Antigen-specific immunotherapies have emerged as important components of curative treatment algorithms for many cancers. In acute myeloid leukemia (AML), success has been less obvious. Nonetheless, among the few drugs shown to improve survival in recent randomized trials is the CD33 antibody–drug conjugate gemtuzumab ozogamicin. Significant antileukemic activity is also well documented for radioimmunoconjugates targeting CD33, CD45, or CD66. These therapeutics can intensify conditioning before hematopoietic cell transplantation, but their effect on patient outcomes needs clarification. Emerging data now suggest clinical antileukemic activity of several novel antibodies and perhaps some adoptive T-cell immunotherapies and vaccines. In parallel, numerous other agents targeting a wider variety of antigens are currently being explored. However, the antigenic heterogeneity characteristic of AML is a considerable limitation for all these therapeutics, and many important questions related to the ideal target antigen(s), disease situation in which to use these therapies, most suitable patient populations, exact treatment modalities, and details of supportive care needs remain open. Addressing such questions in upcoming studies will be required to ensure that antigen-directed therapies become an effective tool in AML, a disease for which outcomes with standard “3 + 7”-based chemotherapy have remained unsatisfactory in many patients.

Learning Objectives
  • To gain an overview of current clinical results with antigen-specific immunotherapies for acute myeloid leukemia

  • To become familiar with recent advances and evolving treatment concepts in the field of antigen-specific immunotherapy for acute myeloid leukemia and to appreciate the challenges in the clinical translation of these therapeutic strategies

Conceptually, antigen-specific immunotherapies offer a versatile means of eliminating tumor cells with limited nonspecific toxicities. For many human cancers, including some hematological malignancies, such therapies have emerged as important components of curative treatment algorithms.1-3  Success has been more modest for acute myeloid leukemia (AML). Nonetheless, among the few drugs that have shown to improve survival in recent randomized trials is the CD33 antibody–drug conjugate gemtuzumab ozogamicin (GO). The clinical experience with GO provides the strongest glimpse thus far of the potential value of antigen-targeted therapies for AML. Here, we review the results of efforts with antibodies and other antigen-directed passive and active immunotherapies in AML and summarize evolving treatment strategies for this disease.

The choice of target antigen is of critical importance for the success of any AML immunotherapeutic.4  Ideally, it is displayed homogeneously at high levels on all AML cells including leukemia stem cells (LSCs), has minimal to no expression in normal tissues, plays an important role in AML pathogenesis, is not shed into the circulation, and is sufficiently immunogenic in the case of active immunization strategies. There are many well-characterized antigens in AML, but finding the perfect target(s) is an unresolved challenge. A main obstacle is antigenic heterogeneity across individual leukemia cells within a patient and between patients because of the clonal and genetic diversity of the disease. As a consequence of the latter, AML-specific antigens (eg, those resulting from chromosomal translocations or gene mutations) are confined to particular patient subsets, limiting their general usefulness and necessitating an individualized treatment strategy. Few antigens on AML cells are restricted to normal embryonic and fetal development (“oncofetal antigens”) or immune-privileged tissues (“cancer–testis antigens”). The vast majority of AML cell antigens described to date are “leukemia associated” and thus also found on nonleukemic cells, especially those of hematopoietic origin, including progenitor and stem cell populations. Toxicity attributable to normal counterpart cell targeting, particularly prolonged cytopenias, is a major concern with all AML-associated antigens and an important factor in the consideration of what constitutes a clinically targetable antigen.

Initially, research on antibody-based therapy in AML has primarily focused on the myeloid differentiation antigen CD33 (SIGLEC-3). CD33 is attractive because it is displayed on at least a subset of leukemic blasts in almost all AML patients,5,6  is possibly present on LSCs in some patients,7  and has endocytic properties that enable its use for targeted delivery of cellular toxins. However, CD33 expression is relatively low, with an average of ∼104 molecules per AML blast and varies over 2-log–fold among patients; thus, some leukemia cells express very few CD33 molecules. For the delivery of radionuclides, CD45 and CD66 have served as alternatives to CD33.8  The number of antigens exploited for immunotherapeutic purposes has recently expanded dramatically. Some are chosen primarily because of their overexpression on AML cells relative to normal cells, including Wilms tumor 1 (WT1) and proteinase 3 (PRTN3). Increasingly, targets are selected because they are important for proliferation, survival and/or drug resistance, or other AML cell functions [eg, CXC chemokine receptor 4 (CXCR4), vascular endothelial growth factor (VEGF), or fms-like tyrosine kinase-3 (FLT3; CD135)] or because they are expressed on putative LSCs [eg, CD44, CD47, CD96, CD123, C-type lectin-like molecule-1 (CLL-1), or interleukin-1 (IL-1) receptor accessory protein (IL1RAP)] or on surrounding cells that mediate host immune responses against AML cells [eg, killer inhibitory receptors (KIRs) on natural killer (NK) cells]. Broadly speaking, the goals pursued with any immunotherapeutic are direct targeting of AML cell antigens, boosting the number and activity of immune effector cells, activating tumor antigen-specific immunity, and/or overcoming inhibitory immune suppression.3 

Although many unconjugated antibodies against a variety of antigens have shown efficacy in preclinical AML models, they typically were unsuccessful when investigated in the clinic (Table 1). Among these were antibodies targeting CD15 or CD52 (alemtuzumab), which were given to only a handful of patients.9,10  Most widely tested was the CD33 antibody lintuzumab (HuM195, SGN-CD33), which produced infrequent complete remissions (CRs) or partial remissions (PRs) confined to patients with low tumor burden even at suprasaturating doses that fully blocked CD33 binding sites for several weeks.11,12  Limited data suggested better efficacy in acute promyelocytic leukemia (APL) in patients with minimal residual disease (MRD) or when given at exceedingly high doses in non-APL AML.13,14  However, the development of lintuzumab was terminated after randomized trials showed no survival improvement when it was added to mitoxantrone, etoposide, and cytarabine (MEC) in patients with relapsed/refractory AML or to low-dose cytarabine (LDAC) in older adults with untreated AML.15,16  Similarly, the VEGF-A antibody bevacizumab did not improve response rates or survival in a randomized trial when added to standard induction chemotherapy in older adults with AML.17  Also unsuccessful were antibodies directed at CD123 (CSL360),18  FLT3 [IMC-EB10 (LY3012218)],19,20  or the oncofetal antigen EphA3 (KB004),21  which lacked antileukemic effects in most patients in phase 1 studies. Conversely, the CXCR4 antibody ulocuplumab combined with MEC yielded responses that compared favorably with historic controls treated with MEC alone among 43 patients with relapsed/refractory AML. Of note, 4 patients achieved a CR or CR with incomplete blood count recovery (CRi) after a single dose of ulocuplumab alone, demonstrating direct cytotoxic activity in a subset of patients.22  Some evidence of clinical antileukemia efficacy was likewise obtained with the KIR antibody IPH2101: at higher doses, this agent was associated with improved survival among 23 older adults with AML in first CR.23  A recombinant form of the same KIR antibody (IPH2102/BMS-986015, lirilumab) is being tested in ongoing clinical trials.

Table 1.

Selected studies with unconjugated antibodies in AML

Selected studies with unconjugated antibodies in AML
Selected studies with unconjugated antibodies in AML

EFS indicates event-free survival; HiDAC, high-dose cytarabine; IPSS, International Prognostic Scoring System; MLFS, morphologic leukemia-free state; ND, newly diagnosed; OS, overall survival; RFS, relapse-free survival; R/R, relapsed/refractory.

Chemical modifications of monoclonal antibodies have long been pursued as a strategy to improve their clinical utility.2  Most widely tested are combinations with cytotoxic drugs (antibody–drug conjugates), toxic proteins from microorganisms or plants (immunotoxins), and radionuclides (radioimmunoconjugates).

Antibody–drug conjugates

The most experience with any antibody-based therapeutic has been with GO, a humanized CD33 antibody conjugated to a calicheamicin-γ1 derivative via a hydrolysable linker. GO was initially granted accelerated US marketing approval in 2000 for adults aged >60 years with relapsed CD33+ AML who were not candidates for cytotoxic chemotherapy. Approval was based on an “overall” response rate [CR/CR with incomplete platelet recovery (CRp)] of 30% in 142 such patients entering phase 2 trials.24  Subsequent studies have confirmed single-agent activity of GO in newly diagnosed and relapsed/refractory non-APL AML, but overall response rates have usually not exceeded 25% to 35% and were occasionally very low.25  GO has higher activity in APL,26  likely because of the typically bright, uniform expression of CD33 on APL blasts, the lack of significant drug transporter activity, and possibly the expression of CD33 on underlying LSCs. Although many of the studies with GO were small and lacked comparative groups, several randomized trials have provided controlled data on the role of GO alone or in combination with other cytotoxic agents in various disease situations (Table 2). Most information comes from 5 studies that investigated GO in addition to intensive chemotherapy in adults with newly diagnosed AML.27-31  The 5 studies used GO in different schedules [3 mg/m2 on day 1 in 2 trials, 6 mg/m2 on day 4 in 2 trials, and 3 mg/m2 (up to a maximum of 5 mg/dose) on days 1, 4, and 7 in one trial]. An individual data meta-analysis of all 3,325 patients on these trials showed that GO significantly reduced relapse risk and improved survival. The reduction in relapse risk appeared greater with the fractionated dosing of GO than the nonfractionated schedule. Survival benefit was greatest in patients with favorable cytogenetics and was also seen in those with intermediate but not adverse cytogenetics. There was no increased 30-day mortality with the 3 mg/m2 dosing schedules, but an increase in early mortality was observed with the 6 mg/m2 dose in the Southwest Oncology Group (SWOG) S0106 trial.32  These findings are complemented by data from a large randomized pediatric trial [Children's Oncology Group (COG) AAML0531] in which the addition of GO to conventional chemotherapy significantly improved event-free survival and relapse risk but not remission rates or overall survival. In contrast to the adult trials, GO was beneficial in all risk groups in AAML0531.33  GO alone also provided a very modest benefit over best supportive care and hydroxyurea in untreated older adults considered “unfit” for intensive chemotherapy.34  Conversely, GO offered no benefit when used sequentially with MEC over MEC alone for older adults with untreated AML and was too toxic for those older than 70 years.35  It also did not improve survival when added to LDAC in this patient population despite nearly doubling the remission rate.36  Moreover, several studies found no benefit of GO during postremission therapy or as maintenance treatment.27,31,37-39  Because of the lack of overall improvement in outcome and concern over increased early mortality in the SWOG S0106 trial, GO was withdrawn from the commercial market in most countries in 2010. Discrepancies between S0106 and the other randomized trials in adult AML have been a topic of discussions in recent years, particularly with regard to the unexpectedly low early death rate in the control arm and the unequal doses of daunorubicin used in the two treatment arms of S0106. Nevertheless, despite repeated calls by many leukemia experts, GO has not (yet) been reintroduced.

Table 2.

Randomized studies of GO in previously untreated AML

Randomized studies of GO in previously untreated AML
Randomized studies of GO in previously untreated AML

ADE indicates cytarabine/daunorubicin/etoposide; ALFA, Acute Leukemia French Association; CI, confidence interval; DA, daunorubicin/cytarabine; DClo, daunorubicin/clofarabine; DFS, disease-free survival; EFS, event-free survival; FLAG-IDA, fludarabine/cytarabine/granulocyte colony-stimulating factor/idarubicin; GOELAMS, Groupe Ouest Est d'Etude des Leucémies aiguës et Autres Maladies du Sang; HR, high risk; IR, intermediate risk; LRF, Leukaemia Research Fund; MA, mitoxantrone/cytarabine; MRC, Medical Research Council; NCRI, National Cancer Research Institute; OR, odds ratio; ORR, overall response rate (CR + CRi); OS, overall survival; RR, relapse risk; TRM, treatment-related mortality.

* Daunorubicin was used at 60 mg/m2/d in the control arm and 45 mg/m2/d in the GO arm.

The effectiveness of GO is curtailed by non-uniform drug conjugation, extrusion of the calicheamicin moiety via drug transporters, the limited amount of cell-surface CD33, and the relatively slow internalization kinetics of CD33/antibody complexes.25  Advancements in conjugation and linker technology and potency of the cytotoxic drug have resulted in SGN-CD33A, a humanized CD33 antibody with engineered cysteines carrying a synthetic DNA crosslinking pyrrolobenzodiazepine dimer via a protease-cleavable linker. Unlike GO, SGN-CD33A maintains activity in the presence of drug transporters in preclinical AML models.40  An ongoing phase 1 study primarily in older adults who either had relapsed/refractory AML or declined intensive therapy for newly diagnosed disease indicates that SGN-CD33A indeed has antileukemic activity: of 17 patients treated at a higher dose, this drug resulted in bone marrow blast clearance in 8 (47%), with 5 (29%) achieving either a CR or CRi.41  In contrast, phase 1 studies found low activity of AVE9633 (huMy9-6-DM4), a humanized CD33 antibody conjugated to a thiol-containing maytansinoid derivative, in adults with relapsed or refractory AML, and its clinical development has been terminated.42 

Immunotoxins

Under optimal experimental conditions, very few molecules of the toxic proteins used in immunotoxins are necessary to kill a eukaryotic cell, but such therapeutics have had limited efficacy in AML (Table 3). For example, HuM195/rGel, a CD33 antibody carrying recombinant gelonin, did not produce any CR or PR among 22 evaluable patients with advanced myeloid malignancies.43  Likewise, SL-401, a CD123-directed immunotoxin consisting of a fusion protein between IL-3 and the catalytic and translocation domains of diphtheria toxin, resulted in only 2 durable CRs and 5 PRs in 59 unselected patients with relapsed/refractory AML.44  Whether this drug is more efficacious in these patients when only minimal amounts of AML are present is currently unknown. More encouraging were results obtained in an expansion arm of that study in adults with blastic plasmacytoid dendritic cell neoplasm (BPDCN), a myeloid malignancy characterized by CD123 overexpression: among 9 evaluable patients, a single course of SL-401 yielded 5 CRs and 2 PRs, with responses lasting for a median of 5 months.45  Although additional studies are needed to elucidate why many immunotoxins are essentially ineffective in leukemia, postulated resistance mechanisms include neutralizing antibodies leading to clearance of circulating drug, limited antigen binding attributable to low-level target antigen expression, inability of the drug to infiltrate the bone marrow niche, and interference with any of the steps required for the toxins to reach the cytosol and exert their effects,46  including lysosomal degradation, which, based on preclinical models, may play a role particularly in multidrug-resistant leukemia cells.47 

Table 3.

Selected nontransplant studies with immunotoxins as monotherapy in AML

Selected nontransplant studies with immunotoxins as monotherapy in AML
Selected nontransplant studies with immunotoxins as monotherapy in AML

BM indicates bone marrow; DLT, dose-limiting toxicity; ND, newly diagnosed; PB, peripheral blood; R/R, relapsed/refractory.

Radioimmunoconjugates

AML is exquisitely radiosensitive. Therefore, radionuclides are a natural choice to arm antibodies. Still, partly because of logistic hurdles and concerns over long-term safety, radioimmunoconjugates are currently not accepted widely as treatment for AML.48-50  The relatively long range of β-emitting radioisotopes (eg, 131I, 90Y, or 188Re) of up to 10 mm provides a “crossfire” effect on nearby cancer cells that do not express the target antigen or are not easily accessible to antibody. The much shorter emission range over just a few cell diameters and higher linear energy transfer of α-emitting radioisotopes (eg, 213Bi, 225Ac, or 211At) allows a more selective leukemia cell kill, but their availability is more limited and their radiolabeling chemistry more challenging. Several trials using radiolabeled antibodies to intensify conditioning before hematopoietic cell transplantation (HCT) have demonstrated tolerable toxicities and antileukemic activity, leading to their incorporation into transplant conditioning regimens even in older adults (Table 4).51-60  Although remissions have been achieved in most patients, the lack of well-controlled studies makes it difficult to know whether inclusion of radiolabeled antibodies indeed improves outcomes of HCT.8,61  More recently, “pretargeted” strategies that separate the targeting vehicle from the subsequently administered radioisotope62,63  (shown in preclinical models to improve tumor/organ ratios of absorbed radioactivity) are clinically investigated as a means to optimize delivery of radiation during transplant conditioning.64  In parallel, limited efforts have been undertaken to use α-emitting antibodies outside the HCT setting (Table 5).65 213Bi-labeled lintuzumab showed moderate antileukemic activity as a single agent but produced some remissions in a subsequent phase 1/2 study when used sequentially with cytarabine.66,67  Likewise, 225Ac-labeled lintuzumab, which can overcome some of the logistic challenges of the short half-life of 213Bi of only 46 min, demonstrated some antileukemic effects as a single agent and is being combined with LDAC in untreated AML.68,69 

Table 4.

Selected transplant studies with radioimmunoconjugates in AML

Selected transplant studies with radioimmunoconjugates in AML
Selected transplant studies with radioimmunoconjugates in AML

ATG indicates anti-thymocyte globulin; Bu, busulfan; Conv, conventional-intensity conditioning; Cy, cyclophosphamide; DFS, disease-free survival; DLT, dose-limiting toxicity; Flu, fludarabine; GVHD, graft-versus-host disease; Mel, melphalan; MUD, matched unrelated donor; NRM, non-relapse mortality; OS, overall survival; RIC, reduced-intensity conditioning, R/R, relapsed/refractory; TBI, total body irradiation; TRM, treatment-related mortality; TT, thiotepa.

Table 5.

Selected nontransplant studies with radioimmunoconjugates in AML

Selected nontransplant studies with radioimmunoconjugates in AML
Selected nontransplant studies with radioimmunoconjugates in AML

BM indicates bone marrow; CMML, chronic myelomonocytic leukemia; CRp, CR with incomplete platelet count recovery; DLT, dose-limiting toxicity; ND, newly diagnosed; PB, peripheral blood; R/R, relapsed/refractory.

The randomized studies with GO validate CD33 as a target for antigen-directed immunotherapies at least for a subset of patients with AML and highlight the potential value of such approaches in this disease. Advances in structural engineering and chemical modification of antibodies give hope that improved antibody-based therapeutics will soon be available that could overcome the limitations of previous generation constructs. Adoptive immunotherapies using genetically modified immune effector cells and vaccination strategies complement the spectrum of evolving antigen-specific immunotherapies for AML. These efforts, summarized in the following sections, are paralleled by efforts to uncover new target antigens.70 

New unconjugated and conjugated monospecific antibodies

For unconjugated antibodies, current efforts are focused on genetic manipulation to increase the interaction with activating Fc receptors, particularly CD16, and to improve antibody-dependent cytotoxicity mediated, for example, by NK cells.71  Fc-engineered antibodies targeting a variety of AML antigens (eg, CD33, CD38, CD96, CD123, CD133, CD135, CD157, CD300F, and T-cell immunoglobulin mucin receptor 3 [TIM-3]) have been preclinically tested recently, and some constructs targeting CD33 (MAb 33.1, BI 836858), CD123 (CSL362), and CD157 (MEN1112) have entered clinical testing.70,72  Instead of targeting AML antigens to elicit direct cytotoxic effects, some antibodies aim to amplify native immune responses, for example via modulation of programmed cell death-1 (PD-1)/programmed death ligand-1 (PD-L1) signaling, which may serve as a negative regulatory mechanism in AML. This possibility has been raised by preclinical data demonstrating that antibodies that block PD-L1 decrease AML burden in experimental mice.73  The PD-1 antibody CT-011 has already been assessed for safety in patients with relapsed/refractory hematologic malignancies including AML,74  and phase 2 trials of the PD-1 antibody nivolumab as monotherapy or CT-011 combined with a cell-based vaccine in patients with AML in remission have been initiated.

For drug-carrying antibodies, interest remains centered on CD33, with IMGN779, a CD33 antibody-drug conjugate that uses a DNA crosslinking indolino-benzodiazepine dimer, being another potential successor to GO. IMGN779 has shown high activity in preclinical AML models, particularly those with FLT3/internal tandem duplication (ITD) mutations,75  and may soon reach the clinic. In contrast, immunotoxins under preclinical development primarily aim at CD123 but also target other antigens (eg, CD64) and explore human toxins (eg, granzymes) to minimize neutralizing antibody formation, but it is unclear whether or when such agents will be introduced in the clinic.

Bispecific antibodies

One long-pursued engineering approach to increase antitumor activity is to create antibodies with dual affinities for a tumor cell antigen and an antigen on immune effector cells, such as cytotoxic T cells,2  directly engaging the latter in the elimination of the former. Initial success was limited by suboptimal effector cell recruitment and challenges with large-scale antibody production.76,77  These shortcomings may be overcome using small molecules consisting of single-chain variable fragments (scFVs), for example, bivalent or tetravalent antibodies that combine the scFVs on one [eg, bispecific T-cell–engaging (BiTE) antibodies] or two [eg, dual-affinity retargeting (DART) antibodies or tandem diabodies] polypeptide chains. These molecules bring polyclonal immune effector cells close to tumor cells and force formation of an immunological lytic synapse that triggers immune cell activation and proliferation and consequent destruction of attached tumor cells at a low effector/target cell ratio.77,78  High efficacy for some patients with acute leukemias was demonstrated in several positive trials with the CD19/CD3 BiTE antibody blinatumomab in chemotherapy-resistant Philadelphia chromosome-negative B lymphoblastic leukemia, which led to recent regulatory approval for this patient population. Efforts in AML primarily focus on CD3-directed agents that target CD33 or CD123, but a variety of alternate targets on tumor cells (eg, CLL-1 and FLT3) as well as immune effector cells (eg, CD16 or NKG2D) are being investigated. Preclinical studies with CD33/CD3-directed molecules, including the BiTE antibody AMG 330, have shown potent lysis of CD33+ AML cells together with healthy donor T cells or autologous T cells from AML patients and broad activity in human AML specimens.6,79-83  A CD123/CD3 DART molecule (MGD006) has entered phase 1 testing, and others, including AMG 330, will follow soon.

Adoptive immunotherapy

Adoptive immunotherapy using T cells expressing chimeric antigen receptors (CARs) or modified T-cell receptor (TCR) genes has drawn major attention in acute leukemias. CARs are hybrid single-chain receptor constructs containing an extracellular tumor antigen-recognizing domain linked to an intracellular component comprising the CD3 zeta chain with or without additional costimulatory endodomains to activate the immune effector cell during tumor cell binding.84  CD19-directed CAR T cells have shown very high remission rates and durable remissions in children and adults with B lymphoblastic leukemia, even among those who previously underwent allogenic HCT, demonstrating the potential of this approach.85,86  In AML, clinical experience is thus far very limited, with a study on 4 adults with relapsed disease treated with cytotoxic drugs followed by CAR T cells directed at Lewis-Y, a carbohydrate antigen overexpressed by malignant myeloid cells: of the 3 patients who had MRD at the time of cell infusion, one had a cytogenetic remission lasting 5 months, whereas the others had stable disease lasting 1.5 and 23 months, but ultimately all experienced disease progression despite persistence of CAR T cells and target antigen expression on myeloblasts.87  Although alternative antigens are explored,88,89  current efforts focus primarily on CD33 and CD123.90-95  Clinical trials testing CAR T cells directed at each of these antigens are ongoing, with one early report available of a patient who had a transient decrease in marrow blasts after receiving CD33-directed CAR T cells.96  Less explored are T cells expressing modified TCR genes. Besides leading to more physiological cellular activation, TCR-modified cells offer the advantage of being able to recognize intracellular epitopes expressed on the cell surface in the context of a peptide/major histocompatibility complex. This strategy is being studied in a phase 1/2 study in which patients with high-risk AML receive donor-derived Ebstein Barr virus- or cytomegalovirus-specific CD8+ T cells transduced with a high-affinity TCR specific for an HLA A*02:01-restricted WT1 peptide together with IL-2 after allogenic HCT. Early results on 9 treated patients, some of whom had measurable disease at the time of T-cell administration, suggest the possibility of antileukemic activity.97 

Vaccines

Antibodies or immune effector cells directed against leukemia cell antigens are present in many AML patients.4  In some cases, these adaptive immune responses may have antileukemic effects.98  Although such observations support the potential value of active immunotherapy as treatment strategy in AML, vaccine-based therapies are still in their infancy. Most vaccination strategies target WT1, proteinase-1 (a specific PRTN3-derived peptide), and hyaluronan-mediated motility receptor (CD168) peptides. Case series and early-phase trials demonstrate the safety of such vaccines in AML patients with active disease, in remission after chemotherapy, and even after allogenic HCT and document immunologic and possible clinical responses in some cases.99-104  By introducing full-length antigens, cell-based vaccines may circumvent some of the limitations of peptide vaccines; still, early trials in which dendritic cells (DCs) were pulsed with leukemic lysates or directly derived from leukemic blasts yielded underwhelming responses.105-107  More effective may be autologous DCs transfected with WT1 mRNA, which yielded clinical responses, including remissions in patients who achieved a partial response with chemotherapy and molecular remissions in patients with residual disease. DC/AML fusion cell vaccines and “off-the-shelf” DC vaccines that are engineered from a myeloid leukemic cell line endogenously expressing a range of AML-associated antigens have also been used in early-phase trials,108-111  but additional studies are needed to demonstrate unequivocally that improved disease control can be achieved with these agents.

With the many antigen-specific immunotherapeutics currently under development, we can be hopeful that we can soon expand the armamentarium for treatment of AML. Nonetheless, a number of important questions remain open at this time and need to be addressed in future investigations. Perhaps foremost is the question of the ideal target antigen(s). It is likely that there is no single optimal antigen in a disease as heterogeneous as AML; the clinical experience with GO is an important reminder that AML cannot be regarded as one disease. For any particular antigen, only a subset of patients may be suitable, and the choice of target antigen may depend on the purpose of the therapy, that is to say whether the primary intent is to reduce the burden of leukemia cells in a patient with active disease or to eradicate MRD and the very rare underlying LSCs. Elimination of LSCs may be an ideal goal for immunotherapies but will require a deepened understanding of the immunophenotypic and functional properties of these elusive cells. LSC-directed therapy may be most successful if the immunophenotypic properties of LSC are clearly distinguishable from normal hematopoietic stem cells; the particular susceptibility of APL to CD33-targeted immunotherapies may support this notion.7  If it is necessary to target more than one antigen for maximum treatment benefit, in what sequence should the therapeutics be used, and how can they be combined with conventional chemotherapeutic or immune-stimulating drugs to achieve optimal effects? Because the display of target antigens may change on AML cells and normal cells during treatment, for example, as the cell cycle status changes, pretreatment antigenic profiles may not adequately reflect expression patterns during therapy. Of course, the exact antigenic profile of some of the targeted cells such as LSCs may not be known at all given their rarity and our current lack of ability to isolate/enrich them effectively. Especially in light of the diversity of explored therapies, another open question is which class of therapeutics will provide optimum outcomes, augmenting the anti-AML effect while minimizing toxicity to normal cells. Life-threatening toxicities with some passive immunotherapies, particularly gene-modified immune effector cells, are now well recognized. Very likely, the ideal format of immunotherapy will depend on many factors, including target antigen, disease stage, and patient characteristics. Finally, if AML-associated antigens such as CD33 or CD123 are targeted, what degree of supportive care is needed? Highly potent therapeutics, such as bispecific antibodies and CAR T cells, will have profound effects on normal hematopoietic cells expressing these antigens and cause prolonged cytopenias. This will put high demands on supportive care measures in a vulnerable patient population. Some therapeutics may even require autologous or allogenic stem cell rescue for safe administration to abrogate their effects on normal hematopoietic cells and/or immune effector cells and therefore could primarily play a role during transplant conditioning. Answers to such questions will ultimately be required to optimize the clinical use of antigen-specific immunotherapies in AML and to ensure that they become an effective tool for this disease for which outcomes have remained unsatisfactory.

We thank Drs. Irwin D. Bernstein, Elihu H. Estey, and Wendy Stock for critical reading of this manuscript.

R.B.W. is a Leukemia & Lymphoma Society Scholar in Clinical Research.

Roland B. Walter, Clinical Research Division, Fred Hutchinson Cancer Research Center; 1100 Fairview Ave N, D2-190, Seattle, WA 98109-1024. Phone: 206-667-3599; Fax: 206-667-6519; e-mail: rwalter@fredhutch.org.

1
Weiner
 
LM
Surana
 
R
Wang
 
S
Monoclonal antibodies: versatile platforms for cancer immunotherapy
Nat Rev Immunol
2010
, vol. 
10
 
5
(pg. 
317
-
327
)
2
Weiner
 
LM
Murray
 
JC
Shuptrine
 
CW
Antibody-based immunotherapy of cancer
Cell
2012
, vol. 
148
 
6
(pg. 
1081
-
1084
)
3
Bachireddy
 
P
Burkhardt
 
UE
Rajasagi
 
M
Wu
 
CJ
Haematological malignancies: at the forefront of immunotherapeutic innovation
Nat Rev Cancer
2015
, vol. 
15
 
4
(pg. 
201
-
215
)
4
Anguille
 
S
Van Tendeloo
 
VF
Berneman
 
ZN
Leukemia-associated antigens and their relevance to the immunotherapy of acute myeloid leukemia
Leukemia
2012
, vol. 
26
 
10
(pg. 
2186
-
2196
)
5
Pollard
 
JA
Alonzo
 
TA
Loken
 
M
, et al. 
Correlation of CD33 expression level with disease characteristics and response to gemtuzumab ozogamicin containing chemotherapy in childhood AML
Blood
2012
, vol. 
119
 
16
(pg. 
3705
-
3711
)
6
Krupka
 
C
Kufer
 
P
Kischel
 
R
, et al. 
CD33 target validation and sustained depletion of AML blasts in long-term cultures by the bispecific T-cell-engaging antibody AMG 330
Blood
2014
, vol. 
123
 
3
(pg. 
356
-
365
)
7
Walter
 
RB
Appelbaum
 
FR
Estey
 
EH
Bernstein
 
ID
Acute myeloid leukemia stem cells and CD33-targeted immunotherapy
Blood
2012
, vol. 
119
 
26
(pg. 
6198
-
6208
)
8
Walter
 
RB
Press
 
OW
Bernstein
 
ID
Andreef
 
M
Antibody-based therapeutics targeting CD33, CD45, and CD66
Targeted Therapy of Acute Myeloid Leukemia
2015
New York, NY
Springer
(pg. 
531
-
555
)
9
Ball
 
ED
Selvaggi
 
K
Hurd
 
D
, et al. 
Phase I clinical trial of serotherapy in patients with acute myeloid leukemia with an immunoglobulin M monoclonal antibody to CD15
Clin Cancer Res
1995
, vol. 
1
 
9
(pg. 
965
-
972
)
10
Tibes
 
R
Keating
 
MJ
Ferrajoli
 
A
, et al. 
Activity of alemtuzumab in patients with CD52-positive acute leukemia
Cancer
2006
, vol. 
106
 
12
(pg. 
2645
-
2651
)
11
Caron
 
PC
Dumont
 
L
Scheinberg
 
DA
Supersaturating infusional humanized anti-CD33 monoclonal antibody HuM195 in myelogenous leukemia
Clin Cancer Res
1998
, vol. 
4
 
6
(pg. 
1421
-
1428
)
12
Feldman
 
E
Kalaycio
 
M
Weiner
 
G
, et al. 
Treatment of relapsed or refractory acute myeloid leukemia with humanized anti-CD33 monoclonal antibody HuM195
Leukemia
2003
, vol. 
17
 
2
(pg. 
314
-
318
)
13
Jurcic
 
JG
DeBlasio
 
T
Dumont
 
L
Yao
 
TJ
Scheinberg
 
DA
Molecular remission induction with retinoic acid and anti-CD33 monoclonal antibody HuM195 in acute promyelocytic leukemia
Clin Cancer Res
2000
, vol. 
6
 
2
(pg. 
372
-
380
)
14
Raza
 
A
Jurcic
 
JG
Roboz
 
GJ
, et al. 
Complete remissions observed in acute myeloid leukemia following prolonged exposure to lintuzumab: a phase 1 trial
Leuk Lymphoma
2009
, vol. 
50
 
8
(pg. 
1336
-
1344
)
15
Feldman
 
EJ
Brandwein
 
J
Stone
 
R
, et al. 
Phase III randomized multicenter study of a humanized anti-CD33 monoclonal antibody, lintuzumab, in combination with chemotherapy, versus chemotherapy alone in patients with refractory or first-relapsed acute myeloid leukemia
J Clin Oncol
2005
, vol. 
23
 
18
(pg. 
4110
-
4116
)
16
Sekeres
 
MA
Lancet
 
JE
Wood
 
BL
, et al. 
Randomized phase IIb study of low-dose cytarabine and lintuzumab versus low-dose cytarabine and placebo in older adults with untreated acute myeloid leukemia
Haematologica
2013
, vol. 
98
 
1
(pg. 
119
-
128
)
17
Ossenkoppele
 
GJ
Stussi
 
G
Maertens
 
J
, et al. 
Addition of bevacizumab to chemotherapy in acute myeloid leukemia at older age: a randomized phase 2 trial of the Dutch-Belgian Cooperative Trial Group for Hemato-Oncology (HOVON) and the Swiss Group for Clinical Cancer Research (SAKK)
Blood
2012
, vol. 
120
 
24
(pg. 
4706
-
4711
)
18
He
 
SZ
Busfield
 
S
Ritchie
 
DS
, et al. 
A Phase 1 study of the safety, pharmacokinetics and anti-leukemic activity of the anti-CD123 monoclonal antibody CSL360 in relapsed, refractory or high-risk acute myeloid leukemia
Leuk Lymphoma
2015
, vol. 
56
 
5
(pg. 
1406
-
1415
)
19
Williams
 
R
Discontinued drugs in 2011: oncology drugs
Expert Opin Investig Drugs
2013
, vol. 
22
 
1
(pg. 
9
-
34
)
20
Sanford
 
D
Blum
 
W
Ravandi
 
R
, et al. 
Efficacy and safety of an anti-FLT3 antibody (LY3012218) in patients with relapsed acute myeloid leukemia [abstract]
J Clin Oncol
2015
, vol. 
33
 
Suppl
pg. 
7059
 
21
Swords
 
RT
Wei
 
AH
Durrant
 
S
, et al. 
KB004, a novel non-fucosylated humaneered antibody, targeting EphA3, is active and well tolerated in a phase I/II study of advanced hematologic malignancies [abstract]
Blood
2014
, vol. 
124
 
21
pg. 
3756
 
22
Becker
 
PS
Foran
 
JM
Altman
 
JK
, et al. 
Targeting the CXCR4 pathway: safety, tolerability and clinical activity of ulocuplumab (BMS-936564), an anti-CXCR4 antibody, in relapsed/refractory acute myeloid leukemia [abstract]
Blood
2014
, vol. 
124
 
21
pg. 
386
 
23
Vey
 
N
Bourhis
 
JH
Boissel
 
N
, et al. 
A phase 1 trial of the anti-inhibitory KIR mAb IPH2101 for AML in complete remission
Blood
2012
, vol. 
120
 
22
(pg. 
4317
-
4323
)
24
Bross
 
PF
Beitz
 
J
Chen
 
G
, et al. 
Approval summary: gemtuzumab ozogamicin in relapsed acute myeloid leukemia
Clin Cancer Res
2001
, vol. 
7
 
6
(pg. 
1490
-
1496
)
25
Cowan
 
AJ
Laszlo
 
GS
Estey
 
EH
Walter
 
RB
Antibody-based therapy of acute myeloid leukemia with gemtuzumab ozogamicin
Front Biosci (Landmark Ed)
2013
, vol. 
18
 
4
(pg. 
1311
-
1334
)
26
Breccia
 
M
Lo-Coco
 
F
Gemtuzumab ozogamicin for the treatment of acute promyelocytic leukemia: mechanisms of action and resistance, safety and efficacy
Expert Opin Biol Ther
2011
, vol. 
11
 
2
(pg. 
225
-
234
)
27
Burnett
 
AK
Hills
 
RK
Milligan
 
D
, et al. 
Identification of patients with acute myeloblastic leukemia who benefit from the addition of gemtuzumab ozogamicin: results of the MRC AML15 trial
J Clin Oncol
2011
, vol. 
29
 
4
(pg. 
369
-
377
)
28
Burnett
 
AK
Russell
 
NH
Hills
 
RK
, et al. 
Addition of gemtuzumab ozogamicin to induction chemotherapy improves survival in older patients with acute myeloid leukemia
J Clin Oncol
2012
, vol. 
30
 
32
(pg. 
3924
-
3931
)
29
Castaigne
 
S
Pautas
 
C
Terré
 
C
, et al. 
Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study
Lancet
2012
, vol. 
379
 
9825
(pg. 
1508
-
1516
)
30
Delaunay
 
J
Recher
 
C
Pigneux
 
A
, et al. 
Addition of gemtuzumab ozogamycin to chemotherapy improves event-free survival but not overall survival of AML patients with intermediate cytogenetics not eligible for allogeneic transplantation. Results of the GOELAMS AML 2006 IR study [abstract]
Blood
2011
, vol. 
118
 
21
(pg. 
37
-
38
)
31
Petersdorf
 
SH
Kopecky
 
KJ
Slovak
 
M
, et al. 
A phase 3 study of gemtuzumab ozogamicin during induction and postconsolidation therapy in younger patients with acute myeloid leukemia
Blood
2013
, vol. 
121
 
24
(pg. 
4854
-
4860
)
32
Hills
 
RK
Castaigne
 
S
Appelbaum
 
FR
, et al. 
Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: a meta-analysis of individual patient data from randomised controlled trials
Lancet Oncol
2014
, vol. 
15
 
9
(pg. 
986
-
996
)
33
Gamis
 
AS
Alonzo
 
TA
Meshinchi
 
S
, et al. 
Gemtuzumab ozogamicin in children and adolescents with de novo acute myeloid leukemia improves event-free survival by reducing relapse risk: results from the randomized phase III Children's Oncology Group trial AAML0531
J Clin Oncol
2014
, vol. 
32
 
27
(pg. 
3021
-
3032
)
34
Amadori
 
S
Suciu
 
S
Selleslag
 
D
, et al. 
Improved overall survival with gemtuzumab ozogamicin (GO) compared with best supportive care (BSC) in elderly patients with untreated acute myeloid leukemia (AML) not considered fit for intensive chemotherapy: final results from the randomized phase III study (AML-19) of the EORTC and Gimema Leukemia Groups [abstract]
Blood
2014
, vol. 
124
 
21
pg. 
619
 
35
Amadori
 
S
Suciu
 
S
Stasi
 
R
, et al. 
Sequential combination of gemtuzumab ozogamicin and standard chemotherapy in older patients with newly diagnosed acute myeloid leukemia: results of a randomized phase III trial by the EORTC and GIMEMA consortium (AML-17)
J Clin Oncol
2013
, vol. 
31
 
35
(pg. 
4424
-
4430
)
36
Burnett
 
AK
Hills
 
RK
Hunter
 
AE
, et al. 
The addition of gemtuzumab ozogamicin to low-dose Ara-C improves remission rate but does not significantly prolong survival in older patients with acute myeloid leukaemia: results from the LRF AML14 and NCRI AML16 pick-a-winner comparison
Leukemia
2013
, vol. 
27
 
1
(pg. 
75
-
81
)
37
Löwenberg
 
B
Beck
 
J
Graux
 
C
, et al. 
Gemtuzumab ozogamicin as postremission treatment in AML at 60 years of age or more: results of a multicenter phase 3 study
Blood
2010
, vol. 
115
 
13
(pg. 
2586
-
2591
)
38
Fernandez
 
HF
Sun
 
Z
Litzow
 
MR
, et al. 
Autologous transplantation gives encouraging results for young adults with favorable-risk acute myeloid leukemia, but is not improved with gemtuzumab ozogamicin
Blood
2011
, vol. 
117
 
20
(pg. 
5306
-
5313
)
39
Hasle
 
H
Abrahamsson
 
J
Forestier
 
E
, et al. 
Gemtuzumab ozogamicin as postconsolidation therapy does not prevent relapse in children with AML: results from NOPHO-AML 2004
Blood
2012
, vol. 
120
 
5
(pg. 
978
-
984
)
40
Kung Sutherland
 
MS
Walter
 
RB
Jeffrey
 
SC
, et al. 
SGN-CD33A: a novel CD33-targeting antibody-drug conjugate using a pyrrolobenzodiazepine dimer is active in models of drug-resistant AML
Blood
2013
, vol. 
122
 
8
(pg. 
1455
-
1463
)
41
Stein
 
EM
Stein
 
A
Walter
 
RB
, et al. 
Interim analysis of a phase 1 trial of SGN-CD33A in patients with CD33-positive acute myeloid leukemia (AML) [abstract]
Blood
2014
, vol. 
124
 
21
pg. 
623
 
42
Lapusan
 
S
Vidriales
 
MB
Thomas
 
X
, et al. 
Phase I studies of AVE9633, an anti-CD33 antibody-maytansinoid conjugate, in adult patients with relapsed/refractory acute myeloid leukemia
Invest New Drugs
2012
, vol. 
30
 
3
(pg. 
1121
-
1131
)
43
Borthakur
 
G
Rosenblum
 
MG
Talpaz
 
M
, et al. 
Phase 1 study of an anti-CD33 immunotoxin, humanized monoclonal antibody M195 conjugated to recombinant gelonin (HUM-195/rGEL), in patients with advanced myeloid malignancies
Haematologica
2013
, vol. 
98
 
2
(pg. 
217
-
221
)
44
Konopleva
 
M
Hogge
 
DE
Rizzieri
 
DA
, et al. 
SL-401, a targeted therapy directed to the interleukin-3 receptor present on leukemia blasts and cancer stem cells, is active as a single agent in patients with advanced AML [abstract]
Blood
2012
, vol. 
120
 
21
pg. 
3625
 
45
Frankel
 
AE
Woo
 
JH
Ahn
 
C
, et al. 
Activity of SL-401, a targeted therapy directed to interleukin-3 receptor, in blastic plasmacytoid dendritic cell neoplasm patients
Blood
2014
, vol. 
124
 
3
(pg. 
385
-
392
)
46
Wayne
 
AS
Fitzgerald
 
DJ
Kreitman
 
RJ
Pastan
 
I
Immunotoxins for leukemia
Blood
2014
, vol. 
123
 
16
(pg. 
2470
-
2477
)
47
McGrath
 
MS
Rosenblum
 
MG
Philips
 
MR
Scheinberg
 
DA
Immunotoxin resistance in multidrug resistant cells
Cancer Res
2003
, vol. 
63
 
1
(pg. 
72
-
79
)
48
Sharkey
 
RM
Goldenberg
 
DM
Cancer radioimmunotherapy
Immunotherapy
2011
, vol. 
3
 
3
(pg. 
349
-
370
)
49
Steiner
 
M
Neri
 
D
Antibody-radionuclide conjugates for cancer therapy: historical considerations and new trends
Clin Cancer Res
2011
, vol. 
17
 
20
(pg. 
6406
-
6416
)
50
Palanca-Wessels
 
MC
Press
 
OW
Advances in the treatment of hematologic malignancies using immunoconjugates
Blood
2014
, vol. 
123
 
15
(pg. 
2293
-
2301
)
51
Appelbaum
 
FR
Matthews
 
DC
Eary
 
JF
, et al. 
The use of radiolabeled anti-CD33 antibody to augment marrow irradiation prior to marrow transplantation for acute myelogenous leukemia
Transplantation
1992
, vol. 
54
 
5
(pg. 
829
-
833
)
52
Burke
 
JM
Caron
 
PC
Papadopoulos
 
EB
, et al. 
Cytoreduction with iodine-131-anti-CD33 antibodies before bone marrow transplantation for advanced myeloid leukemias
Bone Marrow Transplant
2003
, vol. 
32
 
6
(pg. 
549
-
556
)
53
Matthews
 
DC
Appelbaum
 
FR
Eary
 
JF
, et al. 
Phase I study of (131)I-anti-CD45 antibody plus cyclophosphamide and total body irradiation for advanced acute leukemia and myelodysplastic syndrome
Blood
1999
, vol. 
94
 
4
(pg. 
1237
-
1247
)
54
Pagel
 
JM
Appelbaum
 
FR
Eary
 
JF
, et al. 
131I-anti-CD45 antibody plus busulfan and cyclophosphamide before allogeneic hematopoietic cell transplantation for treatment of acute myeloid leukemia in first remission
Blood
2006
, vol. 
107
 
5
(pg. 
2184
-
2191
)
55
Pagel
 
JM
Gooley
 
TA
Rajendran
 
J
, et al. 
Allogeneic hematopoietic cell transplantation after conditioning with 131I-anti-CD45 antibody plus fludarabine and low-dose total body irradiation for elderly patients with advanced acute myeloid leukemia or high-risk myelodysplastic syndrome
Blood
2009
, vol. 
114
 
27
(pg. 
5444
-
5453
)
56
Mawad
 
R
Gooley
 
TA
Rajendran
 
JG
, et al. 
Radiolabeled anti-CD45 antibody with reduced-intensity conditioning and allogeneic transplantation for younger patients with advanced acute myeloid leukemia or myelodysplastic syndrome
Biol Blood Marrow Transplant
2014
, vol. 
20
 
9
(pg. 
1363
-
1368
)
57
Bunjes
 
D
188Re-labeled anti-CD66 monoclonal antibody in stem cell transplantation for patients with high-risk acute myeloid leukemia
Leuk Lymphoma
2002
, vol. 
43
 
11
(pg. 
2125
-
2131
)
58
Klein
 
SA
Hermann
 
S
Dietrich
 
JW
Hoelzer
 
D
Martin
 
H
Transplantation-related toxicity and acute intestinal graft-versus-host disease after conditioning regimens intensified with Rhenium 188-labeled anti-CD66 monoclonal antibodies
Blood
2002
, vol. 
99
 
6
(pg. 
2270
-
2271
)
59
Ringhoffer
 
M
Blumstein
 
N
Neumaier
 
B
, et al. 
188Re or 90Y-labelled anti-CD66 antibody as part of a dose-reduced conditioning regimen for patients with acute leukaemia or myelodysplastic syndrome over the age of 55: results of a phase I-II study
Br J Haematol
2005
, vol. 
130
 
4
(pg. 
604
-
613
)
60
Koenecke
 
C
Hofmann
 
M
Bolte
 
O
, et al. 
Radioimmunotherapy with [(188)Re]-labelled anti-CD66 antibody in the conditioning for allogeneic stem cell transplantation for high-risk acute myeloid leukemia
Int J Hematol
2008
, vol. 
87
 
4
(pg. 
414
-
421
)
61
Walter
 
RB
Pagel
 
JM
Speer
 
TW
Targeted radionuclide therapy for leukemia
Targeted Radionuclide Therapy
2011
Philadelphia, PA
Lippincott Williams & Wilkins
(pg. 
441
-
457
)
62
Walter
 
RB
Press
 
OW
Pagel
 
JM
Pretargeted radioimmunotherapy for hematologic and other malignancies
Cancer Biother Radiopharm
2010
, vol. 
25
 
2
(pg. 
125
-
142
)
63
van de Watering
 
FC
Rijpkema
 
M
Robillard
 
M
Oyen
 
WJ
Boerman
 
OC
Pretargeted imaging and radioimmunotherapy of cancer using antibodies and bioorthogonal chemistry
Front Med (Lausanne)
2014
, vol. 
1
 pg. 
44
 
64
Mawad
 
R
Gooley
 
T
Rajendran
 
JG
, et al. 
Pretargeted radioimmunotherapy using an anti-CD45 antibody-streptavidin conjugate and radiolabeled DOTA-biotin in patients with high-risk acute leukemia or myelodysplastic syndrome undergoing allogeneic hematopoietic cell transplantation [abstract]
Biol Blood Marrow Transplant
2013
, vol. 
19
 
2 Suppl 2
(pg. 
S123
-
S124
)
65
Jurcic
 
JG
Rosenblat
 
TL
Targeted alpha-particle immunotherapy for acute myeloid leukemia
Am Soc Clin Oncol Educ Book
2014
(pg. 
e126
-
e131
)
66
Jurcic
 
JG
Larson
 
SM
Sgouros
 
G
, et al. 
Targeted alpha particle immunotherapy for myeloid leukemia
Blood
2002
, vol. 
100
 
4
(pg. 
1233
-
1239
)
67
Rosenblat
 
TL
McDevitt
 
MR
Mulford
 
DA
, et al. 
Sequential cytarabine and alpha-particle immunotherapy with bismuth-213-lintuzumab (HuM195) for acute myeloid leukemia
Clin Cancer Res
2010
, vol. 
16
 
21
(pg. 
5303
-
5311
)
68
Jurcic
 
JG
Rosenblat
 
TL
McDevitt
 
MR
, et al. 
Phase I trial of the targeted-particle nano-generator actinium 225 (225Ac)-lintuzumab (anti-CD33; HuM195) in acute myeloid leukemia (AML) [abstract]
Blood
2011
, vol. 
118
 
21
pg. 
768
 
69
Jurcic
 
JG
Ravandi
 
F
Pagel
 
JM
, et al. 
Phase I trial of the targeted alpha-particle nano-generator actinium-225 (225Ac)-lintuzumab (anti-CD33) in combination with low-dose cytarabine (LDAC) for older patients with untreated acute myeloid leukemia (AML) [abstract]
Blood
2013
, vol. 
122
 
21
pg. 
1460
 
70
Buckley
 
SA
Walter
 
RB
Update on antigen-specific immunotherapy of acute myeloid leukemia
Current Hematol Malig Rep
2015
, vol. 
10
 
2
(pg. 
65
-
75
)
71
Romain
 
G
Senyukov
 
V
Rey-Villamizar
 
N
, et al. 
Antibody Fc engineering improves frequency and promotes kinetic boosting of serial killing mediated by NK cells
Blood
2014
, vol. 
124
 
22
(pg. 
3241
-
3249
)
72
Smith
 
BD
Roboz
 
GJ
Walter
 
RB
, et al. 
First-in man, phase 1 study of CSL362 (anti-IL3Rα / anti-CD123 monoclonal antibody) in patients with CD123+ acute myeloid leukemia (AML) in CR at high risk for early relapse [abstract]
Blood
2014
, vol. 
124
 
21
pg. 
120
 
73
Zhang
 
L
Gajewski
 
TF
Kline
 
J
PD-1/PD-L1 interactions inhibit antitumor immune responses in a murine acute myeloid leukemia model
Blood
2009
, vol. 
114
 
8
(pg. 
1545
-
1552
)
74
Berger
 
R
Rotem-Yehudar
 
R
Slama
 
G
, et al. 
Phase I safety and pharmacokinetic study of CT-011, a humanized antibody interacting with PD-1, in patients with advanced hematologic malignancies
Clin Cancer Res
2008
, vol. 
14
 
10
(pg. 
3044
-
3051
)
75
Whiteman
 
KR
Noordhuis
 
P
Walker
 
R
, et al. 
The antibody-drug conjugate (ADC) IMGN779 is highly active in vitro and in vivo against acute myeloid leukemia (AML) with FLT3-ITD mutations [abstract]
Blood
2014
, vol. 
124
 
21
pg. 
2321
 
76
Carter
 
P
Improving the efficacy of antibody-based cancer therapies
Nat Rev Cancer
2001
, vol. 
1
 
2
(pg. 
118
-
129
)
77
Riethmüller
 
G
Symmetry breaking: bispecific antibodies, the beginnings, and 50 years on
Cancer Immun
2012
12
pg. 
12
 
78
Baeuerle
 
PA
Reinhardt
 
C
Bispecific T-cell engaging antibodies for cancer therapy
Cancer Res
2009
, vol. 
69
 
12
(pg. 
4941
-
4944
)
79
Arndt
 
C
von Bonin
 
M
Cartellieri
 
M
, et al. 
Redirection of T cells with a first fully humanized bispecific CD33-CD3 antibody efficiently eliminates AML blasts without harming hematopoietic stem cells
Leukemia
2013
, vol. 
27
 
4
(pg. 
964
-
967
)
80
Aigner
 
M
Feulner
 
J
Schaffer
 
S
, et al. 
T lymphocytes can be effectively recruited for ex vivo and in vivo lysis of AML blasts by a novel CD33/CD3-bispecific BiTE antibody construct
Leukemia
2013
, vol. 
27
 
5
(pg. 
1107
-
1115
)
81
Laszlo
 
GS
Gudgeon
 
CJ
Harrington
 
KH
, et al. 
Cellular determinants for preclinical activity of a novel CD33/CD3 bispecific T-cell engager (BiTE) antibody, AMG 330, against human AML
Blood
2014
, vol. 
123
 
4
(pg. 
554
-
561
)
82
Friedrich
 
M
Henn
 
A
Raum
 
T
, et al. 
Preclinical characterization of AMG 330, a CD3/CD33-bispecific T-cell-engaging antibody with potential for treatment of acute myelogenous leukemia
Mol Cancer Ther
2014
, vol. 
13
 
6
(pg. 
1549
-
1557
)
83
Harrington
 
KH
Gudgeon
 
CJ
Laszlo
 
GS
, et al. 
The broad anti-AML activity of the CD33/CD3 bispecific BiTE antibody construct, AMG 330, is impacted by disease stage and risk
PLoS One
2015
, vol. 
10
 
8
pg. 
e0135945
 
84
Fry
 
TJ
Mackall
 
CL
T-cell adoptive immunotherapy for acute lymphoblastic leukemia
Hematology Am Soc Hematol Educ Program
2013
, vol. 
2013
 (pg. 
348
-
353
)
85
Lee
 
DW
Kochenderfer
 
JN
Stetler-Stevenson
 
M
, et al. 
T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial
Lancet
2015
, vol. 
385
 
9967
(pg. 
517
-
528
)
86
Maude
 
SL
Frey
 
N
Shaw
 
PA
, et al. 
Chimeric antigen receptor T cells for sustained remissions in leukemia
N Engl J Med
2014
, vol. 
371
 
16
(pg. 
1507
-
1517
)
87
Ritchie
 
DS
Neeson
 
PJ
Khot
 
A
, et al. 
Persistence and efficacy of second generation CAR T cell against the LeY antigen in acute myeloid leukemia
Mol Ther
2013
, vol. 
21
 
11
(pg. 
2122
-
2129
)
88
Casucci
 
M
Nicolis di Robilant
 
B
Falcone
 
L
, et al. 
CD44v6-targeted T cells mediate potent antitumor effects against acute myeloid leukemia and multiple myeloma
Blood
2013
, vol. 
122
 
20
(pg. 
3461
-
3472
)
89
Lynn
 
RC
Poussin
 
M
Kalota
 
A
, et al. 
Targeting of folate receptor-beta on acute myeloid leukemia blasts with chimeric antigen receptor expressing T cells
Blood
2015
, vol. 
125
 
22
(pg. 
3466
-
3476
)
90
Tettamanti
 
S
Marin
 
V
Pizzitola
 
I
, et al. 
Targeting of acute myeloid leukaemia by cytokine-induced killer cells redirected with a novel CD123-specific chimeric antigen receptor
Br J Haematol
2013
, vol. 
161
 
3
(pg. 
389
-
401
)
91
Mardiros
 
A
Dos Santos
 
C
McDonald
 
T
, et al. 
T cells expressing CD123-specific chimeric antigen receptors exhibit specific cytolytic effector functions and antitumor effects against human acute myeloid leukemia
Blood
2013
, vol. 
122
 
18
(pg. 
3138
-
3148
)
92
Gill
 
S
Tasian
 
SK
Ruella
 
M
, et al. 
Preclinical targeting of human acute myeloid leukemia and myeloablation using chimeric antigen receptor-modified T cells
Blood
2014
, vol. 
123
 
15
(pg. 
2343
-
2354
)
93
Pizzitola
 
I
Anjos-Afonso
 
F
Rouault-Pierre
 
K
, et al. 
Chimeric antigen receptors against CD33/CD123 antigens efficiently target primary acute myeloid leukemia cells in vivo
Leukemia
2014
, vol. 
28
 
8
(pg. 
1596
-
1605
)
94
O'Hear
 
C
Heiber
 
JF
Schubert
 
I
Fey
 
G
Geiger
 
TL
Anti-CD33 chimeric antigen receptor targeting of acute myeloid leukemia
Haematologica
2015
, vol. 
100
 
3
(pg. 
336
-
344
)
95
Kenderian
 
SS
Ruella
 
M
Shestova
 
O
, et al. 
CD33-specific chimeric antigen receptor T cells exhibit potent preclinical activity against human acute myeloid leukemia
Leukemia
2015
, vol. 
29
 
8
(pg. 
1637
-
1647
)
96
Wang
 
QS
Wang
 
Y
Lv
 
HY
, et al. 
Treatment of CD33-directed chimeric antigen receptor-modified T cells in one patient with relapsed and refractory acute myeloid leukemia
Mol Ther
2015
, vol. 
23
 
1
(pg. 
184
-
191
)
97
Bar
 
M
Chapuis
 
AG
Schmitt
 
TM
, et al. 
Transferred donor-derived virus specific CD8+ T cells that have been transduced to express a WT1-specific T cell receptor can persist and provide anti-leukemic activity in AML patients post-transplant [abstract]
Blood
2014
, vol. 
124
 
21
pg. 
3939
 
98
Greiner
 
J
Schneider
 
V
Schmitt
 
M
, et al. 
Immune responses against the mutated region of cytoplasmatic NPM1 might contribute to the favorable clinical outcome of AML patients with NPM1 mutations (NPM1mut)
Blood
2013
, vol. 
122
 
6
(pg. 
1087
-
1088
)
99
Qazilbash
 
MH
Wieder
 
ED
Thall
 
PF
, et al. 
PR1 peptide vaccine-induced immune response is associated with better event-free survival in patients with myeloid leukemia [abstract]
Blood
2007
, vol. 
110
 
11
pg. 
90a
 
100
Rezvani
 
K
Yong
 
AS
Mielke
 
S
, et al. 
Leukemia-associated antigen-specific T-cell responses following combined PR1 and WT1 peptide vaccination in patients with myeloid malignancies
Blood
2008
, vol. 
111
 
1
(pg. 
236
-
242
)
101
Schmitt
 
M
Schmitt
 
A
Rojewski
 
MT
, et al. 
RHAMM-R3 peptide vaccination in patients with acute myeloid leukemia, myelodysplastic syndrome, and multiple myeloma elicits immunologic and clinical responses
Blood
2008
, vol. 
111
 
3
(pg. 
1357
-
1365
)
102
Keilholz
 
U
Letsch
 
A
Busse
 
A
, et al. 
A clinical and immunologic phase 2 trial of Wilms tumor gene product 1 (WT1) peptide vaccination in patients with AML and MDS
Blood
2009
, vol. 
113
 
26
(pg. 
6541
-
6548
)
103
Maslak
 
PG
Dao
 
T
Krug
 
LM
, et al. 
Vaccination with synthetic analog peptides derived from WT1 oncoprotein induces T-cell responses in patients with complete remission from acute myeloid leukemia
Blood
2010
, vol. 
116
 
2
(pg. 
171
-
179
)
104
Rein
 
LA
Chao
 
NJ
WT1 vaccination in acute myeloid leukemia: new methods of implementing adoptive immunotherapy
Expert Opin Investig Drugs
2014
, vol. 
23
 
3
(pg. 
417
-
426
)
105
Lee
 
JJ
Kook
 
H
Park
 
MS
, et al. 
Immunotherapy using autologous monocyte-derived dendritic cells pulsed with leukemic cell lysates for acute myeloid leukemia relapse after autologous peripheral blood stem cell transplantation
J Clin Apher
2004
, vol. 
19
 
2
(pg. 
66
-
70
)
106
Li
 
L
Giannopoulos
 
K
Reinhardt
 
P
, et al. 
Immunotherapy for patients with acute myeloid leukemia using autologous dendritic cells generated from leukemic blasts
Int J Oncol
2006
, vol. 
28
 
4
(pg. 
855
-
861
)
107
Roddie
 
H
Klammer
 
M
Thomas
 
C
, et al. 
Phase I/II study of vaccination with dendritic-like leukaemia cells for the immunotherapy of acute myeloid leukaemia
Br J Haematol
2006
, vol. 
133
 
2
(pg. 
152
-
157
)
108
Van Tendeloo
 
VF
Van de Velde
 
A
Van Driessche
 
A
, et al. 
Induction of complete and molecular remissions in acute myeloid leukemia by Wilms' tumor 1 antigen-targeted dendritic cell vaccination
Proc Natl Acad Sci U S A
2010
, vol. 
107
 
31
(pg. 
13824
-
13829
)
109
Berneman
 
ZN
Van de Velde
 
A
Anguille
 
S
, et al. 
Prevention of relapse in acute myeloid leukemia by dendritic cell vaccination: report on a phase II study with 29 patients [abstract]
Blood
2013
, vol. 
122
 
21
pg. 
236
 
110
Rosenblatt
 
J
Stone
 
RM
Uhl
 
L
, et al. 
Clinical trial evaluating DC/AML fusion cell vaccination in AML patients [abstract]
Blood
2013
, vol. 
122
 
21
pg. 
3928
 
111
van de Loosdrecht
 
AA
van Wetering
 
S
Santegoeds
 
S
, et al. 
Clinical and immunological results of a phase I/IIa study of allogeneic dendritic cell (DC) vaccination, an “off the shelf” treatment to prevent or delay relapse in elderly patients with acute myeloid leukemia [abstract]
Blood
2013
, vol. 
122
 
21
pg. 
2651
 

Competing Interests

Conflict-of-interest disclosures: S.A.B. reports no conflict of interest. R.B.W. has received research funding from Amgen, Amphivena Therapeutics, and Seattle Genetics and has consulted for Covagen, Amphivena Therapeutics, Pfizer, and AstraZeneca.

Author notes

Off-label drug use: Alemtuzumab, bevacizumab, and GO for the treatment of AML.