104 results on '"Jeffrey T. Yap"'
Search Results
2. Supplementary Figure 5 from Bevacizumab plus Ipilimumab in Patients with Metastatic Melanoma
- Author
-
David McDermott, Maria Gargano, Annick D. Van den Abbeele, Nikhil Ramaiya, Jeffrey T. Yap, Pamela J. DiPiro, Sara Russell, Martin C. Mihm, Elsa F. Velazquez, Scott Rodig, George F. Murphy, Keith T. Flaherty, Philip Friedlander, Nageatte Ibrahim, Michael B. Atkins, Anita Giobbie-Hurder, Wanyong Zeng, Tetsuro Sasada, Jun Zhou, Xinqi Wu, Cecilia Lezcano, Donald Lawrence, and F. Stephen Hodi
- Abstract
PDF file - 68K, A. Axial CT section of the chest at baseline demonstrates two nodules in the right lung, the dominant one in the lower lobe (white arrows). B. Follow-up CT 3 months after start of treatment demonstrates resolution of one of the nodules and no change in the other nodule. C. Axial CT section 8 months after start of treatment demonstrates complete resolution of the dominant lung nodule.
- Published
- 2023
3. Supplementary Figure 2 from Bevacizumab plus Ipilimumab in Patients with Metastatic Melanoma
- Author
-
David McDermott, Maria Gargano, Annick D. Van den Abbeele, Nikhil Ramaiya, Jeffrey T. Yap, Pamela J. DiPiro, Sara Russell, Martin C. Mihm, Elsa F. Velazquez, Scott Rodig, George F. Murphy, Keith T. Flaherty, Philip Friedlander, Nageatte Ibrahim, Michael B. Atkins, Anita Giobbie-Hurder, Wanyong Zeng, Tetsuro Sasada, Jun Zhou, Xinqi Wu, Cecilia Lezcano, Donald Lawrence, and F. Stephen Hodi
- Abstract
PDF file - 74K, High-power H&E and CD31 stained post treatment biopsy of intratumoral vessels. The endothelial cells appear morphologically columnar. CD31 staining is concentrated at the interendothelial junctions (arrows).
- Published
- 2023
4. Supplementary Figure Legends from Bevacizumab plus Ipilimumab in Patients with Metastatic Melanoma
- Author
-
David McDermott, Maria Gargano, Annick D. Van den Abbeele, Nikhil Ramaiya, Jeffrey T. Yap, Pamela J. DiPiro, Sara Russell, Martin C. Mihm, Elsa F. Velazquez, Scott Rodig, George F. Murphy, Keith T. Flaherty, Philip Friedlander, Nageatte Ibrahim, Michael B. Atkins, Anita Giobbie-Hurder, Wanyong Zeng, Tetsuro Sasada, Jun Zhou, Xinqi Wu, Cecilia Lezcano, Donald Lawrence, and F. Stephen Hodi
- Abstract
PDF file - 59K
- Published
- 2023
5. Supplementary Figure 3 from Bevacizumab plus Ipilimumab in Patients with Metastatic Melanoma
- Author
-
David McDermott, Maria Gargano, Annick D. Van den Abbeele, Nikhil Ramaiya, Jeffrey T. Yap, Pamela J. DiPiro, Sara Russell, Martin C. Mihm, Elsa F. Velazquez, Scott Rodig, George F. Murphy, Keith T. Flaherty, Philip Friedlander, Nageatte Ibrahim, Michael B. Atkins, Anita Giobbie-Hurder, Wanyong Zeng, Tetsuro Sasada, Jun Zhou, Xinqi Wu, Cecilia Lezcano, Donald Lawrence, and F. Stephen Hodi
- Abstract
PDF file - 64K, Illustrations of the variation in pathologic responses to treatment. Examples of intermediate and no response.
- Published
- 2023
6. Supplementary Figure 1 from Bevacizumab plus Ipilimumab in Patients with Metastatic Melanoma
- Author
-
David McDermott, Maria Gargano, Annick D. Van den Abbeele, Nikhil Ramaiya, Jeffrey T. Yap, Pamela J. DiPiro, Sara Russell, Martin C. Mihm, Elsa F. Velazquez, Scott Rodig, George F. Murphy, Keith T. Flaherty, Philip Friedlander, Nageatte Ibrahim, Michael B. Atkins, Anita Giobbie-Hurder, Wanyong Zeng, Tetsuro Sasada, Jun Zhou, Xinqi Wu, Cecilia Lezcano, Donald Lawrence, and F. Stephen Hodi
- Abstract
PDF file - 76K, CONSORT Diagram.
- Published
- 2023
7. Supplementary Figure 4 from Bevacizumab plus Ipilimumab in Patients with Metastatic Melanoma
- Author
-
David McDermott, Maria Gargano, Annick D. Van den Abbeele, Nikhil Ramaiya, Jeffrey T. Yap, Pamela J. DiPiro, Sara Russell, Martin C. Mihm, Elsa F. Velazquez, Scott Rodig, George F. Murphy, Keith T. Flaherty, Philip Friedlander, Nageatte Ibrahim, Michael B. Atkins, Anita Giobbie-Hurder, Wanyong Zeng, Tetsuro Sasada, Jun Zhou, Xinqi Wu, Cecilia Lezcano, Donald Lawrence, and F. Stephen Hodi
- Abstract
PDF file - 125K, Individual response of CCR7+CD45RO+ and CCD7-CD45RO+ of CD4+ and CD8+ T cells in melanoma patients treated with ipilimumab (3mg/kg), ipilimumab (3mg/kg) plus bevacizumab, or ipilimumab (10mg/kg) plus bevacizumab.
- Published
- 2023
8. Supplementary Table 3 from Bevacizumab plus Ipilimumab in Patients with Metastatic Melanoma
- Author
-
David McDermott, Maria Gargano, Annick D. Van den Abbeele, Nikhil Ramaiya, Jeffrey T. Yap, Pamela J. DiPiro, Sara Russell, Martin C. Mihm, Elsa F. Velazquez, Scott Rodig, George F. Murphy, Keith T. Flaherty, Philip Friedlander, Nageatte Ibrahim, Michael B. Atkins, Anita Giobbie-Hurder, Wanyong Zeng, Tetsuro Sasada, Jun Zhou, Xinqi Wu, Cecilia Lezcano, Donald Lawrence, and F. Stephen Hodi
- Abstract
PDF file - 39K, Summary of grade 3 and grade 4 events incidence at each dose level for all events and those events possibly, probably, or definitively related.
- Published
- 2023
9. Supplementary Table 1 from Bevacizumab plus Ipilimumab in Patients with Metastatic Melanoma
- Author
-
David McDermott, Maria Gargano, Annick D. Van den Abbeele, Nikhil Ramaiya, Jeffrey T. Yap, Pamela J. DiPiro, Sara Russell, Martin C. Mihm, Elsa F. Velazquez, Scott Rodig, George F. Murphy, Keith T. Flaherty, Philip Friedlander, Nageatte Ibrahim, Michael B. Atkins, Anita Giobbie-Hurder, Wanyong Zeng, Tetsuro Sasada, Jun Zhou, Xinqi Wu, Cecilia Lezcano, Donald Lawrence, and F. Stephen Hodi
- Abstract
PDF file - 111K, Patient Demographics.
- Published
- 2023
10. Supplementary Figure 7 from Bevacizumab plus Ipilimumab in Patients with Metastatic Melanoma
- Author
-
David McDermott, Maria Gargano, Annick D. Van den Abbeele, Nikhil Ramaiya, Jeffrey T. Yap, Pamela J. DiPiro, Sara Russell, Martin C. Mihm, Elsa F. Velazquez, Scott Rodig, George F. Murphy, Keith T. Flaherty, Philip Friedlander, Nageatte Ibrahim, Michael B. Atkins, Anita Giobbie-Hurder, Wanyong Zeng, Tetsuro Sasada, Jun Zhou, Xinqi Wu, Cecilia Lezcano, Donald Lawrence, and F. Stephen Hodi
- Abstract
PDF file - 59K, Kaplan-Meier estimates for time-to-progression in the entire treated population (A) and by treatment cohort (B). Kaplan-Meier estimates for overall survival in the entire treated population (C) and by treatment cohort (D).
- Published
- 2023
11. Supplementary Table 2 from Bevacizumab plus Ipilimumab in Patients with Metastatic Melanoma
- Author
-
David McDermott, Maria Gargano, Annick D. Van den Abbeele, Nikhil Ramaiya, Jeffrey T. Yap, Pamela J. DiPiro, Sara Russell, Martin C. Mihm, Elsa F. Velazquez, Scott Rodig, George F. Murphy, Keith T. Flaherty, Philip Friedlander, Nageatte Ibrahim, Michael B. Atkins, Anita Giobbie-Hurder, Wanyong Zeng, Tetsuro Sasada, Jun Zhou, Xinqi Wu, Cecilia Lezcano, Donald Lawrence, and F. Stephen Hodi
- Abstract
PDF file - 139K, Worst grade adverse event for individual patients that occurred within the first twelve weeks of treatment (induction dose-limiting toxicity period). Adverse events recorded multiple times for any patient are reported only once according to the worst grade.
- Published
- 2023
12. Supplementary Table 4 from Bevacizumab plus Ipilimumab in Patients with Metastatic Melanoma
- Author
-
David McDermott, Maria Gargano, Annick D. Van den Abbeele, Nikhil Ramaiya, Jeffrey T. Yap, Pamela J. DiPiro, Sara Russell, Martin C. Mihm, Elsa F. Velazquez, Scott Rodig, George F. Murphy, Keith T. Flaherty, Philip Friedlander, Nageatte Ibrahim, Michael B. Atkins, Anita Giobbie-Hurder, Wanyong Zeng, Tetsuro Sasada, Jun Zhou, Xinqi Wu, Cecilia Lezcano, Donald Lawrence, and F. Stephen Hodi
- Abstract
PDF file - 43K, Best overall response in ipilimumab plus bevacizumab treated patients per RECIST criteria by treatment cohort.
- Published
- 2023
13. Prospective Pilot Study of 18F-Fluoroestradiol PET/CT in Patients With Invasive Lobular Carcinomas
- Author
-
Matthew F. Covington, John M. Hoffman, Kathryn A. Morton, Brandon Buckway, Kenneth M. Boucher, Regina Rosenthal, Jane Porretta, Kirstyn E. Brownson, Cindy B. Matsen, Christos Vaklavas, John H. Ward, Mei Wei, Saundra S. Buys, Namita Chittoria, Ellen D. Yakish, Zane G. Archibald, Lance D. Burrell, Regan I. Butterfield, and Jeffrey T. Yap
- Subjects
Radiology, Nuclear Medicine and imaging ,General Medicine - Published
- 2023
14. Abstract 355: Examination of systemic myokine concentrations with risk of cachexia in non-metastatic colorectal cancer patients - Results from the ColoCare Study
- Author
-
Jennifer Ose, Ellen Beswick, Simon Ta Van, Richard H. Viskochil, Christy A. Warby, Jeffrey T. Yap, Matthew F. Covington, Anne H. Nguyen, Jordan W. Stanford, Tengda Lin, Anita R. Peoples, Sheetal Hardikar, Christopher I. Li, William M. Grady, David Shibatta, Adetunji T. Toriola, Martin Schneider, Jane C. Figueiredo, Daniel Jeong, Erin M. Siegel, Cornelia M. Ulrich, and Biljana Gigic
- Subjects
Cancer Research ,Oncology - Abstract
BACKGROUND: Cachexia is a multifactorial metabolic syndrome associated with higher risk of mortality. The precise molecular mechanisms and biological pathways involved remain poorly characterized. A specific criterion of cachexia is loss of muscle mass. Myokines affect muscle mass and have profound effects on glucose and lipid metabolism, thus contributing to energy homeostasis and potentially cachexia. Only sparse data for patients with non-metastatic colorectal cancer exist. This study aims to investigate associations of systemic myokine concentrations with onset of cachexia in non-metastatic colorectal cancer patients. METHODS: Serum samples from n=125 colorectal cancer patients (stage I-III) recruited from the ColoCare Study site at Huntsman Cancer Institute were collected prior to surgery (baseline). Assays were run with the Milliplex Human Myokine Magnetic panel containing beads such as FABP3, Oncostatin M, and FGF21. Patients were defined as cachectic, pre-cachectic, or non-cachectic based on the criteria by Fearon et al. based on sex, BMI, and weight loss over a period of six months. ANOVA were applied to analyze associations of myokines with cachexia at 12 month after surgery, adjusted by age at diagnosis, sex, tumor stage, and tumor site. RESULTS: At the 12 months follow-up, 11% of patients were diagnosed with cachexia (n=14), 14% of patients with pre-cachexia (n=18), and 74% of patients were defined as non-cachectic. Patients with cachexia were more likely to be diagnosed with rectal cancer (57%) compared to pre-cachectic (17%) or non-cachectic patients (42%; p ONCLUSIONS: Cachectic patients presented higher FABP3 concentrations (p=0.02) which were associated with increased risk of cachexia, however these results were not significant after adjustment. In this study, cachectic patients were more likely to be diagnosed with rectal cancer. In summary, larger studies are needed to further evaluate FABP3 and other myokines as potential prognostic biomarkers for cachexia and consider differences by tumor site. Citation Format: Jennifer Ose, Ellen Beswick, Simon Ta Van, Richard H. Viskochil, Christy A. Warby, Jeffrey T. Yap, Matthew F. Covington, Anne H. Nguyen, Jordan W. Stanford, Tengda Lin, Anita R. Peoples, Sheetal Hardikar, Christopher I. Li, William M. Grady, David Shibatta, Adetunji T. Toriola, Martin Schneider, Jane C. Figueiredo, Daniel Jeong, Erin M. Siegel, Cornelia M. Ulrich, Biljana Gigic. Examination of systemic myokine concentrations with risk of cachexia in non-metastatic colorectal cancer patients - Results from the ColoCare Study [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 355.
