Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
J Vasc Interv Radiol ; 35(6): 809-817.e1, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38219903

ABSTRACT

Traditionally, rodent cancer models have driven preclinical oncology research. However, they do not fully recapitulate characteristics of human cancers, and their size poses challenges when evaluating tools in the interventional oncologists' armamentarium. Pig models, however, have been the gold standard for validating surgical procedures. Their size enables the study of image-guided interventions using human ultrasound (US), computed tomography (CT), and magnetic resonance (MR) imaging platforms. Furthermore, pigs have immunologic features that are similar to those of humans, which can potentially be leveraged for studying immunotherapy. Novel pig models of cancer are being developed, but additional research is required to better understand both the pig immune system and malignancy to enhance the potential for pig models in interventional oncology research. This review aims to address the main advantages and disadvantages of using a pig model for interventional oncology and outline the specific characteristics of pig models that make them more suitable for investigation of locoregional therapies.


Subject(s)
Disease Models, Animal , Immunotherapy , Neoplasms , Animals , Immunotherapy/methods , Neoplasms/therapy , Neoplasms/diagnostic imaging , Neoplasms/immunology , Humans , Swine , Radiography, Interventional , Sus scrofa , Medical Oncology
2.
Cell ; 183(3): 818-834.e13, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33038342

ABSTRACT

Many approaches to identify therapeutically relevant neoantigens couple tumor sequencing with bioinformatic algorithms and inferred rules of tumor epitope immunogenicity. However, there are no reference data to compare these approaches, and the parameters governing tumor epitope immunogenicity remain unclear. Here, we assembled a global consortium wherein each participant predicted immunogenic epitopes from shared tumor sequencing data. 608 epitopes were subsequently assessed for T cell binding in patient-matched samples. By integrating peptide features associated with presentation and recognition, we developed a model of tumor epitope immunogenicity that filtered out 98% of non-immunogenic peptides with a precision above 0.70. Pipelines prioritizing model features had superior performance, and pipeline alterations leveraging them improved prediction performance. These findings were validated in an independent cohort of 310 epitopes prioritized from tumor sequencing data and assessed for T cell binding. This data resource enables identification of parameters underlying effective anti-tumor immunity and is available to the research community.


Subject(s)
Antigens, Neoplasm/immunology , Epitopes/immunology , Neoplasms/immunology , Alleles , Antigen Presentation/immunology , Cohort Studies , Humans , Peptides/immunology , Programmed Cell Death 1 Receptor , Reproducibility of Results
4.
J Steroid Biochem Mol Biol ; 193: 105415, 2019 10.
Article in English | MEDLINE | ID: mdl-31226312

ABSTRACT

Breast cancers (BCs) with expression of estrogen receptor-alpha (ERα) occur in more than 70% of newly-diagnosed patients in the U.S. Endocrine therapy with antiestrogens or aromatase inhibitors is an important intervention for BCs that express ERα, and it remains one of the most effective targeted treatment strategies. However, a substantial proportion of patients with localized disease, and essentially all patients with metastatic BC, become resistant to current endocrine therapies. ERα is present in most resistant BCs, and in many of these its activity continues to regulate BC growth. Fulvestrant represents one class of ERα antagonists termed selective ER downregulators (SERDs). Treatment with fulvestrant causes ERα down-regulation, an event that helps overcome several resistance mechanisms. Unfortunately, full antitumor efficacy of fulvestrant is limited by its poor bioavailability in clinic. We have designed and tested a new generation of steroid-like SERDs. Using ERα-positive BC cells in vitro, we find that these compounds suppress ERα protein levels with efficacy similar to fulvestrant. Moreover, these new SERDs markedly inhibit ERα-positive BC cell transcription and proliferation in vitro even in the presence of estradiol-17ß. In vivo, the SERD termed JD128 significantly inhibited tumor growth in MCF-7 xenograft models in a dose-dependent manner (P < 0.001). Further, our findings indicate that these SERDs also interact with ER-positive immune cells in the tumor microenvironment such as myeloid-derived suppressor cells (MDSC), tumor infiltrating lymphocytes and other selected immune cell subpopulations. SERD-induced inhibition of MDSCs and concurrent actions on CD8+ and CD4 + T-cells promotes interaction of immune checkpoint inhibitors with BC cells in preclinical models, thereby leading to enhanced tumor killing even among highly aggressive BCs such as triple-negative BC that lack ERα expression. Since monotherapy with immune checkpoint inhibitors has not been effective for most BCs, combination therapies with SERDs that enhance immune recognition may increase immunotherapy responses in BC and improve patient survival. Hence, ERα antagonists that also promote ER downregulation may potentially benefit patients who are unresponsive to current endocrine therapies.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Estrogen Antagonists/therapeutic use , Animals , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/immunology , Estrogen Antagonists/pharmacology , Female , Fulvestrant/pharmacology , Fulvestrant/therapeutic use , Humans , Immunotherapy , Lymphocytes, Tumor-Infiltrating/immunology , Mice, Inbred BALB C , Mice, Nude , Receptors, Estrogen/metabolism
5.
Clin Cancer Res ; 25(3): 1000-1011, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30409823

ABSTRACT

PURPOSE: To improve persistence of adoptively transferred T-cell receptor (TCR)-engineered T cells and durable clinical responses, we designed a clinical trial to transplant genetically-modified hematopoietic stem cells (HSCs) together with adoptive cell transfer of T cells both engineered to express an NY-ESO-1 TCR. Here, we report the preclinical studies performed to enable an investigational new drug (IND) application. EXPERIMENTAL DESIGN: HSCs transduced with a lentiviral vector expressing NY-ESO-1 TCR and the PET reporter/suicide gene HSV1-sr39TK and T cells transduced with a retroviral vector expressing NY-ESO-1 TCR were coadministered to myelodepleted HLA-A2/Kb mice within a formal Good Laboratory Practice (GLP)-compliant study to demonstrate safety, persistence, and HSC differentiation into all blood lineages. Non-GLP experiments included assessment of transgene immunogenicity and in vitro viral insertion safety studies. Furthermore, Good Manufacturing Practice (GMP)-compliant cell production qualification runs were performed to establish the manufacturing protocols for clinical use. RESULTS: TCR genetically modified and ex vivo-cultured HSCs differentiated into all blood subsets in vivo after HSC transplantation, and coadministration of TCR-transduced T cells did not result in increased toxicity. The expression of NY-ESO-1 TCR and sr39TK transgenes did not have a detrimental effect on gene-modified HSC's differentiation to all blood cell lineages. There was no evidence of genotoxicity induced by the lentiviral vector. GMP batches of clinical-grade transgenic cells produced during qualification runs had adequate stability and functionality. CONCLUSIONS: Coadministration of HSCs and T cells expressing an NY-ESO-1 TCR is safe in preclinical models. The results presented in this article led to the FDA approval of IND 17471.


Subject(s)
Genetic Therapy/methods , Hematopoietic Stem Cells/immunology , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/genetics , Cells, Cultured , Clinical Trials as Topic , Drugs, Investigational/therapeutic use , HLA-A2 Antigen/genetics , Hematopoietic Stem Cells/metabolism , Humans , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/genetics , Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism
6.
PLoS One ; 13(1): e0191634, 2018.
Article in English | MEDLINE | ID: mdl-29360859

ABSTRACT

For adoptive cell transfer (ACT) immunotherapy of tumor-reactive T cells, an effective therapeutic outcome depends upon cell dose, cell expansion in vivo through a minimally differentiated phenotype, long term persistence, and strong cytolytic effector function. An incomplete understanding of the biological coupling between T cell expansion, differentiation, and response to stimulation hinders the co-optimization of these factors. We report on a biophysical investigation of how the short-term kinetics of T cell functional activation, through molecular stimulation and cell-cell interactions, competes with phenotype differentiation. T cells receive molecular stimulation for a few minutes to a few hours in bulk culture. Following this priming period, the cells are then analyzed at the transcriptional level, or isolated as single cells, with continuing molecular stimulation, within microchambers for analysis via 11-plex secreted protein assays. We resolve a rapid feedback mechanism, promoted by T cell-T cell contact interactions, which strongly amplifies T cell functional performance while yielding only minimal phenotype differentiation. When tested in mouse models of ACT, optimally primed T cells lead to complete tumor eradication. A similar kinetic process is identified in CD8+ and CD4+ T cells collected from a patient with metastatic melanoma.


Subject(s)
Adoptive Transfer , Immunophenotyping , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Female , Flow Cytometry , Heterografts , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic
7.
Proc Natl Acad Sci U S A ; 114(52): 13679-13684, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29229836

ABSTRACT

Continuous BRAF inhibition of BRAF mutant melanomas triggers a series of cell state changes that lead to therapy resistance and escape from immune control before establishing acquired resistance genetically. We used genome-wide transcriptomics and single-cell phenotyping to explore the response kinetics to BRAF inhibition for a panel of patient-derived BRAFV600 -mutant melanoma cell lines. A subset of plastic cell lines, which followed a trajectory covering multiple known cell state transitions, provided models for more detailed biophysical investigations. Markov modeling revealed that the cell state transitions were reversible and mediated by both Lamarckian induction and nongenetic Darwinian selection of drug-tolerant states. Single-cell functional proteomics revealed activation of certain signaling networks shortly after BRAF inhibition, and before the appearance of drug-resistant phenotypes. Drug targeting those networks, in combination with BRAF inhibition, halted the adaptive transition and led to prolonged growth inhibition in multiple patient-derived cell lines.


Subject(s)
Drug Resistance, Neoplasm , Melanoma/genetics , Melanoma/metabolism , Signal Transduction , Single-Cell Analysis , Adaptation, Physiological , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Gene Expression Profiling , Humans , MAP Kinase Signaling System/drug effects , Markov Chains , Melanoma/drug therapy , Melanoma/pathology , NF-kappa B/metabolism , Phenotype , Proteome , Proteomics/methods , Proto-Oncogene Proteins B-raf/genetics
8.
Cell Rep ; 19(6): 1189-1201, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28494868

ABSTRACT

PD-L1 and PD-L2 are ligands for the PD-1 immune inhibiting checkpoint that can be induced in tumors by interferon exposure, leading to immune evasion. This process is important for immunotherapy based on PD-1 blockade. We examined the specific molecules involved in interferon-induced signaling that regulates PD-L1 and PD-L2 expression in melanoma cells. These studies revealed that the interferon-gamma-JAK1/JAK2-STAT1/STAT2/STAT3-IRF1 axis primarily regulates PD-L1 expression, with IRF1 binding to its promoter. PD-L2 responded equally to interferon beta and gamma and is regulated through both IRF1 and STAT3, which bind to the PD-L2 promoter. Analysis of biopsy specimens from patients with melanoma confirmed interferon signature enrichment and upregulation of gene targets for STAT1/STAT2/STAT3 and IRF1 in anti-PD-1-responding tumors. Therefore, these studies map the signaling pathway of interferon-gamma-inducible PD-1 ligand expression.


Subject(s)
B7-H1 Antigen/genetics , Interferon Regulatory Factor-1/metabolism , Melanoma/genetics , Programmed Cell Death 1 Ligand 2 Protein/genetics , Signal Transduction , Transcriptional Activation , B7-H1 Antigen/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Interferon Regulatory Factor-1/genetics , Interferon-beta/metabolism , Interferon-gamma/metabolism , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Melanoma/metabolism , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Promoter Regions, Genetic , Protein Binding , STAT Transcription Factors/metabolism , Up-Regulation
9.
Oncoimmunology ; 5(7): e1052212, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27622011

ABSTRACT

The combination of targeted therapy with BRAF and MEK inhibitors has become the standard of care in patients with BRAF (V600E) mutant melanoma, but responses are not durable. In addition, the impressive clinical benefits with anti-PD-1 and anti-PD-L1 antibodies (Ab) in patients with heavily pretreated metastatic melanoma and the synergistic effect of dabrafenib, trametinib and anti-PD-1 compared with single therapy alone groups support the idea that combining dabrafenib, trametinib and immunotherapy based on PD-1 blockade could be an interesting approach in the treatment of metastatic melanoma. With our mouse model of syngeneic BRAF (V600E) driven melanoma (SM1), we tested whether the addition of an immunostimulatory Ab targeting CD137 (4-1BB) and/or CD134 (OX40) would enhance the antitumor effect of dabrafenib, trametinib and anti-PD-1 or anti-PD-L1 therapy. In vitro studies showed that the combination group of dabrafenib, trametinib and anti-PD-1 increases CD8(+) tumor infiltrating lymphocytes (TILs), as well as CD4(+) T cells and tumor-associated macrophages (TAMs). An upregulation of PD-L1 was observed in the combination of dabrafenib, trametinib and anti-PD-1 therapy. Combination of dabrafenib, trametinib and anti-PD-1, with either anti-CD137 or anti-CD134, showed a superior antitumor effect, but the five-agent combination was not superior to the four-agent combinations. In conclusion, the combination of dabrafenib, trametinib, anti-PD1 or anti-PD-L1 therapy results in robust antitumor activity, which is further improved by adding the immune-stimulating Ab anti-CD137 or anti-CD134. Our findings support the testing of these combinations in patients with BRAF (V600E) mutant metastatic melanoma.

11.
Mol Cancer ; 14: 27, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25645078

ABSTRACT

BACKGROUND: Approximately 20% of melanomas contain a mutation in NRAS. However no direct inhibitor of NRAS is available. One of the main signaling pathways downstream of NRAS is the MAPK pathway. In this study we investigated the possibility of blocking oncogenic signaling of NRAS by inhibiting two signaling points in the MAPK pathway. METHODS: Fourteen NRAS mutated human melanoma cell lines were treated with a pan-RAF inhibitor (PRi, Amgen Compd A), a MEK inhibitor (MEKi, trametinib) or their combination and the effects on proliferation, cell cycle progression, apoptosis, transcription profile and signaling of the cells were investigated. RESULTS: The majority of the cell lines showed a significant growth inhibition, with high levels of synergism of the PRi and MEKi combination. Sensitive cell lines showed induction of apoptosis by the combination treatment and there was a correlation between p-MEK levels and synergistic effect of the combination treatment. Proliferation of sensitive cell lines was blocked by the inhibition of the MAPK pathway, which also blocked expression of cyclin D1. However, in resistant cell lines, proliferation was blocked by combined inhibition of the MAPK pathway and cyclin D3, which is not regulated by the MAPK pathway. Resistant cell lines also showed higher levels of p-GSK3ß and less perturbation of the apoptotic profile upon the treatment in comparison with the sensitive cell lines. CONCLUSIONS: The combination of PRi + MEKi can be an effective regimen for blocking proliferation of NRAS mutant melanomas when there is higher activity of the MAPK pathway and dependence of proliferation and survival on this pathway.


Subject(s)
GTP Phosphohydrolases/genetics , MAP Kinase Signaling System/genetics , Melanoma/genetics , Membrane Proteins/genetics , Mutation/genetics , raf Kinases/genetics , Apoptosis/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin D1/genetics , Cyclin D3/genetics , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Signal Transduction/genetics , Transcription, Genetic/genetics
12.
Oncoimmunology ; 3: e29244, 2014.
Article in English | MEDLINE | ID: mdl-25083336

ABSTRACT

Targeting immune inhibitory receptors has brought excitement, innovation and hope to cancer patients. Our recent work revealed the immunological effects of blocking the CTLA4 and PD-1 immune checkpoints on T cell receptor usage among peripheral blood cells, and further uncovers how the expansion of the T cell repertoire matches the immunotoxicity profile of the therapy.

13.
Mol Cancer ; 13: 194, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25142146

ABSTRACT

BACKGROUND: In melanoma, dysregulation of the MAPK pathway, usually via BRAF(V600) or NRAS(Q61) somatic mutations, leads to constitutive ERK signaling. While BRAF inhibitors are initially effective for BRAF-mutant melanoma, no FDA-approved targeted therapies exist for BRAF-inhibitor-resistant BRAF(V600), NRAS mutant, or wild-type melanoma. METHODS: The 50% inhibitory concentration (IC50) of SCH772984, a novel inhibitor of ERK1/2, was determined in a panel of 50 melanoma cell lines. Effects on MAPK and AKT signaling by western blotting and cell cycle by flow cytometry were determined. RESULTS: Sensitivity fell into three groups: sensitive, 50% inhibitory concentration (IC50) < 1 µM; intermediately sensitive, IC50 1-2 µM; and resistant, >2 µM. Fifteen of 21 (71%) BRAF mutants, including 4 with innate vemurafenib resistance, were sensitive to SCH772984. All three (100%) BRAF/NRAS double mutants, 11 of 14 (78%) NRAS mutants and 5 of 7 (71%) wild-type melanomas were sensitive. Among BRAF(V600) mutants with in vitro acquired resistance to vemurafenib, those with MAPK pathway reactivation as the mechanism of resistance were sensitive to SCH772984. SCH772984 caused G1 arrest and induced apoptosis. CONCLUSIONS: Combining vemurafenib and SCH722984 in BRAF mutant melanoma was synergistic in a majority of cell lines and significantly delayed the onset of acquired resistance in long term in vitro assays. Therefore, SCH772984 may be clinically applicable as a treatment for non-BRAF mutant melanoma or in BRAF-mutant melanoma with innate or acquired resistance, alone or in combination with BRAF inhibitors.


Subject(s)
GTP Phosphohydrolases/antagonists & inhibitors , Indazoles/pharmacology , Membrane Proteins/antagonists & inhibitors , Multiple Myeloma/pathology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Synergism , GTP Phosphohydrolases/genetics , Humans , Indoles/pharmacology , Inhibitory Concentration 50 , Membrane Proteins/genetics , Molecular Targeted Therapy , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/pharmacology , Vemurafenib
14.
Clin Cancer Res ; 20(13): 3446-57, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24812408

ABSTRACT

PURPOSE: PD-L1 is the main ligand for the immune inhibitory receptor PD-1. This ligand is frequently expressed by melanoma cells. In this study, we investigated whether PD-L1 expression is controlled by melanoma driver mutations and modified by oncogenic signaling inhibition. EXPERIMENTAL DESIGN: Expression of PD-L1 was investigated in a panel of 51 melanoma cell lines containing different oncogenic mutations, including cell lines with innate and acquired resistance to BRAF inhibitors (BRAFi). The effects of targeted therapy drugs on expression of PD-L1 by melanoma cells were investigated. RESULTS: No association was found between the level of PD-L1 expression and mutations in BRAF, NRAS, PTEN, or amplification of AKT. Resistance to vemurafenib due to the activation of alternative signaling pathways was accompanied with the induction of PD-L1 expression, whereas the resistance due to the reactivation of the MAPK pathway had no effect on PD-L1 expression. In melanoma cell lines, the effects of BRAF, MEK, and PI3K inhibitors on expression of PD-L1 were variable from reduction to induction, particularly in the presence of INFγ. In PD-L1-exposed lymphocytes, vemurafenib paradoxically restored activity of the MAPK pathway and increased the secretion of cytokines. CONCLUSIONS: In melanoma cell lines, including BRAFi-resistant cells, PD-L1 expression is variably regulated by oncogenic signaling pathways. PD-L1-exposed lymphocytes decrease MAPK signaling, which is corrected by exposure to vemurafenib, providing potential benefits of combining this drug with immunotherapies.


Subject(s)
B7-H1 Antigen/genetics , Melanoma/genetics , Melanoma/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Signal Transduction , B7-H1 Antigen/metabolism , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Cytokines/biosynthesis , Drug Resistance, Neoplasm/genetics , Humans , Indoles/pharmacology , Lymphocytes/immunology , Lymphocytes/metabolism , Melanoma/immunology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mutation , Signal Transduction/drug effects , Sulfonamides/pharmacology , Vemurafenib
15.
Mol Cancer ; 13: 83, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24735930

ABSTRACT

BACKGROUND: The clinical use of BRAF inhibitors for treatment of metastatic melanoma is limited by the development of drug resistance. In this study we investigated whether co-targeting the MAPK and the PI3K-AKT pathway can prevent emergence of resistance or provide additional growth inhibitory effects in vitro. METHODS: Anti-tumor effects of the combination of the BRAF inhibitor (BRAFi) dabrafenib and GSK2141795B (AKTi) in a panel of 23 BRAF mutated melanoma cell lines were evaluated on growth inhibition by an ATP-based luminescent assay, on cell cycle and apoptosis by flow cytometry and on cell signaling by western blot. Moreover, we investigated the possibilities of delaying or reversing resistance or achieving further growth inhibition by combining AKTi with dabrafenib and/or the MEK inhibitor (MEKi) trametinib by using long term cultures. RESULTS: More than 40% of the cell lines, including PTEN-/- and AKT mutants showed sensitivity to AKTi (IC50 < 1.5 µM). The combination of dabrafenib and AKTi synergistically potentiated growth inhibition in the majority of cell lines with IC50 > 5 nM dabrafenib. Combinatorial treatment induced apoptosis only in cell lines sensitive to AKTi. In long term cultures of a PTEN-/- cell line, combinatorial treatment with the MAPK inhibitors, dabrafenib and trametinib, and AKTi markedly delayed the emergence of drug resistance. Moreover, combining AKTi with the MAPK inhibitors from the beginning provided superior growth inhibitory effects compared to addition of AKTi upon development of resistance to MAPK inhibitors in this particular cell line. CONCLUSIONS: AKTi combined with BRAFi-based therapy may benefit patients with tumors harboring BRAF mutations and particularly PTEN deletions or AKT mutations.


Subject(s)
Drug Resistance, Neoplasm/genetics , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-akt/genetics , Apoptosis/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Humans , Imidazoles/administration & dosage , MAP Kinase Kinase Kinases/antagonists & inhibitors , Melanoma/genetics , Melanoma/pathology , Mutation , Oximes/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Signal Transduction , Xenograft Model Antitumor Assays
16.
Mol Oncol ; 8(2): 250-60, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24345644

ABSTRACT

Keratoacanthomas (KAs) and cutaneous squamous cell carcinomas (cuSCCs) develop in 15-30% of patients with BRAF(V600E) metastatic melanoma treated with BRAF inhibitors (BRAFi). These lesions resemble mouse skin tumors induced by the two-stage DMBA/TPA skin carcinogenesis protocol; in this protocol BRAFi accelerates tumor induction. Since prior studies demonstrated cyclooxygenase 2 (COX-2) is necessary for DMBA/TPA tumor induction, we hypothesized that COX-2 inhibition might prevent BRAFi-accelerated skin tumors. Celecoxib, a COX-2 inhibitor, significantly delayed tumor acceleration by the BRAFi inhibitor PLX7420 and decreased tumor number by 90%. Tumor gene expression profiling demonstrated that celecoxib partially reversed the PLX4720-induced gene signature. In PDV cuSCC cells, vemurafenib (a clinically approved BRAFi) increased ERK phosphorylation and soft agar colony formation; both responses were greatly decreased by celecoxib. In clinical trials trametinib, a MEK inhibitor (MEKi) increases BRAFi therapy efficacy in BRAF(V600E) melanomas and reduces BRAFi-induced KA and cuSCC frequency. Trametinib also reduced vemurafenib-induced PDV soft agar colonies, but less efficiently than celecoxib. The trametinb/celecoxib combination was more effective than either inhibitor alone. In conclusion, celecoxib suppressed both BRAFi-accelerated skin tumors and soft-agar colonies, warranting its testing as a chemopreventive agent for non-melanoma skin lesions in patients treated with BRAFi alone or in combination with MEKi.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Indoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Pyrazoles/pharmacology , Pyridones/pharmacology , Pyrimidinones/pharmacology , Skin Neoplasms/drug therapy , Sulfonamides/pharmacology , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Celecoxib , Cyclooxygenase 2/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Keratoacanthoma/diet therapy , Keratoacanthoma/genetics , Keratoacanthoma/metabolism , Keratoacanthoma/pathology , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mice , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Vemurafenib
17.
Cancer Discov ; 3(4): 418-29, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23519018

ABSTRACT

UNLABELLED: Adoptive cell transfer (ACT) of genetically engineered T cells expressing cancer-specific T-cell receptors (TCR) is a promising cancer treatment. Here, we investigate the in vivo functional activity and dynamics of the transferred cells by analyzing samples from 3 representative patients with melanoma enrolled in a clinical trial of ACT with TCR transgenic T cells targeted against the melanosomal antigen MART-1. The analyses included evaluating 19 secreted proteins from individual cells from phenotypically defined T-cell subpopulations, as well as the enumeration of T cells with TCR antigen specificity for 36 melanoma antigens. These analyses revealed the coordinated functional dynamics of the adoptively transferred, as well as endogenous, T cells, and the importance of highly functional T cells in dominating the antitumor immune response. This study highlights the need to develop approaches to maintaining antitumor T-cell functionality with the aim of increasing the long-term efficacy of TCR-engineered ACT immunotherapy. SIGNIFICANCE: A longitudinal functional study of adoptively transferred TCR­engineered lymphocytes yielded revealing snapshots for understanding the changes of antitumor responses over time in ACT immunotherapy of patients with advanced melanoma.


Subject(s)
Immunotherapy, Adoptive , MART-1 Antigen/immunology , Melanoma/therapy , Skin Neoplasms/therapy , T-Lymphocytes/immunology , Adult , Female , Humans , Male , Melanoma/immunology , Middle Aged , Receptors, Antigen, T-Cell/immunology , Skin Neoplasms/immunology
18.
Cancer Res ; 72(16): 3928-37, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22693252

ABSTRACT

Combining immunotherapy with targeted therapy blocking oncogenic BRAFV600 may result in improved treatments for advanced melanoma. In this study, we developed a BRAFV600E-driven murine model of melanoma, SM1, which is syngeneic to fully immunocompetent mice. SM1 cells exposed to the BRAF inhibitor vemurafenib (PLX4032) showed partial in vitro and in vivo sensitivity resulting from the inhibition of MAPK pathway signaling. Combined treatment of vemurafenib plus adoptive cell transfer therapy with lymphocytes genetically modified with a T-cell receptor (TCR) recognizing chicken ovalbumin (OVA) expressed by SM1-OVA tumors or pmel-1 TCR transgenic lymphocytes recognizing gp100 endogenously expressed by SM1 resulted in superior antitumor responses compared with either therapy alone. T-cell analysis showed that vemurafenib did not significantly alter the expansion, distribution, or tumor accumulation of the adoptively transferred cells. However, vemurafenib paradoxically increased mitogen-activated protein kinase (MAPK) signaling, in vivo cytotoxic activity, and intratumoral cytokine secretion by adoptively transferred cells. Taken together, our findings, derived from 2 independent models combining BRAF-targeted therapy with immunotherapy, support the testing of this therapeutic combination in patients with BRAFV600 mutant metastatic melanoma.


Subject(s)
Immunotherapy, Adoptive/methods , Indoles/pharmacology , Melanoma/therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Combined Modality Therapy , Female , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Melanoma/enzymology , Melanoma/immunology , Melanoma, Experimental/enzymology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Cells, Cultured , Vemurafenib
19.
J Invest Dermatol ; 132(1): 163-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21850019

ABSTRACT

Imiquimod is a synthetic Toll-like receptor 7 (TLR7) agonist approved for the topical treatment of actinic keratoses, superficial basal cell carcinoma, and genital warts. Imiquimod leads to an 80-100% cure rate of lentigo maligna; however, studies of invasive melanoma are lacking. We conducted a pilot study to characterize the local, regional, and systemic immune responses induced by imiquimod in patients with high-risk melanoma. After treatment of the primary melanoma biopsy site with placebo or imiquimod cream, we measured immune responses in the treated skin, sentinel lymph nodes (SLNs), and peripheral blood. Treatment of primary melanomas with 5% imiquimod cream was associated with an increase in both CD4+ and CD8+ T cells in the skin, and CD4+ T cells in the SLN. Most of the CD8+ T cells in the skin were CD25 negative. We could not detect any increases in CD8+ T cells specifically recognizing HLA-A(*)0201-restricted melanoma epitopes in the peripheral blood. The findings from this small pilot study demonstrate that topical imiquimod treatment results in enhanced local and regional T-cell numbers in both the skin and SLN. Further research into TLR7 immunomodulating pathways as a basis for effective immunotherapy against melanoma in conjunction with surgery is warranted.


Subject(s)
Aminoquinolines/administration & dosage , Antineoplastic Agents/administration & dosage , Immunologic Factors/administration & dosage , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Administration, Topical , Adult , Combined Modality Therapy , Female , Humans , Imiquimod , Male , Melanoma/epidemiology , Melanoma/surgery , Pilot Projects , Preoperative Care/methods , Prospective Studies , Risk Factors , Skin/drug effects , Skin/pathology , Skin Neoplasms/epidemiology , Skin Neoplasms/surgery , T-Lymphocytes/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 7/metabolism , Treatment Outcome
20.
PLoS One ; 6(12): e28973, 2011.
Article in English | MEDLINE | ID: mdl-22194965

ABSTRACT

BACKGROUND: The sustained clinical activity of the BRAF inhibitor vemurafenib (PLX4032/RG7204) in patients with BRAF(V600) mutant melanoma is limited primarily by the development of acquired resistance leading to tumor progression. Clinical trials are in progress using MEK inhibitors following disease progression in patients receiving BRAF inhibitors. However, the PI3K/AKT pathway can also induce resistance to the inhibitors of MAPK pathway. METHODOLOGY/PRINCIPAL FINDINGS: The sensitivity to vemurafenib or the MEK inhibitor AZD6244 was tested in sensitive and resistant human melanoma cell lines exploring differences in activation-associated phosphorylation levels of major signaling molecules, leading to the testing of co-inhibition of the AKT/mTOR pathway genetically and pharmacologically. There was a high degree of cross-resistance to vemurafenib and AZD6244, except in two vemurafenib-resistant cell lines that acquired a secondary mutation in NRAS. In other cell lines, acquired resistance to both drugs was associated with persistence or increase in activity of AKT pathway. siRNA-mediated gene silencing and combination therapy with an AKT inhibitor or rapamycin partially or completely reversed the resistance. CONCLUSIONS/SIGNIFICANCE: Primary and acquired resistance to vemurafenib in these in vitro models results in frequent cross resistance to MEK inhibitors, except when the resistance is the result of a secondary NRAS mutation. Resistance to BRAF or MEK inhibitors is associated with the induction or persistence of activity within the AKT pathway in the presence of these drugs. This resistance can be potentially reversed by the combination of a RAF or MEK inhibitor with an AKT or mTOR inhibitor. These combinations should be available for clinical testing in patients progressing on BRAF inhibitors.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Melanoma/enzymology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Benzimidazoles/pharmacology , Carrier Proteins/genetics , Cell Line, Tumor , Gene Silencing/drug effects , Humans , Indoles/pharmacology , Inhibitory Concentration 50 , MAP Kinase Signaling System/drug effects , Melanoma/drug therapy , Melanoma/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation/genetics , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Ribosomal Protein S6 Kinases/genetics , Sirolimus/pharmacology , Sulfonamides/pharmacology , TOR Serine-Threonine Kinases/metabolism , Vemurafenib
SELECTION OF CITATIONS
SEARCH DETAIL
...