Network


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

Hotspot


Dive into the research topics where Patricia S. Steeg is active.

Publication


Featured researches published by Patricia S. Steeg.


Cell | 1991

Cancer metastasis and angiogenesis: An imbalance of positive and negative regulation

Lance A. Liotta; Patricia S. Steeg; William G. Stetler-Stevenson

Lance A. Liotta, Patricia S. Steeg, and William G. Stetler-Stevenson Laboratory of Pathology National Cancer Institute National Institutes of Health Bethesda, Maryland 20892 The most life-threatening aspects of the oncogenic pro- cess are invasion and metastasis. Even though the clinical significance of such expression of the malignant pheno- type has been well appreciated, advances in understand- ing the molecular mechanisms involved in metastasis have lagged behind other developments in the cancer field. Progress has been hindered by the sheer complexity of this multistep, tumor-host interaction that also encom- passes angiogenic and immunologic mechanisms. To tackle the problem, investigators have separated invasion and metastasis into a series of defined, sequential steps, and focused on one step at a time. For each step, new ex- perimental models had to be developed, and a combined effort using the disciplines of cell biology, protein bio- chemistry, and molecular genetics has now resulted in a surge of new information. General themes are emerging that yield new strategies for prognosis and therapy of hu- man metastatic cancer. A group of coordinated cellular processes is responsi- ble for metastasis. Furthermore, it is now clear that nega- tive regulatory processes may be just as important as positive ones. Some genetic changes result in an im- balance of growth regulation, leading to uncontrolled proliferation. However, unrestrained growth does not, by itself, result in invasion and metastasis. The latter pheno- type may therefore require additional genetic changes. Thus, tumorigenicity and metastatic potential have both overlapping and separate features. Invasion and metasta- sis can be facilitated by proteins that stimulate tumor cell attachment to host cellular or extracellular matrix deter- minants, tumor cell proteolysis of host barriers such as the basement membrane, tumor cell locomotion, and tumor cell colony formation in the target organ for metastasis. Facilitory proteins may act at many levels intracellularly or extracellularly, but are counterbalanced by factors that can block their production, regulation, or action. A com- mon theme has emerged: in addition to loss of growth control, an imbalanced regulation of motility and proteoly- sis appears to be required for invasion and metastasis. Moreover, these same functions are also necessary for angiogenesis. Angiogenesis by normal endothelial cells and metastasis by tumor cells are functionally similar but differ in their regulation. Metastatic Cells Do Not Constitute a Minor Subpopulation The process of metastasis involves a cascade of linked, sequential steps involving multiple host-tumor interac- tions (Fidler and Hart, 1982; Schirrmacher, 1985; Liotta et


Nature Medicine | 2006

Tumor metastasis: mechanistic insights and clinical challenges.

Patricia S. Steeg

Metastatic disease is the primary cause of death for most cancer patients. Complex and redundant pathways involving the tumor cell and the microenvironment mediate tumor invasion at the primary site, survival and arrest in the bloodstream, and progressive outgrowth at a distant site. Understanding these pathways and their dynamic interactions will help identify promising molecular targets for cancer therapy and key obstacles to their clinical development.


Cell | 1991

Reduced tumor incidence, metastatic potential, and cytokine responsiveness of nm3-transfected melanoma cells

Alvaro Leone; Ursula Flatow; C. Richter King; Mary Ann Sandeen; Inger Margulies; Lance A. Liotta; Patricia S. Steeg

Reduced expression of the nm23 gene in certain rodent model systems and human breast tumors has been correlated with high tumor metastatic potential. To investigate the functional effects of nm23 expression, we have transfected a constitutive murine nm23-1 expression construct into highly metastatic K-1735 TK murine melanoma cells. TK clones expressing the exogenous nm23-1 construct exhibited a reduced incidence of primary tumor formation, significant reductions in tumor metastatic potential independent of tumor cell growth, and altered responses to the cytokine transforming growth factor beta 1 in soft agar colonization assays, compared with control-transfected TK clones. In contrast, nm23-1-transfected TK clones exhibited no significant differences in intrinsic tumor cell growth, i.e., primary tumor size in vivo, anchorage-dependent growth rate in vitro, and anchorage-independent colony formation in soft agar in vitro. The data demonstrate a suppressive effect of nm23 on several aspects of the cancer process, including tumor metastasis.


American Journal of Pathology | 2005

Breast Cancer Metastasis to the Central Nervous System

Robert J. Weil; Diane Palmieri; Julie L. Bronder; Andreas M. Stark; Patricia S. Steeg

Clinically symptomatic metastases to the central nervous system (CNS) occur in approximately 10 to 15% of patients with metastatic beast cancer. CNS metastases are traditionally viewed as a late complication of systemic disease, for which few effective treatment options exist. Recently, patients with Her-2-positive breast tumors who were treated with trastuzumab have been reported to develop CNS metastases at higher rates, often while responding favorably to treatment. The blood:brain barrier and the unique brain microenvironment are hypothesized to promote distinct molecular features in CNS metastases that may require tailored therapeutic approaches. New research approaches using cell lines that reliably and preferentially metastasize in vivo to the brain have been reported. Using such model systems, as well as in vitro analogs of blood-brain barrier penetration and tissue-based studies, new molecular leads into this disease are unfolding.


Cell | 1990

A Drosophila gene that is homologous to a mammalian gene associated with tumor metastasis codes for a nucleoside diphosphate kinase

Joseph Biggs; Evelyn Hersperger; Patricia S. Steeg; Lance A. Liotta; Allen Shearn

The product of the abnormal wing discs (awd) gene of Drosophila is 78% identical to the product of the nm23 gene of mammals, which is differentially expressed in certain metastatic tumors. We present evidence that the awd gene codes for a nucleoside diphosphate kinase (NDP kinase) and that this Awd/NDP kinase is microtubule associated. Neuroblasts in Drosophila larvae homozygous for a null mutation in the awd gene are arrested in metaphase, indicating that microtubule-associated Awd/NDP kinase plays a critical role in spindle microtubule polymerization.


Clinical Cancer Research | 2010

Heterogeneous blood-tumor barrier permeability determines drug efficacy in experimental brain metastases of breast cancer

Paul R. Lockman; Rajendar K. Mittapalli; Kunal S. Taskar; Vinay Rudraraju; Brunilde Gril; Kaci A. Bohn; Chris E. Adkins; Amanda Roberts; Helen R. Thorsheim; Julie A. Gaasch; Suyun Huang; Diane Palmieri; Patricia S. Steeg; Quentin R. Smith

Purpose: Brain metastases of breast cancer appear to be increasing in incidence, confer significant morbidity, and threaten to compromise gains made in systemic chemotherapy. The blood–tumor barrier (BTB) is compromised in many brain metastases; however, the extent to which this influences chemotherapeutic delivery and efficacy is unknown. Herein, we answer this question by measuring BTB passive integrity, chemotherapeutic drug uptake, and anticancer efficacy in vivo in two breast cancer models that metastasize preferentially to brain. Experimental Design:Experimental brain metastasis drug uptake and BTB permeability were simultaneously measured using novel fluorescent and phosphorescent imaging techniques in immune-compromised mice. Drug-induced apoptosis and vascular characteristics were assessed using immunofluorescent microscopy. Results: Analysis of over 2,000 brain metastases from two models (human 231-BR-Her2 and murine 4T1-BR5) showed partial BTB permeability compromise in greater than 89% of lesions, varying in magnitude within and between metastases. Brain metastasis uptake of 14C-paclitaxel and 14C-doxorubicin was generally greater than normal brain but less than 15% of that of other tissues or peripheral metastases, and only reached cytotoxic concentrations in a small subset (∼10%) of the most permeable metastases. Neither drug significantly decreased the experimental brain metastatic ability of 231-BR-Her2 tumor cells. BTB permeability was associated with vascular remodeling and correlated with overexpression of the pericyte protein desmin. Conclusions: This work shows that the BTB remains a significant impediment to standard chemotherapeutic delivery and efficacy in experimental brain metastases of breast cancer. New brain permeable drugs will be needed. Evidence is presented for vascular remodeling in BTB permeability alterations. Clin Cancer Res; 16(23); 5664–78. ©2010 AACR.


Nature | 2015

Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth

Lin Zhang; Siyuan Zhang; Jun Yao; Frank J. Lowery; Qingling Zhang; Wen Chien Huang; Ping Li; Min Li; Xiao Wang; Chenyu Zhang; Hai Wang; Kenneth Ellis; Mujeeburahiman Cheerathodi; Joseph H. McCarty; Diane Palmieri; Jodi M. Saunus; Sunil R. Lakhani; Suyun Huang; Aysegul A. Sahin; Kenneth D. Aldape; Patricia S. Steeg; Dihua Yu

The development of life-threatening cancer metastases at distant organs requires disseminated tumour cells’ adaptation to, and co-evolution with, the drastically different microenvironments of metastatic sites. Cancer cells of common origin manifest distinct gene expression patterns after metastasizing to different organs. Clearly, the dynamic interaction between metastatic tumour cells and extrinsic signals at individual metastatic organ sites critically effects the subsequent metastatic outgrowth. Yet, it is unclear when and how disseminated tumour cells acquire the essential traits from the microenvironment of metastatic organs that prime their subsequent outgrowth. Here we show that both human and mouse tumour cells with normal expression of PTEN, an important tumour suppressor, lose PTEN expression after dissemination to the brain, but not to other organs. The PTEN level in PTEN-loss brain metastatic tumour cells is restored after leaving the brain microenvironment. This brain microenvironment-dependent, reversible PTEN messenger RNA and protein downregulation is epigenetically regulated by microRNAs from brain astrocytes. Mechanistically, astrocyte-derived exosomes mediate an intercellular transfer of PTEN-targeting microRNAs to metastatic tumour cells, while astrocyte-specific depletion of PTEN-targeting microRNAs or blockade of astrocyte exosome secretion rescues the PTEN loss and suppresses brain metastasis in vivo. Furthermore, this adaptive PTEN loss in brain metastatic tumour cells leads to an increased secretion of the chemokine CCL2, which recruits IBA1-expressing myeloid cells that reciprocally enhance the outgrowth of brain metastatic tumour cells via enhanced proliferation and reduced apoptosis. Our findings demonstrate a remarkable plasticity of PTEN expression in metastatic tumour cells in response to different organ microenvironments, underpinning an essential role of co-evolution between the metastatic cells and their microenvironment during the adaptive metastatic outgrowth. Our findings signify the dynamic and reciprocal cross-talk between tumour cells and the metastatic niche; importantly, they provide new opportunities for effective anti-metastasis therapies, especially of consequence for brain metastasis patients.


Clinical Cancer Research | 2007

Nelfinavir, A lead HIV protease inhibitor, is a broad-spectrum, anticancer agent that induces endoplasmic reticulum stress, autophagy, and apoptosis in vitro and in vivo

Joell J. Gills; Jaclyn LoPiccolo; Junji Tsurutani; Robert H. Shoemaker; Carolyn J.M. Best; Mones Abu-Asab; Jennifer P. Borojerdi; Noel A. Warfel; Erin R. Gardner; Matthew Danish; M. Christine Hollander; Shigeru Kawabata; Maria Tsokos; William D. Figg; Patricia S. Steeg; Phillip A. Dennis

Purpose: The development of new cancer drugs is slow and costly. HIV protease inhibitors are Food and Drug Administration approved for HIV patients. Because these drugs cause toxicities that can be associated with inhibition of Akt, an emerging target in cancer, we assessed the potential of HIV protease inhibitors as anticancer agents. Experimental Design: HIV protease inhibitors were screened in vitro using assays that measure cellular proliferation, apoptotic and nonapoptotic cell death, endoplasmic reticulum (ER) stress, autophagy, and activation of Akt. Nelfinavir was tested in non–small cell lung carcinoma (NSCLC) xenografts with biomarker assessment. Results: Three of six HIV protease inhibitors, nelfinavir, ritonavir, and saquinavir, inhibited proliferation of NSCLC cells, as well as every cell line in the NCI60 cell line panel. Nelfinavir was most potent with a mean 50% growth inhibition of 5.2 μmol/L, a concentration achievable in HIV patients. Nelfinavir caused two types of cell death, caspase-dependent apoptosis and caspase-independent death that was characterized by induction of ER stress and autophagy. Autophagy was protective because an inhibitor of autophagy increased nelfinavir-induced death. Akt was variably inhibited by HIV protease inhibitors, but nelfinavir caused the greatest inhibition of endogenous and growth factor–induced Akt activation. Nelfinavir decreased the viability of a panel of drug-resistant breast cancer cell lines and inhibited the growth of NSCLC xenografts that was associated with induction of ER stress, autophagy, and apoptosis. Conclusions: Nelfinavir is a lead HIV protease inhibitor with pleiotropic effects in cancer cells. Given its wide spectrum of activity, oral availability, and familiarity of administration, nelfinavir is a Food and Drug Administration–approved drug that could be repositioned as a cancer therapeutic.


Cancer Research | 2007

Her-2 Overexpression Increases the Metastatic Outgrowth of Breast Cancer Cells in the Brain

Diane Palmieri; Julie L. Bronder; Jeanne M. Herring; Toshiyuki Yoneda; Robert J. Weil; Andreas M. Stark; Raffael Kurek; Eleazar Vega-Valle; Lionel Feigenbaum; Douglas Halverson; Alexander O. Vortmeyer; Seth M. Steinberg; Kenneth D. Aldape; Patricia S. Steeg

Retrospective studies of breast cancer patients suggest that primary tumor Her-2 overexpression or trastuzumab therapy is associated with a devastating complication: the development of central nervous system (brain) metastases. Herein, we present Her-2 expression trends from resected human brain metastases and data from an experimental brain metastasis assay, both indicative of a functional contribution of Her-2 to brain metastatic colonization. Of 124 archival resected brain metastases from breast cancer patients, 36.2% overexpressed Her-2, indicating an enrichment in the frequency of tumor Her-2 overexpression at this metastatic site. Using quantitative real-time PCR of laser capture microdissected epithelial cells, Her-2 and epidermal growth factor receptor (EGFR) mRNA levels in a cohort of 12 frozen brain metastases were increased up to 5- and 9-fold, respectively, over those of Her-2-amplified primary tumors. Co-overexpression of Her-2 and EGFR was also observed in a subset of brain metastases. We then tested the hypothesis that overexpression of Her-2 increases the colonization of breast cancer cells in the brain in vivo. A subclone of MDA-MB-231 human breast carcinoma cells that selectively metastasizes to brain (231-BR) overexpressed EGFR; 231-BR cells were transfected with low (4- to 8-fold) or high (22- to 28-fold) levels of Her-2. In vivo, in a model of brain metastasis, low or high Her-2-overexpressing 231-BR clones produced comparable numbers of micrometastases in the brain as control transfectants; however, the Her-2 transfectants yielded 3-fold greater large metastases (>50 microm(2); P < 0.001). Our data indicate that Her-2 overexpression increases the outgrowth of metastatic tumor cells in the brain in this model system.


Nature Reviews Clinical Oncology | 2008

Metastasis: a therapeutic target for cancer

Patricia S. Steeg; Dan Theodorescu

Metastasis remains the major driver of mortality in patients with cancer. Our growing body of knowledge regarding this process provides the basis for the development of molecularly targeted therapeutics aimed at the tumor cell or its interaction with the host microenvironment. Here we discuss the similarity and differences between primary tumors and metastases, pathways controlling the colonization of a distant organ, and incorporation of antimetastatic therapies into clinical testing.

Collaboration


Dive into the Patricia S. Steeg's collaboration.

Top Co-Authors

Avatar

Diane Palmieri

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Brunilde Gril

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Seth M. Steinberg

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

David J. Liewehr

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Stephan Woditschka

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emily Hua

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yongzhen Qian

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Christine E. Horak

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge