CONTACT FOR REAGENT AND RESOURCE SHARING
Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Jennifer A. Wargo, ([email protected]).
EXPERIMENTAL MODEL AND SUBJECT DETAILS
Patient cohort
Patients with advanced (stage III/IV) melanoma treated at The University of Texas MD Anderson Cancer Center between 01/23/2014 and 08/31/2017 who received at least one dose of ipilimumab in combination with a PD-1 checkpoint blockade agent (either nivolumab or pembrolizumab) as combination immune checkpoint blockade (CICB) were identified from detailed retrospective and prospective review of clinic records (Supplementary Data Table 1). CICB treatment was provided as part of clinical trial or expanded access program protocols (NCT01844505, NCT02186249, NCT02089685, NCT01621490, NCT02519322, NCT02320058) or as standard of care therapy. Due to known differences in underlying biology and immunotherapy responses between melanoma subtypes, only cutaneous melanomas were included for analysis of response as this was the dominant subset. All subtypes (i.e.: cutaneous, mucosal and uveal) were included for toxicity analyses. To enable translational analyses, patients without available biospecimens relevant to the CICB treatment period, or for whom insufficient data were available to determine radiographic responses and toxicity outcomes were excluded.
Mice
All mice experiments were approved by the local institutional board and performed in accordance with government and institutional guidelines and regulations. Female C57Bl/6 and BALB/c were purchased from Harlan (France) and Janvier (France), respectively. Mice were utilized between 8 and 16 weeks of age. All mouse experiments were performed at Gustave Roussy Cancer Campus and mice were housed in specific pathogen-free conditions.
Cell lines
MCA205 and RET melanoma (a transgene-enforced expression of the Ret proto-oncogene under the control of the metallothionein-1 promoter driving spontaneous melanomagenesis, kindly provided by Professor Viktor Umansky) (syngeneic from C57BL/6J mice) were cultured at 37°C under 5% CO2 in RPMI-1640 medium supplemented with 10% heat-inactivated fetal bovine serum (FBS), 1% penicillin/streptomycin, 2 mM L-glutamine and 1% of sodium pyruvate and non-essential amino acids (all from Gibco-Invitrogen), referred herein as complete RPMI medium. Cell lines were regularly tested for mycoplasma contamination and were not used after 10 passages.
METHOD DETAILS
Clinical assessments and biospecimens
Response assessments. Clinical response annotation was performed independently by at least two clinical investigators per patient (MCA, PAP, HT). Treatment responses were defined using the best overall response (BOR) according to RECIST 1.1 criteria33 comparing tumor burden on restaging imaging performed at standard disease re-assessment time points studies with baseline (pre-treatment) studies. Longitudinal restaging scans were evaluated throughout the period of treatment until the initiation of a subsequent line of therapy or last known follow-up date. Imaging modality was matched whenever possible, favoring contrast-enhanced CT of the chest, abdomen and pelvis, contrast-enhanced MRI or CT brain, and imaging of the neck or extremities as indicated by known sites of disease. Patients were classified as “responders” (R) if they achieved objective complete response (CR; 100% reduction in tumor burden) or partial response (PR; ≥30% reduction in tumor burden) attributable to CICB. Patients were classified as non-responders if they achieved a BOR of progressive disease (PD; ≥20% increase in disease burden) or stable disease (SD; not meeting criteria for CR/PR/PD) (Supplementary Data Table 2). Mice were defined as responders (R) if their tumors either regressed or were stable during treatment, or as non-responders (NR) when tumors increased in size over two consecutive measurements.
Toxicity assessments. Immune-related adverse events (irAE) was scored according to the NCI Common Terminology Criteria for Adverse Events (CTCAE) 4.0 criteria and immune-relatedness to CICB therapy (“possible”, “probable”, “definite” association) assigned by consensus opinion of at least two independent clinical investigators (MCA, HT, WSC). Binary toxicity classification was based on whether patients experienced any grade 3 or higher irAE versus less than grade 3 irAE (Supplementary Data Table 2).
Biospecimen collections. Available pre- and on-treatment tumor and peripheral blood samples were identified by querying institutional research biospecimen holdings and, when necessary, archival pathology holdings from diagnostic specimens. Tumor biopsies were obtained as punch, core needle, or excisional biopsies and preserved as snap-frozen (for RNA/DNA extraction) or formalin-fixed paraffin-embedded (FFPE; for immunohistochemistry or DNA extraction) specimens. Peripheral blood samples underwent density-gradient centrifugation to isolate peripheral blood mononuclear cells (PBMC) prior to cryopreservation until required for germline DNA extraction or flow cytometry. Biospecimens were retrieved, collected and analyzed under UT MD Anderson Cancer Center Institutional Review Board-approved protocols in accordance with the Declaration of Helsinki. Fecal samples were obtained on an outpatient basis after detailed in-person explanation and instruction by a treating clinician to facilitate stool capture free of water/urine using a single-use toilet insert. Fecal samples were stabilized immediately using the OMNIgene-GUT Kit (DNA Genotek Inc, Ottawa, Canada) according to the manufacturer’s recommendations, involving contact only with a provided sterile spatula. Stabilized fecal samples were returned in person or by mail to a central laboratory at The University of Texas MD Anderson Cancer Center within 30 days of collection and stored at -80ºC immediately upon receipt. For sequencing, samples were shipped as-is and in bulk to the Alkek Center for Metagenomics and Microbiome Research at Baylor College of Medicine. Patient-level sample utilization is as shown in Supplementary Data Table 3.
Colon biopsies from a cohort of ICB-treated patients taken at the time of clinical grade 3-4 colitis, and from a separate cohort of non-ICB-treated patients without gut symptoms were identified from systematic chart review, as summarized in Supplementary Data Table 7. Archival FFPE material was retrieved and freshly cut sections used to extract RNA for downstream estimation of inflammatory cytokine expression by qPCR.
Genomic analyses
Whole exome sequencing analysis. Whole-exome sequencing (WES) was performed using the same protocol as previously described15. A total of 26 pre-treatment samples were included (19R, 7NR). DNA was extracted from tumor samples after pathological assessment and confirmation of tumor content. Matched peripheral blood leukocytes were collected as germline DNA control. The initial genomic DNA input into the shearing step was 750 ng. End repair, A-base addition, adapter ligation using forked Illumina paired-end adapters, and library enrichment polymerase chain reaction (PCR) was performed using the KAPA Hyper Prep Kit (#KK8504) followed by solid-phase reverse immobilization bead cleanup and cluster generation. Library construction was performed per the manufacturer’s instructions. Target enrichment was performed using the Agilent SureSelectXT Target Enrichment (#5190-8646) protocol as per the manufacturer’s instructions, using 650-750 ng of prepared libraries. Enriched libraries were normalized to equal concentrations using an Eppendorf Mastercycler EP Gradient instrument, pooled to equimolar amounts on the Agilent Bravo B platform and quantified using the KAPA LibraryQuantification Kit (#KK4824). Pooled libraries were adjusted to 2 nM, denatured with 0.2 M NaOH, diluted using Illumina hybridization buffer, and underwent cluster amplification using HiSeq v3 cluster chemistry and the Illumina Multiplexing Sequencing Primer Kit as per manufacturer’s instructions. Pools were then sequenced on an Illumina HiSeq 2000/2500 v3 system using 76 bp paired-end reads, and analyzed using RTA v.1.13 or later. The mean coverage for exome data was 221´ in tumors and 100´ in germ line. Aligned BAM (hg19) files were then processed using Picard and GATK software to identify duplication, realignment and recalibration. Somatic point mutations were identified using MuTect (v1.1.4) and small insertions/deletions identified using Pindel (v0.2.4). Additional post-calling filters were then applied, including: (a) total read count in tumor sample > 30, (b) total read count in matched normal sample > 10, (c) VAF (Variant Allele Frequency) in tumor sample > 0.05, (d) VAF in matched normal sample < 0.01, and (e) SNVs reported in dbSNP129 and 1000 Genomes Project were removed.
Copy number alteration analysis. Copy number alteration analysis was performed as previously described15. Essentially, Sequenza (v2.1.2) algorithm was applied to the aligned BAM data to obtain the log2 copy number ratio (tumor/normal) for each tumor sample. Using R package “CNTools” (v1.24.0), copy number gain (log2 copy ratios > log21.5) and loss (log2 copy ratios < −log21.5) at the gene level were identified. The burden of copy number gain or loss was defined as the total number of genes with copy number gain or loss per sample. To define recurrent CNA, R package “cghMCR” (v1.26.0) was applied to the calculated log2 copy ratios (tumor/normal) to identify genomic regions of recurrent CNAs (minimum common regions, MCRs). To identify genes preferentially lost or gained in responders versus non-responders, Fisher’s exact test was performed at each gene location, and statistical significance was defined by FDR adjusted p<0.05. Genes with CNA in less than 3 samples were excluded.
Neoantigen prediction. Non-synonymous exonic mutations (NSEM) from WES were reviewed and all possible 8- to 12-mer peptides encompassing NSEM were used for neoantigen prediction and compared with wild type peptides. HLA of each case was predicted using PHLAT34. Binding affinity was evaluated, taking into account patient HLA, by the NetMHCpan (v2.8) algorithm35,36. Candidate peptides with a predicted IC50<500 nM were considered HLA-binding.
Immune analyses
Flow cytometry - MDACC. Peripheral blood mononuclear cells (PBMCs) obtained from the study patients were analyzed by members of the MD Anderson Immunotherapy Platform. Pre-treatment and post-treatment blood samples were drawn for immunophenotypic analysis of PBMCs. PBMC samples were available from 20 patients, including 10 patients with ≥Grade 3 irAE, and 10 patients with <Grade 3 irAE. Multiparametric flow cytometry analysis of PBMCs was performed using fluorescently conjugated monoclonal antibodies across several panels: CD4 AF532 (SK3, eBioscience), CD3 PerCP-Cy5.5 (UCHT1, Biolegend) CD8 AF700 (RPA-T8, BD Biosciences), CD127 BV711 (HIL-7R-M21, BD Biosciences), ICOS PE-Cy7 (ISA-3, eBioscience), PD-1 BV650 (EH12.1 BD Biosciences) and FOXP3 PE-e610 (PCH101; eBioscience); CD3 PE-CF594, CD4 Pe-Cy5.5, CD8 AF532, CD45RA BV650 (HI100, Biolegend), CCR7 BV785 (G043H7, Biolegend) CD27 PeCy5 (0323, eBioscience), CD28 APC-e780 (CD28.2 eBioscience), PD-1 BV650 (EH12.1 BD Biosciences), EOMES e660 (WD1928, eBioscience), and TBET BV605 (4B10 Biolegend). Live/Dead fixable yellow stain was obtained from Thermo Fisher Scientific. Samples were run using an LSR Fortessa (BD Biosciences) and analyzed using the FlowJo software program. After appropriate forward/side scatter and live single cell gating, we determined the frequency of total CD3+ T cells, CD8+ T cells (CD3+CD8+) and CD4+ T cells (CD3+CD4+). Among the CD4, CD4+ effector T cells (CD4+FOXP3-) and CD4+ regulatory T cells (CD4+FOXP3+CD127-/low). PD-1 and ICOS expression were evaluated on these populations. CD45RA and CCR7 expression on CD4 and CD8 T cells was used to define naïve, T central memory (TCM), T effector memory (TEM) and effector T (Teff) sub-populations. PD-1, CD28, CD27, EOMES and TBET expression was evaluated in each of these compartments.
Flow cytometry – MSKCC. Peripheral blood mononuclear cells (PBMC) were isolated and cryopreserved from patient whole blood samples. Flow cytometry was performed in the Immune Monitoring Facility at Memorial Sloan Kettering Cancer Center (MSKCC) to examine T cell phenotypic markers. Human peripheral blood mononuclear cells (PBMC) samples were thawed and stained with a fixable viability stain (FVS510, BD Biosciences) and a cocktail of antibodies to the following surface markers: CD45RA-BUV395 (BD, HI100), CD4-BUV496 (BD, SK3), ICOS-BUV563 (BD, DX29), CD25-BUV615 (BD, 2A3), TIM-3-BUV661 (BD, 7D3), CD27-BUV737 (BD, L128), CD8-BUV805 (BD, SK1), CD57-BV421 (BD, NK-1), CXCR5-BV480 (BD, RF8B2), CD14-BV570 (BioLegend, M5E2), CD19-BV570 (BioLegend, HIB19), CCR4-BV605 (BioLegend, L291H4), CCR7-SB645 (eBioscience, 3D12) HLA-DR-BV711 (BD, G46-6), CD3-BV750 (BD, SK7), CD28-BV786 (BD, CD28.2), PD-1-BB515 (BD, MIH4), CD127-BB700 (BD, HIL-7R-M21), CD38-BB790 (BD, HIT2), TIGIT-PE (eBioscience, MBSA43), and GITR-PE-Cy7 (eBioscience, eBioAITR), in the presence of Brilliant Stain Buffer Plus (BD). Cells were next fixed and permeabilized with the FoxP3/Ki-67 Fixation/Permeabilization Concentrate and Diluent (eBioscience), and subsequently stained intracellularly with LAG-3-BB660 (BD, T47-530), Ki-67-AlexaFluor700 (BD, B56), FoxP3-PE-Cy5.5 (eBioscience, PCH101), CTLA-4-PE-Cy5 (BD, BNI3), Eomes-PE-eFluor610 (eBioscience, WD1928), T-bet-APC (eBioscience, ebio4B10), Granzyme B-APC-Fire750 (BioLegend, QA16A02), in the presence of Brilliant Stain Buffer Plus (BD). Stained cells were acquired on a BD Biosciences FACSymphony and analyzed using FlowJo software (FlowJo, LLC).
Immunohistochemistry. A hematoxylin & eosin (H&E) stained slide from each FFPE tumor sample was obtained to confirm the presence of tumor. Heavily pigmented samples were pretreated with melanin bleaching by low concentration hydrogen peroxide. The selected antibody panel included programmed death-ligand 1 (PD-L1) clone E1L3N (1:100, Cell Signaling Technology), PD-1 clone EPR4877 (1:250, Epitomics), CD3 polyclonal (1:100, DAKO), CD4 clone 4B12 (1:80, Leica Biosystems), CD8 clone C8/144B (1:25, Thermo Scientific), FOXP3 clone 206D (1:50, BioLegend) and Granzyme B clone 11F1 (ready to use, Leica Microsystems). IHC staining of a limited antibody panel was performed using a Leica Bond Max automated stainer (Leica Biosystems, Buffalo Grove, IL). The IHC reaction was performed using Leica Bond Polymer Refine detection kit (Leica Biosystems) and diaminobenzidine (DAB) was used as chromogen. Counterstaining was with hematoxylin. All IHC slides were scanned using an Aperio AT Turbo (Leica Biosystems) prior to all downstream IHC analyses. Using the Aperio Image Toolbox analysis software (Leica Biosystems), average values for each markers from five randomly-selected 1mm2 areas within the tumor region were selected for digital analysis as previously described37. PD-L1 expression was evaluated by H-score, which evaluates the percentage of positive cells (0 to 100) and the intensity of staining (0 to 3+), with a total score ranging from 0 to 300. The remaining markers were scored as density of cells.
TCR Sequencing. DNA was extracted from available FFPE tumor tissues (19R, 6NR) and PBMC (15 patients with ≥Grade 3 irAE, and 12 patients with <Grade 3 irAE) using the QIAamp DNA FFPE Tissue Kit (Qiagen). Next generation TCR sequencing of CDR3 variable regions was performed using the ImmunoSeq hsTCRB kit (Adaptive Biotechnologies) followed by sequencing on a MiSeq 150´ (Illumina) and analysis using the ImmunoSeqTM Analyzer software v3.0 (Adaptive Biotechnologies), considering only samples for which a minimum of 1000 unique templates were detected. Clonality is an index inversely correlated with TCR diversity and was measured as 1-(entropy)/log2(# of productive unique sequences). Preferential clonal expansion was defined as the number of T cell clones significantly expanded in post-treatment compared to pre-treatment blood samples.
Murine models
Antibiotic treatments. Mice were treated with an antibiotic solution (ATB) containing ampicillin (1 mg/ml), streptomycin (5 mg/ml), and colistin (1 mg/ml) (Sigma-Aldrich), with or without the addition of vancomycin (0.25 mg/ml) via the drinking water. Solutions and bottles were replaced 3 times and once weekly, respectively. Antibiotic activity was confirmed by cultivating fecal pellets resuspended in BHI+15% glycerol at 0.1 g/ml on COS (Columbia Agar with 5% Sheep Blood) plates for 48 h at 37°C in aerobic and anaerobic conditions. The duration of ATB treatments was slightly different based on the experimental settings. In brief, mice were treated for 2 weeks prior to tumor implantation and continuously throughout the experiment in MCA205 and RET experiments, whilst in experiments where FMT were used, ATB treatment was administered for 3 days prior to fecal microbiota transfer.
Tumor challenge and treatment. Flanks of mice were subcutaneously (s.c.) injected with 0.8 × 106 MCA205 or 0.5 x 106 RET cells. Treatment commenced when tumors reached 20 to 30 mm2. Mice were injected intraperitoneally (i.p) every three days with anti-PD-1 mAb (250 μg/mouse; clone RMP1-14, 6 injections in MCA205, 5 injections in RET) and/or anti-CTLA-4 mAb (100 μg/mouse, clone 9D9, 5 injections in both MCA205 and RET) with or without anti-IL-1R antagonist (anakinra, 500 μg/mouse, injected i.p. three times per week) or respective isotype controls as indicated in figures. All mAbs for in vivo use were obtained from BioXcell (West Lebanon, NH, USA), using the recommended isotype control mAbs except anakinra (Swedish Orphan Biovitrum, Sweden).
Fecal microbiota transfer experiments. After 3 days of ATB treatment, fecal microbiota transfer (FMT) was performed using samples from healthy volunteers whose fecal shot gun sequencing analyses revealed the presence or absence of B. intestinalis. Frozen fecal samples were thawed and thoroughly vortexed. Large particulate material was allowed to settle by gravity. 200 μL of supernatant was administered in a single dose by oral gavage. An additional 100 µL was topically applied onto the fur of each animal. Two weeks after FMT, C57BL/6J mice were inoculated with 1 x 105 RET tumor cells in 100 µL PBS were injected subcutaneously. CICB began 7 days after tumor inoculation and mice were sacrificed at 24 hrs post 1st administration of anti-CTLA4+anti-PD1 ip to harvest ilea and perform qPCR for several gene products (IL-1b, TNFa, IL-6, IL-17).
Gut colonization with dedicated commensal species. Bacteroides intestinalis CSURP836 (provided by Institut hospitalo-universitaire Méditerranée Infection, Marseille, France; isolated from a human sample), B. intestinalis from everImmune (isolated from stools of a lung cancer patient prior to immunotherapy) and B. intestinalis (isolated from a mouse sample) were cultured on COS plates in anaerobic conditions using anaerobic generators (Biomerieux) at 37°C for 24 - 72 hours. Suspensions of 109 CFU/mL were obtained using a spectrophotometer (Eppendorf) at an optical density of 1 measured at 600 nm. Oral gavages of 109 CFU in 100 μL were administered 24 hours prior to antibody treatment and with each antibody treatment. Bacteria were verified using a Matrix-Assisted Laser Desorption/Ionization Time of Flight (MALDI-TOF) mass spectrometer (Microflex LT analyser, Bruker Daltonics, Germany).
Cytokine quantification. Stool samples were collected and stored at -80°C until further processing. Samples were thawed and re-suspended (at 100 mg/mL) in PBS containing 0.1% Tween 20. After a 20 min incubation with shaking at room temperature, samples were centrifuged for 10 min at 12,000 rpm and supernatants were harvested and stored at -20°C until analysis. Lipocalin-2 levels were measured using the mouse Lipocalin-2/NGAL DuoSet ELISA kit (R&D Systems, Minneapolis, MN) following the manufacturer’s instructions.
Immunohistochemistry. Gut tissue was preserved in either formalin fixed paraffin embedded (FFPE) or optimum cutting temperature compound (OCT). At mouse sacrifice the ileum and colon were removed, washed in PBS, cut longitudinally, rolled and fixed in 4% PFA overnight at 4°C or, in some experiments for 2 hours at room temperature. Tissue was then either paraffin-embedded with a Tissue-Tek® VIP® 6 Vacuum Infiltration Processor (Sakura) or rehydrated in 15% sucrose for 1h followed by 30% sucrose overnight, OCT embedded (Sakura) and snap frozen. Longitudinal sections were counterstained with hematoxylin, eosin & safran stain (H&E).
Histological assessment of gut tissue for toxicity. A scoring system was developed with a pathologist (P.O.). Ileum: Inflammatory foci, appearance of the submucosa, length of villi, and the thickness of lamina propria were scored for each section. The score was defined as: 0 = normal, 1 = focal and minor lesions; 2 = diffuse and minor lesions; 3 = diffuse, minor and major lesions; 4 = major lesions with areas containing only connective tissue. Colon: Inflammatory infiltrate, defined as either physiological (0), low (1), moderate (2) and high (3) levels were scored.
immune gene expression by real-time quantitative PCR analysis. RNA was extracted using the RNeasy Mini Kit (Qiagen) and reverse transcribed into cDNA using SuperScript III Reverse Transcriptase and the RNaseOUT™ Recombinant Ribonuclease Inhibitor (Life Technologies) using random primers (Promega, Wisconsin, United States) and the Deoxynucleoside Triphosphate Set, PCR grade (Roche, Basel, Switzerland). Gene expression was analyzed by real-time quantitative PCR (RT-qPCR) using the TaqMan method with TaqMan® Gene Expression Assays and Taqman Universal Master Mix II (Invitrogen) according to the manufacturer’s instructions on the 7500 Fast Real Time PCR system (Applied Biosystems). Expression was normalized to the expression of the housekeeping gene of β-2 microglobulin by means of the 2 −ΔCt method. The following primers were used (all from TaqMan® Gene Expression Assay, ThermoFisher): B2m (Mm00437762_m1), Il1b (Mm00434228_m1), Il6 (Mm00446190_m1), Tnf (Mm00443258_m1), IL1B (Hs01555410_m1), B2M (Hs00187842_m1), IL17A (Hs00174383_m1), TNF (Hs00174128_m1)
Microbiome studies
Patient fecal sample collection. Baseline stool samples were collected using the OMNIgene GUT kit (DNA Genotek, Ottawa, Canada). A total of 54 stool samples were subject to bacterial 16S rRNA gene sequencing, including a cutaneous/unknown primary cohort (29R, 11NR; 24 with ≥Gr3 irAE, 16 with <Gr3 irAE), and for toxicity analyses only, a mucosal cohort (3 with and 5 without ≥Gr3 irAE) and a uveal melanoma cohort (2 with and 4 without ≥Gr3 irAE. Within this cohort, a number of samples obtained early after initiation of CICB were included as surrogate baseline samples, as our parallel study on longitudinal samples collected from patients undergoing immune checkpoint blockade monotherapies showed no significant change in fecal microbiota early after treatment initiation21.
Human fecal bacterial DNA extraction Preparation and sequencing of the human fecal samples was performed in collaboration with the Alkek Center for Metagenomics and Microbiome Research (CMMR), Baylor College of Medicine using methods adapted from the NIH-Human Microbiome Project38,39. Extended details of the analytical pipeline have been reported previously21. Briefly, bacterial genomic DNA extracted using the MO BIO PowerSoil DNA Isolation Kit (MO BIO Laboratories, USA) underwent PCR amplification of the 16S rRNA gene V4 region (2 x 250 bp) and was sequenced using the MiSeq platform (Illumina, Inc, San Diego, CA).
Processing of 16S rRNA gene sequences. Quality filtered sequences with >97% identity were clustered into Operational Taxonomic Units (OTUs) and classified phylogenetically against the NCBI 16S ribosomal RNA sequence database (release date September 1, 2018) using the NCBI-BLAST+ package 2.8.1. 2018). The pipeline involves the following steps:
- The fastq_mergepairs command within VSEARCH40 was used to merge paired-end reads, with a maximum of 10 mismatches to create consensus sequences, followed by dereplication using the derep_fulllength command, sorting by decreasing length (sortbylength command; 200 to 350 bp), and sorting by decreasing cluster size of representative sequences (sortbysize command, minimum 2)
- OTU clustering, selection, and exclusion of chimeras (97%) was done using the cluster_otus command through the UPARSE41 algorithm within USEARCH42
- Representative OTU sequences were then classified using the NCBI 16S database with BLAST (Basic Local Alignment Search Tool). This step was done in R using the blastn UNIX executable and served as the database against which the original merged reads were mapped. At the species level, only OTUs with an unambiguous assignment were classified, whereas all others were annotated as ‘unclassified’ (Supplementary Data Table 4)
- Next the usearch_global command was used to query the database of merged reads for high identity hits using the previously generated representative OTU sequences as reference. The identity threshold used for this step was 0.97. The mapped OTUs were converted into an OTU table using a series of python scripts summarized in uc2otutab.py
- Microbiome indices to estimate of alpha and beta diversity was calculated in QIIME43. In order to estimate the phylogenetic distances among OTUs, sequences were first aligned by the PyNAST44 method using the py command. filter_alignment.py was then used to filter the sequence alignment by removing the highly variable regions
- Next, the py script was used to create the phylogenetic tree from multiple sequence alignment and the beta_diversity.py script was used to estimate beta diversity using Bray-Curtis dissimilarity, Weighted and Unweighted UniFrac distance matrices45.
- In order to estimate alpha diversity, the OTU table was first rarefied using the single_rarefaction.py command in QIIME. The rarefaction cutoff used was the total read count for the sample with the least number of reads. The alpha_diversity.py script in QIIME was then used to estimate alpha diversity
Whole metagenome shotgun sequencing (WMS). DNA extracted for 16S rRNA gene sequencing was also used for WMS to minimize biases introduced in the extraction process. The sequencing was done at CosmosID where samples were quantified using Qubit4 and individual sequencing libraries were prepared using proprietary methods. Pooled libraries were sequenced on the Illumina NextSeq 550 platform in a 300-cycle run. Raw FASTQ files were made available through CosmosID’s client portal and annotated taxonomically using metaphlan246 following exclusion of host reads with kneaddata.
Statistical assessment of microbial biomarkers using LEfSe. The LEfSe method was used to compare abundances of all bacterial clades according to response (i.e.: between R versus NR) and by occurrence of toxicity (i.e.: between patients with ≥Grade 3 irAE versus those with <Grade 3 irAE) using the Kruskal-Wallis test (statistical significance was defined as p<0.05)47. Bacterial taxa with differential abundance between study groups were used as input for the linear discriminant analysis (LDA) to calculate an effect size. LEfSe analysis for murine taxa was performed with Mothur v1.39.5.
Mouse fecal sample collection, DNA extraction and microbiota characterization. At least two longitudinal stool samples were collected from mice (n=71) and stored at -80°C until DNA extraction. Preparation and sequencing of mouse fecal samples was performed at IHU Méditerranée Infection, Marseille, France. Briefly, DNA was extracted using two protocols. The first protocol consisted of physical and chemical lysis, using glass powder and proteinase K respectively, then processing using the Macherey-Nagel DNA Tissue extraction kit (Duren, Germany)48. The second protocol was identical to the first protocol, with the addition of glycoprotein lysis and de-glycosylation steps49. The resulting DNA was sequenced, targeting the V3–V4 regions of the 16S rRNA gene as previously described50. Raw FASTQ files were analyzed with Mothur pipeline v.1.39.5 for quality check and filtering (sequencing errors, chimerae) on a Workstation DELL T7910 (Round Rock, Texas, United States). Raw reads (15512959 in total, on average 125104 per sample) were filtered (6342281 in total, on average 51147 per sample) and clustered into Operational Taxonomic Units (OTUs), followed by elimination of low-populated OTUs (till 5 reads) and by de novo OTU picking at 97% pair-wise identity using standardized parameters and SILVA rDNA Database v.1.19 for alignment. In all, considering RET and MCA samples, 427 bacterial taxa were identified using a prevalence threshold of ≥20% (i.e. present in at least 20% of samples). Sample coverage was computed with Mothur and resulted to be on average higher than 99% for all samples, thus meaning a suitable normalization procedure for subsequent analyses. Bioinformatic and statistical analyses on recognized OTUs were performed with Python v.2.7.11. The most representative and abundant read within each OTU (as evidenced in the previous step with Mothur v.1.39.5) underwent a nucleotide Blast using the National Center for Biotechnology Information (NCBI) Blast software (ncbi-blast-2.3.0) and the latest NCBI 16S Microbial Database accessed at the end of April 2019 (ftp://ftp.ncbi.nlm.nih.gov/blast/db/). A matrix of bacterial relative abundances was built at each taxonomic level (phylum, class, order, family, genus, species) for subsequent multivariate statistical analyses.
Mouse microbiota and OTU-level analyses. For mouse experiments, raw data were firstly normalized then standardized using QuantileTransformer and StandardScaler methods from Sci-Kit learn package v0.20.3. Normalization using the output_distribution='normal' option transforms each variable to a strictly Gaussian-shaped distribution, whilst the standardization results in each normalized variable having a mean of zero and variance of one. These two steps of normalization followed by standardization ensure the proper comparison of variables with different dynamic ranges, such as bacterial relative abundances, tumor size, or colonic infiltrate score. Measurements of α diversity (within sample diversity) such as observed_otus and Shannon index, were calculated at OTU level using the SciKit-learn package v.0.4.1. Exploratory analysis of β-diversity (between sample diversity) was calculated using the Bray-Curtis measure of dissimilarity calculated with Mothur and represented in Principal Coordinate Analyses (PCoA), while for Hierarchical Clustering Analysis (HCA) ‘Bray-Curtis’ metrics and ‘complete linkage’ method were implemented using custom scripts (Python v.2.7.11). We implemented Partial Least Square Discriminant Analysis (PLS-DA) and the subsequent Variable Importance Plot (VIP) as a supervised analysis wherein the VIP values (order of magnitude) are used to identify the most discriminant bacterial species among tumor-bearing and tumor-free mice, and among the different timepoints (T0, T2, T5). As depicted in Figure 2e, bar thickness reports the fold ratio (FR) value of the mean relative abundances for each species among the two cohorts whilst not applicable (NA) refers to comparisons with a group with zero relative abundance. An absent border indicates mean relative abundance of zero in the compared cohort(s). In order to compare the microbiota taxa with gene expression datasets or tumor size and colonic toxicity, a multivariate statistical Spearman (or Pearson for mouse data) correlation analysis (and related P values) was performed with custom Python scripts. Mann-Whitney U and Kruskal-Wallis tests were employed to assess significance for pairwise or multiple comparisons, respectively, considering a p-value <0.05 as significant.
Pairwise comparisons of relative abundances between taxa identified within patient samples were performed using Mann-Whitney tests followed by bootstrapping with 1000 permutations. Only taxa that were present in at least 40% of all samples were considered. Rarefaction limits for the calculation of alpha diversity were set based on the least number of reads in all fecal samples. Taxonomic alpha-diversity of patient samples was estimated using the Inverse Simpson Index calculated as D= (pi is the proportion of the total species S that is comprised by the species i)51, and additional diversity metrics as indicated in figures. Correlations between relative abundance of candidate taxa and peripheral immune markers were estimated using Spearman’s rho. ANalysis Of SIMilarity (ANOSIM, which represents the difference of datasets’ centroids) or, when indicated, Pearson correlation coefficient, were computed with Python 2.7.11.
Quantification of bacteria in fecal samples by qPCR. Genomic DNA was extracted from fecal samples using the QIAamp DNA Stool Mini Kit (Qiagen) following the manufacturer’s instructions. Targeted qPCR systems were applied using either TaqMan technology (for systems targeting All Bacteria domain) or SYBR Green for different Bacteroides species. The following primers and probes were used:
Target
|
PCR system
|
Primers and probes
|
Oligo sequence
|
Reference
|
All bacteria
|
TaqMan
|
Forward
|
CGGTGAATACGTTCCCGG
|
52,53
|
Reverse
|
TACGGCTACCTTGTTACGACTT
|
Probe
|
6 FAM -CTT GTA CAC ACC GCC CGT C-MGB
|
B. intestinalis
|
SYBR Green
|
Forward
|
AGCATGACCTAGCAATAGGTTG
|
54
|
Reverse
|
ACGCATCCCCATCGATTAT
|
B. uniformis
|
SYBR Green
|
Forward
|
TCTTCCGCATGGTAGAACTATTA
|
55
|
Reverse
|
ACCGTGTCTCAGTTCCAATGTG
|
B. fragilis
|
SYBR Green
|
Forward
|
TGATTCCGCATGGTTTCATT
|
54
|
|
Reverse
|
CGACCCATAGAGCCTTCATC
|
|
|
|
|
|
|
|
|
Statistical analyses. Data analyses and representations were performed either with the R software (http://www.R-project.org/), Microsoft Excel (Microsoft Co., 436 Redmont, WA, US) or Prism 5 (GraphPad, San Diego, CA, USA). Patient cohort survival curves were generated using the R package “survival”56. Between-group comparisons of patient cohort genomic and immune parameters were performed using unpaired Mann-Whitney U tests or Fisher’s exact test in the case of low-sample dichotomous variables, taking p<0.05 as statistically significant. All comparisons were two-sided unless a strong a priori hypothesis warranted a one-sided approach (indicated where appropriate). Permutation testing was performed by randomly permuting sample labels for a total of 1000 iterations. Multivariable logistic regression models were built using the best subsets approach to adjust for the effect of clinical prognostic variables. Separate models were built for response and toxicity outcomes and for each model, bacterial candidates identified during the taxonomic discovery phase were considered primary predictors. Abundances estimated from WMS were used as input. All patients were categorized as high or low for a bacterial candidate based on the median relative abundance. We allowed a maximum of two other clinical covariates (given constraints of event rates) from among age at entry, sex, BRAF mutation status (wild type vs mutant), AJCC stage (Stage III and IV vs Stage I and II), baseline LDH (high vs low), and melanoma subtype (uveal/mucosal vs cutaneous).
In murine studies, statistical analyses gathering more than two groups were performed using ANOVA followed by pairwise comparisons with Bonferroni adjustments. Differential enrichment analyses in murine studies were corrected for multiple hypothesis testing using FDR at 10% two-stage Benjamini-Hochberg. ANOSIM and PLS-DA p-values were automatically calculated after 999 permutations. Otherwise, for two groups, statistical analyses were performed using the unpaired t-test. Outliers within a given distribution were tested using Grubbs’ test (https://graphpad.com/quickcalcs/Grubbs1.cfm) with a threshold at p<0.05. All tumor growth curves were analyzed using software developed in Professor Guido Kroemer’s laboratory and information about statistical analyses can be found at this following link: https://kroemerlab.shinyapps.io/TumGrowth/ 57. Briefly, for longitudinal analyses, original tumor measurements were log transformed before statistical testing. When complete regressions of tumors were observed, zeros were imputed by the minimum value divided by 2. An automatic outlier detection at p<0.1 was retained, both for the longitudinal analyses and the Kaplan-Meier curves. Survival curves were estimated using the Cox regression and the multiple testing was taken account using the Bonferroni adjustment. p-values were two-sided with 95% confidence intervals and were considered significant when p<0.05. Symbol significance: *p<0.05, **p<0.01, ***p<0.001.