MICROBIOTA COMPOSITION, AS A MARKER OF RESPONSIVENESS TO ANTI-PD1/PD-L1/PD-L2

Information

  • Patent Application
  • 20220056532
  • Publication Number
    20220056532
  • Date Filed
    September 27, 2019
    4 years ago
  • Date Published
    February 24, 2022
    2 years ago
Abstract
The present invention relates to a method for in vitro determining if an individual having a renal cell cancer (RCC) is likely to respond to a treatment with an anti-PD1/PD-L1/PD-L2 Ab-based therapy, based on the analysis of the microbiota present in a stool sample from said individual. Twelve models useful to perform the above method are disclosed, as well as tools designed to easily perform this method.
Description
FIELD OF THE INVENTION

The present invention relates to the field of anticancer treatment. In particular, the present invention concerns the role of the gut microbiota in the efficacy of treatments comprising administration of immune checkpoint inhibitors (ICI), in the treatment of renal cell cancer (RCC). The present invention provides “metagenomics-based gut oncomicrobiome signatures” (GOMS) at diagnosis prior to PD-1/PDL-1/PDL-2 blockade as novel predictors of response or resistance for the best clinical outcome and at 6 months of therapy of a renal cell cancer. The present invention also provides theranostic methods to identify patients in need of a bacterial compensation treatment before receiving an ICI and/or during the therapy with an ICI.


BACKGROUND AND PRIOR ART

Major conceptual advances in cancer biology have been made over the past decade. The understanding that immune responses are routinely generated against tumor-specific neoantigens expressed by cancer-associated mutations and commonly dampened by the immunosuppressive tumor microenvironment (TME) has been seminal to the development of effective immunotherapies aimed at provoking immune control against tumor progression (Sharma and Allison, 2015a, 2015b). Progress in cancer immunotherapy has resulted in remarkable success in the treatment of a variety of hematological and solid metastatic malignancies such as melanoma, lung, bladder, kidney, Hodgkin's lymphoma, acute B cell leukemia, liver, Merkel-cell carcinoma and head and neck tumors, amongst others (Borghaei et al., 2015; Nghiem et al., 2016; Robert et al., 2011; Rosenberg et al., 2016). To date, therapies that block inhibitory signaling pathways expressed by T lymphocytes (so called “immune checkpoints”) during the initial priming phase (in draining lymph nodes) or the effector phases (in tumor beds) of adaptive anticancer immune responses have demonstrated the greatest clinical benefit in overall survival (Ribas et al., 2016; Topalian et al., 2012). The prototypic example for this success has been the use of monoclonal antibodies (mAbs) targeting PD-1 (expressed by activated/exhausted T cells) or its ligand PD-L1 (commonly expressed by cancer cells or cells of the TME) (Pardoll, 2015). By releasing these molecular brakes, such mAbs reinstate the anticancer adaptive arm of the immune response.


While PD-1 blockade represents the most effective first line therapy in B-RAF wild type melanoma and the best option in second line unresectable lung carcinomas, approximately 60-70% of tumors exhibit primary resistance to this therapeutic strategy. Primary resistance has been attributed to low tumor mutational burden and poor intrinsic antigenicity of tumor cells (Riaz et al., 2016; Rizvi et l., 2015), defective antigen presentation and priming phase (Spranger et al., 2015), limited intratumoral infiltration related to exhausted T cell functions (Smyth et al., 2016), and metabolic immunosuppressive pathways related to adenosine and indoleamine, 2, 3-dioxygenase (Koyama et al., 2016; Smyth et al., 2016). Similarly, primary resistance mechanisms to CTLA-4 blockade have also been elucidated. For example, melanoma presenting with loss of IFN-γ signaling lack response to ipilimumab (Gao et al., 2016).


More recently, secondary resistance mechanisms to chronic inhibition of PD-1 receptors have been reported in approximately 25% of melanoma patients (Koyama et al., 2016; Ribas et al., 2016; Zaretsky et al., 2016). Four out of 74 melanoma patients who progressed for a median follow up of 1.8 years despite continuous therapy with pembrolizumab developed lesions presenting with loss-of-function mutations in janus kinases JAK1 or JAK2, thus leading to decreased STAT1 phosphorylation and reduced sensitivity to the antiproliferative effects of IFNs or alternatively, a mutation within the gene encoding β-2 microglobulin, preventing folding and transport of MHC class I molecules to the cell surface for T cell recognition of tumor cells (Zaretsky et al., 2016). In addition to these darwinian natural selection of genetic (or epigenetic) heritable traits, other acquired resistance mechanisms have been reported in mice. Predominately, adaptive immune resistance resulting from the IFN-γ-inducible expression of PD-L1 (Pardoll, 2012; Taube et al., 2012), a primary ligand of PD-1, TNF-induced loss of antigenic variants (Landsberg et al., 2012) as well as TCR-dependent upregulation of additional exhaustion markers on activated T lymphocytes such as Tim3/HAVCR2 (Koyama et al., 2016; Restifo et al., 2012; Smyth et al., 2016), lymphocyte activation gene 3 (Lag3), T cell immunoreceptor with Ig and ITIM domains (TIGIT), B and T cell lymphocyte attenuator (BTLA), and V-domain Ig suppressor of T cell activation (VISTA).


The inventors' team just reported in Routy et al. Science Jan. 5, 2018, in conjunction with two other reports (Gopalakrishnan V et al and Matson V et al), that primary resistance to PD-1/PDL-1-based immune checkpoint inhibitors (ICI) can be due to an abnormal gut microbiota. Antibiotics (ATB) inhibited the clinical benefit of ICI in advanced lung, kidney and bladder cancer patients. Fecal microbiota transplantation (FMT) from cancer patients who responded to ICI (but not from non-responding patients) into germ-free or ATB-treated mice ameliorated the antitumor effects of PD-1 blockade. Metagenomics of patient stools at diagnosis revealed correlations between clinical responses to ICI and dedicated microbial patterns, with relative increase of Akkermansia muciniphila when examining both lung and kidney cancer patients altogether. The diagnosis of a gut dysbiosis is important since it is amenable to a therapy that restores efficacy of ICI. Indeed, oral supplementation with A. muciniphila or Alistipes indistinctus post-FMT with non-responder feces restored the efficacy of PD-1 blockade in an IL-12-dependent manner, by increasing the recruitment of CCR9+CXCR3+CD4+T lymphocytes into tumor beds.


The results disclosed in the present application show the predictive value of metagenomics-based gut oncomicrobiome signatures (GOMS), calculated at diagnosis from data of a larger cohort of 69 renal cell cancer patients (RCC) patients who received a second line therapy with nivolumab or pembrolizumab for a tyrosine kinase inhibitor or mTOR inhibitor-resistant advanced or metastatic RCC, for the clinical benefit of such a treatment. This study was performed using three RECIST criteria (best outcome, time to progression (TTP) at 3 months, TTP at 6 months), including or excluding 11 RCC patients who took antibiotics during or the 2 months preceeding the first administration of anti-PD1 Abs.


SUMMARY OF THE INVENTION

According to a first aspect, the present invention pertains to a method for in vitro determining if an individual having a renal cell cancer patients (RCC) is likely to respond to a treatment with an anti-PD1/PD-L1/PD-L2 Ab-based therapy, comprising the following steps:

  • (i) from a fecal sample (or ileal or colonic mucosal specimen) of said individual, obtaining an abundances pattern based on the relative abundances of a set of bacterial species comprising at least 8 bacterial species selected from the group of species disclosed in Table 1 below.
  • (ii) using one or several pre-defined equations each corresponding to a model obtained for at least 8 bacterial species from said set of bacterial species, calculating the probability that said individual responds to the treatment (PR) or the probability that said individual resists to the treatment (PNR) with an anti-PD1/PD-L1/PD-L2 Ab-based therapy.


The invention also pertains to 12 models useful to perform the above method, 6 of which were obtained from data of patients who did not take any antibiotics during the last two months, whereas the 6 others can be used in patients whom recent antibiotics uptake is unknown. These models rely on (partly) different subsets of the microbiota (listed in Tables 3, 6, 9 and 12 below) and can be used either alone or combined.


Tools designed to easily perform the above method are also part of the present invention, such as a nucleic acid microarray comprising nucleic acid probes specific for each of the microorganism species to be detected in step (i) of the method, and such as a set of primers comprising primer pairs for amplifying sequences specific for each of the microorganism species to be detected in step (i) of said method.


Theranostic methods for determining whether an individual needs a bacterial compensation with a bacterial composition and/or by FMT before receiving an anti-PD1/PD-L1 Ab-based therapy are also part of the invention.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1. Commensal species associated with best outcome.


The RECIST 1.1 criterium taken into account as predictive for clinical benefit was “best clinical outcome”.


A. Volcano plots (left) and Linear discriminant analysis effect size (LEfSe) (right) analysis to assess putative bacterial biomarkers for building metagenomics-based GOMS in RCC patients' stools regardless antibiotic usage. Volcano plots were generated computing for each bacterial species: i) the log 2 of fold ratio (FR) among the mean relative abundances of R versus NR (x axis); ii) the co-log 10 of P values deriving from Mann-Whitney U test calculated on relative abundances in absolute values (y axis). LEfSe plots were generated with Python 2.7 on output files derived from LEISe pipeline, and all species with LDA score≥2 were considered for subsequent analysis.


B. ROC curves to assess predictability of metagenomic-based GOMS in RCC patients' stools regardless antibiotic usage. Patients were divided into NR and R according to their «resistant» or «responding» clinical phenotype upon ICI treatment. Combinations of selected bacterial species in panel A were performed with Python 2.7 and underwent ROC analysis. Upon 5-fold cross-validation (no noise added), the ROC curve corresponding to the bacterial species consortium having the best AUC was depicted. Specificity (x axis) and sensitivity (y axis) along with their best values were reported for each curve (inset). Diagonal line depicts the absence of predictability for the best clinical outcome.


C-D, Idem as A-B but considering only patients who did not take ATB.



FIG. 2. Commensal species associated with TTP>3 months.


The RECIST 1.1 criterium taken into account as predictive for clinical benefit was “TTP< or >3 months”.


A. Volcano plots (left) and LEfSe (right) analysis to assess putative bacterial biomarkers for building metagenomics-based GOMS in RCC patients' stools regardless antibiotic usage. Volcano plots were generated computing for each bacterial species: i) the log 2 of fold ratio (FR) among the mean relative abundances of R versus NR (x axis); ii) the co-log 10 of P values deriving from Mann-Whitney U test calculated on relative abundances in absolute values (y axis). LEfSe plots were generated with Python 2.7 on output files derived from LEfSe pipeline, and all species with LDA score 2 were considered for subsequent analysis.


B. ROC curves to assess predictability of metagenomic-based GOMS in RCC patients' stools regardless antibiotic usage. Patients were divided into NR and R according to their «resistant» or «responding» clinical phenotype upon ICI treatment. Combinations of selected bacterial species in panel A were performed with Python 2.7 and underwent ROC analysis. Upon 5-fold cross-validation (no noise added), the ROC curve corresponding to the bacterial species consortium having the best AUC was depicted. Specificity (x axis) and sensitivity (y axis) along with their best values were reported for each curve (inset). Diagonal line depicts the absence of predictability for the best clinical outcome.


C-D. Idem as A-B but considering only patients who did not take ATB.



FIG. 3. Commensal species associated with TTP>6 months.


The RECIST 1.1 criterium taken into account as predictive for clinical benefit was “TTP< or >6 months”;


A. Volcano plots (left) and LEfSe (right) analysis to assess putative bacterial biomarkers for building metagenomics-based GOMS in RCC patients' stools regardless antibiotic usage. Volcano plots were generated computing for each bacterial species: i) the log 2 of fold ratio (FR) among the mean relative abundances of R versus NR (x axis); ii) the co-log 10 of P values deriving from Mann-Whitney U test calculated on relative abundances in absolute values (y axis). LEfSe plots were generated with Python 2.7 on output files derived from LEfSe pipeline, and all species with LDA score≥2 were considered for subsequent analysis.


B. ROC curves to assess predictability of metagenomic-based GOMS in RCC patients' stools regardless antibiotic usage. Patients were divided into NR and R according to their «resistant» or «responding» clinical phenotype upon ICI treatment. Combinations of selected bacterial species in panel A were performed with Python 2.7 and underwent ROC analysis. Upon 5-fold cross-validation (no noise added), the ROC curve corresponding to the bacterial species consortium having the best AUC was depicted. Specificity (x axis) and sensitivity (y axis) along with their best values were reported for each curve (inset). Diagonal line depicts the absence of predictability for the best clinical outcome.


C-D. Idem as A-B but considering only patients who did not take ATB.



FIG. 4. TTP6 months allows to segregate R versus NR RCC patients who did not take ATB based on MG composition.


All RCC patients who did not take ATB from the Routy et al. Science 2018 data base were considered unifying discovery and validation cohorts at TTP6. Alfa-diversity, beta-diversity, Volcano and LEfSe plots were generated as described in FIGS. 1, 2, 3 and 4. Pairwise analysis was performed on NR (orange) and R (blue) cohorts and reported are boxplots generated with Python v2.7 only for significant comparisons (Mann-Whitney U test, P≤0.05). Responders tend to have significantly higher alfa-diversity compared to NR, and are significantly divided from NR (beta-diversity, P values are indicated within the ordination plots). LEfSe, volcano and pairwise analysis plots highlights discriminant species within the two cohorts NR and R.



FIG. 5. Logistic regression models based on RCC patients regardless antibiotic usage and OUTCOME_1 criterion (OUTCOME_1 meaning considering all SD among responders).


Logistic regression models were drawn from stool MG of RCC patients regardless antibiotic usage and OUTCOME_1 criterion. Each Model is depicted by three graphs: a feature selection (left), a confusion matrix (center) and a ROC curve (right). Confusion matrices and ROC curves were generated after 5-fold cross-validation. The higher the percentage within the crossing ‘True label’ and ‘Predicted label’ cells, the higher the model predictability. For the ROC curves, both AUC (area under curve) and CV_AUC (cross-validated area under curve) need to be taken into account. At the bottom of each triplet is reported the Model equation described by a logistic regression: exp is the exponent with natural base, CAG is the relative abundance of each CAG species expressed within the closed interval [0:1] and standardized (zero mean and unit variance).



FIG. 6. Logistic regression models based on RCC patients without antibiotic treatment and OUTCOME_1 criterion (OUTCOME_1 meaning considering all SD among responders).


Logistic regression models were drawn from stool MG of RCC patients with no antibiotic usage and OUTCOME_1 criterion. Triplet graph (feature selection, confusion matrix, ROC curve) description as in FIG. 6.



FIG. 7. Logistic regression models based on RCC patients regardless antibiotic usage and OUTCOME_2 criterion (OUTCOME_2 meaning considering only SD>6 months among responders).


Logistic regression models were drawn from stool MG of RCC patients regardless antibiotic usage and OUTCOME_2 criterion. Triplet graph (feature selection, confusion matrix, ROC curve) description as in FIG. 6.



FIG. 8. Logistic regression models based on RCC patients without antibiotic treatment and OUTCOME_2 criterion (OUTCOME_2 meaning considering only SD>6 months among responders).


Logistic regression models were drawn from stool MG of RCC patients with no antibiotic usage and OUTCOME_2 criterion. Triplet graph (feature selection, confusion matrix. ROC curve) description as in FIG. 6.





DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS

In the present text, the following definitions are used:

    • An “immune checkpoint inhibitor” (ICI) designates any drug, molecule or composition which blocks certain proteins made by some types of immune system cells, such as T cells, and some cancer cells. These proteins help keep immune responses in check and can keep T cells from killing cancer cells. When these proteins are blocked, the “brakes” on the immune system are released and T cells are able to kill cancer cells better. Examples of checkpoint proteins found on T cells or cancer cells include PD-1/PD-L1 and CTLA-4/B7-1/B7-2. In particular, ICIs encompass anti-PD1 antibodies (such as Nivolumab or Pembrolizumab), anti-PD-L1 antibodies (such as Atezolizumab or Durvalumab), anti-CTLA-4 antibodies and anti-PD-L2 antibodies. In the scientific literature, ICIs are also designated as “drugs blocking an immune checkpoint”, or “immune checkpoint blockers” or “immune checkpoint blockade drugs”.
    • An “anti-PD1/PD-L1/PD-L2 Ab-based therapy” herein designates any therapy including the use of a drug that antagonizes PD1, PD-L1 or PD-L2. These include therapies mainly based on an ICI such as a drug antagonizing PD1 or PD-L1 or PD-L2, as well as combined therapies using several ICIs and/or additional anticancer drugs such as chemotherapeutic drugs. Non-limitative examples of combined therapies encompassed by the phrase “anti-PD1/PD-L1/PD-L2 Ab-based therapy” include anti-PD1+anti-CTLA4, anti-PD1+polychemotherapy (pemetrexed+carboplatin), anti-Lag3+anti-PD1, anti-NKG2A+anti-PD1, IDO inhibitor+anti-PD1 and anti-ICOS+anti-PD1. Although the currently used drugs antagonizing immune checkpoint proteins are monoclonal antibodies, other molecules specifically binding to PD1, PD-L1, PD-L2 or other proteins could be used for the development of future ICIs such as, for example, antibody fragments or specifically designed aptamers. Of course, the phrase “anti-PD1/PD-L1/PD-L2 Ab-based therapy” encompasses any therapy including an active molecule that antagonizes PD1 or PD-L1 or PD-L2.
    • “NR” defines a non-responder status to PD-1/PDL-1/PDL-2 blockade
    • “R” defines a responder status to PD-1/PDL-1/PDL-2 blockade
    • “NGS” defines any Next Generation Sequencing platform available in the past, present or in the future.
    • In the present text, each “bacterial species” is defined by a Co-Abundance gene Group (“CAC”), which is a group of bacterial genes from the gut microbiome (i.e., the gene repertoire of the gut microbiota), which abundance level varies in the same proportion among different individual samples. In other words, a bacterial species according to the invention is defined by a cluster of bacterial gene sequences which abundance levels in samples from distinct subjects are statistically linked rather than being randomly distributed.


Most current approaches for analyzing metagenomic data rely on comparisons to reference genomes, but the human gut microbiota diversity extends beyond what is currently covered by reference databases. In the results disclosed herein, the inventors used a method based on binning co-abundant genes across a series of metagenomic samples, that enables comprehensive discovery of new microorganisms without the need for reference sequences. In what follows, part of the species identified as likely to play a role in the patients' response to therapies based on antibodies against PD1, PD-L1 or PD-L2 are newly-identified species, not yet precisely referenced in public databases. For each of the identified species (both newly-identified and species very close to already referenced species), the present application discloses a set of 50 bacterial genes which are non-redundant sequences and can be used, alone or in combination, as tracer genes to assess the presence and relative abundance to the corresponding species. Of course, once the species are identified, either by the set of non-redundant genes disclosed herein, or later on by their further identification and/or inclusion into a data base, the skilled in the art can assess their relative abundance by any appropriate means, such as, for example, by measuring the copy number of another non-redundant gene that co-varies with the 50 sequences disclosed in the present application, or even by identifying a signature of this species at the protein level rather than in a nucleic acids sample. Hence, the present invention is not limited to the use of the disclosed sequences to measure the relative abundance of the corresponding species.

    • The “relative abundance” of a definite bacterial is defined as the fraction of the entire bacterial ecosystem belonging to this bacterial species. Throughout the present text, all relative abundances are expressed within the closed interval [0:1], where 1 stands for the maximum fraction available for a single bacterial species (i.e., a bacterial species with a relative abundance equal to 1 means that 100% of the bacteria present in the sample are of the considered species). For NGS technique, the relative abundance of a bacterial species is considered as the number of reads of that selected species divided by the total number of reads representing the overall bacterial community. For the qPCR technique, the relative abundance of a bacterial species is considered as the ΔCt value of that species X (amplified by a pair of primers specific for X) divided by the ΔCt value of the total bacteria (amplified by an universal primers pair able to catch all the eubacteria present in a sample, the pair consisting of primers PRK341F and PRK806R or the pair consisting of primers 27F and 1492R).
    • “predictability” represents the predictability of the overall model done with logistic regression, based on selected bacterial species. Predictability is expressed as a percentage value within the closed interval [0:100].
    • “success rate” is the percentage of subjects recognized as NR or R after 5-fold cross-validation, and is different from model predictability.


When necessary, other definitions are provided later in the present text.


The present invention concerns a method for in vitro determining if a subject having a renal cell cancer is likely to benefit from a cancer treatment with an ICI and more specifically, from a treatment with antibodies (or other inhibiting molecules) directed against immune checkpoint blockers PD1, PD-L1 or PD-L2, alone or together with CTLA4 and/or other drugs as defined above. The responder (NR) or responder (R) status is established following these steps:


(i) from a fecal sample of the said subject (or from an ileal or colonic mucosal specimen), obtaining an “abundances pattern” based on the relative abundances of a set of bacterial species, expressed within the closed interval [0:1];


(ii) using the obtained abundances pattern to calculate, using one (or several) of the complementary models proposed in the present application, the probability that the subject will not respond (NR) or respond (R) to the treatment.


It is important to note here that, as already mentioned above, the present invention is not limited by the technique used to measure the relative abundances of the bacterial species, which can be obtained by NGS (through any past or future NGS platform, from the first generation to the last available on the market and those in development, using any NGS output file provided as fastq, BAM, SAM, or other kind of files extensions) or any other technique such as, for example, qPCR (quantitative polymerase chain reaction) and microarrays to express the relative abundances of selected bacterial species, using the sequences provided herein or any other co-variant sequence. When the relative abundances are assessed by genetic analysis, the data preferably derive from shotgun sequencing, and not 16S targeted sequencing, in order to comply with the bioinformatic pipeline described in Materials and Methods below.


