LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 59

Search options

  1. Article ; Online: The heterogeneity of prostate cancers lacking AR activity will require diverse treatment approaches.

    Labrecque, Mark P / Alumkal, Joshi J / Coleman, Ilsa M / Nelson, Peter S / Morrissey, Colm

    Endocrine-related cancer

    2021  Volume 28, Issue 8, Page(s) T51–T66

    Abstract: The use of androgen deprivation therapy and second-line anti-androgens in prostate cancer has led to the emergence of tumors employing multiple androgen receptor (AR)-dependent and AR-independent mechanisms to resist AR-targeted therapies in castration- ... ...

    Abstract The use of androgen deprivation therapy and second-line anti-androgens in prostate cancer has led to the emergence of tumors employing multiple androgen receptor (AR)-dependent and AR-independent mechanisms to resist AR-targeted therapies in castration-resistant prostate cancer (CRPC). While the AR signaling axis remains the cornerstone for therapeutic development in CRPC, a clearer understanding of the heterogeneous biology of CRPC tumors is needed for innovative treatment strategies. In this review, we discuss the characteristics of CRPC tumors that lack AR activity and the temporal and spatial considerations for the conversion of an AR-dependent to an AR-independent tumor type. We describe the more prevalent treatment-emergent phenotypes arising in the CRPC disease continuum, including amphicrine, AR-low, double-negative, neuroendocrine and small cell phenotypes. We discuss the association between the loss of AR activity and tumor plasticity with a focus on the roles of transcription factors like SOX2, DNA methylation, alternative splicing, and the activity of epigenetic modifiers like EZH2, BRD4, LSD1, and the nBAF complex in conversion to a neuroendocrine or small cell phenotype in CRPC. We hypothesize that only a subset of CRPC tumors have the propensity for tumor plasticity and conversion to the neuroendocrine phenotype and outline how we might target these plastic and emergent phenotypes in CRPC. In conclusion, we assess the current and future avenues for treatment and determine that the heterogeneity of CRPCs lacking AR activity will require diverse treatment approaches.
    MeSH term(s) Androgen Antagonists/therapeutic use ; Cell Cycle Proteins/genetics ; Cell Line, Tumor ; Gene Expression Regulation, Neoplastic ; Humans ; Male ; Nuclear Proteins/genetics ; Prostatic Neoplasms, Castration-Resistant/drug therapy ; Prostatic Neoplasms, Castration-Resistant/genetics ; Prostatic Neoplasms, Castration-Resistant/pathology ; Receptors, Androgen/genetics ; Receptors, Androgen/metabolism ; Signal Transduction ; Transcription Factors/genetics
    Chemical Substances Androgen Antagonists ; BRD4 protein, human ; Cell Cycle Proteins ; Nuclear Proteins ; Receptors, Androgen ; Transcription Factors
    Language English
    Publishing date 2021-07-15
    Publishing country England
    Document type 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. ; Review
    ZDB-ID 1218450-0
    ISSN 1479-6821 ; 1351-0088
    ISSN (online) 1479-6821
    ISSN 1351-0088
    DOI 10.1530/ERC-21-0002
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: Third generation quinoline-3-carboxamide transcriptional disrupter of HDAC4, HIF-1α, and MEF-2 signaling for metastatic castration-resistant prostate cancer.

    Isaacs, John T / Dalrymple, Susan L / Antony, Lizamma / Marc Rosen, David / Coleman, Ilsa M / Nelson, Peter S / Kostova, Maya / Murray, Iain A / Perdew, Gary H / Denmeade, Samuel R / Akinboye, Emmanuel S / Brennen, W Nathaniel

    The Prostate

    2023  Volume 83, Issue 15, Page(s) 1470–1493

    Abstract: Background: The quinoline-3-carboxamide, Tasquinimod (TasQ), is orally active as a maintenance therapy with an on-target mechanism-of-action via allosteric binding to HDAC4. This prevents formation of the HDAC4/NCoR1/HDAC3 complex, disrupting HIF-1α ... ...

    Abstract Background: The quinoline-3-carboxamide, Tasquinimod (TasQ), is orally active as a maintenance therapy with an on-target mechanism-of-action via allosteric binding to HDAC4. This prevents formation of the HDAC4/NCoR1/HDAC3 complex, disrupting HIF-1α transcriptional activation and repressing MEF-2 target genes needed for adaptive survival signaling in the compromised tumor micro environment. In phase 3 clinical testing against metastatic castration-resistant prostate cancer(mCRPC), TasQ (1 mg/day) increased time-to-progression, but not overall survival.
    Methods: TasQ analogs were chemically synthesized and tested for activity compared to the parental compound. These included HDAC4 enzymatic assays, qRT-PCR and western blot analyses of gene and protein expression following treatment, in vitro and in vivo efficacy against multiple prostate cancer models including PDXs, pharmacokinetic analyses,AHR binding and agonist assays, SPR analyses of binding to HDAC4 and NCoR1, RNAseq analysis of in vivo tumors, 3D endothelial sprouting assays, and a targeted kinase screen. Genetic knockout or knockdown controls were used when appropriate.
    Results: Here, we document that, on this regimen (1 mg/day), TasQ blood levels are 10-fold lower than the optimal concentration (≥2 μM) needed for anticancer activity, suggesting higher daily doses are needed. Unfortunately, we also demonstrate that TasQ is an arylhydrocarbon receptor (AHR) agonist, which binds with an EC50 of 1 μM to produce unwanted off-target side effects. Therefore, we screened a library of TasQ analogsto maximize on-target versus off-target activity. Using this approach, we identified ESATA-20, which has ~10-fold lower AHR agonism and 5-fold greater potency against prostate cancer patient-derived xenografts.
    Conclusion: This increased therapeuticindex nominates ESATA-20 as a lead candidate forclinical development as an orally active third generation quinoline-3-carboxamide analog thatretains its on-target ability to disrupt HDAC4/HIF-1α/MEF-2-dependent adaptive survival signaling in the compromisedtumor microenvironment found in mCRPC.
    MeSH term(s) Male ; Humans ; Prostatic Neoplasms, Castration-Resistant/drug therapy ; Prostatic Neoplasms, Castration-Resistant/genetics ; Prostatic Neoplasms, Castration-Resistant/pathology ; Blotting, Western ; Cell Line, Tumor ; Tumor Microenvironment ; Histone Deacetylases/metabolism ; Repressor Proteins/metabolism
    Chemical Substances quinoline-3-carboxamide ; HDAC4 protein, human (EC 3.5.1.98) ; Histone Deacetylases (EC 3.5.1.98) ; Repressor Proteins
    Language English
    Publishing date 2023-08-09
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, U.S. Gov't, Non-P.H.S. ; Research Support, Non-U.S. Gov't
    ZDB-ID 604707-5
    ISSN 1097-0045 ; 0270-4137
    ISSN (online) 1097-0045
    ISSN 0270-4137
    DOI 10.1002/pros.24606
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: Supraphysiological Androgens Promote the Tumor Suppressive Activity of the Androgen Receptor through cMYC Repression and Recruitment of the DREAM Complex.

    Nyquist, Michael D / Coleman, Ilsa M / Lucas, Jared M / Li, Dapei / Hanratty, Brian / Meade, Hannah / Mostaghel, Elahe A / Plymate, Stephen R / Corey, Eva / Haffner, Michael C / Nelson, Peter S

    Cancer research

    2023  Volume 83, Issue 17, Page(s) 2938–2951

    Abstract: The androgen receptor (AR) pathway regulates key cell survival programs in prostate epithelium. The AR represents a near-universal driver and therapeutic vulnerability in metastatic prostate cancer, and targeting AR has a remarkable therapeutic index. ... ...

    Abstract The androgen receptor (AR) pathway regulates key cell survival programs in prostate epithelium. The AR represents a near-universal driver and therapeutic vulnerability in metastatic prostate cancer, and targeting AR has a remarkable therapeutic index. Though most approaches directed toward AR focus on inhibiting AR signaling, laboratory and now clinical data have shown that high dose, supraphysiological androgen treatment (SPA) results in growth repression and improved outcomes in subsets of patients with prostate cancer. A better understanding of the mechanisms contributing to SPA response and resistance could help guide patient selection and combination therapies to improve efficacy. To characterize SPA signaling, we integrated metrics of gene expression changes induced by SPA together with cistrome data and protein-interactomes. These analyses indicated that the dimerization partner, RB-like, E2F, and multivulval class B (DREAM) complex mediates growth repression and downregulation of E2F targets in response to SPA. Notably, prostate cancers with complete genomic loss of RB1 responded to SPA treatment, whereas loss of DREAM complex components such as RBL1/2 promoted resistance. Overexpression of MYC resulted in complete resistance to SPA and attenuated the SPA/AR-mediated repression of E2F target genes. These findings support a model of SPA-mediated growth repression that relies on the negative regulation of MYC by AR leading to repression of E2F1 signaling via the DREAM complex. The integrity of MYC signaling and DREAM complex assembly may consequently serve as determinants of SPA responses and as pathways mediating SPA resistance.
    Significance: Determining the molecular pathways by which supraphysiological androgens promote growth arrest and treatment responses in prostate cancer provides opportunities for biomarker-selected clinical trials and the development of strategies to augment responses.
    MeSH term(s) Male ; Humans ; Androgens/metabolism ; Receptors, Androgen/genetics ; Receptors, Androgen/metabolism ; Proto-Oncogene Proteins c-myc/genetics ; Proto-Oncogene Proteins c-myc/metabolism ; Signal Transduction ; Prostatic Neoplasms/drug therapy ; Prostatic Neoplasms/genetics ; Prostatic Neoplasms/metabolism ; Cell Line, Tumor
    Chemical Substances Androgens ; Receptors, Androgen ; Proto-Oncogene Proteins c-myc
    Language English
    Publishing date 2023-06-21
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 1432-1
    ISSN 1538-7445 ; 0008-5472
    ISSN (online) 1538-7445
    ISSN 0008-5472
    DOI 10.1158/0008-5472.CAN-22-2613
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Cabozantinib can block growth of neuroendocrine prostate cancer patient-derived xenografts by disrupting tumor vasculature.

    Labrecque, Mark P / Brown, Lisha G / Coleman, Ilsa M / Nguyen, Holly M / Lin, Daniel W / Corey, Eva / Nelson, Peter S / Morrissey, Colm

    PloS one

    2021  Volume 16, Issue 1, Page(s) e0245602

    Abstract: With the advent of potent second-line anti-androgen therapy, we and others have observed an increased incidence of androgen receptor (AR)-null small cell or neuroendocrine prostate cancer (SCNPC) in metastatic castration-resistant prostate cancer (mCRPC). ...

    Abstract With the advent of potent second-line anti-androgen therapy, we and others have observed an increased incidence of androgen receptor (AR)-null small cell or neuroendocrine prostate cancer (SCNPC) in metastatic castration-resistant prostate cancer (mCRPC). Our study was designed to determine the effect of cabozantinib, a multi-targeted tyrosine kinase inhibitor that inhibits VEGFR2, MET and RET on SCNPC. Transcriptome analysis of the University of Washington rapid autopsy and SU2C mCRPC datasets revealed upregulated MET and RET expression in SCNPCs relative to adenocarcinomas. Additionally, increased MET expression correlated with attenuated AR expression and activity. In vitro treatment of SCNPC patient-derived xenograft (PDX) cells with the MET inhibitor AMG-337 had no impact on cell viability in LuCaP 93 (MET+/RET+) and LuCaP 173.1 (MET-/RET-), whereas cabozantinib decreased cell viability of LuCaP 93, but not LuCaP 173.1. Notably, MET+/RET+ LuCaP 93 and MET-/RET- LuCaP 173.1 tumor volumes were significantly decreased with cabozantinib treatment in vivo, and this activity was independent of MET or RET expression in LuCaP 173.1. Tissue analysis indicated that cabozantinib did not inhibit tumor cell proliferation (Ki67), but significantly decreased microvessel density (CD31) and increased hypoxic stress and glycolysis (HK2) in LuCaP 93 and LuCaP 173.1 tumors. RNA-Seq and gene set enrichment analysis revealed that hypoxia and glycolysis pathways were increased in cabozantinib-treated tumors relative to control tumors. Our data suggest that the most likely mechanism of cabozantinib-mediated tumor growth suppression in SCNPC PDX models is through disruption of the tumor vasculature. Thus, cabozantinib may represent a potential therapy for patients with metastatic disease in tumor phenotypes that have a significant dependence on the tumor vasculature for survival and proliferation.
    MeSH term(s) Anilides/pharmacology ; Animals ; Carcinoma, Neuroendocrine/blood supply ; Carcinoma, Neuroendocrine/drug therapy ; Carcinoma, Neuroendocrine/metabolism ; Carcinoma, Neuroendocrine/pathology ; Cell Line, Tumor ; Humans ; Male ; Mice ; Mice, SCID ; Neovascularization, Pathologic/drug therapy ; Neovascularization, Pathologic/metabolism ; Prostatic Neoplasms/blood supply ; Prostatic Neoplasms/drug therapy ; Prostatic Neoplasms/metabolism ; Proto-Oncogene Proteins c-met/metabolism ; Proto-Oncogene Proteins c-ret/metabolism ; Pyridines/pharmacology ; Xenograft Model Antitumor Assays
    Chemical Substances Anilides ; Pyridines ; cabozantinib (1C39JW444G) ; MET protein, human (EC 2.7.10.1) ; Proto-Oncogene Proteins c-met (EC 2.7.10.1) ; Proto-Oncogene Proteins c-ret (EC 2.7.10.1) ; RET protein, human (EC 2.7.10.1)
    Language English
    Publishing date 2021-01-20
    Publishing country United States
    Document type 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.
    ISSN 1932-6203
    ISSN (online) 1932-6203
    DOI 10.1371/journal.pone.0245602
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: Targeting the fibroblast growth factor pathway in molecular subtypes of castration-resistant prostate cancer.

