Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Jan 22.
Published in final edited form as: Hematol Oncol Clin North Am. 2011 Dec;25(6):1281–1301. doi: 10.1016/j.hoc.2011.09.015

Cellular Therapies in Acute Lymphoblastic Leukemia

Jae H Park 1,*, Craig Sauter 1,*, Renier Brentjens 1,
PMCID: PMC3551618  NIHMSID: NIHMS335998  PMID: 22093587

Abstract

A majority of adult patients with acute lymphoblastic leukemia (ALL) will die of their disease. While the prognosis for pediatric patients is markedly better, in all cases, the prognosis in patients with relapsed or refractory disease is uniformly poor. Allogeneic hematopoietic stem cell transplantation (HSCT) from a related donor can offer a significant potential therapeutic benefit for some patients. Since most patients lack a suitable related donor, alternative allo-HSCT approaches, including unrelated, umbilical cord blood (UCB), and haploidentical allo-HSCT, have been developed and are increasingly being studied in the clinical setting. Reduced-intensity conditioning further extends access to allo-HSCT for older more comorbid patients. While the use of donor-derived T cell adoptive therapy has a uniformly dismal outcome in patients with relapsed ALL following HSCT, modified adoptive T cell regimens, including the infusion of enriched tumor-targeted donor T cells and genetically targeted T cells, are currently under clinical investigation with promising results. Lastly, natural killer (NK) cells genetically modified to target ALL are also being studied in clinical trials, further expanding therapeutic options for patients with refractory or relapsed ALL. What remains to be seen is whether these novel adoptive cell therapies will ultimately lead to improved clinical outcomes.

Keywords: Acute lymphoblastic leukemia, adoptive cellular therapy, hematopoietic stem cell transplants, chimeric antigen receptor

INTRODUCTION

In general, adult patients diagnosed with acute lymphoblastic leukemia (ALL) have a poor prognosis. Overall, more than 6 out of 10 adult patients diagnosed with ALL will ultimately die of their disease1. In the pediatric population, the prognosis is far more favorable with greater than 8 of 10 patients achieving a long-term survival1, 2. In most cases, up-front therapy involves long-term, toxic, and complex chemotherapy regimens. However, for both adult as well as pediatric patients, a failure to respond to up-front chemotherapy or relapse of disease after achieving a remission portends a dismal prognosis35. These findings suggest that novel approaches to adoptive cell therapies are needed to improve the outcome of patients with ALL. Recent advances in the understanding of tumor biology and immunology, combined with enhanced gene transfer technologies, have made the field of adoptive cell therapy one of great interests to investigators seeking alternative treatment approaches for this disease.

Hematopoietic Stem Cell Transplantation (HSCT)

Allogeneic HSCT (allo-HSCT) is the earliest and most studied form of adoptive cell therapy for leukemia. The original guiding principle of allo-HSCT was that it allows for higher dose chemotherapy with or without additional total body irradiation ideally resulting in consequent ablation of both tumor and normal bone marrow stem cells, the latter of which is subsequently rescued by the infusion of non-malignant HSCs from a healthy allogeneic donor. Clinical studies of allo-HSCT illustrate an additional immunologic benefit of this approach wherein donor T cells may mediate a beneficial graft versus leukemia (GvL) effect through donor T cells recognizing antigens present on residual tumor cells. This GvL effect was first described in patients with acute leukemia, including ALL6, and is best illustrated by higher relapse rates in patients who have received donor grafts from identical twin siblings and patients treated with T cell depleted grafts designed to minimize graft versus host disease (GvHD)7. Consistent with this donor T cell mediated GvL effect is the finding that patients following allo-HSCT who experience acute and/or chronic GvHD are less likely to experience disease relapse when compared to patients who experience little or no GvHD following treatment7. Unfortunately as this GvL benefit is met with the untoward consequences of GvHD and associated morbidity and mortality, the benefit of allo-HSCT remains debatable.

HLA matched donor allo-HSCT in ALL as First Remission Therapy

While a large body of clinical data utilizing myeloablative (MA) matched related donors (MRD) allo-HSCT in patients with ALL exists, there remains debate regarding the utilization of MRD allo-HSCT in the setting of adult patients with ALL as a post-remission therapy. Based on the poor overall prognosis of this disease, there is the contention that all patients with suitable MRD should undergo allo-HSCT. However, this contention should take into account a significant treatment related mortality (TRM) of 20–30% associated with allo-HSCT8, in addition to quality-of-life considerations. Moreover, patients’ age and co-morbidities must be taken into careful consideration to determine transplant-eligibility in order to reach the potential benefit of this modality in terms of overall survival (OS).

The majority of ALL patients (>80%), both adult and pediatric, will achieve disease remission (CR) after one or 2 cycles of induction chemotherapy1. Whether patients in first complete remission (CR1) benefit with MRD HSCT versus chemotherapy alone in the adult ALL setting is a critical question with conflicting answers. Adult patients with ALL have traditionally been divided into standard and high-risk groups based on several clinical and genetic criteria. High-risk patients are variably defined as those patients >35 years of age, with an elevated WBC count at diagnosis, a delayed response (>28 days) following initial induction chemotherapy, and with genetically adverse features including the presence of the Philadelphia chromosome (Ph+), t(1;19) and t(4;11). In high-risk transplant-eligible patients, MA MRD allo-HSCT is currently the consolidation treatment of choice in the setting of CR19, given several large clinical trials as well as a meta-analysis conferring benefit when compared to either chemotherapy alone or autologous HSCT1013. However, in contrast to these findings, data from the PETHEMA ALL-93 and MRC UKALL XII/ECOG E2993 trials failed to demonstrate a similar advantage for patients with high risk disease once more placing the role of MRD HSCT for high risk patients into question14, 15.

An important high-risk category of patients that deserves separate discussion is ALL harboring the Philadelphia chromosome (Ph+) for a multitude of reasons, including: 1) poor prognosis predominately secondary to relative chemo-insensitivity16 , 2) predilection for older patients which may not be able to tolerate intensive therapy and 3) opportunity for targeted tyrosine-kinase inhibitor therapy (TKI: imatinib, dasatinib and nilotinib). Prior to the incorporation of TKIs into treatment regimens, patients with Ph+ ALL fared poorly in the setting of MRD HSCT. However, one recently published report of data generated by the MRC UKALL XII/ECOG E2993 trial demonstrated a significantly improved relapse free survival in Ph+ ALL patients following matched unrelated (MUD) or MRD HSCT when compared to chemotherapy alone, data generated prior to the utilization of TKI17. While this is the largest prospective study evaluating chemotherapy versus allo-HSCT as post-remission consolidation for Ph+ ALL, there are important limitations. Given the age restriction for allo-HSCT on the study (95% of patients receiving allo-HSCT were <50 years of age), the patients that received chemotherapy alone were significantly older than those that received allo-HSCT (p=0.004), which may introduce a potentially large confounder effect as age was a significant prognostic factor in the multivariate analysis. Additionally, further analysis by intent-to-treat (ITT) revealed non-significant differences in the two groups, again speaking to the relative chemo-insensitive nature of Ph+ ALL and the intolerance of therapy of advanced-age patients with this disease phenotype. TKIs in combination with chemotherapy has become an accepted standard of care for remission induction in patients with Ph+ ALL with significant improvements in complete remission rates compared to chemotherapy only historical controls18. Importantly, the relatively low-toxicity of adjunctive TKI with combination chemotherapy appears to offer access to allo-HSCT for this high-risk and typically advanced age patient cohort. Considering ITT models, the efficacy of TKI therapy for patients with Ph+ ALL necessitates reevaluation of the role of allo-HSCT in this disease phenotype. Early results of allo-HSCT in patients with high risk Ph+ disease treated with TKI during induction and consolidation prior to transplantation suggest that the addition of imatinib prior to allo-HSCT results in favorable CR rates and further appears to offer improved disease free survival and overall survival following allo-HSCT transplantation when compared to historical controls19, 20. More recent studies, of a larger Ph+ patient cohort, have confirmed favorable outcomes of TKI in combination with chemotherapy followed by transplantation. In the MRC UKALL XII/ECOG E2993 series, patients randomized to allo-HSCT in the post-TKI era had improved 3-year overall survival (OS) compared to pre-TKI patients randomized to allo-HCT based upon same CR criteria (56% vs 40%)21. Lastly, a study from Japan demonstrated added efficacy of TKI in first CR patients prior to MA allo-HSCT compared to pre-TKI historic controls (3 year OS 65% and 44% respectively, p=0.005)22.