According to a first aspect, the present invention pertains to a method for in vitro determining if an individual having a renal cell cancer (RCC) is likely to respond to a treatment with an ICI-based therapy such as an anti-PD1/PD-L1/PD-L2 Ab-based therapy, comprising the following steps:

    • (i) from a fecal sample of said individual, obtaining an abundances pattern based on the relative abundances of a set of bacterial species comprising at least 8 bacterial species selected from the group consisting of:









TABLE 1







Bacterial species used to assess the R or NR status of a RCC patient. The


third column specifies, for each bacterial species, if its presence/overrepresentation


in the fecal sample is of good (G) or bad (B) prognosis










CAG number
Bacterial species















CAG00008

Clostridium_bolteae_ATCC_BAA_613

B



CAG00013
Clostridiales_bacterium_1_7_47FAA
B



CAG00037

Bacteroides_faecis_MAJ27

G



CAG00048_1

Clostridium_sp_CAG_226

B



CAG00063

Barnesiella_viscericola_DSM_18177

B



CAG00122

Coprococcus_catus_GD_7

B



CAG00140

Subdoligranulum_sp_4_3_54A2FAA

B



CAG00142

Bacteroides_stercoris_ATCC_43183

B



CAG00211
Firmicutes_bacterium_CAG_227
G



CAG00218

Bacteroides_sp_CAG_20

B



CAG00243
Lachnospiraceae_bacterium_1_1_57FAA
B



CAG00300

Prevotella_sp_CAG_891

B



CAG00317

Clostridium_sp_CAG_230

G



CAG00327

Faecalibacterium_sp_CAG_74

B



CAG00341

Eubacterium_sp_CAG_115

B



CAG00346

Eubacterium_rectale_M104_1

B



CAG00357

Bacteroides_ovatus_V975

B



CAG00413

Bacteroides_sp_CAG_144

G



CAG00473

Prevotella_sp_CAG_617

B



CAG00474

Sutterella_wadsworthensis_2_1_59BFAA

G



CAG00487

Prevotella_sp_CAG_279

B



CAG00508

Alistipes_obesi

G



CAG00530

Prevotella_sp_CAG_617

G



CAG00557

Ruminococcus_callidus_ATCC_27760

B



CAG00580

Clostridium_sp_CAG_62

B



CAG00601
uncultured_Faecalibacterium_sp
B



CAG00607

Eubacterium_sp_CAG_251

B



CAG00610

Hungatella_hathewayi_12489931

B



CAG00624
Firmicutes_bacterium_CAG_552
G



CAG00646

Alistipes_sp_CAG_268

G



CAG00650

Dorea_formicigenerans_ATCC_27755

B



CAG00668

Azospirillum_sp_CAG_239

G



CAG00669
Firmicutes_bacterium_CAG_103
G



CAG00676
Firmicutes_bacterium_CAG_176
G



CAG00698

Ruminococcus_sp_CAG_177

B



CAG00713
Firmicutes_bacterium_CAG_270
G



CAG00720

Anaerotruncus_colihominis_DSM_17241

B



CAG00727

Eggerthella_lenta_DSM_2243

B



CAG00766
Firmicutes_bacterium_CAG_176
B



CAG00771

Clostridium_sp_CAG_413

G



CAG00782

Eubacterium_rectale_CAG_36

G



CAG00861

Oscillibacter_sp_CAG_241

B



CAG00873

Butyricimonas_virosa_DSM_23226

B



CAG00880

Subdoligranulum_sp_CAG_314

G



CAG00886

Sutterella_sp_CAG_351

G



CAG00889

Megasphaera_elsdenii_14_14

G



CAG00897
Firmicutes_bacterium_CAG_83
B



CAG00919

Clostridium_methylpentosum_DSM_5476

G



CAG00928

Acidiphilium_sp_CAG_727

B



CAG00937

Clostridium_sp_CAG_7

G



CAG00963

Clostridium_sp_CAG_524

B



CAG01039

Faecalibacterium_cf_prausnitzii_KLE1255

B



CAG01141

Holdemanella_biformis_DSM_3989

B



CAG01144

Lactobacillus_vaginalis_DSM_5837_ATCC_49540

B



CAG01158

Pseudoflavonifractor_capillosus_ATCC_29799

G



CAG01197

Dialister_succinatiphilus_YIT_11850

G



CAG01208

Coprococcus_catus_GD_7

B



CAG01263

Clostridium_clostridioforme_2_1_49FAA

B



CAG01321

Faecalibacterium_prausnitzii_SL3_3

G












    • (ii) using one or several pre-defined equations each corresponding to a model obtained for at least 8 bacterial species from said set of bacterial species, calculating the probability that said individual responds to the treatment (PR) or the probability that said individual resists to the treatment (PNR) with an ICI-based therapy.





In the above method, the “abundances pattern based on the relative abundances of a set of bacterial species” can be, for example, in the form of a vector of said relative abundances. This abundance pattern will be inserted into an executable program (Windows OS environment) by the person performing the method (e.g., a clinician) to obtain the NR or R probability percentage of a subject.


As described in the experimental part below, several models can be obtained from clinical data of a representative cohort to assess the probability, for an individual, to respond (or not to respond) to the treatment. The experimental part describes in detail 12 possible models which, although based on different strategies, all provide very good predictability and success rates.


Other models can possibly be used, keeping in mind that an overrepresentation of bacterial species identified as “good bacteria” in Table 1 above, and/or underrepresentation of bacterial species identified as “bad bacteria” in the same table indicate that the individual is likely to respond to the treatment, whereas an overrepresentation of bacterial species identified as “bad bacteria” in Table 1 above, and/or underrepresentation of bacterial species identified as “good bacteria” in the same table indicate that the individual is likely to be a poor responder to the treatment. For assessing whether a bacterial species is over- or underrepresented in a fecal sample, its relative abundance is compared to a control value corresponding to the relative abundance of the same bacterial species in normal/healthy volunteers (i.e., individuals not having a cancer). For bacterial species that are not detected in the control volunteers, the mere presence of the bacterium is considered as an overrepresentation. Equations are used to “weight” the predictive values of each species' over- or underrepresentation and more precisely calculate the probability that the patient responds to the treatment (PR) or the probability that the patient resists to the treatment (PNR) with an ICI-based therapy.


In step (ii), several equations can be used, each corresponding to a model based on a subset of 8 bacteria from the set recited in step (i). The clinician will then combine the result obtained with each of these equations to more precisely predict the R or NR status of the patient.


According to a particular embodiment of the above method, at least one equation used in step (ii) corresponds to a model for a set of at least 8 bacterial species which are present in the fecal sample of said individual. However, a model based on 8 bacteria which are not all present in the sample can also be used, especially if it is combined to one or several models.


According to another particular embodiment of the above method, the model(s) used in step (ii) lead to an overall predictability of at least 65% and a success rate of at least 70%. The performance of a model will depend on the representativity of the cohort on which it has been obtained and on the relevance of the strategy used to build the model. A person skilled in the art of statistics can easily calculate the overall predictability and success rate of a model and determine whether it satisfies the required predictability and success rate criteria.


According to another particular embodiment of the above method, the Probability score obtained in step (ii) is interpreted as clinically meaningful if it is higher than 75% (fixed threshold) and/or if it is concordant among the different Models used, always taking into account the antibiotic usage (known or unknown).


According to another aspect of the present invention, the above method is combined with another method for determining, from a feces sample from a RCC patient, whether said patient is likely to be a good responder to a treatment with an ICI, based on an animal model. Such a method was already described in a previous application from the inventors's team (WO2016/063263) and comprises the steps of (i) performing a fecal microbial transplantation (FMT) of a feces sample from the patient into germ free (GF) model animals (e.g., GF mice); (ii) at least 7 to 14 days after step (i), inoculating said mice with a transplantable tumor model; (iii) treating the inoculated mice with the ICI; and (iv) measuring the tumor size in the treated animals. The results of step (iv) are illustrative of the response that can be expected for said patient to said treatment. In case the result obtained with the animal differs from the NR or R status predicted by the model(s) (with a probability of X %), the result with the animal model will prevail in the clinician's conclusion.


When the individual's antibiotic regimen exposure during the last two months is unknown, the method according to the invention is preferably performed using, in step (i), a set of bacterial species that comprises at least 8 bacterial species selected from the group consisting of:









TABLE 2







Bacterial species preferably used to assess the R or NR status


(best outcome criterion) of a patient whose antibiotic regimen


exposure during the last two months is unknown.








CAG number
Bacterial species





CAG00008

Clostridium_bolteae_ATCC_BAA_613



CAG00037

Bacteroides_faecis_MAJ27



CAG00063

Barnesiella_viscericola_DSM_18177



CAG00122

Coprococcus_catus_GD_7



CAG00140

Subdoligranulum_sp_4_3_54A2FAA



CAG00243
Lachnospiraceae_bacterium_1_1_57FAA


CAG00317

Clostridium_sp_CAG_230



CAG00327

Faecalibacterium_sp_CAG_74



CAG00357

Bacteroides_ovatus_V975



CAG00413

Bacteroides_sp_CAG_144



CAG00473

Prevotella_sp_CAG_617



CAG00610

Hungatella_hathewayi_12489931



CAG00650

Dorea_formicigenerans_ATCC_27755



CAG00727

Eggerthella_lenta_DSM_2243



CAG00928

Acidiphilium_sp_CAG_727



CAG01039

Faecalibacterium_cf_prausnitzii_KLE1255



CAG01141

Holdemanella_biformis_DSM_3989



CAG01144

Lactobacillus_vaginalis_DSM_5837_ATCC_49540



CAG01263

Clostridium_clostridioforme_2_1_49FAA










According to a preferred embodiment of the method for assessing the R or NR status (best outcome criterion) of a patient whose antibiotic regimen exposure during the last two months is unknown, one, two or three equations are used in step (ii), which correspond to models obtained for the following sets of bacterial species, identified by their CAG numbers:









TABLE 3







sets of bacteria used in Models 1.2, 2.2 and 3.2.











set for model 1.2
set for model 2.2
set for model 3.2







CAG00008
CAG00727
CAG00243



CAG01039
CAG01141
CAG00473



CAG00473
CAG00650
CAG00327



CAG00140
CAG00928
CAG01039



CAG00610
CAG00473
CAG00140



CAG01141
CAG00122
CAG01263



CAG00413
CAG01144
CAG00037



CAG00317
CAG00063
CAG00357










According to a preferred embodiment of the above method, at least one equation used corresponds to a model obtained with a set of bacteria which are all present in the individual's sample. However, the models can be run even if a few bacteria are missing. In such a case, it is preferable to use at least two and preferably at least 3 equations in step (ii).


According to a particular embodiment of the above method based on Models 1.2, 2.2 and/or 3.2, illustrated in the experimental part below, the equations for calculating the probability that said individual responds to the treatment (PR) are as follows:










P
R

=

1
/

[

1
+

exp


-

(




j
=
1

8




β
j



X
j



)


]






[
B
]







wherein Xj (j=1 to 8) are the relative abundances of the bacterial species measured in the individual's sample and βj (j=1 to 8) are the following regression coefficients:









TABLE 4







Regression coefficients for Models 1.2, 2.2 and 3.2.









Model 1.2
Model 2.2
Model 3.2













R

R

R


bacterial
coeffi-
bacterial
coeffi-
bacterial
coeffi-


species
cients
species
cients
species
cients















CAG00008
−1.0323
CAG00727
−1.0791
CAG00243
−1.1735


CAG01039
−1.084
CAG01141
−1.1164
CAG00473
−1.027


CAG00473
−0.9723
CAG00650
−1.2839
CAG00327
−0.9968


CAG00140
−0.9907
CAG00928
−1.1156
CAG01039
−1.0043


CAG00610
−0.6234
CAG00473
−1.014
CAG00140
−0.7456


CAG01141
−0.8468
CAG00122
−0.9394
CAG01263
−0.8736


CAG00413
0.8994
CAG01144
−0.8085
CAG00037
0.7032


CAG00317
0.6041
CAG00063
−0.7679
CAG00357
−0.6073









Of course, these coefficients can be refined by the skilled in the art by performing the same strategies as those described in the present application starting from other clinical data (i.e., clinical data from another cohort in addition to or in replacement of the data used by the inventors), or even merely because the 10000 iterations employed in the logistic regression (see methods) ensure each time a weak ‘floating randomness’ of each coefficient, not surpassing the 0.2 units. The coefficients can also differ if a different technique is used for measuring the relative abundances of the bacterial species (e.g., using qPCR instead of MGS analysis). Importantly, even using different techniques, regression coefficients will retain their positive or negative sign, meaning the positive or negative contribution of a definite CAG species to the overall model.


In case the patient did not take any antibiotic during the last two months, the method according to the invention is preferably performed using, in step (i), a set of bacterial species that comprises at least 8 bacterial species selected from the group consisting of:









TABLE 5







Bacterial species preferably used to assess the R or


NR status (best outcome criterion) of a patient who


did not take antibiotics during the last two months.










CAG number
Bacterial species







CAG00008

Clostridium_bolteae_ATCC_BAA_613




CAG00048_1

Clostridium_sp_CAG_226




CAG00140

Subdoligranulum_sp_4_3_54A2FAA




CAG00243
Lachnospiraceae_bacterium_1_1_57FAA



CAG00300

Prevotella_sp_CAG_891




CAG00327

Faecalibacterium_sp_CAG_74




CAG00413

Bacteroides_sp_CAG_144




CAG00473

Prevotella_sp_CAG_617




CAG00487

Prevotella_sp_CAG_279




CAG00650

Dorea_formicigenerans_ATCC_27755




CAG00698

Ruminococcus_sp_CAG_177




CAG00720

Anaerotruncus_colihominis_DSM_17241




CAG00727

Eggerthella_lenta_DSM_2243




CAG00766
Firmicutes_bacterium_CAG_176



CAG00897
Firmicutes_bacterium_CAG_83



CAG00963

Clostridium_sp_CAG_524




CAG01039

Faecalibacterium_cf_prausnitzii_KLE1255




CAG01141

Holdemanella_biformis_DSM_3989




CAG01208

Coprococcus_catus_GD_7











According to a preferred embodiment of the method for assessing the R or NR status (best outcome criterion) of a patient who did not take antibiotics during the last two months, one, two or three equations are used in step (ii), each of which corresponding to a model obtained for the following sets of bacterial species, identified by their CAG numbers:









TABLE 6







Sets of bacteria used in Models 4.2, 5.2 and 6.2.











set for model 4.2
set for model 5.2
set for model 6.2







CAG00008
CAG00048_1
CAG00243



CAG01039
CAG00300
CAG00008



CAG00473
CAG00473
CAG00698



CAG00487
CAG00650
CAG00473



CAG00140
CAG00720
CAG00327



CAG01141
CAG00727
CAG01039



CAG01208
CAG00963
CAG00897



CAG00413
CAG01141
CAG00766










According to a preferred embodiment of the above method, at least one equation used corresponds to a model obtained with a set of bacteria which are all present in the individual's sample. However, the models can be run even if a few bacteria are missing. In such a case, it is preferable to use at least two and preferably at least 3 equations in step (ii).


According to a particular embodiment of the above method based on Models 4.2, 5.2 and/or 6.2, illustrated in the experimental part below, the equations for calculating the probability that said individual resists (PNR) or responds (PR) to the treatment are as follows:










P

N

R


=


1
/

[

1
+

exp


-

(




j
=
1

8








β
j



X
j



)


]







for





models





4.2





and





5.2





[
A
]







P
R

=


1
/

[

1
+

exp


-

(




j
=
1

8








β
j



X
j



)


]







for





model





6.2





[
B
]







wherein Xj (j=1 to 8) are the relative abundances of the bacterial species measured in the individual's sample and βj (j=1 to 8) are the following regression coefficients:









TABLE 7







Regression coefficients for Models 4.2, 5.2 and 6.2.









Model 4.2
Model 5.2
Model 6.2













NR

NR

R


bacterial
coeffi-
bacterial
coeffi-
bacterial
coeffi-


species
cients
species
cients
species
cients















CAG00008
0.955
CAG00048_1
0.7112
CAG00243
−1.2907


CAG01039
0.8761
CAG00300
0.8931
CAG00008
−0.8848


CAG00473
0.9951
CAG00473
1.0042
CAG00698
−0.9776


CAG00487
0.7609
CAG00650
1.1282
CAG00473
−0.9997


CAG00140
1.1107
CAG00720
1.2569
CAG00327
−1.0697


CAG01141
0.9402
CAG00727
1.2397
CAG01039
−0.7806


CAG01208
0.7187
CAG00963
0.7595
CAG00897
−0.8376


CAG00413
−0.5812
CAG01141
1.0235
CAG00766
−0.7459









As mentioned above, these coefficients can be refined for several reasons or even changed if a different technique is used for measuring the relative abundances of the bacterial species (e.g., using qPCR instead of MGS analysis).


The models 1.2, 2.2, 3.2, 4.2, 5.2 and 6.2 above have been obtained using a criterion of best outcome. Patients who first responded to the treatment but relapsed a few months later were thus considered as responders. In order to assess if an individual is likely to have a long-term benefit from the treatment, defined as a time to progression (TTP) of at least 6 months, the inventors obtained additional models by considering only the stable diseases of more than 6 months (SD>6 months) among responders (OUTCOME_2 in the experimental part). These models are based on partly different subsets of the microbiota composition.


For assessing whether an individual whose antibiotic regimen exposure during the last two months is unknown is likely to have a long-term benefit from a treatment with an anti-PD1/PD-L1/PD-L2 Ab-based therapy, the method according to the invention is preferably performed using, in step (i), a set of bacterial species that comprises at least 8 bacterial species selected from the group consisting of:









TABLE 8







Bacterial species preferably used to assess the R or NR status


(TTP >6 months criterion) of a patient whose antibiotic


regimen exposure during the last two months is unknown.










CAG number
Bacterial species







CAG00013
Clostridiales_bacterium_1_7_47FAA



CAG00211
Firmicutes_bacterium_CAG_227



CAG00474

Sutterella_wadsworthensis_2_1_59BFAA




CAG00557

Ruminococcus_callidus_ATCC_27760




CAG00601
uncultured_Faecalibacterium_sp



CAG00607

Eubacterium_sp_CAG_251




CAG00624
Firmicutes_bacterium_CAG_552



CAG00650

Dorea_formicigenerans_ATCC_27755




CAG00668

Azospirillum_sp_CAG_239




CAG00669
Firmicutes_bacterium_CAG_103



CAG00676
Firmicutes_bacterium_CAG_176



CAG00771

Clostridium_sp_CAG_413




CAG00782

Eubacterium_rectale_CAG_36




CAG00861

Oscillibacter_sp_CAG_241




CAG00873

Butyricimonas_virosa_DSM_23226




CAG00880

Subdoligranulum_sp_CAG_314




CAG00886

Sutterella_sp_CAG_351




CAG00889

Megasphaera_elsdenii_14_14




CAG00937

Clostridium_sp_CAG_7




CAG01141

Holdemanella_biformis_DSM_3989




CAG01197

Dialister_succinatiphilus_YIT_11850




CAG01321

Faecalibacterium_prausnitzii_SL3_3











According to a preferred embodiment of the method for assessing the R or NR status (TTP>6months criterion) of a patient whose antibiotic regimen exposure during the last two months is unknown, one, two or three equations are used in step (ii), which correspond to models obtained for the following sets of bacterial species, identified by their CAG numbers:









TABLE 9







Sets of bacteria used in Models 7.2, 8.2 and 9.2.











set for model 7.2
set for model 8.2
set for model 9.2







CAG00782
CAG00211
CAG00557



CAG00013
CAG00474
CAG00601



CAG00873
CAG00624
CAG00607



CAG01141
CAG00650
CAG00669



CAG00668
CAG00676
CAG00861



CAG00669
CAG00771
CAG00880



CAG00886
CAG01197
CAG00937



CAG00889
CAG01321
CAG01321










According to a preferred embodiment of the above method, at least one equation used in step (ii) corresponds to a model obtained with a set of bacteria which are all present in the individual's sample. However, the models can be run even if a few bacteria are missing. In such a case, it is preferable to use at least two and preferably at least 3 equations in step (ii).


According to a particular embodiment of the above method based on Models 7.2, 8.2 and/or 9.2, illustrated in the experimental part below, the equations for calculating the probability that said individual resists (PNR) to the treatment are as follows:










P

N

R


=

1
/

[

1
+

exp


-

(




j
=
1

8








β
j



X
j



)


]






[
A
]







wherein Xj (j=1 to 8) are the relative abundances of the bacterial species measured in the individual's sample and βj (j=1 to 8) are the following regression coefficients:









TABLE 10







Regression coefficients for Models 7.2, 8.2 and 9.2.









Model 7.2
Model 8.2
Model 9.2













NR

NR

NR


bacterial
coeffi-
bacterial
coeffi-
bacterial
coeffi-


species
cients
species
cients
species
cients















CAG00782
−0.2034
CAG00211
0.6422
CAG00557
0.9341


CAG00013
0.4092
CAG00474
−1.0581
CAG00601
0.8927


CAG00873
0.2708
CAG00624
−0.8984
CAG00607
0.7108


CAG01141
0.7665
CAG00650
1.3189
CAG00669
−1.2341


CAG00668
−0.5899
CAG00676
−0.7324
CAG00861
1.0379


CAG00669
−0.5181
CAG00771
−0.6623
CAG00880
−1.0016


CAG00886
−0.8415
CAG01197
−1.1372
CAG00937
−0.872


CAG00889
−0.7339
CAG01321
−0.8525
CAG01321
−0.8362









As already mentioned, these coefficients can be refined for several reasons or even changed if a different technique is used for measuring the relative abundances of the bacterial species.


In case the patient did not take any antibiotic during the last two months, the method according to the invention for assessing his/her probability of having a long-term benefit (TTP>6 months) from a treatment with an anti-PD1/PD-L1/PD-L2 Ab-based therapy is preferably performed using, in step (i), a set of bacterial species that comprises at least 8 bacterial species selected from the group consisting of:









TABLE 11







Bacterial species preferably used to assess the R


or NR status (best outcome criterion) of a patient who


did not take antibiotics during the last two months.








CAG number
Bacterial species





CAG00013

Clostridiales_bacterium_1_7_47FAA



CAG00142

Bacteroides_stercoris_ATCC_43183



CAG00218

Bacteroides_sp_CAG__20



CAG00341

Eubacterium_sp_CAG_115



CAG00346

Eubacterium_rectale_M104_1



CAG00474

Sutterella_wadsworthensis_2_1_59BFAA



CAG00508

Alistipes_obesi



CAG00530

Prevotella_sp_CAG_617



CAG00580

Clostridium_sp_CAG_62



CAG00601
uncultured_Faecalibacterium_sp_


CAG00607

Eubacterium_sp_CAG_251



CAG00646

Alistipes_sp_CAG_268



CAG00668

Azospirillum_sp_CAG_239



CAG00669

Firmicutes_bacterium_CAG_103



CAG00713

Firmicutes_bacterium_CAG_270



CAG00873

Butyricimonas_virosa_DSM_23226



CAG00880

Subdoligranulum_sp_CAG_314



CAG00886

Sutterella_sp_CAG_351



CAG00889

Megasphaera_elsdenii_14_14



CAG00919

Clostridium_methylpentosum_DSM_5476



CAG01141

Holdemanella_biformis_DSM_3989



CAG01158

Pseudoflavonifractor_capillosus_ATCC_29799



CAG01263

Clostridium_clostridioforme_2_1_49FAA



CAG01321

Faecalibacterium_prausnitzii_SL3_3










According to a preferred embodiment of the method for assessing the R or NR status (TTP>6 months criterion) of a patient who did not take antibiotics during the last two months, one, two or three equations are used in step (ii), each of which corresponding to a model obtained for the following sets of bacterial species, identified by their CAG numbers:









TABLE 12







Sets of bacterial species for Models 10.2, 11.2 and 12.2.











set for model 10.2
set for model 11.2
set for model 12.2







CAG00580
CAG00346
CAG00142



CAG00013
CAG00530
CAG00218



CAG00873
CAG00601
CAG00341



CAG01141
CAG00607
CAG00474



CAG00668
CAG00646
CAG00508



CAG00669
CAG00713
CAG00880



CAG00886
CAG00919
CAG01263



CAG00889
CAG01158
CAG01321










According to a preferred embodiment of the above method, at least one equation used in step (ii) corresponds to a model obtained with a set of bacteria which are all present in the individual's sample. However, the models can be run even if a few bacteria are missing. In such a case, it is preferable to use at least two and preferably at least 3 equations in step (ii).


According to a particular embodiment of the above method based on Models 10.2, 11.2 and/or 12.2, illustrated in the experimental part below, the equations for calculating the probability that said individual resists to the treatment (PNR) or responds (PR) are as follows:










P

N

R


=


1
/

[

1
+

exp


-

(




j
=
1

8




β
j



X
j



)


]







for





model





12.2





[
A
]







P
R

=


1
/

[

1
+

exp


-

(




j
=
1

8








β
j



X
i



)


]







for





models





10.2





and





11.2





[
B
]







wherein Xj (j=1 to 8) are the relative abundances of the bacterial species measured in the individual's sample and βj (j=1 to 8) are the following regression coefficients:









TABLE 13







Regressions coefficients for Models 10.2, 11.2 and 12.2.









Mode 10.2
Mode 11.2
Model 12.2













R

R

NR


bacterial
coeffi-
bacterial
coeffi-
bacterial
coeffi-


species
cients
species
cients
species
cients















CAG00580
−0.1456
CAG00346
−0.8463
CAG00142
1.138


CAG00013
−0.5496
CAG00530
1.13
CAG00218
1.1396


CAG00873
−0.4646
CAG00601
−1.183
CAG00341
−1.0166


CAG01141
−0.5496
CAG00607
−0.9544
CAG00474
−0.8403


CAG00668
0.5684
CAG00646
0.8929
CAG00508
−0.7067


CAG00669
0.5129
CAG00713
1.0919
CAG00880
−1.185


CAG00886
0.6623
CAG00919
0.4881
CAG01263
0.8466


CAG00889
0.7102
CAG01158
1.1606
CAG01321
−0.8895









As for the other models, these coefficients can be refined for several reasons or even changed if a different technique is used for measuring the relative abundances of the bacterial species.


According to a particular embodiment of the method of the invention, the fecal sample is obtained before the first administration of any ICI, such as an anti-PD1/PD-L1/PD-L2 antibody.


According to another particular embodiment, the individual already received a first-line therapy different from PD1/PD-L1/PD-L2 Ab-based therapies for treating his/her RCC. The method of the invention is however not limited to patients receiving a PD1/PD-L1/PD-L2 Ab-based therapy as a second line therapy, and the presently disclosed methods can also be used to assess the responder or non-responder status of RCC patients receiving a first-line PD1/PD-L1/PD-L2 Ab-based therapy either alone or in combination with other antineoplastic drugs (chemotherapy or another ICI such as anti-CTLA-4, as specified above).


According to another particular embodiment, the anti-PD1/PD-L1/PD-L2 Ab-based therapy administered to the patient is a treatment with an anti-PD1 antibody such as nivolumab or pembrolizumab or an anti-PD-L1 antibody such as atezolizumab or durvalumab, for example.


According to another of its aspects, the present invention pertains to a theranostic method for determining if a cancer patient needs a bacterial compensation before administration of an anti-PD1/PD-L1/PD-L2 Ab-based therapy and/or during this therapy, comprising assessing, by a method as above-disclosed, whether the patient is likely to be a good responder to such a therapy, wherein if the patient is not identified as likely to be a good responder, the patient needs a bacterial compensation.


According to this aspect of the invention, the bacterial compensation can be done either by fecal microbiota transplant (FMT), using microbiota from one or several donors (for example, from responders to the treatment), possibly enriched with bacterial strains known to be beneficial in this situation, or by administration of a bacterial composition. The inventors already described bacterial compositions that can be used for such a compensation and restore the ability, for the patient, to respond to the treatment (e.g., in WO 2016/063263 and in WO 2018/115519). Non-limitative examples of bacterial strains which can be beneficial to patients with an initial NR status are: Enterococcus hirae, Akkermansia muciniphila, Blautia strains, Coprococcus comes strains, Alistipes shahii, other Alistipes species (e.g. Alistipes indistinctus and/or onderdonkii and/or finegoldii), Ruminococcacae, Clostridiales species, Bacteroidales species, Actinobacteria, Coriobacteriales species, Methanobrevibacter Burkholderia cepacia, Bacteroides fragilis, Actinotignum schaalii, as well as the following additional bacteria:

    • Clostridiales bacteria of the species Christensenella minuta;
    • Erysipelotrichia of the species Dielma fastidiosa or Erysipelatoclostridium ramosum;
    • Eubacterium limosum;
    • Bacteroidales bacteria of the species Bacteroides salyersiae and/or Bamesiella intestinihominis;
    • Coriobacteriales bacteria of the species Coffinsella intestinalis and/or Collinsella tanakaei;
    • Firmicutes bacteria of the species Flavonifractor plautii.


Examples of compositions which can be beneficial to a patient with an initial NR status assessed by the method according to the invention are:

    • a composition comprising:
      • (i) Enterococcus hirae selected from the group consisting of Enterococcus hirae strain 13144 deposited on Nov. 7, 2013 at the Collection Nationale de Cultures de Microorganismes (CNCM) under the number I-4815, Enterococcus hirae strain IGR7 deposited on Aug. 31, 2017 at the CNCM under the number I-5224, Enterococcus hirae strain IGR4 deposited on Nov. 27, 2017 at the CNCM under the number CNCM I-5260, Enterococcus hirae strain IGR11 deposited on Nov. 27, 2017 at the CNCM under the number CNCM I-5261 and mixtures thereof; and
      • (ii) Akkermansia muciniphila selected from the group consisting of Akkermansia muciniphila strains p2261 and p3415, both deposited at the Collection de souches de I′Unité des Rickettsies (CSUR) and mixtures thereof; and
      • (iii) Eubacterium limosum.
    • a composition comprising:
      • (i) Enterococcus hirae selected from the group consisting of Enterococcus hirae strain 13144 deposited on Nov. 7, 2013 at the Collection Nationale de Cultures de Microorganismes (CNCM) under the number I-4815, Enterococcus hirae strain IGR7 deposited on Aug. 31, 2017 at the CNCM under the number 1-5224, Enterococcus hirae strain IGR4 deposited on Nov. 27, 2017 at the CNCM under the number CNCM I-5260, Enterococcus hirae strain IGR11 deposited on Nov. 27, 2017 at the CNCM under the number CNCM I-5261 and mixtures thereof; and
      • (ii) Barnesiella intestinihominis.
    • a composition comprising:
      • (i) Enterococcus hirae selected from the group consisting of Enterococcus hirae strain 13144 deposited on Nov. 7, 2013 at the Collection Nationale de Cultures de Microorganismes (CNCM) under the number I-4815, Enterococcus hirae strain IGR7 deposited on Aug. 31, 2017 at the CNCM under the number I-5224, Enterococcus hirae strain IGR4 deposited on Nov. 27, 2017 at the CNCM under the number CNCM I-5260, Enterococcus hirae strain IGR11 deposited on Nov. 27, 2017 at the CNCM under the number CNCM I-5261 and mixtures thereof; and
      • (ii) Christensenella minuta.
    • a composition comprising:
      • (i) Enterococcus hirae selected from the group consisting of Enterococcus hirae strain 13144 deposited on Nov. 7, 2013 at the Collection Nationale de Cultures de Microorganismes (CNCM) under the number 1-4815, Enterococcus hirae strain IGR7 deposited on Aug. 31, 2017 at the CNCM under the number 1-5224, Enterococcus hirae strain IGR4 deposited on Nov. 27, 2017 at the CNCM under the number CNCM I-5260, Enterococcus hirae strain IGR11 deposited on Nov. 27, 2017 at the CNCM under the number CNCM I-5261 and mixtures thereof; and
    • (ii) Actinotignum schaalii.


A nucleic acid microarray designed to perform the method according to the invention is also part of the present invention. Such a nucleic acid microarray comprises nucleic acid probes specific for each of the microorganism species to be detected in step (i) of said method (i.e., at least 8 species selected amongst those recited in Table 1). In a specific embodiment, the nucleic acid microarray is an oligonucleotide microarray comprising at least one oligonucleotide specific for at least one sequence selected from SEQ ID NOs: 1-2950. For example, the said microarray comprises at least 8 oligonucleotides, each oligonucleotide being specific for one sequence of a distinct species recited in table 1. The microarray of the invention can of course comprise more oligonucleotides specific for sequences of SEQ ID NOs: 1-2950, for example at least nx96 oligonucleotides, divided into 12 sets of nx8 oligonucleotides corresponding to the 12 models described herein, with n being an integer comprised between 1 and 25 which corresponds to the number of oligonucleotides used to specifically assess the presence of one specific bacterial species. The microarray according to the invention may further comprise at least one oligonucleotide for detecting at least one gene of at least one control bacterial species. A convenient bacterial species may be e.g. a bacterial species the abundance of which does not vary between individuals having a R or a NR status. Preferably, the oligonucleotides are about 50 bases in length. Suitable microarray oligonucleotides specific for any gene of SEQ ID NOs: 1-2950 may be designed, based on the genomic sequence of each gene, using any method of microarray oligonucleotide design known in the art. In particular, any available software developed for the design of microarray oligonucleotides may be used, such as, for instance, the OligoArray software, the GoArrays software, the Array Designer software, the Primer3 software, or the Promide software, all known by the skilled in the art.


As mentioned above, the relative abundance of the recited bacterial species can be measured by techniques different from the metagenomics analysis used herein, especially when the skilled in the art knows which bacterial species are to be measured. A particular technique which can be used to this purpose is qPCR (quantitative PCR). The PCR-based techniques are performed with amplification primers designed to be specific for the sequences which are measured. The present invention hence also pertains to a set of primers suitable for performing the above method, i.e., a set of primers comprising primer pairs for amplifying sequences specific for each of the microorganism species to be detected in step (i) of said method (i.e., at least 8 species selected amongst those recited in Table 1). Such a set of primers comprises a minimum of 16 primers, but it can comprise more primers, for example 30, 40, 50, 60, 70, 80, 100, 200, 300, 500, 1000, 2000 or more. According to a particular embodiment, the set of primers comprises at least one primer pair specifically amplifying part of a sequence selected amongst SEQ ID Nos: 1-2950. Of course, primer sets according to the invention can advantageously comprise 2, 3, 4, 5, 10, 20, 30, 40, 50, 60, 70, 80, 100, 200, 300, 500, 1000 or more pairs of primers each specifically amplifying part of a sequence selected amongst SEQ ID Nos: 1-2950.


Other characteristics of the invention will also become apparent in the course of the description which follows of the biological assays which have been performed in the framework of the invention and which provide it with the required experimental support, without limiting its scope.


EXAMPLES
Materials and Methods

Metagenomic Analysis of Stool Samples


Quantitative metagenomics pipeline developed at MetaGenoPolis (Jouy-en-Josas, France) was employed to reach the species-level description of RCC gut microbiota. DNA was extracted from 69 stool samples through an automated platform (SAMBO, MetaGenoPolis) and subjected to shotgun sequencing using Ion Proton sequencer to reach >20 million short DNA sequence reads (MetaQuant platform, MetaGenoPolis). Reads were filtered (Q>25) and cleaned to eliminate possible contaminants as human reads. The high-quality (HQ) reads were then mapped against the MetaHIT hs_9.9 M genes catalogue and counted through a two-step procedure: 1) using uniquely mapped reads; 2) attributing shared reads according to their mapping ratio based on unique reads. Identity threshold for mapping was set at >95%, in order to overcome gene allelic variants and the non-redundant nature of the catalogue itself. HQ reads were downsized at 13 millions for each sample in order to correct for sequencing depth, then normalized through RPKM (reads per kilo base per million mapped reads) method. A profile matrix of gene frequencies was thus obtained, and used as an input file for MetaOMineR, a suite of R packages developed at MetaGenoPolis.


In order to achieve the species-level description of microbes in RCC stool samples, the hs_9.9 M catalogue was clustered into 1436 MetaGenomic Species (MGS), roughly >500 genes that covary in abundance among hundreds of samples, ultimately belonging to the same microbial species. The taxonomical annotation of MGS was performed using gene homology with previously sequenced organisms using BLASTn against nt and wgs NCBI databases. After taxonomical assignment of each MGS, a matrix of frequency profiles of 50 co-abundant genes (CAG) was built using normalized MGS mean signals, thus the sum of each MGS within a sample resulted equal to 1. With this approach each species was assigned to a definite CAG number.


Statistical Analysis


Multivariate statistics were employed on the matrix of frequency profiles in order to describe the gut microbial composition and diversity. Python 2.7 and related statistical/graphical libraries (Matplotlib, Scikit learn, Pandas, Numpy) were used to compare all MGS through pairwise analysis, linear discriminant analysis effect size (LEfSe) and volcano plots. A P value less than or equal to 0.05 was considered statistically significant. Volcano plots were generated computing for each bacterial species: i) the log 2 of fold ratio (FR) among the mean relative abundances of R versus NR (x axis) and ii) the co-log 10 of P values deriving from Mann-Whitney U test calculated on relative abundances in absolute values (y axis). In order to ease the volcano graphical representation, at TTP6 the chosen P value was less than or equal to 0.7. LEfSe plots were generated with Python 2.7 on output files derived from LEfSe pipeline, and all species with LDA score 2 were considered for subsequent analysis.


Building the Predictive Models


A double strategy was implemented to select the bacterial species used to build the models.


The first one (“human-oriented”) relies on: i) performing Receiver Operating Characteristic (ROC) curves implementing Support Vector Machine (SVM) classifier on all possible combinations of bacterial species, looking for consortia giving the best Area Under Curve (AUC) values; ii) building a logistic regression classifier and a RFECV (recursive feature elimination with cross-validation) feature selection with selected bacterial species; iii) performing metrics as ROC curves and confusion matrices using the train/test subsets deriving from the original dataset.


The second strategy (“machine-learning”) relies on: i) building a logistic regression classifier and a RFECV feature selection on all bacterial species (without permutation), selecting the ones with higher ranking (equal to 1); ii) performing a new feature selection by means of RFE (recursive feature elimination), selecting the 8 species with highest ranking; iii) re-building a new logistic regression classifier plus RFECV feature selection on the 8 selected bacterial species; iv) performing metrics as ROC curves and confusion matrices using the train/test subsets deriving from the original dataset.


The main difference among the first and the second strategies is that the first one allows combinations of all the bacterial species present in the original dataset selecting sub-groups of them (called ‘consortia’) based on the highest AUC, while the second one would be able to select single species due to the natural convergence of the logistic regression model to select all the variables in order to reach the maximum predictability.


The implementations for building the logistic regression classifier and the feature selection models were made in Python (version 2.7) using the methods ‘logisticRegression’, ‘RFE’ and ‘RFECV’ from the Scikit Learn module (version 0.19.1). The implementation for building the SVM classifier (C-Support Vector Classification) was made in Python (version 2.7) using the method SVC from the Scikit Learn module (version 0.19.1). The implementation of combinations for ROC curves in the first strategy was made in Python (version 2.7) using the method ‘combinations’ from the Itertools' module present as a built-in in Python 2.7.


Logistic regression coefficients β were calculated by logistic regression classifier with the following requirements: a constant (also known as intercept) not added to the decision function; a max iteration equal to 10000; a liblinear solver (with L2 penalization). RFECV feature selection model (step=1, cross-validation equal to 3, scoring accuracy) was implemented in Python with logistic regression estimator (no intercept, 10000 max iteration, liblinear solver), providing rankings within the interval [1: (nspecies-1)], and it was used to make a recursive feature selection on all bacterial species, selecting only those species having a rank equal to 1. RFE feature selection model (step=1, feature to select=8) was similarly implemented in Python with the same estimator (logistic regression, no intercept, 10000 max iteration, liblinear solver) of RFECV.


Receiver Operating Characteristic (ROC) curves were built with Python 2.7 using the SVM classifier (linear kernel) and employed to assess the actual predictive meaning of selected bacterial species or consortia. Two different approaches were used to achieve such a selection: 1) species retrieved from LEfSe analysis (with LDA score 2) and combined within all possible combinations (2×2, 3×3, 4×4, 5×5, etc. . . . ) and 2) all species were considered and the first 5 having the best AUC were combined into a consortium. Area Under Curve (AUC), specificity and sensitivity were computed for each best combination, and reported as ROC curve and tabular data. A 5-fold cross-validation was used, with no added noise. Mean relative abundances (±Standard Error of the Mean), Fold Ratio (FR), Log 10 of FR and P value of cohort comparison (R vs NR, non-parametric Mann-Whitney U test) were also reported.


According to the aforementioned methods, bacterial species were chosen in the following manner: i) for the first strategy, taking into account all the combinations for all available species, and especially those derived from LEfSe, we selected the ‘consortium’ for each OUTCOME_1, OUTCOME_2, TTP3 and TTP6 timepoints having the highest AUC value, both for NR and R status; ii) for the second strategy, we selected the bacterial species having a RFECV and then RFE rank equal to 1, regardless negative and positive logistic regression coefficients (β), for OUTCOME_1 and OUTCOME_2.


Taking into account the two aforementioned strategies, a logistic regression classifier was implemented, for both of them, on standardized raw data (removing the mean and scaling to unit variance) in order to calculate regression coefficients (β) to be used in equation [2]. Thus, the relative contribution of each selected bacterial species to each model was weighted by the β coefficient and put into the equation [2] in order to predict through equation [1] the estimation (in probability percentage) of the outcome for a definite new subject.






P=1/(1+exp{circumflex over ( )}−z)  [1]





and






z=β1*x1+ . . . +βn*xn  [2]


For both strategies, two feature selection models (RFECV and then RFE) model were implemented to refine the number of selected species at 8, a number useful to be implemented in future and forthcoming diagnostic kits for clinical practical use. Evaluation of the final Models (built on three different sets of 8 bacterial species) was performed through metrics as ROC curves and confusion matrices using t train/test subsets of the original dataset (implemented in Python 2.7 and Scikit learn 0.19, through the train_test_split method).


For OUTCOME_1, taking into account all the aforementioned embodiments, methods and strategies, and taking into account the dichotomous/complementary nature of logistic regression classifier, in the present invention we propose four equations ([3], [4], [6], [7]) to predict the NR or R status of a definite new subject JD. Two of them ([3], [4]) take into account the eventuality of an unknown antibiotic regimen/treatment status, while the other two ([6], [7]) consider that the individual did not take antibiotics during the past two months.


In order to circumvent the caveats deriving from the natural sources of variability within the subjects' cohort (feces sampling, feces consistencies, sample storage, sequencing biases, etc. . . . ) that could affect relative abundances of under-represented bacterial species, the inventors built two further equations ([5], [8]) based solely on the species having an average relative abundance greater than or equal to 0.001 if expressed in the interval [0:1] (or 0.1% if expressed in the interval [0:100]) for subjects with unknown antibiotic treatment ([5]) and for subjects who did not take antibiotics during the last two months prior sampling ([8]) These pattern of species underwent the second strategy of modeling as described above.


For OUTCOME_2, taking into account all the aforementioned embodiments, methods and strategies, and taking into account the dichotomous/complementary nature of logistic regression classifier, in the present invention we propose four equations ([9], [10], [12], [13]) to predict the NR or R status of a definite new subject JD. Two of them ([9], [10]) take into account the eventuality of an unknown antibiotic regimen/treatment status, while the other two ([12], [13]) consider that the individual did not take antibiotics during the past two months.


In order to circumvent the caveats deriving from the natural sources of variability within the subjects' cohort (feces sampling, feces consistencies, sample storage, sequencing biases, etc. . . . ) that could affect relative abundances of under-represented bacterial species, the inventors built two further equations ([11], [14]) based solely on the species having an average relative abundance greater than or equal to 0.001 if expressed in the interval [0:1] (or 0.1% if expressed in the interval [0:100]) for subjects with unknown antibiotic treatment ([11]) and for subjects who did not take antibiotics during the last two months prior sampling ([14]) These pattern of species underwent the second strategy of modeling as described above.


Example 1: Metagenomics-Based GOMS Associated with Clinical Outcome According to RECIST 1.1 Criteria (FIG. 1)

We explored the composition of the gut microbiota using quantitative metagenomics by shotgun sequencing, reaching >20 million short DNA sequence reads per sample followed by analysis of the results in a 9.9 million-gene reference catalogue (J. Li, Nat. Biotechnol. 32, 834-841 (2014). Total DNA was extracted from 69 patients diagnosed with RCC prior to starting therapy (Table 1). Among the 69 RCC patients, 58 did not take ATB, while 11 had been prescribed ATB before the MG analyses. As reported in Materials and Methods, each metagenomic species (MGS) was assigned to a definite co-abundant gene profile (CAG), and the subsequent taxonomical annotation was performed based on gene homology to previously sequenced organisms (using blastN against the nt and WGS databanks). R (responders) were defined as those patients exhibiting a complete or partial or stable disease >3 months, while NR (non responders) were defined as harbouring a progressive disease









TABLE 14







Patients' characteristics. The clinical parameters relevant for


the 69 RCC included in this study are described in this table (gender,


age, antibiotics, disease status at diagnosis, responses, follow up).









Total (n = 69)



absolute numbers (%)













Age-yr
Median
62



Range
30-82










Age-yr-no.(%)

<65

40
(58)



≥65 <75
22
(32)



≥75
7
(10)


Gender-no.(%)
Male
48
(69)



female
21
(31)


Histology-no.(%)
Clear cell
57
(97)



Non-clear cell
2
(3)


Nephrectomyy-no.(%)
Yes
63
(91)



Mo
6
(9)


IMDC risk group-no.(%)
Good
14
(20)



Intermediate
39
(57)



Poor
14
(20)



Unknown
2
(3)


Number of prior

<2

55
(80)


treatments-no.(%)
 ≥3
14
(20)


ATB-no.(%)
Yes
11
(16)



No
58
(84)





IMDC, International Metastatic Renal Cell Carcinoma Database Consortium (includes: Karnofsky performance status, time from diagnosis to treatment, hemoglobin, serum calcium concentration, neutrophil and platelet counts);


ATB, Antibiotics






When segregating responders (R) from non-responders (NR) in all 69 patients (according to the best clinical response as assessed by RECIST1.1), we failed to observe significant differences with a FDR of 10%.


GAMS Associated with Worse Clinical Outcome


However, the best predictive model allowing to identify the “resistant” patients with AUC value of 0.882, a specificity of 0.778 (true positive cases) and a sensitivity (false positive cases) of 0.860 was obtained when considering LEfSe combination species, and the commensals retained in the model, that were increased in NR (“bad bacteria”) were:

    • Clostridium boltae ATCC BAA 613 (fold increase 13)
    • Faecalibacterium prausnitzii KLE1255-3 (fold increase 4.4)
    • Prevotella sp CAG617 (detectable only in NR)
    • Prevotella copri CAG164 (detectable only in NR)
    • Subdoligranulum sp 4_3_54A2FAA (fold increase 2.6)
    • while those that were decreased in NR (“good bacteria”) were:
    • Bacteroides sp CAG 144 (decreased by 62 fold)
    • Acinetobacter CAG196 (decreased by 3.4 fold)
    • Eubacterium sp CAG38 (decreased by 2.3 fold)


The cut-off values correspond to the means±SEM featuring in Table 2. We kept only those species having a p value <0.05 for the final model, and claim the equilibrium between good and bad bacteria for the best outcome in this patent.


When segregating responders (R) from non-responders (NR) in ROC who did not take ATB (according to the best clinical response as assessed by RECIST1.1), we failed to observe significant differences with a FDR p value <0.05.


However, the best predictive model allowing to identify the “resistant” patients with AUC value of 0.879, a specificity of 0.707 (true positive cases) and a sensitivity (false positive cases) of 0.950 was obtained when considering LEfSe combination species, and the commensals retained in the model that were increased in NR (“bad bacteria”) were:

    • Clostridium boltae ATCC BAA 613 (fold increase 15.7)
    • Prevotella sp CAG617 (detectable only in NR)
    • while those that were decreased in NR (“good bacteria”) were:
    • Bacteroides sp CAG 144 (decreased by 50 fold)
    • Eubacterium sp CAG115 (decreased by 47 fold)
    • Eubacterium siraeum sp CAGBO (decreased by 14 fold)
    • Firmicutes bacterium sp. CAG651 (decreased by 1.9 fold)


The cut-off values correspond to the means±SEM featuring in Table 15. We kept only those species having a p value <0.05 for the final model. According to the present invention, the equilibrium between good and bad bacteria is critical for the best outcome.


GAMS Associated with Best Clinical Outcome


The best predictive model allowing to identify the “responding” patients (R) among all patients with AUC value of 0.863, a specificity of 0.798 (true positive cases) and a sensitivity (false positive cases) of 0.842 was obtained when considering LEfSe combo species: the decrease of the following detrimental commensals retained in the model were:

    • Clostridium boltae ATCC BAA 613 (fold decrease 13)
    • Faecalibacterium prausnitzii KLE1255-3 (fold decrease 15)
    • Prevotella sp CAG617 (infini, detectable only in NR)
    • Subdoligranulum sp 4_3_54A2 FAA (fold decrease 2.6)
    • while those that were increased in R (“good bacteria”) were:
    • Bacteroides sp CAG 144 (increased by 62 fold)
    • Eubacterium sp CAG38 (increased by 2.3 fold)
    • Azospiriilum sp CAG 239 (increased by 3.8 fold)


The cut-off values correspond to the means±SEM featuring in Table 15. We kept only these species (that had a p value <0.05) for the final model and claim the equilibrium between good and bad bacteria for the best outcome in this patent.


The best predictive model allowing to identify the “responding” patients (R) among those who did not take ATB with AUC value of 0.845, Spec=0.990; Sens=0.614 was obtained when considering LEfSe combo species, and the commensals retained in the model were:


Are decreased in R the following ones:

    • Prevotella_sp_CAG_617 (infini, absent in R)
    • Clostridium_clostridioforme_2_1_49FAA_1 (decreased by 6.3 fold in R)


Are increased in R the following ones:

    • Bacteroides plebeius CAG 211 (increased by 327 fold in R)
    • Bacteroides coprophilus DSM 18228 JCM 13818 (present only in R)
    • Eubacterium_siraeum_CAG_80 (increased by 14 fold in R)
    • Eubacterium_sp_CAG_202 (increased by 5.4 fold in R)
    • Eubacterium_sp_CAG_115 (increased by 47 fold in R)


The cut-off values correspond to the means±SEM featuring in Table 15. We kept only these species (that had a p value <0.05) for the final model. According to the present invention, the equilibrium between good and bad bacteria is critical for the best outcome.









TABLE 15







Commensal bacteria associated with best (or worse) clinical outcome). Part A includes all RCC patients while part


B considered only patients who did not take antibiotics. The mean relative abundance with SEM (standard error mean)


of each commensal is indicated, as well as the ratio of this mean comparing responders (R) versus non responders


(NR) as the fold change and its log2 value with the p value (not considered significantly relevant when <0.05).









Commensal bacteria associated with best (or worse) clinical outcome).

















NR Mean

R Mean

Fold Ratio

P value




rel,

rel,

(FR)
log2FR
NR


Species
CAG
abund,
NR Sem
abund,
R Sem
R/NR
R/NR
vs R


















Part A















NR - KIDNEY/OUTCOME_1/AUC = 0.882; Spec = 0.778; Sens = 0.860
















Clostridium_bolteae_ATCC_BAA_613

CAG00008
0.0069
0.00429
0.000531
0.000133
0.0769
−3.7
0.00999



Bacteroides_sp_CAG_144

CAG00413
2.90E−05
2.06E−05
0.00181
0.000585
62.55
5.96
0.0287



Faecalibacterium_cf_prausnitzii_KLE1255_3

CAG01039
0.00301
0.00208
0.000687
0.000199
0.227
−2.133
0.0351



Eubacterium_sp_CAG_38

CAG00178
0.0012
0.000685
0.0028
0.00072
2.336
1.224
0.0181



Prevotella_sp_CAG_617

CAG00473
0.00326
0.00192
0
0
0
−inf
0.0296



Subdoligranulum_sp_4_3_54A2FAA

CAG00140
0.00338
0.00087
0.00128
0.000195
0.379
−1.398
0.0204



Prevotella_copri_CAG_164

CAG01004
0.000576
0.000558
0
0
0
−inf
0.0296



Acinetobacter_sp_CAG_196

CAG00761
0.000557
0.000417
0.00191
0.000695
3.44
1.782
0.0186



Azospirillum_sp_CAG_239

CAG00302
0.000452
0.000331
0.00172
0.00083
3.8
1.926
0.0501







R - KIDNEY/OUTCOME_1/AUC = 0.863; Spec = 0.798; Sens = 0.842
















Clostridium_bolteae_ATCC_BAA_613

CAG00008
0.0069
0.00429
0.000531
0.000133
0.0769
−3.7
0.00999



Bacteroides_sp_CAG_144

CAG00413
2.90E−05
2.06E−05
0.00181
0.000585
62.55
5.96
0.0287



Faecalibacterium_cf_prausnitzii_KLE1255_3

CAG01039
0.00301
0.00208
0.000687
0.000199
0.227
−2.133
0.0351



Eubacterium_sp_CAG_38

CAG00178
0.0012
0.000685
0.0028
0.00072
2.336
1.224
0.0181



Prevotella_sp_CAG_617

CAG00473
0.00326
0.00192
0
0
0
−inf
0.0296



Subdoligranulum_sp_4_3_54A2FAA

CAG00140
0.00338
0.00087
0.00128
0.000195
0.379
−1.398
0.0204



Azospirillum_sp_CAG 239

CAG00302
0.000452
0.000331
0.00172
0.00083
3.8
1.926
0.0501


Part B







NR - KIDNEY_noatb/OUTCOME_1/AUC = 0.879; Spec = 0.707; Sens = 0.950
















Clostridium_bolteae_ATCC_BAA_613

CAG00008
0.00806
0.00605
0.000513
0.000142
0.0636
−3.972
0.00974



Bacteroides_sp_CAG_144

CAG00413
3.88E−05
2.91E−05
0.00195
0.000625
50.203
5.649
0.0368


Firmicutes_bacterium_CAG_65_1
CAG00792
0.00413
0.00234
0.00798
0.00259
1.931
0.949
0.0395



Eubacterium_sp_CAG_115

CAG00341
5.33E−05
3.66E−05
0.00252
0.000927
47.381
5.566
0.0148



Prevotella_sp_CAG_617

CAG00473
0.00378
0.00261
0
0
0
−inf
0.0436



Bacteroides_plebeius_CAG_211

CAG00711
1.66E−05
1.66E−05
0.00544
0.00195
327.612
8.355
0.0557



Eubacterium_siraeum_CAG_80

CAG00653
0.000425
0.000185
0.00608
0.00215
14.277
3.835
0.0232



Bacteroides_coprophilus_DSM_18228_JCM_13818

CAG00132
0
0
0.00234
0.00121
0
−inf
0.0533







R - KIDNEY_noatb/OUTCOME_1/AUC = 0.845; Spec = 0.990; Sens = 0.614
















Eubacterium_sp_CAG_115

CAG00341
5.33E−05
3.66E−05
0.00252
0.000927
47.381
5.566
0.0148



Prevotella_sp_CAG_617

CAG00473
0.00378
0.00261
0
0
0
−inf
0.0436



Clostridium_clostridioforme_2_1_49FAA_1

CAG01263
0.0053
0.00356
0.000839
0.000471
0.158
−2.659
0.0415



Bacteroides_plebeius_CAG_211

CAG00711
1.66E−05
1.66E−05
0.00544
0.00195
327.612
8.355
0.0557



Eubacterium_siraeum_CAG_80

CAG00653
0.000425
0.000185
0.00608
0.00215
14.277
3.835
0.0232



Eubacterium_sp_CAG_202

CAG00796
0.000954
0.00095
0.00514
0.00162
5.39
2.43
0.0402



Bacteroides_coprophilus_DSM_18228_JCM_13818

CAG00132
0
0
0.00234
0.00121
0
−inf
0.0533









Example 2: Metagenomics-Based GOMS Associated with TTP>3 Months According to RECIST 1.1 Criteria (FIG. 2)

Metagenomics-based GOMS associated with TTP>3 months according to RECIST 1.1 criteria (FIG. 2).


When segregating responders (R, TTP>3 months) from non-responders (NR, TTP<3 months) in all 69 patients (according to the best clinical response as assessed by RECIST1.1), we failed to observe significant differences with a FDR p value <0.05. When segregating responders (R, TTP>3 months) from non-responders (NR, TTP<3 months) in RCC who did not take ATB (according to the best clinical response as assessed by RECIST1.1), we failed to observe significant differences with a FDR p value <0.05.


However, the multivariate model allowed to find some GOMS with clinical relevance.


GOMS Associated with TTP<3 Months


Resistance at 3 months was the best clinical readout of our analysis in RCC patients.


The best predictive model allowing to identify the “resistant” patients (NR) among all patients with AUC value of 0.913, a specificity of 0.798 (true positive cases) and a sensitivity (false positive cases) of 0.910 was obtained when considering LEfSe combination, and the commensals retained in the model as “bad bacteria” increased in NR were the following, as shown in Table 16:

    • Clostridium_sp_CAG_71 (increased by 2 fold in NR)
    • Hungatella_hathewayi_12489931_1 (increased by 47 fold in NR)
    • Prevotella_sp_CAG_617 (infinite, detectable only in NR) and
    • Holdemanella_biformis_DSM_3989_1 (increased by 48 fold in NR),
    • whereas in Azospirillum_sp_CAG_239 was decrease by 11 fold in NR (good bacterium)


However, in patients who did not took ATB, the list of bacteria retained in this model which gave an AUC of 0.884, specificity 0.667, sensitivity 1.000 were:


For the bad bacteria, increase of:

    • Clostridium_bolteae_ATCC_BAA_613 (increased by 15 fold in NR)
    • Prevotella_sp_CAG_617 (present only in NR)
    • Holdemanella_biformis_DSM 39891 (increased by 32 fold in NR) and
    • Faecalibacterium_cf_prausnitzii_KLE1255 (increased by 8.8 fold in NR)


For the good bacteria, decrease of:

    • Eubacterium_sp_CAG_38 (decreased by 2 fold in NR)
    • Eubacterium_sp_CAG_115 (decreased by 37 fold in NR)


We did retain the ratio between good and bad bacteria for TTP3 NR in our final model (see Table 16, part B).


GOMS Associated with TTP>3 Months


The best predictive model allowing to identify the “responders” patients (R) among all patients, with AUC=0.876; Spec=0.990; Sens=0.727, was obtained when considering LEfSe combination, and the commensals retained in the model as “bad bacteria” decreased in R were the following, as shown in Table 16:

    • Clostridium_bolteae_ATCC_BAA613 (decreased by 92% in R)
    • Clostridium_sp_CA G_7_1 (decreased by 46%)
    • Faecalibacterium_cf_prausnitzii_KLE12553 (decreased by 83%)
    • Prevotella_sp_CAG_617 (present only in NR)
    • Holdemanella_biformis_DSM_39891 (decreased by 79%)
    • Peptoniphilus_sp_oral_taxon_836_str_F0141 (present only in NR)
    • whereas Azospirillum_sp_CAG_239 was increased in R by 11 fold (good bacterium).


However, in patients who did not took ATB, the list of bacteria retained in this model which gave AUC=0.899; Spec=0.990; Sens=0.836 were:


For the bad bacteria, a decrease in:

    • Clostridium_bolteae_ATCC_BAA_613 (decreased by 93.2% in R)
    • Clostridium_sp_CAG_7_1 (decreased by 44.5%)
    • Prevotella_sp_CAG_617 (present only in NR)
    • Prevotella_copri_CAG_164 (present only in NR)
    • Holdernanella_biformis_DSM_3989_1 (decreased by 96.9%)
    • Prevotella_sp_CAG_2791 (present only in NR)
    • Faecalibacterium_of_prausnitzii_KLE1255 (decreased by 88.6%)


For the good bacteria, an increase in:

    • Akkermansia_muciniphila_CAG_154 (2.5 fold increase in R)
    • Eubacterium_sp_CAG_115 (37 fold increase in R)


We did retain the ratio between good and bad bacteria for TTP3 R in our final model (see Table 16, part B).









TABLE 16







Commensal bacteria associated with TTP> or <3 months. Part A includes all RCC patients while part B considered


only patients who did not take antibiotics. The mean relative abundance with SEM (standard error mean) of each


commensal is indicated, as well as the ratio of this mean comparing responders (R) versus non responders (NR)


as the fold change and its log2 value with the p value (not considered significantly relevant when <0.05).









Commensal bacteria associated with TTP> or <3 months.

















NR Mean

R Mean

Fold Ratio

P value




rel,

rel,

(FR)
log2FR
NR


Species
CAG
abund,
NR Sem
abund,
R Sem
R/NR
R/NR
vs R


















Part A















NR - KIDNEY/TTP3/AUC = 0.913; Spec = 0.798; Sens = 0.910
















Clostridium_sp_CAG_7_1

CAG00047
0.0031
0.000679
0.00166
0.000576
0.537
−0.895
0.00404



Hungatella_hathewayi_12489931_1

CAG00610
0.000963
0.000659
2.15E−05
9.31E−06
0.0223
−5.483
0.0118



Prevotella_sp_CAG_617

CAG00473
0.004
0.00233
0
0
0
−inf
0.0107



Holdemanella_biformis_DSM_3989_1

CAG01141
0.00249
0.0013
5.19E−05
5.19E−05
0.0208
−5.586
0.0154



Azospirillum_sp_CAG_239

CAG00302
0.000157
0.00011
0.00172
0.00076
10.914
 3.448
0.0454







R - KIDNEY/TTP3/AUC = 0.876; Spec = 0.990; Sens = 0.727
















Clostridium_bolteae_ATCC_BAA_613

CAG00008
0.00813
0.00525
0.000635
0.000163
0.0781
−3.678
0.0387



Clostridium_sp_CAG_7_1

CAG00047
0.0031
0.000679
0.00166
0.000576
0.537
−0.895
0.00404



Faecalibacterium_cf_prausnitzii_KLE1255_3

CAG01039
0.00363
0.00254
0.000645
0.00018
0.177
−2.494
0.0242



Prevotella_sp_CAG_617

CAG00473
0.004
0.00233
0
0
0
−inf
0.0107



Holdemanella_biformis_DSM_3989_1

CAG01141
0.00249
0.0013
5.19E−05
5.19E−05
0.0208
−5.586
0.0154



Azospirillum_sp_CAG_239

CAG00302
0.000157
0.00011
0.00172
0.00076
10.914
 3.448
0.0454



Peptoniphilus_sp_oral_taxon_836_str_F0141

CAG01339
6.19E−06
5.00E−06
0
0
0
−inf
0.0393


Part B







NR - KIDNEY_noatb/TTP3/AUC = 0.884; Spec = 0.667; Sens = 1.000
















Clostridium_bolteae_ATCC_BAA_613

CAG00008
0.00919
0.00718
0.000619
0.000181
0.0673
−3.892
0.0242



Eubacterium_sp_CAG_38

CAG00178
0.00141
0.00112
0.00281
0.000721
1.998
 0.998
0.011



Eubacterium_sp_CAG_115

CAG00341
6.32E−05
4.32E−05
0.00234
0.000866
37.05
 5.211
0.0472



Prevotella_sp_CAG_617

CAG00473
0.00449
0.00308
0
0
0
−inf
0.0223



Holdemanella_biformis_DSM_3989_1

CAG01141
0.00187
0.00102
5.81E−05
5.81E−05
0.031
−5.009
0.0265



Faecalibacterium_cf_prausnitzii_KLE1255

CAG00495
0.00342
0.00215
0.00039
0.000277
0.114
−3.132
0.0397







R - KIDNEY_noatb/TTP3/AUC = 0.899; Spec = 0.990; Sens = 0.836
















Clostridium_bolteae_ATCC_BAA_613

CAG00008
0.00919
0.00718
0.000619
0.000181
0.0673
−3.892
0.0242



Clostridium_sp_CAG_7_1

CAG00079
0.00319
0.000675
0.00177
0.000642
0.557
−0.843
0.00285



Akkermansia_muciniphila_CAG_154

CAG00301
0.00228
0.00209
0.00571
0.00159
2.495
 1.319
0.016



Eubacterium_sp_CAG_115

CAG00341
6.32E−05
4.32E−05
0.00234
0.000866
37.05
 5.211
0.0472



Prevotella_sp_CAG_617

CAG00473
0.00449
0.00308
0
0
0
−inf
0.0223



Prevotella_copri_CAG_164

CAG01004
0.000972
0.000941
0
0
0
−inf
0.00461



Holdemanella_biformis DSM_3989_1

CAG01141
0.00187
0.00102
5.81E−05
5.81E−05
0.031
−5.009
0.0265



Prevotella_sp_CAG_279_1

CAG00487
0.000701
0.000568
0
0
0
−inf
0.0223



Faecalibacterium_cf_prausnitzii_KLE1255

CAG00495
0.00342
0.00215
0.00039
0.000277
0.114
−3.132
0.0397









Example 3: Metagenomics-Based GOMS Associated with TTP>6 Months According to RECIST 1.1 Criteria (FIG. 3)

When segregating responders (R, TTP>6 months) from non-responders (NR, TTP<6 months) in all ROC patients (according to the TTP6 as assessed by RECIST1.1), we did not observe significant differences with a FDR p value <0.05 (not shown). When segregating responders (R, TTP>6 months) from non-responders (NR, TTP<6 months) in RCC who did not take ATB (according to the TTP6 as assessed by RECIST1.1), we observed significant differences with a FDR p value <0.05 (FIG. 4). Of note, Subdoligranulum_sp_CAG_314_1 and Eubacterium_sp_CAG_115 were associated with TTP>6 months in RCC patients who took no antibiotics.


We conducted the multivariate model allowing to find very robust GOMS with clinical relevance.


GOMS Associated with TTP>6 Months


The best predictive model allowing to identify the “responding” patients (R) among all patients with AUC value of 0.845, a specificity of 0.889 (true positive cases) and a sensitivity (false positive cases) of 0.800 was obtained when considering LEfSe combination, and the good commensals retained in the model were the following, as shown in Table 17 (part A):

    • Clostridium_sp_CAG_230 (increased by 6.7 fold in R)
    • Clostridium_sp_CAG_167 (increased by 2.8 fold in R)
    • Azospirilium_sp_CAG_239_3 (detectable only in R)
    • Firmicutes_bacterium_CAG_176_12 (increased by 6.9 fold in R)


The bad commensals that should be decreased are:

    • Coprococcus_catus_GD_7_6 (decreased by 65.6%)
    • Oscillibacter_sp_KLE_1745_6 (decreased by 56.6%)


The cut-off values correspond to the means±SEM featuring in Table 17.


However, in patients who did not took ATB, the list of favorable bacteria retained in this model was different and included the following ones, reaching AUC value of 0.804, a specificity of 0.848 (true positive cases) and a sensitivity (false positive cases) of 0.770 was obtained when considering LEfSe combination (Table 17 part B):

    • Eubacterium_sp_CAG_1801 (increased by 3 fold in R)
    • Eubacterium_sp_CAG_115 (increased by 13 fold in R)
    • Eubacterium_siraeum_CAG_80 (increased by 5.4 fold in R)
    • Butyrivibrio_crossotus_DSM_2876 (detectable only in R)


The cut-off values correspond to the means±SEM featuring in Table 17. These favorable and unfavorable commensals are major spp influencing favorable responses to PD1 blockade.


GOMS Associated with TTP<6 Months


As for the bacteria associated with short TTP<6 months in all patients, the list of significant bacteria was very restrained even more without ATB. Very few can be found significantly associated with resistance:

    • Clostridialas_bacterium_VE202_14_1 (undetectable in NR)
    • Clostridium_sp_CAG_230 (6.7 fold increase in R)
    • Azospirillum_sp_CAG 239_1 (undetectable in NR).









TABLE 17







Commensal bacteria associated with TTP> or <6 months. Part A includes all RCC patients while part B considered


only patients who did not take antibiotics. The mean relative abundance with SEM (standard error mean) of each


commensal is indicated, as well as the ratio of this mean comparing responders (R) versus non responders (NR)


as the fold change and its log2 value with the p value (not considered significantly relevant when <0.05).









Commensal bacteria associated with TTP> or <6 months.

















NR Mean

R Mean

Fold Ratio

P value




rel,

rel,

(FR)
log2FR
NR


Species
CAG
abund,
NR Sem
abund,
R Sem
R/NR
R/NR
vs R


















Part A















NR - KIDNEY/TTP6/AUC = 0.796; Spec = 0.828; Sens = 0.688
















Clostridium_bolteae_ATCC_BAA_613

CAG00008
0.00477
0.00285
0.000508
0.000154
0.106
−3.229 
0.0623



Alistipes_sp_CAG_514_1

CAG00827
0
0
0.00618
0.00562
0
−inf
0.0714



Clostridiales_bacterium_VE202_14_1

CAG00821
0
0
1.88E−05
1.13E−05
0
−inf
0.0254



Clostridium_sp_CAG_230

CAG00317
9.10E−05
6.01E−05
0.000612
0.000309
6.733
2.751
0.00989



Azospirillum_sp_CAG_239_1

CAG00472
0
0
0.000412
0.000328
0
−inf
0.0714



Azospirillum_sp_CAG_239_3

CAG00668
0
0
0.000816
0.000574
0
−inf
0.0254



Alistipes_sp_CAG_268

CAG00421
0
0
0.002
0.00193
0
−inf
0.0714







R - KIDNEY/TTP6/AUC = 0.845; Spec = 0.889; Sens = 0.800
















Coprococcus_catus_GD_7_6

CAG01208
0.000926
0.000278
0.000318
9.32E−05
0.343
−1.541 
0.103



Clostridium_sp_CAG_230

CAG00317
9.10E−05
6.01E−05
0.000612
0.000309
6.733
2.751
0.00989



Clostridium_sp_CAG_167

CAG00452
0.000271
0.000164
0.000781
0.00029
2.878
1.525
0.0277



Oscillibacter_sp_KLE_1745_6

CAG00931
0.000795
0.000203
0.000345
7.49E−05
0.433
−1.204 
0.0367



Azospirillum_sp_CAG_239_3

CAG00668
0
0
0.000816
0.000574
0
−inf
0.0254


Firmicutes_bacterium_CAG_176_12
CAG01043
5.85E−05
5.85E−05
0.000404
0.00025
6.912
2.789
0.0501


Part B







NR - KIDNEY_noatb/TTP6/AUC = 0.721; Spec = 0.798; Sens = 0.606















Firmicutes_bacterium_CAG_110_1
CAG00604
0.00166
0.000682
0.00201
0.000735
1.212
0.277
0.207







R - KIDNEY_noatb/TTP6/AUC = 0.804; Spec = 0.848; Sens = 0.770
















Eubacterium_sp_CAG_180_1

CAG00523
0.00221
0.00124
0.00672
0.00195
3.039
1.603
0.0272



Eubacterium_sp_CAG_115

CAG00341
0.000274
0.000241
0.00355
0.0015
12.961
3.696
0.000501



Eubacterium_siraeum_CAG_80

CAG00653
0.00161
0.00095
0.00872
0.00348
5.396
2.431
0.0252



Butyrivibrio_crossotus_DSM_2876

CAG00542
0
0
0.0136
0.00801
0
−inf
0.0324









Example 4: Results for RCC Patients Regardless Antibiotic Usage, OUTCOME 1 Criterion

In this example, three different models were built based on a bundle of 69 “reference patterns”, which are the standardized abundances profiles (zero mean and unit variance) obtained from the 69 samples used in training the logistic regression models. Standardization is necessary to homogenize the dynamic range of bacterial species abundances.


Based on the first strategy described in the Materials and Methods, applied to 69 RCC patients (NR=27; R=42) with unknown antibiotic usage history and OUTCOME_1 criterion (best outcome), we selected 25 bacterial species that gave a logistic regression model overall predictability equal to 89.86% (model 1.1). In order to measure the goodness of the model with 25 species, after a 5-fold cross-validation performed on a subset of the original dataset, the model itself was able to predict 60% of NR and 69% of R subjects, with a CV_AUC=0.917 and an AUC=0.785. Upon RFECV and RFE feature selection, 8 species were selected from the aforementioned 25: the model built on these 8 species gave an overall predictability of 79.71% (model 1.2, FIG. 5), and the summary statistics for these species are reported in Table 18 below. In order to measure the goodness of the model, after a 5-fold cross-validation performed on a subset of the original dataset, the model itself was able to predict 40% of NR and 77% of R subjects, with a CV_AUC=0.635 and an AUC=0.815.


Based on the second strategy (machine-learning) applied to 69 RCC patients (NR=27; R=42) with unknown antibiotic usage history and OUTCOME_1 criterion, we took into account all the bacterial species of the dataset (n=1112). After a first round of feature selection, RFECV gave a rank=1 to 31 species which collectively, after a 5-fold cross-validation, were able to predict 100% of NR and 100% of R subjects, with a CV_AUC=1 and an AUC=1 (model 2.1). Upon a second round of feature selection, RFE gave a rank=1 to 8 species, which were selected for the subsequent logistic regression classifier. The model built on these 8 species gave an overall predictability of 89.86% (model 2.2, FIG. 5), and the summary statistics for these species are reported in Table 19 below. In order to measure the goodness of the model, after a 5-fold cross-validation performed on a subset of the original dataset, the model itself was able to predict 80% of NR and 92% of R subjects, with a CV_AUC=1 and an AUC=0.923.









TABLE 18





Statistics for the species selected in Model 1.2.


























NR (n =
R (n =


NR_Odds



Group
op.
Thr
27) (%)
42) (%)
Pval_Fisher
NR_coeff
Ratio
R_coeff





CAG00008_Clostridium_bolteae_ATCC_BAA_613
>
0
92.6
95.2
0.6415
1.0323
2.0453
−1.0323


CAG01039_Faecalibacterium_cf_prausnitzii_KLE1255
>
0
77.8
47.6
0.0229
1.084
2.1199
−1.084


CAG00473_Prevotella_sp_CAG_617
>
0
11.1
0.0
0.0558
0.9723
1.9619
−0.9723


CAG00140_Subdoligranulum_sp_4_3_54A2FAA
>
0
92.6
100.0
0.1496
0.9907
1.9872
−0.9907


CAG00610_Hungatella_hathewayi_12489931
>
0
51.9
21.4
0.0174
0.6234
1.5406
−0.6234


CAG01141_Holdemanella_biformis_DSM_3989
>
0
14.8
2.4
0.0728
0.8468
1.7986
−0.8468


CAG00413_Bacteroides_sp_CAG_144
>
0
11.1
31.0
0.0803
−0.8994
0.5361
0.8994


CAG00317_Clostridium_sp_CAG_230
>
0
11.1
35.7
0.027
−0.6041
0.6579
0.6041
















R_Odds


NR Mean



Group
Ratio
rank_RFECV
rank_RFE
relabund
NR Sem





CAG00008_Clostridium_bolteae_ATCC_BAA_613
0.4889
1
1
0.0069045
0.00429957


CAG01039_Faecalibacterium_cf_prausnitzii_KLE1255
0.4717
1
1
0.00301567
0.0020844


CAG00473_Prevotella_sp_CAG_617
0.5097
1
1
0.00326379
0.00192


CAG00140_Subdoligranulum_sp_4_3_54A2FAA
0.5032
1
1
0.00338708
0.00087016


CAG00610_Hungatella_hathewayi_12489931
0.6491
1
1
0.00079124
0.00053962


CAG01141_Holdemanella_biformis_DSM_3989
0.556
1
1
0.00203209
0.00107945


CAG00413_Bacteroides_sp_CAG_144
1.8653
1
1
2.895e−05
2.056e−05


CAG00317_Clostridium_sp_CAG_230
1.52
1
1
2.568e−05
1.459e−05


















R Mean

Fold Ratio
log2FR
P value



Group
relabund
R Sem
(FR) R/NR
R/NR
NR vs R







CAG00008_Clostridium_bolteae_ATCC_BAA_613
0.00053101
0.00013359
0.08
−3.7
0.00999



CAG01039_Faecalibacterium_cf_prausnitzii_KLE1255
0.00068723
0.00019997
0.23
−2.13
0.03517



CAG00473_Prevotella_sp_CAG_617
0.0
0.0
0.0
−inf
0.02965



CAG00140_Subdoligranulum_sp_4_3_54A2FAA
0.0012847
0.00019541
0.38
−1.4
0.02047



CAG00610_Hungatella_hathewayi_12489931
2.023e−05
1.029e−05
0.03
−5.29
0.00354



CAG01141_Holdemanella_biformis_DSM_3989
5.809e−05
5.809e−05
0.03
−5.13
0.0489



CAG00413_Bacteroides_sp_CAG_144
0.0018108
0.00058512
62.56
5.97
0.02879



CAG00317_Clostridium_sp_CAG_230
0.00047284
0.00019556
18.41
4.2
0.0214

















TABLE 19





Statistics for the species selected in Model 2.2.


























NR (n =
R (n =


NR_Odds



Group
op.
Thr
27) (%)
42) (%)
Pval_Fisher
NR_coeff
Ratio
R_coeff





CAG00727_Eggerthella_lenta_DSM_2243
>
0
77.8
50.0
0.0251
1.0791
2.1127
−1.0791


CAG01141_Holdemanella_biformis_DSM_3989
>
0
14.8
2.4
0.0728
1.1164
2.1681
−1.1164


CAG00650_Dorea_formicigenerans_ATCC_27755
>
0
81.5
88.1
0.4972
1.2839
2.4349
−1.2839


CAG00928_Acidiphilium_sp_CAG_727
>
0
14.8
4.8
0.2006
1.1156
2.1668
−1.1156


CAG00473_Prevotella_sp_CAG_617
>
0
11.1
0.0
0.0558
1.014
2.0194
−1.014


CAG00122_Coprococcus_catus_GD_7
>
0
11.1
0.0
0.0558
0.9394
1.9177
−0.9394


CAG01144_Lactobacillus_vaginalis_DSM_5837_ATCC_49540
>
0
7.4
0.0
0.1496
0.8085
1.7514
−0.8085


CAG00063_Barnesiella_viscericola_DSM_18177
>
0
7.4
0.0
0.1496
0.7679
1.7028
−0.7679
















R_Odds


NR Mean



Group
Ratio
rank_RFECV
rank_RFE
relabund
NR Sem





CAG00727_Eggerthella_lenta_DSM_2243
0.4733
1
1
0.00062355
0.00025745


CAG01141_Holdemanella_biformis_DSM_3989
0.4612
1
1
0.00203209
0.00107945


CAG00650_Dorea_formicigenerans_ATCC_27755
0.4107
1
1
0.00113438
0.0003259


CAG00928_Acidiphilium_sp_CAG_727
0.4615
1
1
0.00021637
0.00013333


CAG00473_Prevotella_sp_CAG_617
0.4952
1
1
0.00326379
0.00192


CAG00122_Coprococcus_catus_GD_7
0.5214
1
1
0.00015959
0.00010288


CAG01144_Lactobacillus_vaginalis_DSM_5837_ATCC_49540
0.571
1
1
4.76e−06
3.39e−06


CAG00063_Barnesiella_viscericola_DSM_18177
0.5873
1
1
0.00049676
0.00035498


















R Mean


log2FR
P value



Group
relabund
R Sem
FR R/NR
R/NR
NR vs R







CAG00727_Eggerthella_lenta_DSM_2243
9.683e−05
2.923e−05
0.16
−2.69
0.00559



CAG01141_Holdemanella_biformis_DSM_3989
5.809e−05
5.809e−05
0.03
−5.13
0.0489



CAG00650_Dorea_formicigenerans_ATCC_27755
0.00041466
6.136e−05
0.37
−1.45
0.18765



CAG00928_Acidiphilium_sp_CAG_727
 5.87e−06
 4.1e−06
0.03
−5.2
0.14111



CAG00473_Prevotella_sp_CAG_617
0.0
0.0
0.0
−inf
0.02965



CAG00122_Coprococcus_catus_GD_7
0.0
0.0
0.0
−inf
0.02965



CAG01144_Lactobacillus_vaginalis_DSM_5837_ATCC_49540
0.0
0.0
0.0
−inf
0.07916



CAG00063_Barnesiella_viscericola_DSM_18177
0.0
0.0
0.0
−inf
0.07916










Taking into account the bacterial species (n=181, above 1112 total species, 16.3% of the total microbial composition) having an average relative abundance greater than or equal to 0.001 if expressed in the interval [0:1] (thus, the 0.1%) within the overall RCC cohort, we applied the second strategy (machine-learning) for 69 RCC patients (NR=27; R=42) with unknown antibiotic usage history and OUTCOME_1 criterion. After a first round of feature selection, RFECV gave a rank=1 to 21 species which collectively, after a 5-fold cross-validation, were able to predict 100% of NR and 92% of R subjects, with a CV_AUC=0.573 and an AUC=1 (model 3.1). After a second round of feature selection, RFE gave a rank=1 to 8 species which were selected for the subsequent logistic regression classifier. The model built on these 8 species gave an overall predictability of 85.51% (model 3.2, FIG. 5), and the summary statistics for these species are reported in Table 20 below. In order to measure the goodness of the model, after a 5-fold cross-validation performed on a subset of the original dataset, the model itself was able to predict 80% of NR and 85% of R subjects, with a CV_AUC=0.635 and an AUC=0.831. This last model was designed to be more affordable in case of problems in fecal samples storage, sequencing, stiffness, etcetera.


According to the Models 1.2, 2.2 and 3.2, which were modeled on unknown antibiotic usage history and OUTCOME_1 criterion, and are all based on 8 bacterial species so they can be properly compared, it is shown that the Model 2.2 has the highest predictability (89.86%) with the highest CV_AUC and AUC.









TABLE 20





Statistics for the species selected in Model 3.2.


























NR (n =
R (n =






Group
op.
Thr
27) (%)
42) (%)
Pval_Fisher
NR_coeff
NR_OddsRatio
R_coeff





CAG00243_Lachnospiraceae_bacterium_1_1_57FAA
>
0
85.2
90.5
0.7024
1.1735
2.2555
−1.1735


CAG00473_Prevotella_sp_CAG_617
>
0
11.1
0.0
0.0558
1.027
2.0378
−1.027


CAG00327_Faecalibacterium_sp_CAG_74
>
0
44.4
59.5
0.3226
0.9968
1.9956
−0.9968


CAG01039_Faecalibacterium_cf_prausnitzii_KLE1255
>
0
77.8
47.6
0.0229
1.0043
2.006
−1.0043


CAG00140_Subdoligranulum_sp_4_3_54A2FAA
>
0
92.6
100.0
0.1496
0.7456
1.6767
−0.7456


CAG01263_Clostridium_clostridioforme_2_1_49FAA
>
0
81.5
78.6
1.0
0.8736
1.8322
−0.8736


CAG00037_Bacteroides_faecis_MAJ27
>
0
14.8
28.6
0.2478
−0.7032
0.6142
0.7032


CAG00357_Bacteroides_ovatus_V975
>
0
92.6
92.9
1.0
0.6073
1.5234
−0.6073
















R_Odds


NR Mean



Group
Ratio
rank_RFECV
rank_RFE
relabund
NR Sem





CAG00243_Lachnospiraceae_bacterium_1_1_57FAA
0.4434
1
1
0.00861298
0.00346076


CAG00473_Prevotella_sp_CAG_617
0.4907
1
1
0.00326379
0.00192


CAG00327_Faecalibacterium_sp_CAG_74
0.5011
1
1
0.00212113
0.00139362


CAG01039_Faecalibacterium_cf_prausnitzii_KLE1255
0.4985
1
1
0.00301567
0.0020844


CAG00140_Subdoligranulum_sp_4_3_54A2FAA
0.5964
1
1
0.00338708
0.00087016


CAG01263_Clostridium_clostridioforme_2_1_49FAA
0.5458
1
1
0.00452728
0.00253406


CAG00037_Bacteroides_faecis_MAJ27
1.6282
1
1
0.00031624
0.00019911


CAG00357_Bacteroides_ovatus_V975
0.6564
1
1
0.01574035
0.00485731


















R Mean


log2FR
P value



Group
relabund
R Sem
FR R/NR
R/NR
NR vs R







CAG00243_Lachnospiraceae_bacterium_1_1_57FAA
0.00153833
0.00031904
0.18
−2.49
0.26546



CAG00473_Prevotella_sp_CAG_617
0.0
0.0
0.0
−inf
0.02965



CAG00327_Faecalibacterium_sp_CAG_74
0.00043694
0.00012343
0.21
−2.28
0.92773



CAG01039_Faecalibacterium_cf_prausnitzii_KLE1255
0.00068723
0.00019997
0.23
−2.13
0.03517



CAG00140_Subdoligranulum_sp_4_3_54A2FAA
0.0012847
0.00019541
0.38
−1.4
0.02047



CAG01263_Clostridium_clostridioforme_2_1_49FAA
0.0009867
0.00045528
0.22
−2.2
0.05257



CAG00037_Bacteroides_faecis_MAJ27
0.00320497
0.00119881
10.13
3.34
0.12006



CAG00357_Bacteroides_ovatus_V975
0.00872078
0.00225535
0.55
−0.85
0.27645










Example 5: Predictive Models for RCC Patients Regardless Antibiotic Usage, OUTCOME 1 Criterion

According to the results obtained in Example 4, and due to the fact that responders (R) are predicted better than non-responders (NR) in all the three models proposed with 8 species (FIG. 5), we took into consideration the logistic regression β coefficients relative to R.


For the Model 1.2, according to the β coefficients of R (R_coeff), the probability estimate (expressed as a percentage) of belonging to R cohort for a tested subject JD is calculated as follows:






P
R(JD)=1/[1 exp{circumflex over ( )}−(−1.0323*CAG00008+





−1.084*CAG01039+





−0.9723*CAG00473+





−0.9907*CAG00140+





−0.6234*CAG00610+−0.8468*CAG01141+





0.8994*CAG00413+





0.6041*CAG00317)]  [3]


In equation [3], “CAGXXXXX” represents the relative abundance of the bacterial species defined by the recited CAG, in a stool sample from the tested subject JD. These relative abundances need to be expressed as standardized values (zero mean and unit variance). The best option is to standardize the “abundances pattern” of the tested subject with a bundle of known “reference patterns”, i.e., all the relative abundances of each bacterial species for each fecal sample used in training the logistic regression model and the present invention refers a claim of a Python script used to ease the computation of the PR (JD). From the standardized abundance pattern of a tested subject, the equation [3] can be applied to predict his/her belonging to the NR cohort with a model predictability of 79.71% and a success rate of 77%.


For the Model 2.2, according to the β coefficients of R (R_coeff), the probability estimate (expressed as a percentage) of belonging to R cohort for a tested subject JD is calculated as follows:






P
R(JD)=1/[1+exp{circumflex over ( )}−(−1.0791*CAG00727+





−1.1164*CAG01141+





−1.2839*CAG00650+





−1.1156*CAG00928+





−1.014*CAG00473+





−0.9394*CAG00122+





−0.8085*CAG01144+





−0.7679*CAG00063)]  [4]


In equation [4], “CAGXXXXX” represents the relative abundance of the bacterial species defined by the recited CAG, in a stool sample from the tested subject JD. These relative abundances need to be expressed as standardized values (zero mean and unit variance). The best option is to standardize the “abundances pattern” of the tested subject with a bundle of known “reference patterns”, and the present invention refers a claim of a Python script used to ease the computation of the PR (JD). From the standardized abundance pattern of a tested subject, the equation [4] can be applied to predict his/her belonging to the R cohort with a model predictability of 89.86% and a success rate of 92%.


For the Model 3.2, according to the β coefficients of R (R_coeff), the probability estimate (expressed as a percentage) of belonging to R cohort for a tested subject JD is calculated as follows:






P
R(JD)=1/[1+exp{circumflex over ( )}−(−1.1735*CAG00243+





−1.027*CAG00473+





−0.9968*CAG00327+





−1.0043*CAG01039+





−0.7456*CAG00140+





−0.8736*CAG01263+





0.7032*CAG00037+





−0.6073*CAG00357)]  [5]


In equation [5], “CAGXXXXX” represents the relative abundance of the bacterial species defined by the recited CAG, in a stool sample from the tested subject JD. These relative abundances need to be expressed as standardized values (zero mean and unit variance). The best option is to standardize the “abundances pattern” of the tested subject with a bundle of known “reference patterns”, and the present invention refers a claim of a Python script used to ease the computation of the PR (JD). From the standardized abundance pattern of a tested subject, the equation [5] can be applied to predict his/her belonging to the R cohort with a model predictability of 85.51% and a success rate of 85%.


Example 6: Results for RCC Patients with No Antibiotic Usage, OUTCOME 1 Criterion

In this example, three different models were built based on a bundle of 58 “reference patterns”, which are the standardized abundances profiles (zero mean and unit variance) obtained from the 58 samples used in training the logistic regression models. Standardization is necessary to homogenize the dynamic range of bacterial species abundances.


Based on the first strategy applied to 58 RCC patients (NR=19; R=39) with no antibiotic usage history and OUTCOME_1 criterion (best outcome), we selected 25 bacterial species that gave a logistic regression model overall predictability equal to 89.66% (model 4.1). In order to measure the goodness of the model with 25 species, after a 5-fold cross-validation performed on a subset of the original dataset, the model itself was able to predict 67% of NR and 100% of R subjects, with a CV_AUC=1 and an RUC=0.778. Upon RFECV and RFE feature selection, 8 species were selected from the aforementioned 25: the model built on these 8 species gave an overall predictability of 87.93% (model 4.2, FIG. 6), and the summary statistics for these species are reported in Table 21 below. In order to measure the goodness of the model, after a 5-fold cross-validation performed on a subset of the original dataset, the model itself was able to predict 100% of NR and 92% of R subjects, with a CV_AUC=1 and an AUC=1.


Based on the second strategy (machine-learning) applied to 58 RCC patients (NR=19; R=39) with no antibiotic usage history and OUTCOME_1 criterion, we took into account all the bacterial species of the dataset (n=1083). After a first round of feature selection, RFECV gave a rank=1 to 9 species which collectively, after a 5-fold cross-validation, were able to predict 100% of NR and 92% of R subjects, with a CV_AUC=1 and an AUC=1 (model 5.1). Upon a second round of feature selection, RFE gave a rank=1 to 8 species, which were selected for the subsequent logistic regression classifier. The model built on these 8 species gave an overall predictability of 93.1% (model 5.2, FIG. 6), and the summary statistics for these species are reported in Table 22 below. In order to measure the goodness of the model, after a 5-fold cross-validation performed on a subset of the original dataset, the model itself was able to predict 100% of NR and 92% of R subjects, with a CV_AUC=1 and an AUC=1.









TABLE 21





Statistics for the species selected in Model 4.2.


























NR (n =
R (n =


NR_Odds



Group
op.
Thr
19) (%)
39) (%)
Pval_Fisher
NR_coeff
Ratio
R_coeff





CAG00008_Clostridium_bolteae_ATCC_BAA_613
>
0
94.7
94.9
1.0
0.955
1.9386
−0.955


CAG01039_Faecalibacterium_cf_prausnitzii_KLE1255
>
0
73.7
51.3
0.1561
0.8761
1.8354
−0.8761


CAG00473_Prevotella_sp_CAG_617
>
0
10.5
0.0
0.1034
0.9951
1.9932
−0.9951


CAG00487_Prevotella_sp_CAG_279
>
0
10.5
0.0
0.1034
0.7609
1.6945
−0.7609


CAG00140_Subdoligranulum_sp_4_3_54A2FAA
>
0
89.5
100.0
0.1034
1.1107
2.1595
−1.1107


CAG01141_Holdemanella_biformis_DSM_3989
>
0
15.8
2.6
0.0982
0.9402
1.9188
−0.9402


CAG01208_Coprococcus_catus_GD_7
>
0
84.2
69.2
0.3399
0.7187
1.6456
−0.7187


CAG00413_Bacteroides_sp_CAG_144
>
0
10.5
33.3
0.1084
−0.5812
0.6684
0.5812



















NR Mean



Group
R_OddsRatio
rank_RFECV
rank_RFE
relabund
NR Sem





CAG00008_Clostridium_bolteae_ATCC_BAA_613
0.5158
1
1
0.00806075
0.00605804


CAG01039_Faecalibacterium_cf_prausnitzii_KLE1255
0.5449
1
1
0.00364533
0.00295908


CAG00473_Prevotella_sp_CAG_617
0.5017
1
1
0.00378162
0.00261518


CAG00487_Prevotella_sp_CAG_279
0.5901
1
1
0.00059085
0.00047993


CAG00140_Subdoligranulum_sp_4_3_54A2FAA
0.4631
1
1
0.00380225
0.00120314


CAG01141_Holdemanella_biformis_DSM_3989
0.5212
1
1
0.0015759
0.00087372


CAG01208_Coprococcus_catus_GD_7
0.6077
1
1
0.00134292
0.00056097


CAG00413_Bacteroides_sp_CAG_144
1.4961
1
1
3.884e−05
2.907e−05


















R Mean


log2FR
P value



Group
relabund
R Sem
FR R/NR
R/NR
NR vs R







CAG00008_Clostridium_bolteae_ATCC_BAA_613
0.00051335
0.00014219
0.06
−3.97
0.00975



CAG01039_Faecalibacterium_cf_prausnitzii_KLE1255
0.0007401
0.00021316
0.2
−2.3
0.21913



CAG00473_Prevotella_sp_CAG_617
0.0
0.0
0.0
−inf
0.04363



CAG00487_Prevotella_sp_CAG_279
0.0
0.0
0.0
−inf
0.04363



CAG00140_Subdoligranulum_sp_4_3_54A2FAA
0.00128653
0.00020791
0.34
−1.56
0.08193



CAG01141_Holdemanella_biformis_DSM_3989
6.256e−05
6.256e−05
0.04
−4.65
0.05935



CAG01208_Coprococcus_catus_GD_7
0.00040444
7.399e−05
0.3
−1.73
0.36682



CAG00413_Bacteroides_sp_CAG_144
0.00195009
0.00062503
50.2
5.65
0.03684

















TABLE 22





Statistics for the species selected in Model 5.2.


























NR (n =
R (n =


NR_Odds



Group
op.
Thr
19) (%)
39) (%)
Pval_Fisher
NR_coeff
Ratio
R_coeff





CAG00048_1_Clostridium_sp_CAG_226
>
0
21.1
5.1
0.0828
0.7112
1.6372
−0.7112


CAG00300_Prevotella_sp_CAG_891
>
0
15.8
0.0
0.0314
0.8931
1.8571
−0.8931


CAG00473_Prevotella_sp_CAG_617
>
0
10.5
0.0
0.1034
1.0042
2.0058
−1.0042


CAG00650_Dorea_formicigenerans_ATCC_27755
>
0
84.2
92.3
0.3822
1.1282
2.1858
−1.1282


CAG00720_Anaerotruncus_colihominis_DSM_17241
>
0
84.2
69.2
0.3399
1.2569
2.3898
−1.2569


CAG00727_Eggerthella_lenta_DSM_2243
>
0
78.9
48.7
0.0458
1.2397
2.3615
−1.2397


CAG00963_Clostridium_sp_CAG_524
>
0
10.5
0.0
0.1034
0.7595
1.6929
−0.7595


CAG01141_Holdemanella_biformis_DSM_3989
>
0
15.8
2.6
0.0982
1.0235
2.0328
−1.0235
















R_Odds


NR Mean



Group
Ratio
rank RFECV
rank RFE
relabund
NR Sem





CAG00048_1_Clostridium_sp_CAG_226
0.6108
1
1
6.032e−05
3.159e−05


CAG00300_Prevotella_sp_CAG_891
0.5385
1
1
7.957e−05
5.076e−05


CAG00473_Prevotella_sp_CAG_617
0.4986
1
1
0.00378162
0.00261518


CAG00650_Dorea_formicigenerans_ATCC_27755
0.4575
1
1
0.00129344
0.00043426


CAG00720_Anaerotruncus_colihominis_DSM_17241
0.4184
1
1
0.00086591
0.00038685


CAG00727_Eggerthella_lenta_DSM_2243
0.4235
1
1
0.00063799
0.00031339


CAG00963_Clostridium_sp_CAG_524
0.5907
1
1
5.774e−05
4.984e−05


CAG01141_Holdemanella_biformis_DSM_3989
0.4919
1
1
0.0015759 
0.00087372


















R Mean


log2FR
P value



Group
relabund
R Sem
FR R/NR
R/NR
NR vs R







CAG00048_1_Clostridium_sp_CAG_226
 3.2e−06
 2.68e−06
0.05
−4.24
0.05012



CAG00300_Prevotella_sp_CAG_891
0.0
0.0
0.0
−inf
0.01229



CAG00473_Prevotella_sp_CAG_617
0.0
0.0
0.0
−inf
0.04363



CAG00650_Dorea_formicigenerans_ATCC_27755
0.00044294
6.381e−05
0.34
−1.55
0.1252



CAG00720_Anaerotruncus_colihominis_DSM_17241
0.00010478
 2.71e−05
0.12
−3.05
0.01338



CAG00727_Eggerthella_lenta_DSM_2243
9.214e−05
2.989e−05
0.14
−2.79
0.00697



CAG00963_Clostridium_sp_CAG_524
0.0
0.0
0.0
−inf
0.04363



CAG01141_Holdemanella_biformis_DSM_3989
6.256e−05
6.256e−05
0.04
−4.65
0.05935










Taking into account the bacterial species (n=183, above 1083 total species, 16.9% of the total microbial composition) having an average relative abundance greater than or equal to 0.001 if expressed in the interval [0:1] (thus, the 0.1%) within the overall RCC cohort, we applied the second strategy (machine-learning) for 58 RCC patients (NR=19; R=39) with no antibiotic usage history and OUTCOME_1 criterion. After a first round of feature selection, RFECV gave a rank=1 to 20 species which collectively, after a 5-fold cross-validation, were able to predict 100% of NR and 92% of R subjects, with a CV_AUC=0.917 and an AUC=1 (model 6.1). After a second round of feature selection, RFE gave a rank=1 to 8 species which were selected for the subsequent logistic regression classifier. The model built on these 8 species gave an overall predictability of 84.48% (model 6.2, FIG. 6), and the summary statistics for these species are reported in Table 23 below. In order to measure the goodness of the model, after a 5-fold cross-validation performed on a subset of the original dataset, the model itself was able to predict 67% of NR and 100% of R subjects, with a CV_AUC=0.917 and an AUC=0.889. This last model was designed to be more affordable in case of problems in fecal samples storage, sequencing, stiffness, etc.


According to the Models 4.2, 5.2 and 6.2, which were modeled on no antibiotic usage history and OUTCOME_1 criterion and were all based on 8 bacterial species in order to be properly compared, it is shown that the Model 5.2 has the highest predictability (93.1%) with optimal CV_AUC and AUC.









TABLE 23





Statistics for the species selected in Model 6.2.


























NR (n =
R (n =


NR_Odds



Group
op.
Thr
19) (%)
39) (%)
Pval_Fisher
NR_coeff
Ratio
R_coeff





CAG00243_Lachnospiraceae_bacterium_1_1_57FAA
>
0
84.2
89.7
0.6726
1.2907
2.4465
−1.2907


CAG00008_Clostridium_bolteae_ATCC_BAA_613
>
0
94.7
94.9
1.0
0.8848
1.8465
−0.8848


CAG00698_Ruminococcus_sp_CAG_177
>
0
15.8
10.3
0.6726
0.9776
1.9691
−0.9776


CAG00473_Prevotella_sp_CAG_617
>
0
10.5
0.0
0.1034
0.9997
1.9996
−0.9997


CAG00327_Faecalibacterium_sp_CAG_74
>
0
36.8
61.5
0.0973
1.0697
2.099
−1.0697


CAG01039_Faecalibacterium_cf_prausnitzii_KLE1255
>
0
73.7
51.3
0.1561
0.7806
1.7178
−0.7806


CAG00897_Firmicutes_bacterium_CAG_83
>
0
89.5
89.7
1.0
0.8376
1.7871
−0.8376


CAG00766_Firmicutes_bacterium_CAG_176
>
0
36.8
48.7
0.4172
0.7459
1.6771
−0.7459
















R_Odds


NR Mean



Group
Ratio
rank_RFECV
rank_RFE
relabund
NR Sem





CAG00243_Lachnospiraceae_bacterium_1_1_57FAA
0.4087
1
1
0.0104986
0.00482111


CAG00008_Clostridium_bolteae_ATCC_BAA_613
0.5416
1
1
0.00806075
0.00605804


CAG00698_Ruminococcus_sp_CAG_177
0.5078
1
1
0.00683429
0.00584606


CAG00473_Prevotella_sp_CAG_617
0.5001
1
1
0.00378162
0.00261518


CAG00327_Faecalibacterium_sp_CAG_74
0.4764
1
1
0.00262438
0.0019694


CAG01039_Faecalibacterium_cf_prausnitzii_KLE1255
0.5821
1
1
0.00364533
0.00295908


CAG00897_Firmicutes_bacterium_CAG_83
0.5596
1
1
0.00380745
0.00159185


CAG00766_Firmicutes_bacterium_CAG_176
0.5963
1
1
0.01478181
0.00761963


















R Mean

Fold Ratio
log2FR
P value



Group
relabund
R Sem
(FR) R/NR
R/NR
NR vs R







CAG00243_Lachnospiraceae_bacterium_1_1_57FAA
0.00156745
0.00034161
0.15
−2.74
0.42608



CAG00008_Clostridium_bolteae_ATCC_BAA_613
0.00051335
0.00014219
0.06
−3.97
0.00975



CAG00698_Ruminococcus_sp_CAG_177
0.00013458
8.414e−05
0.02
−5.67
0.51949



CAG00473_Prevotella_sp_CAG_617
0.0
0.0
0.0
−inf
0.04363



CAG00327_Faecalibacterium_sp_CAG_74
0.00046816
0.00013167
0.18
−2.49
0.518



CAG01039_Faecalibacterium_cf_prausnitzii_KLE1255
0.0007401
0.00021316
0.2
−2.3 
0.21913



CAG00897_Firmicutes_bacterium_CAG_83
0.00166388
0.00044938
0.44
−1.19
0.35326



CAG00766_Firmicutes_bacterium_CAG_176
0.00510704
0.00148355
0.35
−1.53
0.74374










Example 7: Predictive Models for RCC Patients with No Antibiotic Usage, OUTCOME 1 Criterion

According to the results obtained in Example 6, and due to the fact that non-responders (NR) are predicted better than responders (R) in the first two models proposed with 8 species (FIG. 7), while is the opposite for the last model, we took into consideration the logistic regression β coefficients relative to NR for Models 4.2 and 5.2, and the logistic regression β coefficients relative to R for the Model 6.2.


Regarding the Model 4.2, and according to the g coefficients of NR (NR_coeff), the probability estimate (expressed as a percentage) of belonging to NR cohort for a tested subject JD is calculated as follows:






P
NR(JD)=1/[1+exp{circumflex over ( )}−(0.955*CAG00008+





0.8761*CAG01039+





0.9951*CAG00473+





0.7609*CAG00487+





1.1107*CAG00140+





0.9402*CAG01141+





0.7187*CAG01208+





−0.5812*CAG00413)]  [6]


In equation [6], “CAGXXXXX” represents the relative abundance of the bacterial species defined by the recited CAG, in a stool sample from the tested subject JD. These relative abundances need to be expressed as standardized values (zero mean and unit variance). The best option is to standardize the “abundances pattern” of the tested subject with a bundle of known “reference patterns”, and the present invention refers a claim of a Python script used to ease the computation of the PNR (JD). From the standardized abundance pattern of a tested subject, the equation [6] can be applied to predict his/her belonging to the NR cohort with a model predictability of 87.93% and a success rate of 100%.


Regarding the Model 5.2, and according to the β coefficients of NR (NR_coeff), the probability estimate (expressed as a percentage) of belonging to NR cohort for a tested subject JD is calculated as follows:






P
NR(JD)=1/[1+exp{circumflex over ( )}−(0.7112*CAG00048_1+





0.8931*CAG00300+





1.0042*CAG00473+





1.1282*CAG00650+





1.2569*CAG00720+





1.2397*CAG00727+





0.7595*CAG00963+





1.0235*CAG01141)]  [7]


In equation [7], “CAGXXXXX” represents the relative abundance of the bacterial species defined by the recited CAG, in a stool sample from the tested subject JD. These relative abundances need to be expressed as standardized values (zero mean and unit variance). The best option is to standardize the “abundances pattern” of the tested subject with a bundle of known “reference patterns”, and the present invention refers a claim of a Python script used to ease the computation of the PNR (JD). From the standardized abundance pattern of a tested subject, the equation [7] can be applied to predict his/her belonging to the NR cohort with a model predictability of 93.1% and a success rate of 100%.


Regarding the Model 6.2, and according to the coefficients of R (R_coeff), the probability estimate (expressed as a percentage) of belonging to R cohort for a tested subject JD is calculated as follows:






P
R(JD)=1/[1+exp{circumflex over ( )}−(−1.2907*CAG00243+





−0.8848*CAG00008+





−0.9776*CAG00698+





−0.9997*CAG00473+





−1.0697*CAG00327+





−0.7806*CAG01039+





−0.8376*CAG00897+





−0.7459*CAG00766)]  [8]


In equation [8], “CAGXXXXX” represents the relative abundance of the bacterial species defined by the recited CAG, in a stool sample from the tested subject JD. These relative abundances need to be expressed as standardized values (zero mean and unit variance). The best option is to standardize the “abundances pattern” of the tested subject with a bundle of known “reference patterns”, and the present invention refers a claim of a Python script used to ease the computation of the PR (JD). From the standardized abundance pattern of a tested subject, the equation [8] could be applied to predict his/her belonging to the R cohort with a model predictability of 84.48% and a success rate of 100%.


Example 8: Results for RCC Patients Regardless Antibiotic Usage, OUTCOME 2 Criterion

In this example, three different models were built based on a bundle of 67 “reference patterns”, which are the standardized abundances profiles (zero mean and unit variance) obtained from the 67 samples used in training the logistic regression models. Standardization is necessary to homogenize the dynamic range of bacterial species abundances.


Based on the first strategy applied to 67 RCC patients (NR=37; R=30) with unknown antibiotic usage history and OUTCOME_2 criterion (TPP>6 months), we selected 10 bacterial species that gave a logistic regression model overall predictability equal to 65.22% (model 7.1). In order to measure the goodness of the model with 10 species, after a 5-fold cross-validation performed on a subset of the original dataset, the model itself was able to predict 78% of NR and 44% of R subjects, with a CV_AUC=0.370 and an AUC=0.716. Upon RFECV and RFE feature selection, 8 species were selected from the aforementioned 10: the model built on these 8 species gave an overall predictability of 66.67% (model 7.2, FIG. 7), and the summary statistics for these species are reported in Table 24. In order to measure the goodness of the model, after a 5-fold cross-validation performed on a subset of the original dataset, the model itself was able to predict 78% of NR and 44% of R subjects, with a CV_AUC=0.481 and an AUC=0.704.


Based on the second strategy (machine-learning) applied to 67 RCC patients (NR=37; R=30) with unknown antibiotic usage history and OUTCOME_2 criterion, we took into account all the bacterial species of the dataset (n=1112). After a first round of feature selection, RFECV gave a rank=1 to 1070 species which collectively, after a 5-fold cross-validation, were able to predict 11% of NR and 100% of R subjects, with a CV_AUC=0.667 and an AUC=0.667 (model 8.1). Upon a second round of feature selection, RFE gave a rank=1 to 8 species, which were selected for the subsequent logistic regression classifier. The model built on these 8 species gave an overall predictability of 88.41% (model 8.2, FIG. 7), and the summary statistics for these species are reported in Table 25. In order to measure the goodness of the model, after a 5-fold cross-validation performed on a subset of the original dataset, the model itself was able to predict 100% of NR and 67% of R subjects, with a CV_AUC=0.667 and an AUC=0.926.


Taking into account the bacterial species (n=181, above 1112 total species, 16.3% of the total microbial composition) having an average relative abundance greater than or equal to 0.001 if expressed in the interval [0:1] (thus, the 0.1%) within the overall RCC cohort, we applied the second strategy (machine-learning) for 67 RCC patients (NR=37; R=30) with unknown antibiotic usage history and OUTCOME_2 criterion. After a first round of feature selection, RFECV gave a rank=1 to 175 species which collectively, after a 5-fold cross-validation, were able to predict 33% of NR and 78% of R subjects, with a CV_AUC=0.222 and an AUC=0.605 (model 9.1). After a second round of feature selection, RFE gave a rank=1 to 8 species which were selected for the subsequent logistic regression classifier. The model built on these 8 species gave an overall predictability of 88.41% (model 9.2, FIG. 7), and the summary statistics for these species are reported in Table 26. In order to measure the goodness of the model, after a 5-fold cross-validation performed on a subset of the original dataset, the model itself was able to predict 89% of NR and 78% of R subjects, with a CV_AUC=0.481 and an AUC=0.864. This last model was designed to be more affordable in case of problems in fecal samples storage, sequencing, stiffness, etcetera.


According to all Models, which were modeled on unknown antibiotic usage history and OUTCOME_2 criterion, it is shown that the Model 8.2 has a good predictability (84.41%) with good CV_AUC and AUC, and can be chosen as the most discriminant among NR and R patients.









TABLE 24





Statistics for the species selected in Model 7.2.


























NR (n =
R (n =


NR_Odds



Group
op.
Thr
37) (%)
30) (%)
Pval_Fisher
NR_coeff
Ratio
R_coeff





CAG00782_Eubacterium_rectale_CAG_36
>
0
86.5
83.3
0.7431
−0.2034
0.8685
0.2009


CAG00013_Clostridiales_bacterium_1_7_47FAA
>
0
78.4
76.7
1.0
0.4092
1.328
−0.348


CAG00873_Butyricimonas_virosa_DSM_23226
>
0
70.3
73.3
1.0
0.2708
1.2065
−0.2435


CAG01141_Holdemanella_biformis_DSM_3989
>
0
13.5
0.0
0.0599
0.7665
1.7011
−0.6748


CAG00668_Azospirillum_sp_CAG_239
>
0
0.0
10.0
0.0848
−0.5899
0.6644
0.6848


CAG00669_Firmicutes_bacterium_CAG_103
>
0
32.4
53.3
0.1345
−0.5181
0.6983
0.4599


CAG00886_Sutterella_sp_CAG_351
>
0
0.0
10.0
0.0848
−0.8415
0.5581
0.9265


CAG00889_Megasphaera_elsdenii_14_14
>
0
0.0
13.3
0.0358
−0.7339
0.6013
0.8563
















R_Odds


NR Mean



Group
Ratio
rank_RFECV
rank_RFE
relabund
NR Sem





CAG00782_Eubacterium_rectale_CAG_36
1.1494
3
1
0.01855862
0.00499803


CAG00013_Clostridiales_bacterium_1_7_47FAA
0.7857
1
1
0.00056515
0.00023638


CAG00873_Butyricimonas_virosa_DSM_23226
0.8447
2
1
0.00109418
0.00020006


CAG01141_Holdemanella_biformis_DSM_3989
0.6264
1
1
0.00154882
0.00079727


CAG00668_Azospirillum_sp_CAG_239
1.6075
1
1
0.0
0.0


CAG00669_Firmicutes_bacterium_CAG_103
1.3754
1
1
0.00062774
0.00020461


CAG00886_Sutterella_sp_CAG_351
1.9007
1
1
0.0
0.0


CAG00889_Megasphaera_elsdenii_14_14
1.8104
1
1
0.0
0.0


















R Mean

Fold Ratio
log2FR
P value



Group
relabund
R Sem
(FR) R/NR
R/NR
NR vs R







CAG00782_Eubacterium_rectale_CAG_36
0.0158587
0.00446139
0.85
−0.23
0.84974



CAG00013_Clostridiales_bacterium_1_7_47FAA
0.00030914
0.00011594
0.55
−0.87
0.57258



CAG00873_Butyricimonas_virosa_DSM_23226
0.00063137
0.00012234
0.58
−0.79
0.29268



CAG01141_Holdemanella_biformis_DSM_3989
0.0
0.0
0.0
−inf
0.03925



CAG00668_Azospirillum_sp_CAG_239
0.00068027
0.00048003
0.0
−inf
0.05297



CAG00669_Firmicutes_bacterium_CAG_103
0.00180915
0.00055553
2.88
 1.53
0.0513



CAG00886_Sutterella_sp_CAG_351
0.0007941
0.00060463
0.0
−inf
0.05297



CAG00889_Megasphaera_elsdenii_14_14
0.00133655
0.00079124
0.0
−inf
0.02403

















TABLE 25





Statistics for the species selected in Model 8.2.


























NR (n =
R (n =


NR_Odds



Group
op.
Thr
37) (%)
30) (%)
Pval_Fisher
NR_coeff
Ratio
R_coeff





CAG00211_Firmicutes_bacterium_CAG_227
>
0
16.2
10.0
0.7206
0.6422
1.5607
−0.5746


CAG00474_Sutterella_wadsworthensis_2_1_59BFAA
>
0
8.1
23.3
0.0982
−1.0581
0.4803
1.1768


CAG00624_Firmicutes_bacterium_CAG_552
>
0
0.0
16.7
0.0148
−0.8984
0.5365
1.0055


CAG00650_Dorea_formicigenerans_ATCC_27755
>
0
81.1
90.0
0.4926
1.3189
2.4947
−1.271


CAG00676_Firmicutes_bacterium_CAG_176
>
0
16.2
20.0
0.7553
−0.7324
0.6019
0.0679


CAG00771_Clostridium_sp_CAG_413
>
0
0.0
10.0
0.0848
−0.6623
0.6319
0.4204


CAG01197_Dialister_succinatiphilus_YIT_11850
>
0
8.1
30.0
0.0268
−1.1372
0.4546
0.9425


CAG01321_Faecalibacterium_prausnitzii_SL3_3
>
0
83.8
90.0
0.7206
−0.8525
0.5538
0.8843
















R_Odds


NR Mean



Group
Ratio
rank_RFECV
rank_RFE
relabund
NR Sem





CAG00211_Firmicutes_bacterium_CAG_227
0.6715
1
1
5.982e−05
2.678e−05


CAG00474_Sutterella_wadsworthensis_2_1_59BFAA
2.2608
1
1
0.00023198
0.00014013


CAG00624_Firmicutes_bacterium_CAG_552
2.0076
1
1
0.0
0.0


CAG00650_Dorea_formicigenerans_ATCC_27755
0.4144
1
1
0.00092982
0.0002447


CAG00676_Firmicutes_bacterium_CAG_176
1.0482
1
1
0.0001876
0.00010047


CAG00771_Clostridium_sp_CAG_413
1.3383
1
1
0.0
0.0


CAG01197_Dialister_succinatiphilus_YIT_11850
1.9219
1
1
3.257e−05
2.681e−05


CAG01321_Faecalibacterium_prausnitzii_SL3_3
1.8458
1
1
0.00480104
0.00091279


















R Mean

Fold Ratio
log2FR
P value



Group
relabund
R Sem
(FR) R/NR
R/NR
NR vs R







CAG00211_Firmicutes_bacterium_CAG_227
1.092e−05
 7.11e−06
0.18
−2.45 
0.39482



CAG00474_Sutterella_wadsworthensis_2_1_59BFAA
0.0019427 
0.00086912
8.37
3.07
0.07186



CAG00624_Firmicutes_bacterium_CAG_552
7.419e−05
3.936e−05
0.0
−inf
0.0109



CAG00650_Dorea_formicigenerans_ATCC_27755
0.00039615
7.209e−05
0.43
−1.23 
0.30037



CAG00676_Firmicutes_bacterium_CAG_176
0.00087187
0.00055567
4.65
2.22
0.67128



CAG00771_Clostridium_sp_CAG_413
5.164e−05
3.902e−05
0.0
−inf
0.05297



CAG01197_Dialister_succinatiphilus_YIT_11850
0.00014594
 5.12e−05
4.48
2.16
0.01886



CAG01321_Faecalibacterium_prausnitzii_SL3_3
0.01054466
0.00251204
2.2
1.14
0.13467

















TABLE 26





Statistics for the species selected in Model 9.2.


























NR (n =
R (n =


NR_Odds



Group
op.
Thr
37) (%)
30) (%)
Pval_Fisher
NR_coeff
Ratio
R_coeff





CAG00557_Ruminococcus_callidus_ATCC_27760
>
0
24.3
33.3
0.4303
0.9341
1.9107
−0.8033


CAG00601_uncultured_Faecalibacterium_sp
>
0
70.3
70.0
1.0
0.8927
1.8567
−1.0252


CAG00607_Eubacterium_sp_CAG_251
>
0
37.8
30.0
0.6076
0.7108
1.6367
−0.5952


CAG00669_Firmicutes_bacterium_CAG_103
>
0
32.4
53.3
0.1345
−1.2341
0.4251
1.1675


CAG00861_Oscillibacter_sp_CAG_241
>
0
37.8
26.7
0.4347
1.0379
2.0532
−0.9407


CAG00880_Subdoligranulum_sp_CAG_314
>
0
21.6
36.7
0.188
−1.0016
0.4995
0.8717


CAG00937_Clostridium_sp_CAG_7
>
0
86.5
90.0
0.7225
−0.872
0.5464
0.9665


CAG01321_Faecalibacterium_prausnitzii_SL3_3
>
0
83.8
90.0
0.7206
−0.8362
0.5601
0.8646
















R_Odds


NR Mean



Group
Ratio
rank_RFECV
rank_RFE
relabund
NR Sem





CAG00557_Ruminococcus_callidus_ATCC_27760
0.573
1
1
0.00160652
0.00069973


CAG00601_uncultured_Faecalibacterium_sp
0.4913
1
1
0.00483175
0.0009123


CAG00607_Eubacterium_sp_CAG_251
0.662
1
1
0.00225675
0.00078393


CAG00669_Firmicutes_bacterium_CAG_103
2.2462
1
1
0.00062774
0.00020461


CAG00861_Oscillibacter_sp_CAG_241
0.521
1
1
0.00475511
0.00195861


CAG00880_Subdoligranulum_sp_CAG_314
1.8298
1
1
0.00084578
0.00045115


CAG00937_Clostridium_sp_CAG_7
1.9541
1
1
0.00087447
0.00015342


CAG01321_Faecalibacterium_prausnitzii_SL3_3
1.8208
1
1
0.00480104
0.00091279


















R Mean

Fold Ratio
log2FR
P value



Group
relabund
R Sem
(FR) R/NR
R/NR
NR vs R







CAG00557_Ruminococcus_callidus_ATCC_27760
0.0004874
0.00020579
0.3
−1.72
0.69767



CAG00601_uncultured_Faecalibacterium_sp
0.00223149
0.00050757
0.46
−1.11
0.12832



CAG00607_Eubacterium_sp_CAG_251
0.00059667
0.00025431
0.26
−1.92
0.2972



CAG00669_Firmicutes_bacterium_CAG_103
0.00180915
0.00055553
2.88
1.53
0.0513



CAG00861_Oscillibacter_sp_CAG_241
0.000911
0.00043555
0.19
−2.38
0.26701



CAG00880_Subdoligranulum_sp_CAG_314
0.00390209
0.00155646
4.61
2.21
0.10582



CAG00937_Clostridium_sp_CAG_7
0.00125129
0.00028594
1.43
0.52
0.48764



CAG01321_Faecalibacterium_prausnitzii_SL3_3
0.01054466
0.00251204
2.2
1.14
0.13467










Example 9: Predictive Models for RCC Patients Regardless Antibiotic Usage, OUTCOME 2 Criterion

According to the results obtained in Example 8, we took into consideration the logistic regression β coefficients relative to NR for Models 7.2, 8.2 and 9.2.


For the Model 7.2, according to the β coefficients of non-responders (NR_coeff), the probability estimate (expressed as a percentage) of belonging to NR cohort for a tested subject JD is calculated as follows:






P
NR(JD)=1/[1+exp{circumflex over ( )}−(−0.2034*CAG00782+





0.4092*CAG00013+





0.2708*CAG00873+





0.7665*CAG01141+





−0.5899*CAG00668+





−0.5181*CAG00669+





−0.8415*CAG00886+





−0.7339*CAG00889)]  [9]


In equation [9], “CAGXXXXX” represents the relative abundance of the bacterial species defined by the recited CAG, in a stool sample from the tested subject JD. These relative abundances need to be expressed as standardized values (zero mean and unit variance). The best option is to standardize the “abundances pattern” of the tested subject with a bundle of known “reference patterns”, and the present invention refers a claim of a Python script used to ease the computation of the PNR (JD). From the standardized abundance pattern of a tested subject, the equation [9] can be applied to predict his/her belonging to the NR cohort with a model predictability of 66.67% and a success rate of 78%.


For the Model 8.2, according to the β coefficients of NR (NR_coeff), the probability estimate (expressed as a percentage) of belonging to NR cohort for a tested subject JD is calculated as follows:






P
NR(JD)=1/[1+exp{circumflex over ( )}−(0.6422*CAG00211+





−1.0581*CAG00474+





−0.8984*CAG00624+





1.3189*CAG00650+





−0.7324*CAG00676+





−0.6623*CAG00771+





−1.1372*CAG01197+





−0.8525*CAG01321)]  [10]


In equation [10], “CAGXXXXX” represents the relative abundance of the bacterial species defined by the recited CAG, in a stool sample from the tested subject JD. These relative abundances need to be expressed as standardized values (zero mean and unit variance). The best option is to standardize the “abundances pattern” of the tested subject with a bundle of known “reference patterns”, and the present invention refers a claim of a Python script used to ease the computation of the PNR (JD). From the standardized abundance pattern of a tested subject, the equation [10] can be applied to predict his/her belonging to the NR cohort with a model predictability of 84.41% and a success rate of 100%.


For the Model 9.2, according to the β coefficients of NR (NR_coeff), the probability estimate (expressed as a percentage) of belonging to NR cohort for a tested subject JD is calculated as follows:






P
NR(JD)=1/[1+exp{circumflex over ( )}−(0.9341*CAG00557+





0.8927*CAG00601+





0.7108*CAG00607+





−1.2341*CAG00669+





1.0379*CAG00861+





−1.0016*CAG00880+





−0.872*CAG00937+





−0.8362*CAG01321)]  [11]


In equation [11], “CAGXXXXX” represents the relative abundance of the bacterial species defined by the recited CAG, in a stool sample from the tested subject JD. These relative abundances need to be expressed as standardized values (zero mean and unit variance). The best option is to standardize the “abundances pattern” of the tested subject with a bundle of known “reference patterns”, and the present invention refers a claim of a Python script used to ease the computation of the PNR (JD). From the standardized abundance pattern of a tested subject, the equation [11] can be applied to predict his/her belonging to the NR cohort with a model predictability of 88.41% and a success rate of 89%.


Example 10: Results for RCC Patients with No Antibiotic Usage, OUTCOME 2 Criterion

In this example, three different models were built based on a bundle of 56 “reference patterns”, which are the standardized abundances profiles (zero mean and unit variance) obtained from the 56 samples used in training the logistic regression models. Standardization is necessary to homogenize the dynamic range of bacterial species abundances.


Based on the first strategy applied to 56 RCC patients (NR=29; R=27) with no antibiotic usage history and OUTCOME_2 criterion, we selected 10 bacterial species that gave a logistic regression model overall predictability equal to 72.41% (model 10.1). In order to measure the goodness of the model with 10 species, after a 5-fold cross-validation performed on a subset of the original dataset, the model itself was able to predict 78% of NR and 67% of R subjects, with a CV_AUC=0.667 and an AUC=0.796. Upon RFECV and RFE feature selection, 8 species were selected from the aforementioned 10: the model built on these 8 species gave an overall predictability of 68.97% (model 10.2, FIG. 8), and the summary statistics for these species are reported in Table 27. In order to measure the goodness of the model, after a 5-fold cross-validation performed on a subset of the original dataset, the model itself was able to predict 89% of NR and 100% of R subjects, with a CV_AUC=0.556 and an AUC=0.981.


Based on the second strategy (machine-learning) applied to 56 RCC patients (NR=29; R=27) with no antibiotic usage history and OUTCOME_2 criterion, we took into account all the bacterial species of the dataset (n=1083). After a first round of feature selection, RFECV gave a rank=1 to 51 species which collectively, after a 5-fold cross-validation, were able to predict 89% of NR and 83% of R subjects, with a CV_AUC=0.944 and an AUC=0.944 (model 11.1). Upon a second round of feature selection, RFE gave a rank=1 to 8 species, which were selected for the subsequent logistic regression classifier. The model built on these 8 species gave an overall predictability of 91.38% (model 11.2, FIG. 8), and the summary statistics for these species are reported in Table 28. In order to measure the goodness of the model, after a 5-fold cross-validation performed on a subset of the original dataset, the model itself was able to predict 78% of NR and 100% of R subjects, with a CV_AUC=1 and an AUC=0.981.









TABLE 27





Statistics for the species selected in Model 10.2.


























NR (n =
R (n =


NR_Odds



Group
op.
Thr
29) (%)
27) (%)
Pval_Fisher
NR_coeff
Ratio
R_coeff





CAG00580_Clostridium_sp_CAG_62
>
0
75.9
77.8
1.0
0.2713
1.2069
−0.1456


CAG00013_Clostridiales_bacterium_1_7_47FAA
>
0
79.3
74.1
0.7555
0.662
1.5823
−0.5496


CAG00873_Butyricimonas_virosa_DSM_23226
>
0
72.4
74.1
1.0
0.4991
1.4133
−0.4646


CAG01141_Holdemanella_biformis_DSM_3989
>
0
13.8
0.0
0.1124
0.6615
1.5817
−0.5496


CAG00668_Azospirillum_sp_CAG_239
>
0
0.0
11.1
0.1055
−0.4506
0.7318
0.5684


CAG00669_Firmicutes_bacterium_CAG_103
>
0
27.6
55.6
0.0563
−0.6129
0.6539
0.5129


CAG00886_Sutterella_sp_CAG_351
>
0
0.0
7.4
0.2279
−0.5618
0.6775
0.6623


CAG00889_Megasphaera_elsdenii_14_14
>
0
0.0
11.1
0.1055
−0.5819
0.6681
0.7102
















R_Odds


NR Mean



Group
Ratio
rank_RFECV
rank_RFE
relabund
NR Sem





CAG00580_Clostridium_sp_CAG_62
0.904
2
1
0.00355474
0.00090909


CAG00013_Clostridiales_bacterium_1_7_47FAA
0.6832
1
1
0.00068832
0.00029843


CAG00873_Butyricimonas_virosa_DSM_23226
0.7247
1
1
0.00123868
0.000242


CAG01141_Holdemanella_biformis_DSM_3989
0.6832
1
1
0.00111662
0.00058518


CAG00668_Azospirillum_sp_CAG_239
1.4829
1
1
0.0
0.0


CAG00669_Firmicutes_bacterium_CAG_103
1.4269
1
1
0.00047936
0.00020785


CAG00886_Sutterella_sp_CAG_351
1.5826
1
1
0.0
0.0


CAG00889_Megasphaera_elsdenii_14_14
1.6361
1
1
0.0
0.0


















R Mean

Fold Ratio
log2FR
P value



Group
relabund
R Sem
(FR) R/NR
R/NR
NR vs R







CAG00580_Clostridium_sp_CAG_62
0.00216117
0.00057471
0.61
−0.72
0.43804



CAG00013_Clostridiales_bacterium_1_7_47FAA
0.00017732
6.043e−05
0.26
−1.96
0.27615



CAG00873_Butyricimonas_virosa_DSM_23226
0.00057747
0.00012007
0.47
−1.1 
0.14061



CAG01141_Holdemanella_biformis_DSM_3989
0.0
0.0
0.0
−inf
0.04946



CAG00668_Azospirillum_sp_CAG_239
0.00075585
0.00053233
0.0
−inf
0.0708



CAG00669_Firmicutes_bacterium_CAG_103
0.00194815
0.00061023
4.06
 2.02
0.01602



CAG00886_Sutterella_sp_CAG_351
0.00023671
0.00021212
0.0
−inf
0.14611



CAG00889_Megasphaera_elsdenii_14_14
0.00147009
0.00087685
0.0
−inf
0.0708

















TABLE 28





Statistics for the species selected in Model 11.2.


























NR (n =
R (n =


NR_Odds



Group
op.
Thr
29) (%)
27) (%)
Pval_Fisher
NR_coeff
Ratio
R_coeff





CAG00346_Eubacterium_rectale_M104_1
>
0
10.3
0.0
0.2373
0.9631
1.9495
−0.8463


CAG00530_Prevotella_sp_CAG_617
>
0
10.3
29.6
0.0965
−1.2447
0.422
1.13


CAG00601_uncultured_Faecalibacterium_sp
>
0
79.3
74.1
0.7555
1.0027
2.0038
−1.183


CAG00607_Eubacterium_sp_CAG_251
>
0
37.9
33.3
0.7849
1.181
2.2673
−0.9544


CAG00646_Alistipes_sp_CAG_268
>
0
13.8
25.9
0.3224
−0.7539
0.593
0.8929


CAG00713_Firmicutes_bacterium_CAG_270
>
0
6.9
33.3
0.0184
−0.9526
0.5167
1.0919


CAG00919_Clostridium_methylpentosum_DSM_5476
>
0
17.2
33.3
0.2214
−0.7227
0.606
0.4881


CAG01158_Pseudoflavonifractor_capillosus_ATCC_29799
>
0
3.4
29.6
0.0104
−0.9687
0.511
1.1606
















R_Odds


NR Mean



Group
Ratio
rank_RFECV
rank_RFE
relabund
NR Sem





CAG00346_Eubacterium_rectale_M104_1
0.5562
1
1
2.084e−05
1.172e−05 


CAG00530_Prevotella_sp_CAG_617
2.1886
1
1
0.00032714
0.0002189 


CAG00601_uncultured_Faecalibacterium_sp
0.4404
1
1
0.00595075
0.00106984


CAG00607_Eubacterium_sp_CAG_251
0.5161
1
1
0.00265039
0.00097373


CAG00646_Alistipes_sp_CAG_268
1.8569
1
1
0.00020628
0.00010205


CAG00713_Firmicutes_bacterium_CAG_270
2.1316
1
1
 2.89e−06
2.09e−06


CAG00919_Clostridium_methylpentosum_DSM_5476
1.4026
1
1
1.004e−05
4.17e−06


CAG01158_Pseudoflavonifractor_capillosus_ATCC_29799
2.2355
1
1
 6.3e−06
 6.3e−06


















R Mean

Fold Ratio
log2FR
P value



Group
relabund
R Sem
(FR) R/NR
R/NR
NR vs R







CAG00346_Eubacterium_rectale_M104_1
0.0
0.0
0.0
−inf
0.09282



CAG00530_Prevotella_sp_CAG_617
0.00163373
0.00059881
4.99
2.32
0.06874



CAG00601_uncultured_Faecalibacterium_sp
0.00227143
0.00053733
0.38
−1.39
0.01999



CAG00607_Eubacterium_sp_CAG_251
0.00066297
0.00028011
0.25
−2.0
0.37879



CAG00646_Alistipes_sp_CAG_268
0.00156952
0.00064613
7.61
2.93
0.17786



CAG00713_Firmicutes_bacterium_CAG_270
0.00021793
8.762e−05
75.46
6.24
0.0087



CAG00919_Clostridium_methylpentosum_DSM_5476
7.683e−05
3.389e−05
7.65
2.94
0.16102



CAG01158_Pseudoflavonifractor_capillosus_ATCC_29799
0.00024491
9.486e−05
38.88
5.28
0.00656

















TABLE 29





Statistics for the species selected in Model 12.2.


























NR (n =
R (n =


NR_Odds



Group
op.
Thr
29) (%)
27) (%)
Pval_Fisher
NR_coeff
Ratio
R_coeff





CAG00142_Bacteroides_stercoris_ATCC_43183
>
0
48.3
37.0
0.4303
1.138
2.2007
−0.8803


CAG00218_Bacteroides_sp_CAG_20
>
0
89.7
81.5
0.462
1.1396
2.2031
−0.8724


CAG00341_Eubacterium_sp_CAG_115
>
0
13.8
44.4
0.0174
−1.0166
0.4943
0.6772


CAG00474_Sutterella_wadsworthensis_2_1_59BFAA
>
0
6.9
25.9
0.0733
−0.8403
0.5585
0.8566


CAG00508_Alistipes_obesi
>
0
72.4
92.6
0.0798
−0.7067
0.6127
0.6586


CAG00880_Subdoligranulum_sp_CAG_314
>
0
13.8
33.3
0.1165
−1.185
0.4398
1.0027


CAG01263_Clostridium_clostridioforme_2_1_49FAA
>
0
86.2
74.1
0.3224
0.8466
1.7982
−0.5783


CAG01321_Faecalibacterium_prausnitzii_SL3_3
>
0
79.3
92.6
0.2544
−0.8895
0.5398
0.9053
















R_Odds


NR Mean



Group
Ratio
rank_RFECV
rank_RFE
relabund
NR Sem





CAG00142_Bacteroides_stercoris_ATCC_43183
0.5433
1
1
0.01293929
0.00368976


CAG00218_Bacteroides_sp_CAG_20
0.5462
1
1
0.01321164
0.00169549


CAG00341_Eubacterium_sp_CAG_115
1.599
1
1
0.00031187
0.00027514


CAG00474_Sutterella_wadsworthensis_2_1_59BFAA
1.8108
1
1
0.00020181
0.00015501


CAG00508_Alistipes_obesi
1.5785
1
1
0.00262606
0.00069393


CAG00880_Subdoligranulum_sp_CAG_314
2.0038
1
1
0.00036879
0.00020934


CAG01263_Clostridium_clostridioforme_2_1_49FAA
0.6698
1
1
0.00425323
0.00240791


CAG01321_Faecalibacterium_prausnitzii_SL3_3
1.8729
1
1
0.00465506
0.00103762


















R Mean

Fold Ratio
log2FR
P value



Group
relabund
R Sem
(FR) R/NR
R/NR
NR vs R







CAG00142_Bacteroides_stercoris_ATCC_43183
0.003047
0.00114758
0.24
−2.09
0.14334



CAG00218_Bacteroides_sp_CAG_20
0.00941315
0.00154558
0.71
−0.49
0.07752



CAG00341_Eubacterium_sp_CAG_115
0.00289445
0.00124963
9.28
3.21
0.00822



CAG00474_Sutterella_wadsworthensis_2_1_59BFAA
0.00215856
0.00095824
10.7
3.42
0.04832



CAG00508_Alistipes_obesi
0.00471117
0.0009454
1.79
0.84
0.08724



CAG00880_Subdoligranulum_sp_CAG_314
0.00400279
0.00171271
10.85
3.44
0.05113



CAG01263_Clostridium_clostridioforme_2_1_49FAA
0.0003713
0.00011117
0.09
−3.52
0.05414



CAG01321_Faecalibacterium_prausnitzii_SL3_3
0.01086215
0.00270038
2.33
1.22
0.07615










Taking into account the bacterial species (n=183, above 1083 total species, 16.9% of the total microbial composition) having an average relative abundance greater than or equal to 0.001 if expressed in the interval [0:1] (thus, the 0.1%) within the overall RCC cohort, we applied the second strategy (machine-learning) for 56 RCC patients (NR=29; R=27) with no antibiotic usage history and OUTCOME_2 criterion. After a first round of feature selection, RFECV gave a rank=1 to 20 species which collectively, after a 5-fold cross-validation, were able to predict 78% of NR and 67% of R subjects, with a CV_AUC=0.833 and an AUC=0.852 (model 12.1). After a second round of feature selection, RFE gave a rank=1 to 8 species which were selected for the subsequent logistic regression classifier. The model built on these 8 species gave an overall predictability of 89.66% (model 12.2, FIG. 8), and the summary statistics for these species are reported in Table 29. In order to measure the goodness of the model, after a 5-fold cross-validation performed on a subset of the original dataset, the model itself was able to predict 78% of NR and 50% of R subjects, with a CV_AUC=0.889 and an AUC=0.611. This last model was designed to be more affordable in case of problems in fecal samples storage, sequencing, stiffness, etc.


According to the Models 10.2, 11.2 and 12.2, which were modeled on no antibiotic usage history and OUTCOME_2 criterion, and were all based on 8 bacterial species in order to be properly compared, it is shown that the Model 11.2 has the highest discriminatory power among NR and R, with good predictability (91.38%), and excellent CV_AUC and AUC.


Example 11: Predictive Models for RCC Patients with No Antibiotic Usage, OUTCOME 2 Criterion

According to the results obtained in Example 10, and due to the fact that responders (R) are predicted better than non-responders (NR) in models 10.2 and 11.2, while is the opposite for model 12.2, and that all models were proposed with 8 species (FIG. 7) to make them comparable, we took into consideration the logistic regression β coefficients relative to R for models 10.2 and 11.2, while the logistic regression β coefficients relative to NR for model 12.2.


Regarding the Model 10.2, and according to the coefficients of R (R_coeff), the probability estimate (expressed as a percentage) of belonging to R cohort for a tested subject JD is calculated as follows:






P
R(JD)=1/[1+exp{circumflex over ( )}−(−0.1456*CAG00580+





−0.5496*CAG00013+





−0.4646*CAG00873+





−0.5496*CAG01141+





0.5684*CAG00668+





0.5129*CAG00669+





0.6623*CAG00886+





0.7102*CAG00889)]  [12]


In equation [12], “CAGXXXXX” represents the relative abundance of the bacterial species defined by the recited CAG, in a stool sample from the tested subject JD. These relative abundances need to be expressed as standardized values (zero mean and unit variance). The best option is to standardize the “abundances pattern” of the tested subject with a bundle of known “reference patterns”, and the present invention refers a claim of a Python script used to ease the computation of the PR (JD). From the standardized abundance pattern of a tested subject, the equation [12] can be applied to predict his/her belonging to the R cohort with a model predictability of 68.97% and a success rate of 100%.


Regarding the Model 11.2, and according to the β coefficients of R (R_coeff), the probability estimate (expressed as a percentage) of belonging to R cohort for a tested subject JD is calculated as follows:






P
R(JD)=1/[1+exp{circumflex over ( )}−(−0.8463*CAG00346+





1.13*CAG00530+





−1.183*CAG00601+





−0.9544*CAG00607+





0.8929*CAG00646+





1.0919*CAG00713+





0.4881*CAG00919+





1.1606*CAG01158)]  [13]


In equation [13], “CAGXXXXX” represents the relative abundance of the bacterial species defined by the recited CAG, in a stool sample from the tested subject JD. These relative abundances need to be expressed as standardized values (zero mean and unit variance). The best option is to standardize the “abundances pattern” of the tested subject with a bundle of known “reference patterns”, and the present invention refers a claim of a Python script used to ease the computation of the PR (JD). From the standardized abundance pattern of a tested subject, the equation [13] can be applied to predict his/her belonging to the R cohort with a model predictability of 91.38% and a success rate of 100%.


Regarding the Model 12.2, and according to the β coefficients of NR (NR_coeff), the probability estimate (expressed as a percentage) of belonging to NR cohort for a tested subject JD is calculated as follows:






P
NR(JD)=1/[1+exp{circumflex over ( )}−(1.138*CAG00142+





1.1396*CAG00218+





−1.0166*CAG00341+





−0.8403*CAG00474+





−0.7067*CAG00508+





−1.185*CAG00880+





0.8466*CAG01263+





−0.8895*CAG01321)]  [14]


In equation [14], “CAGXXXXX” represents the relative abundance of the bacterial species defined by the recited CAG, in a stool sample from the tested subject JD. These relative abundances need to be expressed as standardized values (zero mean and unit variance). The best option is to standardize the “abundances pattern” of the tested subject with a bundle of known “reference patterns”, and the present invention refers a claim of a Python script used to ease the computation of the PNR (JD). From the standardized abundance pattern of a tested subject, the equation [14] can be applied to predict his/her belonging to the NR cohort with a model predictability of 89.66% and a success rate of 78%.


Discussion

The method described above relies on the following positive features:

    • (i) it is possible to use different NGS sequencing platforms (454, Illumina, IonTorrent, Nanopore, PacBio) and molecular biology techniques (qPCR, microarray, . . . )
    • (ii) the expression of the relative abundances of model-selected bacterial species within the closed interval [0:1].
    • (iii) if the tested subject does not have one or more selected species belonging to the chosen model, this model retains a high predictability due to its additive nature and the proposed double strategy.
    • (iv) each one of both strategies is split into two different models, one which is limited to individuals who did not take antibiotics during the past two months, while the other one considers the eventuality of an unknown antibiotic regimen/treatment status.


According to the statistical method used to properly fit the “abundances pattern” of a definite subject into one of the proposed models, as described in Materials and Methods, we propose the following steps:

    • (i) a fecal sample taken from the subject undergoes DNA extraction and shotgun sequencing with the preferred NGS platform (as described above), and relative abundances within the closed interval [0:1] are measured through the bioinformatic pipeline described in Materials and Methods.
    • (ii) the predictability score (P) for the subject JD, which represents the probability estimate (expressed as a percentage) of belonging to a definite cohort (NR or R) is defined as follows:






P=1/(1+exp{circumflex over ( )}−z)  [1]





and






z=β1*x1+ . . . +βn*xn  [2]


where z is the log-odds ratio (expressing the natural logarithm of the ratio between the probability that an event will occur to the probability that it will not occur), exp is the exponent in natural base, each β is the regression coefficient pre-computed according to the invention, and each x is the relative abundance of selected bacterial species expressed within the closed interval [0:1], obligatory expressed as standardized data (mean removed and unit variance scaled);

    • (iii) the P score obtained in step (ii) is interpreted as clinically meaningful if it is higher than 75% (fixed threshold) and/or it is concordant among the different Models used, always taking into account the antibiotic usage (known or unknown).


In the above method, the regression coefficients have to be calculated by logistic regression classifier with the following requirements: a constant (also known as intercept) not added to the decision function; a max iteration equal to 10000; a liblinear solver (with L2 penalization). All these parameters for logistic regression are explained and described thoroughly in section 1.1.11. of Sci-Kit Learn library guide.


OUTCOME_2 means one considers only SD>6 months among responders.


Based on the results disclosed herein, we conclude the following notions:


1/GOMS (“gut oncomicrobiome signatures”) can definitely predict good clinical outcome during PD-1 blockade with AUC>0.9, and decent specificity and sensitivity in RCC.


2/Some clinical criteria contrasting R versus NR can be impacted by the composition of the gut oncomicrobiome.


3/The GOMS of R and NR are by and large the same ones when taking into consideration the ‘human-driven’ strategy, and they can predict differently the NR or R status based on logistic regression coefficients.


4/Clinical significance relies on an equilibrium between “favorable” and “unfavorable” commensals, detectable in higher abundance in R feces or NR feces, respectively


5/Some species withstand ATB, mainly members of Firmicutes phylum.


6/Our best predicting minimal ecosystem (called “GOMS”) on the whole population of RCC 2L is based on few MGS (unfavorable Prevotella spp. Faecalibcaterium spp., Coprococcus catus, Eggerthelia lenta, Clostridium boiteae versus favorable Suttereila spp., Akkermansia, Alistipes spp.), as evidenced by pairwise analysis and logistic regression models.


7/Of note, some favorable commensals are part of the differences observed between healthy versus cancer individuals (overexpressed bacteria in HV (vs cancer pts) are also associated with R phenotype in anti-PD1 treated patients)


In the present text, we proposed different GOMS (gut oncomicrobiome signatures) to predict the NR or R status of a definite subject suffering from RCC depending, before an ICI treatment, on two main criteria: i) the presence of an antibiotic treatment/usage within the last two months; ii) the RECIST definition of outcome (OUTCOME_1 or OUTCOME_2). As a first step, we evidenced how GOMS are able to differentiate among NR and R at OUTCOME_1, TTP3 and TTP6, with this latter showing significant alfa- and beta-diversity in the first cohort reported in Routy et al Science 2018. Mean AUC values (among NR and R) for the best descriptors (single bacterial species or consortia) based on ROC curves showed the higher prediction value at TTP3: regardless antibiotic usage we have Mean AUCOUTCOME1=0.872, Mean AUCTTP3=0.894, Mean AUCTTP6=0.840, while with no antibiotic treatment we have Mean AUCOUTCOME1=0.862, Mean AUCTTP3=0.891, Mean AUCTTP6=0.762. From these observations, it seems that in RCC the antibiotic pressure (at least within the two months before ICI treatment) seems to enhance predictability of the GOMS signature especially at TTP6 (+10%), while leaving unalterated the one at OUTCOME1 and TTP3.


In the above study, we deeply analyzed by means of Volcano, LEfSe and pairwise analysis the bacterial signature of NR and R in patients regardless antibiotic usage and of those who did not take antibiotics during the two months preceeding ICI treatment. We found that from the original signature at TTP6 no bacterial species resisted ATB among NR, while 18% resisted ATB among responders (FIG. 3). All these species could be good predictors for a RCC patient with unknown antibiotic regimen. Different logistic regression models are proposed herein, based on the aforementioned GOMS, antibiotic usage, and OUTCOME status following RECIST criteria. Upon a double strategy (as described in Materials and Methods) and after an internal feature selection and 5-fold cross-validation we made six models (Model 10.2, Model 2.2, Model 3.2, Model 7.2, Model 8.2, Model 9.2) for RCC patients regardless ATB, while we made other six models (Model 4.2, Model 5.2, Model 6.2, Model 10.2, Model 11.2, Model 12.2) for RCC patients without ATB treatment (see Table 30). From a first survey, after logistic regression models, it seems that ATB worsens by around 4% the overall model predictability for both OUTCOMES; in fact, we have Mean Pred.OUTCOME1_regardlessATB=85.02%, Mean Pred.OUTCOME1_noATB=88.50%, Mean riedOUTCOME2_regardlessATB=79.83%, and finally Mean Pred.OUTCOME2_noATB=8334%. From these results OUTCOME_1 criterion seems to a best predictor than OUTCOME_2, at least for the overall predictability. Notably, only around 50% of species were in common among models without and regardless antibiotic usage, mainly belonging to Firmicutes phylum (genera Faecalibacterium, Hoidemanella, Subdoligranulum for OUTCOME_1 and OUTCOME_2, while genus Eubacterium only for OUTCOME_2). Members of Proteobacteria phylum (genera Sutterella and Azospirillum) withstanding antibiotic treatment were found only with models generated with OUTCOME_2,









TABLE 31







Specific sequences for each bacterial species.








Bacterial species
Specific sequences





CAG00008_Clostridium_bolteae_ATCC_BAA_613
SEQ ID NO: 1-50


CAG00013_Clostridiales_bacterium_1_7_47FAA
SEQ ID NO: 51-100


CAG00037_Bacteroides_faecis_MAJ27
SEQ ID NO: 101-150


CAG00048_1_Clostridium_sp_CAG_226
SEQ ID NO: 2001-2050


CAG00063_Barnesiella_viscericola_DSM_18177
SEQ ID NO: 151-200


CAG00122_Coprococcus_catus_GD_7
SEQ ID NO: 401-450


CAG00140_Subdoliqranulum_sp_4_3_54A2FAA
SEQ ID NO: 451-500


CAG00142_Bacteroides_stercoris_ATCC_43183
SEQ ID NO: 501-550


CAG00211_Firmicutes_bacterium_CAG_227
SEQ ID NO: 601-650


CAG00218_Bacteroides_sp_CAG_20
SEQ ID NO: 651-700


CAG00243_Lachnospiraceae_bacterium_1_1_57FAA
SEQ ID NO: 551-600


CAG00300_Prevotella_sp_CAG_891
SEQ ID NO: 701-750


CAG00317_Clostridium_sp_CAG_230
SEQ ID NO: 751-800


CAG00327_Faecalibacterium_sp_CAG_74
SEQ ID NO: 801-850


CAG00341_Eubacterium_sp_CAG_115
SEQ ID NO: 851-900


CAG00346_Eubacterium_rectale_M104_1
SEQ ID NO: 901-950


CAG00357_Bacteroides_ovatus_V975
SEQ ID NO: 951-1000


CAG00413_Bacteroides_sp_CAG_144
SEQ ID NO: 1001-1050


CAG00473_Prevotella_sp_CAG_617
SEQ ID NO: 1051-1100


CAG00474_Sutterella_wadsworthensis_2_1_59BFAA
SEQ ID NO: 1101-1150


CAG00487_Prevotella_sp_CAG_279
SEQ ID NO: 351-400


CAG00508_Alistipes_obesi
SEQ ID NO: 1151-1200


CAG00530_Prevotella_sp_CAG_617
SEQ ID NO: 1201-1250


CAG00557_Ruminococcus_callidus_ATCC_27760
SEQ ID NO: 1251-1300


CAG00580_Clostridium_sp_CAG_62
SEQ ID NO: 301-350


CAG00601_uncultured_Faecalibacterium_sp
SEQ ID NO: 201-250


CAG00607_Eubacterium_sp_CAG_251
SEQ ID NO: 1301-1350


CAG00610_Hungatella_hathewayi_12489931
SEQ ID NO: 1351-1400


CAG00624_Firmicutes_bacterium_CAG_552
SEQ ID NO: 1401-1450


CAG00646_Alistipes_sp_CAG_268
SEQ ID NO: 1451-1500


CAG00650_Dorea_formicigenerans_ATCC_27755
SEQ ID NO: 1501-1550


CAG00668_Azospirillum_sp_CAG_239
SEQ ID NO: 1551-1600


CAG00669_Firmicutes_bacterium_CAG_103
SEQ ID NO: 1601-1650


CAG00676_Firmicutes_bacterium_CAG_176
SEQ ID NO: 1651-1700


CAG00698_Ruminococcus_sp_CAG_177
SEQ ID NO: 1701-1750


CAG00713_Firmicutes_bacterium_CAG_270
SEQ ID NO: 1751-1800


CAG00720_Anaerotruncus_colihominis_DSM_17241
SEQ ID NO: 1801-1850


CAG00727_Eggerthella_lenta_DSM_2243
SEQ ID NO: 1851-1900


CAG00766_Firmicutes_bacterium_CAG_176
SEQ ID NO: 251-300


CAG00771_Clostridium_sp_CAG_413
SEQ ID NO: 1901-1950


CAG00782_Eubacterium_rectale_CAG_36
SEQ ID NO: 1951-2000


CAG00861_Oscillibacter_sp_CAG_241
SEQ ID NO: 2051-2100


CAG00873_Butyricimonas_virosa_DSM_23226
SEQ ID NO: 2101-2150


CAG00880_Subdoliqranulum_sp_CAG_314
SEQ ID NO: 2151-2200


CAG00886_Sutterella_sp_CAG_351
SEQ ID NO: 2201-2250


CAG00889_Megasphaera_elsdenii_14_14
SEQ ID NO: 2251-2300


CAG00897_Firmicutes_bacterium_CAG_83
SEQ ID NO: 2301-2350


CAG00919_Clostridium_methylpentosum_DSM_5476
SEQ ID NO: 2351-2400


CAG00928_Acidiphilium_sp_CAG_727
SEQ ID NO: 2401-2450


CAG00937_Clostridium_sp_CAG_7
SEQ ID NO: 2451-2500


CAG00963_Clostridium_sp_CAG_524
SEQ ID NO: 2501-2550


CAG01039_Faecalibacterium_cf_prausnitzii_KLE1255
SEQ ID NO: 2551-2600


CAG01141_Holdemanella_biformis_DSM_3989
SEQ ID NO: 2601-2650


CAG01144_Lactobacillus_vaginalis_DSM_5837
SEQ ID NO: 2651-2700


CAG01158_Pseudoflavonifractor_capillosus
SEQ ID NO: 2701-2750


CAG01197_Dialister_succinatiphilus_YIT_11850
SEQ ID NO: 2751-2800


CAG01208_Coprococcus_catus_GD_7
SEQ ID NO: 2801-2850


CAG01263_Clostridium_clostridioforme_2_1_49FAA
SEQ ID NO: 2851-2900


CAG01321_Faecalibacterium_prausnitzii_SL3_3
SEQ ID NO: 2901-2950









REFERENCES



  • Borghaei, H., Paz-Ares, L., Horn, L., Spigel, D. R., Steins, M., Ready, N. E., Chow, L. Q., Vokes, E. E., Felip, E., Holgado, E., et al. (2015). Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer. N. Engl. J. Med. 373, 1627-1639.

  • Gao, J., Shi, L. Z., Zhao, H., Chen, J., Xiong, L., He, Q., Chen, T., Roszik, J., Bernatchez, C., Woodman, S. E., et al. (2016). Loss of IFN-γ Pathway Genes in Tumor Cells as a Mechanism of Resistance to Anti-CTLA-4 Therapy. Cell 167, 397-404.e9.

  • Gopalakrishnan V, Spencer C N, Nezi L, Reuben A, Andrews M C, Karpinets T V, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018; 359:97-103. doi:10.1126/science.aan4236.

  • Koyama, S., Akbay, E. A., Li, Y. Y., Herter-Sprie, G. S., Buczkowski, K. A., Richards, W. G., Gandhi, L., Redig, A. J., Rodig, S. J., Asahina, H., et al. (2016). Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat. Commun. 7, 10501.

  • Li J.1, Jia H., Cai X., Zhong H., Feng Q., Sunagawa S., Arumugam M., Kultima J. R., Prifti E., Nielsen T. et al. (2014). An integrated catalog of reference genes in the human gut microbiome. Nat Biotechnol. 2014 August; 32(8):834-41.

  • Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre M-L, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018; 359:104-8. doi:10.1126/science.aao3290.

  • Nghiem, P. T., Bhatia, S., Lipson, E. J., Kudchadkar, R. R., Miller, N. J., Annamalai, L., Berry, S., Chartash, E. K., Daud, A., Fling, S. P., et al. (2016). PD-1 Blockade with Pembrolizumab in Advanced Merkel-Cell Carcinoma. N. Engl. J. Med. 374, 2542-2552.

  • Pardoll, D. (2015). Cancer and the Immune System: Basic Concepts and Targets for Intervention. Semin. Oncol. 42, 523-538.

  • Pardoll, D. M. (2012). The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 12, 252-264.

  • Restifo, N. P., Dudley, M. E., and Rosenberg, S. A. (2012). Adoptive immunotherapy for cancer: harnessing the T cell response. Nat. Rev. Immunol. 12, 269-281.

  • Riaz, N., Havel, J. J., Kendall, S. M., Makarov, V., Walsh, L. A., Desrichard, A., Weinhold, N., and Chan, T. A. (2016). Recurrent SERPINB3 and SERPINB4 mutations in patients who respond to anti-CTLA4 immunotherapy. Nat. Genet. 48, 1327-1329.

  • Ribas, A., Hamid, O., Daud, A., Hodi, F. S., Wolchok, J. D., Kefford, R., Joshua, A. M., Patnaik, A., Hwu, W.-J., Weber, J. S., et al. (2016). Association of Pembrolizumab With Tumor Response and Survival Among Patients With Advanced Melanoma. JAMA 315, 1600-1609.

  • Rizvi, N. A., Hellmann, M. D., Snyder, A., Kvistborg, P., Makarov, V., Havel, J. J., Lee, W., Yuan, J., Wong, P., Ho, T. S., et al. (2015). Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348, 124-128.

  • Robert, C., Thomas, L., Bondarenko, I., O'Day, S., Weber, J., Garbe, C., Lebbe, C., Baurain, J.-F., Testori, A., Grob, J.-J., et al. (2011). Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N. Engl. J. Med. 364, 2517-2526.

  • Rosenberg, J. E., Hoffman-Censits, J., Powles, T., van der Heijden, M. S., Balar, A. V., Necchi, A., Dawson, N., O'Donnell, P. H., Balmanoukian, A., Loriot, Y., et al. (2016). Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet Lond. Engl. 387, 1909-1920.

  • Sharma, P., and Allison, J. P. (2015a). Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 161, 205-214.

  • Sharma, P., and Allison, J. P. (2015b). The future of immune checkpoint therapy. Science 348, 56-61.

  • Smyth, M. J., Ngiow, S. F., Ribas, A., and Teng, M. W. L. (2016). Combination cancer immunotherapies tailored to the tumour microenvironment. Nat. Rev. Clin. Oncol. 13, 143-158.

  • Spranger, S., Bao, R., and Gajewski, T. F. (2015). Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 523, 231-235.

  • Taube, J. M., Anders, R. A., Young, G. D., Xu, H., Sharma, R., McMiller, T. L., Chen, S., Klein, A. P., Pardoll, D. M., Topalian, S. L., et al. (2012). Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci. Transl. Med. 4, 127ra37.

  • Topalian, S. L., Hodi, F. S., Brahmer, J. R., Gettinger, S. N., Smith, D. C., McDermott, D. F., Powderly, J. D., Carvajal, R. D., Sosman, J. A., Atkins, M. B., et al. (2012). Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366, 2443-2454.

  • Zaretsky, J. M., Garcia-Diaz, A., Shin, D. S., Escuin-Ordinas, H., Hugo, W., Hu-Lieskovan, S., Torrejon, D. Y., Abril-Rodriguez, G., Sandoval, S., Barthly, L., et al. (2016). Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N. Engl. J. Med. 375, 819-829.

  • Zitvogel, L., Tesniere, A., and Kroemer, G. (2006). Cancer despite immunosurveillance: immunoselection and immunosubversion. Nat. Rev. Immunol. 6, 715-727.X


Claims
  • 1. A method for in vitro determining if an individual having a renal cell cancer (RCC) is likely to respond to a treatment with an immune checkpoint inhibitor (ICI)-based therapy, comprising the following steps: (i) from a fecal sample of said individual, obtaining an abundances pattern based on the relative abundances of a set of bacterial species comprising at least 8 bacterial species selected from the group consisting of:
  • 2-3. (canceled)
  • 4. The method of claim 1, which further comprises a step (iii) of assessing, in an animal model, whether the individual is likely to be a good responder to the treatment with an ICI-based therapy, wherein step (iii) comprises (iiia) performing a fecal microbial transplantation (FMT) of feces from the individual into a germ free (GF) model animal; (iiib) at least 7 to 14 days after step (iiia), inoculating said animal with a transplantable tumor model; (iiic) treating the inoculated animal with the ICI-based therapy; and (iiid) measuring the tumor size in the treated animals, wherein the results of step (iiid) are illustrative of the response that can be expected for said individual to said treatment.
  • 5. The method of claim 1, wherein the individual's antibiotic regimen exposure during the last two months is unknown and wherein, in step (i), the set of bacterial species comprises at least 8 bacterial species selected from the group consisting of:
  • 6. The method of claim 5, wherein in step (ii), one, two or three equations are used, which correspond to models obtained for the following sets of bacterial species, identified by their CAG numbers:
  • 7. The method of claim 6, wherein the equations for calculating the probability that said individual responds to the treatment (PR) are as follows:
  • 8. The method of claim 1, wherein the individual did not take any antibiotic during the last two months and wherein, in step (i), the set of bacterial species comprises at least 8 bacterial species selected from the group consisting of:
  • 9. The method of claim 8, wherein in step (ii), one, two or three equations are used, which correspond to models obtained for the following sets of bacterial species, identified by their CAG numbers:
  • 10. The method of claim 9, wherein the equations for calculating the probability that said individual resists (PNR) or responds (PR) to the treatment are as follows:
  • 11. The method of claim 1, for determining if the individual is likely to have a long-term benefit from a treatment with an ICI-based therapy, wherein the individual's antibiotic regimen exposure during the last two months is unknown and wherein, in step (i), the set of bacterial species comprises at least 8 bacterial species selected from the group consisting of:
  • 12. The method of claim 11, wherein in step (ii), one, two or three equations are used, which correspond to models obtained for the following sets of bacterial species, identified by their CAG numbers:
  • 13. The method of claim 12, wherein the equations for calculating the probability that said individual resists (PNR) to the treatment are as follows:
  • 14. The method of claim 1, for determining if the individual is likely to have a long-term benefit from a treatment with an ICI-based therapy, wherein the individual did not take any antibiotic during the last two months and wherein, in step (i), the set of bacterial species comprises at least 8 bacterial species selected from the group consisting of:
  • 15. The method of claim 14, wherein in step (ii), one, two or three equations are used, which correspond to models obtained for the following sets of bacterial species, identified by their CAG numbers:
  • 16. The method of claim 15, wherein the equations for calculating the probability that said individual resists to the treatment (PNR) or responds (PR) are as follows:
  • 17. The method of claim 1, wherein the fecal sample is obtained before the first administration of ICI.
  • 18. The method of claim 1, wherein the ICI-based therapy is a treatment with an anti-PD1 antibody, an anti-PD-L1 antibody and/or an anti-PD-L2 antibody.
  • 19. (canceled)
  • 20. A theranostic method for determining if a cancer patient needs a bacterial compensation before administration of an ICI-based therapy and/or during such a therapy, comprising assessing, by a method according to claim 1, whether the patient is likely to be a good responder to such a therapy, wherein if the patient is not identified as likely to be a good responder, the patient needs a bacterial compensation.
  • 21. A nucleic acid microarray designed to perform the method of claim 1, characterized in that it comprises nucleic acid probes specific for each of the microorganism species to be detected in said method.
  • 22. A set of primers for performing the method according to claim 1, characterized in that it comprises primer pairs for amplifying sequences specific for each of the microorganism species to be detected in said method.
  • 23. The method of claim 20, further comprising administering a bacterial compensation to the patient.
  • 24. The method of claim 23, further comprising administering an ICI-based therapy to the patient.
Priority Claims (1)
Number Date Country Kind
18306282.7 Sep 2018 EP regional
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2019/076158 9/27/2019 WO 00