Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cancer Cell ; 42(1): 16-34, 2024 01 08.
Article in English | MEDLINE | ID: mdl-38157864

ABSTRACT

Over the last decade, the composition of the gut microbiota has been found to correlate with the outcomes of cancer patients treated with immunotherapy. Accumulating evidence points to the various mechanisms by which intestinal bacteria act on distal tumors and how to harness this complex ecosystem to circumvent primary resistance to immune checkpoint inhibitors. Here, we review the state of the microbiota field in the context of melanoma, the recent breakthroughs in defining microbial modes of action, and how to modulate the microbiota to enhance response to cancer immunotherapy. The host-microbe interaction may be deciphered by the use of "omics" technologies, and will guide patient stratification and the development of microbiota-centered interventions. Efforts needed to advance the field and current gaps of knowledge are also discussed.


Subject(s)
Gastrointestinal Microbiome , Melanoma , Microbiota , Neoplasms , Humans , Melanoma/therapy , Neoplasms/therapy , Immunotherapy , Host Microbial Interactions
2.
Nat Cancer ; 4(8): 1083-1094, 2023 08.
Article in English | MEDLINE | ID: mdl-37525016

ABSTRACT

The intersection of the microbiota and cancer and the mechanisms that define these interactions are a fascinating, rapidly evolving area of cancer biology and therapeutics. Here we present recent insights into the mechanisms by which specific bacteria or their communities contribute to carcinogenesis and discuss the bidirectional interplay between microbiota and host gene or epigenome signaling. We conclude with comments on manipulation of the microbiota for the therapeutic benefit of patients with cancer.


Subject(s)
Microbiota , Neoplasms , Humans , Neoplasms/therapy , Carcinogenesis , Bacteria/genetics
3.
Cancer Metastasis Rev ; 41(2): 335-346, 2022 06.
Article in English | MEDLINE | ID: mdl-35588337

ABSTRACT

The identification of microbes enriched in the healthy lung has led to the compelling discovery that microbes may contribute to lung cancer pathogenesis. Here, we review the recent literature showing microbial associations with lung cancer as well as the functional features that have been identified in human and murine studies. Most biomarker data remain limited due to variable findings. However, multiple studies have found that lung tumors or ipsilateral airway samples have decreased α diversity compared to normal tissue. Specific genera, such as Veillonella and Streptococcus, were also found in association with lung tumors using multiple sampling methodologies. These microbes, which are generally found in the upper respiratory track, are associated with an IL-17 signature in the lung, potentially resulting in a pro-tumorigenic environment. Studies detailing these immune mechanisms are limited, and further investigation is necessary to delineate how these bacteria, their metabolites, and potentially tumor-associated neoantigens modulate the immune response in cancer.


Subject(s)
Lung Neoplasms , Microbiota , Animals , Bacteria , Biomarkers , Humans , Immunity , Lung Neoplasms/pathology , Mice , Microbiota/physiology
4.
Cancer Immunol Immunother ; 71(10): 2405-2420, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35217892

ABSTRACT

Human gut microbial species found to associate with clinical responses to immune checkpoint inhibitors (ICIs) are often tested in mice using fecal microbiota transfer (FMT), wherein tumor responses in recipient mice may recapitulate human responses to ICI treatment. However, many FMT studies have reported only limited methodological description, details of murine cohorts, and statistical methods. To investigate the reproducibility and robustness of gut microbial species that impact ICI responses, we performed human to germ-free mouse FMT using fecal samples from patients with non-small cell lung cancer who had a pathological response or nonresponse after neoadjuvant ICI treatment. R-FMT mice yielded greater anti-tumor responses in combination with anti-PD-L1 treatment compared to NR-FMT, although the magnitude varied depending on mouse cell line, sex, and individual experiment. Detailed investigation of post-FMT mouse microbiota using 16S rRNA amplicon sequencing, with models to classify and correct for biological variables, revealed a shared presence of the most highly abundant taxa between the human inocula and mice, though low abundance human taxa colonized mice more variably after FMT. Multiple Clostridium species also correlated with tumor outcome in individual anti-PD-L1-treated R-FMT mice. RNAseq analysis revealed differential expression of T and NK cell-related pathways in responding tumors, irrespective of FMT source, with enrichment of these cell types confirmed by immunohistochemistry. This study identifies several human gut microbial species that may play a role in clinical responses to ICIs and suggests attention to biological variables is needed to improve reproducibility and limit variability across experimental murine cohorts.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Fecal Microbiota Transplantation , Humans , Mice , Neoadjuvant Therapy , RNA, Ribosomal, 16S/genetics , Reproducibility of Results
5.
Clin Cancer Res ; 27(9): 2571-2583, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33593881

ABSTRACT

PURPOSE: While immune checkpoint inhibitors (ICI) have revolutionized the treatment of cancer by producing durable antitumor responses, only 10%-30% of treated patients respond and the ability to predict clinical benefit remains elusive. Several studies, small in size and using variable analytic methods, suggest the gut microbiome may be a novel, modifiable biomarker for tumor response rates, but the specific bacteria or bacterial communities putatively impacting ICI responses have been inconsistent across the studied populations. EXPERIMENTAL DESIGN: We have reanalyzed the available raw 16S rRNA amplicon and metagenomic sequencing data across five recently published ICI studies (n = 303 unique patients) using a uniform computational approach. RESULTS: Herein, we identify novel bacterial signals associated with clinical responders (R) or nonresponders (NR) and develop an integrated microbiome prediction index. Unexpectedly, the NR-associated integrated index shows the strongest and most consistent signal using a random effects model and in a sensitivity and specificity analysis (P < 0.01). We subsequently tested the integrated index using validation cohorts across three distinct and diverse cancers (n = 105). CONCLUSIONS: Our analysis highlights the development of biomarkers for nonresponse, rather than response, in predicting ICI outcomes and suggests a new approach to identify patients who would benefit from microbiome-based interventions to improve response rates.


Subject(s)
Biomarkers , Computational Biology , Immune Checkpoint Inhibitors/pharmacology , Microbiota/drug effects , Bacteria/classification , Bacteria/genetics , Computational Biology/methods , Gastrointestinal Microbiome , Genome, Bacterial , Humans , Immune Checkpoint Inhibitors/therapeutic use , Metagenomics/methods , Microbiota/genetics , RNA, Ribosomal, 16S , ROC Curve , Reproducibility of Results , Whole Genome Sequencing
6.
Science ; 369(6510): 1427-1428, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32943510
7.
EBioMedicine ; 48: 642-647, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31597596

ABSTRACT

The microbiome is increasingly recognized for its role in multiple aspects of cancer development and treatment, specifically in response to checkpoint inhibitors. While checkpoint inhibitors have revolutionized cancer treatment by producing durable anti-tumor responses, only a minority of patients respond to the available immunotherapy drugs and accurate, sensitive and specific microbiome predictors of response to treatment remain elusive. Additionally, the specific mechanisms linking the microbiome and host immunological responses remain unclear. In this review, we examine the evidence for the gut microbiome's association with anti-tumor responses to checkpoint inhibitors in the treatment of melanoma, non-small cell lung cancer, and renal cell carcinoma. Furthermore, we discuss the current evidence available from murine models seeking to explain the immunological mechanisms that may drive this process. While this work is promising in defining the impact of gut microbiota in cancer treatment, many unanswered questions indicate the need for additional human and experimental studies.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Microbiota , Animals , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/etiology , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/immunology , Humans , Immunomodulation/drug effects , Lung Neoplasms/etiology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Melanoma/etiology , Microbiota/drug effects , Microbiota/immunology , Molecular Targeted Therapy , Prognosis , Treatment Outcome
8.
Am J Surg Pathol ; 40(11): 1496-1506, 2016 11.
Article in English | MEDLINE | ID: mdl-27465786

ABSTRACT

Blockade of the programmed death 1 (PD-1) pathway has emerged as a novel therapy for cancer. Therefore, development of biomarkers for response prediction, such as PD-ligand 1 (PD-L1) expression by immunohistochemistry, may help to stratify patients. Solid tumors with CD8 T-cell rich tumor microenvironment have been implicated to be associated with increased PD-L1 expression. We hypothesized that gastric cancers associated with Epstein-Barr virus infection (EBV+) or microsatellite instability (MSI), both of which are known to harbor such tumor microenvironment, are associated with increased PD-L1 expression. Forty-four resected gastric cancers including 7 EBV+, 16 MSI, and 21 microsatellite stable cancers without EBV (EBV-/MSS) were studied for PD-L1 expression and T-cell subpopulations by immunohistochemistry. Positive PD-L1 expression (PD-L1+), defined as membranous staining in either tumor cells or tumor immune infiltrates, was seen in 32 (72%) gastric cancers. EBV+ or MSI cancers showed significantly higher rates of PD-L1+ compared with EBV-/MSS cancers (7/7, 100%; 14/16, 87%; 11/21, 52%; P=0.013). PD-L1+/EBV+ and PD-L1+/MSI cancers had significantly more CD8 T cells at tumor invasive front than PD-L1+/EBV-/MSS cancers (P<0.001). PD-L1+ was not associated with the depth of invasion or nodal metastasis (P=0.534, 0.288). Multivariate analysis showed PD-L1+ was not an independent predictor of disease-free survival while MSI was (P=0.548, 0.043). In summary, EBV+ or MSI gastric cancers are more likely to express PD-L1 and have increased CD8 T cells at tumor invasive front than EBV-/MSS cancers. Our results suggest EBV infection and MSI should be investigated for predicting response to PD-1 blockade.


Subject(s)
B7-H1 Antigen/biosynthesis , Epstein-Barr Virus Infections/complications , Lymphocytes, Tumor-Infiltrating/pathology , Microsatellite Instability , Stomach Neoplasms , Adult , Aged , Aged, 80 and over , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Disease-Free Survival , Female , Humans , Immunohistochemistry , In Situ Hybridization , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/immunology , Male , Middle Aged , Proportional Hazards Models , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/virology
9.
Hepatobiliary Surg Nutr ; 3(2): 93-4, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24812602

ABSTRACT

MicroRNAs (miRNAs) may play an important role in the development and progression of hepatocellular carcinoma (HCC). Understanding the mechanism of specific miRNAs may provide opportunity for development of biomarkers and novel therapeutics in hepatocellular carcinoma which are desperately needed.

10.
Nanomedicine (Lond) ; 9(11): 1613-24, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24195674

ABSTRACT

AIMS: Antibodies are the principal mediator of immunity against reinfection with viruses. Antibodies typically neutralize viruses by binding to virion particles in solution prior to attachment to susceptible cells. Once viruses enter cells, conventional antibodies cannot inhibit virus infection or replication. It is desirable to develop an efficient and nontoxic method for the introduction of virus-inhibiting antibodies into cells. MATERIALS & METHODS: In this article, we report a new method for the delivery of small recombinant antibody fragments into virus-infected cells using a dendrimer-based molecular transporter. RESULTS & CONCLUSION: The construct penetrated virus-infected cells efficiently and inhibited virus replication. This method provides a novel approach for the immediate delivery of inhibitory antibodies directed to virus proteins that are exposed only in the intracellular environment. This approach circumvents the current and rather complicated expression of inhibitory antibodies in cells following gene transfer.


Subject(s)
Antibodies/chemistry , Nanomedicine/methods , Virion/chemistry , Animals , Antibodies, Monoclonal/chemistry , Biological Transport , Capsid Proteins/chemistry , Cytoplasm/metabolism , Dendrimers/chemistry , Enzyme-Linked Immunosorbent Assay , Gene Transfer Techniques , HIV-1/immunology , Humans , Immunoglobulin Fragments/chemistry , Kidney , Macaca mulatta , Magnetic Resonance Spectroscopy , Microscopy, Confocal , Neutralization Tests , Peptides/chemistry , RNA, Small Interfering/metabolism , Rotavirus/metabolism , Viruses/chemistry
11.
Future Microbiol ; 8(1): 123-31, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23252497

ABSTRACT

Respiratory syncytial virus is a single-stranded RNA virus in the Paramyxoviridae family that preferentially assembles and buds from the apical surface of polarized epithelial cells, forming filamentous structures that contain both viral proteins and the genomic RNA. Recent studies have described both viral and host factors that are involved in ribonucleoprotein assembly and trafficking of viral proteins to the cell surface. At the cell surface, viral proteins assemble into filaments that probably require interactions between viral proteins, host proteins and the cell membrane. Finally, a membrane scission event must occur to release the free virion. This article will review the recent literature describing the mechanisms that drive respiratory syncytial virus assembly and budding.


Subject(s)
Respiratory Syncytial Viruses/physiology , Virus Assembly , Animals , Cell Membrane/metabolism , Cytoskeleton/metabolism , Host-Pathogen Interactions , Humans , Protein Transport , Ribonucleoproteins/metabolism , Viral Fusion Proteins/metabolism , Viral Proteins/metabolism , Virus Release
12.
Virology ; 434(1): 129-36, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-23062737

ABSTRACT

We describe the first example of combining bacterial artificial chromosome (BAC) recombination-mediated mutagenesis with reverse genetics for a negative strand RNA virus. A BAC-based respiratory syncytial virus (RSV) rescue system was established. An important advantage of this system is that RSV antigenomic cDNA was stabilized in the BAC vector. The RSV genotype chosen was A2-line19F, a chimeric strain previously shown to recapitulate in mice key features of RSV pathogenesis. We recovered two RSV reporter viruses, one expressing the red fluorescent protein monomeric Katushka 2 (A2-K-line19F) and one expressing Renilla luciferase (A2-RL-line19F). As proof of principle, we efficiently generated a RSV gene deletion mutant (A2-line19FΔNS1/NS2) and a point mutant (A2-K-line19F-I557V) by recombination-mediated BAC mutagenesis. Together with sequence-optimized helper expression plasmids, BAC-RSV is a stable, versatile, and efficient reverse genetics platform for generation of a recombinant Pneumovirus.


Subject(s)
Mutagenesis , Recombination, Genetic , Respiratory Syncytial Viruses/genetics , Reverse Genetics/methods , Animals , Artificial Gene Fusion , Cell Line , Chromosomes, Artificial, Bacterial , Genes, Reporter , Genetic Vectors , Humans , Luciferases/analysis , Luciferases/genetics , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Red Fluorescent Protein
13.
PLoS One ; 7(7): e40826, 2012.
Article in English | MEDLINE | ID: mdl-22808269

ABSTRACT

Respiratory syncytial virus (RSV) is a single-stranded RNA virus that assembles into viral filaments at the cell surface. Virus assembly often depends on the ability of a virus to use host proteins to accomplish viral tasks. Since the fusion protein cytoplasmic tail (FCT) is critical for viral filamentous assembly, we hypothesized that host proteins important for viral assembly may be recruited by the FCT. Using a yeast two-hybrid screen, we found that filamin A interacted with FCT, and mammalian cell experiments showed it localized to viral filaments but did not affect viral replication. Furthermore, we found that a number of actin-associated proteins also were excluded from viral filaments. Actin or tubulin cytoskeletal rearrangement was not necessary for F trafficking to the cell surface or for viral assembly into filaments, but was necessary for optimal viral replication and may be important for anchoring viral filaments. These findings suggest that RSV assembly into filaments occurs independently of actin polymerization and that viral proteins are the principal drivers for the mechanical tasks involved with formation of complex, structured RSV filaments at the host cell plasma membrane.


Subject(s)
Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Host-Pathogen Interactions , Respiratory Syncytial Viruses/physiology , Virion/metabolism , Virus Assembly/physiology , Animals , Cell Line , Contractile Proteins/genetics , Contractile Proteins/metabolism , Filamins , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Protein Transport , Two-Hybrid System Techniques
14.
mBio ; 3(1)2012.
Article in English | MEDLINE | ID: mdl-22318318

ABSTRACT

UNLABELLED: Respiratory syncytial virus (RSV) is a single-stranded RNA virus in the Paramyxoviridae family that assembles into filamentous structures at the apical surface of polarized epithelial cells. These filaments contain viral genomic RNA and structural proteins, including the fusion (F) protein, matrix (M) protein, nucleoprotein (N), and phosphoprotein (P), while excluding F-actin. It is known that the F protein cytoplasmic tail (FCT) is necessary for filament formation, but the mechanism by which the FCT mediates assembly into filaments is not clear. We hypothesized that the FCT is necessary for interactions with other viral proteins in order to form filaments. In order to test this idea, we expressed the F protein with cytoplasmic tail (CT) truncations or specific point mutations and determined the abilities of these variant F proteins to form filaments independent of viral infection when coexpressed with M, N, and P. Deletion of the terminal three FCT residues (amino acids Phe-Ser-Asn) or mutation of the Phe residue resulted in a loss of filament formation but did not affect F-protein expression or trafficking to the cell surface. Filament formation could be restored by addition of residues Phe-Ser-Asn to an FCT deletion mutant and was unaffected by mutations to Ser or Asn residues. Second, deletion of residues Phe-Ser-Asn or mutation of the Phe residue resulted in a loss of M, N, and P incorporation into virus-like particles. These data suggest that a C-terminal Phe residue in the FCT mediates assembly through incorporation of internal virion proteins into virus filaments at the cell surface. IMPORTANCE: Respiratory syncytial virus (RSV) is a leading cause of bronchiolitis and pneumonia in infants and the elderly worldwide. There is no licensed RSV vaccine and only limited therapeutics for use in infected patients. Many aspects of the RSV life cycle have been studied, but the mechanisms that drive RSV assembly at the cell surface are not well understood. This study provides evidence that a specific residue in the RSV fusion protein cytoplasmic tail coordinates assembly into viral filaments by mediating the incorporation of internal virion proteins. Understanding the mechanisms that drive RSV assembly could lead to targeted development of novel antiviral drugs. Moreover, since RSV exits infected cells in an ESCRT (endosomal sorting complexes required for transport)-independent manner, these studies may contribute new knowledge about a general strategy by which ESCRT-independent viruses mediate outward bud formation using viral protein-mediated mechanisms during assembly and budding.


Subject(s)
Phenylalanine , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/physiology , Viral Fusion Proteins/genetics , Virus Assembly , Amino Acid Sequence , Cell Line , Cell Membrane/metabolism , Cytoplasm/metabolism , Cytoskeleton/ultrastructure , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Point Mutation , RNA, Viral/genetics , RNA, Viral/metabolism , Recombinant Proteins , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/ultrastructure , Sequence Alignment , Viral Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...