The more contentious debate has been the role of allo-HSCT in standard risk adult patients in CR1. The largest prospective randomized trial to attempt to answer this question was the MRC UKALL XII/ECOG E2993. In this trial, 1646 Philadelphia chromosome (Ph)-negative ALL patients underwent a standardized induction and those that achieved CR were “biologically” randomized to allo-HSCT if an MRD was identified and the patient was of appropriate MA allo-HSCT age either <50 years (ECOG) or <55 years (MRC) or chemotherapy/autologous HSCT if no MRD identified. The standard-risk MRD allo-HSCT arm demonstrated a 5-year OS of 62% which was significantly better than 52% in the no donor arm (p=0.02)14. Paradoxically, the high-risk Ph(-) patients did not derive significant advantage from allo-HSCT. In both high and standard risk groups, the relapse risk was significantly abrogated in the MRD allo-HSCT arm, lending credence to a GvL effect. Thus, this study may pose more questions than answered, such as does “intensifying” consolidation with allo-HSCT truly overcomes the poor-prognosis of traditionally-defined Ph(-) high-risk patients. Additionally, one could argue that, given the relative success of intensified pediatric-inspired chemotherapy programs23, the control chemotherapy group in this large randomized ITT study may have been suboptimally treated. Lastly, prognostic modeling has improved in the modern era. Thus, strategies to better risk-adapt patients in hope of potentiating benefit from the toxicity of allo-HSCT consolidation based upon clinical response to induction chemotherapy, i.e. time to CR and minimal residual disease following completion of induction, are being largely adapted in clinical trials in Europe 2426. In addition to identifying the appropriate standard risk patients with which to escalate therapy with allo-HSCT27, high-risk patients may be afforded the opportunity to be spared allo-HSCT if prompt minimal residual disease negative status is attained28. Thus, despite MRC/ECOG randomized data, the role of allo-HSCT in the contemporary era remains a point of discussion.

Unrelated allo-HSCT and ALL

Overall, it would appear that adult patients with ALL in CR1 benefit from allo-HSCT from an MRD, unfortunately only approximately one-third of these patients have an appropriate MRD. Therefore, a majority of patients rely on identification of either an unrelated donor (URD), an umbilical cord blood (UCB) donor, or a haploidentical donor. In all of these settings, one would expect that the risk of TRM secondary to GvHD would be increased, but with a consequently enhanced GvL effect. However, these presumptions are challenged by published data.

Historically, in the MA setting patients with matched unrelated donors (MUD) have fared poorly compared to patients transplanted from MRD secondary to increased transplant-related mortality (TRM) associated with GVHD29, 30. In the contemporary allo-HSCT era of more resolute HLA-matching criteria31, as well as improved supportive care 32, the relative differences in clinical outcome have become less appreciable between allo-HSCT from MRD and MUD3335. Several studies have addressed this question specifically in the setting of MA allo-HSCT for ALL in CR1 demonstrating similar TRM, relapse rate, and ultimately OS when comparing ALL patients who underwent either a MRD or MUD HSCT 3638. A recent study from Japan nicely demonstrated the traditionally increased risk of TRM, but concurrently reduced relapse-rate, in MUD compared to MRD illustrating the enhanced GVL as well as GVHD resulting in comparable OS39. These data support the recommendation that eligible patients in CR1, with either a MRD or MUD available, be given equal consideration for an allo-HSCT.

Alternative donor allo-HSCT

Advances in alternative donor transplantation, i.e. umbilical cord blood (UCB) and haploidentical allo-HSCT, offers a transplantation option for those patients lacking a suitably matched related or unrelated donor. With increasing numbers of public cord blood banks, UCB in adults is becoming an increasingly viable option with burgeoning data emerging in only the last 7 years 40. The early experience demonstrated lower than anticipated degrees of GvHD across greater HLA-barriers compared to traditional volunteer unrelated donor grafts4143. A significant factor in TRM is the total nucleated cell (TNC) dose infused, with patients receiving <2×107/kg TNC exhibiting a higher incidence of graft failure and greater TRM compared to those receiving grafts with >2×107/kg TNC 44, 45. To overcome this limitation, many centers have adopted double unit UCB transplants44, 46. Interestingly, studies have found that double unit recipients appear to fare better than single unit historical controls despite the vast majority of patients engrafting with only one of the 2 infused UCB units47. Theories posited of this benefit include reduced TRM related to larger cell dose and brisk myeloid engraftment46 as well as enhanced GvL and subsequent protection from progression of primary hematologic malignancy48.

Since the field of UCB transplantation is relatively new, most reports pool AML and ALL into a single category of patients with acute leukemia, making recommendations regarding this modality in the specific setting of ALL difficult. That having been said, recently reported outcomes of UCB HSCT in a large registry series compared favorably to bone marrow or peripheral blood stem cell allo-HSCT transplants in adult patients with acute leukemia49. In this series of over 1500 patients, leukemia-free survival in UCB transplant patients mismatched at 0–2 HLA loci were comparable to matched (8/8 HLA-allele matched) or mismatched (7/8 HLA-allele matched) volunteer unrelated donor transplant patients. TRM was significantly greater for UCB patients compared to 8/8 HLA-matched unrelated donors with both peripheral blood stem cell (HR 1.62, 95% CI 1.18–2.23, p=0.003) or bone marrow (1.69, 95% CI 1.19–2.39, p=0.003).

More recent published studies of UCB HSCTs have specifically focused on patients with ALL. In a large retrospective study recently published from Japan, there was no difference in TRM or leukemia-free survival between adult ALL patients that received an UCB graft mismatched at up to two loci and matched or mismatched bone marrow grafts50. Kumar et al compared outcomes of ALL patients receiving MRD HSCT, unrelated matched HSCT, mismatched HSCT, and matched or mismatched UCB transplants. The investigators surprisingly found superior 3 year OS rates in the UCB transplant group when compared to all other treatment groups, as well as improved leukemia-free survival, lower relapse rates, and lower TRM 51. The authors report a statistically significant overall survival in ALL patients treated with UCB transplant when compared to patients treated with URD HSCTs (P=0.01). However, as the authors acknowledge, interpretation of these findings should be tempered by the low numbers of patients analyzed.

A final option for allo-HSCT for patients lacking related, unrelated, or UCB HSCT options, is a haploidentical donor HSCT. In this setting virtually every patient has a suitable related donor (a parent or sibling). Not surprisingly, early studies using haploidentical HSCTs were hampered by significant incidences of GVHD and graft failure52. Over time, modifications in preparative conditioning regimens designed to optimize myeloablation and host immunosuppression, combined with enhanced techniques of T cell depletion of the graft, as well as the infusion of markedly high doses of HSCs generated from the donor through mobilization with recombinant human granulocyte colony-stimulating factor, has led to a high rate of engraftment (>95%) with minimal GvHD even in the absence of immune suppression prophylaxis5255. In a recently published report, patients with high risk acute leukemias were evaluated following haploidentical HSCT. Ciceri et al report a leukemia-free 2 year survival of high risk ALL patients treated with haploidentical HSCTs to be 13% for those undergoing transplantation in CR1, 30% for those undergoing HSCT in CR≥ 2, and 7% undergoing HSCT in non-remission56. Enhanced survival of ALL patients was recently reported in the setting of unmanipulated, non–T cell depleted haploidentical HSCTs. Huang et al report more favorable leukemia free survival in ALL patients treated with un-manipulated haploidentical grafts in ALL patients with a 3 year leukemia free survival of 60% and 25% in standard risk and high risk disease respectively57. However, not surprisingly, these improved survivals were associated with increased incidences of GvHD. Currently, much continued debate surrounds the use of haploidentical graft source as opposed to UCB and vice-versa58, 59.

Reduced-intensity conditioning (RIC) allo-HSCT for adult ALL

Given the typically higher-risk disease in a growing population of older, more infirm, patients with ALL wherein MA conditioning is prohibitively associated with exceedingly high TRM; the need for extending allo-HSCT options with RIC has never been greater. This modality sacrifices disease control with reduced intensity of conditioning to minimize TRM, thus relying more heavily on GvL. Thus, the gravity of disease control prior to allo-HSCT carries greater value to outcomes. The feasibility of this approach has been met with somewhat encouraging results given this high-risk patient population6063. In a recent large retrospective series comparing RIC and MA conditioning for Ph(-) ALL, while there was a trend toward more frequent relapse with RIC (35% vs 28% MA, p=0.08) there was no difference in OS in multivariate analysis (p=0.92)64. The European Group for Blood and Marrow Transplantation registry data demonstrated decreased non-relapse mortality with RIC compared to MA (21% vs 29%, p=0.03) with associated increased frequency of relapse (47% vs 31%, p<0.001) resulting in a trend toward improved estimated 2-year leukemia-free survival with MA compared to RIC (p=0.07) 65. These data illustrates the improved safety of RIC at the expense of diminished disease control. Application of RIC with UCB transplantation to the older ALL patient population may be feasible given data from Brunstein et al reporting on predominately double UCB transplants in older patients with hematologic malignancies. Overall this approach was well tolerated with relatively modest TRM at 3 years follow-up (26%) and promising overall and event-free survival (45% and 38% respectively)66. A more recent publication from the same institution reported the results of 22 ALL patients with 21/22 in CR1, treated with the same reduced intensity conditioning regimen followed by 4 of 22 patients receiving a MRD HSCT while the remaining 18 patients receiving UCB donor grafts. Collectively, these older (median age 49 years) high risk patients (Ph+ (n=14), and ≥ CD2 (n=10)) tolerated therapy well with a TRM of 27%, disease relapse of 36%, and an overall survival at 3 years a very promising 50%67. Again, interpretation of these studies needs to be tempered by the small number of patients reported in this study and requires further confirmation in larger prospective studies.

Novel approaches to lowering risk of GVHD: T-cell depletion (TCD)

With GVHD as the leading cause of TRM in allo-HSCT, several groups have studied T-cell depletion of a conventional donor graft as a means of lowering the frequency of this often fatal complication. The potential risks of this approach include increased risk of relapse with reduced GVL effect and impaired immune reconstitution68 leading to increased risk of infectious complications post-allo-HSCT. The group at Memorial Sloan-Kettering Cancer Center (MSKCC) has recently reported result on 35 adult patients receiving unrelated donor allo-HSCT with ex vivo TCD for hematologic malignancies, including 13 patients with ALL in remission (3 CR1, 7 CR2 and 3 CR≥3)69. Eighteen donors were HLA disparate at 1–3/10 loci. Despite a large proportion of high-risk ALL patients (CR>1), only one patient with ALL relapsed and the relapse incidence of the entire cohort was 6%. The incidence of acute grade II-IV GVHD at 9% and chronic GVHD at 29% were much lower than historical controls with a non-TCD allo-HSCT, especially considering the proportion of mismatched donors in the cohort. There were 5 fatal deaths of the 35 patients69. A British group has recently published there experience with the use of the anti-CD52 antibody alemtuzumab as an in vivo TCD allo-HSCT in 48 high-risk Ph (-) patients in CR1, with one-third of the patients receiving HLA mismatched grafts70. The incidence of acute grade II-IV GVHD and extensive chronic GVHD was 27% and 22% respectively. The OS for the entire group was 61% at 5 years. Both of these key studies demonstrate relatively favorable disease specific outcomes compared with conventional, non-TCD allo-HSCT with a decreased incidence of GVHD. These encouraging results, however, still need to be validated in a randomized prospective fashion.

Allo-HSCT for relapsed and refractory disease

Unfortunately, most patients with relapsed or refractory disease have a less than a 50% chance of responding to salvage chemotherapy71 and prognosis is extremely poor3. In relapsed patients who have not previously received an allo-HSCT, have chemosensitive disease, an appropriate HLA-matched donor, and lack prohibitive comorbidities; an allo-HSCT provides the only chance for long-term disease free survival. For a multitude of reasons, the ability to match these conditions diminishes steeply in the relapsed and refractory setting. A subset analysis from the MRC UKALL XII/ECOG E2993 study, relapsed patients receiving an allo-HSCT from a RD or an unrelated donor had improved OS at 5 years (23% and 16% respectively) versus those that did not proceed to allo-HSCT (4% OS at 5 years)3. Despite the high potential of obvious confounders in this carefully selected subset of patients, it is accepted standard to proceed to allo-HSCT in chemosensitive relapsed and refractory ALL in those who are eligible considering the incurability of chemotherapy alone.

Novel Adoptive Cellular Therapies

Due to conflicting results of these clinical trials, the role of allo-HSCT for patients with ALL remains controversial. Furthermore, novel, less tested approaches to allo-HSCT in ALL, including RIC, UCB and haploidentical HSCTs, lack sufficient numbers or prospective studies to allow for definitive recommendations. While these latter approaches as well as URD HSCT offer viable alternatives for patients requiring allo-HSCT but lacking a MRD, what is equally apparent is the fact that at this time none of these alternative options offers improved outcomes. For this reason, alternative approaches to cell therapies are required for this patient population.

Donor Lymphocyte Infusion (DLI)

DLI has been demonstrated to elicit a good response, mediated through a GvL effect, in patients with chronic myelogenous leukemia (CML) who relapse following allo-HSCT. However, it is rarely successful in relapsed ALL with a long-term disease free survival reported to range between 0% and 13%7276.

The reasons for the suboptimal response with DLI in ALL likely stem from several factors: a lack of adequate T-cell mediated GvL, the delayed effects of DLI in patients with aggressive disease, and/or a lack of costimulatory molecule on the tumor 74. Porter et al addressed the latter issue by treating 7 relapsed ALL patients post allo-HSCT with donor lymphocytes that have been activated and co-stimulated ex vivo using CD3/CD28 agonist antibodies. In this phase I trial, 4 of 7 patients achieved a CR, but 3 of the 4 patients relapsed with only one patient alive in CR at 11+ months77, showing the limitation of the conventional DLI in relapsed ALL.

A modified approach to DLI is the enrichment of donor T cells targeted to antigens over expressed on tumor cells. Wilms tumor-1 antigen (WT-1) is one such antigen which is over expressed on both acute myelogenous leukemia (AML) and ALL tumor cells 78, 79. WT-1 is immunogenic and may represent an attractive target for adoptive T cell therapy, as suggested by a recent study by Rezvani et al, who reported WT-1 specific CD8+ T cell responses following allo-HSCT in 5 of 7 patients and a subsequent molecular disease relapse (i.e. recurrent WT-1 transcript detection) associated with loss of detectable WT-1 specific CD8+ T cells80. Similarly, investigators at MSKCC as well as at the Fred Hutchinson Cancer Research Center (FHCRC) have developed means of enriching for WT-1 specific donor T cell populations through co-culture of donor T cells on antigen presenting cells pulsed with WT-1 peptides. Currently, both MSKCC (NCT00620633) and the FHCRC (NCT00052520) have open phase I clinical trials treating relapsed acute leukemias and myelodysplastic syndrome (MDS) following allo-HSCT with WT-1 specific donor T cells. The completion of these trials will provide more information on the efficacy of tumor-specific, alloreactive T cells in ALL.

Genetically Modified Tumor-Targeted T Cells

Given the limited GvL effect demonstrated with donor T cells in ALL, several investigators have studied a novel form of adoptive cellular therapy by genetically modifying autologous T cells to target specific tumor antigens. One way of genetically modifying T cells is by gene transfer of the α and β chain subunits of the T-cell receptors (TCRs) derived from T-cell clones specific to tumor antigens8183. This approach has been shown to be feasible in clinical trials of metastatic melanoma84, 85, but published data utilizing this approach in hematologic malignancies is limited86, 87. Moreover, because the TCR gene transfer approach can only recognize tumor antigens that are processed and presented by human leukocyte antigen (HLA) molecules, specificity of the TCR is restricted to specific patient HLA phenotypes and therefore lacks universal applicability. In addition, many tumor cells downregulate HLA molecules and/or have dysfunctional antigen-presenting machinery so that the targeted tumor-derived peptides are often not adequately presented on the tumor cell surface88, 89.

One way to circumvent these limitations of TCR gene transfer is the use of chimeric antigen receptors (CARs). CARs are composed of a single-chain variable-fragment (scFv) antibody specific to tumor antigen, fused to a transmembrane domain and a T-cell signaling moiety, most commonly either the CD3-ζ or Fc receptor γ cytoplasmic signaling domains90. The resulting receptor, when expressed on the surface of the T cell, mediates binding to the target tumor antigen through the scFv domain which subsequently mediates an activating signal to the T cell inducing target cell lysis.

The use of T cells engineered to express CARs has several advantages over conventional allo-HSCT. First, since this approach utilizes autologous patient derived T cells, there is no risk of GvHD. Second, tumor specific T cells may be relatively rapidly generated ex vivo in the laboratory91. Third, because CAR recognition of target tumor antigen is HLA independent, CAR-modified T cells can be applied to all HLA types and are less likely to generate resistant tumor cells through down regulation of HLA molecules. And lastly, CARs can be further modified to insert additional genes to express T-cell co-stimulatory molecules or proinflammatory cytokines to enhance anti-tumor efficacy9296.

Genetically Modified T Cells in ALL

While there is limited data regarding the use of TCR gene transfer for genetically targeting of T cells in ALL, several groups including our own have investigated the use of CARs as a means of adoptive cellular therapy for ALL.

The first requirement to redirect CAR-modified T cells toward a selected tumor cell is the identification of an appropriate target molecule which is selectively expressed on cancer cells. With regard to ALL of B-cell origin, CD19 is an ideal target for several reasons: 1) in contrast to CD20, which is the target of current antibody-based immunotherapy, CD19 is expressed on the earliest B-precursor lymphocytes; 2) CD19 expression is retained over the process of neoplastic transformation; and 3) CD19 is absent on pluripotent hematopoietic stem cells94, 97104. Furthermore, a recent report demonstrating the efficacy of bispecific single-chain antibody targeting the CD19 antigen (Blinatumomab) suggests that CD19 is an attractive target for cellular immunotherapy in ALL105.

In fact, human T cells retrovirally modified to express CD19-targeted CAR have been shown to effectively lyse CD19+ tumor cells in vitro and eradicate systemic CD19+ tumors in SCID-Beige mice97. Further studies in our laboratory have shown in vivo efficacy and persistence of these modified T cells is enhanced by co-stimulation97, 98. Since most B-cell tumors fail to express co-stimulatory ligands (CD80 and CD86) required to generate optimal activation and proliferation of T cells, we and others have further modified the CAR to include the signaling domain of the T cell co-stimulatory receptors (e.g. CD28, 4-1BB, OX40). The resulting “second-generation” CARs exhibit in vitro activation and proliferation in the absence of exogenous co-stimulatory ligands, as well as enhanced in vivo anti-tumor efficacy in immunodeficient mice bearing systemic human pre-B cell ALL tumors lacking co-stimulatory CD80 and CD86 ligands 94, 98, 102, 103, 106. More recently, several investigators have constructed and tested “third-generation” CARs containing tandem cytoplasmic signaling domains from two co-stimulatory receptors (i.e., CD28-4-1BB or CD28-OX40) demonstrating potentially enhanced T cell signaling capacity when compared to second generation CARs93, 106108, but these have yet to be studied in the clinical setting.

Clinical Trials with CD19-Targeted Modified T Cells in ALL

These promising preclinical data have led to a robust translation of CD19-targeted CAR+ T cells to the clinical setting for various B cell hematologic malignancies. Currently, there are 11 active and 3 soon-to-open phase I clinical trials targeting CD19 (Table 1). While many of these trials have recently opened, several published preliminary results of these trials suggest that adoptive cellular therapy using autologous CD19-targeted CAR+ T cells is a promising treatment approach for B cell malignancies. For example, investigators at the National Cancer Institute (NCI) reported a dramatic regression of lymphadenopathy lasting 32 weeks in a patient with advanced follicular lymphoma who was treated with a preparative chemotherapy (60mg/kg cyclophosphamide for 2 days and 25mg/m2 fludarabine for 5 days) followed by autologous CD19-targeted CAR+ T cells and Interleukin-2109. We, too, have observed a dramatic reduction of lymphadenopathy that lasted for 9 months in one patient and stable disease in two patients with chemotherapy-refractory relapsed chronic lymphocytic leukemia (CLL) who were treated with 1.5g/m2 cyclophosphamide conditioning one day prior to infusion of autologous CD19-targeted CAR+ T cells110. Investigators from the University of Pennsylvania have recently reported complete responses in 3 patients with advanced stages of indolent B cell lymphomas and CLL, all treated with autologous CD19-targeted CAR+ T cells111, 112.

Table 1.

Summary of Open and Planned Clinical Trials with CD19-Targeted Chimeric T Cell

Cell Source Patient Population Trial Design Accompanying
Lymphodepleting
Cytotoxic Therapy
Trial
Status
Trial Site
Autologous
T cells
Relapsed or refractory CLL Phase I dose-escalation trial Cy Open US,
New York
Autologous
T cells
Relapsed or refractory, or
MRD+ ALL
Phase I dose-escalation trial Cy Open US,
New York
Autologous
T cells
MRD+ or residual disease
following frontline CLL
therapy
Phase I dose escalation
trial; consolidation therapy
following upfront
chemotherapy
Cy Open US,
New York
Autologous
T cells
Relapsed or refractory low-
intermediate grade NHL &
CLL
Phase I dose escalation trial None Open US,
Texas
Autologous
T cells
Relapsed or refractory B-cell
leukemia/lymphoma (ALL,
CLL, FL, MCL, DLBCL)
Phase I dose escalation trial None Open US,
Pennsylvania
Autologous
T cells
Relapsed or refractory FL Phase I Fludarabine +
rituximab
Open US,
California
Autologous
T cells
CD19-expressing B cell
malignancy of any type
Phase I Fludarabine + Cy Open US,
NIH
Autologous
T cells
Relapsed or refractory low-
intermediate grade NHL &
CLL
Phase I; modified T cell
infusion following
autologous-HSCT
Autologous HSCT
regimen (carmustine,
etoposide, cytarabine,
melphalan, rituximab)
Open US,
Texas
Donor-derived
EBV-specific
T cells
MRD+ or relapsed ALL
following allo-HSCT
Phase I dose escalation trial Cy Open US,
New York
Donor-derived
multivirus-
specific T cells
Relapsed B-ALL following
allo-HSCT
Phase I dose-escalation trial None Open US,
Texas
Donor-derived
T cells
Relapsed B-cell
leukemia/lymphoma
following allo-HSCT
Phase I dose-escalation trial N/A Open US,
NIH
UCB-derived
T cells
Relapsed or refractory ALL,
NHL, SLL, CLL
N/A N/A Pending US,
Texas
Autologous T
cells
High-risk, intermediate-
grade NHL
Phase I/II trial; modified T
cell infusion following auto-
HSCT
N/A Pending US,
California
Donor-derived
EBV-specific
T cells
High-risk or relapsed B-ALL
after allo-HSCT
Phase I N/A Pending Europe
(France,
Germany,
Italy, UK)

CLL: chronic lymphocytic leukemia; Cy: cyclophosphamide; MRD: minimal residual disease; ALL: acute lymphocytic leukemia; NHL: non-Hodgkin’s lymphoma; FL: follicular lymphoma; MCL: mantle cell lymphoma; DLBCL: diffuse large B cell lymphoma; HSCT: hematopoietic stem cell transplant; SLL: small lymphocytic lymphoma

While the majority of the clinical experience using CD19-targeted CARs has been in the setting of indolent or chronic B-cell malignancies, 8 of the 14 clinical trials involve patients with ALL: three using autologous T cells, four using donor-derived T cells, and one using UCB-modified T cells. At our center, we are currently conducting a phase I clinical trial in ALL wherein patients with relapsed disease or with minimal residual disease following the initial induction chemotherapy will be treated with cyclophosphamide conditioning followed by autologous CD19-targeted CAR+ T cells (NCT01044069). To date, we have enrolled 3 patients to the trial, and two patients have been treated. Notably, a persistent B cell aplasia was observed in the first treated patient despite prompt recovery of other blood cell counts, which lasted for 8 weeks prior to undergoing allo-HSCT from a related sibling110. The second patient has just received the modified T cells, and the results of the peripheral blood analysis are not yet available. Similarly, investigators at the University of Pennsylvania are conducting a phase I clinical trial with autologous CD19-targeted T cells in patients with B cell malignancies including ALL (NCT01029366).

Other investigators have examined the use of a CD19-targeted CAR to modify donor-derived T cells for patients with relapsed ALL following allo-HSCT. In order to reduce the risk of GvHD associated with infusion of donor-derived T cells and to offer protection against common post-transplant virus infections, the approach using virus-specific T cells has been commonly utilized in this setting. Specifically, investigators at Baylor College of Medicine are conducting a phase I/II dose-escalation trial in ALL wherein patients, following allo-HSCT, will be infused with allogeneic multi-virus targeted T cells, specific to cytomegalovirus (CMV), Epstein-Barr virus (EBV) and adenovirus, further modified to express a CD19 targeted CAR113 (NCT00840853). MSKCC has an open phase I dose-escalation clinical trial wherein patients with relapsed or minimal residual disease-positive ALL will be treated with donor-derived EBV-specific CAR-modified T cells (NCT01430390). Finally, investigators in Europe (NCT01195480) will soon open a phase I trial to evaluate the safety of donor-derived EBV-specific CAR-modified T cells in patients with high-risk or relapsed B-cell ALL after allo-HSCT.

Similarly, UCB cells can be modified to express a CD19-targeted CAR, and preclinical data exploring the CAR-modified UCB cells have been published114, 115. Based on these preclinical data, investigators at MD Anderson Cancer Center are planning a phase I clinical trial wherein patients with B cell malignancies will be infused with CD19-targeted CAR+ T cells derived from UCB at 49 days (+/− 7 days) following UCB transplantation (NCT01362452).

Genetically Modified CD19-Targeted Natural Killer (NK) Cells

NK cells are lymphoid cells of the innate immune system which express CD56 and CD16 but fail to express a TCR (CD3). NK cells express killer immunoglobulin-like receptors (KIRs) which predominantly serve as inhibitory receptors that bind specific matched HLA class I molecules (KIR ligands) on target cells. Expression by target cells of cognate KIR ligands induces a KIR-mediated inhibitory signal to the autologous NK cell, sparing the target cell from NK cell mediated lysis. In the allogeneic setting, however, NK cells encounter target cells with mismatched KIR ligands and as a result trigger NK cell alloreactivity116.

NK cell alloreactivity does not appear to be of benefit in the setting of ALL117. However, genetic modification of NK cells may be a means of overcoming ALL tumor cell resistance to NK cell mediated lysis. To this end, several investigators have successfully expanded large numbers of NK cells through co-culture of peripheral blood mononuclear cells on an irradiated K562 leukemia cell line genetically modified to express the NK cell stimulatory 4-1BB ligand as well as membrane bound IL-15 (K562-mb15-41BBL) 118, and generated ALL targeted NK cells either by retroviral transduction119 or by electroporation120 of a CD19-targeted CAR. These CD19-targeted NK cells effectively lysed NK-cell resistant ALL tumor cells, and further modification of the CAR by incorporation of the 4-1BB receptor cytoplasmic signaling domain enhanced NK-cell killing of CD19+ ALL tumor cell lines and patient derived ALL cells119. Significantly, these investigators at St. Jude Children’s Research Hospital have successfully modified the NK cell transduction and expansion protocol to large scale cGMP conditions, and have opened a phase I dose-escalating clinical trial wherein children with refractory or relapsed ALL are treated with CD19-targeted NK cells from a haploidentical donor. To date, they have enrolled 2 patients at the lowest dose cohort (D. Campana, personal communication).

