Patients
Patients over 20 years old with advanced or recurrent CCR4-negative cancer were enrolled in this study from October 2013 to April 2015. CCR4 expression was determined by immunohistochemistry with anti-CCR4 mAb as described in Immunohistochemistry. Eligibility criteria included a good performance status (an Eastern Cooperative Oncology Group performance status of 0 to 2) and the following laboratory values: absolute neutrophil count ≥ 1,500/μL, hemoglobin ≥ 8.0 g/dL, platelet count ≥ 75,000/μL, total bilirubin ≤ 2.0 mg/dL, AST ≤ 2.5 × the upper limit of the normal range (UNL), ALT ≤ 2.5 × UNL, serum creatinine ≤ 1.5 mg/dL, and arterial blood oxygen saturation ≥ 93%. All patients underwent electrocardiography to confirm the absence of cardiac abnormalities requiring therapeutic intervention and that the left ventricular ejection fraction was at least 50%. Patients were excluded if they had an active infection, a history of organ transplantation, active concurrent cancer, any autoimmune diseases, central nervous system involvement, hepatitis B or C virus infection, HIV infection or previous ICB therapy.
Study design
This multi-institutional, open-label, two-arm, phase Ib study is a part of an investigator-initiated phase Ia/Ib clinical trial of mogamulizumab administration in patients with CCR4-negative advanced or recurrent solid tumors (NCT01929486). The primary objectives were to characterize the safety and the effect of Treg cell depletion in peripheral blood. The secondary objectives were to assess the antitumor activity and to determine the recommended phase II dose. Twenty and nineteen patients were randomly enrolled in cohorts treated with dosages of 1.0 and 0.1 mg/kg mogamulizumab, respectively, and received the drug weekly for 8 weeks, which was followed by monthly intravenous infusion until disease progression was observed. These two dosages of mogamulizumab were determined as the maximum-tolerated dose and minimal dose in our previous phase Ia study 24.
Toxicity was graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events (version 4.0). Clinical responses were evaluated at 12 weeks after the first mogamulizumab administration or at the point of study discontinuation using computed tomography scans. The effects were determined according to the RECIST criteria (version 1.1). Progression-free survival (PFS) was defined from the day of the first mogamulizumab administration until the day of progressive disease (PD) detection. Peripheral blood samples were serially collected, and PBMCs were isolated by density gradient centrifugation with Ficoll-Paque (GE Healthcare, Little Chalfont, UK). To collect TILs, fresh tumor tissues were minced and treated with a gentleMACS Dissociator (Miltenyi Biotec, Bergisch Gladbach, Germany) as previously described 37, 38, and the prepared cells were subjected to immune-monitoring assays.
The protocol was approved by the institutional review boards at each participating site, and all patients provided written informed consent before enrollment in accordance with the Declaration of Helsinki.
Immunohistochemistry
Biopsy samples were formalin-fixed, paraffin-embedded, and sectioned before they were placed onto slides for immunohistochemistry, which was conducted with anti-CCR4 mAb (KM2160; Kyowa Hakko Kirin) as previously reported 24. CCR4 positivity was evaluated by the review committee with central evaluation.
CyTOF analyses
CyTOF staining and analysis were performed as described 39. The antibodies used in the CyTOF analyses are summarized in Supplementary Table 1. Cells were subjected to staining after washing with PBS supplemented with 2% fetal calf serum (FCS, Biosera, Orange, CA, USA) (washing solution). The cells were incubated in 5 μM of Cell-ID rhodium solution (Fluidigm, South San Francisco, CA) in PBS, washed using washing solution, and stained with a mixture of surface-staining antibodies. After washing, the cells were fixed and permeabilized using a Foxp3/Transcription Factor Staining Buffer Set (Thermo Fisher Scientific, Waltham, MA) according to the manufacturer’s instructions. The fixed and permeabilized cells were stained with the intracellular antibodies. After washing twice, the cells were allowed to rest overnight in 125 nM MaxPar Intercalator-Ir (Fluidigm) diluted in PBS solution with 2% paraformaldehyde at 4°C. The cells were then washed once with washing solution and twice with MaxPar water (Fluidigm) and distilled water with minimal heavy element contamination to reduce the background level. The cells were resuspended in MaxPar water supplemented with 10% EQ Four Element Calibration Beads (Fluidigm) and then were applied to the Helios instrument (Fluidigm), and data were acquired at a speed below 300 events/second.
Preprocessing and analysis of CyTOF data
The data from the pre- and post-mogamulizumab administration samples were combined for each patient, and batch effects were removed using ResNet 40. After removing the batch effects, the data were combined, and the UMAP projections were generated using the R package "umap". A modified version of CYBERTRACK was used for clustering the CyTOF data 41. The cluster sizes were determined by the Elbow method, and debris clusters were removed for further analyses.
Flow cytometry analyses
Flow cytometry staining and analyses were performed as described 23, 39, 42. The antibodies used in the flow cytometry analyses are summarized in Supplementary Table 2. Cells (PBMCs and TILs) were washed using washing solution and subjected to staining with surface antibodies. Intracellular staining of FoxP3 was performed with anti-FoxP3 mAb and Foxp3/Transcription Factor Staining Buffer Set (Thermo Fisher Scientific) according to the manufacturer’s instructions. After washing, the cells were analyzed with an LSRFortessa instrument (BD Biosciences, San Jose, CA) and FlowJo software (Treestar, Ashland, OR). The dilution of the staining antibodies was performed according to the manufacturer's instructions.
ATAC-seq data processing
ATAC-seq data reported by Calderon et. al 43 were employed{Calderon, 2019 #1}. Law FASTQ files about naive CD8+ T cells, central-memory CD8+ T cells, effector-memory CD8+ T cells and Treg cells were downloaded from the Gene Expression Omnibus (accession no. GSE118189). Trim-Galore (https://www.bioinformatics.babraham.ac.uk/projects/trim_ galore/) was utilized for trimming adaptor sequence and filtering low quality reads. The filtered FASTQ files were aligned to GRCh38 genome using Bowtie2 v2.4.1 44. The reads mapped to chrM, mapping quality < 30 and with the flags “- F 1804” and “-f 2” were filtered out using samtools v1.9 (http://www.htslib.org), and the duplicated reads were also removed using Picard Tools v1.119 (https://broadinstitute.github.io/picard/). Then, the resultant BAM files for each cell type were merged. Chromatin accessibility signals were calculated form merged BAM files using deepTools2 45. For normalizing signals, we used reads counts data mapped to ATAC peaks regions detected by MACS2 46 under the parameters “-nonodel -nolamda -keep-dup all -call summits”. The MACS2 narrow peak files from each cell type were merged using bedtools2 (https://bedtools.readthedocs.io/en/latest/). The mapped reads in merged peaks region were counted using featureCounts 47. The size factors were calculated using R “DESeq” package, and then ATAC signals were normalized. For visualizing ATAC signals, Integrative Genomics Viewer was used 48.
ChIP-seq data processing
FOXP3 ChIP-seq data reported by Birzele et. al. and Schmldl et. al. were employed 49, 50. Law FASTQ files about Treg cells and naive Treg cells were downloaded from the NCBI SRA (SRP006674) and GEO (GSE43119), respectively. Trim-Galore was used for trimming adaptor sequence and filtering low quality reads. We then aligned filtered FASTQ files to GRCh38 genome using Bowtie2 v2.4.1 44. The duplicated reads and reads having mapping quality < 4 were filtered out using Picard Tools v1.119 and samtools v1.9, respectively. The resultant BAM files for each cell type were merged. FOXP3 ChIP signals were calculated from merged BAM files using deepTools2 under the parameters “-bs=10 –normalize Using CPM --extendReads 200” 45. For visualizing FOXP3 ChIP-seq signals, Integrative Genomics Viewer was used 48.
T cell culture
PBMCs from healthy individuals were cultured with in a round-bottom 96-well plate with medium containing IL-2 (30 U/mL). The indicated dose of mogamulizumab was added to some wells during the entire culture period. After 7 days, the cells were subjected to a BD FACSymphony A3 instrument (BD Biosciences) and FlowJo software (Treestar).
Statistical analysis
The relationships between groups were compared using a t-test or the nonparametric Mann-Whitney U test. For multiple group comparisons, the Bonferroni method was employed. PFS and OS were defined as the time from the initial mogamulizumab administration until the first observation of disease progression and death from any cause, respectively. PFS and OS were investigated with the Kaplan–Meier method and were compared among the groups using the log-rank test or Cox regression proportional hazards analysis. Statistical analysis was performed with GraphPad Prism8 (GraphPad Software, San Diego, CA) or R version 3.1.1 (R Foundation for Statistical Computing, Vienna, Austria). P values less than 0.05 were considered significant.