Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Med Chem ; 66(6): 3852-3865, 2023 03 23.
Article in English | MEDLINE | ID: mdl-36877935

ABSTRACT

Compounds that inhibit glutathione peroxidase 4 (GPX4) hold promise as cancer therapeutics in their ability to induce a form of nonapoptotic cell death called ferroptosis. Our research identified 24, a structural analog of the potent GPX4 inhibitor RSL3, that has much better plasma stability (t1/2 > 5 h in mouse plasma). The bioavailability of 24 provided efficacious plasma drug concentrations with IP dosing, thus enabling in vivo studies to assess tolerability and efficacy. An efficacy study in mouse using a GPX4-sensitive tumor model found that doses of 24 up to 50 mg/kg were tolerated for 20 days but had no effect on tumor growth, although partial target engagement was observed in tumor homogenate.


Subject(s)
Ferroptosis , Neoplasms , Mice , Animals , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Biological Availability
2.
Mol Imaging Biol ; 24(5): 830-841, 2022 10.
Article in English | MEDLINE | ID: mdl-35482146

ABSTRACT

PURPOSE: Bruton's tyrosine kinase (BTK) is a key component of B cell receptor (BCR) signaling, and as such a critical regulator of cell proliferation and survival. Aberrant BCR signaling is important in the pathogenesis of various B cell malignancies and autoimmune disorders. Here, we describe the development of a novel positron emission tomography (PET) tracer for imaging BTK expression and/or occupancy by small molecule therapeutics. METHODS: Radiochemistry was carried out by reacting the precursor with [18F]fluoride on a GE FX-FN TracerLab synthesis module to produce [18F]BTK-1 with a 6% decay-corrected radiochemical yield, 100 ± 6 GBq/µmol molar activity, and a radiochemical purity of 99%. Following intravenous administration of [18F]BTK-1 (3.63 ± 0.59 MBq, 0.084 ± 0.05 µg), 60-min dynamic images were acquired in two xenograft models: REC-1, an efficacious mantle cell lymphoma model, and U87MG, a non-efficacious glioblastoma model. Subsequent studies included vehicle, pretreatment (10 min prior to tracer injection), and displacement (30 min post-tracer injection) studies with different reversible BTK inhibitors to examine BTK binding. Human radiation dosimetry was estimated based on PET imaging in healthy rats. RESULTS: Uptake of [18F]BTK-1 was significantly higher in BTK expressing REC-1 tumors than non-BTK expressing U87MG tumors. Administration of BTK inhibitors prior to tracer administration blocked [18F]BTK-1 binding in the REC-1 tumor model consistent with [18F]BTK-1 binding to BTK. The predicted effective dose in humans was 0.0199 ± 0.0007 mSv/MBq. CONCLUSION: [18F]BTK-1 is a promising PET tracer for imaging of BTK, which could provide valuable information for patient selection, drug dose determination, and improving our understanding of BTK biology in humans.


Subject(s)
Fluorides , Protein Kinase Inhibitors , Humans , Animals , Rats , Adult , Agammaglobulinaemia Tyrosine Kinase/chemistry , Agammaglobulinaemia Tyrosine Kinase/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptors, Antigen, B-Cell , Positron-Emission Tomography
3.
J Am Assoc Lab Anim Sci ; 58(5): 583-588, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31412976

ABSTRACT

In the development of cancer therapeutics, no suitable replacements for the use of animals that are capable of modeling such complex disease processes are currently available. In orthotopic models, surgery is often required to access the target organ for tumor cell inoculation. Historically analgesics have been withheld in such models in light of potential effects on tumor development. The current study evaluated the effect of the opioid buprenorphine on tumor growth of a human ovarian cancer cell line (OVCAR5 OT luc2 mCherry). Female CB17 SCID mice (n = 150) underwent surgery for orthotopic inoculation and were assigned to 1 of 3 treatment groups: vehicle control, 1 dose of buprenorphine, or 2 doses of buprenorphine administered perioperatively. Bioluminescence imaging revealed no significant difference on tumor engraftment rate or growth between control and analgesia-treated groups. These data demonstrate that acute, perioperative analgesia with buprenorphine did not alter tumor growth. Although further research is needed to evaluate potential effects of buprenorphine in other cell lines and mouse strains, the justification for withholding analgesia and the potential influence of pain and stress due to insufficient analgesia in these models should be considered thoroughly.


Subject(s)
Analgesics, Opioid , Buprenorphine , Ovarian Neoplasms , Pain , Animals , Female , Humans , Mice , Analgesia/methods , Analgesics, Opioid/administration & dosage , Buprenorphine/administration & dosage , Laboratory Animal Science , Mice, SCID , Ovarian Neoplasms/complications , Ovarian Neoplasms/veterinary , Pain/drug therapy , Pain/etiology , Pain Management , Pain, Postoperative/drug therapy
4.
Cancer Res ; 78(14): 4059-4072, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29764866

ABSTRACT

Progress in understanding tumor stromal biology has been constrained in part because cancer-associated fibroblasts (CAF) are a heterogeneous population with limited cell-type-specific protein markers. Using RNA expression profiling, we identified the membrane protein leucine-rich repeat containing 15 (LRRC15) as highly expressed in multiple solid tumor indications with limited normal tissue expression. LRRC15 was expressed on stromal fibroblasts in many solid tumors (e.g., breast, head and neck, lung, pancreatic) as well as directly on a subset of cancer cells of mesenchymal origin (e.g., sarcoma, melanoma, glioblastoma). LRRC15 expression was induced by TGFß on activated fibroblasts (αSMA+) and on mesenchymal stem cells. These collective findings suggested LRRC15 as a novel CAF and mesenchymal marker with utility as a therapeutic target for the treatment of cancers with LRRC15-positive stromal desmoplasia or cancers of mesenchymal origin. ABBV-085 is a monomethyl auristatin E (MMAE)-containing antibody-drug conjugate (ADC) directed against LRRC15, and it demonstrated robust preclinical efficacy against LRRC15 stromal-positive/cancer-negative, and LRRC15 cancer-positive models as a monotherapy, or in combination with standard-of-care therapies. ABBV-085's unique mechanism of action relied upon the cell-permeable properties of MMAE to preferentially kill cancer cells over LRRC15-positive CAF while also increasing immune infiltrate (e.g., F4/80+ macrophages) in the tumor microenvironment. In summary, these findings validate LRRC15 as a novel therapeutic target in multiple solid tumor indications and support the ongoing clinical development of the LRRC15-targeted ADC ABBV-085.Significance: These findings identify LRRC15 as a new marker of cancer-associated fibroblasts and cancers of mesenchymal origin and provide preclinical evidence for the efficacy of an antibody-drug conjugate targeting the tumor stroma. Cancer Res; 78(14); 4059-72. ©2018 AACR.


Subject(s)
Antibodies, Monoclonal/pharmacology , Immunoconjugates/pharmacology , Membrane Proteins/metabolism , Neoplasms/drug therapy , Stromal Cells/drug effects , Animals , Cell Line , Cell Line, Tumor , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , HCT116 Cells , Humans , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, SCID , Neoplasms/metabolism , Oligopeptides/pharmacology , Rats , Rats, Sprague-Dawley , Sarcoma/drug therapy , Sarcoma/metabolism , Stromal Cells/metabolism , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays/methods
5.
Mol Cancer Ther ; 17(5): 1039-1050, 2018 05.
Article in English | MEDLINE | ID: mdl-29592882

ABSTRACT

Antiangiogenic therapy is a clinically validated modality in cancer treatment. To date, all approved antiangiogenic drugs primarily inhibit the VEGF pathway. Delta-like ligand 4 (DLL4) has been identified as a potential drug target in VEGF-independent angiogenesis and tumor-initiating cell (TIC) survival. A dual-specific biologic targeting both VEGF and DLL4 could be an attractive strategy to improve the effectiveness of anti-VEGF therapy. ABT-165 was uniquely engineered using a proprietary dual-variable domain immunoglobulin (DVD-Ig) technology based on its ability to bind and inhibit both DLL4 and VEGF. In vivo, ABT-165 induced significant tumor growth inhibition compared with either parental antibody treatment alone, due, in part, to the disruption of functional tumor vasculature. In combination with chemotherapy agents, ABT-165 also induced greater antitumor response and outperformed anti-VEGF treatment. ABT-165 displayed nonlinear pharmacokinetic profiles in cynomolgus monkeys, with an apparent terminal half-life > 5 days at a target saturation dose. In a GLP monkey toxicity study, ABT-165 was well-tolerated at doses up to 200 mg/kg with non-adverse treatment-related histopathology findings limited to the liver and thymus. In summary, ABT-165 represents a novel antiangiogenic strategy that potently inhibits both DLL4 and VEGF, demonstrating favorable in vivo efficacy, pharmacokinetic, and safety profiles in preclinical models. Given these preclinical attributes, ABT-165 has progressed to a phase I study. Mol Cancer Ther; 17(5); 1039-50. ©2018 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Glioblastoma/drug therapy , Immunoglobulins/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Xenograft Model Antitumor Assays , Animals , Antineoplastic Combined Chemotherapy Protocols/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Cell Line, Tumor , Drug Screening Assays, Antitumor/methods , Glioblastoma/metabolism , Glioblastoma/pathology , HT29 Cells , Humans , Immunoglobulins/metabolism , Immunologic Factors/metabolism , Immunologic Factors/pharmacokinetics , Immunologic Factors/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Macaca fascicularis/metabolism , Membrane Proteins/metabolism , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism
6.
Neoplasia ; 19(9): 695-706, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28787674

ABSTRACT

Improving the congruity of preclinical models with cancer as it is manifested in humans is a potential way to mitigate the high attrition rate of new cancer therapies in the clinic. In this regard, three-dimensional (3D) tumor cultures in vitro have recently regained interest as they have been acclaimed to have higher similarity to tumors in vivo than to cells grown in monolayers (2D). To identify cancer functions that are active in 3D rather than in 2D cultures, we compared the transcriptional profiles (TPs) of two non-small cell lung carcinoma cell lines, NCI-H1650 and EBC-1 grown in both conditions to the TP of xenografted tumors. Because confluence, diameter or volume can hypothetically alter TPs, we made intra- and inter-culture comparisons using samples with defined dimensions. As projected by Ingenuity Pathway Analysis (IPA), a limited number of signal transduction pathways operational in vivo were better represented by 3D than by 2D cultures in vitro. Growth of 2D and 3D cultures as well as xenografts induced major changes in the TPs of these 3 modes of culturing. Alterations of transcriptional network activation that were predicted to evolve similarly during progression of 3D cultures and xenografts involved the following functions: hypoxia, proliferation, cell cycle progression, angiogenesis, cell adhesion, and interleukin activation. Direct comparison of TPs of 3D cultures and xenografts to monolayer cultures yielded up-regulation of networks involved in hypoxia, TGF and Wnt signaling as well as regulation of epithelial mesenchymal transition. Differences in TP of 2D and 3D cancer cell cultures are subject to progression of the cultures. The emulation of the predicted cell functions in vivo is therefore not only determined by the type of culture in vitro but also by the confluence or diameter of the 2D or 3D cultures, respectively. Consequently, the successful implementation of 3D models will require phenotypic characterization to verify the relevance of applying these models for drug development.


Subject(s)
Gene Expression Regulation, Neoplastic , Transcriptome , Animals , Cell Culture Techniques , Cell Line, Tumor , Cluster Analysis , Disease Models, Animal , Female , Gene Expression Profiling , Heterografts , Humans , Mice , Spheroids, Cellular
7.
Pharmacol Res Perspect ; 3(5): e00178, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26516589

ABSTRACT

The Bcl-2 family inhibitors venetoclax and navitoclax demonstrated potent antitumor activity in chronic lymphocytic leukemia patients, notably in reducing marrow load and adenopathy. Subsequent trials with venetoclax have been initiated in non-Hodgkin's lymphoma and multiple myeloma patients. Traditional preclinical models fall short either in faithfully recapitulating disease progression within such compartments or in allowing the direct longitudinal analysis of systemic disease. We show that intravenous inoculation of engineered RS4;11 (acute lymphoblastic leukemia) and Granta 519 (mantle cell lymphoma) bioluminescent reporter cell lines result in tumor engraftment of bone marrow, with additional invasion of the central nervous system in the case of Granta 519. Importantly, apoptosis induction and response of these systemically engrafted tumors to Bcl-2 family inhibitors alone or in combination with standard-of-care agents could be monitored longitudinally with optical imaging, and was more accurately reflective of the observed clinical response.

8.
Mol Imaging ; 12(1): 28-38, 2013.
Article in English | MEDLINE | ID: mdl-23348789

ABSTRACT

At present, there is considerable interest in the use of in vivo fluorescence and bioluminescence imaging to track the onset and progression of pathologic processes in preclinical models of human disease. Optical quantitation of such phenomena, however, is often problematic, frequently complicated by the overlying tissue's scattering and absorption of light, as well as the presence of endogenous cutaneous and subcutaneous fluorophores. To partially circumvent this information loss, we report here the development of flexible, surgically implanted, transparent windows that enhance quantitative in vivo fluorescence and bioluminescence imaging of optical reporters. These windows are metal and glass free and thus compatible with computed tomography, magnetic resonance imaging, positron emission tomography, and single-photon emission computed tomography; they also permit visualization of much larger areas with fewer impediments to animal locomotion and grooming than those previously described. To evaluate their utility in preclinical imaging, we surgically implanted these windows in the abdominal walls of female athymic nude mice and subsequently inoculated each animal with 1 × 10(4) to 1 × 10(6) bioluminescent human ovarian cancer cells (SKOV3ip.1-luc). Longitudinal imaging studies of fenestrated animals revealed up to 48-fold gains in imaging sensitivity relative to nonfenestrated animals, with relatively few complications, allowing wide-field in vivo visualization of nascent metastatic ovarian cancer colonization.


Subject(s)
Implants, Experimental , Luminescent Measurements/methods , Molecular Imaging/methods , Optical Imaging/methods , Peritoneum/surgery , Abdomen/surgery , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Kaplan-Meier Estimate , Materials Testing , Mice , Mice, Nude , Polyvinyl Chloride/chemistry
9.
Cancer Chemother Pharmacol ; 69(6): 1669-72, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22327787

ABSTRACT

Linifanib (ABT-869) is a multitargeted receptor tyrosine kinase inhibitor. This work aims to evaluate F-fluorodeoxyglucose-positron emission tomography (FDG-PET) as a pharmacodynamic (PD) biomarker for linifanib treatment utilizing the Calu-6 model of human non-small cell lung (NSCLC) cancer in SCID-beige mice. Animals received either vehicle or 12.5 mg/kg linifanib orally twice a day for the duration of the study. Imaging was performed at -1, 1, 3, and 7 days after beginning treatment (n = 12-14 per group). Linifanib inhibited tumor growth and suppressed tumor metabolic activity. Changes in tumor FDG uptake were observed as early as 1 day after beginning linifanib treatment and were sustained for the duration of the study. This study confirms that linifanib is efficacious in this xenograft model of human NSCLC and confirms FDG-PET is a potential PD biomarker strategy for linifanib therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Fluorodeoxyglucose F18 , Indazoles/therapeutic use , Lung Neoplasms/drug therapy , Phenylurea Compounds/therapeutic use , Positron-Emission Tomography , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Cell Line, Tumor , Humans , Lung Neoplasms/diagnostic imaging , Mice , Mice, SCID , Xenograft Model Antitumor Assays
10.
Clin Exp Metastasis ; 27(3): 185-96, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20229256

ABSTRACT

Despite the potentially crucial contributions of the omentum in the regulation of ovarian cancer metastatic growth, it remains a poorly understood organ. Due to its anatomic location and structural fragility, the omentum presents inherent challenges to mechanism-based in vivo studies. Thus, the availability of an ex vivo omental model would, in part, address some of these difficulties posed. Here we describe a technique for identifying, isolating and maintaining ex vivo cultures of omenta from immune-compromised and -competent mice. Ex vivo culture conditions were developed that maintain tissue viability, architecture, and function for up to 10 days. Further experiments demonstrate that the ex vivo culture conditions allow for the proliferation of ovarian cancer cells in vitro and support a similar pattern of microscopic lesions after either intraperitoneal injection of ovarian cancer cells or co-culture of ovarian cancer cells with the omentum. In agreement with previous studies from our laboratory, histologic evaluation of these specimens found that ovarian cancer cells, as well as other peritoneal cancer cells, preferentially accumulate in, and colonize, omental areas rich in immune cells. We now recognize that these are specific, functional structures referred to as milky spots. In sum, these are foundational studies of a readily accessible model, which is easily manipulated and can be immediately used to study the dynamic process of omental colonization. It is hoped that investigators will use the data herein as a starting point for refinements and modifications which will enable them to tailor the model to the specific needs of the experimental question(s) they wish to pursue.


Subject(s)
Omentum/pathology , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/pathology , Peritoneal Neoplasms/secondary , Animals , Cell Proliferation , Coculture Techniques , Female , Humans , Immunohistochemistry , Injections, Intraperitoneal , Mice , Microscopy , Neoplasm Invasiveness/immunology , Neoplasm Invasiveness/pathology , Omentum/immunology , Omentum/metabolism , Omentum/surgery , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/metabolism , Tumor Cells, Cultured
11.
Urol Oncol ; 27(3): 295-7, 2009.
Article in English | MEDLINE | ID: mdl-19414115

ABSTRACT

There has recently been an explosion in the availability of new technologies to noninvasively detect biological processes in preclinical models. One such modality, optical imaging, comprises using bioluminescent and fluorescent reporters and probes to repetitively interrogate molecular events and monitor disease progression in animal models. This review includes an overview of optical imaging technologies (e.g., hardware, reporters, probes) available for small animal imaging and their application in monitoring disease progression, therapeutic efficacy, and molecular processes such as proliferation, apoptosis, and angiogenesis. Also discussed are some of the challenges associated with in vivo optical imaging and the necessary controls and biological correlates one must include in experimental design and interpretation for successful preclinical studies.


Subject(s)
Diagnostic Imaging/instrumentation , Diagnostic Imaging/methods , Animals , Biomarkers/analysis , Image Processing, Computer-Assisted , Luminescent Measurements/instrumentation , Luminescent Measurements/methods , Luminescent Proteins/chemistry , Molecular Probes/chemistry , Optical Devices , Optical Phenomena , Reproducibility of Results
12.
Clin Exp Metastasis ; 26(1): 67-76, 2009.
Article in English | MEDLINE | ID: mdl-18516689

ABSTRACT

The biochemical and biological mechanisms metastatic cancer cells use to function as communities and thwart internal and external growth control mechanisms remain undefined. In this work, we present the hypothesis that cancer cells may use a Quorum-Sensing mechanism to regulate multicellular functions and control steps in metastatic colonization. Quorum sensing is a bacterial cell-cell communication process used to track increasing cell-population density and, in response to changes in cell number, coordinate gene expression and behavior on a community-wide scale. Important parallels between the behavior of societies of bacterial cells and societies of malignant cancer cells exist in the bacterial literature. Of relevance to metastasis is the finding that pathogenic bacteria use quorum sensing to determine when their population numbers are high enough to collectively form biofilms in or on host organisms. Biofilms are complex, heterogeneous communities of bacterial cells encased within an extracellular matrix attached to a solid surface. Biofilms exacerbate disease and are refractory to a battery of therapies. We suggest that the quorum-sensing-controlled bacterial biofilm formation process closely parallels the steps in metastatic colonization. Cells migrate toward/on target surfaces (organ-specific homing), show cell-cell and cell-matrix interactions (tumor cell-stromal cell crosstalk), remain subclinical until they can mount an effective attack (dormancy), form complex structures with channels for nutrient flow (vascularized lesions), and contain resistant cells which can cause disease recurrence (persistors). Using ovarian cancer as an example, we present data supporting the connection between metastatic colonization and quorum sensing and discuss the implications for understanding and controlling metastasis formation.


Subject(s)
Neoplasm Metastasis , Ovarian Neoplasms/pathology , Female , Humans , Quorum Sensing
13.
Cancer Lett ; 272(1): 12-22, 2008 Dec 08.
Article in English | MEDLINE | ID: mdl-18572308

ABSTRACT

Much work has been done in the 20 years since the discovery of the first metastasis suppressor gene to investigate the diverse biochemical functions of the proteins these genes encode. The function of metastasis suppressors cannot be solely predicted from correlative clinical data or in vitro studies. Instead, careful design of in vivo experiments to test broader hypotheses is necessary to pinpoint the mechanism of action of these novel proteins. Our laboratory identified c-Jun NH2-terminal kinase activating kinase 1 (JNKK1)/Mitogen-activated protein kinase (MAPK) kinase 4 (JNKK1/MKK4) as a metastasis suppressor in prostate and ovarian cancer. JNKK1/MKK4 is a stress activated protein kinase (SAPK) involved in a variety of signaling events, ranging from the regulation of hepatoblast survival during mammalian development to metastasis suppression in adult ovarian and prostate cancers. JNKK1/MKK4 function has typically been associated with the c-Jun NH2-terminal kinase (JNK) signaling pathway, particularly in the immune system where JNK plays a role in inflammatory signaling and apoptosis. However, evidence continues to accumulate that JNKK1/MKK4 is also a physiologic activator of p38 under certain conditions, and that activation of p38 arrests cell cycle progression. This review will provide a historical perspective on the role of JNKK1/MKK4 in SAPK signaling, including some recent findings from our own laboratory that shed light on the complicated role for JNKK1/MKK4 in metastatic colonization.


Subject(s)
MAP Kinase Kinase 4/metabolism , Neoplasms/pathology , Cell Cycle , Cell Death , Cell Division , Homeostasis , Humans , Neoplasm Metastasis/prevention & control , Tumor Suppressor Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Cancer Res ; 68(7): 2166-75, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18381422

ABSTRACT

In many patients without clinical metastases, cancer cells have already escaped from the primary tumor and entered a distant organ. A long-standing question in metastasis research is why some disseminated cancer cells fail to complete steps of metastatic colonization for extended periods of time. Our laboratory identified c-Jun NH(2)-terminal kinase activating kinase 1/mitogen-activated protein kinase kinase 4 (JNKK1/MKK4) as a metastasis suppressor protein in a mouse xenograft model of experimental i.p. ovarian cancer metastasis. In this model, expression of JNKK1/MKK4 via activation of p38 delays formation of >or=1-mm implants and prolongs animal survival. Here, we elucidate the time course of this delay as well as the biological mechanisms underpinning it. Using the Gompertz function to model the net accumulation of experimental omental metastases, we show that MKK4-expressing implants arise, on average, 30 days later than controls. Quantitative real-time PCR shows that MKK4 expression does not have a substantial effect on the number of cancer cells initially adhering to the omentum, and terminal deoxyribonucleotidyl transferase-mediated dUTP nick end labeling analysis shows that there is no increase in apoptosis in these cells. Instead, immunohistochemical quantitation of cell cycle proteins reveals that MKK4-expressing cells fail to proliferate once they reach the omentum and up-regulate p21, a cell cycle inhibitor. Consistent with the time course data, in vitro kinase assays and in vivo passaging of cell lines derived from macroscopic metastases show that the eventual outgrowth of MKK4-expressing cells is not due to a discrete selection event. Rather, the population of MKK4-expressing cells eventually uniformly adapts to the consequences of up-regulated MKK4 signaling.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , MAP Kinase Kinase 4/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Animals , Apoptosis/physiology , Cell Adhesion/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , Hemagglutinins/genetics , Humans , MAP Kinase Kinase 4/biosynthesis , MAP Kinase Kinase 4/genetics , Mice , Mice, Nude , Models, Biological , Neoplasm Metastasis , Omentum/pathology , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Transgenes , Up-Regulation
15.
Cancer Metastasis Rev ; 27(1): 67-73, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18049862

ABSTRACT

Cancer metastasis is a complex, dynamic process that begins with dissemination of cells from the primary tumor and culminates in the formation of clinically detectable, overt metastases at one or more discontinuous secondary sites. Evidence from in vivo video microscopy as well as PCR and immunohistochemical studies suggest that cancer cell dissemination is an early event in tumor progression and that cells may persist in a potentially dormant state for a prolonged period. Similarly, the mechanisms by which these disseminated cells initiate growth and complete the process of metastatic colonization remain largely unknown. Understanding signal transduction pathways regulating this final step of metastasis is therefore critical for successful clinical management. While genetic mutations or epigenetic changes may be required for a cell or group of cells to separate and survive distant from the primary tumor, the microenvironment within secondary tissues plays a substantial role in influencing whether disseminated cells survive and proliferate. Our work is focused on using metastasis suppressor proteins to gain insight into why the majority of disseminated cells, which should be fully malignant, do not proliferate immediately at secondary sites. The translational goal of this work is to identify targets for inhibiting metastatic growth and prolonging disease-free survival.


Subject(s)
MAP Kinase Kinase 4/metabolism , Neoplasm Metastasis/pathology , Neoplasms/metabolism , Animals , Humans , Neoplasms/pathology , Signal Transduction
17.
Cancer Res ; 66(4): 2264-70, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16489030

ABSTRACT

Despite considerable efforts to improve early detection of ovarian cancer, the majority of women at time of diagnosis will have metastatic disease. Understanding and targeting the molecular underpinnings of metastasis continues to be the principal challenge in the clinical management of ovarian cancer. Whereas the multistep process of metastasis development has been well established in both clinical and experimental models, the molecular factors and signaling pathways involved in successful colonization of a secondary site by disseminated cancer cells are not well defined. We have previously identified mitogen-activated protein kinase (MAPK) kinase 4/c-Jun NH2-terminal kinase (JNK)-activating kinase (MKK4/JNKK1/SEK1, hereafter referred to as MKK4) as a metastasis suppressor protein in ovarian carcinoma. In this study, we elucidate key mechanisms of MKK4-mediated metastasis suppression. Through the use of a kinase-inactive mutant, we show that MKK4 kinase activity is essential for metastasis suppression and prolongation of animal survival. Because MKK4 can activate either of two MAPKs, p38 or JNK, we expressed MKK6 or MKK7, specific activators of these MAPKs, respectively, to delineate which MAPK signaling module was involved in MKK4-mediated metastasis suppression. We observed that MKK6 expression suppressed metastatic colonization whereas MKK7 had no effect. Our finding that MKK4 and MKK6 both suppress metastasis points to the p38 pathway as an important regulatory pathway for metastatic colonization in ovarian cancer.


Subject(s)
MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase 6/metabolism , MAP Kinase Signaling System/physiology , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Animals , Cell Line, Tumor , Female , Humans , MAP Kinase Kinase 4/biosynthesis , MAP Kinase Kinase 6/biosynthesis , MAP Kinase Kinase 7/biosynthesis , MAP Kinase Kinase 7/metabolism , Mice , Mice, SCID , Neoplasm Metastasis , Neoplasm Transplantation , Transplantation, Heterologous
18.
Cancer Res ; 65(23): 10984-91, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16322247

ABSTRACT

Advances in clinical, translational, and basic studies of metastasis have identified molecular changes associated with specific facets of the metastatic process. Studies of metastasis suppressor gene function are providing a critical mechanistic link between signaling cascades and biological outcomes. We have previously identified c-Jun NH2-terminal kinase (JNK) kinase 1/mitogen-activated protein kinase (MAPK) kinase 4 (JNKK1/MKK4) as a prostate cancer metastasis suppressor gene. The JNKK1/MKK4 protein is a dual-specificity kinase that has been shown to phosphorylate and activate the JNK and p38 MAPKs in response to a variety of extracellular stimuli. In this current study, we show that the kinase activity of JNKK1/MKK4 is required for suppression of overt metastases and is sufficient to prolong animal survival in the AT6.1 model of spontaneous metastasis. Ectopic expression of the JNK-specific kinase MKK7 suppresses the formation of overt metastases, whereas the p38-specific kinase MKK6 has no effect. In vivo studies show that both JNKK1/MKK4 and MKK7 suppress the formation of overt metastases by inhibiting the ability of disseminated cells to colonize the lung (secondary site). Finally, we show that JNKK1/MKK4 and MKK7 from disseminated tumor cells are active in the lung but not in the primary tumor, providing a biochemical explanation for why their expression specifically suppressed metastasis while exerting no effect on the primary tumor. Taken together, these studies contribute to a mechanistic understanding of the context-dependent function of metastasis regulatory proteins.


Subject(s)
Lung Neoplasms/enzymology , Lung Neoplasms/secondary , MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase 7/metabolism , Animals , Enzyme Activation , Lung Neoplasms/genetics , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 6/genetics , MAP Kinase Kinase 6/metabolism , MAP Kinase Kinase 7/genetics , MAP Kinase Signaling System , Male , Mice , Mice, SCID , Rats , Transfection
19.
Cancer Biol Ther ; 4(8): 805-12, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16082183

ABSTRACT

In the past decade, findings from various disciplines of research have stimulated a reevaluation of fundamental concepts of the biology of metastasis. The convergence of two avenues of research has largely been responsible for this shift. First, clinical and experimental studies of specific steps of the metastatic cascade have shown that cancer cells often disseminate early in the natural history of disease and can persist at secondary sites for extended periods of time. These findings suggest that disseminated cells remain subject to growth regulation at distant sites as "dormant" single cells or microscopic metastases consisting of small numbers of cells. Second, complementary functional, biochemical, and signal transduction studies have identified a specific class of proteins that suppress the formation of overt metastases. These proteins are encoded by metastasis suppressor genes, which are operationally defined as genes that suppress in vivo metastasis without inhibiting primary tumor growth when expressed ectopically in metastatic cell lines. While metastasis suppressor proteins may affect many steps in metastatic development, recent evidence specifically implicates several of these proteins in the regulation of growth of disseminated cells at secondary sites. This review describes the evolving understanding of rate-limiting steps of metastatic growth, and the role of metastasis suppressor proteins in the regulation of these processes. We will give an overview of the studies of metastasis suppressor protein function, which have shifted our attention toward mechanisms of growth control at the secondary site (i.e., "metastatic colonization"). Emphasis is placed upon the complimentary research in the fields of metastasis and signal transduction that has identified signaling pathways controlling metastatic colonization. We also discuss the regulation of metastasis suppressor proteins and the potential biological and biochemical mechanisms responsible for their organ-type specificity. Finally, the implication of these emerging concepts on the development of therapeutic strategies will be presented.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasm Metastasis , Protein Biosynthesis , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , Genes, Tumor Suppressor , Humans , Neoplasm Metastasis/genetics , Protein Biosynthesis/genetics , Signal Transduction
20.
Int J Cancer ; 108(1): 15-22, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14618610

ABSTRACT

Our previous studies demonstrate that introduction of a approximately 70 cM region (now estimated at 63.75 Mb by the Human Genome Project) of human chromosome 12 into the highly metastatic Dunning rat prostate cancer cell line AT6.1 results in >30-fold (>/=90%) reduction in the number of overt metastases in spontaneous metastasis assays. We report the further localization and biological characterization of the metastasis-suppressor activity encoded by a reduced region of chromosome 12. To localize this metastasis-suppressor activity, a panel of AT6.1 microcell hybrids that retain varying portions of human chromosome 12 was constructed and subjected to sequence-tagged site (STS)-based PCR analysis and assessment of in vivo metastatic ability. Data from these complementary approaches localized the metastasis-suppressor activity to a approximately 4.2 Mb portion of human chromosome 12q24.3 comprised of 3 separate regions. Reverse transcriptase-polymerase chain reaction (RT-PCR) and immunoblotting were used for differential expression analyses to identify which characterized genes, predicted gene sequences and expressed sequence tags (ESTs) within this region could be responsible for the observed metastasis suppression. Comprehensive in vivo studies showed that suppressed AT6.1-12 hybrids that retain the metastasis-suppressor region on 12q24.3 are capable of arriving at the secondary site, but are not able to persist there. Thus, unlike other metastasis-suppressor genes characterized to date, the metastasis-suppressor gene encoded by this region appears to utilize a different biologic mechanism to suppress the growth of overt metastases at the secondary site.


Subject(s)
Chromosomes, Human, Pair 12 , Neoplasm Metastasis/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Animals , Chromosome Mapping , Genes, Tumor Suppressor , Humans , Hybrid Cells , Lung Neoplasms/secondary , Male , Mice , Mice, SCID , Neoplasm Metastasis/prevention & control , Neoplasm Transplantation , Rats , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...