LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 16

Search options

  1. Article ; Online: Cancer cell autophagy, reprogrammed macrophages, and remodeled vasculature in glioblastoma triggers tumor immunity.

    Chryplewicz, Agnieszka / Scotton, Julie / Tichet, Mélanie / Zomer, Anoek / Shchors, Ksenya / Joyce, Johanna A / Homicsko, Krisztian / Hanahan, Douglas

    Cancer cell

    2022  Volume 40, Issue 10, Page(s) 1111–1127.e9

    Abstract: Glioblastoma (GBM) is poorly responsive to therapy and invariably lethal. One conceivable strategy to circumvent this intractability is to co-target distinctive mechanistic components of the disease, aiming to concomitantly disrupt multiple capabilities ... ...

    Abstract Glioblastoma (GBM) is poorly responsive to therapy and invariably lethal. One conceivable strategy to circumvent this intractability is to co-target distinctive mechanistic components of the disease, aiming to concomitantly disrupt multiple capabilities required for tumor progression and therapeutic resistance. We assessed this concept by combining vascular endothelial growth factor (VEGF) pathway inhibitors that remodel the tumor vasculature with the tricyclic antidepressant imipramine, which enhances autophagy in GBM cancer cells and unexpectedly reprograms immunosuppressive tumor-associated macrophages via inhibition of histamine receptor signaling to become immunostimulatory. While neither drug is efficacious as monotherapy, the combination of imipramine with VEGF pathway inhibitors orchestrates the infiltration and activation of CD8 and CD4 T cells, producing significant therapeutic benefit in several GBM mouse models. Inclusion up front of immune-checkpoint blockade with anti-programmed death-ligand 1 (PD-L1) in eventually relapsing tumors markedly extends survival benefit. The results illustrate the potential of mechanism-guided therapeutic co-targeting of disparate biological vulnerabilities in the tumor microenvironment.
    MeSH term(s) Animals ; Antidepressive Agents, Tricyclic/metabolism ; Antidepressive Agents, Tricyclic/therapeutic use ; Autophagy ; B7-H1 Antigen/metabolism ; Glioblastoma/pathology ; Imipramine/metabolism ; Imipramine/therapeutic use ; Immune Checkpoint Inhibitors ; Immunotherapy ; Macrophages/metabolism ; Mice ; Neoplasm Recurrence, Local/drug therapy ; Programmed Cell Death 1 Receptor ; Tumor Microenvironment ; Vascular Endothelial Growth Factor A/metabolism
    Chemical Substances Antidepressive Agents, Tricyclic ; B7-H1 Antigen ; Immune Checkpoint Inhibitors ; Programmed Cell Death 1 Receptor ; Vascular Endothelial Growth Factor A ; Imipramine (OGG85SX4E4)
    Language English
    Publishing date 2022-09-15
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2078448-X
    ISSN 1878-3686 ; 1535-6108
    ISSN (online) 1878-3686
    ISSN 1535-6108
    DOI 10.1016/j.ccell.2022.08.014
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: Bispecific PD1-IL2v and anti-PD-L1 break tumor immunity resistance by enhancing stem-like tumor-reactive CD8

    Tichet, Mélanie / Wullschleger, Stephan / Chryplewicz, Agnieszka / Fournier, Nadine / Marcone, Rachel / Kauzlaric, Annamaria / Homicsko, Krisztian / Deak, Laura Codarri / Umaña, Pablo / Klein, Christian / Hanahan, Douglas

    Immunity

    2023  Volume 56, Issue 1, Page(s) 162–179.e6

    Abstract: Immunotherapies have shown remarkable, albeit tumor-selective, therapeutic benefits in the clinic. Most patients respond transiently at best, highlighting the importance of understanding mechanisms underlying resistance. Herein, we evaluated the effects ... ...

    Abstract Immunotherapies have shown remarkable, albeit tumor-selective, therapeutic benefits in the clinic. Most patients respond transiently at best, highlighting the importance of understanding mechanisms underlying resistance. Herein, we evaluated the effects of the engineered immunocytokine PD1-IL2v in a mouse model of de novo pancreatic neuroendocrine cancer that is resistant to checkpoint and other immunotherapies. PD1-IL2v utilizes anti-PD-1 as a targeting moiety fused to an immuno-stimulatory IL-2 cytokine variant (IL2v) to precisely deliver IL2v to PD-1
    MeSH term(s) Animals ; Mice ; B7-H1 Antigen/immunology ; CD8-Positive T-Lymphocytes/immunology ; CD8-Positive T-Lymphocytes/metabolism ; Disease Models, Animal ; Immunotherapy/methods ; Macrophages/immunology ; Macrophages/metabolism ; Neoplasms/therapy ; Tumor Microenvironment ; Antibodies, Bispecific/immunology ; Interleukin-2 ; Programmed Cell Death 1 Receptor/immunology
    Chemical Substances B7-H1 Antigen ; Antibodies, Bispecific ; Interleukin-2 ; Programmed Cell Death 1 Receptor
    Language English
    Publishing date 2023-01-09
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 1217235-2
    ISSN 1097-4180 ; 1074-7613
    ISSN (online) 1097-4180
    ISSN 1074-7613
    DOI 10.1016/j.immuni.2022.12.006
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: ALK7 Signaling Manifests a Homeostatic Tissue Barrier That Is Abrogated during Tumorigenesis and Metastasis.

    Michael, Iacovos P / Saghafinia, Sadegh / Tichet, Mélanie / Zangger, Nadine / Marinoni, Ilaria / Perren, Aurel / Hanahan, Douglas

    Developmental cell

    2019  Volume 49, Issue 3, Page(s) 409–424.e6

    Abstract: Herein, we report that the TGFß superfamily receptor ALK7 is a suppressor of tumorigenesis and metastasis, as revealed by functional studies in mouse models of pancreatic neuroendocrine and luminal breast cancer, complemented by experimental metastasis ... ...

    Abstract Herein, we report that the TGFß superfamily receptor ALK7 is a suppressor of tumorigenesis and metastasis, as revealed by functional studies in mouse models of pancreatic neuroendocrine and luminal breast cancer, complemented by experimental metastasis assays. Activation in neoplastic cells of the ALK7 signaling pathway by its principal ligand activin B induces apoptosis. During tumorigenesis, cancer cells use two different approaches to evade this barrier, either downregulating activin B and/or downregulating ALK7. Suppressing ALK7 expression additionally contributes to the capability for metastatic seeding. ALK7 is associated with shorter relapse-free survival of various human cancers and distant-metastasis-free survival of breast cancer patients. This study introduces mechanistic insights into primary and metastatic tumor development, in the form of a protective barrier that triggers apoptosis in cells that are not "authorized" to proliferate within a particular tissue, by virtue of those cells expressing ALK7 in a tissue microenvironment bathed in its ligand.
    MeSH term(s) Activin Receptors, Type I/metabolism ; Activins/metabolism ; Animals ; Apoptosis/physiology ; Breast Neoplasms/metabolism ; Breast Neoplasms/pathology ; Carcinogenesis ; Cell Line, Tumor ; Cell Transformation, Neoplastic/metabolism ; Female ; Heterografts ; Homeostasis ; Humans ; Male ; Mice ; Mice, Inbred A ; Mice, Inbred C57BL ; Mice, SCID ; Neoplasm Metastasis ; Neoplasms/metabolism ; Neoplasms/pathology ; Pancreatic Neoplasms/metabolism ; Pancreatic Neoplasms/pathology ; Signal Transduction ; Smad2 Protein/metabolism ; Transforming Growth Factor beta/metabolism ; Tumor Microenvironment
    Chemical Substances Smad2 Protein ; Transforming Growth Factor beta ; activin B ; Activins (104625-48-1) ; ACVR1C protein, human (EC 2.7.11.30) ; Activin Receptors, Type I (EC 2.7.11.30)
    Language English
    Publishing date 2019-05-04
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2054967-2
    ISSN 1878-1551 ; 1534-5807
    ISSN (online) 1878-1551
    ISSN 1534-5807
    DOI 10.1016/j.devcel.2019.04.015
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Nanoparticle Conjugation of Human Papillomavirus 16 E7-long Peptides Enhances Therapeutic Vaccine Efficacy against Solid Tumors in Mice.

    Galliverti, Gabriele / Tichet, Mélanie / Domingos-Pereira, Sonia / Hauert, Sylvie / Nardelli-Haefliger, Denise / Swartz, Melody A / Hanahan, Douglas / Wullschleger, Stephan

    Cancer immunology research

    2018  Volume 6, Issue 11, Page(s) 1301–1313

    Abstract: Treatment of patients bearing human papillomavirus (HPV)-related cancers with synthetic long-peptide (SLP) therapeutic vaccines has shown promising results in clinical trials against premalignant lesions, whereas responses against later stage carcinomas ... ...

    Abstract Treatment of patients bearing human papillomavirus (HPV)-related cancers with synthetic long-peptide (SLP) therapeutic vaccines has shown promising results in clinical trials against premalignant lesions, whereas responses against later stage carcinomas have remained elusive. We show that conjugation of a well-documented HPV-E7 SLP to ultra-small polymeric nanoparticles (NP) enhances the antitumor efficacy of therapeutic vaccination in different mouse models of HPV
    MeSH term(s) Animals ; Antibodies/pharmacology ; CD8-Positive T-Lymphocytes/immunology ; Cancer Vaccines/chemistry ; Cancer Vaccines/immunology ; Cancer Vaccines/pharmacology ; Female ; Lung Neoplasms/secondary ; Mice, Inbred C57BL ; Mice, Transgenic ; Nanoparticles/chemistry ; Neoplasm Recurrence, Local ; Neoplasms, Experimental/immunology ; Neoplasms, Experimental/mortality ; Neoplasms, Experimental/therapy ; Papillomavirus E7 Proteins/chemistry ; Papillomavirus E7 Proteins/immunology ; Papillomavirus E7 Proteins/pharmacology ; T-Lymphocytes, Regulatory/immunology ; Treatment Outcome ; Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology ; Vaginal Neoplasms/immunology ; Vaginal Neoplasms/pathology ; Vaginal Neoplasms/prevention & control
    Chemical Substances Antibodies ; Cancer Vaccines ; Papillomavirus E7 Proteins ; Tumor Necrosis Factor Receptor Superfamily, Member 9 ; oncogene protein E7, Human papillomavirus type 16
    Language English
    Publishing date 2018-08-21
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2732489-8
    ISSN 2326-6074 ; 2326-6066
    ISSN (online) 2326-6074
    ISSN 2326-6066
    DOI 10.1158/2326-6066.CIR-18-0166
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: Aberrant hyperexpression of the RNA binding protein FMRP in tumors mediates immune evasion.

    Zeng, Qiqun / Saghafinia, Sadegh / Chryplewicz, Agnieszka / Fournier, Nadine / Christe, Lucine / Xie, Yu-Qing / Guillot, Jeremy / Yucel, Simge / Li, Pumin / Galván, José A / Karamitopoulou, Eva / Zlobec, Inti / Ataca, Dalya / Gallean, Fleuriane / Zhang, Peng / Rodriguez-Calero, José Antonio / Rubin, Mark / Tichet, Mélanie / Homicsko, Krisztian /
    Hanahan, Douglas

    Science (New York, N.Y.)

    2022  Volume 378, Issue 6621, Page(s) eabl7207

    Abstract: Many human cancers manifest the capability to circumvent attack by the adaptive immune system. In this work, we identified a component of immune evasion that involves frequent up-regulation of fragile X mental retardation protein (FMRP) in solid tumors. ... ...

    Abstract Many human cancers manifest the capability to circumvent attack by the adaptive immune system. In this work, we identified a component of immune evasion that involves frequent up-regulation of fragile X mental retardation protein (FMRP) in solid tumors. FMRP represses immune attack, as revealed by cancer cells engineered to lack its expression. FMRP-deficient tumors were infiltrated by activated T cells that impaired tumor growth and enhanced survival in mice. Mechanistically, FMRP's immunosuppression was multifactorial, involving repression of the chemoattractant C-C motif chemokine ligand 7 (CCL7) concomitant with up-regulation of three immunomodulators-interleukin-33 (IL-33), tumor-secreted protein S (PROS1), and extracellular vesicles. Gene signatures associate FMRP's cancer network with poor prognosis and response to therapy in cancer patients. Collectively, FMRP is implicated as a regulator that orchestrates a multifaceted barrier to antitumor immune responses.
    MeSH term(s) Animals ; Humans ; Mice ; Fragile X Mental Retardation Protein/genetics ; Fragile X Mental Retardation Protein/metabolism ; Immune Evasion ; Neoplasms/immunology ; Immune Tolerance ; Chemokine CCL7/metabolism ; Interleukin-33 ; Protein S/metabolism
    Chemical Substances Fragile X Mental Retardation Protein (139135-51-6) ; FMR1 protein, human ; Chemokine CCL7 ; Interleukin-33 ; PROS1 protein, human ; Protein S
    Language English
    Publishing date 2022-11-18
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 128410-1
    ISSN 1095-9203 ; 0036-8075
    ISSN (online) 1095-9203
    ISSN 0036-8075
    DOI 10.1126/science.abl7207
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: Risk for nevus transformation and melanoma proliferation and invasion during natalizumab treatment: four years of dermoscopic follow-up with immunohistological studies and proliferation and invasion assays.

    Pharaon, Momen / Tichet, Mélanie / Lebrun-Frénay, Christine / Tartare-Deckert, Sophie / Passeron, Thierry

    JAMA dermatology

    2014  Volume 150, Issue 8, Page(s) 901–903

    MeSH term(s) Antibodies, Monoclonal, Humanized/pharmacology ; Cell Line, Tumor ; Cell Movement/drug effects ; Cell Proliferation/drug effects ; Cell Transformation, Neoplastic ; Dermoscopy ; Follow-Up Studies ; Humans ; Immunohistochemistry ; Immunologic Factors/pharmacology ; Integrin beta3/analysis ; Melanoma/chemistry ; Melanoma/pathology ; Multiple Sclerosis/drug therapy ; Natalizumab ; Nevus/chemistry ; Nevus/pathology ; Osteonectin/analysis ; Skin Neoplasms/chemistry ; Skin Neoplasms/pathology
    Chemical Substances Antibodies, Monoclonal, Humanized ; Immunologic Factors ; Integrin beta3 ; Natalizumab ; Osteonectin ; SPARC protein, human
    Language English
    Publishing date 2014-08
    Publishing country United States
    Document type Journal Article ; Observational Study ; Research Support, Non-U.S. Gov't
    ZDB-ID 2701761-8
    ISSN 2168-6084 ; 2168-6068
    ISSN (online) 2168-6084
    ISSN 2168-6068
    DOI 10.1001/jamadermatol.2013.9411
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: Myeloid Cells Orchestrate Systemic Immunosuppression, Impairing the Efficacy of Immunotherapy against HPV

    Galliverti, Gabriele / Wullschleger, Stephan / Tichet, Mélanie / Murugan, Dhaarini / Zangger, Nadine / Horton, Wesley / Korman, Alan J / Coussens, Lisa M / Swartz, Melody A / Hanahan, Douglas

    Cancer immunology research

    2019  Volume 8, Issue 1, Page(s) 131–145

    Abstract: Cancers induced by human papillomaviruses (HPV) should be responsive to immunotherapy by virtue of expressing the immunogenic oncoproteins E6/E7. However, advanced forms of cervical cancer, driven by HPV, are poorly responsive to immune response- ... ...

    Abstract Cancers induced by human papillomaviruses (HPV) should be responsive to immunotherapy by virtue of expressing the immunogenic oncoproteins E6/E7. However, advanced forms of cervical cancer, driven by HPV, are poorly responsive to immune response-enhancing treatments involving therapeutic vaccination against these viral neoantigens. Leveraging a transgenic mouse model of HPV-derived cancers, K14HPV16/H2b, we demonstrated that a potent nanoparticle-based E7 vaccine, but not a conventional "liquid" vaccine, induced E7 tumor antigen-specific CD8
    MeSH term(s) Animals ; Antineoplastic Agents, Immunological/pharmacology ; CD8-Positive T-Lymphocytes/immunology ; CTLA-4 Antigen/antagonists & inhibitors ; Disease Models, Animal ; Drug Synergism ; Female ; Humans ; Immunosuppression Therapy ; Immunotherapy/methods ; Mice ; Myeloid Cells/drug effects ; Myeloid Cells/immunology ; Nanoparticles/administration & dosage ; Papillomaviridae/immunology ; Papillomavirus E7 Proteins/immunology ; Papillomavirus E7 Proteins/metabolism ; Papillomavirus Infections/drug therapy ; Papillomavirus Infections/immunology ; Papillomavirus Infections/virology ; Papillomavirus Vaccines/administration & dosage ; Papillomavirus Vaccines/immunology ; Programmed Cell Death 1 Receptor/antagonists & inhibitors ; Tumor Microenvironment ; Uterine Cervical Neoplasms/drug therapy ; Uterine Cervical Neoplasms/immunology ; Uterine Cervical Neoplasms/pathology ; Uterine Cervical Neoplasms/virology
    Chemical Substances Antineoplastic Agents, Immunological ; CTLA-4 Antigen ; Ctla4 protein, mouse ; Papillomavirus E7 Proteins ; Papillomavirus Vaccines ; Pdcd1 protein, mouse ; Programmed Cell Death 1 Receptor
    Language English
    Publishing date 2019-11-26
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2732489-8
    ISSN 2326-6074 ; 2326-6066
    ISSN (online) 2326-6074
    ISSN 2326-6066
    DOI 10.1158/2326-6066.CIR-19-0315
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: Targeting the Proteasome-Associated Deubiquitinating Enzyme USP14 Impairs Melanoma Cell Survival and Overcomes Resistance to MAPK-Targeting Therapies.

    Didier, Robin / Mallavialle, Aude / Ben Jouira, Rania / Domdom, Marie Angela / Tichet, Mélanie / Auberger, Patrick / Luciano, Frédéric / Ohanna, Mickael / Tartare-Deckert, Sophie / Deckert, Marcel

    Molecular cancer therapeutics

    2018  Volume 17, Issue 7, Page(s) 1416–1429

    Abstract: Advanced cutaneous melanoma is one of the most challenging cancers to treat because of its high plasticity, metastatic potential, and resistance to treatment. New targeted therapies and immunotherapies have shown remarkable clinical efficacy. However, ... ...

    Abstract Advanced cutaneous melanoma is one of the most challenging cancers to treat because of its high plasticity, metastatic potential, and resistance to treatment. New targeted therapies and immunotherapies have shown remarkable clinical efficacy. However, such treatments are limited to a subset of patients and relapses often occur, warranting validation of novel targeted therapies. Posttranslational modification of proteins by ubiquitin coordinates essential cellular functions, including ubiquitin-proteasome system (UPS) function and protein homeostasis. Deubiquitinating enzymes (DUB) have been associated to multiple diseases, including cancer. However, their exact involvement in melanoma development and therapeutic resistance remains poorly understood. Using a DUB trap assay to label cellular active DUBs, we have observed an increased activity of the proteasome-associated DUB, USP14 (Ubiquitin-specific peptidase 14) in melanoma cells compared with melanocytes. Our survey of public gene expression databases indicates that high expression of
    MeSH term(s) Animals ; Apoptosis/drug effects ; Cell Line, Tumor ; Cell Survival/drug effects ; Deubiquitinating Enzymes/antagonists & inhibitors ; Deubiquitinating Enzymes/genetics ; Drug Resistance, Neoplasm/genetics ; GTP Phosphohydrolases/genetics ; Gene Expression Regulation, Neoplastic/drug effects ; Humans ; MAP Kinase Kinase 1/genetics ; Melanocytes/drug effects ; Melanoma/drug therapy ; Melanoma/genetics ; Melanoma/pathology ; Membrane Proteins/genetics ; Mice ; Molecular Targeted Therapy ; Proteasome Inhibitors/pharmacology ; Proto-Oncogene Proteins B-raf/genetics ; Reactive Oxygen Species/metabolism ; Tumor Suppressor Protein p53/genetics ; Ubiquitin Thiolesterase/antagonists & inhibitors ; Ubiquitin Thiolesterase/genetics ; Xenograft Model Antitumor Assays
    Chemical Substances Membrane Proteins ; Proteasome Inhibitors ; Reactive Oxygen Species ; TP53 protein, human ; Tumor Suppressor Protein p53 ; USP14 protein, human ; BRAF protein, human (EC 2.7.11.1) ; Proto-Oncogene Proteins B-raf (EC 2.7.11.1) ; MAP Kinase Kinase 1 (EC 2.7.12.2) ; Deubiquitinating Enzymes (EC 3.4.19.12) ; Ubiquitin Thiolesterase (EC 3.4.19.12) ; GTP Phosphohydrolases (EC 3.6.1.-) ; NRAS protein, human (EC 3.6.1.-)
    Language English
    Publishing date 2018-04-27
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2063563-1
    ISSN 1538-8514 ; 1535-7163
    ISSN (online) 1538-8514
    ISSN 1535-7163
    DOI 10.1158/1535-7163.MCT-17-0919
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: PD-1-cis IL-2R agonism yields better effectors from stem-like CD8

    Codarri Deak, Laura / Nicolini, Valeria / Hashimoto, Masao / Karagianni, Maria / Schwalie, Petra C / Lauener, Laura / Varypataki, Eleni Maria / Richard, Marine / Bommer, Esther / Sam, Johannes / Joller, Stefanie / Perro, Mario / Cremasco, Floriana / Kunz, Leo / Yanguez, Emilio / Hüsser, Tamara / Schlenker, Ramona / Mariani, Marisa / Tosevski, Vinko /
    Herter, Sylvia / Bacac, Marina / Waldhauer, Inja / Colombetti, Sara / Gueripel, Xavier / Wullschleger, Stephan / Tichet, Melanie / Hanahan, Douglas / Kissick, Haydn T / Leclair, Stephane / Freimoser-Grundschober, Anne / Seeber, Stefan / Teichgräber, Volker / Ahmed, Rafi / Klein, Christian / Umaña, Pablo

    Nature

    2022  Volume 610, Issue 7930, Page(s) 161–172

    Abstract: Expansion and differentiation of antigen-experienced PD- ... ...

    Abstract Expansion and differentiation of antigen-experienced PD-1
    MeSH term(s) Antibodies, Blocking/immunology ; Antibodies, Blocking/pharmacology ; Antibodies, Blocking/therapeutic use ; B7-H1 Antigen/antagonists & inhibitors ; B7-H1 Antigen/immunology ; CD8-Positive T-Lymphocytes/cytology ; CD8-Positive T-Lymphocytes/drug effects ; CD8-Positive T-Lymphocytes/immunology ; Infections/drug therapy ; Infections/immunology ; Interleukin-2/immunology ; Interleukin-2/pharmacology ; Interleukin-2/therapeutic use ; Interleukin-2 Receptor alpha Subunit/agonists ; Neoplasms/drug therapy ; Neoplasms/immunology ; Programmed Cell Death 1 Receptor/antagonists & inhibitors ; Receptors, Interleukin-2/agonists
    Chemical Substances Antibodies, Blocking ; B7-H1 Antigen ; Interleukin-2 ; Interleukin-2 Receptor alpha Subunit ; Programmed Cell Death 1 Receptor ; Receptors, Interleukin-2
    Language English
    Publishing date 2022-09-28
    Publishing country England
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 120714-3
    ISSN 1476-4687 ; 0028-0836
    ISSN (online) 1476-4687
    ISSN 0028-0836
    DOI 10.1038/s41586-022-05192-0
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article ; Online: Tumour-derived SPARC drives vascular permeability and extravasation through endothelial VCAM1 signalling to promote metastasis.

    Tichet, Mélanie / Prod'Homme, Virginie / Fenouille, Nina / Ambrosetti, Damien / Mallavialle, Aude / Cerezo, Michael / Ohanna, Mickaël / Audebert, Stéphane / Rocchi, Stéphane / Giacchero, Damien / Boukari, Fériel / Allegra, Maryline / Chambard, Jean-Claude / Lacour, Jean-Philippe / Michiels, Jean-François / Borg, Jean-Paul / Deckert, Marcel / Tartare-Deckert, Sophie

    Nature communications

    2015  Volume 6, Page(s) 6993

    Abstract: Disruption of the endothelial barrier by tumour-derived secreted factors is a critical step in cancer cell extravasation and metastasis. Here, by comparative proteomic analysis of melanoma secretomes, we identify the matricellular protein SPARC as a ... ...

    Abstract Disruption of the endothelial barrier by tumour-derived secreted factors is a critical step in cancer cell extravasation and metastasis. Here, by comparative proteomic analysis of melanoma secretomes, we identify the matricellular protein SPARC as a novel tumour-derived vascular permeability factor. SPARC deficiency abrogates tumour-initiated permeability of lung capillaries and prevents extravasation, whereas SPARC overexpression enhances vascular leakiness, extravasation and lung metastasis. SPARC-induced paracellular permeability is dependent on the endothelial VCAM1 receptor and p38 MAPK signalling. Blocking VCAM1 impedes melanoma-induced endothelial permeability and extravasation. The clinical relevance of our findings is highlighted by high levels of SPARC detected in tumour from human pulmonary melanoma lesions. Our study establishes tumour-produced SPARC and VCAM1 as regulators of cancer extravasation, revealing a novel targetable interaction for prevention of metastasis.
    MeSH term(s) Animals ; Capillary Permeability ; Case-Control Studies ; Cell Line, Tumor ; Endothelium, Vascular/metabolism ; Female ; Human Umbilical Vein Endothelial Cells ; Humans ; Lung Neoplasms/secondary ; MAP Kinase Signaling System ; Melanoma/metabolism ; Melanoma/pathology ; Melanoma/secretion ; Mice, Nude ; Neoplasm Metastasis ; Osteonectin/metabolism ; Paracrine Communication ; Vascular Cell Adhesion Molecule-1/metabolism
    Chemical Substances Osteonectin ; SPARC protein, human ; Vascular Cell Adhesion Molecule-1
    Language English
    Publishing date 2015-04-30
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ISSN 2041-1723
    ISSN (online) 2041-1723
    DOI 10.1038/ncomms7993
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top