Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Nucl Med ; 65(5): 761-767, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38514083

ABSTRACT

The membrane protein carbonic anhydrase IX (CAIX) is highly expressed in many hypoxic or von Hippel-Lindau tumor suppressor-mutated tumor types. Its restricted expression in healthy tissues makes CAIX an attractive diagnostic and therapeutic target. DPI-4452 is a CAIX-targeting cyclic peptide with a DOTA cage, allowing radionuclide chelation for theranostic purposes. Here, we report CAIX expression in multiple tumor types and provide in vitro and in vivo evaluations of 68Ga-labeled DPI-4452 ([68Ga]Ga-DPI-4452) and 177Lu-labeled DPI-4452 ([177Lu]Lu-DPI-4452). Methods: CAIX expression was assessed by immunohistochemistry with a panel of tumor and healthy tissues. The molecular interactions of complexed and uncomplexed DPI-4452 with CAIX were assessed by surface plasmon resonance and cell-binding assays. In vivo characterization of radiolabeled and nonradiolabeled DPI-4452 was performed in HT-29 colorectal cancer (CRC) and SK-RC-52 clear cell renal cell carcinoma (ccRCC) human xenograft mouse models and in healthy beagle dogs. Results: Overexpression of CAIX was shown in several tumor types, including ccRCC, CRC, and pancreatic ductal adenocarcinoma. DPI-4452 specifically and selectively bound CAIX with subnanomolar affinity. In cell-binding assays, DPI-4452 displayed comparably high affinities for human and canine CAIX but a much lower affinity for murine CAIX, demonstrating that the dog is a relevant species for biodistribution studies. DPI-4452 was rapidly eliminated from the systemic circulation of beagle dogs. The highest uptake of [68Ga]Ga-DPI-4452 and [177Lu]Lu-DPI-4452 was observed in the small intestine and stomach, 2 organs known to express CAIX. Uptake in other organs (e.g., kidneys) was remarkably low. In HT-29 and SK-RC-52 xenograft mouse models, both [68Ga]Ga-DPI-4452 and [177Lu]Lu-DPI-4452 showed tumor-selective uptake; in addition, [177Lu]Lu-DPI-4452 significantly reduced tumor growth. These results demonstrated the theranostic potential of DPI-4452. Conclusion: DPI-4452 selectively targets CAIX. [68Ga]Ga-DPI-4452 and [177Lu]Lu-DPI-4452 localized to tumors and were well tolerated in mice. [177Lu]Lu-DPI-4452 demonstrated strong tumor growth inhibition in 2 xenograft mouse models. Thus, the 2 agents potentially provide a theranostic approach for selecting and treating patients with CAIX-expressing tumors such as ccRCC, CRC, and pancreatic ductal adenocarcinoma.


Subject(s)
Carbonic Anhydrase IX , Gallium Radioisotopes , Lutetium , Radioisotopes , Carbonic Anhydrase IX/metabolism , Humans , Animals , Mice , Radioisotopes/therapeutic use , Cell Line, Tumor , Tissue Distribution , Ligands , Antigens, Neoplasm/metabolism , Theranostic Nanomedicine , Precision Medicine , Female , Dogs
2.
Cell Death Dis ; 12(7): 641, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162831

ABSTRACT

A major unmet clinical need is a therapeutic capable of removing hepatitis B virus (HBV) genome from the liver of infected individuals to reduce their risk of developing liver cancer. A strategy to deliver such a therapy could utilize the ability to target and promote apoptosis of infected hepatocytes. Presently there is no clinically relevant strategy that has been shown to effectively remove persistent episomal covalently closed circular HBV DNA (cccDNA) from the nucleus of hepatocytes. We used linearized single genome length HBV DNA of various genotypes to establish a cccDNA-like reservoir in immunocompetent mice and showed that clinical-stage orally administered drugs that antagonize the function of cellular inhibitor of apoptosis proteins can eliminate HBV replication and episomal HBV genome in the liver. Primary human liver organoid models were used to confirm the clinical relevance of these results. This study underscores a clinically tenable strategy for the potential elimination of chronic HBV reservoirs in patients.


Subject(s)
Antiviral Agents/pharmacology , Azocines/pharmacology , Benzhydryl Compounds/pharmacology , Genome, Viral , Hepatitis B virus/drug effects , Hepatitis B/drug therapy , Hepatocytes/drug effects , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Liver/drug effects , Thiazoles/pharmacology , Animals , Disease Models, Animal , Hep G2 Cells , Hepatitis B/metabolism , Hepatitis B/pathology , Hepatitis B/virology , Hepatitis B virus/genetics , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatocytes/virology , Host-Pathogen Interactions , Humans , Inhibitor of Apoptosis Proteins/metabolism , Liver/metabolism , Liver/pathology , Liver/virology , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Organoids , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Virus Replication/drug effects
3.
PLoS One ; 15(1): e0227715, 2020.
Article in English | MEDLINE | ID: mdl-31978106

ABSTRACT

The immune checkpoint programmed cell death protein 1 (PD-1) plays a major role in T cell exhaustion in cancer and chronic HIV infection. The inhibitor of apoptosis protein antagonist Debio 1143 (D1143) enhances tumor cell death and synergizes with anti-PD-1 agents to promote tumor immunity and displayed HIV latency reversal activity in vitro. We asked in this study whether D1143 would stimulate the potency of an anti-human PD-1 monoclonal antibody (mAb) to reduce HIV loads in humanized mice. Anti-PD-1 mAb treatment decreased PD-1+ CD8+ cell population by 32.3% after interruption of four weeks treatment, and D1143 co-treatment further reduced it from 32.3 to 73%. Anti-PD-1 mAb administration reduced HIV load in blood by 94%, and addition of D1143 further enhanced this reduction from 94 to 97%. D1143 also more profoundly promoted with the anti-PD-1-mediated reduction of HIV loads in all tissues analyzed including spleen (71 to 96.4%), lymph nodes (64.3 to 80%), liver (64.2 to 94.4), lung (64.3 to 80.1%) and thymic organoid (78.2 to 98.2%), achieving a >5 log reduction of HIV loads in CD4+ cells isolated from tissues 2 weeks after drug treatment interruption. Ex vivo anti-CD3/CD28 stimulation increased the ability to activate exhausted CD8+ T cells in infected mice having received in vivo anti-PD-1 treatment by 7.9-fold (5 to 39.6%), and an additional increase by 1.7-fold upon D1143 co-treatment (39.6 to 67.3%). These findings demonstrate for the first time that an inhibitor of apoptosis protein antagonist enhances in a statistically manner the effects of an immune check point inhibitor on antiviral immunity and on HIV load reduction in tissues of humanized mice, suggesting that the combination of two distinct classes of immunomodulatory agents constitutes a promising anti-HIV immunotherapeutic approach.


Subject(s)
Antibodies, Monoclonal/pharmacology , Azocines/pharmacology , Benzhydryl Compounds/pharmacology , HIV Infections/drug therapy , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Antibodies, Monoclonal/therapeutic use , Azocines/therapeutic use , Benzhydryl Compounds/therapeutic use , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Synergism , Drug Therapy, Combination , Female , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , HIV-1/isolation & purification , Humans , Mice , Mice, Transgenic , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Viral Load/drug effects , Viral Load/immunology
4.
PLoS One ; 14(2): e0211746, 2019.
Article in English | MEDLINE | ID: mdl-30716099

ABSTRACT

Antiretroviral therapy (ART) suppresses HIV replication, but does not cure the infection because replication-competent virus persists within latently infected CD4+ T cells throughout years of therapy. These reservoirs contain integrated HIV-1 genomes and can resupply active virus. Thus, the development of strategies to eliminate the reservoir of latently infected cells is a research priority of global significance. In this study, we tested efficacy of a new inhibitor of apoptosis protein antagonist (IAPa) called Debio 1143 at reversing HIV latency and investigated its mechanisms of action. Debio 1143 activates HIV transcription via NF-kB signaling by degrading the ubiquitin ligase baculoviral IAP repeat-containing 2 (BIRC2), a repressor of the non-canonical NF-kB pathway. Debio 1143-induced BIRC2 degradation results in the accumulation of NF-κB-inducing kinase (NIK) and proteolytic cleavage of p100 into p52, leading to nuclear translocation of p52 and RELB. Debio 1143 greatly enhances the binding of RELB to the HIV-1 LTR. These data indicate that Debio 1143 activates the non-canonical NF-kB signaling pathway by promoting the binding of RELB:p52 complexes to the HIV-1 LTR, resulting in the activation of the LTR-dependent HIV-1 transcription. Importantly, Debio 1143 reverses viral latency in HIV-1 latent T cell lines. Using knockdown (siRNA BIRC2), knockout (CRIPSR NIK) and proteasome machinery neutralization (MG132) approaches, we found that Debio 1143-mediated HIV latency reversal is BIRC2 degradation- and NIK stabilization-dependent. Debio 1143 also reverses HIV-1 latency in resting CD4+ T cells derived from ART-treated patients or HIV-1-infected humanized mice under ART. Interestingly, daily oral administration of Debio 1143 in cancer patients at well-tolerated doses elicited BIRC2 target engagement in PBMCs and induced a moderate increase in cytokines and chemokines mechanistically related to NF-kB signaling. In conclusion, we provide strong evidences that the IAPa Debio 1143, by initially activating the non-canonical NF-kB signaling and subsequently reactivating HIV-1 transcription, represents a new attractive viral latency reversal agent (LRA).


Subject(s)
Anti-HIV Agents/pharmacology , Azocines/pharmacology , Benzhydryl Compounds/pharmacology , CD4-Positive T-Lymphocytes/metabolism , HIV-1/physiology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Ubiquitin-Protein Ligases/antagonists & inhibitors , Virus Latency/drug effects , Animals , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , Female , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Mice , NF-kappa B p52 Subunit/genetics , NF-kappa B p52 Subunit/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factor RelB/genetics , Transcription Factor RelB/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
5.
Clin Cancer Res ; 25(3): 1113-1124, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30352911

ABSTRACT

PURPOSE: Adaptive antitumor immunity following ablative radiotherapy (ART) is attenuated by host myeloid-derived suppressor cell (MDSC), tumor-associated macrophage (TAM), and regulatory T-cell (Treg) infiltrates. We hypothesized treatment with ART and a secondary mitochondrial-derived activators of caspase (SMAC) mimetic could reverse the immunosuppressive lung cancer microenvironment to favor adaptive immunity. EXPERIMENTAL DESIGN: To evaluate for synergy between ART and the SMAC mimetic Debio 1143 and the dependence upon CD8+ T cells and TNFα, we used LLC-OVA syngeneic mouse model of lung cancer and treated them with Debio 1143 and/or ART (30 Gy) with or without anti-CD8, anti-TNFα, or anti-IFNγ antibodies. Tumor-infiltrating OVA-specific CD8+ T cells, Tc1 effector cells, MDSCs, TAMs, and Tregs, were quantified by flow cytometry. Tc1-promoting cytokines TNFα, IFNγ, and IL1ß and the immunosuppressive IL10 and Arg-1 within LLC-OVA tumor tissue or mouse serum were measured by RT-PCR and ELISA. RESULTS: ART delayed tumor growth, and the addition of Debio 1143 greatly enhanced its efficacy, which included several complete responses. These complete responders rejected an LLC-OVA tumor rechallenge. ART and Debio 1143 synergistically induced a tumor-specific, Tc1 cellular and cytokine response while eliminating immunosuppressive cells and cytokines from the tumor microenvironment. Depletion of CD8+ cells, TNFα, and IFNγ with blocking antibody abrogated synergy between ART and Debio 1143 and partially restored tumor-infiltrating MDSCs. CONCLUSIONS: Debio 1143 augments the tumor-specific adaptive immunity induced by ART, while reversing host immunosuppressive cell infiltrates in the tumor microenvironment in a TNFα, IFNγ, and CD8+ T-cell-dependent manner. This provides a novel strategy to enhance the immunogenicity of ART.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Azocines/therapeutic use , Benzhydryl Compounds/therapeutic use , Biomimetic Materials/therapeutic use , Carcinoma, Lewis Lung/therapy , Lung Neoplasms/therapy , Mitochondrial Proteins/metabolism , Radiotherapy/methods , Animals , Apoptosis/drug effects , Apoptosis/immunology , Apoptosis/radiation effects , Azocines/immunology , Benzhydryl Compounds/immunology , Biomimetic Materials/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/radiation effects , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Combined Modality Therapy , Cytokines/blood , Cytokines/genetics , Cytokines/immunology , Female , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/radiation effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects
6.
Sci Rep ; 8(1): 17862, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30552344

ABSTRACT

The poor prognosis of ovarian cancer (it is the leading cause of death from gynecological cancers) is mainly due to the acquisition of resistance to carboplatin. Among the possible resistance pathways, resistance to apoptosis and especially the overexpression of inhibitor of apoptosis proteins (IAP) cIAP1 and X-linked IAP (XIAP), have been implicated. DEBIO 1143, a SMAC (second mitochondria-derived activator of caspase) mimetic, belongs to a new class of targeted agents currently being evaluated in clinical trials, which activate apoptotic cell death and block pro-survival signaling in cancer cells. Here, we demonstrate that DEBIO 1143 in vitro inhibits the cell viability of two carboplatin-sensitive cell lines (IGROV-1 and A2780S) as well as three carboplatin-resistant cell lines (A2780R, SKOV-3 and EFO-21). Of note, DEBIO 1143 is able to reverse resistance to carboplatin by inducing cell death either by apoptosis or necroptosis depending on the cell lines. To identify a biomarker able to predict the sensitivity of the cell lines to DEBIO 1143 treatment we analyzed the expression of the DEBIO 1143 targets cIAP1 and XIAP, and one of their downstream targets, caspase 9. These proteins did not constitute a marker of DEBIO 1143 sensitivity/resistance. Importantly, we confirmed these findings in vivo in SKOV-3 xenograft models where DEBIO 1143 highly potentiated carboplatin treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Azocines/pharmacology , Benzhydryl Compounds/pharmacology , Carboplatin/pharmacology , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Azocines/administration & dosage , Benzhydryl Compounds/administration & dosage , Carboplatin/administration & dosage , Caspase 9/analysis , Cell Line, Tumor , Disease Models, Animal , Female , Heterografts , Humans , Inhibitor of Apoptosis Proteins/analysis , Mice , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/drug therapy , Treatment Outcome , X-Linked Inhibitor of Apoptosis Protein/analysis
7.
Oncotarget ; 6(35): 37410-25, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26485762

ABSTRACT

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


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azepines/pharmacology , Azocines/pharmacology , Benzhydryl Compounds/pharmacology , Camptothecin/analogs & derivatives , Cell Proliferation/drug effects , Lung Neoplasms/drug therapy , Paclitaxel/pharmacology , Taxoids/pharmacology , Topoisomerase Inhibitors/pharmacology , Triazoles/pharmacology , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Camptothecin/pharmacology , Cell Line, Tumor , Docetaxel , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Irinotecan , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/metabolism , Signal Transduction/drug effects , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
8.
Radiother Oncol ; 116(3): 495-503, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26096848

ABSTRACT

BACKGROUND AND PURPOSE: Second mitochondria-derived activator of caspase (SMAC)-mimetics are a new class of targeted drugs that specifically induce apoptotic cancer cell death and block pro-survival signaling by antagonizing selected members of the inhibitor of apoptosis protein (IAP) family. MATERIAL AND METHODS: The present study was designed to investigate the radiosensitizing effect and optimal sequence of administration of the novel SMAC-mimetic Debio 1143 in vitro and in vivo. Apoptosis, alteration of DNA damage repair (DDR), and tumor necrosis factor-alpha (TNF-α) signaling were examined. RESULTS: In vitro, Debio 1143 displayed anti-proliferative activity and enhanced intrinsic radiation sensitivity in 5/6 head and neck squamous cell carcinoma (HNSCC) cell lines in a synergistic manner. In vivo, Debio 1143 dose-dependently radio-sensitized FaDu and SQ20B xenografts, resulting in complete tumor regression in 8/10 FaDu-xenografted mice at the high dose level. At the molecular level, Debio 1143 combined with radiotherapy (RT) induced enhancement of caspase-3 activity, increase in Annexin V-positive cells and karyopyknosis, and increase in TNF-α mRNA levels. Finally, in a neutralization experiment using a TNF-α-blocking antibody and a caspase inhibitor, it was shown that the radiosensitizing effect of Debio 1143 is mediated by caspases and TNF-α. CONCLUSIONS: These results demonstrate that the novel SMAC-mimetic Debio 1143 is a radiosensitizing agent that is worthy of further investigation in clinical trials in combination with radiotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Azocines/pharmacology , Benzhydryl Compounds/pharmacology , Carcinoma, Squamous Cell/therapy , Head and Neck Neoplasms/therapy , Radiation-Sensitizing Agents/pharmacology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Caspase 3/metabolism , Cell Death/drug effects , Cell Line, Tumor , Chemoradiotherapy/methods , Female , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/pharmacology , Mice, Inbred Strains , Mitochondrial Proteins/pharmacology , Neoplasm Transplantation , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck , Transplantation, Heterologous , Xenograft Model Antitumor Assays/methods
9.
EBioMedicine ; 2(12): 1932-43, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26844272

ABSTRACT

BCL9/9L proteins enhance the transcriptional output of the ß-catenin/TCF transcriptional complex and contribute critically to upholding the high WNT signaling level required for stemness maintenance in the intestinal epithelium. Here we show that a BCL9/9L-dependent gene signature derived from independent mouse colorectal cancer (CRC) models unprecedentedly separates patient subgroups with regard to progression free and overall survival. We found that this effect was by and large attributable to stemness related gene sets. Remarkably, this signature proved associated with recently described poor prognosis CRC subtypes exhibiting high stemness and/or epithelial-to-mesenchymal transition (EMT) traits. Consistent with the notion that high WNT signaling is required for stemness maintenance, ablating Bcl9/9l-ß-catenin in murine oncogenic intestinal organoids provoked their differentiation and completely abrogated their tumorigenicity, while not affecting their proliferation. Therapeutic strategies aimed at targeting WNT responses may be limited by intestinal toxicity. Our findings suggest that attenuating WNT signaling to an extent that affects stemness maintenance without disturbing intestinal renewal might be well tolerated and prove sufficient to reduce CRC recurrence and dramatically improve disease outcome.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Neoplasm Proteins/metabolism , Signal Transduction , beta Catenin/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cluster Analysis , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Datasets as Topic , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Mice , Mice, Knockout , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , Phenotype , Prognosis , Sequence Deletion , Transcription Factors , Transcriptome , beta Catenin/antagonists & inhibitors , beta Catenin/genetics
10.
Cancer Res ; 70(16): 6619-28, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20682801

ABSTRACT

Canonical Wnt signaling plays a critical role in stem cell maintenance in epithelial homeostasis and carcinogenesis. Here, we show that in the mouse this role is critically mediated by Bcl9/Bcl9l, the mammalian homologues of Legless, which in Drosophila is required for Armadillo/beta-catenin signaling. Conditional ablation of Bcl9/Bcl9l in the intestinal epithelium, where the essential role of Wnt signaling in epithelial homeostasis and stem cell maintenance is well documented, resulted in decreased expression of intestinal stem cell markers and impaired regeneration of ulcerated colon epithelium. Adenocarcinomas with aberrant Wnt signaling arose with similar incidence in wild-type and mutant mice. However, transcriptional profiles were vastly different: Whereas wild-type tumors displayed characteristics of epithelial-mesenchymal transition (EMT) and stem cell-like properties, these properties were largely abrogated in mutant tumors. These findings reveal an essential role for Bcl9/Bcl9l in regulating a subset of Wnt target genes involved in controlling EMT and stem cell-related features and suggest that targeting the Bcl9/Bcl9l arm of Wnt signaling in Wnt-activated cancers might attenuate these traits, which are associated with tumor invasion, metastasis, and resistance to therapy.


Subject(s)
Adenocarcinoma/pathology , Colonic Neoplasms/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Neoplastic Stem Cells/pathology , Wnt Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Transgenic , Neoplastic Stem Cells/physiology , Transcription Factors , Wnt Proteins/biosynthesis , Wnt Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...