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: The Prostate, Wiley, Vol. 82, No. 2 ( 2022-02), p. 169-181
    Abstract: The 2021 Coffey‐Holden Prostate Cancer Academy (CHPCA) Meeting, “Prostate Cancer Research in the 21st Century,” was held virtually, from June 24–25, 2021. Methods The CHPCA Meeting is organized by the Prostate Cancer Foundation as a unique discussion‐oriented meeting focusing on critical topics in prostate cancer research envisioned to bridge the next major advances in prostate cancer biology and treatment. The 2021 CHPCA Meeting was virtually attended by 89 investigators and included 31 talks over nine sessions. Results Major topic areas discussed at the meeting included: cancer genomics and sequencing, functional genomic approaches to studying mediators of plasticity, emerging signaling pathways in metastatic castration resistant prostate cancer, Wnt signaling biology and the challenges of targeted therapy, clonal hematopoiesis, neuroendocrine cell plasticity and antitumor immunity, cancer immunotherapy and its synergizers, and imaging the tumor microenvironment and metabolism. Discussion This meeting report summarizes the research presented at the 2021 CHPCA Meeting. We hope that publication of this knowledge will accelerate new understandings and the development of new biomarkers and treatments for prostate cancer.
    Type of Medium: Online Resource
    ISSN: 0270-4137 , 1097-0045
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2022
    detail.hit.zdb_id: 1494709-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Science Translational Medicine, American Association for the Advancement of Science (AAAS), Vol. 12, No. 530 ( 2020-02-12)
    Abstract: Solid tumors elicit a detectable immune response including the infiltration of tumor-associated macrophages (TAMs). Unfortunately, this immune response is co-opted into contributing toward tumor growth instead of preventing its progression. We seek to reestablish an antitumor immune response by selectively targeting surface receptors and endogenous signaling processes of the macrophage subtypes driving cancer progression. RP-182 is a synthetic 10-mer amphipathic analog of host defense peptides that selectively induces a conformational switch of the mannose receptor CD206 expressed on TAMs displaying an M2-like phenotype. RP-182–mediated activation of this receptor in human and murine M2-like macrophages elicits a program of endocytosis, phagosome-lysosome formation, and autophagy and reprograms M2-like TAMs to an antitumor M1-like phenotype. In syngeneic and autochthonous murine cancer models, RP-182 suppressed tumor growth, extended survival, and was an effective combination partner with chemo- or immune checkpoint therapy. Antitumor activity of RP-182 was also observed in CD206 high patient-derived xenotransplantation models. Mechanistically, via selective reduction of immunosuppressive M2-like TAMs, RP-182 improved adaptive and innate antitumor immune responses, including increased cancer cell phagocytosis by reprogrammed TAMs.
    Type of Medium: Online Resource
    ISSN: 1946-6234 , 1946-6242
    Language: English
    Publisher: American Association for the Advancement of Science (AAAS)
    Publication Date: 2020
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 9, No. 6 ( 2021-06), p. e001649-
    Abstract: Prostate cancer is the second leading cause of cancer-related death in men in the USA; death occurs when patients progress to metastatic castration-resistant prostate cancer (CRPC). Although immunotherapy with the Food and Drug Administration‐approved vaccine sipuleucel‐T, which targets prostatic acid phosphatase (PAP), extends survival for 2–4 months, the identification of new immunogenic tumor-associated antigens (TAAs) continues to be an unmet need. Methods We evaluated the differential expression profile of castration-resistant prostate epithelial cells that give rise to CRPC from mice following an androgen deprivation/repletion cycle. The expression levels of a set of androgen-responsive genes were further evaluated in prostate, brain, colon, liver, lung, skin, kidney, and salivary gland from murine and human databases. The expression of a novel prostate-restricted TAA was then validated by immunostaining of mouse tissues and analyzed in primary tumors across all human cancer types in The Cancer Genome Atlas. Finally, the immunogenicity of this TAA was evaluated in vitro and in vivo using autologous coculture assays with cells from healthy donors as well as by measuring antigen-specific antibodies in sera from patients with prostate cancer (PCa) from a neoadjuvant clinical trial. Results We identified a set of androgen-responsive genes that could serve as potential TAAs for PCa. In particular, we found transglutaminase 4 (Tgm4) to be highly expressed in prostate tumors that originate from luminal epithelial cells and only expressed at low levels in most extraprostatic tissues evaluated. Furthermore, elevated levels of TGM4 expression in primary PCa tumors correlated with unfavorable prognosis in patients. In vitro and in vivo assays confirmed the immunogenicity of TGM4. We found that activated proinflammatory effector memory CD8 and CD4 T cells were expanded by monocyte-derived dendritic cell (moDCs) pulsed with TGM4 to a greater extent than moDCs pulsed with either PAP or prostate-specific antigen (PSA), and T cells primed with TGM4-pulsed moDCs produce functional cytokines following a prime/boost regiment or in vitro stimulation. An IgG antibody response to TGM4 was detected in 30% of vaccinated patients, while fewer than 8% of vaccinated patients developed antibody responses to PSA or prostate-specific membrane antigen (PSMA). Conclusions These results suggest that TGM4 is an immunogenic, prostate-restricted antigen with the potential for further development as an immunotherapy target.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2021
    detail.hit.zdb_id: 2719863-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2939-2939
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2939-2939
    Abstract: The levels of ceramides, sphingosine and their related metabolites have been reported to play an important role in cell fate by determining the balance between apoptosis and cell survival signaling through what has been called the sphingolipid rheostat. One of sphingosine metabolic product, sphingosine-1-phosphate, has been widely proven to be related with tumor growth, resistance to apoptosis, angiogenesis, metastasis and invasiveness in several tumors. In prostate cancer, sphingosine-1-phosphate levels correlate with risk of progression to castration-resistant prostate cancer (CRPC), PSA levels, Gleason score, positive surgical margins, lymph node extension, metastatic extension and mortality. Hypoxia-inducible factors (HIFs) act as pleiotropic transcription factors which serve as major components for cancer progression, upregulation of HIFs are essential for metastasis and tumor growth once hypoxia has ensued. Several relations between HIFs and ceramides/sphingosine metabolism have been reported recently, suggesting that both mechanisms might be somehow interconnected. Despite decades of research inhibitors to HIF have been few and far between. The DNA-intercalating agent doxorubicin was identified one such inhibitor by a mechanism not yet understood. The major mechanism of doxorubicin-mediated apoptosis has been established to be induced by the transcription factor, p53. The p53 cell cycle arrest is usually through p21 which has been shown to have enhanced expression in response to sphingosine kinase 2, one of the enzymes that mediate the conversion of sphingosine to sphingosine-1-phosphate. Several studies associating chemotherapy with different drugs that attempt to modify sphingosine and ceramides metabolism have been published with some remarkable results, although never complete responses. We hypothesize that the incomplete responses may be due to an incomplete blockade of the conversion of ceramides and sphingosine towards their phosphorylated metabolites, sphingosine-1-phosphate and ceramide-1-phospate respectively. Previous work has focused on single agents to enhance chemotherapy. In this work we are interested in studying the combination of different modifiers in the metabolism of ceramides/sphingosine with doxorubicin in order to achieve a total metabolic blockade in an effort to elicit a complete clinical response for prostate cancer. Citation Format: Gonzalo Torga, Steven Mooney, Jelani C. Zarif, Kenneth J. Pienta. Induction of apoptosis in prostate cancer cells by altering cell metabolism. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2939. doi:10.1158/1538-7445.AM2015-2939
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    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 ...
  • 5
    In: Cancer Epidemiology Biomarkers & Prevention, American Association for Cancer Research (AACR), Vol. 29, No. 7 ( 2020-07), p. 1374-1380
    Type of Medium: Online Resource
    ISSN: 1055-9965 , 1538-7755
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036781-8
    detail.hit.zdb_id: 1153420-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2365-2365
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2365-2365
    Abstract: Prostate cancer is a leading cause of cancer-related deaths of men in the U.S., and in the past year, over 30,000 men died from this disease. While localized prostate cancer is highly treatable by surgical resection and radiation, cancer that has metastasized remains incurable. Alternatively activated macrophages, also known as M2-macrophages, primarily scavenge debris and in the process, promote angiogenesis and wound repair. M2-macrophages are phenotypically similar to M2-tumor associated macrophages (M2-TAMs) have been reported to associate with solid tumors such as prostate cancer to facilitate epithelial to mesenchymal transition (EMT), tumor invasiveness, metastasis, and resistance to therapy. As an invasive species within the tumor microenvironment, this makes M2-TAMs an ideal therapeutic target in prostate cancer. The purpose of this project is to develop novel therapeutics that will directly target M2-TAMs for destruction and subsequently attenuate prostate tumor growth and progression. The central hypothesis of this study is to determine if targeting of M2-TAMs using specific surface antigens will be an effective therapy for lethal prostate cancer and potentially, other cancers. Our study will first elucidate M2-TAM biology, including both a study of known M2-TAM cell surface antigen markers expressed in human prostate cancers as well as rigorous tests to target these markers for anti-tumor cell efficacy using relevant model systems. Functional experiments will then be done in vitro using primary antibodies that are bound by a saporin-conjugated secondary antibody and also, in vivo by utilizing human xenograft mouse models. We will also assess internalization of armed antibodies, M2-TAM cell death, M2-TAM functionality, and EMT status of PCa tumors after targeting. This will be followed by target validation using tissues from rapid autopsies and surgical specimens supplied by the Department of Urology at Johns Hopkins University School of Medicine. By targeting specific markers on M2-TAMs, we predict that this targeting will provide a better prognosis for patients who have been diagnosed with lethal prostate cancer. Modification of targeted ligands and drug combinations will produce a flexible platform for cancer therapy. The studies described here represent an underdeveloped frontier in cancer therapeutics and outline a method of altering the tumor immune microenvironment. If successful, antibodies used against specific M2-TAM surface markers will be constructed into antibody drug conjugates (ADCs). Citation Format: Jelani C. Zarif, James R. Hernandez, Kris F. Sachsenmeier, Robert E. Hollingsworth, Kenneth J. Pienta. Targeting M2-tumor associated macrophages (M2-TAMs) in prostate cancer. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2365. doi:10.1158/1538-7445.AM2015-2365
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    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 ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 4_Supplement ( 2012-02-06), p. A5-A5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 4_Supplement ( 2012-02-06), p. A5-A5
    Abstract: Prostate cancer is the leading cause of cancer deaths in U.S. men and an estimated 240,890 new cases will be diagnosed in 2011. Initially, the malignant tumors arising from this gland rely on the androgen receptor (AR) and its steroid hormone, dihydrotestosterone (DHT) for growth and survival. At this stage, androgen deprivation therapy (ADT) can lead to tumor regression. Unfortunately, patients typically relapse within two years and tumors no longer respond to ADT; this stage is termed castration-resistant prostate cancer (CRPC) for which there is no cure. At this stage, AR expression is either increased or mutationally activated and non-responsive to physiological levels of androgen. Several reports have shown that AR has both genomic signaling within nucleus and non-genomic signaling within the cytoplasm. Using prostate tumor cell line PC-3, expressing either wildtype or mutated AR (PC3-AR), we reported that AR genomic signaling leads to an increase in integrin α6β1 transcription and expression, which in turn increases Bcl-xL expression, a mechanism that regulates prostate cancer cell survival. However, a role for AR and/or integrin α6β1 in prostate cancer metastasis has not been examined. In addition to their role in promoting cell survival, integrins are critically important for cell movement and can activate Src. We observed that in the absence of androgen, PC-3 cells expressing AR have increased Src activity as well as enhanced cell migration and invasion, by PC3-AR, relative to cell not expressing AR, when seeded on laminin to engage integrin α6β1. Thus, we hypothesize that AR-dependent stimulation of integrin α6β1 expression is required to activate Src and promote a metastatic phenotype. We found that knock-down of AR decreased Src activity in PC3-AR cells, as well as in LNCaP cells that retain endogenous AR expression. We also found that inhibiting the expression of either AR or Src blocked the ability of cells to invade in vitro. However, blocking integrin α6 expression did not significantly block cell invasion, nor did it suppress Src activity/expression. We subsequently found that suppression of integrin α6 caused a compensatory increase in another integrin, α3β1, such that depletion of both α6 and α3 integrins was required to suppress cell invasion. To elucidate the mechanism of AR-mediated Src activation, we generated prostate tumor cells that express AR mutants defective in Src binding (ARΔPro) and will compare them to mutants defective in AR-mediated transcription (ARΔNLS). We expect that the ARΔPro mutant will not associate with or activate Src, and there will be decreased tumor cell movement. To determine how Src activity increases invasiveness, we have investigated the role of AR and Src in controlling the expression and/or activity of proteases that degrade proteins in extracellular environment to promote metastasis. The proteases we are currently investigating are matrix metalloproteinases (MMPs) 2 and 9, hepsin, matrilysin, and TMPRSS2, all of which are increased in CRPC. Thus far, we've observed an increase in MMP2 and matrilysin. Based on our in vitro studies, we predict that PC3 cells expressing wildtype or mutant AR (ARΔNLS) may be more aggressive and metastatic than the parental line in vivo. To test this hypothesis, we have created stable clones that express a tetracycline-inducible shRNA targeting either AR or Src, to assess the metastatic and survival potential of these cells in an orthotopic xenograft model. Citation Format: Jelani C. Zarif, Laura E. Lamb, Cindy K. Miranti. Androgen receptor nongenomic regulation of invasion and metastasis of prostate tumor cells [abstract]. In: Proceedings of the AACR Special Conference on Advances in Prostate Cancer Research; 2012 Feb 6-9; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2012;72(4 Suppl):Abstract nr A5.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    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 ...
  • 8
    Online Resource
    Online Resource
    Impact Journals, LLC ; 2015
    In:  Oncotarget Vol. 6, No. 9 ( 2015-03-30), p. 6862-6876
    In: Oncotarget, Impact Journals, LLC, Vol. 6, No. 9 ( 2015-03-30), p. 6862-6876
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2015
    detail.hit.zdb_id: 2560162-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 10, No. 6 ( 2022-06), p. e004400-
    Abstract: The immune suppressive tumor microenvironment (TME) that inhibits T cell infiltration, survival, and antitumor activity has posed a major challenge for developing effective immunotherapies for solid tumors. Chimeric antigen receptor (CAR)-engineered T cell therapy has shown unprecedented clinical response in treating patients with hematological malignancies, and intense investigation is underway to achieve similar responses with solid tumors. Immunologically cold tumors, including prostate cancers, are often infiltrated with abundant tumor-associated macrophages (TAMs), and infiltration of CD163 + M2 macrophages correlates with tumor progression and poor responses to immunotherapy. However, the impact of TAMs on CAR T cell activity alone and in combination with TME immunomodulators is unclear. Methods To model this in vitro, we utilized a novel co-culture system with tumor cells, CAR T cells, and polarized M1 or M2 macrophages from CD14 + peripheral blood mononuclear cells collected from healthy human donors. Tumor cell killing, T cell activation and proliferation, and macrophage phenotypes were evaluated by flow cytometry, cytokine production, RNA sequencing, and functional blockade of signaling pathways using antibodies and small molecule inhibitors. We also evaluated the TME in humanized mice following CAR T cell therapy for validation of our in vitro findings. Results We observed inhibition of CAR T cell activity with the presence of M2 macrophages, but not M1 macrophages, coinciding with a robust induction of programmed death ligand-1 (PD-L1) in M2 macrophages. We observed similar PD-L1 expression in TAMs following CAR T cell therapy in the TME of humanized mice. PD-L1, but not programmed cell death protein-1, blockade in combination with CAR T cell therapy altered phenotypes to more M1-like subsets and led to loss of CD163 + M2 macrophages via interferon-γ signaling, resulting in improved antitumor activity of CAR T cells. Conclusion This study reveals an alternative mechanism by which the combination of CAR T cells and immune checkpoint blockade modulates the immune landscape of solid tumors to enhance therapeutic efficacy of CAR T cells.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2022
    detail.hit.zdb_id: 2719863-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Cell Reports Methods, Elsevier BV, ( 2023-9), p. 100600-
    Type of Medium: Online Resource
    ISSN: 2667-2375
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2023
    detail.hit.zdb_id: 3091714-1
    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...