Network


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

Hotspot


Dive into the research topics where Eric P. Tamm is active.

Publication


Featured researches published by Eric P. Tamm.


Annals of Surgical Oncology | 2006

Borderline Resectable Pancreatic Cancer: Definitions, Management, and Role of Preoperative Therapy

Gauri R. Varadhachary; Eric P. Tamm; James L. Abbruzzese; Henry Q. Xiong; Christopher H. Crane; Huamin Wang; Jeffrey E. Lee; Peter W.T. Pisters; Douglas B. Evans; Robert A. Wolff

With recent advances in pancreatic imaging and surgical techniques, a distinct subset of pancreatic tumors is emerging that blurs the distinction between resectable and locally advanced disease: tumors of “borderline resectability.” In our practice, patients with borderline-resectable pancreatic cancer include those whose tumors exhibit encasement of a short segment of the hepatic artery, without evidence of tumor extension to the celiac axis, that is amenable to resection and reconstruction; tumor abutment of the superior mesenteric artery involving <180° of the circumference of the artery; or short-segment occlusion of the superior mesenteric vein, portal vein, or their confluence with a suitable option available for vascular reconstruction because the veins are normal above and below the area of tumor involvement. With currently available surgical techniques, patients with borderline-resectable pancreatic head cancer are at high risk for a margin-positive resection. Therefore, our approach to these patients is to use preoperative systemic therapy and local-regional chemoradiation to maximize the potential for an R0 resection and to avoid R2 resections. In our experience, patients with favorable responses to preoperative therapy (radiographical evidence of tumor regression and improvement in serum tumor marker levels) are the subset of patients who have the best chance for an R0 resection and a favorable long-term outcome.


Journal of Clinical Oncology | 2008

Preoperative Gemcitabine and Cisplatin Followed by Gemcitabine-Based Chemoradiation for Resectable Adenocarcinoma of the Pancreatic Head

Gauri R. Varadhachary; Robert A. Wolff; Christopher H. Crane; Charlotte C. Sun; Jeffrey E. Lee; Peter W.T. Pisters; Jean Nicolas Vauthey; Eddie K. Abdalla; Huamin Wang; Gregg Staerkel; Jeffrey H. Lee; William A. Ross; Eric P. Tamm; Priya Bhosale; Sunil Krishnan; Prajnan Das; Linus Ho; Henry Xiong; James L. Abbruzzese; Douglas B. Evans

PURPOSE We conducted a phase II trial of preoperative gemcitabine and cisplatin chemotherapy in addition to chemoradiation (Gem-Cis-XRT) and pancreaticoduodenectomy (PD) for patients with stage I/II pancreatic adenocarcinoma. PATIENTS AND METHODS Chemotherapy consisted of gemcitabine (750 mg/m(2)) and cisplatin (30 mg/m(2)) given every 2 weeks for four doses. Chemoradiation consisted of four weekly infusions of gemcitabine (400 mg/m(2)) combined with radiation therapy (30 Gy in 10 fractions administered over 2 weeks) delivered 5 days per week. Patients underwent restaging 4 to 6 weeks after completion of chemoradiation and, in the absence of disease progression, were taken to surgery. RESULTS The study enrolled 90 patients; 79 patients (88%) completed chemo-chemoradiation. Sixty-two (78%) of 79 patients were taken to surgery and 52 (66%) of 79 underwent PD. The median overall survival of all 90 patients was 17.4 months. Median survival for the 79 patients who completed chemo-chemoradiation was 18.7 months, with a median survival of 31 months for the 52 patients who underwent PD and 10.5 months for the 27 patients who did not undergo surgical resection of their primary tumor (P < .001). CONCLUSION Preoperative Gem-Cis-XRT did not improve survival beyond that achieved with preoperative gemcitabine-based chemoradiation (Gem-XRT) alone. The longer preoperative interval required more durable biliary decompression (metal stents) but was not associated with local tumor progression. The gemcitabine-based chemoradiation platform is a reasonable foundation on which to build future phase II multimodality trials for stage I/II pancreatic cancer incorporating emerging systemic therapies.


Gastroenterology | 2012

Frequent Detection of Pancreatic Lesions in Asymptomatic High-Risk Individuals

Marcia I. Canto; Ralph H. Hruban; Elliot K. Fishman; Ihab R. Kamel; Richard D. Schulick; Zhe Zhang; Mark Topazian; Naoki Takahashi; Joel G. Fletcher; Gloria M. Petersen; Alison P. Klein; Jennifer E. Axilbund; Constance A. Griffin; Sapna Syngal; John R. Saltzman; Koenraad J. Mortele; Jeffrey E. Lee; Eric P. Tamm; Raghunandan Vikram; Priya Bhosale; Daniel Margolis; James J. Farrell; Michael Goggins

BACKGROUND & AIMS The risk of pancreatic cancer is increased in patients with a strong family history of pancreatic cancer or a predisposing germline mutation. Screening can detect curable, noninvasive pancreatic neoplasms, but the optimal imaging approach is not known. We determined the baseline prevalence and characteristics of pancreatic abnormalities using 3 imaging tests to screen asymptomatic, high-risk individuals (HRIs). METHODS We screened 225 asymptomatic adult HRIs at 5 academic US medical centers once, using computed tomography (CT), magnetic resonance imaging (MRI), and endoscopic ultrasonography (EUS). We compared results in a blinded, independent fashion. RESULTS Ninety-two of 216 HRIs (42%) were found to have at least 1 pancreatic mass (84 cystic, 3 solid) or a dilated pancreatic duct (n = 5) by any of the imaging modalities. Fifty-one of the 84 HRIs with a cyst (60.7%) had multiple lesions, typically small (mean, 0.55 cm; range, 2-39 mm), in multiple locations. The prevalence of pancreatic lesions increased with age; they were detected in 14% of subjects younger than 50 years old, 34% of subjects 50-59 years old, and 53% of subjects 60-69 years old (P < .0001). CT, MRI, and EUS detected a pancreatic abnormality in 11%, 33.3%, and 42.6% of the HRIs, respectively. Among these abnormalities, proven or suspected neoplasms were identified in 85 HRIs (82 intraductal papillary mucinous neoplasms and 3 pancreatic endocrine tumors). Three of 5 HRIs who underwent pancreatic resection had high-grade dysplasia in less than 3 cm intraductal papillary mucinous neoplasms and in multiple intraepithelial neoplasias. CONCLUSIONS Screening of asymptomatic HRIs frequently detects small pancreatic cysts, including curable, noninvasive high-grade neoplasms. EUS and MRI detect pancreatic lesions better than CT.


Journal of Gastrointestinal Surgery | 2003

Diagnostic accuracy of endoscopic ultrasound-guided fine-needle aspiration in patients with presumed pancreatic cancer

Chandrajit P. Raut; Ana M. Grau; Gregg Staerkel; Madhukar Kaw; Eric P. Tamm; Robert A. Wolff; Jean Nicolas Vauthey; Jeffrey E. Lee; Peter W.T. Pisters; Douglas B. Evans

Endoscopic ultrasound (EUS)-guided fine-needle aspiration (FNA) of the pancreas allows the diagnosis of pancreatic cancer to be established without exploratory surgery. We reviewed our recent experience with EUS-FNA in patients with presumed pancreatic cancer and report the diagnostic accuracy and complications of this procedure. Data were reviewed from all patients who presented with CT evidence of a pancreatic mass or a malignant biliary stricture and underwent EUS-FNA at our institution between November 1, 1999, and October 1, 2001. Based on the findings of contrast-enhanced, multislice CT scanning, patients were categorized as having resectable, locally advanced, or metastatic disease. EUS-FNA was performed in 233 patients. A final diagnosis of cancer was established in 216 patients (93%), 15 patients (6%) were found to have benign disease, and the final diagnosis remains unknown in two patients (1%). The sensitivity, specificity, and accuracy of EUS-FNA for diagnosis of a pancreatic malignancy were 91%, 100%, and 92%, respectively. For the 216 patients subsequently proven to have cancer, the results of EUS-FNA were diagnostic in 197 (91%); 96 (90%) of 107 patients with resectable disease, 62 (97%) of 64 with locally advanced disease, and 39 (87%) of 45 with metastatic disease. Four patients (2%) developed a clinically apparent complication that required hospital admission, including two patients who required surgery for duodenal perforation. There were no EUS-related deaths. We conclude that EUS-FNA can safely and accurately establish a cytologic diagnosis in patients with both early-stage and advanced pancreatic cancer. This enables consideration of all treatment options including protocol-based therapy


Cancer | 2012

Response of borderline resectable pancreatic cancer to neoadjuvant therapy is not reflected by radiographic indicators

Matthew H. Katz; Jason B. Fleming; Priya Bhosale; Gauri R. Varadhachary; Jeffrey E. Lee; Robert A. Wolff; Huamin Wang; James L. Abbruzzese; Peter W.T. Pisters; Jean Nicolas Vauthey; Chusilp Charnsangavej; Eric P. Tamm; Christopher H. Crane; Aparna Balachandran

Experience with preoperative therapy for other cancers has led to an assumption that borderline resectable pancreatic cancers can be converted to resectable cancers with preoperative therapy. In this study, the authors sought to determine the rate at which neoadjuvant therapy is associated with a reduction in the size or stage of borderline resectable tumors.


Journal of Clinical Oncology | 2006

Phase I Trial Evaluating the Safety of Bevacizumab With Concurrent Radiotherapy and Capecitabine in Locally Advanced Pancreatic Cancer

Christopher H. Crane; Lee M. Ellis; James L. Abbruzzese; Henry Q. Xiong; Linus Ho; Douglas B. Evans; Eric P. Tamm; Chaan Ng; Peter W.T. Pisters; Chusilp Charnsangavej; Marc E. Delclos; Michael S. O'Reilly; Jeffrey E. Lee; Robert A. Wolff

PURPOSE To study the safety of bevacizumab with capecitabine-based chemoradiotherapy. PATIENTS AND METHODS Patients with inoperable pancreatic adenocarcinoma received bevacizumab 2 weeks before radiotherapy (50.4 Gy treating the primary tumor and gross adenopathy), every 2 weeks during radiotherapy (12 patients each at 2.5, 5.0, 7.5, and 10 mg/kg), and after radiotherapy until disease progression. Capecitabine was administered on days 14 through 52 (650 mg/m2 orally twice daily for the first six patients; 825 mg/m2 for the remaining patients). RESULTS Significant acute gastrointestinal (43% grade 2; 4% grade 3), hand and foot syndrome (21% grade 2), and transient hematologic (8% grade 3 or greater) events were uncommon with protocol mandated dose reductions of capecitabine grade 2 toxicity (43% of patients). Among the first 30 patients treated, three patients had tumor-associated bleeding duodenal ulcers, and one had a contained duodenal perforation. No additional bleeding events occurred among the final 18 patients after patients with duodenal involvement by tumor were excluded. Nine (20%) of 46 assessable patients had confirmed partial responses until distant progression for a median of 6.2 months. Four patients have undergone pancreaticoduodenectomy without perioperative complication. The median survival was 11.6 months (95% CI, 9.6 to 13.6), from the start of protocol therapy. CONCLUSION Concurrent bevacizumab did not significantly increase the acute toxicity of a relatively well-tolerated chemoradiotherapy regimen. However, ulceration and bleeding in the radiation field possibly related to bevacizumab occurred when tumor involved the duodenal mucosa. The encouraging efficacy end points suggest that the further study of bevacizumab with chemoradiotherapy is warranted.


Radiology | 2014

Pancreatic ductal adenocarcinoma radiology reporting template: Consensus statement of the society of abdominal radiology and the american pancreatic association

Mahmoud M. Al-Hawary; Isaac R. Francis; Suresh T. Chari; Elliot K. Fishman; David M. Hough; David Lu; Michael Macari; Alec J. Megibow; Frank H. Miller; Koenraad J. Mortele; Nipun B. Merchant; Rebecca M. Minter; Eric P. Tamm; Dushyant V. Sahani; Diane M. Simeone

Pancreatic ductal adenocarcinoma is an aggressive malignancy with a high mortality rate. Proper determination of the extent of disease on imaging studies at the time of staging is one of the most important steps in optimal patient management. Given the variability in expertise and definition of disease extent among different practitioners as well as frequent lack of complete reporting of pertinent imaging findings at radiologic examinations, adoption of a standardized template for radiology reporting, using universally accepted and agreed on terminology for solid pancreatic neoplasms, is needed. A consensus statement describing a standardized reporting template authored by a multi-institutional group of experts in pancreatic ductal adenocarcinoma that included radiologists, gastroenterologists, and hepatopancreatobiliary surgeons was developed under the joint sponsorship of the Society of Abdominal Radiologists and the American Pancreatic Association. Adoption of this standardized imaging reporting template should improve the decision-making process for the management of patients with pancreatic ductal adenocarcinoma by providing a complete, pertinent, and accurate reporting of disease staging to optimize treatment recommendations that can be offered to the patient. Standardization can also help to facilitate research and clinical trial design by using appropriate and consistent staging by means of resectability status, thus allowing for comparison of results among different institutions.


Radiologic Clinics of North America | 2012

Imaging of Pancreatic Adenocarcinoma: Update on Staging/Resectability

Eric P. Tamm; Aparna Balachandran; Priya Bhosale; Matthew H. Katz; Jason B. Fleming; Jeffrey H. Lee; Gauri R. Varadhachary

Because of the evolution of treatment strategies staging criteria for pancreatic cancer now emphasize arterial involvement for determining unresectable disease. Preoperative therapy may improve the likelihood of margin negative resections of borderline resectable tumors. Cross-sectional imaging is crucial for correctly staging patients. Magnetic resonance (MR) imaging and computed tomography (CT) are probably comparable, with MR imaging probably offering an advantage for identifying liver metastases. Positron emission tomography/CT and endoscopic ultrasound may be helpful for problem solving. Clear and concise reporting of imaging findings is important. Several national organizations are developing templates to standardize the reporting of imaging findings.


Annals of Surgery | 2001

Patient Evaluation and Management With Selective Use of Magnetic Resonance Cholangiography and Endoscopic Retrograde Cholangiopancreatography Before Laparoscopic Cholecystectomy

Terrence H. Liu; Eileen T. Consorti; Akira Kawashima; Eric P. Tamm; Karen L. Kwong; Brijesh S. Gill; Joseph H. Sellin; Eric K. Peden; David W. Mercer

ObjectiveTo assess the utility of triage guidelines for patients with cholelithiasis and suspected choledocholithiasis, incorporating selective use of magnetic resonance cholangiography (MRC) and endoscopic retrograde cholangiopancreatography (ERCP) before laparoscopic cholecystectomy (LC). Summary Background DataERCP is the most frequently used modality for the diagnosis and resolution of choledocholithiasis before LC. MRC has recently emerged as an accurate, noninvasive modality for the detection of choledocholithiasis. However, useful strategies for implementing this diagnostic modality for patient evaluation before LC have not been investigated. MethodsDuring a 16-month period, the authors prospectively evaluated all patients before LC using triage guidelines incorporating patient information obtained from clinical evaluation, serum chemistry analysis, and abdominal ultrasonography. Patients were then assigned to one of four groups based on the level of suspicion for choledocholithiasis (group I, extremely high; group 2, high; group 3, moderate; group 4, low). Group 1 patients underwent ERCP and clearance of common bile duct stones; group 2 patients underwent MRC; group 3 patients underwent LC with intraoperative cholangiography; and group 4 patients underwent LC without intraoperative cholangiography. ResultsCholedocholithiasis was detected in 43 of 440 patients (9.8%). The occurrence of choledocholithiasis among patients in the four groups were 92.6% (25/27), 32.4% (12/37), 3.8% (2/52), and 0.9% (3/324) for groups 1, 2, 3, and 4, respectively (P < .001). MRC was used for 8.4% (37/440) of patients. Patient triage resulted in the identification of common bile duct stones during preoperative ERCP in 92.3% (36/39) of the patients. Unsuspected common bile duct stones occurred in six patients (1.4%). ConclusionsThe probability of choledocholithiasis can be accurately assessed based on information obtained during the initial noninvasive evaluation. Stratification of risks for choledocholithiasis facilitates patient management with the most appropriate diagnostic studies and interventions, thereby improving patient care and resource utilization.


Journal of Clinical Investigation | 2014

Transport properties of pancreatic cancer describe gemcitabine delivery and response

Eugene J. Koay; Mark J. Truty; Vittorio Cristini; Ryan M. Thomas; Rong Chen; Deyali Chatterjee; Ya’an Kang; Priya Bhosale; Eric P. Tamm; Christopher H. Crane; Milind Javle; Matthew H. Katz; Vijaya Gottumukkala; Marc A. Rozner; Haifa Shen; J. E. Lee; Huamin Wang; Yuling Chen; William Plunkett; James L. Abbruzzese; Robert A. Wolff; Gauri R. Varadhachary; Mauro Ferrari; Jason B. Fleming

BACKGROUND The therapeutic resistance of pancreatic ductal adenocarcinoma (PDAC) is partly ascribed to ineffective delivery of chemotherapy to cancer cells. We hypothesized that physical properties at vascular, extracellular, and cellular scales influence delivery of and response to gemcitabine-based therapy. METHODS We developed a method to measure mass transport properties during routine contrast-enhanced CT scans of individual human PDAC tumors. Additionally, we evaluated gemcitabine infusion during PDAC resection in 12 patients, measuring gemcitabine incorporation into tumor DNA and correlating its uptake with human equilibrative nucleoside transporter (hENT1) levels, stromal reaction, and CT-derived mass transport properties. We also studied associations between CT-derived transport properties and clinical outcomes in patients who received preoperative gemcitabine-based chemoradiotherapy for resectable PDAC. RESULTS Transport modeling of 176 CT scans illustrated striking differences in transport properties between normal pancreas and tumor, with a wide array of enhancement profiles. Reflecting the interpatient differences in contrast enhancement, resected tumors exhibited dramatic differences in gemcitabine DNA incorporation, despite similar intravascular pharmacokinetics. Gemcitabine incorporation into tumor DNA was inversely related to CT-derived transport parameters and PDAC stromal score, after accounting for hENT1 levels. Moreover, stromal score directly correlated with CT-derived parameters. Among 110 patients who received preoperative gemcitabine-based chemoradiotherapy, CT-derived parameters correlated with pathological response and survival. CONCLUSION Gemcitabine incorporation into tumor DNA is highly variable and correlates with multiscale transport properties that can be derived from routine CT scans. Furthermore, pretherapy CT-derived properties correlate with clinically relevant endpoints. TRIAL REGISTRATION Clinicaltrials.gov NCT01276613. FUNDING Lustgarten Foundation (989161), Department of Defense (W81XWH-09-1-0212), NIH (U54CA151668, KCA088084).

Collaboration


Dive into the Eric P. Tamm's collaboration.

Top Co-Authors

Avatar

Priya Bhosale

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jason B. Fleming

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Douglas B. Evans

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Robert A. Wolff

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Gauri R. Varadhachary

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Huamin Wang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jeffrey H. Lee

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Christopher H. Crane

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jeffrey E. Lee

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Matthew H. Katz

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge