Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Nature ; 589(7843): 597-602, 2021 01.
Article in English | MEDLINE | ID: mdl-33361818

ABSTRACT

Isoprenoids are vital for all organisms, in which they maintain membrane stability and support core functions such as respiration1. IspH, an enzyme in the methyl erythritol phosphate pathway of isoprenoid synthesis, is essential for Gram-negative bacteria, mycobacteria and apicomplexans2,3. Its substrate, (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMBPP), is not produced in metazoans, and in humans and other primates it activates cytotoxic Vγ9Vδ2 T cells at extremely low concentrations4-6. Here we describe a class of IspH inhibitors and refine their potency to nanomolar levels through structure-guided analogue design. After modification of these compounds into prodrugs for delivery into bacteria, we show that they kill clinical isolates of several multidrug-resistant bacteria-including those from the genera Acinetobacter, Pseudomonas, Klebsiella, Enterobacter, Vibrio, Shigella, Salmonella, Yersinia, Mycobacterium and Bacillus-yet are relatively non-toxic to mammalian cells. Proteomic analysis reveals that bacteria treated with these prodrugs resemble those after conditional IspH knockdown. Notably, these prodrugs also induce the expansion and activation of human Vγ9Vδ2 T cells in a humanized mouse model of bacterial infection. The prodrugs we describe here synergize the direct killing of bacteria with a simultaneous rapid immune response by cytotoxic γδ T cells, which may limit the increase of antibiotic-resistant bacterial populations.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/immunology , Lymphocyte Activation/drug effects , Microbial Viability/drug effects , Oxidoreductases/antagonists & inhibitors , T-Lymphocytes, Cytotoxic/drug effects , Animals , Drug Resistance, Microbial , Drug Resistance, Multiple , Enzyme Inhibitors/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Female , Half-Life , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/microbiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microbial Sensitivity Tests , Molecular Docking Simulation , Oxidoreductases/deficiency , Oxidoreductases/genetics , Oxidoreductases/metabolism , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Substrate Specificity , Swine/blood , T-Lymphocytes, Cytotoxic/immunology
2.
Nature ; 560(7718): 382-386, 2018 08.
Article in English | MEDLINE | ID: mdl-30089911

ABSTRACT

Tumour cells evade immune surveillance by upregulating the surface expression of programmed death-ligand 1 (PD-L1), which interacts with programmed death-1 (PD-1) receptor on T cells to elicit the immune checkpoint response1,2. Anti-PD-1 antibodies have shown remarkable promise in treating tumours, including metastatic melanoma2-4. However, the patient response rate is low4,5. A better understanding of PD-L1-mediated immune evasion is needed to predict patient response and improve treatment efficacy. Here we report that metastatic melanomas release extracellular vesicles, mostly in the form of exosomes, that carry PD-L1 on their surface. Stimulation with interferon-γ (IFN-γ) increases the amount of PD-L1 on these vesicles, which suppresses the function of CD8 T cells and facilitates tumour growth. In patients with metastatic melanoma, the level of circulating exosomal PD-L1 positively correlates with that of IFN-γ, and varies during the course of anti-PD-1 therapy. The magnitudes of the increase in circulating exosomal PD-L1 during early stages of treatment, as an indicator of the adaptive response of the tumour cells to T cell reinvigoration, stratifies clinical responders from non-responders. Our study unveils a mechanism by which tumour cells systemically suppress the immune system, and provides a rationale for the application of exosomal PD-L1 as a predictor for anti-PD-1 therapy.


Subject(s)
B7-H1 Antigen/immunology , Exosomes/metabolism , Immune Tolerance/immunology , Melanoma/immunology , Programmed Cell Death 1 Receptor/immunology , Tumor Escape/immunology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/blood , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Case-Control Studies , Cell Line, Tumor , Disease Progression , Female , Humans , Immune Tolerance/drug effects , Interferon-gamma/blood , Interferon-gamma/immunology , Melanoma/drug therapy , Melanoma/pathology , Mice , Mice, Nude , Neoplasm Metastasis , Prognosis , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Escape/drug effects , Xenograft Model Antitumor Assays
3.
J Transl Med ; 20(1): 112, 2022 03 07.
Article in English | MEDLINE | ID: mdl-35255925

ABSTRACT

BACKGROUND: Triple negative breast cancer (TNBC) is an aggressive breast cancer for which there is currently no targeted therapy. Tumor-infiltrating B-cells (TIB) have been observed in tumor tissues of TNBC patients, but their functional role is unclear. IgG4 is one of four antibody subclasses of IgG expressed and secreted by B cells. Unlike other IgG isotypes, IgG4 has an immunosuppressive function and is induced by Th2-type cytokines. In cancers such as melanoma, IgG4 has been linked with advanced disease and poor patient survival. Therefore, we sought to determine if IgG4 + B cells are present and determine the mechanisms driving isotype switching in TNBC. METHODS: We performed co-culture assays to examine expression of Th2 cytokines by TNBC cells with and without the presence of B cells. We also performed in vitro class switching experiments with peripheral B cells with and without co-culture with TNBC cells in the presence or absence of an IL-10 blocking antibody. We examined expression of CD20+ TIB, IgG4 and Th2 cytokines by immunohistochemistry in 152 TNBC samples. Statistical analysis was done using Log-Rank and Cox-proportional hazards tests. RESULTS: Our findings indicate that B cells interact with TNBC to drive chronic inflammatory responses through increased expression of inflammatory cytokines including the TH2 cytokines IL-4 and IL-10. In vitro class switching studies show that interactions between TNBC cell lines and B cells drive isotype switching to the IgG4 isotype in an IL-10 dependent manner. In patient tissues, expression of IgG4 correlates with CD20 and tumor expression of IL-10. Both IgG4 and tumor IL-10 are associated to shorter recurrence free survival (RFS) and overall survival (OS) in TNBC. In a multi-variant analysis, IL-10 was associated with poor outcomes indicating that tumor IL-10 may drive immune escape. CONCLUSIONS: These findings indicate that interactions between TIB and TNBC results in activation of chronic inflammatory signals such as IL-10 and IL-4 that drive class switching to an IgG4 + subtype which may suppress antibody driven immune responses. The presence of IgG4 + B cells may serve as a biomarker for poor prognosis.


Subject(s)
Triple Negative Breast Neoplasms , Antibody Formation , Cell Communication , Cell Line, Tumor , Humans , Immunoglobulin Class Switching , Immunoglobulin G/metabolism , Interleukin-10/metabolism , Triple Negative Breast Neoplasms/genetics , Up-Regulation
5.
Cancer Treat Res ; 167: 1-15, 2016.
Article in English | MEDLINE | ID: mdl-26601857

ABSTRACT

Melanoma is among the most aggressive and therapy-resistant human cancers. While great strides in therapy have generated enthusiasm, many challenges remain. Heterogeneity is the most pressing issue for all types of therapy. This chapter summarizes the clinical classification of melanoma, of which the research community now adds additional layers of classifications for better diagnosis and prediction of therapy response. As the search for new biomarkers increases, we expect that biomarker analyses will be essential for all clinical trials to better select patient populations for optimal therapy. While individualized therapy that is based on extensive biomarker analyses is an option, we expect in the future genetic and biologic biomarkers will allow grouping of melanomas in such a way that we can predict therapy outcome. At this time, tumor heterogeneity continues to be the major challenge leading inevitably to relapse. To address heterogeneity therapeutically, we need to develop complex therapies that eliminate the bulk of the tumor and, at the same time, the critical subpopulations.


Subject(s)
Melanoma/classification , Biomarkers, Tumor/analysis , Humans , Melanoma/genetics , Melanoma/pathology , Melanoma/therapy
6.
Mol Cell Proteomics ; 12(12): 3778-92, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24037664

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a key contributor in tumor progression and metastasis. EMT produces cellular heterogeneity within head and neck squamous cell carcinomas (HNSCC) by creating a phenotypically distinct mesenchymal subpopulation that is resistant to conventional therapies. In this study, we systematically characterized differences in the secretomes of E-cadherin high epithelial-like and E-cadherin low mesenchymal-like subpopulations using unbiased and targeted proteomics. A total 1765 proteins showed significant changes with 177 elevated in the epithelial subpopulation and 173 elevated in the mesenchymal cells. Key nodes in affected networks included NFκB, Akt, and ERK, and most implicated cellular components involved various aspects of the extracellular matrix. In particular, large changes were observed in multiple collagens with most affected collagens at much higher abundance levels in the mesenchymal subpopulation. These cells also exhibited a secretome profile resembling that of cancer-associated fibroblastic cells (CAF). S100A4, a commonly used marker for cancer-associated fibroblastic cells, was elevated more than 20-fold in the mesenchymal cells and this increase was further verified at the transcriptome level. S100A4 is a known mediator of EMT, leading to metastasis and EMT has been proposed as a potential source of cancer-associated fibroblastic cells in solid tumors. S100A4 knockdown by small interfering RNA led to decreased expression, secretion and activity of matrix metalloproteinase 2, as verified by quantitative PCR, multiple reaction monitoring and zymography analyses, and reduced invasion in collagen-embedded spheroids. Further confirmation in three-dimensional organotypic reconstructs showed less invasion and advanced differentiation in the S100A4 RNA interference samples. Orthotopic metastasis model, developed to validate the findings in vivo, demonstrated a decrease in spontaneous metastasis and augmented differentiation in the primary tumor in siS100A4 xenografts. These results demonstrate the value of secretome profiling to evaluate phenotypic conversion and identify potential novel therapeutic targets such as S100A4.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cell Transformation, Neoplastic/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , S100 Proteins/genetics , Animals , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Clone Cells , Collagen/genetics , Collagen/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Molecular Sequence Annotation , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Transplantation , Protein Interaction Mapping , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , S100 Calcium-Binding Protein A4 , S100 Proteins/antagonists & inhibitors , S100 Proteins/metabolism , Signal Transduction , Squamous Cell Carcinoma of Head and Neck
7.
Mol Ther ; 20(5): 1056-62, 2012 May.
Article in English | MEDLINE | ID: mdl-22354376

ABSTRACT

Melanomas contain distinct cell subpopulations. Several of these subpopulations, including one expressing CD20, may harbor stem cell-like or tumor-initiating characteristics. We hypothesized that patients at high risk of disease recurrence could benefit from an adjuvant anti-CD20 therapy. Therefore, we initiated a small pilot trial to study the effect of the anti-CD20 antibody rituximab in a group of melanoma patients with stage IV metastatic disease who had been rendered without evident disease by way of surgery, chemotherapy and/or radiation therapy. The major objective was safety, while secondary objectives were description of recurrence-free intervals (RFI) and overall survival (OS). Nine patients received rituximab at 375 mg/m(2) qw for 4 weeks followed by a maintenance therapy every 8 weeks. Treatment was discontinued after 2 years or with disease recurrence. Treatment was well tolerated. After a median observation of 42 months, the median neither of RFI nor of OS has been reached. Despite therapy that ended after 2 years, six out of nine patients are still alive and five of them are recurrence-free. Though the patient number is too small for definitive conclusions, our data may represent a first example of the potential therapeutic value of targeting CD20(+) cell populations-at least for a subset of patients.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antigens, CD20/immunology , Antineoplastic Agents/administration & dosage , Melanoma/therapy , Adult , Aged , Combined Modality Therapy , Disease-Free Survival , Drug Administration Schedule , Female , Humans , Male , Melanoma/immunology , Melanoma/mortality , Melanoma/prevention & control , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Pilot Projects , Risk , Rituximab , Secondary Prevention
8.
J Invest Dermatol ; 142(7): 1882-1892.e5, 2022 07.
Article in English | MEDLINE | ID: mdl-34883044

ABSTRACT

The upregulation of the adaptor protein NUMB triggers melanocytic differentiation from multipotent skin stem cells, which share many properties with aggressive melanoma cells. Although NUMB acts as a tumor suppressor in various human cancer types, little is known about its role in melanoma. In this study, we investigated the role of NUMB in melanoma progression and its regulatory mechanism. Analysis of The Cancer Genome Atlas melanoma datasets revealed that high NUMB expression in melanoma tissues correlates with improved patient survival. Moreover, NUMB expression is downregulated in metastatic melanoma cells. NUMB knockdown significantly increased the invasion potential of melanoma cells in a three-dimensional collagen matrix in vitro and in the lungs of a mouse model in vivo; it also significantly upregulated the expression of the NOTCH target gene CCNE. Previous studies suggested that Wnt signaling increases NUMB expression. By mimicking Wnt stimulation through glycogen synthase kinase-3 inhibition, we increased NUMB expression in melanoma cells. Furthermore, a glycogen synthase kinase-3 inhibitor reduced the invasion of melanoma cells in a NUMB-dependent manner. Together, our results suggest that NUMB suppresses invasion and metastasis in melanoma, potentially through its regulation of the NOTCH‒CCNE axis and that the inhibitors that upregulate NUMB can exert therapeutic effects in melanoma.


Subject(s)
Melanoma , Membrane Proteins , Nerve Tissue Proteins , Animals , Cell Line, Tumor , Glycogen Synthase Kinases/metabolism , Humans , Melanoma/drug therapy , Melanoma/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Wnt Signaling Pathway
9.
J Transl Med ; 9: 33, 2011 Mar 30.
Article in English | MEDLINE | ID: mdl-21450101

ABSTRACT

BACKGROUND: Infiltration of colorectal carcinomas (CRC) with T-cells has been associated with good prognosis. There are some indications that chemokines could be involved in T-cell infiltration of tumors. Selective modulation of chemokine activity at the tumor site could attract immune cells resulting in tumor growth inhibition. In mouse tumor model systems, gene therapy with chemokines or administration of antibody (Ab)-chemokine fusion proteins have provided potent immune mediated tumor rejection which was mediated by infiltrating T cells at the tumor site. To develop such immunotherapeutic strategies for cancer patients, one must identify chemokines and their receptors involved in T-cell migration toward tumor cells. METHODS: To identify chemokine and chemokine receptors involved in T-cell migration toward CRC cells, we have used our previously published three-dimensional organotypic CRC culture system. Organotypic culture was initiated with a layer of fetal fibroblast cells mixed with collagen matrix in a 24 well tissue culture plate. A layer of CRC cells was placed on top of the fibroblast-collagen layer which was followed by a separating layer of fibroblasts in collagen matrix. Anti-CRC specific cytotoxic T lymphocytes (CTLs) mixed with fibroblasts in collagen matrix were placed on top of the separating layer. Excess chemokine ligand (CCL) or Abs to chemokine or chemokine receptor (CCR) were used in migration inhibition assays to identify the chemokine and the receptor involved in CTL migration. RESULTS: Inclusion of excess CCL2 in T-cell layer or Ab to CCL2 in separating layer of collagen fibroblasts blocked the migration of CTLs toward tumor cells and in turn significantly inhibited tumor cell apoptosis. Also, Ab to CCR2 in the separating layer of collagen and fibroblasts blocked the migration of CTLs toward tumor cells and subsequently inhibited tumor cell apoptosis. Expression of CCR2 in four additional CRC patients' lymphocytes isolated from infiltrating tumor tissues suggests their role in migration in other CRC patients. CONCLUSIONS: Our data suggest that CCL2 secreted by tumor cells and CCR2 receptors on CTLs are involved in migration of CTLs towards tumor. Gene therapy of tumor cells with CCL2 or CCL2/anti-tumor Ab fusion proteins may attract CTLs that potentially could inhibit tumor growth.


Subject(s)
Cell Movement , Chemokine CCL2/metabolism , Colonic Neoplasms/pathology , Receptors, CCR2/metabolism , T-Lymphocytes, Cytotoxic/pathology , Animals , Apoptosis , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Humans , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Phenotype , Time Factors
10.
Semin Cancer Biol ; 19(2): 92-6, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19049876

ABSTRACT

Chemokines and chemokine receptors play an important role in immune homeostasis and surveillance. Altered or defective expression of chemokines and/or chemokine receptors could lead to a disease state including autoimmune disorder or cancer. Tumors from glioblastoma, melanoma, and neuroblastoma secrete high levels of chemokines that can promote tumor growth and progression or induce stromal cells present in the tumor microenvironment to produce cytokines or chemokines which, in turn, can regulate angiogenesis, tumor growth, and metastasis. On the other hand, chemokines secreted by tumor or stromal cells can also attract leukocytes such as dendritic cells, macrophages, neutrophils, and lymphocytes which may downmodulate tumor growth. New therapies that are aimed at limiting tumor growth and progression by attracting immune effector cells to the tumor site with chemokines may hold the key to the successful treatment of cancer, although this approach may be hampered by possible tumor growth-stimulating effects of chemokines.


Subject(s)
Chemokines/immunology , Neuroectodermal Tumors/immunology , Animals , Cell Movement/immunology , Humans , Leukocytes/immunology , Neuroectodermal Tumors/pathology , Receptors, Chemokine/immunology , Stromal Cells
11.
J Cancer Biol ; 2(2): 42-43, 2021.
Article in English | MEDLINE | ID: mdl-36283004

ABSTRACT

The complexity of the tumor microenvironment has been a challenge for understanding the mechanisms of therapy resistance. The development of improved animal models that closely mimic human disease is key for understanding and treating diseases. Recently, a new humanized mouse model has been developed that enables the study of human immune cells in tumor host-cell interactions. In this commentary we highlight the critical aspects of mast cells in immune therapy resistance. These mast cells release cytokines that downmodulate HLA class I on the malignant cells making them inaccessible the cytotoxic activity of T cells.

12.
J Invest Dermatol ; 141(6): 1564-1572.e4, 2021 06.
Article in English | MEDLINE | ID: mdl-33333124

ABSTRACT

The SWI/SNF chromatin remodeler family includes the BAF and PBAF complexes. ARID2, encoding a PBAF complex subunit, is frequently mutated in melanoma independently of BRAF/RAS mutations. Emerging evidence shows that SWI/SNF complexes regulate tumor immunity; for instance, the loss of PBRM1, another PBAF complex subunit, enhances susceptibility to immune checkpoint inhibitors in melanoma. Notably, ARID2 mutations are more frequent in melanoma than PBRM1 mutations. However, the role of ARID2 as a modulator of tumor immunity remains unclear. In this study, we show that ARID2 knockout sensitizes melanoma to immune checkpoint inhibitors. Anti‒PD-L1 treatment restricts tumor growth in mice bearing ARID2-knockout melanoma cells, correlating with an increase in the infiltration of cytotoxic CD8+ T cells. Furthermore, ARID2 deficiency leads to signal transducer and activator of transcription 1 upregulation, which subsequently causes increased expression of T-cell‒attracting chemokines such as CXCL9, CXCL10, and CCL5. These results demonstrate that ARID2 is an immunomodulator and a potential biomarker that indicates immune checkpoint inhibitor effectiveness in patients with melanoma.


Subject(s)
Drug Resistance, Neoplasm/genetics , Immune Checkpoint Inhibitors/pharmacology , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Transcription Factors/deficiency , Animals , Disease Models, Animal , Gene Knockout Techniques , Humans , Immune Checkpoint Inhibitors/therapeutic use , Melanoma/genetics , Melanoma/immunology , Melanoma/pathology , Mice , Mutation , Signal Transduction/genetics , Signal Transduction/immunology , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Transcription Factors/genetics
13.
J Immunother Cancer ; 9(12)2021 12.
Article in English | MEDLINE | ID: mdl-34937742

ABSTRACT

BACKGROUND: Gamma delta (γδ) T cells are attractive effector cells for cancer immunotherapy. Vδ2 T cells expanded by zoledronic acid (ZOL) are the most commonly used γδ T cells for adoptive cell therapy. However, adoptive transfer of the expanded Vδ2 T cells has limited clinical efficacy. METHODS: We developed a costimulation method for expansion of Vδ2 T cells in PBMCs by activating γδ T-cell receptor (γδTCR) and Toll-like receptor (TLR) 7/8 using isopentenyl pyrophosphate (IPP) and resiquimod, respectively, and tested the functional markers and antitumoral effects in vitro two-dimensional two-dimensional and three-dimensional spheroid models and in vivo models. Single-cell sequencing dataset analysis and reverse-phase protein array were employed for mechanistic studies. RESULTS: We find that Vδ2 T cells expanded by IPP plus resiquimod showed significantly increased cytotoxicity to tumor cells with lower programmed cell death protein 1 (PD-1) expression than Vδ2 T cells expanded by IPP or ZOL. Mechanistically, the costimulation enhanced the activation of the phosphatidylinositol 3-kinase (PI3K)-protein kinase B (PKB/Akt)-the mammalian target of rapamycin (mTOR) pathway and the TLR7/8-MyD88 pathway. Resiquimod stimulated Vδ2 T-cell expansion in both antigen presenting cell dependent and independent manners. In addition, resiquimod decreased the number of adherent inhibitory antigen-presenting cells (APCs) and suppressed the inhibitory function of APCs by decreasing PD-L1 and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) expression in these cells during in vitro Vδ2 T-cell expansion. Finally, we showed that human Vδ2 T cells can be expanded from PBMCs and spleen of humanized NSG mice using IPP plus resiquimod or ZOL, demonstrating that humanized mice are a promising preclinical model for studying human γδ T-cell development and function. CONCLUSIONS: Vδ2 T cells expanded by IPP and resiquimod demonstrate improved anti-tumor function and have the potential to increase the efficacy of γδ T cell-based therapies.


Subject(s)
Immunotherapy/methods , Melanoma/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , TOR Serine-Threonine Kinases/metabolism , Toll-Like Receptor 7/metabolism , Animals , Humans , Melanoma/pathology , Mice , Mice, Nude
14.
Cancer Res ; 81(20): 5230-5241, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34462276

ABSTRACT

Metastatic melanoma is challenging to clinically address. Although standard-of-care targeted therapy has high response rates in patients with BRAF-mutant melanoma, therapy relapse occurs in most cases. Intrinsically resistant melanoma cells drive therapy resistance and display molecular and biologic properties akin to neural crest-like stem cells (NCLSC) including high invasiveness, plasticity, and self-renewal capacity. The shared transcriptional programs and vulnerabilities between NCLSCs and cancer cells remains poorly understood. Here, we identify a developmental LPAR1-axis critical for NCLSC viability and melanoma cell survival. LPAR1 activity increased during progression and following acquisition of therapeutic resistance. Notably, genetic inhibition of LPAR1 potentiated BRAFi ± MEKi efficacy and ablated melanoma migration and invasion. Our data define LPAR1 as a new therapeutic target in melanoma and highlights the promise of dissecting stem cell-like pathways hijacked by tumor cells. SIGNIFICANCE: This study identifies an LPAR1-axis critical for melanoma invasion and intrinsic/acquired therapy resistance.


Subject(s)
Biomarkers, Tumor/metabolism , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Melanoma/pathology , Neural Crest/pathology , Neural Stem Cells/pathology , Receptors, Lysophosphatidic Acid/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neural Crest/drug effects , Neural Crest/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Prognosis , Receptors, Lysophosphatidic Acid/genetics , Transcriptome , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Nat Commun ; 12(1): 346, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436641

ABSTRACT

Anti-PD-1 therapy is used as a front-line treatment for many cancers, but mechanistic insight into this therapy resistance is still lacking. Here we generate a humanized (Hu)-mouse melanoma model by injecting fetal liver-derived CD34+ cells and implanting autologous thymus in immune-deficient NOD-scid IL2Rγnull (NSG) mice. Reconstituted Hu-mice are challenged with HLA-matched melanomas and treated with anti-PD-1, which results in restricted tumor growth but not complete regression. Tumor RNA-seq, multiplexed imaging and immunohistology staining show high expression of chemokines, as well as recruitment of FOXP3+ Treg and mast cells, in selective tumor regions. Reduced HLA-class I expression and CD8+/Granz B+ T cells homeostasis are observed in tumor regions where FOXP3+ Treg and mast cells co-localize, with such features associated with resistance to anti-PD-1 treatment. Combining anti-PD-1 with sunitinib or imatinib results in the depletion of mast cells and complete regression of tumors. Our results thus implicate mast cell depletion for improving the efficacy of anti-PD-1 therapy.


Subject(s)
Drug Resistance, Neoplasm , Lymphocytes, Tumor-Infiltrating/immunology , Mast Cells/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Drug Resistance, Neoplasm/drug effects , Humans , Immune Checkpoint Inhibitors/pharmacology , Lymphocytes, Tumor-Infiltrating/drug effects , Mast Cells/drug effects , Melanoma/immunology , Melanoma/pathology , Melanoma/therapy , Mice, Transgenic , Programmed Cell Death 1 Receptor/metabolism , Sunitinib/pharmacology , Sunitinib/therapeutic use , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
16.
Int J Cancer ; 127(5): 1124-30, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20027629

ABSTRACT

Immunotherapy of colorectal carcinoma (CRC) has great promise as the presence of T lymphocytes in CRC tissues in situ is correlated with reduced recurrence and increased survival. Thus, identification of the antigens recognized by T cells of CRC patients may permit development of vaccines with potential benefit for these patients. Using expression cloning, we identified the antigen, nucleophosmin (Npm), recognized by an HLA-A1 restricted cytotoxic T lymphocyte (CTL) line derived from the peripheral blood mononuclear cells (PBMC) of a rectal cancer patient. A decamer peptide derived from the Npm sequence sensitized peptide-pulsed HLA-A1 positive cells to lysis by the CTL line. The peptide also induced proliferative and cytotoxic T lymphocytes in the PBMC of 4 of 6 CRC patients, which lysed HLA-A1 positive peptide-pulsed target cells and CRC cells endogenously expressing Npm. Overexpression of Npm by tumors of various histological types, recognition of the antigen by T cells derived from different CRC patients and association of the antigen with poor prognostic outcome make it a promising target for immunotherapeutic intervention in cancer patients.


Subject(s)
HLA-A1 Antigen/immunology , Nuclear Proteins/immunology , Peptide Fragments/pharmacology , Rectal Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigen Presentation , Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cells, Cultured , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Female , Humans , Immunotherapy , Lymphocyte Activation , Melanoma/immunology , Melanoma/pathology , Nucleophosmin , Peptide Fragments/immunology , Rectal Neoplasms/pathology
17.
Nat Commun ; 11(1): 2858, 2020 06 05.
Article in English | MEDLINE | ID: mdl-32504051

ABSTRACT

Metastatic melanoma is challenging to manage. Although targeted- and immune therapies have extended survival, most patients experience therapy resistance. The adaptability of melanoma cells in nutrient- and therapeutically-challenged environments distinguishes melanoma as an ideal model for investigating therapy resistance. In this review, we discuss the current available repertoire of melanoma models including two- and three-dimensional tissue cultures, organoids, genetically engineered mice and patient-derived xenograft. In particular, we highlight how each system recapitulates different features of melanoma adaptability and can be used to better understand melanoma development, progression and therapy resistance.


Subject(s)
Antinematodal Agents/pharmacology , Disease Models, Animal , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Xenograft Model Antitumor Assays/methods , Animals , Antinematodal Agents/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Melanoma/genetics , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Transgenic , Organoids , Skin/cytology , Skin/immunology , Skin/pathology , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Spheroids, Cellular
18.
Cancer Res ; 67(8): 3555-9, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17440064

ABSTRACT

Antigens recognized by T helper (Th) cells in the context of MHC class II molecules have vaccine potential against cancer and infectious agents. We have described previously a melanoma patient's HLA-DR7-restricted Th cell clone recognizing an antigen, which is shared among melanoma and glioma cells derived from various patients. Here, this antigen was cloned using a novel antigen phage display approach. The antigen was identified as the ribosomal protein L8 (RPL8). A peptide of RPL8 significantly stimulated proliferation and/or cytokine expression of the Th cell clone and lymphocytes in four of nine HLA-DR7(+) melanoma patients but not in healthy volunteers. The RPL8 antigen may represent a relevant vaccine target for patients with melanoma, glioma, and breast carcinoma whose tumors express this protein.


Subject(s)
Antigens, Neoplasm/immunology , HLA-DR7 Antigen/immunology , Melanoma/immunology , Ribosomal Proteins/immunology , Animals , Antigens, Neoplasm/genetics , COS Cells , Chlorocebus aethiops , Cloning, Molecular , Epitopes/genetics , Epitopes/immunology , Humans , Lymphocyte Activation , Melanoma/genetics , Peptide Library , Ribosomal Proteins/genetics , T-Lymphocytes, Helper-Inducer/immunology
19.
Nat Commun ; 10(1): 4186, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31519915

ABSTRACT

Tumor associated inflammation predicts response to immune checkpoint blockade in human melanoma. Current theories on regulation of inflammation center on anti-tumor T cell responses. Here we show that tumor associated B cells are vital to melanoma associated inflammation. Human B cells express pro- and anti-inflammatory factors and differentiate into plasmablast-like cells when exposed to autologous melanoma secretomes in vitro. This plasmablast-like phenotype can be reconciled in human melanomas where plasmablast-like cells also express T cell-recruiting chemokines CCL3, CCL4, CCL5. Depletion of B cells in melanoma patients by anti-CD20 immunotherapy decreases tumor associated inflammation and CD8+ T cell numbers. Plasmablast-like cells also increase PD-1+ T cell activation through anti-PD-1 blockade in vitro and their frequency in pretherapy melanomas predicts response and survival to immune checkpoint blockade. Tumor associated B cells therefore orchestrate and sustain melanoma inflammation and may represent a predictor for survival and response to immune checkpoint blockade therapy.


Subject(s)
Inflammation/metabolism , Melanoma/metabolism , Antibodies, Monoclonal/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Chemokine CCL3/metabolism , Chemokine CCL4/metabolism , Chemokine CCL5/metabolism , Humans , Immunotherapy , In Vitro Techniques , Inflammation/immunology , Melanoma/immunology , Melanoma/therapy , Programmed Cell Death 1 Receptor/metabolism
20.
Cancer Discov ; 9(1): 64-81, 2019 01.
Article in English | MEDLINE | ID: mdl-30279173

ABSTRACT

Physical changes in skin are among the most visible signs of aging. We found that young dermal fibroblasts secrete high levels of extracellular matrix (ECM) constituents, including proteoglycans, glycoproteins, and cartilage-linking proteins. The most abundantly secreted was HAPLN1, a hyaluronic and proteoglycan link protein. HAPLN1 was lost in aged fibroblasts, resulting in a more aligned ECM that promoted metastasis of melanoma cells. Reconstituting HAPLN1 inhibited metastasis in an aged microenvironment, in 3-D skin reconstruction models, and in vivo. Intriguingly, aged fibroblast-derived matrices had the opposite effect on the migration of T cells, inhibiting their motility. HAPLN1 treatment of aged fibroblasts restored motility of mononuclear immune cells, while impeding that of polymorphonuclear immune cells, which in turn affected regulatory T-cell recruitment. These data suggest that although age-related physical changes in the ECM can promote tumor cell motility, they may adversely affect the motility of some immune cells, resulting in an overall change in the immune microenvironment. Understanding the physical changes in aging skin may provide avenues for more effective therapy for older patients with melanoma. SIGNIFICANCE: These data shed light on the mechanochemical interactions that occur between aged skin, tumor, and immune cell populations, which may affect tumor metastasis and immune cell infiltration, with implications for the efficacy of current therapies for melanoma.See related commentary by Marie and Merlino, p. 19.This article is highlighted in the In This Issue feature, p. 1.


Subject(s)
Aging , Collagen/metabolism , Melanoma/metabolism , Skin/metabolism , Animals , Cells, Cultured , Extracellular Matrix Proteins/metabolism , Fibroblasts/metabolism , Humans , Immune System , Melanoma/physiopathology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Proteoglycans/metabolism , Skin/physiopathology , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL