145 results on '"Fisher GA"'
Search Results
2. Long-term survivors of gastric cancer: a california population-based study.
- Author
-
Kunz PL, Gubens M, Fisher GA, Ford JM, Lichtensztajn DY, and Clarke CA
- Published
- 2012
- Full Text
- View/download PDF
3. Homicide as a medical outcome: Racial disparity in deaths from assault in us level I and II trauma centers.
- Author
-
Harris AR, Fisher GA, and Thomas SH
- Published
- 2012
- Full Text
- View/download PDF
4. Expression of p16(INK4A) but not hypoxia markers or poly adenosine diphosphate-ribose polymerase is associated with improved survival in patients with pancreatic adenocarcinoma.
- Author
-
Chang DT, Chapman CH, Norton JA, Visser B, Fisher GA, Kunz P, Ford JM, Koong AC, Pai RK, Chang, Daniel T, Chapman, Christopher H, Norton, Jeffrey A, Visser, Brendan, Fisher, George A, Kunz, Pamela, Ford, James M, Koong, Albert C, and Pai, Reetesh K
- Abstract
Background: Pancreatic cancer is associated with mutations in the tumor suppressor gene cyclin-dependent kinase inhibitor 2A (p16(INK4A) ), a regulator of the cell cycle and apoptosis. This study investigates whether immunohistochemical expression of p16(INK4A) as well as hypoxia markers and poly adenosine diphosphate-ribose polymerase (PARP) correlates with survival in patients with resected pancreatic adenocarcinoma.Methods: Seventy-three patients with pancreatic adenocarcinoma who underwent curative resection at Stanford University were included. From the surgical specimens, a tissue microarray was constructed using triplicate tissue cores from the primary tumor and used for immunohistochemical staining for the following markers: carbonic anhydrase IX, dihydrofolate reductase, p16(INK4A) , and PARP1/2. Staining was scored as either positive or negative and percentage positive staining. Staining score was correlated with overall survival (OS) and progression-free survival (PFS).Results: Of the markers tested, only immunohistochemical expression of p16(INK4A) correlated with clinical outcome. On univariate analysis, p16(INK4A) expression in the tumor was associated with improved OS (P = .038) but not PFS (P = .28). The median survival for patients with positive versus negative p16(INK4A) staining was 28.8 months versus 18 months. On multivariate analysis, p16(INK4A) expression was associated with improved OS (P = .026) but not PFS (P = .25). Age (P = .0019) and number of nodes involved (P = .025) were also significant for OS. Adjuvant chemotherapy and margin status did not correlate with OS or PFS.Conclusions: Expression of p16(INK4A) is associated with improved OS in patients with resected pancreatic adenocarcinoma. Further investigation is needed for validation, given conflicting data in the published literature. . [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
5. The effect of oral valspodar (PSC 833), a modulator of multidrug resistance, on the pharmacokinetics of liposomal doxorubicin
- Author
-
Lum, Bl, David Chin, Cho, Cd, Advani, R., Fisher, Ga, Halsey, J., and Sikic, Bi
6. Use of the manual Polybrene test in the routine hospital laboratory
- Author
-
Fisher, GA, primary
- Published
- 1983
- Full Text
- View/download PDF
7. Durable Responses to PD-1 Blockade in Mismatch-Repair Deficient Cancers, Independent of Tissue Origin
- Author
-
Le, DT, primary, Durham, JN, additional, Smith, KN, additional, Wang, H, additional, Bartlett, BR, additional, Aulakh, LK, additional, Lu, S, additional, Kemberling, H, additional, Wilt, C, additional, Luber, BS, additional, Wong, F, additional, Azad, NA, additional, Rucki, AA, additional, Laheru, D, additional, Donehower, R, additional, Zaheer, A, additional, Fisher, GA, additional, Crocenzi, TS, additional, Lee, JJ, additional, Greten, TF, additional, Duffy, AG, additional, Ciombor, KK, additional, Eyring, AD, additional, Lam, BH, additional, Joe, A, additional, Kang, SP, additional, Holdhoff, M, additional, Meyer, C, additional, Zhou, S, additional, Goldberg, RM, additional, Armstrong, D, additional, Bever, KM, additional, Fader, AN, additional, Taube, J, additional, Housseau, F, additional, Spetzler, D, additional, Xiao, N, additional, Pardoll, DM, additional, Papadopoulos, N, additional, Kinzler, KW, additional, Eshleman, JR, additional, Vogelstein, B, additional, Anders, RA, additional, and Diaz Jr, LA, additional
- Full Text
- View/download PDF
8. 18Fluorodeoxyglucose PET is prognostic of progression-free and overall survival in locally advanced pancreas cancer treated with stereotactic radiotherapy.
- Author
-
Schellenberg D, Quon A, Minn AY, Graves EE, Kunz P, Ford JM, Fisher GA, Goodman KA, Koong AC, Chang DT, Schellenberg, Devin, Quon, Andy, Minn, A Yuriko, Graves, Edward E, Kunz, Pamela, Ford, James M, Fisher, George A, Goodman, Karyn A, Koong, Albert C, and Chang, Daniel T
- Abstract
Purpose: This study analyzed the prognostic value of positron emission tomography (PET) for locally advanced pancreas cancer patients undergoing stereotactic body radiotherapy (SBRT).Patients and Methods: Fifty-five previously untreated, unresectable pancreas cancer patients received a single fraction of 25-Gy SBRT sequentially with gemcitabine-based chemotherapy. On the preradiation PET-CT, the tumor was contoured and the maximum standardized uptake value (SUVmax) and metabolic tumor burden (MTB) were calculated using an in-house software application. High-SUVmax and low-SUVmax subgroups were created by categorizing patients above or below the median SUVmax. The analysis was repeated to form high-MTB and low-MTB subgroups as well as clinically relevant subgroups with SUVmax values of <5, 5-10, or >10. Multivariate analysis analyzing SUVmax, MTB, age, chemotherapy cycles, and pretreatment carbohydrate antigen (CA)19-9 was performed.Results: For the entire population, median survival was 12.7 months. Median survival was 9.8 vs.15.3 months for the high- and low- SUVmax subgroups (p <0.01). Similarly, median survival was 10.1 vs. 18.0 months for the high MTB and low MTB subgroups (p <0.01). When clinical SUVmax cutoffs were used, median survival was 6.4 months in those with SUVmax >10, 9.5 months with SUVmax 5.0-10.0, and 17.7 months in those with SUVmax <5 (p <0.01). On multivariate analysis, clinical SUVmax was an independent predictor for overall survival (p = 0.03) and progression-free survival (p = 0.03).Conclusion: PET scan parameters can predict for length of survival in locally advanced pancreas cancer patients. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
9. Circulating KRAS G12D but not G12V is associated with survival in metastatic pancreatic ductal adenocarcinoma.
- Author
-
Till JE, McDaniel L, Chang C, Long Q, Pfeiffer SM, Lyman JP, Padrón LJ, Maurer DM, Yu JX, Spencer CN, Gherardini PF, Da Silva DM, LaVallee TM, Abbott C, Chen RO, Boyle SM, Bhagwat N, Cannas S, Sagreiya H, Li W, Yee SS, Abdalla A, Wang Z, Yin M, Ballinger D, Wissel P, Eads J, Karasic T, Schneider C, O'Dwyer P, Teitelbaum U, Reiss KA, Rahma OE, Fisher GA, Ko AH, Wainberg ZA, Wolff RA, O'Reilly EM, O'Hara MH, Cabanski CR, Vonderheide RH, and Carpenter EL
- Subjects
- Humans, Female, Male, Middle Aged, Aged, Prognosis, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Mutation, Progression-Free Survival, Neoplasm Metastasis, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal mortality, Carcinoma, Pancreatic Ductal blood, Carcinoma, Pancreatic Ductal pathology, Carcinoma, Pancreatic Ductal drug therapy, Proto-Oncogene Proteins p21(ras) genetics, Pancreatic Neoplasms genetics, Pancreatic Neoplasms blood, Pancreatic Neoplasms mortality, Pancreatic Neoplasms pathology, Pancreatic Neoplasms drug therapy, Circulating Tumor DNA blood, Circulating Tumor DNA genetics, Biomarkers, Tumor blood, Biomarkers, Tumor genetics
- Abstract
While high circulating tumor DNA (ctDNA) levels are associated with poor survival for multiple cancers, variant-specific differences in the association of ctDNA levels and survival have not been examined. Here we investigate KRAS ctDNA (ctKRAS) variant-specific associations with overall and progression-free survival (OS/PFS) in first-line metastatic pancreatic ductal adenocarcinoma (mPDAC) for patients receiving chemoimmunotherapy ("PRINCE", NCT03214250), and an independent cohort receiving standard of care (SOC) chemotherapy. For PRINCE, higher baseline plasma levels are associated with worse OS for ctKRAS G12D (log-rank p = 0.0010) but not G12V (p = 0.7101), even with adjustment for clinical covariates. Early, on-therapy clearance of G12D (p = 0.0002), but not G12V (p = 0.4058), strongly associates with OS for PRINCE. Similar results are obtained for the SOC cohort, and for PFS in both cohorts. These results suggest ctKRAS G12D but not G12V as a promising prognostic biomarker for mPDAC and that G12D clearance could also serve as an early biomarker of response., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
10. Advancing clinical trial equity through integration of telehealth and decentralized treatment.
- Author
-
Brown E, Fisher GA Jr, Shelton A, Chang DT, and Pollom E
- Subjects
- Humans, Retrospective Studies, Female, Male, Middle Aged, Travel, Clinical Trials, Phase II as Topic, Health Services Accessibility, Time Factors, Adult, Neoplasms therapy, Neoplasms drug therapy, Antineoplastic Agents economics, Antineoplastic Agents therapeutic use, Antineoplastic Agents administration & dosage, Aged, Telemedicine economics, Telemedicine statistics & numerical data
- Abstract
Innovative strategies to increase clinical trial accessibility and equity are needed. We conducted a retrospective review of a phase II investigator-initiated trial to determine whether the modification of clinical trial design to decentralize study treatment can improve trial accessibility among underrepresented groups. Sociodemographic characteristics, including area deprivation indices, as well as study site travel distance, time, and costs were compared between enrolled participants who received chemotherapy locally and participants who did not. Participants who received chemotherapy locally lived substantially farther from the study site (median = 95.90 vs 25.20 miles, P = .004), faced a greater time burden traveling to the study site (median = 115.00 vs 34.00 minutes, P = .002), and had higher travel-related costs for a single trip to the study site (median = $62.81 vs $16.51, P = .004). This study highlights opportunities for alleviating financial and time burdens associated with clinical trial participation, promoting equity in clinical research. Trial Registration: ClinicalTrials.gov identifier: NCT04380337., (© The Author(s) 2024. Published by Oxford University Press.)
- Published
- 2024
- Full Text
- View/download PDF
11. Inflamed immune phenotype predicts favorable clinical outcomes of immune checkpoint inhibitor therapy across multiple cancer types.
- Author
-
Shen J, Choi YL, Lee T, Kim H, Chae YK, Dulken BW, Bogdan S, Huang M, Fisher GA, Park S, Lee SH, Hwang JE, Chung JH, Kim L, Song H, Pereira S, Shin S, Lim Y, Ahn CH, Kim S, Oum C, Kim S, Park G, Song S, Jung W, Kim S, Bang YJ, Mok TSK, Ali SM, and Ock CY
- Subjects
- Humans, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Retrospective Studies, Biomarkers, Tumor, Phenotype, Tumor Microenvironment, Artificial Intelligence, Brain Neoplasms
- Abstract
Background: The inflamed immune phenotype (IIP), defined by enrichment of tumor-infiltrating lymphocytes (TILs) within intratumoral areas, is a promising tumor-agnostic biomarker of response to immune checkpoint inhibitor (ICI) therapy. However, it is challenging to define the IIP in an objective and reproducible manner during manual histopathologic examination. Here, we investigate artificial intelligence (AI)-based immune phenotypes capable of predicting ICI clinical outcomes in multiple solid tumor types., Methods: Lunit SCOPE IO is a deep learning model which determines the immune phenotype of the tumor microenvironment based on TIL analysis. We evaluated the correlation between the IIP and ICI treatment outcomes in terms of objective response rates (ORR), progression-free survival (PFS), and overall survival (OS) in a cohort of 1,806 ICI-treated patients representing over 27 solid tumor types retrospectively collected from multiple institutions., Results: We observed an overall IIP prevalence of 35.2% and significantly more favorable ORRs (26.3% vs 15.8%), PFS (median 5.3 vs 3.1 months, HR 0.68, 95% CI 0.61 to 0.76), and OS (median 25.3 vs 13.6 months, HR 0.66, 95% CI 0.57 to 0.75) after ICI therapy in IIP compared with non-IIP patients, respectively (p<0.001 for all comparisons). On subgroup analysis, the IIP was generally prognostic of favorable PFS across major patient subgroups, with the exception of the microsatellite unstable/mismatch repair deficient subgroup., Conclusion: The AI-based IIP may represent a practical, affordable, clinically actionable, and tumor-agnostic biomarker prognostic of ICI therapy response across diverse tumor types., Competing Interests: Competing interests: JS and SB received institutional research funding from Lunit, Inc. HS, SP, SSh, YL, CHO, Seulki Kim, CO, Sukjun Kim, GP, SSo, WJ, SA and C-YO are employees of Lunit, Inc. Y-JB. is a Consultant/Advisory Board member for Merck Sharp and Dohme (MSD), Merck Serono, Daiichi-Sankyo, Astellas, Alexo Oncology, Samyang Biopharm, Hanmi, Daewoong, and Amgen, and received institutional research grants for clinical trials from Genentech/Roche, MSD, Merck Serono, Daiichi Sankyo, Astellas, and Amgen in the past 3 years. Other authors declare no potential conflicts of interest., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
12. KRAS Sequence Variation as Prognostic Marker in Patients With Young- vs Late-Onset Colorectal Cancer.
- Author
-
Aljehani MA, Bien J, Lee JSH, Fisher GA Jr, and Lin AY
- Subjects
- Female, Humans, Middle Aged, Adolescent, Proto-Oncogene Proteins p21(ras) genetics, Cross-Sectional Studies, Prognosis, Adenocarcinoma epidemiology, Adenocarcinoma genetics, Colorectal Neoplasms epidemiology, Colorectal Neoplasms genetics
- Abstract
Importance: The understanding of the association between KRAS sequence variation status and clinical outcomes in colorectal cancer (CRC) has evolved over time., Objective: To characterize the association of age at onset, tumor sidedness, and KRAS sequence variation with survival among patients diagnosed with CRC., Design, Setting, and Participants: This cross-sectional study used data extracted from the Surveillance, Epidemiology, and End Results database. Patients diagnosed with adenocarcinoma of the colon or rectum from 2010 through 2015 were included and were classified as having young-onset (YO) cancer if diagnosed between ages 20 to 49 years and late-onset (LO) cancer if diagnosed at age 50 years or older. Data were analyzed from April 2021 through August 2023., Main Outcomes and Measures: CRC cause-specific survival (CSS) was summarized using Fine and Gray cumulative incidence and Kaplan-Meier curves. Estimation of subdistribution hazard ratios (sHRs) for the association of KRAS status, age at onset, and tumor location with CRC CSS was conducted using the Fine and Gray competing risk model. Cox proportional hazards regression was used to estimate and compare HRs., Results: Among 21 661 patients with KRAS sequence variation status (mean [SD] age at diagnosis, 62.50 [13.78] years; 9784 females [45.2%]), 3842 patients had YO CRC, including 1546 patients with KRAS variants, and 17 819 patients had LO CRC, including 7311 patients with KRAS variants. There was a significant difference in median CSS time between patients with variant vs wild-type KRAS (YO: 3.0 years [95% CI, 2.8-3.3 years] vs 3.5 years [95% CI, 3.3-3.9 years]; P = .02; LO: 2.5 years [95% CI, 2.4-2.7 years] vs 3.4 years [95% CI, 3.3-3.6 years]; P < .001). Tumors with variant compared with wild-type KRAS were associated with higher risk of CRC-related death (YO: sHR, 1.09 [95% CI, 1.01-1.18]; P = .03; LO: sHR, 1.06 [95% CI, 1.02-1.09]; P = .002). Among patients with YO cancer, mortality hazards increased by location, from right (sHR, 1.02 [95% CI, 0.88-1.17) to left (sHR, 1.15 [95% CI, 1.02-1.29) and rectum (sHR, 1.16 [95% CI, 0.99-1.36), but no trend by tumor location was seen for LO cancer., Conclusions and Relevance: In this study of patients diagnosed with CRC, KRAS sequence variation was associated with increased mortality among patients with YO and LO tumors. In YO cancer, variant KRAS-associated mortality risk was higher in distal tumors than proximal tumors.
- Published
- 2023
- Full Text
- View/download PDF
13. Paclitaxel With or Without Cixutumumab as Second-Line Treatment of Metastatic Esophageal or Gastroesophageal Junction Cancer: A Randomized Phase II ECOG-ACRIN Trial.
- Author
-
Stockton S, Catalano P, Cohen SJ, Burtness BA, Mitchell EP, Dotan E, Lubner SJ, Kumar P, Mulcahy MF, Fisher GA, Crandall TL, and Benson A
- Subjects
- Humans, Paclitaxel adverse effects, Antibodies, Monoclonal, Humanized therapeutic use, Esophagogastric Junction pathology, Antineoplastic Combined Chemotherapy Protocols adverse effects, Esophageal Neoplasms drug therapy, Esophageal Neoplasms pathology, Stomach Neoplasms drug therapy
- Abstract
Background: Patients with advanced esophageal cancer carry poor prognoses; limited data exist to guide second-line therapy in the metastatic setting. Paclitaxel has been used yet is associated with limited efficacy. There is preclinical evidence of synergy between paclitaxel and cixutumumab, a monoclonal antibody targeting insulin-like growth factor-1 receptor. We conducted a randomized phase II trial of paclitaxel (arm A) versus paclitaxel plus cixutumumab (arm B) in the second-line for patients with metastatic esophageal or gastroesophageal junction (GEJ) cancers., Methods: The primary endpoint was progression-free survival (PFS); 87 patients (43 in arm A, 44 in arm B) were treated., Results: Median PFS was 2.6 months in arm A [90% CL 1.8-3.5] and 2.3 months in arm B [90% 2.0-3.5], P = .86. Stable disease was observed in 29 (33%) patients. Objective response rates for Arms A and B were 12% [90% CI, 5-23%] and 14% [90% CI, 6-25%]. Median overall survival was 6.7 months [90% CL 4.9-9.5] in arm A and 7.2 months [90% CL 4.9-8.1] in arm B, P = 56., Conclusion: The addition of cixutumumab to paclitaxel in second-line therapy of metastatic esophageal/GEJ cancer was well tolerated but did not improve clinical outcomes relative to standard of care (ClinicalTrials.gov Identifier: NCT01142388)., (© The Author(s) 2023. Published by Oxford University Press.)
- Published
- 2023
- Full Text
- View/download PDF
14. A Phase I Trial of the ABCB1 Inhibitor, Oral Valspodar, in Combination With Paclitaxel in Patients With Advanced Solid Tumors.
- Author
-
Fracasso PM, Fisher GA Jr, Goodner SA, Beumer JH, Egorin MJ, Fears CL, Wildi JD, Jones GJ, Pearce TE, and Sikic BI
- Subjects
- Humans, Paclitaxel, ATP Binding Cassette Transporter, Subfamily B therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Neoplasms chemically induced, Cyclosporins adverse effects
- Abstract
Objectives: Multidrug resistance mediated by P-glycoprotein is a potential obstacle to cancer treatment. This phase 1 trial determined the safety of paclitaxel with valspodar, a P-glycoprotein inhibitor, in patients with advanced solid tumors., Methods: Patients were treated with single-agent paclitaxel Q3W 175 mg/m 2 (or 135 mg/m 2 if heavily pretreated) as a 3-hour infusion. If their disease was stable (SD) or progressive (PD), paclitaxel at 30% (52.5 mg/m 2 ), 40% (70 mg/m 2 ), or 50% (87.5 mg/m 2 ) of 175 mg/m 2 (full dose) was administered with valspodar 5 mg/kg orally 4 times daily for 12 doses. Pharmacokinetic sampling (PK) for paclitaxel and valspodar was performed during single-agent and combination therapy., Results: Sixteen patients had SD/PD after one cycle of paclitaxel and then received paclitaxel at 30% (n=3), 40% (n=3), and 50% (n=10) with valspodar. Hematologic adverse events (AEs) including myelosuppression at paclitaxel 40% were comparable to those of full-dose paclitaxel. Non-hematologic AEs consisted of reversible hepatic (hyperbilirubinemia and transaminitis) and neurologic AEs (ataxia and paresthesias). Eleven patients experienced SD with a median of 12.7 weeks (range, 5.4 to 36.0), 4 patients progressed, and 1 was inevaluable. Reduced dose paclitaxel with valspodar resulted in lower plasma peak concentrations of paclitaxel; otherwise, concentrations were similar to single-agent paclitaxel., Conclusion: Paclitaxel at 70 mg/m 2 was administered safely with valspodar. Limited efficacy in hematologic and solid tumors resulted in discontinuation of its clinical development and other transporter inhibitors. Recently, the development of ATP-binding cassette transporter inhibitors has been reconsidered to mitigate resistance to antibody-drug conjugates., Competing Interests: P.M.F. reports previous employment with Bristol Myers Squibb from May 2014 to December 2018 and Adaptimmune LLC from December 2018 to September 2020. She then consulted with Adaptimmune LLC until December 2021. Her work on this clinical study was done while she was in academics at Washington University in St. Louis and occurred prior to her work in pharma. No activities in this submitted work have any relationship to her work at Bristol Myers Squibb or Adaptimmune LLC. G.A.F., Jr. reports that he is the Chair, Data safety Monitoring Board for Astra Zeneca and a member of the Data Safety Monitoring Board of Hutchinson Pharma, and an Advisory Board member for Genentech/Roche and Merck Jan H. Beumer reports no conflicts of interest. Sherry A. Goodner reports previous employment with AbbVie from March 2016 to June 2022. Her work on this clinical trial was done while she was employed at Barnes-Jewish Hospital at Washington University in St. Louis prior to her employment at AbbVie. No activities in this submitted work have any relationship to her work at AbbVie. Jonathan D. Wildi reports no conflicts of interest. Carole L. Fears reports no conflicts of interest. Gary J. Jones reports previous employment with Sandoz/Novartis Pharmaceuticals Corporation from August 1990 to December 2000. He consulted with the company from December 2003 to May 2009 and from February 2012 to February 2013. Tillman E. Pearce was an employee for Novartis during the conduct of this study. He has no other conflicts of interest. B.I.S. reports no conflicts of interest., (Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
15. Validation of a Resectability Scoring System for Prediction of Pancreatic Adenocarcinoma Surgical Outcomes.
- Author
-
Toesca DAS, Susko M, von Eyben R, Baclay JRM, Pollom EL, Jeffrey RB, Poullos PD, Poultsides GA, Fisher GA Jr, Visser BC, Koong AC, Feng M, and Chang DT
- Subjects
- Humans, Retrospective Studies, Treatment Outcome, Pancreatic Neoplasms, Pancreatic Neoplasms diagnostic imaging, Pancreatic Neoplasms surgery, Pancreatic Neoplasms pathology, Adenocarcinoma diagnostic imaging, Adenocarcinoma surgery, Adenocarcinoma pathology
- Abstract
Background: The most used pancreatic cancer (PC) resectability criteria are descriptive in nature or based solely on dichotomous degree of involvement (< 180° or > 180°) of vessels, which allows for a high degree of subjectivity and inconsistency., Methods: Radiographic measurements of the circumferential degree and length of tumor contact with major peripancreatic vessels were retrospectively obtained from pre-treatment multi-detector computed tomography (MDCT) images from PC patients treated between 2001 and 2015 at two large academic institutions. Arterial and venous scores were calculated for each patient, then tested for a correlation with tumor resection and R0 resection., Results: The analysis included 466 patients. Arterial and venous scores were highly predictive of resection and R0 resection in both the training (n = 294) and validation (n = 172) cohorts. A recursive partitioning tree based on arterial and venous score cutoffs developed with the training cohort was able to stratify patients of the validation cohort into discrete groups with distinct resectability probabilities. A refined recursive partitioning tree composed of three resectability groups was generated, with probabilities of resection and R0 resection of respectively 94 and 73% for group A, 61 and 35% for group B, and 4 and 2% for group C. This resectability scoring system (RSS) was highly prognostic, predicting median overall survival times of 27, 18.9, and 13.5 months respectively for patients in RSS groups A, B, and C (p < 0.001)., Conclusions: The proposed RSS was highly predictive of resection, R0 resection, and prognosis for patients with PC when tested against an external dataset., (© 2023. Society of Surgical Oncology.)
- Published
- 2023
- Full Text
- View/download PDF
16. Complete Remission of Widely Metastatic Human Epidermal Growth Factor Receptor 2-Amplified Pancreatic Adenocarcinoma After Precision Immune and Targeted Therapy With Description of Sequencing and Organoid Correlates.
- Author
-
King DA, Smith AR, Pineda G, Nakano M, Michelini F, Goedegebuure SP, Thyparambil S, Liao WL, McCormick A, Ju J, Cioffi M, Zhang X, Hundal J, Griffith M, Grandori C, Pollastro M, Rosati R, Margossian A, Chatterjee P, Ainge T, Flory M, Ocampo P, Chen LM, Poultsides GA, Baron AD, Chang DT, Herman JM, Gillanders WE, Park H, Hoos WA, Nichols M, Fisher GA, and Kuo CJ
- Subjects
- Humans, Receptor, ErbB-2 metabolism, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Adenocarcinoma drug therapy, Adenocarcinoma genetics
- Published
- 2023
- Full Text
- View/download PDF
17. Randomized Study of Temozolomide or Temozolomide and Capecitabine in Patients With Advanced Pancreatic Neuroendocrine Tumors (ECOG-ACRIN E2211).
- Author
-
Kunz PL, Graham NT, Catalano PJ, Nimeiri HS, Fisher GA, Longacre TA, Suarez CJ, Martin BA, Yao JC, Kulke MH, Hendifar AE, Shanks JC, Shah MH, Zalupski MM, Schmulbach EL, Reidy-Lagunes DL, Strosberg JR, O'Dwyer PJ, and Benson AB 3rd
- Subjects
- Humans, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Capecitabine therapeutic use, Dacarbazine therapeutic use, Prospective Studies, Retrospective Studies, Temozolomide therapeutic use, Treatment Outcome, Neuroendocrine Tumors drug therapy, Pancreatic Neoplasms drug therapy
- Abstract
Purpose: Patients with advanced pancreatic neuroendocrine tumors (NETs) have few treatment options that yield objective responses. Retrospective and small prospective studies suggest that capecitabine and temozolomide are associated with high response rates (RRs) and long progression-free survival (PFS)., Patients and Methods: E2211 was a multicenter, randomized, phase II trial comparing temozolomide versus capecitabine/temozolomide in patients with advanced low-grade or intermediate-grade pancreatic NETs. Key eligibility criteria included progression within the preceding 12 months and no prior temozolomide, dimethyl-triazeno-imidazole-carboxamide or dacarbazine, capecitabine or fluorouracil. The primary end point was PFS; secondary endpoints were overall survival, RR, safety, and methylguanine methyltransferase (MGMT) by immunohistochemistry and promoter methylation., Results: A total of 144 patients were enrolled between April 2013 and March 2016 to temozolomide (n = 72) or capecitabine and temozolomide (n = 72); the primary analysis population included 133 eligible patients. At the scheduled interim analysis in January 2018, the median PFS was 14.4 months for temozolomide versus 22.7 months for capecitabine/temozolomide (hazard ratio = 0.58), which was sufficient to reject the null hypothesis for the primary end point (stratified log-rank P = .022). In the final analysis (May 2021), the median overall survival was 53.8 months for temozolomide and 58.7 months for capecitabine/temozolomide (hazard ratio = 0.82, P = .42). MGMT deficiency was associated with response., Conclusion: The combination of capecitabine/temozolomide was associated with a significant improvement in PFS compared with temozolomide alone in patients with advanced pancreatic NETs. The median PFS and RR observed with capecitabine/temozolomide are the highest reported in a randomized study for pancreatic NETs. MGMT deficiency was associated with response, and although routine MGMT testing is not recommended, it can be considered for select patients in need of objective response (ClinicalTrials.gov identifier: NCT01824875).
