Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Oncotarget ; 10(47): 4822-4839, 2019 Aug 06.
Article in English | MEDLINE | ID: mdl-31448050

ABSTRACT

Mesenchymal stem-like (MSL) breast cancers are enriched for cells with tumor reconstituting and mesenchymal characteristics. These cancers are often triple-negative and have a poor prognosis. Few effective targeted treatment options exist for patients with these cancers, and even when targeted therapies exist, resistance often arises and tumors recur, due in part to drug-tolerant persisting tumor cells with self-renewal capability. Effective treatment strategies will combine agents that target the bulk-tumor and reconstituting cells. In order to identify such a combination therapy, we conducted an inhibitor screen using 40 targeted agents at three different doses in all pairwise combinations. Checkpoint Kinase 1 (CHK1) inhibitors were identified as potent inhibitors of MSL breast cancers. When combined with a pro-apoptotic agent/B Cell Lymphoma 2 (BCL2) inhibitor, the effectiveness of the combination regimen was super-additive compared to either treatment alone and was selective for MSL cancers. Treatment of MSL breast cancer cells results in DNA damage, cell-cycle defects characterized by a prolonged S-phase, increased apoptosis and decreased colony forming abilities compared to untreated cells. These data suggest that a combination of a CHK1 and BCL2 inhibitor could be an effective treatment for patients with MSL breast cancer. Several other effective drug combinations were also identified.

2.
Mol Cancer Ther ; 16(6): 1041-1053, 2017 06.
Article in English | MEDLINE | ID: mdl-28292938

ABSTRACT

Pancreatic adenocarcinoma (PDAC) is the fourth most common cause of cancer-related death in the United States. PDAC is difficult to manage effectively, with a five-year survival rate of only 5%. PDAC is largely driven by activating KRAS mutations, and as such, cannot be directly targeted with therapeutic agents that affect the activated protein. Instead, inhibition of downstream signaling and other targets will be necessary to effectively manage PDAC. Here, we describe a tiered single-agent and combination compound screen to identify targeted agents that impair growth of a panel of PDAC cell lines. Several of the combinations identified from the screen were further validated for efficacy and mechanism. Combination of the bromodomain inhibitor JQ1 and the neddylation inhibitor MLN4294 altered the production of reactive oxygen species in PDAC cells, ultimately leading to defects in the DNA damage response. Dual bromodomain/neddylation blockade inhibited in vivo growth of PDAC cell line xenografts. Overall, this work revealed novel combinatorial regimens, including JQ1 plus MLN4294, which show promise for the treatment of RAS-driven PDAC. Mol Cancer Ther; 16(6); 1041-53. ©2017 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , Dose-Response Relationship, Drug , Drug Combinations , Drug Screening Assays, Antitumor/methods , Drug Synergism , High-Throughput Nucleotide Sequencing , Humans , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Targeted Therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Superoxides , Xenograft Model Antitumor Assays , Pancreatic Neoplasms
3.
Cancer Res ; 77(2): 566-578, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27872098

ABSTRACT

Triple-negative breast cancer (TNBC) remains an aggressive disease without effective targeted therapies. In this study, we addressed this challenge by testing 128 FDA-approved or investigational drugs as either single agents or in 768 pairwise drug combinations in TNBC cell lines to identify synergistic combinations tractable to clinical translation. Medium-throughput results were scrutinized and extensively analyzed for sensitivity patterns, synergy, anticancer activity, and were validated in low-throughput experiments. Principal component analysis revealed that a fraction of all upregulated or downregulated genes of a particular targeted pathway could partly explain cell sensitivity toward agents targeting that pathway. Combination therapies deemed immediately tractable to translation included ABT-263/crizotinib, ABT-263/paclitaxel, paclitaxel/JQ1, ABT-263/XL-184, and paclitaxel/nutlin-3, all of which exhibited synergistic antiproliferative and apoptotic activity in multiple TNBC backgrounds. Mechanistic investigations of the ABT-263/crizotinib combination offering a potentially rapid path to clinic demonstrated RTK blockade, inhibition of mitogenic signaling, and proapoptotic signal induction in basal and mesenchymal stem-like TNBC. Our findings provide preclinical proof of concept for several combination treatments of TNBC, which offer near-term prospects for clinical translation. Cancer Res; 77(2); 566-78. ©2016 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Screening Assays, Antitumor/methods , Triple Negative Breast Neoplasms , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Female , Flow Cytometry , Humans , Immunoprecipitation , Principal Component Analysis
4.
Proc Natl Acad Sci U S A ; 113(42): E6409-E6417, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27702896

ABSTRACT

Genetically engineered mouse models (GEMMs) of cancer are increasingly being used to assess putative driver mutations identified by large-scale sequencing of human cancer genomes. To accurately interpret experiments that introduce additional mutations, an understanding of the somatic genetic profile and evolution of GEMM tumors is necessary. Here, we performed whole-exome sequencing of tumors from three GEMMs of lung adenocarcinoma driven by mutant epidermal growth factor receptor (EGFR), mutant Kirsten rat sarcoma viral oncogene homolog (Kras), or overexpression of MYC proto-oncogene. Tumors from EGFR- and Kras-driven models exhibited, respectively, 0.02 and 0.07 nonsynonymous mutations per megabase, a dramatically lower average mutational frequency than observed in human lung adenocarcinomas. Tumors from models driven by strong cancer drivers (mutant EGFR and Kras) harbored few mutations in known cancer genes, whereas tumors driven by MYC, a weaker initiating oncogene in the murine lung, acquired recurrent clonal oncogenic Kras mutations. In addition, although EGFR- and Kras-driven models both exhibited recurrent whole-chromosome DNA copy number alterations, the specific chromosomes altered by gain or loss were different in each model. These data demonstrate that GEMM tumors exhibit relatively simple somatic genotypes compared with human cancers of a similar type, making these autochthonous model systems useful for additive engineering approaches to assess the potential of novel mutations on tumorigenesis, cancer progression, and drug sensitivity.


Subject(s)
Adenocarcinoma/genetics , Cell Transformation, Neoplastic/genetics , ErbB Receptors/genetics , Genes, myc , Genes, ras , Lung Neoplasms/genetics , Mutation , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Carcinogens , DNA Copy Number Variations , DNA Mutational Analysis , Disease Models, Animal , Gene Dosage , Genome-Wide Association Study , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Point Mutation , Proto-Oncogene Mas , ROC Curve , Exome Sequencing
5.
Oncotarget ; 6(35): 37410-25, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26485762

ABSTRACT

Targeting anti-apoptotic proteins can sensitize tumor cells to conventional chemotherapies or other targeted agents. Antagonizing the Inhibitor of Apoptosis Proteins (IAPs) with mimetics of the pro-apoptotic protein SMAC is one such approach. We used sensitization compound screening to uncover possible agents with the potential to further sensitize lung adenocarcinoma cells to the SMAC mimetic Debio 1143. Several compounds in combination with Debio 1143, including taxanes, topoisomerase inhibitors, and bromodomain inhibitors, super-additively inhibited growth and clonogenicity of lung adenocarcinoma cells. Co-treatment with Debio 1143 and the bromodomain inhibitor JQ1 suppresses the expression of c-IAP1, c-IAP2, and XIAP. Non-canonical NF-κB signaling is also activated following Debio 1143 treatment, and Debio 1143 induces the formation of the ripoptosome in Debio 1143-sensitive cell lines. Sensitivity to Debio 1143 and JQ1 co-treatment was associated with baseline caspase-8 expression. In vivo treatment of lung adenocarcinoma xenografts with Debio 1143 in combination with JQ1 or docetaxel reduced tumor volume more than either single agent alone. As Debio 1143-containing combinations effectively inhibited both in vitro and in vivo growth of lung adenocarcinoma cells, these data provide a rationale for Debio 1143 combinations currently being evaluated in ongoing clinical trials and suggest potential utility of other combinations identified here.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azepines/pharmacology , Azocines/pharmacology , Benzhydryl Compounds/pharmacology , Camptothecin/analogs & derivatives , Cell Proliferation/drug effects , Lung Neoplasms/drug therapy , Paclitaxel/pharmacology , Taxoids/pharmacology , Topoisomerase Inhibitors/pharmacology , Triazoles/pharmacology , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Camptothecin/pharmacology , Cell Line, Tumor , Docetaxel , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Irinotecan , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/metabolism , Signal Transduction/drug effects , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
Pigment Cell Melanoma Res ; 28(4): 417-30, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25854919

ABSTRACT

BRAF inhibitors have revolutionized treatment of mutant BRAF metastatic melanomas. However, resistance develops rapidly following BRAF inhibitor treatment. We have found that BRAF-mutant melanoma cell lines are more sensitive than wild-type BRAF cells to the small molecule tyrosine kinase inhibitor dovitinib. Sensitivity is associated with inhibition of a series of known dovitinib targets. Dovitinib in combination with several agents inhibits growth more effectively than either agent alone. These combinations inhibit BRAF-mutant melanoma and colorectal carcinoma cell lines, including cell lines with intrinsic or selected BRAF inhibitor resistance. Hence, combinations of dovitinib with second agents are potentially effective therapies for BRAF-mutant melanomas, regardless of their sensitivity to BRAF inhibitors.


Subject(s)
Benzimidazoles/pharmacology , Melanoma/genetics , Melanoma/pathology , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Quinolones/pharmacology , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Humans , Indoles/pharmacology , Male , Melanoma/enzymology , Mice, Nude , Neoplasm Proteins/metabolism , Skin Neoplasms , Small Molecule Libraries/pharmacology , Sulfonamides/pharmacology , Vemurafenib , Melanoma, Cutaneous Malignant
7.
Cancer Res ; 75(7): 1399-412, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25712345

ABSTRACT

Melanoma development involves members of the AGC kinase family, including AKT, PKC, and, most recently, PDK1, as elucidated recently in studies of Braf::Pten mutant melanomas. Here, we report that PDK1 contributes functionally to skin pigmentation and to the development of melanomas harboring a wild-type PTEN genotype, which occurs in about 70% of human melanomas. The PDK1 substrate SGK3 was determined to be an important mediator of PDK1 activities in melanoma cells. Genetic or pharmacologic inhibition of PDK1 and SGK3 attenuated melanoma growth by inducing G1 phase cell-cycle arrest. In a synthetic lethal screen, pan-PI3K inhibition synergized with PDK1 inhibition to suppress melanoma growth, suggesting that focused blockade of PDK1/PI3K signaling might offer a new therapeutic modality for wild-type PTEN tumors. We also noted that responsiveness to PDK1 inhibition associated with decreased expression of pigmentation genes and increased expression of cytokines and inflammatory genes, suggesting a method to stratify patients with melanoma for PDK1-based therapies. Overall, our work highlights the potential significance of PDK1 as a therapeutic target to improve melanoma treatment.


Subject(s)
Melanoma/enzymology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/enzymology , Animals , Benzoates/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , G1 Phase Cell Cycle Checkpoints , Humans , Immediate-Early Proteins/metabolism , Indazoles/pharmacology , Lymphatic Metastasis , Melanoma/drug therapy , Melanoma/secondary , Mice, Knockout , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Pyrimidines/pharmacology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Skin/enzymology , Skin/pathology , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology
8.
Cancer Cell ; 27(1): 41-56, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25584893

ABSTRACT

Braf(V600E) induces benign, growth-arrested melanocytic nevus development, but also drives melanoma formation. Cdkn2a loss in Braf(V600E) melanocytes in mice results in rare progression to melanoma, but only after stable growth arrest as nevi. Immediate progression to melanoma is prevented by upregulation of miR-99/100, which downregulates mTOR and IGF1R signaling. mTORC1 activation through Stk11 (Lkb1) loss abrogates growth arrest of Braf(V600E) melanocytic nevi, but is insufficient for complete progression to melanoma. Cdkn2a loss is associated with mTORC2 and Akt activation in human and murine melanocytic neoplasms. Simultaneous Cdkn2a and Lkb1 inactivation in Braf(V600E) melanocytes results in activation of both mTORC1 and mTORC2/Akt, inducing rapid melanoma formation in mice. In this model, activation of both mTORC1/2 is required for Braf-induced melanomagenesis.


Subject(s)
Melanoma, Experimental/pathology , Multiprotein Complexes/metabolism , Nevus/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/metabolism , TOR Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Animals , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Melanocytes/metabolism , Melanoma, Experimental/metabolism , Mice , MicroRNAs/metabolism , Molecular Sequence Data , Mutation , Nevus/pathology , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Skin Neoplasms/pathology
9.
Mol Cancer Res ; 12(8): 1140-55, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24829397

ABSTRACT

UNLABELLED: Associations of ErbB4 (ERBB4/HER4), the fourth member of the EGFR family, with cancer are variable, possibly as a result of structural diversity of this receptor. There are multiple structural isoforms of ERBB4 arising by alternative mRNA splicing, and a subset undergo proteolysis that releases membrane-anchored and soluble isoforms that associate with transcription factors and coregulators to modulate transcription. To compare the differential and common signaling activities of full-length (FL) and soluble intracellular isoforms of ERBB4, four JM-a isoforms (FL and soluble intracellular domain (ICD) CYT-1 and CYT-2) were expressed in isogenic MCF10A cells and their biologic activities were analyzed. Both FL and ICD CYT-2 promoted cell proliferation and invasion, and CYT-1 suppressed cell growth. Transcriptional profiling revealed several new and underexplored ERBB4-regulated transcripts, including: proteases/protease inhibitors (MMP3 and SERPINE2), the YAP/Hippo pathway (CTGF, CYR61, and SPARC), the mevalonate/cholesterol pathway (HMGCR, HMGCS1, LDLR, and DHCR7), and cytokines (IL8, CCL20, and CXCL1). Many of these transcripts were subsequently validated in a luminal breast cancer cell line that normally expresses ERBB4. Furthermore, ChIP-seq experiments identified ADAP1, APOE, SPARC, STMN1, and MXD1 as novel molecular targets of ERBB4. These findings clarify the diverse biologic activities of ERBB4 isoforms, and reveal new and divergent functions. IMPLICATIONS: ErbB4 as a regulator of Hippo and mevalonate pathways provides new insight into milk production and anabolic processes in normal mammary epithelia and cancer.


Subject(s)
Epithelial Cells/metabolism , Mammary Glands, Human/metabolism , Protein Isoforms/metabolism , Receptor, ErbB-4/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cells, Cultured , Female , Humans , Protein Isoforms/genetics , Receptor, ErbB-4/genetics , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/genetics
10.
Pigment Cell Melanoma Res ; 26(4): 527-41, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23617806

ABSTRACT

The receptor tyrosine kinase AXL regulates melanoma cell proliferation and migration. We now demonstrate that AXL and the related kinase MERTK are alternately expressed in melanoma and are associated with different transcriptional signatures. MERTK-positive melanoma cells are more proliferative and less migratory than AXL-positive melanoma cells and overexpression of AXL increases cell motility relative to MERTK. MERTK is expressed in up to 50% of melanoma cells and shRNA-mediated knockdown of MERTK reduces colony formation and cell migration in a CDC42-dependent fashion. Targeting MERTK also decreases cell survival and proliferation in an AKT-dependent manner. Finally, we identify a novel mutation in the kinase domain of MERTK, MERTK(P) (802S) , that increases the motility of melanoma cells relative to wild-type MERTK. Together, these data demonstrate that MERTK is a possible therapeutic target in melanoma, that AXL and MERTK are associated with differential cell behaviors, and that mutations in MERTK may contribute to melanoma pathogenesis.


Subject(s)
Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/physiology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Cytophotometry , Gene Expression Profiling , HEK293 Cells , Humans , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Phosphorylation , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction , Skin Neoplasms/metabolism , c-Mer Tyrosine Kinase , cdc42 GTP-Binding Protein/metabolism , Axl Receptor Tyrosine Kinase
11.
Cancer Discov ; 3(1): 52-67, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23239741

ABSTRACT

UNLABELLED: Resistance and partial responses to targeted monotherapy are major obstacles in cancer treatment. Systematic approaches to identify efficacious drug combinations for cancer are not well established, especially in the context of genotype. To address this, we have tested pairwise combinations of an array of small-molecule inhibitors on early-passage melanoma cultures using combinatorial drug screening. Results reveal several inhibitor combinations effective for melanomas with activating RAS or BRAF mutations, including mutant BRAF melanomas with intrinsic or acquired resistance to vemurafenib. Inhibition of both EGF receptor and AKT sensitized treatment-resistant BRAF mutant melanoma cultures to vemurafenib. Melanomas with RAS mutations were more resistant to combination therapies relative to BRAF mutants, but were sensitive to combinations of statins and cyclin-dependent kinase inhibitors in vitro and in vivo. These results show the use of combinatorial drug screening for discovering unique treatment regimens that overcome resistance phenotypes of mutant BRAF- and RAS-driven melanomas. SIGNIFICANCE: We have used drug combinatorial screening to identify effective combinations for mutant BRAF melanomas, including those resistant to vemurafenib, and mutant RAS melanomas that are resistant to many therapies. Mechanisms governing the interactions of the drug combinations are proposed, and in vivo xenografts show the enhanced benefit and tolerability of a mutant RAS -selective combination, which is currently lacking in the clinic.


Subject(s)
Antineoplastic Agents/administration & dosage , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/genetics , ras Proteins/genetics , Animals , Cell Line, Tumor , Drug Interactions , Drug Resistance, Neoplasm , Drug Therapy, Combination , Genes, ras/genetics , High-Throughput Screening Assays , Humans , Melanoma/genetics , Mice , Mice, Nude , Xenograft Model Antitumor Assays
12.
Cancer Cell ; 20(6): 741-54, 2011 Dec 13.
Article in English | MEDLINE | ID: mdl-22172720

ABSTRACT

Malignant melanoma is characterized by frequent metastasis, however, specific changes that regulate this process have not been clearly delineated. Although it is well known that Wnt signaling is frequently dysregulated in melanoma, the functional implications of this observation are unclear. By modulating ß-catenin levels in a mouse model of melanoma that is based on melanocyte-specific Pten loss and Braf(V600E) mutation, we demonstrate that ß-catenin is a central mediator of melanoma metastasis to the lymph nodes and lungs. In addition to altering metastasis, ß-catenin levels control tumor differentiation and regulate both MAPK/Erk and PI3K/Akt signaling. Highly metastatic tumors with ß-catenin stabilization are very similar to a subset of human melanomas. Together these findings establish Wnt signaling as a metastasis regulator in melanoma.


Subject(s)
Lung Neoplasms/secondary , Melanoma, Experimental/secondary , PTEN Phosphohydrolase/deficiency , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/pathology , beta Catenin/metabolism , Animals , Antigens, Differentiation/metabolism , Benzamides , Cell Transformation, Neoplastic , Colorectal Neoplasms/secondary , Enzyme Activation , Gene Knockdown Techniques , Humans , Imatinib Mesylate , Kaplan-Meier Estimate , Lung Neoplasms/metabolism , Lymphatic Metastasis , Melanocytes/metabolism , Melanoma, Experimental/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphorylation , Piperazines/therapeutic use , Protein Stability , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/therapeutic use , Signal Transduction , Skin Neoplasms/metabolism , Splenic Neoplasms/secondary , Transcription, Genetic , Tumor Cells, Cultured , beta Catenin/genetics
13.
Cancer Gene Ther ; 9(10): 813-8, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12224021

ABSTRACT

Salmonella enterica servovar Typhimurium is capable of targeting, colonizing, and eliciting growth suppression of tumors in mice. We examined the effects of mutations on this anticancer phenotype in two Salmonella virulence gene clusters. Salmonella pathogenicity island (SPI)-1 genes promote systemic invasion from the intestine, whereas SPI-2 genes support systemic survival within macrophages and other cells. Disabling SPI-1 (prgH(-)) strongly reduced invasion in vitro, but had no effect on tumor growth suppression in vivo. However, disabling SPI-2 (ssaT(-)) ablated tumor growth suppression. In addition to ssaT(-), mutations in SPI-2 genes sseA, sseB, sseC, sscA, and ssrA also eliminated antitumor activity, whereas mutations in sseF or sseG yielded partial loss of function. Impaired tumor amplification was seen in three SPI-2 mutants tested after intravenous or intratumoral injection. A SPI-2(-) strain was unable to suppress tumor growth in CD18-deficient mice with defective macrophages and neutrophils, suggesting that loss of tumor growth suppression in wild-type mice by SPI-2 mutants was not solely a function of increased susceptibility to immune attack. Thus, SPI-2 is essential for the Salmonella antitumor effects, perhaps by aiding bacterial amplification within tumors, and is the first identified genetic system for this Salmonella phenotype.


Subject(s)
Bacterial Proteins/genetics , Melanoma, Experimental/prevention & control , Membrane Proteins/genetics , Salmonella typhimurium/physiology , Skin Neoplasms/prevention & control , Animals , CD18 Antigens/metabolism , Cell Division , Female , In Vitro Techniques , Injections, Intralesional , Melanoma, Experimental/microbiology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mutation , Neoplasm Invasiveness , Neoplasm Transplantation , Skin Neoplasms/microbiology , Skin Neoplasms/pathology , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...