Network


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

Hotspot


Dive into the research topics where Andrew M. Lowy is active.

Publication


Featured researches published by Andrew M. Lowy.


Cancer Cell | 2003

Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse

Sunil R. Hingorani; Emanuel F. Petricoin; Anirban Maitra; Vinodh N. Rajapakse; Catrina King; Michael A. Jacobetz; Sally Ross; Thomas P. Conrads; Timothey D. Veenstra; Ben A. Hitt; Yoshiya Kawaguchi; Don Johann; Lance A. Liotta; Howard C. Crawford; Mary E. Putt; Tyler Jacks; Christopher V.E. Wright; Ralph H. Hruban; Andrew M. Lowy; David A. Tuveson

To evaluate the role of oncogenic RAS mutations in pancreatic tumorigenesis, we directed endogenous expression of KRAS(G12D) to progenitor cells of the mouse pancreas. We find that physiological levels of Kras(G12D) induce ductal lesions that recapitulate the full spectrum of human pancreatic intraepithelial neoplasias (PanINs), putative precursors to invasive pancreatic cancer. The PanINs are highly proliferative, show evidence of histological progression, and activate signaling pathways normally quiescent in ductal epithelium, suggesting potential therapeutic and chemopreventive targets for the cognate human condition. At low frequency, these lesions also progress spontaneously to invasive and metastatic adenocarcinomas, establishing PanINs as definitive precursors to the invasive disease. Finally, mice with PanINs have an identifiable serum proteomic signature, suggesting a means of detecting the preinvasive state in patients.


Annals of Surgery | 2008

Left-sided pancreatectomy: a multicenter comparison of laparoscopic and open approaches.

David A. Kooby; Theresa W. Gillespie; David J. Bentrem; Attila Nakeeb; Max Schmidt; Nipun B. Merchant; Alexander A. Parikh; Robert C.G. Martin; Charles R. Scoggins; Syed A. Ahmad; Hong Jin Kim; Jaemin Park; Fabian M. Johnston; Matthew J. Strouch; Alex Menze; Jennifer A. Rymer; Rebecca J. McClaine; Steven M. Strasberg; Mark S. Talamonti; Charles A. Staley; Kelly M. McMasters; Andrew M. Lowy; Johnita Byrd-Sellers; William C. Wood; William G. Hawkins

Objectives:To compare perioperative outcomes of laparoscopic left-sided pancreatectomy (LLP) with traditional open left-sided pancreatectomy (OLP) in a multicenter experience. Summary and Background Data:LLP is being performed more commonly with limited data comparing results with outcomes from OLP. Methods:Data from 8 centers were combined for all cases performed between 2002–2006. OLP and LLP cohorts were matched by age, American Society of Anesthesiologists, resected pancreas length, tumor size, and diagnosis. Multivariate analysis was performed using binary logistic regression. Results:Six hundred sixty-seven LPs were performed, with 159 (24%) attempted laparoscopically. Indications were solid lesion in 307 (46%), cystic in 295 (44%), and pancreatitis in 65 (10%) cases. Positive margins occurred in 51 (8%) cases, 335 (50%) had complications, and significant leaks occurred in 108 (16%). Conversion to OLP occurred in 20 (13%) of the LLPs. In the matched comparison, 200 OLPs were compared with 142 LLPs. There were no differences in positive margin rates (8% vs. 7%, P = 0.8), operative times (216 vs. 230 minutes, P = 0.3), or leak rates (18% vs. 11%, P = 0.1). LLP patients had lower average blood loss (357 vs. 588 mL, P < 0.01), fewer complications (40% vs. 57%, P < 0.01), and shorter hospital stays (5.9 vs. 9.0 days, P < 0.01). By MVA, LLP was an independent factor for shorter hospital stay (P < 0.01, odds ratio 0.33, 95% confidence interval 0.19–0.56). Conclusions:In selected patients, LLP is associated with less morbidity and shorter LOS than OLP. Pancreatic fistula rates are similar for OLP and LLP. LLP is appropriate for selected patients with left-sided pancreatic pathology.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Mutant p53 drives metastasis and overcomes growth arrest/senescence in pancreatic cancer

Jennifer P. Morton; Paul Timpson; Saadia A. Karim; Rachel A. Ridgway; Dimitris Athineos; Brendan Doyle; Nigel B. Jamieson; Karin A. Oien; Andrew M. Lowy; Valerie G. Brunton; Margaret C. Frame; T.R. Jeffry Evans; Owen J. Sansom

TP53 mutation occurs in 50–75% of human pancreatic ductal adenocarcinomas (PDAC) following an initiating activating mutation in the KRAS gene. These p53 mutations frequently result in expression of a stable protein, p53R175H, rather than complete loss of protein expression. In this study we elucidate the functions of mutant p53 (Trp53R172H), compared to knockout p53 (Trp53fl), in a mouse model of PDAC. First we find that although KrasG12D is one of the major oncogenic drivers of PDAC, most KrasG12D-expressing pancreatic cells are selectively lost from the tissue, and those that remain form premalignant lesions. Loss, or mutation, of Trp53 allows retention of the KrasG12D-expressing cells and drives rapid progression of these premalignant lesions to PDAC. This progression is consistent with failed growth arrest and/or senescence of premalignant lesions, since a mutant of p53, p53R172P, which can still induce p21 and cell cycle arrest, is resistant to PDAC formation. Second, we find that despite similar kinetics of primary tumor formation, mutant p53R172H, as compared with genetic loss of p53, specifically promotes metastasis. Moreover, only mutant p53R172H-expressing tumor cells exhibit invasive activity in an in vitro assay. Importantly, in human PDAC, p53 accumulation significantly correlates with lymph node metastasis. In summary, by using ‘knock-in’ mutations of Trp53 we have identified two critical acquired functions of a stably expressed mutant form of p53 that drive PDAC; first, an escape from KrasG12D-induced senescence/growth arrest and second, the promotion of metastasis.


Annals of Surgery | 1999

Response to Neoadjuvant Chemotherapy Best Predicts Survival After Curative Resection of Gastric Cancer

Andrew M. Lowy; Paul F. Mansfield; Steven D. Leach; Richard Pazdur; Pamela Dumas; Jaffer A. Ajani

OBJECTIVE In Western populations, long-term survival rates after curative resection of gastric cancer remain extremely poor. The lack of effective adjuvant therapy has prompted the evaluation of neoadjuvant approaches. Since 1988, we have conducted three separate phase II trials using neoadjuvant chemotherapy to treat patients with potentially resectable gastric cancer. The present study was conducted to evaluate whether response to neoadjuvant chemotherapy is predictive of survival in patients with resectable gastric cancer. METHODS Eighty-three patients with pathologically confirmed gastric adenocarcinoma were treated with neoadjuvant chemotherapy before planned surgical resection. Response was assessed by upper gastrointestinal series, endoscopy, computed tomography scan, and pathologic examination. RESULTS For the three phase II trials, clinical response rates ranged from 24% to 38%. Three patients (4%) had a complete pathologic response. Sixty-one patients (73%) underwent a curative resection. Median follow-up was 26 months. Univariate analysis revealed T stage, number of positive nodes, and response to chemotherapy to be significant predictors of overall survival. However, on multivariate analysis, response to chemotherapy was found to be the only independent prognostic factor. CONCLUSIONS Response to neoadjuvant chemotherapy is the single most important predictor of overall survival after neoadjuvant chemotherapy for gastric cancer. These findings support further evaluation of neoadjuvant approaches in the treatment of this disease.


Developmental Cell | 2009

Sox17 Regulates Organ Lineage Segregation of Ventral Foregut Progenitor Cells

Jason R. Spence; Alex W. Lange; Suh Chin J. Lin; Klaus H. Kaestner; Andrew M. Lowy; Injune Kim; Jeffrey A. Whitsett; James M. Wells

The ventral pancreas, biliary system, and liver arise from the posterior ventral foregut, but the cell-intrinsic pathway by which these organ lineages are separated is not known. Here we show that the extrahepatobiliary system shares a common origin with the ventral pancreas and not the liver, as previously thought. These pancreatobiliary progenitor cells coexpress the transcription factors PDX1 and SOX17 at E8.5 and their segregation into a PDX1+ ventral pancreas and a SOX17+ biliary primordium is Sox17-dependent. Deletion of Sox17 at E8.5 results in the loss of biliary structures and ectopic pancreatic tissue in the liver bud and common duct, while Sox17 overexpression suppresses pancreas development and promotes ectopic biliary-like tissue throughout the PDX1+ domain. Restricting SOX17+ biliary progenitor cells to the ventral region of the gut requires the notch effector Hes1. Our results highlight the role of Sox17 and Hes1 in patterning and morphogenetic segregation of ventral foregut lineages.


Journal of Clinical Oncology | 2009

Consensus report of the national cancer institute clinical trials planning meeting on pancreas cancer treatment

Philip A. Philip; Margaret Mooney; Deborah Jaffe; Gail Eckhardt; Malcolm J. Moore; Neal J. Meropol; Leisha A. Emens; Eileen Mary O'Reilly; Murray Korc; Lee M. Ellis; Jacqueline Benedetti; Mace L. Rothenberg; Christopher G. Willett; Margaret A. Tempero; Andrew M. Lowy; James L. Abbruzzese; Diane M. Simeone; Sunil R. Hingorani; Jordan Berlin; Joel E. Tepper

Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer mortality, despite significant improvements in diagnostic imaging and operative mortality rates. The 5-year survival rate remains less than 5% because of microscopic or gross metastatic disease at time of diagnosis. The Clinical Trials Planning Meeting in pancreatic cancer was convened by the National Cancer Institutes Gastrointestinal Cancer Steering Committee to discuss the integration of basic and clinical knowledge in the design of clinical trials in PDAC. Major emphasis was placed on the enhancement of research to identify and validate the relevant targets and molecular pathways in PDAC, cancer stem cells, and the microenvironment. Emphasis was also placed on developing rational combinations of targeted agents and the development of predictive biomarkers to assist selection of patient subsets. The development of preclinical tumor models that are better predictive of human PDAC must be supported with wider availability to the research community. Phase III clinical trials should be implemented only if there is a meaningful clinical signal of efficacy and safety in the phase II setting. The emphasis must therefore be on performing well-designed phase II studies with uniform sets of basic entry and evaluation criteria with survival as a primary endpoint. Patients with either metastatic or locally advanced PDAC must be studied separately.


Annals of Surgical Oncology | 2009

Combined Modality Treatment of Resectable and Borderline Resectable Pancreas Cancer: Expert Consensus Statement

Ross A. Abrams; Andrew M. Lowy; Eileen M. O’Reilly; Robert A. Wolff; Vincent J. Picozzi; Peter W.T. Pisters

Department of Radiation Oncology, Rush University Medical Center, Chicago, IL; Division of Surgical Oncology, University of California at San Diego, San Diego, CA; Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York; Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX; Pancreaticobiliary Cancer, Digestive Diseases Institute, Virginia Mason Medical Center, Seattle, WA; Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX


Nature Cell Biology | 2014

An integrin β 3 –KRAS–RalB complex drives tumour stemness and resistance to EGFR inhibition

Laetitia Seguin; Shumei Kato; Aleksandra Franovic; M. Fernanda Camargo; Jacqueline Lesperance; Kathryn C. Elliott; Mayra Yebra; Ainhoa Mielgo; Andrew M. Lowy; Hatim Husain; Tina Cascone; Lixia Diao; Jing Wang; Ignacio I. Wistuba; John V. Heymach; Scott M. Lippman; Jay S. Desgrosellier; Sudarshan Anand; Sara M. Weis; David A. Cheresh

Tumour cells, with stem-like properties, are highly aggressive and often show drug resistance. Here, we reveal that integrin αvβ3 serves as a marker of breast, lung and pancreatic carcinomas with stem-like properties that are highly resistant to receptor tyrosine kinase inhibitors such as erlotinib. This was observed in vitro and in mice bearing patient-derived tumour xenografts or in clinical specimens from lung cancer patients who had progressed on erlotinib. Mechanistically, αvβ3, in the unliganded state, recruits KRAS and RalB to the tumour cell plasma membrane, leading to the activation of TBK1 and NF-κB. In fact, αvβ3 expression and the resulting KRAS–RalB–NF-κB pathway were both necessary and sufficient for tumour initiation, anchorage independence, self-renewal and erlotinib resistance. Pharmacological targeting of this pathway with bortezomib reversed both tumour stemness and erlotinib resistance. These findings not only identify αvβ3 as a marker/driver of carcinoma stemness but also reveal a therapeutic strategy to sensitize such tumours to RTK inhibition.


BMC Developmental Biology | 2007

Wnt/β-catenin signaling is required for development of the exocrine pancreas

James M. Wells; Farzad Esni; Gregory P. Boivin; Bruce J. Aronow; William D. Stuart; Chelsea Combs; Angela M. Sklenka; Steven D. Leach; Andrew M. Lowy

Backgroundβ-catenin is an essential mediator of canonical Wnt signaling and a central component of the cadherin-catenin epithelial adhesion complex. Dysregulation of β-catenin expression has been described in pancreatic neoplasia. Newly published studies have suggested that β-catenin is critical for normal pancreatic development although these reports reached somewhat different conclusions. In addition, the molecular mechanisms by which loss of β-catenin affects pancreas development are not well understood. The goals of this study then were; 1] to further investigate the role of β-catenin in pancreatic development using a conditional knockout approach and 2] to identify possible mechanisms by which loss of β-catenin disrupts pancreatic development. A Pdx1-cre mouse line was used to delete a floxed β-catenin allele specifically in the developing pancreas, and embryonic pancreata were studied by immunohistochemistry and microarray analysis.ResultsPdx1-cre floxed β-catenin animals were viable but demonstrated small body size and shortened median survival. The pancreata from knockout mice were hypoplastic and histologically demonstrated a striking paucity of exocrine pancreas, acinar to duct metaplasia, but generally intact pancreatic islets containing all lineages of endocrine cells. In animals with extensive acinar hypoplasia, putative hepatocyte transdifferention was occasionally observed. Obvious and uniform pancreatic hypoplasia was observed by embryonic day E16.5. Transcriptional profiling of Pdx1-cre floxed β-catenin embryonic pancreata at E14.5, before there was a morphological phenotype, revealed significant decreases in the β-catenin target gene N-myc, and the basic HLH transcription factor PTF1, and an increase of several pancreatic zymogens compared to control animals. By E16.5, there was a dramatic loss of exocrine markers and an increase in Hoxb4, which is normally expressed anterior to the pancreas.ConclusionWe conclude that β-catenin expression is required for development of the exocrine pancreas, but is not required for development of the endocrine compartment. In contrast, β-catenin/Wnt signaling appears to be critical for proliferation of PTF1+ nascent acinar cells and may also function, in part, to maintain an undifferentiated state in exocrine/acinar cell precursors. Finally, β-catenin may be required to maintain positional identity of the pancreatic endoderm along the anterior-posterior axis. This data is consistent with the findings of frequent β-catenin mutations in carcinomas of acinar cell lineage seen in humans.


Cancer Research | 2010

Integrin α4β1 Signaling Is Required for Lymphangiogenesis and Tumor Metastasis

Barbara Garmy-Susini; Christie J. Avraamides; Michael C. Schmid; Philippe Foubert; Lesley G. Ellies; Leo Barnes; Chloé C. Féral; Thalia Papayannopoulou; Andrew M. Lowy; Sarah L. Blair; David A. Cheresh; Mark H. Ginsberg; Judith A. Varner

Recent studies have shown that lymphangiogenesis or the growth of lymphatic vessels at the periphery of tumors promotes tumor metastasis to lymph nodes. We show here that the fibronectin-binding integrin alpha4beta1 and its ligand fibronectin are novel functional markers of proliferative lymphatic endothelium. Tumors and lymphangiogenic growth factors, such as vascular endothelial growth factor-C (VEGF-C) and VEGF-A, induce lymphatic vessel expression of integrin alpha4beta1. Integrin alpha4beta1 then promotes growth factor and tumor-induced lymphangiogenesis, as genetic loss of integrin alpha4beta1 expression in Tie2Cre+ alpha4(loxp/loxp) mice or genetic loss of alpha4 signaling in alpha4Y991A knock-in mice blocks growth factor and tumor-induced lymphangiogenesis, as well as tumor metastasis to lymph nodes. In addition, antagonists of integrin alpha4beta1 suppress lymphangiogenesis and tumor metastasis. Our studies show that integrin alpha4beta1 and the signals it transduces regulate the adhesion, migration, invasion, and survival of proliferating lymphatic endothelial cells. As suppression of alpha4beta1 expression, signal transduction, or function in tumor lymphatic endothelium not only inhibits tumor lymphangiogenesis but also prevents metastatic disease, these results show that integrin alpha4beta1-mediated tumor lymphangiogenesis promotes metastasis and is a useful target for the suppression of metastatic disease.

Collaboration


Dive into the Andrew M. Lowy's collaboration.

Top Co-Authors

Avatar

Syed A. Ahmad

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Randall French

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dawn Jaquish

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph Kim

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge