51
|
Alexander MS, Wilkes JG, Schroeder SR, Buettner GR, Wagner BA, Du J, Gibson-Corley K, O'Leary BR, Spitz DR, Buatti JM, Berg DJ, Bodeker KL, Vollstedt S, Brown HA, Allen BG, Cullen JJ. Pharmacologic Ascorbate Reduces Radiation-Induced Normal Tissue Toxicity and Enhances Tumor Radiosensitization in Pancreatic Cancer. Cancer Res 2018; 78:6838-6851. [PMID: 30254147 DOI: 10.1158/0008-5472.can-18-1680] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/07/2018] [Accepted: 09/20/2018] [Indexed: 12/23/2022]
Abstract
: Chemoradiation therapy is the mainstay for treatment of locally advanced, borderline resectable pancreatic cancer. Pharmacologic ascorbate (P-AscH-, i.e., intravenous infusions of ascorbic acid, vitamin C), but not oral ascorbate, produces high plasma concentrations capable of selective cytotoxicity to tumor cells. In doses achievable in humans, P-AscH- decreases the viability and proliferative capacity of pancreatic cancer via a hydrogen peroxide (H2O2)-mediated mechanism. In this study, we demonstrate that P-AscH- radiosensitizes pancreatic cancer cells but inhibits radiation-induced damage to normal cells. Specifically, radiation-induced decreases in clonogenic survival and double-stranded DNA breaks in tumor cells, but not in normal cells, were enhanced by P-AscH-, while radiation-induced intestinal damage, collagen deposition, and oxidative stress were also reduced with P-AscH- in normal tissue. We also report on our first-in-human phase I trial that infused P-AscH- during the radiotherapy "beam on." Specifically, treatment with P-AscH- increased median overall survival compared with our institutional average (21.7 vs. 12.7 months, P = 0.08) and the E4201 trial (21.7 vs. 11.1 months). Progression-free survival in P-AscH--treated subjects was also greater than our institutional average (13.7 vs. 4.6 months, P < 0.05) and the E4201 trial (6.0 months). Results indicated that P-AscH- in combination with gemcitabine and radiotherapy for locally advanced pancreatic adenocarcinoma is safe and well tolerated with suggestions of efficacy. Because of the potential effect size and minimal toxicity, our findings suggest that investigation of P-AscH- efficacy is warranted in a phase II clinical trial. SIGNIFICANCE: These findings demonstrate that pharmacologic ascorbate enhances pancreatic tumor cell radiation cytotoxicity in addition to offering potential protection from radiation damage in normal surrounding tissue, making it an optimal agent for improving treatment of locally advanced pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Matthew S Alexander
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Justin G Wilkes
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Samuel R Schroeder
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Garry R Buettner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Brett A Wagner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Juan Du
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Katherine Gibson-Corley
- The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Brianne R O'Leary
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - John M Buatti
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Daniel J Berg
- The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,Division of Hematology, Oncology, and Blood and Marrow Transplantation, Department of Internal Medicine, The University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Kellie L Bodeker
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Sandy Vollstedt
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Heather A Brown
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Bryan G Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Joseph J Cullen
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa. .,Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,The Veterans' Affairs Medical Center, Iowa City, Iowa
| |
Collapse
|
52
|
Wang C, Liu X, Wang X, Wang Y, Cha N. Effects of chemoradiotherapy and chemotherapy on survival of patients with locally advanced pancreatic cancer: A meta-analysis of randomized controlled trials. Medicine (Baltimore) 2018; 97:e12260. [PMID: 30200163 PMCID: PMC6133448 DOI: 10.1097/md.0000000000012260] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
To comparatively evaluate chemoradiotherapy (CRT) and chemotherapy (CT) for the treatment of locally advanced pancreatic cancer (LAPC) by meta-analysis.A literature search was performed until August 2016 to identify comparative studies assessing survival rates and complications. Pooled odds ratios (ORs) and 95% confidence intervals (95% CIs) were determined with the fixed or random effects model.Five randomized controlled trials (RCTs) met the defined inclusion criteria. A total of 593 patients were included, with 295 and 298 treated with CRT and CT, respectively. Overall survival showed no statistically significant difference in patients treated with CRT and CT at 6, 12, 18, and 24 months (respectively: OR = 1.13, 95% CI: 0.60-2.17; OR = 1.15, 95% CI: 0.53-2.52; OR = 1.13, 95% CI: 0.43-2.95; OR = 1.07, 95% CI: 0.67-1.72). Meanwhile, CRT had higher rates of grade 3 to 4 adverse events (nausea and vomiting, OR = 2.74, 95% CI: 1.36-5.52; diarrhea, OR = 4.28, 95% CI: 1.16-15.71).The data are not sufficient to change from CT to CRT in the treatment of patients with LAPC and thus clinical discretion is required until more data is accumulated.
