Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Clin Invest ; 133(13)2023 07 03.
Article in English | MEDLINE | ID: mdl-37200093

ABSTRACT

During emergency hematopoiesis, hematopoietic stem cells (HSCs) rapidly proliferate to produce myeloid and lymphoid effector cells, a response that is critical against infection or tissue injury. If unresolved, this process leads to sustained inflammation, which can cause life-threatening diseases and cancer. Here, we identify a role of double PHD fingers 2 (DPF2) in modulating inflammation. DPF2 is a defining subunit of the hematopoiesis-specific BAF (SWI/SNF) chromatin-remodeling complex, and it is mutated in multiple cancers and neurological disorders. We uncovered that hematopoiesis-specific Dpf2-KO mice developed leukopenia, severe anemia, and lethal systemic inflammation characterized by histiocytic and fibrotic tissue infiltration resembling a clinical hyperinflammatory state. Dpf2 loss impaired the polarization of macrophages responsible for tissue repair, induced the unrestrained activation of Th cells, and generated an emergency-like state of HSC hyperproliferation and myeloid cell-biased differentiation. Mechanistically, Dpf2 deficiency resulted in the loss of the BAF catalytic subunit BRG1 from nuclear factor erythroid 2-like 2-controlled (NRF2-controlled) enhancers, impairing the antioxidant and antiinflammatory transcriptional response needed to modulate inflammation. Finally, pharmacological reactivation of NRF2 suppressed the inflammation-mediated phenotypes and lethality of Dpf2Δ/Δ mice. Our work establishes an essential role of the DPF2-BAF complex in licensing NRF2-dependent gene expression in HSCs and immune effector cells to prevent chronic inflammation.


Subject(s)
Chromatin , Neoplasms , Mice , Animals , Antioxidants , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Chromatin Assembly and Disassembly , Inflammation/genetics , Gene Expression , DNA-Binding Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Cell Rep ; 42(1): 112027, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36848231

ABSTRACT

TET2 haploinsufficiency is a driving event in myeloid cancers and is associated with a worse prognosis in patients with acute myeloid leukemia (AML). Enhancing residual TET2 activity using vitamin C increases oxidized 5-methylcytosine (mC) formation and promotes active DNA demethylation via base excision repair (BER), which slows leukemia progression. We utilize genetic and compound library screening approaches to identify rational combination treatment strategies to improve use of vitamin C as an adjuvant therapy for AML. In addition to increasing the efficacy of several US Food and Drug Administration (FDA)-approved drugs, vitamin C treatment with poly-ADP-ribosyl polymerase inhibitors (PARPis) elicits a strong synergistic effect to block AML self-renewal in murine and human AML models. Vitamin-C-mediated TET activation combined with PARPis causes enrichment of chromatin-bound PARP1 at oxidized mCs and γH2AX accumulation during mid-S phase, leading to cell cycle stalling and differentiation. Given that most AML subtypes maintain residual TET2 expression, vitamin C could elicit broad efficacy as a PARPi therapeutic adjuvant.


Subject(s)
Leukemia , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Humans , Mice , Ascorbic Acid/pharmacology , Ascorbic Acid/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Synthetic Lethal Mutations , Vitamins
3.
NPJ Precis Oncol ; 6(1): 95, 2022 Dec 27.
Article in English | MEDLINE | ID: mdl-36575215

ABSTRACT

Third-generation EGFR tyrosine kinase inhibitors (EGFR-TKIs), including osimertinib, an irreversible EGFR-TKI, are important treatments for non-small cell lung cancer with EGFR-TKI sensitizing or EGFR T790M resistance mutations. While patients treated with osimertinib show clinical benefit, disease progression and drug resistance are common. Emergence of de novo acquired resistance from a drug tolerant persister (DTP) cell population is one mechanism proposed to explain progression on osimertinib and other targeted cancer therapies. Here we profiled osimertinib DTPs using RNA-seq and ATAC-seq to characterize the features of these cells and performed drug screens to identify therapeutic vulnerabilities. We identified several vulnerabilities in osimertinib DTPs that were common across models, including sensitivity to MEK, AURKB, BRD4, and TEAD inhibition. We linked several of these vulnerabilities to gene regulatory changes, for example, TEAD vulnerability was consistent with evidence of Hippo pathway turning off in osimertinib DTPs. Last, we used genetic approaches using siRNA knockdown or CRISPR knockout to validate AURKB, BRD4, and TEAD as the direct targets responsible for the vulnerabilities observed in the drug screen.

4.
JCI Insight ; 6(19)2021 10 08.
Article in English | MEDLINE | ID: mdl-34622806

ABSTRACT

Myelodysplastic syndromes (MDS) are hematopoietic stem and progenitor cell (HSPC) malignancies characterized by ineffective hematopoiesis and an increased risk of leukemia transformation. Epigenetic regulators are recurrently mutated in MDS, directly implicating epigenetic dysregulation in MDS pathogenesis. Here, we identified a tumor suppressor role of the acetyltransferase p300 in clinically relevant MDS models driven by mutations in the epigenetic regulators TET2, ASXL1, and SRSF2. The loss of p300 enhanced the proliferation and self-renewal capacity of Tet2-deficient HSPCs, resulting in an increased HSPC pool and leukemogenicity in primary and transplantation mouse models. Mechanistically, the loss of p300 in Tet2-deficient HSPCs altered enhancer accessibility and the expression of genes associated with differentiation, proliferation, and leukemia development. Particularly, p300 loss led to an increased expression of Myb, and the depletion of Myb attenuated the proliferation of HSPCs and improved the survival of leukemia-bearing mice. Additionally, we show that chemical inhibition of p300 acetyltransferase activity phenocopied Ep300 deletion in Tet2-deficient HSPCs, whereas activation of p300 activity with a small molecule impaired the self-renewal and leukemogenicity of Tet2-deficient cells. This suggests a potential therapeutic application of p300 activators in the treatment of MDS with TET2 inactivating mutations.


Subject(s)
Cell Differentiation/genetics , Cell Proliferation/genetics , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , p300-CBP Transcription Factors/genetics , Animals , DNA-Binding Proteins/genetics , Dioxygenases/genetics , Disease Models, Animal , Disease Progression , Epigenesis, Genetic , Hematopoietic Stem Cells , Leukemia, Myeloid, Acute/metabolism , Mice , Mutation , Myelodysplastic Syndromes/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Repressor Proteins/genetics , Serine-Arginine Splicing Factors/genetics , Survival Rate
5.
Sci Adv ; 6(23): eaaz7249, 2020 06.
Article in English | MEDLINE | ID: mdl-32548262

ABSTRACT

RING1B, a core Polycomb repressive complex 1 subunit, is a histone H2A ubiquitin ligase essential for development. RING1B is overexpressed in patients with luminal breast cancer (BC) and recruited to actively transcribed genes and enhancers co-occupied by the estrogen receptor α (ERα). Whether ERα-induced transcriptional programs are mediated by RING1B is not understood. We show that prolonged estrogen administration induces transcriptional output and chromatin landscape fluctuations. RING1B loss impairs full estrogen-mediated gene expression and chromatin accessibility for key BC transcription factors. These effects were mediated, in part, by RING1B enzymatic activity and nucleosome binding functions. RING1B is recruited in a cyclic manner to ERα, FOXA1, and GRHL2 cobound sites and regulates estrogen-induced enhancers and ERα recruitment. Last, ChIP exo revealed multiple binding events of these factors at single-nucleotide resolution, including RING1B occupancy approximately 10 base pairs around ERα bound sites. We propose RING1B as a key regulator of the dynamic, liganded-ERα transcriptional regulatory circuit in luminal BC.


Subject(s)
Breast Neoplasms , Estrogen Receptor alpha , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Chromatin/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Estrogens/pharmacology , Female , Gene Expression Regulation, Neoplastic , Humans , Polycomb Repressive Complex 1/metabolism
6.
Nucleic Acids Res ; 48(6): 2956-2968, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32025719

ABSTRACT

Protein arginine methyltransferase 5 (PRMT5) catalyzes the symmetric di-methylation of arginine residues in histones H3 and H4, marks that are generally associated with transcriptional repression. However, we found that PRMT5 inhibition or depletion led to more genes being downregulated than upregulated, indicating that PRMT5 can also act as a transcriptional activator. Indeed, the global level of histone H3K27me3 increases in PRMT5 deficient cells. Although PRMT5 does not directly affect PRC2 enzymatic activity, methylation of histone H3 by PRMT5 abrogates its subsequent methylation by PRC2. Treating AML cells with an EZH2 inhibitor partially restored the expression of approximately 50% of the genes that are initially downregulated by PRMT5 inhibition, suggesting that the increased H3K27me3 could directly or indirectly contribute to the transcription repression of these genes. Indeed, ChIP-sequencing analysis confirmed an increase in the H3K27me3 level at the promoter region of a quarter of these genes in PRMT5-inhibited cells. Interestingly, the anti-proliferative effect of PRMT5 inhibition was also partially rescued by treatment with an EZH2 inhibitor in several leukemia cell lines. Thus, PRMT5-mediated crosstalk between histone marks contributes to its functional effects.


Subject(s)
Arginine/metabolism , Histones/metabolism , Polycomb-Group Proteins/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Transcription, Genetic , Animals , Cell Cycle/genetics , Cell Line, Tumor , Gene Deletion , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Methylation , Mice, Knockout , Models, Biological , Nucleosomes/metabolism , Protein-Arginine N-Methyltransferases/antagonists & inhibitors
7.
Nat Commun ; 10(1): 4925, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31664040

ABSTRACT

AML1-ETO (AE) is a fusion transcription factor, generated by the t(8;21) translocation, that functions as a leukemia promoting oncogene. Here, we demonstrate that TATA-Box Binding Protein Associated Factor 1 (TAF1) associates with K43 acetylated AE and this association plays a pivotal role in the proliferation of AE-expressing acute myeloid leukemia (AML) cells. ChIP-sequencing indicates significant overlap of the TAF1 and AE binding sites. Knockdown of TAF1 alters the association of AE with chromatin, affecting of the expression of genes that are activated or repressed by AE. Furthermore, TAF1 is required for leukemic cell self-renewal and its reduction promotes the differentiation and apoptosis of AE+ AML cells, thereby impairing AE driven leukemogenesis. Together, our findings reveal a role of TAF1 in leukemogenesis and identify TAF1 as a potential therapeutic target for AE-expressing leukemia.


Subject(s)
Carcinogenesis/pathology , Core Binding Factor Alpha 2 Subunit/metabolism , Histone Acetyltransferases/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Oncogene Proteins, Fusion/metabolism , RUNX1 Translocation Partner 1 Protein/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Acetylation , Animals , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Cell Self Renewal , Chromatin/metabolism , Gene Expression Regulation, Leukemic , Histone Acetyltransferases/chemistry , Humans , Lysine/metabolism , Mice, Inbred C57BL , Myeloid Cells/pathology , Protein Binding , Protein Domains , TATA-Binding Protein Associated Factors/chemistry , Transcription Factor TFIID/chemistry
8.
Craniomaxillofac Trauma Reconstr ; 12(2): 156-162, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31073367

ABSTRACT

Mandibular fractures are a common injury encountered by facial trauma surgeons. A majority of these cases are in dentate patients and can predictably be treated with several different open or closed techniques. Edentulous mandible fractures can be challenging as maxillomandibular fixation, either as the sole treatment or used for fracture reduction and stabilization prior to internal fixation, is not possible. The atrophic edentulous mandible fracture poses an even greater challenge, as there is more sclerotic bone present and less bone volume for bony contact, both of which can impair healing. In addition, with less bone mass, available plate adaptation and fixation are difficult. In recent years, virtual surgical planning (VSP) has been increasingly used in craniofacial and maxillofacial surgeries as well as in dentistry. Utilizing VSP to fabricate the necessary hardware prior to open reduction and internal fixation of atrophic edentulous mandible fractures can be helpful in treating these cases. Two cases where this method was used are presented.

11.
Cell Rep ; 24(10): 2643-2657, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30184499

ABSTRACT

Protein arginine methyltransferase 5 (PRMT5) is overexpressed in many cancer types and is a promising therapeutic target for several of them, including leukemia and lymphoma. However, we and others have reported that PRMT5 is essential for normal physiology. This dependence may become dose limiting in a therapeutic setting, warranting the search for combinatorial approaches. Here, we report that PRMT5 depletion or inhibition impairs homologous recombination (HR) DNA repair, leading to DNA-damage accumulation, p53 activation, cell-cycle arrest, and cell death. PRMT5 symmetrically dimethylates histone and non-histone substrates, including several components of the RNA splicing machinery. We find that PRMT5 depletion or inhibition induces aberrant splicing of the multifunctional histone-modifying and DNA-repair factor TIP60/KAT5, which selectively affects its lysine acetyltransferase activity and leads to impaired HR. As HR deficiency sensitizes cells to PARP inhibitors, we demonstrate here that PRMT5 and PARP inhibitors have synergistic effects on acute myeloid leukemia cells.


Subject(s)
Protein-Arginine N-Methyltransferases/metabolism , Alternative Splicing/genetics , Alternative Splicing/physiology , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/physiology , Cell Death , Cell Line, Tumor , DNA Repair/genetics , DNA Repair/physiology , Histone Code/genetics , Histone Code/physiology , Histones/metabolism , Humans , Lysine Acetyltransferase 5/genetics , Lysine Acetyltransferase 5/metabolism , Lysine Acetyltransferases/genetics , Lysine Acetyltransferases/metabolism , Protein-Arginine N-Methyltransferases/genetics
12.
Cancer Cell ; 33(6): 1111-1127.e5, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29894694

ABSTRACT

Chromatin-modifying enzymes, and specifically the protein arginine methyltransferases (PRMTs), have emerged as important targets in cancer. Here, we investigated the role of CARM1 in normal and malignant hematopoiesis. Using conditional knockout mice, we show that loss of CARM1 has little effect on normal hematopoiesis. Strikingly, knockout of Carm1 abrogates both the initiation and maintenance of acute myeloid leukemia (AML) driven by oncogenic transcription factors. We show that CARM1 knockdown impairs cell-cycle progression, promotes myeloid differentiation, and ultimately induces apoptosis. Finally, we utilize a selective, small-molecule inhibitor of CARM1 to validate the efficacy of CARM1 inhibition in leukemia cells in vitro and in vivo. Collectively, this work suggests that targeting CARM1 may be an effective therapeutic strategy for AML.


Subject(s)
Gene Expression Regulation, Leukemic , Hematopoiesis/genetics , Leukemia, Myeloid/genetics , Protein-Arginine N-Methyltransferases/genetics , Acute Disease , Animals , Apoptosis/genetics , Cell Cycle/genetics , Cell Line, Tumor , Gene Expression Profiling , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Protein-Arginine N-Methyltransferases/metabolism
13.
Biomaterials ; 157: 1-16, 2018 03.
Article in English | MEDLINE | ID: mdl-29216500

ABSTRACT

The purpose of this work was to investigate new bone formation in macroporous iron foams coated with strontium (FeSr) or bisphosphonate (FeBiP) compared to plain iron foam (Fe) and empty defect in a critical size metaphyseal bone defect model in ovariectomized rats. 60 female rats were subjected to bilateral ovariectomy and multi-deficient diet for 3 months. A 4 mm wedge shaped metaphyseal osteotomy was created, fixed with a mini-plate and subsequently filled with Fe, FeSr, FeBiP or left empty. After 6 weeks, µCt analysis revealed a statistically significant increased bone formation at the implant interface in FeSr compared to FeBiP (p = 0.035) and Fe (p = 0.002), respectively. Increased mineralized tissue was also seen within the pores in FeSr (p = 0.023) compared to Fe. Histomorphometry revealed significantly increased bone formation at the implant interface in FeSr (p < 0.001) and FeBiP (p = 0.006) compared to plain Fe with increased osteoblast and decreased osteoclast activity in combination with increased BMP2 and decreased RANKL/OPG in immunohistochemistry. ToF-SIMS analysis showed overlapping Ca signals with Fe for both FeSr and FeBiP thereby indicating tissue in-growth into the scaffolds. In conclusion, iron foam with strontium or bisphosphonate coating are of further interest in metaphyseal fracture defects in osteopenic bone.


Subject(s)
Diphosphonates/pharmacology , Fracture Healing , Iron/chemistry , Osteogenesis/drug effects , Osteoporotic Fractures/drug therapy , Strontium/pharmacology , Tissue Scaffolds/chemistry , Animals , Bone Density Conservation Agents/pharmacology , Female , Osteoporotic Fractures/etiology , Osteoporotic Fractures/pathology , Ovariectomy/adverse effects , Rats , Rats, Sprague-Dawley
14.
Blood ; 129(20): 2782-2792, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28381396

ABSTRACT

AML1-ETO (AE), a fusion oncoprotein generated by t(8;21), can trigger acute myeloid leukemia (AML) in collaboration with mutations including c-Kit, ASXL1/2, FLT3, N-RAS, and K-RAS. Caspase-3, a key executor among its family, plays multiple roles in cellular processes, including hematopoietic development and leukemia progression. Caspase-3 was revealed to directly cleave AE in vitro, suggesting that AE may accumulate in a Caspase-3-compromised background and thereby accelerate leukemogenesis. Therefore, we developed a Caspase-3 knockout genetic mouse model of AML and found that loss of Caspase-3 actually delayed AML1-ETO9a (AE9a)-driven leukemogenesis, indicating that Caspase-3 may play distinct roles in the initiation and/or progression of AML. We report here that loss of Caspase-3 triggers a conserved, adaptive mechanism, namely autophagy (or macroautophagy), which acts to limit AE9a-driven leukemia. Furthermore, we identify ULK1 as a novel substrate of Caspase-3 and show that upregulation of ULK1 drives autophagy initiation in leukemia cells and that inhibition of ULK1 can rescue the phenotype induced by Caspase-3 deletion in vitro and in vivo. Collectively, these data highlight Caspase-3 as an important regulator of autophagy in AML and demonstrate that the balance and selectivity between its substrates can dictate the pace of disease.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Autophagy , Carcinogenesis/pathology , Caspase 3/metabolism , Leukemia/metabolism , Leukemia/pathology , Oncogene Proteins, Fusion/metabolism , Animals , Antigens, CD34/metabolism , Autophagy-Related Protein-1 Homolog/antagonists & inhibitors , Cell Self Renewal , Disease Models, Animal , Fetus/pathology , Gene Deletion , Gene Knockdown Techniques , Humans , Liver Transplantation , Mice, Inbred C57BL , Mice, Knockout , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Substrate Specificity
15.
AJR Am J Roentgenol ; 204(1): W11-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25539263

ABSTRACT

OBJECTIVE: The purpose of this study was to evaluate the role of CT perfusion in monitoring response to neoadjuvant antiangiogenic and radiation therapy in resectable soft-tissue sarcomas and correlate the findings with tumor size, circulating and tumor biomarkers, and gene expression. SUBJECTS AND METHODS: This phase II clinical trial included 20 patients (13 men and 7 women; mean age, 55 years) with soft-tissue sarcomas who were undergoing treatment with the antiangiogenic drug bevacizumab followed by bevacizumab, radiation, and surgical resection. The patients underwent CT perfusion and diagnostic contrast-enhanced CT at baseline, at 2 weeks after bevacizumab therapy, and after completion of bevacizumab and radiation therapy. Multiple CT perfusion parameters (blood flow, blood volume, mean transit time, and permeability) were correlated with tumor size, circulating and tumor biomarkers, and gene expression. RESULTS: Two weeks after bevacizumab therapy, there was substantial fall in blood volume (31.9% reduction, p = 0.01) with more pronounced reduction in blood flow, blood volume, and permeability after treatment completion (53-64% reduction in blood flow, blood volume, and permeability; p = 0.001), whereas tumor size showed no significant change (p = 0.34). Tumors with higher baseline blood volume and lower baseline tumor size showed superior response to bevacizumab and radiation (p = 0.05). There was also an increase in median plasma vascular endothelial growth factor and placental-derived growth factor concentration after bevacizumab therapy paralleled by a decrease in tumor perfusion depicted by CT perfusion, although this was not statistically significant (p = 0.4). The baseline tumor microvessel density (MVD) correlated with blood flow (p = 0.04). At least 20 different genes were differentially expressed in tumors with higher and lower baseline perfusion. CONCLUSION: CT perfusion is more sensitive than tumor size for monitoring early and late response to bevacizumab and radiation therapy. CT perfusion parameters correlate with MVD, and the gene expression levels of baseline tumors could potentially predict treatment response.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Biomarkers, Tumor/metabolism , Chemoradiotherapy/methods , Perfusion Imaging/methods , Sarcoma/diagnosis , Sarcoma/therapy , Tomography, X-Ray Computed/methods , Adult , Angiogenesis Inhibitors/administration & dosage , Antineoplastic Agents/administration & dosage , Bevacizumab , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Neoadjuvant Therapy , Neoplastic Cells, Circulating/pathology , Neovascularization, Pathologic/diagnosis , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Sarcoma/metabolism , Treatment Outcome
16.
Int J Cancer ; 132(1): 29-41, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22684860

ABSTRACT

Increased levels of hypoxia and hypoxia-inducible factor 1α (HIF-1α) in human sarcomas correlate with tumor progression and radiation resistance. Prolonged antiangiogenic therapy of tumors not only delays tumor growth but may also increase hypoxia and HIF-1α activity. In our recent clinical trial, treatment with the vascular endothelial growth factor A (VEGF-A) antibody, bevacizumab, followed by a combination of bevacizumab and radiation led to near complete necrosis in nearly half of sarcomas. Gene Set Enrichment Analysis of microarrays from pretreatment biopsies found that the Gene Ontology category "Response to hypoxia" was upregulated in poor responders and that the hierarchical clustering based on 140 hypoxia-responsive genes reliably separated poor responders from good responders. The most commonly used chemotherapeutic drug for sarcomas, doxorubicin (Dox), was recently found to block HIF-1α binding to DNA at low metronomic doses. In four sarcoma cell lines, HIF-1α shRNA or Dox at low concentrations blocked HIF-1α induction of VEGF-A by 84-97% and carbonic anhydrase 9 by 83-93%. HT1080 sarcoma xenografts had increased hypoxia and/or HIF-1α activity with increasing tumor size and with anti-VEGF receptor antibody (DC101) treatment. Combining DC101 with HIF-1α shRNA or metronomic Dox had a synergistic effect in suppressing growth of HT1080 xenografts, at least in part via induction of tumor endothelial cell apoptosis. In conclusion, sarcomas respond to increased hypoxia by expressing HIF-1α target genes that may promote resistance to antiangiogenic and other therapies. HIF-1α inhibition blocks this evasive resistance and augments destruction of the tumor vasculature.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Sarcoma/therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Bevacizumab , Carbonic Anhydrase IX , Carbonic Anhydrases/genetics , Carbonic Anhydrases/metabolism , Cell Hypoxia/physiology , Cell Line , Cell Line, Tumor , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Doxorubicin/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Genetic Therapy , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Sarcoma/genetics , Sarcoma/metabolism , Sarcoma/pathology , Vascular Endothelial Growth Factor A/genetics
17.
Gastroenterology ; 141(1): 141-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21440548

ABSTRACT

BACKGROUND & AIMS: A single nucleotide polymorphism 61*G (rs4444903) in the epidermal growth factor (EGF) gene has been associated, in 2 case-control studies, with hepatocellular carcinoma (HCC). We tested associations between demographic, clinical, and genetic data and development of HCC, and developed a simple predictive model in a cohort of patients with chronic hepatitis C and advanced fibrosis. METHODS: Black and white subjects from the Hepatitis C Antiviral Long-term Treatment against Cirrhosis (HALT-C) trial (n=816) were followed up prospectively for development of a definite or presumed case of HCC for a median time period of 6.1 years. We used the Cox proportional hazards regression model to determine the hazard ratio for risk of HCC and to develop prediction models. RESULTS: Subjects with EGF genotype G/G had a higher adjusted risk for HCC than those with genotype A/A (hazard ratio, 2.10; 95% confidence interval, 1.05-4.23; P=.03). After adjusting for EGF genotype, blacks had no increased risk of HCC risk compared with whites. Higher serum levels of EGF were observed among subjects with at least one G allele (P=.08); the subset of subjects with EGF G/G genotype and above-median serum levels of EGF had the highest risk of HCC. We developed a simple prediction model that included the EGF genotype to identify patients at low, intermediate, and high risk for HCC; 6-year cumulative HCC incidences were 2.3%, 10.4%, and 26%, respectively. CONCLUSIONS: We associated the EGF genotype G/G with increased risk for HCC; differences in its frequency among black and white subjects might account for differences in HCC incidence between these groups. We developed a model that incorporates EGF genotype and demographic and clinical variables to identify patients at low, intermediate, and high risk for HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , ErbB Receptors/genetics , Hepatitis C, Chronic/genetics , Liver Cirrhosis/genetics , Liver Neoplasms/genetics , Polymorphism, Single Nucleotide , Adult , Black or African American/genetics , Antiviral Agents/therapeutic use , Carcinoma, Hepatocellular/ethnology , Carcinoma, Hepatocellular/virology , Disease Progression , ErbB Receptors/blood , Female , Gene Frequency , Genetic Predisposition to Disease , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/ethnology , Humans , Incidence , Kaplan-Meier Estimate , Liver Cirrhosis/drug therapy , Liver Cirrhosis/ethnology , Liver Cirrhosis/virology , Liver Neoplasms/ethnology , Liver Neoplasms/virology , Logistic Models , Male , Middle Aged , Phenotype , Proportional Hazards Models , Prospective Studies , Risk Assessment , Risk Factors , Time Factors , United States , White People/genetics
18.
Int J Radiat Oncol Biol Phys ; 81(4): 1081-90, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-20932656

ABSTRACT

PURPOSE: Numerous preclinical studies have demonstrated that angiogenesis inhibitors can increase the efficacy of radiotherapy (RT). We sought to examine the safety and efficacy of bevacizumab (BV) and RT in soft tissue sarcomas and explore biomarkers to help determine the treatment response. METHODS AND MATERIALS: Patients with ≥5 cm, intermediate- or high-grade soft tissue sarcomas at significant risk of local recurrence received neoadjuvant BV alone followed by BV plus RT before surgical resection. Correlative science studies included analysis of the serial blood and tumor samples and serial perfusion computed tomography scans. RESULTS: The 20 patients had a median tumor size of 8.25 cm, with 13 extremity, 1 trunk, and 6 retroperitoneal/pelvis tumors. The neoadjuvant treatment was well tolerated, with only 4 patients having Grade 3 toxicities (hypertension, liver function test elevation). BV plus RT resulted in ≥80% pathologic necrosis in 9 (45%) of 20 tumors, more than double the historical rate seen with RT alone. Three patients had a complete pathologic response. The median microvessel density decreased 53% after BV alone (p <.05). After combination therapy, the median tumor cell proliferation decreased by 73%, apoptosis increased 10.4-fold, and the blood flow, blood volume, and permeability surface area decreased by 62-72% (p <.05). Analysis of gene expression microarrays of untreated tumors identified a 24-gene signature for treatment response. The microvessel density and circulating progenitor cells at baseline and the reduction in microvessel density and plasma soluble c-KIT with BV therapy also correlated with a good pathologic response (p <.05). After a median follow-up of 20 months, only 1 patient had developed local recurrence. CONCLUSIONS: The results from the present exploratory study indicated that BV increases the efficacy of RT against soft tissue sarcomas and might reduce the incidence of local recurrence. Thus, this regimen warrants additional investigation. Gene expression profiles and other tissue and circulating biomarkers showed promising correlations with treatment response.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Sarcoma/therapy , Soft Tissue Neoplasms/therapy , Adult , Aged , Angiogenesis Inhibitors/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , Bevacizumab , Biomarkers, Tumor/blood , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Female , Gene Expression Profiling , Humans , Male , Microvessels/drug effects , Microvessels/pathology , Microvessels/radiation effects , Middle Aged , Neoadjuvant Therapy/adverse effects , Neoadjuvant Therapy/methods , Neoplasm Recurrence, Local/prevention & control , Postoperative Complications , Radiotherapy Dosage , Sarcoma/blood , Sarcoma/blood supply , Sarcoma/pathology , Soft Tissue Neoplasms/blood supply , Soft Tissue Neoplasms/pathology , Treatment Outcome , Tumor Burden/drug effects , Tumor Burden/radiation effects
19.
Cancer Res ; 70(21): 8357-67, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20978198

ABSTRACT

Tumors induce new blood vessel growth primarily from host organ microvascular endothelial cells (EC), and microvasculature differs significantly between the lung and liver. Vascular endothelial growth factor (VEGF or VEGF-A) promotion of tumor angiogenesis is thought to be mediated primarily by VEGF receptor-2 (VEGFR-2). In this study, VEGFR-2 antibody (DC101) inhibited growth of RenCa renal cell carcinoma lung metastases by 26%, whereas VEGFR-1 antibody (MF-1) had no effect. However, VEGFR-2 neutralization had no effect on RenCa liver metastases, whereas VEGFR-1 neutralization decreased RenCa liver metastases by 31%. For CT26 colon carcinoma liver metastases, inhibition of both VEGFR-1 and VEGFR-2 was required to induce growth delay. VEGFR-1 or VEGFR-2 inhibition decreased tumor burden not by preventing the establishment of micrometastases but rather by preventing vascularization and growth of micrometastases by 55% and 43%, respectively. VEGF induced greater phosphorylation of VEGFR-2 in lung ECs and of VEGFR-1 in liver ECs. EC proliferation, migration, and capillary tube formation in vitro were suppressed more by VEGFR-2 inhibition for lung EC and more by VEGFR-1 inhibition for liver EC. Collectively, our results indicate that liver metastases are more reliant on VEGFR-1 than lung metastases to mediate angiogenesis due to differential activity of VEGFRs on liver EC versus lung EC. Thus, therapies inhibiting specific VEGFRs should consider the targeted sites of metastatic disease.


Subject(s)
Antibodies, Neutralizing/pharmacology , Neoplasm Metastasis , Vascular Endothelial Growth Factor Receptor-1/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology , Animals , Blotting, Western , Carcinoma, Renal Cell/secondary , Cell Adhesion , Cell Movement , Cell Proliferation , Cells, Cultured , Colonic Neoplasms/pathology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Fluorescent Antibody Technique , Hemangiosarcoma/metabolism , Hemangiosarcoma/pathology , Hemangiosarcoma/prevention & control , Humans , Immunoenzyme Techniques , Kidney Neoplasms/pathology , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/prevention & control , Organ Specificity , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
20.
Cancer Res ; 70(3): 948-56, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20103648

ABSTRACT

Metastasis relies on angiogenesis for tumor expansion. Tumor angiogenesis is restrained by a variety of endogenous inhibitors, including thrombospondin 1 (TSP1). The principal antiangiogenic activity of TSP1 resides in a domain containing three TSP1 repeats (3TSR), and TSP1 cleavage is regulated, in part, by the metalloproteinase ADAMTS1. In this study, we examined the role of TSP1 and ADAMTS1 in controlling metastatic disease in the liver and lung. TSP1 overexpression inhibited metastatic growth of colon or renal carcinoma cells in liver but not lung. Metastatic melanoma in liver grew more rapidly in Tsp1-null mice compared with controls, whereas in lung grew similarly in Tsp1-null mice or controls. Recombinant TSP1 was cleaved more efficiently in lysates from liver than lung. ADAMTS1 inhibition by neutralizing antibody, small interfering RNA, or genetic deletion abrogated cleavage activity. To confirm that lack of cleavage of TSP1 ablated its antiangiogenic function in the lung, we generated colon cancer cells stably secreting only the 3TSR domain and found that they inhibited formation of both liver and lung metastases. Collectively, our results indicate that the antiangiogenic activity of TSP1 is differentially regulated by ADAMTS1 in the liver and lung, emphasizing the concept that regulation of angiogenesis is varied in different tissue environments.


Subject(s)
ADAM Proteins/metabolism , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Neoplasms, Experimental/pathology , Thrombospondin 1/metabolism , ADAM Proteins/genetics , ADAMTS1 Protein , Animals , Binding Sites , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Proliferation , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Thrombospondin 1/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...