Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 151
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Photochem Photobiol ; 100(4): 910-922, 2024.
Article in English | MEDLINE | ID: mdl-38623955

ABSTRACT

Metastatic melanoma is an aggressive skin cancer with high mortality and recurrence rates. Despite the clinical success of recent immunotherapy approaches, prevailing resistance rates necessitate the continued development of novel therapeutic options. Photoimmunotherapy (PIT) is emerging as a promising immunotherapy strategy that uses photodynamic therapy (PDT) to unleash systemic immune responses against tumor sites while maintaining the superior tumor-specificity and minimally invasive nature of traditional PDT. In this review, we discuss recent advances in PIT and strategies for the management of melanoma using PIT. PIT can strongly induce immunogenic cell death, inviting the concomitant application of immune checkpoint blockade or adoptive cell therapies. PIT can also be leveraged to selectively remove the suppressive immune populations associated with immunotherapy resistance. The modular nature of PIT therapy design combined with the potential for patient-specific antigen selection or drug co-delivery makes PIT an alluring option for future personalized melanoma care.


Subject(s)
Immunotherapy , Melanoma , Photochemotherapy , Humans , Melanoma/therapy , Melanoma/immunology , Immunotherapy/methods , Photochemotherapy/methods , Skin Neoplasms/therapy , Skin Neoplasms/immunology , Photosensitizing Agents/therapeutic use , Animals
3.
J Immunother Cancer ; 9(4)2021 04.
Article in English | MEDLINE | ID: mdl-33883257

ABSTRACT

BACKGROUND: In the tumor microenvironment, tumor cells are able to suppress antitumor immunity by competing for essential nutrients, including amino acids. However, whether amino acid depletion modulates the activity of CD8+ tumor-infiltrating lymphocytes (TILs) is unclear. METHOD: In this study, we evaluated the roles of amino acids and the Rag complex in regulating mammalian target of rapamycin complex 1 (mTORC1) signaling in CD8+ TILs. RESULTS: We discovered that the Rag complex, particularly RagD, was crucial for CD8+ T-cell antitumor immunity. RagD expression was positively correlated with the antitumor response of CD8+ TILs in both murine syngeneic tumor xenografts and clinical human colon cancer samples. On RagD deficiency, CD8+ T cells were rendered more dysfunctional, as demonstrated by attenuation of mTORC1 signaling and reductions in proliferation and cytokine secretion. Amino acids maintained RagD-mediated mTORC1 translocation to the lysosome, thereby achieving maximal mTORC1 activity in CD8+ T cells. Moreover, the limited T-cell access to leucine (LEU), overshadowed by tumor cell amino acid consumption, led to impaired RagD-dependent mTORC1 activity. Finally, combined with antiprogrammed cell death protein 1 antibody, LEU supplementation improved T-cell immunity in MC38 tumor-bearing mice in vivo. CONCLUSION: Our results revealed that robust signaling of amino acids by RagD and downstream mTORC1 signaling were crucial for T-cell receptor-initiated antitumor immunity. The characterization the role of RagD and LEU in nutrient mTORC1 signaling in TILs might suggest potential therapeutic strategies based on the manipulation of RagD and its upstream pathway.


Subject(s)
CD8-Positive T-Lymphocytes/enzymology , Leucine/metabolism , Lymphocytes, Tumor-Infiltrating/enzymology , Mechanistic Target of Rapamycin Complex 1/metabolism , Melanoma, Experimental/enzymology , Monomeric GTP-Binding Proteins/metabolism , Skin Neoplasms/enzymology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Enzyme Activation , HEK293 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Tumor Escape , Tumor Microenvironment
4.
J Immunother Cancer ; 9(3)2021 03.
Article in English | MEDLINE | ID: mdl-33771890

ABSTRACT

Treatment with programmed cell death 1 inhibitors is associated with a wide range of cutaneous immune-related adverse events, with lichenoid eruptions representing one of the major cutaneous toxicities. We describe the case of an 81-year-old man with metastatic melanoma treated with pembrolizumab who subsequently developed a delayed-onset generalized lichenoid dermatitis. After failing multiple lines of systemic immunosuppression, narrowband ultraviolet B (NBUVB) phototherapy three times per week for 17 sessions resulted in a significant clinical response in his cutaneous eruption and was well tolerated. NBUVB is a safe, lower-cost modality that induces local, skin-specific immunosuppression without the toxicities of traditional systemic immunosuppressive agents. To date, this is the first report of use of NBUVB in immune-related lichenoid dermatitis resistant to multiple standard therapies.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Immune Checkpoint Inhibitors/adverse effects , Lichenoid Eruptions/radiotherapy , Melanoma/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Skin Neoplasms/drug therapy , Ultraviolet Therapy , Aged, 80 and over , Humans , Lichenoid Eruptions/chemically induced , Lichenoid Eruptions/immunology , Lichenoid Eruptions/pathology , Male , Melanoma/immunology , Melanoma/secondary , Programmed Cell Death 1 Receptor/immunology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Treatment Outcome
5.
Eur J Immunol ; 51(6): 1473-1481, 2021 06.
Article in English | MEDLINE | ID: mdl-33684223

ABSTRACT

Therapeutic antibodies blocking PD-1-/PD-L1 interaction have achieved remarkable clinical success in cancer. In addition to blocking a target molecule, some isotypes of antibodies can activate complement, NK cells or phagocytes, resulting in death of the cell expressing the antibody's target. Human anti-PD-1 therapeutics use antibody isotypes designed to minimize such antibody-dependent lysis. In contrast, anti-PD-1 reagents used in mice are derived from multiple species, with different isotypes, and are not engineered to reduce target cell death: few studies analyze or discuss how antibody species and isotype may impact data interpretation. We demonstrate here that anti-PD-1 therapy to promote activation and proliferation of murine PD-1-expressing CD8 T cells sometimes led instead to a loss of antigen specific cells. This phenomenon was seen in two tumor models and a model of virus infection, and varied with the clone of anti-PD-1 antibody. Additionally, we compared competition among anti-PD-1 clones to find a combination that allows detection of PD-1-expressing cells despite the presence of blocking anti-PD1 antibodies in vivo. These data bring attention to the possibility of unintended target cell depletion with some commonly used anti-mouse PD-1 clones, and should provide a valuable resource for the design and interpretation of anti-PD-1 studies in mice.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Herpesviridae Infections/immunology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Muromegalovirus/physiology , Sarcoma/immunology , Skin Neoplasms/immunology , Animals , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cell Death , Cell Line, Tumor , Cricetinae , Disease Models, Animal , Drug Evaluation, Preclinical , Herpesviridae Infections/therapy , Humans , Immunoglobulin G/metabolism , Immunoglobulin Isotypes/metabolism , Methylcholanthrene , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Rats , Sarcoma/therapy , Skin Neoplasms/therapy
6.
BMJ Case Rep ; 14(1)2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33509877

ABSTRACT

An 88-year-old Inuit man from Northern Canada presented with an extensive skin rash associated with numerous violaceous skin nodules on his palms and lower extremities. Biopsy of a skin nodule revealed Kaposi's sarcoma (KS), a human herpesvirus 8 (HHV8)-associated malignancy, whereas biopsy of the erythematous skin showed an atypical infiltrate of CD4-positive T-cells that, together with TCR gene rearrangement and presence of clonal T-cells in peripheral blood by flow cytometry, was consistent with a T-cell lymphoma, mycosis fungoides (MF) subtype. Serology was negative for HIV and HTLV-I/II and no immunodeficiency syndrome was identified. The patient was successfully treated with an oral retinoid for KS, and with topical hydrocortisone and ultraviolet B (UVB) phototherapy for MF. This case highlights the existence of HHV8-related lesions in native persons of Northern Canada, and also that MF-induced immunosuppression combined with immunosenescence may play a role in the development of non-HIV-related KS.


Subject(s)
Inuit , Mycosis Fungoides/pathology , Neoplasms, Multiple Primary/pathology , Sarcoma, Kaposi/pathology , Skin Neoplasms/pathology , Acitretin/therapeutic use , Administration, Cutaneous , Aged, 80 and over , Anti-Inflammatory Agents/therapeutic use , Herpesvirus 8, Human , Humans , Hydrocortisone/therapeutic use , Immunocompromised Host , Immunosenescence , Male , Mycosis Fungoides/immunology , Mycosis Fungoides/therapy , Neoplasms, Multiple Primary/immunology , Neoplasms, Multiple Primary/therapy , Sarcoma, Kaposi/drug therapy , Sarcoma, Kaposi/ethnology , Sarcoma, Kaposi/immunology , Skin Neoplasms/immunology , Skin Neoplasms/therapy , Ultraviolet Therapy/methods
7.
Am J Dermatopathol ; 43(4): 259-265, 2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33201017

ABSTRACT

BACKGROUND: Hypopigmented mycosis fungoides (HMF) is an uncommon variant of mycosis fungoides. AIMS: To study the clinical and histopathology presentation in children with HMF. METHOD: We reviewed 9 children diagnosed with HMF. The clinical data were collected and analyzed. RESULT: Eight boys and 1 girl were included, with a median onset age of 7.4 year old and median age of diagnosis of 10.5 year old. Multiple hypopigmented patches were observed in all patients, and 5 patients exhibited multiple scaly erythema at the center of hypopigmented patches. Histopathology showed atypical lymphocytes with hyperchromatic, irregular, and cerebriform nuclei, infiltrated in the epidermis and dermis. Pautrier's microabscesses was noted in 6 of 9 patients, and papillary dermal fibroplasia was noted in 6 of 9 patients. CD8 predominance was detected in 4 of 6 patients. Four patients were simultaneously subjected to skin biopsy on hypopigmented patches and scaly erythema simultaneously. Compared with hypopigmented specimens, erythema biopsy detected deeper and denser infiltration of atypical lymphoid cells in 3 of 4 patients, higher CD4+/CD8+ ratio in 4 of 4 patients, more CD5 loss in 2 of 4 patients, and more CD7 loss in 2 of 4 patients. TCR gene monoclonal rearrangement was detected in 2 of 5 patients. Narrowband ultraviolet B phototherapy was applied in 7 patients. One of 7 patients achieved complete response, and 6 of 7 patients achieved partial response. No recurrence was noted with the median follow-up period of 6 months. CONCLUSION: HMF could occur in young patients, with indolent and benign course. HMF could gradually seem as scaly erythema based on hypopigmented patches. The histopathology indicated a more advanced stage of the scaly erythema lesions than hypopigmented patches.


Subject(s)
Hypopigmentation/pathology , Mycosis Fungoides/pathology , Skin Neoplasms/pathology , Skin Pigmentation , Biomarkers, Tumor/genetics , Child , Child, Preschool , Female , Gene Rearrangement, T-Lymphocyte , Genes, T-Cell Receptor , Humans , Hypopigmentation/genetics , Hypopigmentation/immunology , Hypopigmentation/radiotherapy , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mycosis Fungoides/genetics , Mycosis Fungoides/immunology , Mycosis Fungoides/radiotherapy , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/radiotherapy , Skin Pigmentation/radiation effects , Treatment Outcome , Ultraviolet Therapy
8.
Medicine (Baltimore) ; 99(42): e22786, 2020 Oct 16.
Article in English | MEDLINE | ID: mdl-33080750

ABSTRACT

RATIONALE: Mycosis fungoides (MF) is the most common cutaneous T-cell lymphoma. It appears as patches, plaques, and tumors depending on the stage of the disease, which presents a chronic progressive course. Compared to CD4/CD8 MF, CD4/CD8 dual-positive MF is an uncommon immune phenotype. PATIENT CONCERNS: A 36-year-old male patient presented with dryness and scales on his whole body. DIAGNOSIS: The patient was diagnosed with MF based on results of pathological examination, immunohistochemical staining, and T-cell receptor gene rearrangement test. INTERVENTIONS: The patient was advised to take an herbal medicine orally twice daily and apply a topical moisturizer after showering. OUTCOMES: After treatment and follow-up, the patient's symptoms of dryness and scales improved and his condition stabilized. CONCLUSIONS: While reviewing the literature, we found no previous reports on the treatment of dual-positive MF with Chinese medicine. In this report, we presented the first case of dual-positive MF successfully treated with Chinese medicine. The results suggest that oral ingestion of herbal medicine may be a feasible method for alleviating clinical symptoms of early stage MF. Therefore, the therapy should be explored for clinical use in the future.


Subject(s)
CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Mycosis Fungoides/pathology , Skin Neoplasms/pathology , Adult , Humans , Immunohistochemistry , Male , Mycosis Fungoides/immunology , Skin Neoplasms/immunology
9.
Carbohydr Polym ; 246: 116588, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32747247

ABSTRACT

Hot water extract from biomass of heterotrophic mutant green alga Parachlorella kessleri HY1 (Chlorellaceae) was deproteinised, and three polysaccharidic fractions were obtained by preparative chromatography. The low-molecular fraction (1.5 × 104g mol-1) was defined mainly as branched O-2-ß-xylo-(1→3)-ß-galactofuranan where xylose is partially methylated at O-4. Two high-molecular fractions (3.05 × 105 and 9.84 × 104g mol-1) were complex polysaccharides containing α-l-rhamnan and xylogalactofuranan parts in different ratios. The polysaccharides were well soluble in hot water and, upon cooling, tended to self-segregate. Immunomodulatory activities of the obtained fractions were preliminary tested using ELISA, FACS and ImmunoSpot kits. The polysaccharides increased the TNF-α production in melanoma bearing mice with much higher intensity than in healthy mice. This was in agreement with the FACS results on T and B cells indicating their possibly secondary activation by innate immunity cells.


Subject(s)
B-Lymphocytes/drug effects , Chlorophyta/chemistry , Immunologic Factors/pharmacology , Polysaccharides/pharmacology , T-Lymphocytes/drug effects , Animals , Antigens, CD/genetics , Antigens, CD/immunology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Carbohydrate Sequence , Gene Expression Regulation/drug effects , Immunologic Factors/chemistry , Immunologic Factors/isolation & purification , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Melanoma/immunology , Melanoma/pathology , Methylation , Mice , Mice, Inbred C57BL , Molecular Weight , Plant Extracts/chemistry , Polysaccharides/chemistry , Polysaccharides/isolation & purification , Primary Cell Culture , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Solubility , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Water , Xylose/chemistry , Xylose/isolation & purification
10.
J Dermatol Sci ; 99(3): 146-151, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32600738

ABSTRACT

Skin diseases bring great psychological and physical impacts on patients, however, a considerable number of skin diseases still lack effective treatments, such as psoriasis, systemic lupus erythematosus, melanoma and so on. Receptor-interacting serine threonine kinase 1 (RIPK1) plays an important role in cell death, especially necroptosis, associated with inflammation and tumor. As many molecules modulate the ubiquitination of RIPK1, disruption of this checkpoint can lead to skin diseases, which can be ameliorated by RIPK1 inhibitors. This review will focus on the molecular mechanism of RIPK1 activation in inflammation as well as the current knowledges on the contribution of RIPK1 in skin diseases.


Subject(s)
Dermatitis/immunology , Necroptosis/immunology , Protein Kinase Inhibitors/therapeutic use , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Skin Neoplasms/immunology , Animals , Clinical Trials, Phase II as Topic , Dermatitis/drug therapy , Dermatitis/genetics , Dermatitis/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Half-Life , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Mice , Mice, Knockout , Necroptosis/drug effects , Necroptosis/genetics , Oxazepines/pharmacology , Oxazepines/therapeutic use , Protein Kinase Inhibitors/pharmacology , Rats , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Skin/drug effects , Skin/immunology , Skin/pathology , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Treatment Outcome , Triazoles/pharmacology , Triazoles/therapeutic use , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Ubiquitination/immunology
11.
Cancer Immunol Immunother ; 69(12): 2589-2598, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32591862

ABSTRACT

BACKGROUND: Autologous monocyte-derived mRNA co-electroporated dendritic cells with mRNA encoding CD40 ligand (CD40L), CD70 and a constitutively activated TLR4 (caTLR4) (referred to as TriMixDC-MEL) have anti-tumor activity in advanced melanoma patients. We investigated the safety and activity of adjuvant TriMixDC-MEL in stage III/IV melanoma patients. MATERIALS AND METHODS: Forty-one patients were randomly assigned to treatment with TriMixDC-MEL (n = 21) and standard follow-up (n = 20). "Cross-over" was allowed at the time of non-salvageable recurrence. The primary endpoint was the percentage of patients alive and disease-free at 1-year. For a subset of patients, (formalin-fixed paraffin-embedded), tumor tissue samples were available for mRNA expression profiling and PD-L1 immunohistochemical staining. RESULTS: Baseline characteristics were well balanced. One-year after randomization, 71% of patients in the study arm were alive and free of disease compared to 35% in the control arm. After a median follow-up of 53 months (range 3-67), 23 patients experienced a non-salvageable melanoma recurrence (TriMixDC-Mel arm n = 9 and control arm n = 14).The median time to non-salvageable recurrence was superior in the TriMixDC-MEL arm (median 8 months (range 1-6) vs. not reached; log-rank p 0.044). TriMixDC-MEL-related adverse events (AE) consisted of transient local skin reactions, flu-like symptoms and post-infusion chills. No grade ≥ 3 AE's occurred. The mRNA expression profiling revealed four genes (STAT2, TPSAB1, CD9 and CSF2) as potential predictive biomarkers. CONCLUSION: TriMixDC-MEL id/iv as adjuvant therapy is tolerable and may improve the 1-year disease-free survival rate. Combination of optimized autologous monocyte-derived DC-formulations warrants further investigation in combination with currently approved adjuvant therapy options.


Subject(s)
Dendritic Cells/transplantation , Melanoma/therapy , Neoplasm Recurrence, Local/epidemiology , RNA, Messenger/immunology , Skin Neoplasms/therapy , Adult , Aged , Aged, 80 and over , CD27 Ligand/genetics , CD27 Ligand/immunology , CD40 Ligand/genetics , CD40 Ligand/immunology , Combined Modality Therapy/methods , Dendritic Cells/metabolism , Disease-Free Survival , Electroporation , Female , Follow-Up Studies , Humans , Immunotherapy/methods , Male , Melanoma/immunology , Melanoma/mortality , Melanoma/secondary , Middle Aged , Neoplasm Recurrence, Local/prevention & control , Neoplasm Staging , RNA, Messenger/genetics , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Surgical Procedures, Operative , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Transplantation, Autologous/methods , Young Adult
13.
J Immunother Cancer ; 8(1)2020 04.
Article in English | MEDLINE | ID: mdl-32350119

ABSTRACT

BACKGROUND: Immunogenicity of cancer vaccines is impacted by adjuvants and schedule, but systematic assessments of their effects have not been performed. Montanide ISA-51, an incomplete Freund's adjuvant (IFA), is used in many vaccine trials, but concerns have been raised about negative effects in murine studies. We found in humans that IFA enhances systemic immune responses and that repeat vaccination at one site (same site vaccination (SSV)) creates tertiary lymphoid structures (TLS) in the vaccine site microenvironment (VSME). We hypothesized that vaccination with peptides+IFA+pICLC or SSV×3 with peptides in IFA would create an immunogenic milieu locally at the VSME, with activated dendritic cells (DC), TLS-associated chemokines and a Th1-dominant VSME. METHODS: Biopsies of the VSME were obtained from participants on two clinical trials who were immunized with multiple melanoma peptides (MELITAC 12.1) in adjuvants comprising IFA and/or the TLR3-agonist pICLC. Biopsies were obtained either a week after one vaccine or a week after SSV×3. Controls included normal skin and skin injected with IFA without peptides. Gene expression analysis was performed by RNAseq. RESULTS: VSME samples were evaluated from 27 patients. One vaccine with peptides in pICLC+IFA enhanced expression of CD80, CD83, CD86 (p<0.01), CD40 and CD40L (p<0.0001) over normal skin; these effects were significantly enhanced for SSV with peptides+IFA. Vaccines containing pICLC increased expression of TBX21 (T-bet) but did not decrease GATA3 over normal skin, whereas SSV with peptides in IFA dramatically enhanced TBX21 and decreased GATA3, with high expression of IFNγ and STAT1. SSV with peptides in IFA also reduced arginase-1 (ARG1) expression and enhanced expression of TLR adapter molecules TICAM-1 (TRIF) and MYD88. Furthermore, SSV with IFA and peptides also enhanced expression of chemokines associated with TLS formation. CONCLUSIONS: These findings suggest that SSV with peptides in IFA enhances CD40L expression by CD4 T cells, supports a Th1 microenvironment, with accumulation of activated and mature DC. Increased expression of TLR adaptor proteins after SSV with peptides in IFA might implicate effects of the skin microbiome. Reduced ARG1 may reflect diminished suppressive myeloid activity in the VSME. TRIAL REGISTRATION NUMBER: (NCT00705640, NCT01585350).


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cancer Vaccines/administration & dosage , Freund's Adjuvant/administration & dosage , Lipids/administration & dosage , Melanoma/therapy , Skin Neoplasms/therapy , Vaccination/methods , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/immunology , Arginase/metabolism , Biopsy , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD40 Ligand/immunology , CD40 Ligand/metabolism , Cancer Vaccines/immunology , Clinical Trials, Phase I as Topic , Female , Freund's Adjuvant/immunology , Humans , Immunization, Secondary/methods , Immunogenicity, Vaccine , Injections, Intralesional , Lipids/immunology , Male , Mannitol/administration & dosage , Mannitol/analogs & derivatives , Mannitol/immunology , Melanoma/immunology , Melanoma/pathology , Middle Aged , Oleic Acids/administration & dosage , Oleic Acids/immunology , Randomized Controlled Trials as Topic , Signal Transduction/drug effects , Signal Transduction/immunology , Skin/immunology , Skin/pathology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Th1 Cells/drug effects , Th1 Cells/immunology , Toll-Like Receptors/metabolism , Tumor Microenvironment/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Young Adult
14.
Biochim Biophys Acta Rev Cancer ; 1874(1): 188380, 2020 08.
Article in English | MEDLINE | ID: mdl-32461135

ABSTRACT

Cellular communication through gap junctions and hemichannels formed by connexins and through channels made by pannexins allows for metabolic cooperation and control of cellular activity and signalling. These channel proteins have been described to be tumour suppressors that regulate features such as cell death, proliferation and differentiation. However, they display cancer type-dependent and stage-dependent functions and may facilitate tumour progression through junctional and non-junctional pathways. The accumulated knowledge and emerging strategies to target connexins and pannexins are providing novel clinical opportunities for the treatment of cancer. Here, we provide an updated overview of the role of connexins and pannexins in malignant melanoma. We discuss how targeting of these channel proteins may be used to potentiate antitumour effects in therapeutic settings, including through improved immune-mediated tumour elimination.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Connexins/metabolism , Melanoma/secondary , Skin Neoplasms/pathology , Skin/pathology , Animals , Antineoplastic Agents, Immunological/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/immunology , Carcinogenesis/pathology , Cell Communication/drug effects , Cell Communication/immunology , Cell Line, Tumor , Connexins/agonists , Connexins/antagonists & inhibitors , Disease Models, Animal , Disease Progression , Gap Junctions/drug effects , Gap Junctions/pathology , Host Microbial Interactions/drug effects , Host Microbial Interactions/immunology , Humans , Melanoma/drug therapy , Melanoma/immunology , Melanoma/mortality , Microbiota/immunology , Neoplasm Invasiveness/immunology , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control , Neoplasm Staging , Signal Transduction/drug effects , Signal Transduction/immunology , Skin/cytology , Skin/microbiology , Skin Neoplasms/drug therapy , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
15.
J Intern Med ; 288(6): 625-640, 2020 12.
Article in English | MEDLINE | ID: mdl-32128919

ABSTRACT

The incidence of cutaneous melanoma and the mortality rate of advanced melanoma patients continue to rise globally. Despite the recent success of immunotherapy including ipilimumab and pembrolizumab checkpoint inhibitors, a large proportion of patients are refractory to such treatment modalities. The application of mycobacteria such as Bacillus Calmette-Guérin (BCG) in the treatment of various malignancies, including cutaneous melanoma, has been clearly demonstrated after almost a century of observations and experimentation. Intralesional BCG (IL-BCG) immunotherapy is a highly efficient and cost-effective treatment option for inoperable stage III in-transit melanoma, as recommended in the National Comprehensive Cancer Network Guidelines. IL-BCG has shown great efficacy in the regression of directly injected metastatic melanoma lesions, as well as distal noninjected nodules in immunocompetent patients. Clinical and preclinical studies have shown that BCG serves as a strong immune modulator, inducing the recruitment of various immune cells that contribute to antitumour immunity. However, the specific mechanism of BCG-mediated tumour immunity remains poorly understood. Comparative genome analyses have revealed that different BCG strains exhibit distinct immunological activity and virulence, which might impact the therapeutic response and clinical outcome of patients. In this review, we discuss the immunostimulatory potential of different BCG substrains and highlight clinical studies utilizing BCG immunotherapy for the treatment of cutaneous melanoma. Furthermore, the review focuses on the cellular and molecular mechanisms of the BCG-induced immune responses of both the innate and adaptive arms of the immune system. Furthermore, the review discussed the administration of BCG as a monotherapy or in combination with other immunotherapeutic or chemotherapeutic agents.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , BCG Vaccine/therapeutic use , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Adaptive Immunity , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/therapeutic use , Animals , Antineoplastic Agents, Immunological/adverse effects , BCG Vaccine/adverse effects , BCG Vaccine/immunology , Humans , Immunity, Innate , Melanoma/immunology , Melanoma/pathology , Mycobacterium bovis/immunology , Neoplasm Metastasis , Neoplasm Staging , Neoplasms/drug therapy , Neoplasms/veterinary , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Melanoma, Cutaneous Malignant
16.
Int Immunopharmacol ; 80: 106123, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31927505

ABSTRACT

Accumulating evidence has shown that SHC SH2 domain-binding protein 1 (SHCBP1) functions as an oncogene and participated in the progression of various cancers. Oroxylin A, an active ingredient extracted from Chinese Medicine Scutellaria baicalensis, shows strong anticancer effects on multiple cancers, however, the pharmacological effect of oroxylin A on skin cancer and the regulatory effect of SHCBP1 on this process have never been evaluated. The present study was aimed at elucidating the effect of oroxylin A on carcinogen (DMBA/TPA)-induced skin tumorigenesis, and to further clarify the role of SHCBP1 in oroxylin A induced antitumor effect. Pretreatment with oroxylin A remarkably inhibited DMBA/TPA-induced tumor formation and growth, and significantly reduced tumor incidence and the average number of tumors per mouse. Oroxylin A suppressed DMBA/TPA-induced skin hyperplasia and tumor proliferation. Oroxylin A significantly inhibited the expression of several inflammatory factors in vivo. In vitro experiments found that oroxylin A inhibited TPA-induced cell malignant transformation of skin epidermal JB6 P + cells. Besides, oroxylin A significantly suppressed the levels of TPA-induced inflammatory factors in vitro. Mechanistic studies showed that oroxylin A remarkably inhibited TPA-induced increased expression of SHCBP1. Overexpression of SHCBP1 attenuated the oroxylin A-induced anti-inflammatory effect. In addition, TPA increased the expression of nuclear NF-κB p65, and SHCBP1 siRNA notably decreased the nuclear NF-κB p65 expression in JB6 P + cells. Collectively, the anti-skin cancer effect of oroxylin A may possibly by inhibiting inflammation via suppression of SHCBP1. Oroxylin A might be a potential candidate compound for the treatment of skin cancer.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Flavonoids/therapeutic use , Shc Signaling Adaptor Proteins/immunology , Skin Neoplasms/drug therapy , 9,10-Dimethyl-1,2-benzanthracene , Animals , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Carcinogens , Cell Line , Cytokines/genetics , Cytokines/immunology , Female , Flavonoids/pharmacology , Mice, Inbred ICR , RNA, Small Interfering/genetics , Shc Signaling Adaptor Proteins/genetics , Skin Neoplasms/chemically induced , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate , Transcription Factor RelA/immunology
17.
Front Immunol ; 11: 623052, 2020.
Article in English | MEDLINE | ID: mdl-33613558

ABSTRACT

Humans are exposed to various external environmental factors. Food intake is one of the most influential factors impacting daily lifestyle. Among nutrients obtained from foods, omega-3 polyunsaturated fatty acids (PUFAs) have various beneficial effects on inflammatory diseases. Furthermore, omega-3 PUFA metabolites, including resolvins, are known to demonstrate strong anti-inflammatory effects during allergic and inflammatory diseases; however, little is known regarding the actual impact of these metabolites on skin diseases. In this review, we focused on metabolites that have strong anti-inflammatory actions in various inflammatory diseases, as well as those that present antitumor actions in malignancies, in addition to the actual effect of omega-3 PUFA metabolites on various cells.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Fatty Acids, Omega-3/therapeutic use , Skin Neoplasms/drug therapy , Humans , Skin Neoplasms/immunology , Skin Neoplasms/pathology
18.
Immunology ; 159(4): 373-383, 2020 04.
Article in English | MEDLINE | ID: mdl-31821535

ABSTRACT

The anti-tumor immune response is considered to be due to the T-cell receptor (TCR) binding to tumor antigens, which can be either wild-type, early stem cell proteins, presumably foreign to a developed immune system; or mutant peptides, foreign to the immune system because of a mutant amino acid (aa) or otherwise somatically altered aa sequence. Recently, very large numbers of TCR complementarity-determining region-3 (CDR3) aa sequences obtained from tumor specimens have become available. We developed a novel algorithm for assessing the complementarity of tumor mutant peptides and TCR CDR3s, based on the retrieval of TCR CDR3 aa sequences from both tumor specimen and patient blood exomes and by using an automated process of assessing CDR3 and mutant aa electrical charges. Results indicated many instances where high electrostatic complementarity was associated with a higher survival rate. In particular, our approach led to the identification of specific genes contributing significantly to the complementary, TCR CDR3-mutant aa. These results suggest a novel approach to tumor immunoscoring and may lead to the identification of high-priority neo-antigen, peptide vaccines; or to the identification of ex vivo stimulants of tumor-infiltrating lymphocytes.


Subject(s)
Algorithms , Antigens, Neoplasm/chemistry , Breast Neoplasms/genetics , Complementarity Determining Regions/chemistry , Lung Neoplasms/genetics , Receptor-CD3 Complex, Antigen, T-Cell/chemistry , Skin Neoplasms/genetics , Amino Acid Sequence , Amino Acids , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Binding Sites , Breast Neoplasms/diagnosis , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Exome , Female , Gene Expression , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Male , Mutation , Prognosis , Protein Binding , Receptor-CD3 Complex, Antigen, T-Cell/genetics , Receptor-CD3 Complex, Antigen, T-Cell/immunology , Research Design , Skin Neoplasms/diagnosis , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Static Electricity , Survival Rate , T-Lymphocytes/immunology , T-Lymphocytes/pathology
19.
Int J Cancer ; 146(5): 1409-1420, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31702822

ABSTRACT

Therapeutic success of targeted therapy with BRAF inhibitors (BRAFi) for melanoma is limited by resistance development. Observations from preclinical mouse models and recent insights into the immunological effects caused by BRAFi give promise for future development of combination therapy for human melanoma. In our study, we used the transplantable D4M melanoma mouse model with the BRAFV600E mutation and concomitant PTEN loss in order to characterize alterations in tumor-infiltrating effector immune cells when tumors become resistant to BRAFi. We found that BRAFi-sensitive tumors displayed a pronounced inflammatory milieu characterized by high levels of cytokines and chemokines accompanied by an infiltration of T and NK cells. The tumor-infiltrating effector cells were activated and produced high levels of IFN-γ, TNF-α and granzyme B. When tumors became resistant and progressively grew, they reverted to a low immunogenic state similar to untreated tumors as reflected by low mRNA levels of proinflammatory cytokines and chemokines and fewer tumor-infiltrating T and NK cells. Moreover, these T and NK cells were functionally impaired in comparison to their counterparts in BRAFi-sensitive tumors. Their effector cell function could be restored by additional peritumoral treatment with the TLR7 agonist imiquimod, a clinically approved agent for nonmelanoma skin cancer. Indeed, resistance to BRAFi therapy was delayed and accompanied by high numbers of activated T and NK cells in tumors. Thus, combining BRAFi with an immune stimulating agent such as a TLR ligand could be a promising alternative approach for the treatment of melanoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Melanoma, Experimental/drug therapy , Skin Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor/transplantation , Drug Evaluation, Preclinical , Drug Synergism , Female , Humans , Imiquimod/pharmacology , Imiquimod/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Membrane Glycoproteins/agonists , Membrane Glycoproteins/metabolism , Mice , Mutation , Natural Killer T-Cells , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/metabolism
20.
Eur J Cancer ; 123: 58-71, 2019 12.
Article in English | MEDLINE | ID: mdl-31670077

ABSTRACT

BACKGROUND: Although a myriad of novel treatments entered the treatment paradigm for advanced melanoma, there is lack of head-to-head evidence. We conducted a network meta-analysis (NMA) to estimate each treatment's relative effectiveness and safety. METHODS: A systematic literature review (SLR) was conducted in Embase, MEDLINE and Cochrane to identify all phase III randomised controlled trials (RCTs) with a time frame from January 1, 2010 to March 11, 2019. We retrieved evidence on treatment-related grade III/IV adverse events, progression-free survival (PFS) and overall survival (OS). Evidence was synthesised using a Bayesian fixed-effect NMA. Reference treatment was dacarbazine. In accordance with RCTs, dacarbazine was pooled with temozolomide, paclitaxel and paclitaxel plus carboplatin. To increase homogeneity of the study populations, RCTs were only included if patients were not previously treated with novel treatments. RESULTS: The SLR identified 28 phase III RCTs involving 14,376 patients. Nineteen and seventeen treatments were included in the effectiveness and safety NMA, respectively. For PFS, dabrafenib plus trametinib (hazard ratio [HR] PFS: 0.21) and vemurafenib plus cobimetinib (HR PFS: 0.22) were identified as most favourable treatments. Both had, however, less favourable safety profiles. Five other treatments closely followed (dabrafenib [HR PFS: 0.30], nivolumab plus ipilimumab [HR PFS: 0.34], vemurafenib [HR PFS: 0.38], nivolumab [HR PFS: 0.42] and pembrolizumab [HR PFS: 0.46]). In contrast, for OS, nivolumab plus ipilimumab (HR OS: 0.39), nivolumab (HR OS: 0.46) and pembrolizumab (HR OS: 0.50) were more favourable than dabrafenib plus trametinib (HR OS: 0.55) and vemurafenib plus cobimetinib (HR OS: 0.57). CONCLUSIONS: Our NMA identified the most effective treatment options for advanced melanoma and provided valuable insights into each novel treatment's relative effectiveness and safety. This information may facilitate evidence-based decision-making and may support the optimisation of treatment and outcomes in everyday clinical practice.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cancer Vaccines/therapeutic use , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/therapeutic use , Azetidines/administration & dosage , Azetidines/therapeutic use , Benzimidazoles/administration & dosage , Benzimidazoles/therapeutic use , Carboplatin/administration & dosage , Carboplatin/therapeutic use , Dacarbazine/administration & dosage , Dacarbazine/therapeutic use , Humans , Hydrazines/administration & dosage , Hydrazines/therapeutic use , Imidazoles/administration & dosage , Imidazoles/therapeutic use , Interleukin-2/administration & dosage , Interleukin-2/therapeutic use , Ipilimumab/administration & dosage , Ipilimumab/therapeutic use , Lenalidomide/administration & dosage , Lenalidomide/therapeutic use , Melanoma/immunology , Melanoma/pathology , Network Meta-Analysis , Nitrosourea Compounds/administration & dosage , Nitrosourea Compounds/therapeutic use , Nivolumab/administration & dosage , Nivolumab/therapeutic use , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/therapeutic use , Oximes/administration & dosage , Oximes/therapeutic use , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , Piperidines/administration & dosage , Piperidines/therapeutic use , Progression-Free Survival , Proportional Hazards Models , Pyridones/administration & dosage , Pyridones/therapeutic use , Pyrimidinones/administration & dosage , Pyrimidinones/therapeutic use , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Sorafenib/administration & dosage , Sorafenib/therapeutic use , Survival Rate , Temozolomide/administration & dosage , Temozolomide/therapeutic use , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL