GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Nature Communications, Springer Science and Business Media LLC, Vol. 9, No. 1 ( 2018-12-19)
    Abstract: Myeloid cells are recruited to damaged tissues where they can resolve infections and tumor growth or stimulate wound healing and tumor progression. Recruitment of these cells is regulated by integrins, a family of adhesion receptors that includes integrin CD11b. Here we report that, unexpectedly, integrin CD11b does not regulate myeloid cell recruitment to tumors but instead controls myeloid cell polarization and tumor growth. CD11b activation promotes pro-inflammatory macrophage polarization by stimulating expression of microRNA Let7a . In contrast, inhibition of CD11b prevents Let7a expression and induces cMyc expression, leading to immune suppressive macrophage polarization, vascular maturation, and accelerated tumor growth. Pharmacological activation of CD11b with a small molecule agonist, Leukadherin 1 (LA1), promotes pro-inflammatory macrophage polarization and suppresses tumor growth in animal models of murine and human cancer. These studies identify CD11b as negative regulator of immune suppression and a target for cancer immune therapy.
    Type of Medium: Online Resource
    ISSN: 2041-1723
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2018
    detail.hit.zdb_id: 2553671-0
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Molecular Oncology, Wiley, Vol. 12, No. 4 ( 2018-04), p. 514-528
    Abstract: The interferon‐inducible transcription factor STAT1 is a tumor suppressor in various malignancies. We investigated sex‐specific STAT1 functions in colitis and colitis‐associated colorectal cancer (CRC) using mice with specific STAT1 deletion in intestinal epithelial cells (STAT1 ∆IEC ). Male but not female STAT1 ∆IEC mice were more resistant to DSS‐induced colitis than sex‐matched STAT1 flox/flox controls and displayed reduced intraepithelial infiltration of CD8 + TCRαβ + granzyme B + T cells. Moreover, DSS treatment failed to induce expression of T‐cell‐attracting chemokines in intestinal epithelial cells of male but not of female STAT1 ∆IEC mice. Application of the AOM‐DSS protocol for induction of colitis‐associated CRC resulted in increased intestinal tumor load in male but not in female STAT1 ∆IEC mice. A sex‐specific stratification of human CRC patients corroborated the data obtained in mice and revealed that reduced tumor cell‐intrinsic nuclear STAT1 protein expression is a poor prognostic factor in men but not in women. These data demonstrate that epithelial STAT1 is a male‐specific tumor suppressor in CRC of mice and humans.
    Type of Medium: Online Resource
    ISSN: 1574-7891 , 1878-0261
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2018
    detail.hit.zdb_id: 2322586-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Immunology Research Vol. 8, No. 4_Supplement ( 2020-04-01), p. A86-A86
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 8, No. 4_Supplement ( 2020-04-01), p. A86-A86
    Abstract: Introduction: Tumor-associated macrophages promote immunosuppressive microenvironment in head and neck squamous cell carcinoma (HNSCC). We previously reported that macrophage PI3-kinase γ (PI3Kγ) controls a critical switch between immune stimulation and suppression during inflammation and cancer. The aim of the present study was to investigate the effect of PI3Kγ inhibition on T-cell immune response, especially on T-cell memory and exhaustion status using mouse models of HNSCC. Materials and Methods: Wild-type (WT) or Pik3cg-/- 6- to 8-week-old male syngeneic C57Bl/6J mice were implanted with HPV+ MEER tumor cells (mouse HPV+ HNSCC cell line) by subcutaneous injection. Tumors, draining lymph nodes and spleens were isolated, then analyzed using flow cytometry or mass cytometry. Mice that completely cleared tumors were reinjected with tumor cells and tumor growth was monitored. CD90.2+ T cells or CD19+ B cells that were harvested from spleens of WT or Pik3cg-/- tumor-inoculated mice were mixed 1:1 with viable tumor cells and injected into the flanks of naive WT mice. Results: Mice lacking PI3Kγ exhibited suppressed growth of implanted HPV+ MEER tumors. The proportion of T cells, especially CD8+ T cells, significantly increased in tumors from Pik3c-/- mice. T cells from Pik3cg-/- tumors expressed significantly more granzyme B and less T-cell exhaustion markers. The proportion of CD8+ effector memory T cells significantly increased in spleens from Pik3cg-/- mice. Mice that were implanted with both tumor cells and T cells from spleens of tumor-bearing Pik3cg-/- mice exhibited significant suppression of tumor growth. All mice that had previously cleared tumors dramatically suppressed tumor growth when rechallenged with tumor cells and remained cancer-free. Conclusion: Pik3cg-/- mice showed more activated T-cell immune response and T-cell memory than WT, resulting in significant suppression of tumor growth. These results suggest that PI3Kγ-targeted therapy might enhance the activity of checkpoint inhibitors through the activation of T-cell immune response in patients with HNSCC. Citation Format: Hideyuki Takahashi, Paulina Pathria, Ryan Shepard, Ann Shih, Tiani L. Louis, Judith A. Varner. PI3Kγ inhibition activates T cell memory and relieves T cell exhaustion [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2018 Nov 27-30; Miami Beach, FL. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(4 Suppl):Abstract nr A86.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Gastroenterology, Elsevier BV, Vol. 153, No. 1 ( 2017-07), p. 178-190.e10
    Type of Medium: Online Resource
    ISSN: 0016-5085
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2017
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 13_Supplement ( 2019-07-01), p. 1513-1513
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 1513-1513
    Abstract: We previously reported that macrophage PI3-kinase γ (PI3Kγ) controls a critical switch between immune stimulation and suppression during inflammation and cancer. PI3Kγ inhibition repolarizes tumor-associated macrophages, leading to downregulation of immune suppressive factors such as Arginase and IL10 and upregulation of IL12 and other pro-inflammatory cytokines. This results in recruitment and activation of intratumoral CD8+ T cells, as well as induction of immunological memory (Kaneda et al 2016). In the present study, we investigated the effect of PI3Kγ inhibition on T cell immune responses, including T cell memory induction and T cell exhaustion, in mouse models of head and neck squamous cell carcinomas (HNSCC). We found that Pik3cg−/− mice cleared implanted HPV+ HNSCC tumors; when re-challenged with tumor cells, these mice rapidly cleared secondary tumors and remained cancer-free. The proportion of T cells, especially CD8+ T cells, significantly increased in primary tumors from Pik3cg−/− mice. These CD8+ T cells expressed significantly more granzyme B and interferon and less T cell exhaustion markers than T cells from WT animals, indicating that PI3Kγ inhibition in macrophages results in T cell activation. We found that anti-tumor activity was transferable, as adoptive transfer of splenic T cells from Pik3cg−/− mice to naive WT mice suppressed tumor growth. Accordingly, Pik3cg−/− mice with primary or secondary tumors exhibited more splenic CD62L-CD44+ CD8+ effector memory T cells than WT mice. In summary, Pik3cg−/− mice exhibit a more activated T cell immune response and T cell memory than WT mice, resulting significant suppression of tumor growth. These results suggest that PI3Kγ-targeted therapy may activate durable T cell immune responses in patients with HNSCC. Citation Format: Hideyuki Takahashi, Paulina Pathria, Ann Shih, Ryan M. Shepard, Marc A. Paradise, Judith A. Varner. PI3Kγ inhibition activates T cell memory and relieves T cell exhaustion through the reprogramming of tumor-associated macrophages [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 1513.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: OncoImmunology, Informa UK Limited, Vol. 4, No. 4 ( 2015-04-03), p. e998529-
    Type of Medium: Online Resource
    ISSN: 2162-402X
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2015
    detail.hit.zdb_id: 2645309-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    Elsevier BV ; 2019
    In:  Trends in Immunology Vol. 40, No. 4 ( 2019-04), p. 310-327
    In: Trends in Immunology, Elsevier BV, Vol. 40, No. 4 ( 2019-04), p. 310-327
    Type of Medium: Online Resource
    ISSN: 1471-4906
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2019
    detail.hit.zdb_id: 2040190-5
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 113-113
    Abstract: Macrophages play a key role in promoting tumor growth and resistance to therapy. Here we show that Tissue Resident Macrophages (TRM) as well as Bone Marrow-Derived Macrophages (BMDM) play critical but unique roles in promoting tumor growth. TRM were recently shown to originate in the yolk sac or fetal liver during embryogenesis; these cells self-maintain in post-natal tissues independent of hematopoietic stem cells. We found that BMDM are CD11b+Gr1+F4/80loCX3CR1loCCR2+and are recruited to tumors in a CCR2-dependent manner. In contrast, CD11b+Gr1-F4/80hiCX3CR1hiCCR2- TRMs accumulate in tumors independently of the trafficking receptor CCR2. Gene expression and functional studies indicate that tumor-derived TRM are highly proliferative, immune suppressive and distinct from BMDM. We show that TRM develop from c-Kit/c-KitL - dependent TRM progenitors that are abundant in tumors but not in normal tissues; purified progenitors form macrophages and potently stimulate tumor growth when adoptively transferred into mice. Tumor cells induce the expansion of TRM progenitors by secreting Stem Cell Factor (SCF/c-KitL). Notably, in vitro and in vivo proliferation of TRM progenitors and tumor growth are significantly inhibited by SCF and c-Kit inhibitors, including a novel, allosteric dual inhibitor of cKit and CDK8/19 that dramatically suppresses tumor growth by targeting both TRM and tumor cells. As cKit inhibitors synergize with other immune therapy regimens to suppress tumor growth, our studies identify cKit as a valuable target for immune therapy of solid tumors. Citation Format: Paulina Pathria, Hideyuki Takahashi, Megan Kaneda, Minya Pu, Karen Messer, Ryan M. Shepard, Tiani L. Louis, Ann Shih, Mark Bertagnolli, Wolfgang Wrasidlo, David A. Cheresh, Judith A. Varner. c-Kit-dependent tissue resident macrophage progenitors drive cancer progression [abstract] . In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 113.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. 5006-5006
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 5006-5006
    Abstract: Macrophages play a key role in promoting tumor growth and resistance to therapy. Here we show that tissue-resident as well as bone marrow-derived macrophages play critical roles in promoting tumor growth. Tissue-resident macrophages were recently shown to originate in the yolk sac or fetal liver during embryogenesis; these cells self-maintain in post-natal tissues independent of hematopoietic stem cells. Tissue-resident macrophages and bone marrow-derived macrophages rapidly accumulate in tumors where they play independent roles in promoting tumor growth. Tissue-resident macrophages are CD11b+Gr1-F4/80hiCX3CR1hiCCR2-Ki67+ cells that accumulate in tumors independently of trafficking receptors. In contrast, bone marrow-derived CD11b+Gr1+F4/80loCX3CR1loCCR2+ macrophages accumulate in tumors in an integrin α4β1/αLβ2 and CCR2 or CXCR4-dependent manner. Gene expression studies show that tumor-associated tissue-resident macrophages are highly proliferative, immune-suppressive myeloid cells that are less proangiogenic than bone marrow-derived macrophages. Our studies show that tumor cells induce the expansion of tissue-resident macrophage progenitor cells by secreting stem cell factor and mCSF. Here we identify a Kit/KitL-dependent tissue-resident macrophage progenitor that is abundant in tumors but not in normal tissues. Notably, tumor growth and colony-forming activity are significantly inhibited in mice treated with SCF and Kit inhibitors. Tumors adoptively transferred with tissue-resident macrophage progenitor cells exhibited a significant growth advantage over control tumors. Furthermore, Kit inhibitors synergize with other immune therapy regimens to suppress tumor growth. Our studies show that tissue-resident macrophage progenitors promote aggressive tumor growth that can be targeted by Kit/SCF inhibition. Citation Format: Paulina Pathria, Hideyuki Takahashi, Megan Kaneda, Judith A. Varner. Kit-dependent tissue resident macrophage progenitors drive cancer progression [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5006.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 19 ( 2019-10-01), p. 5048-5059
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 19 ( 2019-10-01), p. 5048-5059
    Abstract: Tumor-associated macrophages (TAM) are highly expressed within the tumor microenvironment of a wide range of cancers, where they exert a protumor phenotype by promoting tumor cell growth and suppressing antitumor immune function. Here, we show that TAM accumulation in human and mouse tumors correlates with tumor cell expression of integrin αvβ3, a known driver of epithelial cancer progression and drug resistance. A monoclonal antibody targeting αvβ3 (LM609) exploited the coenrichment of αvβ3 and TAMs to not only eradicate highly aggressive drug-resistant human lung and pancreas cancers in mice, but also to prevent the emergence of circulating tumor cells. Importantly, this antitumor activity in mice was eliminated following macrophage depletion. Although LM609 had no direct effect on tumor cell viability, it engaged macrophages but not natural killer (NK) cells to induce antibody-dependent cellular cytotoxicity (ADCC) of αvβ3-expressing tumor cells despite their expression of the CD47 “don't eat me” signal. In contrast to strategies designed to eliminate TAMs, these findings suggest that anti-αvβ3 represents a promising immunotherapeutic approach to redirect TAMs to serve as tumor killers for late-stage or drug-resistant cancers. Significance: Therapeutic antibodies are commonly engineered to optimize engagement of NK cells as effectors. In contrast, LM609 targets αvβ3 to suppress tumor progression and enhance drug sensitivity by exploiting TAMs to trigger ADCC.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...