CONCLUSIONS

ALL remains a difficult disease to treat. In the adult setting, most patients will ultimately die of their disease, while in the pediatric setting, relapsed and refractory disease remains a therapeutic challenge. Cellular therapy through allo-HSCT remains an option for these patients and recent advances in alternative forms of allo-HSCT including URD transplants, UCB transplants, and haploidentical transplants have expanded the numbers of patients eligible for allo-HSCT, but to date have not improved outcomes when compared to HLA-matched related allo-HSCTs. In light of this persistent failure, several novel adoptive cellular approaches are currently being investigated to treat patients with ALL. The use of enriched WT-1 specific donor T cells to treat patients with ALL is currently under investigation in phase I trials at several centers. Treatment of ALL with genetically modified T cells targeted to the CD19 antigen through the expression of a CD19-specific CAR have entered phase I clinical trials at several centers as well. Similarly, a clinical trial treating ALL patients with genetically modified NK cells targeted to the CD19 antigen has recently opened for accrual. Collectively, these ongoing and anticipated trials provide promising role for adoptive cellular therapies in the treatment of ALL. What remains to be seen is whether this promise will translate into either improved outcomes for these patients or provide significant insights upon which to design “second generation” adoptive cell therapeutic clinical trials for ALL in the future.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

**

The authors disclose no competing financial interests.

REFERENCES

  • 1.Pui CH, Evans WE. Treatment of acute lymphoblastic leukemia. N Engl J Med. 2006;354(2):166–178. doi: 10.1056/NEJMra052603. [DOI] [PubMed] [Google Scholar]
  • 2.Pui CH, Robison LL, Look AT. Acute lymphoblastic leukaemia. Lancet. 2008;371(9617):1030–1043. doi: 10.1016/S0140-6736(08)60457-2. [DOI] [PubMed] [Google Scholar]
  • 3.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]
  • 4.Gaynon PS. Childhood acute lymphoblastic leukaemia and relapse. Br J Haematol. 2005;131(5):579–587. doi: 10.1111/j.1365-2141.2005.05773.x. [DOI] [PubMed] [Google Scholar]
  • 5.Tavernier E, Boiron JM, Huguet F, et al. Outcome of treatment after first relapse in adults with acute lymphoblastic leukemia initially treated by the LALA-94 trial. Leukemia. 2007;21(9):1907–1914. doi: 10.1038/sj.leu.2404824. [DOI] [PubMed] [Google Scholar]
  • 6.Weiden PL, Flournoy N, Thomas ED, et al. Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts. N Engl J Med. 1979;300(19):1068–1073. doi: 10.1056/NEJM197905103001902. [DOI] [PubMed] [Google Scholar]
  • 7.Porter DL, Antin JH. The graft-versus-leukemia effects of allogeneic cell therapy. Annu Rev Med. 1999;50:369–386. doi: 10.1146/annurev.med.50.1.369. [DOI] [PubMed] [Google Scholar]
  • 8.Gokbuget N, Hoelzer D. Treatment of adult acute lymphoblastic leukemia. Semin Hematol. 2009;46(1):64–75. doi: 10.1053/j.seminhematol.2008.09.003. [DOI] [PubMed] [Google Scholar]
  • 9.Larson R. Allogeneic hematopoietic cell transplantation for adults with ALL. Bone Marrow Transplant. 2008;42(Suppl 1):S18–S24. doi: 10.1038/bmt.2008.107. [DOI] [PubMed] [Google Scholar]
  • 10.Hahn T, Wall D, Camitta B, et al. The role of cytotoxic therapy with hematopoietic stem cell transplantation in the therapy of acute lymphoblastic leukemia in adults: an evidence-based review. Biol Blood Marrow Transplant. 2006;12(1):1–30. doi: 10.1016/j.bbmt.2005.10.018. [DOI] [PubMed] [Google Scholar]
  • 11.Thiebaut A, Vernant JP, Degos L, et al. Adult acute lymphocytic leukemia study testing chemotherapy and autologous and allogeneic transplantation. A follow-up report of the French protocol LALA 87. Hematol Oncol Clin North Am. 2000;14(6):1353–1366. doi: 10.1016/s0889-8588(05)70190-8. x. [DOI] [PubMed] [Google Scholar]
  • 12.Thomas X, Boiron JM, Huguet F, et al. Outcome of treatment in adults with acute lymphoblastic leukemia: analysis of the LALA-94 trial. J Clin Oncol. 2004;22(20):4075–4086. doi: 10.1200/JCO.2004.10.050. [DOI] [PubMed] [Google Scholar]
  • 13.Yanada M, Matsuo K, Suzuki T, et al. 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;106(12):2657–2663. doi: 10.1002/cncr.21932. [DOI] [PubMed] [Google Scholar]
  • 14.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]
  • 15.Ribera JM, Oriol A, Bethencourt C, et al. Comparison of intensive chemotherapy, allogeneic or autologous stem cell transplantation as post-remission treatment for adult patients with high-risk acute lymphoblastic leukemia. Results of the PETHEMA ALL-93 trial. Haematologica. 2005;90(10):1346–1356. [PubMed] [Google Scholar]
  • 16.Fielding AK, Rowe JM, Richards SM, et al. Prospective outcome data on 267 unselected adult patients with Philadelphia chromosome-positive acute lymphoblastic leukemia confirms superiority of allogeneic transplantation over chemotherapy in the pre-imatinib era: results from the International ALL Trial MRC UKALLXII/ECOG2993. Blood. 2009;113(19):4489–4496. doi: 10.1182/blood-2009-01-199380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Fielding AK, Rowe JM, Richards SM, et al. Prospective outcome data on 267 unselected adult patients with Philadelphia-chromosome positive acute lymphoblastic leukaemia confirms superiority of allogeneic transplant over chemotherapy in the pre-imatinib era: Results from the international ALL trial MRC UKALLXII/ECOG2993. Blood. 2009 doi: 10.1182/blood-2009-01-199380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lee HJ, Thompson JE, Wang ES, et al. Philadelphia chromosome-positive acute lymphoblastic leukemia: current treatment and future perspectives. Cancer. 2011;117(8):1583–1594. doi: 10.1002/cncr.25690. [DOI] [PubMed] [Google Scholar]
  • 19.de Labarthe A, Rousselot P, Huguet-Rigal F, et al. Imatinib combined with induction or consolidation chemotherapy in patients with de novo Philadelphia chromosome-positive acute lymphoblastic leukemia: results of the GRAAPH-2003 study. Blood. 2007;109(4):1408–1413. doi: 10.1182/blood-2006-03-011908. [DOI] [PubMed] [Google Scholar]
  • 20.Lee S, Kim DW, Kim YJ, et al. Minimal residual disease-based role of imatinib as a first-line interim therapy prior to allogeneic stem cell transplantation in Philadelphia chromosome-positive acute lymphoblastic leukemia. Blood. 2003;102(8):3068–3070. doi: 10.1182/blood-2003-04-1180. [DOI] [PubMed] [Google Scholar]
  • 21.Fielding AKBG, Lazarus HM, et al. Imatinib Significantly Enhances Long-Term Outcomes In Philadelphia Positive Acute Lymphoblastic Leukaemia; Final Results of the UKALL XII/ECOG 2993 Trial. Blood. 2010;16(21):77. [Google Scholar]
  • 22.Mizuta S, Matsuo K, Yagasaki F, et al. Pre-transplant imatinib-based therapy improves the outcome of allogeneic hematopoietic stem cell transplantation for BCR-ABL-positive acute lymphoblastic leukemia. Leukemia. 2011;25(1):41–47. doi: 10.1038/leu.2010.228. [DOI] [PubMed] [Google Scholar]
  • 23.Huguet F, Leguay T, Raffoux E, et al. Pediatric-inspired therapy in adults with Philadelphia chromosome-negative acute lymphoblastic leukemia: the GRAALL-2003 study. J Clin Oncol. 2009;27(6):911–918. doi: 10.1200/JCO.2008.18.6916. [DOI] [PubMed] [Google Scholar]
  • 24.Bruggemann M, Raff T, Flohr T, et al. Clinical significance of minimal residual disease quantification in adult patients with standard-risk acute lymphoblastic leukemia. Blood. 2006;107(3):1116–1123. doi: 10.1182/blood-2005-07-2708. [DOI] [PubMed] [Google Scholar]
  • 25.Bassan R, Spinelli O, Oldani E, et al. Improved risk classification for risk-specific therapy based on the molecular study of minimal residual disease (MRD) in adult acute lymphoblastic leukemia (ALL) Blood. 2009;113(18):4153–4162. doi: 10.1182/blood-2008-11-185132. [DOI] [PubMed] [Google Scholar]
  • 26.Goekbuget NBM, Arnold R, et al. New definition of treatment response in adult acute lymphoblastic leukemia (ALL): use of molecular marker s for minimal residual disease (MRD) Blood. 2009;114(22):42. [Google Scholar]
  • 27.Beldjord KLV, Boulland M-L, et al. Post-induction minimal residual disease (MRD) defines a large subset of adults with favorable Philadelphia negative acute lymphoblastic leukemia (ALL), who do not benefit from allogeneic stem cell transplantation (SCT) in first complete remission: A GRAALL study. Blood. 2009;114(22):239. [Google Scholar]
  • 28.Ribera J-MOA, Morgades M, et al. Treatment of high-risk (HR) Philadelphia chromosome-negative (Ph-) adult acute lymphoblastic leukemia (ALL) according to baseline risk factors and minimal residual disease (MRD): Results of the PETHEMA ALL-AR-03 trial including the use of propensity score (PS) method to reduce assignment bias. Blood. 2009;114(22):136. [Google Scholar]
  • 29.Marks DI, Cullis JO, Ward KN, et al. Allogeneic bone marrow transplantation for chronic myeloid leukemia using sibling and volunteer unrelated donors. A comparison of complications in the first 2 years. Annals of internal medicine. 1993;119(3):207–214. doi: 10.7326/0003-4819-119-3-199308010-00005. [DOI] [PubMed] [Google Scholar]
  • 30.Beatty PG, Anasetti C, Hansen JA, et al. Marrow transplantation from unrelated donors for treatment of hematologic malignancies: effect of mismatching for one HLA locus. Blood. 1993;81(1):249–253. [PubMed] [Google Scholar]
  • 31.Petersdorf EW, Gooley TA, Anasetti C, et al. Optimizing outcome after unrelated marrow transplantation by comprehensive matching of HLA class I and II alleles in the donor and recipient. Blood. 1998;92(10):3515–3520. [PubMed] [Google Scholar]
  • 32.Gooley TA, Chien JW, Pergam SA, et al. Reduced mortality after allogeneic hematopoietic-cell transplantation. N Engl J Med. 2010;363(22):2091–2101. doi: 10.1056/NEJMoa1004383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ottinger HD, Ferencik S, Beelen DW, et al. Hematopoietic stem cell transplantation: contrasting the outcome of transplantations from HLA-identical siblings, partially HLA-mismatched related donors, and HLA-matched unrelated donors. Blood. 2003;102(3):1131–1137. doi: 10.1182/blood-2002-09-2866. [DOI] [PubMed] [Google Scholar]
  • 34.Walter RB, Pagel JM, Gooley TA, et al. Comparison of matched unrelated and matched related donor myeloablative hematopoietic cell transplantation for adults with acute myeloid leukemia in first remission. Leukemia. 2010;24(7):1276–1282. doi: 10.1038/leu.2010.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Yakoub-Agha I, Mesnil F, Kuentz M, et al. Allogeneic marrow stem-cell transplantation from human leukocyte antigen-identical siblings versus human leukocyte antigen-allelic-matched unrelated donors (10/10) in patients with standard-risk hematologic malignancy: a prospective study from the French Society of Bone Marrow Transplantation and Cell Therapy. J Clin Oncol. 2006;24(36):5695–5702. doi: 10.1200/JCO.2006.08.0952. [DOI] [PubMed] [Google Scholar]
  • 36.Chim CS, Lie AK, Liang R, et al. Long-term results of allogeneic bone marrow transplantation for 108 adult patients with acute lymphoblastic leukemia: favorable outcome with BMT at first remission and HLA-matched unrelated donor. Bone Marrow Transplant. 2007;40(4):339–347. doi: 10.1038/sj.bmt.1705734. [DOI] [PubMed] [Google Scholar]
  • 37.Dahlke J, Kroger N, Zabelina T, et al. Comparable results in patients with acute lymphoblastic leukemia after related and unrelated stem cell transplantation. Bone Marrow Transplant. 2006;37(2):155–163. doi: 10.1038/sj.bmt.1705221. [DOI] [PubMed] [Google Scholar]
  • 38.Kiehl MG, Kraut L, Schwerdtfeger R, et al. Outcome of allogeneic hematopoietic stem-cell transplantation in adult patients with acute lymphoblastic leukemia: no difference in related compared with unrelated transplant in first complete remission. J Clin Oncol. 2004;22(14):2816–2825. doi: 10.1200/JCO.2004.07.130. [DOI] [PubMed] [Google Scholar]
  • 39.Nishiwaki S, Inamoto Y, Sakamaki H, et al. Allogeneic stem cell transplantation for adult Philadelphia chromosome-negative acute lymphocytic leukemia: comparable survival rates but different risk factors between related and unrelated transplantation in first complete remission. Blood. 2010;116(20):4368–4375. doi: 10.1182/blood-2010-02-269571. [DOI] [PubMed] [Google Scholar]
  • 40.Sauter C, Barker JN. Unrelated donor umbilical cord blood transplantation for the treatment of hematologic malignancies. Current opinion in hematology. 2008;15(6):568–575. doi: 10.1097/MOH.0b013e3283136718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Laughlin MJ, Eapen M, Rubinstein P, et al. Outcomes after transplantation of cord blood or bone marrow from unrelated donors in adults with leukemia. N Engl J Med. 2004;351(22):2265–2275. doi: 10.1056/NEJMoa041276. [DOI] [PubMed] [Google Scholar]
  • 42.Rocha V, Labopin M, Sanz G, et al. Transplants of umbilical-cord blood or bone marrow from unrelated donors in adults with acute leukemia. N Engl J Med. 2004;351(22):2276–2285. doi: 10.1056/NEJMoa041469. [DOI] [PubMed] [Google Scholar]
  • 43.Takahashi S, Iseki T, Ooi J, et al. Single-institute comparative analysis of unrelated bone marrow transplantation and cord blood transplantation for adult patients with hematologic malignancies. Blood. 2004;104(12):3813–3820. doi: 10.1182/blood-2004-03-1001. [DOI] [PubMed] [Google Scholar]
  • 44.Barker JN, Rocha V, Scaradavou A. Optimizing unrelated donor cord blood transplantation. Biol Blood Marrow Transplant. 2008;15(1 Suppl):154–161. doi: 10.1016/j.bbmt.2008.10.020. [DOI] [PubMed] [Google Scholar]
  • 45.Barker JN, Byam C, Scaradavou A. How I treat: the selection and acquisition of unrelated cord blood grafts. Blood. 2011;117(8):2332–2339. doi: 10.1182/blood-2010-04-280966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Barker JN, Weisdorf DJ, DeFor TE, et al. Transplantation of 2 partially HLA-matched umbilical cord blood units to enhance engraftment in adults with hematologic malignancy. Blood. 2005;105(3):1343–1347. doi: 10.1182/blood-2004-07-2717. [DOI] [PubMed] [Google Scholar]
  • 47.Haspel RL, Ballen KK. Double cord blood transplants: filling a niche? Stem Cell Rev. 2006;2(2):81–86. doi: 10.1007/s12015-006-0013-z. [DOI] [PubMed] [Google Scholar]
  • 48.Verneris MR, Brunstein CG, Barker J, et al. Relapse risk after umbilical cord blood transplantation: enhanced graft-versus-leukemia effect in recipients of 2 units. Blood. 2009;114(19):4293–4299. doi: 10.1182/blood-2009-05-220525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Eapen M, Rocha V, Sanz G, et al. Effect of graft source on unrelated donor haemopoietic stem-cell transplantation in adults with acute leukaemia: a retrospective analysis. The lancet oncology. 2010;11(7):653–660. doi: 10.1016/S1470-2045(10)70127-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Atsuta Y, Suzuki R, Nagamura-Inoue T, et al. Disease-specific analyses of unrelated cord blood transplantation compared with unrelated bone marrow transplantation in adult patients with acute leukemia. Blood. 2009;113(8):1631–1638. doi: 10.1182/blood-2008-03-147041. [DOI] [PubMed] [Google Scholar]
  • 51.Kumar P, Defor TE, Brunstein C, et al. Allogeneic hematopoietic stem cell transplantation in adult acute lymphocytic leukemia: impact of donor source on survival. Biol Blood Marrow Transplant. 2008;14(12):1394–1400. doi: 10.1016/j.bbmt.2008.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Aversa F. Haploidentical haematopoietic stem cell transplantation for acute leukaemia in adults: experience in Europe and the United States. Bone Marrow Transplant. 2008;41(5):473–481. doi: 10.1038/sj.bmt.1705966. [DOI] [PubMed] [Google Scholar]
  • 53.Aversa F, Tabilio A, Terenzi A, et al. Successful engraftment of T-cell-depleted haploidentical "three-loci" incompatible transplants in leukemia patients by addition of recombinant human granulocyte colony-stimulating factor-mobilized peripheral blood progenitor cells to bone marrow inoculum. Blood. 1994;84(11):3948–3955. [PubMed] [Google Scholar]
  • 54.Aversa F, Tabilio A, Velardi A, et al. Treatment of high-risk acute leukemia with T-cell-depleted stem cells from related donors with one fully mismatched HLA haplotype. N Engl J Med. 1998;339(17):1186–1193. doi: 10.1056/NEJM199810223391702. [DOI] [PubMed] [Google Scholar]
  • 55.Aversa F, Terenzi A, Tabilio A, et al. Full haplotype-mismatched hematopoietic stem-cell transplantation: a phase II study in patients with acute leukemia at high risk of relapse. J Clin Oncol. 2005;23(15):3447–3454. doi: 10.1200/JCO.2005.09.117. [DOI] [PubMed] [Google Scholar]
  • 56.Ciceri F, Labopin M, Aversa F, et al. A survey of fully haploidentical hematopoietic stem cell transplantation in adults with high-risk acute leukemia: a risk factor analysis of outcomes for patients in remission at transplantation. Blood. 2008;112(9):3574–3581. doi: 10.1182/blood-2008-02-140095. [DOI] [PubMed] [Google Scholar]
  • 57.Huang XJ, Liu DH, Liu KY, et al. Treatment of acute leukemia with unmanipulated HLA-mismatched/haploidentical blood and bone marrow transplantation. Biol Blood Marrow Transplant. 2009;15(2):257–265. doi: 10.1016/j.bbmt.2008.11.025. [DOI] [PubMed] [Google Scholar]
  • 58.Barrett J, Gluckman E, Handgretinger R, et al. Point-counterpoint: haploidentical family donors versus cord blood transplantation. Biol Blood Marrow Transplant. 2011;17(1 Suppl):S89–S93. doi: 10.1016/j.bbmt.2010.10.024. [DOI] [PubMed] [Google Scholar]
  • 59.Ballen KK, Spitzer TR. The great debate: haploidentical or cord blood transplant. Bone Marrow Transplant. 2011;46(3):323–329. doi: 10.1038/bmt.2010.260. [DOI] [PubMed] [Google Scholar]
  • 60.Arnold R, Massenkeil G, Bornhauser M, et al. Nonmyeloablative stem cell transplantation in adults with high-risk ALL may be effective in early but not in advanced disease. Leukemia. 2002;16(12):2423–2428. doi: 10.1038/sj.leu.2402712. [DOI] [PubMed] [Google Scholar]
  • 61.Martino R, Giralt S, Caballero MD, et al. Allogeneic hematopoietic stem cell transplantation with reduced-intensity conditioning in acute lymphoblastic leukemia: a feasibility study. Haematologica. 2003;88(5):555–560. [PubMed] [Google Scholar]
  • 62.Ram R, Storb R, Sandmaier BM, et al. Non-myeloablative conditioning with allogeneic hematopoietic cell transplantation for the treatment of high-risk acute lymphoblastic leukemia. Haematologica. 2011;96(8):1113–1120. doi: 10.3324/haematol.2011.040261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Stein AS, Palmer JM, O'Donnell MR, et al. Reduced-intensity conditioning followed by peripheral blood stem cell transplantation for adult patients with high-risk acute lymphoblastic leukemia. Biol Blood Marrow Transplant. 2009;15(11):1407–1414. doi: 10.1016/j.bbmt.2009.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Marks DI, Wang T, Perez WS, et al. The outcome of full-intensity and reduced-intensity conditioning matched sibling or unrelated donor transplantation in adults with Philadelphia chromosome-negative acute lymphoblastic leukemia in first and second complete remission. Blood. 2010;116(3):366–374. doi: 10.1182/blood-2010-01-264077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Mohty M, Labopin M, Volin L, et al. Reduced-intensity versus conventional myeloablative conditioning allogeneic stem cell transplantation for patients with acute lymphoblastic leukemia: a retrospective study from the European Group for Blood and Marrow Transplantation. Blood. 2010;116(22):4439–4443. doi: 10.1182/blood-2010-02-266551. [DOI] [PubMed] [Google Scholar]
  • 66.Brunstein CG, Barker JN, Weisdorf DJ, et al. Umbilical cord blood transplantation after nonmyeloablative conditioning: impact on transplantation outcomes in 110 adults with hematologic disease. Blood. 2007;110(8):3064–3070. doi: 10.1182/blood-2007-04-067215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Bachanova V, Verneris MR, DeFor T, et al. Prolonged survival in adults with acute lymphoblastic leukemia after reduced-intensity conditioning with cord blood or sibling donor transplantation. Blood. 2009;113(13):2902–2905. doi: 10.1182/blood-2008-10-184093. [DOI] [PubMed] [Google Scholar]
  • 68.Small TN, Papadopoulos EB, Boulad F, et al. Comparison of immune reconstitution after unrelated and related T-cell-depleted bone marrow transplantation: effect of patient age and donor leukocyte infusions. Blood. 1999;93(2):467–480. [PubMed] [Google Scholar]
  • 69.Jakubowski AA, Small TN, Kernan NA, et al. T cell-depleted unrelated donor stem cell transplantation provides favorable disease-free survival for adults with hematologic malignancies. Biol Blood Marrow Transplant. 2011;17(9):1335–1342. doi: 10.1016/j.bbmt.2011.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Patel B, Kirkland KE, Szydlo R, et al. Favorable outcomes with alemtuzumab-conditioned unrelated donor stem cell transplantation in adults with high-risk Philadelphia chromosome-negative acute lymphoblastic leukemia in first complete remission. Haematologica. 2009;94(10):1399–1406. doi: 10.3324/haematol.2009.008649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.O'Brien S, Thomas D, Ravandi F, et al. Outcome of adults with acute lymphocytic leukemia after second salvage therapy. Cancer. 2008;113(11):3186–3191. doi: 10.1002/cncr.23919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Kolb HJ, Schattenberg A, Goldman JM, et al. Graft-versus-leukemia effect of donor lymphocyte transfusions in marrow grafted patients. European Group for Blood and Marrow Transplantation Working Party Chronic Leukemia. Blood. 1995;86(5):2041–2050. [PubMed] [Google Scholar]
  • 73.Kolb HJ, Schmid C, Barrett AJ, et al. Graft-versus-leukemia reactions in allogeneic chimeras. Blood. 2004;103(3):767–776. doi: 10.1182/blood-2003-02-0342. [DOI] [PubMed] [Google Scholar]
  • 74.Loren AW, Porter DL. Donor leukocyte infusions for the treatment of relapsed acute leukemia after allogeneic stem cell transplantation. Bone Marrow Transplant. 2008;41(5):483–493. doi: 10.1038/sj.bmt.1705898. [DOI] [PubMed] [Google Scholar]
  • 75.Choi SJ, Lee JH, Lee JH, et al. Treatment of relapsed acute lymphoblastic leukemia after allogeneic bone marrow transplantation with chemotherapy followed by G-CSF-primed donor leukocyte infusion: a prospective study. Bone Marrow Transplant. 2005;36(2):163–169. doi: 10.1038/sj.bmt.1705024. [DOI] [PubMed] [Google Scholar]
  • 76.Collins RH, Jr, Goldstein S, Giralt S, et al. Donor leukocyte infusions in acute lymphocytic leukemia. Bone Marrow Transplant. 2000;26(5):511–516. doi: 10.1038/sj.bmt.1702555. [DOI] [PubMed] [Google Scholar]
  • 77.Porter DL, Levine BL, Bunin N, et al. A phase 1 trial of donor lymphocyte infusions expanded and activated ex vivo via CD3/CD28 costimulation. Blood. 2006;107(4):1325–1331. doi: 10.1182/blood-2005-08-3373. [DOI] [PubMed] [Google Scholar]