- Published
- 2023
- Full Text
- View/download PDF
18. Therapeutic Implications of Oncogenic Missense HER2 ( ERBB2 ) Mutations in Gastric Adenocarcinoma.
- Author
-
King DA, Weiel JJ, Reyes R, Mills M, Itchon A, Fisher GA, Ford JM, and Suarez CJ
- Subjects
- Humans, Mutation, Receptor, ErbB-2 genetics, Adenocarcinoma drug therapy
- Published
- 2023
- Full Text
- View/download PDF
19. Late-Onset Immunotherapy-Induced Myocarditis 2 Years After Checkpoint Inhibitor Initiation.
- Author
-
Nguyen AT, Berry GJ, Witteles RM, Le DT, Wu SM, Fisher GA, and Zhu H
- Abstract
Competing Interests: Dr Zhu is supported by the Sarnoff Scholar Career Development Award through the Sarnoff Cardiovascular Research Foundation. Dr Berry has received a speaker honorarium from Merck Pharmaceuticals. Dr Witteles has served on advisory boards for Pfizer, Alnylam, Eidos, Ionis, Intelia, and Janssen. Dr Le has served on advisory boards for Merck, Bristol Myers Squibb, and Janssen; has received research funding from Merck, Bristol Myers Squibb, Aduro Biotech, Curegenix, Medivir, Nouscom, and Abbvie; has received speaking honoraria from Merck; and is an inventor of licensed intellectual property from and managed by Johns Hopkins University. Dr Wu has received research funding from Sanofi; has a patent on induced pluripotent stem cell cardiomyocytes; and has received other expenses from Amgen. Dr Fisher has served on advisory boards for Merck and Genentech and Data Safety Monitoring Boards for Astra Zeneca, Hutchison Pharma and CytomX. All other authors have reported that they have no relationships relevant to the contents of this paper to disclose.
- Published
- 2022
- Full Text
- View/download PDF
20. A phase II study of sapanisertib (TAK-228) a mTORC1/2 inhibitor in rapalog-resistant advanced pancreatic neuroendocrine tumors (PNET): ECOG-ACRIN EA2161.
- Author
-
Rajdev L, Lee JW, Libutti SK, Benson AB, Fisher GA Jr, Kunz PL, Hendifar AE, Catalano P, and O'Dwyer PJ
- Subjects
- Humans, Mechanistic Target of Rapamycin Complex 1 metabolism, MTOR Inhibitors, Protein Kinase Inhibitors adverse effects, Sirolimus, Neuroendocrine Tumors drug therapy, Neuroendocrine Tumors pathology, Pancreatic Neoplasms pathology, Neuroectodermal Tumors, Primitive drug therapy
- Abstract
This was a two-stage phase II trial of a mTORC1/2 inhibitor (mTORC: mammalian target of rapamycin complex) Sapanisertib (TAK228) in patients with rapalog-resistant pancreatic neuroendocrine tumors (PNETs) (NCT02893930). Approved rapalogs such as everolimus inhibit mTORC1 and have limited clinical activity, possibly due to compensatory feedback loops. Sapanisertib addresses the potential for incomplete inhibition of the mTOR pathway through targeting of both mTORC1 and mTORC2, and thus to reverse resistance to earlier rapamycin analogues. In stage 1, patients received sapanisertib 3 mg by mouth once daily on a continuous dosing schedule in 28-day cycle. This trial adopted a two-stage design with the primary objective of evaluating objective tumor response. The first stage would recruit 13 patients in order to accrue 12 eligible and treated patients. If among the 12 eligible patients at least 1 patient had an objective response to therapy, the study would move to the second stage of accrual where 25 eligible and treated patients would be enrolled. This study activated on February 1, 2017, the required pre-determined number of patients (n = 13) had entered by November 5, 2018 for the first stage response evaluation. The accrual of this trial was formally terminated on December 27, 2019 as no response had been observed after the first stage accrual. Treatment-related grade 3 adverse events were reported in eight (61%) patients with hyperglycemia being the most frequent, in three patients (23%). Other toxicities noted in the trial included fatigue, rash diarrhea, nausea, and vomiting. The median PFS was 5.19 months (95% CI [3.84, 9.30]) and the median OS was 20.44 months (95% CI [5.65, 22.54]). Due to the lack of responses in Stage 1 of the study, the study did not proceed to stage 2. Thus the potential to reverse resistance was not evident., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
21. Evaluation of Liver and Renal Toxicity in Peptide Receptor Radionuclide Therapy for Somatostatin Receptor Expressing Tumors: A 2-Year Follow-Up.
- Author
-
Duan H, Ferri V, Fisher GA, Shaheen S, Davidzon GA, Iagaru A, and Mari Aparici C
- Subjects
- Follow-Up Studies, Humans, Liver pathology, Octreotide adverse effects, Positron-Emission Tomography, Radioisotopes adverse effects, Radionuclide Imaging, Receptors, Somatostatin, Hypoalbuminemia chemically induced, Neuroendocrine Tumors drug therapy, Neuroendocrine Tumors radiotherapy, Renal Insufficiency chemically induced
- Abstract
Background: Peptide receptor radionuclide therapy (PRRT) with radiolabeled somatostatin receptor (SSR) analogs is now an established systemic treatment for neuroendocrine tumors (NET). However, more short- and long-term data about renal and hepatotoxicity is needed. Here we present our experience in this clinical scenario., Methods: Eighty-six patients with progressive SSR-expressing malignancies underwent PRRT with Lu-177 Dotatate and were followed up for up to 2 years. Laboratory tests were done 1 week before each cycle and every 2 months at follow-up. Hepatic and renal toxicity was determined based on NCI CTCAE V5.0., Results: 55/86 (64%) patients completed all 4 cycles of PRRT; 18/86 (20.9%) are currently being treated; 13/86 (15.1%) had to discontinue PRRT: 4/13 (31%) due to hematologic toxicity, 9/13 (69%) due to non-PRRT-related comorbidities. Out of the patients who finished treatment, only transient grade 2 toxicities were observed during PRRT: hypoalbuminemia in 5.5% (3/55), and renal toxicity (serum creatinine and estimated glomerular filtration rate) in 1.8% (1/55). No grade 3 or 4 liver and renal toxicity occurred. Patients presenting with impaired liver or renal function prior to PRRT, either improved or had stable findings. No deterioration was observed., Conclusion: Peptide receptor radionuclide therapy does not have a negative impact on liver and renal function, even in patients with pre-existing impaired parameters. No grade 3 or 4 hepatic or renal toxicity was identified. Only transient grade 2 hypoalbuminemia in 5.5% and nephrotoxicity in 1.8% of patients were seen during PRRT., (© The Author(s) 2022. Published by Oxford University Press.)
- Published
- 2022
- Full Text
- View/download PDF
22. Development and Validation of a Deep Learning CT Signature to Predict Survival and Chemotherapy Benefit in Gastric Cancer: A Multicenter, Retrospective Study.
- Author
-
Jiang Y, Jin C, Yu H, Wu J, Chen C, Yuan Q, Huang W, Hu Y, Xu Y, Zhou Z, Fisher GA Jr, Li G, and Li R
- Subjects
- Aged, Chemotherapy, Adjuvant, Disease-Free Survival, Female, Humans, Male, Middle Aged, Neoplasm Staging, Nomograms, Predictive Value of Tests, Prognosis, Retrospective Studies, Stomach Neoplasms pathology, Deep Learning, Stomach Neoplasms diagnostic imaging, Stomach Neoplasms drug therapy, Tomography, X-Ray Computed
- Abstract
Objective: We aimed to develop a deep learning-based signature to predict prognosis and benefit from adjuvant chemotherapy using preoperative computed tomography (CT) images., Background: Current staging methods do not accurately predict the risk of disease relapse for patients with gastric cancer., Methods: We proposed a novel deep neural network (S-net) to construct a CT signature for predicting disease-free survival (DFS) and overall survival in a training cohort of 457 patients, and independently tested it in an external validation cohort of 1158 patients. An integrated nomogram was constructed to demonstrate the added value of the imaging signature to established clinicopathologic factors for individualized survival prediction. Prediction performance was assessed with respect to discrimination, calibration, and clinical usefulness., Results: The DeLIS was associated with DFS and overall survival in the overall validation cohort and among subgroups defined by clinicopathologic variables, and remained an independent prognostic factor in multivariable analysis (P< 0.001). Integrating the imaging signature and clinicopathologic factors improved prediction performance, with C-indices: 0.792-0.802 versus 0.719-0.724, and net reclassification improvement 10.1%-28.3%. Adjuvant chemotherapy was associated with improved DFS in stage II patients with high-DeLIS [hazard ratio = 0.362 (95% confidence interval 0.149-0.882)] and stage III patients with high- and intermediate-DeLIS [hazard ratio = 0.611 (0.442-0.843); 0.633 (0.433-0.925)]. On the other hand, adjuvant chemotherapy did not affect survival for patients with low-DeLIS, suggesting a predictive effect (Pinteraction = 0.048, 0.016 for DFS in stage II and III disease)., Conclusions: The proposed imaging signature improved prognostic prediction and could help identify patients most likely to benefit from adjuvant chemotherapy in gastric cancer., Competing Interests: The authors report no conflicts of interest., (Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
23. Durable response after immune checkpoint inhibitor-related diabetes in mismatch repair-deficient pancreatic cancer.
- Author
-
Tsang ES, Walker EJ, Carnevale J, Fisher GA, and Ko AH
- Subjects
- DNA Mismatch Repair drug effects, Humans, Male, Middle Aged, Adenocarcinoma drug therapy, Antibodies, Monoclonal, Humanized adverse effects, Carcinoma, Pancreatic Ductal drug therapy, Diabetes Mellitus chemically induced, Immune Checkpoint Inhibitors adverse effects, Pancreatic Neoplasms drug therapy
- Abstract
Mismatch repair protein deficiency occurs in 0.8-2% of pancreatic ductal adenocarcinomas and confers susceptibility to immunotherapy. Herein, we report the case of a patient with Lynch syndrome-associated, locally advanced mismatch repair protein deficiency pancreatic ductal adenocarcinomas who demonstrated a sustained response to second-line treatment with pembrolizumab, but eventually developed immune-related diabetic ketoacidosis requiring discontinuation of treatment. He has since remained in remission, off treatment, over the following 3 years, with regular surveillance showing no clinical or radiographic evidence of disease progression. The patient's unusual disease course raises the question of whether this serious immune-related adverse event affecting the organ of malignant involvement may have predicted his remarkable and durable response.
- Published
- 2021
- Full Text
- View/download PDF
24. Implementation of a cloud-based electronic patient-reported outcome (ePRO) platform in patients with advanced cancer.
- Author
-
Generalova O, Roy M, Hall E, Shah SA, Cunanan K, Fardeen T, Velazquez B, Chu G, Bruzzone B, Cabot A, Fisher GA, Srinivas S, Fan AC, Haraldsdottir S, Wakelee HA, Neal JW, Padda SK, Johnson T, Heestand GM, Hsieh RW, and Ramchandran K
- Abstract
Background: Patient reported outcomes (PROs) have been associated with improved symptom management and quality of life in patients with cancer. However, the implementation of PROs in an academic clinical practice has not been thoroughly described. Here we report on the execution, feasibility and healthcare utilization outcomes of an electronic PRO (ePRO) application for cancer patients at an academic medical center., Methods: We conducted a randomized trial comparing an experimental ePRO arm to standard of care in patients with advanced cancer in the thoracic, gastrointestinal, and genitourinary oncology groups at Stanford Cancer Center from March 2018 to November 2019. We describe the pre-implementation, implementation, and post-implementation phases of the ePRO arm, technological barriers, electronic health record (EHR) integration, clinician burden, and patient data privacy and security. Feasibility was pre-specified to be at least 70% completion of all questionnaires. Acceptability was based on patient and clinician feedback. Ambulatory healthcare utilization was assessed by reviewing numbers of phone messages, electronic portal messages, and referrals for supportive care., Results: Of 617 ePRO questionnaires sent to 72 patients, 445 (72%) were completed. Most clinicians (87.5%) and patients (93%) felt neutral or positive about the ePRO tool's ease of use. Exposure to ePRO did not cause a measurable change in ambulatory healthcare utilization, with a median of less than two phone messages and supportive care referrals, and 5-6 portal messages., Conclusions: Web-based ePRO tools for patients with advanced cancer are feasible and acceptable without increasing clinical burden. Key lessons include the importance of pilot testing, engagement of stakeholders at all levels, and the need for customization by disease group. Future directions for this work include completion of EHR integration, expansion to other centers, and development of integrated workflows for routine clinical practice., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
25. A phase I pharmacokinetic and safety study of Paclitaxel Injection Concentrate for Nano-dispersion (PICN) alone and in combination with carboplatin in patients with advanced solid malignancies and biliary tract cancers.
- Author
-
Ma WW, Zhu M, Lam ET, Diamond JR, Dy GK, Fisher GA, Goff LW, Alberts S, Bui LA, Sanghal A, Kothekar M, Khopade A, Chimote G, Faulkner R, Eckhardt SG, Adjei AA, and Jimeno A
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carboplatin therapeutic use, Cohort Studies, Female, Humans, Male, Maximum Tolerated Dose, Middle Aged, Paclitaxel therapeutic use, Treatment Outcome, Biliary Tract Neoplasms drug therapy, Carboplatin adverse effects, Carboplatin pharmacokinetics, Nanoparticles administration & dosage, Paclitaxel adverse effects, Paclitaxel pharmacokinetics
- Abstract
Purpose: Paclitaxel injection concentrate for nano-dispersion (PICN) is a Cremophor-free, nanotechnology-driven paclitaxel formulation. This phase I study examined the safety, tolerability, pharmacokinetics and maximum tolerated dose (MTD) of PICN alone and in combination with carboplatin. Its early efficacy in unresectable biliary tract cancers (BTCs) was also evaluated., Methods: This multi-center study comprised two parts. Part A contained a dose-escalation cohort following "3 + 3" design using PICN monotherapy in advanced solid tumors (Part A1); Part A2 dose-expansion cohort was then conducted in advanced BTCs due to observed efficacy in Part A1. Part B1 and B2 evaluated escalating dose of PICN with carboplatin in advanced solid tumors. PICN was administered as a 30 min-infusion every 3 weeks without pre-medications for hypersensitivity reactions., Results: Thirty-six patients received PICN monotherapy in Part A and 21 received PICN plus carboplatin in Part B. The MTD of PICN was determined to be 295 mg/m
2 both as a monotherapy and in combination with carboplatin at AUC 5. Dose-proportional exposure in paclitaxel Cmax and AUC was observed overdose range from 175 to 325 mg/m2 for PICN monotherapy and its combination with carboplatin. Carboplatin did not alter PICN exposure. Clinically significant toxicities mainly include neutropenia and peripheral neuropathy. PICN monotherapy yielded a response rate of 20% in unresectable BTCs., Conclusion: This study demonstrated the safety and stable pharmacokinetics of PICN as a monotherapy and in combination with carboplatin. Single-agent PICN showed promising antitumor activity in advanced BTCs, warranting further studies to investigate its role in gastrointestinal cancers.- Published
- 2021
- Full Text
- View/download PDF
26. Pancreatic INI1-deficient undifferentiated rhabdoid carcinoma achieves complete clinical response on gemcitabine and nab-paclitaxel following immediate progression on FOLFIRINOX: a case report.
- Author
-
King DA, Rahalkar S, Bingham DB, and Fisher GA
- Abstract
Introduction: INI1-deficient undifferentiated rhabdoid carcinoma is a rare pancreatic carcinoma for which the optimal treatment is unknown. Pancreatic ductal adenocarcinoma, the most common histology of pancreas cancer, is treated with combination chemotherapy in the advanced setting, a strategy supported by strong evidence in well powered studies. In patients with excellent performance status, first-line treatment usually consists of the three-drug regimen FOLFIRINOX, with the combination of gemcitabine with nab-paclitaxel, typically less toxic than the three-drug regimen, reserved for second-line therapy. Given the lack of published reports describing treatment outcomes for patients with rare forms of pancreatic cancer, the same treatment approach used for pancreatic ductal adenocarcinoma is typically employed., Observation: This case describes a patient with metastatic pancreatic INI1-deficient undifferentiated rhabdoid carcinoma who was primarily resistant to FOLFIRINOX therapy but who then achieved an immediate, marked and sustained response to gemcitabine with nab-paclitaxel., Conclusion: Given the lack of data informing on optimal management of INI1-deficient pancreatic undifferentiated rhabdoid carcinoma, and the exceptional response achieved by gemcitabine with nab-paclitaxel, this case report highlights a surprising and potentially informative anecdote. Additional studies are needed to confirm responses observed in this report which when taken together may strongly influence first-line therapy choice for this rare malignancy. Given the difficult in acquiring sufficient numbers of these rare histologies in any one institution, multi-institution collaboration in studying outcomes of rare pancreatic malignancies is likely essential., Competing Interests: Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at: http://dx.doi.org/10.21037/jgo-20-478). George Fisher reports personal fees from Merck, Taiho, Roche, Terumo and Ipsen; and has served data safety monitoring boards for Astra Zeneca, Hutchison Pharma and Cytom X and Silenseed. The other authors have no conflict of interests to disclose., (2021 Journal of Gastrointestinal Oncology. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
27. Tumour-specific fluorescence-guided surgery for pancreatic cancer using panitumumab-IRDye800CW: a phase 1 single-centre, open-label, single-arm, dose-escalation study.
- Author
-
Lu G, van den Berg NS, Martin BA, Nishio N, Hart ZP, van Keulen S, Fakurnejad S, Chirita SU, Raymundo RC, Yi G, Zhou Q, Fisher GA, Rosenthal EL, and Poultsides GA
- Subjects
- Adult, Aged, Aged, 80 and over, Carcinoma, Pancreatic Ductal diagnostic imaging, Carcinoma, Pancreatic Ductal drug therapy, Female, Humans, Infusions, Intravenous methods, Intraoperative Period, Lymphatic Metastasis diagnostic imaging, Male, Margins of Excision, Middle Aged, Neoplasm Staging methods, Netherlands epidemiology, Peritoneal Neoplasms diagnostic imaging, Peritoneal Neoplasms secondary, Antineoplastic Agents, Immunological administration & dosage, Benzenesulfonates administration & dosage, Carcinoma, Pancreatic Ductal surgery, Indoles administration & dosage, Optical Imaging methods, Pancreatic Neoplasms pathology, Panitumumab administration & dosage
- Abstract
Background: Complete surgical resection remains the primary curative option for pancreatic ductal adenocarcinoma, with positive margins in 30-70% of patients. In this study, we aimed to evaluate the use of intraoperative tumour-specific imaging to enhance a surgeon's ability to detect visually occult cancer in real time., Methods: In this single-centre, open-label, single-arm study, done in the USA, we enrolled patients who had clinically suspicious or biopsy-confirmed pancreatic ductal adenocarcinomas and were scheduled for curative surgery. Eligible patients were 19 years of age or older with a life expectancy of more than 12 weeks and a Karnofsky performance status of at least 70% or an Eastern Cooperative Oncology Group or Zubrod level of one or lower, who were scheduled to undergo curative surgery. Patients were sequentially enrolled into each dosing group and 2-5 days before surgery, patients were intravenously infused with 100 mg of unlabelled panitumumab followed by 25 mg, 50 mg, or 75 mg of the near-infrared fluorescently labelled antibody (panitumumab-IRDye800CW). The primary endpoint was to determine the optimal dose of panitumumab-IRDye800CW in identifying pancreatic ductal adenocarcinomas as measured by tumour-to-background ratio in all patients. The tumour-to-background ratio was defined as the fluorescence signal of the tumour divided by the fluorescence signal of the surrounding healthy tissue. The dose-finding part of this study has been completed. This study is registered with ClinicalTrials.gov, NCT03384238., Findings: Between April, 2018, and July, 2019, 16 patients were screened for enrolment onto the study. Of the 16 screened patients, two (12%) patients withdrew from the study and three (19%) were not eligible; 11 (69%) patients completed the trial, all of whom were clinically diagnosed with pancreatic ductal adenocarcinoma. The mean tumour-to-background ratio of primary tumours was 3·0 (SD 0·5) in the 25 mg group, 4·0 (SD 0·6) in the 50 mg group, and 3·7 (SD 0·4) in the 75 mg group; the optimal dose was identified as 50 mg. Intraoperatively, near-infrared fluorescence imaging provided enhanced visualisation of the primary tumours, metastatic lymph nodes, and small (<2 mm) peritoneal metastasis. Intravenous administration of panitumumab-IRDye800CW at the doses of 25 mg, 50 mg, and 75 mg did not result in any grade 3 or higher adverse events. There were no serious adverse events attributed to panitumumab-IRDye800CW, although four possibly related adverse events (grade 1 and 2) were reported in four patients., Interpretation: To our knowledge, this study presents the first clinical use of panitumumab-IRDye800CW for detecting pancreatic ductal adenocarcinomas and shows that panitumumab-IRDye800CW is safe and feasible to use during pancreatic cancer surgery. Tumour-specific intraoperative imaging might have added value for treatment of patients with pancreatic ductal adenocarcinomas through improved patient selection and enhanced visualisation of surgical margins, metastatic lymph nodes, and distant metastasis., Funding: National Institutes of Health and the Netherlands Organization for Scientific Research., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
28. Immune-Related Adverse Events and Immune Checkpoint Inhibitor Efficacy in Patients with Gastrointestinal Cancer with Food and Drug Administration-Approved Indications for Immunotherapy.
- Author
-
Das S, Ciombor KK, Haraldsdottir S, Pumpalova Y, Sahin IH, Pineda G, Shyr Y, Lin EP, Hsu CY, Chu SK, Goff LW, Cardin DB, Bilen MA, Fisher GA, Wu C, and Berlin J
- Subjects
- Humans, Immunotherapy adverse effects, Prospective Studies, Retrospective Studies, United States, United States Food and Drug Administration, Gastrointestinal Neoplasms drug therapy, Immune Checkpoint Inhibitors
- Abstract
Introduction: Immune-related adverse event (IRAE) onset may represent a clinical biomarker for anti-programmed cell death protein 1 (PD-1) antibody response based on emerging evidence from patients with various advanced malignancies. This phenomenon has not been previously reported in a multidisease cohort of patients with gastrointestinal (GI) cancer with Food and Drug Administration (FDA)-approved indications to receive immune checkpoint inhibitor therapy., Materials and Methods: The study was a multicenter retrospective cohort analysis of 76 patients with GI cancer who had received anti-PD-1 antibodies for FDA-approved indications. The primary and secondary outcomes of the study were progression-free survival (PFS) and overall survival (OS) in patients based upon IRAE presence, respectively. PFS and OS were estimated by the Kaplan-Meier method; a Cox proportional-hazards model adjusted for IRAE onset, patient age, and enrolling institution was used to analyze outcomes., Results: Median PFS and OS were prolonged in patients who experienced IRAEs compared with those who did not experience them (PFS: not reached [NR] vs. 3.9 months [hazard ratio (HR) 0.13, 95% confidence interval (CI) 0.05-0.3, p < .001]; OS: NR vs. 7.4 months [HR 0.11, 95% CI 0.03-0.36, p < .001]). Among patients who experienced IRAEs, there were no significant differences in PFS and OS by either initial IRAE severity, management, or time to onset., Conclusion: Patients with gastrointestinal cancer who experienced IRAEs while on anti-PD-1 antibodies demonstrated significant improvements in PFS and OS compared with their counterparts who did not develop IRAEs. Although these findings add to results from studies in other tumor types, larger prospective studies are needed prior to clinical adoption of IRAE onset as a biomarker for immune checkpoint inhibitor response., Implications for Practice: Predictive clinical biomarkers for immune checkpoint inhibitor response have been understudied in the field of immuno-oncology. Immune-related adverse event onset appears to be one such biomarker. Across tumor types, immune-related adverse event onset has been associated with response to anti-programmed cell death protein 1 (PD-1) antibodies. The results of this study demonstrate this for the first time in patients with gastrointestinal cancer receiving anti-PD-1 antibodies. Before immune-related adverse event onset can be adopted clinically as a predictive biomarker for immune checkpoint inhibitor response, however, larger prospective studies are needed to better understand the nuances between immune-related adverse event characteristics (severity, site, management, timing of onset) and immune checkpoint inhibitor effectiveness., (© AlphaMed Press 2020.)
- Published
- 2020
- Full Text
- View/download PDF
29. Evaluation of Cyclophosphamide/GVAX Pancreas Followed by Listeria-Mesothelin (CRS-207) with or without Nivolumab in Patients with Pancreatic Cancer.
- Author
-
Tsujikawa T, Crocenzi T, Durham JN, Sugar EA, Wu AA, Onners B, Nauroth JM, Anders RA, Fertig EJ, Laheru DA, Reiss K, Vonderheide RH, Ko AH, Tempero MA, Fisher GA, Considine M, Danilova L, Brockstedt DG, Coussens LM, Jaffee EM, and Le DT
- Subjects
- Combined Modality Therapy methods, GPI-Linked Proteins genetics, GPI-Linked Proteins immunology, Kaplan-Meier Estimate, Listeria monocytogenes genetics, Listeria monocytogenes immunology, Mesothelin, Pancreas drug effects, Pancreas immunology, Pancreas pathology, Progression-Free Survival, Response Evaluation Criteria in Solid Tumors, Vaccines, Attenuated administration & dosage, Vaccines, Attenuated adverse effects, Vaccines, Attenuated genetics, Humans, Cancer Vaccines administration & dosage, Cancer Vaccines adverse effects, Cyclophosphamide administration & dosage, Immunotherapy methods, Nivolumab administration & dosage, Nivolumab adverse effects, Pancreatic Neoplasms immunology, Pancreatic Neoplasms mortality, Pancreatic Neoplasms pathology, Pancreatic Neoplasms therapy
- Abstract
Purpose: Two studies in previously treated metastatic pancreatic cancer have been completed combining GVAX pancreas vaccine (GM-CSF-secreting allogeneic pancreatic tumor cells) with cyclophosphamide (Cy) and CRS-207 (live, attenuated Listeria monocytogenes -expressing mesothelin). In the current study, we compared Cy/GVAX followed by CRS-207 with (Arm A) or without nivolumab (Arm B)., Patients and Methods: Patients with pancreatic adenocarcinoma who received one prior therapy for metastatic disease and RECIST measurable disease were randomized 1:1 to receive treatment on Arm A or Arm B. The primary objective was to compare overall survival (OS) between the arms. Additional objectives included assessment of progression-free survival, safety, tumor responses, CA19-9 responses, and immunologic correlates., Results: Ninety-three patients were treated (Arm A, 51; Arm B, 42). The median OS in Arms A and B were 5.9 [95% confidence interval (CI), 4.7-8.6] and 6.1 (95% CI, 3.5-7.0) months, respectively, with an HR of 0.86 (95% CI, 0.55-1.34). Objective responses were seen in 3 patients using immune-related response criteria (4%, 2/51, Arm A; 2%, 1/42, Arm B). The grade ≥3 related adverse event rate, whereas higher in Arm A (35.3% vs. 11.9%) was manageable. Changes in the microenvironment, including increase in CD8
+ T cells and a decrease in CD68+ myeloid cells, were observed in long-term survivors in Arm A only., Conclusions: Although the study did not meet its primary endpoint of improvement in OS of Arm A over Arm B, the OS was comparable with standard therapy. Objective responses and immunologic changes in the tumor microenvironment were evident., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
30. Natural killer cell and stroma abundance are independently prognostic and predict gastric cancer chemotherapy benefit.
- Author
-
Li B, Jiang Y, Li G, Fisher GA Jr, and Li R
- Subjects
- Adenocarcinoma drug therapy, Adult, Aged, Aged, 80 and over, Cohort Studies, Female, Humans, Male, Meta-Analysis as Topic, Middle Aged, Prognosis, Risk Factors, Stomach Neoplasms drug therapy, Young Adult, Adenocarcinoma diagnosis, Endothelial Cells pathology, Fibroblasts pathology, Killer Cells, Natural pathology, Stomach Neoplasms diagnosis, Tumor Microenvironment
- Abstract
BACKGROUNDSpecific features of the tumor microenvironment (TME) may provide useful prognostic information. We conducted a systematic investigation of the cellular composition and prognostic landscape of the TME in gastric cancer.METHODSWe evaluated the prognostic significance of major stromal and immune cells within the TME. We proposed a composite TME-based risk score and tested it in 6 independent cohorts of 1678 patients with gene expression or IHC measurements. Further, we devised a patient classification system based on TME characteristics.RESULTSWe identified NK cells, fibroblasts, and endothelial cells as the most robust prognostic markers. The TME risk score combining these cell types was an independent prognostic factor when adjusted for clinicopathologic variables (gene expression, HR [95% CI], 1.42 [1.22-1.66]; IHC, 1.34 [1.24-1.45], P < 0.0001). Higher TME risk scores consistently associated with worse survival within every pathologic stage (HR range, 2.18-3.11, P < 0.02) and among patients who received surgery only. The TME risk score provided additional prognostic value beyond stage, and combination of the two improved prognostication accuracy (likelihood-ratio test χ2 = 235.4 vs. 187.6, P < 0.0001; net reclassification index, 23%). The TME risk score can predict the survival benefit of adjuvant chemotherapy in nonmetastatic patients (stage I-III) (interaction test, P < 0.02). Patients were divided into 4 TME subtypes that demonstrated distinct genetic and molecular patterns and complemented established genomic and molecular subtypes.CONCLUSIONWe developed and validated a TME-based risk score as an independent prognostic and predictive factor, which has the potential to guide personalized management of gastric cancer.FUNDINGThis project is partially supported by NIH grant 1R01 CA222512.
- Published
- 2020
- Full Text
- View/download PDF
31. Detection of visually occult metastatic lymph nodes using molecularly targeted fluorescent imaging during surgical resection of pancreatic cancer.
- Author
-
Tummers WS, Miller SE, Teraphongphom NT, van den Berg NS, Hasan A, Longacre TA, Fisher GA, Bonsing BA, Vahrmeijer AL, Gambhir SS, Swijnenburg RJ, Rosenthal EL, and Poultsides GA
- Subjects
- Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal secondary, Cetuximab administration & dosage, ErbB Receptors metabolism, Feasibility Studies, Fluorescent Dyes administration & dosage, Humans, Indoles administration & dosage, Lymph Node Excision, Lymph Nodes metabolism, Lymph Nodes surgery, Lymphatic Metastasis, Microscopy, Fluorescence, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Predictive Value of Tests, Prospective Studies, Spectroscopy, Near-Infrared, Treatment Outcome, Carcinoma, Pancreatic Ductal surgery, Intraoperative Care methods, Lymph Nodes pathology, Molecular Imaging, Optical Imaging, Pancreatectomy, Pancreatic Neoplasms surgery
- Abstract
Background: Although most patients with PDAC experience distant failure after resection, a significant portion still present with local recurrence. Intraoperative fluorescent imaging can potentially facilitate the visualization of involved peritumoral LNs and guide the locoregional extent of nodal dissection. Here, the efficacy of targeted intraoperative fluorescent imaging was examined in the detection of metastatic lymph nodes (LNs) during resection of pancreatic ductal adenocarcinoma (PDAC)., Methods: A dose-escalation prospective study was performed to assess feasibility of tumor detection within peripancreatic LNs using cetuximab-IRDye800 in PDAC patients. Fluorescent imaging of dissected LNs was analyzed ex vivo macroscopically and microscopically and fluorescence was correlated with histopathology., Results: A total of 144 LNs (72 in the low-dose and 72 in the high-dose cohort) were evaluated. Detection of metastatic LNs by fluorescence was better in the low-dose (50 mg) cohort, where sensitivity and specificity was 100% and 78% macroscopically, and 91% and 66% microscopically. More importantly, this method was able to detect occult foci of tumor (measuring < 5 mm) with a sensitivity of 88% (15/17 LNs)., Conclusion: This study provides proof of concept that intraoperative fluorescent imaging with cetuximab-IRDye800 can facilitate the detection of peripancreatic lymph nodes often containing subclinical foci of disease., (Copyright © 2018. Published by Elsevier Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
32. Predicting Pancreatic Cancer Resectability and Outcomes Based on an Objective Quantitative Scoring System.
- Author
-
Toesca DAS, Jeffrey RB, von Eyben R, Pollom EL, Poullos PD, Poultsides GA, Fisher GA Jr, Visser BC, Koong AC, and Chang DT
- Subjects
- Adult, Aged, Aged, 80 and over, Decision Making, Female, Humans, Kaplan-Meier Estimate, Male, Margins of Excision, Middle Aged, Pancreatic Neoplasms blood supply, Prognosis, Retrospective Studies, Pancreatectomy methods, Pancreatic Neoplasms diagnostic imaging, Pancreatic Neoplasms surgery, Tomography, X-Ray Computed methods
- Abstract
Objective: To quantitatively assess the probability of tumor resection based on measurements of tumor contact with the major peripancreatic vessels., Methods: This is a retrospective cohort study of pancreatic cancer patients treated between January 2001 and December 2015 in a single academic comprehensive cancer center. Radiographic measurements of the circumferential degree and length of solid tumor contact with major peripancreatic vessels were obtained from diagnostic pancreatic protocol computed tomography images and tested for correlation with tumor resection and margin status., Results: Of 294 patients analyzed, 113 (38%) were resected, with 71 (63%) with negative margins. Based on the individual measurements of vascular involvement, a resectability scoring system (RSS) was created. The RSS correlated strongly with resection (P < 0.0001) and R0 resection (P < 0.0001) probabilities. Moreover, the RSS correlated with overall survival (P < 0.0001) and metastasis-free survival (P < 0.0001), being able to substratify resectable (P = 0.022) and unresectable patients (P = 0.014) into subgroups with different prognosis based on RSS scores., Conclusions: Based on a comprehensive and systematic quantitative approach, we developed a scoring system that demonstrated excellent accuracy to predict tumor resection, surgical margin status, and prognosis.
- Published
- 2019
- Full Text
- View/download PDF
33. First-in-Human, First-in-Class Phase I Trial of the Anti-CD47 Antibody Hu5F9-G4 in Patients With Advanced Cancers.
- Author
-
Sikic BI, Lakhani N, Patnaik A, Shah SA, Chandana SR, Rasco D, Colevas AD, O'Rourke T, Narayanan S, Papadopoulos K, Fisher GA, Villalobos V, Prohaska SS, Howard M, Beeram M, Chao MP, Agoram B, Chen JY, Huang J, Axt M, Liu J, Volkmer JP, Majeti R, Weissman IL, Takimoto CH, Supan D, Wakelee HA, Aoki R, Pegram MD, and Padda SK
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized adverse effects, Antibodies, Monoclonal, Humanized immunology, Antibodies, Monoclonal, Humanized pharmacokinetics, Antineoplastic Agents, Immunological administration & dosage, Antineoplastic Agents, Immunological adverse effects, Antineoplastic Agents, Immunological immunology, Antineoplastic Agents, Immunological pharmacokinetics, Biopsy, CD47 Antigen immunology, Cohort Studies, Female, Humans, Lymphoma immunology, Lymphoma metabolism, Lymphoma pathology, Male, Middle Aged, Neoplasms immunology, Neoplasms metabolism, Neoplasms pathology, Antibodies, Monoclonal, Humanized administration & dosage, Lymphoma drug therapy, Neoplasms drug therapy
- Abstract
Purpose: To evaluate the safety, pharmacokinetics, and pharmacodynamics of Hu5F9-G4 (5F9), a humanized IgG4 antibody that targets CD47 to enable phagocytosis., Patients and Methods: Adult patients with solid tumors were treated in four cohorts: part A, to determine a priming dose; part B, to determine a weekly maintenance dose; part C, to study a loading dose in week 2; and a tumor biopsy cohort., Results: Sixty-two patients were treated: 11 in part A, 14 in B, 22 in C, and 15 in the biopsy cohort. Part A used doses that ranged from 0.1 to 3 mg/kg. On the basis of tolerability and receptor occupancy studies that showed 100% CD47 saturation on RBCs, 1 mg/kg was selected as the priming dose. In subsequent groups, patients were treated with maintenance doses that ranged from 3 to 45 mg/kg, and most toxicities were mild to moderate. These included transient anemia (57% of patients), hemagglutination on peripheral blood smear (36%), fatigue (64%), headaches (50%), fever (45%), chills (45%), hyperbilirubinemia (34%), lymphopenia (34%), infusion-related reactions (34%), and arthralgias (18%). No maximum tolerated dose was reached with maintenance doses up to 45 mg/kg. At doses of 10 mg/kg or more, the CD47 antigen sink was saturated by 5F9, and a 5F9 half-life of approximately 13 days was observed. Strong antibody staining of tumor tissue was observed in a patient at 30 mg/kg. Two patients with ovarian/fallopian tube cancers had partial remissions for 5.2 and 9.2 months., Conclusion: 5F9 is well tolerated using a priming dose at 1 mg/kg on day 1 followed by maintenance doses of up to 45 mg/kg weekly.
- Published
- 2019
- Full Text
- View/download PDF
34. Effect of Lanreotide Depot/Autogel on Urinary 5-Hydroxyindoleacetic Acid and Plasma Chromogranin A Biomarkers in Nonfunctional Metastatic Enteropancreatic Neuroendocrine Tumors.
- Author
-
Pavel ME, Phan AT, Wolin EM, Mirakhur B, Liyanage N, Pitman Lowenthal S, Fisher GA Jr, and Vinik AI
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents therapeutic use, Female, Follow-Up Studies, Gastrointestinal Neoplasms blood, Gastrointestinal Neoplasms drug therapy, Gastrointestinal Neoplasms urine, Humans, International Agencies, Male, Middle Aged, Neuroendocrine Tumors blood, Neuroendocrine Tumors drug therapy, Neuroendocrine Tumors urine, Pancreatic Neoplasms blood, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms urine, Prognosis, Retrospective Studies, Somatostatin therapeutic use, Survival Rate, Biomarkers, Tumor analysis, Chromogranin A blood, Gastrointestinal Neoplasms secondary, Hydroxyindoleacetic Acid urine, Neuroendocrine Tumors secondary, Pancreatic Neoplasms pathology, Peptides, Cyclic therapeutic use, Somatostatin analogs & derivatives
- Abstract
Background: Urinary 5-hydroxyindoleacetic acid (5-HIAA) is an established biomarker in neuroendocrine tumors and carcinoid syndrome; however, its role in nonfunctional neuroendocrine tumors is not defined. We present post hoc data on urinary 5-HIAA and plasma chromogranin A (CgA) from the CLARINET study., Methods: Patients with well- or moderately differentiated, nonfunctioning, locally advanced or metastatic enteropancreatic neuroendocrine tumors were randomized to deep subcutaneous lanreotide depot/autogel 120 mg or placebo once every 28 days for 96 weeks. Tumor response, evaluated centrally (RECIST 1.0), and progression-free survival (PFS) were assessed by treatment and biochemical response, defined as (a) baseline >upper limit of normal (ULN, 41.6 μmol per day 5-HIAA; 98.1 μg/L CgA) and (b) ≥50% decrease from baseline and to ≤ULN value on study., Results: Forty-eight percent (82 of 171; lanreotide, n = 45; placebo, n = 37) and 66% (129 of 195; lanreotide, n = 65; placebo, n = 64) of randomized patients had 5-HIAA and CgA > ULN at baseline. Among patients with >ULN baseline values who did not progress after 96 weeks of treatment, significantly greater reductions in 5-HIAA and CgA were observed in lanreotide-treated versus placebo-treated patients throughout the study (all p < .05). PFS was significantly prolonged among 5-HIAA responders versus nonresponders (median not reached vs. 16.2 months, p < .0001; hazard ratio [HR] = 0.21, 95% confidence interval [CI], 0.09-0.48) and CgA responders versus nonresponders (median not reached vs. 16.2 months, p = .0070; HR = 0.30, 95% CI, 0.12-0.76), regardless of treatment arm. PFS was also significantly prolonged among lanreotide-treated 5-HIAA responders versus nonresponders ( p = .0071) but was not significantly different among placebo-treated 5-HIAA responders versus nonresponders. There were no significant differences in PFS between lanreotide-treated CgA responders versus nonresponders or between placebo-treated CgA responders versus nonresponders., Conclusions: The 5-HIAA findings are noteworthy because they occurred in patients with nonfunctioning enteropancreatic neuroendocrine tumors. Monitoring 5-HIAA and CgA may be useful when treating patients with nonfunctional neuroendocrine tumors., Implications for Practice: Current guidelines focus only on the monitoring of 5-hydroxyindoleacetic acid (5-HIAA) in the diagnosis and management of functional neuroendocrine tumors with carcinoid syndrome. The current post hoc analysis of patients with nonfunctional enteropancreatic neuroendocrine tumors in the CLARINET study demonstrated that measuring and following both 5-HIAA and chromogranin A as biomarkers of disease progression may be useful in the management of patients with nonfunctional neuroendocrine tumors., Competing Interests: Disclosures of potential conflicts of interest may be found at the end of this article., (© AlphaMed Press 2018.)
- Published
- 2019
- Full Text
- View/download PDF
35. Long-Term Survivors of Pancreatic Cancer: A California Population-Based Study.
- Author
-
Kardosh A, Lichtensztajn DY, Gubens MA, Kunz PL, Fisher GA, and Clarke CA
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, California epidemiology, Child, Child, Preschool, Female, Humans, Infant, Infant, Newborn, Kaplan-Meier Estimate, Male, Middle Aged, Young Adult, Adenocarcinoma epidemiology, Pancreatic Neoplasms epidemiology, Population Surveillance methods, Registries statistics & numerical data
- Abstract
Objectives: Pancreatic cancer continues to carry a poor prognosis with survival rates that have had minimal improvement over the past 4 decades. We report a population-based, comprehensive analysis of long-term survivors of pancreatic adenocarcinoma diagnosed in the diverse population of California., Methods: Data from the California Cancer Registry were used to evaluate long-term survival. A total of 70,442 patients diagnosed with pancreatic adenocarcinoma between 1988 and 2009 were identified. Logistic regression was used to identify factors associated with achieving 5-year survival., Results: The overall 5-year survival was 2.5%, with minimal incremental improvements throughout the 3 decades. Age, stage, degree of differentiation, and surgical resection were associated with 5-year survival. Furthermore, younger age and receiving care at a National Cancer Institute-designated cancer center were similarly correlated with 5-year survival regardless of surgical intervention. In addition, we identified stage, differentiation, and adjuvant chemotherapy as significant factors for long-term survival in surgically resected patients. In the unresectable patients, Asian/Pacific islanders and Hispanics were significantly more likely to reach the 5-year milestone than non-Hispanic whites., Conclusions: Although pancreatic cancer mortality remains high, our study highlights baseline characteristics, treatment, biological factors, and ethnicity that are associated with long-term survival. These findings may serve as a springboard for further investigation.
- Published
- 2018
- Full Text
- View/download PDF
36. BIOCHEMICAL RESPONSES IN SYMPTOMATIC AND ASYMPTOMATIC PATIENTS WITH NEUROENDOCRINE TUMORS: POOLED ANALYSIS OF 2 PHASE 3 TRIALS.
- Author
-
Mirakhur B, Pavel ME, Pommier RF, Fisher GA, Phan AT, Massien C, Liyanage N, Lowenthal SP, and Vinik AI
- Abstract
Objective: Neuroendocrine tumors (NETs) are associated with elevated 5-hydroxyindoleacetic acid (5-HIAA) and chromogranin A (CgA) levels. This study aimed to analyze relationships between urinary 5-HIAA and plasma CgA levels and clinical outcomes., Methods: Centrally assessed biomarker levels and correlations with progression-free survival (PFS) and carcinoid syndrome (CS) symptom control were evaluated in a pooled analysis of CLARINET (96-week randomized, double-blind, placebo-controlled) and ELECT (16-week randomized, double-blind, placebo-controlled, 32-week initial open label and ≥2 year long-term extension open label) studies of adults with NETs, with (ELECT) or without (CLARINET) CS at 97 institutions. Patients were treated with subcutaneous lanreotide depot 120 mg monthly., Results: Of 319 pooled patients, 86% and 95% had baseline 5-HIAA and CgA data, respectively, with 47% and 74% having levels greater than the upper limit of normal (ULN). PFS was longer among patients who experienced a decrease in biomarker levels at week 12, with statistical significance reached in the CgA cohort (not reached vs. 14.4 months; P<.0001). A large proportion (87%) of patients without symptoms of CS in the CLARINET study had detectable levels of 5-HIAA (48% >ULN). In ELECT, patients with CS who received lanreotide and experienced a biochemical response (≥50% decrease from baseline) achieved greater symptom control., Conclusion: This pooled analysis of two randomized, placebo-controlled trials demonstrated that 5-HIAA and CgA are secreted as biochemical biomarkers in many patients with NETs, regardless of clinical syndromes. Significant biochemical response was associated with improved clinical outcomes, as measured by improved PFS or improved CS symptom control., Abbreviations: 5-HIAA = 5-hydroxyindoleacetic acid; CgA = chromogranin A; CI = confidence interval; CLARINET = Controlled Study of Lanreotide Antiproliferative Response in Neuroendocrine Tumors; CS = carcinoid syndrome; ELECT = Evaluation of Lanreotide Depot/Autogel Efficacy and Safety as a Carcinoid Syndrome Treatment; HR = hazard ratio; ITT = intention-to-treat; NET = neuroendocrine tumor; PanNET = pancreatic NET; PFS = progression-free survival; PPI = proton pump inhibitor; SSA = somatostatin analogue; ULN = upper limit of normal.
- Published
- 2018
- Full Text
- View/download PDF
37. Intraoperative Pancreatic Cancer Detection using Tumor-Specific Multimodality Molecular Imaging.
- Author
-
Tummers WS, Miller SE, Teraphongphom NT, Gomez A, Steinberg I, Huland DM, Hong S, Kothapalli SR, Hasan A, Ertsey R, Bonsing BA, Vahrmeijer AL, Swijnenburg RJ, Longacre TA, Fisher GA, Gambhir SS, Poultsides GA, and Rosenthal EL
- Subjects
- Antineoplastic Agents, Immunological chemistry, Carcinoma, Pancreatic Ductal diagnostic imaging, Carcinoma, Pancreatic Ductal surgery, Cetuximab chemistry, Cohort Studies, Follow-Up Studies, Humans, Pancreatic Neoplasms diagnostic imaging, Pancreatic Neoplasms surgery, Prognosis, Spectroscopy, Near-Infrared methods, Carcinoma, Pancreatic Ductal pathology, Fluorescent Dyes chemistry, Intraoperative Care, Molecular Imaging methods, Multimodal Imaging methods, Pancreatic Neoplasms pathology
- Abstract
Background: Operative management of pancreatic ductal adenocarcinoma (PDAC) is complicated by several key decisions during the procedure. Identification of metastatic disease at the outset and, when none is found, complete (R0) resection of primary tumor are key to optimizing clinical outcomes. The use of tumor-targeted molecular imaging, based on photoacoustic and fluorescence optical imaging, can provide crucial information to the surgeon. The first-in-human use of multimodality molecular imaging for intraoperative detection of pancreatic cancer is reported using cetuximab-IRDye800, a near-infrared fluorescent agent that binds to epidermal growth factor receptor., Methods: A dose-escalation study was performed to assess safety and feasibility of targeting and identifying PDAC in a tumor-specific manner using cetuximab-IRDye800 in patients undergoing surgical resection for pancreatic cancer. Patients received a loading dose of 100 mg of unlabeled cetuximab before infusion of cetuximab-IRDye800 (50 mg or 100 mg). Multi-instrument fluorescence imaging was performed throughout the surgery in addition to fluorescence and photoacoustic imaging ex vivo., Results: Seven patients with resectable pancreatic masses suspected to be PDAC were enrolled in this study. Fluorescence imaging successfully identified tumor with a significantly higher mean fluorescence intensity in the tumor (0.09 ± 0.06) versus surrounding normal pancreatic tissue (0.02 ± 0.01), and pancreatitis (0.04 ± 0.01; p < 0.001), with a sensitivity of 96.1% and specificity of 67.0%. The mean photoacoustic signal in the tumor site was 3.7-fold higher than surrounding tissue., Conclusions: The safety and feasibilty of intraoperative, tumor-specific detection of PDAC using cetuximab-IRDye800 with multimodal molecular imaging of the primary tumor and metastases was demonstrated.
- Published
- 2018
- Full Text
- View/download PDF
38. Diagnosis of Systemic Capillary Leak Syndrome in a Young Child Treated with Intravenous Immunoglobulin in the Acute Phase.
- Author
-
Asmundsson ASE, Bjorklund AR, and Fisher GA
- Abstract
Systemic capillary leak syndrome (SCLS) is a potentially life-threatening disorder characterized by distributive shock, hypoalbuminemia, and hemoconcentration. It is exceedingly rare in children with less than 20 cases reported to date. The underlying cause for this syndrome remains largely unknown and acute treatment has remained mainly supportive. Prophylaxis with intravenous immunoglobulin (IVIG) has been shown to successfully prevent further episodes in both adults and children. We present a case of a 2-year-old previously healthy male admitted to the pediatric intensive care unit with a clinical course consistent with SCLS. His shock was refractory to aggressive fluid and vasopressor support. Reversal of SCLS with IVIG given in the acute phase had been described in three adult subjects, and for this reason, the decision was made to administer IVIG. Within an hour of administration, hemodynamics stabilized and vasopressor support could be weaned. He has had no further episodes on prophylactic infusions of IVIG. Although the exact mechanism of IVIG in SCLS is unknown, it has proven to be an effective and safe prophylactic therapy, and in our patient, it drastically reversed the acute capillary leak. We suggest that IVIG should be considered as acute therapy in pediatric patients with refractory shock and a clinical course suggestive of SCLS.
- Published
- 2018
- Full Text
- View/download PDF
39. LANREOTIDE THERAPY IN CARCINOID SYNDROME: PROSPECTIVE ANALYSIS OF PATIENT-REPORTED SYMPTOMS IN PATIENTS RESPONSIVE TO PRIOR OCTREOTIDE THERAPY AND PATIENTS NAÏVE TO SOMATOSTATIN ANALOGUE THERAPY IN THE ELECT PHASE 3 STUDY.
- Author
-
Fisher GA Jr, Wolin EM, Liyanage N, Lowenthal SP, Mirakhur B, Pommier RF, Shaheen M, and Vinik AI
- Subjects
- Adult, Aged, Carcinoid Tumor drug therapy, Double-Blind Method, Female, Humans, Male, Middle Aged, Neoadjuvant Therapy, Neuroendocrine Tumors drug therapy, Octreotide therapeutic use, Self Report, Somatostatin therapeutic use, Treatment Outcome, Malignant Carcinoid Syndrome drug therapy, Peptides, Cyclic therapeutic use, Somatostatin analogs & derivatives
- Abstract
Objective: This ELECT prospective analysis examined lanreotide depot/autogel for carcinoid syndrome (CS) symptom control in patients with neuroendocrine tumors (NETs) who were responsive to prior octreotide (prior octreotide group) compared with patients who were naïve to prior somatostatin analogue treatment (de novo group)., Methods: Adults with histopathologically confirmed NET and stable CS (diarrhea and/or flushing) were randomized to subcutaneous (SC) lanreotide 120 mg or placebo every 4 weeks for 16 weeks. Patients reported diarrhea and/or flushing symptom severity and frequency and short-acting SC octreotide rescue therapy daily using an Interactive Voice/Web Response System. To evaluate the efficacy of lanreotide compared with placebo, the novel primary endpoint of patient-determined use of SC octreotide rescue therapy for breakthrough symptoms was used as a surrogate for symptom control. Clinically meaningful patient-reported treatment benefit was examined using daily patient-reported symptoms of diarrhea and flushing., Results: Of the 115 randomized patients, 51 (n = 26 lanreotide, n = 25 placebo) were octreotide-naïve (de novo) and 64 (n = 33 lanreotide; n = 31 placebo) received prior octreotide. Lanreotide versus placebo patients had a lower mean percentage of days of SC octreotide rescue therapy in de novo and prior octreotide groups (least squares [LS] mean difference -19.1, P = .0477 and -6.9, P = .4332, respectively). The mean percentage of days with moderate/severe diarrhea and/or flushing was lower in lanreotide versus placebo patients in de novo and prior octreotide groups (LS mean difference -14.6, P = .0140 and -10.9, P = .0746, respectively). The transition from octreotide to lanreotide was generally well-tolerated., Conclusion: Improvement in CS symptoms occurred with lanreotide treatment, regardless of prior octreotide use., Abbreviations: CI = confidence interval CS = carcinoid syndrome DB = double blind ELECT = Evaluation of Lanreotide depot/autogel Efficacy and safety as a Carcinoid-syndrome Treatment IOL = initial open-label IVRS/IWRS = interactive voice/web response system LS = least square NET = neuroendocrine tumor OR = odds ratio SC = subcutaneous SSA = somatostatin analogue SSTR = somatostatin receptor TEAE = treatment-emergent adverse event.
- Published
- 2018
- Full Text
- View/download PDF
40. Patient-Reported Symptom Control of Diarrhea and Flushing in Patients with Neuroendocrine Tumors Treated with Lanreotide Depot/Autogel: Results from a Randomized, Placebo-Controlled, Double-Blind and 32-Week Open-Label Study.
- Author
-
Fisher GA Jr, Wolin EM, Liyanage N, Pitman Lowenthal S, Mirakhur B, Pommier RF, Shaheen M, and Vinik A
- Subjects
- Double-Blind Method, Follow-Up Studies, Humans, Neuroendocrine Tumors pathology, Prognosis, Prospective Studies, Somatostatin therapeutic use, Antineoplastic Agents therapeutic use, Diarrhea prevention & control, Flushing prevention & control, Gels therapeutic use, Neuroendocrine Tumors drug therapy, Peptides, Cyclic therapeutic use, Somatostatin analogs & derivatives
- Abstract
Background: In the double-blind (DB) ELECT study, lanreotide depot/autogel significantly reduced versus placebo the need for short-acting octreotide for symptomatic carcinoid syndrome (CS) control in neuroendocrine tumor (NET) patients. Here we present patient-reported symptom data during DB and initial open-label (IOL) treatment., Materials and Methods: Adults with NETs and CS history, with/without prior somatostatin analog use, were randomized to 16 weeks' DB lanreotide 120 mg subcutaneous or placebo every 4 weeks, followed by 32 weeks' IOL lanreotide. Patients recorded diarrhea and/or flushing frequency and severity daily by Interactive Voice (Web) Response System for 1 month prior to randomization and throughout the study., Results: Of 115 patients randomized ( n = 59 lanreotide, n = 56 placebo), 56 lanreotide and 45 placebo patients enrolled in the IOL phase. During DB treatment, least square (LS) mean percentages of days with moderate/severe diarrhea and/or flushing were significantly lower for lanreotide (23.4%) versus placebo (35.8%; LS mean difference [95% confidence interval]: -12.4 [-20.73 to -4.07]; p = .004). For DB lanreotide patients, average daily composite (frequency × severity) diarrhea scores improved significantly between DB and IOL treatment (mean difference: -0.71 [-1.20 to -0.22]; p = .005), and remained stable for diarrhea and/or flushing. For DB placebo patients, composite scores for diarrhea, flushing, and diarrhea and/or flushing improved significantly between DB and IOL treatment (mean differences: -1.07 [-1.65 to -0.49]; -1.06 [-1.93 to -0.19]; and -2.13 [-3.35 to -0.91]; all p ≤ .018)., Conclusion: Improved diarrhea and flushing control in CS patients during 16-week lanreotide treatment was sustained during maintenance of lanreotide treatment for the 32-week IOL phase (48 weeks total)., Implications for Practice: This study prospectively collected daily patient-reported data on diarrhea and flushing from the ELECT trial to evaluate the direct impact of lanreotide depot on patients' relief of carcinoid syndrome symptoms. Treatment with lanreotide depot was associated with significant reductions in the percentages of days patients reported symptoms of diarrhea and flushing, as well as reductions in the frequency and severity of daily symptoms compared with placebo during 16 weeks of double-blind treatment. These improvements were sustained for 32 additional weeks of open-label lanreotide treatment (i.e., through week 48 of treatment), resulting in clinically meaningful, long-term symptom reduction., Competing Interests: Disclosures of potential conflicts of interest may be found at the end of this article., (© AlphaMed Press 2017.)
- Published
- 2018
- Full Text
- View/download PDF
41. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade.
- Author
-
Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, Lu S, Kemberling H, Wilt C, Luber BS, Wong F, Azad NS, Rucki AA, Laheru D, Donehower R, Zaheer A, Fisher GA, Crocenzi TS, Lee JJ, Greten TF, Duffy AG, Ciombor KK, Eyring AD, Lam BH, Joe A, Kang SP, Holdhoff M, Danilova L, Cope L, Meyer C, Zhou S, Goldberg RM, Armstrong DK, Bever KM, Fader AN, Taube J, Housseau F, Spetzler D, Xiao N, Pardoll DM, Papadopoulos N, Kinzler KW, Eshleman JR, Vogelstein B, Anders RA, and Diaz LA Jr
- Subjects
- Adult, Aged, Aged, 80 and over, Antigens, Neoplasm immunology, Brain Neoplasms genetics, Brain Neoplasms mortality, Cell Cycle Checkpoints drug effects, Colorectal Neoplasms genetics, Colorectal Neoplasms mortality, DNA Mismatch Repair, Disease-Free Survival, Female, Humans, Male, Middle Aged, Mutation, Neoplastic Syndromes, Hereditary genetics, Neoplastic Syndromes, Hereditary mortality, Programmed Cell Death 1 Receptor immunology, T-Lymphocytes immunology, Young Adult, Antibodies, Monoclonal, Humanized therapeutic use, Biomarkers, Tumor antagonists & inhibitors, Brain Neoplasms immunology, Brain Neoplasms therapy, Colorectal Neoplasms immunology, Colorectal Neoplasms therapy, Neoplastic Syndromes, Hereditary immunology, Neoplastic Syndromes, Hereditary therapy, Programmed Cell Death 1 Receptor antagonists & inhibitors
- Abstract
The genomes of cancers deficient in mismatch repair contain exceptionally high numbers of somatic mutations. In a proof-of-concept study, we previously showed that colorectal cancers with mismatch repair deficiency were sensitive to immune checkpoint blockade with antibodies to programmed death receptor-1 (PD-1). We have now expanded this study to evaluate the efficacy of PD-1 blockade in patients with advanced mismatch repair-deficient cancers across 12 different tumor types. Objective radiographic responses were observed in 53% of patients, and complete responses were achieved in 21% of patients. Responses were durable, with median progression-free survival and overall survival still not reached. Functional analysis in a responding patient demonstrated rapid in vivo expansion of neoantigen-specific T cell clones that were reactive to mutant neopeptides found in the tumor. These data support the hypothesis that the large proportion of mutant neoantigens in mismatch repair-deficient cancers make them sensitive to immune checkpoint blockade, regardless of the cancers' tissue of origin., (Copyright © 2017 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2017
- Full Text
- View/download PDF
42. Safety, tumor trafficking and immunogenicity of chimeric antigen receptor (CAR)-T cells specific for TAG-72 in colorectal cancer.
- Author
-
Hege KM, Bergsland EK, Fisher GA, Nemunaitis JJ, Warren RS, McArthur JG, Lin AA, Schlom J, June CH, and Sherwin SA
- Subjects
- Aged, B-Lymphocytes immunology, B-Lymphocytes pathology, Carcinoembryonic Antigen blood, Colorectal Neoplasms blood, Colorectal Neoplasms immunology, Colorectal Neoplasms pathology, Dose-Response Relationship, Drug, Female, Gene Expression Regulation, Neoplastic drug effects, Gene Expression Regulation, Neoplastic immunology, Glycoproteins antagonists & inhibitors, Humans, Infusions, Intravenous, Interferon-alpha genetics, Interferon-alpha immunology, Male, Middle Aged, Neoplasm Metastasis, Receptors, Antigen, T-Cell antagonists & inhibitors, T-Lymphocytes pathology, Antigens, Neoplasm immunology, Colorectal Neoplasms drug therapy, Glycoproteins immunology, Receptors, Antigen, T-Cell immunology, T-Lymphocytes immunology
- Abstract
Background: T cells engineered to express chimeric antigen receptors (CARs) have established efficacy in the treatment of B-cell malignancies, but their relevance in solid tumors remains undefined. Here we report results of the first human trials of CAR-T cells in the treatment of solid tumors performed in the 1990s., Methods: Patients with metastatic colorectal cancer (CRC) were treated in two phase 1 trials with first-generation retroviral transduced CAR-T cells targeting tumor-associated glycoprotein (TAG)-72 and including a CD3-zeta intracellular signaling domain (CART72 cells). In trial C-9701 and C-9702, CART72 cells were administered in escalating doses up to 10
10 total cells; in trial C-9701 CART72 cells were administered by intravenous infusion. In trial C-9702, CART72 cells were administered via direct hepatic artery infusion in patients with colorectal liver metastases. In both trials, a brief course of interferon-alpha (IFN-α) was given with each CART72 infusion to upregulate expression of TAG-72., Results: Fourteen patients were enrolled in C-9701 and nine in C-9702. CART72 manufacturing success rate was 100% with an average transduction efficiency of 38%. Ten patients were treated in CC-9701 and 6 in CC-9702. Symptoms consistent with low-grade, cytokine release syndrome were observed in both trials without clear evidence of on target/off tumor toxicity. Detectable, but mostly short-term (≤14 weeks), persistence of CART72 cells was observed in blood; one patient had CART72 cells detectable at 48 weeks. Trafficking to tumor tissues was confirmed in a tumor biopsy from one of three patients. A subset of patients had111 Indium-labeled CART72 cells injected, and trafficking could be detected to liver, but T cells appeared largely excluded from large metastatic deposits. Tumor biomarkers carcinoembryonic antigen (CEA) and TAG-72 were measured in serum; there was a precipitous decline of TAG-72, but not CEA, in some patients due to induction of an interfering antibody to the TAG-72 binding domain of humanized CC49, reflecting an anti-CAR immune response. No radiologic tumor responses were observed., Conclusion: These findings demonstrate the relative safety of CART72 cells. The limited persistence supports the incorporation of co-stimulatory domains in the CAR design and the use of fully human CAR constructs to mitigate immunogenicity.- Published
- 2017
- Full Text
- View/download PDF
43. Patient-reported outcomes of a multicenter phase 2 study investigating gemcitabine and stereotactic body radiation therapy in locally advanced pancreatic cancer.
- Author
-
Rao AD, Sugar EA, Chang DT, Goodman KA, Hacker-Prietz A, Rosati LM, Columbo L, O'Reilly E, Fisher GA, Zheng L, Pai JS, Griffith ME, Laheru DA, Iacobuzio-Donahue CA, Wolfgang CL, Koong A, and Herman JM
- Subjects
- Aged, Cancer Pain, Deoxycytidine therapeutic use, Female, Health Status, Humans, Male, Middle Aged, Pancreatic Neoplasms pathology, Surveys and Questionnaires, Gemcitabine, Antimetabolites, Antineoplastic therapeutic use, Deoxycytidine analogs & derivatives, Pancreatic Neoplasms therapy, Patient Reported Outcome Measures, Quality of Life, Radiosurgery methods
- Abstract
Purpose: We previously reported clinical outcomes and physician-reported toxicity of gemcitabine and hypofractionated stereotactic body radiation therapy (SBRT) in locally advanced pancreatic cancer (LAPC). Here we prospectively investigate the impact of gemcitabine and SBRT on patient-reported quality of life (QoL)., Methods and Materials: Forty-nine LAPC patients received 33 Gy SBRT (6.6 Gy daily fractions) upfront or after ≤3 doses of gemcitabine (1000 mg/m
2 ) followed by gemcitabine until progression. European Organization for Research and Treatment of Cancer QoL core cancer (QLQ-C30) and pancreatic cancer-specific (European Organization for Research and Treatment of Cancer QLQ-PAN26) questionnaires were administered to patients pre-SBRT and at 4 to 6 weeks (first follow-up [1FUP]) and 4 months (2FUP) post-SBRT. Changes in QoL scores were deemed clinically relevant if median changes were at least 5 points in magnitude., Results: Forty-three (88%) patients completed pre-SBRT questionnaires. Of these, 88% and 51% completed questionnaires at 1FUP and 2FUP, respectively. There was no change in global QoL from pre-SBRT to 1FUP (P = .17) or 2FUP (P > .99). Statistical and clinical improvements in pancreatic pain (P = .001) and body image (P = .007) were observed from pre-SBRT to 1FUP. Patients with 1FUP and 2FUP questionnaires reported statistically and clinically improved body image (P = .016) by 4 months. Although pancreatic pain initially demonstrated statistical and clinical improvement (P = .020), scores returned to enrollment levels by 2FUP (P = .486). A statistical and clinical decline in role functioning (P = .002) was observed in patients at 2FUP., Conclusions: Global QoL scores are not reduced with gemcitabine and SBRT. In this exploratory analysis, patients experience clinically relevant short-term improvements in pancreatic cancer-specific symptoms. Previously demonstrated acceptable clinical outcomes combined with these favorable QoL data indicate that SBRT can be easily integrated with other systemic therapies and may be a potential standard of care option in patients with LAPC., (Copyright © 2016 American Society for Radiation Oncology. Published by Elsevier Inc. All rights reserved.)- Published
- 2016
- Full Text
- View/download PDF
44. Oxaliplatin-Fluoropyrimidine Chemotherapy Plus Bevacizumab in Advanced Neuroendocrine Tumors: An Analysis of 2 Phase II Trials.
- Author
-
Kunz PL, Balise RR, Fehrenbacher L, Pan M, Venook AP, Fisher GA, Tempero MA, Ko AH, Korn WM, Hwang J, and Bergsland EK
- Subjects
- Antineoplastic Combined Chemotherapy Protocols, Bevacizumab, Capecitabine, Fluorouracil, Humans, Organoplatinum Compounds, Oxaliplatin, Prospective Studies, Neuroendocrine Tumors drug therapy
- Abstract
Objectives: This study aimed to determine the safety and effectiveness of bevacizumab (B) plus FOLFOX or CAPOX in advanced neuroendocrine tumors (NETs) by performing a combined analysis of 2 separate prospective phase II studies., Methods: In the FOLFOX/B study, patients received chemotherapy without scheduled breaks in 3 cohorts: carcinoid, pancreatic NET, and poorly differentiated neuroendocrine carcinomas. In the CAPOX/B study, NET subtypes were pooled, and patients were treated with 4 cycles of CAPOX/B followed by optional maintenance therapy. Primary end points were radiographic response rate (RR) after 12 cycles (FOLFOX/B), progression-free survival (PFS) (CAPOX/B), and toxicity (both)., Results: Seventy-six patients (FOLFOX/B, n = 36; CAPOX/B, n = 40) were included. In FOLFOX/B, RR for carcinoid at 12 cycles 3/22 (13.6%), median PFS 19.3 months; RR for pancreatic NET at 12 cycles 4/12 (41.7%), median PFS 21 months; RR 1/2 (50%) in poorly differentiated neuroendocrine carcinoma; pooled RR 25% and median PFS 21 months (1-year PFS 68%). In CAPOX/B (pooled NET), RR 18% and median PFS 16.7 months (1-year PFS 65%). Predictable toxicity was observed., Conclusions: Neither study met its primary end point, but radiographic responses and prolonged disease stability in previously progressing patients suggest that selected patients with NET may benefit from oxaliplatin-fluoropyrimidine chemotherapy plus bevacizumab and that the combination may warrant further study.
- Published
- 2016
- Full Text
- View/download PDF
45. Neuroendocrine tumors of the pancreas: Degree of cystic component predicts prognosis.
- Author
-
Cloyd JM, Kopecky KE, Norton JA, Kunz PL, Fisher GA, Visser BC, Dua MM, Park WG, and Poultsides GA
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Neoplasm Grading, Neoplasm Invasiveness, Neuroendocrine Tumors mortality, Pancreatectomy, Pancreatic Neoplasms mortality, Retrospective Studies, Survival Rate, Treatment Outcome, Neuroendocrine Tumors pathology, Neuroendocrine Tumors surgery, Pancreatic Neoplasms pathology, Pancreatic Neoplasms surgery
- Abstract
Background: Although most pancreatic neuroendocrine tumors are solid, approximately 10% are cystic. Some studies have suggested that cystic pancreatic neuroendocrine tumors are associated with a more favorable prognosis., Methods: A retrospective review of all patients with pancreatic neuroendocrine tumors who underwent operative resection between 1999 and 2014 at a single academic medical center was performed. Based on cross-sectional imaging performed before operation, pancreatic neuroendocrine tumors were classified according to the size of the cystic component relative to the total tumor size: purely cystic (100%), mostly cystic (≥50%), mostly solid (<50%), and purely solid (0%). Clinicopathologic characteristics and recurrence-free survival were assessed between groups., Results: In the study, 214 patients met inclusion criteria: 8 with purely cystic tumors, 7 with mostly cystic tumors, 15 with mostly solid tumors, and 184 with purely solid tumors. The groups differed in terms of tumor size (1.5 ± 0.5, 3.0 ± 1.7, 3.7 ± 2.6, and 4.0 ± 3.5 cm), lymph node positivity (0%, 0%, 26.7%, and 34.2%), intermediate or high grade (0%, 16.7%, 20.0%, and 31.0%), synchronous liver metastases (0%, 14.3%, 20.0%, and 26.6%) and need for pancreaticoduodenectomy (0%, 0%, 6.7%, and 25.0%), respectively. No cases of purely cystic pancreatic neuroendocrine tumors were associated with synchronous liver or lymph node metastasis, intermediate/high grade, recurrence, or death due to disease. Among patients presenting without metastatic disease, 10-year recurrence-free survival was 100% in patients with purely and mostly cystic tumors versus 53.0% in patients with purely and mostly solid tumors; however, this difference did not reach statistical significance., Conclusion: Pancreatic neuroendocrine tumors demonstrate a spectrum of biologic behavior with an increasing cystic component being associated with more favorable clinicopathologic features and prognosis. Purely cystic pancreatic neuroendocrine tumors may represent 1 subset that can be safely observed without immediate resection., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
46. EVALUATION OF LANREOTIDE DEPOT/AUTOGEL EFFICACY AND SAFETY AS A CARCINOID SYNDROME TREATMENT (ELECT): A RANDOMIZED, DOUBLE-BLIND, PLACEBO-CONTROLLED TRIAL.
- Author
-
Vinik AI, Wolin EM, Liyanage N, Gomez-Panzani E, and Fisher GA
- Subjects
- Aged, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Carcinoid Tumor complications, Double-Blind Method, Drug Compounding, Female, Gels, Humans, Male, Middle Aged, Neuroendocrine Tumors complications, Octreotide administration & dosage, Octreotide adverse effects, Placebos, Somatostatin administration & dosage, Somatostatin adverse effects, Treatment Outcome, Carcinoid Tumor drug therapy, Malignant Carcinoid Syndrome drug therapy, Neuroendocrine Tumors drug therapy, Peptides, Cyclic administration & dosage, Peptides, Cyclic adverse effects, Somatostatin analogs & derivatives
- Abstract
Objective: To evaluate the efficacy and safety of lanreotide depot/autogel 120 mg for the control of carcinoid syndrome (CS) symptoms in patients with neuroendocrine tumors (NETs)., Methods: This was a 16-week, randomized, double-blind, phase 3 trial (Clinicaltrials.gov: NCT00774930). Patients with/without prior somatostatin analog (SSA) use were randomized to lanreotide depot/autogel 120 mg or placebo every 4 weeks, with access to short-acting octreotide as rescue medication. The primary endpoint was the percentage of days in which short-acting octreotide was used, which was assessed from daily diaries using an analysis of covariance including the stratification variables baseline short-acting octreotide use and frequency of diarrhea/flushing. The proportions of patients experiencing treatment success was a supportive analysis. Adverse events were recorded at all visits., Results: A total of 115 patients were enrolled (lanreotide, n = 59; placebo, n = 56). The adjusted mean (95% confidence interval [CI]) percentage of days with rescue octreotide use (primary endpoint) was significantly lower in the lanreotide (33.7%; 95% CI, 25.0%-42.4%) versus the placebo group (48.5%; 95% CI, 39.6%-57.4%), representing an absolute difference of -14.8% (95% CI, -26.8% to -2.8%; P = .017). The odds ratio of full/partial treatment success (≤3 days short-acting octreotide use weeks 12 to 15) was significantly greater with lanreotide than placebo (2.4; 95% CI, 1.1-5.3; P = .036). No new safety concerns were identified, and lanreotide was well tolerated., Conclusion: Lanreotide depot/autogel is effective for the control of CS symptoms in patients (SSA-naïve or experienced) with NETs., Abbreviations: AE = adverse event BMI = body mass index CS = carcinoid syndrome ELECT = Evaluating Lanreotide Efficacy and safety as a Carcinoid-syndrome Treatment HRQoL = health-related quality of life LTOLE = long-term open-label extension NET = neuroendocrine tumor OL = open label SSA = somatostatin analog.
- Published
- 2016
- Full Text
- View/download PDF
47. Appropriate customization of radiation therapy for stage II and III rectal cancer: Executive summary of an ASTRO Clinical Practice Statement using the RAND/UCLA Appropriateness Method.
- Author
-
Goodman KA, Patton CE, Fisher GA, Hoffe SE, Haddock MG, Parikh PJ, Kim J, Baxter NN, Czito BG, Hong TS, Herman JM, Crane CH, and Hoffman KE
- Subjects
- Humans, Neoplasm Staging, Neoadjuvant Therapy methods, Rectal Neoplasms radiotherapy
- Abstract
Purpose: To summarize results of a Clinical Practice Statement on radiation therapy for stage II-III rectal cancer, which addressed appropriate customization of (neo)adjuvant radiation therapy and use of non-surgical therapy for patients who are inoperable or refuse abdominoperineal resection., Methods and Materials: The RAND/University of California, Los Angeles, Appropriateness Method was applied to combine current evidence with multidisciplinary expert opinion. A systematic literature review was conducted and used by the expert panel to rate appropriateness of radiation therapy options for different clinical scenarios. Treatments were categorized by median rating as Appropriate, May Be Appropriate, or Rarely Appropriate., Results: In the neoadjuvant setting, chemoradiation was rated Appropriate and the ratings indicated short-course radiation therapy, chemotherapy alone, and no neoadjuvant therapy are potential options in selected patients. However, neoadjuvant endorectal brachytherapy was rated Rarely Appropriate. For adjuvant therapy, chemoradiation (plus ≥4 months of chemotherapy) was rated Appropriate and chemotherapy alone May Be Appropriate for most scenarios. For medically inoperable patients, definitive external beam radiation therapy and chemotherapy alone were rated May Be Appropriate, whereas endorectal brachytherapy and chemoradiation plus endorectal brachytherapy were possible approaches for some scenarios. The last option, definitive chemoradiation, was rated Appropriate to May Be Appropriate based on performance status. Finally, for patients with low-lying tumors refusing abdominoperineal resection, definitive chemoradiation alone, chemoradiation plus endorectal brachytherapy, and chemoradiation plus external beam radiation therapy were all rated Appropriate., Conclusions: This Clinical Practice Statement demonstrated the central role of radiation therapy in stage II-III rectal cancer management and evaluated ways to better individualize its use in the neoadjuvant, adjuvant, and definitive settings. Ongoing trials may clarify areas of continuing uncertainty and allow further customization., (Copyright © 2015 American Society for Radiation Oncology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
48. CDX2 as a Prognostic Biomarker in Stage II and Stage III Colon Cancer.
- Author
-
Dalerba P, Sahoo D, Paik S, Guo X, Yothers G, Song N, Wilcox-Fogel N, Forgó E, Rajendran PS, Miranda SP, Hisamori S, Hutchison J, Kalisky T, Qian D, Wolmark N, Fisher GA, van de Rijn M, and Clarke MF
- Subjects
- Analysis of Variance, Biomarkers, Tumor genetics, CDX2 Transcription Factor, Chemotherapy, Adjuvant, Colonic Neoplasms drug therapy, Colonic Neoplasms metabolism, Colonic Neoplasms pathology, Computational Biology, Databases, Genetic, Disease-Free Survival, Female, Homeodomain Proteins genetics, Humans, Male, Neoplasm Staging, Oligonucleotide Array Sequence Analysis, Prognosis, RNA, Messenger metabolism, Retrospective Studies, Antineoplastic Agents therapeutic use, Biomarkers, Tumor metabolism, Colonic Neoplasms genetics, Gene Expression, Homeodomain Proteins metabolism
- Abstract
Background The identification of high-risk stage II colon cancers is key to the selection of patients who require adjuvant treatment after surgery. Microarray-based multigene-expression signatures derived from stem cells and progenitor cells hold promise, but they are difficult to use in clinical practice. Methods We used a new bioinformatics approach to search for biomarkers of colon epithelial differentiation across gene-expression arrays and then ranked candidate genes according to the availability of clinical-grade diagnostic assays. With the use of subgroup analysis involving independent and retrospective cohorts of patients with stage II or stage III colon cancer, the top candidate gene was tested for its association with disease-free survival and a benefit from adjuvant chemotherapy. Results The transcription factor CDX2 ranked first in our screening test. A group of 87 of 2115 tumor samples (4.1%) lacked CDX2 expression. In the discovery data set, which included 466 patients, the rate of 5-year disease-free survival was lower among the 32 patients (6.9%) with CDX2-negative colon cancers than among the 434 (93.1%) with CDX2-positive colon cancers (hazard ratio for disease recurrence, 3.44; 95% confidence interval [CI], 1.60 to 7.38; P=0.002). In the validation data set, which included 314 patients, the rate of 5-year disease-free survival was lower among the 38 patients (12.1%) with CDX2 protein-negative colon cancers than among the 276 (87.9%) with CDX2 protein-positive colon cancers (hazard ratio, 2.42; 95% CI, 1.36 to 4.29; P=0.003). In both these groups, these findings were independent of the patient's age, sex, and tumor stage and grade. Among patients with stage II cancer, the difference in 5-year disease-free survival was significant both in the discovery data set (49% among 15 patients with CDX2-negative tumors vs. 87% among 191 patients with CDX2-positive tumors, P=0.003) and in the validation data set (51% among 15 patients with CDX2-negative tumors vs. 80% among 106 patients with CDX2-positive tumors, P=0.004). In a pooled database of all patient cohorts, the rate of 5-year disease-free survival was higher among 23 patients with stage II CDX2-negative tumors who were treated with adjuvant chemotherapy than among 25 who were not treated with adjuvant chemotherapy (91% vs. 56%, P=0.006). Conclusions Lack of CDX2 expression identified a subgroup of patients with high-risk stage II colon cancer who appeared to benefit from adjuvant chemotherapy. (Funded by the National Comprehensive Cancer Network, the National Institutes of Health, and others.).
- Published
- 2016
- Full Text
- View/download PDF
49. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency.
- Author
-
Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, Biedrzycki B, Donehower RC, Zaheer A, Fisher GA, Crocenzi TS, Lee JJ, Duffy SM, Goldberg RM, de la Chapelle A, Koshiji M, Bhaijee F, Huebner T, Hruban RH, Wood LD, Cuka N, Pardoll DM, Papadopoulos N, Kinzler KW, Zhou S, Cornish TC, Taube JM, Anders RA, Eshleman JR, Vogelstein B, and Diaz LA Jr
- Subjects
- Adenocarcinoma drug therapy, Adenocarcinoma genetics, Adenocarcinoma secondary, Adult, Aged, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Agents adverse effects, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Disease-Free Survival, Female, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Neoplasm Metastasis genetics, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents therapeutic use, DNA Mismatch Repair, Neoplasm Metastasis drug therapy, Programmed Cell Death 1 Receptor antagonists & inhibitors
- Abstract
Background: Somatic mutations have the potential to encode "non-self" immunogenic antigens. We hypothesized that tumors with a large number of somatic mutations due to mismatch-repair defects may be susceptible to immune checkpoint blockade., Methods: We conducted a phase 2 study to evaluate the clinical activity of pembrolizumab, an anti-programmed death 1 immune checkpoint inhibitor, in 41 patients with progressive metastatic carcinoma with or without mismatch-repair deficiency. Pembrolizumab was administered intravenously at a dose of 10 mg per kilogram of body weight every 14 days in patients with mismatch repair-deficient colorectal cancers, patients with mismatch repair-proficient colorectal cancers, and patients with mismatch repair-deficient cancers that were not colorectal. The coprimary end points were the immune-related objective response rate and the 20-week immune-related progression-free survival rate., Results: The immune-related objective response rate and immune-related progression-free survival rate were 40% (4 of 10 patients) and 78% (7 of 9 patients), respectively, for mismatch repair-deficient colorectal cancers and 0% (0 of 18 patients) and 11% (2 of 18 patients) for mismatch repair-proficient colorectal cancers. The median progression-free survival and overall survival were not reached in the cohort with mismatch repair-deficient colorectal cancer but were 2.2 and 5.0 months, respectively, in the cohort with mismatch repair-proficient colorectal cancer (hazard ratio for disease progression or death, 0.10 [P<0.001], and hazard ratio for death, 0.22 [P=0.05]). Patients with mismatch repair-deficient noncolorectal cancer had responses similar to those of patients with mismatch repair-deficient colorectal cancer (immune-related objective response rate, 71% [5 of 7 patients]; immune-related progression-free survival rate, 67% [4 of 6 patients]). Whole-exome sequencing revealed a mean of 1782 somatic mutations per tumor in mismatch repair-deficient tumors, as compared with 73 in mismatch repair-proficient tumors (P=0.007), and high somatic mutation loads were associated with prolonged progression-free survival (P=0.02)., Conclusions: This study showed that mismatch-repair status predicted clinical benefit of immune checkpoint blockade with pembrolizumab. (Funded by Johns Hopkins University and others; ClinicalTrials.gov number, NCT01876511.).
- Published
- 2015
- Full Text
- View/download PDF
50. Phase 2 multi-institutional trial evaluating gemcitabine and stereotactic body radiotherapy for patients with locally advanced unresectable pancreatic adenocarcinoma.
- Author
-
Herman JM, Chang DT, Goodman KA, Dholakia AS, Raman SP, Hacker-Prietz A, Iacobuzio-Donahue CA, Griffith ME, Pawlik TM, Pai JS, O'Reilly E, Fisher GA, Wild AT, Rosati LM, Zheng L, Wolfgang CL, Laheru DA, Columbo LA, Sugar EA, and Koong AC
- Subjects
- Adenocarcinoma mortality, Adenocarcinoma pathology, Adult, Aged, Aged, 80 and over, Combined Modality Therapy, Deoxycytidine therapeutic use, Disease Progression, Dose Fractionation, Radiation, Female, Follow-Up Studies, Humans, Male, Middle Aged, Neoplasm Staging, Pancreatic Neoplasms mortality, Pancreatic Neoplasms pathology, Prognosis, Prospective Studies, Quality of Life, Survival Rate, Gemcitabine, Adenocarcinoma therapy, Deoxycytidine analogs & derivatives, Pancreatic Neoplasms therapy, Radiosurgery
- Abstract
Background: This phase 2 multi-institutional study was designed to determine whether gemcitabine (GEM) with fractionated stereotactic body radiotherapy (SBRT) results in acceptable late grade 2 to 4 gastrointestinal toxicity when compared with a prior trial of GEM with single-fraction SBRT in patients with locally advanced pancreatic cancer (LAPC)., Methods: A total of 49 patients with LAPC received up to 3 doses of GEM (1000 mg/m(2)) followed by a 1-week break and SBRT (33.0 gray [Gy] in 5 fractions). After SBRT, patients continued to receive GEM until disease progression or toxicity. Toxicity was assessed using the National Cancer Institute Common Terminology Criteria for Adverse Events [version 4.0] and the Radiation Therapy Oncology Group radiation morbidity scoring criteria. Patients completed the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (QLQ-C30) and pancreatic cancer-specific QLQ-PAN26 module before SBRT and at 4 weeks and 4 months after SBRT., Results: The median follow-up was 13.9 months (range, 3.9-45.2 months). The median age of the patients was 67 years and 84% had tumors of the pancreatic head. Rates of acute and late (primary endpoint) grade ≥ 2 gastritis, fistula, enteritis, or ulcer toxicities were 2% and 11%, respectively. QLQ-C30 global quality of life scores remained stable from baseline to after SBRT (67 at baseline, median change of 0 at both follow-ups; P>.05 for both). Patients reported a significant improvement in pancreatic pain (P = .001) 4 weeks after SBRT on the QLQ-PAN26 questionnaire. The median plasma carbohydrate antigen 19-9 (CA 19-9) level was reduced after SBRT (median time after SBRT, 4.2 weeks; 220 U/mL vs 62 U/mL [P<.001]). The median overall survival was 13.9 months (95% confidence interval, 10.2 months-16.7 months). Freedom from local disease progression at 1 year was 78%. Four patients (8%) underwent margin-negative and lymph node-negative surgical resections., Conclusions: Fractionated SBRT with GEM results in minimal acute and late gastrointestinal toxicity. Future studies should incorporate SBRT with more aggressive multiagent chemotherapy., (© 2014 The Authors. Cancer published by Wiley Periodicals, Inc. on behalf of American Cancer Society.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.