- Published
- 2023
15. Cold‐Activated Brown Adipose Tissue is Associated with Less Cardiometabolic Dysfunction in Young Adults with Obesity
- Author
-
Nicole L. Mihalopoulos, Richard Holubkov, Jeffrey T. Yap, M. Nazeem Nanjee, Britney Beardmore, and John M. Hoffman
- Subjects
Adult ,Male ,medicine.medical_specialty ,Adolescent ,Endocrinology, Diabetes and Metabolism ,Medicine (miscellaneous) ,Adipose tissue ,030209 endocrinology & metabolism ,Article ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Insulin resistance ,Adipose Tissue, Brown ,Fluorodeoxyglucose F18 ,Positron Emission Tomography Computed Tomography ,Internal medicine ,Brown adipose tissue ,medicine ,Humans ,Obesity ,030212 general & internal medicine ,Young adult ,Nutrition and Dietetics ,Adiponectin ,business.industry ,Leptin ,medicine.disease ,medicine.anatomical_structure ,Cardiovascular Diseases ,Female ,business ,Dyslipidemia - Abstract
OBJECTIVE: To test the hypothesis that young adults with obesity and cold-activated brown adipose tissue (BAT) are less likely to have metabolic dysfunction (dyslipidemia, insulin resistance, hypertension) than those without cold-activated BAT. Previous studies have noted a potentially protective effect of BAT and higher adiponectin/leptin ratios, but they acknowledged that the clinical implications of these findings remain uncertain. METHODS: We enrolled 21 females and 23 males with obesity (BMI ≥ 30kg/m(2)) who underwent a 2-hour cooling protocol before FDG-PET/CT scan, to determine the prevalence, volume and FDG uptake of cold-activated BAT. RESULTS: Cold-activated BAT was identified in 43% of participants (11F, 8M); females had greater FDG uptake. Those with cold-activated BAT had lesser degree of metabolic dysfunction. Cold-activated BAT volume correlated with triglycerides (inversely) and adiponectin (concordantly). Body mass adjusted cold-activated BAT activity correlated with HDL cholesterol (concordantly). Males with cold-activated BAT had lower leptin and higher adiponectin/leptin ratios. CONCLUSIONS: We report a high prevalence of cold-activated BAT in our study participants. We suggest that BAT could be important in decreasing metabolic dysfunction among young adults with obesity, making it a potential target for treating metabolically unhealthy obesity. CLINICAL TRIAL REGISTRATION: Trial registry name is STAGES Trial: Study of Adiposity, Growth and Endocrine Stages (STAGES). The clinical trial registration number is NCT01460784. https://clinicaltrials.gov/ct2/show/NCT01460784?term=obesity&cond=brown&draw=8&rank=54
- Published
- 2020
16. The association between body composition, quality of life (QoL), overall survival (OS) and decision to treat (DTT) in patients with metastatic non–small cell lung cancer (mNSCLC)
- Author
-
Hyejung Lee, Ben Haaland, Wallace L. Akerley, Adriana M Coletta, Jeffrey T. Yap, Sonam Puri, and Kathleen Claire Kerrigan
- Subjects
Cancer Research ,Oncology - Abstract
377 Background: Among patients diagnosed with mNSCLC, significant declines in fat and muscle mass are common, yet there is a paucity of data related to body composition, QoL, TD, and survival. Accordingly, we evaluated the relationship between body composition via routine CT scans, patient-reported outcomes (PROs), and overall survival (OS) among a convenience sample of mNSCLC patients. Methods: Data from 80 mNSCLC patients with initial CT scans and NCI PROMIS questionnaires within the first three months of diagnosis were analyzed. Body composition from CT scans (sliceOmatic software) extracted Skeletal muscle (SM), intermuscular adipose tissue (IAT), visceral VAT, and subcutaneous (SAT) in area (cm2) and a discovery set was expressed as HU. PROMIS PROs (pain, fatigue, anxiety, depression, and physical function) were collected in clinic using an iPad as an institutional quality initiative. Results: Median time to OS was 16 months. When stratified by sex, females had longer median survival time (female 25 months, male 14 months). When all body composition variables were considered together, greater amounts of skeletal muscle were linked with a 63% reduction in mortality risk 0.37 (95% CI 0.16, 0.87) in the adjusted model. Per one-unit increase in VAT (area cm2) was linked with a 0.04 unit decrease in pain (95% CI -0.08, -0.01). Associations between HU and clinical were also observed. Greater amount of VAT (mean HU) was linked with 2.57(95% CI 1.10, 6.01) times the odds of death in the crude model. Also, in crude model, one unit of increase in skeletal muscle (mean HU) was associated with 0.3 (95% CI 0.03, 0.57) unit increase in physical functionality, and -0.34(95% CI -0.63, -0.05)) unit decrease in fatigue. Conclusions: Body composition data can be collected from retrospective scans and appears to be prognostic of OS, PRO and TD.
- Published
- 2022
17. Increased risk for other cancers in individuals with Ewing sarcoma and their relatives
- Author
-
Erin L. Young, Jeffrey T. Yap, Diana Abbott, Joshua D. Schiffman, Kevin B. Jones, Stephen L. Lessnick, Lisa A. Cannon-Albright, James M. Farnham, Schuyler O'Brien, and R. Lor Randall
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,Prostate cancer ,0302 clinical medicine ,Risk Factors ,Neoplasms ,Utah ,Registries ,Stomach cancer ,Original Research ,education.field_of_study ,Sarcoma ,Neoplasms, Second Primary ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,relative risk ,Second Primary ,Population Surveillance ,030220 oncology & carcinogenesis ,Cancer Prevention ,medicine.medical_specialty ,Oncology and Carcinogenesis ,Population ,Sarcoma, Ewing ,lcsh:RC254-282 ,Risk Assessment ,03 medical and health sciences ,Breast cancer ,Ewing ,Acute lymphocytic leukemia ,Internal medicine ,medicine ,cancer ,Humans ,Family ,Radiology, Nuclear Medicine and imaging ,UPDB ,education ,Proportional Hazards Models ,business.industry ,Cancer ,medicine.disease ,Cancer registry ,relative ,030104 developmental biology ,Biochemistry and Cell Biology ,business ,Ewing sarcoma - Abstract
Background There are few reports of the association of other cancers with Ewing sarcoma in patients and their relatives. We use a resource combining statewide genealogy and cancer reporting to provide unbiased risks. Methods Using a combined genealogy of 2.3 million Utah individuals and the Utah Cancer Registry (UCR), relative risks (RRs) for cancers of other sites were estimated in 143 Ewing sarcoma patients using a Cox proportional hazards model with matched controls; however, risks in relatives were estimated using internal cohort‐specific cancer rates in first‐, second‐, and third‐degree relatives. Results Cancers of three sites (breast, brain, complex genotype/karyotype sarcoma) were observed in excess in Ewing sarcoma patients. No Ewing sarcoma patients were identified among first‐, second‐, or third‐degree relatives of Ewing sarcoma patients. Significantly increased risk for brain, lung/bronchus, female genital, and prostate cancer was observed in first‐degree relatives. Significantly increased risks were observed in second‐degree relatives for breast cancer, nonmelanoma eye cancer, malignant peripheral nerve sheath cancer, non‐Hodgkin lymphoma, and translocation sarcomas. Significantly increased risks for stomach cancer, prostate cancer, and acute lymphocytic leukemia were observed in third‐degree relatives. Conclusions This analysis of risk for cancer among Ewing sarcoma patients and their relatives indicates evidence for some increased cancer predisposition in this population which can be used to individualize consideration of potential treatment of patients and screening of patients and relatives., We used a unique resource linking genealogy from the mid 1800s to a statewide cancer registry from the mid 1960s to explore the association of cancers of other sites among Ewings sarcoma patients and their relatives. While this rare cancer was not observed to cluster in close relatives, multiple other cancer sites were observed among patients and their relatives and this data could inform the recommended cancer screening for this population.
- Published
- 2019
18. Protective autophagy elicited by RAF→MEK→ERK inhibition suggests a treatment strategy for RAS-driven cancers
- Author
-
Mona Foth, Courtney C. Cavalieri, Lance D. Burrell, Stephanie L. Cutler, Conan G. Kinsey, Kajsa E. Affolter, Jill E. Shea, Amelie M. Boespflug, Kaitrin M. Rehbein, Alana L. Welm, Jeffrey T. Yap, Bryan E. Welm, Michael T. Seipp, Sophia S. Schuman, Jonathan Whisenant, G. Weldon Gilcrease, Amanda Truong, David H. Lum, Katrin P. Guillen, Courtney L. Scaife, Martin McMahon, Eric L. Snyder, and Soledad A. Camolotto
- Subjects
Proto-Oncogene Proteins B-raf ,0301 basic medicine ,MAPK/ERK pathway ,CA-19-9 Antigen ,MAP Kinase Signaling System ,Pyridones ,Mice, SCID ,Pyrimidinones ,medicine.disease_cause ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Pancreatic cancer ,Autophagy ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,Mitogen-Activated Protein Kinase Kinases ,Trametinib ,business.industry ,Melanoma ,Cancer ,Chloroquine ,General Medicine ,medicine.disease ,Xenograft Model Antitumor Assays ,3. Good health ,Pancreatic Neoplasms ,Editorial Commentary ,030104 developmental biology ,030220 oncology & carcinogenesis ,ras Proteins ,Cancer research ,KRAS ,Signal transduction ,business - Abstract
Pancreatic ductal adenocarcinoma (PDA) was responsible for ~ 44,000 deaths in the United States in 2018 and is the epitome of a recalcitrant cancer driven by a pharmacologically intractable oncoprotein, KRAS1–4. Downstream of KRAS, the RAF→MEK→ERK signaling pathway plays a central role in pancreatic carcinogenesis5. However, paradoxically, inhibition of this pathway has provided no clinical benefit to patients with PDA6. Here we show that inhibition of KRAS→RAF→MEK→ERK signaling elicits autophagy, a process of cellular recycling that protects PDA cells from the cytotoxic effects of KRAS pathway inhibition. Mechanistically, inhibition of MEK1/2 leads to activation of the LKB1→AMPK→ULK1 signaling axis, a key regulator of autophagy. Furthermore, combined inhibition of MEK1/2 plus autophagy displays synergistic anti-proliferative effects against PDA cell lines in vitro and promotes regression of xenografted patient-derived PDA tumors in mice. The observed effect of combination trametinib plus chloroquine was not restricted to PDA as other tumors, including patient-derived xenografts (PDX) of NRAS-mutated melanoma and BRAF-mutated colorectal cancer displayed similar responses. Finally, treatment of a patient with PDA with the combination of trametinib plus hydroxychloroquine resulted in a partial, but nonetheless striking disease response. These data suggest that this combination therapy may represent a novel strategy to target RAS-driven cancers. Targeted inhibition of RAF–MEK–ERK signaling induces autophagy through the LKB1–AMPK axis, creating a therapeutic vulnerability that can be exploited for treating patients with pancreatic cancer and potentially other RAS-mutant tumors.
- Published
- 2019
19. MRI of Hypoxia in Primary Central Nervous System Tumors: Part II
- Author
-
Michael Karsy, David L. Gillespie, Kevin Horn, Jonathan B. Harper, Michael Ruesch, Jeffrey T. Yap, and Randy L. Jensen
- Subjects
General Earth and Planetary Sciences ,General Environmental Science - Published
- 2018
20. Radioisotope Imaging and Therapy for Bone Metastasis in Men With Castration-Resistant Prostate Cancer
- Author
-
Umang Swami, Neeraj Agarwal, and Jeffrey T. Yap
- Subjects
Oncology ,Male ,Radioisotopes ,Cancer Research ,medicine.medical_specialty ,business.industry ,MEDLINE ,Bone metastasis ,Bone Neoplasms ,Castration resistant ,medicine.disease ,Prostate cancer ,Prostatic Neoplasms, Castration-Resistant ,Internal medicine ,Benzamides ,Nitriles ,Phenylthiohydantoin ,medicine ,Medical imaging ,Humans ,business ,Randomized Controlled Trials as Topic ,Original Investigation - Abstract
IMPORTANCE: Both α-emitting and β-emitting bone-targeted radioisotopes (RIs) have been developed to treat men with metastatic castration-resistant prostate cancer (CRPC). Only 1 phase 3 randomized clinical trial has demonstrated an overall survival (OS) benefit from an α-emitting RI, radium 223 ((223)Ra), vs standard of care. Yet no head-to-head comparison has been done between α-emitting and β-emitting RIs. OBJECTIVE: To assess OS in men with bone metastases from CRPC treated with bone-targeted RIs and to compare the effects of α-emitting RIs with β-emitting RIs. DATA SOURCES: PubMed, Cochrane Library, ClinicalTrials.gov, and meeting proceedings between January 1993 and June 2013 were reviewed. Key terms included randomized trials, radioisotopes, radiopharmaceuticals, and prostate cancer. Data were collected, checked, and analyzed from February 2017 to October 2018. STUDY SELECTION: Selected trials included patients with prostate cancer, recruited more than 50 patients from January 1993 to June 2013, compared RI use with no RI use (placebo, external radiotherapy, or chemotherapy), and were randomized. Patients were diagnosed with histologically proven prostate cancer and disease progression after both surgical or chemical castration and have evidence of bone metastasis. Nine randomized clinical trials were identified as eligible, but 3 were excluded for insufficient data. DATA EXTRACTION AND SYNTHESIS: Individual patient data were requested for each eligible trial, and all data were checked with a standard procedure. The log-rank test stratified by trial was used to estimate hazard ratios (HRs), and a similar fixed-effects (FE) model was used to estimate odds ratios (ORs). The between-trial heterogeneity of treatment effects was evaluated by Cochran test and I(2) and was accounted by a random-effects (RE) model. MAIN OUTCOMES AND MEASURES: Overall survival; secondary outcomes were symptomatic skeletal event (SSE)–free survival and adverse events. RESULTS: Based on 6 randomized clinical trials including 2081 patients, RI use was significantly associated with OS compared with no RI use (HR, 0.86; 95% CI, 0.77-0.95; P = .004) with high heterogeneity (χ(2)(5) = 24.46; P
- Published
- 2019
21. MRI of Hypoxia in Primary Central Nervous System Tumors: Part I
- Author
-
Jonathan Harper, Jeffrey T. Yap, Michael Karsy, David L. Gillespie, Kevin P. Horn, Randy L. Jensen, and Michael Ruesch
- Subjects
Pathology ,medicine.medical_specialty ,medicine.anatomical_structure ,business.industry ,Central nervous system ,medicine ,General Earth and Planetary Sciences ,Hypoxia (medical) ,medicine.symptom ,business ,General Environmental Science - Published
- 2018
22. The Influential Role of BCL2 Family Members in Synovial Sarcomagenesis
- Author
-
Kyllie Smith-Fry, Dakota Nollner, Kevin B. Jones, Xingming Deng, Asia M. Susko, Jared J. Barrott, Mario R. Capecchi, Ju Fen Zhu, Lance D. Burrell, Lisa A. Cannon-Albright, Jeffrey T. Yap, and Amir Pozner
- Subjects
0301 basic medicine ,Cancer Research ,Carcinogenesis ,bcl-X Protein ,Apoptosis ,Biology ,medicine.disease_cause ,Translocation, Genetic ,Article ,Mice ,Sarcoma, Synovial ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,neoplasms ,Molecular Biology ,Mice, Knockout ,Regulation of gene expression ,Soft tissue ,Cancer ,medicine.disease ,Synovial sarcoma ,Mitochondria ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,030104 developmental biology ,Proto-Oncogene Proteins c-bcl-2 ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Benzimidazoles ,Sarcoma ,biological phenomena, cell phenomena, and immunity ,Signal Transduction - Abstract
Synovial sarcomas are deadly soft tissue malignancies associated with t(X;18) balanced chromosomal translocations. Expression of the apoptotic regulator BCL2 is prominent in synovial sarcomas and has prompted the hypothesis that synovial sarcomagenesis may depend on it. Herein, it is demonstrated that Bcl2 overexpression enhances synovial sarcomagenesis in an animal model. Furthermore, we determined increased familial clustering of human synovial sarcoma patients with victims of other BCL2-associated malignancies in the Utah Population Database. Conditional genetic disruption of Bcl2 in mice also led to reduced sarcomagenesis. Pharmacologic inhibition specific to BCL2 had no demonstrable efficacy against human synovial sarcoma cell lines or mouse tumors. However, targeting BCLxL in human and mouse synovial sarcoma with the small molecule BH3 domain inhibitor, BXI-72, achieved significant cytoreduction and increased apoptotic signaling. Thus, the contributory role of BCL2 in synovial sarcomagenesis does not appear to render it as a therapeutic target, but mitochondrial antiapoptotic BCL2 family members may be. Implications: The association of BCL2 expression with synovial sarcoma is found to fit with a subtle, but significant, impact of its enhanced presence or absence during early tumorigenesis. However, specific pharmacologic inhibition of BCL2 does not demonstrate a persistent dependence in fully developed tumors. Conversely, inhibition of the BCL2 family member BCLxL resulted in nanomolar potency against human synovial sarcoma cell lines and 50% tumor reduction in a genetically engineered mouse model. Mol Cancer Res; 15(12); 1733–40. ©2017 AACR.
- Published
- 2017
23. Oral Administration of
- Author
-
Hedieh, Saffari, Kathryn A, Peterson, Kristin M, Leiferman, Michael G, Stabin, Jeffrey J, Krstyen, Frederic C, Clayton, Leonard F, Pease, Jeffrey T, Yap, John M, Hoffman, and Gerald J, Gleich
- Subjects
Adult ,Male ,Tomography, Emission-Computed, Single-Photon ,Heparin ,Administration, Oral ,Eosinophilic Esophagitis ,Organotechnetium Compounds ,Middle Aged ,Article ,Humans ,Tissue Distribution ,Whole Body Imaging ,Esophagoscopy ,Radiopharmaceuticals - Abstract
To determine if heparin labeled withFreshly preparedOral administration ofThe biodistribution of ingested
- Published
- 2019
24. Linsitinib (OSI-906) for the Treatment of Adult and Pediatric Wild-Type Gastrointestinal Stromal Tumors, a SARC Phase II Study
- Author
-
Suzanne George, Scott M. Schuetze, Douglas B. Flieder, John Crowley, Lori Rink, Jeffrey T. Yap, Jason L. Hornick, Martin G. Belinsky, Michael Heinrich, Samuel Litwin, Amanda Abbott, Annick D. Van den Abbeele, Rashmi Chugh, Margaret von Mehren, Katherine A. Janeway, and Jian Q. Yu
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Linsitinib ,Adolescent ,Gastrointestinal Stromal Tumors ,SDHA ,Phases of clinical research ,PDGFRA ,Gastroenterology ,Article ,Receptor, IGF Type 1 ,03 medical and health sciences ,chemistry.chemical_compound ,Young Adult ,0302 clinical medicine ,Internal medicine ,medicine ,Clinical endpoint ,Humans ,Young adult ,Protein Kinase Inhibitors ,Gastrointestinal Neoplasms ,GiST ,business.industry ,Electron Transport Complex II ,Imidazoles ,Middle Aged ,Clinical trial ,Proto-Oncogene Proteins c-kit ,030104 developmental biology ,Treatment Outcome ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Pyrazines ,Mutation ,Female ,Patient Safety ,business - Abstract
Purpose: Most gastrointestinal stromal tumors (GIST) have activating mutations of KIT, PDGFRA, or uncommonly BRAF. Fifteen percent of adult and 85% of pediatric GISTs are wild type (WT), commonly having high expression of IGF-1R and loss of succinate dehydrogenase (SDH) complex function. We tested the efficacy of linsitinib, an oral TKI IGF-1R inhibitor, in patients with WT GIST. Patients and Methods: A multicenter phase II trial of linsitinib was conducted. The primary endpoint was objective response rate. Secondary endpoints were clinical benefit rate: complete response, partial response, and stable disease (SD) ≥ 9 months, and quantitative 2[18F]fluoro-2-deoxy-D-glucose (FDG) metabolic response (MR) at week 8. Serum levels for glucose, insulin, IGF-1R ligand IGF1, and binding proteins were obtained to explore correlations to patient outcomes and FDG-PET results. Results: Twenty patients were accrued in a 6-month period. Grade 3–4 toxicities possibly related to linsitinib were uncommon (8.5%). No objective responses were seen. Clinical benefit rate (CBR) at 9 months was 40%. Intense FDG uptake was observed at baseline, with partial MR of 12% and stable metabolic disease of 65% at week 8; these patients had RECIST 1.1 SD as their best response. Progression-free survival (PFS) and overall survival Kaplan–Meier estimates at 9 months were 52% and 80%, respectively. SDHA/B loss determined by IHC was seen in 35% and 88% of cases, respectively. Conclusions: Linsitinib is well tolerated in patients with WT GIST. Although the 9-month CBR was 40%, and PFS at 9 months was 52%, no objective responses were observed. Rapid accrual to this study demonstrates that clinical trials of experimental agents in selected subtypes of GIST are feasible.
- Published
- 2019
25. Abstract IA25: Targeting MEK1/2 inhibitor resistance in RAS-mutated cancers
- Author
-
Mona Foth, Soledad A. Camolotto, Stephanie L. Cutler, Jonathan Whisenant, Bryan E. Welm, Amanda Truong, Conan Kinsey, Kaitren Rehbein, Courtney L. Scaife, David H. Lum, Martin McMahon, Eric L. Snyder, Jeffrey T. Yap, Kajsa Afotler, Jill E. Shea, Courtney Cavalier, Michael T. Seipp, Katrin P. Gullien, Lance D. Burrell, Amelie M. Boespflug, and Alana L. Welm
- Subjects
MAPK/ERK pathway ,Trametinib ,Cancer Research ,Combination therapy ,business.industry ,Melanoma ,Autophagy ,Cancer ,medicine.disease ,Oncology ,Pancreatic cancer ,Cancer cell ,Cancer research ,Medicine ,business - Abstract
Although the RAF>MEK>ERK MAP kinase pathway is key to the development of many RAS-mutated cancers, efforts to target this pathway with pharmacologic inhibitors have failed to deliver clinical benefit to patients. Here we show that inhibition of RAS>RAF>MEK>ERK signaling in RAS-mutated cancer cell lines elicits autophagy, a process of cellular recycling that protects cancer cells from the potentially cytotoxic effects of RAF>MEK>ERK pathway inhibition. Furthermore, combined inhibition of MEK1/2 plus autophagy displays synergistic antiproliferative effects against cell lines in vitro and promotes regression of xenografted patient-derived tumors in mice. Finally, treatment of a cancer patient with the combination of trametinib plus hydroxychloroquine resulted in a partial, but nonetheless striking, disease response. These data suggest that this combination therapy may represent a novel strategy to target RAS-driven malignancies such as melanoma, lung, and pancreatic cancer. Citation Format: Conan Kinsey, Soledad Camolotto, Amelie Boespflug, Katrin Gullien, Amanda Truong, Mona Foth, Jill Shea, Michael Seipp, Jeffrey Yap, Lance Burrell, David Lum, Jonathan Whisenant, Courtney Cavalier, Kaitren Rehbein, Stephanie Cutler, Kajsa Afotler, Alana Welm, Bryan Welm, Courtney Scaife, Eric Snyder, Martin McMahon. Targeting MEK1/2 inhibitor resistance in RAS-mutated cancers [abstract]. In: Proceedings of the AACR Special Conference on Melanoma: From Biology to Target; 2019 Jan 15-18; Houston, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(19 Suppl):Abstract nr IA25.
- Published
- 2020
26. Abstract A45: Protective autophagy elicited by RAF→MEK→ERK inhibition suggests a treatment strategy for RAS-driven cancers
- Author
-
Alana L. Welm, Mona Foth, Courtney C. Cavalieri, Martin McMahon, Jill E. Shea, Stephanie L. Cutler, Conan Kinsey, Michael T. Seipp, Weldon Gilcrease, Kajsa E. Affolter, Bryan E. Welm, Eric L. Snyder, Kaitrin M. Rehbein, Jonathan Whisenant, Amelie M. Boespflug, Jeffrey T. Yap, Katrin P. Gullien, Lance D. Burrell, Soledad A. Camolotto, David H. Lum, and Courtney L. Scaife
- Subjects
MAPK/ERK pathway ,Trametinib ,Cancer Research ,Combination therapy ,business.industry ,Autophagy ,Cancer ,medicine.disease ,medicine.disease_cause ,Oncology ,Pancreatic cancer ,medicine ,Cancer research ,KRAS ,Signal transduction ,business ,Molecular Biology - Abstract
Pancreatic ductal adenocarcinoma was responsible for ~43,000 deaths in the USA in 2017 and is the epitome of a recalcitrant cancer driven by a pharmacologically intractable oncoprotein, KRAS. Although the clinical picture remains grim, the mechanisms by which key alterations in tumor suppressors and proto-oncogenes contribute to PDA have been dissected. Downstream of KRAS, the RAF→MEK→ERK signaling pathway plays a central role in pancreatic carcinogenesis. However, to date, pharmacologic inhibition of this pathway has provided no clinical benefit to PDA patients. Here we show that inhibition of KRAS→RAF→MEK→ERK signaling in PDA cell lines elicits autophagy, a process of cellular recycling that protects pancreatic cancer cells from the potentially cytotoxic effects of KRAS pathway inhibition. Furthermore, combined inhibition of MEK1/2 plus autophagy displays synergistic antiproliferative effects against PDA cell lines in vitro, and promotes regression of xenografted patient-derived PDA tumors in mice. Finally, treatment of a KRAS-mutated PDA patient on a compassionate basis with the combination of trametinib plus hydroxychloroquine resulted in a partial but nonetheless striking disease response. These data suggest that this combination therapy may represent a new strategy to target RAS-driven cancers such as PDA. Citation Format: Conan G. Kinsey, Soledad A. Camolotto, Amelie M. Boespflug, Katrin P. Gullien, Mona Foth, Jill E. Shea, Michael T. Seipp, Jeffrey T. Yap, Lance D. Burrell, David H. Lum, Jonathan R. Whisenant, Weldon Gilcrease, Courtney C. Cavalieri, Kaitrin M. Rehbein, Stephanie L. Cutler, Kajsa E. Affolter, Alana L. Welm, Bryan E. Welm, Courtney L. Scaife, Eric L. Snyder, Martin McMahon. Protective autophagy elicited by RAF→MEK→ERK inhibition suggests a treatment strategy for RAS-driven cancers [abstract]. In: Proceedings of the AACR Special Conference on Targeting RAS-Driven Cancers; 2018 Dec 9-12; San Diego, CA. Philadelphia (PA): AACR; Mol Cancer Res 2020;18(5_Suppl):Abstract nr A45.
- Published
- 2020
27. [P2–396]: HOW AMYLOID PET CHANGES COGNITIVE CARE: A BROADER VIEW
- Author
-
Norman L. Foster, Rorie DuPrey, Richard D. King, John M. Hoffman, Jeffrey T. Yap, Satoshi Minoshima, Kevin Horn, and Yue Zhang
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2017
28. [IC‐P‐007]: HOW AMYLOID PET CHANGES COGNITIVE CARE: A BROADER VIEW
- Author
-
Rorie DuPrey, John M. Hoffman, Kevin P. Horn, Yue Zhang, Richard D. King, Jeffrey T. Yap, Satoshi Minoshima, and Norman L. Foster
- Subjects
Cognitive science ,Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Amyloid pet ,Cognition ,Neurology (clinical) ,Geriatrics and Gerontology ,Psychology ,Neuroscience - Published
- 2017
29. Voxelwise single-subject analysis of imaging metabolic response to therapy in neuro-oncology
- Author
-
Armin Schwartzman, Mengye Guo, Annick D. Van den Abbeele, Nan Lin, and Jeffrey T. Yap
- Subjects
Statistics and Probability ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Image registration ,Lapatinib ,Statistical parametric mapping ,medicine.disease ,computer.software_genre ,Bioinformatics ,Clinical trial ,Breast cancer ,Voxel ,Positron emission tomography ,Multiple comparisons problem ,medicine ,Radiology ,Statistics, Probability and Uncertainty ,skin and connective tissue diseases ,business ,computer ,medicine.drug - Abstract
F-18-Fluorodeoxyglucose positron emission tomography (FDG-PET) has been used to evaluate the metabolic response of metastatic brain tumors to treatment by comparing their tumor glucose metabolism before and after treatment. The standard analysis based on regions-of-interest has the advantage of simplicity. However, it is by definition restricted to those regions and is subject to observer variability. In addition, the observed changes in tumor metabolism are often confounded by normal changes in the tissue background, which can be heterogenous. We propose an analysis pipeline for automatically detecting the change at each voxel in the entire brain of a single subject, while adjusting for changes in the background. The complete analysis includes image registration, segmentation, a hierarchical model for background adjustment and voxelwise statistical comparisons. We demonstrate the method’s ability to identify areas of tumor response and/or progression in two subjects enrolled in a clinical trial using FDG-PET to evaluate lapatinib for the treatment of brain metastases in breast cancer patients.
- Published
- 2014
30. Abstract 2183: Protective autophagy elicited by RAF®MEK®ERK inhibition suggests a treatment strategy for pancreatic cancer
- Author
-
Courtney C. Cavalieri, Katrin P. Guillen, Courtney L. Scaife, Kajsa E. Affolter, Lance D. Burrell, Jill E. Shea, Sophia S. Schuman, Alana L. Welm, Martin McMahon, David H. Lum, Jonathan Whisenant, Amanda Truong, Soledad A. Camolotto, Mona Foth, Conan G. Kinsey, Eric L. Snyder, Stephanie L. Cutler, G. Weldon Gilcrease, Amelie M. Boespflug, Kaitrin M. Rehbein, Jeffrey T. Yap, Michael T. Seipp, and Bryan E. Welm
- Subjects
MAPK/ERK pathway ,Trametinib ,Cancer Research ,Combination therapy ,business.industry ,Autophagy ,Cancer ,medicine.disease ,medicine.disease_cause ,Oncology ,Pancreatic cancer ,medicine ,Cancer research ,KRAS ,Signal transduction ,business - Abstract
Pancreatic ductal adenocarcinoma was responsible for ~43,000 deaths in the USA in 2017 and is the epitome of a recalcitrant cancer driven by an, as yet, pharmacologically intractable oncoprotein, KRAS. Although the clinical picture remains grim, the mechanisms by which key alterations in tumor suppressors and proto-oncogenes contribute to PDA have been dissected. Downstream of KRAS, the RAF→MEK→ERK signaling pathway plays a central role in pancreatic carcinogenesis. However, to date, pharmacological inhibition of this pathway has provided no clinical benefit to PDA patients. Here we show that inhibition of KRAS→RAF→MEK→ERK signaling in PDA cell lines elicits autophagy, a process of cellular recycling that protects pancreatic cancer cells from the potentially cytotoxic effects of KRAS pathway inhibition. Furthermore, combined inhibition of MEK1/2 plus autophagy displays synergistic anti-proliferative effects against PDA cell lines in vitro, and promotes regression of xenografted patient-derived PDA tumors in mice. Finally, treatment of a PDA patient with the combination of trametinib plus hydroxychloroquine resulted in a partial, but nonetheless striking disease response. These data suggest that this combination therapy may represent a novel strategy to target RAS-driven cancers such as PDA. Citation Format: Conan G. Kinsey, Soledad A. Camolotto, Amelie M. Boespflug, Katrin P. Guillen, Mona Foth, Amanda Truong, Sophia S. Schuman, Jill E. Shea, Michael T. Seipp, Jeffrey T. Yap, Lance D. Burrell, David H. Lum, Jonathan R. Whisenant, G. Weldon Gilcrease, Courtney C. Cavalieri, Kaitrin M. Rehbein, Stephanie L. Cutler, Kajsa E. Affolter, Alana L. Welm, Bryan E. Welm, Courtney L. Scaife, Eric L. Snyder, Martin McMahon. Protective autophagy elicited by RAF®MEK®ERK inhibition suggests a treatment strategy for pancreatic cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 2183.
- Published
- 2019
31. Publisher Correction: Protective autophagy elicited by RAF→MEK→ERK inhibition suggests a treatment strategy for RAS-driven cancers
- Author
-
Kaitrin M. Rehbein, Mona Foth, Courtney C. Cavalieri, Stephanie L. Cutler, Sophia S. Schuman, Michael T. Seipp, Lance D. Burrell, Jill E. Shea, Alana L. Welm, Conan G. Kinsey, David H. Lum, Amelie M. Boespflug, G. Weldon Gilcrease, Jonathan Whisenant, Kajsa E. Affolter, Amanda Truong, Eric L. Snyder, Soledad A. Camolotto, Jeffrey T. Yap, Katrin P. Guillen, Courtney L. Scaife, Martin McMahon, and Bryan E. Welm
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,business.industry ,Autophagy ,General Medicine ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,Treatment strategy ,business - Abstract
In the version of this article initially published, the label over the bottom schematic in Fig. 1a was “pH > 5.0”; it should have been “pH < 5.0”. Further, the original article misspelt the surname of Katrin P. Guillen as “Gullien”. These errors have been corrected in the print, PDF and HTML versions of the article.
- Published
- 2019
32. Imatinib for Melanomas Harboring Mutationally Activated or Amplified KIT Arising on Mucosal, Acral, and Chronically Sun-Damaged Skin
- Author
-
Jeffrey S. Weber, Jason J. Luke, Thomas F. Gajewski, Kevin B. Kim, Katherine Zukotynski, Meijun Zhu, Philip Friedlander, David E. Fisher, Frances A. Collichio, Keith T. Flaherty, George D. Demetri, Steven J. O'Day, Jonathan A. Fletcher, Rene Gonzalez, Carol Beadling, Annick D. Van den Abbeele, Anita Giobbie-Hurder, F. Stephen Hodi, Christopher L. Corless, Marc S. Ernstoff, Donald P. Lawrence, Jeffrey T. Yap, Adrián Mariño-Enríquez, and Michael Heinrich
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Pathology ,Skin Neoplasms ,Best Overall Response ,Antineoplastic Agents ,Proto-Oncogene Mas ,Gastroenterology ,Piperazines ,GTP Phosphohydrolases ,Internal medicine ,Biomarkers, Tumor ,medicine ,Mutation screening ,Humans ,Melanoma ,neoplasms ,Survival rate ,Aged ,Aged, 80 and over ,Mucous Membrane ,Oncogene ,business.industry ,Gene Amplification ,Membrane Proteins ,Imatinib ,Middle Aged ,Prognosis ,medicine.disease ,Survival Rate ,Proto-Oncogene Proteins c-kit ,Pyrimidines ,Imatinib mesylate ,Oncology ,Benzamides ,Chronic Disease ,Mutation ,Imatinib Mesylate ,Sunlight ,Female ,Neoplasm Recurrence, Local ,business ,Sun damaged skin ,medicine.drug - Abstract
Purpose Amplifications and mutations in the KIT proto-oncogene in subsets of melanomas provide therapeutic opportunities. Patients and Methods We conducted a multicenter phase II trial of imatinib in metastatic mucosal, acral, or chronically sun-damaged (CSD) melanoma with KIT amplifications and/or mutations. Patients received imatinib 400 mg once per day or 400 mg twice per day if there was no initial response. Dose reductions were permitted for treatment-related toxicities. Additional oncogene mutation screening was performed by mass spectroscopy. Results Twenty-five patients were enrolled (24 evaluable). Eight patients (33%) had tumors with KIT mutations, 11 (46%) with KIT amplifications, and five (21%) with both. Median follow-up was 10.6 months (range, 3.7 to 27.1 months). Best overall response rate (BORR) was 29% (21% excluding nonconfirmed responses) with a two-stage 95% CI of 13% to 51%. BORR was significantly greater than the hypothesized null of 5% and statistically significantly different by mutation status (7 of 13 or 54% KIT mutated v 0% KIT amplified only). There were no statistical differences in rates of progression or survival by mutation status or by melanoma site. The overall disease control rate was 50% but varied significantly by KIT mutation status (77% mutated v 18% amplified). Four patients harbored pretreatment NRAS mutations, and one patient acquired increased KIT amplification after treatment. Conclusion Melanomas that arise on mucosal, acral, or CSD skin should be assessed for KIT mutations. Imatinib can be effective when tumors harbor KIT mutations, but not if KIT is amplified only. NRAS mutations and KIT copy number gain may be mechanisms of therapeutic resistance to imatinib.
- Published
- 2013
33. Long-term follow-up results of the multicenter phase II trial of regorafenib in patients with metastatic and/or unresectable GI stromal tumor after failure of standard tyrosine kinase inhibitor therapy
- Author
-
Edwin Choy, Jeffrey T. Yap, M. von Mehren, George D. Demetri, Jonathan A. Fletcher, A. D. Van Den Abbeele, Constance M. Barysauskas, Jeffrey A. Morgan, Christopher L. Corless, Suzanne George, Eytan Ben-Ami, Michael Heinrich, Andrew J. Wagner, Sarah Solomon, and James E. Butrynski
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,medicine.medical_specialty ,Indoles ,Genotype ,Gastrointestinal Stromal Tumors ,Pyridines ,Phases of clinical research ,Disease-Free Survival ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Regorafenib ,medicine ,Sunitinib ,Humans ,Pyrroles ,Progression-free survival ,neoplasms ,Protein Kinase Inhibitors ,Aged ,GiST ,business.industry ,Phenylurea Compounds ,Hematology ,Original Articles ,Middle Aged ,digestive system diseases ,Surgery ,Clinical trial ,Proto-Oncogene Proteins c-kit ,030104 developmental biology ,Imatinib mesylate ,chemistry ,Response Evaluation Criteria in Solid Tumors ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Mutation ,Imatinib Mesylate ,Female ,business ,medicine.drug - Abstract
BACKGROUND This investigator-initiated trial provided the justification for the phase III GRID study resulting in worldwide regulatory approval of regorafenib as a third-line therapy for patients with metastatic gastrointestinal stromal tumors (GIST). We report the genotype analyses, long-term safety, and activity results from this initial trial of regorafenib in GIST. PATIENTS AND METHODS The trial was conducted between February 2010 and January 2014, among adult patients with metastatic GIST, after failure of at least imatinib and sunitinib. Patients received regorafenib orally, 160 mg once daily, days 1-21 of a 28-day cycle. Clinical benefit rate (CBR), defined as complete or partial response (PR), or stable disease lasting ≥16 weeks per RECIST 1.1, progression-free survival (PFS), overall survival (OS), long-term safety data, and metabolic response by functional imaging were assessed. RESULTS Thirty-three patients received at least one dose of regorafenib. The median follow-up was 41 months. CBR was documented in 25 of 33 patients [76%; 95% confidence interval (CI) 58% to 89%], including six PRs. The median PFS was 13.2 months (95% CI 9.2-18.3 months) including four patients who remained progression-free at study closure, each achieving clinical benefit for more than 3 years (range 36.8-43.5 months). The median OS was 25 months (95% CI 13.2-39.1 months). Patients whose tumors harbored a KIT exon 11 mutation demonstrated the longest median PFS (13.4 months), whereas patients with KIT/PDGFRA wild-type, non-SDH-deficient tumors experienced a median 1.6 months PFS (P < 0.0001). Long-term safety profile is consistent with previous reports; hand-foot skin reaction and hypertension were the most common reasons for dose reduction. Notably, regorafenib induced objective responses and durable benefit in SDH-deficient GIST. CONCLUSIONS Long-term follow-up of patients with metastatic GIST treated with regorafenib suggests particular benefit among patients with primary KIT exon 11 mutations and those with SDH-deficient GIST. Dose modifications are frequently required to manage treatment-related toxicities. CLINICAL TRIAL NUMBER NCT01068769.
- Published
- 2016
34. Demonstration of DCE-MRI as an early pharmacodynamic biomarker of response to VEGF Trap in glioblastoma
- Author
-
Allison F. O'Neill, Patrick Y. Wen, Lei Qin, Annick D. Van den Abbeele, Jeffrey T. Yap, and John de Groot
- Subjects
Male ,Vascular Endothelial Growth Factor A ,Cancer Research ,Disease Response ,Angiogenesis ,Recombinant Fusion Proteins ,Brain tumor ,Antineoplastic Agents ,Statistics, Nonparametric ,030218 nuclear medicine & medical imaging ,Lesion ,03 medical and health sciences ,0302 clinical medicine ,Fluorodeoxyglucose F18 ,medicine ,Effective diffusion coefficient ,Humans ,skin and connective tissue diseases ,medicine.diagnostic_test ,business.industry ,Brain Neoplasms ,Magnetic resonance imaging ,medicine.disease ,Magnetic Resonance Imaging ,Survival Analysis ,Receptors, Vascular Endothelial Growth Factor ,Treatment Outcome ,Neurology ,Oncology ,Positron-Emission Tomography ,Dynamic contrast-enhanced MRI ,Biomarker (medicine) ,Female ,Neurology (clinical) ,medicine.symptom ,business ,Nuclear medicine ,Glioblastoma ,030217 neurology & neurosurgery - Abstract
Glioblastoma (GBM) is an incurable brain tumor characterized by the expression of pro-angiogenic cytokines. A recent phase II clinical trial studied VEGF Trap in adult patients with temozolomide-resistant GBM. We sought to explore changes in [18F]Fluorodeoxyglucose positron emission tomography (FDG-PET) or magnetic resonance imaging (MRI) in trial participants correlating these changes with disease response. FDG-PET and MRI images obtained before and after the first dose of VEGF Trap were spatially co-registered. Regions of interest on each image slice were combined to produce a volume of interest representative of the entire tumor. Percent and absolute changes in maximum FDG-avidity, mean apparent diffusion coefficient (ADC), Ktrans, and Ve were calculated per lesion. Among the 12 participants that underwent dynamic contrast enhanced MRI (DCE-MRI), there were large, statistically significant reductions in Ktrans and Ve (median difference = −41.8 %, p
- Published
- 2016
35. Efficacy and Safety of Regorafenib in Patients With Metastatic and/or Unresectable GI Stromal Tumor After Failure of Imatinib and Sunitinib: A Multicenter Phase II Trial
- Author
-
Jonathan A. Fletcher, Sarah Solomon, Jeffrey T. Yap, Jeffrey A. Morgan, Qian Wang, Margaret von Mehren, Judith Manola, William D. Tap, Christopher L. Corless, James E. Butrynski, Meijun Zhu, Michael Heinrich, Andrew J. Wagner, George D. Demetri, Edwin Choy, Annick D. Van den Abbeele, and Suzanne George
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Indoles ,Gastrointestinal Stromal Tumors ,Pyridines ,Antineoplastic Agents ,Disease-Free Survival ,Drug Administration Schedule ,Piperazines ,chemistry.chemical_compound ,Internal medicine ,Regorafenib ,Original Reports ,Sunitinib ,medicine ,Clinical endpoint ,Humans ,Pyrroles ,Neoplasm Metastasis ,Stromal tumor ,Aged ,GiST ,business.industry ,Phenylurea Compounds ,Imatinib ,Middle Aged ,Surgery ,Clinical trial ,Pyrimidines ,Treatment Outcome ,Imatinib mesylate ,chemistry ,Drug Resistance, Neoplasm ,Benzamides ,Imatinib Mesylate ,Female ,business ,medicine.drug - Abstract
Purpose Metastatic GI stromal tumor (GIST) is a life-threatening disease with no therapy of proven efficacy after failure of imatinib and sunitinib. Regorafenib is a structurally unique inhibitor of multiple cancer-associated kinases, including KIT and platelet-derived growth factor receptor (PDGFR), with broad-spectrum anticancer activity in preclinical and early-phase trials. Because KIT and PDGFR-α remain drivers of GIST after resistance to imatinib and sunitinib, we performed a multicenter single-stage phase II trial of regorafenib in patients with advanced GIST after failure of at least imatinib and sunitinib. Patients and Methods Patients received regorafenib orally, 160 mg daily, on days 1 to 21 of a 28-day cycle. Disease assessment was performed every two cycles per RECIST 1.1. Primary end point was clinical benefit rate (CBR), defined as objective responses (ie, complete or partial response [PR] as well as stable disease [SD] ≥ 16 weeks). Serial tumor biopsies were obtained from consenting patients whenever possible. Results From February to December 2010, 34 patients were enrolled at four US centers. As of July 28, 2011, 33 patients had received at least two cycles of regorafenib (range, two to 17 cycles). CBR was 79% (95% CI, 61% to 91%). Four patients achieved PR, and 22 exhibited SD ≥ 16 weeks. Median progression-free survival was 10.0 months. The most common grade 3 toxicities were hypertension and hand-foot-skin reaction. Conclusion Regorafenib has significant activity in patients with advanced GIST after failure of both imatinib and sunitinib. A phase III trial of regorafenib versus placebo is ongoing to define more fully the safety and efficacy of regorafenib in this setting.
- Published
- 2012
36. Evaluating FDG uptake changes between pre and post therapy respiratory gated PET scans
- Author
-
Joseph H. Killoran, Aaron M. Allen, Jeffrey T. Yap, Michalis Aristophanous, Yue Yong, Ross Berbeco, and Aileen B. Chen
- Subjects
Male ,medicine.medical_specialty ,Lung Neoplasms ,medicine.medical_treatment ,Standardized uptake value ,Multimodal Imaging ,Lesion ,Fluorodeoxyglucose F18 ,Region of interest ,Carcinoma, Non-Small-Cell Lung ,medicine ,Carcinoma ,Humans ,Radiology, Nuclear Medicine and imaging ,Four-Dimensional Computed Tomography ,Lung cancer ,Aged ,Aged, 80 and over ,medicine.diagnostic_test ,business.industry ,Hematology ,Middle Aged ,medicine.disease ,Radiation therapy ,Oncology ,Positron emission tomography ,Positron-Emission Tomography ,Female ,Tomography ,Radiology ,Radiopharmaceuticals ,medicine.symptom ,Tomography, X-Ray Computed ,business ,Nuclear medicine - Abstract
Whole body (3D) and respiratory gated (4D) FDG-PET/CT scans performed pre-radiotherapy (pre-RT) and post-radiotherapy (post-RT) were analyzed to investigate the impact of 4D PET in evaluating 18F-fluorodeoxyglucose (FDG) uptake changes due to therapy, relative to traditional 3D PET.3D and 4D sequential FDG-PET/CT scans were acquired pre-RT and approximately one month post-RT for patients with non-small cell lung cancer (NSCLC). The lesions of high uptake targeted with radiotherapy were identified on the pre-RT scan of each patient. Each lesion on the 3D and each of the five phases of the 4D scan were analyzed using a region of interest (ROI). For each patient the ROIs of the pre-RT scans were used to locate the areas of initial FDG uptake on the post-RT scans following rigid registration. Post-RT ROIs were drawn and the FDG uptake was compared with that of the pre-RT scans.Sixteen distinct lesions from 12 patients were identified and analyzed. Standardized uptake value (SUV) maxima were significantly higher (p-value0.005) for the lesions as measured on the 4D compared to 3D PET. Comparison of serial pre and post-RT scans showed a mean 62% decrease in SUV with the 3D PET scan (range 36-89%), and a 67% decrease with the 4D PET scan (range 30-89%). The mean absolute difference in SUV change on 3D versus 4D scans was 4.9%, with a range 0-15% (p-value = 0.07).Signal recovery with 4D PET results in higher SUVs when compared to standard 3D PET. Consequently, differences in the evaluation of SUV changes between pre and post-RT plans were observed. Such difference can have a significant impact in PET-based response assessment.
