Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Susan L. Dalrymple is active.

Publication


Featured researches published by Susan L. Dalrymple.


Cancer Research | 2006

Low-Calcium Serum-Free Defined Medium Selects for Growth of Normal Prostatic Epithelial Stem Cells

Ivan V. Litvinov; Donald J. Vander Griend; Yi Xu; Lizamma Antony; Susan L. Dalrymple; John T. Isaacs

Stage-specific differentiation markers were used to evaluate the cellular composition and the origin of nonimmortalized (PrEC) and immortalized (PZ-HPV7, CA-HPV10, RWPE-1, and 957E/hTERT) human prostate cell lines. These studies documented that immortalized and nonimmortalized prostate epithelial cells established and maintained in low (i.e., <300 micromol/L) Ca(2+) serum-free defined (SFD) medium were all derived from normal nonmalignant prostate tissues and contain CD133(+)/ABCG2(+)/alpha(2)beta(1)(Hi)/p63(-)/PSCA(-)/AR(-)/PSA(-) prostate stem cells. In these cultures, prostate stem cells are able to self-renew and generate two distinct cell lineages: the minor proliferatively quiescent neuroendocrine lineage and the major transit-amplifying cell lineage. Subsequently, CD133(-)/ABCG2(-)/alpha(2)beta(1)(Hi)/p63(+)/PSCA(-)/AR(-)/PSA(-) transit-amplifying cells proliferate frequently and eventually mature into proliferatively quiescent CD133(-)/ABCG2(-)/alpha(2)beta(1)(Lo)/p63(-)/PSCA(+)/AR(-)/PSA(-) intermediate cells. Such proliferatively quiescent intermediate cells, however, do not complete their full maturation into CD133(-)/ABCG2(-)/alpha(2)beta(1)(Lo)/p63(-)/PSCA(-)/AR(+)/PSA(+) luminal-secretory cells in low Ca(2+) SFD medium. Addition of universal type I IFN and synthetic androgen (R1881) to culture medium resulted in up-regulation of androgen receptor protein expression. However, it failed to induce full differentiation of intermediate cells into AR(+)/PSA(+) luminal-secretory cells. Our results indicate that such inability of prostate epithelial cells to complete their differentiation is due to continuous expression of Notch-1 receptor and its downstream effector, Hey-1 protein, which actively suppresses differentiation via its ability to transcriptionally repress a series of genes, including the GATA family of transcription factors.


Science Translational Medicine | 2012

Engineering a prostate-specific membrane antigen-activated tumor endothelial cell prodrug for cancer therapy

Samuel R. Denmeade; Annastasiah Mhaka; D. Marc Rosen; W. Nathaniel Brennen; Susan L. Dalrymple; Ingrid Dach; Claus Olesen; Bora Gurel; Angelo M. DeMarzo; George Wilding; Michael A. Carducci; Craig A. Dionne; Jesper V. Møller; Poul Nissen; S. Brøgger Christensen; John T. Isaacs

A prostate-specific membrane antigen–activated prodrug selectively kills cancer cells and is being tested in patients with advanced cancer. An Old Approach Is New Again In the 1995 film The Last Supper, a group of graduate students invite a diverse cast of characters for a series of Sunday dinners. After one guest threatens the lives of several of the students, subsequent dinners turn deadly. If the guest holds views that the group considers toxic to society, then the house wine is made poisonous and served only to the unwanted houseguest, who promptly dies. In a related scenario, Denmeade et al. use a prodrug to seek out and selectively poison unsavory guests that are toxic to the body—namely, cancer cells. The new work describes the development of a thapsigargin (TG) prodrug that is activated in the vasculature of solid tumors by tumor endothelial cells. The carboxypeptidase prostate-specific membrane antigen (PSMA)—which is selectively expressed on the surface of prostate cancer cells, including metastatic ones, and tumor, but not normal, endothelial cells—cleaves and activates the prodrug extracellularly in the tumor microenvironment. The activated cytotoxic moiety then poisons neighboring cancer cells within sites of metastases by entering the cells and inhibiting the sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA) pump, which is essential to the function of all normal and tumor cell types. The authors showed that treatment with the prodrug caused significant tumor regression in two mouse xenograft models of human prostate cancer and one model of human breast cancer with relatively little toxicity—less than that of the maximally tolerated dose of the widely used cancer drug docetaxel. Although the targeted prodrug concept is not new, the current approach has several features that make it superior to many previous ones. First, unlike most cytotoxic cancer drugs, TG is not cell cycle–dependent and thus can kill nondividing cancer cells. Furthermore, drug toxicity is expected to be low, because the PSMA substrate in the prodrug is cleaved primarily by prostate cancer cells and in the vicinity of tumor endothelial cells. In fact, the authors report that studies in cynomolgus monkeys showed minimal toxic effects except in the kidney, and even that renal toxicity was minimal to mild and reversible at the low drug dose. As with all cancer drugs, the new findings will require clinical validation in ongoing studies. However, this unusual therapeutic approach has the potential to be an effective and selective ouster of unwanted invaders that threaten their hosts. Heterogeneous expression of drug target proteins within tumor sites is a major mechanism of resistance to anticancer therapies. We describe a strategy to selectively inhibit, within tumor sites, the function of a critical intracellular protein, the sarcoplasmic/endoplasmic reticulum calcium adenosine triphosphatase (SERCA) pump, whose proper function is required by all cell types for viability. To achieve targeted inhibition, we took advantage of the unique expression of the carboxypeptidase prostate-specific membrane antigen (PSMA) by tumor endothelial cells within the microenvironment of solid tumors. We generated a prodrug, G202, consisting of a PSMA-specific peptide coupled to an analog of the potent SERCA pump inhibitor thapsigargin. G202 produced substantial tumor regression against a panel of human cancer xenografts in vivo at doses that were minimally toxic to the host. On the basis of these data, a phase 1 dose-escalation clinical trial has been initiated with G202 in patients with advanced cancer.


Clinical Cancer Research | 2006

Pharmacologic Basis for the Enhanced Efficacy of Dutasteride against Prostatic Cancers

Yi Xu; Susan L. Dalrymple; Robyn E. Becker; Samuel R. Denmeade; John T. Isaacs

Purpose: Prostatic dihydrotestosterone (DHT) concentration is regulated by precursors from systemic circulation and prostatic enzymes of androgen metabolism, particularly 5α-reductases (i.e., SRD5A1 and SRD5A2). Therefore, the levels of expression SRD5A1 and SRD5A2 and the antiprostatic cancer growth response to finasteride, a selective SRD5A2 inhibitor, versus the dual SRD5A1 and SRD5A2 inhibitor, dutasteride, were compared. Experimental Design: Real-time PCR and enzymatic assays were used to determine the levels of SRD5A1 and SRD5A2 in normal versus malignant rat and human prostatic tissues. Rats bearing the Dunning R-3327H rat prostate cancer and nude mice bearing LNCaP or PC-3 human prostate cancer xenografts were used as model systems. Tissue levels of testosterone and DHT were determined using liquid chromatography-mass spectrometry. Results: Prostate cancer cells express undetectable to low levels of SRD5A2 but elevated levels of SRD5A1 activity compared with nonmalignant prostatic tissue. Daily oral treatment of rats with the SRD5A2 selective inhibitor, finasteride, reduces prostate weight and DHT content but did not inhibit R-3327H rat prostate cancer growth or DHT content in intact (i.e., noncastrated) male rats. In contrast, daily oral treatment with even a low 1 mg/kg/d dose of the dual SRD5A1 and SRD5A2 inhibitor, dutasteride, reduces both normal prostate and H tumor DHT content and weight in intact rats while elevating tissue testosterone. Daily oral treatment with finasteride significantly (P < 0.05) inhibits growth of LNCaP human prostate cancer xenografts in intact male nude mice, but this inhibition is not as great as that by equimolar oral dosing with dutasteride. This anticancer efficacy is not equivalent, however, to that produced by castration. Only combination of dutasteride and castration produces a greater tumor inhibition (P < 0.05) than castration monotherapy against androgen-responsive LNCaP cancers. In contrast, no response was induced by dutasteride in nude mice bearing androgen-independent PC-3 human prostatic cancer xenografts. Conclusions: These results document that testosterone is not as potent as DHT but does stimulate prostate cancer growth, thus combining castration with dutasteride enhances therapeutic efficacy.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Androgen receptor as a licensing factor for DNA replication in androgen-sensitive prostate cancer cells

Ivan V. Litvinov; Donald J. Vander Griend; Lizamma Antony; Susan L. Dalrymple; Angelo M. De Marzo; Charles G. Drake; John T. Isaacs

Androgen receptor (AR) protein expression and function are critical for survival and proliferation of androgen-sensitive (AS) prostate cancer cells. Besides its ability to function as a transcription factor, experimental observations suggest that AR becomes a licensing factor for DNA replication in AS prostate cancer cells and thus must be degraded during each cell cycle in these cells to allow reinitiation of DNA replication in the next cell cycle. This possibility was tested by using the AS human prostate cancer cell lines, LNCaP, CWR22Rv1, and LAPC-4. These studies demonstrated that AR levels fluctuate both within and between various phases of the cell cycle in each of these AS lines. Consistent with its licensing ability, AR is degraded during mitosis via a proteasome-dependent pathway in these AS prostate cancer cells. In contrast, proteasome-dependent degradation of AR during mitosis is not observed in AR-expressing but androgen-insensitive human prostate stromal cells, in which AR does not function as a licensing factor for DNA replication. To evaluate mitotic degradation of AR in vivo, the same series of human AS prostate cancers growing as xenografts in nude mice and malignant tissues obtained directly from prostate cancer patients were evaluated by dual Ki-67 and AR immunohistochemistry for AR expression in mitosis. These results document that AR is also down-regulated during mitosis in vivo. Thus, AS prostate cancer cells do not express AR protein during mitosis, either in vitro or in vivo, consistent with AR functioning as a licensing factor for DNA replication in AS prostate cancer cells.


The Journal of Nuclear Medicine | 2009

Detection and Quantification of the Evolution Dynamics of Apoptosis Using the PET Voltage Sensor 18F-Fluorobenzyl Triphenyl Phosphonium

Igal Madar; Yi Huang; Hayden T. Ravert; Susan L. Dalrymple; Nancy E. Davidson; John T. Isaacs; Robert F. Dannals; J. James Frost

Apoptosis is a key mechanism in numerous pathologies. However, there are no effective noninvasive means available for an early detection and quantitative assessment of evolution dynamics of the apoptotic process. Here, we have characterized the ability of the novel PET voltage sensor 18F-fluorobenzyl triphenyl phosphonium (18F-FBnTP) to quantify the time-dependent apoptotic action of the taxanes paclitaxel and docetaxel in vitro and in vivo. Methods: The duration-dependent treatment effect of paclitaxel on 18F-FBnTP uptake was assayed in human MDA-MB-231 breast carcinoma cells. The expression of the proapoptotic Bax and antiapoptotic Bcl-2 mitochondrial proteins, release of the apoptogen cytochrome c, and activation of executioner caspase-3 were determined by Western blotting. The fraction of viable cells was determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide. The effect of docetaxel on 18F-FBnTP and 18F-FDG uptake in orthotopic prostate tumors in mice was compared. Results: 18F-FBnTP cellular uptake in viable cells declined linearly with the increasing duration of paclitaxel treatment, from 3 to 24 h, and plateaued at 48 h. The extent of decrease of 18F-FBnTP correlated strongly with the Bax-to-Bcl-2 ratio (R2 = 0.83) and release of cytochrome c (R2 = 0.92), but preceded in time the caspase-3 cleavage. The P-glycoprotein blocker verapamil did not interfere with 18F-FBnTP cellular uptake. 18F-FBnTP prostate tumor contrast was greater than 18F-FDG prostate tumor contrast. Docetaxel caused a marked decrease (52.4%) of 18F-FBnTP tumor uptake, within 48 h, whereas 18F-FDG was much less affected (12%). Conclusion: The voltage sensor 18F-FBnTP is a viable means for quantification of paclitaxel pharmacodynamics. 18F-FBnTP permits the detection of paclitaxel apoptotic action in vivo earlier than does 18F-FDG. 18F-FBnTP may afford a novel approach for early detection and quantitative assessment of the cumulative-effect kinetics of proapoptotic drugs and conditions using PET.


Cancer Research | 2013

Tasquinimod is an Allosteric Modulator of HDAC4 Survival Signaling within the Compromised Cancer Microenvironment

John T. Isaacs; Lizamma Antony; Susan L. Dalrymple; W. Nathaniel Brennen; Stephanie Gerber; Hans J. Hammers; Michel D. Wissing; Sushant Kachhap; Jun Luo; Li Xing; Per Björk; Anders Olsson; Anders Björk; Tomas Leanderson

Tasquinimod is an orally active antiangiogenic drug that is currently in phase III clinical trials for the treatment of castration-resistant prostate cancer. However, the target of this drug has remained unclear. In this study, we applied diverse strategies to identify the histone deacetylase HDAC4 as a target for the antiangiogenic activity of tasquinimod. Our comprehensive analysis revealed allosteric binding (Kd 10-30 nmol/L) to the regulatory Zn(2+) binding domain of HDAC4 that locks the protein in a conformation preventing HDAC4/N-CoR/HDAC3 complex formation. This binding inhibited colocalization of N-CoR/HDAC3, thereby inhibiting deacetylation of histones and HDAC4 client transcription factors, such as HIF-1α, which are bound at promoter/enhancers where epigenetic reprogramming is required for cancer cell survival and angiogenic response. Through this mechanism, tasquinimod is effective as a monotherapeutic agent against human prostate, breast, bladder, and colon tumor xenografts, where its efficacy could be further enhanced in combination with a targeted thapsigargin prodrug (G202) that selectively kills tumor endothelial cells. Together, our findings define a mechanism of action of tasquinimod and offer a perspective on how its clinical activity might be leveraged in combination with other drugs that target the tumor microenvironment. Cancer Res; 73(4); 1386-99. ©2012 AACR.


The Prostate | 2014

Androgen receptor (AR) suppresses normal human prostate epithelial cell proliferation via AR/β-catenin/TCF-4 complex inhibition of c-MYC transcription.

Lizamma Antony; Freek van der Schoor; Susan L. Dalrymple; John T. Isaacs

Physiologic testosterone continuously stimulates prostate stromal cell secretion of paracrine growth factors (PGFs), which if unopposed would induce hyperplastic overgrowth of normal prostate epithelial cells (PrECs).


Molecular Cancer Therapeutics | 2009

Amino acid containing thapsigargin analogues deplete androgen receptor protein via synthesis inhibition and induce the death of prostate cancer cells

Donald J. Vander Griend; Lizamma Antony; Susan L. Dalrymple; Yi Xu; S. Brøgger Christensen; Samuel R. Denmeade; John T. Isaacs

There are quantitative and/or qualitative mechanisms allowing androgen receptor (AR) growth signaling in androgen ablation refractory prostate cancer cells. Regardless of the mechanism, agents that deplete AR protein expression prevent such AR growth signaling. Thapsigargin (TG) is a highly cell-penetrant sequiterpene-lactone that once inside cells inhibits (IC50, ∼10 nmol/L) critically important housekeeping SERCA 2b calcium pumps in the endoplasmic reticulum. Using a series of five genetically diverse androgen ablation refractory human prostate cancer lines (LNCaP, LAPC-4, VCaP, MDA-PCa-2b, and CWR22Rv1), TG inhibition of SERCA pumps consistently results in depletion of the endoplasmic reticulum Ca+2 coupled with μmol/L elevation in the intracellular free Ca+2 initiating a molecular cascade that: (a) inhibits Cap-dependent AR protein synthesis resulting in 90% depletion of AR protein by 24 hours of TG exposure, (b) arrests the cells in G0, and (c) induces their apoptotic death. Unfortunately, due to its highly lipophilic nature, TG is not deliverable as a systemic agent without host toxicity. Therefore, TG analogues containing amino acids were developed, which retain ability to deplete AR protein and induce cell death and which can be covalently linked to peptide carriers producing water soluble prodrugs for systemic delivery. Specific amino acid sequences are used to restrict the liberation of cytotoxic amino acid containing TG analogues from the peptide prodrug by prostate-specific proteases, such as prostate-specific antigen and prostate-specific membrane antigen, or cancer-specific proteases, such as fibroblast activation protein, so that toxicity of these prodrugs is selectively targeted to metastatic sites of prostate cancer. Based on these results, these prodrugs are undergoing clinical development. [Mol Cancer Ther 2008;8(5):1340–9]


Oncotarget | 2016

Assessing angiogenic responses induced by primary human prostate stromal cells in a three-dimensional fibrin matrix assay

W. Nathaniel Brennen; Huong Nguyen; Susan L. Dalrymple; Stephanie Reppert-Gerber; Jeesun Kim; John T. Isaacs; Hans J. Hammers

Accurate modeling of angiogenesis in vitro is essential for guiding the preclinical development of novel anti-angiogenic agents and treatment strategies. The formation of new blood vessels is a multifactorial and multi-stage process dependent upon paracrine factors produced by stromal cells in the local microenvironment. Mesenchymal stem cells (MSCs) are multipotent cells in adults that can be recruited to sites of inflammation and tissue damage where they aid in wound healing through regenerative, trophic, and immunomodulatory properties. Primary stromal cultures derived from human bone marrow, normal prostate, or prostate cancer tissue are highly enriched in MSCs and stromal progenitors. Using conditioned media from these primary cultures, a robust pro-angiogenic response was observed in a physiologically-relevant three-dimensional fibrin matrix assay. To evaluate the utility of this assay, the allosteric HDAC4 inhibitor tasquinimod and the anti-VEGF monoclonal antibody bevacizumab were used as model compounds with distinct mechanisms of action. While both agents had a profound inhibitory effect on endothelial sprouting, only bevacizumab induced significant regression of established vessels. Additionally, the pro-angiogenic properties of MSCs derived from prostate cancer patients provides further evidence that selective targeting of this population may be of therapeutic benefit.


Cancer Research | 2012

Abstract 4701: Tasquinimod suppresses the adaptive stress response within sites of prostate cancer via disrupting zinc-dependent Histone Deacetylase 4 signaling.

Lizamma Antony; Susan L. Dalrymple; Stephanie Gerber; Hans J. Hammers; Sushant Kachhap; Per Björk; Anders Olsson; Tomas Leanderson; John T. Isaacs

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL Tasquinimod (i.e., N-ethyl-N-phenyl-5-chloro-1,2-dihydro-4-hydroxy-1-methyl-2-oxo-3-quinoline-carboxamide) is a novel orally active potent inhibitor of tumor angiogenesis with single agent efficacy which also enhances androgen ablation, taxane-based chemotherapies, or fractionated radiation in pre-clinical prostate cancer models [Expert Opin Investig Drugs (2010). 19: 1235-43]. Based upon its anti-angiogenic efficacy, tasquinimod is in clinical development and has successfully completed phase I/II testing and is presently in multi-center Phase III evaluation for the treatment of castration resistant prostate cancer at an oral dose of up to1mg per day. To clarify the molecular targets for tasquinimods anti-angiogenic efficacy, a library of quinoline-3-carboxamide analogs was screened based upon inhibition of human endothelial cell sprouting in a 3-dimensional assay to identify the active pharmacophore. These structure activity relationship (SAR) studies document that the two carbonyl oxygens in the quinoline-2-position and 3-position carboxamide side chain are essential for activity. These SAR studies raised the issue of whether these two carbonyl oxygens might function in bidentate metal binding. To test this possibility surface plasmon resonance was used to evaluate binding of tasquinimod to Zn bound proteins. These studies document that tasquinimod binds with low nM affinity to Zn bound proteins. A combination of tumor, cell, and molecular biology assays identified HDAC4 as one of the Zn bound proteins to which tasquinimod binds. Using shRNA knock-down and subsequent replacement knock-in, both human prostate cancer cells and human endothelial cells require a critical level of nuclear HDAC4 function to survive in both cell culture and xenograft assays under cellular stress conditions like acidic pH and hypoxia characteristic of cancer growth. Survival under these stress conditions requires formation of HDAC4/N-CoR/HDAC3 complexes which bind to and deacetylate transcription factors, like HIF-1α, altering transcription. Such HDAC4/N-CoR/HDAC3 complex formation occurs through a structural Zn binding domain in HDAC4. While tasquinimod does not inhibit HDAC enzymatic activity directly, it binds HDAC4 and inhibits Zn-dependent formation of HDAC4/N-CoR/HDAC3 complex thus inhibiting deacetylation of HIF-1α preventing the adaptive transcription response needed for survival under acidic/hypoxia conditions. The fact that tasquinimod phenocopies the response of prostate cancer and endothelial cells to HDAC4 knock-down identifies HDAC4 as one of the critical targets for tasquinimods mechanism of action. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 4701. doi:1538-7445.AM2012-4701

Collaboration


Dive into the Susan L. Dalrymple's collaboration.

Top Co-Authors

Avatar

John T. Isaacs

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Lizamma Antony

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Samuel R. Denmeade

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yi Xu

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

D. Marc Rosen

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge