Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1086102, 2023.
Article in English | MEDLINE | ID: mdl-36891296

ABSTRACT

V-domain Ig suppressor of T-cell activation (VISTA) is a B7 family member that plays key roles in maintaining T cell quiescence and regulation of myeloid cell populations, which together establish it as a novel immunotherapy target for solid tumors. Here we review the growing literature on VISTA expression in relation to various malignancies to better understand the role of VISTA and its interactions with both tumor cells and immune cells expressing other checkpoint molecules within the tumor microenvironment (TME). The biology of VISTA creates several mechanisms to maintain the TME, including supporting the function of myeloid-derived suppressor cells, regulating natural killer cell activation, supporting the survival of regulatory T cells, limiting antigen presentation on antigen-presenting cells and maintaining T cells in a quiescent state. Understanding these mechanisms is an important foundation of rational patient selection for anti-VISTA therapy. We provide a general framework to describe distinct patterns of VISTA expression in correlation with other known predictive immunotherapy biomarkers (programmed cell death ligand 1 and tumor-infiltrating lymphocytes) across solid tumors to facilitate investigation of the most efficacious TMEs for VISTA-targeted treatment as a single agent and/or in combination with anti-programmed death 1/anti-cytotoxic T lymphocyte antigen-4 therapies.


Subject(s)
B7 Antigens , Neoplasms , Humans , B7 Antigens/metabolism , Biomarkers , Neoplasms/therapy , Patient Selection , T-Lymphocytes, Regulatory , Tumor Microenvironment
2.
Oncol Rep ; 46(4)2021 Oct.
Article in English | MEDLINE | ID: mdl-34468011

ABSTRACT

The selection of effective therapeutic agents is critical for improving the survival of patients with renal cell carcinoma (RCC). The aim of the present study was to develop an ex vivo drug testing assay using patient­derived tumor organoid (TO) cultures. For this purpose, surgical tumor specimens were obtained from 20 patients with RCC. TOs were developed ex vivo from freshly resected RCC tumors, and their histopathological and molecular characteristics were evaluated using histological staining and whole­exome sequencing (WES). Using a cell viability assay, the therapeutic efficacy of standard of care tyrosine kinase inhibitors in RCC TOs was determined. It was found that TOs recapitulated the histological features of primary RCC tumors. Using WES, a strong concordance was identified at the genetic level between the primary tumors and their corresponding TOs. Using patient­derived TO models, a prototype of an ex vivo drug testing assay was developed, and it was found that RCC TOs exhibited differential responses to sunitinib, pazopanib, cabozantinib, axitinib and sorafenib treatment. On the whole, although the predictive value of the current assay has to be tested and validated in future clinical studies, the findings of the present study demonstrate a novel approach for ex vivo drug testing in patient­derived TO models, which may have potential for use in the personalized treatment of cancer patients.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Organoids/drug effects , Protein Kinase Inhibitors/pharmacology , Cell Line, Tumor , Humans , Receptor Protein-Tyrosine Kinases
4.
Int J Mol Med ; 45(2): 315-323, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31894292

ABSTRACT

Glycogen synthase kinase­3 (GSK­3), a serine/threonine kinase, is involved in a broad range of pathological processes including cancer. GSK­3 has two isoforms, GSK­3α and GSK­3ß, and GSK­3ß has been recognized as a therapeutic target for the development of new anticancer drugs. The present study aimed to investigate the antitumor effects of 9­ING­41, which is a maleimide­based ATP­competitive small molecule GSK­3ß inhibitor active in patients with advanced cancer. In renal cancer cell lines, treatment with 9­ING­41 alone induced cell cycle arrest and apoptosis, and autophagy inhibitors increased the antitumor effects of 9­ING­41 when used in combination. Treatment with 9­ING­41 potentiated the antitumor effects of targeted therapeutics and increased the cytotoxic effects of cytokine­activated immune cells on renal cancer cell lines. These results provided a compelling rationale for the inclusion of patients with renal cancer in studies of 9­ING­41, both as a single agent and in combination with current standard therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Maleimides/pharmacology , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Kidney Neoplasms/metabolism , Maleimides/chemistry , Protein Kinase Inhibitors/chemistry
5.
Sci Rep ; 9(1): 19977, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31882719

ABSTRACT

Glycogen synthase kinase-3 beta (GSK-3ß), a serine/threonine kinase, has been identified as a potential therapeutic target in human bladder cancer. In the present study, we investigated the antitumor effect of a small molecule GSK-3ß inhibitor, 9-ING-41, currently in clinical studies in patients with advanced cancer, in bladder cancer cell lines. We found that treatment with 9-ING-41 leads to cell cycle arrest, autophagy and apoptosis in bladder cancer cells. The autophagy inhibitor chloroquine potentiated the antitumor effects of 9-ING-41 when tested in combination studies. Our findings also demonstrate that 9-ING-41 enhanced the growth inhibitory effects of gemcitabine or cisplatin when used in combination in bladder cancer cells. Finally, we found that 9-ING-41 sensitized bladder cancer cells to the cytotoxic effects of human immune effector cells. Our results provide a rationale for the inclusion of patients with advanced bladder cancer in clinical studies of 9-ING-41.


Subject(s)
Antineoplastic Agents/pharmacology , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Indoles/pharmacology , Maleimides/pharmacology , Protein Kinase Inhibitors/pharmacology , Autophagy/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Drug Synergism , Humans , Urinary Bladder Neoplasms
6.
Clin Cancer Res ; 25(21): 6452-6462, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31533931

ABSTRACT

PURPOSE: Pancreatic ductal adenocarcinoma (PDAC) is a predominantly fatal common malignancy with inadequate treatment options. Glycogen synthase kinase 3ß (GSK-3ß) is an emerging target in human malignancies including PDAC.Experimental Design: Pancreatic cancer cell lines and patient-derived xenografts were treated with a novel GSK-3 inhibitor 9-ING-41 alone or in combination with chemotherapy. Activation of the DNA damage response pathway and S-phase arrest induced by gemcitabine were assessed in pancreatic tumor cells with pharmacologic inhibition or siRNA depletion of GSK-3 kinases by immunoblotting, flow cytometry, and immunofluorescence. RESULTS: 9-ING-41 treatment significantly increased pancreatic tumor cell killing when combined with chemotherapy. Inhibition of GSK-3 by 9-ING-41 prevented gemcitabine-induced S-phase arrest suggesting an impact on the ATR-mediated DNA damage response. Both 9-ING-41 and siRNA depletion of GSK-3 kinases impaired the activation of ATR leading to the phosphorylation and activation of Chk1. Mechanistically, depletion or knockdown of GSK-3 kinases resulted in the degradation of the ATR-interacting protein TopBP1, thus limiting the activation of ATR in response to single-strand DNA damage. CONCLUSIONS: These data identify a previously unknown role for GSK-3 kinases in the regulation of the TopBP1/ATR/Chk1 DNA damage response pathway. The data also support the inclusion of patients with PDAC in clinical studies of 9-ING-41 alone and in combination with gemcitabine.


Subject(s)
Adenocarcinoma/drug therapy , Ataxia Telangiectasia Mutated Proteins/genetics , Carcinoma, Pancreatic Ductal/drug therapy , Carrier Proteins/genetics , DNA-Binding Proteins/genetics , Glycogen Synthase Kinase 3 beta/genetics , Nuclear Proteins/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Checkpoint Kinase 1 , DNA Damage/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Humans , Indoles/pharmacology , Maleimides/pharmacology , Mice , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Gemcitabine
7.
Clin Cancer Res ; 25(4): 1331-1342, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30420445

ABSTRACT

PURPOSE: Response to toxicity in chemotherapies varies considerably from tissue to tissue and from patient to patient. An ability to monitor the tissue damage done by chemotherapy may have a profound impact on treatment and prognosis allowing for a proactive management in understanding and mitigating such events. For the first time, we investigated the feasibility of using whole-body imaging to map chemotherapeutic drug-induced toxicity on an individual basis. EXPERIMENTAL DESIGN: In a preclinical proof-of-concept, rats were treated with a single clinical dose of cyclophosphamide, methotrexate, or cisplatin. In vivo whole-body imaging data were acquired using 99mTc-duramycin, which identifies dead and dying cells as an unambiguous marker for tissue injury in susceptible organs. Imaging results were cross-validated using quantitative ex vivo measurements and histopathology and compared with standard blood and serum panels for toxicology. RESULTS: The in vivo whole-body imaging data detected widespread changes, where spatially heterogeneous toxic effects were identified across different tissues, within substructures of organs, as well as among different individuals. The signal changes were consistent with established toxicity profiles of these chemotherapeutic drugs. Apart from generating a map of susceptible tissues, this in vivo imaging approach was more sensitive compared with conventional blood and serum markers used in toxicology. Also, repeated imaging during the acute period after drug treatment captured different kinetics of tissue injury among susceptible organs in males and females. CONCLUSIONS: This novel and highly translational imaging approach shows promise in optimizing therapeutic decisions by detecting and managing drug toxicity on a personalized basis.Toxicity to normal tissues is a significant limitation in chemotherapies. This work demonstrated an in vivo imaging-based approach for characterizing toxicity-induced tissue injury in a systemic, dynamic, and near-real time fashion. This novel approach shows promise in optimizing therapeutic decisions by monitoring drug toxicity on a personalized basis.


Subject(s)
Apoptosis/drug effects , Bacteriocins/pharmacology , Drug-Related Side Effects and Adverse Reactions/diagnostic imaging , Organotechnetium Compounds/pharmacology , Whole Body Imaging , Animals , Cell Death/drug effects , Cisplatin/pharmacology , Cyclophosphamide/pharmacology , Drug-Related Side Effects and Adverse Reactions/pathology , Humans , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Rats
8.
Mol Cancer Res ; 17(1): 70-83, 2019 01.
Article in English | MEDLINE | ID: mdl-30171177

ABSTRACT

Patient-derived pancreatic ductal adenocarcinoma (PDAC) organoid systems show great promise for understanding the biological underpinnings of disease and advancing therapeutic precision medicine. Despite the increased use of organoids, the fidelity of molecular features, genetic heterogeneity, and drug response to the tumor of origin remain important unanswered questions limiting their utility. To address this gap in knowledge, primary tumor- and patient-derived xenograft (PDX)-derived organoids, and 2D cultures for in-depth genomic and histopathologic comparisons with the primary tumor were created. Histopathologic features and PDAC representative protein markers (e.g., claudin 4 and CA19-9) showed strong concordance. DNA- and RNA-sequencing (RNAseq) of single organoids revealed patient-specific genomic and transcriptomic consistency. Single-cell RNAseq demonstrated that organoids are primarily a clonal population. In drug response assays, organoids displayed patient-specific sensitivities. In addition, the in vivo PDX response to FOLFIRINOX and gemcitabine/abraxane treatments were examined, which was recapitulated in vitro with organoids. This study has demonstrated that organoids are potentially invaluable for precision medicine as well as preclinical drug treatment studies because they maintain distinct patient phenotypes and respond differently to drug combinations and dosage. IMPLICATIONS: The patient-specific molecular and histopathologic fidelity of organoids indicate that they can be used to understand the etiology of the patient's tumor and the differential response to therapies and suggests utility for predicting drug responses.


Subject(s)
Adenocarcinoma/genetics , Organoids/metabolism , Pancreatic Neoplasms/genetics , Animals , Humans , Mice
9.
Oncol Lett ; 16(5): 6437-6444, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30405781

ABSTRACT

Glycogen Synthase Kinase-3ß (GSK-3ß), a serine/threonine protein kinase, has been implicated as a potential therapeutic target in human cancer. The objective of the present study was to evaluate aberrant expression of GSK-3ß as a potential biomarker in human breast and head and neck cancers. Nuclear/cytosolic fractionation, immunoblotting and immunohistochemical staining was used to study the expression of GSK-3ß in human breast and head and neck cancer. Aberrant nuclear accumulation of GSK-3ß in five human breast cancer cell lines was demonstrated and in 89/128 (70%) human breast carcinomas, whereas no detectable expression of GSK-3ß was found in benign breast tissue. Nuclear GSK-3ß expression was associated with HER-2 positive tumors (P=0.02) and non-triple negative breast carcinomas (P=0.0001), although nuclear GSK-3ß was observed in some samples across all breast cancer subtypes. Aberrant nuclear expression of GSK-3ß was found in 11/15 (73%) squamous cell head and neck carcinomas, whereas weak or no detectable expression of GSK-3ß was found in benign salivary gland and other benign head and neck tissues. These results support the hypothesis that aberrant nuclear GSK-3ß may represent a potential target for the clinical treatment of human breast and squamous cell carcinoma.

10.
Anticancer Drugs ; 29(8): 717-724, 2018 09.
Article in English | MEDLINE | ID: mdl-29846250

ABSTRACT

Advanced stage neuroblastoma is a very aggressive pediatric cancer with limited treatment options and a high mortality rate. Glycogen synthase kinase-3ß (GSK-3ß) is a potential therapeutic target in neuroblastoma. Using immunohistochemical staining, we observed positive GSK-3ß expression in 67% of human neuroblastomas (34 of 51 cases). Chemically distinct GSK-3 inhibitors (AR-A014418, TDZD-8, and 9-ING-41) suppressed the growth of neuroblastoma cells, whereas 9-ING-41, a clinically relevant small-molecule GSK-3ß inhibitor with broad-spectrum preclinical antitumor activity, being the most potent. Inhibition of GSK-3 resulted in a decreased expression of the antiapoptotic molecule XIAP and an increase in neuroblastoma cell apoptosis. Mouse xenograft studies showed that the combination of clinically relevant doses of CPT-11 and 9-ING-41 led to greater antitumor effect than was observed with either agent alone. These data support the inclusion of patients with advanced neuroblastoma in clinical studies of 9-ING-41, especially in combination with CPT-11.


Subject(s)
Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Indoles/pharmacology , Maleimides/pharmacology , Neuroblastoma/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Growth Processes/drug effects , Cell Line, Tumor , Drug Synergism , Female , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Indoles/administration & dosage , Irinotecan/administration & dosage , Irinotecan/pharmacology , Maleimides/administration & dosage , Mice , Mice, Nude , Neuroblastoma/enzymology , Neuroblastoma/pathology , Xenograft Model Antitumor Assays
11.
Transl Oncol ; 10(4): 669-678, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28672195

ABSTRACT

Resistance to chemotherapy remains a major challenge in the treatment of human glioblastoma (GBM). Glycogen synthase kinase-3ß (GSK-3ß), a positive regulator of NF-κB-mediated survival and chemoresistance of cancer cells, has been identified as a potential therapeutic target in human GBM. Our objective was to determine the antitumor effect of GSK-3 inhibitor 9-ING-41 in combination with chemotherapy in patient-derived xenograft (PDX) models of human GBM. We utilized chemoresistant PDX models of GBM, GBM6 and GBM12, to study the effect of 9-ING-41 used alone and in combination with chemotherapy on tumor progression and survival. GBM6 and GBM12 were transfected by reporter constructs to enable bioluminescence imaging, which was used to stage animals prior to treatment and to follow intracranial GBM tumor growth. Immunohistochemical staining, apoptosis assay, and immunoblotting were used to assess the expression of GSK-3ß and the effects of treatment in these models. We found that 9-ING-41 significantly enhanced 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (CCNU) antitumor activity in staged orthotopic GBM12 (no response to CCNU) and GBM6 (partial response to CCNU) PDX models, as indicated by a decrease in tumor bioluminescence in mouse brain and a significant increase in overall survival. Treatment with the combination of CCNU and 9-ING-41 resulted in histologically confirmed cures in these studies. Our results demonstrate that the GSK-3 inhibitor 9-ING-41, a clinical candidate currently in Investigational New Drug (IND)-enabling development, significantly enhances the efficacy of CCNU therapy for human GBM and warrants consideration for clinical evaluation in this difficult-to-treat patient population.

12.
Clin Cancer Res ; 23(8): 1891-1897, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28053024

ABSTRACT

Glycogen synthase kinase-3ß (GSK-3ß), a serine/threonine protein kinase, is a complex regulator of numerous cellular functions. GSK-3ß is a unique kinase which is constitutively active in resting and nonstimulated cells. GSK-3ß has been implicated in a wide range of diseases including neurodegeneration, inflammation and fibrosis, noninsulin-dependent diabetes mellitus, and cancer. It is a regulator of NF-κB-mediated survival of cancer cells, which provided a rationale for the development of GSK-3 inhibitors targeting malignant tumors. Recent studies, many of them reported over the past decade, have identified GSK-3ß as a potential therapeutic target in more than 15 different types of cancer. Whereas only active GSK-3ß is expressed in cancer cell nucleus, aberrant nuclear accumulation of GSK-3ß has been identified as a hallmark of cancer cells in malignant tumors of different origin. This review focuses on the preclinical and clinical development of GSK-3 inhibitors and the potential therapeutic impact of targeting GSK-3ß in human cancer. Clin Cancer Res; 23(8); 1891-7. ©2017 AACR.


Subject(s)
Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Molecular Targeted Therapy/methods , Neoplasms/enzymology , Animals , Antineoplastic Agents/pharmacology , Humans , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology
13.
Oncotarget ; 8(70): 114924-114934, 2017 Dec 29.
Article in English | MEDLINE | ID: mdl-29383130

ABSTRACT

The complexities of GSK-3ß function and interactions with PI3K/AKT/mTOR signaling, cell cycling, and apoptotic pathways are poorly understood in the context of lymphomagenesis and cancer therapeutics. In this study, we explored the anti-tumor effects of the GSK-3ß inhibitor, 9-ING-41, in lymphoma cell lines as a single agent and in combination with novel agents comprising BCL-2 inhibitor (Venetoclax), CDK-9 inhibitor (BAY-1143572) and p110δ-PI3K inhibitor (Idelalisib). Treatment of Daudi, SUDHL-4, Karpas 422, KPUM-UH1, and TMD8 lymphoma cell lines with 1 µM 9-ING-41 reduced cell viability by 40-70% (p<0.05) and halted proliferation. Luminex analysis of apoptotic pathways revealed a significant increase in active caspase 3 in all lymphoma cell lines (p<0.001) except TMD8 cells. Co-treating SUDHL-4 and KPUM-UH1 lymphoma cells with 0.5 µM 9-ING-41 showed 8-and 2-fold reduction in IC50 values of Venetoclax, respectively. No significant benefit for this combination was seen in other lymphoma cells tested. The combination of BAY-1143572 with 0.5 µM 9-ING-41 showed an 8-fold reduction in the IC50 value of the former in SUDHL-4 lymphoma cells alone. No significant changes in IC50 values of Idelalisib were measured across all cell lines for the combination of 9-ING-41 and Idelalisib. Further, signaling analysis via Western blot in the double-hit lymphoma cell line, KPUM-UH1, suggests that phospho-c-MYC is modified with 9-ING-41 treatment. Altogether, our data show that 9-ING-41 results in increased apoptosis and decreased proliferation in aggressive B-cell lymphoma cells and enhances the antitumor effects of BCL-2 and CDK-9 antagonists.

14.
Nat Biomed Eng ; 1(11): 902-913, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29450107

ABSTRACT

Many human diseases result from the dysregulation of the complex interactions between tens to thousands of genes. However, approaches for the transcriptional modulation of many genes simultaneously in a predictive manner are lacking. Here, through the combination of simulations, systems modelling and in vitro experiments, we provide a physical regulatory framework based on chromatin packing-density heterogeneity for modulating the genomic information space. Because transcriptional interactions are essentially chemical reactions, they depend largely on the local physical nanoenvironment. We show that the regulation of the chromatin nanoenvironment allows for the predictable modulation of global patterns in gene expression. In particular, we show that the rational modulation of chromatin density fluctuations can lead to a decrease in global transcriptional activity and intercellular transcriptional heterogeneity in cancer cells during chemotherapeutic responses to achieve near-complete cancer cell killing in vitro. Our findings represent a 'macrogenomic engineering' approach to modulating the physical structure of chromatin for whole-scale transcriptional modulation.

15.
Cancer Lett ; 380(2): 384-392, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27424289

ABSTRACT

Glycogen Synthase Kinase-3ß (GSK-3ß), a serine/threonine protein kinase, is an emerging therapeutic target in the treatment of human breast cancer. In this study, we demonstrate that the pharmacological inhibition of GSK-3 by two novel small molecule GSK-3 inhibitors, 9-ING-41 and 9-ING-87, reduced the viability of breast cancer cells but had little effect on non-tumorigenic cell growth. Moreover, treatment with 9-ING-41 enhanced the antitumor effect of irinotecan (CPT-11) against breast cancer cells in vitro. We next established two patient-derived xenograft tumor models (BC-1 and BC-2) from metastatic pleural effusions obtained from patients with progressive, chemorefractory breast cancer and demonstrated that 9-ING-41 also potentiated the effect of the chemotherapeutic drug CPT-11 in vivo, leading to regression of established BC-1 and BC-2 tumors in mice. Our results suggest that the inhibition of GSK-3 is a promising therapeutic approach to overcome chemoresistance in human breast cancer, and identify the GSK-3 inhibitor 9-ING-41 as a candidate targeted agent for metastatic breast cancer therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Camptothecin/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Indoles/pharmacology , Maleimides/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Camptothecin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Design , Female , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Irinotecan , Mice, Inbred NOD , Mice, SCID , Molecular Targeted Therapy , Signal Transduction/drug effects , Time Factors , Tumor Burden/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Cancer Res ; 76(8): 2265-76, 2016 04 15.
Article in English | MEDLINE | ID: mdl-26921338

ABSTRACT

Loss of pigment epithelium-derived factor (PEDF, SERPINF1) in cancer cells is associated with poor prognosis and metastasis, but the contribution of stromal PEDF to cancer evolution is poorly understood. Therefore, we investigated the role of fibroblast-derived PEDF in melanoma progression. We demonstrate that normal dermal fibroblasts expressing high PEDF levels attenuated melanoma growth and angiogenesis in vivo, whereas PEDF-depleted fibroblasts exerted tumor-promoting effects. Accordingly, mice with global PEDF knockout were more susceptible to melanoma metastasis. We also demonstrate that normal fibroblasts in close contact with PEDF-null melanoma cells lost PEDF expression and tumor-suppressive properties. Further mechanistic investigations underlying the crosstalk between tumor and stromal cells revealed that melanoma cells produced PDGF-BB and TGFß, which blocked PEDF production in fibroblasts. Notably, cancer-associated fibroblasts (CAF) isolated from patient-derived tumors expressed markedly low levels of PEDF. Treatment of patient CAF and TGFß-treated normal fibroblasts with exogenous PEDF decreased the expression of CAF markers and restored PEDF expression. Finally, expression profiling of PEDF-depleted fibroblasts revealed induction of IL8, SERPINB2, hyaluronan synthase-2, and other genes associated with tumor promotion and metastasis. Collectively, our results demonstrate that PEDF maintains tumor-suppressive functions in fibroblasts to prevent CAF conversion and illustrate the mechanisms by which melanoma cells silence stromal PEDF to promote malignancy. Cancer Res; 76(8); 2265-76. ©2016 AACR.


Subject(s)
Cell Proliferation , Eye Proteins/biosynthesis , Fibroblasts/metabolism , Melanoma/pathology , Nerve Growth Factors/biosynthesis , Serpins/biosynthesis , Animals , Cell Line, Tumor , Humans , Melanoma/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Transforming Growth Factor beta/metabolism
17.
ChemMedChem ; 11(1): 81-92, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26592932

ABSTRACT

The histone deacetylases (HDACs) occur in 11 different isoforms, and these enzymes regulate the activity of a large number of proteins involved in cancer initiation and progression. The discovery of isoform-selective HDAC inhibitors (HDACIs) is desirable, as it is likely that such compounds would avoid some of the undesirable side effects found with the first-generation inhibitors. A series of HDACIs previously reported by us were found to display some selectivity for HDAC6 and to induce cell-cycle arrest and apoptosis in pancreatic cancer cells. In the present work, we show that structural modification of these isoxazole-based inhibitors leads to high potency and selectivity for HDAC6 over HDAC1-3 and HDAC10, while unexpectedly abolishing their ability to block cell growth. Three inhibitors with lower HDAC6 selectivity inhibit the growth of cell lines BxPC3 and L3.6pl, and they only induce apoptosis in L3.6pl cells. We conclude that HDAC6 inhibition alone is insufficient for disruption of cell growth, and that some degree of class 1 HDAC inhibition is required. Moreover, the highly selective HDAC6Is reported herein that are weakly cytotoxic may find use in cancer immune system reactivation.


Subject(s)
Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Pancreatic Neoplasms/pathology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Histone Deacetylase 6 , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Humans , Molecular Structure , Pancreatic Neoplasms/enzymology , Structure-Activity Relationship
18.
Neoplasia ; 17(9): 735-741, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26476081

ABSTRACT

Patient-derived xenograft (PDX) tumor models have emerged as a new approach to evaluate the effects of cancer drugs on patients' personalized tumor grafts enabling to select the best treatment for the cancer patient and providing a new tool for oncology drug developers. Here, we report that human tumors engrafted in immunodeficient mice are susceptible to formation of B-and T-cell PDX tumors. We xenografted human primary and metastatic tumor samples into immunodeficient mice and found that a fraction of PDX tumors generated from patients' samples of breast, colon, pancreatic, bladder and renal cancer were histologically similar to lymphocytic neoplasms. Moreover, we found that the first passage of breast and pancreatic cancer PDX tumors after initial transplantation of the tumor pieces from the same human tumor graft could grow as a lymphocytic tumor in one mouse and as an adenocarcinoma in another mouse. Whereas subcutaneous PDX tumors resembling human adenocarcinoma histology were slow growing and non-metastatic, we found that subcutaneous PDX lymphocytic tumors were fast growing and formed large metastatic lesions in mouse lymph nodes, liver, lungs, and spleen. PDX lymphocytic tumors were comprised of B-cells which were Epstein-Barr virus positive and expressed CD45 and CD20. Because B-cells are typically present in malignant solid tumors, formation of B-cell tumor may evolve in a wide range of PDX tumor models. Although PDX tumor models show great promise in the development of personalized therapy for cancer patients, our results suggest that confidence in any given PDX tumor model requires careful screening of lymphocytic markers.


Subject(s)
B-Lymphocytes/pathology , Lymphoma/pathology , Xenograft Model Antitumor Assays/methods , Animals , B-Lymphocytes/immunology , Humans , Lymphoma/etiology , Lymphoma/immunology , Mice , Transplantation, Heterologous/methods
19.
Nanomedicine ; 11(3): 671-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25461281

ABSTRACT

This paper reports an in vivo evaluation of toxicology and biodistribution of a highly anisotropic Au nanoconstruct composed of a gold nanostar (AuNS) core and a ligand shell of a G-quadruplex DNA aptamer AS1411 (Apt) supporting both targeting and therapy capabilities. We examined the toxicity of the nanoconstructs (Apt-AuNS) at four different injected concentrations. At the highest dose tested (48 mg/kg), maximal tolerated dose was not reached. Clinical pathology showed no apparent signs of acute toxicity. Interestingly, the nanoconstructs circulated longer in female rats compared to male rats. In two different tumor models, the biodistribution of Apt-AuNS, especially tumor accumulation, was different. Accumulation of Apt-AuNS was 5 times higher in invasive breast cancer tumors compared to fibrosarcoma tumors. These results provide insight on identifying a tumor model and nanoconstruct for in vivo studies, especially when an in vitro therapeutic response is observed in multiple cancer cell lines. From the clinical editor: This study investigated the toxicity and distribution of aptamer loaded gold nanostars in a rodent model of invasive breast cancer and fibrosarcoma. Acute toxicity was not identified even in the highest studied doses. Fivefold accumulation was demonstrated in the breast cancer model compared to the fibrosarcoma model. Studies like this are critically important in further clarifying the potential therapeutic use of these nanoconstructs, especially when ex vivo effects are clearly demonstrated.


Subject(s)
Aptamers, Nucleotide , Fibrosarcoma/drug therapy , Gold , Mammary Neoplasms, Experimental/drug therapy , Metal Nanoparticles , Animals , Aptamers, Nucleotide/adverse effects , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/pharmacokinetics , Aptamers, Nucleotide/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Gold/adverse effects , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , Male , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Metal Nanoparticles/adverse effects , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice , Mice, Nude , Rats , Sex Characteristics
20.
PLoS One ; 9(1): e85349, 2014.
Article in English | MEDLINE | ID: mdl-24465541

ABSTRACT

The urokinase plasminogen activator receptor (uPAR) plays a role in tumor progression and has been proposed as a target for the treatment of cancer. We recently described the development of a novel humanized monoclonal antibody that targets uPAR and has anti-tumor activity in multiple xenograft animal tumor models. This antibody, ATN-658, does not inhibit ligand binding (i.e. uPA and vitronectin) to uPAR and its mechanism of action remains unclear. As a first step in understanding the anti-tumor activity of ATN-658, we set out to identify the epitope on uPAR to which ATN-658 binds. Guided by comparisons between primate and human uPAR, epitope mapping studies were performed using several orthogonal techniques. Systematic site directed and alanine scanning mutagenesis identified the region of aa 268-275 of uPAR as the epitope for ATN-658. No known function has previously been attributed to this epitope Structural insights into epitope recognition were obtained from structural studies of the Fab fragment of ATN-658 bound to uPAR. The structure shows that the ATN-658 binds to the DIII domain of uPAR, close to the C-terminus of the receptor, corroborating the epitope mapping results. Intriguingly, when bound to uPAR, the complementarity determining region (CDR) regions of ATN-658 closely mimic the binding regions of the integrin CD11b (αM), a previously identified uPAR ligand thought to be involved in leukocyte rolling, migration and complement fixation with no known role in tumor progression of solid tumors. These studies reveal a new functional epitope on uPAR involved in tumor progression and demonstrate a previously unrecognized strategy for the therapeutic targeting of uPAR.


Subject(s)
Antibodies, Monoclonal/chemistry , CD11b Antigen/chemistry , Epitopes/chemistry , Receptors, Urokinase Plasminogen Activator/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , CD11b Antigen/immunology , Cell Line, Tumor , Chlorocebus aethiops , Dogs , Drosophila melanogaster , Epitope Mapping , Epitopes/immunology , Humans , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding/drug effects , Receptors, Urokinase Plasminogen Activator/antagonists & inhibitors , Receptors, Urokinase Plasminogen Activator/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Sequence Homology, Amino Acid , Urokinase-Type Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/immunology , Vitronectin/chemistry , Vitronectin/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...