Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 467
Filter
1.
Skin Res Technol ; 30(8): e70017, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39167029

ABSTRACT

BACKGROUND: Melanoma is an aggressive malignancy primarily impacting the skin, mucous membranes, and pigment epithelium. The tumor microbial microenvironment encompasses both the microorganisms inhabiting the tumor vicinity and the environmental factors influencing their interactions. Emerging evidence highlights the pivotal role of the microbial immune microenvironment in melanoma. METHODS: We conducted an extensive review of scholarly works published from 2012 to 2022, utilizing The Web of Science Core Collection. Subsequently, we employed analytical tools such as VOSviewer, CiteSpace, and the R programming language to scrutinize prevailing research patterns within this domain. RESULTS: A sum of 513 articles were pinpointed, with notable input coming from the United States and China. Harvard University stood out as the top-contributing institution, while the journal Science received the most citations. Current research within this sphere chiefly focuses on two principal domains: the gut microbiota and the PD-L1 pathway concerning melanoma treatment. CONCLUSION: The study offers an extensive analysis and overview of the worldwide research landscape concerning the immune microenvironment with a focus on microbes in melanoma. It underscores the promising prospects for harnessing the microbial immune microenvironment's potential in melanoma. These findings furnish valuable insights and guidance for advancing scientific inquiry and refining clinical approaches within this dynamic field.


Subject(s)
Bibliometrics , Melanoma , Skin Neoplasms , Tumor Microenvironment , Melanoma/immunology , Melanoma/microbiology , Humans , Tumor Microenvironment/immunology , Skin Neoplasms/immunology , Skin Neoplasms/microbiology , Gastrointestinal Microbiome , Biomedical Research
2.
Cancer Res Commun ; 4(8): 1978-1990, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39015091

ABSTRACT

Emerging evidence supports the important role of the tumor microbiome in oncogenesis, cancer immune phenotype, cancer progression, and treatment outcomes in many malignancies. In this study, we investigated the metastatic melanoma tumor microbiome and its potential roles in association with clinical outcomes, such as survival, in patients with metastatic disease treated with immune checkpoint inhibitors (ICI). Baseline tumor samples were collected from 71 patients with metastatic melanoma before treatment with ICIs. Bulk RNA sequencing (RNA-seq) was conducted on the formalin-fixed, paraffin-embedded and fresh frozen tumor samples. Durable clinical benefit (primary clinical endpoint) following ICIs was defined as overall survival >24 months and no change to the primary drug regimen (responders). We processed RNA-seq reads to carefully identify exogenous sequences using the {exotic} tool. The age of the 71 patients with metastatic melanoma ranged from 24 to 83 years, 59% were male, and 55% survived >24 months following the initiation of ICI treatment. Exogenous taxa were identified in the tumor RNA-seq, including bacteria, fungi, and viruses. We found differences in gene expression and microbe abundances in immunotherapy-responsive versus nonresponsive tumors. Responders showed significant enrichment of bacteriophages in the phylum Uroviricota, and nonresponders showed enrichment of several bacteria, including Campylobacter jejuni. These microbes correlated with immune-related gene expression signatures. Finally, we found that models for predicting prolonged survival with immunotherapy using both microbe abundances and gene expression outperformed models using either dataset alone. Our findings warrant further investigation and potentially support therapeutic strategies to modify the tumor microbiome in order to improve treatment outcomes with ICIs. SIGNIFICANCE: We analyzed the tumor microbiome and interactions with genes and pathways in metastatic melanoma treated with immunotherapy and identified several microbes associated with immunotherapy response and immune-related gene expression signatures. Machine learning models that combined microbe abundances and gene expression outperformed models using either dataset alone in predicting immunotherapy responses.


Subject(s)
Immune Checkpoint Inhibitors , Melanoma , Microbiota , Humans , Melanoma/drug therapy , Melanoma/microbiology , Melanoma/immunology , Melanoma/secondary , Male , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Female , Middle Aged , Aged , Adult , Microbiota/drug effects , Aged, 80 and over , Young Adult , Treatment Outcome , Skin Neoplasms/drug therapy , Skin Neoplasms/microbiology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Neoplasm Metastasis , Prognosis
3.
Skin Res Technol ; 30(6): e13804, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38895789

ABSTRACT

OBJECTIVE: Research has previously established connections between the intestinal microbiome and the progression of some cancers. However, there is a noticeable gap in the literature in regard to using Mendelian randomisation (MR) to delve into potential causal relationships between the gut microbiota (GM) and basal cell carcinoma (BCC). Therefore, the purpose of our study was to use MR to explore the causal relationship between four kinds of GM (Bacteroides, Streptococcus, Proteobacteria and Lachnospiraceae) and BCC. METHODS: We used genome-wide association study (GWAS) data and MR to explore the causal relationship between four kinds of GM and BCC. This study primarily employed the random effect inverse variance weighted (IVW) model for analysis, as complemented by additional methods including the simple mode, weighted median, weighted mode and MR‒Egger methods. We used heterogeneity and horizontal multiplicity to judge the reliability of each analysis. MR-PRESSO was mainly used to detect and correct outliers. RESULTS: The random-effects IVW results showed that Bacteroides (OR = 0.936, 95% CI = 0.787-1.113, p = 0.455), Streptococcus (OR = 0.974, 95% CI = 0.875-1.083, p = 0.629), Proteobacteria (OR = 1.113, 95% CI = 0.977-1.267, p = 0.106) and Lachnospiraceae (OR = 1.027, 95% CI = 0.899-1.173, p = 0.688) had no genetic causal relationship with BCC. All analyses revealed no horizontal pleiotropy, heterogeneity or outliers. CONCLUSION: We found that Bacteroides, Streptococcus, Proteobacteria and Lachnospiraceae do not increase the incidence of BCC at the genetic level, which provides new insight for the study of GM and BCC.


Subject(s)
Carcinoma, Basal Cell , Gastrointestinal Microbiome , Genome-Wide Association Study , Mendelian Randomization Analysis , Skin Neoplasms , Humans , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/microbiology , Gastrointestinal Microbiome/genetics , Skin Neoplasms/genetics , Skin Neoplasms/microbiology , Streptococcus/genetics , Proteobacteria/genetics , Bacteroides/genetics , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide
4.
J Invest Dermatol ; 144(9): 1954-1962, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38703171

ABSTRACT

The etiology of CTCL is a subject of extensive investigation. Researchers have explored links between CTCL and environmental chemical exposures, such as aromatic hydrocarbons (eg, pesticides and benzene), as well as infectious factors, including various viruses (eg, human T-lymphotropic virus [HTLV]-I and HTLV-II) and bacteria (eg, Staphylococcus aureus). There has been growing emphasis on the role of malignant inflammation in CTCL development. In this review, we synthesize studies of environmental and infectious exposures, along with research on the aryl hydrocarbon receptor and the involvement of pathogens in disease etiology, providing insight into the pathogenesis of CTCL.


Subject(s)
Lymphoma, T-Cell, Cutaneous , Skin Neoplasms , Humans , Lymphoma, T-Cell, Cutaneous/microbiology , Lymphoma, T-Cell, Cutaneous/pathology , Skin Neoplasms/microbiology , Skin Neoplasms/pathology , Skin Neoplasms/etiology , Inflammation/microbiology , Environmental Exposure/adverse effects , Receptors, Aryl Hydrocarbon/metabolism , Animals
5.
Front Immunol ; 15: 1255859, 2024.
Article in English | MEDLINE | ID: mdl-38646524

ABSTRACT

Cutaneous T-cell lymphomas (CTCL) are a group of lymphoproliferative disorders of skin-homing T cells causing chronic inflammation. These disorders cause impairment of the immune environment, which leads to severe infections and/or sepsis due to dysbiosis. In this study, we elucidated the host-microbial interaction in CTCL that occurs during the phototherapeutic treatment regime and determined whether modulation of the skin microbiota could beneficially affect the course of CTCL. EL4 T-cell lymphoma cells were intradermally grafted on the back of C57BL/6 mice. Animals were treated with conventional therapeutics such as psoralen + UVA (PUVA) or UVB in the presence or absence of topical antibiotic treatment (neomycin, bacitracin, and polymyxin B sulphate) as an adjuvant. Microbial colonisation of the skin was assessed to correlate with disease severity and tumour growth. Triple antibiotic treatment significantly delayed tumour occurrence (p = 0.026), which prolonged the survival of the mice (p = 0.033). Allocation to phototherapeutic agents PUVA, UVB, or none of these, along with antibiotic intervention, reduced the tumour growth significantly (p = 0.0327, p ≤ 0.0001, p ≤ 0.0001 respectively). The beta diversity indices calculated using the Bray-Curtis model showed that the microbial population significantly differed after antibiotic treatment (p = 0.001). Upon modulating the skin microbiome by antibiotic treatment, we saw an increase in commensal Clostridium species, e.g., Lachnospiraceae sp. (p = 0.0008), Ruminococcaceae sp. (p = 0.0001)., Blautia sp. (p = 0.007) and a significant reduction in facultative pathogens Corynebacterium sp. (p = 0.0009), Pelomonas sp. (p = 0.0306), Streptococcus sp. (p ≥ 0.0001), Pseudomonas sp. (p = 0.0358), and Cutibacterium sp. (p = 0.0237). Intriguingly, we observed a significant decrease in Staphylococcus aureus frequency (p = 0.0001) but an increase in the overall detection frequency of the Staphylococcus genus, indicating that antibiotic treatment helped regain the microbial balance and increased the number of non-pathogenic Staphylococcus populations. These study findings show that modulating microbiota by topical antibiotic treatment helps to restore microbial balance by diminishing the numbers of pathogenic microbes, which, in turn, reduces chronic inflammation, delays tumour growth, and increases survival rates in our CTCL model. These findings support the rationale to modulate the microbial milieu during the disease course of CTCL and indicate its therapeutic potential.


Subject(s)
Lymphoma, T-Cell, Cutaneous , Mice, Inbred C57BL , Microbiota , Skin Neoplasms , Skin , Animals , Microbiota/drug effects , Mice , Skin/microbiology , Skin/pathology , Skin/immunology , Skin/drug effects , Skin Neoplasms/microbiology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Lymphoma, T-Cell, Cutaneous/microbiology , Lymphoma, T-Cell, Cutaneous/pathology , Lymphoma, T-Cell, Cutaneous/drug therapy , Lymphoma, T-Cell, Cutaneous/therapy , Disease Models, Animal , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/administration & dosage , Cell Line, Tumor , Female , Humans
6.
mSphere ; 9(4): e0055523, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38530017

ABSTRACT

Human cutaneous squamous cell carcinomas (SCCs) and actinic keratoses (AK) display microbial dysbiosis with an enrichment of staphylococcal species, which have been implicated in AK and SCC progression. SCCs are common in both felines and canines and are often diagnosed at late stages leading to high disease morbidity and mortality rates. Although recent studies support the involvement of the skin microbiome in AK and SCC progression in humans, there is no knowledge of this in companion animals. Here, we provide microbiome data for SCC in cats and dogs using culture-independent molecular profiling and show a significant decrease in microbial alpha diversity on SCC lesions compared to normal skin (P ≤ 0.05). Similar to human skin cancer, SCC samples had an elevated abundance of staphylococci relative to normal skin-50% (6/12) had >50% staphylococci, as did 16% (4/25) of perilesional samples. Analysis of Staphylococcus at the species level revealed an enrichment of the pathogenic species Staphylococcus felis in cat SCC samples, a higher prevalence of Staphylococcus pseudintermedius in dogs, and a higher abundance of Staphylococcus aureus compared to normal skin in both companion animals. Additionally, a comparison of previously published human SCC and perilesional samples against the present pet samples revealed that Staphylococcus was the most prevalent genera across human and companion animals for both sample types. Similarities between the microbial profile of human and cat/dog SCC lesions should facilitate future skin cancer research. IMPORTANCE: The progression of precancerous actinic keratosis lesions (AK) to cutaneous squamous cell carcinoma (SCC) is poorly understood in humans and companion animals, despite causing a significant burden of disease. Recent studies have revealed that the microbiota may play a significant role in disease progression. Staphylococcus aureus has been found in high abundance on AK and SCC lesions, where it secretes DNA-damaging toxins, which could potentiate tumorigenesis. Currently, a suitable animal model to investigate this relationship is lacking. Thus, we examined the microbiome of cutaneous SCC in pets, revealing similarities to humans, with increased staphylococci and reduced commensals on SCC lesions and peri-lesional skin compared to normal skin. Two genera that were in abundance in SCC samples have also been found in human oral SCC lesions. These findings suggest the potential suitability of pets as a model for studying microbiome-related skin cancer progression.


Subject(s)
Carcinoma, Squamous Cell , Cat Diseases , Dog Diseases , Microbiota , Skin Neoplasms , Skin , Staphylococcus , Cats , Dogs , Animals , Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/veterinary , Skin Neoplasms/microbiology , Skin Neoplasms/veterinary , Skin Neoplasms/pathology , Skin/microbiology , Skin/pathology , Cat Diseases/microbiology , Staphylococcus/isolation & purification , Staphylococcus/genetics , Staphylococcus/classification , Staphylococcus/pathogenicity , Dog Diseases/microbiology , Keratosis, Actinic/microbiology , Keratosis, Actinic/veterinary , Keratosis, Actinic/pathology
8.
Melanoma Res ; 34(3): 225-233, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38469881

ABSTRACT

Currently, numerous studies suggest a potential association between the gut microbiota and the progression of melanoma. Hence, our objective was to examine the genetic impact of the gut microbiota on melanoma through the utilization of the Mendelian randomization (MR) approach. This research employed Bacteroides, Streptococcus, Proteobacteria, and Lachnospiraceae as exposure variables and cutaneous melanoma (CM) as the outcome in a two-sample MR analysis. In this MR research, the primary analytical approach was the random-effects inverse-variance weighting (IVW) model. Complementary methods included weighted median, MR Egger, and basic and weighted models. We assessed both heterogeneity and horizontal pleiotropy in our study, scrutinizing whether the analysis results were affected by any individual SNP. The random-effects IVW outcomes indicated that Streptococcus, Bacteroides, Lachnospiraceae and Proteobacteria had no causal relationship with CM, with odds ratios of 1.001 [95% confidence interval (CI) = 0.998-1.004, P  = 0.444], 0.999 (95% CI = 0.996-1.002, P  = 0.692), 1.001 (95% CI = 0.998-1.003, P  = 0.306), and 0.999 (95% CI = 0.997-1.002, P  = 0.998), respectively. No analyses exhibited heterogeneity, horizontal pleiotropy, or deviations. Our research determined that Bacteroides, Streptococcus, Proteobacteria, and Lachnospiraceae do not induce CM at the genetic level. However, we cannot dismiss the possibility that these four gut microbiotas might influence CM through other mechanisms.


Subject(s)
Gastrointestinal Microbiome , Melanoma , Mendelian Randomization Analysis , Skin Neoplasms , Humans , Melanoma/genetics , Melanoma/microbiology , Skin Neoplasms/genetics , Skin Neoplasms/microbiology , Melanoma, Cutaneous Malignant
10.
J Invest Dermatol ; 143(9): 1757-1768.e3, 2023 09.
Article in English | MEDLINE | ID: mdl-36889662

ABSTRACT

Staphylococcus aureus is suspected to fuel disease activity in cutaneous T-cell lymphomas. In this study, we investigate the effect of a recombinant, antibacterial protein, endolysin (XZ.700), on S. aureus skin colonization and malignant T-cell activation. We show that endolysin strongly inhibits the proliferation of S. aureus isolated from cutaneous T-cell lymphoma skin and significantly decreases S. aureus bacterial cell counts in a dose-dependent manner. Likewise, ex vivo colonization of both healthy and lesional skin by S. aureus is profoundly inhibited by endolysin. Moreover, endolysin inhibits the patient-derived S. aureus induction of IFNγ and the IFNγ-inducible chemokine CXCL10 in healthy skin. Whereas patient-derived S. aureus stimulates activation and proliferation of malignant T cells in vitro through an indirect mechanism involving nonmalignant T cells, endolysin strongly inhibits the effects of S. aureus on activation (reduced CD25 and signal transducer and activator of transcription 5 phosphorylation) and proliferation (reduced Ki-67) of malignant T cells and cell lines in the presence of nonmalignant T cells. Taken together, we provide evidence that endolysin XZ.700 inhibits skin colonization, chemokine expression, and proliferation of pathogenic S. aureus and blocks their potential tumor-promoting effects on malignant T cells.


Subject(s)
Lymphoma, T-Cell, Cutaneous , Skin Neoplasms , Staphylococcal Infections , Humans , Staphylococcus aureus , Skin/microbiology , Staphylococcal Infections/microbiology , Lymphoma, T-Cell, Cutaneous/drug therapy , Recombinant Proteins , T-Lymphocytes , Skin Neoplasms/drug therapy , Skin Neoplasms/microbiology
11.
JCI Insight ; 7(2)2022 01 25.
Article in English | MEDLINE | ID: mdl-35076024

ABSTRACT

Besides promoting inflammation by mobilizing lipid mediators, group IIA secreted phospholipase A2 (sPLA2-IIA) prevents bacterial infection by degrading bacterial membranes. Here, we show that, despite the restricted intestinal expression of sPLA2-IIA in BALB/c mice, its genetic deletion leads to amelioration of cancer and exacerbation of psoriasis in distal skin. Intestinal expression of sPLA2-IIA is reduced after treatment with antibiotics or under germ-free conditions, suggesting its upregulation by gut microbiota. Metagenome, transcriptome, and metabolome analyses have revealed that sPLA2-IIA deficiency alters the gut microbiota, accompanied by notable changes in the intestinal expression of genes related to immunity and metabolism, as well as in the levels of various blood metabolites and fecal bacterial lipids, suggesting that sPLA2-IIA contributes to shaping of the gut microbiota. The skin phenotypes in Pla2g2a-/- mice are lost (a) when they are cohoused with littermate WT mice, resulting in the mixing of the microbiota between the genotypes, or (b) when they are housed in a more stringent pathogen-free facility, where Pla2g2a expression in WT mice is low and the gut microbial compositions in both genotypes are nearly identical. Thus, our results highlight a potentially new aspect of sPLA2-IIA as a modulator of gut microbiota, perturbation of which affects distal skin responses.


Subject(s)
Gastrointestinal Microbiome/immunology , Group II Phospholipases A2/metabolism , Psoriasis , Skin Neoplasms , Animals , Carcinogenesis/immunology , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation/immunology , Inflammation/microbiology , Mice , Mice, Inbred BALB C , Pathology, Molecular/methods , Psoriasis/immunology , Psoriasis/microbiology , Skin Neoplasms/immunology , Skin Neoplasms/microbiology
12.
Genome Biol ; 22(1): 187, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162397

ABSTRACT

BACKGROUND: The human microbiome plays an important role in cancer. Accumulating evidence indicates that commensal microbiome-derived DNA may be represented in minute quantities in the cell-free DNA of human blood and could possibly be harnessed as a new cancer biomarker. However, there has been limited use of rigorous experimental controls to account for contamination, which invariably affects low-biomass microbiome studies. RESULTS: We apply a combination of 16S-rRNA-gene sequencing and droplet digital PCR to determine if the specific detection of cell-free microbial DNA (cfmDNA) is possible in metastatic melanoma patients. Compared to matched stool and saliva samples, the absolute concentration of cfmDNA is low but significantly above the levels detected from negative controls. The microbial community of plasma is strongly influenced by laboratory and reagent contaminants introduced during the DNA extraction and sequencing processes. Through the application of an in silico decontamination strategy including the filtering of amplicon sequence variants (ASVs) with batch dependent abundances and those with a higher prevalence in negative controls, we identify known gut commensal bacteria, such as Faecalibacterium, Bacteroides and Ruminococcus, and also other uncharacterised ASVs. We analyse additional plasma samples, highlighting the potential of this framework to identify differences in cfmDNA between healthy and cancer patients. CONCLUSIONS: Together, these observations indicate that plasma can harbour a low yet detectable level of cfmDNA. The results highlight the importance of accounting for contamination and provide an analytical decontamination framework to allow the accurate detection of cfmDNA for future biomarker studies in cancer and other diseases.


Subject(s)
Cell-Free Nucleic Acids/genetics , DNA, Bacterial/genetics , Melanoma/microbiology , Microbiota/genetics , Skin Neoplasms/microbiology , Bacteroides/classification , Bacteroides/genetics , Bacteroides/isolation & purification , Cell-Free Nucleic Acids/blood , DNA Contamination , DNA, Bacterial/blood , Faecalibacterium/classification , Faecalibacterium/genetics , Faecalibacterium/isolation & purification , Feces/microbiology , Humans , Melanoma/diagnosis , Melanoma/pathology , Neoplasm Metastasis , Neoplasm Staging , Polymerase Chain Reaction/methods , RNA, Ribosomal, 16S/genetics , Ruminococcus/classification , Ruminococcus/genetics , Ruminococcus/isolation & purification , Saliva/microbiology , Skin Neoplasms/diagnosis , Skin Neoplasms/pathology , Symbiosis/physiology
13.
Eur J Cancer ; 151: 25-34, 2021 07.
Article in English | MEDLINE | ID: mdl-33962358

ABSTRACT

OBJECTIVE: The gut microbiome plays an important role in systemic inflammation and immune response. Microbes can translocate and reside in tumour niches. However, it is unclear how the intratumour microbiome affects immunity in human cancer. The purpose of this study was to investigate the association between intratumour bacteria, infiltrating CD8+ T cells and patient survival in cutaneous melanoma. METHODS: Using The Cancer Genome Altas's cutaneous melanoma RNA sequencing data, levels of intratumour bacteria and infiltrating CD8+ T cells were determined. Correlation between intratumour bacteria and infiltrating CD8+ T cells or chemokine gene expression and survival analysis of infiltrating CD8+ T cells and Lachnoclostridium in cutaneous melanoma were performed. RESULTS: Patients with low levels of CD8+ T cells have significantly shorter survival than those with high levels. The adjusted hazard ratio was 1.57 (low vs high) (95% confidence interval: 1.17-2.10, p = 0.002). Intratumour bacteria of the Lachnoclostridium genus ranked top in a positive association with infiltrating CD8+ T cells (correlation coefficient = 0.38, p = 9.4 × 10-14), followed by Gelidibacter (0.31, p = 1.13 × 10-9), Flammeovirga (0.29, p = 1.96 × 10-8) and Acinetobacter (0.28, p = 8.94 × 10-8). These intratumour genera positively correlated with chemokine CXCL9, CXCL10 and CCL5 expression. The high Lachnoclostridium load significantly reduced the mortality risk (p = 0.0003). However, no statistically significant correlation was observed between intratumour Lachnoclostridium abundance and the levels of either NK, B or CD4+ T cells. CONCLUSION: Intratumour-residing gut microbiota could modulate chemokine levels and affect CD8+ T-cell infiltration, consequently influencing patient survival in cutaneous melanoma. Manipulating the intratumour gut microbiome may benefit patient outcomes for those undergoing immunotherapy.


Subject(s)
Bacteria/growth & development , Bacterial Translocation , Gastrointestinal Microbiome , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Melanoma/microbiology , Skin Neoplasms/immunology , Skin Neoplasms/microbiology , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Bacterial Load , Chemokines/genetics , Chemokines/metabolism , Clostridiales/growth & development , Cytotoxicity, Immunologic , Female , Humans , Lymphocyte Count , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Melanoma/metabolism , Melanoma/mortality , Middle Aged , Prognosis , Skin Neoplasms/metabolism , Skin Neoplasms/mortality , T-Lymphocytes, Cytotoxic/metabolism , Young Adult
14.
Am J Clin Dermatol ; 22(3): 301-314, 2021 May.
Article in English | MEDLINE | ID: mdl-33765322

ABSTRACT

The management and prognosis of BRAF-mutant metastatic melanoma have changed drastically following the introduction of immune checkpoint inhibitors and molecularly targeted agents. These treatment options present different mechanisms of action and toxicities but also totally distinct kinetics of their response, including a "relatively" short-lasting benefit in subsets of patients treated with BRAF/MEK inhibitors and a lower response rate in patients treated with immune checkpoint inhibitors. BRAF/MEK inhibitors, when administered prior to or concurrently with immune checkpoint inhibitors, at least transiently alter some immunosuppressive parameters of the tumor microenvironment and theoretically improve sensitivity to immunotherapy. Preclinical data from mouse models with oncogene-addicted melanoma confirmed this beneficial immune/targeted synergy and supported the clinical testing of combinations of BRAF/MEK inhibitors and immune checkpoint inhibitors to improve the activity of upfront anti-melanoma therapies. The first positive phase III results were published in 2020, and triggered the discussion about the benefits, the limitations, as well as the possible implications of combining or sequencing targeted therapies with immune checkpoint inhibitors in everyday practice. Beginning from the interplay of immune/targeted agents within the melanoma microenvironment, this review outlines available information from the retrospective experience up to the late-stage randomized evidence on combinatorial treatments. Many clinical trials are currently underway exploring open questions about optimal timing, new immune biomarkers, and eligible patient subsets for these immune/targeted regimens. Awaiting these results, decision making in the first-line setting for BRAF-mutant melanoma is still guided by the patients' characteristics and the biological aspects of melanoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Melanoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Skin Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Clinical Decision-Making , Clinical Trials as Topic , Humans , Immune Checkpoint Inhibitors/pharmacology , Melanoma/genetics , Melanoma/immunology , Melanoma/secondary , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Molecular Targeted Therapy/methods , Mutation , Patient Selection , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/genetics , Skin Neoplasms/microbiology , Skin Neoplasms/pathology , Treatment Outcome , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
16.
Cancer Med ; 9(18): 6791-6801, 2020 09.
Article in English | MEDLINE | ID: mdl-32750218

ABSTRACT

BACKGROUND: A growing number of studies show that intestinal microbiota affect the therapeutic effects of antineoplastic agents. Disulfiram (tetraethylthiuram disulfide, DSF) is an old alcohol-aversion drug that has been shown to be effective against various types of cancers in preclinical studies, while few studies are carried out to explore its mechanism. METHODS: A mice model of melanoma xenograft was generated and treated with antibiotics (Abx), disulfiram/copper (DSF/Cu2+ ), Abx + DSF/Cu2+ , and the tumor volume and survival curve were observed. Hematoxylin-eosin (HE) staining and western blotting (WB) were used to observe the protein changes related to cell morphology, inflammation, and apoptosis in tumor tissues. Quantitative real time polymerase chain reaction (qPCR) was used to detect the expression of pro-inflammatory cytokines in tumors. High-throughput sequencing was used to detect the effects of Abx and DSF/Cu2+ on intestinal microbiota. RESULTS: The DSF/Cu2+ and Abx + DSF/Cu2+ markedly delayed tumor progression and prolonged mice survival, of which the combination of Abx and DSF/Cu2+ possessed the best anti-tumor effect. Abx + DSF/Cu2+ significantly reduced the pro-inflammatory cytokines Interleukin-1ß (IL-1ß), IL-6 and tumor necrosis factor α (TNF-α) in tumors, and significantly reduced the expression of phosphorylated-protein kinase B (p-AKT)/protein kinase B (AKT), toll-like receptors 4 (TLR-4), and phosphorylated- nuclear factor kappa-B (p-NFκB)/NFκB in tumors. Moreover our high-throughput sequencing first indicated that the sound anti-cancer effect of Abx + DSF/Cu2+ had a strong connection with the increased abundance of intestinal beneficial bacteria Akkermansia, as well as the reduced abundance of opportunistic pathogenic bacteria Campylobacterales, Helicobacteraceae, and Coriobacteriaceae. CONCLUSIONS: The disturbed intestinal microbiota (increased abundance of opportunistic pathogens Campylobacterales, Helicobacteraceae, and Coriobacteriaceae) and the over-activated TLR4/NF-κB signaling pathway in tumor tissues deteriorated the cancer development, and the using of antibiotics is benefit to enhance the therapeutic effect of DSF on tumors via inhibiting the growth of opportunistic pathogenic bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bacteria/drug effects , Disulfiram/pharmacology , Gastrointestinal Microbiome/drug effects , Gluconates/pharmacology , Intestines/microbiology , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Bacteria/growth & development , Cell Line, Tumor , Drug Synergism , Female , Host-Pathogen Interactions , Melanoma/microbiology , Melanoma/pathology , Mice, Inbred C57BL , Skin Neoplasms/microbiology , Skin Neoplasms/pathology
17.
Crit Rev Microbiol ; 46(4): 433-449, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32692305

ABSTRACT

Recently, research has been deeply focusing on the role of the microbiota in numerous diseases, either affecting the skin or other organs. What it is well established is that its dysregulation promotes several cutaneous disorders (i.e. psoriasis and atopic dermatitis). To date, little is known about its composition, mediators and role in the genesis, progression and response to therapy of Non-Melanoma Skin Cancer (NMSC). Starting from a bibliographic study, we classified the selected articles into four sections: i) normal skin microbiota; ii) in vitro study models; iii) microbiota and NMSC and iv) probiotics, antibiotics and NMSC. What has emerged is how skin microflora changes, mainly represented by increases of Staphylococcus aureus, Streptococcus pyogenes and Pseudomonas aeruginosa strains, modifications in the mutual quantity of ß-Human papillomavirus genotypes, of Epstein Barr Virus and Malassezia or candidiasis, may contribute to the induction of a state of chronic self-maintaining inflammation, leading to cancer. In this context, the role of S. aureus and that of specific antimicrobial peptides look to be prominent. Moreover, although antibiotics may contribute to carcinogenesis, due to their ability to influence the microbiota balance, specific probiotics, such as Lacticaseibacillus rhamnosus GG, Lactobacillus johnsonii NCC 533 and Bifidobacteria spp., may be protective.


Subject(s)
Bacteria/isolation & purification , Microbiota , Skin Neoplasms/microbiology , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/growth & development , Humans , Skin/microbiology , Skin/virology , Skin Neoplasms/virology , Viruses/classification , Viruses/genetics , Viruses/isolation & purification
19.
Clin Exp Dermatol ; 45(6): 722-726, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32304588

ABSTRACT

Symptomatic cutaneous metastases are associated with discharge, malodour, pruritus and pain, all of which may negatively impact quality of life and cutaneous health. We conducted a retrospective chart review of patients referred to the Dermatology Service at Memorial Sloan Kettering Cancer Center between August 2006 and June 2015, and characterized the microbial flora and antimicrobial management of cutaneous metastases in 64 patients. We detected pathogenic and/or opportunistic bacteria in 50% of skin lesions. The most commonly isolated organisms were Staphylococcus aureus and Pseudomonas aeruginosa. Patients treated with oral antibiotics, alone or in combination with topical agents, had a statistically significant better improvement in infectious symptoms than those treated without oral antibiotics. Our findings suggest that the normal skin microbial flora is disrupted in patients with symptomatic skin metastases. Oral antibiotics may provide benefit when used as first-line therapy for infected skin lesions in patients with symptomatic cutaneous metastases.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Gram-Negative Bacteria/isolation & purification , Gram-Positive Bacteria/isolation & purification , Skin Diseases, Bacterial/drug therapy , Skin Neoplasms/microbiology , Skin Neoplasms/secondary , Skin/microbiology , Administration, Oral , Administration, Topical , Aspergillus flavus/isolation & purification , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Retrospective Studies , Skin Diseases, Bacterial/etiology , Skin Neoplasms/complications
SELECTION OF CITATIONS
SEARCH DETAIL