ph-positive ALL (n = 47) treated with induction chemotherapy and TKI (87% with imatinib) had superior OS compared with patients with Ph-negative ALL treated with similar chemotherapy alone (median 12 vs 8 months; p = 0.037) [25].
For patients unfit for intensive chemotherapy, TKIs can be combined with steroids alone or with low-intensity chemotherapy regimens. Most patients achieve complete hematologic responses with such an approach but remissions are usually short-lived (Table 2) [27,28]. TKIs also provide deeper responses and better outcomes when combined with intensive chemotherapy regimens [29]. In a study by the GMALL group of 55 elderly patients with Ph-positive ALL, induction with imatinib was associated with significantly higher CR rates than with multiagent chemotherapy (96 vs 50%) and had lower toxicity [30]. In the LAL0201 trial, 30 newly diagnosed elderly patients with Ph-positive ALL (median age 69 years, range 61–83) received induction with imatinib plus prednisone for 45 days. Imatinib was then continued in responding patients. All evaluable patients achieved complete hematologic responses. The 1-year OS and disease-free survival (DFS) rates were 74 and 48%, respectively. Treatment was well tolerated with no major toxicities [27]. The GRAAPH-2005 study randomized 268 patients younger than 60 years with Ph-positive ALL to receive imatinib combined with hyper-CVAD or imatinib combined with dexamethasone and vincristine, with both groups receiving high-dose methotrexate and cytarabine in combination with imatinib as a second cycle. Higher CR rates were achieved with imatinib in combination with low-intensity chemotherapy (98 vs 91%), with no significant difference in event-free survival (EFS) and OS at 3 years between the two groups [31].
Table 2. . Selected studies in older patients with Ph-positive acute lymphoblastic leukemia.
Year† | Group/study | Treatment | Age | No. of patients | CR rate % | OS rate % | |
---|---|---|---|---|---|---|---|
Induction | Postinduction | ||||||
2006 | GRAALL AFR09 [76] | CTX | Imatinib + CTX | 58–78 | 30 | 72 | 66 (1 year) |
2007 | GIMEMA LAL0201-B [27] | Imatinib + pred | Imatinib + PC | 61–83 | 30 | 100 | 74 (1 year) |
2007 | GMALL [30] | Imatinib | Imatinib + CTX | 54–79 | 28 | 96 | 57 (1.5 years) |
CTX | Imatinib + CTX | 58–78 | 27 | 50 | 41 (1.5 years) | ||
2011 | GIMEMA LAL1205 [28] | Dasatinib + pred | Dasatinib + PC | 24–77‡ | 54 | 100 | 69 (1.5 years) |
2015 | MD Anderson [22] | Dasatinib + CTX | Dasatinib + CTX | 21–80§ | 72 | 96 | 46 (5 years) |
2015 | MD Anderson [33,34] | Ponatinib + CTX | Ponatinib + CTX | 21–80 | 64 | 98 | 76 (3 years) |
2016 | EWALL-PH-01 [32] | Dasatinib + CTX | Dasatinib + CTX | 59–83 | 71 | 96 | 36 (5 years) |
†Year of study publication.
‡12 patients >60 years.
§46 patients >50 years.
ALL: Acute lymphoblastic leukemia; CR: Complete response; CTX: Chemotherapy; OS: Overall survival; PC: Physician choice; Pred: Prednisone.
Clinical trials using the second-generation TKI, dasatinib, in patients with newly diagnosed Ph-positive ALL followed. In the LAL1205 study, 53 patients were treated with dasatinib for 12 weeks plus prednisone (12 patients were older than 60 years). All patients achieved complete hematologic response with more than half of them achieving major molecular response (MMR) by the end of the treatment. The treatment was well tolerated but more than half of the patients relapsed by 2 years [28]. Subsequently, the EWALL-PH-01 study combined dasatinib with low-intensity chemotherapy (induction with vincristine and dexamethasone and consolidation with methotrexate, asparaginase and 6-mercaptopurine) in 71 patients (median age 69 years, range 59–83) [32]. Dasatinib was continued until relapse or death. The CR rate was 96% with 65% of the patients achieving MMR. The treatment-related mortality rate was 4%, while the 1-year relapse-free survival (RFS) was 58% and 5-year OS was 36% [32].
A single-institution study evaluated the combination of dasatinib with hyper-CVAD in 72 patients with a median age of 55 years (64% of patients were older than 50 years). Patients who achieved CR were started on maintenance therapy with dasatinib, vincristine, and prednisone. The combination was highly effective with a CR rate of 96% and a complete molecular response rate of 65%. The median DFS and OS were 31 and 47 months, respectively. However, more than half of the patients could not receive the eight planned induction chemotherapy cycles due to poor tolerance (43%) or early death (13%). Serious infections occurred in more than 60% of the patients during induction and in more than 90% during consolidation [22].
Ponatinib is a potent third generation TKI that has activity in patients harboring the T315I BCR-ABL mutation which has been implicated in the development of resistance to other TKIs at the time of relapse [22,32]. A single-center Phase II trial evaluated ponatinib in combination with hyper-CVAD in 64 patients (the median age was 48 years, range 21–80). The CR rate was 98% with remarkably high MMR and complete molecular response rates of 97 and 77%, respectively. The 3-year OS rate was 76%. Ponatinib dose had to be reduced due to serious vascular events (three cases of myocardial infarction with two deaths). Other serious toxicities included pancreatitis in 19% and other thrombotic events in 6% [33,34]. In a propensity score analysis of patients treated on the previously mentioned Phase II trials of hyper-CVAD with either dasatinib or ponatinib, the 3-year EFS and OS rates were significantly higher in patients treated with ponatinib (for EFS: 69 vs 46% and for OS: 83 vs 56%). Ponatinib achieved deeper molecular responses and higher minimal residual disease (MRD) negativity [35].
Postremission therapy
The optimal postremission treatment strategy for older patients with ALL is not well established. Options include consolidation/intensification plus maintenance chemotherapy, autologous hematopoietic stem cell transplantation (SCT) or allogeneic SCT. Toxicity and poor tolerance often preclude the use of intensive and prolonged postremission chemotherapy in older patients. Different protocols incorporate different chemotherapeutic agents but they are, in general, similar to those used for induction albeit given at lesser intensities. For patients with Ph-positive ALL, TKIs are typically continued indefinitely, particularly for those who do not proceed to allogeneic SCT.
Autologous SCT
For patients with Ph-negative ALL, autologous SCT is not recommended as postremission therapy as it has not been shown to be associated with better outcomes compared with chemotherapy alone or allogeneic SCT [36]. However, for Ph-positive ALL, incorporating TKIs with autologous SCT seems to be an effective treatment strategy and may achieve outcomes comparable to those achieved with allogeneic SCT. This was shown in a study of 59 patients with Ph-positive ALL with an upper age limit of 66 years who were treated with imatinib plus chemotherapy. Patients in CR were eligible to receive allogeneic SCT or autologous SCT followed by maintenance with imatinib. The 5-year OS was similar between the allogeneic SCT (n = 45) and autologous SCT plus maintenance (n = 9) groups [37]. Similarly, the GRAAPH-2005 study of patients younger than 60 years with Ph-positive ALL showed no significant difference in RFS and OS in patients who received autologous (n = 35) or allogeneic (n = 161) SCT [31]. A smaller study by the CALGB group demonstrated similar findings (19 patients received autologous and 15 allogeneic SCT) [38]. Treatment using a TKI in combination with low-intensity chemotherapy initially, then more intensified chemotherapy in subsequent cycles followed by consolidation with autologous SCT is emerging as a promising approach, but more data are still needed given the relatively small numbers of patients treated in clinical trials so far [31,38].
Allogeneic SCT
Few studies have looked at the role of consolidative allogeneic SCT in older patients with ALL. A small Phase II study of 30 patients with a median age of 58 years (range 51–72) showed no survival advantage for the 12 patients who received allogeneic SCT over consolidation with chemotherapy [39]. A retrospective study of 80 patients older than 40 years with Ph-negative ALL showed no difference in the 3-year DFS and OS between patients who received chemotherapy only or allogeneic SCT in first remission. While allogeneic SCT was associated with a lower risk of disease relapse, the benefit was abrogated by the higher nonrelapse mortality rate [40]. Similarly, a meta-analysis that included more than 2900 patients with Ph-negative ALL showed a survival benefit with matched sibling donor myeloablative allogeneic SCT only in patients younger than 35 years. The lack of benefit in the older patients was due to the associated high nonrelapse mortality rates [41]. The GRAAPH-2005 study previously mentioned showed a significant survival benefit with the use of allogeneic SCT versus chemotherapy alone in patients with Ph-positive ALL in first CR (hazard ratio = 0.64; p = 0.02); however, only patients younger than 60 years were included [31]. Similar studies in older patients with Ph-positive ALL are lacking.
The role of allogeneic SCT in patients with ALL in first remission remains a topic of debate. With the better outcomes achieved with current induction strategies, especially the use of TKIs in Ph-positive ALL, more patients are able to achieve remission and with less toxicity [31]. In addition, the use of reduced intensity conditioning regimens and the improvement in supportive care have extended the base of eligibility for transplantation [31]. On the other hand, the limited data available do not show a survival advantage with the use of allogeneic SCT for older patients [39–41]. MRD assessment is emerging as an important tool for risk stratification and determining the need for allogeneic SCT. Two studies of younger patients (less than 55–60 years of age) with high risk, Ph-negative ALL in first CR have indicated that allogeneic SCT might not be needed in patients with negative MRD [42,43]. Such studies are lacking in patients with Ph-positive ALL, but the deep remissions reported with the combination of chemotherapy and TKIs suggest the same [35], though this is yet to be confirmed. However, it is important to keep in mind that the definition of negative MRD and when to test for it are not yet standardized, and that older patients were not included in these studies. MRD assessment has become a key element in ALL clinical trials, and its role in treatment selection remains an area of active research.
Treatment of relapsed disease
The relapse rate for patients with ALL remains high and achieving a second remission is difficult [44]. This is even more challenging for elderly patients where the treatment options with conventional chemotherapy are very limited by the poor outcomes and excessive toxicities [44]. Novel immune-based therapies targeting various surface antigens expressed on ALL cells have demonstrated very encouraging results in this setting and have changed the treatment paradigm of ALL.
Anti-CD19 bispecific t-cell engager: blinatumomab
Blinatumomab is a bispecific T-cell engager antibody against CD3 and CD19 that is designed to direct cytotoxic T cells to CD19-expressing B cells [45]. Blinatumomab is currently approved by the US FDA for relapsed/refractory Ph-negative and Ph-positive ALL, and the EMA for relapsed/refractory Ph-negative ALL. The large Phase III multicenter TOWER study compared blinatumomab with standard-of-care chemotherapy in patients with relapsed/refractory Ph-negative B cell ALL. The mean age was 41 years (range 18–80). Blinatumomab resulted in significantly higher remission rates (44 vs 25%) and improvement in 6-month EFS (31 vs 12%) and OS (7.7 vs 4 months) [46]. The single-arm, multicenter ALCANTARA trial evaluated the role of blinatumomab in 45 patients with Ph-positive ALL who had relapsed or were refractory to TKIs. Half of the patients were older than 55 years. The CR rate was 36% with 88% of responders achieving MRD negativity. The median duration of response was 6.7 months [47].
Notably, blinatumomab seems to have similar clinical efficacy irrespective of age. In a pooled analysis from two Phase II studies of patients with relapsed/refractory B cell ALL treated with blinatumomab, older patients had CR/CR with incomplete hematologic recovery (CRh) and MRD negativity rates similar to younger patients [48]. Cytokine release syndrome and neurological toxicities including encephalopathy and seizures are the main serious toxicities of blinatumomab, and are more common in older patients [47,48].
With these very promising results in patients with relapsed/refractory B-cell ALL, blinatumomab is currently being studied in the upfront setting. An ongoing Phase III randomized trial (ECOG E1910, NCT02003222) is evaluating the role of consolidation with blinatumomab following induction with intensive chemotherapy in patients between the ages of 30 and 70 years with newly diagnosed Ph-negative ALL. A Phase II trial (SWOG S1318, NCT02143414) is currently evaluating the role of frontline therapy with blinatumomab in patients 65 years or older who are not candidates for intensive chemotherapy. In this trial, patients with Ph-negative ALL receive induction and postremission therapy with blinatumomab followed by maintenance with POMP, while those with Ph-positive ALL receive induction with dasatinib and prednisone, postremission therapy with blinatumomab followed by maintenance with dasatinib and prednisone.
Anti-CD22 antibody–drug conjugate: inotuzumab
CD22 is expressed in more than 90% of patients with B-cell ALL making it an attractive therapeutic target [49]. Inotuzumab ozogamicin is an anti-CD22 monoclonal antibody conjugated to calicheamicin, a cytotoxic antibiotic. It is currently approved for the treatment of relapsed/refractory B cell ALL by the EMA and the FDA. The INO-VATE ALL trial was a large Phase III study that randomized patients with relapsed/refractory B-cell ALL to receive either inotuzumab or standard intensive chemotherapy [50]. Inotuzumab was shown to be associated with significantly higher CR rates (81 vs 29%), higher MRD negativity (for patients in CR: 78 vs 28%) and longer PFS (5 vs 2 months). Importantly, more than a third of the patients were older than 55 years, and in those treated with inotuzumab, the CR and MRD negativity rates were similar to those in younger patients [50]. In two single-center Phase II trials of patients with Ph-negative, CD22 positive ALL, inotuzumab was combined with lower-intensity or ‘mini’ hyper-CVAD with or without rituximab in the frontline and salvage settings [51,52]. In newly diagnosed patients who were 60 years or older (n = 43), the combination resulted in CR/CR with incomplete platelet recovery rate of 95% and MRD negativity in 96%. The 3-year OS was 54%, which compared favorably with historical controls treated with hyper-CVAD alone (3-year OS of 31%) [51]. In the salvage setting (n = 59, median age of 35 years, range 18–87), the overall response rate was 78% with 59% of the patients achieving CR. For patients in CR, the rate of MRD negativity was 82%. The median RFS and OS were 8 and 11 months, respectively. Using a propensity score analysis to compare the results with historical controls treated with inotuzumab monotherapy, the combination resulted in significantly higher overall response rate (75 vs 63%) and 1-year OS (43 vs 27%) [52]. Veno-occlusive liver disease has been reported in 10–15% of patients treated with inotuzumab, with higher risk in patients undergoing allogeneic SCT [50–52].
Other therapies
Vincristine sulfate liposome injection
Vincristine sulfate liposome injection (VSLI) is a modified formulation of vincristine with enhanced stability and better tissue penetration compared with the standard formulation [53,54]. Despite being given at a higher dose (2.25 mg/ml [2]) and without dose capping, its toxicities, mainly peripheral neuropathy and constipation, do not occur at a frequency higher than with the standard formulation. Further, it does not typically cause bone marrow suppression and is given on a once-per-week basis. In a Phase II trial of 65 patients with ALL with second or greater relapse, who were previously treated with standard vincristine, the CR/CRh rate with VSLI was 20%, with an overall response rate of 35% [55]. VSLI is used off label as monotherapy in the frontline setting for unfit patients.
Nelarabine
Nelarabine, a purine nucleoside analog, has single agent activity in both adults and children with T-cell ALL, and is currently approved in the USA for the treatment of patients with relapsed/refractory T cell ALL. In two Phase II trials of adult patients with relapsed/refractory T-cell ALL or lymphoblastic lymphoma treated with nelarabine monotherapy, the overall response rate was 41–46%, with a 1-year OS rate of 24–28% [56,57]. Neurotoxicity, including neuropathy, mental status change, and seizures, has been reported in up to 16% of patients, but is usually mild and reversible (grades 3 and 4 in less than 5%) [56,57]. In a Phase II trial of 40 adult patients with T-cell ALL or lymphoblastic lymphoma (median age of 38 years, range 19–78), the combination of nelarabine and hyper-CVAD as frontline therapy resulted in a CR rate of 91% with a 3-year DFS of 63% [58].
Anti-CD20 monoclonal antibodies: rituximab & ofatumumab
CD20 expression of more than 20% is found in approximately 30–40% of patients with B-ALL and is associated with a poor prognosis [49,59]. The anti-CD20 monoclonal antibody, rituximab, has shown clinical benefit when combined with intensive induction chemotherapy regimens in patients younger than 60 years with newly diagnosed, CD20 positive, Ph-negative ALL (4-year EFS of 55% with the combination vs 43% with chemotherapy alone) [60]. However, similar clinical trials are lacking in elderly patients. In fact, a single-center study of patients with CD20 positive ALL showed no benefit from adding rituximab to modified hyper-CVAD in patients older than 60 years when compared with historical controls treated with the standard hyper-CVAD regimen alone [61]. Rituximab is therefore not recommended as part of induction or maintenance therapy for elderly patients. Ofatumumab is a second generation anti-CD20 that binds with greater avidity than rituximab. A single-center Phase II study combined ofatumumab with hyper-CVAD in 55 patients with newly diagnosed CD20 positive ALL (median age 41 years, range 18–71). Interim analysis showed an MRD negativity in 93% of the patients and 3-year CR and OS rates of 78 and 68%, respectively [62]. These results compare favorably with those achieved historically with rituximab, and suggest a potential role for ofatumumab and other next generation anti-CD20 antibodies in the treatment of ALL.
Chimeric antigen receptor T cells
The chimeric antigen receptor (CAR) is composed primarily of a single-chain antibody specific to tumor antigen, fused to T-cell activation and co-stimulatory domains. The resulting receptor, when expressed on the surface of the T-cell, mediates binding to the target tumor antigen, independent of MHC recognition, and subsequently activates the T-cell and induces target cell lysis [63]. CAR T-cell therapy has been explored in multiple hematologic malignancies with the most mature data in ALL, where it has been approved by the FDA for the treatment of pediatric and young (<25 years) adult patients with relapsed/refractory B-cell ALL. The approval was based on the updated results of the Phase II clinical trial, ELIANA, of pediatric/young adult patients with relapsed/refractory B cell ALL treated with CTL019 (tisagenlecleucel) CAR T cells. Of the 75 evaluable patients, 81% achieved CR/CRh within 3 months of CAR T-cell infusion, with all patients achieving MRD negativity. The 1-year EFS and OS rates were 50 and 76%, respectively [64,65]. A Phase I trial in adult patients with heavily pretreated relapsed/refractory ALL treated with 19–28z CAR T cells has also shown very promising results, although the number of treated patients is relatively small and longer follow-up is needed [66]. Of the 53 patients treated with CAR T cells and evaluated for safety and efficacy (83 patients were enrolled onto the trial), 44 patients (83%) achieved CR, and 67% achieved MRD negativity (48 patients had sufficient bone marrow samples for evaluation of MRD status). With a median follow-up of 29 months, the median EFS and OS were 6 and 13 months, respectively. There was no significant difference in the rate of CR in the 8 patients older than 60 years compared with younger patients (the median age for treated patients was 44 years, range 23–74). Severe cytokine release syndrome and neurotoxic effects occurred in 26 and 42% of the patients, respectively, and were not more common in older patients [66].
Ph-like ALL
A subset of patients with Ph-negative ALL has a gene expression profile similar to those with Ph-positive ALL. This entity of Ph-like ALL is associated with poor outcomes [67,68]. The prevalence of Ph-like ALL in patients 60 years or older is around 25%. Kinase-activating alterations are present in the majority of patients and include CRLF2 rearrangements, ABL class fusions, JAK2 or EPOR rearrangements and other JAK-STAT pathway mutations [67,68]. Importantly, the activation of the ABL1 and JAK/STAT pathway might be a therapeutic target for several ABL inhibitors, such as imatinib and dasatinib and JAK kinase inhibitors, such as ruxolitinib. Clinical trials in this area are ongoing.
Conclusion & future perspective
The introduction of several novel antibody-based, targeted, and cellular therapies has changed the treatment paradigm of ALL, with many more agents targeting the diverse molecular and genetic alterations in ALL in development. Elderly patients, in particular, are in dire need for novel therapies as treatment with conventional chemotherapy has been associated with poor outcomes and unacceptable toxicities. The high remission rates achieved with second and third generation TKIs cast doubt on the need to use intensive chemotherapy and transplant in Ph-positive ALL, particularly for older patients. Several challenges exist including finding the appropriate sequencing of different therapies, knowing how to incorporate MRD assessment in the decision making process, and management of a new set of toxicities that might be more common in elderly patients. For these reasons, the need to enroll older patients with ALL on appropriate and well-designed clinical trials cannot be overstressed enough.
Footnotes
Financial & competing interests disclosure
Yazeed Sawalha declares no financial or competing interests. Anjali Advani declares consulting for and honoraria from Pfizer and Novartis, and research support from Amgen. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.
No writing assistance was utilized in the production of this manuscript.
Open access
This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/
References
- 1.Toft N, Schmiegelow K, Klausen TW, Birgens H. Adult acute lymphoblastic leukaemia in Denmark. A national population-based retrospective study on acute lymphoblastic leukaemia in Denmark 1998–2008. Br. J. Haematol. 2012;157(1):97–104. doi: 10.1111/j.1365-2141.2011.09020.x. [DOI] [PubMed] [Google Scholar]
- 2.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;115(2):206–214. doi: 10.1182/blood-2009-07-232124. [DOI] [PubMed] [Google Scholar]
- 3.Dinmohamed AG, Szabo A, van der Mark M, et al. Improved survival in adult patients with acute lymphoblastic leukemia in the Netherlands: a population-based study on treatment, trial participation and survival. Leukemia. 2016;30(2):310–317. doi: 10.1038/leu.2015.230. [DOI] [PubMed] [Google Scholar]
- 4.Howlader NNA, Krapcho M, Miller D, et al. SEER Cancer Statistics Review, 1975–2014. National Cancer Institute; MD, USA: https://seer.cancer.gov/csr/1975_2014/ [Google Scholar]
- 5.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;113(7):1408–1411. doi: 10.1182/blood-2008-06-164863. [DOI] [PubMed] [Google Scholar]
- 6.Legrand O, Marie JP, Marjanovic Z, et al. Prognostic factors in elderly acute lymphoblastic leukaemia. Br. J. Haematol. 1997;97(3):596–602. doi: 10.1046/j.1365-2141.1997.952909.x. [DOI] [PubMed] [Google Scholar]
- 7.Juliusson G, Karlsson K, Hallböök H. Population-based analyses in adult acute lymphoblastic leukemia. Blood. 2010;116(6):1011. doi: 10.1182/blood-2010-03-272724. [DOI] [PubMed] [Google Scholar]
- 8.Extermann M, Hurria A. Comprehensive geriatric assessment for older patients with cancer. J. Clin. Oncol. 2007;25(14):1824–1831. doi: 10.1200/JCO.2007.10.6559. [DOI] [PubMed] [Google Scholar]
- 9.Li D, Soto-Perez-de-Celis E, Hurria A. Geriatric assessment and tools for predicting treatment toxicity in older adults with cancer. Cancer J. 2017;23(4):206–210. doi: 10.1097/PPO.0000000000000269. [DOI] [PubMed] [Google Scholar]
- 10.Klepin HD, Geiger AM, Tooze JA, et al. The feasibility of inpatient geriatric assessment for older adults receiving induction chemotherapy for acute myelogenous leukemia. J. Am. Geriatr. Soc. 2011;59(10):1837–1846. doi: 10.1111/j.1532-5415.2011.03614.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Klepin HD, Geiger AM, Tooze JA, et al. Geriatric assessment predicts survival for older adults receiving induction chemotherapy for acute myelogenous leukemia. Blood. 2013;121(21):4287–4294. doi: 10.1182/blood-2012-12-471680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Sherman AE, Motyckova G, Fega KR, et al. Geriatric assessment in older patients with acute myeloid leukemia: a retrospective study of associated treatment and outcomes. Leuk. Res. 2013;37(9):998–1003. doi: 10.1016/j.leukres.2013.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.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. doi: 10.1182/blood-2006-07-035006. [DOI] [PubMed] [Google Scholar]
- 14.Sive JI, Buck G, Fielding A, et al. Outcomes in older adults with acute lymphoblastic leukaemia (ALL): results from the international MRC UKALL XII/ECOG2993 trial. Br. J. Haematol. 2012;157(4):463–471. doi: 10.1111/j.1365-2141.2012.09095.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Patel B, Kirkwood AA, Dey A, et al. Pegylated-asparaginase during induction therapy for adult acute lymphoblastic leukaemia: toxicity data from the UKALL14 trial. Leukemia. 2016;31:58. doi: 10.1038/leu.2016.219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.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;52(12):2237–2253. doi: 10.3109/10428194.2011.596963. [DOI] [PubMed] [Google Scholar]
- 17.Sancho JM, Ribera JM, Xicoy B, et al. Results of the PETHEMA ALL-96 trial in elderly patients with Philadelphia chromosome-negative acute lymphoblastic leukemia. Eur. J. Haematol. 2007;78(2):102–110. doi: 10.1111/j.1600-0609.2006.00778.x. [DOI] [PubMed] [Google Scholar]
- 18.Goekbuget N, Beck J, Brueggemann M, et al. Moderate intensive chemotherapy including CNS-prophylaxis with liposomal cytarabine is feasible and effective in older patients with Ph-negative acute lymphoblastic leukemia (ALL): results of a prospective trial from the German multicenter study group for adult ALL (GMALL) Blood. 2012;120(21):1493. [Google Scholar]
- 19.O'Brien S, Thomas DA, Ravandi F, Faderl S, Pierce S, Kantarjian H. Results of the hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone regimen in elderly patients with acute lymphocytic leukemia. Cancer. 2008;113(8):2097–2101. doi: 10.1002/cncr.23819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Larson RA, Dodge RK, Linker CA, et al. A randomized controlled trial of filgrastim during remission induction and consolidation chemotherapy for adults with acute lymphoblastic leukemia: CALGB study 9111. Blood. 1998;92(5):1556–1564. [PubMed] [Google Scholar]
- 21.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;112(4):1005–1012. doi: 10.1182/blood-2008-02-140665. [DOI] [PubMed] [Google Scholar]
- 22.Ravandi F, O'Brien SM, Cortes JE, et al. Long-term follow-up of a Phase II study of chemotherapy plus dasatinib for the initial treatment of patients with Philadelphia chromosome-positive acute lymphoblastic leukemia. Cancer. 2015;121(23):4158–4164. doi: 10.1002/cncr.29646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Wieduwilt MJ, Yin J, Wetzler M, et al. A Phase II study of dasatinib and dexamethasone as primary therapy followed by hematopoietic cell transplantation for adults with Philadelphia chromosome-positive acute lymphoblastic leukemia: CALGB Study 10701 (Alliance) Blood. 2016;128(22):2782. [Google Scholar]
- 24.Burmeister T, Schwartz S, Bartram CR, Gokbuget N, Hoelzer D, Thiel E. Patients’ age and BCR-ABL frequency in adult B-precursor ALL: a retrospective analysis from the GMALL study group. Blood. 2008;112(3):918–919. doi: 10.1182/blood-2008-04-149286. [DOI] [PubMed] [Google Scholar]
- 25.Byun JM, Koh Y, Shin DY, et al. Korean Adult ALL Working Party, Korean Society of Hematology. BCR-ABL translocation as a favorable prognostic factor in elderly patients with acute lymphoblastic leukemia in the era of potent tyrosine kinase inhibitors. Haematologica. 2017;102(5):e187–e190. doi: 10.3324/haematol.2016.159988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kozlowski P, Lennmyr E, Ahlberg L, et al. Age but not Philadelphia positivity impairs outcome in older/elderly patients with acute lymphoblastic leukemia in Sweden. Eur. J. Haematol. 2017;99(2):141–149. doi: 10.1111/ejh.12896. [DOI] [PubMed] [Google Scholar]
- 27.Vignetti M, Fazi P, Cimino G, et al. Imatinib plus steroids induces complete remissions and prolonged survival in elderly Philadelphia chromosome-positive patients with acute lymphoblastic leukemia without additional chemotherapy: results of the Gruppo Italiano Malattie Ematologiche dell'Adulto (GIMEMA) LAL0201-B protocol. Blood. 2007;109(9):3676–3678. doi: 10.1182/blood-2006-10-052746. [DOI] [PubMed] [Google Scholar]
- 28.Foà R, Vitale A, Vignetti M, et al. Dasatinib as first-line treatment for adult patients with Philadelphia chromosome–positive acute lymphoblastic leukemia. Blood. 2011;118(25):6521–6528. doi: 10.1182/blood-2011-05-351403. [DOI] [PubMed] [Google Scholar]
- 29.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;123(6):843–850. doi: 10.1182/blood-2013-09-529008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ottmann OG, Wassmann B, Pfeifer H, et al. Imatinib compared with chemotherapy as front-line treatment of elderly patients with Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ALL) Cancer. 2007;109(10):2068–2076. doi: 10.1002/cncr.22631. [DOI] [PubMed] [Google Scholar]
- 31.Chalandon Y, Thomas X, Hayette S, et al. Randomized study of reduced-intensity chemotherapy combined with imatinib in adults with Ph-positive acute lymphoblastic leukemia. Blood. 2015;125(24):3711–3719. doi: 10.1182/blood-2015-02-627935. [DOI] [PubMed] [Google Scholar]
- 32.Rousselot P, Coude MM, Gokbuget N, et al. Dasatinib and low-intensity chemotherapy in elderly patients with Philadelphia chromosome-positive ALL. Blood. 2016;128(6):774–782. doi: 10.1182/blood-2016-02-700153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Short NJ, Kantarjian HM, Ravandi F, et al. Frontline hyper-CVAD plus ponatinib for patients with Philadelphia chromosome-positive acute lymphoblastic leukemia: updated results of a Phase II study. J. Clin. Oncol. 2017;35(Suppl. 15):7013. [Google Scholar]
- 34.Jabbour E, Kantarjian H, Ravandi F, et al. Combination of hyper-CVAD with ponatinib as first-line therapy for patients with Philadelphia chromosome-positive acute lymphoblastic leukaemia: a single-centre, Phase II study. Lancet Oncol. 2015;16(15):1547–1555. doi: 10.1016/S1470-2045(15)00207-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Sasaki K, Jabbour EJ, Ravandi F, et al. Hyper-CVAD plus ponatinib versus hyper-CVAD plus dasatinib as frontline therapy for patients with Philadelphia chromosome-positive acute lymphoblastic leukemia: a propensity score analysis. Cancer. 2016;122(23):3650–3656. doi: 10.1002/cncr.30231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.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. doi: 10.1182/blood-2007-10-116582. [DOI] [PubMed] [Google Scholar]
- 37.Bassan R, Rossi G, Pogliani EM, et al. Chemotherapy-phased imatinib pulses improve long-term outcome of adult patients with Philadelphia chromosome-positive acute lymphoblastic leukemia: Northern Italy Leukemia Group protocol 09/00. J. Clin. Oncol. 2010;28(22):3644–3652. doi: 10.1200/JCO.2010.28.1287. [DOI] [PubMed] [Google Scholar]
- 38.Wetzler M, Watson D, Stock W, et al. Autologous transplantation for Philadelphia chromosome-positive acute lymphoblastic leukemia achieves outcomes similar to allogeneic transplantation: results of CALGB Study 10001 (Alliance) Haematologica. 2014;99(1):111–115. doi: 10.3324/haematol.2013.085811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Fathi AT, DeAngelo DJ, Stevenson KE, et al. Phase II study of intensified chemotherapy and allogeneic hematopoietic stem cell transplantation for older patients with acute lymphoblastic leukemia. Cancer. 2016;122(15):2379–2388. doi: 10.1002/cncr.30037. [DOI] [PubMed] [Google Scholar]
- 40.Wolach O, Stevenson KE, Wadleigh M, et al. Allogeneic transplantation is not superior to chemotherapy in most patients over 40 years of age with Philadelphia-negative acute lymphoblastic leukemia in first remission. Am. J. Hematol. 2016;91(8):793–799. doi: 10.1002/ajh.24410. [DOI] [PubMed] [Google Scholar]
- 41.Gupta V, Richards S, Rowe J. Allogeneic, but not autologous, hematopoietic cell transplantation improves survival only among younger adults with acute lymphoblastic leukemia in first remission: an individual patient data meta-analysis. Blood. 2013;121(2):339–350. doi: 10.1182/blood-2012-07-445098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Dhedin N, Huynh A, Maury S, et al. Role of allogeneic stem cell transplantation in adult patients with Ph-negative acute lymphoblastic leukemia. Blood. 2015;125(16):2486–2496. doi: 10.1182/blood-2014-09-599894. quiz 586. [DOI] [PubMed] [Google Scholar]
- 43.Ribera JM, Oriol A, Morgades M, et al. Treatment of high-risk Philadelphia chromosome-negative acute lymphoblastic leukemia in adolescents and adults according to early cytologic response and minimal residual disease after consolidation assessed by flow cytometry: final results of the PETHEMA ALL-AR-03 trial. J. Clin. Oncol. 2014;32(15):1595–1604. doi: 10.1200/JCO.2013.52.2425. [DOI] [PubMed] [Google Scholar]
- 44.Fielding AK, Richards SM, Chopra R, et al. Outcome of 609 adults after relapse of acute lymphoblastic leukemia (ALL); an MRC UKALL12/ECOG 2993 study. Blood. 2007;109(3):944–950. doi: 10.1182/blood-2006-05-018192. [DOI] [PubMed] [Google Scholar]
- 45.Bargou R, Leo E, Zugmaier G, et al. Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science. 2008;321(5891):974–977. doi: 10.1126/science.1158545. [DOI] [PubMed] [Google Scholar]
- 46.Kantarjian H, Stein A, Gokbuget N, et al. Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia. N. Engl. J. Med. 2017;376(9):836–847. doi: 10.1056/NEJMoa1609783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Martinelli G, Boissel N, Chevallier P, et al. Complete hematologic and molecular response in adult patients with relapsed/refractory philadelphia chromosome – positive B-precursor acute lymphoblastic leukemia following treatment with blinatumomab: results from a Phase II, single-arm, multicenter study. J. Clin. Oncol. 2017;35(16):1795–1802. doi: 10.1200/JCO.2016.69.3531. [DOI] [PubMed] [Google Scholar]
- 48.Kantarjian HM, Stein AS, Bargou RC, et al. Blinatumomab treatment of older adults with relapsed/refractory B-precursor acute lymphoblastic leukemia: results from 2 Phase II studies. Cancer. 2016;122(14):2178–2185. doi: 10.1002/cncr.30031. 2016. [DOI] [PubMed] [Google Scholar]
- 49.Hoelzer D, Gokbuget N. Chemoimmunotherapy in acute lymphoblastic leukemia. Blood Rev. 2012;26(1):25–32. doi: 10.1016/j.blre.2011.08.001. [DOI] [PubMed] [Google Scholar]
- 50.Kantarjian HM, DeAngelo DJ, Stelljes M, et al. Inotuzumab ozogamicin versus standard therapy for acute lymphoblastic leukemia. N. Engl. J. Med. 2016;375(8):740–753. doi: 10.1056/NEJMoa1509277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Short NJ, Kantarjian HM, O'Brien SM, et al. Updated results of a Phase I/II study of inotuzumab ozogamicin in combination with low-intensity chemotherapy (mini-hyper-CVD) as frontline therapy for older patients with acute lymphoblastic leukemia. J. Clin. Oncol. 2017;35(Suppl. 15):7014. [Google Scholar]
- 52.Jabbour E, Ravandi F, Kebriaei P, et al. Salvage chemoimmunotherapy with inotuzumab ozogamicin combined with mini-hyper-cvd for patients with relapsed or refractory Philadelphia chromosome-negative acute lymphoblastic leukemia: a Phase II clinical trial. JAMA Oncol. 2018;4(2):230–234. doi: 10.1001/jamaoncol.2017.2380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Webb MS, Harasym TO, Masin D, Bally MB, Mayer LD. Sphingomyelin-cholesterol liposomes significantly enhance the pharmacokinetic and therapeutic properties of vincristine in murine and human tumour models. Br. J. Cancer. 1995;72(4):896–904. doi: 10.1038/bjc.1995.430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Webb MS, Logan P, Kanter PM, et al. Preclinical pharmacology, toxicology and efficacy of sphingomyelin/cholesterol liposomal vincristine for therapeutic treatment of cancer. Cancer Chemother. Pharmacol. 1998;42(6):461–470. doi: 10.1007/s002800050846. [DOI] [PubMed] [Google Scholar]
- 55.O'Brien S, Schiller G, Lister J, et al. High-dose vincristine sulfate liposome injection for advanced, relapsed, and refractory adult Philadelphia chromosome-negative acute lymphoblastic leukemia. J. Clin. Oncol. 2013;31(6):676–683. doi: 10.1200/JCO.2012.46.2309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.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. doi: 10.1182/blood-2006-11-056754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.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. doi: 10.1182/blood-2011-01-329441. [DOI] [PubMed] [Google Scholar]
- 58.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. doi: 10.1038/leu.2013.312. [DOI] [PubMed] [Google Scholar]
- 59.Thomas DA, O'Brien S, Jorgensen JL, et al. Prognostic significance of CD20 expression in adults with de novo precursor B-lineage acute lymphoblastic leukemia. Blood. 2009;113(25):6330–6337. doi: 10.1182/blood-2008-04-151860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Maury S, Chevret S, Thomas X, et al. Rituximab in B-lineage adult acute lymphoblastic leukemia. N. Engl. J. Med. 2016;375(11):1044–1053. doi: 10.1056/NEJMoa1605085. [DOI] [PubMed] [Google Scholar]
- 61.Thomas DA, O'Brien S, Faderl S, et al. Chemoimmunotherapy with a modified hyper-CVAD and rituximab regimen improves outcome in de novo Philadelphia chromosome-negative precursor B-lineage acute lymphoblastic leukemia. J. Clin. Oncol. 2010;28(24):3880–3889. doi: 10.1200/JCO.2009.26.9456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Sasaki K, Kantarjian HM, Ravandi F, et al. Frontline ofatumumab in combination with hyper-CVAD for adult patients with CD-20 positive acute lymphoblastic leukemia (ALL): interim result of a Phase II clinical trial. Blood. 2016;128(22):2783. [Google Scholar]
- 63.Park JH, Sauter C, Brentjens R. Cellular therapies in acute lymphoblastic leukemia. Hematol. Oncol. Clin. North Am. 2011;25(6):1281–1301. doi: 10.1016/j.hoc.2011.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Buechner J, Grupp SA, Maude SL, et al. Global registration trial of efficacy and safety of CTL019 in pediatric and young adult patients with relapsed/refractory (R/R) acute lymphoblastic leukemia (ALL): update to the interim analysis. Clin. Lymphoma Myeloma and Leuk. 2017;17(Suppl. 2):S263–S264. [Google Scholar]
- 65.Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N. Engl. J. Med. 2018;378(5):439–448. doi: 10.1056/NEJMoa1709866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Park JH, Rivière I, Gonen M, et al. Long-term follow-up of cd19 car therapy in acute lymphoblastic leukemia. N. Engl. J. Med. 2018;378(5):449–459. doi: 10.1056/NEJMoa1709919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Roberts KG, Gu Z, Payne-Turner D, et al. High frequency and poor outcome of Philadelphia chromosome-like acute lymphoblastic leukemia in adults. J. Clin. Oncol. 2017;35(4):394–401. doi: 10.1200/JCO.2016.69.0073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Jain N, Roberts KG, Jabbour E, et al. Ph-like acute lymphoblastic leukemia: a high-risk subtype in adults. Blood. 2017;129(5):572–581. doi: 10.1182/blood-2016-07-726588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Kantarjian HM, O'Brien S, Smith TL, et al. Results of treatment with hyper-CVAD, a dose-intensive regimen, in adult acute lymphocytic leukemia. J. Clin. Oncol. 2000;18(3):547–561. doi: 10.1200/JCO.2000.18.3.547. [DOI] [PubMed] [Google Scholar]
- 70.Petersdorf SH, Kopecky KJ, Head DR, et al. Comparison of the L10M consolidation regimen to an alternative regimen including escalating methotrexate/L-asparaginase for adult acute lymphoblastic leukemia: a Southwest Oncology Group Study. Leukemia. 2001;15(2):208–216. doi: 10.1038/sj.leu.2402006. [DOI] [PubMed] [Google Scholar]
- 71.Annino L, Vegna ML, Camera A, et al. Treatment of adult acute lymphoblastic leukemia (ALL): long-term follow-up of the GIMEMA ALL 0288 randomized study. Blood. 2002;99(3):863–871. doi: 10.1182/blood.v99.3.863. [DOI] [PubMed] [Google Scholar]
- 72.Larson RA. Acute lymphoblastic leukemia: older patients and newer drugs. Hematology Am. Soc. Hematol. Educ. Program. 2005;2005:131–136. doi: 10.1182/asheducation-2005.1.131. [DOI] [PubMed] [Google Scholar]
- 73.Pullarkat V, Slovak ML, Kopecky KJ, Forman SJ, Appelbaum FR. Impact of cytogenetics on the outcome of adult acute lymphoblastic leukemia: results of Southwest Oncology Group 9400 study. Blood. 2008;111(5):2563–2572. doi: 10.1182/blood-2007-10-116186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Goekbuget N, Leguay T, Hunault M, et al. First European chemotherapy schedule for elderly patients with acute lymphoblastic leukemia: promising remission rate and feasible moderate dose intensity consolidation. Blood. 2008;112(11):304. [Google Scholar]
- 75.Ribera JM, Garcia O, Oriol A, et al. Feasibility and results of subtype-oriented protocols in older adults and fit elderly patients with acute lymphoblastic leukemia: results of three prospective parallel trials from the PETHEMA group. Leuk. Res. 2016;41:12–20. doi: 10.1016/j.leukres.2015.11.012. [DOI] [PubMed] [Google Scholar]
- 76.Delannoy A, Delabesse E, Lheritier V, et al. Imatinib and methylprednisolone alternated with chemotherapy improve the outcome of elderly patients with Philadelphia-positive acute lymphoblastic leukemia: results of the GRAALL AFR09 study. Leukemia. 2006;20(9):1526–1532. doi: 10.1038/sj.leu.2404320. [DOI] [PubMed] [Google Scholar]