Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
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.
Eur J Nucl Med Mol Imaging ; 50(9): 2621-2635, 2023 07.
Article in English | MEDLINE | ID: mdl-37086273

ABSTRACT

PURPOSE: FAP is a membrane-bound protease under investigation as a pan-cancer target, given its high levels in tumors but limited expression in normal tissues. FAP-2286 is a radiopharmaceutical in clinical development for solid tumors that consists of two functional elements: a FAP-targeting peptide and a chelator used to attach radioisotopes. Preclinically, we evaluated the immune modulation and anti-tumor efficacy of FAP-2287, a murine surrogate for FAP-2286, conjugated to the radionuclide lutetium-177 (177Lu) as a monotherapy and in combination with a PD-1 targeting antibody. METHODS: C57BL/6 mice bearing MCA205 mouse FAP-expressing tumors (MCA205-mFAP) were treated with 177Lu-FAP-2287, anti-PD-1, or both. Tumor uptake of 177Lu- FAP-2287 was assessed by SPECT/CT scanning, while therapeutic efficacy was measured by tumor volume and survival. Immune profiling of tumor infiltrates was evaluated through flow cytometry, RNA expression, and immunohistochemistry analyses. RESULTS: 177Lu-FAP-2287 rapidly accumulated in MCA205-mFAP tumors leading to significant tumor growth inhibition (TGI) and longer survival time. Significant TGI was also observed from anti-PD-1 and the combination. In flow cytometry analysis of tumors, 177Lu-FAP-2287 increased CD8+ T cell infiltration which was maintained in the combination with anti-PD-1. The increase in CD8+ T cells was accompanied by an induction of STING-mediated type I interferon response and higher levels of co-stimulatory molecules such as CD86. CONCLUSION: In a preclinical model, FAP-targeted radiotherapy enhanced anti-PD-1-mediated TGI by modulating the TME and increasing the recruitment of tumor-infiltrating CD8+ T cells. These findings provide a rationale for clinical studies of combined 177Lu-FAP-2286 radiotherapy and immune checkpoint inhibition in FAP-positive tumors.


Subject(s)
CD8-Positive T-Lymphocytes , Immune Checkpoint Inhibitors , Animals , Mice , Tumor Microenvironment , Cell Line, Tumor , Mice, Inbred C57BL , Fibroblasts
3.
Eur J Nucl Med Mol Imaging ; 49(11): 3651-3667, 2022 09.
Article in English | MEDLINE | ID: mdl-35608703

ABSTRACT

PURPOSE: Fibroblast activation protein (FAP) is a membrane-bound protease that has limited expression in normal adult tissues but is highly expressed in the tumor microenvironment of many solid cancers. FAP-2286 is a FAP-binding peptide coupled to a radionuclide chelator that is currently being investigated in patients as an imaging and therapeutic agent. The potency, selectivity, and efficacy of FAP-2286 were evaluated in preclinical studies. METHODS: FAP expression analysis was performed by immunohistochemistry and autoradiography on primary human cancer specimens. FAP-2286 was assessed in biochemical and cellular assays and in in vivo imaging and efficacy studies, and was further evaluated against FAPI-46, a small molecule-based FAP-targeting agent. RESULTS: Immunohistochemistry confirmed elevated levels of FAP expression in multiple tumor types including pancreatic, breast, and sarcoma, which correlated with FAP binding by FAP-2286 autoradiography. FAP-2286 and its metal complexes demonstrated high affinity to FAP recombinant protein and cell surface FAP expressed on fibroblasts. Biodistribution studies in mice showed rapid and persistent uptake of 68Ga-FAP-2286, 111In-FAP-2286, and 177Lu-FAP-2286 in FAP-positive tumors, with renal clearance and minimal uptake in normal tissues. 177Lu-FAP-2286 exhibited antitumor activity in FAP-expressing HEK293 tumors and sarcoma patient-derived xenografts, with no significant weight loss. In addition, FAP-2286 maintained longer tumor retention and suppression in comparison to FAPI-46. CONCLUSION: In preclinical models, radiolabeled FAP-2286 demonstrated high tumor uptake and retention, as well as potent efficacy in FAP-positive tumors. These results support clinical development of 68Ga-FAP-2286 for imaging and 177Lu-FAP-2286 for therapeutic use in a broad spectrum of FAP-positive tumors.


Subject(s)
Gallium Radioisotopes , Sarcoma , Adult , Animals , Cell Line, Tumor , Fibroblasts , HEK293 Cells , Humans , Mice , Radionuclide Imaging , Tissue Distribution , Tumor Microenvironment
4.
J Nucl Med ; 63(3): 415-423, 2022 03.
Article in English | MEDLINE | ID: mdl-34168013

ABSTRACT

Fibroblast activation protein (FAP) is a promising target for diagnosis and therapy of numerous malignant tumors. FAP-2286 is the conjugate of a FAP-binding peptide, which can be labeled with radionuclides for theranostic applications. We present the first-in-humans results using 177Lu-FAP-2286 for peptide-targeted radionuclide therapy (PTRT). Methods: PTRT using 177Lu-FAP-2286 was performed on 11 patients with advanced adenocarcinomas of the pancreas, breast, rectum, or ovary after prior confirmation of uptake on 68Ga-FAP-2286 or 68Ga-FAPI-04 PET/CT. Results: Administration of 177Lu-FAP-2286 (5.8 ± 2.0 GBq; range, 2.4-9.9 GBq) was well tolerated, with no adverse symptoms or clinically detectable pharmacologic effects being noticed or reported in any of the patients. The whole-body effective dose was 0.07 ± 0.02 Gy/GBq (range, 0.04-0.1 Gy/GBq). The mean absorbed doses for kidneys and red marrow were 1.0 ± 0.6 Gy/GBq (range, 0.4-2.0 Gy/GBq) and 0.05 ± 0.02 Gy/GBq (range, 0.03-0.09 Gy/GBq), respectively. Significant uptake and long tumor retention of 177Lu-FAP-2286 resulted in high absorbed tumor doses, such as 3.0 ± 2.7 Gy/GBq (range, 0.5-10.6 Gy/GBq) in bone metastases. No grade 4 adverse events were observed. Grade 3 events occurred in 3 patients-1 with pancytopenia, 1 with leukocytopenia, and 1 with pain flare-up; 3 patients reported a pain response. Conclusion:177Lu-FAP-2286 PTRT, applied in a broad spectrum of cancers, was relatively well tolerated, with acceptable side effects, and demonstrated long retention of the radiopeptide. Prospective clinical studies are warranted.


Subject(s)
Adenocarcinoma , Positron Emission Tomography Computed Tomography , Feasibility Studies , Female , Gallium Radioisotopes , Humans , Peptides , Prospective Studies , Radioisotopes/therapeutic use , Tissue Distribution
5.
Methods Mol Biol ; 1884: 269-281, 2019.
Article in English | MEDLINE | ID: mdl-30465210

ABSTRACT

Clinically apparent tumors have often established an immunosuppressive tumor microenvironment which renders them "cold," meaning that there are low numbers of immune cells within the tumor. Consequently, novel immunotherapy approaches such as checkpoint inhibitors fail to reactivate the tumor-targeted immune cells. Here we describe the generation of heterotypic tumor-stroma spheroids to study various approaches aiming at the reactivation of cancer immunosurveillance. These spheroids allow to investigate whether a certain immunotherapy or a combination treatment is able to stimulate antitumor immunity in poorly immunological ("cold") tumors, by increasing the number of tumor-infiltrating immune cells ("hot" tumors).


Subject(s)
Cell Culture Techniques/methods , Immunologic Surveillance , Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Aptamers, Nucleotide/pharmacology , Blood Buffy Coat/cytology , Cell Culture Techniques/instrumentation , Chemokine CXCL12/antagonists & inhibitors , Chemokine CXCL12/genetics , Chemokine CXCL12/immunology , Flow Cytometry/instrumentation , Flow Cytometry/methods , HT29 Cells , Healthy Volunteers , Humans , Immunotherapy, Adoptive/methods , Leukocytes, Mononuclear/immunology , Mice , Neoplasms/drug therapy , Neoplasms/pathology , Receptors, Chimeric Antigen/immunology , Spheroids, Cellular/drug effects , Spheroids, Cellular/immunology , Stromal Cells/drug effects , Stromal Cells/immunology , Tumor Microenvironment/drug effects
6.
Cancer Immunol Res ; 5(11): 950-956, 2017 11.
Article in English | MEDLINE | ID: mdl-28963140

ABSTRACT

Immune checkpoint inhibitors promote T cell-mediated killing of cancer cells; however, only a subset of patients benefit from the treatment. A possible reason for this limitation may be that the tumor microenvironment (TME) is immune privileged, which may exclude cytotoxic T cells from the vicinity of cancer cells. The chemokine CXCL12 is key to the TME-driven immune suppression. In this study, we investigated the potential of CXCL12 inhibition by use of the clinical-stage l-RNA-aptamer NOX-A12 (olaptesed pegol) to increase the number of tumor-infiltrating lymphocytes. We used heterotypic tumor-stroma spheroids that mimic a solid tumor with a CXCL12-abundant TME. NOX-A12 enhanced the infiltration of T and NK cells in a dose-dependent manner. NOX-A12 and PD-1 checkpoint inhibition synergistically activated T cells in the spheroids, indicating that the agents complement each other. The findings were validated in vivo in a syngeneic murine model of colorectal cancer in which the addition of NOX-A12 improved anti-PD-1 therapy. Taken together, our work shows that CXCL12 inhibition can break the immune-privileged status of the TME by paving the way for immune effector cells to enter into the tumor, thereby broadening the applicability of checkpoint inhibitors in cancer patients. Cancer Immunol Res; 5(11); 950-6. ©2017 AACR.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Aptamers, Nucleotide/therapeutic use , Chemokine CXCL12/antagonists & inhibitors , Colonic Neoplasms/drug therapy , Lymphocytes, Tumor-Infiltrating/drug effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents, Immunological/pharmacology , Aptamers, Nucleotide/pharmacology , Cell Line, Tumor , Chemokine CXCL12/immunology , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Female , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Mice, Inbred BALB C , Tumor Microenvironment/immunology
7.
Cell Rep ; 9(1): 118-128, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25263552

ABSTRACT

Bone marrow (BM) metastasis remains one of the main causes of death associated with solid tumors as well as multiple myeloma (MM). Targeting the BM niche to prevent or modulate metastasis has not been successful to date. Here, we show that stromal cell-derived factor-1 (SDF-1/CXCL12) is highly expressed in active MM, as well as in BM sites of tumor metastasis and report on the discovery of the high-affinity anti-SDF-1 PEGylated mirror-image l-oligonucleotide (olaptesed-pegol). In vivo confocal imaging showed that SDF-1 levels are increased within MM cell-colonized BM areas. Using in vivo murine and xenograft mouse models, we document that in vivo SDF-1 neutralization within BM niches leads to a microenvironment that is less receptive for MM cells and reduces MM cell homing and growth, thereby inhibiting MM disease progression. Targeting of SDF-1 represents a valid strategy for preventing or disrupting colonization of the BM by MM cells.


Subject(s)
Bone Marrow/pathology , Chemokine CXCL12/antagonists & inhibitors , Multiple Myeloma/therapy , Oligonucleotides/pharmacology , Animals , Bone Marrow/metabolism , Bone Marrow Neoplasms/metabolism , Bone Marrow Neoplasms/secondary , Boronic Acids/pharmacology , Bortezomib , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/genetics , Female , Gene Knockdown Techniques , Humans , Male , Mice , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neoplasm Metastasis , Oligonucleotides/chemistry , Oligonucleotides/genetics , Polyethylene Glycols/chemistry , Pyrazines/pharmacology
8.
Biochem J ; 462(1): 153-62, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24832383

ABSTRACT

The sphingolipid S1P (sphingosine 1-phosphate) is known to be involved in a number of pathophysiological conditions such as cancer, autoimmune diseases and fibrosis. It acts extracellularly through a set of five G-protein-coupled receptors, but its intracellular actions are also well documented. Employing in vitro selection techniques, we identified an L-aptamer (Spiegelmer®) to S1P designated NOX-S93. The binding affinity of NOX-S93 to S1P had a Kd value of 4.3 nM. The Spiegelmer® shows equal binding to dihydro-S1P, but no cross-reactivity to the related lipids sphingosine, lysophosphatidic acid, ceramide, ceramide-1-phosphate or sphingosine phosphocholine. In stably transfected CHO (Chinese-hamster ovary) cell lines expressing the S1P receptors S1PR1 or S1PR3, NOX-S93 inhibits S1P-mediated ß-arrestin recruitment and intracellular calcium release respectively, with IC50 values in the low nanomolar range. The pro-angiogenic activity of S1P, and of the growth factors VEGF-A (vascular endothelial growth factor-A), FGF-2 (fibroblast growth factor-2) and IGF-1 (insulin-like growth factor-1), was effectively blocked by NOX-S93 in a cellular angiogenesis assay employing primary human endothelial cells. These data provide further evidence for the relevance of extracellular S1P as a central mediator of angiogenesis, suggesting pharmacological S1P neutralization as a promising treatment alternative to current anti-angiogenesis approaches.


Subject(s)
Angiogenesis Inhibitors/isolation & purification , Aptamers, Nucleotide/pharmacology , Lysophospholipids/antagonists & inhibitors , Sphingosine/analogs & derivatives , Angiogenesis Inhibitors/pharmacology , Animals , Arrestins/metabolism , CHO Cells , Calcium/metabolism , Cricetulus , Fibroblast Growth Factor 2/antagonists & inhibitors , Humans , Insulin-Like Growth Factor I/antagonists & inhibitors , Neovascularization, Pathologic , Receptors, Lysosphingolipid/metabolism , Sphingosine/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors , beta-Arrestins
9.
Blood ; 123(7): 1032-9, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24277076

ABSTRACT

The CXC chemokine ligand (CXCL12, or stromal cell-derived factor-1 as previously known) plays a critical role for homing and retention of chronic lymphocytic leukemia (CLL) cells in tissues such as the bone marrow (BM). In tissues, stromal cells constitutively secrete and present CXCL12 via cell-surface-bound glycosaminoglycans (GAGs), thereby attracting CLL cells and protecting them from cytotoxic drugs, a mechanism that may account for residual disease after conventional CLL therapy. NOX-A12, an RNA oligonucleotide in L-configuration (Spiegelmer) that binds and neutralizes CXCL12, was developed for interference with CXCL12 in the tumor microenvironment and for cell mobilization. Here, we examined effects of NOX-A12 on CLL cell migration and drug sensitivity. We found that NOX-A12 effectively inhibited CXCL12-induced chemotaxis of CLL cells. In contrast, NOX-A12 increased CLL migration underneath a confluent layer of BM stromal cells (BMSCs) due to interference with the CXCL12 gradient established by BMSCs. In particular, NOX-A12 competes with GAGs such as heparin for CXCL12 binding, leading to the release of CXCL12 from stromal cell-surface-bound GAGs, and thereby to neutralization of the chemokine. Furthermore, NOX-A12 sensitizes CLL cells toward bendamustine and fludarabine in BMSC cocultures. These data demonstrate that NOX-A12 effectively interferes with CLL cell migration and BMSC-mediated drug resistance, and establishes a rationale for clinical development of NOX-A12 in combination with conventional agents in CLL.


Subject(s)
Antineoplastic Agents/pharmacology , Aptamers, Nucleotide/pharmacology , Cell Movement/drug effects , Chemokine CXCL12/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Cells, Cultured , Chemokine CXCL12/pharmacology , Drug Evaluation, Preclinical , Drug Synergism , Humans , Jurkat Cells , Lymphocytes/drug effects , Lymphocytes/physiology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , Recombinant Proteins/pharmacology , Transcellular Cell Migration/drug effects
10.
Neuro Oncol ; 16(1): 21-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24335554

ABSTRACT

BACKGROUND: Tumor irradiation blocks local angiogenesis, forcing any recurrent tumor to form new vessels from circulating cells. We have previously demonstrated that the post-irradiation recurrence of human glioblastomas in the brains of nude mice can be delayed or prevented by inhibiting circulating blood vessel-forming cells by blocking the interaction of CXCR4 with its ligand stromal cell-derived factor (SDF)-1 (CXCL12). In the present study we test this strategy by directly neutralizing SDF-1 in a clinically relevant model using autochthonous brain tumors in immune competent hosts. METHODS: We used NOX-A12, an l-enantiomeric RNA oligonucleotide that binds and inhibits SDF-1 with high affinity. We tested the effect of this inhibitor on the response to irradiation of brain tumors in rat induced by n-ethyl-N-nitrosourea. RESULTS: Rats treated in utero with N-ethyl-N-nitrosourea began to die of brain tumors from approximately 120 days of age. We delivered a single dose of whole brain irradiation (20 Gy) on day 115 of age, began treatment with NOX-A12 immediately following irradiation, and continued with either 5 or 20 mg/kg for 4 or 8 weeks, doses and times equivalent to well-tolerated human exposures. We found a marked prolongation of rat life span that was dependent on both drug dose and duration of treatment. In addition we treated tumors only when they were visible by MRI and demonstrated complete regression of the tumors that was not achieved by irradiation alone or with the addition of temozolomide. CONCLUSIONS: Inhibition of SDF-1 following tumor irradiation is a powerful way of improving tumor response of glioblastoma multiforme.


Subject(s)
Brain Neoplasms/prevention & control , Chemokine CXCL12/antagonists & inhibitors , Neoplasm Recurrence, Local/prevention & control , Oligonucleotides, Antisense/pharmacology , X-Ray Therapy , Alkylating Agents/toxicity , Animals , Brain Neoplasms/chemically induced , Brain Neoplasms/metabolism , Cell Proliferation , Cells, Cultured , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Ethylnitrosourea/toxicity , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/radiation effects , Humans , Magnetic Resonance Imaging , Mice , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/mortality , Rats , Rats, Sprague-Dawley , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR/metabolism , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Survival Rate
11.
Blood ; 121(12): 2311-5, 2013 Mar 21.
Article in English | MEDLINE | ID: mdl-23349391

ABSTRACT

Anemia of chronic inflammation is the most prevalent form of anemia in hospitalized patients. A hallmark of this disease is the intracellular sequestration of iron. This is a consequence of hepcidin-induced internalization and subsequent degradation of ferroportin, the hepcidin receptor and only known iron-export protein. This study describes the characterization of novel anti-hepcidin compound NOX-H94, a structured L-oligoribonucleotide that binds human hepcidin with high affinity (Kd = 0.65 ± 0.06 nmol/L). In J774A.1 macrophages, NOX-H94 blocked hepcidin-induced ferroportin degradation and ferritin expression (half maximal inhibitory concentration = 19.8 ± 4.6 nmol/L). In an acute cynomolgus monkey model of interleukin 6 (IL-6)-induced hypoferremia, NOX-H94 inhibited serum iron reduction completely. In a subchronic model of IL-6-induced anemia, NOX-H94 inhibited the decrease in hemoglobin concentration. We conclude that NOX-H94 protects ferroportin from hepcidin-induced degradation. Therefore, this pharmacologic approach may represent an interesting treatment option for patients suffering from anemia of chronic inflammation.


Subject(s)
Anemia/drug therapy , Anemia/etiology , Inflammation/complications , Inflammation/drug therapy , Oligoribonucleotides/therapeutic use , Anemia/pathology , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antimicrobial Cationic Peptides/antagonists & inhibitors , Cells, Cultured , Disease Models, Animal , Drug Evaluation, Preclinical , Erythrocytes/drug effects , Erythrocytes/metabolism , Hemoglobins/analysis , Hemoglobins/drug effects , Hepcidins , Interleukin-6/administration & dosage , Interleukin-6/adverse effects , Iron/blood , Iron/metabolism , Iron Metabolism Disorders/chemically induced , Macaca fascicularis , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Oligoribonucleotides/administration & dosage , Oligoribonucleotides/pharmacology
12.
Mol Cancer ; 9(1): 65, 2010 Mar 22.
Article in English | MEDLINE | ID: mdl-20307265

ABSTRACT

BACKGROUND: Functional loss of the tumor suppressor Smad4 is involved in pancreatic and colorectal carcinogenesis and has been associated with the acquisition of invasiveness. We have previously demonstrated that the heterotrimeric basement membrane protein laminin-332 is a Smad4 target. Namely, Smad4 functions as a positive transcriptional regulator of all three genes encoding laminin-332; its loss is thus implicated in the reduced or discontinuous deposition of the heterotrimeric basement membrane molecule as evident in carcinomas. Uncoupled expression of laminin genes, on the other hand, namely overexpression of the laminin-gamma2 chain is an impressive marker at invasive edges of carcinomas where tumor cells are maximally exposed to signals from stromal cell types like macrophages. As Smad4 is characterized as an integrator of multiple extracellular stimuli in a strongly contextual manner, we asked if loss of Smad4 may also be involved in uncoupled expression of laminin genes in response to altered environmental stimuli. Here, we address Smad4 dependent effects of the prominent inflammatory cytokine TNFalpha on tumor cells. RESULTS: Smad4-reconstituted colon carcinoma cells like adenoma cells respond to TNFalpha with an increased expression of all three chains encoding laminin-332; coincubation with TGFbeta and TNFalpha leads to synergistic induction and to the secretion of large amounts of the heterotrimer. In contrast, in Smad4-deficient cells TNFalpha can induce expression of the gamma2 and beta3 but not the alpha3 chain. Surprisingly, this uncoupled induction of laminin-332 chains in Smad4-negative cells rather than causing intracellular accumulation is followed by the release of gamma2 into the medium, either in a monomeric form or in complexes with as yet unknown proteins. Soluble gamma2 is associated with increased cell migration. CONCLUSIONS: Loss of Smad4 may lead to uncoupled induction of laminin-gamma2 in response to TNFalpha and may therefore represent one of the mechanisms which underlie accumulation of laminin-gamma2 at the invasive margin of a tumor. The finding, that gamma2 is secreted from tumor cells in significant amounts and is associated with increased cell migration may pave the way for further investigation to better understand its functional relevance for tumor progression.


Subject(s)
Colorectal Neoplasms/metabolism , Laminin/metabolism , Smad4 Protein/deficiency , Tumor Necrosis Factor-alpha/pharmacology , Adenoma/metabolism , Amino Acid Sequence , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Movement/physiology , Drug Synergism , Gene Knockdown Techniques , Humans , Laminin/chemistry , Mass Spectrometry , Molecular Sequence Data , NF-kappa B/metabolism , Promoter Regions, Genetic , Protein Conformation , Smad4 Protein/genetics , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Kalinin
13.
BMC Cancer ; 8: 215, 2008 Jul 29.
Article in English | MEDLINE | ID: mdl-18664273

ABSTRACT

BACKGROUND: Functional inactivation of the tumor suppressor Smad4 in colorectal and pancreatic carcinogenesis occurs coincident with the transition to invasive growth. Breaking the basement membrane (BM) barrier, a prerequisite for invasive growth, can be due to tumor induced proteolytic tissue remodeling or to reduced synthesis of BM molecules by incipient tumor cells. Laminin-332 (laminin-5), a heterotrimeric BM component composed of alpha 3-, beta 3- and gamma 2-chains, has recently been identified as a target structure of Smad4 and represents the first example for expression control of an essential BM component by a tumor and invasion suppressor. Biochemically Smad4 is a transmitter of signals of the TGFbeta superfamily of cytokines. We have reported previously, that Smad4 functions as a positive transcriptional regulator of constitutive and of TGFbeta-induced transcription of all three genes encoding Laminin-332, LAMA3, LAMB3 and LAMC2. METHODS: Promoter-reporter constructs harboring 4 kb upstream regions, each of the three genes encoding Laminin-322 as well as deletion and mutations constructs were established. Promoter activities and TGFbeta induction were assayed through transient transfections in Smad4-negative human cancer cells and their stable Smad4-positive derivatives. Functionally relevant binding sites were subsequently confirmed through chromatin immunoprecipitation. RESULTS: Herein, we report that Smad4 mediates transcriptional regulation through three different mechanisms, namely through Smad4 binding to a functional SBE site exclusively in the LAMA3 promoter, Smad4 binding to AP1 (and Sp1) sites presumably via interaction with AP1 family components and lastly a Smad4 impact on transcription of AP1 factors. Whereas Smad4 is essential for positive regulation of all three genes, the molecular mechanisms are significantly divergent between the LAMA3 promoter as compared to the LAMB3 and LAMC2 promoters. CONCLUSION: We hypothesize that this divergence in modular regulation of the three promoters may lay the ground for uncoupled regulation of Laminin-332 in Smad4-deficient tumor cells in response to stromally expressed cytokines acting on budding tumor cells.


Subject(s)
Basement Membrane/metabolism , Cell Adhesion Molecules/chemistry , Gene Expression Regulation, Neoplastic , Smad4 Protein/metabolism , Carcinoma/metabolism , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Cytokines/biosynthesis , Cytokines/metabolism , Humans , Laminin/metabolism , Pancreatic Neoplasms/metabolism , Promoter Regions, Genetic , Transcription Factor AP-1/metabolism , Transcription, Genetic , Transforming Growth Factor beta/metabolism , Kalinin
SELECTION OF CITATIONS
SEARCH DETAIL
...