Cutaneous T-cell lymphoma (CTCL) is a heterogeneous group of neoplastic disorders characterized by clonally derived and skin-homing malignant T cells that express high level of chemokine receptor CCR4, which is associated with their skin-homing capacity. CCR4 is also highly expressed on T-regulatory cells (Tregs) that can migrate to several different types of chemotactic ligand CCL17- and CCL22-secreting tumors to facilitate tumor cell evasion from immune surveillance. Thus, its high-level expression on CTCL cells and Tregs makes CCR4 a potential ideal target for antibody-based immunotherapy for CTCL and other types of solid tumors. Here, we conducted humanization and affinity optimization of a murine anti-CCR4 monoclonal antibody (mAb), mAb1567, that recognizes both the N-terminal and extracellular domains of CCR4 with high affinity and inhibits chemotaxis of CCR4+ CTCL cells. In a mouse CTCL tumor model, mAb1567 exhibited a potent antitumor effect and in vitro mechanistic studies showed that both complement-dependent cytotoxicity (CDC) and neutrophil-mediated antibody-dependent cellular cytotoxicity (ADCC) likely mediated this effect. mAb1567 also exerts human NK cell–mediated ADCC activity in vitro. Moreover, mAb1567 also effectively inhibits chemotaxis of CD4+CD25high Tregs via CCL22 and abrogates Treg suppression activity in vitro. An affinity-optimized variant of humanized mAb1567, mAb2-3, was selected for further preclinical development based on its higher binding affinity and more potent ADCC and CDC activities. Taken together, this high-affinity humanized mAb2-3 with potent antitumor effect and a broad range of mechanisms of action may provide a novel immunotherapy for CTCL and other solid tumors. Mol Cancer Ther; 11(11); 2451–61. ©2012 AACR.

This article is featured in Highlights of This Issue, p. 2309

Cutaneous T-cell lymphoma (CTCL) is the second most common extranodal non–Hodgkin lymphoma in adults, characterized by primary accumulation of clonally derived malignant CD4+ T cells in the skin. There are 13 clinically and histologically distinct types of CTCL, and the majority falls into 3 classes: mycosis fungoides, the most common type of CTCL, accounts for almost 50% of all primary cutaneous lymphomas; primary cutaneous CD30+ lymphoproliferative disorders—more specifically primary cutaneous anaplastic large cell lymphoma (PC-ALCL)—the second most common CTCL, account for circa 30%; and Sézary syndrome, the most aggressive type of CTCL, makes up circa 5% (1, 2). Other forms of CTCL include adult T-cell leukemia/lymphoma (2%–5%) and peripheral T-cell lymphoma (PTCL; 2%). Although CTCL usually has indolent clinical behavior, in advanced stages, it can progress into an aggressive phenotype with poor prognosis and survival (1, 3, 4) with severe immunodeficiency characteristically developing during disease progression (5). Current hypotheses maintain that the malignant T cells drive this evolving immunodeficiency through the constitutive secretion of immunosuppressive cytokines, dysregulated expression of immunoregulatory proteins by the malignant T cells, and loss of T-cell receptor repertoire complexity due to competitive replacement of normal T cells by the clonally expanded malignant T cells. In addition, a subset of the malignant T cells may act as T-regulatory cells (Tregs) to suppress antitumor responses in some patients with CTCL (6–8). Current therapies for CTCLs do not prevent new lesions from emerging (9) and durable long-term remissions are rare (10). Therefore, disease-specific and more effective therapeutics that can decrease toxicity profiles and induce durable responses will greatly benefit patients with CTCL.

Normal skin-homing CD4+ T cells express cutaneous lymphocyte–associated antigen and CC chemokine receptors (CCR) CCR4, CCR6, CCR7, and CCR10 (11–14). Among these, only CCR4 is universally expressed at high levels on the malignant skin-homing T cells, and its surface expression is closely associated with the enhanced skin-homing characteristics of CTCL cells and unfavorable disease outcome (15, 16). Tregs also selectively express high levels of CCR4 compared with other T-cell subsets. The 2 specific ligands for CCR4, chemokines CCL17, and CCL22, produced by tumor cells and cells of the tumor microenvironment, attract CCR4+ Tregs to the tumor, where they suppress host immune responses against tumor and create a favorable environment for cancer cells to grow (17, 18). Thus, the high-level expression of CCR4 on CTCL cells and its preferential expression on Tregs (19) make CCR4 not only a potential ideal therapeutic target for CTCLs, but also for other type of cancers for which CCR4+ Tregs are involved in their immune evasion.

In this study, we characterized and humanized a mouse anti-CCR4 monoclonal antibody, mAb1567, that recognizes both the N-terminal (NT) and the extracellular domains of CCR4. The antibody exhibited potent antitumor effects in a CTCL mouse model and its mechanism(s) of action, including complement-dependent cytotoxicity (CDC), neutrophil,- and NK-mediated antibody-dependent cellular cytotoxicity (ADCC), were elucidated by a number of in vitro studies. In addition, mAb1567 could not only inhibit Tregs migration toward CCR4 ligand, CCL22, but also abrogate suppression by Tregs in the T-cell proliferation assay. Finally, after the affinity maturation of humanized mAb1567, the resulting mAb2-3 was further improved in affinity and showed stronger CDC and ADCC activities against CCR4+ tumor cells.

Cells

Mac-1 cell line was isolated from a patient with PC-ALCL, one of CTCLs (20), obtained from Dr. Thomas S. Kupper (Brigham and Women's Hospital, Harvard Medical School, Boston, MA) and cultured in 10% FBS RPMI-1640. Luciferase-expressed Mac-1 cells were stably transduced with a luciferase reporter retrovirus and authenticated by detecting luminescence. 293F cell line was purchased from Invitrogen. 293T (CRL-11268) and Cf2Th (CRL-1430) cell lines were purchased from American Type Culture Collection and incubated in 10% FBS Dulbecco's Modified Eagle's Medium. No additional authentication of these cell lines was conducted by the authors.

Antibodies and flow cytometry analysis

mAb1567 was purchased from R&D systems and the other 1567 variant antibodies were produced as described previously (21). Briefly, scFv-Fcs were constructed by cloning the single-chain variable region (scFv) into pcDNA3.1-Hinge vector in frame with human IgG1 Fc region. IgG1 was generated by cloning heavy-chain variable region (VH) and light-chain variable region (VL) into TCAE5.3 vector (22). Antibodies were produced in 293T or 293F cells and purified by proteinA-Sepharose (Amersham) affinity chromatography. For staining, Mac-1 was stained with anti-CCR4 antibodies, detected by fluorescein isothiocynate-conjugated goat–anti-human IgG or anti-mouse IgG antibodies (Sigma), and analyzed with FACSCalibur and CellQuest software.

Chemotaxis

Mac-1 cells (1 × 106 cells/well) were placed in Transwell migration wells (Corning) with or without mAb1567 for 3 hours at 37°C. Migrated cells harvested from the bottom chamber containing 50 ng/mL human CCL17 or CCL22 (R&D Systems) were enumerated by fluorescence-activated cell sorting (FACS) analysis. Human CD4+ T cells were isolated by CD4+ T-Cell Isolation Kit (Miltenyi Biotech) and placed in Transwell migration assays with c1567IgG. Migrated cells (CD4+CD25high) were enumerated as above in response to 100 ng/mL CCL22. Percentages of migrated cells were calculated by dividing the number of transmigrated Mac-1 or CD4+CD25high cells by the number of input cells.

Antibody-dependent cell cytotoxicity assay

For lactate dehydrogenase (LDH) release assay, SCID/Beige mouse neutrophils, human peripheral blood mononucleated cells (PBMC), or human NK cells and neutrophils were used as effector cells and Mac-1, Cf2Th-CCR4, or Cf2Th were used as target cells. Target cells (1 × 104 cells/well) were plated into 96-well plates and antibodies were added. After 1 hour, effector cells were added at an appropriate effector/target (E/T) ratio and incubated (PBMCs, NK, and neutrophils for 4, 16, and 6 hours, respectively). The supernatants were recovered by centrifugation at 300 × g and measured using nonradioactive cytotoxicity assay kits (Promega) at 490 nm. For 51Cr release assay, 1 × 106 Mac-1 cells were labeled with 100 μCi (3.7 MBq) of Na51Cr (Amersham International), washed, and used as targets. 51Cr-labeled target cells (5,000 cells/well) were seeded into 96-well plates and the release of 51Cr into supernatants was determined. The cytotoxicity was calculated by the following formula:

where E is released LDH from E/T culture with antibody; SE, spontaneous released LDH from effectors; ST, spontaneous released LDH from targets; and M, the maximum released LDH from lysed targets.

Complement-dependent cytotoxicity assay

The 5 × 104 Mac-1 cells per well, resuspended with medium containing rabbit serum (Cedarlane Laboratories) or mouse serum (IMS-COMPL; Innovative Research), were plated in 96-well plates with anti-CCR4 antibodies. After 4-hour incubation, the supernatants were recovered and detected by LDH release assay, calculating as: |$\% \,\,{\rm Cytotoxicity} = {\rm 100} \times \left({{\rm E} - S_{\rm E} - S_{\rm T}} \right){\rm /}\left({{\rm M} - S_{\rm T}} \right)$|⁠.

Regulatory T-cell suppression assay

CD4+CD25high and CD4+CD25 T cells were sorted by Beckman CoulterMoFlo sorter using mouse-anti-human CD4-PE-Cy5 (RPA-T4) and antihuman CD25-PE (M-A251) antibodies (BD Pharmigen). CD4+CD25 Teffs (2,500) were cultured with or without CD4+CD25high Tregs (1,250) in 96-well plates with 25,000 irradiated (3,000 rad) CD3-depleted PBMCs. Cells were stimulated with 0.05 μg/mL plate-bound anti-CD3 (UCHT1) and 1 μg/mL soluble anti-CD28 (CD28.2) antibodies (BD Pharmigen). Anti-CCR4 antibodies were included in the appropriate cultures. The cultures were pulsed on day 5 after culture initiation with 1 μCi 3H-labeled thymidine/well (Perkin Elmer). Proliferation of cultures was measured in terms of incorporation of 3H-thymidine by reading counts in a scintillation counter (Perkin Elmer).

CCR4+ CTCL tumor-bearing mouse model

2 × 106 luciferase-Mac-1 or 1 × 107 Mac-1 cells were injected subcutaneously into the dorsolateral flank in 6-week SCID/Beige mice (Charles River). After 24 hours of injection, mice were randomly assigned into different groups and treated with 3 mg/kg of mAb1567 and mouse IgG2b (twice a week for 3 weeks) or 5 mg/kg of control-scFv-Fc, c1567-scFv-Fc, and h1567-scFv-Fc (twice a week for 4 weeks) by intraperitoneal injection. Body weight and tumor size were measured using digital caliper and Xenogen imaging. Tumor volumes were calculated as length × (width)2 × 0.52. Animal care was carried out in accordance with the guidelines of Animal Care and Use Committee of Dana-Farber Cancer Institute (Boston, MA).

Statistical analyses

Data were analyzed using 2-sided unpaired Student t test. *, **, and *** indicate P < 0.05, 0.01, and 0.001, respectively. All values and bars are represented as mean ± SD.

Characterization of a mouse anti-CCR4 mAb and mAb1567, in vitro and in vivo

CCR4 has 4 regions exposed at the cell surface: the NT (∼30–50 aa) and 3 extracellular domains loops (ECL, each of ∼10–30 aa), which are important for ligand binding, intracellular signaling, and other biologic functions. In this study, 2 commercially available murine anti-CCR4 mAbs, mAb1567 (R&D Systems), and 1G1 (BD Pharmingen), both generated by immunizing mouse with full-length human CCR4 (hCCR4)-expressing cells (13, 23), were initially selected for evaluation. Both mAb1567 and 1G1 showed specific binding activity in FACS analysis to hCCR4-expressing Cf2Th-CCR4 cells but not to the parental Cf2Th cells. In comparison, mAb1567 had relatively higher affinity than 1G1 under the same antibody concentrations tested (data not shown). Therefore, we selected only mAb1567 for further characterization.

Binding of mAb1567 was further tested using the CCR4+ Mac-1 cells by FACS analysis and the half maximal effective binding concentration (EC50) is about 0.45 nmol/L (Fig. 1A). Chemotaxis inhibition assay showed that mAb1567 effectively inhibited chemotaxis of Mac-1 cells in a dose-dependent manner toward both CCR4 ligands, CCL17 and CCL22 (Fig. 1B). We next examined the epitope recognized by mAb1567, in particular, whether it recognizes solely the NT or a nonlinear conformational-dependent epitope comprising of both NT and ECL, by using hCCR4 and hCCR8 NT swapping chimeras that either contained CCR8-NT/CCR4-ECLs (Chi#1) or CCR4-NT/CCR8-ECLs (Chi#2; ref. 24). As shown in Supplementary Fig. S1A, all constructs encoding wild-type or chimeras CCR4 and CCR8 expressed similar levels on the cell surface as validated by antibody staining against the hemagglutinin tag. mAb1567 specifically recognized cell surface full-length hCCR4 but not hCCR8. It bound to Chi#1 and Chi#2 in a similar level as WT CCR4, indicating that the epitope of mAb1567 is not solely a linear epitope on NT of CCR4, rather both ECLs and NT contribute to the binding of mAb1567 with CCR4. However, the CCR4-Nt alone is also sufficient for some degree of mAb1567 binding to CCR4-Nt-Fc as determined in ELISA studies to plate-bound mAb1567 (Supplementary Fig. S1B–S1D). Moreover, mAb1567 showed high specificity for CCR4 rather than CCR5, the most similar CCR molecule to CCR4 (Supplementary Fig. S1E).

Figure 1.

Overexpression of functional CCR4 on cutaneous T-cell lymphoma and mouse anti-CCR4 mAb1567 inhibits tumor formation. A, dose-dependent binding curve of mAb1567 to CCR4+ Mac-1 cells by FACS analysis. The EC50 was generated using SigmaPlot software. B, mAb1567 effectively inhibited chemotaxis of Mac-1 cells to CCR4 ligands, CCRL22 and CCL17. C, the antitumor effect of mAb1567 in SCID/Beige mice–bearing Mac-1 xenografts.

Figure 1.

Overexpression of functional CCR4 on cutaneous T-cell lymphoma and mouse anti-CCR4 mAb1567 inhibits tumor formation. A, dose-dependent binding curve of mAb1567 to CCR4+ Mac-1 cells by FACS analysis. The EC50 was generated using SigmaPlot software. B, mAb1567 effectively inhibited chemotaxis of Mac-1 cells to CCR4 ligands, CCRL22 and CCL17. C, the antitumor effect of mAb1567 in SCID/Beige mice–bearing Mac-1 xenografts.

Close modal

We then tested the antitumor effect of mAb1567 in vivo in a CTCL model using immunodeficient SCID/Beige mice that lack T and B cells and have defective NK function. SCID/Beige mice implanted with Mac-1 cells can efficiently form subcutaneous tumors (25). As shown in Fig. 1C, the tumor size in the mAb1567-treated group was 3- to 4-fold smaller than seen in the control group. None of the mice showed mAb1567 treatment–related toxicity.

mAb1567 mediates killing of Mac-1 cells by both CDC and neutrophil ADCC

To further understand the mechanism underlying the antitumor effect of mAb1567 seen in the SCID/Beige mice, we tested whether mAb1567 can mediate CDC and/or neutrophil-mediated ADCC effects against CCR4+ tumor cells in vitro. mAb1567 induced a significant lysis of Mac-1 cells in a dose-dependent manner in the presence of mouse complement as compared with the mouse IgG2b isotype control antibody (Fig. 2A). Rabbit complement was also tested and mAb1567 mediated a much more potent CDC activity and reached 80% of the target cell lysis (Fig. 2B). Next, neutrophils isolated from the SCID/Beige mice were tested in an in vitro ADCC assay. As shown in Fig. 2C, mAb1567 specifically mediated approximately 20% lysis via mouse neutrophils as compared with control at E/T (neutrophils/Mac-1) ratio of 80:1. These results show that mAb1567 can directly mediate not only CDC but also mouse neutrophil-induced ADCC activities.

Figure 2.

mAb1567 mediates killing of Mac-1 cells by both CDC and neutrophil-ADCC. A, mAb1567-mediated CDC activity via mouse complement. Figure shown is one experiment that is representative of at least 3 independent experiments. B, mAb1567-mediated CDC activity with rabbit complement. C, neutrophils from SCID-Beige mice–mediated mAb1567-dependent ADCC.

Figure 2.

mAb1567 mediates killing of Mac-1 cells by both CDC and neutrophil-ADCC. A, mAb1567-mediated CDC activity via mouse complement. Figure shown is one experiment that is representative of at least 3 independent experiments. B, mAb1567-mediated CDC activity with rabbit complement. C, neutrophils from SCID-Beige mice–mediated mAb1567-dependent ADCC.

Close modal

Cloning, expression, and activity of chimeric mAb1567

To humanize mAb1567 for further preclinical studies, the cDNAs encoding the VH and VL genes from the hybridoma cell line were individually recovered by RT-PCR using primers specific for mouse antibody variable genes. The VH and VL of mAb1567 belong to mouse VH1 (IGHV1S56*01) and VK8 (IGKV8-27*01) families and were rearranged with the JH1 (IGHJ1*01) and JK2 (IGKJ2*01) segments, respectively. The cloned and rearranged VH and VL genes were then assembled as a single-chain antibody variable region fragment (scFv) using a (G4S)3 linker. Binding of the recombinant mAb1567 to CCR4 was verified in both scFv-Fc IgG1 minibody (c1567-scFv-Fc; Fig. 3A) and full-length chimeric IgG1 form (c1567-IgG; data not shown).

Figure 3.

Humanization of mAb1567 and function analysis. A, comparative binding analysis of c1567 and h1567 scFv-Fcs. B and C, ADCC activity mediated by c1567 scFv-Fc. Either PBMCs (B) or NK cells (C) from healthy donors were used as effector cells. Target cell lysis was measured either by Cr51 (B) or LDH release (C). Data were calculated from triplicate wells of one experiment and are representative of 3 independent experiments. D, amino acid sequence alignment of the rearranged mouse and humanized variable heavy (VH) and variable light (VK) κ domains. Residues in magenta indicate framework residues that were changed for humanization.

Figure 3.

Humanization of mAb1567 and function analysis. A, comparative binding analysis of c1567 and h1567 scFv-Fcs. B and C, ADCC activity mediated by c1567 scFv-Fc. Either PBMCs (B) or NK cells (C) from healthy donors were used as effector cells. Target cell lysis was measured either by Cr51 (B) or LDH release (C). Data were calculated from triplicate wells of one experiment and are representative of 3 independent experiments. D, amino acid sequence alignment of the rearranged mouse and humanized variable heavy (VH) and variable light (VK) κ domains. Residues in magenta indicate framework residues that were changed for humanization.

Close modal

As NK cell–mediated ADCC is one of the most important mechanisms of action for immunotherapy with human IgG1 antibodies, we further tested whether recombinant mAb1567 can mediate ADCC via NK cells. Chimeric 1567 in both scFv-Fc or IgG1 forms were highly effective in killing Mac-1 cells in an in vitro ADCC assay using human PBMCs (Fig. 3B) or purified NK (CD56+CD16+) cells (Fig. 3C) from healthy donors as effector cells at different E/T ratios.

Humanization of mAb1567 and related biologic studies

Next, the structure-guided complementarity-determining region (CDR) grafting approach was used to humanize mAb1567. Homology 3-dimensional modeling of the VH and VL chains of mAb1567 using Web Antibody Modeling program (26) was generated to known antibody structures in the PDB database. For selecting the human acceptor framework template for CDR-grafting, the VH and VL amino acid sequences of mAb1567 were separately compared with human Ab sequences in the IGBLAST database to identify the most similar human Ab and Ig germline VH and VL sequences (Fig 3D). The human VH (McAb Ctm01, PDB:1ae6H) and VL (GenBank #ABG38372) share 82% and 84% amino acid sequence homology to the VH and VL of mAb1567, respectively; the best matched human Ig germline V sequences are IGHV1-3*01 (67% homology to mAb1567-VH) and IGKV4-1*01 (83% homology to mAb1567-VL). The framework residues of mAb1567 were manually changed to the selected human framework residues to generate the humanized mAb1567 (h1567). GROMOS force field energy minimization parameter was then applied to homology model h1567 using DeepView program (27). Examination of this energy minimized homology model of h1567 was carried out to ensure that no residues had distorted geometry or steric clashes with other residues.

The h1567 sequence shown in Fig. 3D has 21 and 11 amino acid differences in the framework regions compared with the mouse VH and VL, respectively. The humanized VH and VL gene were de novo synthesized and codon-optimized for mammalian cell expression. The binding affinity of h1567 and c1567 scFv-Fcs to CCR4 was then compared by FACS with Mac-1 cells. The h1567 had approximately 2-fold decrease in binding as compared with c1567 but both are in the nanomolar range, with EC50 of 2.2 and 1 nmol/L, respectively (Fig. 3A). The humanized h1567 scFv-Fc maintained potent NK-mediated ADCC killing of Mac-1 cells compared with c1567 (Fig 3C).

To test the in vivo antitumor effect of h1567, the luciferase-expressing Mac-1 cells were subcutaneously implanted into the dorsolateral flank of SCID/Beige mice, and mice were intraperitoneally treated with 5 mg/kg of control-scFv-Fc, c1567-scFv-Fc, h1567-scFv-Fc, or equivalent volumes of saline. Tumor growth in mice was monitored for luciferase intensity by IVIS imaging. All mice were sacrificed on day 28 and tumors were excised for photographing and measuring tumor weight. As shown in Fig. 4, tumors were significantly reduced in size at day 21 in c1567- and h1567-treated groups but not in control-scFv-Fc or PBS-treated groups as measured by IVIS imaging (Fig. 4A, top, and C), size of the excised tumors (Fig. 4A, bottom), tumor volume (Fig. 4B), and tumor weight (Fig. 4D).

Figure 4.

Humanized 1567 in tumor treatment. A, mice-bearing Mac-1 tumors were imaged using an IVIS imaging system. Luciferase signal (top) and tumor size (bottom) in mice treated with anti-CCR4 antibodies. Color scale: blue, luminescent signal intensity; red, least intense signal; most intense signal. Bar scale, 1 cm. Tumor sizes (B), image intensity (C), and tumor weight (D) were measured.

Figure 4.

Humanized 1567 in tumor treatment. A, mice-bearing Mac-1 tumors were imaged using an IVIS imaging system. Luciferase signal (top) and tumor size (bottom) in mice treated with anti-CCR4 antibodies. Color scale: blue, luminescent signal intensity; red, least intense signal; most intense signal. Bar scale, 1 cm. Tumor sizes (B), image intensity (C), and tumor weight (D) were measured.

Close modal

ADCC and CDC activities of higher affinity h1567 variants

Although the h1567 exhibited similar biologic activity as its murine counterpart in both in vitro and in vivo, the relative apparent binding affinity of h1567 is 2-fold lower than c1567 (Fig. 3A). To further affinity mature the h1567, we conducted VL-chain shuffling and alanine scanning to identify key residues in CDRs, followed by selection and screening of phage display library constructed by random mutagenesis of key residues in the CDRs (see Supplementary Method and SupplementaryFig. S2 for details).

The 2 affinity-improved h1567 variants, mAbs 1-44 and 2-3, that showed higher binding affinity to Mac-1 cells than parental h1567 with EC50 of 1.47 and 1.39 nmol/L, respectively (Supplementary Fig. S2F) were further evaluated for their capacity to mediate ADCC activity using human NK cells. The result showed that improvement in ADCC activity of the h1567 variants is correlated with their binding affinity, 2-3-scFv-Fc exhibited the best human NK-mediated ADCC activity for both Mac-1 cells (Fig. 5A) and Cf2Th-CCR4 cells but not to negative control Cf2Th (Supplementary Fig. S3). Moreover, because parental mAb1567 could induce mouse neutrophil-mediated ADCC, h1567 and 2-3 were tested for human neutrophil-mediated ADCC assay and mAb2-3 showed enhanced cytotoxic activity (Fig. 5B) compared with h1567. Furthermore, slightly improved CDC activity against Mac-1 cells was also observed for both 1-44 and 2-3 variants, but more for the 2-3-scFv-Fc (Supplementary Fig. S4).

Figure 5.

Humanized 1567 variants with improved binding affinity, ADCC, and CDC activity. A, H1567 and its variants 1-44 and 2-3 were tested in the ADCC activity. Data shown in the box and whiskers graph represent 3 independent experiments and each was carried out with NK cells from a different healthy donor; the box extends from lowest percentile to the highest percentile, with a line at the median. The whiskers above and below the box indicate the 95th and 5th percentiles. B, ADCC activities on Mac-1 with human neutrophils. C, ADCC activities on Mac-1 with human NK cells. D, CDC of wild-type Fc antibodies (scFv-Fcs) and mutant Fc antibodies (scFv-mFcs) against Mac-1.

Figure 5.

Humanized 1567 variants with improved binding affinity, ADCC, and CDC activity. A, H1567 and its variants 1-44 and 2-3 were tested in the ADCC activity. Data shown in the box and whiskers graph represent 3 independent experiments and each was carried out with NK cells from a different healthy donor; the box extends from lowest percentile to the highest percentile, with a line at the median. The whiskers above and below the box indicate the 95th and 5th percentiles. B, ADCC activities on Mac-1 with human neutrophils. C, ADCC activities on Mac-1 with human NK cells. D, CDC of wild-type Fc antibodies (scFv-Fcs) and mutant Fc antibodies (scFv-mFcs) against Mac-1.

Close modal

Fc engineering was also conducted on mutant antibodies 1-44 and 2-3 to further enhance ADCC activity by mutating 3 residues (S239D, A330L, and I332E) in CH2 domain, which have been shown to increase ADCC effect of human IgG1 antibody (28). As shown in Fig. 5C, ADCC-mediated by 1–44- and 2-3-scFv-mFcs was significantly enhanced as compared with their WT Fc counterparts or the WT h1567. However, as the A330L mutation in the Fc domain can ablate CDC function (29), we also tested and confirmed that CDC activity for the scFv-mFcs forms of 1-44 and 2-3 scFv-mFcs was completely abolished (Fig. 5D). Taken together, the affinity optimized variants of humanized 1567, in particular the 2-3 variant, showed improved ADCC and CDC effector functions, and NK cell–mediated ADCC activity can be further enhanced through Fc engineering.

mAb1567 inhibits Treg chemotaxis and partially abrogates Treg suppression on Teffs in vitro

Finally, as the majority (94%) of freshly isolated CD4+CD25high Tregs from peripheral blood express high level of surface CCR4 (30) and they have been reported to migrate to tumors secreting CCL22 (31), we investigated whether Ab1567 could have an antitumor role by modulating the chemotactic recruitment and suppressive activity of human CD4+ Tregs. First, we confirmed that CD4+CD25high Tregs migrated toward CCL22 much more effectively than CD4+CD25 T cells (Fig. 6A). Next, using peripheral blood CD4+ T cells in combination with examining the Treg phenotype of the migrated cells, we confirmed that c1567 completely inhibited the migration of CD4+CD25high Tregs in a transwell chemotaxis assay at concentrations greater than 2 μg/mL (Fig. 6B).

Figure 6.

Anti-CCR4 antibody abrogates suppression by Tregs. A, CD4+CD25high T cells showed demonstrable chemotactic responses toward CCL22. B, chimeric mAb1567 effectively inhibited chemotaxis of Tregs to CCL22. C, the effect of anti-CCR4 antibodies on proliferation of Teffs and the abrogation of the suppressive function of Tregs. The percent proliferation was normalized to CD4+CD25 Teffs without antibody treatment.

Figure 6.

Anti-CCR4 antibody abrogates suppression by Tregs. A, CD4+CD25high T cells showed demonstrable chemotactic responses toward CCL22. B, chimeric mAb1567 effectively inhibited chemotaxis of Tregs to CCL22. C, the effect of anti-CCR4 antibodies on proliferation of Teffs and the abrogation of the suppressive function of Tregs. The percent proliferation was normalized to CD4+CD25 Teffs without antibody treatment.

Close modal

In addition, as we are unaware of any published data on the role of CCR4 in Treg function, we also examined whether 1567 engagement of CCR4 could result in modulation of Treg suppression activity in an in vitro Treg suppression assay. As shown in Fig. 6C, the proliferation of CD4+ T-effector cells (Teffs alone, lane 1) was inhibited by highly purified CD4+CD25high Tregs (1:2 ratio) by 78% (lane 7), which is our typical Treg suppression effect on Teffs (32). Surprisingly, in the presence of c1567IgG or h1567scFv-Fc, the proliferation of Teff was stimulated to 183% and 207%, respectively (lanes 3 and 5), but there was no stimulatory effect on Treg (lanes 4 and 6). In the Treg/Teff coculture (1:2 ratio), the proliferation of Teff was inhibited directly (lane 7) and there was no reversal of this inhibition by control mAb (lane 8). Moreover, T-cell proliferation was restored with a net positive response to 258% and 221% (lanes 9 and 10) in the presence of c1567IgG or h156scFv-Fc, respectively.

The increase in CD4+CD25 T-cell proliferation in the presence of anti-CCR4 antibodies was further observed by flow cytometry. We detected CD4+CD25 T-cell proliferation on the basis of carboxyfluorescein succinimidyl ester fluorescence intensity of labeled CD4+CD25 T cells in the Treg/Teff coculture. The analysis revealed that over the 7-day study, CD4+CD25 T-cell proliferation responded to stimulation of anti-CCR4 antibodies in a time-dependent manner and was approximately 50% higher than with control antibody–treated cells that showed no proliferation (Supplementary Fig. S5). Importantly, Treg-mediated suppression of Teff proliferation occurred even in the presence of anti-CD3/CD28 costimulation but could be reversed in the presence of anti-CCR4 antibody resulting in an increased proliferative capacity of CD4+CD25 T cells.

In this study, we humanized a mouse anti-CCR4 antibody, mAb1567, which recognizes both the NT and the ECLs of CCR4 with high affinity and inhibits migration of CCR4+ tumor cells toward its 2 ligands, CCL22 and CCL17. The antibody exhibited potent antitumor effect in a CTCL mouse model with the mechanisms of CDC and neutrophil-mediated ADCC likely involved (Figs. 1 and 2). The chimeric or humanized mAb1567 also showed potent CDC and human NK cell–mediated ADCC activity in vitro and therapeutic effects in vivo (Figs. 3 and 4). In addition, this antibody effectively inhibited the chemotaxis of CD4+CD25high Tregs to CCL22. Interestingly, it also stimulated CD4+CD25 cell proliferation and inhibited Tregs' immune-suppressive activity (Fig. 6 and Supplementary Fig. S5). The affinity of h1567 was further improved by employing a targeted mutagenesis strategy in combination with phage-display library selection and the affinity optimized h1567 variant mAb2-3 showed better CDC and ADCC activities against CCR4+ tumor cells in vitro (Fig. 5). These studies support that the affinity selected mAb2-3 may provide a novel immunotherapy option not only to directly kill the CCR4+ tumor cells, but also may have a role in other cancers by suppressing Treg trafficking and overcoming the suppressive effect of CCR4+ Tregs to enhance host antitumor immune responses.

A number of studies have been reported on a humanized anti-CCR4 antibody, KM-0761 (33, 34). These studies have not only showed KM-0761′s effect in CTCL tumor animal models and its mechanism of action, but also convincing support that CCR4 is an important antibody-based immunotherapy target for relapsed CCR4+ ATL or PTCL (35, 36). The KM-0761 antibody was originally raised in mouse by immunizing with the CCR4′s NT(12–29) peptide (34, 37), it works in killing tumor cells by ADCC mainly through NK and/or macrophages (33, 38, 39), and augmentation of FcγR engagement and ADCC killing is achieved by defucosylation (34). In contrast, mAb1567 was generated with full-length CCR4, it recognizes an epitope comprising of both NT and ECLs of CCR4, and ADCC-enhanced killing is achieved through both affinity maturation and Fc engineering (29). In addition, mAb1567 showed both CDC and ADCC activity. The disparity in CDC activities between these 2 IgG1 antibodies is likely due to different recognition of the CCR4 epitope(s). CDC is often dependent on the distance between the plasma membrane and the constant region of the sensitizing antibody that mediates effector functions (40). It is possible that the CDC activity mediated by mAb1567 is related to the position and orientation of the recognized epitope, when bound, the mAb1567 can position its Fc region more proximal to the membrane to recruit complement efficiently. Another possibility is that binding to the different epitope may promote more efficient cross-linking of CCR4, and thus, increasing the binding avidity (41).

Antitumor effect mediated by cancer cell–directed antibodies can generally be attributed to ADCC, CDC, or direct antiproliferation. We found that mAb1567 did not show any inhibition of cell proliferation (data not shown). Effector cells that can mediate ADCC are NK cells, neutrophils, and monocytes/macrophages. The SCID/Biege mice used in our in vivo studies not only lack T- and B-lymphocytes but are also NK cell defective. Thus, we surmised that the antitumor activity of mAb1567 seen in SCID/Beige mice might be due to effector cells other than NK cell–mediated ADCC and/or to CDC. In vitro CDC experiments indeed showed that mAb1567 had CDC activity as compared with control Ab, although the level of lysis target Mac-1 cells was low, upto approxiamtely 20%. Most mouse strains (including the mouse stains tested here) have exceptionally low complement activity, much lower than that of humans or other animals, including rabbits, guinea pigs, and hamsters (42). CDC assay with rabbit complement indeed showed dramatically improved lysis of target cells by mAb1567 (Fig. 2B) upto approximately 80%. Furthermore, mAb1567 could mediate ADCC activity (approximately 25%) via neutrophils isolated from SCID/Beige mice. Because of limited number of neutrophils available, only a single dose of mAb1567 and isotype control mAb at 50 μg/mL was tested (Fig. 2C). These results suggest that the antitumor activity of mAb1567 in the mouse model is likely due to a combination of both CDC and through neutrophil-mediated ADCC, although it remains to be tested if mouse monocytes/macrophages may also play a role.

Many therapies for cancer are accompanied by adverse side effects and dose-dependent toxicities. The development of more effective cancer immunotherapy with better discrimination between tumor cells and normal cells is the most important goal of current anticancer research. CCR4 is universally expressed at high levels among most CTCL cells; however, a subset of normal T cells also can express CCR4. It was reported that the CCR4 is selectively expressed on T-helper 2 (TH2) cells, but not TH1 cells, which are the precursors of memory T cells (43). Of particular importance to the in vivo expression of CCR4 on TH2 cells would be a concern of bystander cytotoxicity in response to anti-CCR4 antibody treatment. However, potential bystander effects can be managed as shown in the recent clinical trial reports on the anti-CCR4 mAb, KW-0761, for treatment of adult T-cell leukemia/lymphoma and PTCL, where good clinical activity was seen without serious side effects (35, 36). This may be due, at least in part, to the CCR4-expressing CTCL patient cells showing a significantly 4-fold higher expression level than T cells from healthy donors (44). This high CCR4 expression in CTCL patients may result in most anti-CCR4 antibody binding to tumor T cells but not normal T cells. We are also aware of reports that CCR4 is expressed on platelets (45, 46) but again in the KW-0761 clinical trial (35), grade 3 or 4 thrombocytopenia was only seen in 5 of 28 patients (18%) and not associated with bleeding.

CCR4 is also highly expressed on the majority (94%) of CD4+CD25highFoxP3+ Tregs (30) that are considered the most potent inhibitors of antitumor immunity and the greatest barrier to successful immunotherapy (31, 47). CCR4 also plays an important role in Treg recruitment to the site of action. In some solid tumors including breast cancer (17, 48), ovarian cancer (18), and oral squamous cell carcinoma (49), increased numbers of recruited Tregs that are chemoattracted through the CCR4–CCL22 axis likely play a significant role in the suppression of host antitumor immunity. This trafficking pattern makes CCR4 an even more attractive therapeutic target for a broader range of tumors. mAb1567 was tested for inhibition of Treg migration and showed complete inhibition of chemotaxis of CD4+CD25high Tregs (Fig. 6A). Interestingly, it appeared that mAb1567 can also abrogate the immune suppressive function of CD4+CD25high Tregs while stimulating the proliferation of CD4+CD25 Teffs (Fig. 6C and Supplementary Fig. S5). It has been proposed that Tregs exert their function by multiple suppressive mechanisms including cell–cell contact, including competitive consumption of IL-2, production of the immunosuppressive cytokines IL-10 and TGF-β, cytolysis, metabolic disruption, and modulation of the function of antigen-presenting cells (5, 47). However, CCR4 is not known to play a role in CD4+CD25 cell proliferation and although a previous study described that CCR4−/− Tregs are defective in suppressive activity in a model of colitis, this was due to their lack of chemotactic recruitment to the mesenteric lymph nodes because their in vitro suppressive function was intact (50). It will be important to conduct additional studies to further confirm these findings and elucidate the mechanism(s) by which anti-CCR4 mAb1567 could act through CCR4 on these cells to exercise different functions—proliferative effect on CD4+CD25 Teffs and partial to complete abrogation of Tregs' suppression.

In summary, we have humanized and affinity maturated murine anti-CCR4 antibody 1567 and showed its in vitro and in vivo antitumor activity against CTCL cells. The mechanisms of tumor cell killing by mAb1567 seem to be multiple including CDC as well as neutrophil- and NK cell–mediated ADCC. The affinity-matured mAb2-3 promoted more potent CTCL lysis by augmentation of both NK cell–mediated ADCC and CDC activities. Remarkably, the activity of mAb1567 also extended to the normal T-cell compartment with demonstrable proliferative effect on CD4+CD25 Teffs and abrogation of Treg suppression. These findings support further evaluation of our anti-CCR4 antibody as an immunotherapy for CTCL and other cancers where augmentation of Teff functions against the tumor cells are likely to have a therapeutic benefit.

No potential conflicts of interest were disclosed.

Conception and design: J. Sui, M. Bai, R. C. Fuhlbrigge, Q. Zhu, T. S. Kupper, W. A. Marasco

Development of methodology: D.-K. Chang, J. Sui, S. Geng, A. Muvaffak, M. Bai, R. C. Fuhlbrigge, A. S. Lo, Q. Zhu, W. A. Marasco

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S. Geng, M. Bai, A. Yammanuru, L. Hubbard, J. Sheehan, J. J. Campbell, Q. Zhu, T. S. Kupper

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): D.-K. Chang, J. Sui, S. Geng, M. Bai, A. Yammanuru, Q. Zhu, T. S. Kupper, W. A. Marasco

Writing, review, and/or revision of the manuscript: D.-K. Chang, J. Sui, R. C. Fuhlbrigge, J. Sheehan, Q. Zhu, T. S. Kupper, W. A. Marasco

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): D.-K. Chang, J. Sheehan

Study supervision: J. Sui, Q. Zhu, W. A. Marasco

The authors thank Maryam Ali, Hong Tao, and Erin M. McConocha for technical support, and National Foundation of Cancer Research (CFCR) for contribution of equipment used in this study.

This work was funded by Skin Cancer Score project 2P50CA093683 to T. S. Kupper, J. Campbell, and W. A. Marasco and NIH AI058804 to Q. Zhu.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Willemze
R
,
Jaffe
ES
,
Burg
G
,
Cerroni
L
,
Berti
E
,
Swerdlow
SH
, et al
WHO-EORTC classification for cutaneous lymphomas
.
Blood
2005
;
105
:
3768
85
.
2.
Clark
RA
,
Watanabe
R
,
Teague
JE
,
Schlapbach
C
,
Tawa
MC
,
Adams
N
, et al
Skin effector memory T-cells do not recirculate and provide immune protection in alemtuzumab-treated CTCL patients
.
Sci Transl Med
2012
;
4
:
117ra7
.
3.
Kim
YH
,
Liu
HL
,
Mraz-Gernhard
S
,
Varghese
A
,
Hoppe
RT
. 
Long-term outcome of 525 patients with mycosis fungoides and Sezary syndrome: clinical prognostic factors and risk for disease progression
.
Arch Dermatol
2003
;
139
:
857
66
.
4.
Kadin
ME
,
Carpenter
C
. 
Systemic and primary cutaneous anaplastic large cell lymphomas
.
Semin Hematol
2003
;
40
:
244
56
.
5.
Krejsgaard
T
,
Odum
N
,
Geisler
C
,
Wasik
MA
,
Woetmann
A
. 
Regulatory T-cells and immunodeficiency in mycosis fungoides and Sezary syndrome
.
Leukemia
2011
;
26
:
424
32
.
6.
Zhang
Q
,
Nowak
I
,
Vonderheid
EC
,
Rook
AH
,
Kadin
ME
,
Nowell
PC
, et al
Activation of Jak/STAT proteins involved in signal transduction pathway mediated by receptor for interleukin 2 in malignant T lymphocytes derived from cutaneous anaplastic large T-cell lymphoma and Sezary syndrome
.
Proc Natl Acad Sci U S A
1996
;
93
:
9148
53
.
7.
Kasprzycka
M
,
Zhang
Q
,
Witkiewicz
A
,
Marzec
M
,
Potoczek
M
,
Liu
X
, et al
Gamma c-signaling cytokines induce a regulatory T-cell phenotype in malignant CD4+ T lymphocytes
.
J Immunol
2008
;
181
:
2506
12
.
8.
Yawalkar
N
,
Ferenczi
K
,
Jones
DA
,
Yamanaka
K
,
Suh
KY
,
Sadat
S
, et al
Profound loss of T-cell receptor repertoire complexity in cutaneous T-cell lymphoma
.
Blood
2003
;
102
:
4059
66
.
9.
Liu
HL
,
Hoppe
RT
,
Kohler
S
,
Harvell
JD
,
Reddy
S
,
Kim
YH
. 
CD30+ cutaneous lymphoproliferative disorders: the Stanford experience in lymphomatoid papulosis and primary cutaneous anaplastic large cell lymphoma
.
J Am Acad Dermatol
2003
;
49
:
1049
58
.
10.
Querfeld
C
,
Rosen
ST
,
Guitart
J
,
Kuzel
TM
. 
The spectrum of cutaneous T-cell lymphomas: new insights into biology and therapy
.
Curr Opin Hematol
2005
;
12
:
273
8
.
11.
Kupper
TS
,
Fuhlbrigge
RC
. 
Immune surveillance in the skin: mechanisms and clinical consequences
.
Nat Rev Immunol
2004
;
4
:
211
22
.
12.
Kunkel
EJ
,
Boisvert
J
,
Murphy
K
,
Vierra
MA
,
Genovese
MC
,
Wardlaw
AJ
, et al
Expression of the chemokine receptors CCR4, CCR5, and CXCR3 by human tissue-infiltrating lymphocytes
.
Am J Pathol
2002
;
160
:
347
55
.
13.
Campbell
JJ
,
Haraldsen
G
,
Pan
J
,
Rottman
J
,
Qin
S
,
Ponath
P
, et al
The chemokine receptor CCR4 in vascular recognition by cutaneous but not intestinal memory T-cells
.
Nature
1999
;
400
:
776
80
.
14.
Campbell
JJ
,
Clark
RA
,
Watanabe
R
,
Kupper
TS
. 
Sezary syndrome and mycosis fungoides arise from distinct T-cell subsets: a biologic rationale for their distinct clinical behaviors
.
Blood
2010
;
116
:
767
71
.
15.
Wu
XS
,
Lonsdorf
AS
,
Hwang
ST
. 
Cutaneous T-cell lymphoma: roles for chemokines and chemokine receptors
.
J Invest Dermatol
2009
;
129
:
1115
9
.
16.
Tokura
Y
,
Sugita
K
,
Yagi
H
,
Shimauchi
T
,
Kabashima
K
,
Takigawa
M
. 
Primary cutaneous anaplastic large cell lymphoma with fatal leukemic outcome in association with CLA and CCR4-negative conversion
.
J Am Acad Dermatol
2007
;
57
:
S92
6
.
17.
Olkhanud
PB
,
Baatar
D
,
Bodogai
M
,
Hakim
F
,
Gress
R
,
Anderson
RL
, et al
Breast cancer lung metastasis requires expression of chemokine receptor CCR4 and regulatory T-cells
.
Cancer Res
2009
;
69
:
5996
6004
.
18.
Curiel
TJ
,
Coukos
G
,
Zou
L
,
Alvarez
X
,
Cheng
P
,
Mottram
P
, et al
Specific recruitment of regulatory T-cells in ovarian carcinoma fosters immune privilege and predicts reduced survival
.
Nat Med
2004
;
10
:
942
9
.
19.
Ishida
T
,
Iida
S
,
Akatsuka
Y
,
Ishii
T
,
Miyazaki
M
,
Komatsu
H
, et al
The CC chemokine receptor 4 as a novel specific molecular target for immunotherapy in adult T-cell leukemia/lymphoma
.
Clin Cancer Res
2004
;
10
:
7529
39
.
20.
Kadin
ME
,
Cavaille-Coll
MW
,
Gertz
R
,
Massague
J
,
Cheifetz
S
,
George
D
. 
Loss of receptors for transforming growth factor beta in human T-cell malignancies
.
Proc Natl Acad Sci U S A
1994
;
91
:
6002
6
.
21.
Sui
J
,
Li
W
,
Murakami
A
,
Tamin
A
,
Matthews
LJ
,
Wong
SK
, et al
Potent neutralization of severe acute respiratory syndrome (SARS) coronavirus by a human mAb to S1 protein that blocks receptor association
.
Proc Natl Acad Sci U S A
2004
;
101
:
2536
41
.
22.
Reff
ME
,
Carner
K
,
Chambers
KS
,
Chinn
PC
,
Leonard
JE
,
Raab
R
, et al
Depletion of B cells in vivo by a chimeric mouse human monoclonal antibody to CD20
.
Blood
1994
;
83
:
435
45
.
24.
Jopling
LA
,
Sabroe
I
,
Andrew
DP
,
Mitchell
TJ
,
Li
Y
,
Hodge
MR
, et al
The identification, characterization, and distribution of guinea pig CCR4 and epitope mapping of a blocking antibody
.
J Biol Chem
2002
;
277
:
6864
73
.
25.
Pfeifer
W
,
Levi
E
,
Petrogiannis-Haliotis
T
,
Lehmann
L
,
Wang
Z
,
Kadin
ME
. 
A murine xenograft model for human CD30+ anaplastic large cell lymphoma. Successful growth inhibition with an anti-CD30 antibody (HeFi-1)
.
Am J Pathol
1999
;
155
:
1353
9
.
26.
Whitelegg
NR
,
Rees
AR
. 
WAM: an improved algorithm for modelling antibodies on the WEB
.
Protein Eng
2000
;
13
:
819
24
.
27.
Daura
X
,
Oliva
B
,
Querol
E
,
Aviles
FX
,
Tapia
O
. 
On the sensitivity of MD trajectories to changes in water-protein interaction parameters: the potato carboxypeptidase inhibitor in water as a test case for the GROMOS force field
.
Proteins
1996
;
25
:
89
103
.
28.
Carter
PJ
. 
Potent antibody therapeutics by design
.
Nat Rev Immunol
2006
;
6
:
343
57
.
29.
Lazar
GA
,
Dang
W
,
Karki
S
,
Vafa
O
,
Peng
JS
,
Hyun
L
, et al
Engineered antibody Fc variants with enhanced effector function
.
Proc Natl Acad Sci U S A
2006
;
103
:
4005
10
.
30.
Baatar
D
,
Olkhanud
P
,
Sumitomo
K
,
Taub
D
,
Gress
R
,
Biragyn
A
. 
Human peripheral blood T regulatory cells (Tregs), functionally primed CCR4+ Tregs and unprimed CCR4- Tregs, regulate effector T cells using FasL
.
J Immunol
2007
;
178
:
4891
900
.
31.
Byrne
WL
,
Mills
KH
,
Lederer
JA
,
O'Sullivan
GC
. 
Targeting regulatory T-cells in cancer
.
Cancer Res
2011
;
71
:
6915
20
.
32.
Hirahara
K
,
Liu
L
,
Clark
RA
,
Yamanaka
K
,
Fuhlbrigge
RC
,
Kupper
TS
. 
The majority of human peripheral blood CD4+CD25highFoxp3+ regulatory T-cells bear functional skin-homing receptors
.
J Immunol
2006
;
177
:
4488
94
.
33.
Ishida
T
,
Ishii
T
,
Inagaki
A
,
Yano
H
,
Kusumoto
S
,
Ri
M
, et al
The CCR4 as a novel-specific molecular target for immunotherapy in Hodgkin lymphoma
.
Leukemia
2006
;
20
:
2162
8
.
34.
Niwa
R
,
Shoji-Hosaka
E
,
Sakurada
M
,
Shinkawa
T
,
Uchida
K
,
Nakamura
K
, et al
Defucosylated chimeric anti-CC chemokine receptor 4 IgG1 with enhanced antibody-dependent cellular cytotoxicity shows potent therapeutic activity to T-cell leukemia and lymphoma
.
Cancer Res
2004
;
64
:
2127
33
.
35.
Ishida
T
,
Joh
T
,
Uike
N
,
Yamamoto
K
,
Utsunomiya
A
,
Yoshida
S
, et al
Defucosylated anti-CCR4 monoclonal antibody (KW-0761) for relapsed adult T-cell leukemia-lymphoma: a multicenter phase II study
.
J Clin Oncol
2012
;
30
:
837
42
.
36.
Yamamoto
K
,
Utsunomiya
A
,
Tobinai
K
,
Tsukasaki
K
,
Uike
N
,
Uozumi
K
, et al
Phase I study of KW-0761, a defucosylated humanized anti-CCR4 antibody, in relapsed patients with adult T-cell leukemia-lymphoma and peripheral T-cell lymphoma
.
J Clin Oncol
2010
;
28
:
1591
8
.
37.
Ishida
T
,
Utsunomiya
A
,
Iida
S
,
Inagaki
H
,
Takatsuka
Y
,
Kusumoto
S
, et al
Clinical significance of CCR4 expression in adult T-cell leukemia/lymphoma: its close association with skin involvement and unfavorable outcome
.
Clin Cancer Res
2003
;
9
:
3625
34
.
38.
Yano
H
,
Ishida
T
,
Inagaki
A
,
Ishii
T
,
Ding
J
,
Kusumoto
S
, et al
Defucosylated anti-CC chemokine receptor 4 monoclonal antibody combined with immunomodulatory cytokines: a novel immunotherapy for aggressive/refractory mycosis fungoides and Sezary syndrome
.
Clin Cancer Res
2007
;
13
:
6494
500
.
39.
Yano
H
,
Ishida
T
,
Imada
K
,
Sakai
T
,
Ishii
T
,
Inagaki
A
, et al
Augmentation of antitumour activity of defucosylated chimeric anti-CCR4 monoclonal antibody in SCID mouse model of adult T-cell leukaemia/lymphoma using G-CSF
.
Br J Haematol
2008
;
140
:
586
9
.
40.
Bindon
CI
,
Hale
G
,
Bruggemann
M
,
Waldmann
H
. 
Human monoclonal IgG isotypes differ in complement activating function at the level of C4 as well as C1q
.
J Exp Med
1988
;
168
:
127
42
.
41.
Teeling
JL
,
Mackus
WJ
,
Wiegman
LJ
,
van den Brakel
JH
,
Beers
SA
,
French
RR
, et al
The biological activity of human CD20 monoclonal antibodies is linked to unique epitopes on CD20
.
J Immunol
2006
;
177
:
362
71
.
42.
Ong
GL
,
Mattes
MJ
. 
Mouse strains with typical mammalian levels of complement activity
.
J Immunol Methods
1989
;
125
:
147
58
.
43.
Lloyd
CM
,
Delaney
T
,
Nguyen
T
,
Tian
J
,
Martinez
AC
,
Coyle
AJ
, et al
CC chemokine receptor (CCR)3/eotaxin is followed by CCR4/monocyte-derived chemokine in mediating pulmonary T helper lymphocyte type 2 recruitment after serial antigen challenge in vivo
.
J Exp Med
2000
;
191
:
265
74
.
44.
Ferenczi
K
,
Fuhlbrigge
RC
,
Pinkus
J
,
Pinkus
GS
,
Kupper
TS
. 
Increased CCR4 expression in cutaneous T-cell lymphoma
.
J Invest Dermatol
2002
;
119
:
1405
10
.
45.
Gear
AR
,
Camerini
D
. 
Platelet chemokines and chemokine receptors: linking hemostasis, inflammation, and host defense
.
Microcirculation
2003
;
10
:
335
50
.
46.
Abi-Younes
S
,
Si-Tahar
M
,
Luster
AD
. 
The CC chemokines MDC and TARC induce platelet activation via CCR4
.
Thromb Res
2001
;
101
:
279
89
.
47.
Zou
W
. 
Regulatory T-cells, tumour immunity and immunotherapy
.
Nat Rev Immunol
2006
;
6
:
295
307
.
48.
Hong
H
,
Gu
Y
,
Zhang
H
,
Simon
AK
,
Chen
X
,
Wu
C
, et al
Depletion of CD4+CD25+ regulatory T cells enhances natural killer T cell-mediated anti-tumour immunity in a murine mammary breast cancer model
.
Clin Exp Immunol
2010
;
159
:
93
9
.
49.
Watanabe
Y
,
Katou
F
,
Ohtani
H
,
Nakayama
T
,
Yoshie
O
,
Hashimoto
K
. 
Tumor-infiltrating lymphocytes, particularly the balance between CD8(+) T-cells and CCR4(+) regulatory T-cells, affect the survival of patients with oral squamous cell carcinoma
.
Oral Surg Oral Med Oral Pathol Oral Radiol Endod
2010
;
109
:
744
52
.
50.
Yuan
Q
,
Bromley
SK
,
Means
TK
,
Jones
KJ
,
Hayashi
F
,
Bhan
AK
, et al
CCR4-dependent regulatory T-cell function in inflammatory bowel disease
.
J Exp Med
2007
;
204
:
1327
34
.