    Labrecque, Mark P / Brown, Lisha G / Coleman, Ilsa M / Nguyen, Holly M / Dalrymple, Susan / Brennen, W Nathaniel / Isaacs, John T / Li, Dapei / Lakely, Bryce / DeLucia, Diana C / Lee, John K / Schweizer, Michael T / Lin, Daniel W / Corey, Eva / Nelson, Peter S / Morrissey, Colm

    The Prostate

    2023  Volume 84, Issue 1, Page(s) 100–110

    Abstract: Background: Androgen receptor (AR) pathway inhibition remains the cornerstone for prostate cancer therapies. However, castration-resistant prostate cancer (CRPC) tumors can resist AR signaling inhibitors through AR amplification and AR splice variants ... ...

    Abstract Background: Androgen receptor (AR) pathway inhibition remains the cornerstone for prostate cancer therapies. However, castration-resistant prostate cancer (CRPC) tumors can resist AR signaling inhibitors through AR amplification and AR splice variants in AR-positive CRPC (ARPC), and conversion to AR-null phenotypes, such as double-negative prostate cancer (DNPC) and small cell or neuroendocrine prostate cancer (SCNPC). We have shown previously that DNPC can bypass AR-dependence through fibroblast growth factor receptor (FGFR) signaling. However, the role of the FGFR pathway in other CRPC phenotypes has not been elucidated.
    Methods: RNA-Seq analysis was conducted on patient metastases, LuCaP patient-derived xenograft (PDX) models, and CRPC cell lines. Cell lines (C4-2B, VCaP, and 22Rv1) and ex vivo LuCaP PDX tumor cells were treated with enzalutamide (ENZA) and FGFR inhibitors (FGFRi) alone or in combination and sensitivity was determined using cell viability assays. In vivo efficacy of FGFRi in ARPC, DNPC, and SCNPC were evaluated using PDX models.
    Results: RNA-Seq analysis of FGFR signaling in metastatic specimens, LuCaP PDX models, and CRPC cell lines revealed significant FGF pathway activation in AR-low PC (ARLPC), DNPC, and SCNPC tumors. In vitro/ex vivo analysis of erdafitinib and CH5183284 demonstrated robust and moderate growth suppression of ARPC, respectively. In vivo studies using four ARPC PDX models showed that combination ENZA and CH5183284 significantly suppressed tumor growth. Additional in vivo studies using four ARPC PDX models revealed that erdafitinib monotherapy was as effective as ENZA in suppressing tumor growth, and there was limited combination benefit. Furthermore, two of three DNPC models and two of four SCNPC models responded to CH5183284 monotherapy, suggesting FGFRi responses were model dependent. RNA-Seq and gene set enrichment analysis of end-of-study ARPC tumors treated with FGFRi displayed decreased expression of E2F and MYC target genes and suppressed G2M checkpoint genes, whereas end-of-study SCNPC tumors had heterogeneous transcriptional responses.
    Conclusions: Although FGFRi treatments suppressed tumor growth across CRPC phenotypes, our analyses did not identify a single pathway or biomarker that would identify tumor response to FGFRi. This is very likely due to the array of FGFR1-4 expression and tumor phenotypes present in CRPC. Nevertheless, our data nominate the FGFR pathway as a clinically actionable target that promotes tumor growth in diverse phenotypes of treatment-refractory metastatic CRPC.
    MeSH term(s) Male ; Humans ; Prostatic Neoplasms, Castration-Resistant/drug therapy ; Prostatic Neoplasms, Castration-Resistant/genetics ; Prostatic Neoplasms, Castration-Resistant/metabolism ; Fibroblast Growth Factors/genetics ; Fibroblast Growth Factors/pharmacology ; Receptors, Androgen/genetics ; Receptors, Androgen/metabolism ; Androgen Receptor Antagonists/pharmacology ; Androgens/pharmacology ; Signal Transduction ; Cell Line, Tumor ; Nitriles/pharmacology
    Chemical Substances Fibroblast Growth Factors (62031-54-3) ; Receptors, Androgen ; Androgen Receptor Antagonists ; Androgens ; Nitriles
    Language English
    Publishing date 2023-10-05
    Publishing country United States
    Document type Journal Article
    ZDB-ID 604707-5
    ISSN 1097-0045 ; 0270-4137
    ISSN (online) 1097-0045
    ISSN 0270-4137
    DOI 10.1002/pros.24630
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: Therapeutic Implications for Intrinsic Phenotype Classification of Metastatic Castration-Resistant Prostate Cancer.

    Coleman, Ilsa M / DeSarkar, Navonil / Morrissey, Colm / Xin, Li / Roudier, Martine P / Sayar, Erolcan / Li, Dapei / Corey, Eva / Haffner, Michael C / Nelson, Peter S

    Clinical cancer research : an official journal of the American Association for Cancer Research

    2022  Volume 28, Issue 14, Page(s) 3127–3140

    Abstract: Purpose: To determine whether metastatic castration-resistant prostate cancers (mCRPC) partition into molecular phenotypes corresponding to intrinsic differentiation states and ascertain whether these subtypes exhibit specific druggable features and ... ...

    Abstract Purpose: To determine whether metastatic castration-resistant prostate cancers (mCRPC) partition into molecular phenotypes corresponding to intrinsic differentiation states and ascertain whether these subtypes exhibit specific druggable features and associate with treatment outcomes.
    Experimental design: We used RNAseq, digital spatial profiling, and histological assessments from metastatic biopsies and patient-derived xenografts to segregate mCRPCs into subtypes defined by the PAM50 breast cancer classification algorithm. Subtype associations with treatment responses in preclinical models and patients were determined.
    Results: Using the PAM50 algorithm, we partitioned 270 mCRPC tumors into LumA (42%), LumB (24%), and Basal (34%) subtypes with classification largely driven by proliferation rates and androgen receptor (AR) activity. Most neuroendocrine tumors classified as Basal. Pathways enriched in the LumA subtype include TGFß and NOTCH signaling. LumB subtype tumors were notable for elevated MYC activity. Basal subtype tumors exhibited elevated IL6-STAT3 signaling and features of adult stem cell states. In patients where multiple tumors were evaluated, the majority had concordant PAM50 subtype determination, though a subset exhibited marked inter- and intratumor heterogeneity, including divergent classifications between primary and metastatic sites. In preclinical models, LumA subtype tumors were highly responsive to androgen deprivation and docetaxel chemotherapy whereas Basal tumors were largely resistant. In clinical cohorts patients with Basal subtype tumors demonstrated a shorter time on treatment with AR signaling inhibitors and docetaxel relative to patients with luminal subtypes.
    Conclusions: Subtyping of mCRPC based on cell differentiation states has potential clinical utility for identifying patients with divergent expression of treatment targets and responses to systemic therapy.
    MeSH term(s) Androgen Antagonists/pharmacology ; Androgen Antagonists/therapeutic use ; Breast Neoplasms/pathology ; Docetaxel/therapeutic use ; Humans ; Male ; Phenotype ; Prostatic Neoplasms, Castration-Resistant/drug therapy ; Prostatic Neoplasms, Castration-Resistant/genetics ; Prostatic Neoplasms, Castration-Resistant/pathology
    Chemical Substances Androgen Antagonists ; Docetaxel (15H5577CQD)
    Language English
    Publishing date 2022-04-28
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Research Support, N.I.H., Extramural
    ZDB-ID 1225457-5
    ISSN 1557-3265 ; 1078-0432
    ISSN (online) 1557-3265
    ISSN 1078-0432
    DOI 10.1158/1078-0432.CCR-21-4289
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: Imaging Fibroblast Activation Protein Alpha Improves Diagnosis of Metastatic Prostate Cancer with Positron Emission Tomography.

    Hintz, Hallie M / Gallant, Joseph P / Vander Griend, Donald J / Coleman, Ilsa M / Nelson, Peter S / LeBeau, Aaron M

    Clinical cancer research : an official journal of the American Association for Cancer Research

    2020  Volume 26, Issue 18, Page(s) 4882–4891

    Abstract: Purpose: Metastatic castration-resistant prostate cancer (mCRPC) is a lethal, heterogeneous disease with few therapeutic strategies that significantly prolong survival. Innovative therapies for mCRPC are needed; however, the development of new therapies ...

    Abstract Purpose: Metastatic castration-resistant prostate cancer (mCRPC) is a lethal, heterogeneous disease with few therapeutic strategies that significantly prolong survival. Innovative therapies for mCRPC are needed; however, the development of new therapies relies on accurate imaging to assess metastasis and monitor response. Standard imaging modalities for prostate cancer require improvement and there remains a need for selective and sensitive imaging probes that can be widely used in patients with mCRPC.
    Experimental design: We evaluated the transmembrane protease fibroblast activation protein alpha (FAP) as a targetable cell surface antigen for mCRPC. Genomic and IHC analyses were performed to investigate FAP expression in prostate cancer. Our FAP-targeted antibody imaging probe, [
    Results: Analysis of patient data documented FAP overexpression in metastatic disease across tumor subtypes. PET imaging with [
    Conclusions: Our study reveals FAP as a target for imaging the tumor microenvironment of prostate cancer. Validation of [
    MeSH term(s) Animals ; Cell Line, Tumor ; Endopeptidases/metabolism ; HEK293 Cells ; Humans ; Immunoconjugates/administration & dosage ; Immunoconjugates/pharmacokinetics ; Male ; Membrane Proteins/antagonists & inhibitors ; Membrane Proteins/metabolism ; Mice ; Molecular Imaging/methods ; Positron Emission Tomography Computed Tomography/methods ; Prostate/diagnostic imaging ; Prostate/pathology ; Prostatic Neoplasms, Castration-Resistant/diagnosis ; Prostatic Neoplasms, Castration-Resistant/pathology ; RNA-Seq ; Radioisotopes/administration & dosage ; Radioisotopes/pharmacokinetics ; Radiopharmaceuticals/administration & dosage ; Radiopharmaceuticals/pharmacokinetics ; Tissue Distribution ; Tumor Microenvironment ; X-Ray Microtomography ; Zirconium/administration & dosage ; Zirconium/pharmacokinetics
    Chemical Substances Immunoconjugates ; Membrane Proteins ; Radioisotopes ; Radiopharmaceuticals ; Zirconium (C6V6S92N3C) ; Endopeptidases (EC 3.4.-) ; fibroblast activation protein alpha (EC 3.4.21.-) ; Zirconium-89 (NTM296JU95)
    Language English
    Publishing date 2020-07-07
    Publishing country United States
    Document type Journal Article ; Observational Study ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 1225457-5
    ISSN 1557-3265 ; 1078-0432
    ISSN (online) 1557-3265
    ISSN 1078-0432
    DOI 10.1158/1078-0432.CCR-20-1358
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: A comparison of prostate cancer cell transcriptomes in 2D monoculture vs 3D xenografts identify consistent gene expression alterations associated with tumor microenvironments.

    Brady, Lauren / Gil da Costa, Rui M / Coleman, Ilsa M / Matson, Clinton K / Risk, Michael C / Coleman, Roger T / Nelson, Peter S

    The Prostate

    2020  Volume 80, Issue 6, Page(s) 491–499

    Abstract: Background: Prostate cancer (PC) research has relied heavily on patient-derived cell lines, which may be used for in vitro (two-dimensional [2D]) studies or cultivated as three-dimensional (3D) xenografts in mice. These approaches are likely to have ... ...

    Abstract Background: Prostate cancer (PC) research has relied heavily on patient-derived cell lines, which may be used for in vitro (two-dimensional [2D]) studies or cultivated as three-dimensional (3D) xenografts in mice. These approaches are likely to have differential impacts on cell phenotypes, with implications for experimental outcomes. Therefore, defining and comparing the transcriptional signatures associated with 2D and 3D approaches may be useful for designing experiments and interpreting research results.
    Methods: In this study, LNCaP, VCaP, and 22Rv1 human PC cells were either cultivated in monolayers or as xenografts in NOD SCID mice, and their gene transcription profiles were quantitated and compared using microarray and real-time polymerase chain reaction techniques. Immunohistochemistry was used to evaluate protein expression in cancer cell xenografts.
    Results: Comparisons of gene expression profiles of tumor cells grown in 2D vs 3D environments identified gene sets featuring similar expression patterns in all three cancer cell lines and unique transcriptional signatures associated with 3D vs 2D growth. Pathways related to cell-cell interactions, differentiation, and the extracellular matrix were enriched in 3D conditions. Immunohistochemical analyses confirmed that gene upregulation in xenografts occurred in implanted cancer cells and not in mouse stromal cells. Cultivating cells in vitro in the presence of mouse, rather than bovine serum failed to elicit the gene transcription profile observed in xenografts, further supporting the hypothesis that this profile reflects 3D growth and enhanced microenvironmental interactions, rather than exposure to species-specific serum factors.
    Conclusions: Overall, these findings define the expression profiles observed in PC cells cultivated in 2D monolayers and in 3D xenografts, highlighting differentially regulated pathways in each setting and providing information for interpreting research results in model systems.
    MeSH term(s) Animals ; Cell Line, Tumor ; Gene Expression Regulation, Neoplastic ; Genome, Human ; Heterografts ; Humans ; Immunohistochemistry ; Male ; Mice ; Mice, Inbred NOD ; Mice, SCID ; Oligonucleotide Array Sequence Analysis/methods ; Prostatic Neoplasms/genetics ; Prostatic Neoplasms/pathology ; Transcriptome ; Tumor Cells, Cultured ; Tumor Microenvironment/genetics
    Language English
    Publishing date 2020-02-18
    Publishing country United States
    Document type Comparative Study ; Journal Article ; Research Support, N.I.H., Extramural ; Research Support, U.S. Gov't, Non-P.H.S.
    ZDB-ID 604707-5
    ISSN 1097-0045 ; 0270-4137
    ISSN (online) 1097-0045
    ISSN 0270-4137
    DOI 10.1002/pros.23963
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: Multiplexed functional genomic analysis of 5' untranslated region mutations across the spectrum of prostate cancer.

    Lim, Yiting / Arora, Sonali / Schuster, Samantha L / Corey, Lukas / Fitzgibbon, Matthew / Wladyka, Cynthia L / Wu, Xiaoying / Coleman, Ilsa M / Delrow, Jeffrey J / Corey, Eva / True, Lawrence D / Nelson, Peter S / Ha, Gavin / Hsieh, Andrew C

    Nature communications

    2021  Volume 12, Issue 1, Page(s) 4217

    Abstract: The functional consequences of genetic variants within 5' untranslated regions (UTRs) on a genome-wide scale are poorly understood in disease. Here we develop a high-throughput multi-layer functional genomics method called PLUMAGE (Pooled full-length UTR ...

    Abstract The functional consequences of genetic variants within 5' untranslated regions (UTRs) on a genome-wide scale are poorly understood in disease. Here we develop a high-throughput multi-layer functional genomics method called PLUMAGE (Pooled full-length UTR Multiplex Assay on Gene Expression) to quantify the molecular consequences of somatic 5' UTR mutations in human prostate cancer. We show that 5' UTR mutations can control transcript levels and mRNA translation rates through the creation of DNA binding elements or RNA-based cis-regulatory motifs. We discover that point mutations can simultaneously impact transcript and translation levels of the same gene. We provide evidence that functional 5' UTR mutations in the MAP kinase signaling pathway can upregulate pathway-specific gene expression and are associated with clinical outcomes. Our study reveals the diverse mechanisms by which the mutational landscape of 5' UTRs can co-opt gene expression and demonstrates that single nucleotide alterations within 5' UTRs are functional in cancer.
    MeSH term(s) 5' Untranslated Regions/genetics ; Cell Line, Tumor ; DNA Mutational Analysis/methods ; Gene Expression Regulation, Neoplastic ; Genomics/methods ; HEK293 Cells ; High-Throughput Screening Assays ; Humans ; Male ; Point Mutation ; Prostate/pathology ; Prostatic Neoplasms/genetics ; Prostatic Neoplasms/pathology ; Protein Biosynthesis/genetics ; RNA-Seq
    Chemical Substances 5' Untranslated Regions
    Language English
    Publishing date 2021-07-09
    Publishing country England
    Document type Journal Article ; Observational Study ; 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 2553671-0
    ISSN 2041-1723 ; 2041-1723
    ISSN (online) 2041-1723
    ISSN 2041-1723
    DOI 10.1038/s41467-021-24445-6
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article ; Online: Defining the challenges and opportunities for using patient-derived models in prostate cancer research.

    Brennen, W Nathaniel / Le Magnen, Clémentine / Karkampouna, Sofia / Anselmino, Nicolas / Bock, Nathalie / Choo, Nicholas / Clark, Ashlee K / Coleman, Ilsa M / Dolgos, Robin / Ferguson, Alison M / Goode, David L / Krutihof-de Julio, Marianna / Navone, Nora M / Nelson, Peter S / O'Neill, Edward / Porter, Laura H / Ranasinghe, Weranja / Sunada, Takuro / Williams, Elizabeth D /
    Butler, Lisa M / Corey, Eva / van Weerden, Wytske M / Taylor, Renea A / Risbridger, Gail P / Lawrence, Mitchell G

    The Prostate

    2024  Volume 84, Issue 7, Page(s) 623–635

    Abstract: Background: There are relatively few widely used models of prostate cancer compared to other common malignancies. This impedes translational prostate cancer research because the range of models does not reflect the diversity of disease seen in clinical ... ...

    Abstract Background: There are relatively few widely used models of prostate cancer compared to other common malignancies. This impedes translational prostate cancer research because the range of models does not reflect the diversity of disease seen in clinical practice. In response to this challenge, research laboratories around the world have been developing new patient-derived models of prostate cancer, including xenografts, organoids, and tumor explants.
    Methods: In May 2023, we held a workshop at the Monash University Prato Campus for researchers with expertise in establishing and using a variety of patient-derived models of prostate cancer. This review summarizes our collective ideas on how patient-derived models are currently being used, the common challenges, and future opportunities for maximizing their usefulness in prostate cancer research.
    Results: An increasing number of patient-derived models for prostate cancer are being developed. Despite their individual limitations and varying success rates, these models are valuable resources for exploring new concepts in prostate cancer biology and for preclinical testing of potential treatments. Here we focus on the need for larger collections of models that represent the changing treatment landscape of prostate cancer, robust readouts for preclinical testing, improved in vitro culture conditions, and integration of the tumor microenvironment. Additional priorities include ensuring model reproducibility, standardization, and replication, and streamlining the exchange of models and data sets among research groups.
    Conclusions: There are several opportunities to maximize the impact of patient-derived models on prostate cancer research. We must develop large, diverse and accessible cohorts of models and more sophisticated methods for emulating the intricacy of patient tumors. In this way, we can use the samples that are generously donated by patients to advance the outcomes of patients in the future.
    MeSH term(s) Male ; Humans ; Reproducibility of Results ; Prostatic Neoplasms/therapy ; Prostatic Neoplasms/pathology ; Prostate/pathology ; Organoids/pathology ; Heterografts ; Tumor Microenvironment
    Language English
    Publishing date 2024-03-07
    Publishing country United States
    Document type Journal Article ; Review
    ZDB-ID 604707-5
    ISSN 1097-0045 ; 0270-4137
    ISSN (online) 1097-0045
    ISSN 0270-4137
    DOI 10.1002/pros.24682
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top