- Published
- 2012
37. P2-09-07: Metabolic Response by FDG-PET in Patients (pts) Receiving Trastuzumab (T) and Lapatinib (L) for HER2+ Metastatic Breast Cancer (MBC): Correlative Analysis of TBCRC 003
- Author
-
A. D. Van Den Abbeele, Elizabeth Claire Dees, Yoko Franchetti, Rita Nanda, Nu Lin, EP Winer, J Berkowitz, Rebecca Gelman, Mothaffar F. Rimawi, Julie Najita, Carla I. Falkson, Timothy J. Hobday, Ingrid A. Mayer, Antonio C. Wolff, Jeffrey T. Yap, and Tricia Locascio
- Subjects
Cancer Research ,medicine.medical_specialty ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Standardized uptake value ,Context (language use) ,medicine.disease ,Lapatinib ,Gastroenterology ,Metastatic breast cancer ,Surgery ,Oncology ,Trastuzumab ,Internal medicine ,Cohort ,medicine ,business ,Progressive disease ,medicine.drug - Abstract
Background We evaluated the safety and efficacy of L+T in pts with 0–2 prior lines of chemotherapy (CT) for HER2+ MBC. In the context of this phase II trial, we evaluated metabolic response by FDG-PET and explored the relationship between metabolic response and clinical outcomes. Methods: Pts with measurable, HER2+ MBC were eligible. Cohort 1: No prior T, L, or CT +T for MBC, and >1 yr from adjuvant T, if received. Cohort 2: 1–2 prior lines of CT for MBC, including T, or relapse within 1 yr of adjuvant T. Pts received L 1,000 mg QD + T (2 mg/kg weekly or 6 mg/kg Q3W). Staging studies were done with CT or MRI at baseline (BL) and every 2 cycles (1 cycle=4 weeks [wks]). Objective response was assessed by local investigator according to RECIST 1.0. FDG-PET/CT was performed at BL, Wk 1, and Wk 8 per NCI guidelines. Central quality assurance, review, and analysis were performed on FDG-PET studies. Up to 5 target lesions were identified on BL FDG-PET images based on hypermetabolic uptake. Percent change in the summed maximum standardized uptake value (SUVmax) of target lesions was calculated at Wk 1 or Wk 8, compared to BL. Metabolic response was assessed according to EORTC criteria for % change in SUVmax (progressive disease [PD]: ≥25% increase; partial response [PR]: ≥25% decrease; stable disease [SD]: Results: 87 pts were registered to the study. Of these, one pt did not begin protocol therapy and one pt did not have MBC on further testing, and are not included. 81/85 pts had FDG-PET data at Wk 1; 75/85 had data at Wk 8. Metabolic PR at Wk 1 was observed in 28/39 (72%) pts in Cohort 1 and 20/42 (48%) pts in Cohort 2. Metabolic PR at Wk 8 was observed in 27/34 (79%) pts in Cohort 1 and 18/41 (44%) pts in Cohort 2. Wk 1 and Wk 8 metabolic responses were similar. In cohort 1, 18/28 (64%) pts who achieved Wk 1 metabolic PR had clinical benefit by RECIST. Of pts with Wk 1 metabolic SD, 2/9 (22%) had clinical benefit. In cohort 2, 9/20 (45%) pts who achieved Wk 1 metabolic PR had clinical benefit; 5/22 (23%) who achieved Week 1 metabolic SD had clinical benefit. Exploratory analysis of progression-free survival (PFS) showed that pts in Cohort 1 who achieved Wk 1 metabolic PR experienced a median PFS of 9.3 months ([mos]; 95% CI 5.6−22.3); for pts with metabolic SD, median PFS was 1.9 mos (95% CI 0.8−5.5). For pts in Cohort 2, Wk 1 metabolic PR was associated with median PFS of 5.6 mos (95% CI 3.7−7.8), whereas for pts with metabolic SD, median PFS was 3.7 mos (95% CI 1.8−5.5). Conclusions: L+T is associated with a high rate of early and sustained metabolic response by FDG-PET. Exploratory analyses suggest that metabolic PR may be associated with clinical benefit and longer PFS. Citation Information: Cancer Res 2011;71(24 Suppl):Abstract nr P2-09-07.
- Published
- 2011
38. Four-Dimensional Positron Emission Tomography: Implications for Dose Painting of High-Uptake Regions
- Author
-
Ross Berbeco, Aileen B. Chen, Joseph H. Killoran, Michalis Aristophanous, and Jeffrey T. Yap
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,medicine.medical_treatment ,Planning target volume ,Adenocarcinoma ,Fluorodeoxyglucose F18 ,Dose painting ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Aged ,Aged, 80 and over ,Fluorodeoxyglucose ,Radiation ,medicine.diagnostic_test ,business.industry ,Radiotherapy Dosage ,Middle Aged ,Tumor Burden ,High uptake ,Radiation therapy ,Oncology ,Positron emission tomography ,Positron-Emission Tomography ,Carcinoma, Squamous Cell ,Female ,Radiology ,Tomography ,Radiopharmaceuticals ,Tomography, X-Ray Computed ,business ,Nuclear medicine ,Algorithms ,Emission computed tomography ,medicine.drug - Abstract
To investigate the behavior of tumor subvolumes of high [18F]-fluorodeoxyglucose (FDG) uptake as seen on clinical four-dimensional (4D) FDG-positron emission tomography (PET) scans.Four-dimensional FDG-PET/computed tomography scans from 13 patients taken before radiotherapy were available. The analysis was focused on regions of high uptake that are potential dose-painting targets. A total of 17 lesions (primary tumors and lymph nodes) were analyzed. On each one of the five phases of the 4D scan a classification algorithm was applied to obtain the region of highest uptake and segment the tumor volume. We looked at the behavior of both the high-uptake subvolume, called "Boost," and the segmented tumor volume, called "Target." We measured several quantities that characterize the Target and Boost volumes and quantified correlations between them.The behavior of the Target could not always predict the behavior of the Boost. The shape deformation of the Boost regions was on average 133% higher than that of the Target. The gross to internal target volume expansion was on average 27.4% for the Target and 64% for the Boost, a statistically significant difference (p0.05). Finally, the inhale-to-exhale phase (20%) had the highest shape deformation for the Boost regions.A complex relationship between the measured quantities for the Boost and Target volumes is revealed. The results suggest that in cases in which advanced therapy techniques such as dose painting are being used, a close examination of the 4D PET scan should be performed.
- Published
- 2011
39. Comparison of Four Early Posttherapy Imaging Changes (EPTIC; RECIST 1.0, Tumor Shrinkage, Computed Tomography Tumor Density, Choi Criteria) in Assessing Outcome to Vascular Endothelial Growth Factor–Targeted Therapy in Patients With Advanced Renal Cell Carcinoma
- Author
-
Katherine M. Krajewski, David F. McDermott, Mengye Guo, Toni K. Choueiri, Ivan Pedrosa, Jyothi P. Jagannathan, Michael B. Atkins, Fabio A.B. Schutz, Daniel Y.C. Heng, Jeffrey T. Yap, Nikhil H. Ramaiya, and Annick D. Van den Abbeele
- Subjects
Niacinamide ,Vascular Endothelial Growth Factor A ,Sorafenib ,Indoles ,Time Factors ,Bevacizumab ,Pyridines ,Urology ,Angiogenesis Inhibitors ,Kaplan-Meier Estimate ,Antibodies, Monoclonal, Humanized ,Predictive Value of Tests ,Renal cell carcinoma ,Sunitinib ,Humans ,Medicine ,Pyrroles ,Molecular Targeted Therapy ,Carcinoma, Renal Cell ,Neoplasm Staging ,Retrospective Studies ,Observer Variation ,Academic Medical Centers ,business.industry ,Phenylurea Compounds ,Standard treatment ,Benzenesulfonates ,Antibodies, Monoclonal ,Reproducibility of Results ,Middle Aged ,medicine.disease ,Kidney Neoplasms ,Tumor Burden ,Treatment Outcome ,ROC Curve ,Response Evaluation Criteria in Solid Tumors ,Predictive value of tests ,Tomography, X-Ray Computed ,business ,Nuclear medicine ,Kidney cancer ,Boston ,medicine.drug - Abstract
Background Vascular endothelial growth factor (VEGF)-targeted therapy has become standard treatment for patients with metastatic renal cell cancer (mRCC). Since these therapies can induce tumor necrosis and minimal tumor shrinkage, Response Evaluation Criteria in Solid Tumors (RECIST) may not be optimal for predicting clinical outcome. Objective To systematically determine the optimal early posttherapy imaging changes (EPTIC) to separate responders and nonresponders at the first posttreatment follow-up computed tomography (CT). Design, setting, and participants Seventy mRCC patients with 155 target lesions treated with first-line sunitinib, sorafenib, or bevacizumab at academic medical centers underwent contrast-enhanced thoracic and abdominal CT at baseline and first follow-up after therapy initiation (median: 78 d after therapy initiation; range: 31–223 d). Measurements Evaluations were performed according to (1) RECIST 1.0; (2) Choi criteria; (3) tumor shrinkage (TS) of ≥10% decrease in sum of the longest unidimensional diameter (SLD); and (4) 15% or 20% decrease in mean CT tumor density. Correlation with time to treatment failure (TTF) and overall survival (OS) were compared and stratified by response to each of the radiologic criteria. Results and limitations Eleven patients were considered responders by RECIST 1.0; 49 based on Choi criteria; 31 patients had ≥10% decrease in the SLD; and 36 and 32 patients had ≥15% and ≥20% decrease, respectively, in mean tumor density on CT. Only the threshold of 10% decrease in the SLD was statistically significant in predicting TTF (10.4 vs 5.1 mo; p =0.02) and OS (32.5 vs 15.8 mo; p =0.002). Receiver operating characteristic analysis yielded a 10% decrease in SLD as the optimal size change threshold for responders. The retrospective nature of the study and measurements by a single oncoradiologist are inherent limitations. Conclusions In the retrospectively analyzed study population of mRCC patients receiving VEGF-targeted agents, a 10% reduction in the SLD on the first follow-up CT was an optimal early predictor of outcome.
- Published
- 2011
40. CT Tumor Volume Measurement in Advanced Non-small-cell Lung Cancer
- Author
-
David M. Jackman, Annick D. Van den Abbeele, Hiroto Hatabu, Mizuki Nishino, Pasi A. Jänne, Pamela J. DiPiro, Tak Kei Ho, Jeffrey T. Yap, Bruce E. Johnson, and Mengye Guo
- Subjects
medicine.medical_specialty ,Cone beam computed tomography ,Lung ,business.industry ,Concordance ,Atelectasis ,medicine.disease ,Clinical trial ,medicine.anatomical_structure ,Volume measurement ,medicine ,Radiology, Nuclear Medicine and imaging ,Radiology ,Iohexol ,Lung cancer ,business ,medicine.drug - Abstract
Rationale and Objectives Determine inter- and intraobserver variability of computed tomography (CT) tumor volume measurements in advanced non-small-cell lung cancer (NSCLC) patients treated in a Phase II clinical trial using chest CT. Materials and Methods Twenty-three advanced NSCLC patients with a total of 53 measurable lung lesions enrolled in a Phase II, multicenter, open-label clinical trial of erlotinib were retrospectively studied with institutional review board approval. Two radiologists independently measured the tumor size, volume, and CT attenuation coefficient using commercially available volume analysis software. Concordance correlation coefficients (CCCs) and Bland-Altman plots were used to assess inter- and intraobserver agreement. Results High CCCs (0.949–0.990) were observed in all types of measurements for interobserver agreement. The 95% limits of agreements for volume, unidimensional, and bidimensional measurements were (−26.0%, 18.6%), (−23.1%, 24.4%), and (−34.0%, 48.6%), respectively. Volume measurement had slightly higher CCC and narrower 95% limits of agreement compared to uni- and bidimensional measurements. CCCs for intraobserver agreement were high (range, 0.946–0.996) with CCC for volume being slightly higher than CCCs of uni- and bidimensional measurements. The smaller the tumor volume was, the larger the interobserver difference of CT attenuation. Location, morphology, or adjacent atelectasis had no significant impact on inter- or intraobserver variability. Conclusion CT tumor volume measurement in advanced NSCLC patients using clinical chest CT and commercially available software demonstrated high inter- and intraobserver agreement, indicating that the method may be used routinely in clinical practice.
- Published
- 2011
41. New Response Evaluation Criteria in Solid Tumors (RECIST) Guidelines for Advanced Non–Small Cell Lung Cancer: Comparison With Original RECIST and Impact on Assessment of Tumor Response to Targeted Therapy
- Author
-
Leigh Anne Cioffredi, David M. Jackman, Annick D. Van den Abbeele, Pasi A. Jänne, Hiroto Hatabu, Mizuki Nishino, Jeffrey T. Yap, Bruce E. Johnson, and Beow Y. Yeap
- Subjects
Male ,medicine.medical_specialty ,Lung Neoplasms ,medicine.medical_treatment ,Article ,Targeted therapy ,Erlotinib Hydrochloride ,Carcinoma, Non-Small-Cell Lung ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Lung cancer ,Protein Kinase Inhibitors ,Aged ,Retrospective Studies ,Aged, 80 and over ,PET-CT ,business.industry ,General Medicine ,Guideline ,medicine.disease ,Clinical trial ,Treatment Outcome ,Response Evaluation Criteria in Solid Tumors ,Practice Guidelines as Topic ,Disease Progression ,Quinazolines ,Female ,Radiology ,Erlotinib ,Tomography, X-Ray Computed ,business ,medicine.drug - Abstract
The purpose of this article is to compare the recently published revised Response Evaluation Criteria in Solid Tumors (RECIST) guidelines (version 1.1) to the original guidelines (RECIST 1.0) for advanced non-small cell lung cancer (NSCLC) after erlotinib therapy and to evaluate the impact of the new CT tumor measurement guideline on response assessment.Forty-three chemotherapy-naive patients with advanced NSCLC treated with erlotinib in a single-arm phase 2 multicenter open-label clinical trial were retrospectively studied. CT tumor measurement records using RECIST 1.0 that were generated as part of the prospective clinical trial were reviewed. A second set of CT tumor measurements was generated from the records to meet RECIST 1.1 guidelines. The number of target lesions, best response, and time to progression were compared between RECIST 1.1 and RECIST 1.0.The number of target lesions according to RECIST 1.1 decreased in 22 patients (51%) and did not change in 21 patients (49%) compared with the number according to RECIST 1.0 (p0.0001, paired Student's t test). Almost perfect agreement was observed between best responses using RECIST 1.1 and RECIST 1.0 (weighted kappa = 0.905). Two patients with stable disease according to RECIST 1.0 had progressive disease according to RECIST 1.1 criteria because of new lesions found on PET/CT. There was no significant difference in time to progression between RECIST 1.1 and RECIST 1.0 (p = 1.000, sign test).RECIST 1.1 provided almost perfect agreement in response assessment after erlotinib therapy compared with RECIST 1.0. Assessment with PET/CT was a major factor that influenced the difference in best response assessment between RECIST 1.1 and RECIST 1.0.
- Published
- 2010
42. Performance Evaluation of the microPET®—FOCUS-F120
- Author
-
D.F. Newport, D. Longford, Stefan Siegel, Jeffrey T. Yap, and Richard Laforest
- Subjects
Nuclear and High Energy Physics ,medicine.medical_specialty ,Scanner ,medicine.diagnostic_test ,business.industry ,Computer science ,Image quality ,Bandwidth (signal processing) ,Detector ,Coincidence ,Imaging phantom ,Optics ,Nuclear Energy and Engineering ,Positron emission tomography ,medicine ,Medical physics ,Electrical and Electronic Engineering ,business ,Image resolution - Abstract
microPETreg-Focus-F120 is the latest model of dedicated small animal PET scanners from CTI-Concorde Microsystems LLC, (Knoxville, TN). This scanner, based on the geometry of the microPET-R4, takes advantage of several detector modifications to the coincidence processing electronics that improve the image resolution, sensitivity, and counting rate performance as compared to the predecessor models. This work evaluates the performance of the Focus-F120 system and shows its improvement over the earlier models. In particular, the spatial resolution is shown to improve from 2.32 to 1.69 mm at 5 mm radial distance and the peak absolute sensitivity increases from 4.1% to 7.1% compared to the microPET-R4. The counting rate capability, expressed in noise equivalent counting rate (NEC-1R), was shown to peak at over 800 kcps at 88 MBq for both systems using a mouse phantom. For this small phantom, the NECR counting rate is limited by the data transmission bandwidth between the scanner and the acquisition console. The rat-like phantom showed peak NEC-1R value at 300 kcps at 140 MBq. Evaluation of image quality and quantitation accuracy was also performed using specially designed phantoms and animal experiments
- Published
- 2007
43. Phase II Study of Lapatinib in Combination With Trastuzumab in Patients With Human Epidermal Growth Factor Receptor 2-Positive Metastatic Breast Cancer: Clinical Outcomes and Predictive Value of Early [18F]Fluorodeoxyglucose Positron Emission Tomography Imaging (TBCRC 003)
- Author
-
Eric P. Winer, Ingrid A. Mayer, Julie Najita, Jeffrey T. Yap, Annick D. Van den Abbeele, Nan Lin, Mothaffar F. Rimawi, Ian E. Krop, Carla I. Falkson, E. Claire Dees, Carlos L. Arteaga, N. Ryabin, William T. Barry, Hao Guo, Antonio C. Wolff, Timothy J. Hobday, Rita Nanda, and Andrea L. Richardson
- Subjects
Oncology ,Adult ,Cancer Research ,medicine.medical_specialty ,Receptor, ErbB-2 ,medicine.medical_treatment ,Phases of clinical research ,Breast Neoplasms ,Lapatinib ,Antibodies, Monoclonal, Humanized ,Cohort Studies ,Trastuzumab ,Fluorodeoxyglucose F18 ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Humans ,Neoplasm Metastasis ,Radionuclide Imaging ,Aged ,Aged, 80 and over ,Chemotherapy ,business.industry ,ORIGINAL REPORTS ,Middle Aged ,medicine.disease ,Metastatic breast cancer ,Surgery ,Clinical trial ,Treatment Outcome ,Positron-Emission Tomography ,Cohort ,Quinazolines ,Female ,business ,medicine.drug - Abstract
Purpose Lapatinib plus trastuzumab improves outcomes relative to lapatinib alone in heavily pretreated, human epidermal growth factor receptor 2–positive metastatic breast cancer (MBC). We tested the combination in the earlier-line setting and explored the predictive value of [18F]fluorodeoxyglucose positron emission tomography ([18F]FDG-PET) for clinical outcomes. Patients and Methods Two cohorts were enrolled (cohort 1: no prior trastuzumab for MBC and ≥ 1 year from adjuvant trastuzumab, if given; cohort 2: one to two lines of chemotherapy including trastuzumab for MBC and/or recurrence < 1 year from adjuvant trastuzumab). The primary end point was objective response rate by RECIST v1.0; secondary end points included clinical benefit rate (complete response plus partial response plus stable disease ≥ 24 weeks) and progression-free survival. [18F]FDG-PET scans were acquired at baseline, week 1, and week 8. Associations between metabolic response and clinical outcomes were explored. Results Eighty-seven patients were registered (85 were evaluable for efficacy). The confirmed objective response rate was 50.0% (95% CI, 33.8% to 66.2%) in cohort 1 and 22.2% (95% CI, 11.3% to 37.3%) in cohort 2. Clinical benefit rate was 57.5% (95% CI, 40.9% to 73.0%) in cohort 1 and 40.0% (95% CI, 25.7% to 55.7%) in cohort 2. Median progression-free survival was 7.4 and 5.3 months, respectively. Lack of week-1 [18F]FDG-PET/computed tomography ([18F]FDG-PET/CT) response was associated with failure to achieve an objective response by RECIST (negative predictive value, 91% [95% CI, 74% to 100%] for cohort 1 and 91% [95% CI, 79% to 100%] for cohort 2). Conclusion Early use of lapatinib and trastuzumab is active in human epidermal growth factor receptor 2–positive MBC. Week-1 [18F]FDG-PET/CT may allow selection of patients who can be treated with targeted regimens and spared the toxicity of chemotherapy.
- Published
- 2015
44. Assessment of Pharmacodynamic Vascular Response in a Phase I Trial of Combretastatin A4 Phosphate
- Author
-
Helen Anderson, Terry Jones, Patricia M Price, Mathew P. Miller, Adele Robbins, and Jeffrey T. Yap
- Subjects
Cancer Research ,Chemotherapy ,Kidney ,medicine.diagnostic_test ,business.industry ,medicine.medical_treatment ,Blood volume ,chemistry.chemical_compound ,medicine.anatomical_structure ,Oncology ,chemistry ,Positron emission tomography ,Pharmacodynamics ,medicine ,Vascular-targeting agent ,Combretastatin A-4 phosphate ,Nuclear medicine ,business ,Perfusion - Abstract
Purpose: Clinical evaluation of novel agents that target tumor blood vessels requires pharmacodynamic end points that measure vascular damage. Positron emission tomography (PET) was used to measure the effects of the vascular targeting agent combretastatin A4 phosphate (CA4P) on tumor and normal tissue perfusion and blood volume. Patients and Methods: Patients with advanced solid tumors were enrolled onto part of a phase I, accelerated-titration, dose-escalation study. The effects of 5 to 114 mg/m2 CA4P on tumor, spleen, and kidney were investigated. Tissue perfusion was measured using oxygen-15 (15O)–labeled water and blood volume was measured using 15O-labeled carbon monoxide (C15O). Scans were performed immediately before, and 30 minutes and 24 hours after the first infusion of each dose level of CA4P. All statistical tests were two sided. Results: PET data were obtained for 13 patients with intrapatient dose escalation. Significant dose-dependent reductions were seen in tumor perfusion 30 minutes after CA4P administration (mean change, −49% at ≥ 52 mg/m2; P = .0010). Significant reductions were also seen in tumor blood volume (mean change, −15% at ≥ 52 mg/m2; P = .0070). Although by 24 hours there was tumor vascular recovery, for doses ≥ 52 mg/m2 the reduction in perfusion remained significant (P = .013). Thirty minutes after CA4P administration borderline significant changes were seen in spleen perfusion (mean change, −35%; P = .018), spleen blood volume (mean change, −18%; P = .022), kidney perfusion (mean change, −6%; P = .026), and kidney blood volume (mean change, −6%; P = .014). No significant changes were seen at 24 hours in spleen or kidney. Conclusion: CA4P produces rapid changes in the vasculature of human tumors that can be assessed using PET measurements of tumor perfusion.
- Published
- 2003
45. Measurement of renal tumour and normal tissue perfusion using positron emission tomography in a phase II clinical trial of razoxane
- Author
-
Paula Wells, Jeffrey T. Yap, Patricia M Price, H Anderson, Adrian L. Harris, M P Miller, and D Propper
- Subjects
Cancer Research ,positron emission tomography ,Antineoplastic Agents ,Blood volume ,Neovascularization ,Clinical ,Oxygen Radioisotopes ,In vivo ,medicine ,Humans ,Carcinoma, Renal Cell ,Volume of distribution ,Carbon Monoxide ,Kidney ,Neovascularization, Pathologic ,medicine.diagnostic_test ,business.industry ,tumour perfusion ,Water ,antiangiogenic ,medicine.disease ,Kidney Neoplasms ,medicine.anatomical_structure ,Oncology ,Regional Blood Flow ,Positron emission tomography ,razoxane ,medicine.symptom ,Nuclear medicine ,business ,Perfusion ,Tomography, Emission-Computed ,Kidney disease - Abstract
Measurement of tumour and normal tissue perfusion in vivo in cancer patients will aid the clinical development of antiangiogenic and antivascular agents. We investigated the potential antiangiogenic effects of the drug razoxane by measuring the changes in parameters estimated from H(2)(15)O and C(15)O positron emission tomography (PET) to indicate alterations in vascular physiology. The study comprised 12 patients with primary or metastatic renal tumours >3 cm in diameter enrolled in a Phase II clinical trial of oral razoxane. Perfusion, fractional volume of distribution of water (VD) and blood volume (BV) were measured in tumour and normal tissue before and 4-8 weeks after treatment with 125 mg twice-daily razoxane. Renal tumour perfusion was variable but lower than normal tissue: mean 0.87 ml min(-1) ml(-1) (range 0.33-1.67) compared to renal parenchyma: mean 1.65 ml min(-1) ml(-1) (range 1.16-2.88). In eight patients, where parallel measurements were made during the same scan session, renal tumour perfusion was significantly lower than in normal kidney (P=0.0027). There was no statistically significant relationship between pretreatment perfusion and tumour size (r=0.32, n=13). In six patients scanned before and after razoxane administration, there was no statistically significant change in tumour perfusion: mean perfusion pretreatment was 0.81 ml min(-1) ml(-1) (range 0.46-1.26) and perfusion post-treatment was 0.72 ml min(-1) ml(-1) (range 0.51-1.15, P=0.15). Tumour VD and BV did not change significantly following treatment: mean pretreatment VD=0.66 (range 0.50-0.87), post-treatment VD=0.71 (range 0.63-0.82, P=0.22); pretreatment BV=0.18 ml ml(-1) (range 0.10-0.25), post-treatment BV=0.167 ml ml(-1) (range 0.091-0.24, P=0.55). Tumour perfusion, VD and BV did not change significantly with tumour progression. This study has shown that H(2)(15)O and C(15)O PET provide useful in vivo physiological measurements, that even highly angiogenic renal cancers have poor perfusion compared to surrounding normal tissue, and that PET can provide valuable information on the in vivo biology of angiogenesis in man and can assess the effects of antiangiogenic therapy.
- Published
- 2003
46. Method for detecting voxelwise changes in fluorodeoxyglucose-positron emission tomography brain images via background adjustment in cancer clinical trials
- Author
-
Nezamoddin N. Kachouie, Jeffrey T. Yap, K McCall, Lei Qin, and Armin Schwartzman
- Subjects
medicine.diagnostic_test ,business.industry ,Image Processing ,Brain tumor ,Image registration ,computer.software_genre ,medicine.disease ,Imaging phantom ,Lesion ,03 medical and health sciences ,0302 clinical medicine ,Neuroimaging ,Positron emission tomography ,Voxel ,030220 oncology & carcinogenesis ,medicine ,Radiology, Nuclear Medicine and imaging ,Tomography ,medicine.symptom ,Nuclear medicine ,business ,computer ,030217 neurology & neurosurgery - Abstract
An important challenge to using fluorodeoxyglucose-positron emission tomography (FDG-PET) in clinical trials of brain tumor patients is to identify malignant regions whose metabolic activity shows significant changes between pretreatment and a posttreatment scans in the presence of high normal brain background metabolism. This paper describes a semiautomated processing and analysis pipeline that is able to detect such changes objectively with a given false detection rate. Image registration and voxelwise comparison of the pre- and posttreatment images were performed. A key step is adjustment of the observed difference by the estimated background change at each voxel, thereby overcoming the confounding effect of spatially heterogeneous metabolic activity in the brain. Components of the proposed method were validated via phantom experiments and computer simulations. It achieves a false response volume accuracy of 0.4% at a significance threshold of 3 standard deviations. It is shown that the proposed methodology can detect lesion response with 100% accuracy with a tumor-to-background-ratio as low as 1.5, and it is not affected by the background brain glucose metabolism change. We also applied the method to FDG-PET patient images from a clinical trial to assess treatment effects of lapatinib, which demonstrated significant changes in metabolism corresponding to tumor regions.
- Published
- 2017
47. Association of genomically unstable Ewing sarcoma tumors with HOTAIR overexpression and clinical outcome
- Author
-
Jeffrey T. Yap, James F. Amatruda, Evangelia L Lazaris, Joshua D. Schiffman, Xiaomeng Huang, R. Lor Randall, Lisa M. Abegglen, Kevin B. Jones, and Jamie D. Gardiner
- Subjects
Cancer Research ,Oncology ,Bone cancer ,business.industry ,Cancer research ,medicine ,HOTAIR ,Sarcoma ,medicine.disease ,business ,Fusion protein - Abstract
11035 Background: Ewing sarcoma (ES) is the second most common bone cancer in children, characterized by the EWS-FLI1 fusion protein. Like most translocation-driven pediatric cancers, ES is a genomically “quiet” cancer with a low mutational burden and strong epigenetic regulation. However, recurring copy number alterations (CNAs) still arise in some ES tumors (e.g., chromosome 1q gain, 8 gain, 12 gain, and 16q loss) while other ES tumors completely lack genomic CNAs. We hypothesized that clinical, molecular, and epigenetic differences exist between these two unstable vs. stable genomic subtypes of ES. Methods: We performed CNA analysis on over 200 ES FFPE tumors. 30% of ES tumors had stable genomes while 60% had one or more CNAs across the genome. We performed gene expression and methylation microarray analyses on 24 ES FFPE tumors (11 stable, 13 unstable). Expression and methylation signatures were compared between stable vs. unstable ES and combined with clinical outcome. Results: Patients with unstable vs. stable ES tumors revealed worse 5-year overall (OS) and event-free survival (EFS) (38% vs. 67% EFS, p = 0.005; 58% vs. 84% OS, p = 0.0016). The subtypes had distinct expression and methylation signatures and clustered by methylation patterns. We identified differentially expressed and methylated genes, including upregulation (p = 0.0005) and unmethylation (p = 0.0009) of the long non-coding RNA HOTAIR in unstable vs. stable ES tumors. HOTAIR expression is higher in metastatic ES tumors compared to primary ES tumors (p = 0.02). Per the Cancer Cell Line Encyclopedia (Broad), ES cell lines have increased HOTAIR expression compared to 36 other cancers. Conclusions: ES genomic profiling through copy number, gene expression, and methylation identified at least two subtypes of ES (stable and unstable) that differ in outcome, gene expression and methylation. This data suggests HOTAIR’s involvement in ES pathogenesis, particularly in unstable tumors that have worse prognosis. Investigation is ongoing for HOTAIR expression as a prognostic and therapeutic target for ES, including a marker for LSD-inhibitor response.
- Published
- 2017
48. A phase I study of everolimus and docetaxel in patients with castration-resistant prostate cancer
- Author
-
William Oh, Philip W. Kantoff, Judith Manola, Carmen Priolo, Mary-Ellen Taplin, Annick D. Van den Abbeele, Jeffrey T. Yap, Tomasz M. Beer, Massimo Loda, Aymen Elfiky, Christopher W. Ryan, Kevin D. Courtney, and Robert S. Ross
- Subjects
Oncology ,Male ,medicine.medical_specialty ,Combination therapy ,Maximum Tolerated Dose ,Urology ,Antineoplastic Agents ,Progressive Metabolic Disease ,Docetaxel ,urologic and male genital diseases ,Drug Administration Schedule ,Article ,Prostate cancer ,Fluorodeoxyglucose F18 ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Biomarkers, Tumor ,Humans ,Everolimus ,Aged ,business.industry ,Middle Aged ,medicine.disease ,Prostatic Neoplasms, Castration-Resistant ,Treatment Outcome ,Tolerability ,Positron-Emission Tomography ,Biomarker (medicine) ,Taxoids ,business ,medicine.drug - Abstract
Background The PTEN tumor suppressor is frequently lost in CRPC, with activation of Akt-mTOR signaling, driving growth. We conducted a phase I trial of the mTOR inhibitor, everolimus, and docetaxel in CRPC. Patients and Methods Eligible patients had progressive, metastatic, chemotherapy-naive CRPC. Patients received everolimus 10 mg daily for 2 weeks and underwent a restaging FDG-PET/computed tomography scan. Patient cohorts were subsequently treated at 3 dose levels of everolimus with docetaxel: 5 mg to 60 mg/m2, 10 mg to 60 mg/m2, and 10 mg to 70 mg/m2. The primary end point was the safety and tolerability of combination therapy. Results Accrual was 4 patients at level 1, 3 patients at level 2, and 8 patients at level 3. Common toxicities were hematologic and fatigue. Serum concentrations of everolimus when administered with docetaxel were 1.5 to 14.8 ng/mL in patients receiving 5 mg everolimus and 4.5 to 55.4 ng/mL in patients receiving 10 mg everolimus. Four patients had partial metabolic response (PMR) using FDG-PET, 12 had stable metabolic disease, and 2 had progressive metabolic disease after a 2-week treatment with everolimus alone. Five of 12 evaluable patients experienced a prostate-specific antigen (PSA) reduction ≥ 50% during treatment with everolimus together with docetaxel. All 4 patients with a PMR according to PET imaging experienced a PSA reduction in response to everolimus with docetaxel, and 3 of 4 had PSA declines ≥ 50%. Conclusion Everolimus 10 mg daily and docetaxel 60 mg/m2 was safe in CRPC patients and these were the recommended doses in combination. FDG-PET response might serve as a biomarker for target inhibition by mTOR inhibitors.
- Published
- 2014
49. Bevacizumab plus ipilimumab in patients with metastatic melanoma
- Author
-
Jeffrey T. Yap, Anita Giobbie-Hurder, F. Stephen Hodi, Jun Zhou, Martin C. Mihm, Elsa F. Velazquez, Philip Friedlander, Tetsuro Sasada, Nageatte Ibrahim, George F. Murphy, Annick D. Van den Abbeele, Sara Russell, Pamela J. DiPiro, Keith T. Flaherty, Xinqi Wu, Wanyong Zeng, Scott J. Rodig, Maria Gargano, Cecilia Lezcano, Donald P. Lawrence, David F. McDermott, Michael B. Atkins, and Nikhil H. Ramaiya
- Subjects
Adult ,Male ,Cancer Research ,Bevacizumab ,Angiogenesis ,Antibodies, Neoplasm ,Lymphocyte ,Immunology ,Ipilimumab ,Antibodies, Monoclonal, Humanized ,Drug Administration Schedule ,Article ,chemistry.chemical_compound ,Lymphocytes, Tumor-Infiltrating ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Melanoma ,Aged ,Neoplasm Staging ,Aged, 80 and over ,Dose-Response Relationship, Drug ,Neovascularization, Pathologic ,business.industry ,Antibodies, Monoclonal ,Middle Aged ,medicine.disease ,Immune checkpoint ,Blockade ,Vascular endothelial growth factor ,medicine.anatomical_structure ,Treatment Outcome ,chemistry ,Cancer research ,Female ,business ,medicine.drug - Abstract
Ipilimumab improves survival in advanced melanoma and can induce immune-mediated tumor vasculopathy. Besides promoting angiogenesis, vascular endothelial growth factor (VEGF) suppresses dendritic cell maturation and modulates lymphocyte endothelial trafficking. This study investigated the combination of CTLA4 blockade with ipilimumab and VEGF inhibition with bevacizumab. Patients with metastatic melanoma were treated in four dosing cohorts of ipilimumab (3 or 10 mg/kg) with four doses at 3-week intervals and then every 12 weeks, and bevacizumab (7.5 or 15 mg/kg) every 3 weeks. Forty-six patients were treated. Inflammatory events included giant cell arteritis (n = 1), hepatitis (n = 2), and uveitis (n = 2). On-treatment tumor biopsies revealed activated vessel endothelium with extensive CD8+ and macrophage cell infiltration. Peripheral blood analyses demonstrated increases in CCR7+/−/CD45RO+ cells and anti-galectin antibodies. Best overall response included 8 partial responses, 22 instances of stable disease, and a disease-control rate of 67.4%. Median survival was 25.1 months. Bevacizumab influences changes in tumor vasculature and immune responses with ipilimumab administration. The combination of bevacizumab and ipilimumab can be safely administered and reveals VEGF-A blockade influences on inflammation, lymphocyte trafficking, and immune regulation. These findings provide a basis for further investigating the dual roles of angiogenic factors in blood vessel formation and immune regulation, as well as future combinations of antiangiogenesis agents and immune checkpoint blockade. Cancer Immunol Res; 2(7); 632–42. ©2014 AACR.
- Published
- 2014
50. Image reconstruction for dynamic PET based on low-order approximation and restoration of the sinogram
- Author
-
Miles N. Wernick, Jeffrey T. Yap, Chien-Min Kao, and Jogeshwar Mukherjee
- Subjects
Radiological and Ultrasound Technology ,medicine.diagnostic_test ,Receptors, Dopamine D2 ,Image quality ,Computer science ,business.industry ,Noise reduction ,Brain ,Iterative reconstruction ,Macaca mulatta ,Computer Science Applications ,Positron emission tomography ,Image Processing, Computer-Assisted ,medicine ,Animals ,Projections onto convex sets ,Computer vision ,Artificial intelligence ,Noise (video) ,Electrical and Electronic Engineering ,business ,Image resolution ,Software ,Smoothing ,Tomography, Emission-Computed - Abstract
Many image-reconstruction methods have been proposed to improve the spatial resolution of positron emission tomography (PET) images and, thus, to produce better quantification. However, these techniques, which are designed for static images, may be inadequate for good reconstruction from dynamic data. The authors present a simple, but effective, reconstruction approach intended specifically for dynamic studies. First, the level of noise in dynamic PET data is reduced by smoothing along the time axis using a low-order approximation. Next, the denoised sinograms are restored spatially by the method of projections onto convex sets. Finally, images are reconstructed from the restored sinograms by ordinary filtered backprojection. The authors present experimental results that demonstrate substantial improvements in region-of-interest quantification in actual and simulated dopamine D-2 neuroreceptor-imaging studies of a monkey brain.
- Published
- 1997
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.