  • 78.Oka Y, Elisseeva OA, Tsuboi A, et al. Human cytotoxic T-lymphocyte responses specific for peptides of the wild-type Wilms' tumor gene (WT1 ) product. Immunogenetics. 2000;51(2):99–107. doi: 10.1007/s002510050018. [DOI] [PubMed] [Google Scholar]
  • 79.Oka Y, Udaka K, Tsuboi A, et al. Cancer immunotherapy targeting Wilms' tumor gene WT1 product. J Immunol. 2000;164(4):1873–1880. doi: 10.4049/jimmunol.164.4.1873. [DOI] [PubMed] [Google Scholar]
  • 80.Rezvani K, Yong AS, Savani BN, et al. Graft-versus-leukemia effects associated with detectable Wilms tumor-1 specific T lymphocytes after allogeneic stem-cell transplantation for acute lymphoblastic leukemia. Blood. 2007;110(6):1924–1932. doi: 10.1182/blood-2007-03-076844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Brentjens RJ. Novel Approaches to Immunotherapy for B-cell Malignancies. Curr Oncol Rep. 2004;6(5):339–347. doi: 10.1007/s11912-004-0059-5. [DOI] [PubMed] [Google Scholar]
  • 82.Sadelain M, Brentjens R, Riviere I. The promise and potential pitfalls of chimeric antigen receptors. Curr Opin Immunol. 2009 doi: 10.1016/j.coi.2009.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Sadelain M, Riviere I, Brentjens R. Targeting tumours with genetically enhanced T lymphocytes. Nat Rev Cancer. 2003;3(1):35–45. doi: 10.1038/nrc971. [DOI] [PubMed] [Google Scholar]
  • 84.Johnson LA, Morgan RA, Dudley ME, et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood. 2009;114(3):535–546. doi: 10.1182/blood-2009-03-211714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Morgan RA, Dudley ME, Wunderlich JR, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science. 2006;314(5796):126–129. doi: 10.1126/science.1129003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Dossett ML, Teague RM, Schmitt TM, et al. Adoptive immunotherapy of disseminated leukemia with TCR-transduced, CD8+ T cells expressing a known endogenous TCR. Mol Ther. 2009;17(4):742–749. doi: 10.1038/mt.2008.300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Xue SA, Gao L, Hart D, et al. Elimination of human leukemia cells in NOD/SCID mice by WT1-TCR gene-transduced human T cells. Blood. 2005;106(9):3062–3067. doi: 10.1182/blood-2005-01-0146. [DOI] [PubMed] [Google Scholar]
  • 88.Khong HT, Restifo NP. Natural selection of tumor variants in the generation of "tumor escape" phenotypes. Nat Immunol. 2002;3(11):999–1005. doi: 10.1038/ni1102-999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Gottschalk S, Ng CY, Perez M, et al. An Epstein-Barr virus deletion mutant associated with fatal lymphoproliferative disease unresponsive to therapy with virus-specific CTLs. Blood. 2001;97(4):835–843. doi: 10.1182/blood.v97.4.835. [DOI] [PubMed] [Google Scholar]
  • 90.Park JH, Brentjens RJ. Adoptive immunotherapy for B-cell malignancies with autologous chimeric antigen receptor modified tumor targeted T cells. Discovery medicine. 2010;9(47):277–288. [PMC free article] [PubMed] [Google Scholar]
  • 91.Hollyman D, Stefanski J, Przybylowski M, et al. Manufacturing validation of biologically functional T cells targeted to CD19 antigen for autologous adoptive cell therapy. J Immunother. 2009;32(2):169–180. doi: 10.1097/CJI.0b013e318194a6e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Singh H, Figliola MJ, Dawson MJ, et al. Reprogramming CD19-specific T cells with IL-21 signaling can improve adoptive immunotherapy of B-lineage malignancies. Cancer Res. 2011;71(10):3516–3527. doi: 10.1158/0008-5472.CAN-10-3843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Tammana S, Huang X, Wong M, et al. 4-1BB and CD28 signaling plays a synergistic role in redirecting umbilical cord blood T cells against B-cell malignancies. Hum Gene Ther. 2010;21(1):75–86. doi: 10.1089/hum.2009.122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Kowolik CM, Topp MS, Gonzalez S, et al. CD28 costimulation provided through a CD19-specific chimeric antigen receptor enhances in vivo persistence and antitumor efficacy of adoptively transferred T cells. Cancer Res. 2006;66(22):10995–11004. doi: 10.1158/0008-5472.CAN-06-0160. [DOI] [PubMed] [Google Scholar]
  • 95.Hoyos V, Savoldo B, Quintarelli C, et al. Engineering CD19-specific T lymphocytes with interleukin-15 and a suicide gene to enhance their anti-lymphoma/leukemia effects and safety. Leukemia. 2010;24(6):1160–1170. doi: 10.1038/leu.2010.75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Chmielewski M, Kopecky C, Hombach AA, et al. IL-12 Release by Engineered T Cells Expressing Chimeric Antigen Receptors Can Effectively Muster an Antigen-Independent Macrophage Response on Tumor Cells That Have Shut Down Tumor Antigen Expression. Cancer Res. 2011;71(17):5697–5706. doi: 10.1158/0008-5472.CAN-11-0103. [DOI] [PubMed] [Google Scholar]
  • 97.Brentjens RJ, Latouche JB, Santos E, et al. Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat Med. 2003;9(3):279–286. doi: 10.1038/nm827. [DOI] [PubMed] [Google Scholar]
  • 98.Brentjens RJ, Santos E, Nikhamin Y, et al. Genetically targeted T cells eradicate systemic acute lymphoblastic leukemia xenografts. Clin Cancer Res. 2007;13(18 Pt 1):5426–5435. doi: 10.1158/1078-0432.CCR-07-0674. [DOI] [PubMed] [Google Scholar]
  • 99.Cooper LJ, Al-Kadhimi Z, DiGiusto D, et al. Development and application of CD19-specific T cells for adoptive immunotherapy of B cell malignancies. Blood Cells Mol Dis. 2004;33(1):83–89. doi: 10.1016/j.bcmd.2004.03.003. [DOI] [PubMed] [Google Scholar]
  • 100.Cooper LJ, Al-Kadhimi Z, Serrano LM, et al. Enhanced antilymphoma efficacy of CD19-redirected influenza MP1-specific CTLs by cotransfer of T cells modified to present influenza MP1. Blood. 2005;105(4):1622–1631. doi: 10.1182/blood-2004-03-1208. Epub 2004 Oct 1626. [DOI] [PubMed] [Google Scholar]
  • 101.Cooper LJ, Topp MS, Serrano LM, et al. T-cell clones can be rendered specific for CD19: toward the selective augmentation of the graft-versus-B-lineage leukemia effect. Blood. 2003;101(4):1637–1644. doi: 10.1182/blood-2002-07-1989. [DOI] [PubMed] [Google Scholar]
  • 102.Imai C, Mihara K, Andreansky M, et al. Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia. 2004;18(4):676–684. doi: 10.1038/sj.leu.2403302. [DOI] [PubMed] [Google Scholar]
  • 103.Loskog A, Giandomenico V, Rossig C, et al. Addition of the CD28 signaling domain to chimeric T-cell receptors enhances chimeric T-cell resistance to T regulatory cells. Leukemia. 2006;20(10):1819–1828. doi: 10.1038/sj.leu.2404366. [DOI] [PubMed] [Google Scholar]
  • 104.Rossig C, Bollard CM, Nuchtern JG, et al. Epstein-Barr virus-specific human T lymphocytes expressing antitumor chimeric T-cell receptors: potential for improved immunotherapy. Blood. 2002;99(6):2009–2016. doi: 10.1182/blood.v99.6.2009. [DOI] [PubMed] [Google Scholar]
  • 105.Topp MS, Kufer P, Gokbuget N, et al. Targeted therapy with the T-cell-engaging antibody blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival. J Clin Oncol. 2011;29(18):2493–2498. doi: 10.1200/JCO.2010.32.7270. [DOI] [PubMed] [Google Scholar]
  • 106.Milone MC, Fish JD, Carpenito C, et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol Ther. 2009;17(8):1453–1464. doi: 10.1038/mt.2009.83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Kochenderfer JN, Feldman SA, Zhao Y, et al. Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptor. J Immunother. 2009;32(7):689–702. doi: 10.1097/CJI.0b013e3181ac6138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Wang J, Jensen M, Lin Y, et al. Optimizing adoptive polyclonal T cell immunotherapy of lymphomas, using a chimeric T cell receptor possessing CD28 and CD137 costimulatory domains. Hum Gene Ther. 2007;18(8):712–725. doi: 10.1089/hum.2007.028. [DOI] [PubMed] [Google Scholar]
  • 109.Kochenderfer JN, Wilson WH, Janik JE, et al. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood. 2010;116(20):4099–4102. doi: 10.1182/blood-2010-04-281931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Brentjens RJ, Riviere I, Park JH, et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood. 2011 doi: 10.1182/blood-2011-04-348540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Porter DL, Levine BL, Kalos M, et al. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365(8):725–733. doi: 10.1056/NEJMoa1103849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Kalos M, Levine BL, Porter DL, et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Science translational medicine. 2011;3(95):95ra73. doi: 10.1126/scitranslmed.3002842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Savoldo B, Mickllethwaite K, Cooper L, et al. Monoculture-derived T lymphocytes providing multiple virus specificity and anti-leukemia activity for recipients of hematopoietic stem cells or umbilical cord blood transplants. Blood. 2008;112(11) Abstract 3909. [Google Scholar]
  • 114.Serrano LM, Pfeiffer T, Olivares S, et al. Differentiation of naive cord-blood T cells into CD19-specific cytolytic effectors for posttransplantation adoptive immunotherapy. Blood. 2006;107(7):2643–2652. doi: 10.1182/blood-2005-09-3904. Epub 2005 Dec 2613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Micklethwaite KP, Savoldo B, Hanley PJ, et al. Derivation of human T lymphocytes from cord blood and peripheral blood with antiviral and antileukemic specificity from a single culture as protection against infection and relapse after stem cell transplantation. Blood. 2010;115(13):2695–2703. doi: 10.1182/blood-2009-09-242263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Grzywacz B, Miller JS, Verneris MR. Use of natural killer cells as immunotherapy for leukaemia. Best Pract Res Clin Haematol. 2008;21(3):467–483. doi: 10.1016/j.beha.2008.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Ruggeri L, Capanni M, Urbani E, et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science. 2002;295(5562):2097–2100. doi: 10.1126/science.1068440. [DOI] [PubMed] [Google Scholar]
  • 118.Fujisaki H, Kakuda H, Shimasaki N, et al. Expansion of Highly Cytotoxic Human Natural Killer Cells for Cancer Cell Therapy. Cancer Res. 2009 doi: 10.1158/0008-5472.CAN-08-3712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Imai C, Iwamoto S, Campana D. Genetic modification of primary natural killer cells overcomes inhibitory signals and induces specific killing of leukemic cells. Blood. 2005;106(1):376–383. doi: 10.1182/blood-2004-12-4797. Epub 2005 Mar 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Li L, Liu LN, Feller S, et al. Expression of chimeric antigen receptors in natural killer cells with a regulatory-compliant non-viral method. Cancer Gene Ther. 2010;17(3):147–154. doi: 10.1038/cgt.2009.61. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES