LIVIVO - Das Suchportal für Lebenswissenschaften

switch to English language
Erweiterte Suche

Ihre letzten Suchen

  1. AU="Ng, Mei R"
  2. AU=Button Brian AU=Button Brian
  3. AU=Ramiro Sofia
  4. AU="Marsh, Henry"
  5. AU="Wang, Guixuan"
  6. AU="Luga, Erion"
  7. AU=Patel Shilpan M AU=Patel Shilpan M
  8. AU="Cherniack, Ariella R"
  9. AU=Lai A C K
  10. AU="Cortrezzi, Sylvia"
  11. AU="Fan, Kathy"
  12. AU="Proshkina, Anastasia"
  13. AU="Abdel-Hady, Enas A"
  14. AU="George, Sherly A"
  15. AU=Nicholas Tendongfor
  16. AU="Robert J Coffey"
  17. AU=Aboubakr Hamada A AU=Aboubakr Hamada A
  18. AU="Daniela Brunner"
  19. AU="Abdi Ghavidel, Afshin"
  20. AU="Sagheb, K"
  21. AU="Vincent, B"
  22. AU="Vitale, Carmine"
  23. AU="Trasino, Steven E" AU="Trasino, Steven E"
  24. AU="Joachim Schüz"
  25. AU="Taghizadieh, Ali"
  26. AU=Patel Alpesh A
  27. AU="Uchida, Hiroki"
  28. AU="Veldtman, Gruschen R"
  29. AU="Taiba, C"
  30. AU="Xu-wen Liu"
  31. AU="Chakraborty, Pulak"
  32. AU="Jinhua Xu"
  33. AU="Lai, Tsz-Wah"
  34. AU="Bhudia, Nisha"
  35. AU="Gee, Bruce"
  36. AU="Baogang Sun"
  37. AU="Suszycki, L"
  38. AU="Edward G. Clark"
  39. AU="Regan, Anthony"
  40. AU="Hammoudi Halat, Dalal"
  41. AU="Lanata, Claudio F"
  42. AU="Bao, Ho The"
  43. AU="Morin, Cory W"
  44. AU="Deniskina, N."
  45. AU="Filipović, Nenad R"
  46. AU="Stülke, Jörg"
  47. AU="Tran Kiem, Cécile"
  48. AU="Trivedi, Pritesh S"
  49. AU="Seemann, Stefan E"
  50. AU="Lotfi Chouchane"
  51. AU="Fay, Bianca"
  52. AU="Dahlke, Marc-H"
  53. AU="Foell, Jens"
  54. AU="Bruno Alessandro Rivieccio"
  55. AU="Cristea, Alexandra I"
  56. AU="Sakr, Hader I"
  57. AU="Shikora, Scott A."
  58. AU="Raza Naqvi"
  59. AU="Chin Fatt, Cherise R"
  60. AU="Leena Sahlström"
  61. AU="Karkare, Siddharth"
  62. AU="Corrêa, Diogo G"
  63. AU="Choi, Sun Ha"

Suchergebnis

Treffer 1 - 7 von insgesamt 7

Suchoptionen

  1. Artikel ; Online: Phase II and Biomarker Study of Cabozantinib in Metastatic Triple-Negative Breast Cancer Patients.

    Tolaney, Sara M / Ziehr, David R / Guo, Hao / Ng, Mei R / Barry, William T / Higgins, Michaela J / Isakoff, Steven J / Brock, Jane E / Ivanova, Elena V / Paweletz, Cloud P / Demeo, Michelle K / Ramaiya, Nikhil H / Overmoyer, Beth A / Jain, Rakesh K / Winer, Eric P / Duda, Dan G

    The oncologist

    2021  Band 26, Heft 8, Seite(n) e1483

    Sprache Englisch
    Erscheinungsdatum 2021-05-12
    Erscheinungsland United States
    Dokumenttyp Published Erratum
    ZDB-ID 1409038-7
    ISSN 1549-490X ; 1083-7159
    ISSN (online) 1549-490X
    ISSN 1083-7159
    DOI 10.1002/onco.13809
    Datenquelle MEDical Literature Analysis and Retrieval System OnLINE

    Zusatzmaterialien

    Kategorien

  2. Artikel ; Online: Losartan controls immune checkpoint blocker-induced edema and improves survival in glioblastoma mouse models.

    Datta, Meenal / Chatterjee, Sampurna / Perez, Elizabeth M / Gritsch, Simon / Roberge, Sylvie / Duquette, Mark / Chen, Ivy X / Naxerova, Kamila / Kumar, Ashwin S / Ghosh, Mitrajit / Emblem, Kyrre E / Ng, Mei R / Ho, William W / Kumar, Pragya / Krishnan, Shanmugarajan / Dong, Xinyue / Speranza, Maria C / Neagu, Martha R / Iorgulescu, J Bryan /
    Huang, Raymond Y / Youssef, Gilbert / Reardon, David A / Sharpe, Arlene H / Freeman, Gordon J / Suvà, Mario L / Xu, Lei / Jain, Rakesh K

    Proceedings of the National Academy of Sciences of the United States of America

    2023  Band 120, Heft 6, Seite(n) e2219199120

    Abstract: Immune checkpoint blockers (ICBs) have failed in all phase III glioblastoma trials. Here, we found that ICBs induce cerebral edema in some patients and mice with glioblastoma. Through single-cell RNA sequencing, intravital imaging, and ... ...

    Abstract Immune checkpoint blockers (ICBs) have failed in all phase III glioblastoma trials. Here, we found that ICBs induce cerebral edema in some patients and mice with glioblastoma. Through single-cell RNA sequencing, intravital imaging, and CD8
    Mesh-Begriff(e) Animals ; Mice ; Glioblastoma/pathology ; Losartan/pharmacology ; Losartan/therapeutic use ; Immune Checkpoint Inhibitors/adverse effects ; CD8-Positive T-Lymphocytes ; Edema ; Tumor Microenvironment
    Chemische Substanzen Losartan (JMS50MPO89) ; Immune Checkpoint Inhibitors
    Sprache Englisch
    Erscheinungsdatum 2023-02-01
    Erscheinungsland United States
    Dokumenttyp Journal Article ; Research Support, Non-U.S. Gov't ; Research Support, N.I.H., Extramural ; Research Support, U.S. Gov't, Non-P.H.S.
    ZDB-ID 209104-5
    ISSN 1091-6490 ; 0027-8424
    ISSN (online) 1091-6490
    ISSN 0027-8424
    DOI 10.1073/pnas.2219199120
    Datenquelle MEDical Literature Analysis and Retrieval System OnLINE

    Zusatzmaterialien

    Kategorien

  3. Artikel ; Online: Blocking CXCR4 alleviates desmoplasia, increases T-lymphocyte infiltration, and improves immunotherapy in metastatic breast cancer.

    Chen, Ivy X / Chauhan, Vikash P / Posada, Jessica / Ng, Mei R / Wu, Michelle W / Adstamongkonkul, Pichet / Huang, Peigen / Lindeman, Neal / Langer, Robert / Jain, Rakesh K

    Proceedings of the National Academy of Sciences of the United States of America

    2019  Band 116, Heft 10, Seite(n) 4558–4566

    Abstract: Metastatic breast cancers (mBCs) are largely resistant to immune checkpoint blockade, but the mechanisms remain unclear. Primary breast cancers are characterized by a dense fibrotic stroma, which is considered immunosuppressive in multiple malignancies, ... ...

    Abstract Metastatic breast cancers (mBCs) are largely resistant to immune checkpoint blockade, but the mechanisms remain unclear. Primary breast cancers are characterized by a dense fibrotic stroma, which is considered immunosuppressive in multiple malignancies, but the stromal composition of breast cancer metastases and its role in immunosuppression are largely unknown. Here we show that liver and lung metastases of human breast cancers tend to be highly fibrotic, and unlike primary breast tumors, they exclude cytotoxic T lymphocytes (CTLs). Unbiased analysis of the The Cancer Genome Atlas database of human breast tumors revealed a set of genes that are associated with stromal T-lymphocyte exclusion. Among these, we focused on
    Mesh-Begriff(e) Animals ; Breast Neoplasms/pathology ; Breast Neoplasms/therapy ; Female ; Humans ; Immunotherapy ; Mice ; Neoplasm Metastasis ; Receptors, CXCR4/antagonists & inhibitors ; T-Lymphocytes/cytology ; Tumor Microenvironment
    Chemische Substanzen CXCR4 protein, human ; Receptors, CXCR4
    Sprache Englisch
    Erscheinungsdatum 2019-01-30
    Erscheinungsland United States
    Dokumenttyp Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't ; Research Support, U.S. Gov't, Non-P.H.S.
    ZDB-ID 209104-5
    ISSN 1091-6490 ; 0027-8424
    ISSN (online) 1091-6490
    ISSN 0027-8424
    DOI 10.1073/pnas.1815515116
    Datenquelle MEDical Literature Analysis and Retrieval System OnLINE

    Zusatzmaterialien

    Kategorien

  4. Artikel ; Online: A cerebellopontine angle mouse model for the investigation of tumor biology, hearing, and neurological function in NF2-related vestibular schwannoma.

    Chen, Jie / Landegger, Lukas D / Sun, Yao / Ren, Jun / Maimon, Nir / Wu, Limeng / Ng, Mei R / Chen, John W / Zhang, Na / Zhao, Yingchao / Gao, Xing / Fujita, Takeshi / Roberge, Sylvie / Huang, Peigen / Jain, Rakesh K / Plotkin, Scott R / Stankovic, Konstantina M / Xu, Lei

    Nature protocols

    2019  Band 14, Heft 2, Seite(n) 541–555

    Abstract: Neurofibromatosis type II (NF2) is a disease that lacks effective therapies. NF2 is characterized by bilateral vestibular schwannomas (VSs) that cause progressive and debilitating hearing loss, leading to social isolation and increased rates of ... ...

    Abstract Neurofibromatosis type II (NF2) is a disease that lacks effective therapies. NF2 is characterized by bilateral vestibular schwannomas (VSs) that cause progressive and debilitating hearing loss, leading to social isolation and increased rates of depression. A major limitation in NF2 basic and translational research is the lack of animal models that allow the full spectrum of research into the biology and molecular mechanisms of NF2 tumor progression, as well as the effects on neurological function. In this protocol, we describe how to inject schwannoma cells into the mouse brain cerebellopontine angle (CPA) region. We also describe how to apply state-of-the-art intravital imaging and hearing assessment techniques to study tumor growth and hearing loss. In addition, ataxia, angiogenesis, and tumor-stroma interaction assays can be applied, and the model can be used to test the efficacy of novel therapeutic approaches. By studying the disease from every angle, this model offers the potential to unravel the basic biological underpinnings of NF2 and to develop novel therapeutics to control this devastating disease. Our protocol can be adapted to study other diseases within the CPA, including meningiomas, lipomas, vascular malformations, hemangiomas, epidermoid cysts, cerebellar astrocytomas, and metastatic lesions. The entire surgical procedure takes ~45 min per mouse and allows for subsequent longitudinal imaging, as well as neurological and hearing assessment, for up to 2 months.
    Mesh-Begriff(e) Animals ; Cell Line, Tumor ; Cerebellopontine Angle/metabolism ; Cerebellopontine Angle/pathology ; Cerebellopontine Angle/surgery ; Diagnostic Imaging/methods ; Disease Models, Animal ; Gene Expression ; Genes, Reporter ; Hearing/physiology ; Hearing Loss/genetics ; Hearing Loss/pathology ; Hearing Loss/physiopathology ; Hearing Tests ; Humans ; Injections, Intraventricular ; Luciferases/genetics ; Luciferases/metabolism ; Mice ; Mice, Knockout ; Neurofibromatosis 2/genetics ; Neurofibromatosis 2/pathology ; Neurofibromatosis 2/physiopathology ; Neurofibromin 2/deficiency ; Neurofibromin 2/genetics ; Neuroma, Acoustic/genetics ; Neuroma, Acoustic/pathology ; Neuroma, Acoustic/physiopathology ; Stereotaxic Techniques
    Chemische Substanzen Neurofibromin 2 ; Luciferases (EC 1.13.12.-)
    Schlagwörter covid19
    Sprache Englisch
    Erscheinungsdatum 2019-01-07
    Erscheinungsland England
    Dokumenttyp Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't ; Research Support, U.S. Gov't, Non-P.H.S.
    ZDB-ID 2244966-8
    ISSN 1750-2799 ; 1754-2189
    ISSN (online) 1750-2799
    ISSN 1754-2189
    DOI 10.1038/s41596-018-0105-7
    Datenquelle MEDical Literature Analysis and Retrieval System OnLINE

    Zusatzmaterialien

    Kategorien

  5. Artikel ; Online: Phase II and Biomarker Study of Cabozantinib in Metastatic Triple-Negative Breast Cancer Patients.

    Tolaney, Sara M / Ziehr, David R / Guo, Hao / Ng, Mei R / Barry, William T / Higgins, Michaela J / Isakoff, Steven J / Brock, Jane E / Ivanova, Elena V / Paweletz, Cloud P / Demeo, Michelle K / Ramaiya, Nikhil H / Overmoyer, Beth A / Jain, Rakesh K / Winer, Eric P / Duda, Dan G

    The oncologist

    2016  Band 22, Heft 1, Seite(n) 25–32

    Abstract: Currently, no targeted therapies are available for metastatic triplenegative breast cancer (mTNBC). We evaluated the safety, efficacy, and biomarkers of response to cabozantinib, a multikinase inhibitor, in patients with mTNBC. We conducted a single arm ... ...

    Abstract Currently, no targeted therapies are available for metastatic triplenegative breast cancer (mTNBC). We evaluated the safety, efficacy, and biomarkers of response to cabozantinib, a multikinase inhibitor, in patients with mTNBC. We conducted a single arm phase II and biomarker study that enrolled patients with measurable mTNBC. Patients received cabozantinib (60 mg daily) on a 3-week cycle and were restaged after 6 weeks and then every 9 weeks. The primary endpoint was objective response rate. Predefined secondary endpoints included progression-free survival (PFS), toxicity, and tissue and blood circulating cell and protein biomarkers. Of 35 patients who initiated protocol therapy, 3 (9% [95% confidence interval (CI): 2, 26]) achieved a partial response (PR). Nine patients achieved stable disease (SD) for at least 15 weeks, and thus the clinical benefit rate (PR+SD) was 34% [95% CI: 19, 52]. Median PFS was 2.0 months [95% CI: 1.3, 3.3]. The most common toxicities were fatigue, diarrhea, mucositis, and palmar-plantar erythrodysesthesia. There were no grade 4 toxicities, but 12 patients (34%) required dose reduction. Two patients had TNBCs with MET amplification. During cabozantinib therapy, there were significant and durable increases in plasma placental growth factor, vascular endothelial growth factor (VEGF), VEGF-D, stromal cell-derived factor 1a, and carbonic anhydrase IX, and circulating CD3 + cells and CD8 + T lymphocytes, and decreases in plasma soluble VEGF receptor 2 and CD14+ monocytes (all p < .05). Higher baseline concentrations of soluble MET (sMET) associated with longer PFS (p = .03). In conclusion, cabozantinib showed encouraging safety and efficacy signals but did not meet the primary endpoint in pretreated mTNBC. Exploratory analyses of circulating biomarkers showed that cabozantinib induces systemic changes consistent with activation of the immune system and antiangiogenic activity, and that sMET should be further evaluated a potential biomarker of response.
    Implications for practice: Triple-negative breast cancer (TNBC)-a disease with a dearth of effective therapies-often overexpress MET, which is associated with poor clinical outcomes. However, clinical studies of agents targeting MET and VEGF pathways-alone or in combination-have shown disappointing results. This study of cabozantinib (a dual VEGFR2/MET) in metastatic TNBC, while not meeting its prespecified endpoint, showed that treatment is associated with circulating biomarker changes, and is active in a subset of patients. Furthermore, this study demonstrates that cabozantinib therapy induces a systemic increase in cytotoxic lymphocyte populations and a decrease in immunosuppressive myeloid populations. This supports the testing of combinations of cabozantinib with immunotherapy in future studies in breast cancer patients.
    Mesh-Begriff(e) Adult ; Aged ; Anilides/administration & dosage ; Anilides/adverse effects ; Biomarkers, Tumor/blood ; Disease-Free Survival ; Female ; Humans ; Kaplan-Meier Estimate ; Middle Aged ; Molecular Targeted Therapy ; Neoplasm Metastasis ; Placenta Growth Factor/blood ; Protein Kinase Inhibitors/administration & dosage ; Protein Kinase Inhibitors/adverse effects ; Proto-Oncogene Proteins c-met/antagonists & inhibitors ; Proto-Oncogene Proteins c-met/genetics ; Pyridines/administration & dosage ; Pyridines/adverse effects ; Triple Negative Breast Neoplasms/blood ; Triple Negative Breast Neoplasms/drug therapy ; Triple Negative Breast Neoplasms/pathology ; Vascular Endothelial Growth Factor D/blood ; Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors ; Vascular Endothelial Growth Factor Receptor-2/genetics
    Chemische Substanzen Anilides ; Biomarkers, Tumor ; PGF protein, human ; Protein Kinase Inhibitors ; Pyridines ; VEGFD protein, human ; Vascular Endothelial Growth Factor D ; Placenta Growth Factor (144589-93-5) ; cabozantinib (1C39JW444G) ; KDR protein, human (EC 2.7.10.1) ; MET protein, human (EC 2.7.10.1) ; Proto-Oncogene Proteins c-met (EC 2.7.10.1) ; Vascular Endothelial Growth Factor Receptor-2 (EC 2.7.10.1)
    Sprache Englisch
    Erscheinungsdatum 2016-10-27
    Erscheinungsland England
    Dokumenttyp Clinical Trial, Phase II ; Journal Article
    ZDB-ID 1409038-7
    ISSN 1549-490X ; 1083-7159
    ISSN (online) 1549-490X
    ISSN 1083-7159
    DOI 10.1634/theoncologist.2016-0229
    Datenquelle MEDical Literature Analysis and Retrieval System OnLINE

    Zusatzmaterialien

    Kategorien

  6. Artikel ; Online: CXCR4 inhibition in tumor microenvironment facilitates anti-programmed death receptor-1 immunotherapy in sorafenib-treated hepatocellular carcinoma in mice.

    Chen, Yunching / Ramjiawan, Rakesh R / Reiberger, Thomas / Ng, Mei R / Hato, Tai / Huang, Yuhui / Ochiai, Hiroki / Kitahara, Shuji / Unan, Elizabeth C / Reddy, Tejaswini P / Fan, Christopher / Huang, Peigen / Bardeesy, Nabeel / Zhu, Andrew X / Jain, Rakesh K / Duda, Dan G

    Hepatology (Baltimore, Md.)

    2015  Band 61, Heft 5, Seite(n) 1591–1602

    Abstract: Unlabelled: Sorafenib, a broad tyrosine kinase inhibitor, is the only approved systemic therapy for advanced hepatocellular carcinoma (HCC) but provides limited survival benefits. Recently, immunotherapy has emerged as a promising treatment strategy, ... ...

    Abstract Unlabelled: Sorafenib, a broad tyrosine kinase inhibitor, is the only approved systemic therapy for advanced hepatocellular carcinoma (HCC) but provides limited survival benefits. Recently, immunotherapy has emerged as a promising treatment strategy, but its role remains unclear in HCCs, which are associated with decreased cytotoxic CD8(+) T-lymphocyte infiltration in both murine and human tumors. Moreover, in mouse models after sorafenib treatment intratumoral hypoxia is increased and may fuel evasive resistance. Using orthotopic HCC models, we now show that increased hypoxia after sorafenib treatment promotes immunosuppression, characterized by increased intratumoral expression of the immune checkpoint inhibitor programmed death ligand-1 and accumulation of T-regulatory cells and M2-type macrophages. We also show that the recruitment of immunosuppressive cells is mediated in part by hypoxia-induced up-regulation of stromal cell-derived 1 alpha. Inhibition of the stromal cell-derived 1 alpha receptor (C-X-C receptor type 4 or CXCR4) using AMD3100 prevented the polarization toward an immunosuppressive microenvironment after sorafenib treatment, inhibited tumor growth, reduced lung metastasis, and improved survival. However, the combination of AMD3100 and sorafenib did not significantly change cytotoxic CD8(+) T-lymphocyte infiltration into HCC tumors and did not modify their activation status. In separate experiments, antibody blockade of the programmed death ligand-1 receptor programmed death receptor-1 (PD-1) showed antitumor effects in treatment-naive tumors in orthotopic (grafted and genetically engineered) models of HCC. However, anti-PD-1 antibody treatment had additional antitumor activity only when combined with sorafenib and AMD3100 and not when combined with sorafenib alone.
    Conclusion: Anti-PD-1 treatment can boost antitumor immune responses in HCC models; when used in combination with sorafenib, anti-PD-1 immunotherapy shows efficacy only with concomitant targeting of the hypoxic and immunosuppressive microenvironment with agents such as CXCR4 inhibitors.
    Mesh-Begriff(e) Animals ; Antineoplastic Agents/therapeutic use ; Carcinoma, Hepatocellular/drug therapy ; Carcinoma, Hepatocellular/immunology ; Humans ; Immunotherapy/methods ; Liver Neoplasms/drug therapy ; Liver Neoplasms/immunology ; Mice ; Niacinamide/analogs & derivatives ; Niacinamide/therapeutic use ; Phenylurea Compounds/therapeutic use ; Programmed Cell Death 1 Receptor/antagonists & inhibitors ; Protein Kinase Inhibitors/therapeutic use ; Receptors, CXCR4/antagonists & inhibitors ; Sorafenib ; Tumor Microenvironment/drug effects ; Tumor Microenvironment/immunology
    Chemische Substanzen Antineoplastic Agents ; CXCR4 protein, human ; Phenylurea Compounds ; Programmed Cell Death 1 Receptor ; Protein Kinase Inhibitors ; Receptors, CXCR4 ; Niacinamide (25X51I8RD4) ; Sorafenib (9ZOQ3TZI87)
    Sprache Englisch
    Erscheinungsdatum 2015-03-20
    Erscheinungsland United States
    Dokumenttyp Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 604603-4
    ISSN 1527-3350 ; 0270-9139
    ISSN (online) 1527-3350
    ISSN 0270-9139
    DOI 10.1002/hep.27665
    Datenquelle MEDical Literature Analysis and Retrieval System OnLINE

    Zusatzmaterialien

    Kategorien

  7. Artikel ; Online: Obesity-Induced Inflammation and Desmoplasia Promote Pancreatic Cancer Progression and Resistance to Chemotherapy.

    Incio, Joao / Liu, Hao / Suboj, Priya / Chin, Shan M / Chen, Ivy X / Pinter, Matthias / Ng, Mei R / Nia, Hadi T / Grahovac, Jelena / Kao, Shannon / Babykutty, Suboj / Huang, Yuhui / Jung, Keehoon / Rahbari, Nuh N / Han, Xiaoxing / Chauhan, Vikash P / Martin, John D / Kahn, Julia / Huang, Peigen /
    Desphande, Vikram / Michaelson, James / Michelakos, Theodoros P / Ferrone, Cristina R / Soares, Raquel / Boucher, Yves / Fukumura, Dai / Jain, Rakesh K

    Cancer discovery

    2016  Band 6, Heft 8, Seite(n) 852–869

    Abstract: Unlabelled: It remains unclear how obesity worsens treatment outcomes in patients with pancreatic ductal adenocarcinoma (PDAC). In normal pancreas, obesity promotes inflammation and fibrosis. We found in mouse models of PDAC that obesity also promotes ... ...

    Abstract Unlabelled: It remains unclear how obesity worsens treatment outcomes in patients with pancreatic ductal adenocarcinoma (PDAC). In normal pancreas, obesity promotes inflammation and fibrosis. We found in mouse models of PDAC that obesity also promotes desmoplasia associated with accelerated tumor growth and impaired delivery/efficacy of chemotherapeutics through reduced perfusion. Genetic and pharmacologic inhibition of angiotensin-II type-1 receptor reverses obesity-augmented desmoplasia and tumor growth and improves response to chemotherapy. Augmented activation of pancreatic stellate cells (PSC) in obesity is induced by tumor-associated neutrophils (TAN) recruited by adipocyte-secreted IL1β. PSCs further secrete IL1β, and inactivation of PSCs reduces IL1β expression and TAN recruitment. Furthermore, depletion of TANs, IL1β inhibition, or inactivation of PSCs prevents obesity-accelerated tumor growth. In patients with pancreatic cancer, we confirmed that obesity is associated with increased desmoplasia and reduced response to chemotherapy. We conclude that cross-talk between adipocytes, TANs, and PSCs exacerbates desmoplasia and promotes tumor progression in obesity.
    Significance: Considering the current obesity pandemic, unraveling the mechanisms underlying obesity-induced cancer progression is an urgent need. We found that the aggravation of desmoplasia is a key mechanism of obesity-promoted PDAC progression. Importantly, we discovered that clinically available antifibrotic/inflammatory agents can improve the treatment response of PDAC in obese hosts. Cancer Discov; 6(8); 852-69. ©2016 AACR.See related commentary by Bronte and Tortora, p. 821This article is highlighted in the In This Issue feature, p. 803.
    Mesh-Begriff(e) Adipose Tissue/metabolism ; Angiotensin II Type 1 Receptor Blockers/pharmacology ; Animals ; Body Mass Index ; Carcinoma, Pancreatic Ductal/drug therapy ; Carcinoma, Pancreatic Ductal/pathology ; Combined Modality Therapy ; Diet, High-Fat/adverse effects ; Disease Models, Animal ; Disease Progression ; Drug Resistance, Neoplasm/drug effects ; Fibrosis ; Genetic Predisposition to Disease ; Humans ; Inflammation/etiology ; Inflammation/pathology ; Interleukin-1beta/metabolism ; Male ; Mice ; Mice, Knockout ; Models, Biological ; Neutrophils/immunology ; Neutrophils/metabolism ; Obesity/complications ; Obesity/etiology ; Pancreatic Neoplasms/drug therapy ; Pancreatic Neoplasms/etiology ; Pancreatic Neoplasms/pathology ; Receptor, Angiotensin, Type 1/metabolism ; Signal Transduction/drug effects ; Tumor Burden ; Tumor Microenvironment
    Chemische Substanzen Angiotensin II Type 1 Receptor Blockers ; Interleukin-1beta ; Receptor, Angiotensin, Type 1
    Sprache Englisch
    Erscheinungsdatum 2016-05-31
    Erscheinungsland United States
    Dokumenttyp Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 2625242-9
    ISSN 2159-8290 ; 2159-8274
    ISSN (online) 2159-8290
    ISSN 2159-8274
    DOI 10.1158/2159-8290.CD-15-1177
    Datenquelle MEDical Literature Analysis and Retrieval System OnLINE

    Zusatzmaterialien

    Kategorien

Zum Seitenanfang