4,215
Views
27
CrossRef citations to date
0
Altmetric
Editorial

The role of CD33 as therapeutic target in acute myeloid leukemia

, MD PhD MS (Epi)

Abstract

CD33 is a myeloid differentiation antigen that is displayed on acute myeloid leukemia (AML) blasts in most patients and, possibly, leukemic stem cells in some, and has thus served as target for antibody-based therapies for many years. Validation for this approach comes from the antibody–drug conjugate, gemtuzumab ozogamicin, which improves survival of some patients with AML when added to induction chemotherapy. Still, CD33 is a challenging target because of its low expression and slow internalization; these characteristics limit antibody-dependent cell-mediated cytotoxicity and intracellular drug accumulation and, consequently, the activity of unlabeled and toxin-carrying antibodies. Very promising preclinical data are now available from an improved antibody–drug conjugate and CD33-targeted strategies that redirect immune effector cells to eradicate the leukemia, most notably bispecific antibodies and chimeric antigen receptor T-cell immunotherapy. In parallel to their clinical testing, efforts will be needed to identify the patients that most likely benefit from such agents and the disease stage in which they are most efficacious. With enhanced activity of CD33-directed therapies, toxic effects on normal hematopoiesis will increase and require excellent supportive care measures, or even rescue with donor cells, to minimize morbidity and mortality from expected cytopenias and to optimize treatment outcomes with these therapeutics.

1. Introduction

CD33 is a myeloid differentiation antigen that can be displayed on some normal B-cells and activated T- and natural killer cells but is not expressed on pluripotent hematopoietic stem cells or outside the hematopoietic system Citation[1]. It is found on at least a subset of blasts in nearly all acute myeloid leukemias (AMLs). With an average of ∼ 104 molecules/leukemic cell, CD33 is not highly abundant but levels vary considerably across individual patients. Besides its broad expression on AML blasts, a main impetus to pursue CD33 as therapeutic target emanated from the notion that some AMLs may predominantly or entirely involve committed CD33+ myeloid precursors, suggesting that this antigen could serve to eradicate malignant stem cells in such leukemias, with acute promyelocytic leukemia (APL) being one possible example Citation[1].

Although crosslinking of CD33 can induce AML cell apoptosis in vitro, the results with unconjugated CD33 antibodies have so far been disappointing. For instance, the best-studied molecule (lintuzumab) has very modest activity as single agent even at supra-saturating doses and failed to improve survival when added to intensive or non-intensive chemotherapy in two randomized trials Citation[1]. Because of the endocytic property of CD33, it was suspected early that adding a toxic payload might improve the efficacy of CD33 antibodies. Indeed, gemtuzumab ozogamicin (GO), a CD33 immunoconjugate carrying a calicheamicin-γ1 derivative, induces remissions in a subset of AML patients Citation[1]. More importantly, several randomized studies have demonstrated that addition of GO to conventional chemotherapy reduces the risk of relapse and improves survival of previously untreated patients with favorable-risk and, to a lesser degree, intermediate-risk AML Citation[2], justifying its continued availability for the treatment of these patients and validating CD33 as therapeutic target.

2. Emerging strategies to improve CD33-directed therapy

Regardless, CD33 is a challenging target even for armed antibodies: the development of a CD33 antibody conjugated to a thiol-containing maytansinoid derivative (AVE9633) was terminated because of modest single agent activity Citation[1], and similarly modest activity was observed with an immunoconjugate carrying recombinant gelonin (HuM-195/rGel) Citation[3]. Two potential reasons for the poor clinical activity of CD33 antibodies are the oftentimes relatively low cell surface density of CD33 and the slow internalization kinetic of CD33/antibody complexes, which limit antibody-dependent cell-mediated cytotoxicity (ADCC) and intracellular accumulation of antibody-delivered payloads. The latter may then be extruded from the AML cell before exerting any cytotoxic effects. Such limitations may explain why GO is ineffective in many patients even though a highly potent toxin is delivered Citation[1]. Nonetheless, the survival improvement seen in some patients with GO, together with the fact that GO is no longer available in most countries, provides a strong impetus to develop novel CD33-directed drugs that might be more efficacious than this first-generation antibody–drug conjugate.

2.1 Unconjugated antibodies

Efforts are ongoing to generate engineered CD33 antibodies with better ADCC than lintuzumab, and a fully human antibody with these properties (mAb 33.1) is currently tested clinically (NCT01690624).

2.2 Antibody–drug conjugates

Besides cellular extrusion of the calicheamicin-1 derivative, another factor limiting GO's activity was heterogeneous and incomplete drug loading Citation[1]. Obvious technological advancements would therefore include improvements of conjugation and linker technology as well as use of highly potent toxins that are poor substrates for drug transporters. One such candidate is SGN-CD33A, a humanized CD33 antibody with engineered cysteines carrying a synthetic DNA cross-linking pyrrolobenzodiazepine dimer Citation[4]. After preclinical studies have demonstrated that SGN-CD33A is more potent than GO against AML cell lines and primary AML cells and maintains activity in models of multidrug-resistant disease, this drug has recently entered clinical testing (NCT01902329).

2.3 Radiolabeled antibodies

Because of the radiosensitivity of AML, radionuclides are an attractive means to augment the efficacy of therapeutic antibodies. However, the low CD33 abundance limits how much radiation can be delivered via antibodies, and immunoconjugate metabolization shortens the intracellular retention of radio-isotopes. Nevertheless, studies with 131I demonstrated that radiolabeled CD33 antibodies could be used as single agent or part of pretransplant conditioning regimens Citation[5]. Later studies with α-emitters similarly found these to be safe and to have anti-leukemia activity Citation[5], and trials with an 225Ac-labeled antibody are ongoing (NCT00672165, NCT01756677).

2.4 Bispecific antibodies

A long-pursued strategy to improve antitumor antibodies encompasses bispecific constructs that harness the immune system in the elimination of cancer cells. Many types have been explored over the years, but their success was limited by suboptimal effector cell recruitment and challenges with antibody production Citation[6]. As evidenced by recent data on T-cell-directed constructs, such shortcomings may not pertain to small single-chain constructs that only contain the minimal binding domains of the two antibodies. By binding CD3, they bring polyclonal T-cells in close proximity of targeted tumor cells and force formation of a synapse that triggers lymphocyte activation and serial destruction of attached tumor cells through perforin/granzyme-mediated apoptosis at low effector-to-target (E:T) ratio. Therapeutic utility of these molecules for acute leukemias is suggested by emerging results from small studies with the CD19/CD3 bispecific T-cell engager (BiTE) antibody, blinatumomab, showing a 70 – 80% response rate and high relapse-free survival rates among adults with CD19+ acute lymphoblastic leukemia (ALL) that persisted or relapsed after chemotherapy Citation[7].

Developing small bispecific antibodies targeting CD33 is a logical consequence of the experience with GO and the encouraging experience with blinatumomab. We and others have recently demonstrated that a CD33-directed candidate built on the BiTE platform (AMG 330) causes highly potent cytolysis of CD33+ AML cell lines and primary AML cells in the presence of healthy donor or patient T-cells at low E:T ratio in vitro and in immunodeficient mice Citation[8-10]. Target antigen density, antibody dose and E:T ratio are critical determinants for the drug's activity, whereas cytolysis is not affected by drug transporter activity Citation[10]. Other CD33/CD3-targeting single-chain antibodies confirm the high anti-AML activity of these constructs Citation[11]. None of these bispecific antibodies have been brought to the clinic yet. Despite their high preclinical potency, however, further improvements seem possible. For example, a modular targeting system that not only brings CD33+ AML cells together with CD3+ T-cells but also provides a co-stimulatory signal yielded more efficient cytolysis of AML cells expressing low levels of CD33 Citation[12]. Moreover, several groups explore whether targeting a second tumor antigen simultaneously with CD33, or targeting additional/alternative immune effector cell antigens, could provide superior anti-AML efficacy.

2.5 Chimeric antigen receptor T-cell immunotherapy

An alternative strategy to redirect the immune system in the eradication of leukemia cells is adoptive immunotherapy using T-cells genetically engineered to express chimeric antigen receptors (CARs). These hybrid single-chain receptor constructs contain an extracellular tumor antigen-recognizing domain linked to an intracellular component composed of the CD3 zeta chain as primary domain with or without additional co-stimulatory endodomains that, together, activate immune effector cells upon binding to the tumor antigen Citation[13]. Complete responses were observed in small series of ALL patients treated with CAR T-cells recognizing CD19 Citation[13]. Various types of T-cells modified to express CD33-directed CARs have now been generated and shown to very efficiently reduce the burden of AML cells in preclinical models Citation[14-16]. A first clinical trial testing CD33-directed CAR T-cells is currently ongoing (NCT01864902).

3. Conclusion

Considering that survival improvements were observed with a CD33 immunoconjugate that had significant limitations with regard to drug labeling and susceptibility to drug extrusion, the true potential of CD33-targeted therapies may not have been elucidated yet. The emerging preclinical data from several novel CD33 antibody-based therapeutics indeed suggest that they might be active against a much broader subset of leukemias than GO. It is thus conceivable that these agents could benefit a high proportion of AML patients. Nevertheless, to avoid the mistake made with GO where little attention was paid to the heterogeneity of AML, efforts should be made early to identify the patient subsets that most likely respond to such therapies to optimize the clinical outcome of these targeted treatments.

With the almost universal expression of CD33 on AML blasts of individual patients, CD33 antibody-based therapeutics may be useful for tumor debulking. On the other hand, it currently remains debated whether this antigen is displayed on AML stem and progenitor cells, and CD33-directed therapies may eradicate such cells only in a subset of leukemias (e.g., APL). Resolving this controversy will be instrumental for the understanding in which disease stages CD33-targeted agents may be beneficial.

As CD33 is widely expressed on hematopoietic progenitor cells, effects on these cells are likely substantial if the CD33-targeted drug is highly efficacious even at low CD33 expression levels or in the presence of drug transporter activity. Patients may thus experience very prolonged cytopenias and require excellent supportive care or even rescue with donor hematopoietic cells to minimize morbidity and mortality. Anticipated cytopenias may prove a particular hurdle for CD33-directed CAR T-cells as forming myeloid cells could be continuously destroyed, which could lead to permanent cytopenias without the use of suicide genes or myeloablative conditioning to limit their long-term persistence in patients.

Undoubtedly, with the various new CD33 antibody-based therapeutics on the horizon, we are entering a new treatment era. Many questions remain open, but these agents offer exciting new prospects for a disease for which the outcomes remain unsatisfactory for many patients.

Declaration of interest

The author was supported by research funding from Amgen, Inc., and Seattle Genetics, Inc., and has served as a consultant for Seattle Genetics, Inc. The author has no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Bibliography

  • Cowan AJ, Laszlo GS, Estey EH, et al. Antibody-based therapy of acute myeloid leukemia with gemtuzumab ozogamicin. Front Biosci (Landmark Ed) 2013;18(4):1311-34
  • Hills RK, Petersdorf S, Estey EH, et al. The addition of gemtuzumab ozogamicin (GO) to induction chemotherapy reduces relapse and improves survival in patients without adverse risk karyotype: results of an individual patient meta-analysis of the five randomised trials [abstract]. Blood 2013;122(21):356
  • 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;98(2):217-21
  • 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;122(8):1455-63
  • Walter RB, Press OW, Bernstein ID. Antibody-based therapeutics targeting CD33, CD45, and CD66. In: Andreeff M, editor. Targeted therapy of acute myeloid leukemia. 1st edition. Springer, New York, NJ; 2014; In press
  • Carter P. Improving the efficacy of antibody-based cancer therapies. Nat Rev Cancer 2001;1(2):118-29
  • Walter RB. Biting back: BiTE antibodies as a promising therapy for acute myeloid leukemia. Expert Rev Hematol 2014; In press
  • 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;27(5):1107-15
  • 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;123(3):356-65
  • 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;123(4):554-61
  • 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;27(4):964-7
  • Arndt C, Feldmann A, von Bonin M, et al. Costimulation improves the killing capability of T cells redirected to tumor cells expressing low levels of CD33: description of a novel modular targeting system. Leukemia 2014;28(1):59-69
  • Fry TJ, Mackall CL. T-cell adoptive immunotherapy for acute lymphoblastic leukemia. Hematology Am Soc Hematol Educ Program 2013;2013:348-53
  • Marin V, Pizzitola I, Agostoni V, et al. Cytokine-induced killer cells for cell therapy of acute myeloid leukemia: improvement of their immune activity by expression of CD33-specific chimeric receptors. Haematologica 2010;95(12):2144-52
  • Dutour A, Marin V, Pizzitola I, et al. In vitro and in vivo antitumor effect of anti-CD33 chimeric receptor-expressing EBV-CTL against CD33+ acute myeloid leukemia. Adv Hematol 2012;2012:683065
  • 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; In press

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.