D. Laheru
Johns Hopkins University School of Medicine
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by D. Laheru.
Journal of Clinical Oncology | 2009
Christine A. Iacobuzio-Donahue; Baojin Fu; Shinichi Yachida; Mingde Luo; Hisashi Abe; Clark Henderson; Felip Vilardell; Zheng Wang; Jesse Keller; Priya Banerjee; Joseph M. Herman; John L. Cameron; Charles J. Yeo; Marc K. Halushka; James R. Eshleman; Marian Raben; Alison P. Klein; Ralph H. Hruban; Manuel Hidalgo; D. Laheru
PURPOSEnContrary to the extensive data accumulated regarding pancreatic carcinogenesis, the clinical and molecular features characteristic of advanced stage (stage III and IV) disease are unknown. A comprehensive study of pancreatic cancers from patients who have succumbed to their disease has the potential to greatly expand our understanding of the most lethal stage of this disease and identify novel areas for intervention.nnnMATERIALS AND METHODSnRapid autopsies were performed on 76 patients with documented pancreatic cancer. The histologic features of end stage disease were determined and correlated to the stage at initial diagnosis, patterns of failure (locally destructive v metastatic disease) and the status of the KRAS2, TP53, and DPC4 genes.nnnRESULTSnAt autopsy, 30% of patients died with locally destructive pancreatic cancer, and 70% died with widespread metastatic disease. These divergent patterns of failure found at autopsy (locally destructive v metastatic) were unrelated to clinical stage at initial presentation, treatment history, or histopathologic features. However, Dpc4 immunolabeling status of carcinoma tissues harvested at autopsy, a sensitive marker of DPC4 genetic status, was highly correlated with the presence of widespread metastasis but not with locally destructive tumors (P = .007).nnnCONCLUSIONnPancreatic cancers are represented by distinct genetic subtypes with significantly different patterns of failure. Determinations of DPC4 status at initial diagnosis may be of value in stratifying patients into treatment regimens related to local control versus systemic therapy.
Radiation Oncology | 2011
Amol K. Narang; Robert C. Miller; Charles C. Hsu; Sumita Bhatia; Timothy M. Pawlik; D. Laheru; Ralph H. Hruban; J. Zhou; Jordan M. Winter; Michael G. Haddock; John H. Donohue; Richard D. Schulick; Christopher L. Wolfgang; John L. Cameron; Joseph M. Herman
BackgroundThe role of adjuvant chemoradiation therapy for ampullary carcinoma is unknown. Previous literature suggests that certain populations with high risk factors for recurrence may benefit from adjuvant chemoradiation. We combined the experience of two institutions to better delineate which patients may benefit from adjuvant chemoradiation.MethodsPatients who underwent curative surgery for ampullary carcinoma at the Johns Hopkins Hospital (n = 290; 1992-2007) and at the Mayo Clinic (n = 130; 1977-2005) were reviewed. Patients with <60 days of follow-up, metastatic disease at surgery, or insufficient pathologic data were excluded. The final combined study consisted of 186 patients (n = 104 Johns Hopkins, n = 82 Mayo). Most patients received 5-FU based chemoradiation with conformal radiation. Cox proportional hazards models were used for survival analysis.ResultsMedian overall-survival was 39.9 months with 2- and 5-year survival rates of 62.4% and 39.1%. On univariate analysis, adverse prognostic factors for overall survival included T3/T4 stage disease (RR = 1.86, p = 0.002), node positive status (RR = 3.18, p < 0.001), and poor histological grade (RR = 1.69, p = 0.011). Patients who received adjuvant chemoradiation (n = 66) vs. surgery alone (n = 120) showed a higher rate of T3/T4 stage disease (57.6% vs. 30.8%, P < 0.001), lymph node involvement (72.7% vs. 30.0%, P < 0.001), and close or positive margins (4.6% vs. 0.0%, P = 0.019). Five year survival rates among node negative and node positive patients were 58.7% and 18.4% respectively. When compared with surgery alone, use of adjuvant chemoradiation improved survival among node positive patients (mOS 32.1 vs. 15.7 mos, 5 yr OS: 27.5% vs. 5.9%; RR = 0.47, P = 0.004). After adjusting for adverse prognostic factors on multivariate analysis, patients treated with adjuvant chemoradiation demonstrated a significant survival benefit (RR = 0.40, P < 0.001). Disease relapse occurred in 37.1% of all patients, most commonly metastatic disease in the liver or peritoneum.ConclusionsNode-positive patients with resected ampullary adenocarcinoma may benefit from 5-FU based adjuvant chemoradiation. Since a significant proportion of patients develop metastatic disease, there is a need for more effective systemic treatment.
Annals of Surgical Oncology | 2015
Shalini Moningi; Avani S. Dholakia; Siva P. Raman; Amanda Blackford; John L. Cameron; Dung T. Le; Ana De Jesus-Acosta; Amy Hacker-Prietz; Lauren M. Rosati; Ryan K. Assadi; Shirl Dipasquale; Timothy M. Pawlik; Lei Zheng; Matthew J. Weiss; D. Laheru; Christopher L. Wolfgang; Joseph M. Herman
BackgroundStereotactic body radiation therapy (SBRT) is a promising option for patients with pancreatic cancer (PCA); however, limited data support its efficacy. This study reviews our institutional experience of SBRT in the treatment of locally advanced (LAPC) and borderline resectable (BRPC) PCA.MethodsCharts of all PCA patients receiving SBRT at our institution from 2010 to 2014 were reviewed. Most patients received pre-SBRT chemotherapy.xa0Primary endpoints included overall survival (OS) and local progression-free survival (LPFS). Patients received a total dose of 25–33xa0Gy in five fractions.ResultsA total of 88 patients were included in the analysis, 74 with LAPC and 14 with BRPC. The median age at diagnosis was 67.2xa0years, and median follow-up from date of diagnosis for LAPC and BRPC patients was 14.5 and 10.3xa0months, respectively. Median OS from date of diagnosis was 18.4xa0months (LAPC, 18.4 mo; BRPC, 14.4 mo) and median PFS was 9.8xa0months (95xa0% CI 8.0–12.3). Acute toxicity was minimal with only three patients (3.4xa0%) experiencing acute grade ≥3 toxicity. Late grade ≥2 gastrointestinal toxicity was seen in five patients (5.7xa0%). Of the 19 patients (21.6xa0%) who underwent surgery, 79xa0% were LAPC patients and 84xa0% had margin-negative resections.ConclusionsChemotherapy followed by SBRT in patients with LAPC and BRPC resulted in minimal acute and late toxicity. A large proportion of patients underwent surgical resection despite limited radiographic response to therapy. Further refinements in the integration of chemotherapy, SBRT, and surgery might offer additional advancements toward optimizing patient outcomes.
Investigational New Drugs | 2014
Ana De Jesus-Acosta; D. Laheru; Anirban Maitra; John J. Arcaroli; Michelle A. Rudek; Arvind Dasari; Patrick J. Blatchford; Kevin Quackenbush; Wells A. Messersmith
SummaryPurpose The notch pathway is overexpressed in pancreatic adenocarcinoma. RO4929097, an oral inhibitor of the γ-secretase enzyme has been safely given as a single agent in patients with advanced solid tumors. We aimed to evaluate the efficacy of RO4929097 in patients with pancreatic adenocarcinoma (PDA). Methods A two-stage, single-arm Phase II trial was conducted in patients with previously treated metastatic PDA. RO4929097 was administered at a dose of 20xa0mg daily on days 1–3, 8–10 and 15–17 of 21-dayxa0cycles. The primary endpoint was survival at 6xa0months. Secondary endpoints included overall survival (OS), response rate, toxicities, pharmacokinetic and pharmacodynamic analyses. Results Eighteen patients were recruited, 17 in the first stage and one in the 2nd stage. It was decided to stop further enrollment after RO4929097 was discontinued by the sponsor and was no longer a development candidate. Three (25xa0%) of 12 evaluable patients achieved stable disease. The 6-month survival rate was 27.8xa0% (95xa0% CI 9.7–53.5). The median OS was 4.1xa0months (95xa0% CI 2.7–5.8xa0months) and median progression-free survival was 1.5xa0months (95xa0% CI 1.3–1.6xa0months). Pharmacokinetic properties of RO4929097 in patients (nu2009=u20095) with PDA was similar to that previously reported in other patient populations. There was a trend towards a decrease in HeyL (pu2009=u20090.08) gene expression in three patients following study drug administration. Conclusions RO4929097 was well-tolerated in patients with previously treated PDA. Development of RO4929097 has been discontinued, but development of other notch-targeting agents in pancreatic cancer is continuing.
International Journal of Radiation Oncology Biology Physics | 2014
Avani S. Dholakia; Muhammad Chaudhry; Jeffrey Leal; Daniel T. Chang; Siva P. Raman; Amy Hacker-Prietz; Zheng Su; J. Pai; Katharine Oteiza; Mary E. Griffith; Richard Wahl; Erik Tryggestad; Timothy M. Pawlik; D. Laheru; Christopher L. Wolfgang; Albert C. Koong; Joseph M. Herman
PURPOSEnAlthough previous studies have demonstrated the prognostic value of positron emission tomography (PET) parameters in other malignancies, the role of PET in pancreatic cancer has yet to be well established. We analyzed the prognostic utility of PET for patients with locally advanced pancreatic cancer (LAPC) undergoing fractionated stereotactic body radiation therapy (SBRT).nnnMATERIALS AND METHODSnThirty-two patients with LAPC in a prospective clinical trial received up to 3 doses of gemcitabine, followed by 33 Gy in 5 fractions of 6.6 Gy, using SBRT. All patients received a baseline PET scan prior to SBRT (pre-SBRT PET). Metabolic tumor volume (MTV), total lesion glycolysis (TLG), and maximum and peak standardized uptake values (SUVmax and SUVpeak) on pre-SBRT PET scans were calculated using custom-designed software. Disease was measured at a threshold based on the liver SUV, using the equation Livermean + [2 × Liversd]. Median values of PET parameters were used as cutoffs when assessing their prognostic potential through Cox regression analyses.nnnRESULTSnOf the 32 patients, the majority were male (n=19, 59%), 65 years or older (n=21, 66%), and had tumors located in the pancreatic head (n=27, 84%). Twenty-seven patients (84%) received induction gemcitabine prior to SBRT. Median overall survival for the entire cohort was 18.8 months (95% confidence interval [CI], 15.7-22.0). An MTV of 26.8 cm(3) or greater (hazard ratio [HR] 4.46, 95% CI 1.64-5.88, P<.003) and TLG of 70.9 or greater (HR 3.08, 95% CI 1.18-8.02, P<.021) on pre-SBRT PET scan were associated with inferior overall survival on univariate analysis. Both pre-SBRT MTV (HR 5.13, 95% CI 1.19-22.21, P=.029) and TLG (HR 3.34, 95% CI 1.07-10.48, P=.038) remained independently associated with overall survival in separate multivariate analyses.nnnCONCLUSIONSnPre-SBRT MTV and TLG are potential predictive factors for overall survival in patients with LAPC and may assist in tailoring therapy.
Clinical Cancer Research | 2017
Sonya Parpart-Li; Bjarne Bartlett; Maria Popoli; Vilmos Adleff; Laura Tucker; Rebecca Steinberg; Andrew Georgiadis; Jillian Phallen; Julie R. Brahmer; Nilofer A Azad; Ilene Browner; D. Laheru; Victor E. Velculescu; Mark Sausen; Luis A. Diaz
Purpose: Analysis of genomic alterations in cell-free DNA (cfDNA) is evolving as an approach to detect, monitor, and genotype malignancies. Methods to separate the liquid from the cellular fraction of whole blood for circulating tumor DNA (ctDNA) analyses have been largely unstudied, although these may be a critical consideration for assay performance. Experimental Design: To evaluate the influence of blood processing on cfDNA and ctDNA quality and yield, we compared the cfDNA levels in serum with those in plasma. Given the limitations of serum for ctDNA analyses, we evaluated the effects of two plasma processing approaches, K2EDTA and Cell-Free DNA BCT (BCT) tubes, on cfDNA and ctDNA recovery. A total of 45 samples from nine patients with cancer were collected in both tube types. Once collected, blood was processed into plasma immediately or kept at room temperature and processed into plasma at 1, 3, 5, or 7 days. Results: As early as 24 hours after collection, plasma isolated from blood collected in K2EDTA tubes contained an elevated level of cfDNA that increased over time compared with BCT tubes where no significant increase in cfDNA levels was observed. When samples from an additional six patients with cancer, collected in the same manner, were stored at 4°C in K2EDTA tubes over the course of 3 days, total cfDNA and ctDNA levels were comparable between samples collected in BCT tubes. At day 3, there was a trend toward a decrease in ctDNA levels in both tubes that was more pronounced when measuring the mutant allele fraction for cases stored at 4°C in K2EDTA tubes. Conclusions: In summary, methods of blood processing have a strong influence on cfDNA and ctDNA levels and should be a consideration when evaluating ctDNA in peripheral circulation. Clin Cancer Res; 23(10); 2471–7. ©2016 AACR.
Journal of Radiation Oncology | 2013
Avani S. Dholakia; Amy Hacker-Prietz; Aaron T. Wild; Siva P. Raman; Laura D. Wood; Peng Huang; D. Laheru; Lei Zheng; Ana De Jesus-Acosta; Dung T. Le; Richard D. Schulick; Barish H. Edil; Susannah G. Ellsworth; Timothy M. Pawlik; Christine A. Iacobuzio-Donahue; Ralph H. Hruban; John L. Cameron; Elliot K. Fishman; Christopher L. Wolfgang; Joseph M. Herman
ObjectiveNeoadjuvant therapy increases rates of margin-negative resection of borderline resectable pancreatic ductal adenocarcinoma (BL-PDAC). Criteria for BL-PDAC resection following neoadjuvant chemotherapy and radiation therapy (NCRT) have not been clearly defined.MethodsFifty consecutive patients with BL-PDAC who received NCRT from 2007 to 2012 were identified. Computed tomography (CT) scans pre- and post-treatment were centrally reviewed.ResultsTwenty-nine patients (58xa0%) underwent resection following NCRT, while 21 (42xa0%) remained unresected. Patients selected for and successfully undergoing resection were more likely to have better performance status and absence of the following features on pre- and post-treatment CT: superior mesenteric vein/portal vein encasement, superior mesenteric artery involvement, tumor involvement of two or more vessels, and questionable/overt metastases (all pu2009<u20090.05). Tumor volume and degree of tumor–vessel involvement did not significantly change in both groups after NCRT (all pu2009>u20090.05). The median overall survival was 22.9xa0months in resected versus 13.0xa0months in unresected patients (pu2009<u20090.001). Of patients undergoing resection, 93xa0% were margin-negative, 72xa0% were node-negative, and 54xa0% demonstrated moderate pathologic response to NCRT.ConclusionApparent radiographic extent of vascular involvement does not change significantly after NCRT. Patients without metastatic disease should be chosen for surgical exploration based on adequate performance status and lack of disease progression.
International Journal of Radiation Oncology Biology Physics | 2013
Avani S. Dholakia; Rachit Kumar; Siva P. Raman; Joseph A. Moore; Susannah G. Ellsworth; T.R. McNutt; D. Laheru; Elizabeth M. Jaffee; John L. Cameron; Phuoc T. Tran; R. Hobbs; Christopher L. Wolfgang; Joseph M. Herman
PURPOSEnTo generate a map of local recurrences after pancreaticoduodenectomy (PD) for patients with resectable pancreatic ductal adenocarcinoma (PDA) and to model an adjuvant radiation therapy planning treatment volume (PTV) that encompasses a majority of local recurrences.nnnMETHODS AND MATERIALSnConsecutive patients with resectable PDA undergoing PD and 1 or more computed tomography (CT) scans more than 60 days after PD at our institution were reviewed. Patients were divided into 3 groups: no adjuvant treatment (NA), chemotherapy alone (CTA), or chemoradiation (CRT). Cross-sectional scans were centrally reviewed, and local recurrences were plotted to scale with respect to the celiac axis (CA), superior mesenteric artery (SMA), and renal veins on 1 CT scan of a template post-PD patient. An adjuvant clinical treatment volume comprising 90% of local failures based on standard expansions of the CA and SMA was created and simulated on 3 post-PD CT scans to assess the feasibility of this planning approach.nnnRESULTSnOf the 202 patients in the study, 40 (20%), 34 (17%), and 128 (63%) received NA, CTA, and CRT adjuvant therapy, respectively. The rate of margin-positive resections was greater in CRT patients than in CTA patients (28% vs 9%, P=.023). Local recurrence occurred in 90 of the 202 patients overall (45%) and in 19 (48%), 22 (65%), and 49 (38%) in the NA, CTA, and CRT groups, respectively. Ninety percent of recurrences were within a 3.0-cm right-lateral, 2.0-cm left-lateral, 1.5-cm anterior, 1.0-cm posterior, 1.0-cm superior, and 2.0-cm inferior expansion of the combined CA and SMA contours. Three simulated radiation treatment plans using these expansions with adjustments to avoid nearby structures were created to demonstrate the use of this treatment volume.nnnCONCLUSIONSnModified PTVs targeting high-risk areas may improve local control while minimizing toxicities, allowing dose escalation with intensity-modulated or stereotactic body radiation therapy.
Practical radiation oncology | 2013
Aaron T. Wild; Daniel T. Chang; Karyn A. Goodman; D. Laheru; Lei Zheng; Siva P. Raman; Laurie Ann Columbo; Christopher L. Wolfgang; Albert C. Koong; Joseph M. Herman
patients had pre-SBRT FDG-PET/CT scans with documented pretreatment SUVmax. Treatment fractionation consisted of 60Gy in 3 fractions with a median treatment time of six days (range 3-21 days). Local, regional, and distant failures specified from the last day of treatment were evaluated independently according to the terms of RTOG 1021. Tumor control, overalland progression-free survivals were estimated by the Kaplan-Meier method. Cox proportional hazards regression was performed to determine whether SUVmax, age, KPS, gender, tumor size/T stage, or smoking history influenced outcomes. SUVmax was evaluated as both a continuous and as a dichotomous variable using a cutoff of b5 and ≥5. Results: With a median follow-up of 15 months, median OS and PFS were 25.3 and 40.3 months, respectively. In the univariate analysis, pretreatment SUVmax with a cutoff value of 5 predicted for OS and PFS (p = 0.024, each) but did not achieve significance for LC (p = 0.256). On Cox univariate regression analysis, SUV as a dichotomous variable predicted for both OS and PFS (p = 0.027 and p = 0.030, respectively). Defined as a continuous variable, SUVmax continued to predict for OS and PFS (p = 0.003, each), but also predicted LC (p = 0.045) and trended toward significance for DC (p = 0.064). Tumor stage, histology, KPS, and age were significant for OS on univariate analysis and were included in multivariate analyses. On ANOVA test, tumor T-stage and histology were both significantly correlated to SUVmax (p = 0.013 and p b 0.001, respectively). Tumor stage remained a significant predictor of OS on multivariate analysis (p = 0.05), though SUVmax did not predict for OS as a dichotomous or continuous variable (p = 0.209 and p = 0.223, respectively). It did, however, predict for PFS as a continuous variable (p = 0.009) and neared significance as a dichotomous variable (p = 0.053). SUVmax also trended toward significance for local control as a continuous variable (p = 0.076). Conclusions: SUVmax appears to be a statistically and clinically significant independent prognostic marker for progression-free survival in patients with Stage 1 NSCLC treated with SBRT. Prospective studies to more accurately define the role of tumor FDG uptake in the prognosis of NSCLC are warranted.
Radiotherapy and Oncology | 2012
Haoming Qiu; Aaron T. Wild; Hao Wang; Elliot K. Fishman; Ralph H. Hruban; D. Laheru; Rachit Kumar; Amy Hacker-Prietz; Richard Tuli; Erik Tryggestad; Richard D. Schulick; John L. Cameron; Barish H. Edil; Timothy M. Pawlik; Christopher L. Wolfgang; Joseph M. Herman
BACKGROUND AND PURPOSEnThis study seeks to: (a) quantify radiologic-pathologic discrepancy for pancreatic adenocarcinoma by comparing tumor size on conventional computed tomography (C-CT) and 3-dimensional CT (3D-CT) to corresponding pathologic specimens; and (b) to identify clinico-pathologic characteristics predictive of radiologic-pathologic discrepancy to assist radiotherapy planning.nnnMATERIALS AND METHODSnSixty-three patients with pancreatic adenocarcinoma and preoperative C-CT and volume-rendered 3D-CT imaging within 6 weeks of resection were identified. Maximum tumor diameter (MTD) was measured on pathology, C-CT, and 3D-CT and compared for each patient as well as among different clinico-pathologic subgroups.nnnRESULTSnThere was a trend toward C-CT underestimation of MTD compared to final pathology (p=0.08), but no significant difference between 3D-CT MTD and pathology (p=0.54). Pathologic tumor size was significantly underestimated by C-CT in patients with larger pathologic tumor size (>3.0 cm, p=0.0001), smaller tumor size on C-CT (<3.0 cm, p=0.003), higher CA19-9 (>90 U/mL, p=0.008), and location in the pancreatic head (p=0.015). A model for predicting pathologic MTD using C-CT MTD and CA19-9 level was generated.nnnCONCLUSIONSn3D-CT may allow for more accurate contouring of pancreatic tumors than C-CT. Patients with the above clinico-pathologic characteristics may require expanded margins relative to tumor size estimates on C-CT during radiotherapy planning.