PROCESSED MICROBIAL EXTRACELLULAR VESICLES

Information

  • Patent Application
  • 20220296654
  • Publication Number
    20220296654
  • Date Filed
    June 11, 2020
    4 years ago
  • Date Published
    September 22, 2022
    a year ago
Abstract
Provided herein are methods and pharmaceutical compositions related to processed microbial extracellular vesicles (pmEVs) that can be useful as therapeutic agents.
Description
SUMMARY

As disclosed herein, certain types of microbial extracellular vesicles (mEVs), such as processed microbial extracellular vesicles (pmEVs) obtained from microbes (such as bacteria) have therapeutic effects and are useful for the treatment and/or prevention of disease and/or health disorders.


In some embodiments, a pharmaceutical composition provided herein can contain mEVs (such as pmEVs) from one or more microbe source, e.g., one or more bacterial strain. In some embodiments, a pharmaceutical composition provided herein can contain mEVs from one microbe source, e.g., one bacterial strain. The bacterial strain used as a source of mEVs may be selected based on the properties of the bacteria (e.g., growth characteristics, yield, ability to modulate an immune response in an assay or a subject). A pharmaceutical composition comprising mEVs can contain pmEVs. The pharmaceutical composition can comprise a pharmaceutically acceptable excipient.


In some embodiments, a pharmaceutical composition provided herein comprising mEVs (such as pmEVs) can be used for the treatment or prevention of a disease and/or a health disorder, e.g., in a subject (e.g., human).


In some embodiments, a pharmaceutical composition provided herein comprising mEVs (such as pmEVs) can be prepared as powder (e.g., for resuspension) or as a solid dose form, such as a tablet, a minitablet, a capsule, a pill, or a powder; or a combination of these forms (e.g., minitablets comprised in a capsule). The solid dose form can comprise a coating (e.g., enteric coating).


In some embodiments, a pharmaceutical composition provided herein can comprise lyophilized mEVs (such as pmEVs). The lyophilized mEVs (such as pmEVs) can be formulated into a solid dose form, such as a tablet, a minitablet, a capsule, a pill, or a powder; or can be resuspended in a solution.


In some embodiments, a pharmaceutical composition provided herein can comprise gamma irradiated mEVs (such as pmEVs). The gamma irradiated mEVs (such as pmEVs) can be formulated into a solid dose form, such as a tablet, a minitablet, a capsule, a pill, or a powder; or can be resuspended in a solution.


In some embodiments, a pharmaceutical composition provided herein comprising mEVs (such as pmEVs) can be orally administered.


In some embodiments, a pharmaceutical composition provided herein comprising mEVs (such as pmEVs) can be administered intravenously.


In some embodiments, a pharmaceutical composition provided herein comprising mEVs (such as pmEVs) can be administered intratumorally or subtumorally, e.g., to a subject who has a tumor.


In certain aspects, provided herein are pharmaceutical compositions comprising mEVs (such as pmEVs) useful for the treatment and/or prevention of a disease or a health disorder (e.g., adverse health disorders) (e.g., a cancer, an autoimmune disease, an inflammatory disease, a dysbiosis, or a metabolic disease), as well as methods of making and/or identifying such mEVs, and methods of using such pharmaceutical compositions (e.g., for the treatment of a cancer, an autoimmune disease, an inflammatory disease, a dysbiosis, or a metabolic disease, either alone or in combination with other therapeutics). In some embodiments, the pharmaceutical compositions comprise both mEVs and whole microbes from which they were obtained, such as bacteria, (e.g., live bacteria, killed bacteria, attenuated bacteria). In some embodiments, the pharmaceutical compositions comprise mEVs in the absence of microbes from which they were obtained, such as bacteria (e.g., over about 95% (or over about 99%) of the microbe-sourced content of the pharmaceutical composition comprises mEVs).


In some embodiments, the pharmaceutical compositions comprise mEVs from one or more of the bacteria strains or species listed in Table 1, Table 2 and/or Table 3.


In some embodiments, the pharmaceutical composition comprises isolated mEVs (e.g., from one or more strains of bacteria (e.g., bacteria of interest) (e.g., a therapeutically effective amount thereof). E.g., wherein at least 50%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% of the content of the pharmaceutical composition is isolated mEV of bacteria (e.g., bacteria of interest).


In some embodiments, the pharmaceutical composition comprises isolated mEVs (e.g., from one strain of bacteria (e.g., bacteria of interest) (e.g., a therapeutically effective amount thereof). E.g., wherein at least 50%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% of the content of the pharmaceutical composition is isolated mEV of bacteria (e.g., bacteria of interest).


In some embodiments, the pharmaceutical composition comprises processed mEVs (pmEVs).


In some embodiments, the pharmaceutical composition comprises pmEVs and the pmEVs are produced from bacteria that have been gamma irradiated, UV irradiated, heat inactivated, acid treated, or oxygen sparged.


In some embodiments, the pharmaceutical composition comprises pmEVs and the pmEVs are produced from live bacteria.


In some embodiments, the pharmaceutical composition comprises pmEVs and the pmEVs are produced from dead bacteria.


In some embodiments, the pharmaceutical composition comprises pmEVs and the pmEVs are produced from non-replicating bacteria.


In some embodiments, the pharmaceutical composition comprises mEVs and the mEVs are from one strain of bacteria.


In some embodiments, the pharmaceutical composition comprises mEVs and the mEVs are from one strain of bacteria.


In some embodiments, the mEVs are lyophilized (e.g., the lyophilized product further comprises a pharmaceutically acceptable excipient).


In some embodiments, the mEVs are gamma irradiated.


In some embodiments, the mEVs are UV irradiated.


In some embodiments, the mEVs are heat inactivated (e.g., at 50° C. for two hours or at 90° C. for two hours).


In some embodiments, the mEVs are acid treated.


In some embodiments, the mEVs are oxygen sparged (e.g., at 0.1 vvm for two hours).


In some embodiments, the mEVs are from Gram positive bacteria.


In some embodiments, the mEVs are from Gram negative bacteria.


In some embodiments, the Gram negative bacteria belong to class Negativicutes.


In some embodiments, the mEVs are from aerobic bacteria.


In some embodiments, the mEVs are from anaerobic bacteria.


In some embodiments, the mEVs are from acidophile bacteria.


In some embodiments, the mEVs are from alkaliphile bacteria.


In some embodiments, the mEVs are from neutralophile bacteria.


In some embodiments, the mEVs are from fastidious bacteria.


In some embodiments, the mEVs are from nonfastidious bacteria.


In some embodiments, the mEVs are from a bacterial strain listed in Table 1, Table 2, or Table 3.


In some embodiments, the Gram negative bacteria belong to class Negativicutes.


In some embodiments, the Gram negative bacteria belong to family Veillonellaceae, Selenomonadaceae, Acidaminococcaceae, or Sporomusaceae.


In some embodiments, the mEVs are from bacteria of the genus Megasphaera, Selenomonas, Propionospora, or Acidaminococcus.


In some embodiments, the mEVs are Megasphaera sp., Selenomonas felix, Acidaminococcus intestine, or Propionospora sp. bacteria.


In some embodiments, the mEVs are from bacteria of the genus Lactococcus, Prevotella, Bifidobacterium, or Veillonella.


In some embodiments, the mEVs are from Lactococcus lactis cremoris bacteria.


In some embodiments, the mEVs are from Prevotella histicola bacteria.


In some embodiments, the mEVs are from Bifidobacterium animalis bacteria.


In some embodiments, the mEVs are from Veillonella parvula bacteria.


In some embodiments, the mEVs are from Lactococcus lactis cremoris bacteria. In some embodiments, the Lactococcus lactis cremoris bacteria are from a strain comprising at least 90% (or at least 97%) genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Lactococcus lactis cremoris Strain A (ATCC designation number PTA-125368). In some embodiments, the Lactococcus bacteria are from a strain comprising at least 99% genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Lactococcus lactis cremoris Strain A (ATCC designation number PTA-125368). In some embodiments, the Lactococcus bacteria are from Lactococcus lactis cremoris Strain A (ATCC designation number PTA-125368).


In some embodiments, the mEVs are from Prevotella bacteria. In some embodiments, the Prevotella bacteria are from a strain comprising at least 90% (or at least 97%) genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Prevotella Strain B 50329 (NRRL accession number B 50329). In some embodiments, the Prevotella bacteria are from a strain comprising at least 99% genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Prevotella Strain B 50329 (NRRL accession number B 50329). In some embodiments, the Prevotella bacteria are from Prevotella Strain B 50329 (NRRL accession number B 50329).


In some embodiments, the mEVs are from Bifidobacterium bacteria. In some embodiments, the Bifidobacterium bacteria are from a strain comprising at least 90% (or at least 97%) genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Bifidobacterium bacteria deposited as ATCC designation number PTA-125097. In some embodiments, the Bifidobacterium bacteria are from a strain comprising at least 99% genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Bifidobacterium bacteria deposited as ATCC designation number PTA-125097. In some embodiments, the Bifidobacterium bacteria are from Bifidobacterium bacteria deposited as ATCC designation number PTA-125097.


In some embodiments, the mEVs are from Veillonella bacteria. In some embodiments, the Veillonella bacteria are from a strain comprising at least 90% (or at least 97%) genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Veillonella bacteria deposited as ATCC designation number PTA-125691. In some embodiments, the Veillonella bacteria are from a strain comprising at least 99% genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Veillonella bacteria deposited as ATCC designation number PTA-125691. In some embodiments, the Veillonella bacteria are from Veillonella bacteria deposited as ATCC designation number PTA-125691.


In some embodiments, the mEVs are from Ruminococcus gnavus bacteria. In some embodiments, the Ruminococcus gnavus bacteria are from a strain comprising at least 90% (or at least 97%) genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Ruminococcus gnavus bacteria deposited as ATCC designation number PTA-126695. In some embodiments, the Ruminococcus gnavus bacteria are from a strain comprising at least 99% genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Ruminococcus gnavus bacteria deposited as ATCC designation number PTA-126695. In some embodiments, the Ruminococcus gnavus bacteria are from Ruminococcus gnavus bacteria deposited as ATCC designation number PTA-126695.


In some embodiments, the mEVs are from Megasphaera sp. bacteria. In some embodiments, the Megasphaera sp. bacteria are from a strain comprising at least 90% (or at least 97%) genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Megasphaera sp. bacteria deposited as ATCC designation number PTA-126770. In some embodiments, the Megasphaera sp. bacteria are from a strain comprising at least 99% genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Megasphaera sp. bacteria deposited as ATCC designation number PTA-126770. In some embodiments, the Megasphaera sp. bacteria are from Megasphaera sp. bacteria deposited as ATCC designation number PTA-126770.


In some embodiments, the mEVs are from Fournierella massiliensis bacteria. In some embodiments, the Fournierella massiliensis bacteria are from a strain comprising at least 90% (or at least 97%) genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Fournierella massiliensis bacteria deposited as ATCC designation number PTA-126694. In some embodiments, the Fournierella massiliensis bacteria are from a strain comprising at least 99% genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Fournierella massiliensis bacteria deposited as ATCC designation number PTA-126694. In some embodiments, the Fournierella massiliensis bacteria are from Fournierella massiliensis bacteria deposited as ATCC designation number PTA-126694.


In some embodiments, the mEVs are from Harryflintia acetispora bacteria. In some embodiments, the Harryflintia acetispora bacteria are from a strain comprising at least 90% (or at least 97%) genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Harryflintia acetispora bacteria deposited as ATCC designation number PTA-126696. In some embodiments, the Harryflintia acetispora bacteria are from a strain comprising at least 99% genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Harryflintia acetispora bacteria deposited as ATCC designation number PTA-126696. In some embodiments, the Harryflintia acetispora bacteria are from Harryflintia acetispora bacteria deposited as ATCC designation number PTA-126696.


In some embodiments, the mEVs are from bacteria of the genus Akkermansia, Christensenella, Blautia, Enterococcus, Eubacterium, Roseburia, Bacteroides, Parabacteroides, or Erysipelatoclostridium.


In some embodiments, the mEVs are from Blautia hydrogenotrophica, Blautia stercoris, Blautia wexlerae, Eubacterium faecium, Eubacterium contortum, Eubacterium rectale, Enterococcus faecalis, Enterococcus durans, Enterococcus villorum, Enterococcus gallinarum; Bifidobacterium lacus, Bifidobacterium bifidium, Bifidobacterium longum, Bifidobacterium animalis, or Bifidobacterium breve bacteria.


In some embodiments, the mEVs are from BCG (bacillus Calmette-Guerin), Parabacteroides, Blautia, Veillonella, Lactobacillus salivarius, Agathobaculum, Ruminococcus gnavus, Paraclostridium benzoelyticum, Turicibacter sanguinus, Burkholderia, Klebsiella quasipneumoniae ssp similpneumoniae, Klebsiella oxytoca, Tyzzerela nexilis, or Neisseria bacteria.


In some embodiments, the mEVs are from Blautia hydrogenotrophica bacteria.


In some embodiments, the mEVs are from Blautia stercoris bacteria.


In some embodiments, the mEVs are from Blautia wexlerae bacteria.


In some embodiments, the mEVs are from Enterococcus gallinarum bacteria.


In some embodiments, the mEVs are from Enterococcus faecium bacteria.


In some embodiments, the mEVs are from Bifidobacterium bifidium bacteria.


In some embodiments, the mEVs are from Bifidobacterium breve bacteria.


In some embodiments, the mEVs are from Bifidobacterium longum bacteria.


In some embodiments, the mEVs are from Roseburia hominis bacteria.


In some embodiments, the mEVs are from Bacteroides thetaiotaomicron bacteria.


In some embodiments, the mEVs are from Bacteroides coprocola bacteria.


In some embodiments, the mEVs are from Erysipelatoclostridium ramosum bacteria.


In some embodiments, the mEVs are from Megasphera massiliensis bacteria.


In some embodiments, the mEVs are from Eubacterium bacteria.


In some embodiments, the mEVs are from Parabacteroides distasonis bacteria.


In certain aspects, the mEVs (such as pmEVs) are obtained from bacteria that have been selected based on certain desirable properties, such as reduced toxicity and adverse effects (e.g., by removing or deleting lipopolysaccharide (LPS)), enhanced oral delivery (e.g., by improving acid resistance, muco-adherence and/or penetration and/or resistance to bile acids, resistance to anti-microbial peptides and/or antibody neutralization), target desired cell types (e.g., M-cells, goblet cells, enterocytes, dendritic cells, macrophages), improved bioavailability systemically or in an appropriate niche (e.g., mesenteric lymph nodes, Peyer's patches, lamina propria, tumor draining lymph nodes, and/or blood), enhanced immunomodulatory and/or therapeutic effect (e.g., either alone or in combination with another therapeutic agent), enhanced immune activation , and/or manufacturing attributes (e.g., growth characteristics, yield, greater stability, improved freeze-thaw tolerance, shorter generation times).


In certain aspects, the mEVs are from engineered bacteria that are modified to enhance certain desirable properties. In some embodiments, the engineered bacteria are modified so that mEVs (such as pmEVs) produced therefrom will have reduced toxicity and adverse effects (e.g., by removing or deleting lipopolysaccharide (LPS)), enhanced oral delivery (e.g., by improving acid resistance, muco-adherence and/or penetration and/or resistance to bile acids, resistance to anti-microbial peptides and/or antibody neutralization), target desired cell types (e.g., M-cells, goblet cells, enterocytes, dendritic cells, macrophages), improved bioavailability systemically or in an appropriate niche (e.g., mesenteric lymph nodes, Peyer's patches, lamina propria, tumor draining lymph nodes, and/or blood), enhanced immunomodulatory and/or therapeutic effect (e.g., either alone or in combination with another therapeutic agent), enhanced immune activation, and/or improved manufacturing attributes (e.g., growth characteristics, yield, greater stability, improved freeze-thaw tolerance, shorter generation times). In some embodiments, provided herein are methods of making such mEVs (such as pmEVs).


In certain aspects, provided herein are pharmaceutical compositions comprising mEVs (such as pmEVs) useful for the treatment and/or prevention of a disease or a health disorder (e.g., a cancer, an autoimmune disease, an inflammatory disease, or a metabolic disease), as well as methods of making and/or identifying such mEVs, and methods of using such pharmaceutical compositions (e.g., for the treatment of a cancer, an autoimmune disease, an inflammatory disease, or a metabolic disease), either alone or in combination with one or more other therapeutics.


Pharmaceutical compositions containing mEVs (such pmEVs) can provide potency comparable to or greater than pharmaceutical compositions that contain the whole microbes from which the mEVs were obtained. For example, at the same dose of mEVs (e.g., based on particle count or protein content), a pharmaceutical composition containing mEVs can provide potency comparable to or greater than a comparable pharmaceutical composition that contains whole microbes of the same bacterial strain from which the mEVs were obtained. Such mEV containing pharmaceutical compositions can allow the administration of higher doses and elicit a comparable or greater (e.g., more effective) response than observed with a comparable pharmaceutical composition that contains whole microbes of the same bacterial strain from which the mEVs were obtained.


As a further example, at the same dose (e.g., based on particle count or protein content), a pharmaceutical composition containing mEVs may contain less microbially-derived material (based on particle count or protein content), as compared to a pharmaceutical composition that contains the whole microbes of the same bacterial strain from which the mEVs were obtained, while providing an equivalent or greater therapeutic benefit to the subject receiving such pharmaceutical composition.


As a further example, mEVs can be administered at doses e.g., of about 1×10-about 1×1015 particles, e.g., as measured by NTA.


As another example, mEVs can be administered at doses e.g., of about 5 mg to about 900 mg total protein, e.g., as measured by Bradford assay. As another example, mEVs can be administered at doses e.g., of about 5 mg to about 900 mg total protein, e.g., as measured by BCA assay.


In certain embodiments, provided herein are methods of treating a subject who has cancer comprising administering to the subject a pharmaceutical composition described herein. In certain embodiments, provided herein are methods of treating a subject who has an immune disorder (e.g., an autoimmune disease, an inflammatory disease, an allergy) comprising administering to the subject a pharmaceutical composition described herein. In certain embodiments, provided herein are methods of treating a subject who has a metabolic disease comprising administering to the subject a pharmaceutical composition described herein. In certain embodiments, provided herein are methods of treating a subject who has a neurologic disease comprising administering to the subject a pharmaceutical composition described herein.


In some embodiments, the method further comprises administering to the subject an antibiotic. In some embodiments, the method further comprises administering to the subject one or more other cancer therapies (e.g., surgical removal of a tumor, the administration of a chemotherapeutic agent, the administration of radiation therapy, and/or the administration of a cancer immunotherapy, such as an immune checkpoint inhibitor, a cancer-specific antibody, a cancer vaccine, a primed antigen presenting cell, a cancer-specific T cell, a cancer-specific chimeric antigen receptor (CAR) T cell, an immune activating protein, and/or an adjuvant). In some embodiments, the method further comprises the administration of another therapeutic bacterium and/or mEVs (such as pmEVs) from one or more other bacterial strains (e.g., therapeutic bacterium). In some embodiments, the method further comprises the administration of an immune suppressant and/or an anti-inflammatory agent. In some embodiments, the method further comprises the administration of a metabolic disease therapeutic agent.


In certain aspects, provided herein is a pharmaceutical composition comprising mEVs (such as pmEVs) for use in the treatment and/or prevention of a disease (e.g., a cancer, an autoimmune disease, an inflammatory disease, a dysbiosis, or a metabolic disease) or a health disorder, either alone or in combination with one or more other therapeutic agent.


In certain embodiments, provided herein is a pharmaceutical composition comprising mEVs (such as pmEVs) for use in treating and/or preventing a cancer in a subject (e.g., human). The pharmaceutical composition can be used either alone or in combination with one or more other therapeutic agent for the treatment of the cancer. In certain embodiments, provided herein is a pharmaceutical composition comprising mEVs (such as pmEVs) for use in treating and/or preventing an immune disorder (e.g., an autoimmune disease, an inflammatory disease, an allergy) in a subject (e.g., human). The pharmaceutical composition can be used either alone or in combination with one or more other therapeutic agent for the treatment of the immune disorder. In certain embodiments, provided herein is a pharmaceutical composition comprising mEVs (such as pmEVs) for use in treating and/or preventing a dysbiosis in a subject (e.g., human). The pharmaceutical composition can be used either alone or in combination with therapeutic agent for the treatment of the dysbiosis. In certain embodiments, provided herein is a pharmaceutical composition comprising mEVs (such as pmEVs) for use in treating and/or preventing a metabolic disease in a subject (e.g., human). The pharmaceutical composition can be used either alone or in combination with therapeutic agent for the treatment of the metabolic disease. In certain embodiments, provided herein is a pharmaceutical composition comprising mEVs (such as pmEVs) for use in treating and/or preventing a neurologic disease in a subject (e.g., human). The pharmaceutical composition can be used either alone or in combination with one or more other therapeutic agent for treatment of the neurologic disorder.


In some embodiments, the pharmaceutical composition comprising mEVs can be for use in combination with an antibiotic. In some embodiments, the pharmaceutical composition comprising mEVs can be for use in combination with one or more other cancer therapies (e.g., surgical removal of a tumor, the use of a chemotherapeutic agent, the use of radiation therapy, and/or the use of a cancer immunotherapy, such as an immune checkpoint inhibitor, a cancer-specific antibody, a cancer vaccine, a primed antigen presenting cell, a cancer-specific T cell, a cancer-specific chimeric antigen receptor (CAR) T cell, an immune activating protein, and/or an adjuvant). In some embodiments, the pharmaceutical composition comprising mEVs can be for use in combination with another therapeutic bacterium and/or mEVs obtained from one or more other bacterial strains (e.g., therapeutic bacterium). In some embodiments, the pharmaceutical composition comprising mEVs can be for use in combination with one or more immune suppressant(s) and/or an anti-inflammatory agent(s). In some embodiments, the pharmaceutical composition comprising mEVs can be for use in combination with one or more other metabolic disease therapeutic agents.


In certain aspects, provided herein is use of a pharmaceutical composition comprising mEVs (such as pmEVs) for the preparation of a medicament for the treatment and/or prevention of a disease (e.g., a cancer, an autoimmune disease, an inflammatory disease, a dysbiosis, or a metabolic disease), either alone or in combination with another therapeutic agent. In some embodiments, the use is in combination with another therapeutic bacterium and/or mEVs obtained from one or more other bacterial strains (e.g., therapeutic bacterium).


In certain embodiments, provided herein is use of a pharmaceutical composition comprising mEVs (such as pmEVs) for the preparation of a medicament for treating and/or preventing a cancer in a subject (e.g., human). The pharmaceutical composition can be for use either alone or in combination with another therapeutic agent for the cancer. In certain embodiments, provided herein is use of a pharmaceutical composition comprising mEVs (for the preparation of a medicament for treating and/or preventing an immune disorder (e.g., an autoimmune disease, an inflammatory disease, an allergy) in a subject (e.g., human). The pharmaceutical composition can be for use either alone or in combination with another therapeutic agent for the immune disorder. In certain embodiments, provided herein is use of a pharmaceutical composition comprising mEVs (such as pmEVs) for the preparation of a medicament for treating and/or preventing a dysbiosis in a subject (e.g., human). The pharmaceutical composition can be for use either alone or in combination with another therapeutic agent for the dysbiosis. In certain embodiments, provided herein is use of a pharmaceutical composition comprising mEVs (such as pmEVs) for the preparation of a medicament for treating and/or preventing a metabolic disease in a subject (e.g., human). The pharmaceutical composition can be for use either alone or in combination with another therapeutic agent for the metabolic disease. In certain embodiments, provided herein is use of a pharmaceutical composition comprising mEVs (such as pmEVs) for the preparation of a medicament for treating and or preventing a neurologic disease in a subject (e.g., human). The pharmaceutical composition can be for use either alone or in combination with another therapeutic agent for the neurologic disorder.


In some embodiments, the pharmaceutical composition comprising mEVs can be for use in combination with an antibiotic. In some embodiments, the pharmaceutical composition comprising mEVs can for use in combination with one or more other cancer therapies (e.g., surgical removal of a tumor, the use of a chemotherapeutic agent, the use of radiation therapy, and/or the use of a cancer immunotherapy, such as an immune checkpoint inhibitor, a cancer-specific antibody, a cancer vaccine, a primed antigen presenting cell, a cancer-specific T cell, a cancer-specific chimeric antigen receptor (CAR) T cell, an immune activating protein, and/or an adjuvant). In some embodiments, the pharmaceutical composition comprising mEVs can be for use in combination with another therapeutic bacterium and/or mEVs obtained from one or more other bacterial strains (e.g., therapeutic bacterium). In some embodiments, the pharmaceutical composition comprising mEVs can be for use in combination with one or more other immune suppressant(s) and/or an anti-inflammatory agent(s). In some embodiments, the pharmaceutical composition can be for use in combination with one or more other metabolic disease therapeutic agent(s).


A pharmaceutical composition, e.g., as described herein, comprising mEVs (such as pmEVs) can provide a therapeutically effective amount of mEVs to a subject, e.g., a human.


A pharmaceutical composition, e.g., as described herein, comprising mEVs (such as pmEVs) can provide a non-natural amount of the therapeutically effective components (e.g., present in the mEVs (such as pmEVs) to a subject, e.g., a human.


A pharmaceutical composition, e.g., as described herein, comprising mEVs (such as pmEVs) can provide unnatural quantity of the therapeutically effective components (e.g., present in the mEVs (such as pmEVs) to a subject, e.g., a human.


A pharmaceutical composition, e.g., as described herein, comprising mEVs (such as pmEVs) can bring about one or more changes to a subject, e.g., human, e.g., to treat or prevent a disease or a health disorder.


A pharmaceutical composition, e.g., as described herein, comprising mEVs (such as pmEVs) has potential for significant utility, e.g., to affect a subject, e.g., a human, e.g., to treat or prevent a disease or a health disorder.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1 shows the efficacy of i.v. administered processed microbial extracellular vesicles (pmEVs) from B. animalis ssp. lactis compared to that of i.p. administered anti-PD-1 or vehicle in a mouse colorectal carcinoma model at day 11.



FIG. 2 shows the efficacy of i.v. administered pmEVs from Anaerostipes hadrus compared to that of i.p. administered anti-PD-1 or vehicle in a mouse colorectal carcinoma model at day 11.



FIG. 3 shows the efficacy of i.v. administered pmEVs from S. pyogenes compared to that of i.p. administered anti-PD-1 or vehicle in a mouse colorectal carcinoma model at day 11.



FIG. 4 shows the efficacy of i.v. administered pmEVs from P. benzoelyticum compared to that of i.p. administered anti-PD-1 or vehicle in a mouse colorectal carcinoma model at day 11.



FIG. 5 shows the efficacy of i.v. administered pmEVs from Hungatella sp. compared to that of i.p. administered anti-PD-1 or vehicle in a mouse colorectal carcinoma model at day 11.



FIG. 6 shows the efficacy of i.v. administered pmEVs from S. aureus compared to that of i.p. administered anti-PD-1 or vehicle in a mouse colorectal carcinoma model at day 11.



FIG. 7 shows the efficacy of i.v. administered pmEVs from R. gnavus compared to that of i.p. administered anti-PD-1 or vehicle in a mouse colorectal carcinoma model at day 11.



FIG. 8 shows the efficacy of i.v. administered pmEVs from B. animalis ssp. lactis and Megasphaera massiliensis compared to that of i.p. administered anti-PD-1 or vehicle in a mouse colorectal carcinoma model at day 11.



FIG. 9 shows the efficacy of i.v. administered pmEVs from R. gnavus compared to that of intraperitoneally (i.p.) administered anti-PD-1 or vehicle in a mouse colorectal carcinoma model at day 9.



FIG. 10 shows the efficacy of i.v. administered pmEVs from R. gnavus compared to that of i.p. administered anti-PD-1 or vehicle in a mouse colorectal carcinoma model at day 11.



FIG. 11 shows the efficacy of i.v. administered pmEVs from B. animalis ssp. lactis alone or in combination with anti-PD-1 compared to that of anti-PD-1 (alone) or vehicle in a mouse colorectal carcinoma model at day 9.



FIG. 12 shows the efficacy of i.v. administered pmEVs from B. animahs ssp. lactis alone or in combination with anti-PD-1 compared to that of anti-PD-1 (alone) or vehicle in a mouse colorectal carcinoma model at day 11.



FIG. 13 shows the efficacy of i.v. administered pmEVs from P. distasonis compared to that of i.p. administered anti-PD-1 or vehicle in a mouse colorectal carcinoma model at day 9.



FIG. 14 shows the efficacy of i.v. administered pmEVs from P. distasonis compared to that of i.p. administered anti-PD-1 or vehicle in a mouse colorectal carcinoma model at day 11.



FIG. 15 shows the efficacy of orally-gavaged pmEVs from P. histicola compared to dexamethasone. pmEVs from P. histicola were tested at low (6.0E+07), medium (6.0E+09), and high (6.0E+11) dosages.



FIG. 16 shows the efficacy of i.v. administered smEVs from V. parvula compared to that of i.p. administered anti-PD-1 or vehicle in a mouse colorectal carcinoma model at day 11.



FIG. 17 shows the efficacy of i.v. administered smEVs from V. parvula compared to that of i.p. administered anti-PD-1 or vehicle in a mouse colorectal carcinoma model at day 11. smEVs from V. parvula were tested at 2 ug/dose, 5 ug/dose, and 10 ug/dose.



FIG. 18 shows the efficacy of i.v. administered smEVs from V. atypica compared to that of i.p. administered anti-PD-1 or vehicle in a mouse colorectal carcinoma model at day 11. smEVs from V. atypica were tested at 2.0e+11PC, 7.0e+10PC, and 1.5e+10PC.



FIG. 19 shows the efficacy of i.v. administered smEVs from V. tobetsuensis compared to that of i.p. administered anti-PD-1 or vehicle in a mouse colorectal carcinoma model at day 11. smEVs from V. tobetsuensis were tested at 2 ug/dose, 5 ug/dose, and 10 ug/dose.



FIG. 20 shows the efficacy of orally administered smEVs and lyophilized smEVs from Prevotella histicola at high (6.0 e+11 particle count), medium (6.0 e+9 particle count), and low (6.0 e+7 particle count) concentrations in reducing antigen-specific ear swelling (ear thickness) at 24 hours compared to vehicle (negative control) and dexamethasone (positive control) following antigen challenge in a KLH-based delayed type hypersensitivity model.



FIG. 21 shows the efficacy (as determined by 24-hour ear measurements) of three doses (low, mid, and high) of pmEVs and lyophilized pmEVs from a Prevotella histicola (P. histicola) strain as compared to the efficacy of powder from the same Prevotella histicola strain in reducing ear thickness at a 24-hour time point in a DTH model. Dexamethasone was used as a positive control.



FIG. 22 shows the efficacy (as determined by 24-hour ear measurements) of three doses (low, mid, and high) of smEVs from a Veillonella parvula (V. parvula) strain and of pmEVs and gamma irradiated (GI) pmEVs from the same Veillonella parvula strain as compared to the efficacy of gamma irradiated (GI) powder from the same Veillonella parvula strain in reducing ear thickness at a 24-hour time point in a DTH model. Dexamethasone was used as a positive control.



FIG. 23 shows the efficacy (as determined by 24-hour ear measurements) of two doses (low and high) of smEVs from Megasphaera Sp. Strain A.



FIG. 24 shows the efficacy (as determined by 24-hour ear measurements) of two doses (low and high) of smEVs from Megasphaera Sp. Strain B.



FIG. 25 shows shows the efficacy (as determined by 24-hour ear measurements) of two doses (low and high) of smEVs from Selenomonas felix.



FIG. 26 shows smEVs from Megasphaera Sp. Strain A induce cytokine production from PMA-differentiated U937 cells. U937 cells were treated with smEV at 1×106-1×109 concentrations as well as TLR2 (FSL) and TLR4 (LPS) agonist controls for 24 hrs and cytokine production was measured. “Blank” indicates the medium control.



FIGS. 27A and 27B show Day 22 Tumor Volume Summary (FIG. 27A) and Tumor Volume Curves (FIG. 27B) comparing Megasphaera sp. Strain A smEV (2e11) against a negative control (Vehicle PBS), and positive control (anti-PD-1).



FIGS. 28A and 28B show Day 23 Tumor Volume Summary (FIG. 28A) and Tumor Volume Curves (FIG. 28B) comparing Megasphaera sp. Strain A smEV smEVs at 3 doses (2e11, 2e9, and 2e7) BID, as well as Megasphaera sp. smEVs (2e11) QD against a negative control (Vehicle PBS), and positive control (anti-PD-1).



FIG. 29 shows tumor volumes after d10 tumors were dosed once daily for 14 days with pmEVs from E. gallinarum Strains A and B.



FIG. 30 shows EVs from Megasphaera Sp. Strain A induce cytokine production from PMA-differentiated U937 cells. Cytokine release was measured by MSD ELISA. TLR2 (FSL) and TLR4 (LPS) agonists were used as controls. Blank indicates the media control.



FIG. 31 shows EVs from Megasphaera Sp. Strain B induce cytokine production from PMA-differentiated U937 cells. Cytokine release was measured by MSD ELISA. TLR2 (FSL) and TLR4 (LPS) agonists were used as controls. Blank indicates the media control.



FIG. 32 shows EVs from Selenomonas felix induce cytokine production from PMA-differentiated U937 cells. Cytokine release was measured by MSD ELISA. TLR2 (FSL) and TLR4 (LPS) agonists were used as controls. Blank indicates the media control.



FIG. 33 shows EVs from Acidaminococcus intestini induce cytokine production from PMA-differentiated U937 cells. Cytokine release was measured by MSD ELISA. TLR2 (FSL) and TLR4 (LPS) agonists were used as controls. Blank indicates the media control.



FIG. 34 shows EVs from Propionospora sp. induce cytokine production from PMA-differentiated U937 cells. Cytokine release was measured by MSD ELISA. TLR2 (FSL) and TLR4 (LPS) agonists were used as controls. Blank indicates the media control.





DETAILED DESCRIPTION
Definitions

“Adjuvant” or “Adjuvant therapy” broadly refers to an agent that affects an immunological or physiological response in a patient or subject (e.g., human). For example, an adjuvant might increase the presence of an antigen over time or to an area of interest like a tumor, help absorb an antigen presenting cell antigen, activate macrophages and lymphocytes and support the production of cytokines. By changing an immune response, an adjuvant might permit a smaller dose of an immune interacting agent to increase the effectiveness or safety of a particular dose of the immune interacting agent. For example, an adjuvant might prevent T cell exhaustion and thus increase the effectiveness or safety of a particular immune interacting agent.


“Administration” broadly refers to a route of administration of a composition (e.g., a pharmaceutical composition) to a subject. Examples of routes of administration include oral administration, rectal administration, topical administration, inhalation (nasal) or injection. Administration by injection includes intravenous (IV), intramuscular (IM), intratumoral (IT) and subcutaneous (SC) administration. A pharmaceutical composition described herein can be administered in any form by any effective route, including but not limited to intratumoral, oral, parenteral, enteral, intravenous, intraperitoneal, topical, transdermal (e.g., using any standard patch), intradermal, ophthalmic, (intra)nasally, local, non-oral, such as aerosol, inhalation, subcutaneous, intramuscular, buccal, sublingual, (trans)rectal, vaginal, intra-arterial, and intrathecal, transmucosal (e.g., sublingual, lingual, (trans)buccal, (trans)urethral, vaginal (e.g., trans- and perivaginally), implanted, intravesical, intrapulmonary, intraduodenal, intragastrical, and intrabronchial. In preferred embodiments, a pharmaceutical composition described herein is administered orally, rectally, intratumorally, topically, intravesically, by injection into or adjacent to a draining lymph node, intravenously, by inhalation or aerosol, or subcutaneously. In another preferred embodiment, a pharmaceutical composition described herein is administered orally, intratumorally, or intravenously.


As used herein, the term “antibody” may refer to both an intact antibody and an antigen binding fragment thereof. Intact antibodies are glycoproteins that include at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds. Each heavy chain includes a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. Each light chain includes a light chain variable region (abbreviated herein as VL) and a light chain constant region. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The term “antibody” includes, for example, monoclonal antibodies, polyclonal antibodies, chimeric antibodies, humanized antibodies, human antibodies, multispecific antibodies (e.g., bispecific antibodies), single-chain antibodies and antigen-binding antibody fragments.


The terms “antigen binding fragment” and “antigen-binding portion” of an antibody, as used herein, refer to one or more fragments of an antibody that retain the ability to bind to an antigen. Examples of binding fragments encompassed within the term “antigen-binding fragment” of an antibody include Fab, Fab′, F(ab′)2, Fv, scFv, disulfide linked Fv, Fd, diabodies, single-chain antibodies, NANOBODIES®, isolated CDRH3, and other antibody fragments that retain at least a portion of the variable region of an intact antibody. These antibody fragments can be obtained using conventional recombinant and/or enzymatic techniques and can be screened for antigen binding in the same manner as intact antibodies.


“Cancer” broadly refers to an uncontrolled, abnormal growth of a host's own cells leading to invasion of surrounding tissue and potentially tissue distal to the initial site of abnormal cell growth in the host. Major classes include carcinomas which are cancers of the epithelial tissue (e.g., skin, squamous cells); sarcomas which are cancers of the connective tissue (e.g., bone, cartilage, fat, muscle, blood vessels, etc.); leukemias which are cancers of blood forming tissue (e.g., bone marrow tissue); lymphomas and myelomas which are cancers of immune cells; and central nervous system cancers which include cancers from brain and spinal tissue. “Cancer(s) and” “neoplasm(s)” are used herein interchangeably. As used herein, “cancer” refers to all types of cancer or neoplasm or malignant tumors including leukemias, carcinomas and sarcomas, whether new or recurring. Specific examples of cancers are: carcinomas, sarcomas, myelomas, leukemias, lymphomas and mixed type tumors. Non-limiting examples of cancers are new or recurring cancers of the brain, melanoma, bladder, breast, cervix, colon, head and neck, kidney, lung, non-small cell lung, mesothelioma, ovary, prostate, sarcoma, stomach, uterus and medulloblastoma. In some embodiments, the cancer comprises a solid tumor. In some embodiments, the cancer comprises a metastasis.


A “carbohydrate” refers to a sugar or polymer of sugars. The terms “saccharide,” “polysaccharide,” “carbohydrate,” and “oligosaccharide” may be used interchangeably. Most carbohydrates are aldehydes or ketones with many hydroxyl groups, usually one on each carbon atom of the molecule. Carbohydrates generally have the molecular formula CnH2nOn. A carbohydrate may be a monosaccharide, a disaccharide, trisaccharide, oligosaccharide, or polysaccharide. The most basic carbohydrate is a monosaccharide, such as glucose, galactose, mannose, ribose, arabinose, xylose, and fructose. Disaccharides are two joined monosaccharides. Exemplary disaccharides include sucrose, maltose, cellobiose, and lactose. Typically, an oligosaccharide includes between three and six monosaccharide units (e.g., raffinose, stachyose), and polysaccharides include six or more monosaccharide units. Exemplary polysaccharides include starch, glycogen, and cellulose. Carbohydrates may contain modified saccharide units such as 2′-deoxyribose wherein a hydroxyl group is removed, 2′-fluororibose wherein a hydroxyl group is replaced with a fluorine, or N-acetylglucosamine, a nitrogen-containing form of glucose (e.g., 2′-fluororibose, deoxyribose, and hexose). Carbohydrates may exist in many different forms, for example, conformers, cyclic forms, acyclic forms, stereoisomers, tautomers, anomers, and isomers.


“Cellular augmentation” broadly refers to the influx of cells or expansion of cells in an environment that are not substantially present in the environment prior to administration of a composition and not present in the composition itself. Cells that augment the environment include immune cells, stromal cells, bacterial and fungal cells. Environments of particular interest are the microenvironments where cancer cells reside or locate. In some instances, the microenvironment is a tumor microenvironment or a tumor draining lymph node. In other instances, the microenvironment is a pre-cancerous tissue site or the site of local administration of a composition or a site where the composition will accumulate after remote administration.


“Clade” refers to the OTUs or members of a phylogenetic tree that are downstream of a statistically valid node in a phylogenetic tree. The clade comprises a set of terminal leaves in the phylogenetic tree that is a distinct monophyletic evolutionary unit and that share some extent of sequence similarity.


A “combination” of mEVs (such as smEVs) from two or more microbial strains includes the physical co-existence of the microbes from which the mEVs (such as smEVs) are obtained, either in the same material or product or in physically connected products, as well as the temporal co-administration or co-localization of the mEVs (such as smEVs) from the two strains.


“Dysbiosis” refers to a state of the microbiota or microbiome of the gut or other body area, including, e.g., mucosal or skin surfaces (or any other microbiome niche) in which the normal diversity and/or function of the host gut microbiome ecological networks (“microbiome”) are disrupted. A state of dysbiosis may result in a diseased state, or it may be unhealthy under only certain conditions or only if present for a prolonged period. Dysbiosis may be due to a variety of factors, including, environmental factors, infectious agents, host genotype, host diet and/or stress. A dysbiosis may result in: a change (e.g., increase or decrease) in the prevalence of one or more bacteria types (e.g., anaerobic), species and/or strains, change (e.g., increase or decrease) in diversity of the host microbiome population composition; a change (e.g., increase or reduction) of one or more populations of symbiont organisms resulting in a reduction or loss of one or more beneficial effects; overgrowth of one or more populations of pathogens (e.g., pathogenic bacteria); and/or the presence of, and/or overgrowth of, symbiotic organisms that cause disease only when certain conditions are present.


The term “decrease” or “deplete” means a change, such that the difference is, depending on circumstances, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 1/100, 1/1000, 1/10,000, 1/100,000, 1/1,000,000 or undetectable after treatment when compared to a pre-treatment state. Properties that may be decreased include the number of immune cells, bacterial cells, stromal cells, myeloid derived suppressor cells, fibroblasts, metabolites; the level of a cytokine; or another physical parameter (such as ear thickness (e.g., in a DTH animal model) or tumor size (e.g., in an animal tumor model)).


The term “ecological consortium” is a group of bacteria which trades metabolites and positively co-regulates one another, in contrast to two bacteria which induce host synergy through activating complementary host pathways for improved efficacy.


As used herein, “engineered bacteria” are any bacteria that have been genetically altered from their natural state by human activities, and the progeny of any such bacteria. Engineered bacteria include, for example, the products of targeted genetic modification, the products of random mutagenesis screens and the products of directed evolution.


The term “epitope” means a protein determinant capable of specific binding to an antibody or T cell receptor. Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains. Certain epitopes can be defined by a particular sequence of amino acids to which an antibody is capable of binding.


The term “gene” is used broadly to refer to any nucleic acid associated with a biological function. The term “gene” applies to a specific genomic sequence, as well as to a cDNA or an mRNA encoded by that genomic sequence.


“Identity” as between nucleic acid sequences of two nucleic acid molecules can be determined as a percentage of identity using known computer algorithms such as the “FASTA” program, using for example, the default parameters as in Pearson et al. (1988) Proc. Natl. Acad. Sci. USA 85:2444 (other programs include the GCG program package (Devereux, J., et al., Nucleic Acids Research 12(I):387 (1984)), BLASTP, BLASTN, FASTA Atschul, S. F., et al., J Molec Biol 215:403 (1990); Guide to Huge Computers, Mrtin J. Bishop, ed., Academic Press, San Diego, 1994, and Carillo et al. (1988) SIAM J Applied Math 48:1073). For example, the BLAST function of the National Center for Biotechnology Information database can be used to determine identity. Other commercially or publicly available programs include, DNAStar “MegAlign” program (Madison, Wis.) and the University of Wisconsin Genetics Computer Group (UWG) “Gap” program (Madison Wis.)).


As used herein, the term “immune disorder” refers to any disease, disorder or disease symptom caused by an activity of the immune system, including autoimmune diseases, inflammatory diseases and allergies. Immune disorders include, but are not limited to, autoimmune diseases (e.g., psoriasis, atopic dermatitis, lupus, scleroderma, hemolytic anemia, vasculitis, type one diabetes, Grave's disease, rheumatoid arthritis, multiple sclerosis, Goodpasture's syndrome, pernicious anemia and/or myopathy), inflammatory diseases (e.g., acne vulgaris, asthma, celiac disease, chronic prostatitis, glomerulonephritis, inflammatory bowel disease, pelvic inflammatory disease, reperfusion injury, rheumatoid arthritis, sarcoidosis, transplant rejection, vasculitis and/or interstitial cystitis), and/or an allergies (e.g., food allergies, drug allergies and/or environmental allergies).


“Immunotherapy” is treatment that uses a subject's immune system to treat disease (e.g., immune disease, inflammatory disease, metabolic disease, cancer) and includes, for example, checkpoint inhibitors, cancer vaccines, cytokines, cell therapy, CAR-T cells, and dendritic cell therapy.


The term “increase” means a change, such that the difference is, depending on circumstances, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 2-fold, 4-fold, 10-fold, 100-fold, 10{circumflex over ( )}3 fold, 10{circumflex over ( )}4 fold, 10{circumflex over ( )}5 fold, 10{circumflex over ( )}6 fold, and/or 10{circumflex over ( )}7 fold greater after treatment when compared to a pre-treatment state. Properties that may be increased include the number of immune cells, bacterial cells, stromal cells, myeloid derived suppressor cells, fibroblasts, metabolites; the level of a cytokine; or another physical parameter (such as ear thickness (e.g., in a DTH animal model) or tumor size (e.g., in an animal tumor model).


“Innate immune agonists” or “immuno-adjuvants” are small molecules, proteins, or other agents that specifically target innate immune receptors including Toll-Like Receptors (TLR), NOD receptors, RLRs, C-type lectin receptors, STING-cGAS Pathway components, inflammasome complexes. For example, LPS is a TLR-4 agonist that is bacterially derived or synthesized and aluminum can be used as an immune stimulating adjuvant. Immuno-adjuvants are a specific class of broader adjuvant or adjuvant therapy. Examples of STING agonists include, but are not limited to, 2′3′-cGAMP, 3′3′-cGAMP, c-di-AMP, c-di-GMP, 2′2′-cGAMP, and 2′3′-cGAM(PS)2 (Rp/Sp) (Rp, Sp-isomers of the bis-phosphorothioate analog of 2′3′-cGAMP). Examples of TLR agonists include, but are not limited to, TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10 and TLR11. Examples of NOD agonists include, but are not limited to, N-acetylmuramyl-L-alanyl-D-isoglutamine (muramyldipeptide (MDP)), gamma-D-glutamyl-meso-diaminopimelic acid (iE-DAP), and desmuramylpeptides (DMP).


The “internal transcribed spacer” or “ITS” is a piece of non-functional RNA located between structural ribosomal RNAs (rRNA) on a common precursor transcript often used for identification of eukaryotic species in particular fungi. The rRNA of fungi that forms the core of the ribosome is transcribed as a signal gene and consists of the 8S, 5.8S and 28S regions with ITS4 and 5 between the 8S and 5.8S and 5.8S and 28S regions, respectively. These two intercistronic segments between the 18S and 5.8S and 5.8S and 28S regions are removed by splicing and contain significant variation between species for barcoding purposes as previously described (Schoch et al Nuclear ribosomal internal transcribed spacer (ITS) region as a universal DNA barcode marker for Fungi. PNAS 109:6241-6246. 2012). 18S rDNA is traditionally used for phylogenetic reconstruction however the ITS can serve this function as it is generally highly conserved but contains hypervariable regions that harbor sufficient nucleotide diversity to differentiate genera and species of most fungus.


The term “isolated” or “enriched” encompasses a microbe, an mEV (such as an smEV) or other entity or substance that has been (1) separated from at least some of the components with which it was associated when initially produced (whether in nature or in an experimental setting), and/or (2) produced, prepared, purified, and/or manufactured by the hand of man. Isolated microbes or mEVs may be separated from at least about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or more of the other components with which they were initially associated. In some embodiments, isolated microbes or mEVs are more than about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% pure, e.g., substantially free of other components. The terms “purify,” “purifying” and “purified” refer to a microbe or mEV or other material that has been separated from at least some of the components with which it was associated either when initially produced or generated (e.g., whether in nature or in an experimental setting), or during any time after its initial production. A microbe or a microbial population or mEV may be considered purified if it is isolated at or after production, such as from a material or environment containing the microbe or microbial population or mEV, and a purified microbe or microbial or mEV population may contain other materials up to about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or above about 90% and still be considered “isolated.” In some embodiments, purified microbes or mEVs or microbial population are more than about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% pure. In the instance of microbial compositions provided herein, the one or more microbial types present in the composition can be independently purified from one or more other microbes produced and/or present in the material or environment containing the microbial type. Microbial compositions and the microbial components such as mEVs thereof are generally purified from residual habitat products.


As used herein a “lipid” includes fats, oils, triglycerides, cholesterol, phospholipids, fatty acids in any form including free fatty acids. Fats, oils and fatty acids can be saturated, unsaturated (cis or trans) or partially unsaturated (cis or trans).


The term “LPS mutant or lipopolysaccharide mutant” broadly refers to selected bacteria that comprises loss of LPS. Loss of LPS might be due to mutations or disruption to genes involved in lipid A biosynthesis, such as lpxA, lpxC, and lpxD. Bacteria comprising LPS mutants can be resistant to aminoglycosides and polymyxins (polymyxin B and colistin).


“Metabolite” as used herein refers to any and all molecular compounds, compositions, molecules, ions, co-factors, catalysts or nutrients used as substrates in any cellular or microbial metabolic reaction or resulting as product compounds, compositions, molecules, ions, co-factors, catalysts or nutrients from any cellular or microbial metabolic reaction.


“Microbe” refers to any natural or engineered organism characterized as a archaeaon, parasite, bacterium, fungus, microscopic alga, protozoan, and the stages of development or life cycle stages (e.g., vegetative, spore (including sporulation, dormancy, and germination), latent, biofilm) associated with the organism. Examples of gut microbes include: Actinomyces graevenitzii, Actinomyces odontolyticus, Akkermansia muciniphila, Bacteroides caccae, Bacteroides fragilis, Bacteroides putredinis, Bacteroides thetaiotaomicron, Bacteroides vultagus, Bifidobacterium adolescentis, Bifidobacterium bifidum, Bilophila wadsworthia, Blautia, Butyrivibrio, Campylobacter gracilis, Clostridia cluster III, Clostridia cluster IV, Clostridia cluster IX (Acidaminococcaceae group), Clostridia cluster XI, Clostridia cluster XIII (Peptostreptococcus group), Clostridia cluster XIV, Clostridia cluster XV, Collinsella aerofaciens, Coprococcus, Corynebacterium sunsvallense, Desulfomonas pigra, Dorea formicigenerans, Dorea longicatena, Escherichia coli, Eubacterium hadrum, Eubacterium rectale, Faecalibacteria prausnitzii, Gemella, Lactococcus, Lanchnospira, Mollicutes cluster XVI, Mollicutes cluster XVIII, Prevotella, Rothia mucilaginosa, Ruminococcus callidus, Ruminococcus gnavus, Ruminococcus torques, and Streptococcus.


“Microbial extracellular vesicles” (mEVs) can be obtained from microbes such as bacteria, archaea, fungi, microscopic algae, protozoans, and parasites. In some embodiments, the mEVs are obtained from bacteria. mEVs include secreted microbial extracellular vesicles (smEVs) and processed microbial extracellular vesicles (pmEVs). “Secreted microbial extracellular vesicles” (smEVs) are naturally-produced vesicles derived from microbes. smEVs are comprised of microbial lipids and/or microbial proteins and/or microbial nucleic acids and/or microbial carbohydrate moieties, and are isolated from culture supernatant. The natural production of these vesicles can be artificially enhanced (e.g., increased) or decreased through manipulation of the environment in which the bacterial cells are being cultured (e.g., by media or temperature alterations). Further, smEV compositions may be modified to reduce, increase, add, or remove microbial components or foreign substances to alter efficacy, immune stimulation, stability, immune stimulatory capacity, stability, organ targeting (e.g., lymph node), absorption (e.g., gastrointestinal), and/or yield (e.g., thereby altering the efficacy). As used herein, the term “purified smEV composition” or “smEV composition” refers to a preparation of smEVs that have been separated from at least one associated substance found in a source material (e.g., separated from at least one other microbial component) or any material associated with the smEVs in any process used to produce the preparation. It can also refer to a composition that has been significantly enriched for specific components. “Processed microbial extracellular vesicles” (pmEVs) are a non-naturally-occurring collection of microbial membrane components that have been purified from artificially lysed microbes (e.g., bacteria) (e.g., microbial membrane components that have been separated from other, intracellular microbial cell components), and which may comprise particles of a varied or a selected size range, depending on the method of purification. A pool of pmEVs is obtained by chemically disrupting (e.g., by lysozyme and/or lysostaphin) and/or physically disrupting (e.g., by mechanical force) microbial cells and separating the microbial membrane components from the intracellular components through centrifugation and/or ultracentrifugation, or other methods. The resulting pmEV mixture contains an enrichment of the microbial membranes and the components thereof (e.g., peripherally associated or integral membrane proteins, lipids, glycans, polysaccharides, carbohydrates, other polymers), such that there is an increased concentration of microbial membrane components, and a decreased concentration (e.g., dilution) of intracellular contents, relative to whole microbes. For gram-positive bacteria, pmEVs may include cell or cytoplasmic membranes. For gram-negative bacteria, a pmEV may include inner and outer membranes. Gram-negative bacteria may belong to the class Negativicutes. pmEVs may be modified to increase purity, to adjust the size of particles in the composition, and/or modified to reduce, increase, add or remove, microbial components or foreign substances to alter efficacy, immune stimulation, stability, immune stimulatory capacity, stability, organ targeting (e.g., lymph node), absorption (e.g., gastrointestinal), and/or yield (e.g., thereby altering the efficacy). pmEVs can be modified by adding, removing, enriching for, or diluting specific components, including intracellular components from the same or other microbes. As used herein, the term “purified pmEV composition” or “pmEV composition” refers to a preparation of pmEVs that have been separated from at least one associated substance found in a source material (e.g., separated from at least one other microbial component) or any material associated with the pmEVs in any process used to produce the preparation. It can also refer to a composition that has been significantly enriched for specific components.


“Microbiome” broadly refers to the microbes residing on or in body site of a subject or patient. Microbes in a microbiome may include bacteria, viruses, eukaryotic microorganisms, and/or viruses. Individual microbes in a microbiome may be metabolically active, dormant, latent, or exist as spores, may exist planktonically or in biofilms, or may be present in the microbiome in sustainable or transient manner. The microbiome may be a commensal or healthy-state microbiome or a disease-state microbiome. The microbiome may be native to the subject or patient, or components of the microbiome may be modulated, introduced, or depleted due to changes in health state (e.g., precancerous or cancerous state) or treatment conditions (e.g., antibiotic treatment, exposure to different microbes). In some aspects, the microbiome occurs at a mucosal surface. In some aspects, the microbiome is a gut microbiome. In some aspects, the microbiome is a tumor microbiome.


A “microbiome profile” or a “microbiome signature” of a tissue or sample refers to an at least partial characterization of the bacterial makeup of a microbiome. In some embodiments, a microbiome profile indicates whether at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more bacterial strains are present or absent in a microbiome. In some embodiments, a microbiome profile indicates whether at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more cancer-associated bacterial strains are present in a sample. In some embodiments, the microbiome profile indicates the relative or absolute amount of each bacterial strain detected in the sample. In some embodiments, the microbiome profile is a cancer-associated microbiome profile. A cancer-associated microbiome profile is a microbiome profile that occurs with greater frequency in a subject who has cancer than in the general population. In some embodiments, the cancer-associated microbiome profile comprises a greater number of or amount of cancer-associated bacteria than is normally present in a microbiome of an otherwise equivalent tissue or sample taken from an individual who does not have cancer.


“Modified” in reference to a bacteria broadly refers to a bacteria that has undergone a change from its wild-type form. Bacterial modification can result from engineering bacteria. Examples of bacterial modifications include genetic modification, gene expression modification, phenotype modification, formulation modification, chemical modification, and dose or concentration. Examples of improved properties are described throughout this specification and include, e.g., attenuation, auxotrophy, homing, or antigenicity. Phenotype modification might include, by way of example, bacteria growth in media that modify the phenotype of a bacterium such that it increases or decreases virulence.


An “oncobiome” as used herein comprises tumorigenic and/or cancer-associated microbiota, wherein the microbiota comprises one or more of a virus, a bacterium, a fungus, a protist, a parasite, or another microbe.


“Oncotrophic” or “oncophilic” microbes and bacteria are microbes that are highly associated or present in a cancer microenvironment. They may be preferentially selected for within the environment, preferentially grow in a cancer microenvironment or hone to a said environment.


“Operational taxonomic units” and “OTU(s)” refer to a terminal leaf in a phylogenetic tree and is defined by a nucleic acid sequence, e.g., the entire genome, or a specific genetic sequence, and all sequences that share sequence identity to this nucleic acid sequence at the level of species. In some embodiments the specific genetic sequence may be the 16S sequence or a portion of the 16S sequence. In other embodiments, the entire genomes of two entities are sequenced and compared. In another embodiment, select regions such as multilocus sequence tags (MLST), specific genes, or sets of genes may be genetically compared. For 16S, OTUs that share ≥97% average nucleotide identity across the entire 16S or some variable region of the 16S are considered the same OTU. See e.g., Claesson M J, Wang Q, O'Sullivan O, Greene-Diniz R, Cole J R, Ross R P, and O'Toole P W. 2010. Comparison of two next-generation sequencing technologies for resolving highly complex microbiota composition using tandem variable 16S rRNA gene regions. Nucleic Acids Res 38: e200. Konstantinidis K T, Ramette A, and Tiedje J M. 2006. The bacterial species definition in the genomic era. Philos Trans R Soc Lond B Biol Sci 361: 1929-1940. For complete genomes, MLSTs, specific genes, other than 16S, or sets of genes OTUs that share ≥95% average nucleotide identity are considered the same OTU. See e.g., Achtman M, and Wagner M. 2008. Microbial diversity and the genetic nature of microbial species. Nat. Rev. Microbiol. 6: 431-440. Konstantinidis K T, Ramette A, and Tiedje J M. 2006. The bacterial species definition in the genomic era. Philos Trans R Soc Lond B Biol Sci 361: 1929-1940. OTUs are frequently defined by comparing sequences between organisms. Generally, sequences with less than 95% sequence identity are not considered to form part of the same OTU. OTUs may also be characterized by any combination of nucleotide markers or genes, in particular highly conserved genes (e.g., “house-keeping” genes), or a combination thereof. Operational Taxonomic Units (OTUs) with taxonomic assignments made to, e.g., genus, species, and phylogenetic clade are provided herein.


As used herein, a gene is “overexpressed” in a bacteria if it is expressed at a higher level in an engineered bacteria under at least some conditions than it is expressed by a wild-type bacteria of the same species under the same conditions. Similarly, a gene is “underexpressed” in a bacteria if it is expressed at a lower level in an engineered bacteria under at least some conditions than it is expressed by a wild-type bacteria of the same species under the same conditions.


The terms “polynucleotide”, and “nucleic acid” are used interchangeably. They refer to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides, or analogs thereof. Polynucleotides may have any three-dimensional structure, and may perform any function. The following are non-limiting examples of polynucleotides: coding or non-coding regions of a gene or gene fragment, loci (locus) defined from linkage analysis, exons, introns, messenger RNA (mRNA), micro RNA (miRNA), silencing RNA (siRNA), transfer RNA, ribosomal RNA, ribozymes, cDNA, recombinant polynucleotides, branched polynucleotides, plasmids, vectors, isolated DNA of any sequence, isolated RNA of any sequence, nucleic acid probes, and primers. A polynucleotide may comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs. If present, modifications to the nucleotide structure may be imparted before or after assembly of the polymer. A polynucleotide may be further modified, such as by conjugation with a labeling component. In all nucleic acid sequences provided herein, U nucleotides are interchangeable with T nucleotides.


As used herein, a substance is “pure” if it is substantially free of other components. The terms “purify,” “purifying” and “purified” refer to an mEV (such as an smEV) preparation or other material that has been separated from at least some of the components with which it was associated either when initially produced or generated (e.g., whether in nature or in an experimental setting), or during any time after its initial production. An mEV (such as an smEV) preparation or compositions may be considered purified if it is isolated at or after production, such as from one or more other bacterial components, and a purified microbe or microbial population may contain other materials up to about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or above about 90% and still be considered “purified.” In some embodiments, purified mEVs (such as smEVs) are more than about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% pure. mEV (such as an smEV) compositions (or preparations) are, e.g., purified from residual habitat products.


As used herein, the term “purified mEV composition” or “mEV composition” refers to a preparation that includes mEVs (such as smEVs) that have been separated from at least one associated substance found in a source material (e.g., separated from at least one other bacterial component) or any material associated with the mEVs (such as smEVs) in any process used to produce the preparation. It also refers to a composition that has been significantly enriched or concentrated. In some embodiments, the mEVs (such as smEVs) are concentrated by 2 fold, 3-fold, 4-fold, 5-fold, 10-fold, 100-fold, 1000-fold, 10,000-fold or more than 10,000 fold.


“Residual habitat products” refers to material derived from the habitat for microbiota within or on a subject. For example, fermentation cultures of microbes can contain contaminants, e.g., other microbe strains or forms (e.g., bacteria, virus, mycoplasm, and/or fungus). For example, microbes live in feces in the gastrointestinal tract, on the skin itself, in saliva, mucus of the respiratory tract, or secretions of the genitourinary tract (i.e., biological matter associated with the microbial community). Substantially free of residual habitat products means that the microbial composition no longer contains the biological matter associated with the microbial environment on or in the culture or human or animal subject and is 100% free, 99% free, 98% free, 97% free, 96% free, or 95% free of any contaminating biological matter associated with the microbial community. Residual habitat products can include abiotic materials (including undigested food) or it can include unwanted microorganisms. Substantially free of residual habitat products may also mean that the microbial composition contains no detectable cells from a culture contaminant or a human or animal and that only microbial cells are detectable. In one embodiment, substantially free of residual habitat products may also mean that the microbial composition contains no detectable viral (including bacteria, viruses (e.g., phage)), fungal, mycoplasmal contaminants. In another embodiment, it means that fewer than 1×10−2%, 1×10−3%, 1×10−4%, 1×10−5%, 1×10−6%, 1×10−7%, 1×10−8% of the viable cells in the microbial composition are human or animal, as compared to microbial cells. There are multiple ways to accomplish this degree of purity, none of which are limiting. Thus, contamination may be reduced by isolating desired constituents through multiple steps of streaking to single colonies on solid media until replicate (such as, but not limited to, two) streaks from serial single colonies have shown only a single colony morphology. Alternatively, reduction of contamination can be accomplished by multiple rounds of serial dilutions to single desired cells (e.g., a dilution of 10−8 or 10−9), such as through multiple 10-fold serial dilutions. This can further be confirmed by showing that multiple isolated colonies have similar cell shapes and Gram staining behavior. Other methods for confirming adequate purity include genetic analysis (e.g., PCR, DNA sequencing), serology and antigen analysis, enzymatic and metabolic analysis, and methods using instrumentation such as flow cytometry with reagents that distinguish desired constituents from contaminants.


As used herein, “specific binding” refers to the ability of an antibody to bind to a predetermined antigen or the ability of a polypeptide to bind to its predetermined binding partner. Typically, an antibody or polypeptide specifically binds to its predetermined antigen or binding partner with an affinity corresponding to a KD of about 10−7 M or less, and binds to the predetermined antigen/binding partner with an affinity (as expressed by KD) that is at least 10 fold less, at least 100 fold less or at least 1000 fold less than its affinity for binding to a non-specific and unrelated antigen/binding partner (e.g., BSA, casein). Alternatively, specific binding applies more broadly to a two component system where one component is a protein, lipid, or carbohydrate or combination thereof and engages with the second component which is a protein, lipid, carbohydrate or combination thereof in a specific way.


“Strain” refers to a member of a bacterial species with a genetic signature such that it may be differentiated from closely-related members of the same bacterial species. The genetic signature may be the absence of all or part of at least one gene, the absence of all or part of at least on regulatory region (e.g., a promoter, a terminator, a riboswitch, a ribosome binding site), the absence (“curing”) of at least one native plasmid, the presence of at least one recombinant gene, the presence of at least one mutated gene, the presence of at least one foreign gene (a gene derived from another species), the presence at least one mutated regulatory region (e.g., a promoter, a terminator, a riboswitch, a ribosome binding site), the presence of at least one non-native plasmid, the presence of at least one antibiotic resistance cassette, or a combination thereof. Genetic signatures between different strains may be identified by PCR amplification optionally followed by DNA sequencing of the genomic region(s) of interest or of the whole genome. In the case in which one strain (compared with another of the same species) has gained or lost antibiotic resistance or gained or lost a biosynthetic capability (such as an auxotrophic strain), strains may be differentiated by selection or counter-selection using an antibiotic or nutrient/metabolite, respectively.


The terms “subject” or “patient” refers to any mammal. A subject or a patient described as “in need thereof” refers to one in need of a treatment (or prevention) for a disease. Mammals (i.e., mammalian animals) include humans, laboratory animals (e.g., primates, rats, mice), livestock (e.g., cows, sheep, goats, pigs), and household pets (e.g., dogs, cats, rodents). The subject may be a human. The subject may be a non-human mammal including but not limited to of a dog, a cat, a cow, a horse, a pig, a donkey, a goat, a camel, a mouse, a rat, a guinea pig, a sheep, a llama, a monkey, a gorilla or a chimpanzee. The subject may be healthy, or may be suffering from a cancer at any developmental stage, wherein any of the stages are either caused by or opportunistically supported of a cancer associated or causative pathogen, or may be at risk of developing a cancer, or transmitting to others a cancer associated or cancer causative pathogen. In some embodiments, a subject has lung cancer, bladder cancer, prostate cancer, plasmacytoma, colorectal cancer, rectal cancer, Merkel Cell carcinoma, salivary gland carcinoma, ovarian cancer, and/or melanoma. The subject may have a tumor. The subject may have a tumor that shows enhanced macropinocytosis with the underlying genomics of this process including Ras activation. In other embodiments, the subject has another cancer. In some embodiments, the subject has undergone a cancer therapy.


As used herein, the term “treating” a disease in a subject or “treating” a subject having or suspected of having a disease refers to administering to the subject to a pharmaceutical treatment, e.g., the administration of one or more agents, such that at least one symptom of the disease is decreased or prevented from worsening. Thus, in one embodiment, “treating” refers inter alia to delaying progression, expediting remission, inducing remission, augmenting remission, speeding recovery, increasing efficacy of or decreasing resistance to alternative therapeutics, or a combination thereof. As used herein, the term “preventing” a disease in a subject refers to administering to the subject to a pharmaceutical treatment, e.g., the administration of one or more agents, such that onset of at least one symptom of the disease is delayed or prevented.


Bacteria

In certain aspects, provided herein are pharmaceutical compositions that comprise mEVs (such as smEVs) obtained from bacteria.


In some embodiments, the bacteria from which the mEVs (such as smEVs) are obtained are modified to reduce toxicity or other adverse effects, to enhance delivery) (e.g., oral delivery) of the mEVs (such as smEVs) (e.g., by improving acid resistance, muco-adherence and/or penetration and/or resistance to bile acids, digestive enzymes, resistance to anti-microbial peptides and/or antibody neutralization), to target desired cell types (e.g., M-cells, goblet cells, enterocytes, dendritic cells, macrophages), to enhance their immunomodulatory and/or therapeutic effect of the mEVs (such as smEVs) (e.g., either alone or in combination with another therapeutic agent), and/or to enhance immune activation or suppression by the mEVs (such as smEVs) (e.g., through modified production of polysaccharides, pili, fimbriae, adhesins). In some embodiments, the engineered bacteria described herein are modified to improve mEV (such as smEV) manufacturing (e.g., higher oxygen tolerance, stability, improved freeze-thaw tolerance, shorter generation times). For example, in some embodiments, the engineered bacteria described include bacteria harboring one or more genetic changes, such change being an insertion, deletion, translocation, or substitution, or any combination thereof, of one or more nucleotides contained on the bacterial chromosome or endogenous plasmid and/or one or more foreign plasmids, wherein the genetic change may results in the overexpression and/or underexpression of one or more genes. The engineered bacteria may be produced using any technique known in the art, including but not limited to site-directed mutagenesis, transposon mutagenesis, knock-outs, knock-ins, polymerase chain reaction mutagenesis, chemical mutagenesis, ultraviolet light mutagenesis, transformation (chemically or by electroporation), phage transduction, directed evolution, or any combination thereof.


Examples of species and/or strains of bacteria that can be used as a source of mEVs (such as smEVs) described herein are provided in Table 1, Table 2, and/or Table 3 and elsewhere throughout the specification. In some embodiments, the bacterial strain is a bacterial strain having a genome that has at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8% or 99.9% sequence identity to a strain listed in Table 1, Table 2, and/or Table 3. In some embodiments, the mEVs are from an oncotrophic bacteria. In some embodiments, the mEVs are from an immunostimulatory bacteria. In some embodiments, the mEVs are from an immunosuppressive bacteria. In some embodiments, the mEVs are from an immunomodulatory bacteria. In certain embodiments, mEVs are generated from a combination of bacterial strains provided herein. In some embodiments, the combination is a combination of at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45 or 50 bacterial strains. In some embodiments, the combination includes mEVs from bacterial strains listed in Table 1, Table 2, and/or Table 3 and/or bacterial strains having a genome that has at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8% or 99.9% sequence identity to a strain listed in Table 1, Table 2, and/or Table 3.


In some embodiments, the mEVs are obtained from Gram negative bacteria.


In some embodiments, the Gram negative bacteria belong to the class Negativicutes. The Negativicutes represent a unique class of microorganisms as they are the only diderm members of the Firmicutes phylum. These anaerobic organisms can be found in the environment and are normal commensals of the oral cavity and GI tract of humans. Because these organisms have an outer membrane, the yields of smEVs from this class were investigated. It was found that on a per cell basis these microbes produce a high number of vesicles (10-150 EVs/cell). The smEVs from these organisms are broadly stimulatory and highly potent in in vitro assays. Investigations into their therapeutic applications in several oncology and inflammation in vivo models have shown their therapeutic potential. The class Negativicutes includes the families Veillonellaceae, Selenomonadaceae, Acidaminococcaceae, and Sporomusaceae. The class Negativicutes includes the genera Megasphaera, Selenomonas, Propionospora, and Acidaminococcus. Exemplary Negativicutes species include, but are not limited to, Megasphaera sp., Selenomonas felix, Acidaminococcus intestine, and Propionospora sp.


In some embodiments, the mEVs are obtained from Gram positive bacteria.


In some embodiments, the mEVs are obtained from aerobic bacteria.


In some embodiments, the mEVs are obtained from anaerobic bacteria.


In some embodiments, the mEVs are obtained from acidophile bacteria.


In some embodiments, the mEVs are obtained from alkaliphile bacteria.


In some embodiments, the mEVs are obtained from neutralophile bacteria.


In some embodiments, the mEVs are obtained from fastidious bacteria.


In some embodiments, the mEVs are obtained from nonfastidious bacteria.


In some embodiments, bacteria from which mEVs are obtained are lyophilized.


In some embodiments, bacteria from which mEVs are obtained are gamma irradiated (e.g., at 17.5 or 25 kGy).


In some embodiments, bacteria from which mEVs are obtained are UV irradiated.


In some embodiments, bacteria from which mEVs are obtained are heat inactivated (e.g., at 50° C. for two hours or at 90° C. for two hours).


In some embodiments, bacteria from which mEVs are obtained are acid treated.


In some embodiments, bacteria from which mEVs are obtained are oxygen sparged (e.g., at 0.1 vvm for two hours).


In some embodiments, the mEVs are lyophilized.


In some embodiments, the mEVs are gamma irradiated (e.g., at 17.5 or 25 kGy).


In some embodiments, the mEVs are UV irradiated.


In some embodiments, the mEVs are heat inactivated (e.g., at 50° C. for two hours or at 90° C. for two hours).


In some embodiments, the mEVs are acid treated.


In some embodiments, the mEVs are oxygen sparged (e.g., at 0.1 vvm for two hours).


The phase of growth can affect the amount or properties of bacteria and/or smEVs produced by bacteria. For example, in the methods of smEVs preparation provided herein, smEVs can be isolated, e.g., from a culture, at the start of the log phase of growth, midway through the log phase, and/or once stationary phase growth has been reached.









TABLE 1







Exemplary Bacterial Strains









Public DB


OTU
Accession






Abiotrophia defectiva

ACIN02000016



Abiotrophia para_adiacens

AB022027



Abiotrophia sp. oral clone P4PA_155 P1

AY207063



Acetanaerobacterium elongatum

NR_042930



Acetivibrio cellulolyticus

NR_025917



Acetivibrio ethanolgignens

FR749897



Acetobacter aceti

NR_026121



Acetobacter fabarum

NR_042678



Acetobacter lovaniensis

NR_040832



Acetobacter malorum

NR_025513



Acetobacter orientalis

NR_028625



Acetobacter pasteurianus

NR_026107



Acetobacter pomorum

NR_042112



Acetobacter syzygii

NR_040868



Acetobacter tropicalis

NR_036881


Acetobacteraceae bacterium AT_5844
AGEZ01000040



Acholeplasma laidlawii

NR_074448



Achromobacter denitrificans

NR_042021



Achromobacter piechaudii

ADMS01000149



Achromobacter xylosoxidans

ACRC01000072



Acidaminococcus fermentans

CP001859



Acidaminococcus intestini

CP003058



Acidaminococcus sp. D21

ACGB01000071



Acidilobus saccharovorans

AY350586



Acidithiobacillus ferrivorans

NR_074660



Acidovorax sp. 98_63833

AY258065



Acinetobacter baumannii

ACYQ01000014



Acinetobacter calcoaceticus

AM157426



Acinetobacter genomosp. C1

AY278636



Acinetobacter haemolyticus

ADMT01000017



Acinetobacter johnsonii

ACPL01000162



Acinetobacter junii

ACPM01000135



Acinetobacter lwoffii

ACPN01000204



Acinetobacter parvus

AIEB01000124



Acinetobacter radioresistens

ACVR01000010



Acinetobacter schindleri

NR_025412



Acinetobacter sp. 56A1

GQ178049



Acinetobacter sp. CIP 101934

JQ638573



Acinetobacter sp. CIP 102143

JQ638578



Acinetobacter sp. CIP 53.82

JQ638584



Acinetobacter sp. M16_22

HM366447



Acinetobacter sp. RUH2624

ACQF01000094



Acinetobacter sp. SH024

ADCH01000068



Actinobacillus actinomycetemcomitans

AY362885



Actinobacillus minor

ACFT01000025



Actinobacillus pleuropneumoniae

NR_074857



Actinobacillus succinogenes

CP000746



Actinobacillus ureae

AEVG01000167



Actinobaculum massiliae

AF487679



Actinobaculum schaalii

AY957507



Actinobaculum sp. BM#101342

AY282578



Actinobaculum sp. P2P_19 P1

AY207066



Actinomyces cardiffensis

GU470888



Actinomyces europaeus

NR_026363



Actinomyces funkei

HQ906497



Actinomyces genomosp. C1

AY278610



Actinomyces genomosp. C2

AY278611



Actinomyces genomosp. P1 oral clone MB6_C03

DQ003632



Actinomyces georgiae

GU561319



Actinomyces israelii

AF479270



Actinomyces massiliensis

AB545934



Actinomyces meyeri

GU561321



Actinomyces naeslundii

X81062



Actinomyces nasicola

AJ508455



Actinomyces neuii

X71862



Actinomyces odontolyticus

ACYT01000123



Actinomyces oricola

NR_025559



Actinomyces orihominis

AJ575186



Actinomyces oris

BABV01000070



Actinomyces sp. 7400942

EU484334



Actinomyces sp. c109

AB16723 9



Actinomyces sp. CCUG 37290

AJ234058



Actinomyces sp. ChDC Bl97

AF543275



Actinomyces sp. GEJ15

GU561313



Actinomyces sp. HKU31

HQ335393



Actinomyces sp. ICM34

HQ616391



Actinomyces sp. ICM41

HQ616392



Actinomyces sp. ICM47

HQ616395



Actinomyces sp. ICM54

HQ616398



Actinomyces sp. M2231_94_1

AJ234063



Actinomyces sp. oral clone GU009

AY349361



Actinomyces sp. oral clone GU067

AY349362



Actinomyces sp. oral clone IO076

AY349363



Actinomyces sp. oral clone IO077

AY349364



Actinomyces sp. oral clone IP073

AY349365



Actinomyces sp. oral clone IP081

AY349366



Actinomyces sp. oral clone JA063

AY349367



Actinomyces sp. oral taxon 170

AFBL01000010



Actinomyces sp. oral taxon 171

AECW01000034



Actinomyces sp. oral taxon 178

AEUH01000060



Actinomyces sp. oral taxon 180

AEPP01000041



Actinomyces sp. oral taxon 848

ACUY01000072



Actinomyces sp. oral taxon C55

HM099646



Actinomyces sp. TeJ5

GU561315



Actinomyces urogenitalis

ACFH01000038



Actinomyces viscosus

ACRE01000096



Adlercreutzia equolifaciens

AB306661



Aerococcus sanguinicola

AY837833



Aerococcus urinae

CP002512



Aerococcus urinaeequi

NR_043443



Aerococcus viridans

ADNT01000041



Aeromicrobium marinum

NR_025681



Aeromicrobium sp. JC14

JF824798



Aeromonas allosaccharophila

S39232



Aeromonas enteropelogenes

X71121



Aeromonas hydrophila

NC_008570



Aeromonas jandaei

X60413



Aeromonas salmonicida

NC_009348



Aeromonas trota

X60415



Aeromonas veronii

NR_044845



Afipia genomosp. 4

EU117385



Aggregatibacter actinomycetemcomitans

CP001733



Aggregatibacter aphrophilus

CP001607



Aggregatibacter segnis

AEPS01000017



Agrobacterium radiobacter

CP000628



Agrobacterium tumefaciens

AJ3 89893



Agrococcus jenensis

NR_026275



Akkermansia muciniphila

CP001071



Alcaligenes faecalis

AB680368



Alcaligenes sp. CO14

DQ643040



Alcaligenes sp. S3

HQ262549



Alicyclobacillus acidocaldarius

NR_074721



Alicyclobacillus acidoterrestris

NR_040844



Alicyclobacillus contaminans

NR_041475



Alicyclobacillus cycloheptanicus

NR_024754



Alicyclobacillus herbarius

NR_024753



Alicyclobacillus pomorum

NR_024801



Alicyclobacillus sp. CCUG 53762

HE613268



Alistipes finegoldii

NR_043064



Alistipes indistinctus

AB490804



Alistipes onderdonkii

NR_043318



Alistipes putredinis

ABFK02000017



Alistipes shahii

FP929032



Alistipes sp. HGB5

AENZ01000082



Alistipes sp. JC50

JF824804



Alistipes sp. RMA 9912

GQ140629



Alkaliphilus metalliredigenes

AY137848



Alkaliphilus oremlandii

NR_043674



Alloscardovia omnicolens

NR_042583



Alloscardovia sp. OB7196

AB425070



Anaerobaculum hydrogeniformans

ACJX02000009



Anaerobiospirillum succiniciproducens

NR_026075



Anaerobiospirillum thomasii

AJ420985



Anaerococcus hydrogenalis

ABXA01000039



Anaerococcus lactolyticus

ABYO01000217



Anaerococcus octavius

NR_026360



Anaerococcus prevotii

CP001708



Anaerococcus sp. 8404299

HM587318



Anaerococcus sp. 8405254

HM587319



Anaerococcus sp. 9401487

HM587322



Anaerococcus sp. 9403502

HM587325



Anaerococcus sp. gpac104

AM176528



Anaerococcus sp. gpac126

AM176530



Anaerococcus sp. gpac155

AM176536



Anaerococcus sp. gpac199

AM176539



Anaerococcus sp. gpac215

AM176540



Anaerococcus tetradius

ACGC01000107



Anaerococcus vaginalis

ACXU01000016



Anaerofustis stercorihominis

ABIL02000005



Anaeroglobus geminatus

AGCJ01000054



Anaerosporobacter mobilis

NR_042953



Anaerostipes caccae

ABAX03000023



Anaerostipes sp. 3_2_56FAA

ACWB01000002



Anaerotruncus colihominis

ABGD02000021



Anaplasma marginale

ABOR01000019



Anaplasma phagocytophilum

NC_007797



Aneurinibacillus aneurinilyticus

AB101592



Aneurinibacillus danicus

NR_028657



Aneurinibacillus migulanus

NR_036799



Aneurinibacillus terranovensis

NR_042271



Aneurinibacillus thermoaerophilus

NR_029303



Anoxybacillus contaminans

NR_029006



Anoxybacillus flavithermus

NR_074667



Arcanobacterium haemolyticum

NR_025347



Arcanobacterium pyogenes

GU585578



Arcobacter butzleri

AEPT01000071



Arcobacter cryaerophilus

NR_025905



Arthrobacter agilis

NR_026198



Arthrobacter arilaitensis

NR_074608



Arthrobacter bergerei

NR_025612



Arthrobacter globiformis

NR_026187



Arthrobacter nicotianae

NR_026190



Atopobium minutum

HM007583



Atopobium parvulum

CP001721



Atopobium rimae

ACFE01000007



Atopobium sp. BS2

HQ616367



Atopobium sp. F0209

EU592966



Atopobium sp. ICM42b10

HQ616393



Atopobium sp. ICM57

HQ616400



Atopobium vaginae

AEDQ01000024



Aurantimonas coralicida

AY065627



Aureimonas altamirensis

FN658986



Auritibacter ignavus

FN554542



Averyella dalhousiensis

DQ481464



Bacillus aeolius

NR_025557



Bacillus aerophilus

NR_042339



Bacillus aestuarii

GQ980243



Bacillus alcalophilus

X76436



Bacillus amyloliquefaciens

NR_075005



Bacillus anthracis

AAEN01000020



Bacillus atrophaeus

NR_075016



Bacillus badius

NR_036893



Bacillus cereus

ABDJ01000015



Bacillus circulans

AB271747



Bacillus clausii

FN397477



Bacillus coagulans

DQ297928



Bacillus firmus

NR_025842



Bacillus flexus

NR_024691



Bacillus fordii

NR_025786



Bacillus gelatini

NR_025595



Bacillus halmapalus

NR_026144



Bacillus halodurans

AY144582



Bacillus herbersteinensis

NR_042286



Bacillus horti

NR_036860



Bacillus idriensis

NR_043268



Bacillus lentus

NR_040792



Bacillus licheniformis

NC_006270



Bacillus megaterium

GU252124



Bacillus nealsonii

NR_044546



Bacillus niabensis

NR_043334



Bacillus niacini

NR_024695



Bacillus pocheonensis

NR_041377



Bacillus pumilus

NR_074977



Bacillus safensis

JQ624766



Bacillus simplex

NR_042136



Bacillus sonorensis

NR_025130



Bacillus sp. 10403023 MM10403188

CAET01000089



Bacillus sp. 2_A_57_CT2

ACWD01000095



Bacillus sp. 2008724126

GU252108



Bacillus sp. 2008724139

GU252111



Bacillus sp. 7_16AIA

FN397518



Bacillus sp. 9_3AIA

FN397519



Bacillus sp. AP8

JX101689



Bacillus sp. B27(2008)

EU362173



Bacillus sp. BT1B_CT2

ACWC01000034



Bacillus sp. GB1.1

FJ897765



Bacillus sp. GB9

FJ897766



Bacillus sp. HU19.1

FJ897769



Bacillus sp. HU29

FJ897771



Bacillus sp. HU33.1

FJ897772



Bacillus sp. JC6

JF824800



Bacillus sp. oral taxon F26

HM099642



Bacillus sp. oral taxon F28

HM099650



Bacillus sp. oral taxon F79

HM099654



Bacillus sp. SRC_DSF1

GU797283



Bacillus sp. SRC_DSF10

GU797292



Bacillus sp. SRC_DSF2

GU797284



Bacillus sp. SRC_DSF6

GU797288



Bacillus sp. tc09

HQ844242



Bacillus sp. zh168

FJ851424



Bacillus sphaericus

DQ286318



Bacillus sporothermodurans

NR_026010



Bacillus subtilis

EU627588



Bacillus thermoamylovorans

NR_029151



Bacillus thuringiensis

NC_008600



Bacillus weihenstephanensis

NR_074926


Bacteroidales bacterium ph8
JN837494


Bacteroidales genomosp. P1
AY341819


Bacteroidales genomosp. P2 oral clone MB1_G13
DQ003613


Bacteroidales genomosp. P3 oral clone MB1_G34
DQ003615


Bacteroidales genomosp. P4 oral clone MB2_G17
DQ003617


Bacteroidales genomosp. P5 oral clone MB2_P04
DQ003619


Bacteroidales genomosp. P6 oral clone MB3_C19
DQ003634


Bacteroidales genomosp. P7 oral clone MB3_P19
DQ003623


Bacteroidales genomosp. P8 oral clone MB4_G15
DQ003626



Bacteroides acidifaciens

NR_028607



Bacteroides barnesiae

NR_041446



Bacteroides caccae

EU136686



Bacteroides cellulosilyticus

ACCH01000108



Bacteroides clarus

AFBM01000011



Bacteroides coagulans

AB547639



Bacteroides coprocola

ABIY02000050



Bacteroides coprophilus

ACBW01000012



Bacteroides dorei

ABWZ01000093



Bacteroides eggerthii

ACWG01000065



Bacteroides faecis

GQ496624



Bacteroides finegoldii

AB222699



Bacteroides fluxus

AFBN01000029



Bacteroides fragilis

AP006841



Bacteroides galacturonicus

DQ497994



Bacteroides helcogenes

CP002352



Bacteroides heparinolyticus

JN867284



Bacteroides intestinalis

ABJL02000006



Bacteroides massiliensis

AB200226



Bacteroides nordii

NR_043017



Bacteroides oleiciplenus

AB547644



Bacteroides ovatus

ACWH01000036



Bacteroides pectinophilus

ABVQ01000036



Bacteroides plebeius

AB200218



Bacteroides pyogenes

NR_041280



Bacteroides salanitronis

CP002530



Bacteroides salyersiae

EU136690



Bacteroides sp. 1_1_14

ACRP01000155



Bacteroides sp. 1_1_30

ADCL01000128



Bacteroides sp. 1_1_6

ACIC01000215



Bacteroides sp. 2_1_22

ACPQ01000117



Bacteroides sp. 2_1_56FAA

ACWI01000065



Bacteroides sp. 2_2_4

ABZZ01000168



Bacteroides sp. 20_3

ACRQ01000064



Bacteroides sp. 3_1_19

ADCJ01000062



Bacteroides sp. 3_1_23

ACRS01000081



Bacteroides sp. 3_1_33FAA

ACPS01000085



Bacteroides sp. 3_1_40A

ACRT01000136



Bacteroides sp. 3_2_5

ACIB01000079



Bacteroides sp. 315_5

FJ848547



Bacteroides sp. 31SF15

AJ583248



Bacteroides sp. 31SF18

AJ583249



Bacteroides sp. 35AE31

AJ583244



Bacteroides sp. 35AE37

AJ583245



Bacteroides sp. 35BE34

AJ583246



Bacteroides sp. 35BE35

AJ583247



Bacteroides sp. 4_1_36

ACTC01000133



Bacteroides sp. 4_3_47FAA

ACDR02000029



Bacteroides sp. 9_1_42FAA

ACAA01000096



Bacteroides sp. AR20

AF139524



Bacteroides sp. AR29

AF139525



Bacteroides sp. B2

EU722733



Bacteroides sp. D1

ACAB02000030



Bacteroides sp. D2

ACGA01000077



Bacteroides sp. D20

ACPT01000052



Bacteroides sp. D22

ADCK01000151



Bacteroides sp. F_4

AB470322



Bacteroides sp. NB_8

AB117565



Bacteroides sp. WH2

AY895180



Bacteroides sp. XB12B

AM230648



Bacteroides sp. XB44A

AM230649



Bacteroides stercoris

ABFZ02000022



Bacteroides thetaiotaomicron

NR_074277



Bacteroides uniforms

AB050110



Bacteroides ureolyticus

GQ167666



Bacteroides vulgatus

CP000139



Bacteroides xylanisolvens

ADKP01000087


Bacteroidetes bacterium oral taxon D27
HM099638


Bacteroidetes bacterium oral taxon F31
HM099643


Bacteroidetes bacterium oral taxon F44
HM099649



Bamesiella intestinihominis

AB370251



Bamesiella viscericola

NR_041508



Bartonella bacilliformis

NC_008783



Bartonella grahamii

CP001562



Bartonella henselae

NC_005956



Bartonella quintana

BX897700



Bartonella tamiae

EF672728



Bartonella washoensis

FJ719017



Bdellovibrio sp. MPA

AY294215


Bifidobacteriaceae genomosp. C1
AY278612



Bifidobacterium adolescentis

AAXD02000018



Bifidobacterium angulatum

ABYS02000004



Bifidobacterium animalis

CP001606



Bifidobacterium bifidum

ABQP01000027



Bifidobacterium breve

CP002743



Bifidobacterium catenulatum

ABXY01000019



Bifidobacterium dentium

CP001750



Bifidobacterium gallicum

ABXB03000004



Bifidobacterium infantis

AY151398



Bifidobacterium kashiwanohense

AB491757



Bifidobacterium longum

ABQQ01000041



Bifidobacterium pseudocatenulatum

ABXX02000002



Bifidobacterium pseudolongum

NR_043442



Bifidobacterium scardovii

AJ307005



Bifidobacterium sp. HM2

AB425276



Bifidobacterium sp. HMLN12

JF519685



Bifidobacterium sp. M45

HM626176



Bifidobacterium sp. MSX5B

HQ616382



Bifidobacterium sp. TM_7

AB218972



Bifidobacterium thermophilum

DQ340557



Bifidobacterium urinalis

AJ278695



Bilophila wadsworthia

ADCP01000166


Bisgaard Taxon
AY683487


Bisgaard Taxon
AY683489


Bisgaard Taxon
AY683491


Bisgaard Taxon
AY683492



Blastomonas natatoria

NR_040824



Blautia coccoides

AB571656



Blautia glucerasea

AB588023



Blautia glucerasei

AB439724



Blautia hansenii

ABYU02000037



Blautia hydrogenotrophica

ACBZ01000217



Blautia luti

AB691576



Blautia producta

AB600998



Blautia schinkii

NR_026312



Blautia sp. M25

HM626178



Blautia stercoris

HM626177



Blautia wexlerae

EF036467



Bordetella bronchiseptica

NR_025949



Bordetella holmesii

AB683187



Bordetella parapertussis

NR_025950



Bordetella pertussis

BX640418



Borrelia afzelii

ABCU01000001



Borrelia burgdorferi

ABGI01000001



Borrelia crocidurae

DQ057990



Borrelia duttonii

NC_011229



Borrelia garinii

ABJV01000001



Borrelia hermsii

AY597657



Borrelia hispanica

DQ057988



Borrelia persica

HM161645



Borrelia recurrentis

AF107367



Borrelia sp. NE49

AJ224142



Borrelia spielmanii

ABKB01000002



Borrelia turicatae

NC_008710



Borrelia valaisiana

ABCY01000002



Brachybacterium alimentarium

NR_026269



Brachybacterium conglomeratum

AB537169



Brachybacterium tyrofermentans

NR_026272



Brachyspira aalborgi

FM178386



Brachyspira pilosicoli

NR_075069



Brachyspira sp. HIS3

FM178387



Brachyspira sp. HIS4

FM178388



Brachyspira sp. HIS5

FM178389



Brevibacillus agri

NR_040983



Brevibacillus brevis

NR_041524



Brevibacillus centrosporus

NR_043414



Brevibacillus choshinensis

NR_040980



Brevibacillus invocatus

NR_041836



Brevibacillus laterosporus

NR_037005



Brevibacillus parabrevis

NR_040981



Brevibacillus reuszeri

NR_040982



Brevibacillus sp. phR

JN837488



Brevibacillus thermoruber

NR_026514



Brevibacterium aurantiacum

NR_044854



Brevibacterium casei

JF951998



Brevibacterium epidermidis

NR_029262



Brevibacterium frigoritolerans

NR_042639



Brevibacterium linens

AJ315491



Brevibacterium mcbrellneri

ADNU01000076



Brevibacterium paucivorans

EU086796



Brevibacterium sanguinis

NR_028016



Brevibacterium sp. H15

AB 177640



Brevibacterium sp. JC43

JF824806



Brevundimonas subvibrioides

CP002102



Brucella abortus

ACBJ01000075



Brucella canis

NR_044652



Brucella ceti

ACJD01000006



Brucella melitensis

AE009462



Brucella microti

NR_042549



Brucella ovis

NC_009504



Brucella sp. 83_13

ACBQ01000040



Brucella sp. BO1

EU053207



Brucella suis

ACBK01000034



Bryantella formatexigens

ACCL02000018



Buchnera aphidicola

NR_074609



Bulleidia extructa

ADFR01000011



Burkholderia ambifaria

AAUZ01000009



Burkholderia cenocepacia

AAEH01000060



Burkholderia cepacia

NR_041719



Burkholderia mallei

CP000547



Burkholderia multivorans

NC_010086



Burkholderia oklahomensis

DQ108388



Burkholderia pseudomallei

CP001408



Burkholderia rhizoxinica

HQ005410



Burkholderia sp. 383

CP000151



Burkholderia xenovorans

U86373


Burkholderiales bacterium 1_1_47
ADCQ01000066



Butyricicoccus pullicaecorum

HH793440



Butyricimonas virosa

AB443949



Butyrivibrio crossotus

ABWN01000012



Butyrivibrio fibrisolvens

U41172



Caldimonas manganoxidans

NR_040787



Caminicella sporogenes

NR_025485



Campylobacter coli

AAFL01000004



Campylobacter concisus

CP000792



Campylobacter curvus

NC_009715



Campylobacter fetus

ACLG01001177



Campylobacter gracilis

ACYG01000026



Campylobacter hominis

NC_009714



Campylobacter jejuni

AL139074



Campylobacter lari

CP000932



Campylobacter rectus

ACFU01000050



Campylobacter showae

ACVQ01000030



Campylobacter sp. FOBRC14

HQ616379



Campylobacter sp. FOBRC15

HQ616380



Campylobacter sp. oral clone BB120

AY005038



Campylobacter sputorum

NR_044839



Campylobacter upsaliensis

AEPU01000040



Candidatus Arthromitus sp. SFB_mouse_Yit

NR_074460



Candidatus Sulcia muelleri

CP002163



Capnocytophaga canimorsus

CP002113



Capnocytophaga genomosp. C1

AY278613



Capnocytophaga gingivalis

ACLQ01000011



Capnocytophaga granulosa

X97248



Capnocytophaga ochracea

AEOH01000054



Capnocytophaga sp. GEJ8

GU561335



Capnocytophaga sp. oral clone AH015

AY005074



Capnocytophaga sp. oral clone ASCH05

AY923149



Capnocytophaga sp. oral clone ID062

AY349368



Capnocytophaga sp. oral strain A47ROY

AY005077



Capnocytophaga sp. oral strain S3

AY005073



Capnocytophaga sp. oral taxon 338

AEXX01000050



Capnocytophaga sp. S1b

U42009



Capnocytophaga sputigena

ABZV01000054



Cardiobacterium hominis

ACKY01000036



Cardiobacterium valvarum

NR_028847



Camobacterium divergens

NR_044706



Camobacterium maltaromaticum

NC_019425



Catabacter hongkongensis

AB671763



Catenibacterium mitsuokai

AB030224



Catonella genomosp. P1 oral clone MB5_P12

DQ003629



Catonella morbi

ACIL02000016



Catonella sp. oral clone FL037

AY349369



Cedecea davisae

AF493976



Cellulosimicrobium funkei

AY501364



Cetobacterium somerae

AJ438155



Chlamydia muridarum

AE002160



Chlamydia psittaci

NR_036864



Chlamydia trachomatis

U68443


Chlamydiales bacterium NS11
JN606074


Chlamydiales bacterium NS13
JN606075


Chlamydiales bacterium NS16
JN606076



Chlamydophila pecorum

D88317



Chlamydophila pneumoniae

NC_002179



Chlamydophila psittaci

D85712


Chloroflexi genomosp. P1
AY331414



Christensenella minuta

AB490809



Chromobacterium violaceum

NC_005085



Chryseobacterium anthropi

AM982793



Chryseobacterium gleum

ACKQ02000003



Chryseobacterium hominis

NR_042517



Citrobacter amalonaticus

FR870441



Citrobacter braakii

NR_028687



Citrobacter farmeri

AF025371



Citrobacter freundii

NR_028894



Citrobacter gillenii

AF025367



Citrobacter koseri

NC_009792



Citrobacter murliniae

AF025369



Citrobacter rodentium

NR_074903



Citrobacter sedlakii

AF025364



Citrobacter sp. 30_2

ACDJ01000053



Citrobacter sp. KMSI_3

GQ468398



Citrobacter werkmanii

AF025373



Citrobacter youngae

ABWL02000011



Cloacibacillus evryensis

GQ258966


Clostridiaceae bacterium END_2
EF451053


Clostridiaceae bacterium JC13
JF824807


Clostridiales bacterium 1_7_47FAA
ABQR01000074


Clostridiales bacterium 9400853
HM587320


Clostridiales bacterium 9403326
HM587324


Clostridiales bacterium oral clone P4PA_66 P1
AY207065


Clostridiales bacterium oral taxon 093
GQ422712


Clostridiales bacterium oral taxon F32
HM099644


Clostridiales bacterium ph2
JN837487


Clostridiales bacterium SY8519
AB477431


Clostridiales genomosp. BVAB3
CP001850


Clostridiales sp. SM4_1
FP929060


Clostridiales sp. SS3_4
AY305316


Clostridiales sp. SSC_2
FP929061



Clostridium acetobutylicum

NR_074511



Clostridium aerotolerans

X76163



Clostridium aldenense

NR_043680



Clostridium aldrichii

NR_026099



Clostridium algidicamis

NR_041746



Clostridium algidixylanolyticum

NR_028726



Clostridium aminovalericum

NR_029245



Clostridium amygdalinum

AY353957



Clostridium argentinense

NR_029232



Clostridium asparagiforme

ACCJ01000522



Clostridium baratii

NR_029229



Clostridium bartlettii

ABEZ02000012



Clostridium beijerinckii

NR_074434



Clostridium bifermentans

X73437



Clostridium bolteae

ABCC02000039



Clostridium botulinum

NC_010723



Clostridium butyricum

ABDT01000017



Clostridium cadaveris

AB542932



Clostridium carboxidivorans

FR733710



Clostridium carnis

NR_044716



Clostridium celatum

X77844



Clostridium celerecrescens

JQ246092



Clostridium cellulosi

NR_044624



Clostridium chauvoei

EU106372



Clostridium citroniae

ADLJ01000059



Clostridium clariflavum

NR_041235



Clostridium clostridiiformes

M59089



Clostridium clostridioforme

NR_044715



Clostridium coccoides

EF025906



Clostridium cochlearium

NR_044717



Clostridium cocleatum

NR_026495



Clostridium colicanis

FJ957863



Clostridium colinum

NR_026151



Clostridium difficile

NC_013315



Clostridium disporicum

NR_026491



Clostridium estertheticum

NR_042153



Clostridium fallax

NR_044714



Clostridium favososporum

X76749



Clostridium felsineum

AF270502



Clostridium frigidicamis

NR_024919



Clostridium gasigenes

NR_024945



Clostridium ghonii

AB542933



Clostridium glycolicum

FJ384385



Clostridium glycyrrhizinilyticum

AB233029



Clostridium haemolyticum

NR_024749



Clostridium hathewayi

AY552788



Clostridium hiranonis

AB023970



Clostridium histolyticum

HF558362



Clostridium hylemonae

AB023973



Clostridium indolis

AF028351



Clostridium innocuum

M23732



Clostridium irregulare

NR_029249



Clostridium isatidis

NR_026347



Clostridium kluyveri

NR_074165



Clostridium lactatifermentans

NR_025651



Clostridium lavalense

EF564277



Clostridium leptum

AJ305238



Clostridium limosum

FR870444



Clostridium magnum

X77835



Clostridium malenominatum

FR749893



Clostridium mayombei

FR733682



Clostridium methylpentosum

ACEC01000059



Clostridium nexile

X73443



Clostridium novyi

NR_074343



Clostridium orbiscindens

Y18187



Clostridium oroticum

FR749922



Clostridium paraputrificum

AB536771



Clostridium perfringens

ABDW01000023



Clostridium phytofermentans

NR_074652



Clostridium piliforme

D14639



Clostridium putrefaciens

NR_024995



Clostridium quinii

NR_026149



Clostridium ramosum

M23731



Clostridium rectum

NR_029271



Clostridium saccharogumia

DQ100445



Clostridium saccharolyticum

CP002109



Clostridium sardiniense

NR_041006



Clostridium sariagoforme

NR_026490



Clostridium scindens

AF262238



Clostridium septicum

NR_026020



Clostridium sordellii

AB448946



Clostridium sp. 7_2_43FAA

ACDK01000101



Clostridium sp. D5

ADBG01000142



Clostridium sp. HGF2

AENW01000022



Clostridium sp. HPB_46

AY862516



Clostridium sp. JC122

CAEV01000127



Clostridium sp. L2_50

AAYW02000018



Clostridium sp. LMG 16094

X95274



Clostridium sp. M62_1

ACFX02000046



Clostridium sp. MLG055

AF304435



Clostridium sp. MT4 E

FJ159523



Clostridium sp. NMBHI_1

JN093130



Clostridium sp. NML 04A032

EU815224



Clostridium sp. SS2_1

ABGC03000041



Clostridium sp. SY8519

AP012212



Clostridium sp. TM_40

AB249652



Clostridium sp. YIT 12069

AB491207



Clostridium sp. YIT 12070

AB491208



Clostridium sphenoides

X73449



Clostridium spiroforme

X73441



Clostridium sporogenes

ABKW02000003



Clostridium sporosphaeroides

NR_044835



Clostridium stercorarium

NR_025100



Clostridium sticklandii

L04167



Clostridium straminisolvens

NR_024829



Clostridium subterminale

NR_041795



Clostridium sulfidigenes

NR_044161



Clostridium symbiosum

ADLQ01000114



Clostridium tertium

Y18174



Clostridium tetani

NC_004557



Clostridium thermocellum

NR_074629



Clostridium tyrobutyricum

NR_044718



Clostridium viride

NR_026204



Clostridium xylanolyticum

NR_037068



Collinsella aerofaciens

AAVN02000007



Collinsella intestinalis

ABXH02000037



Collinsella stercoris

ABXJ01000150



Collinsella tanakaei

AB490807


Comamonadaceae bacterium NML000135
JN585335


Comamonadaceae bacterium NML790751
JN585331


Comamonadaceae bacterium NML910035
JN585332


Comamonadaceae bacterium NML910036
JN585333


Comamonadaceae bacterium oral taxon F47
HM099651



Comamonas sp. NSP5

AB076850



Conchiformibius kuhniae

NR_041821



Coprobacillus cateniformis

AB030218



Coprobacillus sp. 29_1

ADKX01000057



Coprobacillus sp. D7

ACDT01000199



Coprococcus catus

EU266552



Coprococcus comes

ABVR01000038



Coprococcus eutactus

EF031543



Coprococcus sp. ART55_1

AY350746


Coriobacteriaceae bacterium BV3Ac1
JN809768


Coriobacteriaceae bacterium JC110
CAEM01000062


Coriobacteriaceae bacterium phI
JN837493



Corynebacterium accolens

ACGD01000048



Corynebacterium ammoniagenes

ADNS01000011



Corynebacterium amycolatum

ABZU01000033



Corynebacterium appendicis

NR_028951



Corynebacterium argentoratense

EF463055



Corynebacterium atypicum

NR_025540



Corynebacterium aurimucosum

ACLH01000041



Corynebacterium bovis

AF537590



Corynebacterium canis

GQ871934



Corynebacterium casei

NR_025101



Corynebacterium confusum

Y15886



Corynebacterium coyleae

X96497



Corynebacterium diphtheriae

NC_002935



Corynebacterium durum

Z97069



Corynebacterium efficiens

ACLI01000121



Corynebacterium falsenii

Y13024



Corynebacterium flavescens

NR_037040



Corynebacterium genitalium

ACLJ01000031



Corynebacterium glaucum

NR_028971



Corynebacterium glucuronolyticum

ABYP01000081



Corynebacterium glutamicum

BA000036



Corynebacterium hansenii

AM946639



Corynebacterium imitans

AF537597



Corynebacterium jeikeium

ACYW01000001



Corynebacterium kroppenstedtii

NR_026380



Corynebacterium lipophiloflavum

ACHJ01000075



Corynebacterium macginleyi

AB359393



Corynebacterium mastitidis

AB359395



Corynebacterium matruchotii

ACSH02000003



Corynebacterium minutissimum

X82064



Corynebacterium mucifaciens

NR_026396



Corynebacterium propinquum

NR_037038



Corynebacterium pseudodiphtheriticum

X84258



Corynebacterium pseudogenitalium

ABYQ01000237



Corynebacterium pseudotuberculosis

NR_037070



Corynebacterium pyruviciproducens

FJ185225



Corynebacterium renale

NR_037069



Corynebacterium resistens

ADGN01000058



Corynebacterium riegelii

EU848548



Corynebacterium simulans

AF537604



Corynebacterium singulare

NR_026394



Corynebacterium sp. 1 ex sheep

Y13427



Corynebacterium sp. L_2012475

HE575405



Corynebacterium sp. NML 93_0481

GU238409



Corynebacterium sp. NML 97_0186

GU238411



Corynebacterium sp. NML 99_0018

GU238413



Corynebacterium striatum

ACGE01000001



Corynebacterium sundsvallense

Y09655



Corynebacterium tuberculostearicum

ACVP01000009



Corynebacterium tuscaniae

AY677186



Corynebacterium ulcerans

NR_074467



Corynebacterium urealyticum

X81913



Corynebacterium ureicelerivorans

AM397636



Corynebacterium variabile

NR_025314



Corynebacterium xerosis

FN179330



Coxiella burnetii

CP000890



Cronobacter malonaticus

GU122174



Cronobacter sakazakii

NC_009778



Cronobacter turicensis

FN543093



Cryptobacterium curtum

GQ422741



Cupriavidus metallidurans

GU230889



Cytophaga xylanolytica

FR733683


Deferribacteres sp. oral clone JV001
AY349370


Deferribacteres sp. oral clone JV006
AY349371


Deferribacteres sp. oral clone JV023
AY349372



Deinococcus radiodurans

AE000513



Deinococcus sp. R_43890

FR682752



Delftia acidovorans

CP000884



Dermabacter hominis

FJ263375



Dermacoccus sp. Ellin185

AEIQ01000090



Desmospora activa

AM940019



Desmospora sp. 8437

AFHT01000143



Desulfitobacterium frappieri

AJ276701



Desulfitobacterium hafniense

NR_074996



Desulfobulbus sp. oral clone CH031

AY005036



Desulfotomaculum nigrificans

NR_044832



Desulfovibrio desulfuricans

DQ092636



Desulfovibrio fairfieldensis

U42221



Desulfovibrio piger

AF192152



Desulfovibrio sp. 3_1_syn3

ADDR01000239



Desulfovibrio vulgaris

NR_074897



Dialister invisus

ACIM02000001



Dialister micraerophilus

AFBB01000028



Dialister microaerophilus

AENT01000008



Dialister pneumosintes

HM596297



Dialister propionicifaciens

NR_043231



Dialister sp. oral taxon 502

GQ422739



Dialister succinatiphilus

AB370249



Dietzia natronolimnaea

GQ870426



Dietzia sp. BBDP51

DQ337512



Dietzia sp. CA149

GQ870422



Dietzia timorensis

GQ870424



Dorea formicigenerans

AAXA02000006



Dorea longicatena

AJ132842



Dysgonomonas gadei

ADLV01000001



Dysgonomonas mossii

ADLW01000023



Edwardsiella tarda

CP002154



Eggerthella lenta

AF292375



Eggerthella sinensis

AY321958



Eggerthella sp. 1_3_56FAA

ACWN01000099



Eggerthella sp. HGA1

AEXR01000021



Eggerthella sp. YY7918

AP012211



Ehrlichia chaffeensis

AAIF01000035



Eikenella corrodens

ACEA01000028



Enhydrobacter aerosaccus

ACYI01000081



Enterobacter aerogenes

AJ251468



Enterobacter asburiae

NR_024640



Enterobacter cancerogenus

Z96078



Enterobacter cloacae

FP929040



Enterobacter cowanii

NR_025566



Enterobacter hormaechei

AFHR01000079



Enterobacter sp. 247BMC

HQ122932



Enterobacter sp. 638

NR_074777



Enterobacter sp. JC163

JN657217



Enterobacter sp. SCSS

HM007811



Enterobacter sp. TSE38

HM156134


Enterobacteriaceae bacterium 9_2_54FAA
ADCU01000033


Enterobacteriaceae bacterium CF01Ent_1
AJ489826


Enterobacteriaceae bacterium Smarlab 3302238
AY538694



Enterococcus avium

AF133535



Enterococcus caccae

AY943820



Enterococcus casseliflavus

AEWT01000047



Enterococcus durans

AJ276354



Enterococcus faecalis

AE016830



Enterococcus faecium

AM157434



Enterococcus gallinarum

AB269767



Enterococcus gilvus

AY033814



Enterococcus hawaiiensis

AY321377



Enterococcus hirae

AF061011



Enterococcus italicus

AEPV01000109



Enterococcus mundtii

NR_024906



Enterococcus raffinosus

FN600541



Enterococcus sp. BV2CASA2

JN809766



Enterococcus sp. CCRI_16620

GU457263



Enterococcus sp. F95

FJ463817



Enterococcus sp. RfL6

AJ133478



Enterococcus thailandicus

AY321376



Eremococcus coleocola

AENN01000008



Erysipelothrix inopinata

NR_025594



Erysipelothrix rhusiopathiae

ACLK01000021



Erysipelothrix tonsillarum

NR_040871


Erysipelotrichaceae bacterium 3_1_53
ACTJ01000113


Erysipelotrichaceae bacterium 5_2_54FAA
ACZW01000054



Escherichia albertii

ABKX01000012



Escherichia coli

NC_008563



Escherichia fergusonii

CU928158



Escherichia hermannii

HQ407266



Escherichia sp. 1_1_43

ACID0100003 3



Escherichia sp. 4_1_40B

ACDM02000056



Escherichia sp. B4

EU722735



Escherichia vulneris

NR_041927



Ethanoligenens harbinense

AY675965


Eubacteriaceae bacterium P4P_50 P4
AY207060



Eubacterium barkeri

NR_044661



Eubacterium biforme

ABYT01000002



Eubacterium brachy

U13038



Eubacterium budayi

NR_024682



Eubacterium callanderi

NR_026330



Eubacterium cellulosolvens

AY178842



Eubacterium contortum

FR749946



Eubacterium coprostanoligenes

HM037995



Eubacterium cylindroides

FP929041



Eubacterium desmolans

NR_044644



Eubacterium dolichum

L34682



Eubacterium eligens

CP001104



Eubacterium fissicatena

FR749935



Eubacterium hadrum

FR749933



Eubacterium hallii

L34621



Eubacterium infirmum

U13039



Eubacterium limosum

CP002273



Eubacterium moniliforme

HF558373



Eubacterium multiforme

NR_024683



Eubacterium nitritogenes

NR_024684



Eubacterium nodatum

U13041



Eubacterium ramulus

AJ011522



Eubacterium rectale

FP929042



Eubacterium ruminantium

NR_024661



Eubacterium saburreum

AB525414



Eubacterium saphenum

NR_026031



Eubacterium siraeum

ABCA03000054



Eubacterium sp. 3_1_31

ACTL01000045



Eubacterium sp. AS15b

HQ616364



Eubacterium sp. OBRC9

HQ616354



Eubacterium sp. oral clone GI038

AY349374



Eubacterium sp. oral clone IR009

AY349376



Eubacterium sp. oral clone JH012

AY349373



Eubacterium sp. oral clone JI012

AY349379



Eubacterium sp. oral clone JN088

AY349377



Eubacterium sp. oral clone JS001

AY349378



Eubacterium sp. oral clone OH3A

AY947497



Eubacterium sp. WAL 14571

FJ687606



Eubacterium tenue

M59118



Eubacterium tortuosum

NR_044648



Eubacterium ventriosum

L34421



Eubacterium xylanophilum

L34628



Eubacterium yurii

AEES01000073



Ewingella americana

JN175329



Exiguobacterium acetylicum

FJ970034



Facklamia hominis

Y10772



Faecalibacterium prausnitzii

ACOP02000011



Filifactor alocis

CP002390



Filifactor villosus

NR_041928



Finegoldia magna

ACHM02000001


Flavobacteriaceae genomosp. C1
AY278614



Flavobacterium sp. NF2_1

FJ195988



Flavonifractor plautii

AY724678



Flexispira rappini

AY126479



Flexistipes sinusarabici

NR_074881



Francisella novicida

ABSS01000002



Francisella philomiragia

AY928394



Francisella tularensis

ABAZ01000082



Fulvimonas sp. NML 060897

EF589680



Fusobacterium canifelinum

AY162222



Fusobacterium genomosp. C1

AY278616



Fusobacterium genomosp. C2

AY278617



Fusobacterium gonidiaformans

ACET01000043



Fusobacterium mortiferum

ACDB02000034



Fusobacterium naviforme

HQ223106



Fusobacterium necrogenes

X55408



Fusobacterium necrophorum

AM905356



Fusobacterium nucleatum

ADVK01000034



Fusobacterium periodonticum

ACJY01000002



Fusobacterium russii

NR_044687



Fusobacterium sp. 1_1_41FAA

ADGG01000053



Fusobacterium sp. 11_3_2

ACUO01000052



Fusobacterium sp. 12_1B

AGWJ01000070



Fusobacterium sp. 2_1_31

ACDC02000018



Fusobacterium sp. 3_1_27

ADGF01000045



Fusobacterium sp. 3_1_33

ACQE01000178



Fusobacterium sp. 3_1_36A2

ACPU01000044



Fusobacterium sp. 3_1_5R

ACDD01000078



Fusobacterium sp. AC18

HQ616357



Fusobacterium sp. ACB2

HQ616358



Fusobacterium sp. AS2

HQ616361



Fusobacterium sp. CM1

HQ616371



Fusobacterium sp. CM21

HQ616375



Fusobacterium sp. CM22

HQ616376



Fusobacterium sp. D12

ACDG02000036



Fusobacterium sp. oral clone ASCF06

AY923141



Fusobacterium sp. oral clone ASCF11

AY953256



Fusobacterium ulcerans

ACDH01000090



Fusobacterium varium

ACIE01000009



Gardnerella vaginalis

CP001849



Gemella haemolysans

ACDZ02000012



Gemella morbillorum

NR_025904



Gemella morbillorum

ACRX01000010



Gemella sanguinis

ACRY01000057



Gemella sp. oral clone ASCE02

AY923133



Gemella sp. oral clone ASCF04

AY923139



Gemella sp. oral clone ASCF12

AY923143



Gemella sp. WAL 1945J

EU427463



Gemmiger formicilis

GU562446



Geobacillus kaustophilus

NR_074989



Geobacillus sp. E263

DQ647387



Geobacillus sp. WCH70

CP001638



Geobacillus stearothermophilus

NR_040794



Geobacillus thermocatenulatus

NR_043020



Geobacillus thermodenitrificans

NR_074976



Geobacillus thermoglucosidasius

NR_043022



Geobacillus thermoleovorans

NR_074931



Geobacter bemidjiensis

CP001124



Gloeobacter violaceus

NR_074282



Gluconacetobacter azotocaptans

NR_028767



Gluconacetobacter diazotrophicus

NR_074292



Gluconacetobacter entanii

NR_028909



Gluconacetobacter europaeus

NR_026513



Gluconacetobacter hansenii

NR_026133



Gluconacetobacter johannae

NR_024959



Gluconacetobacter oboediens

NR_041295



Gluconacetobacter xylinus

NR_074338



Gordonia bronchialis

NR_027594



Gordonia polyisoprenivorans

DQ385609



Gordonia sp. KTR9

DQ068383



Gordonia sputi

FJ536304



Gordonia terrae

GQ848239



Gordonibacter pamelaeae

AM886059



Gordonibacter pamelaeae

FP929047



Gracilibacter thermotolerans

NR_043559



Gramella forsetii

NR_074707



Granulicatella adiacens

ACKZ01000002



Granulicatella elegans

AB252689



Granulicatella paradiacens

AY879298



Granulicatella sp. M658_99_3

AJ271861



Granulicatella sp. oral clone ASC02

AY923126



Granulicatella sp. oral clone ASCA05

DQ341469



Granulicatella sp. oral clone ASCB09

AY953251



Granulicatella sp. oral clone ASCG05

AY923146



Grimontia hollisae

ADAQ01000013



Haematobacter sp. BC14248

GU396991



Haemophilus aegyptius

AFBC01000053



Haemophilus ducreyi

AE017143



Haemophilus genomosp. P2 oral clone MB3_C24

DQ003621



Haemophilus genomosp. P3 oral clone MB3_C38

DQ003635



Haemophilus haemolyticus

JN175335



Haemophilus influenzae

AADP01000001



Haemophilus parahaemolyticus

GU561425



Haemophilus parainfluenzae

AEWU01000024



Haemophilus paraphrophaemolyticus

M75076



Haemophilus parasuis

GU226366



Haemophilus somnus

NC_008309



Haemophilus sp. 70334

HQ680854



Haemophilus sp. HK445

FJ685624



Haemophilus sp. oral clone ASCA07

AY923117



Haemophilus sp. oral clone ASCG06

AY923147



Haemophilus sp. oral clone BJ021

AY005034



Haemophilus sp. oral clone BJ095

AY005033



Haemophilus sp. oral clone JM053

AY349380



Haemophilus sp. oral taxon 851

AGRK01000004



Haemophilus sputorum

AFNK01000005



Hafnia alvei

DQ412565



Halomonas elongata

NR_074782



Halomonas johnsoniae

FR775979



Halorubrum lipolyticum

AB477978



Helicobacter bilis

ACDN01000023



Helicobacter canadensis

ABQS01000108



Helicobacter cinaedi

ABQT01000054



Helicobacter pullorum

ABQU01000097



Helicobacter pylori

CP000012



Helicobacter sp. None

U44756



Helicobacter winghamensis

ACDO01000013



Heliobacterium modesticaldum

NR_074517



Herbaspirillum seropedicae

CP002039



Herbaspirillum sp. JC206

JN657219



Histophilus somni

AF549387



Holdemania filiformis

Y11466



Hydrogenoanaerobacterium saccharovorans

NR_044425



Hyperthermus butylicus

CP000493



Hyphomicrobium sulfonivorans

AY468372



Hyphomonas neptunium

NR_074092



Ignatzschineria indica

HQ823562



Ignatzschineria sp. NML 95_0260

HQ823559



Ignicoccus islandicus

X99562



Inquilinus limosus

NR_029046



Janibacter limosus

NR_026362



Janibacter melonis

EF063716



Janthinobacterium sp. SY12

EF455530



Johnsonella ignava

X87152



Jonquetella anthropi

ACOO02000004



Kerstersia gyiorum

NR_025669



Kingella denitrificans

AEWV01000047



Kingella genomosp. P1 oral cone MB2_C20

DQ003616



Kingella kingae

AFHS01000073



Kingella oralis

ACJW02000005



Kingella sp. oral clone ID059

AY349381



Klebsiella oxytoca

AY292871



Klebsiella pneumoniae

CP000647



Klebsiella sp. AS10

HQ616362



Klebsiella sp. Co9935

DQ068764



Klebsiella sp. enrichment culture clone SRC_DSD25

HM195210



Klebsiella sp. OBRC7

HQ616353



Klebsiella sp. SP_BA

FJ999767



Klebsiella sp. SRC_DSD1

GU797254



Klebsiella sp. SRC_DSD11

GU797263



Klebsiella sp. SRC_DSD12

GU797264



Klebsiella sp. SRC_DSD15

GU797267



Klebsiella sp. SRC_DSD2

GU797253



Klebsiella sp. SRC_DSD6

GU797258



Klebsiella variicola

CP001891



Kluyvera ascorbata

NR_028677



Kluyvera cryocrescens

NR_028803



Kocuria marina

GQ260086



Kocuria palustris

EU333884



Kocuria rhizophila

AY030315



Kocuria rosea

X87756



Kocuria varians

AF542074



Lachnobacterium bovis

GU324407



Lachnospira multipara

FR733699



Lachnospira pectinoschiza

L14675


Lachnospiraceae bacterium 1_1_57FAA
ACTM01000065


Lachnospiraceae bacterium 1_4_56FAA
ACTN01000028


Lachnospiraceae bacterium 2_1_46FAA
ADLB01000035


Lachnospiraceae bacterium 2_1_58FAA
ACTO01000052


Lachnospiraceae bacterium 3_1_57FAA_CT1
ACTP01000124


Lachnospiraceae bacterium 4_1_37FAA
ADCR01000030


Lachnospiraceae bacterium 5_1_57FAA
ACTR01000020


Lachnospiraceae bacterium 5_1_63FAA
ACTS01000081


Lachnospiraceae bacterium 6_1_63FAA
ACTV01000014


Lachnospiraceae bacterium 8_1_57FAA
ACWQ01000079


Lachnospiraceae bacterium 9_1_43BFAA
ACTX01000023


Lachnospiraceae bacterium A4
DQ789118


Lachnospiraceae bacterium DJF VP30
EU728771


Lachnospiraceae bacterium ICM62
HQ616401


Lachnospiraceae bacterium MSX33
HQ616384


Lachnospiraceae bacterium oral taxon 107
ADDS01000069


Lachnospiraceae bacterium oral taxon F15
HM099641


Lachnospiraceae genomosp. C1
AY278618



Lactobacillus acidipiscis

NR_024718



Lactobacillus acidophilus

CP000033



Lactobacillus alimentarius

NR_044701



Lactobacillus amylolyticus

ADNY01000006



Lactobacillus amylovorus

CP002338



Lactobacillus antri

ACLL01000037



Lactobacillus brevis

EU194349



Lactobacillus buchneri

ACGH01000101



Lactobacillus casei

CP000423



Lactobacillus catenaformis

M23729



Lactobacillus coleohominis

ACOH01000030



Lactobacillus coryniformis

NR_044705



Lactobacillus crispatus

ACOG01000151



Lactobacillus curvatus

NR_042437



Lactobacillus delbrueckii

CP002341



Lactobacillus dextrinicus

NR_036861



Lactobacillus farciminis

NR_044707



Lactobacillus fermentum

CP002033



Lactobacillus gasseri

ACOZ01000018



Lactobacillus gastricus

AICN01000060



Lactobacillus genomosp. C1

AY278619



Lactobacillus genomosp. C2

AY278620



Lactobacillus helveticus

ACLM01000202



Lactobacillus hilgardii

ACGP01000200



Lactobacillus hominis

FR681902



Lactobacillus iners

AEKJ01000002



Lactobacillus jensenii

ACQD01000066



Lactobacillus johnsonii

AE017198



Lactobacillus kalixensis

NR_029083



Lactobacillus kefiranofaciens

NR_042440



Lactobacillus kefiri

NR_042230



Lactobacillus kimchii

NR_025045



Lactobacillus leichmannii

JX986966



Lactobacillus mucosae

FR693800



Lactobacillus murinus

NR_042231



Lactobacillus nodensis

NR_041629



Lactobacillus oeni

NR_043095



Lactobacillus oris

AEKL01000077



Lactobacillus parabrevis

NR_042456



Lactobacillus parabuchneri

NR_041294



Lactobacillus paracasei

ABQV01000067



Lactobacillus parakefiri

NR_029039



Lactobacillus pentosus

JN813103



Lactobacillus perolens

NR_029360



Lactobacillus plantarum

ACGZ02000033



Lactobacillus pontis

HM218420



Lactobacillus reuteri

ACGW02000012



Lactobacillus rhamnosus

ABWJ01000068



Lactobacillus rogosae

GU269544



Lactobacillus ruminis

ACGS02000043



Lactobacillus sakei

DQ989236



Lactobacillus salivarius

AEBA01000145



Lactobacillus saniviri

AB602569



Lactobacillus senioris

AB602570



Lactobacillus sp. 66c

FR681900



Lactobacillus sp. BT6

HQ616370



Lactobacillus sp. KLDS 1.0701

EU600905



Lactobacillus sp. KLDS 1.0702

EU600906



Lactobacillus sp. KLDS 1.0703

EU600907



Lactobacillus sp. KLDS 1.0704

EU600908



Lactobacillus sp. KLDS 1.0705

EU600909



Lactobacillus sp. KLDS 1.0707

EU600911



Lactobacillus sp. KLDS 1.0709

EU600913



Lactobacillus sp. KLDS 1.0711

EU600915



Lactobacillus sp. KLDS 1.0712

EU600916



Lactobacillus sp. KLDS 1.0713

EU600917



Lactobacillus sp. KLDS 1.0716

EU600921



Lactobacillus sp. KLDS 1.0718

EU600922



Lactobacillus sp. KLDS 1.0719

EU600923



Lactobacillus sp. oral clone HT002

AY349382



Lactobacillus sp. oral clone HT070

AY349383



Lactobacillus sp. oral taxon 052

GQ422710



Lactobacillus tucceti

NR_042194



Lactobacillus ultunensis

ACGU01000081



Lactobacillus vaginalis

ACGV01000168



Lactobacillus vini

NR_042196



Lactobacillus vitulinus

NR_041305



Lactobacillus zeae

NR_037122



Lactococcus garvieae

AF061005



Lactococcus lactis

CP002365



Lactococcus raffinolactis

NR_044359



Lactonifactor longoviformis

DQ100449



Laribacter hongkongensis

CP001154



Lautropia mirabilis

AEQP01000026



Lautropia sp. oral clone AP009

AY005030



Legionella hackeliae

M36028



Legionella longbeachae

M36029



Legionella pneumophila

NC_002942



Legionella sp. D3923

JN380999



Legionella sp. D4088

JN381012



Legionella sp. H63

JF831047



Legionella sp. NML 93L054

GU062706



Legionella steelei

HQ398202



Leminorella grimontii

AJ233421



Leminorella richardii

HF558368



Leptospira borgpetersenii

NC_008508



Leptospira broomii

NR_043200



Leptospira interrogans

NC_005823



Leptospira licerasiae

EF612284



Leptotrichia buccalis

CP001685



Leptotrichia genomosp. C1

AY278621



Leptotrichia goodfellowii

ADAD01000110



Leptotrichia hofstadii

ACVB02000032



Leptotrichia shahii

AY029806



Leptotrichia sp. neutropenicPatient

AF189244



Leptotrichia sp. oral clone GT018

AY349384



Leptotrichia sp. oral clone GT020

AY349385



Leptotrichia sp. oral clone HE012

AY349386



Leptotrichia sp. oral clone IK040

AY349387



Leptotrichia sp. oral clone P2PB_51 P1

AY207053



Leptotrichia sp. oral taxon 223

GU408547



Leuconostoc carnosum

NR_040811



Leuconostoc citreum

AM157444



Leuconostoc gasicomitatum

FN822744



Leuconostoc inhae

NR_025204



Leuconostoc kimchii

NR_075014



Leuconostoc lactis

NR_040823



Leuconostoc mesenteroides

ACKV01000113



Leuconostoc pseudomesenteroides

NR_040814



Listeria grayi

ACCR02000003



Listeria innocua

JF967625



Listeria ivanovii

X56151



Listeria monocytogenes

CP002003



Listeria welshimeri

AM263198



Luteococcus sanguinis

NR_025507



Lutispora thermophila

NR_041236



Lysinibacillus fusiformis

FN397522



Lysinibacillus sphaericus

NR_074883



Macrococcus caseolyticus

NR_074941



Mannheimia haemolytica

ACZX01000102



Marvinbryantia formatexigens

AJ505973



Massilia sp. CCUG 43427A

FR773700



Megamonas funiformis

AB300988



Megamonas hypermegale

AJ420107



Megasphaera elsdenii

AY038996



Megasphaera genomosp. C1

AY278622



Megasphaera genomosp. type_1

ADGP01000010



Megasphaera micronuciformis

AECS01000020



Megasphaera sp. BLPYG_07

HM990964



Megasphaera sp. UPII 199_6

AFIJ01000040



Metallosphaera sedula

D26491



Methanobacterium formicicum

NR_025028



Methanobrevibacter acididurans

NR_028779



Methanobrevibacter arboriphilus

NR_042783



Methanobrevibacter curvatus

NR_044796



Methanobrevibacter cuticularis

NR_044776



Methanobrevibacter filiformis

NR_044801



Methanobrevibacter gottschalkii

NR_044789



Methanobrevibacter millerae

NR_042785



Methanobrevibacter olleyae

NR_043024



Methanobrevibacter oralis

HE654003



Methanobrevibacter ruminantium

NR_042784



Methanobrevibacter smithii

ABYV02000002



Methanobrevibacter thaueri

NR_044787



Methanobrevibacter woesei

NR_044788



Methanobrevibacter wolinii

NR_044790



Methanosphaera stadtmanae

AY196684



Methylobacterium extorquens

NC_010172



Methylobacterium podarium

AY468363



Methylobacterium radiotolerans

GU294320



Methylobacterium sp. 1sub

AY468371



Methylobacterium sp. MM4

AY468370



Methylocella silvestris

NR_074237



Methylophilus sp. ECd5

AY436794



Microbacterium chocolatum

NR_037045



Microbacterium flavescens

EU714363



Microbacterium gubbeenense

NR_025098



Microbacterium lacticum

EU714351



Microbacterium oleivorans

EU714381



Microbacterium oxydans

EU714348



Microbacterium paraoxydans

AJ491806



Microbacterium phyllosphaerae

EU714359



Microbacterium schleiferi

NR_044936



Microbacterium sp. 768

EU714378



Microbacterium sp. oral strain C24KA

AF287752



Microbacterium testaceum

EU714365



Micrococcus antarcticus

NR_025285



Micrococcus luteus

NR_075062



Micrococcus lylae

NR_026200



Micrococcus sp. 185

EU714334



Microcystis aeruginosa

NC_010296



Mitsuokella jalaludinii

NR_028840



Mitsuokella multacida

ABWK02000005



Mitsuokella sp. oral taxon 521

GU413658



Mitsuokella sp. oral taxon G68

GU432166



Mobiluncus curtisii

AEPZ01000013



Mobiluncus mulieris

ACKW01000035



Moellerella wisconsensis

JN175344



Mogibacterium diversum

NR_027191



Mogibacterium neglectum

NR_027203



Mogibacterium pumilum

NR_028608



Mogibacterium timidum

Z36296


Mollicutes bacterium pACH93
AY297808



Moorella thermoacetica

NR_075001



Moraxella catarrhalis

CP002005



Moraxella lincolnii

FR822735



Moraxella osloensis

JN175341



Moraxella sp. 16285

JF682466



Moraxella sp. GM2

JF837191



Morganella morganii

AJ301681



Morganella sp. JB_T16

AJ781005



Morococcus cerebrosus

JN175352



Moryella indoligenes

AF527773



Mycobacterium abscessus

AGQU01000002



Mycobacterium africanum

AF480605



Mycobacterium alsiensis

AJ938169



Mycobacterium avium

CP000479



Mycobacterium chelonae

AB548610



Mycobacterium colombiense

AM062764



Mycobacterium elephantis

AF385898



Mycobacterium gordonae

GU142930



Mycobacterium intracellulare

GQ153276



Mycobacterium kansasii

AF480601



Mycobacterium lacus

NR_025175



Mycobacterium leprae

FM211192



Mycobacterium lepromatosis

EU203590



Mycobacterium mageritense

FR798914



Mycobacterium mantenii

FJ042897



Mycobacterium marinum

NC_010612



Mycobacterium microti

NR_025234



Mycobacterium neoaurum

AF268445



Mycobacterium parascrofulaceum

ADNV01000350



Mycobacterium paraterrae

EU919229



Mycobacterium phlei

GU142920



Mycobacterium seoulense

DQ536403



Mycobacterium smegmatis

CP000480



Mycobacterium sp. 1761

EU703150



Mycobacterium sp. 1776

EU703152



Mycobacterium sp. 1781

EU703147



Mycobacterium sp. 1791

EU703148



Mycobacterium sp. 1797

EU703149



Mycobacterium sp. AQ1GA4

HM210417



Mycobacterium sp. B10_07.09.0206

HQ174245



Mycobacterium sp. GN_10546

FJ497243



Mycobacterium sp. GN_10827

FJ497247



Mycobacterium sp. GN_11124

FJ652846



Mycobacterium sp. GN_9188

FJ497240



Mycobacterium sp. GR_2007_210

FJ555538



Mycobacterium sp. HE5

AJ012738



Mycobacterium sp. NLA001000736

HM627011



Mycobacterium sp. W

DQ437715



Mycobacterium tuberculosis

CP001658



Mycobacterium ulcerans

AB548725



Mycobacterium vulneris

EU834055



Mycoplasma agalactiae

AF010477



Mycoplasma amphoriforme

AY531656



Mycoplasma arthritidis

NC_011025



Mycoplasma bovoculi

NR_025987



Mycoplasma faucium

NR_024983



Mycoplasma fermentans

CP002458



Mycoplasma flocculare

X62699



Mycoplasma genitalium

L43967



Mycoplasma hominis

AF443616



Mycoplasma orale

AY796060



Mycoplasma ovipneumoniae

NR_025989



Mycoplasma penetrans

NC_004432



Mycoplasma pneumoniae

NC_000912



Mycoplasma putrefaciens

U26055



Mycoplasma salivarium

M24661


Mycoplasmataceae genomosp. P1 oral clone
DQ003614


MB1_G23



Myroides odoratimimus

NR_042354



Myroides sp. MY15

GU253339



Neisseria bacilliformis

AFAY01000058



Neisseria cinerea

ACDY01000037



Neisseria elongata

ADBF01000003



Neisseria flavescens

ACQV01000025



Neisseria genomosp. P2 oral clone MB5_P15

DQ003630



Neisseria gonorrhoeae

CP002440



Neisseria lactamica

ACEQ01000095



Neisseria macacae

AFQE01000146



Neisseria meningitidis

NC_003112



Neisseria mucosa

ACDX01000110



Neisseria pharyngis

AJ239281



Neisseria polysaccharea

ADBE01000137



Neisseria sicca

ACKO02000016



Neisseria sp. KEM232

GQ203291



Neisseria sp. oral clone API32

AY005027



Neisseria sp. oral clone JC012

AY349388



Neisseria sp. oral strain B33KA

AY005028



Neisseria sp. oral taxon 014

ADEA01000039



Neisseria sp. SMC_A9199

FJ763637



Neisseria sp. TM10_1

DQ279352



Neisseria subflava

ACEO01000067



Neorickettsia risticii

CP001431



Neorickettsia sennetsu

NC_007798



Nocardia brasiliensis

AIHV01000038



Nocardia cyriacigeorgica

HQ009486



Nocardia farcinica

NC_006361



Nocardia puris

NR_028994



Nocardia sp. 01_Je_025

GU574059



Nocardiopsis dassonvillei

CP002041



Novosphingobium aromaticivorans

AAAV03000008



Oceanobacillus caeni

NR_041533



Oceanobacillus sp. Ndiop

CAER01000083



Ochrobactrum anthropi

NC_009667



Ochrobactrum intermedium

ACQA01000001



Ochrobactrum pseudintermedium

DQ365921



Odoribacter laneus

AB490805



Odoribacter splanchnicus

CP002544



Okadaella gastrococcus

HQ699465



Oligella ureolytica

NR_041998



Oligella urethralis

NR_041753



Olsenella genomosp. C1

AY278623



Olsenella profusa

FN178466



Olsenella sp. F0004

EU592964



Olsenella sp. oral taxon 809

ACVE01000002



Olsenella uli

CP002106



Opitutus terrae

NR_074978



Oribacterium sinus

ACKX01000142



Oribacterium sp. ACB1

HM120210



Oribacterium sp. ACB7

HM120211



Oribacterium sp. CM12

HQ616374



Oribacterium sp. ICM51

HQ616397



Oribacterium sp. OBRC12

HQ616355



Oribacterium sp. oral taxon 078

ACIQ02000009



Oribacterium sp. oral taxon 102

GQ422713



Oribacterium sp. oral taxon 108

AFIH01000001



Orientia tsutsugamushi

AP008981



Ornithinibacillus bavariensis

NR_044923



Omithinibacillus sp. 7_10AIA

FN397526



Oscillibacter sp. G2

HM626173



Oscillibacter valericigenes

NR_074793



Oscillospira guilliermondii

AB040495



Oxalobacter formigenes

ACDQ01000020



Paenibacillus barcinonensis

NR_042272



Paenibacillus barengoltzii

NR_042756



Paenibacillus chibensis

NR_040885



Paenibacillus cookii

NR_025372



Paenibacillus durus

NR_037017



Paenibacillus glucanolyticus

D78470



Paenibacillus lactis

NR_025739



Paenibacillus lautus

NR_040882



Paenibacillus pabuli

NR_040853



Paenibacillus polymyxa

NR_037006



Paenibacillus popilliae

NR_040888



Paenibacillus sp. CIP 101062

HM212646



Paenibacillus sp. HGF5

AEXS01000095



Paenibacillus sp. HGF7

AFDH01000147



Paenibacillus sp. JC66

JF824808



Paenibacillus sp. oral taxon F45

HM099647



Paenibacillus sp. R_27413

HE586333



Paenibacillus sp. R_27422

HE586338



Paenibacillus timonensis

NR_042844



Pantoea agglomerans

AY335552



Pantoea ananatis

CP001875



Pantoea brenneri

EU216735



Pantoea citrea

EF688008



Pantoea conspicua

EU216737



Pantoea septica

EU216734



Papillibacter cinnamivorans

NR_025025



Parabacteroides distasonis

CP000140



Parabacteroides goldsteinii

AY974070



Parabacteroides gordonii

AB470344



Parabacteroides johnsonii

ABYH01000014



Parabacteroides merdae

EU136685



Parabacteroides sp. D13

ACPW01000017



Parabacteroides sp. NS31_3

JN029805



Parachlamydia sp. UWE25

BX908798



Paracoccus denitrificans

CP000490



Paracoccus marcusii

NR_044922



Paraprevotella clara

AFFY01000068



Paraprevotella xylaniphila

AFBR01000011



Parascardovia denticolens

ADEB01000020



Parasutterella excrementihominis

AFBP01000029



Parasutterella secunda

AB491209



Parvimonas micra

AB729072



Parvimonas sp. oral taxon 110

AFII01000002



Pasteurella bettyae

L06088



Pasteurella dagmatis

ACZR01000003



Pasteurella multocida

NC_002663



Pediococcus acidilactici

ACXB01000026



Pediococcus pentosaceus

NR_075052



Peptococcus niger

NR_029221



Peptococcus sp. oral clone JM048

AY349389



Peptococcus sp. oral taxon 167

GQ422727



Peptoniphilus asaccharolyticus

D14145



Peptoniphilus duerdenii

EU526290



Peptoniphilus harei

NR_026358



Peptoniphilus indolicus

AY153431



Peptoniphilus ivorii

Y07840



Peptoniphilus lacrimalis

ADDO01000050



Peptoniphilus sp. gpac007

AM176517



Peptoniphilus sp. gpac018A

AM176519



Peptoniphilus sp. gpac077

AM176527



Peptoniphilus sp. gpac148

AM176535



Peptoniphilus sp. JC140

JF824803



Peptoniphilus sp. oral taxon 386

ADCS01000031



Peptoniphilus sp. oral taxon 836

AEAA01000090


Peptostreptococcaceae bacterium ph1
JN837495



Peptostreptococcus anaerobius

AY326462



Peptostreptococcus micros

AM176538



Peptostreptococcus sp. 9succ1

X90471



Peptostreptococcus sp. oral clone AP24

AB175072



Peptostreptococcus sp. oral clone FJ023

AY349390



Peptostreptococcus sp. P4P_31 P3

AY207059



Peptostreptococcus stomatis

ADGQ01000048



Phascolarctobacterium faecium

NR_026111



Phascolarctobacterium sp. YIT 12068

AB490812



Phascolarctobacterium succinatutens

AB490811



Phenylobacterium zucineum

AY628697



Photorhabdus asymbiotica

Z76752



Pigmentiphaga daeguensis

JN585327



Planomicrobium koreense

NR_025011



Plesiomonas shigelloides

X60418


Porphyromonadaceae bacterium NML 060648
EF184292



Porphyromonas asaccharolytica

AENO01000048



Porphyromonas endodontalis

ACNN01000021



Porphyromonas gingivalis

AE015924



Porphyromonas levii

NR_025907



Porphyromonas macacae

NR_025908



Porphyromonas somerae

AB547667



Porphyromonas sp. oral clone BB134

AY005068



Porphyromonas sp. oral clone F016

AY005069



Porphyromonas sp. oral clone P2PB_52 P1

AY207054



Porphyromonas sp. oral clone P4GB_100 P2

AY207057



Porphyromonas sp. UQD 301

EU012301



Porphyromonas uenonis

ACLR01000152



Prevotella albensis

NR_025300



Prevotella amnii

AB547670



Prevotella bergensis

ACKS01000100



Prevotella bivia

ADFO01000096



Prevotella brevis

NR_041954



Prevotella buccae

ACRB01000001



Prevotella buccalis

JN867261



Prevotella copri

ACBX02000014



Prevotella corporis

L16465



Prevotella dentalis

AB547678



Prevotella denticola

CP002589



Prevotella disiens

AEDO01000026



Prevotella genomosp. C1

AY278624



Prevotella genomosp. C2

AY278625



Prevotella genomosp. P7 oral clone MB2_P31

DQ003620



Prevotella genomosp. P8 oral clone MB3_P13

DQ003622



Prevotella genomosp. P9 oral clone MB7_G16

DQ003633



Prevotella heparinolytica

GQ422742



Prevotella histicola

JN867315



Prevotella intermedia

AF414829



Prevotella loescheii

JN867231



Prevotella maculosa

AGEK01000035



Prevotella marshii

AEEI01000070



Prevotella melaninogenica

CP002122



Prevotella micans

AGWK01000061



Prevotella multiformis

AEWX01000054



Prevotella multisaccharivorax

AFJE01000016



Prevotella nanceiensis

JN867228



Prevotella nigrescens

AFPX01000069



Prevotella oralis

AEPE01000021



Prevotella oris

ADDV01000091



Prevotella oulorum

L16472



Prevotella pallens

AFPY01000135



Prevotella ruminicola

CP002006



Prevotella salivae

AB108826



Prevotella sp. BI_42

AJ581354



Prevotella sp. CM38

HQ610181



Prevotella sp. ICM1

HQ616385



Prevotella sp. ICM55

HQ616399



Prevotella sp. JCM 6330

AB547699



Prevotella sp. oral clone AA020

AY005057



Prevotella sp. oral clone ASCG10

AY923148



Prevotella sp. oral clone ASCG12

DQ272511



Prevotella sp. oral clone AU069

AY005062



Prevotella sp. oral clone CY006

AY005063



Prevotella sp. oral clone DA058

AY005065



Prevotella sp. oral clone FL019

AY349392



Prevotella sp. oral clone FU048

AY349393



Prevotella sp. oral clone FW035

AY349394



Prevotella sp. oral clone GI030

AY349395



Prevotella sp. oral clone GI032

AY349396



Prevotella sp. oral clone GI059

AY349397



Prevotella sp. oral clone GU027

AY349398



Prevotella sp. oral clone HF050

AY349399



Prevotella sp. oral clone ID019

AY349400



Prevotella sp. oral clone IDR_CEC_0055

AY550997



Prevotella sp. oral clone IK053

AY349401



Prevotella sp. oral clone IK062

AY349402



Prevotella sp. oral clone P4PB_83 P2

AY207050



Prevotella sp. oral taxon 292

GQ422735



Prevotella sp. oral taxon 299

ACWZ01000026



Prevotella sp. oral taxon 300

GU409549



Prevotella sp. oral taxon 302

ACZK01000043



Prevotella sp. oral taxon 310

GQ422737



Prevotella sp. oral taxon 317

ACQH01000158



Prevotella sp. oral taxon 472

ACZS01000106



Prevotella sp. oral taxon 781

GQ422744



Prevotella sp. oral taxon 782

GQ422745



Prevotella sp. oral taxon F68

HM099652



Prevotella sp. oral taxon G60

GU432133



Prevotella sp. oral taxon G70

GU432179



Prevotella sp. oral taxon G71

GU432180



Prevotella sp. SEQ053

JN867222



Prevotella sp. SEQ065

JN867234



Prevotella sp. SEQ072

JN867238



Prevotella sp. SEQ116

JN867246



Prevotella sp. SG12

GU561343



Prevotella sp. sp24

AB003384



Prevotella sp. sp34

AB003385



Prevotella stercorea

AB244774



Prevotella tannerae

ACIJ02000018



Prevotella timonensis

ADEF01000012



Prevotella veroralis

ACVA01000027



Prevotella jejuni, Prevotella aurantiaca,




Prevotella baroniae, Prevotella colorans,




Prevotella corporis, Prevotella dentasini,




Prevotella enoeca, Prevotella falsenii, Prevotella




fusca, Prevotella heparinolytica, Prevotella




loescheii, Prevotella multisaccharivorax,




Prevotella nanceiensis, Prevotella oryzae,




Prevotella paludivivens, Prevotella pleuritidis,




Prevotella ruminicola, Prevotella




saccharolytica, Prevotella scopos, Prevotella




shahii, Prevotella zoogleoformans



Prevotellaceae bacterium P4P_62 P1
AY207061



Prochlorococcus marinus

CP000551


Propionibacteriaceae bacterium NML 02_0265
EF599122



Propionibacterium acidipropionici

NC_019395



Propionibacterium acnes

ADJM01000010



Propionibacterium avidum

AJ003055



Propionibacterium freudenreichii

NR_036972



Propionibacterium granulosum

FJ785716



Propionibacterium jensenii

NR_042269



Propionibacterium propionicum

NR_025277



Propionibacterium sp. 434_HC2

AFIL01000035



Propionibacterium sp. H456

AB177643



Propionibacterium sp. LG

AY354921



Propionibacterium sp. oral taxon 192

GQ422728



Propionibacterium sp. S555a

AB264622



Propionibacterium thoenii

NR_042270



Proteus mirabilis

ACLE01000013



Proteus penneri

ABVP01000020



Proteus sp. HS7514

DQ512963



Proteus vulgaris

AJ233425



Providencia alcalifaciens

ABXW01000071



Providencia rettgeri

AM040492



Providencia rustigianii

AM040489



Providencia stuartii

AF008581



Pseudoclavibacter sp. Timone

FJ375951



Pseudoflavonifractor capillosus

AY136666



Pseudomonas aeruginosa

AABQ07000001



Pseudomonas fluorescens

AY622220



Pseudomonas gessardii

FJ943496



Pseudomonas mendocina

AAUL01000021



Pseudomonas monteilii

NR_024910



Pseudomonas poae

GU188951



Pseudomonas pseudoalcaligenes

NR_037000



Pseudomonas putida

AF094741



Pseudomonas sp. 2_1_26

ACWU01000257



Pseudomonas sp. G1229

DQ910482



Pseudomonas sp. NP522b

EU723211



Pseudomonas stutzeri

AM905854



Pseudomonas tolaasii

AF320988



Pseudomonas viridiflava

NR_042764



Pseudoramibacter alactolyticus

AB036759



Psychrobacter arcticus

CP000082



Psychrobacter cibarius

HQ698586



Psychrobacter cryohalolentis

CP000323



Psychrobacter faecalis

HQ698566



Psychrobacter nivimaris

HQ698587



Psychrobacter pulmonis

HQ698582



Psychrobacter sp. 13983

HM212668



Pyramidobacter piscolens

AY207056



Ralstonia pickettii

NC_010682



Ralstonia sp. 5_7_47FAA

ACUF01000076



Raoultella omithinolytica

AB364958



Raoultella planticola

AF129443



Raoultella terrigena

NR_037085



Rhodobacter sp. oral taxon C30

HM099648



Rhodobacter sphaeroides

CP000144



Rhodococcus corynebacterioides

X80615



Rhodococcus equi

ADNW01000058



Rhodococcus erythropolis

ACNO01000030



Rhodococcus fascians

NR_037021



Rhodopseudomonas palustris

CP000301



Rickettsia akari

CP000847



Rickettsia conorii

AE008647



Rickettsia prowazekii

M21789



Rickettsia rickettsii

NC_010263



Rickettsia slovaca

L36224



Rickettsia typhi

AE017197



Robinsoniella peoriensis

AF445258



Roseburia cecicola

GU233441



Roseburia faecalis

AY804149



Roseburia faecis

AY305310



Roseburia hominis

AJ270482



Roseburia intestinalis

FP929050



Roseburia inulinivorans

AJ270473



Roseburia sp. 11SE37

FM954975



Roseburia sp. 11SE38

FM954976



Roseiflexus castenholzii

CP000804



Roseomonas cervicalis

ADVL01000363



Roseomonas mucosa

NR_028857



Roseomonas sp. NML94_0193

AF533357



Roseomonas sp. NML97_0121

AF533359



Roseomonas sp. NML98_0009

AF533358



Roseomonas sp. NML98_0157

AF533360



Rothia aeria

DQ673320



Rothia dentocariosa

ADDW01000024



Rothia mucilaginosa

ACVO01000020



Rothia nasimurium

NR_025310



Rothia sp. oral taxon 188

GU470892



Ruminobacter amylophilus

NR_026450


Ruminococcaceae bacterium D16
ADDX01000083



Ruminococcus albus

AY445600



Ruminococcus bromii

EU266549



Ruminococcus callidus

NR_029160



Ruminococcus champanellensis

FP929052



Ruminococcus flavefaciens

NR_025931



Ruminococcus gnavus

X94967



Ruminococcus hansenii

M59114



Ruminococcus lactaris

ABOU02000049



Ruminococcus obeum

AY169419



Ruminococcus sp. 18P13

AJ515913



Ruminococcus sp. 5_1_39BFAA

ACII01000172



Ruminococcus sp. 9SE51

FM954974



Ruminococcus sp. ID8

AY960564



Ruminococcus sp. K_1

AB222208



Ruminococcus torques

AAVP02000002



Saccharomonospora viridis

X54286



Salmonella bongori

NR_041699



Salmonella enterica

NC_011149



Salmonella enterica

NC_011205



Salmonella enterica

DQ344532



Salmonella enterica

ABEH02000004



Salmonella enterica

ABAK02000001



Salmonella enterica

NC_011080



Salmonella enterica

EU118094



Salmonella enterica

NC_011094



Salmonella enterica

AE014613



Salmonella enterica

ABFH02000001



Salmonella enterica

ABEM01000001



Salmonella enterica

ABAM02000001



Salmonella typhimurium

DQ344533



Salmonella typhimurium

AF170176



Sarcina ventriculi

NR_026146



Scardovia inopinata

AB029087



Scardovia wiggsiae

AY278626



Segniliparus rotundus

CP001958



Segniliparus rugosus

ACZI01000025



Selenomonas artemidis

HM596274



Selenomonas dianae

GQ422719



Selenomonas flueggei

AF287803



Selenomonas genomosp. C1

AY278627



Selenomonas genomosp. C2

AY278628



Selenomonas genomosp. P5

AY341820



Selenomonas genomosp. P6 oral clone MB3_C41

DQ003636



Selenomonas genomosp. P7 oral clone MB5_C08

DQ003627



Selenomonas genomosp. P8 oral clone MB5_P06

DQ003628



Selenomonas infelix

AF287802



Selenomonas noxia

GU470909



Selenomonas ruminantium

NR_075026



Selenomonas sp. FOBRC9

HQ616378



Selenomonas sp. oral clone FT050

AY349403



Selenomonas sp. oral clone GI064

AY349404



Selenomonas sp. oral clone GT010

AY349405



Selenomonas sp. oral clone HU051

AY349406



Selenomonas sp. oral clone IK004

AY349407



Selenomonas sp. oral clone IQ048

AY349408



Selenomonas sp. oral clone JI021

AY349409



Selenomonas sp. oral clone JS031

AY349410



Selenomonas sp. oral clone OH4A

AY947498



Selenomonas sp. oral clone P2PA_80 P4

AY207052



Selenomonas sp. oral taxon 137

AENV01000007



Selenomonas sp. oral taxon 149

AEEJ01000007



Selenomonas sputigena

ACKP02000033



Serratia fonticola

NR_025339



Serratia liquefaciens

NR_042062



Serratia marcescens

GU826157



Serratia odorifera

ADBY01000001



Serratia proteamaculans

AAUN01000015



Shewanella putrefaciens

CP002457



Shigella boydii

AAKA01000007



Shigella dysenteriae

NC_007606



Shigella flexneri

AE005674



Shigella sonnei

NC_007384



Shuttleworthia satelles

ACIP02000004



Shuttleworthia sp. MSX8B

HQ616383



Shuttleworthia sp. oral taxon G69

GU432167



Simonsiella muelleri

ADCY01000105



Slackia equolifaciens

EU377663



Slackia exigua

ACUX01000029



Slackia faecicanis

NR_042220



Slackia heliotrinireducens

NR_074439



Slackia isoflavoniconvertens

AB566418



Slackia piriformis

AB490806



Slackia sp. NATTS

AB505075



Solobacterium moorei

AECQ01000039



Sphingobacterium faecium

NR_025537



Sphingobacterium mizutaii

JF708889



Sphingobacterium multivorum

NR_040953



Sphingobacterium spiritivorum

ACHA02000013



Sphingomonas echinoides

NR_024700



Sphingomonas sp. oral clone FI012

AY349411



Sphingomonas sp. oral clone FZ016

AY349412



Sphingomonas sp. oral taxon A09

HM099639



Sphingomonas sp. oral taxon F71

HM099645



Sphingopyxis alaskensis

CP000356



Spiroplasma insolitum

NR_025705



Sporobacter termitidis

NR_044972



Sporolactobacillus inulinus

NR_040962



Sporolactobacillus nakayamae

NR_042247



Sporosarcina newyorkensis

AFPZ01000142



Sporosarcina sp. 2681

GU994081


Staphylococcaceae bacterium NML 92_0017
AY841362



Staphylococcus aureus

CP002643



Staphylococcus auricularis

JQ624774



Staphylococcus capitis

ACFR01000029



Staphylococcus caprae

ACRH01000033



Staphylococcus camosus

NR_075003



Staphylococcus cohnii

JN175375



Staphylococcus condimenti

NR_029345



Staphylococcus epidermidis

ACHE01000056



Staphylococcus equorum

NR_027520



Staphylococcus fleurettii

NR_041326



Staphylococcus haemolyticus

NC_007168



Staphylococcus hominis

AM157418



Staphylococcus lugdunensis

AEQA01000024



Staphylococcus pasteuri

FJ189773



Staphylococcus pseudintermedius

CP002439



Staphylococcus saccharolyticus

NR_029158



Staphylococcus saprophyticus

NC_007350



Staphylococcus sciuri

NR_025520



Staphylococcus sp. clone bottae7

AF467424



Staphylococcus sp. H292

AB177642



Staphylococcus sp. H780

AB177644



Staphylococcus succinus

NR_028667



Staphylococcus vitulinus

NR_024670



Staphylococcus wameri

ACPZ01000009



Staphylococcus xylosus

AY395016



Stenotrophomonas maltophilia

AAVZ01000005



Stenotrophomonas sp. FG_6

EF017810



Streptobacillus moniliformis

NR_027615



Streptococcus agalactiae

AAJO01000130



Streptococcus alactolyticus

NR_041781



Streptococcus anginosus

AECT01000011



Streptococcus australis

AEQR01000024



Streptococcus bovis

AEEL01000030



Streptococcus canis

AJ413203



Streptococcus constellatus

AY277942



Streptococcus cristatus

AEVC01000028



Streptococcus downei

AEKN01000002



Streptococcus dysgalactiae

AP010935



Streptococcus equi

CP001129



Streptococcus equinus

AEVB01000043



Streptococcus gallolyticus

FR824043



Streptococcus genomosp. C1

AY278629



Streptococcus genomosp. C2

AY278630



Streptococcus genomosp. C3

AY278631



Streptococcus genomosp. C4

AY278632



Streptococcus genomosp. C5

AY278633



Streptococcus genomosp. C6

AY278634



Streptococcus genomosp. C7

AY278635



Streptococcus genomosp. C8

AY278609



Streptococcus gordonii

NC_009785



Streptococcus infantarius

ABJK02000017



Streptococcus infantis

AFNN01000024



Streptococcus intermedius

NR_028736



Streptococcus lutetiensis

NR_037096



Streptococcus massiliensis

AY769997



Streptococcus milleri

X81023



Streptococcus mitis

AM157420



Streptococcus mutans

AP010655



Streptococcus oligofermentans

AY099095



Streptococcus oralis

ADMV01000001



Streptococcus parasanguinis

AEKM01000012



Streptococcus pasteurianus

AP012054



Streptococcus peroris

AEVF01000016



Streptococcus pneumoniae

AE008537



Streptococcus porcinus

EF121439



Streptococcus pseudopneumoniae

FJ827123



Streptococcus pseudoporcinus

AENS01000003



Streptococcus pyogenes

AE006496



Streptococcus ratti

X58304



Streptococcus salivarius

AGBV01000001



Streptococcus sanguinis

NR_074974



Streptococcus sinensis

AF432857



Streptococcus sp. 16362

JN590019



Streptococcus sp. 2_1_36FAA

ACOI01000028



Streptococcus sp. 2285_97

AJ131965



Streptococcus sp. 69130

X78825



Streptococcus sp. AC15

HQ616356



Streptococcus sp. ACS2

HQ616360



Streptococcus sp. AS20

HQ616366



Streptococcus sp. BS35a

HQ616369



Streptococcus sp. C150

ACRI01000045



Streptococcus sp. CM6

HQ616372



Streptococcus sp. CM7

HQ616373



Streptococcus sp. ICM10

HQ616389



Streptococcus sp. ICM12

HQ616390



Streptococcus sp. ICM2

HQ616386



Streptococcus sp. ICM4

HQ616387



Streptococcus sp. ICM45

HQ616394



Streptococcus sp. M143

ACRK01000025



Streptococcus sp. M334

ACRL01000052



Streptococcus sp. OBRC6

HQ616352



Streptococcus sp. oral clone ASB02

AY923121



Streptococcus sp. oral clone ASCA03

DQ272504



Streptococcus sp. oral clone ASCA04

AY923116



Streptococcus sp. oral clone ASCA09

AY923119



Streptococcus sp. oral clone ASCB04

AY923123



Streptococcus sp. oral clone ASCB06

AY923124



Streptococcus sp. oral clone ASCC04

AY923127



Streptococcus sp. oral clone ASCC05

AY923128



Streptococcus sp. oral clone ASCC12

DQ272507



Streptococcus sp. oral clone ASCD01

AY923129



Streptococcus sp. oral clone ASCD09

AY923130



Streptococcus sp. oral clone ASCD10

DQ272509



Streptococcus sp. oral clone ASCE03

AY923134



Streptococcus sp. oral clone ASCE04

AY953253



Streptococcus sp. oral clone ASCE05

DQ272510



Streptococcus sp. oral clone ASCE06

AY923135



Streptococcus sp. oral clone ASCE09

AY923136



Streptococcus sp. oral clone ASCE10

AY923137



Streptococcus sp. oral clone ASCE12

AY923138



Streptococcus sp. oral clone ASCF05

AY923140



Streptococcus sp. oral clone ASCF07

AY953255



Streptococcus sp. oral clone ASCF09

AY923142



Streptococcus sp. oral clone ASCG04

AY923145



Streptococcus sp. oral clone BW009

AY005042



Streptococcus sp. oral clone CH016

AY005044



Streptococcus sp. oral clone GK051

AY349413



Streptococcus sp. oral clone GM006

AY349414



Streptococcus sp. oral clone P2PA_41 P2

AY207051



Streptococcus sp. oral clone P4PA_30 P4

AY207064



Streptococcus sp. oral taxon 071

AEEP01000019



Streptococcus sp. oral taxon G59

GU432132



Streptococcus sp. oral taxon G62

GU432146



Streptococcus sp. oral taxon G63

GU432150



Streptococcus sp. SHV515

Y07601



Streptococcus suis

FM252032



Streptococcus thermophilus

CP000419



Streptococcus uberis

HQ391900



Streptococcus urinalis

DQ303194



Streptococcus vestibularis

AEKO01000008



Streptococcus viridans

AF076036



Streptomyces albus

AJ697941



Streptomyces griseus

NR_074787



Streptomyces sp. 1 AIP_2009

FJ176782



Streptomyces sp. SD 511

EU544231



Streptomyces sp. SD 524

EU544234



Streptomyces sp. SD 528

EU544233



Streptomyces sp. SD 534

EU544232



Streptomyces thermoviolaceus

NR_027616



Subdoligranulum variabile

AJ518869



Succinatimonas hippei

AEVO01000027



Sutterella morbirenis

AJ832129



Sutterella parvirubra

AB300989



Sutterella sanguinus

AJ748647



Sutterella sp. YIT 12072

AB491210



Sutterella stercoricanis

NR_025600



Sutterella wadsworthensis

ADMF01000048



Synergistes genomosp. C1

AY278615



Synergistes sp. RMA 14551

DQ412722


Synergistetes bacterium ADV897
GQ258968


Synergistetes bacterium LBVCM1157
GQ258969


Synergistetes bacterium oral taxon 362
GU410752


Synergistetes bacterium oral taxon D48
GU430992



Syntrophococcus sucromutans

NR_036869


Syntrophomonadaceae genomosp. P1
AY341821



Tannerella forsythia

CP003191



Tannerella sp. 6_1_58FAA_CT1

ACWX01000068



Tatlockia micdadei

M36032



Tatumella ptyseos

NR_025342



Tessaracoccus sp. oral taxon F04

HM099640



Tetragenococcus halophilus

NR_075020



Tetragenococcus koreensis

NR_043113



Thermoanaerobacter pseudethanolicus

CP000924



Thermobifida fusca

NC_007333



Thermofilum pendens

X14835



Thermus aquaticus

NR_025900



Tissierella praeacuta

NR_044860



Trabulsiella guamensis

AY373830



Treponema denticola

ADEC01000002



Treponema genomosp. P1

AY341822



Treponema genomosp. P4 oral clone MB2_G19

DQ003618



Treponema genomosp. P5 oral clone MB3_P23

DQ003624



Treponema genomosp. P6 oral clone MB4_G11

DQ003625



Treponema lecithinolyticum

NR_026247



Treponema pallidum

CP001752



Treponema parvum

AF302937



Treponema phagedenis

AEFH01000172



Treponema putidum

AJ543428



Treponema refringens

AF426101



Treponema socranskii

NR_024868



Treponema sp. 6:H:D15A_4

AY005083



Treponema sp. clone DDKL_4

Y08894



Treponema sp. oral clone JU025

AY349417



Treponema sp. oral clone JU031

AY349416



Treponema sp. oral clone P2PB_53 P3

AY207055



Treponema sp. oral taxon 228

GU408580



Treponema sp. oral taxon 230

GU408603



Treponema sp. oral taxon 231

GU408631



Treponema sp. oral taxon 232

GU408646



Treponema sp. oral taxon 235

GU408673



Treponema sp. oral taxon 239

GU408738



Treponema sp. oral taxon 247

GU408748



Treponema sp. oral taxon 250

GU408776



Treponema sp. oral taxon 251

GU408781



Treponema sp. oral taxon 254

GU408803



Treponema sp. oral taxon 265

GU408850



Treponema sp. oral taxon 270

GQ422733



Treponema sp. oral taxon 271

GU408871



Treponema sp. oral taxon 508

GU413616



Treponema sp. oral taxon 518

GU413640



Treponema sp. oral taxon G85

GU432215



Treponema sp. ovine footrot

AJO10951



Treponema vincentii

ACYH01000036



Tropheryma whipplei

BX251412



Trueperella pyogenes

NR_044858



Tsukamurella paurometabola

X80628



Tsukamurella tyrosinosolvens

AB478958



Turicibacter sanguinis

AF349724



Ureaplasma parvum

AE002127



Ureaplasma urealyticum

AAYN01000002



Ureibacillus composti

NR_043746



Ureibacillus suwonensis

NR_043232



Ureibacillus terrenus

NR_025394



Ureibacillus thermophilus

NR_043747



Ureibacillus thermosphaericus

NR_040961



Vagococcus fluvialis

NR_026489



Veillonella atypica

AEDS01000059



Veillonella dispar

ACIK02000021



Veillonella genomosp. P1 oral clone MB5_P17

DQ003631



Veillonella montpellierensis

AF473836



Veillonella parvula

ADFU01000009



Veillonella sp. 3_1_44

ADCV01000019



Veillonella sp. 6_1_27

ADCW01000016



Veillonella sp. ACP1

HQ616359



Veillonella sp. AS16

HQ616365



Veillonella sp. BS32b

HQ616368



Veillonella sp. ICM51a

HQ616396



Veillonella sp. MSA12

HQ616381



Veillonella sp. NVG 100cf

EF108443



Veillonella sp. OK11

JN695650



Veillonella sp. oral clone ASCA08

AY923118



Veillonella sp. oral clone ASCB03

AY923122



Veillonella sp. oral clone ASCG01

AY923144



Veillonella sp. oral clone ASCG02

AY953257



Veillonella sp. oral clone OH1A

AY947495



Veillonella sp. oral taxon 158

AENU01000007


Veillonellaceae bacterium oral taxon 131
GU402916


Veillonellaceae bacterium oral taxon 155
GU470897



Vibrio cholerae

AAUR01000095



Vibrio fluvialis

X76335



Vibrio furnissii

CP002377



Vibrio mimicus

ADAF01000001



Vibrio parahaemolyticus

AAWQ01000116



Vibrio sp. RC341

ACZT01000024



Vibrio vulnificus

AE016796


Victivallaceae bacterium NML 080035
FJ394915



Victivallis vadensis

ABDE02000010



Virgibacillus proomii

NR_025308



Weissella beninensis

EU439435



Weissella cibaria

NR_036924



Weissella confusa

NR_040816



Weissella hellenica

AB680902



Weissella kandleri

NR_044659



Weissella koreensis

NR_075058



Weissella paramesenteroides

ACKU01000017



Weissella sp. KLDS 7.0701

EU600924



Wolinella succinogenes

BX571657


Xanthomonadaceae bacterium NML 03_0222
EU313791



Xanthomonas campestris

EF101975



Xanthomonas sp. kmd_489

EU723184



Xenophilus aerolatus

JN585329



Yersinia aldovae

AJ871363



Yersinia aleksiciae

AJ627597



Yersinia bercovieri

AF366377



Yersinia enterocolitica

FR729477



Yersinia frederiksenii

AF366379



Yersinia intermedia

AF366380



Yersinia kristensenii

ACCA01000078



Yersinia mollaretii

NR_027546



Yersinia pestis

AE013632



Yersinia pseudotuberculosis

NC_009708



Yersinia rohdei

ACCD01000071



Yokenella regensburgei

AB273739



Zimmermannella bifida

AB012592



Zymomonas mobilis

NR_074274
















TABLE 2







Exemplary Oncophilic Bacteria









Genera
Species
Tumor Association






Mycoplasma


hyorhinis

Gastric Carcinoma



Propionibacterium


Acnes

Prostate Cancer



Mycoplasma


genitalium

Prostate Cancer



Methylophilus

sp.
Prostate Cancer



Chlamydia


trachomatis

Prostate Cancer



Helicobacter


pylori

Gastric MALT



Listeria


welshimeri

Renal Cancer



Streptococcus


pneumoniae

Lymphoma and Leukemia



Haemophilus


influenzae

Lymphoma and Leukemia



Staphylococcus


aureus

Breast Cancer



Listeria


monocytogenes

Breast Cancer



Methylobacterium


radiotolerans

Breast Cancer



Shingomonas


yanoikuyae

breast Cancer



Fusobacterium

sp
Larynx cancer



Provetelis

sp
Larynx cancer



streptococcus


pneumoniae

Larynx cancer



Gemella

sp
Larynx cancer



Bordetella


Pertussis

Larynx cancer



Corumebacterium


tuberculosteraricum

Oral squamous cell carcinoma



Micrococcus


luteus

Oral squamous cell carcinoma



Prevotella


melaninogenica

Oral squamous cell carcinoma



Exiguobacterium


oxidotolerans

Oral squamous cell carcinoma



Fusobacterium


naviforme

Oral squamous cell carcinoma



Veillonella


parvula

Oral squamous cell carcinoma



Streptococcus


salivarius

Oral squamous cell carcinoma



Streptococcus


mitis/oralis

Oral squamous cell carcinoma



veillonella


dispar

Oral squamous cell carcinoma



Peptostreptococcus


stomatis

Oral squamous cell carcinoma



Streptococcus


gordonii

Oral squamous cell carcinoma



Gemella


Haemolysans

Oral squamous cell carcinoma



Gemella


morbillorum

Oral squamous cell carcinoma



Johnsonella


ignava

Oral squamous cell carcinoma



Streptococcus


parasanguins

Oral squamous cell carcinoma



Granulicatella


adiacens

Oral squamous cell carcinoma



Mycobacteria


marinum

lung infection



Campylobacter


concisus

Barrett's Esophagus



Campylobacter


rectus

Barrett's Esophagus



Oribacterium

sp
Esophageal adenocarcinoma



Catonella

sp
Esophageal adenocarcinoma



Peptostreptococcus

sp
Esophageal adenocarcinoma



Eubacterium

sp
Esophageal adenocarcinoma



Dialister

sp
Esophageal adenocarcinoma



Veillonella

sp
Esophageal adenocarcinoma



Anaeroglobus

sp
Esophageal adenocarcinoma



Megasphaera

sp
Esophageal adenocarcinoma



Atoppbium

sp
Esophageal adenocarcinoma



Solobacterium

sp
Esophageal adenocarcinoma



Rothia

sp
Esophageal adenocarcinoma



Actinomyces

sp
Esophageal adenocarcinoma



Fusobacterium

sp
Esophageal adenocarcinoma



Sneathia

sp
Esophageal adenocarcinoma



Leptotrichia

sp
Esophageal adenocarcinoma



Capnocytophaga

sp
Esophageal adenocarcinoma



Prevotella

sp
Esophageal adenocarcinoma



Porphyromonas

sp
Esophageal adenocarcinoma



Campylobacter

sp
Esophageal adenocarcinoma



Haemophilus

sp
Esophageal adenocarcinoma



Neisseria

sp
Esophageal adenocarcinoma


TM7
sp
Esophageal adenocarcinoma



Granulicatella

sp
Esophageal adenocarcinoma



Variovorax

sp
Psuedomyxoma Peritonei



Escherichia


Shigella

Psuedomyxoma Peritonei



Pseudomonas

sp
Psuedomyxoma Peritonei



Tessaracoccus

sp
Psuedomyxoma Peritonei



Acinetobacter

sp
Psuedomyxoma Peritonei



Helicobacter


hepaticus

Breast cancer



Chlamydia


psittaci

MALT lymphoma



Borrelia


burgdorferi

B cell lymphoma skin



Escherichia


Coli NC101

Colorectal Cancer



Salmonella


typhimurium

Tool



Eterococcus


faecalis

blood



Streptococcus


mitis

blood



Streptococcus


sanguis

blood



Streptococcus


anginosus

blood



Streptococcus


salvarius

blood



Staphylococcus


epidermidis

blood



Streptococcus


gallolyticus

Colorectal Cancer



Campylobacter


showae CC57C

Colorectal Cancer



Leptotrichia

sp
Colorectal Cancer









In certain embodiments, the mEVs (such as smEVs) described herein are obtained from obligate anaerobic bacteria. Examples of obligate anaerobic bacteria include gram-negative rods (including the genera of Bacteroides, Prevotella, Porphyromonas, Fusobacterium, Bilophila and Sutterella spp.), gram-positive cocci (primarily Peptostreptococcus spp.), gram-positive spore-forming (Clostridium spp.), non-spore-forming bacilli (Actinomyces, Propionibacterium, Eubacterium, Lactobacillus and Bifidobacterium spp.), and gram-negative cocci (mainly Veillonella spp.). In some embodiments, the obligate anaerobic bacteria are of a genus selected from the group consisting of Agathobaculum, Atopobium, Blautia, Burkholderia, Dielma, Longicatena, Paraclostridium, Turicibacter, and Tyzzerella.


In some embodiments, the mEVs (such as smEVs) described herein are obtained from bacterium of a genus selected from the group consisting of Escherichia, Klebsiella, Lactobacillus, Shigella, and Staphylococcus.


In some embodiments, the mEVs (such as smEVs) described herein are obtained from a species selected from the group consisting of Blautia massiliensis, Paraclostridium benzoelyticum, Dielma fastidiosa, Longicatena caecimuris, Lactococcus lactis cremoris, Tyzzerella nexilis, Hungatella effluvia, Klebsiella quasipneumoniae subsp. Similipneumoniae, Klebsiella oxytoca, and Veillonella tobetsuensis.


In some embodiments, the mEVs (such as smEVs) described herein are obtained from a Prevotella bacteria selected from the group consisting of Prevotella albensis, Prevotella amnii, Prevotella bergensis, Prevotella bivia, Prevotella brevis, Prevotella bryantii, Prevotella buccae, Prevotella buccalis, Prevotella copri, Prevotella dentalis, Prevotella denticola, Prevotella disiens, Prevotella histicola, Prevotella intermedia, Prevotella maculosa, Prevotella marshii, Prevotella melaninogenica, Prevotella micans, Prevotella multiformis, Prevotella nigrescens, Prevotella oralis, Prevotella oris, Prevotella oulorum, Prevotella pallens, Prevotella salivae, Prevotella stercorea, Prevotella tannerae, Prevotella timonensis, Prevotella jejuni, Prevotella aurantiaca, Prevotella baroniae, Prevotella colorans, Prevotella corporis, Prevotella dentasini, Prevotella enoeca, Prevotella falsenii, Prevotella fusca, Prevotella heparinolytica, Prevotella loescheii, Prevotella multisaccharivorax, Prevotella nanceiensis, Prevotella oryzae, Prevotella paludivivens, Prevotella pleuritidis, Prevotella ruminicola, Prevotella saccharolytica, Prevotella scopos, Prevotella shahii, Prevotella zoogleoformans, and Prevotella veroralis.


In some embodiments, the mEVs (such as smEVs) described herein are obtained from a strain of bacteria comprising a genomic sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity (e.g., at least 99.5% sequence identity, at least 99.6% sequence identity, at least 99.7% sequence identity, at least 99.8% sequence identity, at least 99.9% sequence identity) to the genomic sequence of the strain of bacteria deposited with the A TCC Deposit number as provided in Table 3. In some embodiments, the mEVs (such as smEVs) described herein are obtained from a strain of bacteria comprising a 16S sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity (e.g., at least 99.5% sequence identity, at least 99.6% sequence identity, at least 99.7% sequence identity, at least 99.8% sequence identity, at least 99.9% sequence identity) to the 16S sequence as provided in Table 3.









TABLE 3







Exemplary Bacterial Strains










SEQ





ID

Deposit
16S 


No.
Strain
Number
Sequence







Parabacteroides







goldsteinii Strain A










Bifidobacterium

PTA-125097





animalis ssp. lactis






Strain A









Bifidobacterium







animalis ssp. lactis






Strain B









Bifidobacterium







animalis ssp. lactis






Strain C









BlautiaMassiliensis

PTA-125134




Strain A









Prevotella Strain B

NRRL accession





Number B 50329








PrevotellaHisticola






Strain A









Prevotella







melanogenica Strain






A








Blautia Strain A
PTA-125346








Lactococcuslactis

PTA-125368





cremoris Strain A










Lactococcuslactis







cremoris Strain B










Runtinococcus

PTA-125706





gnavus strain










Tyzzerellanexilis

PTA-125707




strain









Clostridium


>S10-19-contig




symbiosum S10-19


CAGCGACGCCGCGTGAGTGAAGAAGTATTTC





GGTATGTAAAGCTCTATCAGCAGGGAAGAAA





ATGACGGTACCTGACTAAGAAGCCCCGGCTA





ACTACGTGCCAGCAGCCGCGGTAATACGTAG





GGGGCAAGCGTTATCCGGATTTACTGGGTGTA





AAGGGAGCGTAGACGGTAAAGCAAGTCTGAA





GTGAAAGCCCGCGGCTCAACTGCGGGACTGC





TTTGGAAACTGTTTAACTGGAGTGTCGGAGAG





GTAAGTGGAATTCCTAGTGTAGCGGTGAAAT





GCGTAGATATTAGGAGGAACACCAGTGGCGA





AGGCGACTTACTGGACGATAACTGACGTTGA





GGCTCGAAAGCGTGGGGAGCAAACAGGATTA





GATACCCTGGTAGTCCACGCCGTAAACGATG





AATACTAGGTGTTGGGGAGCAAAGCTCTTCG





GTGCCGTCGCAAACGCAGTAAGTATTCCACCT





GGGGAGTACGTTCGCAAGAATGAAACTCAAA





GGAATTGACGGGGACCCGCACAAGCGGTGGA





GCATGTGGTTTAATTCGAAGCAACGCGAAGA





ACCTTACCAGGTCTTGACATCGATCCGACGGG





GGAGTAACGTCCCCTTCCCTTCGGGGCGGAG





AAGACAGGTGGTGCATGGTTGTCGTCAGCTC





GTGTCGTGAGATGTTGGGTTAAGTCCCGCAAC





GAGCGCAACCCTTATTCTAAGTAGCCAGCGGT





TCGGCCGGGAACTCTTGGGAGACTGCCAGGG





ATAACCTGGAGGAAGGTGGGGATGACGTCAA





ATCATCATGCCCCTTATGATCTGGGCTACACA





CGTGCTACAATGGCGTAAACAAAGAGAAGCA





AGACCGCGAGGTGGAGCAAATCTCAAAAATA





ACGTCTCAGTTCGGACTGCAGGGTGCAACTCG





CCTGCACGAAGCTGGAATCGCTAGTAATCGC





GAATCAGAATGTCGCGGTGAATACGTTCCCG





GGTCTTGTACACACCGCCCGTCACACCATGGG





AGTCAGTAACGCCCGAAGTCAGTGACCCAAC





CGCAAGG







Clostridium


>S6-202-contig




symbiosum S6-202


GATGCAGCGACGCCGCGTGAGTGAAGAAGTA





TTTCGGTATGTAAAGCTCTATCAGCAGGGAAG





AAAATGACGGTACCTGACTAAGAAGCCCCGG





CTAACTACGTGCCAGCAGCCGCGGTAATACG





TAGGGGGCAAGCGTTATCCGGATTTACTGGGT





GTAAAGGGAGCGTAGACGGTAAAGCAAGTCT





GAAGTGAAAGCCCGCGGCTCAACTGCGGGAC





TGCTTTGGAAACTGTTTAACTGGAGTGTCGGA





GAGGTAAGTGGAATTCCTAGTGTAGCGGTGA





AATGCGTAGATATTAGGAGGAACACCAGTGG





CGAAGGCGACTTACTGGACGATAACTGACGT





TOAGGCTCGAAAGCGTGGGGAGCAAACAGGA





TTAGATACCCTGGTAGTCCACGCCGTAAACGA





TGAATACTAGGTGTTGGGGAGCAAAGCTCTTC





GGTGCCGTCGCAAACGCAGTAAGTATTCCAC





CTGGGGAGTACGTTCGCAAGAATGAAACTCA





AAGGAATTGACGGGGACCCGCACAAGCGGTG





GAGCATGTGGTTTAATTCGAAGCAACGCGAA





GAACCTTACCAGGTCTTGACATCGATCCGACG





GGGGAGTAACGTCCCCTTCCCTTCGGGGCGG





AGAAGACAGGTGGTGCATGGTTGTCGTCAGC





TCGTGTCGTGAGATGTTGGGTTAAGTCCCGCA





ACGAGCGCAACCCTTATTCTAAGTAGCCAGC





GGTTCGGCCGGGAACTCTTGGGAGACTGCCA





GGGATAACCTGGAGGAAGGTGGGGATGACGT





CAAATCATCATGCCCCTTATGATCTGGGCTAC





ACACGTGCTACAATGGCGTAAACAAAGAGAA





GCAAGACCGCGAGGTGGAGCAAATCTCAAAA





ATAACGTCTCAGTTCGGACTGCAGGCTGCAAC





TCGCCTGCACGAAGCTGGAATCGCTAGTAATC





GCGAATCAGAATGTCGCGGTGAATACGTTCC





CGGGTCTTGTACACACCGCCCGTCACACCATG





GGAGTCAGTAACGCCCGAAGTCAGTGACCCA





ACCGCAAGGAGGG







Clostridium


>consensus sequence




symbiosum S10-257


TGACTAAGAAGCCCCGGCTAACTACGTGCCA





GCAGCCGCGGTAATACGTAGGGGGCAAGCGT





TATCCGGATTTACTGGGTGTAAAGGGAGCGT





AGACGGTAAAGCAAGTCTGAAGTGAAAGCCC





GCGGCTCAACTGCGGGACTGCTTTGGAAACT





GTTTAACTGGAGTGTCGGAGAGGTAAGTGGA





ATTCCTAGTGTAGCGGTGAAATGCGTAGATAT





TAGGAGGAACACCAGTGGCGAAGGCGACTTA





CTGGACGATAACTGACGTTGAGGCTCGAAAG





CGTGGGGAGCAAACAGGATTAGATACCCTGG





TAGTCCACGCCGTAAACGATGAATACTAGGT





GTTGGGGAGCAAAGCTCTTCGGTGCCGTCGC





AAACGCAGTAAGTATTCCACCTGGGGAGTAC





GTTCGCAAGAATGAAACTCAAAGGAATTGAC





GGGGACCCGCACAAGCGGTGGAGCATGTGGT





TTAATTCGAAGCAACGCGAAGAACCTTACCA





GGTCTTGACATCGATCCGACGGGGGAGTAAC





GTCCCCTTCCCTTCGGGGCGGAGAAGACAGG





TGGTGCATGGTTGTCGTCAGCTCGTGTCGTGA





GATGTTGGGTTAAGTCCCGCAACGAGCGCAA





CCCTTATTCTAAGTAGCCAGCGGTTCGGCCGG





GAACTCTTGGGAGACTGCCAGGGATAACCTG





GAGGAAGGTGGGGATGACGTCAAATCATCAT





GCCCCTTATGATCTGGGCTACACACGTGCTAC





AATGGCGTAAACAAAGAGAAGCAAGACCGCG





AGGTGGAGCAAATCTCAAAAATAACGTCTCA





GTTCGGACTGCAGGCTGCAACTCGCCTGCACG





AAGCTGGAATCGCTAGTAATCGCGAATCAGA





ATGTCGCGGTGAATACGTTCCC







Clostridium


>10-552 consensus sequence




symbiosum S10-552


CGTATTCACCGCGACATTCTGATTCGC





GATTACTAGCGATTCCAGCTTCGTGCAGGCGA





GTTGCAGCCTGCAGTCCGAACTGAGACGTTAT





TTTTGAGATTTGCTCCACCTCGCGGTCTTGCTT





CTCTTTGTTTACGCCATTGTAGCACGTGTGTA





GCCCAGATCATAAGGGGCATGATGATTTGAC





GTCATCCCCACCTTCCTCCAGGTTATCCCTGG





CAGTCTCCCAAGAGTTCCCGGCCGAACCGCTG





GCTACTTAGAATAAGGGTTGCGCTCGTTGCGG





GACTTAACCCAACATCTCACGACACGAGCTG





ACGACAACCATGCACCACCTGTCTTCTCCGCC





CCGAAGGGAAGGGGACGTTACTCCCCCGTCG





GATCGATGTCAAGACCTGGTAAGGTTCTTCGC





GTTGCTTCGAATTAAACCACATGCTCCACCGC





TTGTGCGGGTCCCCGTCAATTCCTTTGAGTTT





CATTCTTGCGAACGTACTCCCCAGGTGGAATA





CnACTGCGTTTGCGACGGCACCGAAGAGCTT





TGCTCCCCAACACCTAGTATTCATCGTTTACG





GCGTGGACTACCAGGGTATCTAATCCTGTTTG





CTCCCCACGCTTTCGAGCCTCAACGTCAGTTA





TCGTCCAGTAAGTCGCCTTCGCCACTGGTGTT





CCTCCTAATATCTACGCATTTCACCGCTACAC





TAGGAATTCCACTTACCTCTCCGACACTCCAG





TTAAACAGTTTCCAAAGCAGTCCCGCAGTTGA





GCCGCGGGCTTTCACTTCAGACTTGCTTTACC





GTCTACGCTCCCTTTACACCCAGTAAATCCGG





ATAACGCTTGCCCCCTACGTATTACCGCGGCT





GCTGGCACGTAGTTAGCCGGGGCTTCTTAGT







Clostridium


>10-511_consensus_sequence 2




symbiosum S10-551


reads from 10-511





ACTAAGAAGCCCCGGCTAACTACGTGCCAGC





AGCCGCGGTAATACGTAGGGGGCAAGCGTTA





TCCGGATTTACTGGGTGTAAAGGGAGCGTAG





ACGGTAAAGCAAGTCTGAAGTGAAAGCCCGC





GGCTCAACTGCGGGACTGCTTTGGAAACTGTT





TAACTGGAGTGTCGGAGAGGTAAGTGGAATT





CCTAGTGTAGCGGTGAAATGCGTAGATATTA





GGAGGAACACCAGTGGCGAAGGCGACTTACT





GGACGATAACTGACGTTGAGGCTCGAAAGCG





TGGGGAGCAAACAGGATTAGATACCCTGGTA





GTCCACGCCGTAAACGATGAATACTAGGTGTT





GGGGAGCAAAGCTCTTCGGTGCCGTCGCAAA





CGCAGTAAGTATTCCACCTGGGGAGTACGTTC





GCAAGAATGAAACTCAAAGGAATTGACGGGG





ACCCGCACAAGCGGTGGAGCATGTGGTTTAA





TTCGAAGCAACGCGAAGAACCTTACCAGGTC





TTGACATCGATCCGACGGGGGAGTAACGTCC





CCTTCCCTTCGGGGCGGAGAAGACAGGTGGT





GCATGGTTGTCGTCAGCTCGTGTCGTGAGATG





TTGGGTTAAGTCCCGCAACGAGCGCAACCCTT





ATTCTAAGTAGCCAGCGGTTCGGCCGGGAAC





TCTTGGGAGACTGCCAGGGATAACCTGGAGG





AAGGTGGGGATGACGTCAAATCATCATGCCC





CTTATGATCTGGGCTACACACGTGCTACAATG





GCGTAAACAAAGAGAAGCAAGACCGCGAGGT





GGAGCAAATCTCAAAAATAACGTCTCAGTTC





GGACTGCAGGCTGCAACTCGCCTGCACGAAG





CTGGAATCGCTAGTAATCGCGAATCAGAATG





TCGCGGTGAATACGTTCCC







Clostridium


>10-530




symbiosum S10-530


GAAAATGACGGTACCTGACTAAGAAGCCC





CGGCTAACTACGTGCCAGCAGCCGCGGTAAT





ACGTAGGGGGCAAGCGTTATCCGGATTTACT





GGGTGTAAAGGGAGCGTAGACGGTAAAGCAA





GTCTGAAGTGAAAGCCCGCGGCTCAACTGCG





GGACTGCTTTGGAAACTGTTTAACTGGAGTGT





CGGAGAGGTAAGTGGAATTCCTAGTGTAGCG





GTGAAATGCGTAGATATTAGGAGGAACACCA





GTGGCGAAGGCGACTTACTGGACGATAACTG





ACGTTGAGGCTCGAAAGCGTGGGGAGCAAAC





AGGATTAGATACCCTGGTAGTCCACGCCGTA





AACGATGAATACTAGGTGTTGGGGAGCAAAG





CTCTTCGGTGCCGTCGCAAACGCAGTAAGTAT





TCCACCTGGGGAGTACGTTCGCAAGAATGAA





ACTCAAAGGAATTGACGGGGACCCGCACAAG





CGGTGGAGCATGTGGTTTAATTCGAAGCAAC





GCGAAGAACCTTACCAGGTCTTGACATCGATC





CGACGGGGGAGTAACGTCCCCTTCCCTTCGGG





GCGGA







Clostridium


>10-533_consensus_sequence 2




symbiosum S10-533


reads from 10-533





GAACGTATTCACCGCGACATTCTGATTCGCGA





TTACTAGCGATTCCAGCTTCGTGCAGGCGAGT





TGCAGCCTGCAGTCCGAACTGAGACGTTATTT





TTGAGATTTGCTCCACCTCGCGGTCTTGCTT





CTCTTTGTTTACGCCATTGTAGCACGTGTGTA





GCCCAGATCATAAGGGGCATGATGATTTGAC





GTCATCCCCACCTTCCTCCAGGTTATCCCTGG





CAGTCTCCCAAGAGTTCCCGGCCGAACCGCTG





GCTACTTAGAATAAGGGTTGCGCTCGTTGCGG





GACTTAACCCAACATCTCACGACACGAGCTG





ACGACAACCATGCACCACCTGTCTTCTCCGCC





CCGAAGGGAAGGGGACGTTACTCCCCCGTCG





GATCGATGTCAAGACCTGGTAAGGTTCTTCGC





GTTGCTTCGAATTAAACCACATGCTCCACCGC





TTGTGCGGGTCCCCGTCAATTCCTTTGAGTTT





CATTCTTGCGAACGTACTCCCCAGGTGGAATA





CTTACTGCGTTTGCGACGGCACCGAAGAGCTT





TGCTCCCCAACACCTAGTATTCATCGTTTACG





GCGTGGACTACCAGGGTATCTAATCCTGTTTG





CTCCCCACGCTTTCGAGCCTCAACGTCAGTTA





TCGTCCAGTAAGTCGCCTTCGCCACTGGTGTT





CCTCCTAATATCTACGCATTTCACCGCTACAC





TAGGAATTCCACTTACCTCTCCGACACTCCAG





TTAAACAGTTTCCAAAGCAGTCCCGCAGTTGA





GCCGCGGGCTTTCACTTCAGACTTGCTTTACC





GTCTACGCTCCCTTTACACCCAGTAAATCCGG





ATAACGCTTGCCCCCTACGTATTACCGCGGCT





GCTGGCACGTAGTTAGCCGGGGCTTCTTAG







Clostridium


>10-537_consensus_sequence 2




symbiosum S10-537


reads from 10-537





ACTAAGAAGCCCCGGCTAACTACGTGCCA





GCAGCCGCGGTAATACGTAGGGGGCAAGCGT





TATCCGGATTTACTGGGTGTAAAGGGAGCGT





AGACGGTAAAGCAAGTCTGAAGTGAAAGCCC





GCGGCTCAACTGCGGGACTGCTTTGGAAACT





GTTTAACTGGAGTGTCGGAGAGGTAAGTGGA





ATTCCTAGTGTAGCGGTGAAATGCGTAGATAT





TAGGAGGAACACCAGTGGCGAAGGCGACTTA





CTGGACGATAACTGACGTTGAGGCTCGAAAG





CGTGGGGAGCAAACAGGATTAGATACCCTGG





TAGTCCACGCCGTAAACGATGAATACTAGGT





GTTGGGGAGCAAAGCTCTTCGGTGCCGTCGC





AAACGCAGTAAGTATTCCACCTGGGGAGTAC





GTTCGCAAGAATGAAACTCAAAGGAATTGAC





GGGGACCCGCACAAGCGGTGGAGCATGTGGT





TTAATTCGAAGCAACGCGAAGAACCTTACCA





GGTCTTGACATCGATCCGACGGGGGAGTAAC





GTCCCCTTCCCTTCGGGGCGGAGAAGACAGG





TGGTGCATGGTTGTCGTCAGCTCGTGTCGTGA





GATGTTGGGTTAAGTCCCGCAACGAGCGCAA





CCCTTATTCTAAGTAGCCAGCGGTTCGGCCGG





GAACTCTTGGGAGACTGCCAGGGATAACCTG





GAGGAAGGTGGGGATGACGTCAAATCATCAT





GCCCCTTATGATCTGGGCTACACACGTGCTAC





AATGGCGTAAACAAAGAGAAGCAAGACCGCG





AGGTGGAGCAAATCTCAAAAATAACGTCTCA





GTTCGGACTGCAGGCTGCAACTCGCCTGCACG





AAGCTGGAATCGCTAGTAATCGCGAATCAGA





ATGTCGCGGTGAATACGTT







Clostridium


>10-544




symbiosum S10-544


ATGACGGTACCTGACTAAGAAGCCCCGGC





TAACTACGTGCCAGCAGCCGCGGTAATACGT





AGGGGGCAAGCGTTATCCGGATTTACTGGGT





GTAAAGGGAGCGTAGACGGTAAAGCAAGTCT





GAAGTGAAAGCCCGCGGCTCAACTGCGGGAC





TGCTTTGGAAACTGTTTAACTGGAGTGTCGGA





GAGGTAAGTGGAATTCCTAGTGTAGCGGTGA





AATGCGTAGATATTAGGAGGAACACCAGTGG





CGAAGGCGACTTACTGGACGATAACTGACGT





TGAGGCTCGAAAGCGTGGGGAGCAAACAGGA





TTAGATACCCTGGTAGTCCACGCCGTAAACGA





TGAATACTAGGTGTTGGGGAGCAAAGCTCTTC





GGTGCCGTCGCAAACGCAGTAAGTATTCCAC





CTGGGGAGTACGTTCGCAAGAATGAAACTCA





AAGGAATTGACGGGGACCCGCACAAGCGGTG





GAGCATGTGGTTTAATTCGAAGCAACGCGAA





GAACCTTACCAGGTCTTGACATCGATCCGACG





GGGGAGTAACGTCCCCTTCCCTTCGGGGCGG





AGAAGACAGGTGGTGCATGGTTGTCGTCAGC





TCGTGTCGTGAGATGTTGGGTTAAGTCCCGCA





ACGAGCGCAACCCTTATTCTAAGTAGCCAGC





GGTTCGGCCGGGAACTCTTGGGAGACTGCCA





GGGATAACCTG







Clostridium


>10-547




symbiosum S10-547


GGGAAGAAAATGACGGTACCTGACTAAGA





AGCCCCGGCTAACTACGTGCCAGCAGCCGCG





GTAATACGTAGGGGGCAAGCGTTATCCGGAT





TTACTGGGTGTAAAGGGAGCGTAGACGGTAA





AGCAAGTCTGAAGTGAAAGCCCGCGGCTCAA





CTGCGGGACTGCTTTGGAAACTGTTTAACTGG





AGTGTCGGAGAGGTAAGTGGAATTCCTAGTG





TAGCGGTGAAATGCGTAGATATTAGGAGGAA





CACCAGTGGCGAAGGCGACTTACTGGACGAT





AACTGACGTTGAGGCTCGAAAGCGTGGGGAG





CAAACAGGATTAGATACCCTGGTAGTCCACG





CCGTAAACGATGAATACTAGGTGTTGGGGAG





CAAAGCTCTTCGGTGCCGTCGCAAACGCAGT





AAGTATTCCACCTGGGGAGTACGTTCGCAAG





AATGAAACTCAAAGGAATTGACGGGGACCCG





CACAAGCGGTGGAGCATGTGGTTTAATTCGA





AGCAACGCGAAGAACCTTACCAGGTCTTGAC





ATCGATCCGACGGGGGAGTAACGTCCCCTTCC





CTTCGGGGCGGAGAAGACAGGTGGTGCATGG





TTGTCGTCAGCTCGTGTCGTGAGATGTTGGGT





TAAGTCCCGCAACGAGCGCAACCCTTATTCTA





AGTAGCCAGCGGTTCGGCCGGGAACTC







Clostridium


>10-548_consensus_sequence 2




symbiosum S10-548


reads from 10-548





AAGAAGCCCCGGCTAACTACGTGCCAGCA





GCCGCGGTAATACGTAGGGGGCAAGCGTTAT





CCGGATTTACTGGGTGTAAAGGGAGCGTAGA





CGGTAAAGCAAGTCTGAAGTGAAAGCCCGCG





GCTCAACTGCGGGACTGCTTTGGAAACTGTTT





AACTGGAGTGTCGGAGAGGTAAGTGGAATTC





CTAGTGTAGCGGTGAAATGCGTAGATATTAG





GAGGAACACCAGTGGCGAAGGCGACTTACTG





GACGATAACTGACGTTGAGGCTCGAAAGCGT





GGGGAGCAAACAGGATTAGATACCCTGGTAG





TCCACGCCGTAAACGATGAATACTAGGTGTTG





GGGAGCAAAGCTCTTCGGTGCCGTCGCAAAC





GCAGTAAGTATTCCACCTGGGGAGTACGTTCG





CAAGAATGAAACTCAAAGGAATTGACGGGGA





CCCGCACAAGCGGTGGAGCATGTGGTTTAATT





CGAAGCAACGCGAAGAACCTTACCAGGTCTT





GACATCGATCCGACGGGGGAGTAACGTCCCC





TTCCCTTCGGGGCGGAGAAGACAGGTGGTGC





ATGGTTGTCGTCAGCTCGTGTCGTGAGATGTT





GGGTTAAGTCCCGCAACGAGCGCAACCCTTA





TTCTAAGTAGCCAGCGGTTCGGCCGGGAACTC





TTGGGAGACTGCCAGGGATAACCTGGAGGAA





GGTGGGGATGACGTCAAATCATCATGCCCCTT





ATGATCTGGGCTACACACGTGCTACAATGGC





GTAAACAAAGAGAAGCAAGACCGCGAGGTG





GAGCAAATCTCAAAAATAACGTCTCAGTTCG





GACTGCAGGCTGCAACTCGCCTGCACGAAGC





TGGAATCGCTAGTAATCGCGAATCAGAATGT





CGCGGTGAATACGTT







Clostridium sp. S7-


>S7-203-357F



203

TGATGCAGCGACGCCGCGTGAGTGAAGAAGT





ATTTCGGTATGTAAAGCTCTATCAGCAGGGAA





GAAAATGACGGTACCTGACTAAGAAGCCCCG





GCTAACTACGTGCCAGCAGCCGCGGTAATAC





GTAGGGGGCAAGCGTTATCCGGATTTACTGG





GTGTAAAGGGAGCGTAGACGGTAAAGCAAGT





CTGAAGTGAAAGCCCGCGGCTCAACTGCGGG





ACTGCTTTGGAAACTGTTTAACTGGAGTGTCG





GAGAGGTAAGTGGAATTCCTAGTGTAGCGGT





GAAATGCGTAGATATTAGGAGGAACACCAGT





GGCGAAGGCGACTTACTGGACGATAACTGAC





GTTGAGGCTCGAAAGCGTGGGGAGCAAACAG





GATTAGATACCCTGGTAGTCCACGCCGTAAAC





GATGAATACTAGGTGTTGGGGAGCAAAGCTC





TTCGGTGCCGTCGCAAACGCAGTAAGTATTCC





ACCTGGGGAGTACGTTCGCAAGAATGAAACT





CAAAGGAATTGACGGGGACCCGCACAAGCGG





TGGAGCATGTGGTTTAATTCGAAGCAACGCG





AAGAACCTTACCAGGTCTTGACATCGATCCGA





CGGGGGAGTAACGTCCCCTTCCCTTCGGGGCG





GAGAAGACAGGTGGTGCATGGTTGTCGTCAG





CTCGTGTCGTGAGATGTTGGGTTAAGTCCCGC





AACGAGCGCAACCCTTATTCTAAGTAGCCAG





CGGTTCGGCCGGGAACTCTTGGGAGACTGCC





AGGGATAACCTGGAGGAAGGTGGGGATGACG





TCAAATCATCATGCCCCT







Clostridium sp.


GCCGCGTGAGTGAAGAAGTATTTCGGTATGT



36A7-1014

AAAGCTCTATCAGCAGGGAAGAAAATGACGG





TACCTGACTAAGAAGCCCCGGCTAACTACGT





GCCAGCAGCCGCGGTAATACGTAGGGGGCAA





GCGTTATCCGGATTTACTGGGTGTAAAGGGA





GCGTAGACGGTAAAGCAAGTCTGAAGTGAAA





GCCCGCGGCTCAACTGCGGGACTGCTTTGGA





AACTGTTTAACTGGAGTGTCGGAGAGGTAAG





TGGAATTCCTAGTGTAGCGGTGAAATGCGTA





GATATTAGGAGGAACACCAGTGGCGAAGGCG





ACTTACTGGACGATAACTGACGTTGAGGCTCG





AAAGCGTGGGGAGCAAACAGGATTAGATACC





CTGGrAGtCCACGCCGTAAACGATGAATACT





AGGTGTTGGGGAGCAAAGCTCTTCGGTGCCG





TCGCAAACGCAGTAAGTATTCCACCTGGGGA





GTACGTTCGCAAGAATGAAACTCAAAGGAAT





TGACGGGGACCCGCACAAGCGGTGGAGCATG





TGGTTTAATTCGAAGCAACGCGAAGAACCTT





ACCAGGTCTTGACATCGATCCGACGGGGGAG





TAACGTCCCCTTCCCTTCGGGGCGGAGAAGAC





AGGTGGTGCATGGTTGTCGTCAGCTCGTGTCG





TGAGATGTTGGGTTAAGTCCCGCAACQAGCG





CAACCCTTATTCTAAGTAGCCAGCGGTTC







Clostridium sp


>4-31-contig



S4-31

GCCTGATGCAGCGACGCCGCGTGAGTGAAGA





AGTATTTCGGTATGTAAAGCTCTATCAGCAGG





GAAGAAAATGACGGTACCTGACTAAGAAGCC





CCGGCTAACTACGTGCCAGCAGCCGCGGTAA





TACGTAGGGGGCAAGCGTTATCCGGATTTACT





GGGTGTAAAGGGAGCGTAGACGGTAAAGCAA





GTCTGAAGTGAAAGCCCGCGGCTCAACTGCG





GGACTGCTTTGGAAAdGTTTAACTGGAGTGT





CGGAGAGGTAAGTGGAATTCCTAGTGTAGCG





GTGAAATGCGTAGATATTAGGAGGAACACCA





GTGGCGAAGGCGACTTACTGGACGATAACTG





ACGTTGAGGCTCGAAAGCGTGGGGAGCAAAC





AGGATTAGATACCCTGGTAGTCCACGCCGTA





AACGATGAATACTAGGTGTTGGGGAGCAAAG





CTCTTCGGTGCCGTCGCAAACGCAGTAAGTAT





TCCACCTGGGGAGTACGTTCGCAAGAATGAA





ACTCAAAGGAATTGACGGGGACCCGCACAAG





CGGTGGAGCATGTGGTTTAATTCGAAGCAAC





GCGAAGAACCTTACCAGGTCTTGACATCGATC





CGACGGGGGAGTAACGTCCCCTTCCCTTCGGG





GCGGAGAAGACAGGTGGTGCATGGTTGTCGT





CAGCTCGTGTCGTGAGATGTTGGGTTAAGTCC





CGCAACGAGCGCAACCCTTATTCTAAGTAGCC





AGCGGTTCGGCCGGGAACTCTTGGGAGACTG





CCAGGGATAACCTGGAGGAAGGTGGGGATGA





CGTCAAATCATCATGCCCCTTATGATCTGGGC





TACACACGTGCTACAATGGCGTAAACAAAGA





GAAGCAAGACCGCGAGGTGGAGCAAATCTCA





AAAATAACGTCTCAGTTCGGACTGCAGGCTG





CAACTCGCCTGCACGAAGCTGGAATCGCTAG





TAATCGCGAATCAGAATGTCGCGGTGAATAC





GTTCCCGGGTCTTGTACACACCGCCCGTCACA





CCATGGGAGTCAGTAACGCCCGAAGTCAGTG





ACCCAACCGCAAGGAGGGAGCTG







Clostridium sp


>210-133-Contig



S210-133

TTCGGTATGTAAAGCTCTATCAGCAGGGAAG





AAAATGACGGTACCTGACTAAGAAGCCCCGG





CTAACTACGTGCCAGCAGCCGCGGTAATACG





TAGGGGGCAAGCGTTATCCGGATTTACTGGGT





GTAAAGGGAGCGTAGACGGTAAAGCAAGTCT





GAAGTGAAAGCCCGCGGCTCAACTGCGGGAC





TGCTTTGGAAACTGTTTAACTGGAGTGTCGGA





GAGGTAAGTGGAATTCCTAGTGTAGCGGTGA





AATGCGTAGATATTAGGAGGAACACCAGTGG





CGAAGGCGACTTACTGGACGATAACTGACGT





TGAGGCTCGAAAGCGTGGGGAGCAAACAGGA





TTAGATACCCTGGTAGTCCACGCCGTAAACGA





TGAATACTAGGTGTTGGGGAGCAAAGCTCTTC





GGTGCCGTCGCAAACGCAGTAAGTATTCCAC





CTGGGGAGTACGTTCGCAAGAATGAAACTCA





AAGGAATTGACGGGGACCCGCACAAGCGGTG





GAGCATGTGGTTTAATTCGAAGCAACGCGAA





GAACCTTACCAGGTCTTGACATCGATCCGACG





GGGGAGTAACGTCCCCTTCCCTTCGGGGCGG





AGAAGACAGGTGGTGCATGGTTGTCGTCAGC





TCGTGTCGTGAGATGTTGGGTTAAGTCCCGCA





ACGAGCGCAACCCTTATTCTAAGTAGCCAGC





GGTTCGGCCGGGAACTCTTGGGAGACTGCCA





GGGATAACCTGGAGGAAGGTGGGGGATGACG





TCAAATCATCATGCCCCTTATGATCTGGGCTA





CACACGTGCTACAATGGCGTAAACAAAGAGA





AGCAAGACCGCGAGGTGGAGCAAATCTCAAA





AATAACGTCTCAGTTCGGACTGCAGGCTGCA





ACTCGCCTGCACGAAGCTGGAATCGCTAGTA





ATCGCGAATCAGAATGTCGCGGTGAATACGT





TCCCGGGTCTTGTACACACCGCCCGTCACACC





ATGGGAGTCAGTAACGCCCGAAGTCAGTGAC





CCA







Clostridium


>10-534_consensus_sequence 2




symbiosum S10-534


reads from 10-534





ACTAAGAAGCCCCGGCTAACTACGTGCCA





GCAGCCGCGGTAATACGTAGGGGGCAAGCGT





TATCCGGATTTACTGGGTGTAAAGGGAGCGT





AGACGGTAAAGCAAGTCTGAAGTGAAAGCCC





GCGGCTCAACTGCGGGACTGCTTTGGAAACT





GTTTAACTGGAGTGTCGGAGAGGTAAAGTGG





AATTCCTAGTGTAGCGGTGAAATGCGTAGAT





ATTAGGAGGAACACCAGTGGCGAAGGCGACT





TACTGGACGATAACTGACGTTGAGGCTCGAA





AGCGTGGGGAGCAAACAGGATTAGATACCCT





GGTAGTCCACGCCGTAAACGATGAATACTAG





GTGTTGGGGAGCAAAGCTCTTCGGTGCCGTCG





CAAACGCAGTAAGTATTCCACCTGGGGAGTA





CGTTCGCAAGAATGAAACTCAAAGGAATTGA





CGGGGACCCGCACAAGCGGTGGAGCATGTGG





TTTAATTCGAAGCAACGCGAAGAACCTTACC





AGGTCTTGACATCGATCCGACGGGGGAGTAA





CGTCCCCTTCCCTTCGGGGCGGAGAAGACAG





GTGGTGCATGGTTGTCGTCAGCTCGTGTCGTG





AGATGTTGGGTTAAGTCCCGCAACGAGCGCA





ACCCTTATTCTAAGTAGCCAGCGGTTCGGCCG





GGAACTCTTGGGAGACTGCCAGGGATAACCT





GGAGGAAGGTGGGGATGACGTCAAATCATCA





TGCCCCTTATGATCTGGGCTACACACGTGCTA





CAATGGCGTAAACAAAGAGAAGCAAGACCGC





GAGGTGGAGCAAATCTCAAAAATAACGTCTC





AGTTCGGACTGCAGGCTGCAACTCGCCTGCAC





GAAGCTGGAATCGCTAGTAATCGCGAATCAG





AATGTCGCGGTGAATACGTTCC







Clostridium sp. S4-


>4-44-contig



44

CTGATGCAGCGACGCCGCGTGAGTGAAGAAG





TAGTTTCGGTATGTAAAGCTCTATCAGCAGGG





AAGAAAATGACGGTACCTGACTAAGAAGCCC





CGGCTAACTACGTGCCAGCAGCCGCGGTAAT





ACGTAGGGGGCAAGCGTTATCCGGATTTACT





GGGTGTAAAGGGAGCGTAGACGGTAAAGCAA





GTCTGAAGTGAAAGCCCGCGGCTCAACTGCG





GGACTGCTTTGGAAACTGTTTAACTGGAGTGT





CGGAGAGGTAAGTGGAATTCCTAGTGTAGCG





GTGAAATGCGTAGATATTAGGAGGAACACCA





GTGGCGAAGGCGACTTACTGGACGATAACTG





ACGTTGAGGCTCGAAAGCGTGGGGAGCAAAC





AGGATTAGATACCCTGGTAGTCCACGCCGTA





AACGATGAATACTAGGTGTTGGGGAGCAAAG





CTCTTCGGTGCCGTCGCAAACGCAGTAAGTAT





TCCACCTGGGGAGTACGTTCGCAAGAATGAA





ACTCAAAGGAATTGACGGGGACCCGCACAAG





CGGTGGAGCATGTGGTTTAATTCGAAGCAAC





GCGAAGAACCTTACCAGGTCTTGACATCGATC





CGACGGGGGAGTAACGTCCCCTTCCCTTCGGG





GCGGAGAAGACAGGTGGTGCATGGTTGTCGT





CAGCTCGTGTCGTGAGATGTTGGGTTAAGTCC





CGCAACGAGCGCAACCCTTATTCTAAGTAGCC





AGCGGTTCGGCCGGGAACTCTTGGGAGACTG





CCAGGGATAACCTGGAGGAAGGTGGGGGATG





ACGTCAAATCATCATGCCCCTTATGATCTGGG





CTACACACGTGCTACAATGGCGTAAACAAAG





AGAAGCAAGACCGCGAGGTGGAGCAAATCTC





AAAAATAACGTCTCAGTTCGGACTGCAGGCT





GCAACTCGCCTGCACGAAGCTGGAATCGCTA





GTAATCGCGAATCAGAATGTCGCGGTGAATA





CGTTCCCGGGTCTTGTACACACCGCCCGTCAC





ACCATGGGAGTCAGTAACGCCCGAAGTCAGT





GACCCAACCGCAAGGAGGGAGCTGCCGA







Hungatella


GAAGTATTTCGGTATGTAAAGCTCTATCAGCA




hathewayi or


GGGAAGAAAATGACGGTACCTGACTAAGAAG



[Clostridium]

CCCCGGCTAACTACGTGCCAGCAGCCGCGGT




hathewayi 34D2-


AATACGTAGGGGGCAAGCGTTATCCGGATTT



1004

ACTGGGTGTAAAGGGAGCGTAGACGGTTTAG





CAAGTCTGAAGTGAAAGCCCGGGGCTCAACC





CCGGTACTGCTTTGGAAACTGTTAGACTTGAG





TGCAGGAGAGGTAAGTGGAATTCCTAGTGTA





GCGGTGAAATGCGTAGATATTAGGAGGAACA





CCAGTGGCGAAGGCGGCTTACTGGACTGTAA





CTGACGTTGAGGCTCGAAAGCGTGGGGAGCA





AACAGGATTAGATACCCTGGTAGTCCACGCC





GTAAACGATGAATACTAGGTGTCGGGGGGCA





AAGCCCTTCGGTGCCGCCGCAAACGCAATAA





GTATTCCACCTGGGGAGTACGTTCGCAAGAAT





GAAACTCAAAGGAATTGACGGGGACCCGCAC





AAGCGGTGGAGCATGTGGTTTAATTCGAAGC





AACGCGAAGAACCTTACCAAGTCTTGACATC







Hungatella


TTCGGTATGTAAAGCTCTATCAGCAGGGAAG




hathewayi or


AAAATGACGGTACCTGACTAAGAAGCCCCGG



[Clostridium]

CTAACTACGTGCCAGCAGCCGCGGTAATACG




hathewayi 34H6-


TAGGGGGCAAGCGTTATCCGGATTTACTGGGT



1004

GTAAAGGGAGCGTAGACGGTTTAGCAAGTCT





GAAGTGAAAGCCCGGGGCTCAACCCCGGTAC





TGCTTTGGAAACTGTTAGACTTGAGTGCAGGA





GAGGTAAGTGGAATTCCTAGTGTAGCGGTGA





AATGCGTAGATATTAGGAGGAACACCAGTGG





CGAAGGCGGCTTACTGGACTGTAACTGACGTT





GAGGCTCGAAAGCGTGGGGAGCAAACAGGAT





TAGATACCCTGGTAGTCCACGCCGTAAACGAT





GAATACTAGGTGTCGGGGGGCAAAGCCCTTC





GGTGCCGCCGCAAACGCAATAAGTATTCCAC





CTGGGGAGTACGTTCGCAAGAATGAAACTCA





AAGGAATTGACGGGGACCCGCACAAGCGGTG





GAGCATGTGGTTTAATTCGAAGCAACGCGAA





GAACCTTACCAAGTCTTGACATCCCA







Hungatellaeffluvia


GCCGCGTGAGTGAAGAAGTATTTCGGTATGT



36B10-1014

AAAGCTCTATCAGCAGGGAAGAAAATGACGG





TACCTGACTAAGAAGCCCCGGCTAACTACGT





GCCAGCAGCCGCGGTAATACGTAGGGGGCAA





GCGTTATCCGGATTTACTGGGTGTAAAGGGA





GCGTAGACGGTTAAGCAAGTCTGAAGTGAAA





GCCCGGGGCTCAACCCCGGTACTGCTTTGGAA





ACTGTTTGACTTGAGTGCAGGAGAGGTAAGT





GGAATTCCTAGTGTAGCGGTGAAATGCGTAG





ATATTAGGAGGAACACCAGTGGCGAAGGCGG





CTTACTGGACTGTAACTGACGTTGAGGCTCGA





AAGCGTGGGGAGCAAACAGGATTAGATACCC





TGGTAGTCCACGCCGTAAACGATGAATACTA





GGTGTCGGGGGACAAAGTCCTTCGGTGCCGC





CGCTAACGCAATAAGTATTCCACCTGGGGAG





TACGTTCGCAAGAATGAAACTCAAAGGAATT





GACGGGGACCCGCACAAGCGGTGGAGCATGT





GGTTTAATTCGAAGCAACGCGAAGAACCTTA





CCAAGTCTTGACATCCCATTGAAAATCATTTA





ACCG







Hungatellaeffluvia


GCCGCGTGAGTGAAGAAGTATTTCGGTATGT



36C4-1014

AAAGCTCTATCAGCAGGGAAGAAAATGACGG





TACCTGACTAAGAAGCCCCGGCTAACTACGT





GCCAGCAGCCGCGGTAATACGTAGGGGGCAA





GCGTTATCCGGATTTACTGGGTGTAAAGGGA





GCGTAGACGGTTAAGCAAGTCTGAAGTGAAA





GCCCGGGGCTCAACCCCGGTACTGCTTTGGAA





ACTGTTTGACTTGAGTGCAGGAGAGGTAAGT





GGAATTCCTAGTGTAGCGGTGAAATGCGTAG





ATATTAGGAGGAACACCAGTGGCGAAGGCGG





CTTACTGGACTGTAACTGACGTTGAGGCTCGA





AAGCGTGGGGAGCAAACAGGATTAGATACCC





TGGTAGTCCACGCCGTAAACGATGAATACTA





GGTGTCGGGGGACAAAGTCCTTCGGTGCCGC





CGCTAACGCAATAAGTATTCCACCTGGGGAG





TACGTTCGCAAGAATGAAACTCAAAGGAATT





GACGGGGACCCGCACAAGCGGTGGAGCATGT





GGTTTAATTCGAAGCAACGCGAAGAACCTTA





CCAAGTCTTGACATCCCATTGAAAA







Hungatellaeffluvii


GCCGCGTGAGTGAAGAAGTATTTCGGTATGT



36F7-1014

AAAGCTCTATCAGCAGGGAAGAAAATGACGG





TACCTGACTAAGAAGCCCCGGCTAACTACGT





GCCAGCAGCCGCGGTAATACGTAGGGGGCAA





GCGTTATCCGGATTTACTGGGTGTAAAGGGA





GCGTAGACGGTTAAGCAAGTCTGAAGTGAAA





GCCCGGGGCTCAACCCCGGTACTGCTTTGGAA





ACTGTTTGACTTGAGTGCAGGAGAGGTAAGT





GGAATTCCTAGTGTAGCGGTGAAATGCGTAG





ATATTAGGAGGAACACCAGTGGCGAAGGCGG





CTTACTGGACTGTAACTGACGTTGAGGCTCGA





AAGCGTGGGGAGCAAACAGGATTAGATACCC





TGGTAGTCCACGCCGTAAACGATGAATACTA





GGTGTCGGGGGACAAAGTCCTTCGGTGCCGC





CGCTAACGCAATAAGTATTCCACCTGGGGAG





TACGTTCGCAAGAATGAAACTCAAAGGAATT





GACGGGGACCCGCACAAGCGGTGGAGCATGT





GGTTTAATTCGAAGCAACGCGAAGAACCTTA





CCAAGTCTTGACATCCCATTGAA







Lachnospiraceac sp


GACGGTACCTGACTAAGAAGCCCCGGCTAAC



or [Clostridium]

TACGTGCCAGCAGCCGCGGTAATACGTAGGG




Citroniae 39A7-


GGCAAGCGTTATCCGGATTTACTGGGTGTAAA



1014

GGGAGCGTAGACGGCGAAGCAAGTCTGGAGT





GAAAACCCAGGGCTCAACCCTGGGACTGCTT





TGGAAACTGTTTTGCTAGAGTGTCGGAGAGGT





AAGTGGAATTCCTAGTGTAGCGGTGAAATGC





GTAGATATTAGGAGGAACACCAGTGGCGAAG





GCGGCTTACTGGACGATAACTGACGTTGAGG





CTCGAAAGCGTGGGGAGCAAACAGGATTAGA





TACCCTGGTAGTCCACGCCGTAAACGATGAAT





GCTAGGTGTTGGGGGG







Lachnospiraceae sp


GACGGTACCTGACTAAGAAGCCCCGGCTAAC



or [Clostridium]

TACGTGCCAGCAGCCGCGGTAATACGTAGGG




citroniae 39A8-1014


GGCAAGCGTTATCCGGATTTACTGGGTGTAAA





GGGAGCGTAGACGGCGAAGCAAGTCTGGAGT





GAAAACCCAGGGCTCAACCCTGGGACTGCTT





TGGAAACTGTTTTGCTAGAGTGTCGGAGAGGT





AAGTGGAATTCCTAGTGTAGCGGTGAAATGC





GTAGATATTAGGAGGAACACCAGTGGCGAAG





GCGGCTTACTGGACGATAACTGACGTTGAGG





CTCGAAAGCGTGGGGAGCAAACAGGATTAGA





TACCCTGGTAGTCCACGCCGTAAACGATGAAT





GCTAGGTGTTGGGGGG







Lachnospiraceae sp


GCCGCGTGAGTGAAGAAGTATTTCGGTATGT



or [Clostridium]

AAAGCTCTATCAGCAGGGAAGAAACTGACGG




citroniae 36A6-1014


TACCTGACTAAGAAGCCCCGGCTAACTACGT





GCCAGCAGCCGCGGTAATACGTAGGGGGCAA





GCGTTATCCGGATTTACTGGGTGTAAAGGGA





GCGTAGACGGCGAAGCAAGTCTGGAGTGAAA





ACCCAGGGCTCAACCCTGGGACTGCTTTGGA





AACTGTTTTGCTAGAGTGTCGGAGAGGTAAGT





GGAATTCCTAGTGTAGCGGTGAAATGCGTAG





ATATTAGGAGGAACACCAGTGGCGAAGGCGG





CTTACTGGACGATAACTGACGTTGAGGCTCGA





AAGCGTGGGGAGCAAACAGGATTAGATACCC





TGGTAGTCCACGCCGTAAACGATGAATGCTA





GGTGTTGGGGGGCAAAGCCCTTC







Lachnospiraceae sp


GAAGTATTTCGGTATGTAAACTTCTATCAGCA



or [Clostridium] sp

GGGAAGAAAATGACGGTACCTGACTAAGAAG



36C9-1014

CCCCGGCTAACTACGTGCCAGCAGCCGCGGT





AATACGTAGGGGGCAAGCGTTATCCGGATTT





ACTGGGTGTAAAGGGAGCGTAGACGGCAGTG





CAAGTCTGAAGTGAAAGCCCGGGGCTCAACC





CCGGGACTGCTTTGGAAACTGTGCAGCTAGA





GTGTCGGAGAGGCAAGCGGAATTCCTAGTGT





AGCGGTGAAATGCGTAGATATTAGGAGGAAC





ACCAGTGGCGAAGGCGGCTTGCTGGACGATG





ACTGACGTTGAGGCTCGAAAGCGTGGGGAGC





AAACAGGATTAGATACCCTGGTAGTCCACGC





CGTAAACGATGACTACTAGGTGTCGGGGAGC





AAAGCTCTTCGGTGCCGCAGCCAACGCAATA





AGTAGTCCACCTGGGGAGTACGTTCGCAAGA





ATGAAACTCAAAGGAATTGACGGGGACCCGC





ACAAGCGGTGGAGCATGTGGTTTAATTCGAA





GCAACGCGAAGAACCTTACCTGCTCTTGACAT





CCCTCTGACCG






[Clostridium]

>S10-121-contig




bolteae S10-21


GATGCAGCGACGCCGCGTGAGTGAAGAAGTA





TTTCGGTATGTAAAGCTCTATCAGCAGGGAAG





AAAATGACGGTACCTGACTAAGAAGCCCCGG





CTAACTACGTGCCAGOAGCCGCGGTAATACG





TAGGGGGCAAGCGTTATCCGGATTTACTGGGT





GTAAAGGGAGCGTAGACGGCGAAGCAAGTCT





GAAGTGAAAACCCAGGGCTCAACCCTGGGAC





TGCTTTGGAAACTGTTTTGCTAGAGTGTCGGA





GAGGTAAGTGGAATTCCTAGTGTAGCGGTGA





AATGCGTAGATATTAGGAGGAACACCAGTGG





CGAAGGCGGCTTACTGGACGATAACTGACGT





TGAGGCTCGAAAGCGTGGGGAGCAAACAGGA





TTAGATACCCTGGTAGTCCACGCCGTAAACGA





TGAATGCTAGGTGTTGGGGGGCAAAGCCCTT





CGGTGCCGTCGCAAACGCAGTAAGCATTCCA





CCTGGGGAGTACGTTCGCAAGAATGAAACTC





AAAGGAATTGACGGGGACCCGCACAAGCGGT





GGAGCATGTGGTTTAATTCGAAGCAACGCGA





AGAACCTTACCAAGTCTTGACATCCTCTTGAC





CGGCGTGTAACGGCGCCTTCCCTTCGGGGCAG





GAGAGACAGGTGGTGCATGGTTGTCGTCAGC





TCGTGTCGTGAGATGTTGGGTTAAGTCCCGCA





ACGAGCGCAACCCTTATCCTTAGTAGCCAGCA





GGTAAAGCTGGGCACTCTAGGGAGACTGCCA





GGGATAACCTGGAGGAAGGTGGGGATGACGT





CAAATCATCATGCCCCTTATGATTTGGGCTAC





ACACGTGCTACAATGGCGTAAACAAAGGGAA





GCAAGACAGTGATGTGGAGCAAATCCCAAAA





ATAACGTCCCAGTTCGGACTGTAGTCTGCAAC





CCGACTACACGAAGCTGGAATCGCTAGTAAT





CGCGAATCAGAATGTCGCGGTGAATACGTTC





CCGGGTCTTGTACACACCGCCCGTCACACCAT





GGGAGTCAGCAACGCCCGAAGTCAGTGACCC





AACTCGCAAGAGAGGG







Ruminococcus

PTA-126695
CCTTAGCGGTTGGGTCACTGACTTCGGGCGTT




gnavus Strain A


ACTGACTCCCATGGTGTGACGGGCGGTGTGTA





CAAGACCCGGGAACGTATTCACCGCGACATT





CTGATTCGCGATTACTAGCGATTCCAGCTTCA





TGTAGTCGAGTTGCAGACTACAATCCGAACTG





AGACGTTATTTTTGGGATTTGCTCCCCCTCGC





GGGCTCGCTTCCCTTTGTTTACGCCATTGTAG





CACGTGTGTAGCCCTGGTCATAAGGGGCATG





ATGATTTGACGTCATCCCCACCTTCCTCCAGG





TTATCCCTGGCAGTCTCTCTAGAGTGCCCATC





CTAAATGCTGGCTACTAAAGATAGGGGTTGC





GCTCGTTGCGGGACTTAACCCAACATCTCACG





ACACGAGCTGACGACAACCATGCACCACCTG





TCTCCTCTGTCCCGAAGGAAAGCTCCGATTAA





AGAGCGGTCAGAGGGATGTCAAGACCAGGTA





AGGTTCTTCGCGTTGCTTCGAATTAAACCACA





TGCTCCACCGCTTGTGCGGGTCCCCGTCAATT





CCTTTGAGTTTCATTCTTGCGAACGTACTCCC





CAGGTGGAATACTTATTGCGTTTGCTGCGGCA





CCGAATGGCTTTGCCACCCGACACCTAGTATT





CATCGTTTACGGCGTGGACTACCAGGGTATCT





AATCCTGTTTGCTCCCCACGCTTTCGAGCCTC





AACGTCAGTCATCGTCCAGAAAGCCGCCTTCG





CCACTGGTGTTCCTCCTAATATCTACGCATTT





CACCGCTACACTAGGAATTCCGCTTTCCTCTC





CGACACTCTAGCCTGACAGTTCCAAATGCAGT







Tyzzerellanexilis


>T.nexilis S10-231 consensus



Strain A

sequence





GGCTAAATACGTGCCAGCAGCCGCGGTAATA





CGTATGGTGCAAGCGTTATCCGGATTTACTGG





GTGTAAAGGGAGCGTAGACGGTTGTGTAAGT





CTGATGTGAAAGCCCGGGGCTCAACCCCGGG





ACTGCATTGGAAACTATGTAACTAGAGTGTCG





GAGAGGTAAGCGGAATTCCTAGTGTAGCGGT





GAAATGCGTAGATATTAGGAGGAACACCAGT





GGCGAAGGCGGCTTACTGGACGATCACTGAC





GTTGAGGCTCGAAAGCGTGGGGAGCAAACAG





GATTAGATACCCTGGTAGTCCACGCCGTAAAC





GATGACTACTAGGTGTCGGGGAGCAAAGCTC





TTCGGTGCCGCAGCAAACGCAATAAGTAGTC





CACCTGGGGAGTACGTTCGCAAGAATGAAAC





TCAAAGGAATTGACGGGGACCCGCACAAGCG





GTGGAGCATGTGGTTTAATTCGAAGCAACGC





GAAGAACCTTACCTGGTCTTGACATCCCTCTG





ACCGCTCTTTAATCGGAGTTTTCCTTCGGGAC





AGAGGAGACAGGTGGTGCATGGTTGTCGTCA





GCTCGTGTCGTGAGATGTTGGGTTAAGTCCCG





CAACGAGCGCAACCCCTATCTTCAGTAGCCA





GCATTTAAGGTGGGCACTCTGGAGAGACTGC





CAGGGATAACCTGGAGGAAGGTGGGGATGAC





GTCAAATCATCATGCCCCTTATGACCAGGGCT





ACACACGTGCTACAATGGCGTAAACAAAGGG





AAGCGAACCTGTGAGGGGAAGCAAATCTCAA





AAATAACGTCTCAGTTCGGATTGTAGTCTGCA





ACTCGACTACATGAAGCTGGAATCGCTAGTA





ATCGCGAATCAGCATGTCGCGGTGAATACGTT





CCCGGGTCTTGTACACACCGCCCGTC







Veillonella


>S11-T9-357F




tobetsuensis


AGCAACGCCGCGTGAGTGATGACGGCCTTCG





GGTTGTAAAGCTCTGTTAATCGGGACGAAAG





GCCTTCTTGCGAATAGTTAGAAGGATTGACGG





TACCGGAATAGAAAGCCACGGCTAACTACGT





GCCAGCAGCCGCGGTAATACGTAGGTGGCAA





GCGTTGTCCGGAATTATTGGGCGTAAAGCGC





GCGCAGGCGGATCGGTCAGTCTGTCTTAAAA





GTTCGGGGCTTAACCCCGTGAGGGGATGGAA





ACTGCTGATCTAGAGTATCGGAGAGGAAAGT





GGAATTCCTAGTGTAGCGGTGAAATGCGTAG





ATATTAGGAAGAACACCAGTGGCGAAGGCGA





CTTTCTGGACGAAAACTGACGCTGAGGCGCG





AAAGCCAGGGGAGCGAACGGGATTAGATACC





CCGGTAGTCCTGGCCGTAAACGATGGGTACT





AGGTGTAGGAGGTATCGACCCCTTCTGTGCCG





GAGTTAACGCAATAAGTACCCCGCCTGGGGA





GTACGACCGCAAGGTTGAAACTCAAAGGAAT





TGACGGGGGCCCGCACAAGCGGTGGAGTATG





TGGTTTAATTCGACGCAACGCGAAGAACCTTA





CCAGGTCTTGACATTGATGGACAGAACTAGA





GATAGTTCCTCTTCTTCGGAAGCCAGAAAACA





GGTGGTGCACGGTTGTCGTCAGCTCGTGTCGT





GAGATGTTGGGTTAAGTCCCGCAACGAGCGC





AACCCCTATCTTATGTTGCCAGCACTTCGGGT





GGGAACTCAT







Veillonellaparvula


>S14-201





Contig





GAGTGATGACGGCCTTCGGGTTGTAAAGCTCT





GTTAATCGGGACGAAAGGCCTTCTTGCGAAT





AGTGAGAAGGATTGACGGTACCGGAATAGAA





AGCCACGGCTAACTACGTGCCAGCAGCCGCG





GTAATACGTAGGTGGCAAGCGTTGTCCGGAA





TTATTGGGCGTAAAGCGCGCGCAGGCGGATA





GGTCAGTCTGTCTTAAAAGTTCGGGGCTTAAC





CCCGTGATGGGATGGAAACTGCCAATCTAGA





GTATCGGAGAGGAAAGTGGAATTCCTAGTGT





AGCGGTGAAATGCGTAGATATTAGGAAGAAC





ACCAGTGGCGAAGGCGACTTTCTGGACGAAA





ACTGACGCTGAGGCGCGAAAGCCAGGGGAGC





GAACGGGATTAGATACCCCGGTAGTCCTGGC





CGTAAACGATGGGTACTAGGTGTAGGAGGTA





TCGACCCCTTCTGTGCCGGAGTTAACGCAATA





AGTACCCCGCCTGGGGAGTACGACCGCAAGG





TTGAAACTCAAAGGAATTGACGGGGGCCCGC





ACAAGCGGTGGAGTATGTGGTTTAATTCGAC





GCAACGCGAAGAACCTTACCAGGTCTTGACA





TTGATGGACAGAACCAGAGATGGTTCCTCTTC





TTCGGAAGCCAGAAAACAGGTGGTGCACGGT





TGTCGTCAGCTCGTGTCGTGAGATGTTGGGTT





AAGTCCCGCAACGAGCGCAACCCCTATCTTAT





GTTGCCAGCACTTTGGGTGGGGACTCATGAG





AGACTGCCGCAGACAATGCGGAGGAAGGCGG





GGATGACGTCAAATCATCATGCCCCTTATGAC





CTGGGCTACACACGTACTACAATGGGAGTTA





ATAGACGGAAGCGAGATCGCGAGATGGAGCA





AACCCGAGAAACACTCTCTCAGTTCGGATCGT





AGGCTGCAACTCGCCTACGTGAAGTCGGAAT





CGCTAGTAATCGCAGGTCAGCATACTGCGGT





GAATACGTTCCCGGGCCTTGTACACACCGCCC





GTCACACCACGAAAGTCGGAAGTGCCCAAAG





CCGGTGGGGTAACCTTC







Veillonellaparvula


>S14-205 Contig





GAGTGATGACGGCCTTCGGGTTGTAAAGCTCT





GTTAATCGGGACGAAAGGCCTTCTTGCGAAT





AGTGAGAAGGATTGACGGTACCGGAATAGAA





AGCCACGGCTAACTACGTGCCAGCAGCCGCG





GTAATACGTAGGTGGCAAGCGTTGTCCGGAA





TTATTGGGCGTAAAGCGCGCGCAGGCGGATA





GGTCAGTCTGTCTTAAAAGTTCGGGGCTTAAC





CCCGTGATGGGATGGAAACTGCCAATCTAGA





GTATCGGAGAGGAAAGTGGAATTCCTAGTGT





AGCGGTGAAATGCGTAGATATTAGGAAGAAC





ACCAGTGGCGAAGGCGACTTTCTGGACGAAA





ACTGACGCTGAGGCGCGAAAGCCAGGGGAGC





GAACGGGATTAGATACCCCGGTAGTCCTGGC





CGTAAACGATGGGTACTAGGTGTAGGAGGTA





TCGACCCCTTCTGTGCCGGAGTTAACGCAATA





AGTACCCCGCCTGGGGAGTACGACCGCAAGG





TTGAAACTCAAAGGAATTGACGGGGGCCCGC





ACAAGCGGTGGAGTATGTGGTTTAATTCGAC





GCAACGCGAAGAACCTTACCAGGTCTTGACA





TTGATGGACAGAACCAGAGATGGTTCCTCTTC





TTCGGAAGCCAGAAAACAGGTGGTGCACGGT





TGTCGTCAGCTCGTGTCGTGAGATGTTGGGTT





AAGTCCCGCAACGAGCGCAACCCCTATCTTAT





GTTGCCAGCACTTTGGGTGGGGACTCATGAG





AGACTGCCGCAGACAATGCGGAGGAAGGCGG





GGATGACGTCAAATCATCATGCCCCTTATGAC





CTGGGCTACACACGTACTACAATGGGAGTTA





ATAGACGGAAGCGAGATCGCGAGATGGAGCA





AACCCGAGAAACACTCTCTCAGTTCGGATCGT





AGGCTGCAACTCGCCTACGTGAAGTCGGAAT





CGCTAGTAATCGCAGGTCAGCATACTGCGGT





GAATACGTTCCCGGGCCTTGTACACACCGCCC





GTCACACCACGAAAGTCGGAAGTGCCCAAAG





CCGGTG







Veillonellaatypica

PTA-125709




Strain A









Veillonellaatypica

PTA-125711




Strain B









Veillonelladispar










Veillonellaparvula

PTA-125691




Strain A









Veillonellaparvula

PTA-125711




Strain B









Veillonella

PTA-125 708





tobetsuensis Strain






A









Veillonella







tobetsuensis Strain B










Lactobacillus


ATGGAGCAACGCCGCGTGAGTGAAGAAGGTC




salivarius Strain A


TTCGGATCGTAAAACTCTGTTGTTAGAGAAGA





ACACGAGTGAGAGTAACTGTTCATTCGATGA





CGGTATCTAACCAGCAAGTCACGGCTAACTA





CGTGCCAGCAGCCGCGGTAATACGTAGGTGG





CAAGCGTTGTCCGGATTTATTGGGCGTAAAGG





GAACGCAGGCGGTCTTTTAAGTCTGATGTGAA





AGCCTTCGGCTTAACCGGAGTAGTGCATTGGA





AACTGGAAGACTTGAGTGCAGAAGAGGAGAG





TGGAACTCCATGTGTAGCGGTGAAATGCGTA





GATATATGGAAGAACACCAGTGGCGAAAGCG





GCTCTCTGGTCTGTAACTGACGCTGAGGTTCG





AAAGCGTGGGTAGCAAACAGGATTAGATACC





CTGGTAGTCCACGCCGTAAACGATGAATGCT





AGGTGTTGGAGGGTTTCCGCCCTTCAGTGCCG





CAGCTAACGCAATAAGCATTCCGCCTGGGGA





GTACGACCGCAAGGTTGAAACTCAAAGGAAT





TGACGGGGGCCCGCACAAGCGGTGGAGCATG





TGGTTTAATTCGAAGCAACGCGAAGAACCTT





ACCAGGTCTTGACATCCTTTGACCACCTAAGA





GATTAGGCTTTCCCTTCGGGGACAAAGTGACA





GGTGGTGCATGGCTGTCGTCAGCTCGTGTCGT





GAGATGTTGGGTTAAGTCCCGCAACGAGCGC





AACCCTTGTTGTCAGTTGCCAGCATTAAGTTG





GGCACTCTGGCGAGACTGCCGGTGACAAACC





GGAGGAAGGTGGGGACGACGTCAAGTCATCA





TGCCCCTTATGACCTGGGCTACACACGTGCTA





CAATGGACGGTACAACGAGTCGCGAGACCGC





GAGGTTTAGCTAATCTCTTAAAGCCGTTCTCA





GTTCGGATTGTAGGCTGCAACTCGCCTACATG





AAGTCGGAATCGCTAGTAATCGCGAATCAGC





ATGTCGCGGTGAATACGTTCCCGGGCCTTGTA





CACACCGCCCGTCACACCATGAGAGTTTGTAA





CACCCAAAGCCGGTGGGGTAACCGCAAGGAG





CCAGCCG







Agathobaculum


CCGCGTGATTGAAGAAGGCCTNTCGGGTTGT



Strain A

AAAGATCTTTAATTCGGGACGAAAAATGACG





GTACCGAAAGAATAAGCTCCGGCTAACTACG





TGCCAGCAGCCGCGGTAATACGTAGGGAGCA





AGCGTTATCCGGATTTACTGGGTGTAAAGGGC





GCGCAGGCGGGCTGGCAAGTTGGAAGTGAAA





TCTAGGGGCTTAACCCCTAAACTGCTTTCAAA





ACTGCTGGTCTTGAGTGATGGAGAGGCAGGC





GGAATTCCGTGTGTAGCGGTGAAATGCGTAG





ATATACGGAGGAACACCAGTGGCGAAGGCGG





CCTGGTGGACATTAACTGACGCTGAGGGGCG





AAAGCGTGGGGAGCAAACAGGATTAGATACC





CTGGTAGTCCACGCCGTAAACGATGGATACT





AGGTGTGGGAGGTATTGACCCCTTCCGTGCCG





CAGTTAACACAATAAGTATCCCACCTGGGGA





GTACGGCCGCAAGGTTGAAACTCAAAGGAAT





TGACGGGGGCCCGCACAAGCAGTGGAGTATG





TGGTTTAATTCGAAGCAACGCGAAGAACCTT





ACCAGGCCTTGACATCCCGATGACCGGTCTAG





AGATAGACCTTCTCTTCGGAGCATCGGTGACA





GGTGGTGCATGGTTGTCGTCAGCTCGTGTCGT





GAGATGTTGGGTTAAGTCCCGCAACGAGCGC





AACCCTTACGGTTAGTTGATACGCAAGATCAC





TCTAGCCGGACTGCCGTTGACAAAACGGAGG





AAGGTGGGGACGACGTCAAATCATCATGCCC





CTTATGGCCTGGGCTACACACGTACTACAATG





GCAGTCATACAGAGGGAAGCAAAGCTGTGAG





GCGGAGCAAATCCCTAAAAGCTGTCCCAGTT





CAGATTGCAGGCTGCAACCCGCCTGCATGAA





GTCGGAATTGCTAGTAATCGCGGATCAGCAT





GCCGCGGTGAATACGTTCCCGGGCCTTGTACA





CACCGCCCGTCACACCATGAGAGCCGTCAAT





ACCCGAAGTCCGTAGCCTAACCGCAAG







Paraclostridium


GAATTACTGGGCGTAAAGGGTGCGTAGGTGG




benzoelyticum


TTTTTTAAGTCAGAAGTGAAAGGCTACGGCTC



Strain A

AACCGTAGTAAGCTTTTGAAACTAGAGAACTT





GAGTGCAGGAGAGGAGAGTAGAATTCCTAGT





GTAGCGGTGAAATGCGTAGATATTAGGAGGA





ATACCAGTAGCGAAGGCGGCTCTCTGGACTG





TAACTGACACTGAGGCACGAAAGCGTGGGGA





GCAAACAGGATTAGATACCCTGGTAGTCCAC





GCCGTAAACGATGAGTACTAGGTGTCGGGGG





TTACCCCCCTCGGTGCCGCAGCTAACGCATTA





AGTACTCCGCCTGGGAAGTACGCTCGCAAGA





GTGAAACTCAAAGGAATTGACGGGGACCCGC





ACAAGTAGCGGAGCATGTGGTTTAATTCGAA





GCAACGCGAAGAACCTTACCTAAGCTTGACA





TCCCACTGACCTCTCCCTAATCGGAGATTTCC





CTTCGGGGACAGTGGTGACAGGTGGTGCATG





GTTGTCGTCAGCTCGTGTCGTGAGATGTTGGG





TTAAGTCCCGCAACGAGCGCAACCCTTGCCTT





TAGTTGCCAGCATTAAGTTGGGCACTCTAGAG





GGACTGCCGAGGATAACTCGGAGGAAGGTGG





GGATGACGTCAAATCATCATGCCCCTTATGCT





TAGGGCTACACACGTGCTACAATGGGTGGTA





CAGAGGGTTGCCAAGCCGCGAGGTGGAGCTA





ATCCCTTAAAGCCATTCTCAGTTCGGATTGTA





GGCTGAAACTCGCCTACATGAAGCTGGAGTT





ACTAGTAATCGCAGATCAGAATGCTGCGGTG





AATGCGTTCCCGGGTCTTGTACACACCGCCCG





TCACACCATGGAAGTTGGGGGCGCCCGAAGC





CGGTTAGCTAACCTTTTAGGAAGCGGCCGT







Turicibacter


ATGGCTAGAGTGTGACGGTACCTTATGAGAA




sanguinis Strain A


AGCCACGGCTAACTACGTGCCAGCAGCCGCG





GTAATACGTAGGTGGCGAGCGTTATCCGGAA





TTATTGGGCGTAAAGAGCGCGCAGGTGGTTG





ATTAAGTCTGATGTGAAAGCCCACGGCTTAAC





CGTGGAGGGTCATTGGAAACTGGTCAACTTG





AGTGCAGAAGAGGGAAGTGGAATTCCATGTG





TAGCGGTGAAATGCGTAGAGATATGGAGGAA





CACCAGTGGCGAAGGCGGCTTCCTGGTCTGTA





ACTGACACTGAGGCGCGAAAGCGTGGGGAGC





AAACAGGATTAGATACCCTGGTAGTCCACGC





CGTAAACGATGAGTGCTAAGTGTTGGGGGTC





GAACCTCAGTGCTGAAGTTAACGCATTAAGC





ACTCCGCCTGGGGAGTACGGTCGCAAGACTG





AAACTCAAAGGAATTGACGGGGACCCGCACA





AGCGGTGGAGCATGTGGTTTAATTCGAAGCA





ACGCGAAGAACCTTACCAGGTCTTGACATAC





CAGTGACCGTCCTAGAGATAGGATTTTCCCT





TCGGGGACAATGGATACAGGTGGTGCATGGT





TGTCGTCAGCTCGTGTCGTGAGATGTTGGGTT





AAGTCCCGCAACGAGCGCAACCCCTGTCGTT





AGTTGCCAGCATTCAGTTGGGGACTCTAACGA





GACTGCCAGTGACAAACTGGAGGAAGGTGGG





GATGACGTCAAATCATCATGCCCCTTATGACC





TGGGCTACACACGTGCTACAATGGTTGGTACA





AAGAGAAGCGAAGCGGTGACGTGGAGCAAA





CCTCATAAAGCCAATCTCAGTTCGGATTGTAG





GCTGCAACTCGCCTACATGAAGTTGGAATCGC





TAGTAATCGCGAATCAGCATGTCGCGGTGAA





TACGTT







Burkholderia







pseudomallei










Klebsiella







quasipneumoniae






subsp.






similipneumoniae










Klebsiellaoxytoca






Strain A









Megasphaera Sp.

PTA-126770
TATCAATTCGAGTGGCAAACGGGTGA



Strain A

GTAACGCGTAAGCAACCTGCCCTTCA





GATGGGGACAACAGCTGGAAACGGCT





GCTAATACCGAATACGTTCTTTCCGCC





GCATGACGGGATGAAGAAAGGGAGG





CCTTCGGGCTTTCGCTGGAGGAGGGG





CTTGCGTCTGATTAGCTAGTTGGAGG





GGTAACGGCCCACCAAGGCGACGATC





AGTAGCCGGTCTGAGAGGATGAACGG





CCACATTGGGACTGAGACACGGCCCA





GACTCCTACGGGAGGCAGCAGTGGGG





AATCTTCCGCAATGGACGAAAGTCTG





ACGGAGCAACGCCGCGTGAACGATGA





CGGCCTTCGGGTTGTAAAGTTCTGTTA





TATGGGACGAACAGGATAGCGGTCAA





TACCCGTTATCCCTGACGGTACCGTAA





GAGAAAGCCACGGCTAACTACGTGCC





AGCAGCCGCGGTAATACGTAGGTGGC





AAGCGTTGTCCGGAATTATTGGGCGT





AAAGGGCGCGCAGGCGGCATCGCAA





GTCGGTCTTAAAAGTGCGGGGCTTAA





CCCCGTGAGGGGACCGAAACTGTGAA





GCTCGAGTGTCGGAGAGGAAAGCGGA





ATTCCTAGTGTAGCGGTGAAATGCGT





AGATATTAGGAGGAACACCAGTGGCG





AAAGCGGCTTTCTGGACGACAACTGA





CGCTGAGGCGCGAAAGCCAGGGGAG





CAAACGGGATTAGATACCCCGGTAGT





CCTGGCCGTAAACGATGGATACTAGG





TGTAGGAGGTATCGACTCCTTCTGTGC





CGGAGTTAACGCAATAAGTATCCCGC





CTGGGGAGTACGGCCGCAAGGCTGAA





ACTCAAAGGAATTGACGGGGGCCCGC





ACAAGCGGTGGAGTATGTGGTTTAAT





TCGACGCAACGCGAAGAACCTTACCA





AGCCTTGACATTGATTGCTACGGAAA





GAGATTTCCGGTTCTTCTTCGGAAGAC





AAGAAAACAGGTGGTGCACGGCTGTC





GTCAGCTCGTGTCGTGAGATGTTGGG





TTAAGTCCCGCAACGAGCGCAACCCC





TATCTTCTGTTGCCAGCACTAAGGGTG





GGGACTCAGAAGAGACTGCCGCAGAC





AATGCGGAGGAAGGCGGGGATGACGT





CAAGTCATCATGCCCCTTATGGCTTG





GGCTACACACGTACTACAATGGCTCT





TAATAGAGGGAAGCGAAGGAGCGAT





CCGGAGCAAACCCCAAAAACAGAGTC





CCAGTTCGGATTGCAGGCTGCAACTC





GCCTGCATGAAGCAGGAATCGCTAGT





AATCGCAGGTCAGCATACTGCGGTGA





ATACGTTCCCGGGCCTTGTACACACC





GCCCGTCACACCACGAAAGTCATTCA





CACCCGAAGCCGGTGAGGCAACCGCA





AGGAACCAGCCGTCGAAGGTGGGGGC





GATGATTGGGGTGAAGTCGTAACAAG





GTAGCCGTATCGGAAGGTGCGGCTGG





ATCACCTCCTTT







Megasphaera Sp.


ATGGAGAGTTTGATCCTGGCTCAGGA



Strain B

CGAACGCTGGCGGCGTGCTTAACACA





TGCAAGTCGAACGAGAAGAGATGAGA





AGCTTGCTTCTTATCAATTCGAGTGG





CAAACGGGTGAGTAACGCGTAAGCAA





CCTGCCCTTCAGATGGGGACAACAGC





TGGAAACGGCTGCTAATACCGAATAC





GTTCTTTCCGCCGCATGACGGGATGA





AGAAAGGGAGGCCTTCGGGCTTTCGC





TGGAGGAGGGGCTTGCGTCTGATTAG





CTAGTTGGAGGGGTAACGGCCCACCA





AGGCGACGATCAGTAGCCGGTCTGAG





AGGATGAACGGCCACATTGGGACTGA





GACACGGCCCAGACTCCTACGGGAGG





CAGCAGTGGGGAATCTTCCGCAATGG





ACGAAAGTCTGACGGAGCAACGCCGC





GTGAACGATGACGGCCTTCGGGTTGT





AAAGTTCTGTTATATGGGACGAACAG





GATAGCGGTCAATACCCGTTATCCCT





GACGGTACCGTAAGAGAAAGCCACGG





CTAACTACGTGCCAGCAGCCGCGGTA





ATACGTAGGTGGCAAGCGTTGTCCGG





AATTATTGGGCGTAAAGGGCGCGCAG





GCGGCATCGCAAGTCGGTCTTAAAAG





TGCGGGGCTTAACCCCGTGAGGGGAC





CGAAACTGTGAAGCTCGAGTGTCGGA





GAGGAAAGCGGAATTCCTAGTGTAGC





GGTGAAATGCGTAGATATTAGGAGGA





ACACCAGTGGCGAAAGCGGCTTTCTG





GACGACAACTGACGCTGAGGCGCGAA





AGCCAGGGGAGCAAACGGGATTAGAT





ACCCCGGTAGTCCTGGCCGTAAACGA





TGGATACTAGGTGTAGGAGGTATCGA





CTCCTTCTGTGCCGGAGTTAACGCAAT





AAGTATCCCGCCTGGGGAGTACGGCC





GCAAGGCTGAAACTCAAAGGAATTGA





CGGGGGCCCGCACAAGCGGTGGAGTA





TGTGGTTTAATTCGACGCAACGCGAA





GAACCTTACCAAGCCTTGACATTGATT





GCTACGGAAAGAGATTTCCGGTTCTT





CTTCGGAAGACAAGAAAACAGGTGGT





GCACGGCTGTCGTCAGCTCGTGTCGT





GAGATGTTGGGTTAAGTCCCGCAACG





AGCGCAACCCCTATCTTCTGTTGCCAG





CACTAAGGGTGGGGACTCAGAAGAGA





CTGCCGCAGACAATGCGGAGGAAGGC





GGGGATGACGTCAAGTCATCATGCCC





CTTATGGCTTGGGCTACACACGTACTA





CAATGGCTCTTAATAGAGGGAAGCGA





AGGAGCGATCCGGAGCAAACCCCAAA





AACAGAGTCCCAGTTCGGATTGCAGG





CTGCAACTCGCCTGCATGAAGGAGGA





ATCGCTAGTAATCGCAGGTCAGCATA





CTGCGGTGAATACGTTCCCGGGCCTT





GTACACACCGCCCGTCACACCACGAA





AGTCATTCACACCCGAAGCCGGTGAG





GCAACCGCAAGGAACCAGCCGTCGAA





GGTGGGGGCGATGATTGGGGTGAAGT





CGTAACAAGGTAGCCGTATCGGAAGG





TGCGGCTGGATCACCTCCTTT







Selenomonasfelix


GTTGGTGAGGTAACGGCTCACCAAGG





CGACGATCAGTAGCCGGTCTGAGAGG





ATGAACGGCCACATTGGGACTGAGAC





ACGGCCCAGACTCCTACGGGAGGCAG





CAGTGGGGAATCTTCCGCAATGGGCG





CAAGCCTGACGGAGCAACGCCGCGTG





AGTGAAGAAGGTCTTCGGATCGTAAA





GCTCTGTTGACGGGGACGAACGTGCG





GAGTGCGAATAGCGCTTTGTAATGAC





GGTACCTGTCGAGGAAGCCACGGCTA





ACTACGTGCCAGCAGCCGCGGTAATA





CGTAGGTGGCGAGCGTTGTCCGGAAT





CATTGGGCGTAAAGGGAGCGCAGGCG





GGCCGGTAAGTCTTACTTAAAAGTGC





GGGGCTCAACCCCGTGATGGGAGAGA





AACTATCGGTCTTGAGTACAGGAGAG





GAAAGCGGAATTCCCAGTGTAGCGGT





GAAATGCGTAGATATTGGGAAGAACA





CCAGTGGCGAAGGCGGCTTTCTGGAC





TGCAACTGACGCTGAGGCTCGAAAGC





CAGGGGAGCGAACGGGATTAGATACC





CCGGTAGTCCTGGCCGTAAACGATGG





ATACTAGGTGTGGGAGGTATCGACCC





CTACCGTGCCGGAGTTAACGCAATAA





GTATCCCGCCTGGGGAGTACGGCCGC





AAGGCTGAAACTCAAAGGAATTGACG





GGGACCCGCACAAGCGGTGGAGTATG





TGGTTTAATTCGAAGCAACGCGAAGA





ACCTTACCAGGCCTTGACATTGACTG





AAAGCACTAGAGATAGTGCCCTCTCT





TCGGAGACAGGAAAACAGGTGGTGCA





TGGCTGTCGTCAGCTCGTGTCGTGAG





ATGTTGGGTTAAGTCCCGCAACGAGC





GCAACCCCTGTTCTTTGTTGCCATCAG





GTAAAGCTGGGCACTCAAAGGAGACT





GCCGCGGAGAACGCGGAGGAAGGCG





GGGATGACGTCAAGTCATCATGCCCC





TTATGGCCTGGGCTACACACGTACTA





CAATGGAACGGACAGAGAGCAGCGA





ACCCGCGAGGGCAAGCGAACCTCAAA





AACCGTTTCCCAGTTCGGATTGCAGG





CTGCAACCCGCCTGCATGAAGTCGGA





ATCGCTAGTAATCGCAGGTCAGCATA





CTGCGGTGAATACGTTCCCGGGTCTTG





TACACACCGCCCGTCACACCACGGAA





GTCATTCACACCCGAAGCCGGCGCAG





CCGTCTAAGGTGGGGAAGGTGACTGG





GGTGAAGTCGTAACAAGGTAGCCGTA





TCGGAAGGTGCGGCTGGATCACCTCC





TTT







Enterococcus


CTGACCGAGCACGCCGCGTGAGTGAA




gallinarum Strain A


GAAGGTTTTCGGATCGTAAAACTCTG





TTGTTAGAGAAGAACAAGGATGAGAG





TAAAACGTTCATCCCTTGACGGTATCT





AACCAGAAAGCCACGGCTAACTACGT





GCCAGCAGCCGCGGTAATACGTAGGT





GGCAAGCGTTGTCCGGATTTATTGGG





CGTAAAGCGAGCGCAGGCGGTTTCTT





AAGTCTGATGTGAAAGCCCCCGGCTC





AACCGGGGAGGGTCATTGGAAACTGG





GAGACTTGAGTGCAGAAGAGGAGAGT





GGAATTCCATGTGTAGCGGTGAAATG





CGTAGATATATGGAGGAACACCAGTG





GCGAAGGCGGCTCTCTGGTCTGTAAC





TGACGCTGAGGCTCGAAAGCGTGGGG





AGCGAACAGGATTAGATACCCTGGTA





GTCCACGCCGTAAACGATGAGTGCTA





AGTGTTGGAGGGTTTCCGCCCTTCAGT





GCTGCAGCAAACGCATTAAGCACTCC





GCCTGGGGAGTACGACCGCAAGGTTG





AAACTCAAAGGAATTGACGGGGGCCC





GCACAAGCGGTGGAGCATGTGGTTTA





ATTCGAAGCAACGCGAAGAACCTTAC





CAGGTCTTGACATCCTTTGACCACTCT





AGAGATAGAGCTTCCCCTTCGGGGGC





AAAGTGACAGGTGGTGCATGGTTGTC





GTCAGCTCGTGTCGTGAGATGTTGGG





TTAAGTCCCGCAACGAGCGCAACCCT





TATTGTTAGTTGCCATCATTTAGTTGG





GCACTCTAGCGAGACTGCCGGTGACA





AACCGGAGGAAGGTGGGGATGACGTC





AAATCATCATGCCCCTTATGACCTGG





GCTACACACGTGCTACAATGGGAAGT





ACAACGAGTTGCGAAGTCGCGAGGCT





AAGCTAATCTCTTAAAGCTTCTCTCAG





TTCGGATTGTAGGCTGCAACTCGCCTA





CATGAAGCCGGAATCGCTAGTAATCG





CGGATCAGCACGCCGCGGTGAATACG





TTCCCGGGCCTTGTACACACCGCCCGT





CACACCACGAGAGTTTGTAACACCCG





AAGTCGGTGAGGTAACCTTT







Enterococcus


CGCGTGAGTGAAGAAGGTTTTCGGAT




Gallinarum Strain B


CGTAAAACTCTGTTGTTAGAGAAGAA





CAAGGATGAGAGTAGAACGTTCATCC





CTTGACGGTATCTAACCAGAAAGCCA





CGGCTAACTACGTGCCAGCAGCCGCG





GTAATACGTAGGTGGCAAGCGTTGTC





CGGATTTATTGGGCGTAAAGCGAGCG





CAGGCGGTTTCTTAAGTCTGATGTGA





AAGCCCCCGGCTCAACCGGGGAGGGT





CATTGGAAACTGGGAGACTTGAGTGC





AGAAGAGGAGAGTGGAATTCCATGTG





TAGCGGTGAAATGCGTAGATATATGG





AGGAACACCAGTGGCGAAGGCGGCTC





TCTGGTCTGTAACTGACGCTGAGGCTC





GAAAGCGTGGGGAGCGAACAGGATT





AGATACCCTGGTAGTCCACGCCGTAA





ACGATGAGTGCTAAGTGTTGGAGGGT





TTCCGCCCTTCAGTGCTGCAGCAAAC





GCATTAAGCACTCCGCCTGGGGAGTA





CGACCGCAAGGTTGAAACTCAAAGGA





ATTGACGGGGGCCCGCACAAGCGGTG





GAGCATGTGGTTTAATTCGAAGCAAC





GCGAAGAACCTTACCAGGTCTTGACA





TCCTTTGACCACTCTAGAGATAGAGCT





TCCCCTTCGGGGGCAAAGTGACAGGT





GGTGCATGGTTGTCGTCAGCTCGTGTC





GTGAGATGTTGGGTTAAGTCCCGCAA





CGAGCGCAACCCTTATTGTTAGTTGCC





ATCATTTAGTTGGGCACTCTAGCGAG





ACTGCCGGTGACAAACCGGAGGAAGG





TGGGGATGACGTCAAATCATCATGCC





CCTTATGACCTGGGCTACACACGTGCT





ACAATGGGAAGTACAACGAGTTGCGA





AGTCGCGAGGCTAAGCTAATCTCTTA





AAGCTTCTCTCAGTTCGGATTGTAGGC





TGCAACTCGCCTACATGAAGCCGGAA





TCGCTAGTAATCGCGGATCAGCACGC





CGCGGTGAATACGTTCCCGGGCCTTG





TACACACCGCCCGTCACACCACGAGA





GTTTGTAACACCCGAAGTCGGTGAGG





TAACCTTTTNGGAGCCAGCCGC






Fournierella

PTA-126694


Fournierellamassiliensis




massiliensis










Harryflintia

PTA-126696


Harryflintiaacetispora




acetispora






In some embodiments, the mEVs from one or more of the following bacteria:



Akkermansia,
Christensenella,
Blautia,
Enterococcus,
Eubacterium,
Roseburia,
Bacteroides,
Parabacteroides,
or
Erysipelatoclostridium




Blautia
hydrogenotrophica,
Blautia
stercoris,
Blautia
wexlerae,
Eubacterium
faecium,
Eubacterium
contortum,
Eubacterium
rectale,
Enterococcus
faecalis,
Enterococcus
durans,
Enterococcus
villorum,
Enterococcus
gallinarum;
Bifidobacterium
lactis,
Bifidobacterium
bifidium,
Bifidobacterium
longam,
Bifidobacterium
animalis,
or
Bifidobacterium
breve




BCG,
Parabacteroides,
Blautia,
Veillonella,
Lactobacillus
salivarius,
Agathobaculum,
Ruminococcus
gnavus,
Paraclostridium
benzoelyticum,
Turicibacter
sanguinus,
Burkholderia,
Klebsiella
quasi
pneumoniae ssp similpneumoniae,Klebsiellaoxytoca,Tyzzerelanexilis,orNeisseria




Blautia
hydrogenotrophica




Blautia
stercoris




Blautia
wexlerae




Enterococcus
gallinarum




Enterococcus
faecium




Bifidobacterium
bifidium




Bifidobacterium
breve




Bifidobacterium
longum




Roseburia
hominis




Bacteroides
thetaiotaomicron




Bacteroides
coprocola




Erysipelatoclostridium
ramosum




Megasphera,
including
Megasphera
massiliensis




Parabacteroides
distasonis




Eubacterium
contortum




Eubacterium
hallii




Intestimonas
butyriciproducens




Streptococcus
australis




Eubacterium
eligens




Faecalibacterium
prausnitzii




Anaerostipes
caccae




Erysipelotrichaceae




Rikenellaceae




Lactococcus,
Prevotella,
Bifidobacterium,
Veillonella




Lactococcus
lactis
cremoris




Prevotella
histicola




Bifidobacterium
animalis
lactis




Veillonella
parvula







In some embodiments, the mEVs are from Lactococcus lactis cremoris bacteria, e.g., from a strain comprising at least 90% or at least 99% genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Lactococcus lactis cremoris Strain A (ATCC designation number PTA-125368). In some embodiments, the mEVs are from Lactococcus bacteria, e.g., from Lactococcus lactis cremoris Strain A (ATCC designation number PTA-125368).


In some embodiments, the mEVs are from Prevotella bacteria, e.g., from a strain comprising at least 90% or at least 99% genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Prevotella Strain B 50329 (NRRL accession number B 50329). In some embodiments, the mEVs are from Prevotella bacteria, e.g., from Prevotella Strain B 50329 (NRRL accession number B 50329).


In some embodiments, the mEVs are from Bifidobacterium bacteria, e.g., from a strain comprising at least 90% or at least 99% genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Bifidobacterium bacteria deposited as ATCC designation number PTA-125097. In some embodiments, the mEVs are from Bifidobacterium bacteria, e.g., from Bifidobacterium bacteria deposited as ATCC designation number PTA-125097.


In some embodiments, the mEVs are from Veillonella bacteria, e.g., from a strain comprising at least 90% or at least 99% genomic, 16S and/or CRISPR sequence identity to the nucleotide sequence of the Veillonella bacteria deposited as ATCC designation number PTA-125691. In some embodiments, the mEVs are from Veillonella bacteria, e.g., from Veillonella bacteria deposited as ATCC designation number PTA-125691.


Modified mEVs


In some aspects, the mEVs (such as smEVs) described herein are modified such that they comprise, are linked to, and/or are bound by a therapeutic moiety.


In some embodiments, the therapeutic moiety is a cancer-specific moiety. In some embodiments, the cancer-specific moiety has binding specificity for a cancer cell (e.g., has binding specificity for a cancer-specific antigen). In some embodiments, the cancer-specific moiety comprises an antibody or antigen binding fragment thereof. In some embodiments, the cancer-specific moiety comprises a T cell receptor or a chimeric antigen receptor (CAR). In some embodiments, the cancer-specific moiety comprises a ligand for a receptor expressed on the surface of a cancer cell or a receptor-binding fragment thereof. In some embodiments, the cancer-specific moiety is a bipartite fusion protein that has two parts: a first part that binds to and/or is linked to the bacterium and a second part that is capable of binding to a cancer cell (e.g., by having binding specificity for a cancer-specific antigen). In some embodiments, the first part is a fragment of or a full-length peptidoglycan recognition protein, such as PGRP. In some embodiments the first part has binding specificity for the mEV (e.g., by having binding specificity for a bacterial antigen). In some embodiments, the first and/or second part comprises an antibody or antigen binding fragment thereof. In some embodiments, the first and/or second part comprises a T cell receptor or a chimeric antigen receptor (CAR). In some embodiments, the first and/or second part comprises a ligand for a receptor expressed on the surface of a cancer cell or a receptor-binding fragment thereof In certain embodiments, co-administration of the cancer-specific moiety with the mEVs (either in combination or in separate administrations) increases the targeting of the mEVs to the cancer cells.


In some embodiments, the mEVs described herein are modified such that they comprise, are linked to, and/or are bound by a magnetic and/or paramagnetic moiety (e.g., a magnetic bead). In some embodiments, the magnetic and/or paramagnetic moiety is comprised by and/or directly linked to the bacteria. In some embodiments, the magnetic and/or paramagnetic moiety is linked to and/or a part of an mEV-binding moiety that that binds to the mEV. In some embodiments, the mEV-binding moiety is a fragment of or a full-length peptidoglycan recognition protein, such as PGRP. In some embodiments the mEV-binding moiety has binding specificity for the mEV (e.g., by having binding specificity for a bacterial antigen). In some embodiments, the mEV-binding moiety comprises an antibody or antigen binding fragment thereof. In some embodiments, the mEV-binding moiety comprises a T cell receptor or a chimeric antigen receptor (CAR). In some embodiments, the mEV-binding moiety comprises a ligand for a receptor expressed on the surface of a cancer cell or a receptor-binding fragment thereof In certain embodiments, co-administration of the magnetic and/or paramagnetic moiety with the mEVs (either together or in separate administrations) can be used to increase the targeting of the mEVs (e.g., to cancer cells and/or a part of a subject where cancer cells are present.


Production of Secreted Microbial Extracellular Vesicles (smEVs)


In certain aspects, the smEVs described herein can be prepared using any method known in the art.


In some embodiments, the smEVs are prepared without an smEV purification step. For example, in some embodiments, bacteria described herein are killed using a method that leaves the smEVs intact and the resulting bacterial components, including the smEVs, are used in the methods and compositions described herein. In some embodiments, the bacteria are killed using an antibiotic (e.g., using an antibiotic described herein). In some embodiments, the bacteria are killed using UV irradiation. In some embodiments, the bacteria are heat-killed.


In some embodiments, the smEVs described herein are purified from one or more other bacterial components. Methods for purifying smEVs from bacteria are known in the art. In some embodiments, smEVs are prepared from bacterial cultures using methods described in S. Bin Park, et al. PLoS ONE. 6(3):e17629 (2011) or G. Norheim, et al. PLoS ONE. 10(9): e0134353 (2015) or Jeppesen, et al. Cell 177:428 (2019), each of which is hereby incorporated by reference in its entirety. In some embodiments, the bacteria are cultured to high optical density and then centrifuged to pellet bacteria (e.g., at 10,000×g for 30 min at 4° C., at 15,500×g for 15 min at 4° C.). In some embodiments, the culture supernatants are then passed through filters to exclude intact bacterial cells (e.g., a 0.22 μm filter). In some embodiments, the supernatants are then subjected to tangential flow filtration, during which the supernatant is concentrated, species smaller than 100 kDa are removed, and the media is partially exchanged with PBS. In some embodiments, filtered supernatants are centrifuged to pellet bacterial smEVs (e.g., at 100,000-150,000×g for 1-3 hours at 4° C., at 200,000×g for 1-3 hours at 4° C.). In some embodiments, the smEVs are further purified by resuspending the resulting smEV pellets (e.g., in PBS), and applying the resuspended smEVs to an Optiprep (iodixanol) gradient or gradient (e.g., a 30-60% discontinuous gradient, a 0-45% discontinuous gradient), followed by centrifugation (e.g., at 200,000×g for 4-20 hours at 4° C.). smEV bands can be collected, diluted with PBS, and centrifuged to pellet the smEVs (e.g., at 150,000×g for 3 hours at 4° C., at 200,000×g for 1 hour at 4° C.). The purified smEVs can be stored, for example, at −80° C. or −20° C. until use. In some embodiments, the smEVs are further purified by treatment with DNase and/or proteinase K.


For example, in some embodiments, cultures of bacteria can be centrifuged at 11,000×g for 20-40 min at 4° C. to pellet bacteria. Culture supernatants may be passed through a 0.22 μm filter to exclude intact bacterial cells. Filtered supernatants may then be concentrated using methods that may include, but are not limited to, ammonium sulfate precipitation, ultracentrifugation, or filtration. For example, for ammonium sulfate precipitation, 1.5-3 M ammonium sulfate can be added to filtered supernatant slowly, while stirring at 4° C. Precipitations can be incubated at 4° C. for 8-48 hours and then centrifuged at 11,000×g for 20-40 min at 4° C. The resulting pellets contain bacteria smEVs and other debris. Using ultracentrifugation, filtered supernatants can be centrifuged at 100,000-200,000×g for 1-16 hours at 4° C. The pellet of this centrifugation contains bacteria smEVs and other debris such as large protein complexes. In some embodiments, using a filtration technique, such as through the use of an Amicon Ultra spin filter or by tangential flow filtration, supernatants can be filtered so as to retain species of molecular weight>50 or 100 kDa.


Alternatively, smEVs can be obtained from bacteria cultures continuously during growth, or at selected time points during growth, for example, by connecting a bioreactor to an alternating tangential flow (ATF) system (e.g., XCell ATF from Repligen). The ATF system retains intact cells (>0.22 um) in the bioreactor, and allows smaller components (e.g., smEVs, free proteins) to pass through a filter for collection. For example, the system may be configured so that the <0.22 um filtrate is then passed through a second filter of 100 kDa, allowing species such as smEVs between 0.22 um and 100 kDa to be collected, and species smaller than 100 kDa to be pumped back into the bioreactor. Alternatively, the system may be configured to allow for medium in the bioreactor to be replenished and/or modified during growth of the culture. smEVs collected by this method may be further purified and/or concentrated by ultracentrifugation or filtration as described above for filtered supernatants.


smEVs obtained by methods provided herein may be further purified by size-based column chromatography, by affinity chromatography, by ion-exchange chromatography, and by gradient ultracentrifugation, using methods that may include, but are not limited to, use of a sucrose gradient or Optiprep gradient. Briefly, using a sucrose gradient method, if ammonium sulfate precipitation or ultracentrifugation were used to concentrate the filtered supernatants, pellets are resuspended in 60% sucrose, 30 mM Tris, pH 8.0. If filtration was used to concentrate the filtered supernatant, the concentrate is buffer exchanged into 60% sucrose, 30 mM Tris, pH 8.0, using an Amicon Ultra column. Samples are applied to a 35-60% discontinuous sucrose gradient and centrifuged at 200,000×g for 3-24 hours at 4° C. Briefly, using an Optiprep gradient method, if ammonium sulfate precipitation or ultracentrifugation were used to concentrate the filtered supernatants, pellets are resuspended in PBS and 3 volumes of 60% Optiprep are added to the sample. In some embodiments, if filtration was used to concentrate the filtered supernatant, the concentrate is diluted using 60% Optiprep to a final concentration of 35% Optiprep. Samples are applied to a 0-45% discontinuous Optiprep gradient and centrifuged at 200,000×g for 3-24 hours at 4° C., e.g., 4-24 hours at 4° C.


In some embodiments, to confirm sterility and isolation of the smEV preparations, smEVs are serially diluted onto agar medium used for routine culture of the bacteria being tested, and incubated using routine conditions. Non-sterile preparations are passed through a 0.22 um filter to exclude intact cells. To further increase purity, isolated smEVs may be DNase or proteinase K treated.


In some embodiments, for preparation of smEVs used for in vivo injections, purified smEVs are processed as described previously (G. Norheim, et al. PLoS ONE. 10(9): e0134353 (2015)). Briefly, after sucrose gradient centrifugation, bands containing smEVs are resuspended to a final concentration of 50 μg/mL in a solution containing 3% sucrose or other solution suitable for in vivo injection known to one skilled in the art. This solution may also contain adjuvant, for example aluminum hydroxide at a concentration of 0-0.5% (w/v). In some embodiments, for preparation of smEVs used for in vivo injections, smEVs in PBS are sterile-filtered to <0.22 um.


In certain embodiments, to make samples compatible with further testing (e.g., to remove sucrose prior to TEM imaging or in vitro assays), samples are buffer exchanged into PBS or 30 mM Tris, pH 8.0 using filtration (e.g., Amicon Ultra columns), dialysis, or ultracentrifugation (200,000×g, ≥3 hours, 4° C.) and resuspension.


In some embodiments, the sterility of the smEV preparations can be confirmed by plating a portion of the smEVs onto agar medium used for standard culture of the bacteria used in the generation of the smEVs and incubating using standard conditions.


In some embodiments, select smEVs are isolated and enriched by chromatography and binding surface moieties on smEVs. In other embodiments, select smEVs are isolated and/or enriched by fluorescent cell sorting by methods using affinity reagents, chemical dyes, recombinant proteins or other methods known to one skilled in the art.


The smEVs can be analyzed, e.g., as described in Jeppesen, et al. Cell 177:428 (2019).


In some embodiments, smEVs are lyophilized.


In some embodiments, smEVs are gamma irradiated (e.g., at 17.5 or 25 kGy).


In some embodiments, smEVs are UV irradiated.


In some embodiments, smEVs are heat inactivated (e.g., at 50° C. for two hours or at 90° C. for two hours).


In some embodiments, smEVs s are acid treated.


In some embodiments, smEVs are oxygen sparged (e.g., at 0.1 vvm for two hours).


The phase of growth can affect the amount or properties of bacteria and/or smEVs produced by bacteria. For example, in the methods of smEV preparation provided herein, smEVs can be isolated, e.g., from a culture, at the start of the log phase of growth, midway through the log phase, and/or once stationary phase growth has been reached.


The growth environment (e.g., culture conditions) can affect the amount of smEVs produced by bacteria. For example, the yield of smEVs can be increased by an smEV inducer, as provided in Table 4.









TABLE 4







Culture Techniques to Increase smEV Production









smEV
smEV



inducement
inducer
Acts on





Temperature
Heat
stress response



RT to 37° C. temp change
simulates infection



37 to 40° C. temp change
febrile infection


ROS
Plumbagin
oxidative stress response



Cumene hydroperoxide
oxidative stress response



Hydrogen Peroxide
oxidative stress response


Antibiotics
Ciprofloxacin
bacterial SOS response



Gentamycin
protein synthesis



Polymyxin B
outer membrane



D-cylcloserine
cell wall


Osmolyte
NaCl
osmotic stress


Metal Ion
Iron Chelation
iron levels


Stress
EDTA
removes divalent cations



Low Hemin
iron levels


Media additives


or removal


Other
Lactate
growth


mechanisms
Amino acid deprivation
stress



Hexadecane
stress



Glucose
growth



Sodium bicarbonate
ToxT induction



PQS
vesiculator



Diamines + DFMO
(from bacteria)



High nutrients
membrane anchoring



Low nutrients
(negativicutes only)



Oxygen
enhanced growth



No Cysteine
oxygen stress in anaerobe



Inducing biofilm or
oxygen stress in anaerobe



floculation



Diauxic Growth



Phage



Urea









In the methods of smEVs preparation provided herein, the method can optionally include exposing a culture of bacteria to an smEV inducer prior to isolating smEVs from the bacterial culture. The culture of bacteria can be exposed to an smEV inducer at the start of the log phase of growth, midway through the log phase, and/or once stationary phase growth has been reached.


Pharmaceutical Compositions

In certain embodiments, provided herein are pharmaceutical compositions comprising mEVs (such as smEVs) (e.g., an mEV composition (e.g., an smEV composition)). In some embodiments, the mEV composition comprises mEVs (such as smEVs) and/or a combination of mEVs (such as smEVs) described herein and a pharmaceutically acceptable carrier. In some embodiments, the smEV composition comprises smEVs and/or a combination of smEVs described herein and a pharmaceutically acceptable carrier.


In some embodiments, the pharmaceutical compositions comprise mEVs (such as smEVs) substantially or entirely free of whole bacteria (e.g., live bacteria, killed bacteria, attenuated bacteria). In some embodiments, the pharmaceutical compositions comprise both mEVs and whole bacteria (e.g., live bacteria, killed bacteria, attenuated bacteria). In some embodiments, the pharmaceutical compositions comprise mEVs from one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) of the bacteria strains or species listed in Table 1, Table 2, and/or Table 3. In some embodiments, the pharmaceutical compositions comprise mEVs from one of the bacteria strains or species listed in Table 1, Table 2, and/or Table 3. In some embodiments, the pharmaceutical composition comprises lyophilized mEVs (such as smEVs). In some embodiments, the pharmaceutical composition comprises gamma irradiated mEVs (such as smEVs). The mEVs (such as smEVs) can be gamma irradiated after the mEVs are isolated (e.g., prepared).


In some embodiments, to quantify the numbers of mEVs (such as smEVs) and/or bacteria present in a bacterial sample, electron microscopy (e.g., EM of ultrathin frozen sections) can be used to visualize the mEVs (such as smEVs) and/or bacteria and count their relative numbers. Alternatively, nanoparticle tracking analysis (NTA), Coulter counting, or dynamic light scattering (DLS) or a combination of these techniques can be used. NTA and the Coulter counter count particles and show their sizes. DLS gives the size distribution of particles, but not the concentration. Bacteria frequently have diameters of 1-2 um (microns). The full range is 0.2-20 um. Combined results from Coulter counting and NTA can reveal the numbers of bacteria and/or mEVs (such as smEVs) in a given sample. Coulter counting reveals the numbers of particles with diameters of 0.7-10 um. For most bacterial and/or mEV (such as smEV) samples, the Coulter counter alone can reveal the number of bacteria and/or mEVs (such as smEVs) in a sample. For NTA, a Nanosight instrument can be obtained from Malvern Pananlytical. For example, the NS300 can visualize and measure particles in suspension in the size range 10-2000 nm. NTA allows for counting of the numbers of particles that are, for example, 50-1000 nm in diameter. DLS reveals the distribution of particles of different diameters within an approximate range of 1 nm-3 um.


mEVs can be characterized by analytical methods known in the art (e.g., Jeppesen, et al. Cell 177:428 (2019)).


In some embodiments, the mEVs may be quantified based on particle count. For example, total protein content of an mEV preparation can be measured using NTA.


In some embodiments, the mEVs may be quantified based on the amount of protein, lipid, or carbohydrate. For example, total protein content of an mEV preparation can be measured using the Bradford assay.


In some embodiments, the mEVs are isolated away from one or more other bacterial components of the source bacteria. In some embodiments, the pharmaceutical composition further comprises other bacterial components.


In certain embodiments, the mEV preparation obtained from the source bacteria may be fractionated into subpopulations based on the physical properties (e.g., sized, density, protein content, binding affinity) of the subpopulations. One or more of the mEV subpopulations can then be incorporated into the pharmaceutical compositions of the invention.


In certain aspects, provided herein are pharmaceutical compositions comprising mEVs (such as smEVs) useful for the treatment and/or prevention of disease (e.g., a cancer, an autoimmune disease, an inflammatory disease, or a metabolic disease), as well as methods of making and/or identifying such mEVs, and methods of using such pharmaceutical compositions (e.g., for the treatment of a cancer, an autoimmune disease, an inflammatory disease, or a metabolic disease, either alone or in combination with other therapeutics). In some embodiments, the pharmaceutical compositions comprise both mEVs (such as smEVs), and whole bacteria (e.g., live bacteria, killed bacteria, attenuated bacteria). In some embodiments, the pharmaceutical compositions comprise mEVs (such as smEVs) in the absence of bacteria. In some embodiments, the pharmaceutical compositions comprise mEVs (such as smEVs) and/or bacteria from one or more of the bacteria strains or species listed in Table 1, Table 2, and/or Table 3. In some embodiments, the pharmaceutical compositions comprise mEVs (such as smEVs) and/or bacteria from one of the bacteria strains or species listed in Table 1, Table 2, and/or Table 3.


In certain aspects, provided are pharmaceutical compositions for administration to a subject (e.g., human subject). In some embodiments, the pharmaceutical compositions are combined with additional active and/or inactive materials in order to produce a final product, which may be in single dosage unit or in a multi-dose format. In some embodiments, the pharmaceutical composition is combined with an adjuvant such as an immuno-adjuvant (e.g., a STING agonist, a TLR agonist, or a NOD agonist).


In some embodiments, the pharmaceutical composition comprises at least one carbohydrate.


In some embodiments, the pharmaceutical composition comprises at least one lipid. In some embodiments the lipid comprises at least one fatty acid selected from lauric acid (12:0), myristic acid (14:0), palmitic acid (16:0), palmitoleic acid (16:1), margaric acid (17:0), heptadecenoic acid (17:1), stearic acid (18:0), oleic acid (18:1), linoleic acid (18:2), linolenic acid (18:3), octadecatetraenoic acid (18:4), arachidic acid (20:0), eicosenoic acid (20:1), eicosadienoic acid (20:2), eicosatetraenoic acid (20:4), eicosapentaenoic acid (20:5) (EPA), docosanoic acid (22:0), docosenoic acid (22:1), docosapentaenoic acid (22:5), docosahexaenoic acid (22:6) (DHA), and tetracosanoic acid (24:0).


In some embodiments, the pharmaceutical composition comprises at least one supplemental mineral or mineral source. Examples of minerals include, without limitation: chloride, sodium, calcium, iron, chromium, copper, iodine, zinc, magnesium, manganese, molybdenum, phosphorus, potassium, and selenium. Suitable forms of any of the foregoing minerals include soluble mineral salts, slightly soluble mineral salts, insoluble mineral salts, chelated minerals, mineral complexes, non-reactive minerals such as carbonyl minerals, and reduced minerals, and combinations thereof.


In some embodiments, the pharmaceutical composition comprises at least one supplemental vitamin. The at least one vitamin can be fat-soluble or water soluble vitamins. Suitable vitamins include but are not limited to vitamin C, vitamin A, vitamin E, vitamin B12, vitamin K, riboflavin, niacin, vitamin D, vitamin B6, folic acid, pyridoxine, thiamine, pantothenic acid, and biotin. Suitable forms of any of the foregoing are salts of the vitamin, derivatives of the vitamin, compounds having the same or similar activity of the vitamin, and metabolites of the vitamin.


In some embodiments, the pharmaceutical composition comprises an excipient. Non-limiting examples of suitable excipients include a buffering agent, a preservative, a stabilizer, a binder, a compaction agent, a lubricant, a dispersion enhancer, a disintegration agent, a flavoring agent, a sweetener, and a coloring agent.


In some embodiments, the excipient is a buffering agent. Non-limiting examples of suitable buffering agents include sodium citrate, magnesium carbonate, magnesium bicarbonate, calcium carbonate, and calcium bicarbonate.


In some embodiments, the excipient comprises a preservative. Non-limiting examples of suitable preservatives include antioxidants, such as alpha-tocopherol and ascorbate, and antimicrobials, such as parabens, chlorobutanol, and phenol.


In some embodiments, the pharmaceutical composition comprises a binder as an excipient. Non-limiting examples of suitable binders include starches, pregelatinized starches, gelatin, polyvinylpyrolidone, cellulose, methylcellulose, sodium carboxymethylcellulose, ethylcellulose, polyacrylamides, polyvinyloxoazolidone, polyvinylalcohols, C12-C18 fatty acid alcohol, polyethylene glycol, polyols, saccharides, oligosaccharides, and combinations thereof.


In some embodiments, the pharmaceutical composition comprises a lubricant as an excipient. Non-limiting examples of suitable lubricants include magnesium stearate, calcium stearate, zinc stearate, hydrogenated vegetable oils, sterotex, polyoxyethylene monostearate, talc, polyethyleneglycol, sodium benzoate, sodium lauryl sulfate, magnesium lauryl sulfate, and light mineral oil.


In some embodiments, the pharmaceutical composition comprises a dispersion enhancer as an excipient. Non-limiting examples of suitable dispersants include starch, alginic acid, polyvinylpyrrolidones, guar gum, kaolin, bentonite, purified wood cellulose, sodium starch glycolate, isoamorphous silicate, and microcrystalline cellulose as high HLB emulsifier surfactants.


In some embodiments, the pharmaceutical composition comprises a disintegrant as an excipient. In some embodiments the disintegrant is a non-effervescent disintegrant. Non-limiting examples of suitable non-effervescent disintegrants include starches such as corn starch, potato starch, pregelatinized and modified starches thereof, sweeteners, clays, such as bentonite, micro-crystalline cellulose, alginates, sodium starch glycolate, gums such as agar, guar, locust bean, karaya, pectin, and tragacanth. In some embodiments the disintegrant is an effervescent disintegrant. Non-limiting examples of suitable effervescent disintegrants include sodium bicarbonate in combination with citric acid, and sodium bicarbonate in combination with tartaric acid.


In some embodiments, the pharmaceutical composition is a food product (e.g., a food or beverage) such as a health food or beverage, a food or beverage for infants, a food or beverage for pregnant women, athletes, senior citizens or other specified group, a functional food, a beverage, a food or beverage for specified health use, a dietary supplement, a food or beverage for patients, or an animal feed. Specific examples of the foods and beverages include various beverages such as juices, refreshing beverages, tea beverages, drink preparations, jelly beverages, and functional beverages; alcoholic beverages such as beers; carbohydrate-containing foods such as rice food products, noodles, breads, and pastas; paste products such as fish hams, sausages, paste products of seafood; retort pouch products such as curries, food dressed with a thick starchy sauces, and Chinese soups; soups; dairy products such as milk, dairy beverages, ice creams, cheeses, and yogurts; fermented products such as fermented soybean pastes, yogurts, fermented beverages, and pickles; bean products; various confectionery products, including biscuits, cookies, and the like, candies, chewing gums, gummies, cold desserts including jellies, cream caramels, and frozen desserts; instant foods such as instant soups and instant soy-bean soups; microwavable foods; and the like. Further, the examples also include health foods and beverages prepared in the forms of powders, granules, tablets, capsules, liquids, pastes, and jellies.


In some embodiments, the pharmaceutical composition is a food product for animals, including humans. The animals, other than humans, are not particularly limited, and the composition can be used for various livestock, poultry, pets, experimental animals, and the like. Specific examples of the animals include pigs, cattle, horses, sheep, goats, chickens, wild ducks, ostriches, domestic ducks, dogs, cats, rabbits, hamsters, mice, rats, monkeys, and the like, but the animals are not limited thereto.


Dose Forms

A pharmaceutical composition comprising mEVs (such as smEVs) can be formulated as a solid dose form, e.g., for oral administration. The solid dose form can comprise one or more excipients, e.g., pharmaceutically acceptable excipients. The mEVs in the solid dose form can be isolated mEVs. Optionally, the mEVs in the solid dose form can be lyophilized. Optionally, the mEVs in the solid dose form are gamma irradiated. The solid dose form can comprise a tablet, a minitablet, a capsule, a pill, or a powder; or a combination of these forms (e.g., minitablets comprised in a capsule).


The solid dose form can comprise a tablet (e.g., >4 mm).


The solid dose form can comprise a mini tablet (e.g., 1-4 mm sized minitablet, e.g., a 2 mm minitablet or a 3 mm minitablet).


The solid dose form can comprise a capsule, e.g., a size 00, size 0, size 1, size 2, size 3, size 4, or size 5 capsule; e.g., a size 0 capsule.


The solid dose form can comprise a coating. The solid dose form can comprise a single layer coating, e.g., enteric coating, e.g., a Eudragit-based coating, e.g., EUDRAGIT L30 D-55, triethylcitrate, and talc. The solid dose form can comprise two layers of coating. For example, an inner coating can comprise, e.g., EUDRAGIT L30 D-55, triethylcitrate, talc, citric acid anhydrous, and sodium hydroxide, and an outer coating can comprise, e.g., EUDRAGIT L30 D-55, triethylcitrate, and talc. EUDRAGIT is the brand name for a diverse range of polymethacrylate-based copolymers. It includes anionic, cationic, and neutral copolymers based on methacrylic acid and methacrylic/acrylic esters or their derivatives. Eudragits are amorphous polymers having glass transition temperatures between 9 to >150° C. Eudragits are non-biodegradable, nonabsorbable, and nontoxic. Anionic Eudragit L dissolves at pH>6 and is used for enteric coating, while Eudragit S, soluble at pH>7 is used for colon targeting. Eudragit RL and RS, having quaternary ammonium groups, are water insoluble, but swellable/permeable polymers which are suitable for the sustained release film coating applications. Cationic Eudragit E, insoluble at pH≥5, can prevent drug release in saliva.


The solid dose form (e.g., a capsule) can comprise a single layer coating, e.g., a non-enteric coating such as HPMC (hydroxyl propyl methyl cellulose) or gelatin.


A pharmaceutical composition comprising mEVs (such as smEVs) can be formulated as a suspension, e.g., for oral administration or for injection. Administration by injection includes intravenous (IV), intramuscular (IM), intratumoral (IT) and subcutaneous (SC) administration. For a suspension, mEVs can be in a buffer, e.g., a pharmaceutically acceptable buffer, e.g., saline or PBS. The suspension can comprise one or more excipients, e.g., pharmaceutically acceptable excipients. The suspension can comprise, e.g., sucrose or glucose. The mEVs in the suspension can be isolated mEVs. Optionally, the mEVs in the suspension can be lyophilized. Optionally, the mEVs in the suspension can be gamma irradiated.


Dosage

For oral administration to a human subject, the dose of mEVs (such as smEVs) can be, e.g., about 2×106-about 2×1016 particles. The dose can be, e.g., about 1×107-about 1×1015, about 1×108-about 1×1014, about 1×109-about 1×1013, about 1×1010-about 1×1014, or about 1×108-about 1×1012 particles. The dose can be, e.g., about 2×106, about 2×107, about 2×108, about 2×109, about 1×1010, about 2×1010, about 2×1112, about 2×1012, about 2×1013, about 2×1014, or about 1×1015 particles. The dose can be, e.g., about 2×1014 particles. The dose can be, e.g., about 2×1012 particles. The dose can be, e.g., about 2×1010 particles. The dose can be, e.g., about 1×1010 particles. Particle count can be determined, e.g., by NTA.


For oral administration to a human subject, the dose of mEVs (such as smEVs) can be, e.g., based on total protein. The dose can be, e.g., about 5 mg to about 900 mg total protein. The dose can be, e.g., about 20 mg to about 800 mg, about 50 mg to about 700 mg, about 75 mg to about 600 mg, about 100 mg to about 500 mg, about 250 mg to about 750 mg, or about 200 mg to about 500 mg total protein. The dose can be, e.g., about 10 mg, about 25 mg, about 50 mg, about 75 mg, about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 400 mg, about 500 mg, about 600 mg, or about 750 mg total protein. Total protein can be determined, e.g., by Bradford assay.


For administration by injection (e.g., intravenous administration) to a human subject, the dose of mEVs (such as smEVs) can be, e.g., about 1×106-about 1×1016 particles. The dose can be, e.g., about 1×107-about 1×1015, about 1×108-about 1×1014, about 1×109-about 1×1013, about 1×1010°-about 1×1014, or about 1×108-about 1×1012 particles. The dose can be, e.g., about 2×106, about 2×107, about 2×108, about 2×109, about 1×1010, about 2×1010, about 2×1011, about 2×1012, about 2×1013, about 2×1014, or about 1×1015 particles. The dose can be, e.g., about 1×1015 particles. The dose can be, e.g., about 2×1014 particles. The dose can be, e.g., about 2×1013 particles. Particle count can be determined, e.g., by NTA.


For administration by injection (e.g., intravenous administration), the dose of mEVs (such as smEVs) can be, e.g., about 5 mg to about 900 mg total protein. The dose can be, e.g., about 20 mg to about 800 mg, about 50 mg to about 700 mg, about 75 mg to about 600 mg, about 100 mg to about 500 mg, about 250 mg to about 750 mg, or about 200 mg to about 500 mg total protein. The dose can be, e.g., about 10 mg, about 25 mg, about 50 mg, about 75 mg, about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 400 mg, about 500 mg, about 600 mg, or about 750 mg total protein. The dose can be, e.g., about 700 mg total protein. The dose can be, e.g., about 350 mg total protein. The dose can be, e.g., about 175 mg total protein. Total protein can be determined, e.g., by Bradford assay.


Gamma-Irradiation

Powders (e.g., of mEVs (such as smEVs)) can be gamma-irradiated at 17.5 kGy radiation unit at ambient temperature.


Frozen biomasses (e.g., of mEVs (such as smEVs)) can be gamma-irradiated at 25 kGy radiation unit in the presence of dry ice.


Additional Therapeutic Agents

In certain aspects, the methods provided herein include the administration to a subject of a pharmaceutical composition described herein either alone or in combination with an additional therapeutic agent. In some embodiments, the additional therapeutic agent is an immunosuppressant, an anti-inflammatory agent, a steroid, and/or a cancer therapeutic.


In some embodiments, the pharmaceutical composition comprising mEVs (such as smEVs) is administered to the subject before the additional therapeutic agent is administered (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 hours before or at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 days before). In some embodiments , the pharmaceutical composition comprising mEVs (such as smEVs) is administered to the subject after the additional therapeutic agent is administered (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 hours after or at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 days after). In some embodiments, the pharmaceutical composition comprising mEVs (such as smEVs) and the additional therapeutic agent are administered to the subject simultaneously or nearly simultaneously (e.g., administrations occur within an hour of each other).


In some embodiments, an antibiotic is administered to the subject before the pharmaceutical composition comprising mEVs (such as smEVs) is administered to the subject (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 hours before or at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 days before). In some embodiments, an antibiotic is administered to the subject after pharmaceutical composition comprising mEVs (such as smEVs) is administered to the subject (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 hours before or at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 days after). In some embodiments, the pharmaceutical composition comprising mEVs (such as smEVs) and the antibiotic are administered to the subject simultaneously or nearly simultaneously (e.g., administrations occur within an hour of each other).


In some embodiments, the additional therapeutic agent is a cancer therapeutic. In some embodiments, the cancer therapeutic is a chemotherapeutic agent. Examples of such chemotherapeutic agents include, but are not limited to, alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CB1-TM1); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin, especially calicheamicin gamma1I and calicheamicin omega1I; dynemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores, aclacinomysins, actinomycin, authrarnycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elformithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK polysaccharide complex); razoxane; rhizoxin; sizofuran; spirogermanium; tenuazonic acid; triaziquone; 2,2′,2″-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside (“Ara-C”); cyclophosphamide; thiotepa; taxoids, e.g., paclitaxel and doxetaxel; chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum coordination complexes such as cisplatin, oxaliplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitoxantrone; vincristine; vinorelbine; novantrone; teniposide; edatrexate; daunomycin; aminopterin; xeloda; ibandronate; irinotecan (e.g., CPT-11); topoisomerase inhibitor RFS 2000; difluoromethylomithine (DMFO); retinoids such as retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above.


In some embodiments, the cancer therapeutic is a cancer immunotherapy agent. Immunotherapy refers to a treatment that uses a subject's immune system to treat cancer, e.g., checkpoint inhibitors, cancer vaccines, cytokines, cell therapy, CAR-T cells, and dendritic cell therapy. Non-limiting examples of immunotherapies are checkpoint inhibitors include Nivolumab (BMS, anti-PD-1), Pembrolizumab (Merck, anti-PD-1), Ipilimumab (BMS, anti-CTLA-4), MEDI4736 (AstraZeneca, anti-PD-L1), and MPDL3280A (Roche, anti-PD-L1). Other immunotherapies may be tumor vaccines, such as Gardail, Cervarix, BCG, sipulencel-T, Gp100:209-217, AGS-003, DCVax-L, Algenpantucel-L, Tergenpantucel-L, TG4010, ProstAtak, Prostvac-V/R-TRICOM, Rindopepimul, E75 peptide acetate, IMA901, POL-103A, Belagenpumatucel-L, GSK1572932A, MDX-1279, GV1001, and Tecemotide. The immunotherapy agent may be administered via injection (e.g., intravenously, intratumorally, subcutaneously, or into lymph nodes), but may also be administered orally, topically, or via aerosol. Immunotherapies may comprise adjuvants such as cytokines.


In some embodiments, the immunotherapy agent is an immune checkpoint inhibitor. Immune checkpoint inhibition broadly refers to inhibiting the checkpoints that cancer cells can produce to prevent or downregulate an immune response. Examples of immune checkpoint proteins include, but are not limited to, CTLA4, PD-1, PD-L1, PD-L2, A2AR, B7-H3, B7-H4, BTLA, KIR, LAG3, TIM-3 or VISTA. Immune checkpoint inhibitors can be antibodies or antigen binding fragments thereof that bind to and inhibit an immune checkpoint protein. Examples of immune checkpoint inhibitors include, but are not limited to, nivolumab, pembrolizumab, pidilizumab, AMP-224, AMP-514, STI-A1110, TSR-042, RG-7446, BMS-936559, MEDI-4736, MSB-0010718C (avelumab), AUR-012 and STI-A1010.


In some embodiments, the methods provided herein include the administration of a pharmaceutical composition described herein in combination with one or more additional therapeutic agents. In some embodiments, the methods disclosed herein include the administration of two immunotherapy agents (e.g., immune checkpoint inhibitor). For example, the methods provided herein include the administration of a pharmaceutical composition described herein in combination with a PD-1 inhibitor (such as pemrolizumab or nivolumab or pidilizumab) or a CLTA-4 inhibitor (such as ipilimumab) or a PD-L1 inhibitor (such as avelumab).


In some embodiments, the immunotherapy agent is an antibody or antigen binding fragment thereof that, for example, binds to a cancer-associated antigen. Examples of cancer-associated antigens include, but are not limited to, adipophilin, AIM-2, ALDH1A1, alpha-actinin-4, alpha-fetoprotein (“AFP”), ARTC1, B-RAF, BAGE-1, BCLX (L), BCR-ABL fusion protein b3a2, beta-catenin, BING-4, CA-125, CALCA, carcinoembryonic antigen (“CEA”), CASP-5, CASP-8, CD274, CD45, Cdc27, CDK12, CDK4, CDKN2A, CEA, CLPP, COA-1, CPSF, CSNK1A1, CTAG1, CTAG2, cyclin D1, Cyclin-A1, dek-can fusion protein, DKK1, EFTUD2, Elongation factor 2, ENAH (hMena), Ep-CAM, EpCAM, EphA3, epithelial tumor antigen (“ETA”), ETV6-AML1 fusion protein, EZH2, FGF5, FLT3-ITD, FN1, G250/MN/CAIX, GAGE-1,2,8, GAGE-3,4,5,6,7, GAS7, glypican-3, GnTV, gp100/Pme117, GPNMB, HAUS3, Hepsin, HER-2/neu, HERV-K-MEL, HLA-A11, HLA-A2, HLA-DOB, hsp70-2, IDO1, IGF2B3, IL13Ralpha2, Intestinal carboxyl esterase, K-ras, Kallikrein 4, KIF20A, KK-LC-1, KKLC1, KM-HN-1, KMHN1 also known as CCDC110, LAGE-1, LDLR-fucosyltransferaseAS fusion protein, Lengsin, M-CSF, MAGE-A1, MAGE-A10, MAGE-A12, MAGE-A2, MAGE-A3, MAGE-A4, MAGE-A6, MAGE-A9, MAGE-C1, MAGE-C2, malic enzyme, mammaglobin-A, MART2, MATN, MC1R, MCSP, mdm-2, ME1, Melan-A/MART-1, Meloe, Midkine, MMP-2, MUC1, MUC5AC, mucin, MUM-1, MUM-2, MUM-3, Myosin, Myosin class I, N-raw, NA88-A, neo-PAP, NFYC, NY-BR-1, NY-ESO-1/LAGE-2, OA1, OGT, OS-9, P polypeptide, p53, PAP, PAX5, PBF, pml-RARalpha fusion protein, polymorphic epithelial mucin (“PEM”), PPP1R3B, PRAME, PRDX5, PSA, PSMA, PTPRK, RAB38/NY-MEL-1, RAGE-1, RBAF600, RGS5, RhoC, RNF43, RU2AS, SAGE, secernin 1, SIRT2, SNRPD1, SOX10, Sp17, SPA17, SSX-2, SSX-4, STEAP1, survivin, SYT-SSX1 or -SSX2 fusion protein, TAG-1, TAG-2, Telomerase, TGF-betaRH, TPBG, TRAG-3, Triosephosphate isomerase, TRP-1/gp75, TRP-2, TRP2-INT2, tyrosinase, tyrosinase (“TYR”), VEGF, WT1, XAGE-1b/GAGED2a. In some embodiments, the antigen is a neo-antigen.


In some embodiments, the immunotherapy agent is a cancer vaccine and/or a component of a cancer vaccine (e.g., an antigenic peptide and/or protein). The cancer vaccine can be a protein vaccine, a nucleic acid vaccine or a combination thereof. For example, in some embodiments, the cancer vaccine comprises a polypeptide comprising an epitope of a cancer-associated antigen. In some embodiments, the cancer vaccine comprises a nucleic acid (e.g., DNA or RNA, such as mRNA) that encodes an epitope of a cancer-associated antigen. Examples of cancer-associated antigens include, but are not limited to, adipophilin, AIM-2, ALDH1A1, alpha-actinin-4, alpha-fetoprotein (“AFP”), ARTC1, B-RAF, BAGE-1, BCLX (L), BCR-ABL fusion protein b3a2, beta-catenin, BING-4, CA-125, CALCA, carcinoembryonic antigen (“CEA”), CASP-5, CASP-8, CD274, CD45, Cdc27, CDK12, CDK4, CDKN2A, CEA, CLPP, COA-1, CPSF, CSNK1A1, CTAG1, CTAG2, cyclin D1, Cyclin-A1, dek-can fusion protein, DKK1, EFTUD2, Elongation factor 2, ENAH (hMena), Ep-CAM, EpCAM, EphA3, epithelial tumor antigen (“ETA”), ETV6-AML1 fusion protein, EZH2, FGF5, FLT3-ITD, FN1, G250/MN/CAIX, GAGE-1,2,8, GAGE-3,4,5,6,7, GAS7, glypican-3, GnTV, gp100/Pmel17, GPNMB, HAUS3, Hepsin, HER-2/neu, HERV-K-MEL, HLA-A11, HLA-A2, HLA-DOB, hsp70-2, IDO1, IGF2B3, IL13Ralpha2, Intestinal carboxyl esterase, K-ras, Kallikrein 4, KIF20A, KK-LC-1, KKLC1, KM-HN-1, KMHN1 also known as CCDC110, LAGE-1, LDLR-fucosyltransferaseAS fusion protein, Lengsin, M-CSF, MAGE-A1, MAGE-A10, MAGE-A12, MAGE-A2, MAGE-A3, MAGE-A4, MAGE-A6, MAGE-A9, MAGE-C1, MAGE-C2, malic enzyme, mammaglobin-A, MART2, MATN, MC1R, MCSP, mdm-2, MEL Melan-A/MART-1, Meloe, Midkine, MMP-2, MMP-7, MUC1, MUC5AC, mucin, MUM-1, MUM-2, MUM-3, Myosin, Myosin class I, N-raw, NA88-A, neo-PAP, NFYC, NY-BR-1, NY-ESO-1/LAGE-2, OA1, OGT, OS-9, P polypeptide, p53, PAP, PAX5, PBF, pml-RARalpha fusion protein, polymorphic epithelial mucin (“PEM”), PPP1R3B, PRAME, PRDX5, PSA, PSMA, PTPRK, RAB38/NY-MEL-1, RAGE-1, RBAF600, RGS5, RhoC, RNF43, RU2AS, SAGE, secernin 1, SIRT2, SNRPD1, SOX10, Sp17, SPA17, SSX-2, SSX-4, STEAP1, survivin, SYT-SSX1 or -SSX2 fusion protein, TAG-1, TAG-2, Telomerase, TGF-betaRII, TPBG, TRAG-3, Triosephosphate isomerase, TRP-1/gp75, TRP-2, TRP2-INT2, tyrosinase, tyrosinase (“TYR”), VEGF, WT1, XAGE-1b/GAGED2a. In some embodiments, the antigen is a neo-antigen. In some embodiments, the cancer vaccine is administered with an adjuvant. Examples of adjuvants include, but are not limited to, an immune modulatory protein, Adjuvant 65, α-GalCer, aluminum phosphate, aluminum hydroxide, calcium phosphate, β-Glucan Peptide, CpG ODN DNA, GPI-0100, lipid A, lipopolysaccharide, Lipovant, Montanide, N-acetyl-muramyl-L-alanyl-D-isoglutamine, Pam3CSK4, quil A, cholera toxin (CT) and heat-labile toxin from enterotoxigenic Escherichia coli (LT) including derivatives of these (CM, mmCT, CTA1-DD, LTB, LTK63, LTR72, dmLT) and trehalose dimycolate.


In some embodiments, the immunotherapy agent is an immune modulating protein to the subject. In some embodiments, the immune modulatory protein is a cytokine or chemokine. Examples of immune modulating proteins include, but are not limited to, B lymphocyte chemoattractant (“BLC”), C—C motif chemokine 11 (“Eotaxin-1”), Eosinophil chemotactic protein 2 (“Eotaxin-2”), Granulocyte colony-stimulating factor (“G-CSF”), Granulocyte macrophage colony-stimulating factor (“GM-CSF”), 1-309, Intercellular Adhesion Molecule 1 (“ICAM-1”), Interferon alpha (“IFN-alpha”), Interferon beta (“IFN-beta”) Interferon gamma (“IFN-gamma”), Interlukin-1 alpha (“IL-1 alpha”), Interlukin-1 beta (“IL-1 beta”), Interleukin 1 receptor antagonist (“IL-1 ra”), Interleukin-2 (“IL-2”), Interleukin-4 (“IL-4”), Interleukin-5 (“IL-5”), Interleukin-6 (“IL-6”), Interleukin-6 soluble receptor (“IL-6 sR”), Interleukin-7 (“IL-7”), Interleukin-8 (“IL-8”), Interleukin-10 (“IL-10”), Interleukin-11 (“IL-11”), Subunit beta of Interleukin-12 (“IL-12 p40” or “IL-12 p70”), Interleukin-13 (“IL-13”), Interleukin-15 (“IL-15”), Interleukin-16 (“IL-16”), Interleukin-17A-F (“IL-17A-F”), Interleukin-18 (“IL-18”), Interleukin-21 (“IL-21”), Interleukin-22 (“IL-22”), Interleukin-23 (“IL-23”), Interleukin-33 (“IL-33”), Chemokine (C—C motif) Ligand 2 (“MCP-1”), Macrophage colony-stimulating factor (“M-CSF”), Monokine induced by gamma interferon (“MIG”), Chemokine (C—C motif) ligand 2 (“MIP-1 alpha”), Chemokine (C—C motif) ligand 4 (“MIP-1 beta”), Macrophage inflammatory protein-1-delta (“MIP-1 delta”), Platelet-derived growth factor subunit B (“PDGF-BB”), Chemokine (C—C motif) ligand 5, Regulated on Activation, Normal T cell Expressed and Secreted (“RANTES”), TIMP metallopeptidase inhibitor 1 (“TIMP-1”), TIMP metallopeptidase inhibitor 2 (“TIMP-2”), Tumor necrosis factor, lymphotoxin-alpha (“TNF alpha”), Tumor necrosis factor, lymphotoxin-beta (“TNF beta”), Soluble TNF receptor type 1 (“sTNFRI”), sTNFRIIAR, Brain-derived neurotrophic factor (“BDNF”), Basic fibroblast growth factor (“bFGF”), Bone morphogenetic protein 4 (“BMP-4”), Bone morphogenetic protein 5 (“BMP-5”), Bone morphogenetic protein 7 (“BMP-7”), Nerve growth factor (“b-NGF”), Epidermal growth factor (“EGF”), Epidermal growth factor receptor (“EGFR”), Endocrine-gland-derived vascular endothelial growth factor (“EG-VEGF”), Fibroblast growth factor 4 (“FGF-4”), Keratinocyte growth factor (“FGF-7”), Growth differentiation factor 15 (“GDF-15”), Glial cell-derived neurotrophic factor (“GDNF”), Growth Hormone, Heparin-binding EGF-like growth factor (“HB-EGF”), Hepatocyte growth factor (“HGF”), Insulin-like growth factor binding protein 1 (“IGFBP-1”), Insulin-like growth factor binding protein 2 (“IGFBP-2”), Insulin-like growth factor binding protein 3 (“IGFBP-3”), Insulin-like growth factor binding protein 4 (“IGFBP-4”), Insulin-like growth factor binding protein 6 (“IGFBP-6”), Insulin-like growth factor 1 (“IGF-1”), Insulin, Macrophage colony-stimulating factor (“M-CSF R”), Nerve growth factor receptor (“NGF R”), Neurotrophin-3 (“NT-3”), Neurotrophin-4 (“NT-4”), Osteoclastogenesis inhibitory factor (“Osteoprotegerin”), Platelet-derived growth factor receptors (“PDGF-AA”), Phosphatidylinositol-glycan biosynthesis (“PIGF”), Skp, Cullin, F-box containing comples (“SCF”), Stem cell factor receptor (“SCF R”), Transforming growth factor alpha (“TGFalpha”), Transforming growth factor beta-1 (“TGF beta 1”), Transforming growth factor beta-3 (“TGF beta 3”), Vascular endothelial growth factor (“VEGF”), Vascular endothelial growth factor receptor 2 (“VEGFR2”), Vascular endothelial growth factor receptor 3 (“VEGFR3”), VEGF-D 6Ckine, Tyrosine-protein kinase receptor UFO (“Axl”), Betacellulin (“BTC”), Mucosae-associated epithelial chemokine (“CCL28”), Chemokine (C—C motif) ligand 27 (“CTACK”), Chemokine (C—X—C motif) ligand 16 (“CXCL16”), C—X—C motif chemokine 5 (“ENA-78”), Chemokine (C—C motif) ligand 26 (“Eotaxin-3”), Granulocyte chemotactic protein 2 (“GCP-2”), GRO, Chemokine (C—C motif) ligand 14 (“HCC-1”), Chemokine (C—C motif) ligand 16 (“HCC-4”), Interleukin-9 (“IL-9”), Interleukin-17 F (“IL-17F”), Interleukin-18-binding protein (“IL-18 BPa”), Interleukin-28 A (“IL-28A”), Interleukin 29 (“IL-29”), Interleukin 31 (“IL-31”), C—X—C motif chemokine 10 (“IP-10”), Chemokine receptor CXCR3 (“I-TAC”), Leukemia inhibitory factor (“LIF”), Light, Chemokine (C motif) ligand (“Lymphotactin”), Monocyte chemoattractant protein 2 (“MCP-2”), Monocyte chemoattractant protein 3 (“MCP-3”), Monocyte chemoattractant protein 4 (“MCP-4”), Macrophage-derived chemokine (“MDC”), Macrophage migration inhibitory factor (“MIF”), Chemokine (C—C motif) ligand 20 (“MIP-3 alpha”), C—C motif chemokine 19 (“MIP-3 beta”), Chemokine (C—C motif) ligand 23 (“MPIF-1”), Macrophage stimulating protein alpha chain (“MSPalpha”), Nucleosome assembly protein 1-like 4 (“NAP-2”), Secreted phosphoprotein 1 (“Osteopontin”), Pulmonary and activation-regulated cytokine (“PARC”), Platelet factor 4 (“PF4”), Stroma cell-derived factor-1 alpha (“SDF-1 alpha”), Chemokine (C—C motif) ligand 17 (“TARC”), Thymus-expressed chemokine (“TECK”), Thymic stromal lymphopoietin (“TSLP 4-IBB”), CD 166 antigen (“ALCAM”), Cluster of Differentiation 80 (“B7-1”), Tumor necrosis factor receptor superfamily member 17 (“BCMA”), Cluster of Differentiation 14 (“CD14”), Cluster of Differentiation 30 (“CD30”), Cluster of Differentiation 40 (“CD40 Ligand”), Carcinoembryonic antigen-related cell adhesion molecule 1 (biliary glycoprotein) (“CEACANI-1”), Death Receptor 6 (“DR6”), Deoxythymidine kinase (“Dtk”), Type 1 membrane glycoprotein (“Endoglin”), Receptor tyrosine-protein kinase erbB-3 (“ErbB3”), Endothelial-leukocyte adhesion molecule 1 (“E-Selectin”), Apoptosis antigen 1 (“Fas”), Fms-like tyrosine kinase 3 (“Flt-3L”), Tumor necrosis factor receptor superfamily member 1 (“GITR”), Tumor necrosis factor receptor superfamily member 14 (“HVEM”), Intercellular adhesion molecule 3 (“ICAM-3”), IL-1 R4, IL-1 RI, IL-10 Rbeta, IL-17R, IL-2Rgamma, IL-21R, Lysosome membrane protein 2 (“LIMPII”), Neutrophil gelatinase-associated lipocalin (“Lipocalin-2”), CD62L (“L-Selectin”), Lymphatic endothelium (“LYVE-1”), MHC class I polypeptide-related sequence A (“MICA”), MHC class I polypeptide-related sequence B (“MICB”), NRG1-beta1, Beta-type platelet-derived growth factor receptor (“PDGF Rbeta”), Platelet endothelial cell adhesion molecule (“PECANI-1”), RAGE, Hepatitis A virus cellular receptor 1 (“TIM-1”), Tumor necrosis factor receptor superfamily member IOC (“TRAIL R3”), Trappin protein transglutaminase binding domain (“Trappin-2”), Urokinase receptor (“uPAR”), Vascular cell adhesion protein 1 (“VCAM-1”), XEDARActivin A, Agouti-related protein (“AgRP”), Ribonuclease 5 (“Angiogenin”), Angiopoietin 1, Angiostatin, Catheprin S, CD40, Cryptic family protein IB (“Cripto-1”), DAN, Dickkopf-related protein 1 (“DKK-1”), E-Cadherin, Epithelial cell adhesion molecule (“EpCAM”), Fas Ligand (FasL or CD95L), Fcg MIB/C, Follistatin, Galectin-7, Intercellular adhesion molecule 2 (“ICAM-2”), IL-13 R1, IL-13R2, IL-17B, IL-2 Ra, IL-2 Rb, IL-23, LAP, Neuronal cell adhesion molecule (“NrCAM”), Plasminogen activator inhibitor-1 (“PM-1”), Platelet derived growth factor receptors (“PDGF-AB”), Resistin, stromal cell-derived factor 1 (“SDF-1 beta”), sgp130, Secreted frizzled-related protein 2 (“ShhN”), Sialic acid-binding immunoglobulin-type lectins (“Siglec-5”), ST2, Transforming growth factor-beta 2 (“TGF beta 2”), Tie-2, Thrombopoietin (“TPO”), Tumor necrosis factor receptor superfamily member 10D (“TRAIL R4”), Triggering receptor expressed on myeloid cells 1 (“TREM-1”), Vascular endothelial growth factor C (“VEGF-C”), VEGFR1Adiponectin, Adipsin (“AND”), Alpha-fetoprotein (“AFP”), Angiopoietin-like 4 (“ANGPTL4”), Beta-2-microglobulin (“B2M”), Basal cell adhesion molecule (“BCAM”), Carbohydrate antigen 125 (“CA125”), Cancer Antigen 15-3 (“CA15-3”), Carcinoembryonic antigen (“CEA”), cAMP receptor protein (“CRP”), Human Epidermal Growth Factor Receptor 2 (“ErbB2”), Follistatin, Follicle-stimulating hormone (“FSH”), Chemokine (C—X—C motif) ligand 1 (“GRO alpha”), human chorionic gonadotropin (“beta HCG”), Insulin-like growth factor 1 receptor (“IGF-1 sR”), IL-1 sRII, IL-3, IL-18 Rb, IL-21, Leptin, Matrix metalloproteinase-1 (“MMP-1”), Matrix metalloproteinase-2 (“MMP-2”), Matrix metalloproteinase-3 (“MMP-3”), Matrix metalloproteinase-8 (“MMP-8”), Matrix metalloproteinase-9 (“MMP-9”), Matrix metalloproteinase-10 (“MMP-10”), Matrix metalloproteinase-13 (“MMP-13”), Neural Cell Adhesion Molecule (“NCAM-1”), Entactin (“Nidogen-1”), Neuron specific enolase (“NSE”), Oncostatin M (“OSM”), Procalcitonin, Prolactin, Prostate specific antigen (“PSA”), Sialic acid-binding Ig-like lectin 9 (“Siglec-9”), ADAM 17 endopeptidase (“TACE”), Thyroglobulin, Metalloproteinase inhibitor 4 (“TIMP-4”), TSH2B4, Disintegrin and metalloproteinase domain-containing protein 9 (“ADAM-9”), Angiopoietin 2, Tumor necrosis factor ligand superfamily member 13/Acidic leucine-rich nuclear phosphoprotein 32 family member B (“APRIL”), Bone morphogenetic protein 2 (“BMP-2”), Bone morphogenetic protein 9 (“BMP-9”), Complement component 5a (“C5a”), Cathepsin L, CD200, CD97, Chemerin, Tumor necrosis factor receptor superfamily member 6B (“DcR3”), Fatty acid-binding protein 2 (“FABP2”), Fibroblast activation protein, alpha (“FAP”), Fibroblast growth factor 19 (“FGF-19”), Galectin-3, Hepatocyte growth factor receptor (“HGF R”), IFN-gammalpha/beta R2, Insulin-like growth factor 2 (“IGF-2”), Insulin-like growth factor 2 receptor (“IGF-2 R”), Interleukin-1 receptor 6 (“IL-1R6”), Interleukin 24 (“IL-24”), Interleukin 33 (“IL-33”, Kallikrein 14, Asparaginyl endopeptidase (“Legumain”), Oxidized low-density lipoprotein receptor 1 (“LOX-1”), Mannose-binding lectin (“MBL”), Neprilysin (“NEP”), Notch homolog 1, translocation-associated (Drosophila) (“Notch-1”), Nephroblastoma overexpressed (“NOV”), Osteoactivin, Programmed cell death protein 1 (“PD-1”), N-acetylmuramoyl-L-alanine amidase (“PGRP-5”), Serpin A4, Secreted frizzled related protein 3 (“sFRP-3”), Thrombomodulin, Tolllike receptor 2 (“TLR2”), Tumor necrosis factor receptor superfamily member 10A (“TRAIL R1”), Transferrin (“TRF”), WIF-1ACE-2, Albumin, AMICA, Angiopoietin 4, B-cell activating factor (“BAFF”), Carbohydrate antigen 19-9 (“CA19-9”), CD 163, Clusterin, CRT AM, Chemokine (C—X—C motif) ligand 14 (“CXCL14”), Cystatin C, Decorin (“DCN”), Dickkopf-related protein 3 (“Dkk-3”), Delta-like protein 1 (“DLL1”), Fetuin A, Heparin-binding growth factor 1 (“aFGF”), Folate receptor alpha (“FOLR1”), Furin, GPCR-associated sorting protein 1 (“GASP-1”), GPCR-associated sorting protein 2 (“GASP-2”), Granulocyte colony-stimulating factor receptor (“GCSF R”), Serine protease hepsin (“HAI-2”), Interleukin-17B Receptor (“IL-17B R”), Interleukin 27 (“IL-27”), Lymphocyte-activation gene 3 (“LAG-3”), Apolipoprotein A-V (“LDL R”), Pepsinogen I, Retinol binding protein 4 (“RBP4”), SOST, Heparan sulfate proteoglycan (“Syndecan-1”), Tumor necrosis factor receptor superfamily member 13B (“TACI”), Tissue factor pathway inhibitor (“TFPI”), TSP-1, Tumor necrosis factor receptor superfamily, member 10b (“TRAIL R2”), TRANCE, Troponin I, Urokinase Plasminogen Activator (“uPA”), Cadherin 5, type 2 or VE-cadherin (vascular endothelial) also known as CD144 (“VE-Cadherin”), WNT1-inducible-signaling pathway protein 1 (“WISP-1”), and Receptor Activator of Nuclear Factor κB (“RANK”).


In some embodiments, the cancer therapeutic is an anti-cancer compound. Exemplary anti-cancer compounds include, but are not limited to, Alemtuzumab (Campath®), Alitretinoin (Panretin®), Anastrozole (Arimidex®), Bevacizumab (Avastin®), Bexarotene (Targretin®), Bortezomib (Velcade®), Bosutinib (Bosulif®), Brentuximab vedotin (Adcetris®), Cabozantinib (Cometriq™), Carfilzomib (Kyprolis™), Cetuximab (Erbitux®), Crizotinib (Xalkori®), Dasatinib (Sprycel®), Denileukin diftitox (Ontak®), Erlotinib hydrochloride (Tarceva®), Everolimus (Afinitor®), Exemestane (Aromasin®), Fulvestrant (Faslodex®), Gefitinib (Iressa®), Ibritumomab tiuxetan (Zevalin®), Imatinib mesylate (Gleevec®), Ipilimumab (Yervoy™), Lapatinib ditosylate (Tykerb®), Letrozole (Femara®), Nilotinib (Tasigna®), Ofatumumab (Arzerra®), Panitumumab (Vectibix®), Pazopanib hydrochloride (Votrient®), Pertuzumab (Perjeta™), Pralatrexate (Folotyn®), Regorafenib (Stivarga®), Rituximab (Rituxan®), Romidepsin (Istodax®), Sorafenib tosylate (Nexavar®), Sunitinib malate (Sutent®), Tamoxifen, Temsirolimus (Torisel®), Toremifene (Fareston®), Tositumomab and 131I-tositumomab (Bexxar®), Trastuzumab (Herceptin®), Tretinoin (Vesanoid®), Vandetanib (Caprelsa®), Vemurafenib (Zelboraf®), Vorinostat (Zolinza®), and Ziv-aflibercept (Zaltrap®).


Exemplary anti-cancer compounds that modify the function of proteins that regulate gene expression and other cellular functions (e.g., HDAC inhibitors, retinoid receptor ligants) are Vorinostat (Zolinza®), Bexarotene (Targretin®) and Romidepsin (Istodax®), Alitretinoin (Panretin®), and Tretinoin (Vesanoid®).


Exemplary anti-cancer compounds that induce apoptosis (e.g., proteasome inhibitors, antifolates) are Bortezomib (Velcade®), Carfilzomib (Kyprolis™), and Pralatrexate (Folotyn®).


Exemplary anti-cancer compounds that increase anti-tumor immune response (e.g., anti CD20, anti CD52; anti-cytotoxic T-lymphocyte-associated antigen-4) are Rituximab (Rituxan®), Alemtuzumab (Campath®), Ofatumumab (Arzerra®), and Ipilimumab (Yervoy™).


Exemplary anti-cancer compounds that deliver toxic agents to cancer cells (e.g., anti-CD20-radionuclide fusions; IL-2-diphtheria toxin fusions; anti-CD30-monomethylauristatin E (MMAE)-fusions) are Tositumomab and 131I-tositumomab (Bexxar®) and Ibritumomab tiuxetan (Zevalin®), Denileukin diftitox (Ontak®), and Brentuximab vedotin (Adcetris®).


Other exemplary anti-cancer compounds are small molecule inhibitors and conjugates thereof of, e.g., Janus kinase, ALK, Bcl-2, PARP, PI3K, VEGF receptor, Braf, MEK, CDK, and HSP90.


Exemplary platinum-based anti-cancer compounds include, for example, cisplatin, carboplatin, oxaliplatin, satraplatin, picoplatin, Nedaplatin, Triplatin, and Lipoplatin. Other metal-based drugs suitable for treatment include, but are not limited to ruthenium-based compounds, ferrocene derivatives, titanium-based compounds, and gallium-based compounds.


In some embodiments, the cancer therapeutic is a radioactive moiety that comprises a radionuclide. Exemplary radionuclides include, but are not limited to Cr-51, Cs-131, Ce-134, Se-75, Ru-97, I-125, Eu-149, Os-189m, Sb-119, I-123, Ho-161, Sb-117, Ce-139, In-111, Rh-103m, Ga-67, Tl-201, Pd-103, Au-195, Hg-197, Sr-87m, Pt-191, P-33, Er-169, Ru-103, Yb-169, Au-199, Sn-121, Tm-167, Yb-175, In-113m, Sn-113, Lu-177, Rh-105, Sn-117m, Cu-67, Sc-47, Pt-195m, Ce-141, I-131, Tb-161, As-77, Pt-197, Sm-153, Gd-159, Tm-173, Pr-143, Au-198, Tm-170, Re-186, Ag-111, Pd-109, Ga-73, Dy-165, Pm-149, Sn-123, Sr-89, Ho-166, P-32, Re-188, Pr-142, Ir-194, In-114m/In-114, and Y-90.


In some embodiments, the cancer therapeutic is an antibiotic. For example, if the presence of a cancer-associated bacteria and/or a cancer-associated microbiome profile is detected according to the methods provided herein, antibiotics can be administered to eliminate the cancer-associated bacteria from the subject. “Antibiotics” broadly refers to compounds capable of inhibiting or preventing a bacterial infection. Antibiotics can be classified in a number of ways, including their use for specific infections, their mechanism of action, their bioavailability, or their spectrum of target microbe (e.g., Gram-negative vs. Gram-positive bacteria, aerobic vs. anaerobic bacteria, etc.) and these may be used to kill specific bacteria in specific areas of the host (“niches”) (Leekha, et al 2011. General Principles of Antimicrobial Therapy. Mayo Clin Proc. 86(2): 156-167). In certain embodiments, antibiotics can be used to selectively target bacteria of a specific niche. In some embodiments, antibiotics known to treat a particular infection that includes a cancer niche may be used to target cancer-associated microbes, including cancer-associated bacteria in that niche. In other embodiments, antibiotics are administered after the pharmaceutical composition comprising mEVs (such as smEVs). In some embodiments, antibiotics are administered before pharmaceutical composition comprising mEVs (such as smEVs).


In some aspects, antibiotics can be selected based on their bactericidal or bacteriostatic properties. Bactericidal antibiotics include mechanisms of action that disrupt the cell wall (e.g., β-lactams), the cell membrane (e.g., daptomycin), or bacterial DNA (e.g., fluoroquinolones). Bacteriostatic agents inhibit bacterial replication and include sulfonamides, tetracyclines, and macrolides, and act by inhibiting protein synthesis. Furthermore, while some drugs can be bactericidal in certain organisms and bacteriostatic in others, knowing the target organism allows one skilled in the art to select an antibiotic with the appropriate properties. In certain treatment conditions, bacteriostatic antibiotics inhibit the activity of bactericidal antibiotics. Thus, in certain embodiments, bactericidal and bacteriostatic antibiotics are not combined.


Antibiotics include, but are not limited to aminoglycosides, ansamycins, carbacephems, carbapenems, cephalosporins, glycopeptides, lincosamides, lipopeptides, macrolides, monobactams, nitrofurans, oxazolidonones, penicillins, polypeptide antibiotics, quinolones, fluoroquinolone, sulfonamides, tetracyclines, and anti-mycobacterial compounds, and combinations thereof.


Aminoglycosides include, but are not limited to Amikacin, Gentamicin, Kanamycin, Neomycin, Netilmicin, Tobramycin, Paromomycin, and Spectinomycin. Aminoglycosides are effective, e.g., against Gram-negative bacteria, such as Escherichia coli, Klebsiella, Pseudomonas aeruginosa, and Francisella tularensis, and against certain aerobic bacteria but less effective against obligate/facultative anaerobes. Aminoglycosides are believed to bind to the bacterial 30S or 50S ribosomal subunit thereby inhibiting bacterial protein synthesis.


Ansamycins include, but are not limited to, Geldanamycin, Herbimycin, Rifamycin, and Streptovaricin. Geldanamycin and Herbimycin are believed to inhibit or alter the function of Heat Shock Protein 90.


Carbacephems include, but are not limited to, Loracarbef. Carbacephems are believed to inhibit bacterial cell wall synthesis.


Carbapenems include, but are not limited to, Ertapenem, Doripenem, Imipenem/Cilastatin, and Meropenem. Carbapenems are bactericidal for both Gram-positive and Gram-negative bacteria as broad-spectrum antibiotics. Carbapenems are believed to inhibit bacterial cell wall synthesis.


Cephalosporins include, but are not limited to, Cefadroxil, Cefazolin, Cefalotin, Cefalothin, Cefalexin, Cefaclor, Cefamandole, Cefoxitin, Cefprozil, Cefuroxime, Cefixime, Cefdinir, Cefditoren, Cefoperazone, Cefotaxime, Cefpodoxime, Ceftazidime, Ceftibuten, Ceftizoxime, Ceftriaxone, Cefepime, Ceftaroline fosamil, and Ceftobiprole. Selected Cephalosporins are effective, e.g., against Gram-negative bacteria and against Gram-positive bacteria, including Pseudomonas, certain Cephalosporins are effective against methicillin-resistant Staphylococcus aureus (MRSA). Cephalosporins are believed to inhibit bacterial cell wall synthesis by disrupting synthesis of the peptidoglycan layer of bacterial cell walls.


Glycopeptides include, but are not limited to, Teicoplanin, Vancomycin, and Telavancin. Glycopeptides are effective, e.g., against aerobic and anaerobic Gram-positive bacteria including MRSA and Clostridium difficile. Glycopeptides are believed to inhibit bacterial cell wall synthesis by disrupting synthesis of the peptidoglycan layer of bacterial cell walls.


Lincosamides include, but are not limited to, Clindamycin and Lincomycin. Lincosamides are effective, e.g., against anaerobic bacteria, as well as Staphylococcus, and Streptococcus. Lincosamides are believed to bind to the bacterial 50S ribosomal subunit thereby inhibiting bacterial protein synthesis.


Lipopeptides include, but are not limited to, Daptomycin. Lipopeptides are effective, e.g., against Gram-positive bacteria. Lipopeptides are believed to bind to the bacterial membrane and cause rapid depolarization.


Macrolides include, but are not limited to, Azithromycin, Clarithromycin, Dirithromycin, Erythromycin, Roxithromycin, Troleandomycin, Telithromycin, and Spiramycin. Macrolides are effective, e.g., against Streptococcus and Mycoplasma. Macrolides are believed to bind to the bacterial or 50S ribosomal subunit, thereby inhibiting bacterial protein synthesis.


Monobactams include, but are not limited to, Aztreonam. Monobactams are effective, e.g., against Gram-negative bacteria. Monobactams are believed to inhibit bacterial cell wall synthesis by disrupting synthesis of the peptidoglycan layer of bacterial cell walls.


Nitrofurans include, but are not limited to, Furazolidone and Nitrofurantoin.


Oxazolidonones include, but are not limited to, Linezolid, Posizolid, Radezolid, and Torezolid. Oxazolidonones are believed to be protein synthesis inhibitors.


Penicillins include, but are not limited to, Amoxicillin, Ampicillin, Azlocillin, Carbenicillin, Cloxacillin, Dicloxacillin, Flucloxacillin, Mezlocillin, Methicillin, Nafcillin, Oxacillin, Penicillin G, Penicillin V, Piperacillin, Temocillin and Ticarcillin. Penicillins are effective, e.g., against Gram-positive bacteria, facultative anaerobes, e.g., Streptococcus, Borrelia, and Treponema. Penicillins are believed to inhibit bacterial cell wall synthesis by disrupting synthesis of the peptidoglycan layer of bacterial cell walls.


Penicillin combinations include, but are not limited to, Amoxicillin/clavulanate, Ampicillin/sulbactam, Piperacillin/tazobactam, and Ticarcillin/clavulanate.


Polypeptide antibiotics include, but are not limited to, Bacitracin, Colistin, and Polymyxin B and E. Polypeptide Antibiotics are effective, e.g., against Gram-negative bacteria. Certain polypeptide antibiotics are believed to inhibit isoprenyl pyrophosphate involved in synthesis of the peptidoglycan layer of bacterial cell walls, while others destabilize the bacterial outer membrane by displacing bacterial counter-ions.


Quinolones and Fluoroquinolone include, but are not limited to, Ciprofloxacin, Enoxacin, Gatifloxacin, Gemifloxacin, Levofloxacin, Lomefloxacin, Moxifloxacin, Nalidixic acid, Norfloxacin, Ofloxacin, Trovafloxacin, Grepafloxacin, Sparfloxacin, and Temafloxacin. Quinolones/Fluoroquinol one are effective, e.g., against Streptococcus and Neisseria. Quinolones/Fluoroquinolone are believed to inhibit the bacterial DNA gyrase or topoisomerase IV, thereby inhibiting DNA replication and transcription.


Sulfonamides include, but are not limited to, Mafenide, Sulfacetamide, Sulfadiazine, Silver sulfadiazine, Sulfadimethoxine, Sulfamethizole, Sulfamethoxazole, Sulfanilimide, Sulfasalazine, Sulfisoxazole, Trimethoprim-Sulfamethoxazole (Co-trimoxazole), and Sulfonamidochrysoidine. Sulfonamides are believed to inhibit folate synthesis by competitive inhibition of dihydropteroate synthetase, thereby inhibiting nucleic acid synthesis.


Tetracyclines include, but are not limited to, Demeclocycline, Doxycycline, Minocycline, Oxytetracycline, and Tetracycline. Tetracyclines are effective, e.g., against Gram-negative bacteria. Tetracyclines are believed to bind to the bacterial 30S ribosomal subunit thereby inhibiting bacterial protein synthesis.


Anti-mycobacterial compounds include, but are not limited to, Clofazimine, Dapsone, Capreomycin, Cycloserine, Ethambutol, Ethionamide, Isoniazid, Pyrazinamide, Rifampicin, Rifabutin, Rifapentine, and Streptomycin.


Suitable antibiotics also include arsphenamine, chloramphenicol, fosfomycin, fusidic acid, metronidazole, mupirocin, platensimycin, quinupristin/dalfopristin, tigecycline, tinidazole, trimethoprim amoxicillin/clavulanate, ampicillin/sulbactam, amphomycin ristocetin, azithromycin, bacitracin, buforin II, carbomycin, cecropin Pl, clarithromycin, erythromycins, furazolidone, fusidic acid, Na fusidate, gramicidin, imipenem, indolicidin, josamycin, magainan II, metronidazole, nitroimidazoles, mikamycin, mutacin B-Ny266, mutacin B-JHl 140, mutacin J-T8, nisin, nisin A, novobiocin, oleandomycin, ostreogrycin, piperacillin/tazobactam, pristinamycin, ramoplanin, ranalexin, reuterin, rifaximin, rosamicin, rosaramicin, spectinomycin, spiramycin, staphylomycin, streptogramin, streptogramin A, synergistin, taurolidine, teicoplanin, telithromycin, ticarcillin/clavulanic acid, triacetyloleandomycin, tylosin, tyrocidin, tyrothricin, vancomycin, vemamycin, and virginiamycin.


In some embodiments, the additional therapeutic agent is an immunosuppressive agent, a DMARD, a pain-control drug, a steroid, a non-steroidal antiinflammatory drug (NSAID), or a cytokine antagonist, and combinations thereof. Representative agents include, but are not limited to, cyclosporin, retinoids, corticosteroids, propionic acid derivative, acetic acid derivative, enolic acid derivatives, fenamic acid derivatives, Cox-2 inhibitors, lumiracoxib, ibuprophen, cholin magnesium salicylate, fenoprofen, salsalate, difunisal, tolmetin, ketoprofen, flurbiprofen, oxaprozin, indomethacin, sulindac, etodolac, ketorolac, nabumetone, naproxen, valdecoxib, etoricoxib, MK0966; rofecoxib, acetominophen, Celecoxib, Diclofenac, tramadol, piroxicam, meloxicam, tenoxicam, droxicam, lornoxicam, isoxicam, mefanamic acid, meclofenamic acid, flufenamic acid, tolfenamic, valdecoxib, parecoxib, etodolac, indomethacin, aspirin, ibuprophen, firocoxib, methotrexate (MTX), antimalarial drugs (e.g., hydroxychloroquine and chloroquine), sulfasalazine, Leflunomide, azathioprine, cyclosporin, gold salts, minocycline, cyclophosphamide, D-penicillamine, minocycline, auranofin, tacrolimus, myocrisin, chlorambucil, TNF alpha antagonists (e.g., TNF alpha antagonists or TNF alpha receptor antagonists), e.g., ADALIMUMAB (Humira®), ETANERCEPT (Enbrel®), INFLIXIMAB (Remicade®; TA-650), CERTOLIZUMAB PEGOL (Cimzia®; CDP870), GOLIMUMAB (Simpom®; CNTO 148), ANAKINRA (Kineret®), RITUXIMAB (Rituxan®; MabThera®), ABATACEPT (Orencia®), TOCILIZUMAB (RoActemra/Actemra®), integrin antagonists (TYSABRI® (natalizumab)), IL-1 antagonists (ACZ885 (Ilaris)), Anakinra (Kineret®)), CD4 antagonists, IL-23 antagonists, IL-20 antagonists, IL-6 antagonists, BLyS antagonists (e.g., Atacicept, Benlystag/LymphoStat-B® (belimumab)), p38 Inhibitors, CD20 antagonists (Ocrelizumab, Ofatumumab (Arzerra®)), interferon gamma antagonists (Fontolizumab), prednisolone, Prednisone, dexamethasone, Cortisol, cortisone, hydrocortisone, methylprednisolone, betamethasone, triamcinolone, beclometasome, fludrocortisone, deoxycorticosterone, aldosterone, Doxycycline, vancomycin, pioglitazone, SBI-087, SCIO-469, Cura-100, Oncoxin+Viusid, TwHF, Methoxsalen, Vitamin D—ergocalciferol, Milnacipran, Paclitaxel, rosig tazone, Tacrolimus (Prograf®), RADOO1, rapamune, rapamycin, fostamatinib, Fentanyl, XOMA 052, Fostamatinib disodium,rosightazone, Curcumin (Longvida™), Rosuvastatin, Maraviroc, ramipnl, Milnacipran, Cobiprostone, somatropin, tgAAC94 gene therapy vector, MK0359, GW856553, esomeprazole, everolimus, trastuzumab, JAK1 and JAK2 inhibitors, pan JAK inhibitors, e.g., tetracyclic pyridone 6 (P6), 325, PF-956980, denosumab, IL-6 antagonists, CD20 antagonistis, CTLA4 antagonists, IL-8 antagonists, IL-21 antagonists, IL-22 antagonist, integrin antagonists (Tysarbri® (natalizumab)), VGEF antagnosits, CXCL antagonists, MMP antagonists, defensin antagonists, IL-1 antagonists (including IL-1 beta antagonsits), and IL-23 antagonists (e.g., receptor decoys, antagonistic antibodies, etc.).


In some embodiments, the additional therapeutic agent is an immunosuppressive agent. Examples of immunosuppressive agents include, but are not limited to, corticosteroids, mesalazine, mesalamine, sulfasalazine, sulfasalazine derivatives, immunosuppressive drugs, cyclosporin A, mercaptopurine, azathiopurine, prednisone, methotrexate, antihistamines, glucocorticoids, epinephrine, theophylline, cromolyn sodium, anti-leukotrienes, anti-cholinergic drugs for rhinitis, TLR antagonists, inflammasome inhibitors, anti-cholinergic decongestants, mast-cell stabilizers, monoclonal anti-IgE antibodies, vaccines (e.g., vaccines used for vaccination where the amount of an allergen is gradually increased), cytokine inhibitors, such as anti-IL-6 antibodies, TNF inhibitors such as infliximab, adalimumab, certolizumab pegol, golimumab, or etanercept, iand combinations thereof.


Administration

In certain aspects, provided herein is a method of delivering a pharmaceutical composition described herein (e.g., a pharmaceutical composition comprising mEVs (such as smEVs) to a subject. In some embodiments of the methods provided herein, the pharmaceutical composition is administered in conjunction with the administration of an additional therapeutic agent. In some embodiments, the pharmaceutical composition comprises mEVs (such as smEVs) co-formulated with the additional therapeutic agent. In some embodiments, the pharmaceutical composition comprising mEVs (such as smEVs) is co-administered with the additional therapeutic agent. In some embodiments, the additional therapeutic agent is administered to the subject before administration of the pharmaceutical composition that comprises mEVs (such as smEVs) (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or 55 minutes before, about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22 or 23 hours before, or about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days before). In some embodiments, the additional therapeutic agent is administered to the subject after administration of the pharmaceutical composition that comprises mEVs (such as smEVs) (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or 55 minutes after, about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22 or 23 hours after, or about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days after). In some embodiments, the same mode of delivery is used to deliver both the pharmaceutical composition that comprises mEVs (such as smEVs) and the additional therapeutic agent. In some embodiments, different modes of delivery are used to administer the pharmaceutical composition that comprises mEVs (such as smEVs) and the additional therapeutic agent. For example, in some embodiments the pharmaceutical composition that comprises mEVs (such as smEVs) is administered orally while the additional therapeutic agent is administered via injection (e.g., an intravenous, intramuscular and/or intratumoral injection).


In some embodiments, the pharmaceutical composition described herein is administered once a day. In some embodiments, the pharmaceutical composition described herein is administered twice a day. In some embodiments, the pharmaceutical composition described herein is formulated for a daily dose. In some embodiments, the pharmaceutical composition described herein is formulated for twice a day dose, wherein each dose is half of the daily dose.


In certain embodiments, the pharmaceutical compositions and dosage forms described herein can be administered in conjunction with any other conventional anti-cancer treatment, such as, for example, radiation therapy and surgical resection of the tumor. These treatments may be applied as necessary and/or as indicated and may occur before, concurrent with or after administration of the pharmaceutical composition that comprises mEVs (such as smEVs) or dosage forms described herein.


The dosage regimen can be any of a variety of methods and amounts, and can be determined by one skilled in the art according to known clinical factors. As is known in the medical arts, dosages for any one patient can depend on many factors, including the subject's species, size, body surface area, age, sex, immunocompetence, and general health, the particular microorganism to be administered, duration and route of administration, the kind and stage of the disease, for example, tumor size, and other compounds such as drugs being administered concurrently or near-concurrently. In addition to the above factors, such levels can be affected by the infectivity of the microorganism, and the nature of the microorganism, as can be determined by one skilled in the art. In the present methods, appropriate minimum dosage levels of microorganisms can be levels sufficient for the microorganism to survive, grow and replicate. The dose of a pharmaceutical composition that comprises mEVs (such as smEVs) described herein may be appropriately set or adjusted in accordance with the dosage form, the route of administration, the degree or stage of a target disease, and the like. For example, the general effective dose of the agents may range between 0.01 mg/kg body weight/day and 1000 mg/kg body weight/day, between 0.1 mg/kg body weight/day and 1000 mg/kg body weight/day, 0.5 mg/kg body weight/day and 500 mg/kg body weight/day, 1 mg/kg body weight/day and 100 mg/kg body weight/day, or between 5 mg/kg body weight/day and 50 mg/kg body weight/day. The effective dose may be 0.01, 0.05, 0.1, 0.5, 1, 2, 3, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, or 1000 mg/kg body weight/day or more, but the dose is not limited thereto.


In some embodiments, the dose administered to a subject is sufficient to prevent disease (e.g., autoimmune disease, inflammatory disease, metabolic disease, or cancer), delay its onset, or slow or stop its progression, or relieve one or more symptoms of the disease. One skilled in the art will recognize that dosage will depend upon a variety of factors including the strength of the particular agent (e.g., therapeutic agent) employed, as well as the age, species, condition, and body weight of the subject. The size of the dose will also be determined by the route, timing, and frequency of administration as well as the existence, nature, and extent of any adverse side-effects that might accompany the administration of a particular therapeutic agent and the desired physiological effect.


Suitable doses and dosage regimens can be determined by conventional range-finding techniques known to those of ordinary skill in the art. Generally, treatment is initiated with smaller dosages, which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached. An effective dosage and treatment protocol can be determined by routine and conventional means, starting e.g., with a low dose in laboratory animals and then increasing the dosage while monitoring the effects, and systematically varying the dosage regimen as well. Animal studies are commonly used to determine the maximal tolerable dose (“MTD”) of bioactive agent per kilogram weight. Those skilled in the art regularly extrapolate doses for efficacy, while avoiding toxicity, in other species, including humans.


In accordance with the above, in therapeutic applications, the dosages of the therapeutic agents used in accordance with the invention vary depending on the active agent, the age, weight, and clinical condition of the recipient patient, and the experience and judgment of the clinician or practitioner administering the therapy, among other factors affecting the selected dosage. For example, for cancer treatment, the dose should be sufficient to result in slowing, and preferably regressing, the growth of a tumor and most preferably causing complete regression of the cancer, or reduction in the size or number of metastases As another example, the dose should be sufficient to result in slowing of progression of the disease for which the subject is being treated, and preferably amelioration of one or more symptoms of the disease for which the subject is being treated.


Separate administrations can include any number of two or more administrations, including two, three, four, five or six administrations. One skilled in the art can readily determine the number of administrations to perform or the desirability of performing one or more additional administrations according to methods known in the art for monitoring therapeutic methods and other monitoring methods provided herein. Accordingly, the methods provided herein include methods of providing to the subject one or more administrations of a pharmaceutical composition, where the number of administrations can be determined by monitoring the subject, and, based on the results of the monitoring, determining whether or not to provide one or more additional administrations. Deciding on whether or not to provide one or more additional administrations can be based on a variety of monitoring results.


The time period between administrations can be any of a variety of time periods. The time period between administrations can be a function of any of a variety of factors, including monitoring steps, as described in relation to the number of administrations, the time period for a subject to mount an immune response. In one example, the time period can be a function of the time period for a subject to mount an immune response; for example, the time period can be more than the time period for a subject to mount an immune response, such as more than about one week, more than about ten days, more than about two weeks, or more than about a month; in another example, the time period can be less than the time period for a subject to mount an immune response, such as less than about one week, less than about ten days, less than about two weeks, or less than about a month.


In some embodiments, the delivery of an additional therapeutic agent in combination with the pharmaceutical composition described herein reduces the adverse effects and/or improves the efficacy of the additional therapeutic agent.


The effective dose of an additional therapeutic agent described herein is the amount of the additional therapeutic agent that is effective to achieve the desired therapeutic response for a particular subject, composition, and mode of administration, with the least toxicity to the subject. The effective dosage level can be identified using the methods described herein and will depend upon a variety of pharmacokinetic factors including the activity of the particular compositions or agents administered, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the subject being treated, and like factors well known in the medical arts. In general, an effective dose of an additional therapeutic agent will be the amount of the additional therapeutic agent which is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above.


The toxicity of an additional therapeutic agent is the level of adverse effects experienced by the subject during and following treatment. Adverse events associated with additional therapy toxicity can include, but are not limited to, abdominal pain, acid indigestion, acid reflux, allergic reactions, alopecia, anaphylasix, anemia, anxiety, lack of appetite, arthralgias, asthenia, ataxia, azotemia, loss of balance, bone pain, bleeding, blood clots, low blood pressure, elevated blood pressure, difficulty breathing, bronchitis, bruising, low white blood cell count, low red blood cell count, low platelet count, cardiotoxicity, cystitis, hemorrhagic cystitis, arrhythmias, heart valve disease, cardiomyopathy, coronary artery disease, cataracts, central neurotoxicity, cognitive impairment, confusion, conjunctivitis, constipation, coughing, cramping, cystitis, deep vein thrombosis, dehydration, depression, diarrhea, dizziness, dry mouth, dry skin, dyspepsia, dyspnea, edema, electrolyte imbalance, esophagitis, fatigue, loss of fertility, fever, flatulence, flushing, gastric reflux, gastroesophageal reflux disease, genital pain, granulocytopenia, gynecomastia, glaucoma, hair loss, hand-foot syndrome, headache, hearing loss, heart failure, heart palpitations, heartburn, hematoma, hemorrhagic cystitis, hepatotoxicity, hyperamylasemia, hypercalcemia, hyperchloremia, hyperglycemia, hyperkalemia, hyperlipasemia, hypermagnesemia, hypernatremia, hyperphosphatemia, hyperpigmentation, hypertriglyceridemia, hyperuricemia, hypoalbuminemia, hypocalcemia, hypochloremia, hypoglycemia, hypokalemia, hypomagnesemia, hyponatremia, hypophosphatemia, impotence, infection, injection site reactions, insomnia, iron deficiency, itching, joint pain, kidney failure, leukopenia, liver dysfunction, memory loss, menopause, mouth sores, mucositis, muscle pain, myalgias, myelosuppression, myocarditis, neutropenic fever, nausea, nephrotoxicity, neutropenia, nosebleeds, numbness, ototoxi city, pain, palmar-plantar erythrodysesthesia, pancytopenia, pericarditis, peripheral neuropathy, pharyngitis, photophobia, photosensitivity, pneumonia, pneumonitis, proteinuria, pulmonary embolus, pulmonary fibrosis, pulmonary toxicity, rash, rapid heart beat, rectal bleeding, restlessness, rhinitis, seizures, shortness of breath, sinusitis, thrombocytopenia, tinnitus, urinary tract infection, vaginal bleeding, vaginal dryness, vertigo, water retention, weakness, weight loss, weight gain, and xerostomia. In general, toxicity is acceptable if the benefits to the subject achieved through the therapy outweigh the adverse events experienced by the subject due to the therapy.


Immune Disorders

In some embodiments, the methods and pharmaceutical compositions described herein relate to the treatment or prevention of a disease or disorder associated a pathological immune response, such as an autoimmune disease, an allergic reaction and/or an inflammatory disease. In some embodiments, the disease or disorder is an inflammatory bowel disease (e.g., Crohn's disease or ulcerative colitis). In some embodiments, the disease or disorder is psoriasis. In some embodiments, the disease or disorder is atopic dermatitis.


The methods described herein can be used to treat any subject in need thereof. As used herein, a “subject in need thereof” includes any subject that has a disease or disorder associated with a pathological immune response (e.g., an inflammatory bowel disease), as well as any subject with an increased likelihood of acquiring a such a disease or disorder.


The pharmaceutical compositions described herein can be used, for example, as a pharmaceutical composition for preventing or treating (reducing, partially or completely, the adverse effects of) an autoimmune disease, such as chronic inflammatory bowel disease, systemic lupus erythematosus, psoriasis, muckle-wells syndrome, rheumatoid arthritis, multiple sclerosis, or Hashimoto's disease; an allergic disease, such as a food allergy, pollenosis, or asthma; an infectious disease, such as an infection with Clostridium difficile; an inflammatory disease such as a TNF-mediated inflammatory disease (e.g., an inflammatory disease of the gastrointestinal tract, such as pouchitis, a cardiovascular inflammatory condition, such as atherosclerosis, or an inflammatory lung disease, such as chronic obstructive pulmonary disease); a pharmaceutical composition for suppressing rejection in organ transplantation or other situations in which tissue rejection might occur; a supplement, food, or beverage for improving immune functions; or a reagent for suppressing the proliferation or function of immune cells.


In some embodiments, the methods provided herein are useful for the treatment of inflammation. In certain embodiments, the inflammation of any tissue and organs of the body, including musculoskeletal inflammation, vascular inflammation, neural inflammation, digestive system inflammation, ocular inflammation, inflammation of the reproductive system, and other inflammation, as discussed below.


Immune disorders of the musculoskeletal system include, but are not limited, to those conditions affecting skeletal joints, including joints of the hand, wrist, elbow, shoulder, jaw, spine, neck, hip, knew, ankle, and foot, and conditions affecting tissues connecting muscles to bones such as tendons. Examples of such immune disorders, which may be treated with the methods and compositions described herein include, but are not limited to, arthritis (including, for example, osteoarthritis, rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, acute and chronic infectious arthritis, arthritis associated with gout and pseudogout, and juvenile idiopathic arthritis), tendonitis, synovitis, tenosynovitis, bursitis, fibrositis (fibromyalgia), epicondylitis, myositis, and osteitis (including, for example, Paget's disease, osteitis pubis, and osteitis fibrosa cystic).


Ocular immune disorders refers to a immune disorder that affects any structure of the eye, including the eye lids. Examples of ocular immune disorders which may be treated with the methods and compositions described herein include, but are not limited to, blepharitis, blepharochalasis, conjunctivitis, dacryoadenitis, keratitis, keratoconjunctivitis sicca (dry eye), scleritis, trichiasis, and uveitis


Examples of nervous system immune disorders which may be treated with the methods and compositions described herein include, but are not limited to, encephalitis, Guillain-Barre syndrome, meningitis, neuromyotonia, narcolepsy, multiple sclerosis, myelitis and schizophrenia. Examples of inflammation of the vasculature or lymphatic system which may be treated with the methods and compositions described herein include, but are not limited to, arthrosclerosis, arthritis, phlebitis, vasculitis, and lymphangitis.


Examples of digestive system immune disorders which may be treated with the methods and pharmaceutical compositions described herein include, but are not limited to, cholangitis, cholecystitis, enteritis, enterocolitis, gastritis, gastroenteritis, inflammatory bowel disease, ileitis, and proctitis. Inflammatory bowel diseases include, for example, certain art-recognized forms of a group of related conditions. Several major forms of inflammatory bowel diseases are known, with Crohn's disease (regional bowel disease, e.g., inactive and active forms) and ulcerative colitis (e.g., inactive and active forms) the most common of these disorders. In addition, the inflammatory bowel disease encompasses irritable bowel syndrome, microscopic colitis, lymphocytic-plasmocytic enteritis, coeliac disease, collagenous colitis, lymphocytic colitis and eosinophilic enterocolitis. Other less common forms of IBD include indeterminate colitis, pseudomembranous colitis (necrotizing colitis), ischemic inflammatory bowel disease, Behcet's disease, sarcoidosis, scleroderma, IBD-associated dysplasia, dysplasia associated masses or lesions, and primary sclerosing cholangitis.


Examples of reproductive system immune disorders which may be treated with the methods and pharmaceutical compositions described herein include, but are not limited to, cervicitis, chorioamnionitis, endometritis, epididymitis, omphalitis, oophoritis, orchitis, salpingitis, tubo-ovarian abscess, urethritis, vaginitis, vulvitis, and vulvodynia.


The methods and pharmaceutical compositions described herein may be used to treat autoimmune conditions having an inflammatory component. Such conditions include, but are not limited to, acute disseminated alopecia universalise, Behcet's disease, Chagas' disease, chronic fatigue syndrome, dysautonomia, encephalomyelitis, ankylosing spondylitis, aplastic anemia, hidradenitis suppurativa, autoimmune hepatitis, autoimmune oophoritis, celiac disease, Crohn's disease, diabetes mellitus type 1, giant cell arteritis, goodpasture's syndrome, Grave's disease, Guillain-Barre syndrome, Hashimoto's disease, Henoch-Schonlein purpura, Kawasaki's disease, lupus erythematosus, microscopic colitis, microscopic polyarteritis, mixed connective tissue disease, Muckle-Wells syndrome, multiple sclerosis, myasthenia gravis, opsoclonus myoclonus syndrome, optic neuritis, ord's thyroiditis, pemphigus, polyarteritis nodosa, polymyalgia, rheumatoid arthritis, Reiter's syndrome, Sjögren's syndrome, temporal arteritis, Wegener's granulomatosis, warm autoimmune haemolytic anemia, interstitial cystitis, Lyme disease, morphea, psoriasis, sarcoidosis, scleroderma, ulcerative colitis, and vitiligo.


The methods and pharmaceutical compositions described herein may be used to treat T-cell mediated hypersensitivity diseases having an inflammatory component. Such conditions include, but are not limited to, contact hypersensitivity, contact dermatitis (including that due to poison ivy), uticaria, skin allergies, respiratory allergies (hay fever, allergic rhinitis, house dustmite allergy) and gluten-sensitive enteropathy (Celiac disease).


Other immune disorders which may be treated with the methods and pharmaceutical compositions include, for example, appendicitis, dermatitis, dermatomyositis, endocarditis, fibrositis, gingivitis, glossitis, hepatitis, hidradenitis suppurativa, iritis, laryngitis, mastitis, myocarditis, nephritis, otitis, pancreatitis, parotitis, percarditis, peritonoitis, pharyngitis, pleuritis, pneumonitis, prostatistis, pyelonephritis, and stomatisi, transplant rejection (involving organs such as kidney, liver, heart, lung, pancreas (e.g., islet cells), bone marrow, cornea, small bowel, skin allografts, skin homografts, and heart valve xengrafts, sewrum sickness, and graft vs host disease), acute pancreatitis, chronic pancreatitis, acute respiratory distress syndrome, Sexary's syndrome, congenital adrenal hyperplasis, nonsuppurative thyroiditis, hypercalcemia associated with cancer, pemphigus, bullous dermatitis herpetiformis, severe erythema multiforme, exfoliative dermatitis, seborrheic dermatitis, seasonal or perennial allergic rhinitis, bronchial asthma, contact dermatitis, atopic dermatitis, drug hypersensistivity reactions, allergic conjunctivitis, keratitis, herpes zoster ophthalmicus, iritis and oiridocyclitis, chorioretinitis, optic neuritis, symptomatic sarcoidosis, fulminating or disseminated pulmonary tuberculosis chemotherapy, idiopathic thrombocytopenic purpura in adults, secondary thrombocytopenia in adults, acquired (autoimmune) haemolytic anemia, leukaemia and lymphomas in adults, acute leukaemia of childhood, regional enteritis, autoimmune vasculitis, multiple sclerosis, chronic obstructive pulmonary disease, solid organ transplant rejection, sepsis. Preferred treatments include treatment of transplant rejection, rheumatoid arthritis, psoriatic arthritis, multiple sclerosis, Type 1 diabetes, asthma, inflammatory bowel disease, systemic lupus erythematosus, psoriasis, chronic obstructive pulmonary disease, and inflammation accompanying infectious conditions (e.g., sepsis).


Metabolic Disorders

In some embodiments, the methods and pharmaceutical compositions described herein relate to the treatment or prevention of a metabolic disease or disorder a, such as type II diabetes, impaired glucose tolerance, insulin resistance, obesity, hyperglycemia, hyperinsulinemia, fatty liver, non-alcoholic steatohepatitis, hypercholesterolemia, hypertension, hyperlipoproteinemia, hyperlipidemia, hypertriglylceridemia, ketoacidosis, hypoglycemia, thrombotic disorders, dyslipidemia, non-alcoholic fatty liver disease (NAFLD), Nonalcoholic Steatohepatitis (NASH) or a related disease. In some embodiments, the related disease is cardiovascular disease, atherosclerosis, kidney disease, nephropathy, diabetic neuropathy, diabetic retinopathy, sexual dysfunction, dermatopathy, dyspepsia, or edema. In some embodiments, the methods and pharmaceutical compositions described herein relate to the treatment of Nonalcoholic Fatty Liver Disease (NAFLD) and Nonalcoholic Steatohepatitis (NASH).


The methods described herein can be used to treat any subject in need thereof. As used herein, a “subject in need thereof” includes any subject that has a metabolic disease or disorder, as well as any subject with an increased likelihood of acquiring a such a disease or disorder.


The pharmaceutical compositions described herein can be used, for example, for preventing or treating (reducing, partially or completely, the adverse effects of) a metabolic disease, such as type II diabetes, impaired glucose tolerance, insulin resistance, obesity, hyperglycemia, hyperinsulinemia, fatty liver, non-alcoholic steatohepatitis, hypercholesterolemia, hypertension, hyperlipoproteinemia, hyperlipidemia, hypertriglylceridemia, ketoacidosis, hypoglycemia, thrombotic disorders, dyslipidemia, non-alcoholic fatty liver disease (NAFLD), Nonalcoholic Steatohepatitis (NASH), or a related disease. In some embodiments, the related disease is cardiovascular disease, atherosclerosis, kidney disease, nephropathy, diabetic neuropathy, diabetic retinopathy, sexual dysfunction, dermatopathy, dyspepsia, or edema.


Cancer

In some embodiments, the methods and pharmaceutical compositions described herein relate to the treatment of cancer. In some embodiments, any cancer can be treated using the methods described herein. Examples of cancers that may treated by methods and pharmaceutical compositions described herein include, but are not limited to, cancer cells from the bladder, blood, bone, bone marrow, brain, breast, colon, esophagus, gastrointestine, gum, head, kidney, liver, lung, nasopharynx, neck, ovary, prostate, skin, stomach, testis, tongue, or uterus. In addition, the cancer may specifically be of the following histological type, though it is not limited to these: neoplasm, malignant; carcinoma; carcinoma, undifferentiated; giant and spindle cell carcinoma; small cell carcinoma; papillary carcinoma; squamous cell carcinoma; lymphoepitheli al carcinoma; basal cell carcinoma; pilomatrix carcinoma; transitional cell carcinoma; papillary transitional cell carcinoma; adenocarcinoma; gastrinoma, malignant; cholangiocarcinoma; hepatocellular carcinoma; combined hepatocellular carcinoma and cholangiocarcinoma; trabecular adenocarcinoma; adenoid cystic carcinoma; adenocarcinoma in adenomatous polyp; adenocarcinoma, familial polyposis coli; solid carcinoma; carcinoid tumor, malignant; branchiolo-alveolar adenocarcinoma; papillary adenocarcinoma; chromophobe carcinoma; acidophil carcinoma; oxyphilic adenocarcinoma; basophil carcinoma; clear cell adenocarcinoma; granular cell carcinoma; follicular adenocarcinoma; papillary and follicular adenocarcinoma; nonencapsulating sclerosing carcinoma; adrenal cortical carcinoma; endometroid carcinoma; skin appendage carcinoma; apocrine adenocarcinoma; sebaceous adenocarcinoma; ceruminous adenocarcinoma; mucoepidermoid carcinoma; cystadenocarcinoma; papillary cystadenocarcinoma; papillary serous cystadenocarcinoma; mucinous cystadenocarcinoma; mucinous adenocarcinoma; signet ring cell carcinoma; infiltrating duct carcinoma; medullary carcinoma; lobular carcinoma; inflammatory carcinoma; paget's disease, mammary; acinar cell carcinoma; adenosquamous carcinoma; adenocarcinoma w/squamous metaplasia; thymoma, malignant; ovarian stromal tumor, malignant; thecoma, malignant; granulosa cell tumor, malignant; and roblastoma, malignant; sertoli cell carcinoma; leydig cell tumor, malignant; lipid cell tumor, malignant; paraganglioma, malignant; extra-mammary paraganglioma, malignant; pheochromocytoma; glomangiosarcoma; malignant melanoma; amelanotic melanoma; superficial spreading melanoma; malig melanoma in giant pigmented nevus; epithelioid cell melanoma; blue nevus, malignant; sarcoma; fibrosarcoma; fibrous histiocytoma, malignant; myxosarcoma; liposarcoma; leiomyosarcoma; rhabdomyosarcoma; embryonal rhabdomyosarcoma; alveolar rhabdomyosarcoma; stromal sarcoma; mixed tumor, malignant; mullerian mixed tumor; nephroblastoma; hepatoblastoma; carcinosarcoma; mesenchymoma, malignant; brenner tumor, malignant; phyllodes tumor, malignant; synovial sarcoma; mesothelioma, malignant; dysgerminoma; embryonal carcinoma; teratoma, malignant; struma ovarii, malignant; choriocarcinoma; mesonephroma, malignant; hemangiosarcoma; hemangioendothelioma, malignant; kaposi's sarcoma; hemangiopericytoma, malignant; lymphangiosarcoma; osteosarcoma; juxtacortical osteosarcoma; chondrosarcoma; chondroblastoma, malignant; mesenchymal chondrosarcoma; giant cell tumor of bone; ewing's sarcoma; odontogenic tumor, malignant; am el oblastic odontosarcoma; ameloblastoma, malignant; ameloblastic fibrosarcoma; pineal oma, malignant; chordoma; glioma, malignant; ependymoma; astrocytoma; protoplasmic astrocytoma; fibrillary astrocytoma; astroblastoma; glioblastoma; oligodendroglioma; oligodendroblastoma; primitive neuroectodermal; cerebellar sarcoma; ganglioneuroblastoma; neuroblastoma; retinoblastoma; olfactory neurogenic tumor; meningioma, malignant; neurofibrosarcoma; neurilemmoma, malignant; granular cell tumor, malignant; malignant lymphoma; Hodgkin's disease; Hodgkin's lymphoma; paragranuloma; malignant lymphoma, small lymphocytic; malignant lymphoma, large cell, diffuse; malignant lymphoma, follicular; mycosis fungoides; other specified non-Hodgkin's lymphomas; malignant histiocytosis; multiple myeloma; mast cell sarcoma; immunoproliferative small intestinal disease; leukemia; lymphoid leukemia; plasma cell leukemia; erythroleukemia; lymphosarcoma cell leukemia; myeloid leukemia; basophilic leukemia; eosinophilic leukemia; monocytic leukemia; mast cell leukemia; megakaryoblastic leukemia; myeloid sarcoma; and hairy cell leukemia.


In some embodiments, the methods and pharmaceutical compositions provided herein relate to the treatment of a leukemia. The term “leukemia” includes broadly progressive, malignant diseases of the hematopoietic organs/systems and is generally characterized by a distorted proliferation and development of leukocytes and their precursors in the blood and bone marrow. Non-limiting examples of leukemia diseases include, acute nonlymphocytic leukemia, chronic lymphocytic leukemia, acute granulocytic leukemia, chronic granulocytic leukemia, acute promyelocytic leukemia, adult T-cell leukemia, aleukemic leukemia, a leukocythemic leukemia, basophilic leukemia, blast cell leukemia, bovine leukemia, chronic myelocytic leukemia, leukemia cutis, embryonal leukemia, eosinophilic leukemia, Gross' leukemia, Rieder cell leukemia, Schilling's leukemia, stem cell leukemia, subleukemic leukemia, undifferentiated cell leukemia, hairy-cell leukemia, hemoblastic leukemia, hemocytoblastic leukemia, histiocytic leukemia, stem cell leukemia, acute monocytic leukemia, leukopenic leukemia, lymphatic leukemia, lymphoblastic leukemia, lymphocytic leukemia, lymphogenous leukemia, lymphoid leukemia, lymphosarcoma cell leukemia, mast cell leukemia, megakaryocytic leukemia, micromyeloblastic leukemia, monocytic leukemia, myeloblastic leukemia, myelocytic leukemia, myeloid granulocytic leukemia, myelomonocytic leukemia, Naegeli leukemia, plasma cell leukemia, plasmacytic leukemia, and promyelocytic leukemia.


In some embodiments, the methods and pharmaceutical compositions provided herein relate to the treatment of a carcinoma. The term “carcinoma” refers to a malignant growth made up of epithelial cells tending to infiltrate the surrounding tissues, and/or resist physiological and non-physiological cell death signals and gives rise to metastases. Non-limiting exemplary types of carcinomas include, acinar carcinoma, acinous carcinoma, adenocystic carcinoma, adenoid cystic carcinoma, carcinoma adenomatosum, carcinoma of adrenal cortex, alveolar carcinoma, alveolar cell carcinoma, basal cell carcinoma, carcinoma basocellulare, basaloid carcinoma, basosquamous cell carcinoma, bronchioalveolar carcinoma, bronchiolar carcinoma, bronchogenic carcinoma, cerebriform carcinoma, cholangiocellular carcinoma, chorionic carcinoma, colloid carcinoma, comedo carcinoma, corpus carcinoma, cribriform carcinoma, carcinoma en cuirasse, carcinoma cutaneum, cylindrical carcinoma, cylindrical cell carcinoma, duct carcinoma, carcinoma durum, embryonal carcinoma, encephaloid carcinoma, epiennoid carcinoma, carcinoma epitheliale adenoides, exophytic carcinoma, carcinoma ex ulcere, carcinoma fibrosum, gelatiniform carcinoma, gelatinous carcinoma, giant cell carcinoma, signet-ring cell carcinoma, carcinoma simplex, small-cell carcinoma, solanoid carcinoma, spheroidal cell carcinoma, spindle cell carcinoma, carcinoma spongiosum, squamous carcinoma, squamous cell carcinoma, string carcinoma, carcinoma telangiectaticum, carcinoma telangiectodes, transitional cell carcinoma, carcinoma tuberosum, tuberous carcinoma, verrucous carcinoma, carcinoma villosum, carcinoma gigantocellulare, glandular carcinoma, granulosa cell carcinoma, hair-matrix carcinoma, hematoid carcinoma, hepatocellular carcinoma, Hurthle cell carcinoma, hyaline carcinoma, hypernephroid carcinoma, infantile embryonal carcinoma, carcinoma in situ, intraepidermal carcinoma, intraepithelial carcinoma, Krompecher's carcinoma, Kulchitzky-cell carcinoma, large-cell carcinoma, lenticular carcinoma, carcinoma lenticulare, lipomatous carcinoma, lymphoepithelial carcinoma, carcinoma medullare, medullary carcinoma, melanotic carcinoma, carcinoma molle, mucinous carcinoma, carcinoma muciparum, carcinoma mucocellulare, mucoepidermoid carcinoma, carcinoma mucosum, mucous carcinoma, carcinoma myxomatodes, naspharyngeal carcinoma, oat cell carcinoma, carcinoma ossificans, osteoid carcinoma, papillary carcinoma, periportal carcinoma, preinvasive carcinoma, prickle cell carcinoma, pultaceous carcinoma, renal cell carcinoma of kidney, reserve cell carcinoma, carcinoma sarcomatodes, schneiderian carcinoma, scirrhous carcinoma, and carcinoma scroti.


In some embodiments, the methods and pharmaceutical compositions provided herein relate to the treatment of a sarcoma. The term “sarcoma” generally refers to a tumor which is made up of a substance like the embryonic connective tissue and is generally composed of closely packed cells embedded in a fibrillar, heterogeneous, or homogeneous substance. Sarcomas include, but are not limited to, chondrosarcoma, fibrosarcoma, lymphosarcoma, melanosarcoma, myxosarcoma, osteosarcoma, endometrial sarcoma, stromal sarcoma, Ewing's sarcoma, fascial sarcoma, fibroblastic sarcoma, giant cell sarcoma, Abemethy's sarcoma, adipose sarcoma, liposarcoma, alveolar soft part sarcoma, ameloblastic sarcoma, botryoid sarcoma, chloroma sarcoma, chorio carcinoma, embryonal sarcoma, Wilms' tumor sarcoma, granulocytic sarcoma, Hodgkin's sarcoma, idiopathic multiple pigmented hemorrhagic sarcoma, immunoblastic sarcoma of B cells, lymphoma, immunoblastic sarcoma of T-cells, Jensen's sarcoma, Kaposi's sarcoma, Kupffer cell sarcoma, angiosarcoma, leukosarcoma, malignant mesenchymoma sarcoma, parosteal sarcoma, reticulocytic sarcoma, Rous sarcoma, serocystic sarcoma, synovial sarcoma, and telangiectaltic sarcoma.


Additional exemplary neoplasias that can be treated using the methods and pharmaceutical compositions described herein include Hodgkin's Disease, Non-Hodgkin's Lymphoma, multiple myeloma, neuroblastoma, breast cancer, ovarian cancer, lung cancer, rhabdomyosarcoma, primary thrombocytosis, primary macroglobulinemia, small-cell lung tumors, primary brain tumors, stomach cancer, colon cancer, malignant pancreatic insulanoma, malignant carcinoid, premalignant skin lesions, testicular cancer, lymphomas, thyroid cancer, neuroblastoma, esophageal cancer, genitourinary tract cancer, malignant hypercalcemia, cervical cancer, endometrial cancer, plasmacytoma, colorectal cancer, rectal cancer, and adrenal cortical cancer.


In some embodiments, the cancer treated is a melanoma. The term “melanoma” is taken to mean a tumor arising from the melanocytic system of the skin and other organs. Non-limiting examples of melanomas are Harding-Passey melanoma, juvenile melanoma, lentigo maligna melanoma, malignant melanoma, acral-lentiginous melanoma, amelanotic melanoma, benign juvenile melanoma, Cloudman's melanoma, S91 melanoma, nodular melanoma subungal melanoma, and superficial spreading melanoma.


In some embodiments, the cancer comprises breast cancer (e.g., triple negative breast cancer).


In some embodiments, the cancer comprises colorectal cancer (e.g., microsatellite stable (MSS) colorectal cancer).


In some embodiments, the cancer comprises renal cell carcinoma.


In some embodiments, the cancer comprises lung cancer (e.g., non small cell lung cancer).


In some embodiments, the cancer comprises bladder cancer.


In some embodiments, the cancer comprises gastroesophageal cancer.


Particular categories of tumors that can be treated using methods and pharmaceutical compositions described herein include lymphoproliferative disorders, breast cancer, ovarian cancer, prostate cancer, cervical cancer, endometrial cancer, bone cancer, liver cancer, stomach cancer, colon cancer, pancreatic cancer, cancer of the thyroid, head and neck cancer, cancer of the central nervous system, cancer of the peripheral nervous system, skin cancer, kidney cancer, as well as metastases of all the above. Particular types of tumors include hepatocellular carcinoma, hepatoma, hepatoblastoma, rhabdomyosarcoma, esophageal carcinoma, thyroid carcinoma, ganglioblastoma, fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, Ewing's tumor, leimyosarcoma, rhabdotheliosarcoma, invasive ductal carcinoma, papillary adenocarcinoma, melanoma, pulmonary squamous cell carcinoma, basal cell carcinoma, adenocarcinoma (well differentiated, moderately differentiated, poorly differentiated or undifferentiated), bronchioloalveolar carcinoma, renal cell carcinoma, hypernephroma, hypernephroid adenocarcinoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms' tumor, testicular tumor, lung carcinoma including small cell, non-small and large cell lung carcinoma, bladder carcinoma, glioma, astrocyoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, retinoblastoma, neuroblastoma, colon carcinoma, rectal carcinoma, hematopoietic malignancies including all types of leukemia and lymphoma including: acute myelogenous leukemia, acute myelocytic leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, mast cell leukemia, multiple myeloma, myeloid lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, plasmacytoma, colorectal cancer, and rectal cancer.


Cancers treated in certain embodiments also include precancerous lesions, e.g., actinic keratosis (solar keratosis), moles (dysplastic nevi), acitinic chelitis (farmer's lip), cutaneous horns, Barrett's esophagus, atrophic gastritis, dyskeratosis congenita, sideropenic dysphagia, lichen planus, oral submucous fibrosis, actinic (solar) elastosis and cervical dysplasia.


Cancers treated in some embodiments include non-cancerous or benign tumors, e.g., of endodermal, ectodermal or mesenchymal origin, including, but not limited to cholangioma, colonic polyp, adenoma, papilloma, cystadenoma, liver cell adenoma, hydatidiform mole, renal tubular adenoma, squamous cell papilloma, gastric polyp, hemangioma, osteoma, chondroma, lipoma, fibroma, lymphangioma, leiomyoma, rhabdomyoma, astrocytoma, nevus, meningioma, and ganglioneuroma.


Other Diseases and Disorders

In some embodiments, the methods and pharmaceutical compositions described herein relate to the treatment of liver diseases. Such diseases include, but are not limited to, Alagille Syndrome, Alcohol-Related Liver Disease, Alpha-1 Antitrypsin Deficiency, Autoimmune Hepatitis, Benign Liver Tumors, Biliary Atresia, Cirrhosis, Galactosemia, Gilbert Syndrome, Hemochromatosis, Hepatitis A, Hepatitis B, Hepatitis C, Hepatic Encephalopathy, Intrahepatic Cholestasis of Pregnancy (ICP), Lysosomal Acid Lipase Deficiency (LAL-D), Liver Cysts, Liver Cancer, Newborn Jaundice, Primary Biliary Cholangitis (PBC), Primary Sclerosing Cholangitis (PSC), Reye Syndrome, Type I Glycogen Storage Disease, and Wilson Disease.


The methods and pharmaceutical compositions described herein may be used to treat neurodegenerative and neurological diseases. In certain embodiments, the neurodegenerative and/or neurological disease is Parkinson's disease, Alzheimer's disease, prion disease, Huntington's disease, motor neuron diseases (MND), spinocerebellar ataxia, spinal muscular atrophy, dystonia, idiopathicintracranial hypertension, epilepsy, nervous system disease, central nervous system disease, movement disorders, multiple sclerosis, encephalopathy, peripheral neuropathy or post-operative cognitive dysfunction.


Dysbiosis

The gut microbiome (also called the “gut microbiota”) can have a significant impact on an individual's health through microbial activity and influence (local and/or distal) on immune and other cells of the host (Walker, W. A., Dysbiosis. The Microbiota in Gastrointestinal Pathophysiology. Chapter 25. 2017; Weiss and Thierry, Mechanisms and consequences of intestinal dysbiosis. Cellular and Molecular Life Sciences. (2017) 74(16):2959-2977. Zurich Open Repository and Archive, doi: https://doi.org/10.1007/s00018-017-2509-x)).


A healthy host-gut microbiome homeostasis is sometimes referred to as a “eubiosis” or “normobiosis,” whereas a detrimental change in the host microbiome composition and/or its diversity can lead to an unhealthy imbalance in the microbiome, or a “dysbiosis” (Hooks and O'Malley. Dysbiosis and its discontents. American Society for Microbiology. October 2017. Vol. 8. Issue 5. mBio 8:e01492-17. https://doi.org/10.1128/mBio.01492-17). Dysbiosis, and associated local or distal host inflammatory or immune effects, may occur where microbiome homeostasis is lost or diminished, resulting in: increased susceptibility to pathogens; altered host bacterial metabolic activity; induction of host proinflammatory activity and/or reduction of host anti-inflammatory activity. Such effects are mediated in part by interactions between host immune cells (e.g., T cells, dendritic cells, mast cells, NK cells, intestinal epithelial lymphocytes (IEC), macrophages and phagocytes) and cytokines, and other substances released by such cells and other host cells.


A dysbiosis may occur within the gastrointestinal tract (a “gastrointestinal dysbiosis” or “gut dysbiosis”) or may occur outside the lumen of the gastrointestinal tract (a “distal dysbiosis”). Gastrointestinal dysbiosis is often associated with a reduction in integrity of the intestinal epithelial barrier, reduced tight junction integrity and increased intestinal permeability. Citi, S. Intestinal Barriers protect against disease, Science 359:1098-99 (2018); Srinivasan et al., TEER measurement techniques for in vitro barrier model systems. J. Lab. Autom. 20:107-126 (2015). A gastrointestinal dysbiosis can have physiological and immune effects within and outside the gastrointestinal tract.


The presence of a dysbiosis can be associated with a wide variety of diseases and conditions including: infection, cancer, autoimmune disorders (e.g., systemic lupus erythematosus (SLE)) or inflammatory disorders (e.g., functional gastrointestinal disorders such as inflammatory bowel disease (IBD), ulcerative colitis, and Crohn's disease), neuroinflammatory diseases (e.g., multiple sclerosis), transplant disorders (e.g., graft-versus-host disease), fatty liver disease, type I diabetes, rheumatoid arthritis, Sjögren's syndrome, celiac disease, cystic fibrosis, chronic obstructive pulmonary disorder (COPD), and other diseases and conditions associated with immune dysfunction. Lynch et al., The Human Microbiome in Health and Disease, N. Engl. J. Med. 375:2369-79 (2016), Carding et al., Dysbiosis of the gut microbiota in disease. Microb. Ecol. Health Dis. (2015); 26: 10: 3402/mehd.v26.2619; Levy et al, Dysbiosis and the Immune System, Nature Reviews Immunology 17:219 (April 2017)


In certain embodiments, exemplary pharmaceutical compositions disclosed herein can treat a dysbiosis and its effects by modifying the immune activity present at the site of dysbiosis. As described herein, such compositions can modify a dysbiosis via effects on host immune cells, resulting in, e.g., an increase in secretion of anti-inflammatory cytokines and/or a decrease in secretion of pro-inflammatory cytokines, reducing inflammation in the subject recipient or via changes in metabolite production.


Exemplary pharmaceutical compositions disclosed herein that are useful for treatment of disorders associated with a dysbiosis contain one or more types of mEVs (microbial extracellular vesicles) derived from immunomodulatory bacteria (e.g., anti-inflammatory bacteria). Such compositions are capable of affecting the recipient host's immune function, in the gastrointestinal tract, and/or a systemic effect at distal sites outside the subject's gastrointestinal tract.


Exemplary pharmaceutical compositions disclosed herein that are useful for treatment of disorders associated with a dysbiosis contain a population of immunomodulatory bacteria of a single bacterial species (e.g., a single strain) (e.g., anti-inflammatory bacteria) and/or a population of mEVs derived from immunomodulatory bacteria of a single bacterial species (e.g., a single strain) (e.g., anti-inflammatory bacteria). Such compositions are capable of affecting the recipient host's immune function, in the gastrointestinal tract, and/or a systemic effect at distal sites outside the subject's gastrointestinal tract.


In one embodiment, pharmaceutical compositions containing an isolated population of mEVs derived from immunomodulatory bacteria (e.g., anti-inflammatory bacterial cells) are administered (e.g., orally) to a mammalian recipient in an amount effective to treat a dysbiosis and one or more of its effects in the recipient. The dysbiosis may be a gastrointestinal tract dysbiosis or a distal dysbiosis.


In another embodiment, pharmaceutical compositions of the instant invention can treat a gastrointestinal dysbiosis and one or more of its effects on host immune cells, resulting in an increase in secretion of anti-inflammatory cytokines and/or a decrease in secretion of pro-inflammatory cytokines, reducing inflammation in the subject recipient.


In another embodiment, the pharmaceutical compositions can treat a gastrointestinal dysbiosis and one or more of its effects by modulating the recipient immune response via cellular and cytokine modulation to reduce gut permeability by increasing the integrity of the intestinal epithelial barrier.


In another embodiment, the pharmaceutical compositions can treat a distal dysbiosis and one or more of its effects by modulating the recipient immune response at the site of dysbiosis via modulation of host immune cells.


Other exemplary pharmaceutical compositions are useful for treatment of disorders associated with a dysbiosis, which compositions contain one or more types of bacteria or mEVs capable of altering the relative proportions of host immune cell subpopulations, e.g., subpopulations of T cells, immune lymphoid cells, dendritic cells, NK cells and other immune cells, or the function thereof, in the recipient.


Other exemplary pharmaceutical compositions are useful for treatment of disorders associated with a dysbiosis, which compositions contain a population of mEVs of a single immunomodulatory bacterial (e.g., anti-inflammatory bacterial cells) species (e.g., a single strain) capable of altering the relative proportions of immune cell subpopulations, e.g., T cell subpopulations, immune lymphoid cells, NK cells and other immune cells, or the function thereof, in the recipient subject.


In one embodiment, the invention provides methods of treating a gastrointestinal dysbiosis and one or more of its effects by orally administering to a subject in need thereof a pharmaceutical composition which alters the microbiome population existing at the site of the dysbiosis. The pharmaceutical composition can contain one or more types of mEVs from immunomodulatory bacteria or a population of mEVs of a single immunomodulatory bacterial species (e.g., anti-inflammatory bacterial cells) (e.g., a single strain).


In one embodiment, the invention provides methods of treating a distal dysbiosis and one or more of its effects by orally administering to a subject in need thereof a pharmaceutical composition which alters the subject's immune response outside the gastrointestinal tract. The pharmaceutical composition can contain one or more types of mEVs from immunomodulatory bacteria (e.g., anti-inflammatory bacterial cells) or a population of mEVs of a single immunomodulatory bacterial (e.g., anti-inflammatory bacterial cells) species (e.g., a single strain).


In exemplary embodiments, pharmaceutical compositions useful for treatment of disorders associated with a dysbiosis stimulate secretion of one or more anti-inflammatory cytokines by host immune cells. Anti-inflammatory cytokines include, but are not limited to, IL-10, IL-13, IL-9, IL-4, IL-5, TGFβ, and combinations thereof. In other exemplary embodiments, pharmaceutical compositions useful for treatment of disorders associated with a dysbiosis that decrease (e.g., inhibit) secretion of one or more pro-inflammatory cytokines by host immune cells. Pro-inflammatory cytokines include, but are not limited to, IFNγ, IL-12p70, IL-1α, IL-6, IL-8, MCP1, MIP1α, MIP1β, TNFα, and combinations thereof. Other exemplary cytokines are known in the art and are described herein.


In another aspect, the invention provides a method of treating or preventing a disorder associated with a dysbiosis in a subject in need thereof, comprising administering (e.g., orally administering) to the subject a therapeutic composition in the form of a probiotic or medical food comprising bacteria or mEVs in an amount sufficient to alter the microbiome at a site of the dysbiosis, such that the disorder associated with the dysbiosis is treated.


In another embodiment, a therapeutic composition of the instant invention in the form of a probiotic or medical food may be used to prevent or delay the onset of a dysbiosis in a subject at risk for developing a dysbiosis.


Methods of Making Enhanced Bacteria

In certain aspects, provided herein are methods of making engineered bacteria for the production of the mEVs (such as smEVs) described herein. In some embodiments, the engineered bacteria are modified to enhance certain desirable properties. For example, in some embodiments, the engineered bacteria are modified to enhance the immunomodulatory and/or therapeutic effect of the mEVs (such as smEVs) (e.g., either alone or in combination with another therapeutic agent), to reduce toxicity and/or to improve bacterial and/or mEV (such as smEV) manufacturing (e.g., higher oxygen tolerance, improved freeze-thaw tolerance, shorter generation times). The engineered bacteria may be produced using any technique known in the art, including but not limited to site-directed mutagenesis, transposon mutagenesis, knock-outs, knock-ins, polymerase chain reaction mutagenesis, chemical mutagenesis, ultraviolet light mutagenesis, transformation (chemically or by electroporation), phage transduction, directed evolution, CRISPR/Cas9, or any combination thereof.


In some embodiments of the methods provided herein, the bacterium is modified by directed evolution. In some embodiments, the directed evolution comprises exposure of the bacterium to an environmental condition and selection of bacterium with improved survival and/or growth under the environmental condition. In some embodiments, the method comprises a screen of mutagenized bacteria using an assay that identifies enhanced bacterium. In some embodiments, the method further comprises mutagenizing the bacteria (e.g., by exposure to chemical mutagens and/or UV radiation) or exposing them to a therapeutic agent (e.g., antibiotic) followed by an assay to detect bacteria having the desired phenotype (e.g., an in vivo assay, an ex vivo assay, or an in vitro assay).


EXAMPLES
Example 1
Purification and Preparation of Membranes from Bacteria to Obtain Processed Microbial Extracellular Vesicles (pmEVs)
Purification

Processed microbial extracellular vesicles (pmEVs) are purified and prepared from bacterial cultures (e.g., bacteria listed in Table 1, Table 2, and/or Table 3) using methods known to those skilled in the art (Thein et al, 2010. Efficient subfractionation of gram-negative bacteria for proteomics studies. J. Proteome Res. 2010 Dec. 3; 9(12): 6135-47. Doi: 10.1021/pr1002438. Epub 2010 Oct. 28; Sandrini et al. 2014. Fractionation by Ultracentrifugation of Gram negative cytoplasmic and membrane proteins. Bio-Protocol. Vol. 4 (21) Doi: 10.21769/BioProtoc.1287).


Alternatively, pmEVs are purified by methods adapted from Thein et al. For example, bacterial cultures are centrifuged at 10,000-15,500×g for 10-30 minutes at room temperature or at 4° C. Supernatants are discarded and cell pellets are frozen at −80° C. Cell pellets are thawed on ice and resuspended in 100 mM Tris-HCl, pH 7.5, and may be supplemented with 1 mg/mL DNase I and/or 100 mM NaCl. Thawed cells are incubated in 500 ug/ml lysozyme, 40 ug/ml lyostaphin, and/or 1 mg/ml DNasel for 40 minutes to facilitate cell lysis. Additional enzymes may be used to facilitate the lysing process (e.g., EDTA (5 mM), PMSF (Sigma Aldrich), and/or benzamidine (Sigma Aldrich). Cells are then lysed using an Emulsiflex C-3 (Avestin, Inc.) under conditions recommended by the manufacturer. Alternatively, pellets may be frozen at −80° C. and thawed again prior to lysis. Debris and unlysed cells are pelleted by centrifugation at 10,000-12,500×g for 15 minutes at 4° C. Supernatants are then centrifuged at 120,000×g for 1 hour at 4° C. Pellets are resuspended in ice-cold 100 mM sodium carbonate, pH 11, incubated with agitation for 1 hour at 4° C. Alternatively, pellets are centrifuged at 120,000×g for 1 hour at 4° C. in sodium carbonate immediately following resuspension. Pellets are resuspended in 100 mM Tris-HCl, pH 7.5 supplemented with 100 mM NaCl re-centrifuged at 120,000×g for 20 minutes at 4° C., and then resuspended in 100 mM Tris-HCl, pH 7.5 supplemented with up to or around 100 mM NaCl or in PBS. Samples are stored at −20° C. To protect the pmEV preparation during the freeze/thaw steps, 250 mM sucrose and up to 500 mM NaCl may be added to the final preparation to stabilize the vesicles in the pmEV preparation.


Alternatively, pmEVs are obtained by methods adapted from Sandrini et al, 2014. After, bacterial cultures are centrifuged at 10,000-15,500×g for 10-15 minutes at room temperature or at 4° C., cell pellets are frozen at −80° C. and supernatants are discarded. Then, cell pellets are thawed on ice and resuspended in 10 mM Tris-HCl, pH 8.0, 1 mM EDTA supplemented with 0.1 mg/mL lysozyme. Samples are then incubated with mixing at room temperature or at 37° C. for 30 min. In an optional step, samples are re-frozen at −80° C. and thawed again on ice. DNase I is added to a final concentration of 1.6 mg/mL and MgCl2 to a final concentration of 100 mM. Samples are sonicated using a QSonica Q500 sonicator with 7 cycles of 30 sec on and 30 sec off. Debris and unlysed cells are pelleted by centrifugation at 10,000×g for 15 min. at 4° C. Supernatants are then centrifuged at 110,000×g for 15 minutes at 4° C. Pellets are resuspended in 10 mM Tris-HCl, pH 8.0 and incubated 30-60 minutes with mixing at room temperature. Samples are centrifuged at 110,000×g for 15 minutes at 4° C. Pellets are resuspended in PBS and stored at −20° C.


Optionally, pmEVs can be separated from other bacterial components and debris using methods known in the art. Size-exclusion chromatography or fast protein liquid chromatography (FPLC) may be used for pmEV purification. Additional separation methods that could be used include field flow fractionation, microfluidic filtering, contact-free sorting, and/or immunoaffinity enrichment chromatography. Alternatively, high resolution density gradient fractionation could be used to separate pmEV particles based on density.


Preparation

Bacterial cultures are centrifuged at 10,000-15,500×g for 10-30 minutes at room temperature or at 4° C. Supernatants are discarded and cell pellets are frozen at −80° C. Cell pellets are thawed on ice and resuspended in 100 mM Tris-HCl, pH 7.5, 100 mM NaCl, 500 ug/ml lysozyme and/or 40 ug/ml Lysostaphin to facilitate cell lysis; up to 0.5 mg/ml DNasel to reduce genomic DNA size, and EDTA (5 mM), PMSF (1 mM, Sigma Aldrich), and Benzamidine (1 mM, Sigma Aldrich) to inhibit proteases. Cells are then lysed using an Emulsiflex C-3 (Avestin, Inc.) under conditions recommended by the manufacturer. Alternatively, pellets may be frozen at −80° C. and thawed again prior to lysis. Debris and unlysed are pelleted by centrifugation at 10,000-12,500×g at for 15 minutes at 4° C. Supernatants are subjected to size exclusion chromatography (Sepharose 4 FF, GE Healthcare) using an FPLC instrument (AKTA Pure 150, GE Healthcare) with PBS and running buffer supplemented with up to 0.3M NaCl. Pure pmEVs are collected in the column void volume, concentrated and stored at −20° C. Concentration may be performed by a number of methods. For example, ultra-centrifugation may be used (1401×g, 1 hour, 4° C., followed by resuspension in small volume of PBS). To protect the pmEV preparation during the freeze-thaw steps, 250 mM sucrose and up to 500 mM NaCl may be added to the final preparation to stabilize the vesicles in the pmEV preparation. Additional separation methods that could be used include field flow fractionation, microfluidic filtering, contact-free sorting, and/or immunoaffinity enrichment chromatography. Other techniques that may be employed using methods known in the arts include Whipped Film Evaporation, Molecular Distillation, Short Pass Distillation, and/or Tangential Flow Filtration.


In some instances, pmEVs are weighed and are administered at varying doses (in ug/ml). Optionally, pmEVs are assessed for particle count and size distribution using Nanoparticle Tracking Analysis (NTA), using methods known in the art. For example, a Malvern NS300 instrument may be used according to manufacturer's instructions or as described by Bachurski et al. 2019. Journal of Extracellular Vesicles. Vol. 8(1). Alternatively, for the pmEVs, total protein may be measured using Bio-rad assays (Cat #5000205) performed per manufacturer's instructions and administered at varying doses based on protein content/dose.


For all of the studies described below, the pmEVs may be irradiated, heated, and/or lyophilized prior to administration (as described in Example 49).


Example 2
A Colorectal Carcinoma Model

To study the efficacy of pmEVs in a tumor model, one of many cancer cell lines may be used according to rodent tumor models known in the art.


For example, female 6-8 week old Balb/c mice are obtained from Taconic (Germantown, N.Y.) or other vendor. 100,000 CT-26 colorectal tumor cells (ATCC CRL-2638) are resuspended in sterile PBS and inoculated in the presence of 50% Matrigel. CT-26 tumor cells are subcutaneously injected into one hind flank of each mouse. When tumor volumes reach an average of 100 mm3 (approximately 10-12 days following tumor cell inoculation), animals are distributed into various treatment groups (e.g., Vehicle; Veillonella pmEVs, Bifidobacteria pmEVs, with or without anti-PD-1 antibody). Antibodies are administered intraperitoneally (i.p.) at 200 μg/mouse (100 μl final volume) every four days, starting on day 1, for a total of 3 times (Q4D×3), and pmEVs are administered orally or intravenously and at varied doses and varied times. For example, pmEVs (5 μg) are intravenously (i.v.) injected every third day, starting on day 1 for a total of 4 times (Q3D×4) and mice are assessed for tumor growth.


Alternatively, when tumor volumes reach an average of 100 mm3 (approximately 10-12 days following tumor cell inoculation), animals are distributed into the following groups: 1) Vehicle; 2) Neisseria Meningitidis pmEVs isolated from the Bexsero® vaccine; and 3) anti-PD-1 antibody. Antibodies are administered intraperitoneally (i.p.) at 200 ug/mouse (100 ul final volume) every four days, starting on day 1, and Neisseria Meningitidis pmEVs are administered intraperitoneally (i.p.) daily, starting on day 1 until the conclusion of the study.


When tumor volumes reached an average of 100 mm3 (approximately 10-12 days following tumor cell inoculation), animals were distributed into the following groups: 1) Vehicle; 2) anti-PD-1 antibody; 3) pmEV B. animalis ssp. lactis (7.0 e+10 particle count); 4) pmEV Anaerostipes hadrus (7.0 e+10 particle count); 5) pmEV S. pyogenes (3.0 e+10 particle count); 6) pmEV P. benzoelyticum (3.0 e+10 particle count); 7) pmEV Hungatella sp. (7.0 e+10 particle count); 8) pmEV S. aureus (7.0 e+10 particle count); and 9) pmEV R. gnavus (7.0 e+10 particle count). Antibodies were administered intraperitoneally (i.p.) at 200 μg/mouse (100 μl final volume) every four days, starting on day 1, and pmEVs were intravenously (i.v.) injected daily, starting on day 1 until the conclusion of the study and tumors measured for growth. At day 11, all of the pmEV groups exhibited tumor growth inhibition (FIGS. 1-7). The pmEV B. animalis ssp. lactis (FIG. 1), pmEV Anaerostipes hadrus (FIG. 2), pmEV S. pyogenes (FIG. 3), pmEV P. benzoelyticum (FIG. 4), and pmEV Hungatella sp. (FIG. 5) groups all showed tumor growth inhibition comparable to the anti-PD-1 group, while the pmEV S. aureus and pmEV R. gnavus groups showed tumor growth inhibition better than that seen in the anti-PD-1 group (FIGS. 6 and 7). In a similar dose-response study, the highest dose of pmEV B. animalis lactis demonstrated the greatest efficacy, although pmEV Megasphaera massiliensis showed significant efficacy at a lower dose (FIG. 8). Welch's test is performed for treatment versus vehicle.


Yet another study demonstrated significant efficacy of pmEVs earlier than on day 11. The pmEV R. gnavus 7.0E+10 (FIGS. 9 and 10), pmEV B. animalis ssp. lactis 2.0E+11 (FIGS. 11 and 12), and pmEV P. distasonis groups 7.0E+10 (FIGS. 13 and 14) all showed efficacy as early as day 9.


Example 3
Administering pmEV Compositions to Treat Mouse Tumor Models

As described in Example 2, a mouse model of cancer is generated by subcutaneously injecting a tumor cell line or patient-derived tumor sample and allowing it to engraft into healthy mice. The methods provided herein may be performed using one of several different tumor cell lines including, but not limited to: B16-F10 or B16-F10-SIY cells as an orthotopic model of melanoma, Panc02 cells as an orthotopic model of pancreatic cancer (Maletzki et al., 2008, Gut 57:483-491), LLC1 cells as an orthotopic model of lung cancer, and RM-1 as an orthotopic model of prostate cancer. As an example, but without limitation, methods for studying the efficacy of pmEVs in the B16-F10 model are provided in depth herein.


A syngeneic mouse model of spontaneous melanoma with a very high metastatic frequency is used to test the ability of bacteria to reduce tumor growth and the spread of metastases. The pmEVs chosen for this assay are compositions that may display enhanced activation of immune cell subsets and stimulate enhanced killing of tumor cells in vitro. The mouse melanoma cell line B16-F10 is obtained from ATCC. The cells are cultured in vitro as a monolayer in RPMI medium, supplemented with 10% heat-inactivated fetal bovine serum and 1% penicillin/streptomycin at 37□ in an atmosphere of 5% CO2 in air. The exponentially growing tumor cells are harvested by trypsinization, washed three times with cold 1× PBS, and a suspension of 5E6 cells/ml is prepared for administration. Female C57BL/6 mice are used for this experiment. The mice are 6-8 weeks old and weigh approximately 16-20 g. For tumor development, each mouse is injected SC into the flank with 100 μl of the B16-F10 cell suspension. The mice are anesthetized by ketamine and xylazine prior to the cell transplantation. The animals used in the experiment may be started on an antibiotic treatment via instillation of a cocktail of kanamycin (0.4 mg/ml), gentamicin, (0.035 mg/ml), colistin (850 U/ml), metronidazole (0.215 mg/ml) and vancomycin (0.045 mg/ml) in the drinking water from day 2 to 5 and an intraperitoneal injection of clindamycin (10 mg/kg) on day 7 after tumor injection.


The size of the primary flank tumor is measured with a caliper every 2-3 days and the tumor volume is calculated using the following formula: tumor volume=the tumor width×tumor length×0.5. After the primary tumor reaches approximately 100 mm3, the animals are sorted into several groups based on their body weight. The mice are then randomly taken from each group and assigned to a treatment group. pmEV compositions are prepared as previously described. The mice are orally inoculated by gavage with approximately 7.0e+09 to 3.0e+12 pmEV particles. Alternatively, pmEVs are administered intravenously. Mice receive pmEVs daily, weekly, bi-weekly, monthly, bi-monthly, or on any other dosing schedule throughout the treatment period. Mice may be IV injected with pmEVs in the tail vein, or directly injected into the tumor. Mice can be injected with pmEVs, with or without live bacteria, with or without inactivated/weakened or killed bacteria. Mice can be injected or orally gavaged weekly or once a month. Mice may receive combinations of purified pmEVs and live bacteria to maximize tumor-killing potential. All mice are housed under specific pathogen-free conditions following approved protocols. Tumor size, mouse weight, and body temperature are monitored every 3-4 days and the mice are humanely sacrificed 6 weeks after the B16-F10 mouse melanoma cell injection or when the volume of the primary tumor reaches 1000 mm3. Blood draws are taken weekly and a full necropsy under sterile conditions is performed at the termination of the protocol.


Cancer cells can be easily visualized in the mouse B16-F10 melanoma model due to their melanin production. Following standard protocols, tissue samples from lymph nodes and organs from the neck and chest region are collected and the presence of micro- and macro-metastases is analyzed using the following classification rule. An organ is classified as positive for metastasis if at least two micro-metastatic and one macro-metastatic lesion per lymph node or organ are found. Micro-metastases are detected by staining the paraffin-embedded lymphoid tissue sections with hematoxylin-eosin following standard protocols known to one skilled in the art. The total number of metastases is correlated to the volume of the primary tumor and it is found that the tumor volume correlates significantly with tumor growth time and the number of macro- and micro-metastases in lymph nodes and visceral organs and also with the sum of all observed metastases. Twenty-five different metastatic sites are identified as previously described (Bobek V., et al., Syngeneic lymph-node-targeting model of green fluorescent protein-expressing Lewis lung carcinoma, Clin. Exp. Metastasis, 2004; 21(8):705-8).


The tumor tissue samples are further analyzed for tumor infiltrating lymphocytes. The CD8+ cytotoxic T cells can be isolated by FACS and can then be further analyzed using customized p/MHC class I microarrays to reveal their antigen specificity (see e.g., Deviren G., et al., Detection of antigen-specific T cells on p/MHC microarrays, J. Mol. Recognit., 2007 January-February; 20(1):32-8). CD4+ T cells can be analyzed using customized p/MHC class II microarrays.


At various timepoints, mice are sacrificed and tumors, lymph nodes, or other tissues may be removed for ex vivo flow cytometric analysis using methods known in the art. For example, tumors are dissociated using a Miltenyi tumor dissociation enzyme cocktail according to the manufacturer's instructions. Tumor weights are recorded and tumors are chopped then placed in 15 ml tubes containing the enzyme cocktail and placed on ice. Samples are then placed on a gentle shaker at 37° C. for 45 minutes and quenched with up to 15 ml complete RPMI. Each cell suspension is strained through a 70 μm filter into a 50 ml falcon tube and centrifuged at 1000 rpm for 10 minutes. Cells are resuspended in FACS buffer and washed to remove remaining debris. If necessary, samples are strained again through a second 70 μm filter into a new tube. Cells are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-MHCII, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Ror□t, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, MHCII, CD206, CD40, CSF1R, PD-L1, Gr-1). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out immune cells obtained from lymph nodes or other tissue, and/or on purified CD45+ tumor-infiltrated immune cells obtained ex vivo. Finally, immunohistochemistry is carried out on tumor sections to measure T cells, macrophages, dendritic cells, and checkpoint molecule protein expression.


The same experiment is also performed with a mouse model of multiple pulmonary melanoma metastases. The mouse melanoma cell line B16-BL6 is obtained from ATCC and the cells are cultured in vitro as described above. Female C57BL/6 mice are used for this experiment. The mice are 6-8 weeks old and weigh approximately 16-20 g. For tumor development, each mouse is injected into the tail vein with 100 μl of a 2E6 cells/ml suspension of B16-BL6 cells. The tumor cells that engraft upon IV injection end up in the lungs.


The mice are humanely killed after 9 days. The lungs are weighed and analyzed for the presence of pulmonary nodules on the lung surface. The extracted lungs are bleached with Fekete's solution, which does not bleach the tumor nodules because of the melanin in the B16 cells though a small fraction of the nodules is amelanotic (i.e. white). The number of tumor nodules is carefully counted to determine the tumor burden in the mice. Typically, 200-250 pulmonary nodules are found on the lungs of the control group mice (i.e. PBS gavage).


The percentage tumor burden is calculated for the three treatment groups. Percentage tumor burden is defined as the mean number of pulmonary nodules on the lung surfaces of mice that belong to a treatment group divided by the mean number of pulmonary nodules on the lung surfaces of the control group mice.


The tumor biopsies and blood samples are submitted for metabolic analysis via LCMS techniques or other methods known in the art. Differential levels of amino acids, sugars, lactate, among other metabolites, between test groups demonstrate the ability of the microbial composition to disrupt the tumor metabolic state.


RNA Seq to Determine Mechanism of Action

Dendritic cells are purified from tumors, Peyers patches, and mesenteric lymph nodes. RNAseq analysis is carried out and analyzed according to standard techniques known to one skilled in the art (Z. Hou. Scientific Reports. 5(9570):doi:10.1038/srep09570 (2015)). In the analysis, specific attention is placed on innate inflammatory pathway genes including TLRs, CLRs, NLRB, and STING, cytokines, chemokines, antigen processing and presentation pathways, cross presentation, and T cell co-stimulation.


Rather than being sacrificed, some mice may be rechallenged with tumor cell injection into the contralateral flank (or other area) to determine the impact of the immune system's memory response on tumor growth.


Example 4
Administering pmEVs to Treat Mouse Tumor Models in Combination with PD-1 or PD-L1 Inhibition

To determine the efficacy of pmEVs in tumor mouse models, in combination with PD-1 or PD-L1 inhibition, a mouse tumor model may be used as described above.


pmEVs are tested for their efficacy in the mouse tumor model, either alone or in combination with whole bacterial cells and with or without anti-PD-1 or anti-PD-L1. pmEVs, bacterial cells, and/or anti-PD-1 or anti-PD-L1 are administered at varied time points and at varied doses. For example, on day 10 after tumor injection, or after the tumor volume reaches 100 mm3, the mice are treated with pmEVs alone or in combination with anti-PD-1 or anti-PD-L1.


Mice may be administered pmEVs orally, intravenously, or intratumorally. For example, some mice are intravenously injected with anywhere between 7.0e+09 to 3.0e+12 pmEV particles. While some mice receive pmEVs through i.v. injection, other mice may receive pmEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, oral gavage, or other means of administration. Some mice may receive pmEVs every day (e.g., starting on day 1), while others may receive pmEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of pmEVs and bacterial cells. For example, the composition may comprise pmEV particles and whole bacteria in a ratio from 1:1 (pmEVs:bacterial cells) to 1-1×1012:1 (pmEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the pmEV administration. As with the pmEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the pmEVs. Some groups of mice are also injected with effective doses of checkpoint inhibitor. For example, mice receive 100 μg anti-PD-L1 mAB (clone 10f.9g2, BioXCell) or another anti-PD-1 or anti-PD-L1 mAB in 100 μl PBS, and some mice receive vehicle and/or other appropriate control (e.g., control antibody). Mice are injected with mABs 3, 6, and 9 days after the initial injection. To assess whether checkpoint inhibition and pmEV immunotherapy have an additive anti-tumor effect, control mice receiving anti-PD-1 or anti-PD-L1 mABs are included to the standard control panel. Primary (tumor size) and secondary (tumor infiltrating lymphocytes and cytokine analysis) endpoints are assessed, and some groups of mice may be rechallenged with a subsequent tumor cell inoculation to assess the effect of treatment on memory response.


Example 5
pmEVs in a Mouse Model of Delayed-Type Hypersensitivity (DTH)

Delayed-type hypersensitivity (DTH) is an animal model of atopic dermatitis (or allergic contact dermatitis), as reviewed by Petersen et al. (In vivo pharmacological disease models for psoriasis and atopic dermatitis in drug discovery. Basic & Clinical Pharm & Toxicology. 2006. 99(2): 104-115; see also Irving C. Allen (ed.) Mouse Models of Innate Immunity: Methods and Protocols, Methods in Molecular Biology, 2013. vol. 1031, DOI 10.1007/978-1-62703-481-4_13). Several variations of the DTH model have been used and are well known in the art (Irving C. Allen (ed.). Mouse Models of Innate Immunity: Methods and Protocols, Methods in Molecular Biology. Vol. 1031, DOI 10.1007/978-1-62703-481-4_13, Springer Science+Business Media, LLC 2013).


DTH can be induced in a variety of mouse and rat strains using various haptens or antigens, for example an antigen emulsified with an adjuvant. DTH is characterized by sensitization as well as an antigen-specific T cell-mediated reaction that results in erythema, edema, and cellular infiltration—especially infiltration of antigen presenting cells (APCs), eosinophils, activated CD4+ T cells, and cytokine-expressing Th2 cells.


Generally, mice are primed with an antigen administered in the context of an adjuvant (e.g., Complete Freund's Adjuvant) in order to induce a secondary (or memory) immune response measured by swelling and antigen-specific antibody titer.


Dexamethasone, a corticosteroid, is a known anti-inflammatory that ameliorates DTH reactions in mice and serves as a positive control for suppressing inflammation in this model (Taube and Carlsten, Action of dexamethasone in the suppression of delayed-type hypersensitivity in reconstituted SCID mice. Inflamm Res. 2000. 49(10): 548-52). For the positive control group, a stock solution of 17 mg/mL of Dexamethasone is prepared on Day 0 by diluting 6.8 mg Dexamethasone in 400 μL 96% ethanol. For each day of dosing, a working solution is prepared by diluting the stock solution 100× in sterile PBS to obtain a final concentration of 0.17 mg/mL in a septum vial for intraperitoneal dosing. Dexamethasone-treated mice receive 100 μL Dexamethasone i.p. (5 mL/kg of a 0.17 mg/mL solution). Frozen sucrose serves as the negative control (vehicle). In the study described below, vehicle, Dexamethasone (positive control) and pmEVs were dosed daily.


pmEVs are tested for their efficacy in the mouse model of DTH, either alone or in combination with whole bacterial cells, with or without the addition of other anti-inflammatory treatments. For example, 6-8 week old C57Bl/6 mice are obtained from Taconic (Germantown, N.Y.), or other vendor. Groups of mice are administered four subcutaneous (s.c.) injections at four sites on the back (upper and lower) of antigen (e.g., Ovalbumin (OVA) or Keyhole Limpet Hemocyanin (KLH)) in an effective dose (e.g., 50 ul total volume per site). For a DTH response, animals are injected intradermally (i.d.) in the ears under ketamine/xylazine anesthesia (approximately 50 mg/kg and 5 mg/kg, respectively). Some mice serve as control animals. Some groups of mice are challenged with 10 ul per ear (vehicle control (0.01% DMSO in saline) in the left ear and antigen (21.2 ug (12 nmol) in the right ear) on day 8. To measure ear inflammation, the ear thickness of manually restrained animals is measured using a Mitutoyo micrometer. The ear thickness is measured before intradermal challenge as the baseline level for each individual animal. Subsequently, the ear thickness is measured two times after intradermal challenge, at approximately 24 hours and 48 hours (i.e., days 9 and 10).


Treatment with pmEVs is initiated at some point, either around the time of priming or around the time of DTH challenge. For example, pmEVs may be administered at the same time as the subcutaneous injections (day 0), or they may be administered prior to, or upon, intradermal injection. pmEVs are administered at varied doses and at defined intervals. For example, some mice are intravenously injected with pmEVs at 10, 15, or 20 ug/mouse. Other mice may receive 25, 50, or 100 mg of pmEVs per mouse. Alternatively, some mice receive between 7.0e+09 to 3.0e+12 pmEV particles per dose. While some mice receive pmEVs through i.v. injection, other mice may receive pmEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, oral gavage, topical administration, intradermal (i.d.) injection, or other means of administration. Some mice may receive pmEVs every day (e.g., starting on day 0), while others may receive pmEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of pmEVs and bacterial cells. For example, the composition may comprise pmEV particles and whole bacteria in a ratio from 1:1 (pmEVs:bacterial cells) to 1-1×1012:1 (pmEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the pmEV administration. As with the pmEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the pmEVs.


For the pmEVs, total protein is measured using Bio-rad assays (Cat #5000205) performed per manufacturer's instructions.


An emulsion of Keyhole Limpet Hemocyanin (KLH) and Complete Freund's Adjuvant (CFA) was prepared freshly on the day of immunization (day 0). To this end, 8 mg of KLH powder is weighed and is thoroughly re-suspended in 16 mL saline. An emulsion was prepared by mixing the KLH/saline with an equal volume of CFA solution (e.g., 10 mL KLH/saline+10 mL CFA solution) using syringes and a luer lock connector. KLH and CFA were mixed vigorously for several minutes to form a white-colored emulsion to obtain maximum stability. A drop test was performed to check if a homogenous emulsion was obtained.


On day 0, C57Bl/6J female mice, approximately 7 weeks old, were primed with KLH antigen in CFA by subcutaneous immunization (4 sites, 50 μL per site). Orally-gavaged P. histicola pmEVs were tested at low (6.0E+07), medium (6.0E+09), and high (6.0E+11) dosages.


On day 8, mice were challenged intradermally (i.d.) with 10 mg KLH in saline (in a volume of 10 μL) in the left ear. Ear pinna thickness was measured at 24 hours following antigen challenge (FIG. 15). As determined by ear thickness, P. histicola pmEVs were efficacious at suppressing inflammation.


For future inflammation studies, some groups of mice may be treated with anti-inflammatory agent(s) (e.g., anti-CD154, blockade of members of the TNF family, or other treatment), and/or an appropriate control (e.g., vehicle or control antibody) at various timepoints and at effective doses.


At various timepoints, serum samples may be taken. Other groups of mice may be sacrificed and lymph nodes, spleen, mesenteric lymph nodes (MLN), the small intestine, colon, and other tissues may be removed for histology studies, ex vivo histological, cytokine and/or flow cytometric analysis using methods known in the art. Some mice are exsanguinated from the orbital plexus under O2/CO2 anesthesia and ELISA assays performed.


Tissues may be dissociated using dissociation enzymes according to the manufacturer's instructions. Cells are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Rory-gamma-t, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, MHCII, CD206, CD40, CSF1R, PD-L1, Gr-1, F4/80). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified CD45+ infiltrated immune cells obtained ex vivo. Finally, immunohistochemistry is carried out on various tissue sections to measure T cells, macrophages, dendritic cells, and checkpoint molecule protein expression.


Ears may be removed from the sacrificed animals and placed in cold EDTA-free protease inhibitor cocktail (Roche). Ears are homogenized using bead disruption and supernatants analyzed for various cytokines by Luminex kit (EMD Millipore) as per manufacturer's instructions. In addition, cervical lymph nodes are dissociated through a cell strainer, washed, and stained for FoxP3 (PE-FJK-16s) and CD25 (FITC-PC61.5) using methods known in the art.


In order to examine the impact and longevity of DTH protection, rather than being sacrificed, some mice may be rechallenged with the challenging antigen at a later time and mice analyzed for susceptibility to DTH and severity of response.


Example 6
pmEVs in a Mouse Model of Experimental Autoimmune Encephalomyelitis (EAE)

EAE is a well-studied animal model of multiple sclerosis, as reviewed by Constantinescu et al., (Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). Br J Pharmacol. 2011 October; 164(4): 1079-1106). It can be induced in a variety of mouse and rat strains using different myelin-associated peptides, by the adoptive transfer of activated encephalitogenic T cells, or the use of TCR transgenic mice susceptible to EAE, as discussed in Mangalam et al., (Two discreet subsets of CD8+ T cells modulate PLP91-110 induced experimental autoimmune encephalomyelitis in HLA-DR3 transgenic mice. J Autoimmun. 2012 June; 38(4): 344-353).


pmEVs are tested for their efficacy in the rodent model of EAE, either alone or in combination with whole bacterial cells, with or without the addition of other anti-inflammatory treatments. Additionally, pmEVs may be administered orally or via intravenous administration. For example, female 6-8 week old C57Bl/6 mice are obtained from Taconic (Germantown, N.Y.). Groups of mice are administered two subcutaneous (s.c.) injections at two sites on the back (upper and lower) of 0.1 ml myelin oligodentrocyte glycoprotein 35-55 (MOG35-55; 100 ug per injection; 200 ug per mouse (total 0.2 ml per mouse)), emulsified in Complete Freund's Adjuvant (CFA; 2-5 mg killed mycobacterium tuberculosis H37Ra/ml emulsion). Approximately 1-2 hours after the above, mice are intraperitoneally (i.p.) injected with 200 ng Pertussis toxin (PTx) in 0.1 ml PBS (2 ug/ml). An additional IP injection of PTx is administered on day 2. Alternatively, an appropriate amount of an alternative myelin peptide (e.g., proteolipid protein (PLP)) is used to induce EAE. Some animals serve as naïve controls. EAE severity is assessed and a disability score is assigned daily beginning on day 4 according to methods known in the art (Mangalam et al. 2012).


Treatment with pmEVs is initiated at some point, either around the time of immunization or following EAE immunization. For example, pmEVs may be administered at the same time as immunization (day 1), or they may be administered upon the first signs of disability (e.g., limp tail), or during severe EAE. pmEVs are administered at varied doses and at defined intervals. For example, some mice are intravenously injected with pmEVs at 10, 15, or 20 ug/mouse. Other mice may receive 25, 50, or 100 mg of pmEVs per mouse. Alternatively, some mice receive between 7.0e+09 to 3.0e+12 pmEV particles per dose. While some mice receive pmEVs through i.v. injection, other mice may receive pmEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, oral gavage, or other means of administration. Some mice may receive pmEVs every day (e.g., starting on day 1), while others may receive pmEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of pmEVs and bacterial cells. For example, the composition may comprise pmEV particles and whole bacteria in a ratio from 1:1 (pmEVs:bacterial cells) to 1-1×1012:1 (pmEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the pmEV administration. As with the pmEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the pmEVs.


Some groups of mice may be treated with additional anti-inflammatory agent(s) or EAE therapeutic(s) (e.g., anti-CD154, blockade of members of the TNF family, Vitamin D, steroids, anti-inflammatory agents, or other treatment(s)), and/or an appropriate control (e.g., vehicle or control antibody) at various time points and at effective doses.


In addition, some mice are treated with antibiotics prior to treatment. For example, vancomycin (0.5 g/L), ampicillin (1.0 g/L), gentamicin (1.0 g/L) and amphotericin B (0.2 g/L) are added to the drinking water, and antibiotic treatment is halted at the time of treatment or a few days prior to treatment. Some immunized mice are treated without receiving antibiotics.


At various timepoints, mice are sacrificed and sites of inflammation (e.g., brain and spinal cord), lymph nodes, or other tissues may be removed for ex vivo histological, cytokine and/or flow cytometric analysis using methods known in the art. For example, tissues are dissociated using dissociation enzymes according to the manufacturer's instructions. Cells are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-MITCH, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Roryt, Granzyme B, CD69, PD-1, C′TLA-4), and macrophage/myeloid markers (CD11b, MHCII, CD206, CD40, CSF1R, PD-L1, Gr-1, F4/80). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified CD45+ central nervous system (CNS)-infiltrated immune cells obtained ex vivo. Finally, immunohistochemistry is carried out on various tissue sections to measure T cells, macrophages, dendritic cells, and checkpoint molecule protein expression.


In order to examine the impact and longevity of disease protection, rather than being sacrificed, some mice may be rechallenged with a disease trigger (e.g., activated encephalitogenic T cells or re-injection of EAE-inducing peptides). Mice are analyzed for susceptibility to disease and EAE severity following rechallenge.


Example 7
pmEVs in a Mouse Model of Collagen-Induced Arthritis (CIA)

Collagen-induced arthritis (CIA) is an animal model commonly used to study rheumatoid arthritis (RA), as described by Caplazi et al. (Mouse models of rheumatoid arthritis. Veterinary Pathology. Sep. 1, 2015. 52(5): 819-826) (see also Brand et al. Collagen-induced arthritis. Nature Protocols. 2007. 2: 1269-1275; Pietrosimone et al. Collagen-induced arthritis: a model for murine autoimmune arthritis. Bio Protoc. 2015 Oct. 20; 5(20): e1626).


Among other versions of the CIA rodent model, one model involves immunizing HLA-DQ8 Tg mice with chick type II collagen as described by Taneja et al. (J. Immunology. 2007. 56: 69-78; see also Taneja et al. J. Immunology 2008. 181: 2869-2877; and Taneja et al. Arthritis Rheum., 2007. 56: 69-78). Purification of chick CII has been described by Taneja et al. (Arthritis Rheum., 2007. 56: 69-78). Mice are monitored for CIA disease onset and progression following immunization, and severity of disease is evaluated and “graded” as described by Wooley, J. Exp. Med. 1981. 154: 688-700.


Mice are immunized for CIA induction and separated into various treatment groups. pmEVs are tested for their efficacy in CIA, either alone or in combination with whole bacterial cells, with or without the addition of other anti-inflammatory treatments.


Treatment with pmEVs is initiated either around the time of immunization with collagen or post-immunization. For example, in some groups, pmEVs may be administered at the same time as immunization (day 1), or pmEVs may be administered upon first signs of disease, or upon the onset of severe symptoms. pmEVs are administered at varied doses and at defined intervals. For example, some mice are intravenously injected with pmEVs at 10, 15, or 20 ug/mouse. Other mice may receive 25, 50, or 100 mg of pmEVs per mouse. Alternatively, some mice receive between 7.0e+09 to 3.0e+12 pmEV particles per dose. While some mice receive pmEVs through oral gavage or i.v. injection, while other groups of mice may receive pmEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, or other means of administration. Some mice may receive pmEVs every day (e.g., starting on day 1), while others may receive pmEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of pmEVs and bacterial cells. For example, the composition may comprise pmEV particles and whole bacteria in a ratio from 1:1 (pmEVs:bacterial cells) to 1-1×1012:1 (pmEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the pmEV administration. As with the pmEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the pmEVs.


Some groups of mice may be treated with additional anti-inflammatory agent(s) or CIA therapeutic(s) (e.g., anti-CD154, blockade of members of the TNF family, Vitamin D, steroid(s), anti-inflammatory agent(s), and/or other treatment), and/or an appropriate control (e.g., vehicle or control antibody) at various timepoints and at effective doses.


In addition, some mice are treated with antibiotics prior to treatment. For example, vancomycin (0.5 g/L), ampicillin (1.0 g/L), gentamicin (1.0 g/L) and amphotericin B (0.2 g/L) are added to the drinking water, and antibiotic treatment is halted at the time of treatment or a few days prior to treatment. Some immunized mice are treated without receiving antibiotics.


At various timepoints, serum samples are obtained to assess levels of anti-chick and anti-mouse CII IgG antibodies using a standard ELISA (Batsalova et al. Comparative analysis of collagen type II-specific immune responses during development of collagen-induced arthritis in two B10 mouse strains. Arthritis Res Ther. 2012. 14(6): R237). Also, some mice are sacrificed and sites of inflammation (e.g., synovium), lymph nodes, or other tissues may be removed for ex vivo histological, cytokine and/or flow cytometric analysis using methods known in the art. The synovium and synovial fluid are analyzed for plasma cell infiltration and the presence of antibodies using techniques known in the art. In addition, tissues are dissociated using dissociation enzymes according to the manufacturer's instructions to examine the profiles of the cellular infiltrates. Cells are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Roryt, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, MHCII, CD206, CD40, CSF1R, PD-L1, Gr-1, F4/80). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified CD45+ synovium-infiltrated immune cells obtained ex vivo. Finally, immunohistochemistry is carried out on various tissue sections to measure T cells, macrophages, dendritic cells, and checkpoint molecule protein expression.


In order to examine the impact and longevity of disease protection, rather than being sacrificed, some mice may be rechallenged with a disease trigger (e.g., activated re-injection with CIA-inducing peptides). Mice are analyzed for susceptibility to disease and CIA severity following rechallenge.


Example 8
pmEVs in a Mouse Model of Colitis

Dextran sulfate sodium (DSS)-induced colitis is a well-studied animal model of colitis, as reviewed by Randhawa et al. (A review on chemical-induced inflammatory bowel disease models in rodents. Korean J Physiol Pharmacol. 2014. 18(4): 279-288; see also Chassaing et al. Dextran sulfate sodium (DSS)-induced colitis in mice. Curr Protoc Immunol. 2014 Feb. 4; 104: Unit 15.25).


pmEVs are tested for their efficacy in a mouse model of DSS-induced colitis, either alone or in combination with whole bacterial cells, with or without the addition of other anti-inflammatory agents.


Groups of mice are treated with DSS to induce colitis as known in the art (Randhawa et al. 2014; Chassaing et al. 2014; see also Kim et al. Investigating intestinal inflammation in DSS-induced model of IBD. J Vis Exp. 2012. 60: 3678). For example, male 6-8 week old C57Bl/6 mice are obtained from Charles River Labs, Taconic, or other vendor. Colitis is induced by adding 3% DSS (MP Biomedicals, Cat. #0260110) to the drinking water. Some mice do not receive DSS in the drinking water and serve as naïve controls. Some mice receive water for five (5) days. Some mice may receive DSS for a shorter duration or longer than five (5) days. Mice are monitored and scored using a disability activity index known in the art based on weight loss (e.g., no weight loss (score 0); 1-5% weight loss (score 1); 5-10% weight loss (score 2)); stool consistency (e.g., normal (score 0); loose stool (score 2); diarrhea (score 4)); and bleeding (e.g., no blood (score 0), hemoccult positive (score 1); hemoccult positive and visual pellet bleeding (score 2); blood around anus, gross bleeding (score 4).


Treatment with pmEVs is initiated at some point, either on day 1 of DSS administration, or sometime thereafter. For example, pmEVs may be administered at the same time as DSS initiation (day 1), or they may be administered upon the first signs of disease (e.g., weight loss or diarrhea), or during the stages of severe colitis. Mice are observed daily for weight, morbidity, survival, presence of diarrhea and/or bloody stool.


pmEVs are administered at various doses and at defined intervals. For example, some mice receive between 7.0e+09 and 3.0e+12 pmEV particles. While some mice receive pmEVs through oral gavage or i.v. injection, while other groups of mice may receive pmEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, or other means of administration. Some mice may receive pmEVs every day (e.g., starting on day 1), while others may receive pmEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of pmEVs and bacterial cells. For example, the composition may comprise pmEV particles and whole bacteria in a ratio from 1:1 (pmEVs:bacterial cells) to 1-1×1012:1 (pmEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the pmEV administration. As with the pmEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the pmEVs.


Some groups of mice may be treated with additional anti-inflammatory agent(s) (e.g., anti-CD154, blockade of members of the TNF family, or other treatment), and/or an appropriate control (e.g., vehicle or control antibody) at various timepoints and at effective doses.


In addition, some mice are treated with antibiotics prior to treatment. For example, vancomycin (0.5 g/L), ampicillin (1.0 g/L), gentamicin (1.0 g/L) and amphotericin B (0.2 g/L) are added to the drinking water, and antibiotic treatment is halted at the time of treatment or a few days prior to treatment. Some mice receive DSS without receiving antibiotics beforehand.


At various timepoints, mice undergo video endoscopy using a small animal endoscope (Karl Storz Endoskipe, Germany) under isoflurane anesthesia. Still images and video are recorded to evaluate the extent of colitis and the response to treatment. Colitis is scored using criteria known in the art. Fecal material is collected for study.


At various timepoints, mice are sacrificed and the colon, small intestine, spleen, and lymph nodes (e.g., mesenteric lymph nodes) are collected. Additionally, blood is collected into serum separation tubes. Tissue damage is assessed through histological studies that evaluate, but are not limited to, crypt architecture, degree of inflammatory cell infiltration, and goblet cell depletion.


The gastrointestinal (GI) tract, lymph nodes, and/or other tissues may be removed for ex vivo histological, cytokine and/or flow cytometric analysis using methods known in the art. For example, tissues are harvested and may be dissociated using dissociation enzymes according to the manufacturer's instructions. Cells are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-MHCII, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Roryt, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, MI-ICII, CD206, CD40, CSF1R, PD-L1, Gr-1, F4/80). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified CD45+ GI tract-infiltrated immune cells obtained ex vivo. Finally, immunohistochemistry is carried out on various tissue sections to measure T cells, macrophages, dendritic cells, and checkpoint molecule protein expression.


In order to examine the impact and longevity of disease protection, rather than being sacrificed, some mice may be rechallenged with a disease trigger. Mice are analyzed for susceptibility to colitis severity following rechallenge.


Example 9
pmEVs in a Mouse Model of Type 1 Diabetes (T1D)

Type 1 diabetes (T1D) is an autoimmune disease in which the immune system targets the islets of Langerhans of the pancreas, thereby destroying the body's ability to produce insulin.


There are various models of animal models of T1D, as reviewed by Belle et al. (Mouse models for type 1 diabetes. Drug Discov Today Dis Models. 2009; 6(2): 41-45; see also Aileen JF King. The use of animal models in diabetes research. Br J Pharmacol. 2012 June; 166(3): 877-894. There are models for chemically-induced T1D, pathogen-induced T1D, as well as models in which the mice spontaneously develop T1D.


pmEVs are tested for their efficacy in a mouse model of T1D, either alone or in combination with whole bacterial cells, with or without the addition of other anti-inflammatory treatments.


Depending on the method of T1D induction and/or whether T1D development is spontaneous, treatment with pmEVs is initiated at some point, either around the time of induction or following induction, or prior to the onset (or upon the onset) of spontaneously-occurring T1D. pmEVs are administered at varied doses and at defined intervals. For example, some mice are intravenously injected with pmEVs at 10, 15, or 20 ug/mouse. Other mice may receive 25, 50, or 100 mg of pmEVs per mouse. Alternatively, some mice receive between 7.0e+09 to 3.0e+12 pmEV particles per dose. While some mice receive pmEVs through oral gavage or i.v. injection, while other groups of mice may receive pmEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, or other means of administration. Some mice may receive pmEVs every day, while others may receive pmEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of pmEVs and bacterial cells. For example, the composition may comprise pmEV particles and whole bacteria in a ratio from 1:1 (pmEVs:bacterial cells) to 1-1×1012:1 (pmEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the pmEV administration. As with the pmEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the pmEVs.


Some groups of mice may be treated with additional treatments and/or an appropriate control (e.g., vehicle or control antibody) at various timepoints and at effective doses.


In addition, some mice are treated with antibiotics prior to treatment. For example, vancomycin (0.5 g/L), ampicillin (1.0 g/L), gentamicin (1.0 g/L) and amphotericin B (0.2 g/L) are added to the drinking water, and antibiotic treatment is halted at the time of treatment or a few days prior to treatment. Some immunized mice are treated without receiving antibiotics.


Blood glucose is monitored biweekly prior to the start of the experiment. At various timepoints thereafter, nonfasting blood glucose is measured. At various timepoints, mice are sacrificed and site the pancreas, lymph nodes, or other tissues may be removed for ex vivo histological, cytokine and/or flow cytometric analysis using methods known in the art. For example, tissues are dissociated using dissociation enzymes according to the manufacturer's instructions. Cells are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-MHCII, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Roryt, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, MHCII, CD206, CD40, CSF1R, PD-L1, Gr-1, F4/80). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified tissue-infiltrating immune cells obtained ex vivo. Finally, immunohistochemistry is carried out on various tissue sections to measure T cells, macrophages, dendritic cells, and checkpoint molecule protein expression. Antibody production may also be assessed by ELISA.


In order to examine the impact and longevity of disease protection, rather than being sacrificed, some mice may be rechallenged with a disease trigger, or assessed for susceptibility to relapse. Mice are analyzed for susceptibility to diabetes onset and severity following rechallenge (or spontaneously-occurring relapse).


Example 10
pmEVs in a Mouse Model of Primary Sclerosing Cholangitis (PSC)

Primary Sclerosing Cholangitis (PSC) is a chronic liver disease that slowly damages the bile ducts and leads to end-stage cirrhosis. It is associated with inflammatory bowel disease (IBD).


There are various animal models for PSC, as reviewed by Fickert et al. (Characterization of animal models for primary sclerosing cholangitis (PSC). J Hepatol. 2014 June. 60(6): 1290-1303; see also Pollheimer and Fickert. Animal models in primary biliary cirrhosis and primary sclerosing cholangitis. Clin Rev Allergy Immunol. 2015 June. 48(2-3): 207-17). Induction of disease in PSC models includes chemical induction (e.g., 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-induced cholangitis), pathogen-induced (e.g., Cryptosporidium parvum), experimental biliary obstruction (e.g., common bile duct ligation (CBDL)), and transgenic mouse model of antigen-driven biliary injury (e.g., Ova-Bil transgenic mice). For example, bile duct ligation is performed as described by Georgiev et al. (Characterization of time-related changes after experimental bile duct ligation. Br J Surg. 2008. 95(5): 646-56), or disease is induced by DCC exposure as described by Fickert et al. (A new xenobiotic-induced mouse model of sclerosing cholangitis and biliary fibrosis. Am J Path. Vol 171(2): 525-536.


pmEVs are tested for their efficacy in a mouse model of PSC, either alone or in combination with whole bacterial cells, with or without the addition of some other therapeutic agent.


DCC-Induced Cholangitis

For example, 6-8 week old C57bl/6 mice are obtained from Taconic or other vendor. Mice are fed a 0.1% DCC-supplemented diet for various durations. Some groups receive DCC-supplement food for 1 week, others for 4 weeks, others for 8 weeks. Some groups of mice may receive a DCC-supplemented diet for a length of time and then be allowed to recover, thereafter receiving a normal diet. These mice may be studied for their ability to recover from disease and/or their susceptibility to relapse upon subsequent exposure to DCC. Treatment with pmEVs is initiated at some point, either around the time of DCC-feeding or subsequent to initial exposure to DCC. For example, pmEVs may be administered on day 1, or they may be administered sometime thereafter. pmEVs are administered at varied doses and at defined intervals. For example, some mice are intravenously injected with pmEVs at 10, 15, or 20 ug/mouse. Alternatively, some mice may receive between 7.0e+09 and 3.0e+12 pmEV particles. While some mice receive pmEVs through oral gavage or i.v. injection, while other groups of mice may receive pmEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, or other means of administration. Some mice may receive pmEVs every day (e.g., starting on day 1), while others may receive pmEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of pmEVs and bacterial cells. For example, the composition may comprise pmEV particles and whole bacteria in a ratio from 1:1 (pmEVs:bacterial cells) to 1-1×1012:1 (pmEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the pmEV administration. As with the pmEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the pmEVs.


Some groups of mice may be treated with additional agents and/or an appropriate control (e.g., vehicle or antibody) at various timepoints and at effective doses.


In addition, some mice are treated with antibiotics prior to treatment. For example, vancomycin (0.5 g/L), ampicillin (1.0 g/L), gentamicin (1.0 g/L) and amphotericin B (0.2 g/L) are added to the drinking water, and antibiotic treatment is halted at the time of treatment or a few days prior to treatment. Some immunized mice are treated without receiving antibiotics. At various timepoints, serum samples are analyzed for ALT, AP, bilirubin, and serum bile acid (BA) levels.


At various timepoints, mice are sacrificed, body and liver weight are recorded, and sites of inflammation (e.g., liver, small and large intestine, spleen), lymph nodes, or other tissues may be removed for ex vivo histolomorphological characterization, cytokine and/or flow cytometric analysis using methods known in the art (see Fickert et al. Characterization of animal models for primary sclerosing cholangitis (PSC)). J Hepatol. 2014. 60(6): 1290-1303). For example, bile ducts are stained for expression of ICAM-1, VCAM-1, MadCAM-1. Some tissues are stained for histological examination, while others are dissociated using dissociation enzymes according to the manufacturer's instructions. Cells are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-MHCII, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Roryt, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, MHCII, CD206, CD40, CSF1R, PD-L1, Gr-1, F4/80), as well as adhesion molecule expression (ICAM-1, VCAM-1, MadCAM-1). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified CD45+ bile duct-infiltrated immune cells obtained ex vivo.


Liver tissue is prepared for histological analysis, for example, using Sirius-red staining followed by quantification of the fibrotic area. At the end of the treatment, blood is collected for plasma analysis of liver enzymes, for example, AST or ALT, and to determine Bilirubin levels. The hepatic content of Hydroxyproline can be measured using established protocols. Hepatic gene expression analysis of inflammation and fibrosis markers may be performed by qRT-PCR using validated primers. These markers may include, but are not limited to, MCP-1, alpha-SMA, Coll1a1, and TIMP. Metabolite measurements may be performed in plasma, tissue and fecal samples using established metabolomics methods. Finally, immunohistochemistry is carried out on liver sections to measure neutrophils, T cells, macrophages, dendritic cells, or other immune cell infiltrates.


In order to examine the impact and longevity of disease protection, rather than being sacrificed, some mice may be rechallenged with DCC at a later time. Mice are analyzed for susceptibility to cholangitis and cholangitis severity following rechallenge.


BDL-Induced Cholangitis

Alternatively, pmEVs are tested for their efficacy in BDL-induced cholangitis. For example, 6-8 week old C57Bl/6J mice are obtained from Taconic or other vendor. After an acclimation period the mice are subjected to a surgical procedure to perform a bile duct ligation (BDL). Some control animals receive a sham surgery. The BDL procedure leads to liver injury, inflammation and fibrosis within 7-21 days.


Treatment with pmEVs is initiated at some point, either around the time of surgery or some time following the surgery. pmEVs are administered at varied doses and at defined intervals. For example, some mice are intravenously injected with pmEVs at 10, 15, or 20 ug/mouse. Other mice may receive 25, 50, or 100 mg of pmEVs per mouse. Alternatively, some mice receive between 7.0e+09 to 3.0e+12 pmEV particles per dose. While some mice receive pmEVs through oral gavage or i.v. injection, while other groups of mice may receive pmEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, or other means of administration. Some mice receive pmEVs every day (e.g., starting on day 1), while others may receive pmEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of pmEVs and bacterial cells. For example, the composition may comprise pmEV particles and whole bacteria in a ratio from 1:1 (pmEVs:bacterial cells) to 1-1×1012:1 (pmEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the pmEV administration. As with the pmEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the pmEVs.


Some groups of mice may be treated with additional agents and/or an appropriate control (e.g., vehicle or antibody) at various timepoints and at effective doses.


In addition, some mice are treated with antibiotics prior to treatment. For example, vancomycin (0.5 g/L), ampicillin (1.0 g/L), gentamicin (1.0 g/L) and amphotericin B (0.2 g/L) are added to the drinking water, and antibiotic treatment is halted at the time of treatment or a few days prior to treatment. Some immunized mice are treated without receiving antibiotics. At various timepoints, serum samples are analyzed for ALT, AP, bilirubin, and serum bile acid (BA) levels.


At various timepoints, mice are sacrificed, body and liver weight are recorded, and sites of inflammation (e.g., liver, small and large intestine, spleen), lymph nodes, or other tissues may be removed for ex vivo histolomorphological characterization, cytokine and/or flow cytometric analysis using methods known in the art (see Fickert et al. Characterization of animal models for primary sclerosing cholangitis (PSC)). J Hepatol. 2014. 60(6): 1290-1303). For example, bile ducts are stained for expression of ICAM-1, VCAM-1, MadCAM-1. Some tissues are stained for histological examination, while others are dissociated using dissociation enzymes according to the manufacturer's instructions. Cells are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-MHCII, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Roryt, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, MHCII, CD206, CD40, CSF1R, PD-L1, Gr-1, F4/80), as well as adhesion molecule expression (ICAM-1, VCAM-1, MadCAM-1). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified CD45+ bile duct-infiltrated immune cells obtained ex vivo.


Liver tissue is prepared for histological analysis, for example, using Sirius-red staining followed by quantification of the fibrotic area. At the end of the treatment, blood is collected for plasma analysis of liver enzymes, for example, AST or ALT, and to determine Bilirubin levels. The hepatic content of Hydroxyproline can be measured using established protocols. Hepatic gene expression analysis of inflammation and fibrosis markers may be performed by qRT-PCR using validated primers. These markers may include, but are not limited to, MCP-1, alpha-SMA, Coll1a1, and TIMP. Metabolite measurements may be performed in plasma, tissue and fecal samples using established metabolomics methods. Finally, immunohistochemistry is carried out on liver sections to measure neutrophils, T cells, macrophages, dendritic cells, or other immune cell infiltrates.


In order to examine the impact and longevity of disease protection, rather than being sacrificed, some mice may be analyzed for recovery.


Example 11
pmEVs in a Mouse Model of Nonalcoholic Steatohepatitis (NASH)

Nonalcoholic Steatohepatitis (NASH) is a severe form of Nonalcoholic Fatty Liver Disease (NAFLD), where buildup of hepatic fat (steatosis) and inflammation lead to liver injury and hepatocyte cell death (ballooning).


There are various animal models of NASH, as reviewed by Ibrahim et al. (Animal models of nonalcoholic steatohepatitis: Eat, Delete, and Inflame. Dig Dis Sci. 2016 May. 61(5): 1325-1336; see also Lau et al. Animal models of non-alcoholic fatty liver disease: current perspectives and recent advances 2017 January. 241(1): 36-44).


pmEVs are tested for their efficacy in a mouse model of NASH, either alone or in combination with whole bacterial cells, with or without the addition of another therapeutic agent. For example, 8-10 week old C57Bl/6J mice, obtained from Taconic (Germantown, N.Y.), or other vendor, are placed on a methionine choline deficient (MCD) diet for a period of 4-8 weeks during which NASH features develop, including steatosis, inflammation, ballooning and fibrosis.



P. histicola pmEVs are tested for their efficacy in a mouse model of NASH, either alone or in combination with each other, in varying proportions, with or without the addition of another therapeutic agent. For example, 8 week old C57Bl/6J mice, obtained from Charles River (France), or other vendor, are acclimated for a period of 5 days, randomized intro groups of 10 mice based on body weight, and placed on a methionine choline deficient (MCD) diet for example A02082002B from Research Diets (USA), for a period of 4 weeks during which NASH features developed, including steatosis, inflammation, ballooning and fibrosis. Control chow mice are fed a normal chow diet, for example RM1 (E) 801492 from SDS Diets (UK). Control chow, MCD diet, and water are provided ad libitum.


An NAS scoring system adapted from Kleiner et al. (Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology. 2005 June. 41(6): 1313-1321) is used to determine the degree of steatosis (scored 0-3), lobular inflammation (scored 0-3), hepatocyte ballooning (scored 0-3), and fibrosis (scored 0-4). An individual mouse NAS score may be calculated by summing the score for steatosis, inflammation, ballooning, and fibrosis (scored 0-13). In addition, the levels of plasma AST and ALT are determined using a Pentra 400 instrument from Horiba (USA), according to manufacturer's instructions. The levels of hepatic total cholesterol, triglycerides, fatty acids, alanine aminotransferase, and aspartate aminotransferase are also determined using methods known in the art.


In other studies, hepatic gene expression analysis of inflammation, fibrosis, steatosis, ER stress, or oxidative stress markers may be performed by qRT-PCR using validated primers. These markers may include, but are not limited to, IL-1β, TNF-α, MCP-1, α-SMA, Coll1a1, CHOP, and NRF2.


In other studies, hepatic gene expression analysis of inflammation, fibrosis, steatosis, ER stress, or oxidative stress markers may be performed by qRT-PCR using validated primers. These markers may include, but are not limited to, IL-1β, TNF-α, MCP-1, α-SMA, Coll1a1, CHOP, and NRF2.


Treatment with pmEVs is initiated at some point, either at the beginning of the diet, or at some point following diet initiation (for example, one week after). For example, pmEVs may be administered starting in the same day as the initiation of the MCD diet. pmEVs are administered at varied doses and at defined intervals. For example, some mice are intravenously injected with pmEVs at 10, 15, or 20 ug/mouse. Other mice may receive 25, 50, or 100 mg of pmEVs per mouse. Alternatively, some mice receive between 7.0e+09 to 3.0e+12 pmEV particles per dose. While some mice receive pmEVs through oral gavage or i.v. injection, while other groups of mice may receive pmEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, or other means of administration. Some mice may receive pmEVs every day (e.g., starting on day 1), while others may receive pmEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of pmEVs and bacterial cells. For example, the composition may comprise pmEV particles and whole bacteria in a ratio from 1:1 (pmEVs:bacterial cells) to 1-1×1012:1 (pmEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the pmEV administration. As with the pmEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the pmEVs.


Some groups of mice may be treated with additional NASH therapeutic(s) (e.g., FXR agonists, PPAR agonists, CCR2/5 antagonists or other treatment) and/or appropriate control at various timepoints and effective doses.


At various timepoints and/or at the end of the treatment, mice are sacrificed and liver, intestine, blood, feces, or other tissues may be removed for ex vivo histological, biochemical, molecular or cytokine and/or flow cytometry analysis using methods known in the art. For example, liver tissues are weighed and prepared for histological analysis, which may comprise staining with H&E, Sirius Red, and determination of NASH activity score (NAS). At various timepoints, blood is collected for plasma analysis of liver enzymes, for example, AST or ALT, using standards assays. In addition, the hepatic content of cholesterol, triglycerides, or fatty acid acids can be measured using established protocols. Hepatic gene expression analysis of inflammation, fibrosis, steatosis, ER stress, or oxidative stress markers may be performed by qRT-PCR using validated primers. These markers may include, but are not limited to, IL-6, MCP-1, alpha-SMA, Coll1a1, CHOP, and NRF2. Metabolite measurements may be performed in plasma, tissue and fecal samples using established biochemical and mass-spectrometry-based metabolomics methods. Serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified CD45+ bile duct-infiltrated immune cells obtained ex vivo. Finally, immunohistochemistry is carried out on liver or intestine sections to measure neutrophils, T cells, macrophages, dendritic cells, or other immune cell infiltrates.


In order to examine the impact and longevity of disease protection, rather than being sacrificed, some mice may be analyzed for recovery.


Example 12
pmEVs in a Mouse Model of Psoriasis

Psoriasis is a T-cell-mediated chronic inflammatory skin disease. So-called “plaque-type” psoriasis is the most common form of psoriasis and is typified by dry scales, red plaques, and thickening of the skin due to infiltration of immune cells into the dermis and epidermis. Several animal models have contributed to the understanding of this disease, as reviewed by Gudjonsson et al. (Mouse models of psoriasis. J Invest Derm. 2007. 127: 1292-1308; see also van der Fits et al. Imiquimod-induced psoriasis-like skin inflammation in mice is mediated via the IL-23/IL-17 axis. J. Immunol. 2009 May 1. 182(9): 5836-45).


Psoriasis can be induced in a variety of mouse models, including those that use transgenic, knockout, or xenograft models, as well as topical application of imiquimod (IMQ), a TLR7/8 ligand.


pmEVs are tested for their efficacy in the mouse model of psoriasis, either alone or in combination with whole bacterial cells, with or without the addition of other anti-inflammatory treatments. For example, 6-8 week old C57Bl/6 or Balb/c mice are obtained from Taconic (Germantown, N.Y.), or other vendor. Mice are shaved on the back and the right ear. Groups of mice receive a daily topical dose of 62.5 mg of commercially available IMQ cream (5%) (Aldara; 3M Pharmaceuticals). The dose is applied to the shaved areas for 5 or 6 consecutive days. At regular intervals, mice are scored for erythema, scaling, and thickening on a scale from 0 to 4, as described by van der Fits et al. (2009). Mice are monitored for ear thickness using a Mitutoyo micrometer.


Treatment with pmEVs is initiated at some point, either around the time of the first application of IMQ, or something thereafter. For example, pmEVs may be administered at the same time as the subcutaneous injections (day 0), or they may be administered prior to, or upon, application. pmEVs are administered at varied doses and at defined intervals. For example, some mice are intravenously injected with pmEVs at 10, 15, or 20 ug/mouse. Other mice may receive 25, 50, or 100 mg of pmEVs per mouse. Alternatively, some mice receive between 7.0e+09 to 3.0e+12 pmEV particles per dose. While some mice receive pmEVs through oral gavage or i.v. injection, while other groups of mice may receive pmEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, or other means of administration. Some mice may receive pmEVs every day (e.g., starting on day 0), while others may receive pmEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of pmEVs and bacterial cells. For example, the composition may comprise pmEV particles and whole bacteria in a ratio from 1:1 (pmEVs:bacterial cells) to 1-1×1012:1 (pmEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the pmEV administration. As with the pmEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the pmEVs.


Some groups of mice may be treated with anti-inflammatory agent(s) (e.g., anti-CD154, blockade of members of the TNF family, or other treatment), and/or an appropriate control (e.g., vehicle or control antibody) at various timepoints and at effective doses.


In addition, some mice are treated with antibiotics prior to treatment. For example, vancomycin (0.5 g/L), ampicillin (1.0 g/L), gentamicin (1.0 g/L) and amphotericin B (0.2 g/L) are added to the drinking water, and antibiotic treatment is halted at the time of treatment or a few days prior to treatment. Some immunized mice are treated without receiving antibiotics.


At various timepoints, samples from back and ear skin are taken for cryosection staining analysis using methods known in the art. Other groups of mice are sacrificed and lymph nodes, spleen, mesenteric lymph nodes (MLN), the small intestine, colon, and other tissues may be removed for histology studies, ex vivo histological, cytokine and/or flow cytometric analysis using methods known in the art. Some tissues may be dissociated using dissociation enzymes according to the manufacturer's instructions. Cryosection samples, tissue samples, or cells obtained ex vivo are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-MHCII, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Roryt, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, MHCII, CD206, CD40, CSF1R, PD-L1, Gr-1, F4/80). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified CD45+ skin-infiltrated immune cells obtained ex vivo. Finally, immunohistochemistry is carried out on various tissue sections to measure T cells, macrophages, dendritic cells, and checkpoint molecule protein expression.


In order to examine the impact and longevity of psoriasis protection, rather than being sacrificed, some mice may be studied to assess recovery, or they may be rechallenged with IMQ. The groups of rechallenged mice are analyzed for susceptibility to psoriasis and severity of response.


Example 13
pmEVs in a Mouse Model of Obesity (DIO)

There are various animal models of DIO, as reviewed by Tschop et al. (A guide to analysis of mouse energy metabolism. Nat. Methods. 2012; 9(1):57-63) and Ayala et al. (Standard operating procedures for describing and performing metabolic tests of glucose homeostasis in mice. Disease Models and Mechanisms. 2010; 3:525-534) and provided by Physiogenex.


pmEVs are tested for their efficacy in a mouse model of DIO, either alone or in combination with other whole bacterial cells (live, killed, irradiated, and/or inactivated, etc) with or without the addition of other anti-inflammatory treatments.


Depending on the method of DIO induction and/or whether DIO development is spontaneous, treatment with pmEVs is initiated at some point, either around the time of induction or following induction, or prior to the onset (or upon the onset) of spontaneously-occurring T1D. pmEVs are administered at varied doses and at defined intervals. For example, some mice are intravenously injected with pmEVs at 10, 15, or 20 ug/mouse. Other mice may receive 25, 50, or 100 mg of pmEVs per mouse. Alternatively, some mice receive between 7.0e+09 to 3.0e+12 pmEV particles per dose. While some mice receive pmEVs through i.v. injection, other mice may receive pmEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, oral gavage, or other means of administration. Some mice may receive pmEVs every day, while others may receive pmEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of pmEVs and bacterial cells. For example, the composition may comprise pmEV particles and whole bacteria in a ratio from 1:1 (pmEVs:bacterial cells) to 1-1×1012:1 (pmEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the pmEV administration. As with the pmEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the pmEVs.


Some groups of mice may be treated with additional treatments and/or an appropriate control (e.g., vehicle or control antibody) at various timepoints and at effective doses.


In addition, some mice are treated with antibiotics prior to treatment. For example, vancomycin (0.5 g/L), ampicillin (1.0 g/L), gentamicin (1.0 g/L) and amphotericin B (0.2 g/L) are added to the drinking water, and antibiotic treatment is halted at the time of treatment or a few days prior to treatment. Some immunized mice are treated without receiving antibiotics.


Blood glucose is monitored biweekly prior to the start of the experiment. At various timepoints thereafter, nonfasting blood glucose is measured. At various timepoints, mice are sacrificed and site the pancreas, lymph nodes, or other tissues may be removed for ex vivo histological, cytokine and/or flow cytometric analysis using methods known in the art. For example, tissues are dissociated using dissociation enzymes according to the manufacturer's instructions. Cells are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-MHCII, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Roryt, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, MHCII, CD206, CD40, CSF1R, PD-L1, Gr-1, F4/80). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified tissue-infiltrating immune cells obtained ex vivo. Finally, immunohistochemistry is carried out on various tissue sections to measure T cells, macrophages, dendritic cells, and checkpoint molecule protein expression. Antibody production may also be assessed by ELISA.


In order to examine the impact and longevity of disease protection, rather than being sacrificed, some mice may be rechallenged with a disease trigger, or assessed for susceptibility to relapse. Mice are analyzed for susceptibility to diabetes onset and severity following rechallenge (or spontaneously-occurring relapse).


Example 14
Labeling Bacterial pmEVs

pmEVs may be labeled in order to track their biodistribution in viva and to quantify and track cellular localization in various preparations and in assays conducted with mammalian cells. For example, pmEVs may be radio-labeled, incubated with dyes, fluorescently labeled, luminescently labeled, or labeled with conjugates containing metals and isotopes of metals.


For example, pmEVs may be incubated with dyes conjugated to functional groups such as NHS-ester, click-chemistry groups, streptavidin or biotin. The labeling reaction may occur at a variety of temperatures for minutes or hours, and with or without agitation or rotation. The reaction may then be stopped by adding a reagent such as bovine serum albumin (BSA), or similar agent, depending on the protocol, and free or unbound dye molecule removed by ultra-centrifugation, filtration, centrifugal filtration, column affinity purification or dialysis. Additional washing steps involving wash buffers and vortexing or agitation may be employed to ensure complete removal of free dyes molecules such as described in Su Chul Jang et al, Small. 11, No. 4, 456-461(2017).


Optionally, pmEVs may be concentrated to 5.0 E12 particle/ml (300 ug) and diluted up to 1.8 mo using 2× concentrated PBS buffer pH 8.2 and pelleted by centrifugation at 165,000×g at 4 C using a benchtop ultracentrifuge. The pellet is resuspended in 300 ul 2× PBS pH 8.2 and an NHS-ester fluorescent dye is added at a final concentration of 0.2 mM from a 10 mM dye stock (dissolved in DMSO). The sample is gently agitated at 24° C. for 1.5 hours, and then incubated overnight at 4° C. Free non-reacted dye is removed by 2 repeated steps of dilution/pelleting as described above, using 1× PBS buffer, and resuspending in 300 ul final volume.


Fluorescently labeled pmEVs are detected in cells or organs, or in in vitro and/or ex vivo samples by confocal microscopy, nanoparticle tracking analysis, flow cytometry, fluorescence activated cell sorting (FACs) or fluorescent imaging system such as the Odyssey CLx LICOR (see e.g., Wiklander et al. 2015. J. Extracellular Vesicles. 4:10.3402/j ev.v4.26316). Additionally, fluorescently labeled pmEVs are detected in whole animals and/or dissected organs and tissues using an instrument such as the IVIS spectrum CT (Perkin Elmer) or Pearl Imager, as in H-I. Choi, et al. Experimental & Molecular Medicine. 49: e330 (2017).


pmEVs may be labeled with conjugates containing metals and isotopes of metals using the protocols described above. Metal-conjugated pmEVs may be administered in vivo to animals. Cells may then be harvested from organs at various time-points, and analyzed ex vivo. Alternatively, cells derived from animals, humans, or immortalized cell lines may be treated with metal-labelled pmEVs in vitro and cells subsequently labelled with metal-conjugated antibodies and phenotyped using a Cytometry by Time of Flight (CyTOF) instrument such as the Helios CyTOF (Fluidigm) or imaged and analyzed using and Imaging Mass Cytometry instrument such as the Hyperion Imaging System (Fluidigm). Additionally, pmEVs may be labelled with a radioisotope to track the pmEVs biodistribution (see, e.g., Miller et al., Nanoscale. 2014 May 7; 6(9):4928-35).


Example 15
Transmission Electron Microscopy to Visualize Bacterial pmEVs

pmEVs are prepared from bacteria batch cultures. Transmission electron microscopy (TEM) may be used to visualize purified bacterial pmEVs (S. Bin Park, et al. PLoS ONE. 6(3):e17629 (2011). pmEVs are mounted onto 300- or 400-mesh-size carbon-coated copper grids (Electron Microscopy Sciences, USA) for 2 minutes and washed with deionized water. pmEVs are negatively stained using 2% (w/v) uranyl acetate for 20 sec-1 min. Copper grids are washed with sterile water and dried. Images are acquired using a transmission electron microscope with 100-120 kV acceleration voltage. Stained pmEVs appear between 20-600 nm in diameter and are electron dense. 10-50 fields on each grid are screened.


Example 16
Profiling pmEV Composition and Content

pmEVs may be characterized by any one of various methods including, but not limited to, NanoSight characterization, SDS-PAGE gel electrophoresis, Western blot, ELISA, liquid chromatography-mass spectrometry and mass spectrometry, dynamic light scattering, lipid levels, total protein, lipid to protein ratios, nucleic acid analysis and/or zeta potential.


NanoSight Characterization of pmEVs


Nanoparticle tracking analysis (NTA) is used to characterize the size distribution of purified bacterial pmEVs. Purified pmEV preparations are run on a NanoSight machine (Malvern Instruments) to assess pmEV size and concentration.


SDS-PAGE Gel Electrophoresis

To identify the protein components of purified pmEVs, samples are run on a gel, for example a Bolt Bis-Tris Plus 4-12% gel (Thermo-Fisher Scientific), using standard techniques. Samples are boiled in 1× SDS sample buffer for 10 minutes, cooled to 4° C., and then centrifuged at 16,000×g for 1 min. Samples are then run on a SDS-PAGE gel and stained using one of several standard techniques (e.g., Silver staining, Coomassie Blue, Gel Code Blue) for visualization of bands.


Western Blot Analysis

To identify and quantify specific protein components of purified pmEVs, pmEV proteins are separated by SDS-PAGE as described above and subjected to Western blot analysis (Cvjetkovic et al., Sci. Rep. 6, 36338 (2016)) and are quantified via ELISA.


pmEV Proteomics and Liquid Chromatography-Mass Spectrometry (LC-MS/MS) and Mass Spectrometry (MS)


Proteins present in pmEVs are identified and quantified by Mass Spectrometry techniques. pmEV proteins may be prepared for LC-MS/MS using standard techniques including protein reduction using dithiotreitol solution (DTT) and protein digestion using enzymes such as LysC and trypsin as described in Erickson et al, 2017 (Molecular Cell, VOLUME 65, ISSUE 2, P361-370, JAN. 19, 2017). Alternatively, peptides are prepared as described by Liu et al. 2010 (JOURNAL OF BACTERIOLOGY, June 2010, p. 2852-2860 Vol. 192, No. 11), Kieselbach and Oscarsson 2017 (Data Brief. 2017 February; 10: 426-431.), Vildhede et al, 2018 (Drug Metabolism and Disposition Feb. 8, 2018). Following digestion, peptide preparations are run directly on liquid chromatography and mass spectrometry devices for protein identification within a single sample. For relative quantitation of proteins between samples, peptide digests from different samples are labeled with isobaric tags using the iTRAQ Reagent-8plex Multiplex Kit (Applied Biosystems, Foster City, Calif.) or TMT 10plex and 11plex Label Reagents (Thermo Fischer Scientific, San Jose, Calif., USA). Each peptide digest is labeled with a different isobaric tag and then the labeled digests are combined into one sample mixture. The combined peptide mixture is analyzed by LC-MS/MS for both identification and quantification. A database search is performed using the LC-MS/MS data to identify the labeled peptides and the corresponding proteins. In the case of isobaric labeling, the fragmentation of the attached tag generates a low molecular mass reporter ion that is used to obtain a relative quantitation of the peptides and proteins present in each pmEV.


Additionally, metabolic content is ascertained using liquid chromatography techniques combined with mass spectrometry. A variety of techniques exist to determine metabolomic content of various samples and are known to one skilled in the art involving solvent extraction, chromatographic separation and a variety of ionization techniques coupled to mass determination (Roberts et al 2012 Targeted Metabolomics. Curr Protoc Mol Biol. 30: 1-24; Dettmer et al 2007, Mass spectrometry-based metabolomics. Mass Spectrom Rev. 26(1):51-78). As a non-limiting example, a LC-MS system includes a 4000 QTRAP triple quadrupole mass spectrometer (AB SCIEX) combined with 1100 Series pump (Agilent) and an HTS PAL autosampler (Leap Technologies). Media samples or other complex metabolic mixtures (˜10 μL) are extracted using nine volumes of 74.9:24.9:0.2 (v/v/v) acetonitrile/methanol/formic acid containing stable isotope-labeled internal standards (valine-d8, Isotec; and phenylalanine-d8, Cambridge Isotope Laboratories). Standards may be adjusted or modified depending on the metabolites of interest. The samples are centrifuged (10 minutes, 9,000 g, 4° C.), and the supernatants (10 μL) are submitted to LCMS by injecting the solution onto the HILIC column (150×2.1 mm, 3 μm particle size). The column is eluted by flowing a 5% mobile phase [10 mM ammonium formate, 0.1% formic acid in water] for 1 minute at a rate of 250 uL/minute followed by a linear gradient over 10 minutes to a solution of 40% mobile phase [acetonitrile with 0.1% formic acid]. The ion spray voltage is set to 4.5 kV and the source temperature is 450° C.


The data are analyzed using commercially available software like Multiquant 1.2 from AB SCIEX for mass spectrum peak integration. Peaks of interest should be manually curated and compared to standards to confirm the identity of the peak. Quantitation with appropriate standards is performed to determine the number of metabolites present in the initial media, after bacterial conditioning and after tumor cell growth. A non-targeted metabolomics approach may also be used using metabolite databases, such as but not limited to the NIST database, for peak identification.


Dynamic Light Scattering (DLS)

DLS measurements, including the distribution of particles of different sizes in different pmEV preparations are taken using instruments such as the DynaPro NanoStar (Wyatt Technology) and the Zetasizer Nano ZS (Malvern Instruments).


Lipid Levels

Lipid levels are quantified using FM4-64 (Life Technologies), by methods similar to those described by A. J. McBroom et al. J Bacteriol 188:5385-5392, and A. Frias, et al. Microb Ecol. 59:476-486 (2010). Samples are incubated with FM4-64 (3.3 μg/mL in PBS for 10 minutes at 37° C. in the dark). After excitation at 515 nm, emission at 635 nm is measured using a Spectramax M5 plate reader (Molecular Devices). Absolute concentrations are determined by comparison of unknown samples to standards (such as palmitoyloleoylphosphatidylglycerol (POPG) vesicles) of known concentrations. Lipidomics can be used to identify the lipids present in the pmEVs.


Total Protein

Protein levels are quantified by standard assays such as the Bradford and BCA assays. The Bradford assays are run using Quick Start Bradford 1× Dye Reagent (Bio-Rad), according to manufacturer's protocols. BCA assays are run using the Pierce BCA Protein Assay Kit (Thermo-Fisher Scientific). Absolute concentrations are determined by comparison to a standard curve generated from BSA of known concentrations. Alternatively, protein concentration can be calculated using the Beer-Lambert equation using the sample absorbance at 280 nm (A280) as measured on a Nanodrop spectrophotometer (Thermo-Fisher Scientific). In addition, proteomics may be used to identify proteins in the sample.


Lipid:Protein Ratios

Lipid:protein ratios are generated by dividing lipid concentrations by protein concentrations. These provide a measure of the purity of vesicles as compared to free protein in each preparation.


Nucleic Acid Analysis

Nucleic acids are extracted from pmEVs and quantified using a Qubit fluorometer. Size distribution is assessed using a BioAnalyzer and the material is sequenced.


Zeta Potential

The zeta potential of different preparations are measured using instruments such as the Zetasizer ZS (Malvern Instruments).


Example 17
In Vitro Screening of pmEVs for Enhanced Activation of Dendritic Cells

In vitro immune responses are thought to simulate mechanisms by which immune responses are induced in vivo, e.g., as in response to a cancer microenvironment. Briefly, PBMCs are isolated from heparinized venous blood from healthy donors by gradient centrifugation using Lymphoprep (Nycomed, Oslo, Norway), or from mouse spleens or bone marrow using the magnetic bead-based Human Blood Dendritic cell isolation kit (Miltenyi Biotech, Cambridge, Mass.). Using anti-human CD14 mAb, the monocytes are purified by Moflo and cultured in cRPMI at a cell density of 5e5 cells/ml in a 96-well plate (Costar Corp) for 7 days at 37° C. For maturation of dendritic cells, the culture is stimulated with 0.2 ng/mL IL-4 and 1000 U/ml GM-CSF at 37° C. for one week. Alternatively, maturation is achieved through incubation with recombinant GM-CSF for a week, or using other methods known in the art. Mouse DCs can be harvested directly from spleens using bead enrichment or differentiated from hematopoietic stem cells. Briefly, bone marrow may be obtained from the femurs of mice. Cells are recovered and red blood cells lysed. Stem cells are cultured in cell culture medium in 20 ng/ml mouse GMCSF for 4 days. Additional medium containing 20 ng/ml mouse GM-CSF is added. On day 6 the medium and non-adherent cells are removed and replaced with fresh cell culture medium containing 20 ng/ml GMCSF. A final addition of cell culture medium with 20 ng/ml GM-CSF is added on day 7. On day 10, non-adherent cells are harvested and seeded into cell culture plates overnight and stimulated as required. Dendritic cells are then treated with various doses of pmEVs with or without antibiotics. For example, 25-75 ug/mL pmEVs for 24 hours with antibiotics. pmEV compositions tested may include pmEVs from a single bacterial species or strain, or a mixture of pmEVs from one or more genus, 1 or more species, or 1 or more strains (e.g., one or more strains within one species). PBS is included as a negative control and LPS, anti-CD40 antibodies, from Bifidobacterium spp. are used as positive controls. Following incubation, DCs are stained with anti CD11b, CD11c, CD103, CD8a, CD40, CD80, CD83, CD86, MHCI and MHCII, and analyzed by flow cytometry. DCs that are significantly increased in CD40, CD80, CD83, and CD86 as compared to negative controls are considered to be activated by the associated bacterial pmEV composition. These experiments are repeated three times at minimum.


To screen for the ability of pmEV-activated epithelial cells to stimulate DCs, the above protocol is followed with the addition of a 24-hour epithelial cell pmEV co-culture prior to incubation with DCs. Epithelial cells are washed after incubation with pmEVs and are then co-cultured with DCs in an absence of pmEVs for 24 hours before being processed as above. Epithelial cell lines may include Int407, HEL293, HT29, T84 and CACO2.


As an additional measure of DC activation, 100 μl of culture supernatant is removed from wells following 24-hour incubation of DCs with pmEVs or pmEV-treated epithelial cells and is analyzed for secreted cytokines, chemokines, and growth factors using the multiplexed Luminex Magpix. Kit (EMD Millipore, Darmstadt, Germany). Briefly, the wells are pre-wet with buffer, and 25 μl of 1× antibody-coated magnetic beads are added and 2×200 μl of wash buffer are performed in every well using the magnet. 50 μl of Incubation buffer, 50 μl of diluent and 50 μl of samples are added and mixed via shaking for 2 hrs at room temperature in the dark. The beads are then washed twice with 200 μl wash buffer. 100 μl of 1× biotinylated detector antibody is added and the suspension is incubated for 1 hour with shaking in the dark. Two, 200 μl washes are then performed with wash buffer. 100 μl of 1× SAV-RPE reagent is added to each well and is incubated for 30 min at RT in the dark. Three 200 μl washes are performed and 125 μl of wash buffer is added with 2-3 min shaking occurs. The wells are then submitted for analysis in the Luminex xMAP system.


Standards allow for careful quantitation of the cytokines including GM-CSF, IFN-g, IFN-a, IFN-B, IL-la, IL-1B, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-13, IL-12 (p40/p70), IL-17A, IL-17F, IL-21, IL-22 IL-23, IL-25, IP-10, KC, MCP-1, MIG, MIP1a, TNFa, and VEGF. These cytokines are assessed in samples of both mouse and human origin. Increases in these cytokines in the bacterial treated samples indicate enhanced production of proteins and cytokines from the host. Other variations on this assay examining specific cell types ability to release cytokines are assessed by acquiring these cells through sorting methods and are recognized by one of ordinary skill in the art. Furthermore, cytokine mRNA is also assessed to address cytokine release in response to an pmEV composition.


This DC stimulation protocol may be repeated using combinations of purified pmEVs and live bacterial strains to maximize immune stimulation potential.


Example 18
In Vitro Screening of pmEVs for Enhanced Activation of CD8+ T Cell Killing when Incubated with Tumor Cells

In vitro methods for screening pmEVs that can activate CD8+ T cell killing of tumor cells are described. Briefly, DCs are isolated from human PBMCs or mouse spleens, using techniques known in the art, and incubated in vitro with single-strain pmEVs, mixtures of pmEVs, and/or appropriate controls. In addition, CD8+ T cells are obtained from human PBMCs or mouse spleens using techniques known in the art, for example the magnetic bead-based Mouse CD8a+ T Cell Isolation Kit and the magnetic bead-based Human CD8+ T Cell Isolation Kit (both from Miltenyi Biotech, Cambridge, Mass.). After incubation of DCs with pmEVs for some time (e.g., for 24-hours), or incubation of DCs with pmEV-stimulated epithelial cells, pmEVs are removed from the cell culture with PBS washes and 100 ul of fresh media with antibiotics is added to each well, and 200,000 T cells are added to each experimental well in the 96-well plate. Anti-CD3 antibody is added at a final concentration of 2 ug/ml. Co-cultures are then allowed to incubate at 37° C. for 96 hours under normal oxygen conditions.


For example, approximately 72 hours into the coculture incubation, tumor cells are plated for use in the assay using techniques known in the art. For example, 50,000 tumor cells/well are plated per well in new 96-well plates. Mouse tumor cell lines used may include B16.F10, SIY+B16.F10, and others. Human tumor cell lines are HLA-matched to donor, and can include PANC-1, UNKPC960/961, UNKC, and HELA cell lines. After completion of the 96-hour co-culture, 100 μl of the CD8+ T cell and DC mixture is transferred to wells containing tumor cells. Plates are incubated for 24 hours at 37° C. under normal oxygen conditions. Staurospaurine may be used as negative control to account for cell death.


Following this incubation, flow cytometry is used to measure tumor cell death and characterize immune cell phenotype. Briefly, tumor cells are stained with viability dye. FACS analysis is used to gate specifically on tumor cells and measure the percentage of dead (killed) tumor cells. Data are also displayed as the absolute number of dead tumor cells per well. Cytotoxic CD8+ T cell phenotype may be characterized by the following methods: a) concentration of supernatant granzyme B, IFNy and TNFa in the culture supernatant as described below, b) CD8+ T cell surface expression of activation markers such as DC69, CD25, CD154, PD-1, gamma/delta TCR, Foxp3, T-bet, granzyme B, c) intracellular cytokine staining of IFNy, granzyme B, TNFa in CD8+ T cells. CD4+ T cell phenotype may also be assessed by intracellular cytokine staining in addition to supernatant cytokine concentration including INFy, TNFa, IL-12, IL-4, IL-5, IL-17, IL-10, chemokines etc.


As an additional measure of CD8+ T cell activation, 100 μl of culture supernatant is removed from wells following the 96-hour incubation of T cells with DCs and is analyzed for secreted cytokines, chemokines, and growth factors using the multiplexed Luminex Magpix. Kit (EMD Millipore, Darmstadt, Germany). Briefly, the wells are pre-wet with buffer, and 25 μl of 1× antibody-coated magnetic beads are added and 2×200 μl of wash buffer are performed in every well using the magnet. 50 μl of Incubation buffer, 50 μl of diluent and 50 μl of samples are added and mixed via shaking for 2 hrs at room temperature in the dark. The beads are then washed twice with 200 μl wash buffer. 100 μl of 1× biotinylated detector antibody is added and the suspension is incubated for 1 hour with shaking in the dark. Two, 200 μl washes are then performed with wash buffer. 100 μl of 1× SAV-RPE reagent is added to each well and is incubated for 30 min at RT in the dark. Three 200 μl washes are performed and 125 μl of wash buffer is added with 2-3 min shaking occurs. The wells are then submitted for analysis in the Luminex xMAP system.


Standards allow for careful quantitation of the cytokines including GM-CSF, IFN-g, IFN-a, IFN-B IL-la, IL-1B, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-13, IL-12 (p40/p70), IL-17, IL-23, IP-10, KC, MCP-1, MIG, MIP1a, TNFa, and VEGF. These cytokines are assessed in samples of both mouse and human origin. Increases in these cytokines in the bacterial treated samples indicate enhanced production of proteins and cytokines from the host. Other variations on this assay examining specific cell types ability to release cytokines are assessed by acquiring these cells through sorting methods and are recognized by one of ordinary skill in the art. Furthermore, cytokine mRNA is also assessed to address cytokine release in response to an pmEV composition. These changes in the cells of the host stimulate an immune response similarly to in vivo response in a cancer microenvironment.


This CD8+ T cell stimulation protocol may be repeated using combinations of purified pmEVs and live bacterial strains to maximize immune stimulation potential.


Example 19
In Vitro Screening of pmEVs for Enhanced Tumor Cell Killing by PBMCs

Various methods may be used to screen pmEVs for the ability to stimulate PBMCs, which in turn activate CD8+ T cells to kill tumor cells. For example, PBMCs are isolated from heparinized venous blood from healthy human donors by ficoll-paque gradient centrifugation for mouse or human blood, or with Lympholyte Cell Separation Media (Cedarlane Labs, Ontario, Canada) from mouse blood. PBMCs are incubated with single-strain pmEVs, mixtures of pmEVs, and appropriate controls. In addition, CD8+ T cells are obtained from human PBMCs or mouse spleens. After the 24-hour incubation of PBMCs with pmEVs, pmEVs are removed from the cells using PBS washes. 100 ul of fresh media with antibiotics is added to each well. An appropriate number of T cells (e.g., 200,000 T cells) are added to each experimental well in the 96-well plate. Anti-CD3 antibody is added at a final concentration of 2 ug/ml. Co-cultures are then allowed to incubate at 37° C. for 96 hours under normal oxygen conditions.


For example, 72 hours into the coculture incubation, 50,000 tumor cells/well are plated per well in new 96-well plates. Mouse tumor cell lines used include B16.F10, SIY+B16.F10, and others. Human tumor cell lines are HLA-matched to donor, and can include PANC-1, UNKPC960/961, UNKC, and HELA cell lines. After completion of the 96-hour co-culture, 100 μl of the CD8+ T cell and PBMC mixture is transferred to wells containing tumor cells. Plates are incubated for 24 hours at 37° C. under normal oxygen conditions. Staurospaurine is used as negative control to account for cell death.


Following this incubation, flow cytometry is used to measure tumor cell death and characterize immune cell phenotype. Briefly, tumor cells are stained with viability dye. FACS analysis is used to gate specifically on tumor cells and measure the percentage of dead (killed) tumor cells. Data are also displayed as the absolute number of dead tumor cells per well. Cytotoxic CD8+ T cell phenotype may be characterized by the following methods: a) concentration of supernatant granzyme B, IFNy and TNFa in the culture supernatant as described below, b) CD8+ T cell surface expression of activation markers such as DC69, CD25, CD154, PD-1, gamma/delta TCR, Foxp3, T-bet, granzyme B, c) intracellular cytokine staining of IFNy, granzyme B, TNFa in CD8+ T cells. CD4+ T cell phenotype may also be assessed by intracellular cytokine staining in addition to supernatant cytokine concentration including INFy, TNFa, IL-12, IL-4, IL-5, IL-17, IL-10, chemokines etc.


As an additional measure of CD8+ T cell activation, 100 μl of culture supernatant is removed from wells following the 96-hour incubation of T cells with DCs and is analyzed for secreted cytokines, chemokines, and growth factors using the multiplexed Luminex Magpix. Kit (EMD Millipore, Darmstadt, Germany). Briefly, the wells are pre-wet with buffer, and 25 μl of 1× antibody-coated magnetic beads are added and 2×200 μl of wash buffer are performed in every well using the magnet. 50 μl of Incubation buffer, 50 μl of diluent and 50 μl of samples are added and mixed via shaking for 2 hrs at room temperature in the dark. The beads are then washed twice with 200 μl wash buffer. 100 μl of 1× biotinylated detector antibody is added and the suspension is incubated for 1 hour with shaking in the dark. Two, 200 μl washes are then performed with wash buffer. 100 μl of 1× SAV-RPE reagent is added to each well and is incubated for 30 min at RT in the dark. Three 200 μl washes are performed and 125 μl of wash buffer is added with 2-3 min shaking occurs. The wells are then submitted for analysis in the Luminex xMAP system.


Standards allow for careful quantitation of the cytokines including GM-CSF, IFN-g, IFN-a, IFN-B IL-la, IL-1B, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-13, IL-12 (p40/p70), IL-17, IL-23, IP-10, KC, MCP-1, MIG, MIP1a, TNFa, and VEGF. These cytokines are assessed in samples of both mouse and human origin. Increases in these cytokines in the bacterial treated samples indicate enhanced production of proteins and cytokines from the host. Other variations on this assay examining specific cell types ability to release cytokines are assessed by acquiring these cells through sorting methods and are recognized by one of ordinary skill in the art. Furthermore, cytokine mRNA is also assessed to address cytokine release in response to an pmEV composition. These changes in the cells of the host stimulate an immune response similarly to in vivo response in a cancer microenvironment.


This PBMC stimulation protocol may be repeated using combinations of purified pmEVs with or without combinations of live, dead, or inactivated/weakened bacterial strains to maximize immune stimulation potential.


Example 20
In Vitro Detection of pmEVs in Antigen-Presenting Cells

Dendritic cells in the lamina propria constantly sample live bacteria, dead bacteria, and microbial products in the gut lumen by extending their dendrites across the gut epithelium, which is one way that pmEVs produced by bacteria in the intestinal lumen may directly stimulate dendritic cells. The following methods represent a way to assess the differential uptake of pmEVs by antigen-presenting cells. Optionally, these methods may be applied to assess immunomodulatory behavior of pmEVs administered to a patient.


Dendritic cells (DCs) are isolated from human or mouse bone marrow, blood, or spleens according to standard methods or kit protocols (e.g., Inaba K, Swiggard W J, Steinman R M, Romani N, Schuler G, 2001. Isolation of dendritic cells. Current Protocols in Immunology. Chapter 3:Unit3.7).


To evaluate pmEV entrance into and/or presence in DCs, 250,000 DCs are seeded on a round cover slip in complete RPMI-1640 medium and are then incubated with pmEVs from single bacterial strains or combinations pmEVs at various ratios. Purified pmEVs may be labeled with fluorochromes or fluorescent proteins. After incubation for various timepoints (e.g., 1 hour, 2 hours), the cells are washed twice with ice-cold PBS and detached from the plate using trypsin. Cells are either allowed to remain intact or are lysed. Samples are then processed for flow cytometry. Total internalized pmEVs are quantified from lysed samples, and percentage of cells that uptake pmEVs is measured by counting fluorescent cells. The methods described above may also be performed in substantially the same manner using macrophages or epithelial cell lines (obtained from the ATCC) in place of DCs.


Example 21
In Vitro Screening of pmEVs with an Enhanced Ability to Activate NK Cell Killing when Incubated with Target Cells

To demonstrate the ability of the selected pmEV compositions to elicit potent NK cell cytotoxicity to tumor cells, the following in vitro assay is used. Briefly, mononuclear cells from heparinized blood are obtained from healthy human donors. Optionally, an expansion step to increase the numbers of NK cells is performed as previously described (e.g., see Somanschi et al., J Vis Exp. 2011; (48):2540). The cells may be adjusted to a concentration of cells/ml in RPMI-1640 medium containing 5% human serum. The PMNC cells are then labeled with appropriate antibodies and NK cells are isolated through FACS as CD3−/CD56+ cells and are ready for the subsequent cytotoxicity assay. Alternatively, NK cells are isolated using the autoMACs instrument and NK cell isolation kit following manufacturer's instructions (Miltenyl Biotec).


NK cells are counted and plated in a 96 well format with 20,000 or more cells per well, and incubated with single-strain pmEVs, with or without addition of antigen presenting cells (e.g., monocytes derived from the same donor), pmEVs from mixtures of bacterial strains, and appropriate controls. After 5-24 hours incubation of NK cells with pmEVs, pmEVs are removed from cells with PBS washes, NK cells are resuspended in 10 mL fresh media with antibiotics and are added to 96-well plates containing 20,000 target tumor cells/well. Mouse tumor cell lines used include B16.F10, SIY+B16.F10, and others. Human tumor cell lines are HLA-matched to donor, and can include PANC-1, UNKPC960/961, UNKC, and HELA cell lines. Plates are incubated for 2-24 hours at 37° C. under normal oxygen conditions. Staurospaurine is used as negative control to account for cell death.


Following this incubation, flow cytometry is used to measure tumor cell death using methods known in the art. Briefly, tumor cells are stained with viability dye. FACS analysis is used to gate specifically on tumor cells and measure the percentage of dead (killed) tumor cells. Data are also displayed as the absolute number of dead tumor cells per well.


This NK cell stimulation protocol may be repeated using combinations of purified pmEVs and live bacterial strains to maximize immune stimulation potential.


Example 22
Using In Vitro Immune Activation Assays to Predict In Vivo Cancer Immunotherapy Efficacy of pmEV Compositions

In vitro immune activation assays identify pmEVs that are able to stimulate dendritic cells, which in turn activate CD8+ T cell killing. Therefore, the in vitro assays described above are used as a predictive screen of a large number of candidate pmEVs for potential immunotherapy activity. pmEVs that display enhanced stimulation of dendritic cells, enhanced stimulation of CD8+ T cell killing, enhanced stimulation of PBMC killing, and/or enhanced stimulation of NK cell killing, are preferentially chosen for in vivo cancer immunotherapy efficacy studies.


Example 23
Determining the Biodistribution of pmEVs when Delivered Orally to Mice

Wild-type mice (e.g., C57BL/6 or BALB/c) are orally inoculated with the pmEV composition of interest to determine the in vivo biodistibution profile of purified pmEVs. pmEVs are labeled to aide in downstream analyses. Alternatively, tumor-bearing mice or mice with some immune disorder (e.g., systemic lupus erythematosus, experimental autoimmune encephalomyelitis, NASH) may be studied for in vivo distribution of pmEVs over a given time-course.


Mice can receive a single dose of the pmEV (e.g., 25-100 μg) or several doses over a defined time course (25-100 μg). Alternatively, pmEVs dosages may be administered based on particle count (e.g., 7e+08 to 6e+11 particles). Mice are housed under specific pathogen-free conditions following approved protocols. Alternatively, mice may be bred and maintained under sterile, germ-free conditions. Blood, stool, and other tissue samples can be taken at appropriate time points.


The mice are humanely sacrificed at various time points (i.e., hours to days) post administration of the pmEV compositions, and a full necropsy under sterile conditions is performed. Following standard protocols, lymph nodes, adrenal glands, liver, colon, small intestine, cecum, stomach, spleen, kidneys, bladder, pancreas, heart, skin, lungs, brain, and other tissue of interest are harvested and are used directly or snap frozen for further testing. The tissue samples are dissected and homogenized to prepare single-cell suspensions following standard protocols known to one skilled in the art. The number of pmEVs present in the sample is then quantified through flow cytometry. Quantification may also proceed with use of fluorescence microscopy after appropriate processing of whole mouse tissue (Vankelecom H., Fixation and paraffin-embedding of mouse tissues for GFP visualization, Cold Spring Harb. Proloc., 2009). Alternatively, the animals may be analyzed using live-imaging according to the pmEV labeling technique.


Biodistribution may be performed in mouse models of cancer such as but not limited to CT-26 and B16 (see, e.g., Kim et al., Nature Communications vol. 8, no. 626 (2017)) or autoimmunity such as but not limited to EAE and DTH (see, e.g., Turjeman et al., PLoS One 10(7): e0130442 (20105).


Example 24
Purification and Preparation of Secreted Microbial Extracellular Vesicles (smEVs) from Bacteria
Purification

Secreted microbial extracellular vesicles (smEVs) are purified and prepared from bacterial cultures (e.g., bacteria from Table 1, Table 2, and/or Table 3) using methods known to those skilled in the art (S. Bin Park, et al. PLoS ONE. 6(3):e17629 (2011)).


For example, bacterial cultures are centrifuged at 10,000-15,500×g for 10-40 min at 4° C. or room temperature to pellet bacteria. Culture supernatants are then filtered to include material≤0.22 μm (for example, via a 0.22 μm or 0.45 μm filter) and to exclude intact bacterial cells. Filtered supernatants are concentrated using methods that may include, but are not limited to, ammonium sulfate precipitation, ultracentrifugation, or filtration. Briefly, for ammonium sulfate precipitation, 1.5-3 M ammonium sulfate is added to filtered supernatant slowly, while stirring at 4° C. Precipitations are incubated at 4° C. for 8-48 hours and then centrifuged at 11,000×g for 20-40 min at 4° C. The pellets contain smEVs and other debris. Briefly, using ultracentrifugation, filtered supernatants are centrifuged at 100,000-200,000×g for 1-16 hours at 4° C. The pellet of this centrifugation contains smEVs and other debris. Briefly, using a filtration technique, using an Amicon Ultra spin filter or by tangential flow filtration, supernatants are filtered so as to retain species of molecular weight>50, 100, 300, or 500 kDa.


Alternatively, smEVs are obtained from bacterial cultures continuously during growth, or at selected time points during growth, by connecting a bioreactor to an alternating tangential flow (ATF) system (e.g., XCell ATF from Repligen) according to manufacturer's instructions. The ATF system retains intact cells (>0.22 um) in the bioreactor, and allows smaller components (e.g., smEVs, free proteins) to pass through a filter for collection. For example, the system may be configured so that the <0.22 um filtrate is then passed through a second filter of 100 kDa, allowing species such as smEVs between 0.22 um and 100 kDa to be collected, and species smaller than 100 kDa to be pumped back into the bioreactor. Alternatively, the system may be configured to allow for medium in the bioreactor to be replenished and/or modified during growth of the culture. smEVs collected by this method may be further purified and/or concentrated by ultracentrifugation or filtration as described above for filtered supernatants.


smEVs obtained by methods described above may be further purified by gradient ultracentrifugation, using methods that may include, but are not limited to, use of a sucrose gradient or Optiprep gradient. Briefly, using a sucrose gradient method, if ammonium sulfate precipitation or ultracentrifugation were used to concentrate the filtered supernatants, pellets are resuspended in 60% sucrose, 30 mM Tris, pH 8.0. If filtration was used to concentrate the filtered supernatant, the concentrate is buffer exchanged into 60% sucrose, 30 mM Tris, pH 8.0, using an Amicon Ultra column. Samples are applied to a 35-60% discontinuous sucrose gradient and centrifuged at 200,000×g for 3-24 hours at 4° C. Briefly, using an Optiprep gradient method, if ammonium sulfate precipitation or ultracentrifugation were used to concentrate the filtered supernatants, pellets are resuspended in 45% Optiprep in PBS. If filtration was used to concentrate the filtered supernatant, the concentrate is diluted using 60% Optiprep to a final concentration of 45% Optiprep. Samples are applied to a 0-45% discontinuous sucrose gradient and centrifuged at 200,000×g for 3-24 hours at 4° C. Alternatively, high resolution density gradient fractionation could be used to separate smEVs based on density.


Preparation

To confirm sterility and isolation of the smEV preparations, smEVs are serially diluted onto agar medium used for routine culture of the bacteria being tested and incubated using routine conditions. Non-sterile preparations are passed through a 0.22 um filter to exclude intact cells. To further increase purity, isolated smEVs may be DNase or proteinase K treated.


Alternatively, for preparation of smEVs used for in vivo injections, purified smEVs are processed as described previously (G. Norheim, et al. PLoS ONE. 10(9): e0134353 (2015)). Briefly, after sucrose gradient centrifugation, bands containing smEVs are resuspended to a final concentration of 50 μg/mL in a solution containing 3% sucrose or other solution suitable for in vivo injection known to one skilled in the art. This solution may also contain adjuvant, for example aluminum hydroxide at a concentration of 0-0.5% (w/v).


To make samples compatible with further testing (e.g., to remove sucrose prior to TEM imaging or in vitro assays), samples are buffer exchanged into PBS or 30 mM Tris, pH 8.0 using filtration (e.g., Amicon Ultra columns), dialysis, or ultracentrifugation (following 15-fold or greater dilution in PBS, 200,000×g, 1-3 hours, 4° C.) and resuspension in PBS.


For all of these studies, smEVs may be heated, irradiated, and/or lyophilized prior to administration (as described in Example 49).


Example 25
Manipulating Bacteria Through Stress to Produce Various Amounts of smEVs and/or to Vary Content of smEVs

Stress, and in particular envelope stress, has been shown to increase production of smEVs by some bacterial strains (I. MacDonald, M. Kuehn. J Bacteriol 195(13): doi: 10/1128/JB.02267-12). In order to vary production of smEVs by bacteria, bacteria are stressed using various methods.


Bacteria may be subjected to single stressors or stressors in combination. The effects of different stressors on different bacteria is determined empirically by varying the stress condition and determining the IC50 value (the conditions required to inhibit cell growth by 50%). smEV purification, quantification, and characterization occurs. smEV production is quantified (1) in complex samples of bacteria and smEVs by nanoparticle tracking analysis (NTA) or transmission electron microscopy (TEM); or (2) following smEV purification by NTA, lipid quantification, or protein quantification. smEV content is assessed following purification by methods described above.


Antibiotic Stress


Bacteria are cultivated under standard growth conditions with the addition of sublethal concentrations of antibiotics. This may include 0.1-1 μg/mL chloramphenicol, or 0.1-0.3 μg/mL gentamicin, or similar concentrations of other antibiotics (e.g., ampicillin, polymyxin B). Host antimicrobial products such as lysozyme, defensins, and Reg proteins may be used in place of antibiotics. Bacterially-produced antimicrobial peptides, including bacteriocins and microcins may also be used.


Temperature Stress


Bacteria are cultivated under standard growth conditions, but at higher or lower temperatures than are typical for their growth. Alternatively, bacteria are grown under standard conditions, and then subjected to cold shock or heat shock by incubation for a short period of time at low or high temperatures respectively. For example, bacteria grown at 37° C. are incubated for 1 hour at 4° C.-18° C. for cold shock or 42° C.-50° C. for heat shock.


Starvation and Nutrient Limitation


To induce nutritional stress, bacteria are cultivated under conditions where one or more nutrients are limited. Bacteria may be subjected to nutritional stress throughout growth or shifted from a rich medium to a poor medium. Some examples of media components that are limited are carbon, nitrogen, iron, and sulfur. An example medium is M9 minimal medium (Sigma-Aldrich), which contains low glucose as the sole carbon source. Particularly for Prevotella spp., iron availability is varied by altering the concentration of hemin in media and/or by varying the type of porphyrin or other iron carrier present in the media, as cells grown in low hemin conditions were found to produce greater numbers of smEVs (S. Stubbs et al. Letters in Applied Microbiology. 29:31-36 (1999). Media components are also manipulated by the addition of chelators such as EDTA and deferoxamine.


Saturation


Bacteria are grown to saturation and incubated past the saturation point for various periods of time. Alternatively, conditioned media is used to mimic saturating environments during exponential growth. Conditioned media is prepared by removing intact cells from saturated cultures by centrifugation and filtration, and conditioned media may be further treated to concentrate or remove specific components.


Salt Stress


Bacteria are cultivated in or exposed for brief periods to medium containing NaCl, bile salts, or other salts.


UV Stress


UV stress is achieved by cultivating bacteria under a UV lamp or by exposing bacteria to UV using an instrument such as a Stratalinker (Agilent). UV may be administered throughout the entire cultivation period, in short bursts, or for a single defined period following growth.


Reactive Oxygen Stress


Bacteria are cultivated in the presence of sublethal concentrations of hydrogen peroxide (250-1,000 μM) to induce stress in the form of reactive oxygen species. Anaerobic bacteria are cultivated in or exposed to concentrations of oxygen that are toxic to them.


Detergent Stress


Bacteria are cultivated in or exposed to detergent, such as sodium dodecyl sulfate (SDS) or deoxycholate.


pH Stress


Bacteria are cultivated in or exposed for limited times to media of different pH.


Example 26
Preparation of smEV-Free Bacteria

Bacterial samples containing minimal amounts of smEVs are prepared. smEV production is quantified (1) in complex samples of bacteria and extracellular components by NTA or TEM; or (2) following smEV purification from bacterial samples, by NTA, lipid quantification, or protein quantification.


a. Centrifugation and washing: Bacterial cultures are centrifuged at 11,000×g to separate intact cells from supernatant (including free proteins and vesicles). The pellet is washed with buffer, such as PBS, and stored in a stable way (e.g., mixed with glycerol, flash frozen, and stored at −80° C.).


b. ATF: Bacteria and smEVs are separated by connection of a bioreactor to an ATF system. smEV-free bacteria are retained within the bioreactor, and may be further separated from residual smEVs by centrifugation and washing, as described above.


c. Bacteria are grown under conditions that are found to limit production of smEVs. Conditions that may be varied.


Example 27
A Colorectal Carcinoma Model

To study the efficacy of smEVs in a tumor model, one of many cancer cell lines may be used according to rodent tumor models known in the art. smEVs may be generated from any one of several bacterial species, for instance Veillonella parvula or V. atypica.


For example, female 6-8 week old Balb/c mice are obtained from Taconic (Germantown, N.Y.) or other vendor. 100,000 CT-26 colorectal tumor cells (ATCC CRL-2638) are resuspended in sterile PBS and inoculated in the presence of 50% Matrigel. CT-26 tumor cells are subcutaneously injected into one hind flank of each mouse. When tumor volumes reach an average of 100 mm3 (approximately 10-12 days following tumor cell inoculation), animals are distributed into various treatment groups (e.g., Vehicle; Veillonella smEVs, Bifidobacteria smEVs, with or without anti-PD-1 antibody). Antibodies are administered intraperitoneally (i.p.) at 200 μg/mouse (100 μl final volume) every four days, starting on day 1, for a total of 3 times (Q4D×3), and smEVs are administered orally or intravenously and at varied doses and varied times. For example, smEVs (5 μg) are intravenously (i.v.) injected every third day, starting on day 1 for a total of 4 times (Q3D×4) and mice are assessed for tumor growth. Some mice may be intravenously injected with smEVs at 10, 15, or 20 ug smEVs/mouse. Other mice may receive 25, 50, or 100 mg of smEVs per mouse. Alternatively, some mice receive between 7.0e+09 to 3.0e+12 smEV particles per dose.


Alternatively, when tumor volumes reach an average of 100 mm3 (approximately 10-12 days following tumor cell inoculation), animals are distributed into the following groups: 1) Vehicle; 2) Neisseria Meningitidis smEVs isolated from the Bexsero® vaccine; and 3) anti-PD-1 antibody. Antibodies are administered intraperitoneally (i.p.) at 200 ug/mouse (100 ul final volume) every four days, starting on day 1, and Neisseria Meningitidis smEVs are administered intraperitoneally (i.p.) daily, starting on day 1 until the conclusion of the study.


When tumor volumes reached an average of 100 mm3 (approximately 10-12 days following tumor cell inoculation), animals were distributed into the following groups: 1) Vehicle; 2) anti-PD-1 antibody; and 3) smEV V. parvula (7.0 e+10 particle count). Antibodies were administered intraperitoneally (i.p.) at 200 μg/mouse (100 μl final volume) every four days, starting on day 1, and smEVs were intravenously (i.v.) injected daily, starting on day 1 until the conclusion of the study and tumors measured for growth. At day 11, the smEV V. parvula group exhibited tumor growth inhibition that was significantly better than that seen in the anti-PD-1 group (FIG. 16). Welch's test is performed for treatment vs. vehicle. In a study looking at dose-response of smEVs purified from V. parvula and V. atypica, the highest dose of smEVs demonstrated the greatest efficacy (FIGS. 17 and 18), although in a study with smEVs from V. tobetsuensis, higher doses do not necessarily correspond to greater efficacy (FIG. 19).


Example 28
Administering smEV Compositions to Treat Mouse Tumor Models

As described in Example 27 a mouse model of cancer is generated by subcutaneously injecting a tumor cell line or patient-derived tumor sample and allowing it to engraft into healthy mice. The methods provided herein may be performed using one of several different tumor cell lines including, but not limited to: B16-F10 or B16-F10-SIY cells as an orthotopic model of melanoma, Panc02 cells as an orthotopic model of pancreatic cancer (Maletzki et al., 2008, Gut 57:483-491), LLC1 cells as an orthotopic model of lung cancer, and RM-1 as an orthotopic model of prostate cancer. As an example, but without limitation, methods for studying the efficacy of smEVs in the B16-F10 model are provided in depth herein.


A syngeneic mouse model of spontaneous melanoma with a very high metastatic frequency is used to test the ability of bacteria to reduce tumor growth and the spread of metastases. The smEVs chosen for this assay are compositions that may display enhanced activation of immune cell subsets and stimulate enhanced killing of tumor cells in vitro. The mouse melanoma cell line B16-F10 is obtained from ATCC. The cells are cultured in vitro as a monolayer in RPMI medium, supplemented with 10% heat-inactivated fetal bovine serum and 1% penicillin/streptomycin at 37□ in an atmosphere of 5% CO2 in air. The exponentially growing tumor cells are harvested by trypsinization, washed three times with cold 1× PBS, and a suspension of 5E6 cells/ml is prepared for administration. Female C57BL/6 mice are used for this experiment. The mice are 6-8 weeks old and weigh approximately 16-20 g. For tumor development, each mouse is injected SC into the flank with 100 μl of the B16-F10 cell suspension. The mice are anesthetized by ketamine and xylazine prior to the cell transplantation. The animals used in the experiment may be started on an antibiotic treatment via instillation of a cocktail of kanamycin (0.4 mg/ml), gentamicin, (0.035 mg/ml), colistin (850 U/ml), metronidazole (0.215 mg/ml) and vancomycin (0.045 mg/ml) in the drinking water from day 2 to 5 and an intraperitoneal injection of clindamycin (10 mg/kg) on day 7 after tumor injection.


The size of the primary flank tumor is measured with a caliper every 2-3 days and the tumor volume is calculated using the following formula: tumor volume=the tumor width×tumor length×0.5. After the primary tumor reaches approximately 100 mm3, the animals are sorted into several groups based on their body weight. The mice are then randomly taken from each group and assigned to a treatment group. smEV compositions are prepared as previously described. The mice are orally inoculated by gavage with approximately 7.0e+09 to 3.0e+12 smEV particles. Alternatively, smEVs are administered intravenously. Mice receive smEVs daily, weekly, bi-weekly, monthly, bi-monthly, or on any other dosing schedule throughout the treatment period. Mice may be IV injected with smEVs in the tail vein, or directly injected into the tumor. Mice can be injected with smEVs, with or without live bacteria, and/or smEVs with or without inactivated/weakened or killed bacteria. Mice can be injected or orally gavaged weekly or once a month. Mice may receive combinations of purified smEVs and live bacteria to maximize tumor-killing potential. All mice are housed under specific pathogen-free conditions following approved protocols. Tumor size, mouse weight, and body temperature are monitored every 3-4 days and the mice are humanely sacrificed 6 weeks after the B16-F10 mouse melanoma cell injection or when the volume of the primary tumor reaches 1000 mm3. Blood draws are taken weekly and a full necropsy under sterile conditions is performed at the termination of the protocol.


Cancer cells can be easily visualized in the mouse B16-F10 melanoma model due to their melanin production. Following standard protocols, tissue samples from lymph nodes and organs from the neck and chest region are collected and the presence of micro- and macro-metastases is analyzed using the following classification rule. An organ is classified as positive for metastasis if at least two micro-metastatic and one macro-metastatic lesion per lymph node or organ are found. Micro-metastases are detected by staining the paraffin-embedded lymphoid tissue sections with hematoxylin-eosin following standard protocols known to one skilled in the art. The total number of metastases is correlated to the volume of the primary tumor and it is found that the tumor volume correlates significantly with tumor growth time and the number of macro- and micro-metastases in lymph nodes and visceral organs and also with the sum of all observed metastases. Twenty-five different metastatic sites are identified as previously described (Bobek V., et al., Syngeneic lymph-node-targeting model of green fluorescent protein-expressing Lewis lung carcinoma, Clin. Exp. Metastasis, 2004; 21(8):705-8).


The tumor tissue samples are further analyzed for tumor infiltrating lymphocytes. The CD8+ cytotoxic T cells can be isolated by FACS and can then be further analyzed using customized p/MHC class I microarrays to reveal their antigen specificity (see e.g., Deviren G., et al., Detection of antigen-specific T cells on p/MEIC microarrays, J. Mol. Recognit., 2007 January-February; 20(1):32-8). CD4+ T cells can be analyzed using customized p/MHC class II microarrays.


At various timepoints, mice are sacrificed and tumors, lymph nodes, or other tissues may be removed for ex vivo flow cytometric analysis using methods known in the art. For example, tumors are dissociated using a Miltenyi tumor dissociation enzyme cocktail according to the manufacturer's instructions. Tumor weights are recorded and tumors are chopped then placed in 15 ml tubes containing the enzyme cocktail and placed on ice. Samples are then placed on a gentle shaker at 37° C. for 45 minutes and quenched with up to 15 ml complete RPMI. Each cell suspension is strained through a 70 μm filter into a 50 ml falcon tube and centrifuged at 1000 rpm for 10 minutes. Cells are resuspended in FACS buffer and washed to remove remaining debris. If necessary, samples are strained again through a second 70 μm filter into a new tube. Cells are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-MHCII, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Ror□t, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, MHCII, CD206, CD40, CSF1R, PD-L1, Gr-1). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, RANTES, and MCP-1. Cytokine analysis may be carried out immune cells obtained from lymph nodes or other tissue, and/or on purified CD45+ tumor-infiltrated immune cells obtained ex vivo. Finally, immunohistochemistry is carried out on tumor sections to measure T cells, macrophages, dendritic cells, and checkpoint molecule protein expression.


The same experiment is also performed with a mouse model of multiple pulmonary melanoma metastases. The mouse melanoma cell line B16-BL6 is obtained from ATCC and the cells are cultured in vitro as described above. Female C57BL/6 mice are used for this experiment. The mice are 6-8 weeks old and weigh approximately 16-20 g. For tumor development, each mouse is injected into the tail vein with 100 μl of a 2E6 cells/ml suspension of B16-BL6 cells. The tumor cells that engraft upon IV injection end up in the lungs.


The mice are humanely killed after 9 days. The lungs are weighed and analyzed for the presence of pulmonary nodules on the lung surface. The extracted lungs are bleached with Fekete's solution, which does not bleach the tumor nodules because of the melanin in the B16 cells though a small fraction of the nodules is amelanotic (i.e. white). The number of tumor nodules is carefully counted to determine the tumor burden in the mice. Typically, 200-250 pulmonary nodules are found on the lungs of the control group mice (i.e. PBS gavage).


The percentage tumor burden is calculated for the various treatment groups. Percentage tumor burden is defined as the mean number of pulmonary nodules on the lung surfaces of mice that belong to a treatment group divided by the mean number of pulmonary nodules on the lung surfaces of the control group mice.


The tumor biopsies and blood samples are submitted for metabolic analysis via LCMS techniques or other methods known in the art. Differential levels of amino acids, sugars, lactate, among other metabolites, between test groups demonstrate the ability of the microbial composition to disrupt the tumor metabolic state.


RNA Seq to Determine Mechanism of Action

Dendritic cells are purified from tumors, Peyers patches, and mesenteric lymph nodes. RNAseq analysis is carried out and analyzed according to standard techniques known to one skilled in the art (Z. Hou. Scientific Reports. 5(9570):doi:10.1038/srep09570 (2015)). In the analysis, specific attention is placed on innate inflammatory pathway genes including TLRs, CLRs, NLRs, and STING, cytokines, chemokines, antigen processing and presentation pathways, cross presentation, and T cell co-stimulation.


Rather than being sacrificed, some mice may be rechallenged with tumor cell injection into the contralateral flank (or other area) to determine the impact of the immune system's memory response on tumor growth.


Example 29
Administering smEVs to Treat Mouse Tumor Models in Combination with PD-1 or PD-L1 Inhibition

To determine the efficacy of smEVs in tumor mouse models in combination with PD-1 or PD-L1 inhibition, a mouse tumor model may be used as described above.


smEVs are tested for their efficacy in the mouse tumor model, either alone or in combination with whole bacterial cells and with or without anti-PD-1 or anti-PD-L1. smEVs, bacterial cells, and/or anti-PD-1 or anti-PD-L1 are administered at varied time points and at varied doses. For example, on day 10 after tumor injection, or after the tumor volume reaches 100 mm3, the mice are treated with smEVs alone or in combination with anti-PD-1 or anti-PD-L1.


Mice may be administered smEVs orally, intravenously, or intratumorally. For example, some mice are intravenously injected with anywhere between 7.0e+09 to 3.0e+12 smEV particles. While some mice receive smEVs through i.v. injection, other mice may receive smEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, oral gavage, or other means of administration. Some mice may receive smEVs every day (e.g., starting on day 1), while others may receive smEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of smEVs and bacterial cells. For example, the composition may comprise smEV particles and whole bacteria in a ratio from 1:1 (smEVs:bacterial cells) to 1-1×1012:1 (smEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the smEV administration. As with the smEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the smEVs.


Some groups of mice are also injected with effective doses of checkpoint inhibitor. For example, mice receive 100 μg anti-PD-L1 mAB (clone 10f.9g2, BioXCell) or another anti-PD-1 or anti-PD-L1 mAB in 100 μl PBS, and some mice receive vehicle and/or other appropriate control (e.g., control antibody). Mice are injected with mABs 3, 6, and 9 days after the initial injection. To assess whether checkpoint inhibition and smEV immunotherapy have an additive anti-tumor effect, control mice receiving anti-PD-1 or anti-PD-L1 mABs are included to the standard control panel. Primary (tumor size) and secondary (tumor infiltrating lymphocytes and cytokine analysis) endpoints are assessed, and some groups of mice may be rechallenged with a subsequent tumor cell inoculation to assess the effect of treatment on memory response.


Example 30
smEVs in a Mouse Model of Delayed-Type Hypersensitivity (DTH)

Delayed-type hypersensitivity (DTH) is an animal model of atopic dermatitis (or allergic contact dermatitis), as reviewed by Petersen et al. (In vivo pharmacological disease models for psoriasis and atopic dermatitis in drug discovery. Basic & Clinical Pharm & Toxicology. 2006. 99(2): 104-115; see also Irving C. Allen (ed.) Mouse Models of Innate Immunity: Methods and Protocols, Methods in Molecular Biology, 2013. vol. 1031, DOI 10.1007/978-1-62703-481-4_13). Several variations of the DTH model have been used and are well known in the art (Irving C. Allen (ed.). Mouse Models of Innate Immunity: Methods and Protocols, Methods in Molecular Biology. Vol. 1031, DOI 10.1007/978-1-62703-481-4_13, Springer Science+Business Media, LLC 2013).


DTH can be induced in a variety of mouse and rat strains using various haptens or antigens, for example an antigen emulsified with an adjuvant. DTH is characterized by sensitization as well as an antigen-specific T cell-mediated reaction that results in erythema, edema, and cellular infiltration—especially infiltration of antigen presenting cells (APCs), eosinophils, activated CD4+ T cells, and cytokine-expressing Th2 cells.


Generally, mice are primed with an antigen administered in the context of an adjuvant (e.g., Complete Freund's Adjuvant) in order to induce a secondary (or memory) immune response measured by swelling and antigen-specific antibody titer.


Dexamethasone, a corticosteroid, is a known anti-inflammatory that ameliorates DTH reactions in mice and serves as a positive control for suppressing inflammation in this model (Taube and Carlsten, Action of dexamethasone in the suppression of delayed-type hypersensitivity in reconstituted SCID mice. Inflamm Res. 2000. 49(10): 548-52). For the positive control group, a stock solution of 17 mg/mL of Dexamethasone is prepared on Day 0 by diluting 6.8 mg Dexamethasone in 400 μL 96% ethanol. For each day of dosing, a working solution is prepared by diluting the stock solution 100× in sterile PBS to obtain a final concentration of 0.17 mg/mL in a septum vial for intraperitoneal dosing. Dexamethasone-treated mice receive 100 μL Dexamethasone i.p. (5 mL/kg of a 0.17 mg/mL solution). Frozen sucrose serves as the negative control (vehicle). In the study described below, vehicle, Dexamethasone (positive control) and smEVs were dosed daily.


smEVs are tested for their efficacy in the mouse model of DTH, either alone or in combination with whole bacterial cells, with or without the addition of other anti-inflammatory treatments. For example, 6-8 week old C57Bl/6 mice are obtained from Taconic (Germantown, N.Y.), or other vendor. Groups of mice are administered four subcutaneous (s.c.) injections at four sites on the back (upper and lower) of antigen (e.g., Ovalbumin (OVA) or Keyhole Limpet Hemocyanin (KLH)) in an effective dose (e.g., 50 ul total volume per site). For a DTH response, animals are injected intradermally (i.d.) in the ears under ketamine/xylazine anesthesia (approximately 50 mg/kg and 5 mg/kg, respectively). Some mice serve as control animals. Some groups of mice are challenged with 10 ul per ear (vehicle control (0.01% DMSO in saline) in the left ear and antigen (21.2 ug (12 nmol) in the right ear) on day 8. To measure ear inflammation, the ear thickness of manually restrained animals is measured using a Mitutoyo micrometer. The ear thickness is measured before intradermal challenge as the baseline level for each individual animal. Subsequently, the ear thickness is measured two times after intradermal challenge, at approximately 24 hours and 48 hours (i.e., days 9 and 10).


Treatment with smEVs is initiated at some point, either around the time of priming or around the time of DTH challenge. For example, smEVs may be administered at the same time as the subcutaneous injections (day 0), or they may be administered prior to, or upon, intradermal injection. smEVs are administered at varied doses and at defined intervals. For example, some mice are intravenously injected with smEVs at 10, 15, or 20 ug/mouse. Other mice may receive 25, 50, or 100 mg of smEVs per mouse. Other mice may receive 25, 50, or 100 mg of smEVs per mouse. Alternatively, some mice receive between 7.0e+09 to 3.0e+12 smEV particles per dose.


While some mice receive smEVs through i.v. injection, other mice may receive smEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, oral gavage, topical administration, intradermal (i.d.) injection, or other means of administration. Some mice may receive smEVs every day (e.g., starting on day 0), while others may receive smEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of smEVs and bacterial cells. For example, the composition may comprise smEV particles and whole bacteria in a ratio from 1:1 (smEVs:bacterial cells) to 1-1×1012:1 (smEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the smEV administration. As with the smEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the smEVs.


For the smEVs, total protein is measured using Bio-rad assays (Cat #5000205) performed per manufacturer's instructions.


An emulsion of Keyhole Limpet Hemocyanin (KLH) and Complete Freund's Adjuvant (CFA) was prepared freshly on the day of immunization (day 0). To this end, 8 mg of KLH powder is weighed and is thoroughly re-suspended in 16 mL saline. An emulsion was prepared by mixing the KLH/saline with an equal volume of CFA solution (e.g., 10 mL KLH/saline+10 mL CFA solution) using syringes and a luer lock connector. KLH and CFA were mixed vigorously for several minutes to form a white-colored emulsion to obtain maximum stability. A drop test was performed to check if a homogenous emulsion was obtained.


On day 0, C57Bl/6J female mice, approximately 7 weeks old, were primed with KLH antigen in CFA by subcutaneous immunization (4 sites, 50 μL per site). P. histicola smEVs and lyophilized P. histicola smEVs were tested by oral gavage at low (6.0E+07), medium (6.0E+09), and high (6.0E+11) dosages.


On day 8, mice were challenged intradermally (i.d.) with 10 μg KLH in saline (in a volume of 10 μL) in the left ear. Ear pinna thickness was measured at 24 hours following antigen challenge (FIG. 20). As determined by ear thickness, P. histicola smEVs were efficacious at suppressing inflammation in both their non-lyophilized and lyophilized forms.


For future inflammation studies, some groups of mice may be treated with anti-inflammatory agent(s) (e.g., anti-CD154, blockade of members of the TNF family, or other treatment), and/or an appropriate control (e.g., vehicle or control antibody) at various timepoints and at effective doses.


At various timepoints, serum samples may be taken. Other groups of mice may be sacrificed and lymph nodes, spleen, mesenteric lymph nodes (MLN), the small intestine, colon, and other tissues may be removed for histology studies, ex vivo histological, cytokine and/or flow cytometric analysis using methods known in the art. Some mice are exsanguinated from the orbital plexus under O2/CO2 anesthesia and ELISA assays performed.


Tissues may be dissociated using dissociation enzymes according to the manufacturer's instructions. Cells are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-MHCII, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Rory-gamma-t, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, MHCII, CD206, CD40, CSF1R, PD-L1, Gr-1, F4/80). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified CD45+ infiltrated immune cells obtained ex vivo. Finally, immunohistochemistry is carried out on various tissue sections to measure T cells, macrophages, dendritic cells, and checkpoint molecule protein expression.


Ears may be removed from the sacrificed animals and placed in cold EDTA-free protease inhibitor cocktail (Roche). Ears are homogenized using bead disruption and supernatants analyzed for various cytokines by Luminex kit (EMD Millipore) as per manufacturer's instructions. In addition, cervical lymph nodes are dissociated through a cell strainer, washed, and stained for FoxP3 (PE-FJK-165) and CD25 (FITC-PC61.5) using methods known in the art.


In order to examine the impact and longevity of DTH protection, rather than being sacrificed, some mice may be rechallenged with the challenging antigen at a later time and mice analyzed for susceptibility to DTH and severity of response.


Example 31
smEVs in a Mouse Model of Experimental Autoimmune Encephalomyelitis (EAE)

EAE is a well-studied animal model of multiple sclerosis, as reviewed by Constantinescu et al., (Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). Br J Pharmacol. 2011 October; 164(4): 1079-1106). It can be induced in a variety of mouse and rat strains using different myelin-associated peptides, by the adoptive transfer of activated encephalitogenic T cells, or the use of TCR transgenic mice susceptible to EAE, as discussed in Mangalam et al., (Two discreet subsets of CD8+ T cells modulate PLP91-110 induced experimental autoimmune encephalomyelitis in HLA-DR3 transgenic mice. J Autoimmun. 2012 June; 38(4): 344-353).


smEVs are tested for their efficacy in the rodent model of EAE, either alone or in combination with whole bacterial cells, with or without the addition of other anti-inflammatory treatments. Additionally, smEVs may be administered orally or via intravenous administration. For example, female 6-8 week old C57Bl/6 mice are obtained from Taconic (Germantown, N.Y.). Groups of mice are administered two subcutaneous (s.c.) injections at two sites on the back (upper and lower) of 0.1 ml myelin oligodentrocyte glycoprotein 35-55 (MOG35-55; 100 ug per injection; 200 ug per mouse (total 0.2 ml per mouse)), emulsified in Complete Freund's Adjuvant (CFA; 2-5 mg killed mycobacterium tuberculosis H37Ra/ml emulsion). Approximately 1-2 hours after the above, mice are intraperitoneally (i.p.) injected with 200 ng Pertussis toxin (PTx) in 0.1 ml PBS (2 ug/ml). An additional IP injection of PTx is administered on day 2. Alternatively, an appropriate amount of an alternative myelin peptide (e.g., proteolipid protein (PLP)) is used to induce EAE. Some animals serve as naïve controls. EAE severity is assessed and a disability score is assigned daily beginning on day 4 according to methods known in the art (Mangalam et al. 2012).


Treatment with smEVs is initiated at some point, either around the time of immunization or following EAE immunization. For example, smEVs may be administered at the same time as immunization (day 1), or they may be administered upon the first signs of disability (e.g., limp tail), or during severe EAE. smEVs are administered at varied doses and at defined intervals. For example, some mice are intravenously injected with smEVs at 10, 15, or 20 ug/mouse. Other mice may receive 25, 50, or 100 mg of smEVs per mouse. Alternatively, some mice receive between 7.0e+09 to 3.0e+12 smEV particles per dose. While some mice receive smEVs through i.v. injection, other mice may receive smEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, oral gavage, or other means of administration. Some mice may receive smEVs every day (e.g., starting on day 1), while others may receive smEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of smEVs and bacterial cells. For example, the composition may comprise smEV particles and whole bacteria in a ratio from 1:1 (smEVs:bacterial cells) to 1-1×1012:1 (smEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the smEV administration. As with the smEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the smEVs.


Some groups of mice may be treated with additional anti-inflammatory agent(s) or EAE therapeutic(s) (e.g., anti-CD154, blockade of members of the TNF family, Vitamin D, steroids, anti-inflammatory agents, or other treatment(s)), and/or an appropriate control (e.g., vehicle or control antibody) at various time points and at effective doses.


In addition, some mice are treated with antibiotics prior to treatment. For example, vancomycin (0.5 g/L), ampicillin (1.0 g/L), gentamicin (1.0 g/L) and amphotericin B (0.2 g/L) are added to the drinking water, and antibiotic treatment is halted at the time of treatment or a few days prior to treatment. Some immunized mice are treated without receiving antibiotics.


At various timepoints, mice are sacrificed and sites of inflammation (e.g., brain and spinal cord), lymph nodes, or other tissues may be removed for ex vivo histological, cytokine and/or flow cytometric analysis using methods known in the art. For example, tissues are dissociated using dissociation enzymes according to the manufacturer's instructions. Cells are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-MHCII, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Roryt, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, MHCII, CD206, CD40, CSF IR, PD-L1, Gr-1, F4/80). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL 12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified CD45+ central nervous system (CNS)-infiltrated immune cells obtained ex vivo. Finally, immunohistochemistry is carried out on various tissue sections to measure T cells, macrophages, dendritic cells, and checkpoint molecule protein expression.


In order to examine the impact and longevity of disease protection, rather than being sacrificed, some mice may be rechallenged with a disease trigger (e.g., activated encephalitogenic T cells or re-injection of EAE-inducing peptides). Mice are analyzed for susceptibility to disease and EAE severity following rechallenge.


Example 32
smEVs in a Mouse Model of Collagen-Induced Arthritis (CIA)

Collagen-induced arthritis (CIA) is an animal model commonly used to study rheumatoid arthritis (RA), as described by Caplazi et al. (Mouse models of rheumatoid arthritis. Veterinary Pathology. Sep. 1, 2015. 52(5): 819-826) (see also Brand et al. Collagen-induced arthritis. Nature Protocols. 2007. 2: 1269-1275; Pietrosimone et al. Collagen-induced arthritis: a model for murine autoimmune arthritis. Bio Protoc. 2015 Oct. 20; 5(20): e1626).


Among other versions of the CIA rodent model, one model involves immunizing HLA-DQ8 Tg mice with chick type II collagen as described by Taneja et al. (J. Immunology. 2007. 56: 69-78; see also Taneja et al. J. Immunology 2008. 181: 2869-2877; and Taneja et al. Arthritis Rheum., 2007. 56: 69-78). Purification of chick CII has been described by Taneja et al. (Arthritis Rheum., 2007. 56: 69-78). Mice are monitored for CIA disease onset and progression following immunization, and severity of disease is evaluated and “graded” as described by Wooley, J. Exp. Med. 1981. 154: 688-700.


Mice are immunized for CIA induction and separated into various treatment groups. smEVs are tested for their efficacy in CIA, either alone or in combination with whole bacterial cells, with or without the addition of other anti-inflammatory treatments.


Treatment with smEVs is initiated either around the time of immunization with collagen or post-immunization. For example, in some groups, smEVs may be administered at the same time as immunization (day 1), or smEVs may be administered upon first signs of disease, or upon the onset of severe symptoms. smEVs are administered at varied doses and at defined intervals. For example, some mice are intravenously injected with smEVs at 10, 15, or 20 ug/mouse. Other mice may receive 25, 50, or 100 mg of smEVs per mouse. Alternatively, some mice receive between 7.0e+09 to 3.0e+12 smEV particles per dose. While some mice receive smEVs through oral gavage or i.v. injection, while other groups of mice may receive smEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, or other means of administration. Some mice may receive smEVs every day (e.g., starting on day 1), while others may receive smEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of smEVs and bacterial cells. For example, the composition may comprise smEV particles and whole bacteria in a ratio from 1:1 (smEVs:bacterial cells) to 1-1×1012:1 (smEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the smEV administration. As with the smEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the smEVs.


Some groups of mice may be treated with additional anti-inflammatory agent(s) or CIA therapeutic(s) (e.g., anti-CD1 54, blockade of members of the TNF family, Vitamin D, steroid(s), anti-inflammatory agent(s), and/or other treatment), and/or an appropriate control (e.g., vehicle or control antibody) at various timepoints and at effective doses.


In addition, some mice are treated with antibiotics prior to treatment. For example, vancomycin (0.5 g/L), ampicillin (1.0 g/L), gentamicin (1.0 g/L) and amphotericin B (0.2 g/L) are added to the drinking water, and antibiotic treatment is halted at the time of treatment or a few days prior to treatment. Some immunized mice are treated without receiving antibiotics.


At various timepoints, serum samples are obtained to assess levels of anti-chick and anti-mouse CII IgG antibodies using a standard ELISA (Batsalova et al. Comparative analysis of collagen type II-specific immune responses during development of collagen-induced arthritis in two B10 mouse strains. Arthritis Res Ther. 2012. 14(6): R237). Also, some mice are sacrificed and sites of inflammation (e.g., synovium), lymph nodes, or other tissues may be removed for ex vivo histological, cytokine and/or flow cytometric analysis using methods known in the art. The synovium and synovial fluid are analyzed for plasma cell infiltration and the presence of antibodies using techniques known in the art. In addition, tissues are dissociated using dissociation enzymes according to the manufacturer's instructions to examine the profiles of the cellular infiltrates. Cells are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-MHCII, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Roryt, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, WICK CD206, CD40, CSF1R, PD-L1, Gr-1, F4/80). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified CD45+ synovium-infiltrated immune cells obtained ex vivo. Finally, immunohistochemistry is carried out on various tissue sections to measure T cells, macrophages, dendritic cells, and checkpoint molecule protein expression.


In order to examine the impact and longevity of disease protection, rather than being sacrificed, some mice may be rechallenged with a disease trigger (e.g., activated re-injection with CIA-inducing peptides). Mice are analyzed for susceptibility to disease and CIA severity following rechallenge.


Example 33
smEVs in a Mouse Model of Colitis

Dextran sulfate sodium (DSS)-induced colitis is a well-studied animal model of colitis, as reviewed by Randhawa et al. (A review on chemical-induced inflammatory bowel disease models in rodents. Korean J Physiol Pharmacol. 2014. 18(4): 279-288; see also Chassaing et al. Dextran sulfate sodium (DSS)-induced colitis in mice. Curr Protoc Immunol. 2014 Feb. 4; 104: Unit 15.25).


smEVs are tested for their efficacy in a mouse model of DSS-induced colitis, either alone or in combination with whole bacterial cells, with or without the addition of other anti-inflammatory agents.


Groups of mice are treated with DSS to induce colitis as known in the art (Randhawa et al. 2014; Chassaing et al. 2014; see also Kim et al. Investigating intestinal inflammation in DSS-induced model of IBD. J Vis Exp. 2012. 60: 3678). For example, male 6-8 week old C57Bl/6 mice are obtained from Charles River Labs, Taconic, or other vendor. Colitis is induced by adding 3% DSS (pmEV Biomedicals, Cat. #0260110) to the drinking water. Some mice do not receive DSS in the drinking water and serve as naïve controls. Some mice receive water for five (5) days. Some mice may receive DSS for a shorter duration or longer than five (5) days. Mice are monitored and scored using a disability activity index known in the art based on weight loss (e.g., no weight loss (score 0); 1-5% weight loss (score 1); 5-10% weight loss (score 2)); stool consistency (e.g., normal (score 0); loose stool (score 2); diarrhea (score 4)); and bleeding (e.g., no blood (score 0), hemoccult positive (score 1); hemoccult positive and visual pellet bleeding (score 2); blood around anus, gross bleeding (score 4).


Treatment with smEVs is initiated at some point, either on day 1 of DSS administration, or sometime thereafter. For example, smEVs may be administered at the same time as DSS initiation (day 1), or they may be administered upon the first signs of disease (e.g., weight loss or diarrhea), or during the stages of severe colitis. Mice are observed daily for weight, morbidity, survival, presence of diarrhea and/or bloody stool.


smEVs are administered at various doses and at defined intervals. For example, some mice receive between 7.0e+09 and 3.0e+12 smEV particles. While some mice receive smEVs through oral gavage or i.v. injection, while other groups of mice may receive smEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, or other means of administration. Some mice may receive smEVs every day (e.g., starting on day 1), while others may receive smEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of smEVs and bacterial cells. For example, the composition may comprise smEV particles and whole bacteria in a ratio from 1:1 (smEVs:bacterial cells) to 1-1×1012:1 (smEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the smEV administration. As with the smEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the smEVs.


Some groups of mice may be treated with additional anti-inflammatory agent(s) (e.g., anti-CD154, blockade of members of the TNF family, or other treatment), and/or an appropriate control (e.g., vehicle or control antibody) at various timepoints and at effective doses.


In addition, some mice are treated with antibiotics prior to treatment. For example, vancomycin (0.5 g/L), ampicillin (1.0 g/L), gentamicin (1.0 g/L) and amphotericin B (0.2 g/L) are added to the drinking water, and antibiotic treatment is halted at the time of treatment or a few days prior to treatment. Some mice receive DSS without receiving antibiotics beforehand.


At various timepoints, mice undergo video endoscopy using a small animal endoscope (Karl Storz Endoskipe, Germany) under isoflurane anesthesia. Still images and video are recorded to evaluate the extent of colitis and the response to treatment. Colitis is scored using criteria known in the art. Fecal material is collected for study.


At various timepoints, mice are sacrificed and the colon, small intestine, spleen, and lymph nodes (e.g., mesenteric lymph nodes) are collected. Additionally, blood is collected into serum separation tubes. Tissue damage is assessed through histological studies that evaluate, but are not limited to, crypt architecture, degree of inflammatory cell infiltration, and goblet cell depletion.


The gastrointestinal (GI) tract, lymph nodes, and/or other tissues may be removed for ex vivo histological, cytokine and/or flow cytometric analysis using methods known in the art. For example, tissues are harvested and may be dissociated using dissociation enzymes according to the manufacturer's instructions. Cells are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Roryt, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, MHCII, CD206, CD40, CSF1R, PD-L Gr-1, F4/80). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified CD45+ GI tract-infiltrated immune cells obtained ex vivo. Finally, immunohistochemistry is carried out on various tissue sections to measure T cells, macrophages, dendritic cells, and checkpoint molecule protein expression.


In order to examine the impact and longevity of disease protection, rather than being sacrificed, some mice may be rechallenged with a disease trigger. Mice are analyzed for susceptibility to colitis severity following rechallenge.


Example 34
smEVs in a Mouse Model of Type 1 Diabetes (T1D)

Type 1 diabetes (T1D) is an autoimmune disease in which the immune system targets the islets of Langerhans of the pancreas, thereby destroying the body's ability to produce insulin.


There are various models of animal models of T1D, as reviewed by Belle et al. (Mouse models for type 1 diabetes. Drug Discov Today Dis Models. 2009; 6(2): 41-45; see also Aileen J F King. The use of animal models in diabetes research. Br J Pharmacol. 2012 June; 166(3): 877-894. There are models for chemically-induced T1D, pathogen-induced T1D, as well as models in which the mice spontaneously develop T1D.


smEVs are tested for their efficacy in a mouse model of T1D, either alone or in combination with whole bacterial cells, with or without the addition of other anti-inflammatory treatments.


Depending on the method of T1D induction and/or whether T1D development is spontaneous, treatment with smEVs is initiated at some point, either around the time of induction or following induction, or prior to the onset (or upon the onset) of spontaneously-occurring T1D. smEVs are administered at varied doses and at defined intervals. For example, some mice are intravenously injected with smEVs at 10, 15, or 20 ug/mouse. Other mice may receive 25, 50, or 100 mg of smEVs per mouse. Alternatively, some mice receive between 7.0e+09 to 3.0e+12 smEV particles per dose. While some mice receive smEVs through oral gavage or i.v. injection, while other groups of mice may receive smEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, or other means of administration. Some mice may receive smEVs every day, while others may receive smEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of smEVs and bacterial cells. For example, the composition may comprise smEV particles and whole bacteria in a ratio from 1:1 (smEVs:bacterial cells) to 1-1×1012:1 (smEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the smEV administration. As with the smEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the smEVs.


Some groups of mice may be treated with additional treatments and/or an appropriate control (e.g., vehicle or control antibody) at various timepoints and at effective doses.


In addition, some mice are treated with antibiotics prior to treatment. For example, vancomycin (0.5 g/L), ampicillin (1.0 g/L), gentamicin (1.0 g/L) and amphotericin B (0.2 g/L) are added to the drinking water, and antibiotic treatment is halted at the time of treatment or a few days prior to treatment. Some immunized mice are treated without receiving antibiotics.


Blood glucose is monitored biweekly prior to the start of the experiment. At various timepoints thereafter, nonfasting blood glucose is measured. At various timepoints, mice are sacrificed and site the pancreas, lymph nodes, or other tissues may be removed for ex vivo histological, cytokine and/or flow cytometric analysis using methods known in the art. For example, tissues are dissociated using dissociation enzymes according to the manufacturer's instructions. Cells are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-MHCII, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Roryt, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, MHCII, CD206, CD40, CSF1R, PD-L1, Gr-1, F4/80). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified tissue-infiltrating immune cells obtained ex vivo. Finally, immunohistochemistry is carried out on various tissue sections to measure T cells, macrophages, dendritic cells, and checkpoint molecule protein expression. Antibody production may also be assessed by ELISA.


In order to examine the impact and longevity of disease protection, rather than being sacrificed, some mice may be rechallenged with a disease trigger, or assessed for susceptibility to relapse. Mice are analyzed for susceptibility to diabetes onset and severity following rechallenge (or spontaneously-occurring relapse).


Example 35
smEVs in a Mouse Model of Primary Sclerosing Cholangitis (PSC)

Primary Sclerosing Cholangitis (PSC) is a chronic liver disease that slowly damages the bile ducts and leads to end-stage cirrhosis. It is associated with inflammatory bowel disease (IBD).


There are various animal models for PSC, as reviewed by Fickert et al. (Characterization of animal models for primary sclerosing cholangitis (PSC). J Hepatol. 2014 June. 60(6): 1290-1303; see also Pollheimer and Fickert. Animal models in primary biliary cirrhosis and primary sclerosing cholangitis. Clin Rev Allergy Immunol. 2015 June. 48(2-3): 207-17). Induction of disease in PSC models includes chemical induction (e.g., 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-induced cholangitis), pathogen-induced (e.g., Cryptosporidium parvum), experimental biliary obstruction (e.g., common bile duct ligation (CBDL)), and transgenic mouse model of antigen-driven biliary injury (e.g., Ova-Bil transgenic mice). For example, bile duct ligation is performed as described by Georgiev et al. (Characterization of time-related changes after experimental bile duct ligation. Br J Surg. 2008. 95(5): 646-56), or disease is induced by DCC exposure as described by Fickert et al. (A new xenobiotic-induced mouse model of sclerosing cholangitis and biliary fibrosis. Am J Path. Vol 171(2): 525-536.


smEVs are tested for their efficacy in a mouse model of PSC, either alone or in combination with whole bacterial cells, with or without the addition of some other therapeutic agent.


DCC-Induced Cholangitis

For example, 6-8 week old C57bl/6 mice are obtained from Taconic or other vendor. Mice are fed a 0.1% DCC-supplemented diet for various durations. Some groups receive DCC-supplement food for 1 week, others for 4 weeks, others for 8 weeks. Some groups of mice may receive a DCC-supplemented diet for a length of time and then be allowed to recover, thereafter receiving a normal diet. These mice may be studied for their ability to recover from disease and/or their susceptibility to relapse upon subsequent exposure to DCC. Treatment with smEVs is initiated at some point, either around the time of DCC-feeding or subsequent to initial exposure to DCC. For example, smEVs may be administered on day 1, or they may be administered sometime thereafter. smEVs are administered at varied doses and at defined intervals. For example, some mice are intravenously injected with smEVs at 10, 15, or 20 ug/mouse. Alternatively, some mice may receive between 7.0e+09 and 3.0e+12 smEV particles. While some mice receive smEVs through oral gavage or i.v. injection, while other groups of mice may receive smEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, or other means of administration. Some mice may receive smEVs every day (e.g., starting on day 1), while others may receive smEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of smEVs and bacterial cells. For example, the composition may comprise smEV particles and whole bacteria in a ratio from 1:1 (smEVs:bacterial cells) to 1-1×1012:1 (smEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the smEV administration. As with the smEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the smEVs.


Some groups of mice may be treated with additional agents and/or an appropriate control (e.g., vehicle or antibody) at various timepoints and at effective doses.


In addition, some mice are treated with antibiotics prior to treatment. For example, vancomycin (0.5 g/L), ampicillin (1.0 g/L), gentamicin (1.0 g/L) and amphotericin B (0.2 g/L) are added to the drinking water, and antibiotic treatment is halted at the time of treatment or a few days prior to treatment. Some immunized mice are treated without receiving antibiotics. At various timepoints, serum samples are analyzed for ALT, AP, bilirubin, and serum bile acid (BA) levels.


At various timepoints, mice are sacrificed, body and liver weight are recorded, and sites of inflammation (e.g., liver, small and large intestine, spleen), lymph nodes, or other tissues may be removed for ex vivo histolomorphological characterization, cytokine and/or flow cytometric analysis using methods known in the art (see Fickert et al. Characterization of animal models for primary sclerosing cholangitis (PSC)). J Hepatol. 2014. 60(6): 1290-1303). For example, bile ducts are stained for expression of ICAM-1, VCAM-1, MadCAM-1. Some tissues are stained for histological examination, while others are dissociated using dissociation enzymes according to the manufacturer's instructions. Cells are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-MHCII, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Roryt, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, MHCII, CD206, CD40, CSF1R, PD-L1, Gr-1, F4/80), as well as adhesion molecule expression (ICAM-1, VCAM-1, MadCAM-1). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified CD45+ bile duct-infiltrated immune cells obtained ex vivo.


Liver tissue is prepared for histological analysis, for example, using Sirius-red staining followed by quantification of the fibrotic area. At the end of the treatment, blood is collected for plasma analysis of liver enzymes, for example, AST or ALT, and to determine Bilirubin levels. The hepatic content of Hydroxyproline can be measured using established protocols. Hepatic gene expression analysis of inflammation and fibrosis markers may be performed by qRT-PCR using validated primers. These markers may include, but are not limited to, MCP-1, alpha-SMA, Coll1a1, and TIMP-. Metabolite measurements may be performed in plasma, tissue and fecal samples using established metabolomics methods. Finally, immunohistochemistry is carried out on liver sections to measure neutrophils, T cells, macrophages, dendritic cells, or other immune cell infiltrates.


In order to examine the impact and longevity of disease protection, rather than being sacrificed, some mice may be rechallenged with DCC at a later time. Mice are analyzed for susceptibility to cholangitis and cholangitis severity following rechallenge.


BDL-Induced Cholangitis

Alternatively, smEVs are tested for their efficacy in BDL-induced cholangitis. For example, 6-8 week old C57Bl/6J mice are obtained from Taconic or other vendor. After an acclimation period the mice are subjected to a surgical procedure to perform a bile duct ligation (BDL). Some control animals receive a sham surgery. The BDL procedure leads to liver injury, inflammation and fibrosis within 7-21 days.


Treatment with smEVs is initiated at some point, either around the time of surgery or some time following the surgery. smEVs are administered at varied doses and at defined intervals. For example, some mice are intravenously injected with smEVs at 10, 15, or 20 ug/mouse. Other mice may receive 25, 50, or 100 mg of smEVs per mouse. Alternatively, some mice receive between 7.0e+09 to 3.0e+12 smEV particles per dose. While some mice receive smEVs through oral gavage or i.v. injection, while other groups of mice may receive smEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, or other means of administration. Some mice receive smEVs every day (e.g., starting on day 1), while others may receive smEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of smEVs and bacterial cells. For example, the composition may comprise smEV particles and whole bacteria in a ratio from 1:1 (smEVs:bacterial cells) to 1-1×1012:1 (smEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the smEV administration. As with the smEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the smEVs.


Some groups of mice may be treated with additional agents and/or an appropriate control (e.g., vehicle or antibody) at various timepoints and at effective doses.


In addition, some mice are treated with antibiotics prior to treatment. For example, vancomycin (0.5 g/L), ampicillin (1.0 g/L), gentamicin (1.0 g/L) and amphotericin B (0.2 g/L) are added to the drinking water, and antibiotic treatment is halted at the time of treatment or a few days prior to treatment. Some immunized mice are treated without receiving antibiotics. At various timepoints, serum samples are analyzed for ALT, AP, bilirubin, and serum bile acid (BA) levels.


At various timepoints, mice are sacrificed, body and liver weight are recorded, and sites of inflammation (e.g., liver, small and large intestine, spleen), lymph nodes, or other tissues may be removed for ex vivo histolomorphological characterization, cytokine and/or flow cytometric analysis using methods known in the art (see Fickert et al. Characterization of animal models for primary sclerosing cholangitis (PSC)). J Hepatol. 2014. 60(6): 1290-1303). For example, bile ducts are stained for expression of ICAM-1, VCAM-1, MadCAM-1. Some tissues are stained for histological examination, while others are dissociated using dissociation enzymes according to the manufacturer's instructions. Cells are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-MHCII, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Roryt, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, MITCH, CD206, CD40, CSF1R, PD-L1, Gr-1, F4/80), as well as adhesion molecule expression (ICAM-1, VCAM-1, MadCAM-1). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified CD45+ bile duct-infiltrated immune cells obtained ex vivo.


Liver tissue is prepared for histological analysis, for example, using Sirius-red staining followed by quantification of the fibrotic area. At the end of the treatment, blood is collected for plasma analysis of liver enzymes, for example, AST or ALT, and to determine Bilirubin levels. The hepatic content of Hydroxyproline can be measured using established protocols. Hepatic gene expression analysis of inflammation and fibrosis markers may be performed by qRT-PCR using validated primers. These markers may include, but are not limited to, MCP-1, alpha-SMA, Coll1a1, and TIMP. Metabolite measurements may be performed in plasma, tissue and fecal samples using established metabolomics methods. Finally, immunohistochemistry is carried out on liver sections to measure neutrophils, T cells, macrophages, dendritic cells, or other immune cell infiltrates.


In order to examine the impact and longevity of disease protection, rather than being sacrificed, some mice may be analyzed for recovery.


Example 36
smEVs in a Mouse Model of Nonalcoholic Steatohepatitis (NASH)

Nonalcoholic Steatohepatitis (NASH) is a severe form of Nonalcoholic Fatty Liver Disease (NAFLD), where buildup of hepatic fat (steatosis) and inflammation lead to liver injury and hepatocyte cell death (ballooning).


There are various animal models of NASH, as reviewed by Ibrahim et al. (Animal models of nonalcoholic steatohepatitis: Eat, Delete, and Inflame. Dig Dis Sci. 2016 May. 61(5): 1325-1336; see also Lau et al. Animal models of non-alcoholic fatty liver disease: current perspectives and recent advances 2017 January. 241(1): 36-44).


smEVs are tested for their efficacy in a mouse model of NASH, either alone or in combination with whole bacterial cells, with or without the addition of another therapeutic agent. For example, 8-10 week old C57Bl/6J mice, obtained from Taconic (Germantown, N.Y.), or other vendor, are placed on a methionine choline deficient (MCD) diet for a period of 4-8 weeks during which NASH features develop, including steatosis, inflammation, ballooning and fibrosis.



P. histicola-derived smEVs are tested for their efficacy in a mouse model of NASH, either alone or in combination with each other, in varying proportions, with or without the addition of another therapeutic agent. For example, 8 week old C57Bl/6J mice, obtained from Charles River (France), or other vendor, are acclimated for a period of 5 days, randomized intro groups of 10 mice based on body weight, and placed on a methionine choline deficient (MCD) diet for example A02082002B from Research Diets (USA), for a period of 4 weeks during which NASH features developed, including steatosis, inflammation, ballooning and fibrosis. Control chow mice are fed a normal chow diet, for example RM1 (E) 801492 from SDS Diets (UK). Control chow, MCD diet, and water are provided ad libitum.


An NAS scoring system adapted from Kleiner et al. (Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology. 2005 June. 41(6): 1313-1321) is used to determine the degree of steatosis (scored 0-3), lobular inflammation (scored 0-3), hepatocyte ballooning (scored 0-3), and fibrosis (scored 0-4). An individual mouse NAS score may be calculated by summing the score for steatosis, inflammation, ballooning, and fibrosis (scored 0-13). In addition, the levels of plasma AST and ALT are determined using a Pentra 400 instrument from Horiba (USA), according to manufacturer's instructions. The levels of hepatic total cholesterol, triglycerides, fatty acids, alanine aminotransferase, and aspartate aminotransferase are also determined using methods known in the art.


In other studies, hepatic gene expression analysis of inflammation, fibrosis, steatosis, ER stress, or oxidative stress markers may be performed by qRT-PCR using validated primers. These markers may include, but are not limited to, IL-1β, TNF-α, MCP-1, α-SMA, Coll1a1, CHOP, and NRF2.


Treatment with smEVs is initiated at some point, either at the beginning of the diet, or at some point following diet initiation (for example, one week after). For example, smEVs may be administered starting in the same day as the initiation of the MCD diet. smEVs are administered at varied doses and at defined intervals. For example, some mice are intravenously injected with smEVs at 10, 15, or 20 ug/mouse. Other mice may receive 25, 50, or 100 mg of smEVs per mouse. Alternatively, some mice receive between 7.0e+09 to 3.0e+12 smEV particles per dose. While some mice receive smEVs through oral gavage or i.v. injection, while other groups of mice may receive smEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, or other means of administration. Some mice may receive smEVs every day (e.g., starting on day 1), while others may receive smEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of smEVs and bacterial cells. For example, the composition may comprise smEV particles and whole bacteria in a ratio from 1:1 (smEVs:bacterial cells) to 1-1×1012:1 (smEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the smEV administration. As with the smEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the smEVs.


Some groups of mice may be treated with additional NASH therapeutic(s) (e.g., FXR agonists, PPAR agonists, CCR2/5 antagonists or other treatment) and/or appropriate control at various timepoints and effective doses.


At various timepoints and/or at the end of the treatment, mice are sacrificed and liver, intestine, blood, feces, or other tissues may be removed for ex vivo histological, biochemical, molecular or cytokine and/or flow cytometry analysis using methods known in the art. For example, liver tissues are weighed and prepared for histological analysis, which may comprise staining with H&E, Sirius Red, and determination of NASH activity score (NAS). At various timepoints, blood is collected for plasma analysis of liver enzymes, for example, AST or ALT, using standards assays. In addition, the hepatic content of cholesterol, triglycerides, or fatty acid acids can be measured using established protocols. Hepatic gene expression analysis of inflammation, fibrosis, steatosis, ER stress, or oxidative stress markers may be performed by qRT-PCR using validated primers. These markers may include, but are not limited to, IL-6, MCP-1, alpha-SMA, Coll1a1, CHOP, and NRF2. Metabolite measurements may be performed in plasma, tissue and fecal samples using established biochemical and mass-spectrometry-based metabolomics methods. Serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-ib, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified CD45+ bile duct-infiltrated immune cells obtained ex vivo. Finally, immunohistochemistry is carried out on liver or intestine sections to measure neutrophils, T cells, macrophages, dendritic cells, or other immune cell infiltrates.


In order to examine the impact and longevity of disease protection, rather than being sacrificed, some mice may be analyzed for recovery.


Example 37
smEVs in a Mouse Model of Psoriasis

Psoriasis is a T-cell-mediated chronic inflammatory skin disease. So-called “plaque-type” psoriasis is the most common form of psoriasis and is typified by dry scales, red plaques, and thickening of the skin due to infiltration of immune cells into the dermis and epidermis. Several animal models have contributed to the understanding of this disease, as reviewed by Gudjonsson et al. (Mouse models of psoriasis. J Invest Derm. 2007. 127: 1292-1308; see also van der Fits et al. Imiquimod-induced psoriasis-like skin inflammation in mice is mediated via the IL-23/IL-17 axis. J. Immunol. 2009 May 1. 182(9): 5836-45).


Psoriasis can be induced in a variety of mouse models, including those that use transgenic, knockout, or xenograft models, as well as topical application of imiquimod (IMQ), a TLR7/8 ligand.


smEVs are tested for their efficacy in the mouse model of psoriasis, either alone or in combination with whole bacterial cells, with or without the addition of other anti-inflammatory treatments. For example, 6-8 week old C57Bl/6 or Balb/c mice are obtained from Taconic (Germantown, N.Y.), or other vendor. Mice are shaved on the back and the right ear. Groups of mice receive a daily topical dose of 62.5 mg of commercially available IMQ cream (5%) (Aldara; 3M Pharmaceuticals). The dose is applied to the shaved areas for 5 or 6 consecutive days. At regular intervals, mice are scored for erythema, scaling, and thickening on a scale from 0 to 4, as described by van der Fits et al. (2009). Mice are monitored for ear thickness using a Mitutoyo micrometer.


Treatment with smEVs is initiated at some point, either around the time of the first application of IMQ, or something thereafter. For example, smEVs may be administered at the same time as the subcutaneous injections (day 0), or they may be administered prior to, or upon, application. smEVs are administered at varied doses and at defined intervals. For example, some mice are intravenously injected with smEVs at 10, 15, or 20 ug/mouse. Other mice may receive 25, 50, or 100 mg of smEVs per mouse. Alternatively, some mice receive between 7.0e+09 to 3.0e+12 smEV particles per dose. While some mice receive smEVs through oral gavage or i.v. injection, while other groups of mice may receive smEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, or other means of administration. Some mice may receive smEVs every day (e.g., starting on day 0), while others may receive smEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of smEVs and bacterial cells. For example, the composition may comprise smEV particles and whole bacteria in a ratio from 1:1 (smEVs:bacterial cells) to 1-1×1012:1 (smEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the smEV administration. As with the smEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the smEVs.


Some groups of mice may be treated with anti-inflammatory agent(s) (e.g., anti-CD154, blockade of members of the TNF family, or other treatment), and/or an appropriate control (e.g., vehicle or control antibody) at various timepoints and at effective doses.


In addition, some mice are treated with antibiotics prior to treatment. For example, vancomycin (0.5 g/L), ampicillin (1.0 g/L), gentamicin (1.0 g/L) and amphotericin B (0.2 g/L) are added to the drinking water, and antibiotic treatment is halted at the time of treatment or a few days prior to treatment. Some immunized mice are treated without receiving antibiotics.


At various timepoints, samples from back and ear skin are taken for cryosection staining analysis using methods known in the art. Other groups of mice are sacrificed and lymph nodes, spleen, mesenteric lymph nodes (MLN), the small intestine, colon, and other tissues may be removed for histology studies, ex vivo histological, cytokine and/or flow cytometric analysis using methods known in the art. Some tissues may be dissociated using dissociation enzymes according to the manufacturer's instructions. Cryosection samples, tissue samples, or cells obtained ex vivo are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-MHCII, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Roryt, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, MCHII, CD206, CD40, CSF1R, PD-L1, Gr-1, F4/80). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified CD45+ skin-infiltrated immune cells obtained ex vivo. Finally, immunohistochemistry is carried out on various tissue sections to measure T cells, macrophages, dendritic cells, and checkpoint molecule protein expression.


In order to examine the impact and longevity of psoriasis protection, rather than being sacrificed, some mice may be studied to assess recovery, or they may be rechallenged with IMQ. The groups of rechallenged mice are analyzed for susceptibility to psoriasis and severity of response.


Example 38
smEVs in a Mouse Model of Obesity (DIO)

There are various animal models of DIO, as reviewed by Tschop et al. (A guide to analysis of mouse energy metabolism. Nat. Methods. 2012; 9(1):57-63) and Ayala et al. (Standard operating procedures for describing and performing metabolic tests of glucose homeostasis in mice. Disease Models and Mechanisms. 2010; 3:525-534) and provided by Physiogenex.


smEVs are tested for their efficacy in a mouse model of DIO, either alone or in combination with other whole bacterial cells (live, killed, irradiated, and/or inactivated, etc) with or without the addition of other anti-inflammatory treatments.


Depending on the method of DIO induction and/or whether DIO development is spontaneous, treatment with smEVs is initiated at some point, either around the time of induction or following induction, or prior to the onset (or upon the onset) of spontaneously-occurring T1D. smEVs are administered at varied doses and at defined intervals. For example, some mice are intravenously injected with smEVs at 10, 15, or 20 ug/mouse. Other mice may receive 25, 50, or 100 mg of smEVs per mouse. Alternatively, some mice receive between 7.0e+09 to 3.0e+12 smEV particles per dose. While some mice receive smEVs through i.v. injection, other mice may receive smEVs through intraperitoneal (i.p.) injection, subcutaneous (s.c.) injection, nasal route administration, oral gavage, or other means of administration. Some mice may receive smEVs every day, while others may receive smEVs at alternative intervals (e.g., every other day, or once every three days). Groups of mice may be administered a pharmaceutical composition of the invention comprising a mixture of smEVs and bacterial cells. For example, the composition may comprise smEV particles and whole bacteria in a ratio from 1:1 (smEVs:bacterial cells) to 1-1×1012:1 (smEVs:bacterial cells).


Alternatively, some groups of mice may receive between 1×104 and 5×109 bacterial cells in an administration separate from, or comingled with, the smEV administration. As with the smEVs, bacterial cell administration may be varied by route of administration, dose, and schedule. The bacterial cells may be live, dead, or weakened. The bacterial cells may be harvested fresh (or frozen) and administered, or they may be irradiated or heat-killed prior to administration with the smEVs.


Some groups of mice may be treated with additional treatments and/or an appropriate control (e.g., vehicle or control antibody) at various timepoints and at effective doses.


In addition, some mice are treated with antibiotics prior to treatment. For example, vancomycin (0.5 g/L), ampicillin (1.0 g/L), gentamicin (1.0 g/L) and amphotericin B (0.2 g/L) are added to the drinking water, and antibiotic treatment is halted at the time of treatment or a few days prior to treatment. Some immunized mice are treated without receiving antibiotics.


Blood glucose is monitored biweekly prior to the start of the experiment. At various timepoints thereafter, nonfasting blood glucose is measured. At various timepoints, mice are sacrificed and site the pancreas, lymph nodes, or other tissues may be removed for ex vivo histological, cytokine and/or flow cytometric analysis using methods known in the art. For example, tissues are dissociated using dissociation enzymes according to the manufacturer's instructions. Cells are stained for analysis by flow cytometry using techniques known in the art. Staining antibodies can include anti-CD11c (dendritic cells), anti-CD80, anti-CD86, anti-CD40, anti-MHCII, anti-CD8a, anti-CD4, and anti-CD103. Other markers that may be analyzed include pan-immune cell marker CD45, T cell markers (CD3, CD4, CD8, CD25, Foxp3, T-bet, Gata3, Roryt, Granzyme B, CD69, PD-1, CTLA-4), and macrophage/myeloid markers (CD11b, MCHII, CD206, CD40, CSF1R, PD-L1, Gr-1, F4/80). In addition to immunophenotyping, serum cytokines can be analyzed including, but not limited to, TNFa, IL-17, IL-13, IL-12p70, IL12p40, IL-10, IL-6, IL-5, IL-4, IL-2, IL-1b, IFNy, GM-CSF, G-CSF, M-CSF, MIG, IP10, MIP1b, RANTES, and MCP-1. Cytokine analysis may be carried out on immune cells obtained from lymph nodes or other tissue, and/or on purified tissue-infiltrating immune cells obtained ex vivo. Finally, immunohistochemistry is carried out on various tissue sections to measure T cells, macrophages, dendritic cells, and checkpoint molecule protein expression. Antibody production may also be assessed by ELISA.


In order to examine the impact and longevity of disease protection, rather than being sacrificed, some mice may be rechallenged with a disease trigger, or assessed for susceptibility to relapse. Mice are analyzed for susceptibility to diabetes onset and severity following rechallenge (or spontaneously-occurring relapse).


Example 39
Labeling Bacterial smEVs

smEVs may be labeled in order to track their biodistribution in vivo and to quantify and track cellular localization in various preparations and in assays conducted with mammalian cells. For example, smEVs may be radio-labeled, incubated with dyes, fluorescently labeled, luminescently labeled, or labeled with conjugates containing metals and isotopes of metals.


For example, smEVs may be incubated with dyes conjugated to functional groups such as NHS-ester, click-chemistry groups, streptavidin or biotin. The labeling reaction may occur at a variety of temperatures for minutes or hours, and with or without agitation or rotation. The reaction may then be stopped by adding a reagent such as bovine serum albumin (BSA), or similar agent, depending on the protocol, and free or unbound dye molecule removed by ultra-centrifugation, filtration, centrifugal filtration, column affinity purification or dialysis. Additional washing steps involving wash buffers and vortexing or agitation may be employed to ensure complete removal of free dyes molecules such as described in Su Chul Jang et al, Small. 11, No. 4, 456-461(2017).


Fluorescently labeled smEVs are detected in cells or organs, or in in vitro and/or ex vivo samples by confocal microscopy, nanoparticle tracking analysis, flow cytometry, fluorescence activated cell sorting (FACs) or fluorescent imaging system such as the Odyssey CLx LICOR (see e.g., Wiklander et al. 2015. J. Extracellular Vesicles. 4:10.3402/jev.v4.26316). Additionally, fluorescently labeled smEVs are detected in whole animals and/or dissected organs and tissues using an instrument such as the IVIS spectrum CT (Perkin Elmer) or Pearl Imager, as in H-I. Choi, et al. Experimental & Molecular Medicine. 49: e330 (2017).


smEVs may be labeled with conjugates containing metals and isotopes of metals using the protocols described above. Metal-conjugated smEVs may be administered in vivo to animals. Cells may then be harvested from organs at various time-points, and analyzed ex vivo. Alternatively, cells derived from animals, humans, or immortalized cell lines may be treated with metal-labelled smEVs in vitro and cells subsequently labelled with metal-conjugated antibodies and phenotyped using a Cytometry by Time of Flight (CyTOF) instrument such as the Helios CyTOF (Fluidigm) or imaged and analyzed using and Imaging Mass Cytometry instrument such as the Hyperion Imaging System (Fluidigm). Additionally, smEVs may be labelled with a radioisotope to track the smEVs biodistribution (see, e.g., Miller et al., Nanoscale. 2014 May 7; 6(9):4928-35).


Example 40
Transmission Electron Microscopy to Visualize Purified Bacterial smEVs

smEVs are purified from bacteria batch cultures. Transmission electron microscopy (TEM) may be used to visualize purified bacterial smEVs (S. Bin Park, et al. PLoS ONE. 6(3):e17629 (2011). smEVs are mounted onto 300- or 400-mesh-size carbon-coated copper grids (Electron Microscopy Sciences, USA) for 2 minutes and washed with deionized water. smEVs are negatively stained using 2% (w/v) uranyl acetate for 20 sec-1 min. Copper grids are washed with sterile water and dried. Images are acquired using a transmission electron microscope with 100-120 kV acceleration voltage. Stained smEVs appear between 20-600 nm in diameter and are electron dense. 10-50 fields on each grid are screened.


Example 41
Profiling smEV Composition and Content

smEVs may be characterized by any one of various methods including, but not limited to, NanoSight characterization, SDS-PAGE gel electrophoresis, Western blot, ELISA, liquid chromatography-mass spectrometry and mass spectrometry, dynamic light scattering, lipid levels, total protein, lipid to protein ratios, nucleic acid analysis and/or zeta potential.


NanoSight Characterization of smEVs


Nanoparticle tracking analysis (NTA) is used to characterize the size distribution of purified smEVs. Purified smEV preparations are run on a NanoSight machine (Malvern Instruments) to assess smEV size and concentration.


SDS-PAGE Gel Electrophoresis

To identify the protein components of purified smEVs, samples are run on a gel, for example a Bolt Bis-Tris Plus 4-12% gel (Thermo-Fisher Scientific), using standard techniques. Samples are boiled in 1× SDS sample buffer for 10 minutes, cooled to 4° C., and then centrifuged at 16,000×g for 1 min. Samples are then run on a SDS-PAGE gel and stained using one of several standard techniques (e.g., Silver staining, Coomassie Blue, Gel Code Blue) for visualization of bands.


Western Blot Analysis

To identify and quantify specific protein components of purified smEVs, smEV proteins are separated by SDS-PAGE as described above and subjected to Western blot analysis (Cvjetkovic et al., Sci. Rep. 6, 36338 (2016)) and are quantified via ELISA.


smEV Proteomics and Liquid Chromatography-Mass Spectrometry (LC-MS/MS) and Mass Spectrometry (MS)


Proteins present in smEVs are identified and quantified by Mass Spectrometry techniques. smEV proteins may be prepared for LC-MS/MS using standard techniques including protein reduction using dithiotreitol solution (DTT) and protein digestion using enzymes such as LysC and trypsin as described in Erickson et al, 2017 (Molecular Cell, VOLUME 65, ISSUE 2, P361-370, JAN. 19, 2017). Alternatively, peptides are prepared as described by Liu et al. 2010 (JOURNAL OF BACTERIOLOGY, June 2010, p. 2852-2860 Vol. 192, No. 11), Kieselbach and Oscarsson 2017 (Data Brief. 2017 February; 10: 426-431.), Vildhede et al, 2018 (Drug Metabolism and Disposition Feb. 8, 2018). Following digestion, peptide preparations are run directly on liquid chromatography and mass spectrometry devices for protein identification within a single sample. For relative quantitation of proteins between samples, peptide digests from different samples are labeled with isobaric tags using the iTRAQ Reagent-8plex Multiplex Kit (Applied Biosystems, Foster City, Calif.) or TMT 10plex and 11plex Label Reagents (Thermo Fischer Scientific, San Jose, Calif., USA). Each peptide digest is labeled with a different isobaric tag and then the labeled digests are combined into one sample mixtur. The combined peptide mixture is analyzed by LC-MS/MS for both identification and quantification. A database search is performed using the LC-MS/MS data to identify the labeled peptides and the corresponding proteins. In the case of isobaric labeling, the fragmentation of the attached tag generates a low molecular mass reporter ion that is used to obtain a relative quantitation of the peptides and proteins present in each smEV.


Additionally, metabolic content is ascertained using liquid chromatography techniques combined with mass spectrometry. A variety of techniques exist to determine metabolomic content of various samples and are known to one skilled in the art involving solvent extraction, chromatographic separation and a variety of ionization techniques coupled to mass determination (Roberts et al 2012 Targeted Metabolomics. Curr Protoc Mol Biol. 30: 1-24; Dettmer et al 2007, Mass spectrometry-based metabolomics. Mass Spectrom Rev. 26(1):51-78). As a non-limiting example, a LC-MS system includes a 4000 QTRAP triple quadrupole mass spectrometer (AB SCIEX) combined with 1100 Series pump (Agilent) and an HTS PAL autosampler (Leap Technologies). Media samples or other complex metabolic mixtures (˜10 μL) are extracted using nine volumes of 74.9:24.9:0.2 (v/v/v) acetonitrile/methanol/formic acid containing stable isotope-labeled internal standards (valine-d8, Isotec; and phenylalanine-d8, Cambridge Isotope Laboratories). Standards may be adjusted or modified depending on the metabolites of interest. The samples are centrifuged (10 minutes, 9,000 g, 4° C.), and the supernatants (10 μL) are submitted to LCMS by injecting the solution onto the HILIC column (150×2.1 mm, 3 μm particle size). The column is eluted by flowing a 5% mobile phase [10 mM ammonium formate, 0.1% formic acid in water] for 1 minute at a rate of 250 uL/minute followed by a linear gradient over 10 minutes to a solution of 40% mobile phase [acetonitrile with 0.1% formic acid]. The ion spray voltage is set to 4.5 kV and the source temperature is 450° C.


The data are analyzed using commercially available software like Multiquant 1.2 from AB SCIEX for mass spectrum peak integration. Peaks of interest should be manually curated and compared to standards to confirm the identity of the peak. Quantitation with appropriate standards is performed to determine the number of metabolites present in the initial media, after bacterial conditioning and after tumor cell growth. A non-targeted metabolomics approach may also be used using metabolite databases, such as but not limited to the NIST database, for peak identification.


Dynamic Light Scattering (DLS)


DLS measurements, including the distribution of particles of different sizes in different smEV preparations are taken using instruments such as the DynaPro NanoStar (Wyatt Technology) and the Zetasizer Nano ZS (Malvern Instruments).


Lipid Levels

Lipid levels are quantified using FM4-64 (Life Technologies), by methods similar to those described by A. J. McBroom et al. J Bacteriol 188:5385-5392. and A. Frias, et al. Microb Ecol. 59:476-486 (2010). Samples are incubated with FM4-64 (3.3 μg/mL in PBS for 10 minutes at 37° C. in the dark). After excitation at 515 nm, emission at 635 nm is measured using a Spectramax M5 plate reader (Molecular Devices). Absolute concentrations are determined by comparison of unknown samples to standards (such as palmitoyloleoylphosphatidylglycerol (POPG) vesicles) of known concentrations. Lipidomics can be used to identify the lipids present in the smEVs.


Total Protein

Protein levels are quantified by standard assays such as the Bradford and BCA assays. The Bradford assays are run using Quick Start Bradford 1× Dye Reagent (Bio-Rad), according to manufacturer's protocols. BCA assays are run using the Pierce BCA Protein Assay Kit (Thermo-Fisher Scientific). Absolute concentrations are determined by comparison to a standard curve generated from BSA of known concentrations. Alternatively, protein concentration can be calculated using the Beer-Lambert equation using the sample absorbance at 280 nm (A280) as measured on a Nanodrop spectrophotometer (Thermo-Fisher Scientific),In addition, proteomics may be used to identify proteins in the sample.


Lipid:Protein Ratios

Lipid:protein ratios are generated by dividing lipid concentrations by protein concentrations. These provide a measure of the purity of vesicles as compared to free protein in each preparation.


Nucleic Acid Analysis

Nucleic acids are extracted from smEVs and quantified using a Qubit fluorometer. Size distribution is assessed using a BioAnalyzer and the material is sequenced.


Zeta Potential

The zeta potential of different preparations are measured using instruments such as the Zetasizer ZS (Malvern Instruments).


Example 42
In Vitro Screening of smEVs for Enhanced Activation of Dendritic Cells

In vitro immune responses are thought to simulate mechanisms by which immune responses are induced in vivo, e.g., as in response to a cancer microenvironment. Briefly, PBMCs are isolated from heparinized venous blood from healthy donors by gradient centrifugation using Lymphoprep (Nycomed, Oslo, Norway), or from mouse spleens or bone marrow using the magnetic bead-based Human Blood Dendritic cell isolation kit (Miltenyi Biotech, Cambridge, Mass.). Using anti-human CD14 mAb, the monocytes are purified by Moflo and cultured in cRPMI at a cell density of 5e5 cells/ml in a 96-well plate (Costar Corp) for 7 days at 37° C. For maturation of dendritic cells, the culture is stimulated with 0.2 ng/mL IL-4 and 1000 U/ml GM-CSF at 37° C. for one week. Alternatively, maturation is achieved through incubation with recombinant GM-CSF for a week, or using other methods known in the art. Mouse DCs can be harvested directly from spleens using bead enrichment or differentiated from hematopoietic stem cells. Briefly, bone marrow may be obtained from the femurs of mice. Cells are recovered and red blood cells lysed. Stem cells are cultured in cell culture medium in 20 ng/ml mouse GMCSF for 4 days. Additional medium containing 20 ng/ml mouse GM-CSF is added. On day 6 the medium and non-adherent cells are removed and replaced with fresh cell culture medium containing 20 ng/ml GMCSF. A final addition of cell culture medium with 20 ng/ml GM-CSF is added on day 7. On day 10, non-adherent cells are harvested and seeded into cell culture plates overnight and stimulated as required. Dendritic cells are then treated with various doses of smEVs with or without antibiotics. For example, 25-75 ug/mL smEVs for 24 hours with antibiotics. smEV compositions tested may include smEVs from a single bacterial species or strain, or a mixture of smEVs from one or more genus, 1 or more species, or 1 or more strains (e.g., one or more strains within one species). PBS is included as a negative control and LPS, anti-CD40 antibodies, and/or smEVs from Bifidobacterium spp. are used as positive controls. Following incubation, DCs are stained with anti CD11b, CD11c, CD103, CD8a, CD40, CD80, CD83, CD86, MHCI and MHCII, and analyzed by flow cytometry. DCs that are significantly increased in CD40, CD80, CD83, and CD86 as compared to negative controls are considered to be activated by the associated bacterial smEV composition. These experiments are repeated three times at minimum.


To screen for the ability of smEV-activated epithelial cells to stimulate DCs, the above protocol is followed with the addition of a 24-hour epithelial cell smEV co-culture prior to incubation with DCs. Epithelial cells are washed after incubation with smEVs and are then co-cultured with DCs in an absence of smEVs for 24 hours before being processed as above. Epithelial cell lines may include Int407, HEL293, HT29, T84 and CACO2.


As an additional measure of DC activation, 100 μl of culture supernatant is removed from wells following 24-hour incubation of DCs with smEVs or smEV-treated epithelial cells and is analyzed for secreted cytokines, chemokines, and growth factors using the multiplexed Luminex Magpix. Kit (EMD Millipore, Darmstadt, Germany). Briefly, the wells are pre-wet with buffer, and 25 μl of 1× antibody-coated magnetic beads are added and 2×200 μl of wash buffer are performed in every well using the magnet. 50 μl of Incubation buffer, 50 μl of diluent and 50 μl of samples are added and mixed via shaking for 2 hrs at room temperature in the dark. The beads are then washed twice with 200 μl wash buffer. 100 μl of 1× biotinylated detector antibody is added and the suspension is incubated for 1 hour with shaking in the dark. Two, 200 μl washes are then performed with wash buffer. 100 μl of 1× SAV-RPE reagent is added to each well and is incubated for 30 min at RT in the dark. Three 200 μl washes are performed and 125 μl of wash buffer is added with 2-3 min shaking occurs. The wells are then submitted for analysis in the Luminex xMAP system.


Standards allow for careful quantitation of the cytokines including GM-CSF, IFN-g, IFN-a, IFN-B, IL-1a, IL-1B, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-13, IL-12 (p40/p70), IL-17A, IL-17F, IL-21, IL-22 IL-23, IL-25, IP-10, KC, MCP-1, MIG, MIP1a, TNFa, and VEGF. These cytokines are assessed in samples of both mouse and human origin. Increases in these cytokines in the bacterial treated samples indicate enhanced production of proteins and cytokines from the host. Other variations on this assay examining specific cell types ability to release cytokines are assessed by acquiring these cells through sorting methods and are recognized by one of ordinary skill in the art. Furthermore, cytokine mRNA is also assessed to address cytokine release in response to an smEV composition.


This DC stimulation protocol may be repeated using combinations of purified smEVs and live bacterial strains to maximize immune stimulation potential.


Example 43
In Vitro Screening of smEVs for Enhanced Activation of CD8+ T Cell Killing when Incubated with Tumor Cells

In vitro methods for screening smEVs that can activate CD8+ T cell killing of tumor cells are described. Briefly, DCs are isolated from human PBMCs or mouse spleens, using techniques known in the art, and incubated in vitro with single-strain smEVs, mixtures of smEVs, and/or appropriate controls. In addition, CD8+ T cells are obtained from human PBMCs or mouse spleens using techniques known in the art, for example the magnetic bead-based Mouse CD8a+ T Cell Isolation Kit and the magnetic bead-based Human CD8+ T Cell Isolation Kit (both from Miltenyi Biotech, Cambridge, Mass.). After incubation of DCs with smEVs for some time (e.g., for 24-hours), or incubation of DCs with smEV-stimulated epithelial cells, smEVs are removed from the cell culture with PBS washes and 100 ul of fresh media with antibiotics is added to each well, and 200,000 T cells are added to each experimental well in the 96-well plate. Anti-CD3 antibody is added at a final concentration of 2 ug/ml. Co-cultures are then allowed to incubate at 37° C. for 96 hours under normal oxygen conditions.


For example, approximately 72 hours into the coculture incubation, tumor cells are plated for use in the assay using techniques known in the art. For example, 50,000 tumor cells/well are plated per well in new 96-well plates. Mouse tumor cell lines used may include B16.F10, SIY+B16.F10, and others. Human tumor cell lines are HLA-matched to donor, and can include PANC-1, UNKPC960/961, UNKC, and HELA cell lines. After completion of the 96-hour co-culture, 100 μl of the CD8+ T cell and DC mixture is transferred to wells containing tumor cells. Plates are incubated for 24 hours at 37° C. under normal oxygen conditions. Staurospaurine may be used as negative control to account for cell death.


Following this incubation, flow cytometry is used to measure tumor cell death and characterize immune cell phenotype. Briefly, tumor cells are stained with viability dye. FACS analysis is used to gate specifically on tumor cells and measure the percentage of dead (killed) tumor cells. Data are also displayed as the absolute number of dead tumor cells per well. Cytotoxic CD8+ T cell phenotype may be characterized by the following methods: a) concentration of supernatant granzyme B, IFNy and TNFa in the culture supernatant as described below, b) CD8+ T cell surface expression of activation markers such as DC69, CD25, CD154, PD-1, gamma/delta TCR, Foxp3, T-bet, granzyme B, c) intracellular cytokine staining of IFNy, granzyme B, TNFa in CD8+ T cells. CD4+ T cell phenotype may also be assessed by intracellular cytokine staining in addition to supernatant cytokine concentration including INFy, TNFa, IL-12, IL-4, IL-5, IL-17, IL-10, chemokines etc.


As an additional measure of CD8+ T cell activation, 100 μl of culture supernatant is removed from wells following the 96-hour incubation of T cells with DCs and is analyzed for secreted cytokines, chemokines, and growth factors using the multiplexed Luminex Magpix. Kit (EMD Millipore, Darmstadt, Germany). Briefly, the wells are pre-wet with buffer, and 25 μl of 1× antibody-coated magnetic beads are added and 2×200 μl of wash buffer are performed in every well using the magnet. 50 μl of Incubation buffer, 50 μl of diluent and 50 μl of samples are added and mixed via shaking for 2 hrs at room temperature in the dark. The beads are then washed twice with 200 μl wash buffer. 100 μl of 1× biotinylated detector antibody is added and the suspension is incubated for 1 hour with shaking in the dark. Two, 200 μl washes are then performed with wash buffer. 100 μl of 1× SAV-RPE reagent is added to each well and is incubated for 30 min at RT in the dark. Three 200 μl washes are performed and 125 μl of wash buffer is added with 2-3 min shaking occurs. The wells are then submitted for analysis in the Luminex xMAP system.


Standards allow for careful quantitation of the cytokines including GM-CSF, IFN-g, IFN-a, IFN-B IL-la, IL-1B, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-13, IL-12 (p40/p70), IL-17, IL-23, IP-10, KC, MCP-1, MIG, MIP1a, TNFa, and VEGF. These cytokines are assessed in samples of both mouse and human origin. Increases in these cytokines in the bacterial treated samples indicate enhanced production of proteins and cytokines from the host. Other variations on this assay examining specific cell types ability to release cytokines are assessed by acquiring these cells through sorting methods and are recognized by one of ordinary skill in the art. Furthermore, cytokine mRNA is also assessed to address cytokine release in response to an smEV composition. These changes in the cells of the host stimulate an immune response similarly to in vivo response in a cancer microenvironment.


This CD8+ T cell stimulation protocol may be repeated using combinations of purified smEVs and live bacterial strains to maximize immune stimulation potential.


Example 44
In Vitro Screening of smEVs for Enhanced Tumor Cell Killing by PBMCs

Various methods may be used to screen smEVs for the ability to stimulate PBMCs, which in turn activate CD8+ T cells to kill tumor cells. For example, PBMCs are isolated from heparinized venous blood from healthy human donors by ficoll-paque gradient centrifugation for mouse or human blood, or with Lympholyte Cell Separation Media (Cedarlane Labs, Ontario, Canada) from mouse blood. PBMCs are incubated with single-strain smEVs, mixtures of smEVs, and appropriate controls. In addition, CD8+ T cells are obtained from human PBMCs or mouse spleens. After the 24-hour incubation of PBMCs with smEVs, smEVs are removed from the cells using PBS washes. 100 ul of fresh media with antibiotics is added to each well. An appropriate number of T cells (e.g., 200,000 T cells) are added to each experimental well in the 96-well plate. Anti-CD3 antibody is added at a final concentration of 2 ug/ml. Co-cultures are then allowed to incubate at 37° C. for 96 hours under normal oxygen conditions.


For example, 72 hours into the coculture incubation, 50,000 tumor cells/well are plated per well in new 96-well plates. Mouse tumor cell lines used include B16.F10, SIY+B16.F10, and others. Human tumor cell lines are HLA-matched to donor, and can include PANC-1, UNKPC960/961, UNKC, and HELA cell lines. After completion of the 96-hour co-culture, 100 μl of the CD8+ T cell and PBMC mixture is transferred to wells containing tumor cells. Plates are incubated for 24 hours at 37° C. under normal oxygen conditions. Staurospaurine is used as negative control to account for cell death.


Following this incubation, flow cytometry is used to measure tumor cell death and characterize immune cell phenotype. Briefly, tumor cells are stained with viability dye. FACS analysis is used to gate specifically on tumor cells and measure the percentage of dead (killed) tumor cells. Data are also displayed as the absolute number of dead tumor cells per well. Cytotoxic CD8+ T cell phenotype may be characterized by the following methods: a) concentration of supernatant granzyme B, IFNy and TNFa in the culture supernatant as described below, b) CD8+ T cell surface expression of activation markers such as DC69, CD25, CD154, PD-1, gamma/delta TCR, Foxp3, T-bet, granzyme B, c) intracellular cytokine staining of IFNy, granzyme B, TNFa in CD8+ T cells. CD4+ T cell phenotype may also be assessed by intracellular cytokine staining in addition to supernatant cytokine concentration including INFy, TNFa, IL-12, IL-4, IL-5, IL-17, IL-10, chemokines etc.


As an additional measure of CD8+ T cell activation, 100 μl of culture supernatant is removed from wells following the 96-hour incubation of T cells with DCs and is analyzed for secreted cytokines, chemokines, and growth factors using the multiplexed Luminex Magpix. Kit (EMD Millipore, Darmstadt, Germany). Briefly, the wells are pre-wet with buffer, and 25 μl of 1× antibody-coated magnetic beads are added and 2×200 μl of wash buffer are performed in every well using the magnet. 50 μl of Incubation buffer, 50 μl of diluent and 50 μl of samples are added and mixed via shaking for 2 hrs at room temperature in the dark. The beads are then washed twice with 200 μl wash buffer. 100 μl of 1× biotinylated detector antibody is added and the suspension is incubated for 1 hour with shaking in the dark. Two, 200 μl washes are then performed with wash buffer. 100 μl of 1× SAV-RPE reagent is added to each well and is incubated for 30 min at RT in the dark. Three 200 μl washes are performed and 125 μl of wash buffer is added with 2-3 min shaking occurs. The wells are then submitted for analysis in the Luminex xMAP system.


Standards allow for careful quantitation of the cytokines including GM-CSF, IFN-g, IFN-a, IFN-B IL-1a, IL-1B, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-13, IL-12 (p40/p70), IL-17, IL-23, IP-10, KC, MCP-1, MIG, MIP1a, TNFa, and VEGF. These cytokines are assessed in samples of both mouse and human origin. Increases in these cytokines in the bacterial treated samples indicate enhanced production of proteins and cytokines from the host. Other variations on this assay examining specific cell types ability to release cytokines are assessed by acquiring these cells through sorting methods and are recognized by one of ordinary skill in the art. Furthermore, cytokine mRNA is also assessed to address cytokine release in response to an smEV composition. These changes in the cells of the host stimulate an immune response similarly to in vivo response in a cancer microenvironment.


This PBMC stimulation protocol may be repeated using combinations of purified smEVs with or without combinations of live, dead, or inactivated/weakened bacterial strains to maximize immune stimulation potential.


Example 45
In Vitro Detection of smEVs in Antigen-Presenting Cells

Dendritic cells in the lamina propria constantly sample live bacteria, dead bacteria, and microbial products in the gut lumen by extending their dendrites across the gut epithelium, which is one way that smEVs produced by bacteria in the intestinal lumen may directly stimulate dendritic cells. The following methods represent a way to assess the differential uptake of smEVs by antigen-presenting cells. Optionally, these methods may be applied to assess immunomodulatory behavior of smEVs administered to a patient.


Dendritic cells (DCs) are isolated from human or mouse bone marrow, blood, or spleens according to standard methods or kit protocols (e.g., Inaba K, Swiggard W J, Steinman R M, Romani N, Schuler G, 2001. Isolation of dendritic cells. Current Protocols in Immunology. Chapter 3:Unit3.7).


To evaluate smEV entrance into and/or presence in DCs, 250,000 DCs are seeded on a round cover slip in complete RPMI-1640 medium and are then incubated with smEVs from single bacterial strains or combinations smEVs at various ratios. Purified smEVs may be labeled with fluorochromes or fluorescent proteins. After incubation for various timepoints (e.g., 1 hour, 2 hours), the cells are washed twice with ice-cold PBS and detached from the plate using trypsin. Cells are either allowed to remain intact or are lysed. Samples are then processed for flow cytometry. Total internalized smEVs are quantified from lysed samples, and percentage of cells that uptake smEVs is measured by counting fluorescent cells. The methods described above may also be performed in substantially the same manner using macrophages or epithelial cell lines (obtained from the ATCC) in place of DCs.


Example 46
In Vitro Screening of smEVs with an Enhanced Ability to Activate NK Cell Killing when Incubated with Target Cells

To demonstrate the ability of the selected smEV compositions to elicit potent NK cell cytotoxicity to tumor cells, the following in vitro assay is used. Briefly, mononuclear cells from heparinized blood are obtained from healthy human donors. Optionally, an expansion step to increase the numbers of NK cells is performed as previously described (e.g., see Somanschi et al., J Vis Exp. 2011; (48):2540). The cells may be adjusted to a concentration of cells/ml in RPMI-1640 medium containing 5% human serum. The PMNC cells are then labeled with appropriate antibodies and NK cells are isolated through FACS as CD3−/CD56+ cells and are ready for the subsequent cytotoxicity assay. Alternatively, NK cells are isolated using the autoMACs instrument and NK cell isolation kit following manufacturer's instructions (Miltenyl Biotec).


NK cells are counted and plated in a 96 well format with 20,000 or more cells per well, and incubated with single-strain smEVs, with or without addition of antigen presenting cells (e.g., monocytes derived from the same donor), smEVs from mixtures of bacterial strains, and appropriate controls. After 5-24 hours incubation of NK cells with smEVs, smEVs are removed from cells with PBS washes, NK cells are resuspended in 10 mL fresh media with antibiotics and are added to 96-well plates containing 20,000 target tumor cells/well. Mouse tumor cell lines used include B16.F10, SIY+B16.F10, and others. Human tumor cell lines are HLA-matched to donor, and can include PANC-1, UNKPC960/961, UNKC, and HELA cell lines. Plates are incubated for 2-24 hours at 37° C. under normal oxygen conditions. Staurospaurine is used as negative control to account for cell death.


Following this incubation, flow cytometry is used to measure tumor cell death using methods known in the art. Briefly, tumor cells are stained with viability dye. FACS analysis is used to gate specifically on tumor cells and measure the percentage of dead (killed) tumor cells. Data are also displayed as the absolute number of dead tumor cells per well.


This NK cell stimulation protocol may be repeated using combinations of purified smEVs and live bacterial strains to maximize immune stimulation potential.


Example 47
Using In Vitro Immune Activation Assays to Predict In Vivo Cancer Immunotherapy Efficacy of smEV Compositions

In vitro immune activation assays identify smEVs that are able to stimulate dendritic cells, which in turn activate CD8+ T cell killing. Therefore, the in vitro assays described above are used as a predictive screen of a large number of candidate smEVs for potential immunotherapy activity. smEVs that display enhanced stimulation of dendritic cells, enhanced stimulation of CD8+ T cell killing, enhanced stimulation of PBMC killing, and/or enhanced stimulation of NK cell killing, are preferentially chosen for in vivo cancer immunotherapy efficacy studies.


Example 48
Determining the Biodistribution of smEVs when Delivered Orally to Mice

Wild-type mice (e.g., C57BL/6 or BALB/c) are orally inoculated with the smEV composition of interest to determine the in vivo biodistibution profile of purified smEVs. smEVs are labeled to aide in downstream analyses. Alternatively, tumor-bearing mice or mice with some immune disorder (e.g., systemic lupus erythematosus, experimental autoimmune encephalomyelitis, NASH) may be studied for in vivo distribution of smEVs over a given time-course.


Mice can receive a single dose of the smEV (e.g., 25-100 μg) or several doses over a defined time course (25-100 μg). Alternatively, smEVs dosages may be administered based on particle count (e.g., 7e+08 to 6e+11 particles). Mice are housed under specific pathogen-free conditions following approved protocols. Alternatively, mice may be bred and maintained under sterile, germ-free conditions. Blood, stool, and other tissue samples can be taken at appropriate time points.


The mice are humanely sacrificed at various time points (i.e., hours to days) post administration of the smEV compositions, and a full necropsy under sterile conditions is performed. Following standard protocols, lymph nodes, adrenal glands, liver, colon, small intestine, cecum, stomach, spleen, kidneys, bladder, pancreas, heart, skin, lungs, brain, and other tissue of interest are harvested and are used directly or snap frozen for further testing. The tissue samples are dissected and homogenized to prepare single-cell suspensions following standard protocols known to one skilled in the art. The number of smEVs present in the sample is then quantified through flow cytometry. Quantification may also proceed with use of fluorescence microscopy after appropriate processing of whole mouse tissue (Vankelecom H., Fixation and paraffin-embedding of mouse tissues for GFP visualization, Cold Spring Harb. Proloc., 2009). Alternatively, the animals may be analyzed using live-imaging according to the smEV labeling technique.


Biodistribution may be performed in mouse models of cancer such as but not limited to CT-26 and B16 (see, e.g., Kim et al., Nature Communications vol. 8, no. 626 (2017)) or autoimmunity such as but not limited to EAE and DTH (see, e.g., Turjeman et al., PLoS One 10(7): e0130442 (20105).


Example 49
Manufacturing Conditions

Enriched media is used to grow and prepare the bacteria for in vitro and in vivo use and, ultimately, for pmEV and smEV preparations. For example, media may contain sugar, yeast extracts, plant-based peptones, buffers, salts, trace elements, surfactants, anti-foaming agents, and vitamins. Composition of complex components such as yeast extracts and peptones may be undefined or partially defined (including approximate concentrations of amino acids, sugars etc.). Microbial metabolism may be dependent on the availability of resources such as carbon and nitrogen. Various sugars or other carbon sources may be tested. Alternatively, media may be prepared and the selected bacterium grown as shown by Saarela et al., J. Applied Microbiology. 2005. 99: 1330-1339, which is hereby incorporated by reference. Influence of fermentation time, cryoprotectant and neutralization of cell concentrate on freeze-drying survival, storage stability, and acid and bile exposure of the selected bacterium produced without milk-based ingredients.


At large scale, the media is sterilized. Sterilization may be accomplished by Ultra High Temperature (UHT) processing. The UHT processing is performed at very high temperature for short periods of time. The UHT range may be from 135-180° C. For example, the medium may be sterilized from between 10 to 30 seconds at 135° C.


Inoculum can be prepared in flasks or in smaller bioreactors and growth is monitored. For example, the inoculum size may be between approximately 0.5 and 3% of the total bioreactor volume. Depending on the application and need for material, bioreactor volume can be at least 2 L, 10 L, 80 L, 100 L, 250 L, 1000 L, 2500 L, 5000 L, 10,000 L.


Before the inoculation, the bioreactor is prepared with medium at desired pH, temperature, and oxygen concentration. The initial pH of the culture medium may be different that the process set-point. pH stress may be detrimental at low cell centration; the initial pH could be between pH 7.5 and the process set-point. For example, pH may be set between 4.5 and 8.0. During the fermentation, the pH can be controlled through the use of sodium hydroxide, potassium hydroxide, or ammonium hydroxide. The temperature may be controlled from 25° C. to 45° C., for example at 37° C. Anaerobic conditions are created by reducing the level of oxygen in the culture broth from around 8 mg/L to 0 mg/L. For example, nitrogen or gas mixtures (N2, CO2, and H2) may be used in order to establish anaerobic conditions. Alternatively, no gases are used and anaerobic conditions are established by cells consuming remaining oxygen from the medium. Depending on strain and inoculum size, the bioreactor fermentation time can vary. For example, fermentation time can vary from approximately 5 hours to 48 hours.


Reviving microbes from a frozen state may require special considerations. Production medium may stress cells after a thaw; a specific thaw medium may be required to consistently start a seed train from thawed material. The kinetics of transfer or passage of seed material to fresh medium, for the purposes of increasing the seed volume or maintaining the microbial growth state, may be influenced by the current state of the microbes (ex. exponential growth, stationary growth, unstressed, stressed).


Inoculation of the production fermenter(s) can impact growth kinetics and cellular activity. The initial state of the bioreactor system must be optimized to facilitate successful and consistent production. The fraction of seed culture to total medium (e.g., a percentage) has a dramatic impact on growth kinetics. The range may be 1-5% of the fermenter's working volume. The initial pH of the culture medium may be different from the process set-point. pH stress may be detrimental at low cell concentration; the initial pH may be between pH 7.5 and the process set-point. Agitation and gas flow into the system during inoculation may be different from the process set-points. Physical and chemical stresses due to both conditions may be detrimental at low cell concentration.


Process conditions and control settings may influence the kinetics of microbial growth and cellular activity. Shifts in process conditions may change membrane composition, production of metabolites, growth rate, cellular stress, etc. Optimal temperature range for growth may vary with strain. The range may be 20-40° C. Optimal pH for cell growth and performance of downstream activity may vary with strain. The range may be pH 5-8. Gasses dissolved in the medium may be used by cells for metabolism. Adjusting concentrations of O2, CO2, and N2 throughout the process may be required. Availability of nutrients may shift cellular growth. Microbes may have alternate kinetics when excess nutrients are available.


The state of microbes at the end of a fermentation and during harvesting may impact cell survival and activity. Microbes may be preconditioned shortly before harvest to better prepare them for the physical and chemical stresses involved in separation and downstream processing. A change in temperature (often reducing to 20-5° C.) may reduce cellular metabolism, slowing growth (and/or death) and physiological change when removed from the fermenter. Effectiveness of centrifugal concentration may be influenced by culture pH. Raising pH by 1-2 points can improve effectiveness of concentration but can also be detrimental to cells. Microbes may be stressed shortly before harvest by increasing the concentration of salts and/or sugars in the medium. Cells stressed in this way may better survive freezing and lyophilization during downstream.


Separation methods and technology may impact how efficiently microbes are separated from the culture medium. Solids may be removed using centrifugation techniques. Effectiveness of centrifugal concentration can be influenced by culture pH or by the use of flocculating agents. Raising pH by 1-2 points may improve effectiveness of concentration but can also be detrimental to cells. Microbes may be stressed shortly before harvest by increasing the concentration of salts and/or sugars in the medium. Cells stressed in this way may better survive freezing and lyophilization during downstream. Additionally, Microbes may also be separated via filtration. Filtration is superior to centrifugation techniques for purification if the cells require excessive g-minutes to successfully centrifuge. Excipients can be added before after separation. Excipients can be added for cryo protection or for protection during lyophilization. Excipients can include, but are not limited to, sucrose, trehalose, or lactose, and these may be alternatively mixed with buffer and anti-oxidants. Prior to lyophilization, droplets of cell pellets mixed with excipients are submerged in liquid nitrogen.


Harvesting can be performed by continuous centrifugation. Product may be resuspended with various excipients to a desired final concentration. Excipients can be added for cryo protection or for protection during lyophilization. Excipients can include, but are not limited to, sucrose, trehalose, or lactose, and these may be alternatively mixed with buffer and anti-oxidants. Prior to lyophilization, droplets of cell pellets mixed with excipients are submerged in liquid nitrogen.


Lyophilization of material, including live bacteria, vesicles, or other bacterial derivative includes a freezing, primary drying, and secondary drying phase. Lyophilization begins with freezing. The product material may or may not be mixed with a lyoprotectant or stabilizer prior to the freezing stage. A product may be frozen prior to the loading of the lyophilizer, or under controlled conditions on the shelf of the lyophilizer. During the next phase, the primary drying phase, ice is removed via sublimation. Here, a vacuum is generated and an appropriate amount of heat is supplied to the material. The ice will sublime while keeping the product temperature below freezing, and below the material's critical temperature (Tc). The temperature of the shelf on which the material is loaded and the chamber vacuum can be manipulated to achieve the desired product temperature. During the secondary drying phase, product-bound water molecules are removed. Here, the temperature is generally raised higher than in the primary drying phase to break any physico-chemical interactions that have formed between the water molecules and the product material. After the freeze-drying process is complete, the chamber may be filled with an inert gas, such as nitrogen. The product may be sealed within the freeze dryer under dry conditions, in a glass vial or other similar container, preventing exposure to atmospheric water and contaminates.


Example 50
Oral Prevotella Histicola and Veillonella Parvula smEVs and pmEVs: DTH Studies

I. Female 5 week old C57BL/6 mice were purchased from Taconic Biosciences and acclimated at a vivarium for one week. Mice were primed with an emulsion of KLH and CFA (1:1) by subcutaneous immunization on day 0. Mice were orally gavaged daily with pmEVs or powder of whole microbe of the indicated strain or dosed intraperitoneally with dexamethasone at 1 mg/kg from days 1-8. After dosing on day 8, mice were anaesthetized with isoflurane, left ears were measured for baseline measurements with Fowler calipers and the mice were challenged intradermally with KLH in saline (10 μl) in the left ear and ear thickness measurements were taken at 24 hours.


The 24 hour ear measurement results are shown in FIG. 21. The efficacy of P. histicola pmEVs at three doses (high: 6.0E+11, mid: 6.0E+09 and low: 6.0E+07) was tested in comparison to lyophilized P. histicola pmEVs at the same doses and to 10 mg of powder (with total cell count 3.13E+09). The results show that the high dose of pmEVs displayed comparable efficacy to the 10 mg dose of powder. The efficacy of P. histicola pmEVs is not affected by lyophilization.


II. Female 5 week old C57BL/6 mice were purchased from Taconic Biosciences and acclimated at a vivarium for one week. Mice were primed with an emulsion of KLH and CFA (1:1) by subcutaneous immunization on day 0. Mice were orally gavaged daily with smEVs, pmEVs, gamma irradiated (GI) pmEVs, or gamma irradiated (GI) powder (of whole microbe) of the indicated strain or dosed intraperitoneally with dexamethasone at 1 mg/kg from days 1-8. After dosing on day 8, mice were anaesthetized with isoflurane, left ears were measured for baseline measurements with Fowler calipers and the mice were challenged intradermally with KLH in saline (10 μl) in the left ear and ear thickness measurements were taken at 24 hours.


The 24 hour ear measurement results are shown in FIG. 22. The efficacy of V. parvula smEVs, pmEVs and gamma-irradiated (GI) pmEVs were tested head-to-head at three doses (high: 3.0E+11, mid: 3.0E+09 and low: 3.0E+07). There was not a significant difference between the highest dose of each group. V. parvula pmEVs, both gamma-irradiated and non-gamma-irradiated, are just as efficacious as smEVs.


Example 51
smEV and pmEV Preparation

For the studies described in Example 50, the smEVs and pmEVs were prepared as follows.


smEVs: Downstream processing of smEVs began immediately following harvest of the bioreactor. Centrifugation at 20,000 g was used to remove the cells from the broth. The resulting supernatant was clarified using 0.22 μm filter. The smEVs were concentrated and washed using tangential flow filtration (TFF) with flat sheet cassettes ultrafiltration (UF) membranes with 100 kDa molecular weight cutoff (MWCO). Diafiltration (DF) was used to washout small molecules and small proteins using 5 volumes of phosphate buffer solution (PBS). The retentate from TFF was spun down in an ultracentrifuge at 200,000 g for 1 hour to form a pellet rich in smEVs called a high-speed pellet (HSP). The pellet was resuspended with minimal PBS and a gradient was prepared with Optiprep™ density gradient medium and ultracentrifuged at 200,000 g for 16 hours. Of the resulting fractions, 2 middle bands contained smEVs. The fractions were washed with 15 fold PBS and the smEVs spun down at 200,000 g for 1 hr to create the fractionated HSP or fHSP. It was subsequently resuspended with minimal PBS, pooled, and analyzed for particles per mL and protein content. Dosing was prepared from the particle/mL count to achieve desired concentration. The smEVs were characterized using a NanoSight NS300 by Malvern Panalytical in scatter mode using the 532 nm laser.



Prevotella Histicola pmEVs:


Cell pellets were removed from freezer and placed on ice. Pellet weights were noted.


Cold 100 mM Tris-HCl pH 7.5 was added to the frozen pellets and the pellets were thawed rotating at 4° C.


10 mg/ml DNase stock was added to the thawed pellets to a final concentration of 1 mg/mL.


The pellets were incubated on the inverter for 40 min at RT (room temperature).


The sample was filtered in a 70 um cell strainer before running through the Emulsiflex.


The samples were lysed using the Emulsiflex with 8 discrete cycles at 22,000 psi.


To remove the cellular debris from the lysed sample, the sample was centrifuged at 12,500×g, 15 min, 4° C.


The sample was centrifuged two additional times at 12,500×g, 15 min, 4° C., each time moving the supernatant to a fresh tube.


To pellet the membrane proteins, the sample was centrifuged at 120,000×g, 1 hr, 4° C.


The pellet was resuspended in 10 mL ice-cold 0.1 M sodium carbonate pH 11. The sample was incubated on the inverter at 4° C. for 1 hour.


The sample was centrifuged at 120,000×g, 1 hr, 4° C.


10 mL 100 mM Tris-HCl pH 7.5 was added to pellet and incubate O/N (overnight) at 4° C.


The pellet was resuspended and the sample was centrifuged at 120,000×g for 1 hour at 4° C.


The supernatant was discarded and the pellet was resuspended in a minimal volume of PBS.



Veillonella Parvula pmEVs:


The V. parvula pmEVs used in the studies in Example 50 came from three different isolations (isolations 1, 2 and 3). There were small variations in protocol.


Cell pellets were removed from freezer and place on ice. Pellet weights were noted.


Cold MP Buffer (100 mM Tris-HCl pH 7.5) was added to the frozen pellets and the pellets were thawed rotating at RT.


10 mg/ml DNase stock was added to the thawed pellets from isolations 1 and 2 to a final concentration of 1 mg/mL and incubate. The pellets were incubated an additional 40′ on the inverter.


The samples were lysed using the Emulsiflex with 8 discrete cycles at 20,000-30,000 psi.


For isolations 1 and 2, the samples were filtered in a 70 um cell strainer before running through the Emulsiflex to remove clumps.


For isolation 3, 1 mM PMSF (Phenylmethylsulfonyl fluoride, Sigma) and 1 mM Benzamidine (Sigma) were added immediately prior to passage through the Emulsiflex and the sample was first cycled through the Emulsiflex continuously for 1.5 minutes at 15,000 psi to break up large clumps.


To remove the cellular debris from the cell lysate, the samples were centrifuged at 12,500×g, 15 min, 4° C.


The supernatant from isolation 3 was centrifuged one additional time while the supernatants from isolations 1 and 2 were cycled two additional times at 12,500×g, 15 min, 4° C. After each centrifugation the supernatant was moved to a fresh tube.


The final supernatant was centrifuged 120,000×g, 1 hr, 4° C.


The membrane pellet was resuspended in 10 mL ice-cold 0.1 M sodium carbonate pH 11. For isolations 1 and 2, the samples were incubated in sodium carbonate for 1 hour prior to high speed spin.


The samples were spun at 120,000×g, 1 hr, 4° C.


10 mL 100 mM Tris-HCl pH 7.5 was added to the pellet and the pellet was resuspended.


The sample was centrifuged at 120,000×g for 1 hour at 4° C.


The supernatant was discarded and the pellets were in a minimal volume of in PBS (isolations 1 and 2) or PBS containing 250 mM sucrose (isolation 3).


Dosing pmEVs was based on particle counts, as assessed by Nanoparticle Tracking Analysis (NTA) using a NanoSight NS300 (Malvern Panalytical) according to manufacturer instructions. Counts for each sample were based on at least three videos of 30 sec duration each, counting 40-140 particles per frame.


Gamma irradiation: For gamma irradiation, V. parvula pmEVs were prepared in frozen form and gamma irradiated on dry ice at 25 kGy radiation dose; V. parvula whole microbe lyophilized powder was gamma irradiated at ambient temperature at 17.5 kGy radiation dose.


Lyophilization: Samples were placed in lyophilization equipment and frozen at −45° C. The lyophilization cycle included a hold step at −45° C. for 10 min. The vacuum began and was set to 100 mTorr and the sample was held at −45° C. for another 10 min. Primary drying began with a temperature ramp to −25° C. over 300 minutes and it was held at this temperature for 4630 min. Secondary drying started with a temperature ramp to 20° C. over 200 min while the vacuum was decreased to 20 mTorr. It was held at this temperature and pressure for 1200 min. The final step increased the temperature from 20 to 25° C. where it remained at a vacuum of 20 mTorr for 10 min.


Example 52
smEV Isolation and Enumeration

The equipment used in smEV isolation includes a Sorvall RC-5C centrifuge with SLA-3000 rotor; an Optima XE-90 Ultracentrifuge by Beckman-Coulter 45Ti rotor; a Sorvall wX+ Ultra Series Centrifuge by Thermo Scientific; and a Fiberlite F37L-8×100 rotor.


Microbial Supernatant Collection and Filtration

Microbes must be pelleted and filtered away from supernatant in order to recover smEVs and not microbes.


Pellet microbial culture is generated by using a Sorvall RC-5C centrifuge with the SLA-3000 rotor and centrifuge culture for a minimum of 15 min at a minimum of 7,000 rpm. And then decanting the supernatant into new and sterile container.


The supernatant is filtered through a 0.2 um filter. For supernatants with poor filterability (less than 300 ml of supernatant pass through filter) a 0.45 um capsule filter is attached ahead of the 0.2 um vacuum filter. The filtered supernatant is stored atat 4° C. The filtered supernatant can then be concentrated using TFF.


Isolation of smEVs Using Ultracentrifugation


Concentrated supernatant is centrifuged in the ultracentrifuge to pellet smEVs and isolate the smEVs from smaller biomolecules. The speed is for 200,000 g, time for 1 hour, and temperature at 4° C. When rotor has stopped, tubes are removed from the ultracentrifuge and the supernatant is gently poured off. More supernatant is added the tubes are centrifuged again. After all concentrated supernatant has been centrifuged, the pellets generated are referred to as ‘crude’ smEV pellets. Sterile 1× PBS is added to pellets, which are placed in a container. The container is placed on a shaker set at speed 70, in a 4° C. fridge overnight or longer. The smEV pellets are resuspended with additional sterile 1× PBS. The resuspended crude EV samples are stored at 4° C. or at −80° C.


smEV Purification Using Density Gradients


Density gradients are used for smEV purification. During ultracentrifugation, particles in the sample will move, and separate, within the graded density medium based on their ‘buoyant’ densities. In this way smEVs are separated from other particles, such as sugars, lipids, or other proteins, in the sample.


For smEV purification, four different percentages of the density medium (60% Optiprep) are used, a 45% layer, a 35% layer, a 25%, and a 15% layer. This will create the graded layers. A 0% layer is added at the top consisting of sterile 1× PBS. The 45% gradient layer should contain the crude smEV sample. 5 ml of sample is added to 15 ml of Optiprep. If crude smEV sample is less than 5 ml, bring up to volume using sterile 1× PBS.


Using a serological pipette, the 45% gradient mixture is pipetted up and down to mix. The sample is then pipetted into a labeled clean and sterile ultracentrifuge tube. Next, a 10 ml serological pipette is used to slowly add 13 ml of 35% gradient mixture. Next 13 ml of the 25% gradient mixture is added, followed by 13 ml of the 15% mixture and finally 6 ml of sterile 1× PBS. The ultracentrifuge tubes are balanced with sterile 1× PBS. The gradients are carefully placed in a rotor and the ultracentrifuge is set for for 200,000 g and 4° C. The gradients are centrifuged for a minimum of 16 hours.


A clean pipette is used to remove fraction(s) of interest, which are added to 15 ml conical tube. These ‘purified’ smEV samples are kept at 4° C.


In order to clean and remove residual optiprep from smEVs, 10× volume of PBS are added to purified smEVs. The ultracentrifuge is set for 200,000 g and 4° C. Centrifuge and spun for 1 hour. The tubes are carefully removed from ultracentrifuge and the supernatant decanted. The purified EVs are washed until all sample has been pelleted. 1× PBS is added to the purified pellets, which are placed in a container. The container is placed on a shaker set at speed 70 in a 4° C. fridge overnight or longer. The ‘purified’ smEV pellets are resuspended with additional sterile 1× PBS. The resuspended purified smEV samples are stored at 4° C. or at −80° C.


Example 53
KLH DTH Study

Female 5 week old C57BL/6 mice were purchased from Taconic Biosciences and acclimated at a vivarium for one week. Mice were primed with an emulsion of KLH and CFA (1:1) by subcutaneous immunization on day 0. Mice were orally gavaged daily with smEVs or dosed intraperitoneally with dexamethasone at 1 mg/kg from days 1-8. After dosing on day 8, mice were anaesthetized with isoflurane, left ears were measured for baseline measurements with Fowler calipers and the mice were challenged intradermally with KLH in saline (10 μl) in the left ear and ear thickness measurements were taken at 24 hours. Dose was determined by particle count by NTA.


The 24 hour ear measurement results are shown in FIG. 23. smEVs made from Megasphaera Sp. Strain A were compared at two doses, 2E+11 and 2E+07 (based on particles per dose). The smEVs were efficacious, showing decreased ear inflammation 24 hours after challenge.


The 24 hour ear measurement results are shown in FIG. 24. smEVs made from Megasphaera Sp. Strain B were compared at two doses, 2E+11 and 2E+07 (based on particles per dose). The smEVs were efficacious, showing decreased ear inflammation 24 hours after challenge.


The 24 hour ear measurement results are shown in FIG. 25. smEVs made from Selenomonas felix were compared at two doses, 2E+11 and 2E+07 (based on particles per dose). The smEVs were efficacious, showing decreased ear inflammation 24 hours after challenge.


Example 54
smEV and Gamma-Irradiated Whole Bacterium U937 Testing Protocol

Cell line preparation: The U937 Monocyte cell line (ATCC) was propagated in RPM1 medium with added FBS HEPES, sodium pyruvate, and antibiotic. at 37° C. with 5% CO2. Cells were enumerated using a cellometer with live/dead staining to determine viability. Next, Cells were diluted to a concentration of 5×105 cells per ml in RPMI medium with 20 nM phorbol-12-myristate-13-acetate (PMA) to differentiate the monocytes into macrophage-like cells. Next, 200 microliters of cell suspension was added to each well of a 96-well plate and incubated 37° C. with 5% CO2 for 72 hrs. The adherent, differentiated cells were washed and incubated in fresh medium without PMA for 24 hrs before experimentation.


Experimental Setup: smEVs were diluted to the appropriate concentration in RPMI medium without antibiotics (typically)1×105-1×1010). Treatment-free and TLR 2 and 4 agonist control samples were also prepared. The 96-well plate containing the differentiated U937 cells was washed with fresh medium without antibiotics, to remove residual antibiotics. Next, the suspension of smEVs was added to the washed plate. The plate was incubated for 24 hrs at 37° C. with 5% CO2.


Experimental Endpoints: After 24 hrs of coincubation, the supernatants were removed from the U937 cells into a separate 96-well plate. The cells were observed for any obvious lysis (plaques) in the wells. Two treatment-free wells did not have the supernatants removed and Lysis buffer was added to the wells and incubated at 37° C. for 30 minutes to lyse cells (maximum lysis control). 50 microliters of each supernatant or maximum lysis control was added to a new 96-well plate and cell lysis was determined (CytoTox 96® Non-Radioactive Cytotoxicity Assay, Promega) per manufacturer's instructions. Cytokines were measured from the supernatants using U-plex MSD plates (Meso Scale Discovery) per manufacturer's instructions.


Results are shown in FIG. 26. smEVs from Megasphaera Sp. Strain A induce cytokine production from PMA-differentiated U937 cells. U937 cells were treated with smEV at 1×106-1×109 concentrations as well as TLR2 (FSL) and TLR4 (LPS) agonist controls for 24 hrs and cytokine production was measured. “Blank” indicates the medium control.


Example 55
Oral Delivery of Megasphaera sp. smEVs in CT26 Tumor Studies, First Representative Oncology Study

Female 8 week old BALB/c mice were acquired from Taconic Biosciences and allowed to acclimate at a vivarium for 3 weeks. On Day 0, mice were anesthetized with isoflurane, and inoculated subcutaneously on the left flank with 1.0e5 CT-26 cells (0.1 mL) prepared in PBS and Corning (GFR) Phenol Red-Free Matrigel (1:1). Mice were allowed to rest for 9 days post CT-26 inoculation to allow formation of palpable tumors. On Day 9, tumors were measured using a sliding digital caliper to collect length and width in measurements (in millimeters) to calculate estimated tumor volume ((L×W×W)/2)=TVmm3)). Mice were randomized into different treatment groups with a total of 9 or 10 mice per group. Randomization was done to balance all treatment groups, allowing each group to begin treatment with a similar average tumor volume and standard deviation. Dosing began on Day 10, and ended on Day 22 for 13 consecutive days of dosing. Mice were orally dosed BID with Megasphaera sp. Strain AsmEVs, or Q4D intraperitoneally with 200 ug anti-mouse PD-1 antibody. Body weight and tumor measurements were collected on a MWF (Monday-Wednesday-Friday) schedule. Dose of smEVs was determined by particle count by NTA. Two mice from the Megasphaera sp. smEV group were censored out of the study due to mortality caused by dosing injury.


Results are shown in FIGS. 27A and 27B. The Day 22 Tumor Volume Summary compares Megasphaera sp. smEV (2e11) against a negative control (Vehicle PBS), and positive control (anti-PD-1). Megasphaera sp. smEV (2e11) compared to Vehicle PBS showed statistically significant efficacy and is not significantly different than anti-PD-1. The Tumor Volume Curves show similar growth trends Megasphaera sp. smEVs and anti-PD-1, along with sustained efficacy after 13 days of treatment.


Example 56
Oral Delivery of Megasphaera sp. smEVs in CT26 Tumor Studies, Second Representative Oncology Study

Female 8 week old BALB/c mice were acquired from Taconic Biosciences and allowed to acclimate at a vivarium for 1 week. On Day 0, mice were anesthetized with isoflurane, and inoculated subcutaneously on the left flank with 1.0e5 CT-26 cells (0.1 mL) prepared in PBS and Coming (GFR) Phenol Red-Free Matrigel (1:1). Mice were allowed to rest for 9 days post CT-26 inoculation to allow formation of palpable tumors. On Day 9, tumors were measured using a sliding digital caliper to collect length and width in measurements (in millimeters) to calculate estimated tumor volume ((L×W×W)/2)=TVmm3)). Mice were randomized into different treatment groups with a total of 9 mice per group. Randomization was done to balance all treatment groups, allowing each group to begin treatment with a similar average tumor volume and standard deviation. Dosing began on Day 10, and ended on Day 23 for 14 consecutive days of dosing. Mice were orally dosed BID and QD with Megasphaera sp. Strain A smEVs, or Q4D intraperitoneally with 200 ug anti-mouse PD-1 antibody. Body weight and tumor measurements were collected on a MWF schedule. Dose of smEVs was determined by particle count by NTA.


Results are shown in FIGS. 28A and 28B. The Day 23 Tumor Volume Summary compares Megasphaera sp. smEVs at 3 doses (2e11, 2e9, and 2e7) BID, as well as Megasphaera sp. smEVs (2e11) QD against a negative control (Vehicle PBS), and positive control (anti-PD-1). All Megasphaera sp. smEV treatment groups compared to Vehicle PBS show statistically significant efficacy compared to Vehicle (PBS). All Megasphaera sp. smEV doses tested are not significantly different than anti-PD-1. The Tumor Growth Curve shows sustained efficacy of Megasphaera sp. smEV treatment groups over 14 days of treatment similar to anti-PD-1.


Example 57
Isolation of pmEVs from Enterococcus Gallinarum Strains

pmEVs from both Enterococcus gallinarum strains were prepared as follows: Cold MP Buffer (50 mM Tris-HCl pH 7.5 with 100 mM NaCl) was added to frozen cell pellets and pellets were thawed rotating at RT (room temperature) or 4° C. Cells were lysed on the Emulsiflex. The samples were lysd on the Emulsiflex with 4 discrete passes at 24,000 psi. Immediately prior to lysis a proteinase inhibitors, phenylmethylsulfonyl fluoride (PMSF) and benzamidine were added to the sample to a final concentration of 1 mM each. Debris and unlysed cells were pelleted: 6,000×g, 30 min, 40 C.


pmEVs were purified by FPLC from Low Speed Supernatant (LSS) Setup: A large column (GE ,CK 26/70) packed with Captocore 700 was used for pmEV purification: 70% EtOH for sterilization; 0.1× PBS for running buffer; Milli-Q water for washing; 20% EtOH w/0.1 M NaOH for cleaning and storage. Benzonase was added to LSS sample and incubate at RT for 30 minutes while rotating (Final concentration of 100 U/ml Benzonase and 1 mM MgCl). LSS from bacterial lysis was kept on ice and at 4 C until ready to load into the Superloop.


FPLC purification was run: Flow rate was set to 5 ml/min and set delta column pressure to 0.25 psi. Throughout the purification process, the UV absorbance, pressure, and flow rate were monitored. Run was started and sample (Superloop) was manually loaded. When the sample became visible on the chromatogram (˜50 mAU), the fraction collector was engaged. The entire sample peak was collected.


Final pmEV sample was concentrated: Final pmEV fractions were added to clean ultracentrifuge tubes and balance. Tubes were spun at 120,000×g for 1 hour at 40 C. Supernatant was discarded and pellets were resuspended in a minimal volume of sterile PBS.


Example 58
In Vivo Data Generated with pmEVs

Female 8 week old BALB/c mice were allowed to acclimate at a vivarium for 1 week. On Day 0, mice were anesthetized with isoflurane, and inoculated subcutaneously on the left flank with 1×105 CT-26 cells (0.1 mL) prepared in PBS and Corning (GFR) Phenol Red-Free Matrigel (1:1). Mice were allowed to rest for 9 days post CT-26 inoculation to allow formation of palpable tumors. On Day 9, tumors were measured using a sliding digital caliper to collect length and width in measurements (in millimeters) to calculate estimated tumor volume ((L×W×W)/2)=TVmm3)). Mice were randomized into different treatment groups with a total of (9) mice per group. Randomization was done to balance all treatment groups, allowing begin each group to begin treatment with a similar average tumor volume and standard deviation. Dosing began on Day 10, and ended on Day 23 for 14 consecutive days of dosing. Mice were orally dosed once daily with the Enterococcus gallinarum pmEVs, or Q4D intraperitoneally with 200 μg anti-mouse PD-1. Body weight and tumor measurements were collected on a MWF schedule.


pmEVs were prepared from two strains of Enterococcus gallinarum. One strain was obtained from a JAX mouse; one strain was obtained from a human source. The dose particle count for the pmEVs was 2×1011. The dose was determined as particle count by NTA.



FIG. 29 shows tumor volumes after d10 tumors were dosed once daily for 14 days with pmEVs from E. gallinarum Strain A.


Example 59
Negativicutes U937 Results

To demonstrate the therapeutic utility of the Negativicutes as a class, representatives from each family in Table 5 were selected and EVs were harvested from culture supernatants. The EVs were added to PMA-differentiated U937 cells and incubated for 24 hrs. Cytokine release was measured by MSD ELISA.


The results are shown in FIGS. 30-34. The broad robust stimulation exhibited by each strain's EVs follows a similar profile between strains. TLR2 (FSL) and TLR4 (LPS) agonists were used as controls. Blank indicates the media control.












TABLE 5







Strain Name
Family within Negativicutes Class










Megasphaera sp. Strain A

Veillonellaceae




Megasphaera sp. Strain B

Veillonellaceae




Selenomonas felix

Selenomonadaceae




Acidaminococcus intestini

Acidaminococcaceae




Propionospora sp.

Sporomusaceae










INCORPORATION BY REFERENCE

All publications patent applications mentioned herein are hereby incorporated by reference in their entirety as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference. In case of conflict, the present application, including any definitions herein, will control.


Equivalents

Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims
  • 1. A pharmaceutical composition comprising isolated processed microbial extracellular vesicles (pmEVs).
  • 2. The pharmaceutical composition of claim 1, wherein at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% of the microbial-derived content of the pharmaceutical composition is pmEVs.
  • 3. The pharmaceutical composition of claim 1 or claim 2 for use in the treatment of a disease via immune suppression.
  • 4. The pharmaceutical composition of claim 1 or claim 2 for use in the treatment of a disease via immune activation.
  • 5. The pharmaceutical composition of claim 1 or claim 2 for use in the treatment of a disease via activation or enhancement of one or more immune responses in the subject.
  • 6. The pharmaceutical composition of claim 1 or claim 2 for use in the treatment of a disease via promotion of immune suppression in the subject.
  • 7. The pharmaceutical composition of any one of claims 2 to 6, wherein the disease is a cancer, an autoimmune disease, an inflammatory disease, or a metabolic disease.
  • 8. The pharmaceutical composition of any one of claims 1 to 7, comprising a therapeutically effective amount of the pmEVs.
  • 9. The pharmaceutical composition of any one of claims 1 to 8, wherein the composition activates innate antigen presenting cells.
  • 10. The pharmaceutical composition of any one of claims 1 to 9, wherein the composition has one or more beneficial immune effects outside the gastrointestinal tract when orally administered.
  • 11. The pharmaceutical composition of any one of claims 1 to 10, wherein the composition modulates immune effects outside the gastrointestinal tract in the subject when orally administered.
  • 12. The pharmaceutical composition of any one of claims 1 to 11, wherein the composition comprises pmEVs from one strain of bacteria.
  • 13. The pharmaceutical composition of any one of claims 1 to 12, wherein the pmEVs are lyophilized (e.g., the lyophilized product further comprises a pharmaceutically acceptable excipient).
  • 14. The pharmaceutical composition of any one of claims 1 to 13, wherein the pmEVs are gamma irradiated.
  • 15. The pharmaceutical composition of any one of claims 1 to 14, wherein the pmEVs are UV irradiated.
  • 16. The pharmaceutical composition of any one of claims 1 to 15, wherein the pmEVs are heat inactivated.
  • 17. The pharmaceutical composition of claim 16, wherein the pmEVs are heat inactivated at about 50° C. for two hours or at about 90° C. for two hours.
  • 18. The pharmaceutical composition of any one of claims 1 to 17, wherein the pmEVs are acid treated.
  • 19. The pharmaceutical composition of any one of claims 1 to 18, wherein the pmEVs are oxygen sparge.
  • 20. The pharmaceutical composition of claim 19, wherein the pmEVs are ozygen sparged at about 0.1 vvm for at least two hours.
  • 21. The pharmaceutical composition of any one of claims 1 to 20, wherein the dose of pmEVs is about 2×106 to about 2×1016 particles.
  • 22. The pharmaceutical composition of any one of claims 1 to 21, wherein the dose of pmEVs is about 5 mg to about 900 mg total protein.
  • 23. The pharmaceutical composition of any one of claims 1 to 22, wherein the pharmaceutical composition is a solid dose form.
  • 24. The pharmaceutical composition of claim 23, wherein the solid dose form comprises a tablet, a minitablet, a capsule, a pill, or a powder, or a combination of the foregoing.
  • 25. The pharmaceutical composition of claim 23 or 24, wherein the solid dose form further comprises a pharmaceutically acceptable excipient.
  • 26. The pharmaceutical composition of any one of claims 23 to 25, wherein the solid dose form comprises an enteric coating.
  • 27. The pharmaceutical composition of any one of claims 23 to 26, wherein the solid dose form is formulated for oral administration.
  • 28. The pharmaceutical composition of any one of claims 1 to 22, wherein the pharmaceutical composition is in the form of a suspension.
  • 29. The pharmaceutical composition of claim 28, wherein the suspension is formulated for oral administration.
  • 30. The pharmaceutical composition of claim 29, wherein the suspension comprises PBS, and optionally, sucrose or glucose.
  • 31. The pharmaceutical composition of claim 28, wherein the suspension is formulated for intravenous, intraperitoneal, or intratumoral administration.
  • 32. The pharmaceutical composition of claim 31, wherein the suspension comprises PBS.
  • 33. The pharmaceutical composition of any one of claims 28 to 32, wherein the suspension further comprises a pharmaceutically acceptable excipient or a buffer.
  • 34. The pharmaceutical composition of any one of claims 1 to 33, wherein the pmEvs are from Gram positive bacteria.
  • 35. The pharmaceutical composition of any one of claims 1 to 33, wherein the pmEvs are from Gram negative bacteria.
  • 36. The pharmaceutical composition of claim 35, wherein the Gram negative bacteria belongs to the class Negativicutes.
  • 37. The pharmaceutical composition of any one of claims 1 to 36, wherein the pmEVs are from aerobic bacteria, anaerobic bacteria, acidophile bacteria, alkaliniphile bacteria, neutralophile bacteria, fastidious bacteria, nonfastidiouius bacteria, or a combination thereof.
  • 38. The pharmaceutical composition of any one of claims 1 to 37, wherein the pmEVs are from one or more bacterial strain listed in Table 1, Table 2 or Table 3.
  • 39. The pharmaceutical composition of any one of claims 1 to 38, wherein the composition further comprises one or more additional therapeutic agents.
  • 40. Use of a pharmaceutical composition of any one of claims 1 to 39 for the preparation of a medicament for the treatment of a disease.
  • 41. The use of claim 49, wherein the disease is a cancer, an autoimmune disease, an inflammatory disease, a dysbiosis, and/or a metabolic disease.
  • 42. A method of treating a subject comprising administering to the subject a pharmaceutical composition of any one of claims 1 to 41.
  • 43. The method of claim 42, wherein the pmEVs are from bacteria that have been gamma irradiated, UV irradiated, heat inactivated, acid treated, oxygen sparged, or a combination thereof.
  • 44. The method of claim 42, wherein the pmEVs are from live bacteria.
  • 45. The method of any one of claims 42 to 44, wherein the composition activates or enhances of one or more immune responses in the subject.
  • 46. The method of claim 45, wherein the one or more immune responses comprises a systemic immune response.
  • 47. The method of any one of claims 42 to 44, wherein the composition suppresses an immune response in the subject.
  • 48. The method of any one of claims 42 to 44, wherein the composition promotes immune activation in the subject.
  • 49. The method of any one of claims 42 to 48, wherein the pharmaceutical composition comprising the pmEVs has comparable potency or increased potency compared to a pharmaceutical composition that contains whole microbes from the same bacterial strain from which the pmEVs were produced).
  • 50. The method of any one of claims 42 to 48, wherein the pharmaceutical composition comprising the pmEVs has more therapeutically active microbial material compared to a pharmaceutical composition that contains whole microbesfrom which the pmEVs were obtained.
  • 51. The method of any one of claims 42 to 50, wherein the subject is in need of treatment for a cancer.
  • 52. The method of any one of claims 42 to 50, wherein the subject is in need of treatment for an autoimmune disease and/or an inflammatory disease.
  • 53. The method of any one of claims 42 to 50, wherein the subject is in need of treatment for a dysbiosis.
  • 54. The method of any one of claims 42 to 50, wherein the subject is in need of treatment for a metabolic disease.
  • 55. The method of any one of claims 42 to 50, wherein the pharmaceutical composition is administered in combination with an additional therapeutic agent.
  • 56. The method of any one of claims 42 to 55, wherein the composition comprises pmEVs from one strain of bacteria.
  • 57. The method of any one of claims 42 to 56, wherein the pmEVs are lyophilized.
  • 58. The method of any one of claims 42 to 57, wherein the pharmaceutical composition is orally administered.
  • 59. The method of any one of claims 42 to 57, wherein the pharmaceutical composition is administered intravenously.
  • 60. The method of any one of claims 42 to 57, wherein the pharmaceutical composition is administered intratumorally.
  • 61. The method of any one of claims 42 to 57, wherein the pharmaceutical composition is administered subtumorally.
  • 62. The method of any one of claims 42 to 57, wherein the pharmaceutical composition is administered by injection.
  • 63. A method for preparing a pharmaceutical composition comprising pmEVs in a suspension, the method comprising: combining pmEVs with a pharmaceutically acceptable buffer, thereby preparing the pharmaceutical composition.
  • 64. The method of claim 63, wherein the pharmaceutically acceptable buffer comprises PBS.
  • 65. The method of claim 63 or 64, wherein the suspension further comprises sucrose or glucose.
  • 66. The method of any one of claims 63 to 65, wherein the pmEVs comprise about 2×106 to about 2×1016 particles of pmEVs.
  • 67. The method of any one of claims 63 to 66, wherein the pmEVs comprise about 5 mg to about 900 mg total protein.
  • 68. A pharmaceutical composition prepared by the method of any one of claims 62 to 67.
  • 69. A method for preparing a solid dose form of pharmaceutical composition comprising pmEVs (e.g., a therapeutically effective amount thereof) in a solid dose form, the method comprising: a) combining pmEVs with a pharmaceutically acceptable excipient; andb) compressing the combined pmEVs and pharmaceutically acceptable excipient; thereby preparing a solid dose form of a pharmaceutical composition.
  • 70. The method of claim 69, further comprising enterically coating the solid dose form.
  • 71. The method of claim 69 or 70, wherein the solid dose form comprises a tablet or a minitablet.
  • 72. The method of any one of claims 69 to 71, wherein the composition comprises pmEVs from one strain of bacteria.
  • 73. The method of any one of claims 69 to 72, wherein the pmEVs are lyophilized.
  • 74. The method of any one of claims 69 to 73, wherein the pmEVs comprise about 2×106 to about 2×1016 particles.
  • 75. The method of any one of claims 69 to 74, wherein the pmEVs comprise about 5 mg to about 900 mg total protein.
  • 76. A pharmaceutical composition prepared by the method of any one of claims 69 to 75.
RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Patent Application No. 62/860,029, filed Jun. 11, 2019; U.S. Provisional Patent Application No. 62/860,049, filed Jun. 11, 2019; U.S. Provisional Patent Application No. 62/979,545, filed Feb. 21, 2020; and U.S. Provisional Patent Application No. 62/991,767, filed Mar. 19, 2020, the contents of each of which are hereby incorporated by reference in their entirety.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2020/037210 6/11/2020 WO
Provisional Applications (4)
Number Date Country
62991767 Mar 2020 US
62979545 Feb 2020 US
62860029 Jun 2019 US
62860049 Jun 2019 US