Cancer Letters

Cancer Letters

Volume 303, Issue 2, 28 April 2011, Pages 128-139
Cancer Letters

Heterodimeric bispecific antibody-derivatives against CD19 and CD16 induce effective antibody-dependent cellular cytotoxicity against B-lymphoid tumor cells

https://doi.org/10.1016/j.canlet.2011.01.020Get rights and content

Abstract

Bispecific scFv antibody-derivatives (bsscFvs) recruiting natural killer (NK) cells for the lysis of malignant cells have therapeutic potential. However, a bsscFv specific for the B-lymphoid tumor antigen CD19 and the trigger molecule CD16 on NK cells had similar affinities for both antigens (42 and 58 nM, respectively) and was not optimal for cytotoxicity. Therefore, a bispecific tribody (bsTb) was constructed with two binding sites for CD19 and one for CD16. This bsTb contained a CD19-specific Fab fragment carrying a CD16-specific scFv fused to its light chain and a CD19-specific scFv fused to its heavy chain. The bsTb was compared with a bispecific bibody (bsBb) lacking the CD19-specific scFv. The bsTb had 3-fold greater avidity for CD19 than the bsBb (8 and 24 nM, respectively), while both had equal affinity for CD16 (56 nM). Both molecules mediated antibody-dependent cellular cytotoxicity (ADCC) of leukemia-derived SEM cells and primary cells from leukemia patients. The bsTb showed half-maximum effective concentrations (EC50) of 55 pM and promoted equal lysis as the bsBb and the bsscFv at 6- and 12-fold lower concentrations, respectively. Among these three molecules the bsTb showed the most promising in vitro properties which are anticipated to be displayed also in vivo.

Introduction

The cell surface antigen CD19 is a 95 kDa transmembrane glycoprotein belonging to the immunoglobulin superfamily [1], [2]. It is exclusively expressed on B-lymphoid cells in nearly all developmental stages from the early pro-B cell on, but no longer on plasma cells. The protein is abundantly expressed on the majority of B-lineage acute lymphoblastic leukemias (B-ALL) and different types of non-Hodgkin-lymphomas (NHL) [3], [4], and its presence on leukemia repopulating cells (leukemia stem cells) of certain types of B-ALL has recently been reported [5], [6], [7]. The antigen is neither shed from the surface nor lost from the tumor cells upon antibody treatment and is weakly internalized after binding of antibodies and antibody fragments [8], [9], [10]. Thus, CD19 is an attractive target for the treatment of different types of B-cell malignancies, and possibly certain chronic inflammatory diseases and autoimmune disorders with antibody-derived agents [9], [11].

Although unconjugated monoclonal antibodies (mAbs) against CD19 showed encouraging results in pre-clinical tests [11], [12], [13], they displayed only modest therapeutic effects in clinical trials [14]. Therefore, different antibody-based strategies were followed to target CD19 more efficiently, including both immunotoxins and antibodies with improved effector cell-mediated functions [15], [16]. The recruitment of immune effector cells and the induction of antibody-dependent cellular cytotoxicity (ADCC) by engagement of Fc receptors (FcRs) are key events in vitro and in vivo, as demonstrated for the CD20-specific antibody Rituximab and others [17], [18], [19], [20]. Both functions are mediated by the antibody Fc-portion, and therefore, it was anticipated that optimization of the Fc-portion of CD19 antibodies may improve their therapeutic performance. Indeed, the pre-clinical efficacy of full-length CD19 antibodies was increased by strengthening the interaction between their Fc-portion and FcRs either through glyco-engineering or through amino acid substitutions achieved by mutagenesis [10], [21].

A strong improvement of effector functions was also obtained through the development of bispecific antibodies [22]. These molecules combine one binding site for a target antigen on tumor cells with a second site for an activating trigger molecule on effector cells, such as CD3 or CD28 on T-cells and CD16 (FcγRIII) on NK cells and macrophages [22], [23], [24]. In some cases, bispecific antibodies demonstrated more favorable cytotoxic properties in vitro than the parental mAbs and offered the opportunity to selectively address a desirable effector cell population [22]. In addition, the choice of antigen-combining domains from particular mAbs specific for non Fc-binding epitopes on trigger molecules allowed to avoid competition with circulating endogenous immunoglobulin (Ig) G, a known competitor of mAbs [25].

Bispecific recombinant antibody-derivatives targeting CD19 have been designed in a variety of formats [26]. Most of them used single-chain variable fragments (scFvs) as binding domains and avoided the unspecific uptake by non-activating FcRs on various cell types due to the lack of Fc-portions. Among them are tandem bispecific scFvs (bsscFvs), diabodies and tandem diabodies addressing CD16 on NK cells or CD3 on T cells [26], [27], [28]. The most advanced bispecific molecule is a tandem bsscFv with specificities for CD19 and CD3 (Blinatumomab™), which produced promising results in a phase I clinical study with NHL patients and established clinical potency for bispecific antibody-derivatives [29], [30]. However, in some clinical settings, such as the treatment of ALL patients after stem cell transplantation, NK cells may represent a more favorable population of effector cells than T cells [31]. Therefore it is still important to establish potent strategies activating NK-cells for efficient tumor cell lysis.

Although tandem bsscFvs and some of the alternative formats mentioned above offer distinct advantages, they are associated with certain limitations that might affect their therapeutic efficacy. One critical parameter for the potency of bispecific proteins is the strength of binding to the tumor cells, which is determined by the intrinsic affinity of the individual combining site and the overall functional avidity of the molecules. In one approach, an important improvement was achieved through in vitro affinity maturation of the tumor cell-directed scFv-component, which resulted in an increased cytotoxic potential in vitro [32]. In another approach, the ADCC potential of a bispecific antibody-derivative has been enhanced by including a second binding site for the tumor antigen: a heterodimeric bispecific minibody containing two scFvs for the tumor antigen Her2/neu and one scFv for CD16 was more effective in mediating ADCC than a control protein with monovalent binding for both antigens [33].

Fab fragments, assembling a binding site from an antibody light chain (L) and the Fd-fragment of the heavy chain (H), have been used as hetero-dimerization scaffolds to engineer recombinant Fab–scFv fusion proteins [34], [35], [36]. Such molecules were constructed by genetic fusion of an scFv with a different specificity to the C-terminus of either the L- or the Fd-chain of a Fab fragment. Co-expression of the extended chain with its corresponding heterologous counterpart lead to an efficient production and efficient hetero-dimerization in eukaryotic cells mediated by the complementarity of the contact zones of the L- and Fd-components and a disulfide bridge formed between them [34], [35]. Through this assembly, the binding-site of the Fab fragment was reconstituted and bispecific proteins were formed, so-called bibodies (Fig. 1A). Both chains of Fab fragments have further been extended by fusion with scFvs to build so-called tribodies (Fig. 1A). Tribodies and bibodies have intermediate molecular masses of approximately 110 and 75 kDa, respectively, and were initially designed to achieve longer plasma retention times in vivo than the smaller bsscFv fragments [34]. BsscFvs have molecular masses of only 55–65 kDa, close to the renal exclusion limit, and molecules with masses below this limit are rapidly cleared from the blood-stream by renal filtration [37]. Finally, the use of Fab fragments as binding domains in bispecific proteins is attractive, because Fab fragments in some cases had a higher affinity and a greater stability than the corresponding scFvs derived from the same hybridoma antibody [38], [39].

Our group has previously generated a disulfide-stabilized (ds) tandem bsscFv with monovalent binding for the tumor antigen CD19 and the trigger molecule CD16, ds[19 × 16] [27]. This molecule displayed strong cytotoxicity for CD19-positive human leukemic cells in ADCC reactions with mononuclear cells (MNCs) in vitro. However, monovalent targeting of tumor cells and almost similar affinities for the target antigen and the trigger molecule were considered limiting. To overcome this limitation we have designed a CD19 × CD16 tribody with one Fab and one scFv binding site for CD19 and an scFv binding site for CD16 and, for comparison, a CD19 × CD16 bibody lacking the CD19-specific scFv (Fig. 1). The tribody had the predicted increased overall avidity for CD19 over the bibody and the bsscFv, accompanied by a corresponding gain in functional ADCC activity. These results provide strong motivation to also test the in vivo properties of this molecule in future experiments.

Section snippets

Cell lines and hybridomas

Chinese hamster ovary (CHO) cells stably expressing human CD16A were from Dr. J. van de Winkel (University Medical Centre, Utrecht, The Netherlands). The 4G7 hybridoma (CD19; IgG1) [40] was from Dr. R. Levy (Stanford University, Palo Alto, CA, USA) and the hybridoma 3G8 (FcγRIII, CD16; IgG1) [41] from the American Type Culture Collection (ATCC, Manassas, VA, USA). CHO cells, the hybridomas, and the leukemia-derived cell lines SEM [42] and CEM (ATCC) were cultured in Roswell Park Memorial

Construction of Fab–scFv fusion proteins

The Fab–scFv fusion proteins were built using the CD19-directed Fab fragment from the chimeric CD19 antibody and the ds CD19 and CD16 scFvs [21], [27]. The latter were used because they were more stable than the respective scFvs containing the wild-type sequences [27]. For the simultaneous expression of both polypeptide chains building the entire molecules, double gene expression vectors were constructed (Fig. 1B). Briefly, the coding sequence for the ds CD16 scFv was fused to the 3′ end of the

Discussion

CD19 is a tumor-antigen with highly favorable properties for the design of antibody-derived therapeutics. Its attractive features have been recognized for more than 20 years, and many investigators in academic institutions and the pharmaceutical industry have attempted to target this antigen [8], [9]. However, until recently, these efforts have not produced therapeutic molecules of comparable efficacy as the CD20 antibody Rituximab (Mabthera™) or the Her2/neu antibody Trastuzumab (Herceptin™).

Conflict of interest

The authors declare no conflicts of interests.

Acknowledgements

The authors thank Dr. R. Levy for the 4G7 hybridoma, Dr. J.G. van de Winkel for the CD16 transfected CHO cells, D. Saul and B. Bock for excellent technical assistance. Th. Lange is gratefully acknowledged for administrative assistance. This work was supported by grants from Deutsche Krebshilfe e.V. (No. 108242), the association “Kaminkehrer helfen krebskranken Kindern e.V.”, the association of Friends of the University of Erlangen Children’s Hospital (bequest of Dr. Wilhelmine Fey), the

References (52)

  • D. Elsasser et al.

    HLA class II as potential target antigen on malignant B cells for therapy with bispecific antibodies in combination with granulocyte colony-stimulating factor

    Blood

    (1996)
  • S.M. Kipriyanov et al.

    Bispecific tandem diabody for tumor therapy with improved antigen binding and pharmacokinetics

    J. Mol. Biol.

    (1999)
  • T.F. Tedder et al.

    Isolation of cDNAs encoding the CD19 antigen of human and mouse B lymphocytes. A new member of the immunoglobulin superfamily

    J. Immunol.

    (1989)
  • L.M. Nadler et al.

    B4, a human B lymphocyte-associated antigen expressed on normal, mitogen-activated, and malignant B lymphocytes

    J. Immunol.

    (1983)
  • L. Ginaldi et al.

    Levels of expression of CD19 and CD20 in chronic B cell leukaemias

    J. Clin. Pathol.

    (1998)
  • A. Castor et al.

    Distinct patterns of hematopoietic stem cell involvement in acute lymphoblastic leukemia

    Nat. Med.

    (2005)
  • Y. Kong et al.

    CD34+CD38+CD19+ as well as CD34+CD38−CD19+ cells are leukemia-initiating cells with self-renewal capacity in human B-precursor ALL

    Leukemia

    (2008)
  • R.H. Scheuermann et al.

    CD19 antigen in leukemia and lymphoma diagnosis and immunotherapy

    Leuk. Lymphoma

    (1995)
  • H.M. Horton et al.

    Potent in vitro and in vivo activity of an Fc-engineered anti-CD19 monoclonal antibody against lymphoma and leukemia

    Cancer Res.

    (2008)
  • N. Yazawa et al.

    Immunotherapy using unconjugated CD19 monoclonal antibodies in animal models for B lymphocyte malignancies and autoimmune disease

    Proc. Natl. Acad. Sci. USA

    (2005)
  • G.A. Pietersz et al.

    In vitro and in vivo antitumor activity of a chimeric anti-CD19 antibody

    Cancer Immunol. Immunother.

    (1995)
  • A. Hekman et al.

    Initial experience with treatment of human B cell lymphoma with anti-CD19 monoclonal antibody

    Cancer Immunol. Immunother.

    (1991)
  • M. Schwemmlein et al.

    A CD19-specific single-chain immunotoxin mediates potent apoptosis of B-lineage leukemic cells

    Leukemia

    (2007)
  • D.J. Flavell et al.

    The anti-CD20 antibody rituximab augments the immunospecific therapeutic effectiveness of an anti-CD19 immunotoxin directed against human B-cell lymphoma

    Br. J. Haematol.

    (2006)
  • R.A. Clynes et al.

    Inhibitory Fc receptors modulate in vivo cytoxicity against tumor targets

    Nat. Med.

    (2000)
  • W.K. Weng et al.

    Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma

    J. Clin. Oncol.

    (2003)
  • Cited by (41)

    • Natural killer cells as a double-edged sword in cancer immunotherapy: A comprehensive review from cytokine therapy to adoptive cell immunotherapy

      2020, Pharmacological Research
      Citation Excerpt :

      By linking these anti-CD16 fragments of variable region (Fv) domains with other molecules, different types of tumor cells can be targeted. For example, linking the Fv domain of CD16 to anti-CD19 antibody and anti-HLA II antibodies targets CD19 and B-cell neoplasia with highly expressed HLA II [214,215]. When Fv domain of CD16 is linked to anti-CD30 antibody, it targets CD30 high expression in Hodgkin's lymphoma, and linked to human epidermal growth factor receptor-2 (HER2) antibody targets Her2+breast cancer [216,217].

    • Therapeutic Antibodies and Immunologic Conjugates

      2019, Abeloff’s Clinical Oncology
    • Natural killer cells unleashed: Checkpoint receptor blockade and BiKE/TriKE utilization in NK-mediated anti-tumor immunotherapy

      2017, Seminars in Immunology
      Citation Excerpt :

      The final step is protein purification, achieved through size exclusion chromatography or column isolation with an affinity tag. Currently, bi-specific antibody constructs have been generated to engage CD16 in conjunction with: CD20/CD19 on B cell Non-Hodgkin’s lymphomas [163–171], CD19/CD33 on mixed lineage leukemia [172], CD33 or CD33/CD123 on acute myelogenous leukemia (AML) [173–175], HLA Class II on lymphoma [176], CD30 on Hodgkin’s lymphoma [177–186], EGF-R on EGF-R+ tumors [187,188], HER2/neu on metastatic breast cancer and other HER2 expressing tumors [156,189,190] and MOV19 on ovarian cancer [191]. Our group has generated and tested BiKEs and TriKEs that engage CD16 along with: CD19/CD22 on B cell Non-Hodgkin’s lymphomas [192], CD33 on AML [193] and MDS/MDSCs [194], EpCAM on prostate, breast, colon, head, and neck carcinomas [195] and CD133 on cancer stem cells or a combination of EpCAM/CD133 for a broad spectrum molecule [196,197].

    • Exploitation of natural killer cells for the treatment of acute leukemia

      2016, Blood
      Citation Excerpt :

      Because the CD19 antigen is widely expressed in BCP-ALL, an Fc-optimized anti-CD19 antibody was constructed and used posttransplant in selected high-risk patients with pediatric BCP-ALL.94 Other approaches to activate NK cells against BCP-ALL or AML in either the autologous or allogeneic setting, independent of their alloreactive status, might be the future clinical use of the CD16xCD1995 or CD16xCD33 bispecific killer engager96 or the trispecific killer engager CD16xCD19xCD22.97 NK cells and established NK cell lines such as NK-92 can also be redirected toward target antigens expressed on leukemic blasts by chimeric antigen receptors (CARs).98

    • Adoptive Immunotherapies After Allogeneic Hematopoietic Stem Cell Transplantation in Patients With Hematologic Malignancies

      2015, Transfusion Medicine Reviews
      Citation Excerpt :

      Genetic reprogramming of Tregs [71,72] or engineered T-cell receptors that redirect Tregs specificity [73] could improve Tregs potency. Creating either bispecific or trispecific killer engagers to redirect NK or CIK cytotoxicity toward tumor cells [74-76] or redirecting NK, CIK, CTLs, and γδ T cells toward tumor targets through a chimeric antigen receptor can effectively increase these cell reactivities and specificities of recognitions [77-81]. Third party–derived T-cell infusion is being currently investigated as potential solution to overcome the availability limitation; other solutions (eg, rapid T-cell generation, simplify and shorten CTL production) should also continue to be tested to further broaden the application of this approach [82].

    • γδ T cell activation by bispecific antibodies

      2015, Cellular Immunology
      Citation Excerpt :

      In an attempt to improve such small bispecific antibody fragments, formats with three binding sites have been developed to increase avidity to the tumor antigen, or to target two different antigens simultaneously [6–8]. For example, so-called tribodies are heterodimeric proteins consisting of a fragment antigen binding (Fab) and two scFv and exhibit enhanced cytotoxic potency relative to bsscFv or IgG1 antibodies [9–11]. Bispecific antibodies may also be applied as a support for cellular therapies such as stem cell transplantation or adoptive T cell transfer.

    View all citing articles on Scopus
    View full text