Collapse
|
53
|
Di Maso LD, Huang J, Bassetti MF, DeWerd LA, Miller JR. Investigating a novel split-filter dual-energy CT technique for improving pancreas tumor visibility for radiation therapy. J Appl Clin Med Phys 2018; 19:676-683. [PMID: 30117641 PMCID: PMC6123148 DOI: 10.1002/acm2.12435] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/17/2018] [Accepted: 07/21/2018] [Indexed: 12/17/2022] Open
Abstract
Purpose Tumor delineation using conventional CT images can be a challenge for pancreatic adenocarcinoma where contrast between the tumor and surrounding healthy tissue is low. This work investigates the ability of a split‐filter dual‐energy CT (DECT) system to improve pancreatic tumor contrast and contrast‐to‐noise ratio (CNR) for radiation therapy treatment planning. Materials and methods Multiphasic scans of 20 pancreatic tumors were acquired using a split‐filter DECT technique with iodinated contrast medium, OMNIPAQUETM. Analysis was performed on the pancreatic and portal venous phases for several types of DECT images. Pancreatic gross target volume (GTV) contrast and CNR were calculated and analyzed from mixed 120 kVp‐equivalent images and virtual monoenergetic images (VMI) at 57 and 40 keV. The role of iterative reconstruction on DECT images was also investigated. Paired t‐tests were used to assess the difference in GTV contrast and CNR among the different images. Results The VMIs at 40 keV had a 110% greater image noise compared to the mixed 120 kVp‐equivalent images (P < 0.0001). VMIs at 40 keV increased GTV contrast from 15.9 ± 19.9 HU to 93.7 ± 49.6 HU and CNR from 1.37 ± 2.05 to 3.86 ± 2.78 in comparison to the mixed 120 kVp‐equivalent images. The iterative reconstruction algorithm investigated decreased noise in the VMIs by about 20% and improved CNR by about 30%. Conclusions Pancreatic tumor contrast and CNR were significantly improved using VMIs reconstructed from the split‐filter DECT technique, and the use of iterative reconstruction further improved CNR. This gain in tumor contrast may lead to more accurate tumor delineation for radiation therapy treatment planning.
Collapse
Affiliation(s)
- Lianna D Di Maso
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, 53716, USA
| | - Jessie Huang
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, 53716, USA
| | - Michael F Bassetti
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, 53716, USA
| | - Larry A DeWerd
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, 53716, USA
| | - Jessica R Miller
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, 53716, USA
| |
Collapse
|
54
|
Chuong M, Badiyan SN, Yam M, Li Z, Langen K, Regine W, Morris C, Snider J, Mehta M, Huh S, Rutenberg M, Nichols RC. Pencil beam scanning versus passively scattered proton therapy for unresectable pancreatic cancer. J Gastrointest Oncol 2018; 9:687-693. [PMID: 30151265 DOI: 10.21037/jgo.2018.03.14] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Background With an increasing number of proton centers capable of delivering pencil beam scanning (PBS), understanding the dosimetric differences in PBS compared to passively scattered proton therapy (PSPT) for pancreatic cancer is of interest. Methods Optimized PBS plans were retrospectively generated for 11 patients with locally advanced pancreatic cancer previously treated with PSPT to 59.4 Gy on a prospective trial. The primary tumor was targeted without elective nodal coverage. The same treatment couch, target coverage and normal tissue dose objectives were used for all plans. A Wilcoxon t-test was performed to compare various dosimetric points between the two plans for each patient. Results All target volume coverage goals were met in all PBS and passive scattering (PS) plans, except for the planning target volume (PTV) coverage goal (V100% >95%) which was not met in one PS plan (range, 81.8-98.9%). PBS was associated with a lower median relative dose (102.4% vs. 103.8%) to 10% of the PTV (P=0.001). PBS plans had a lower median duodenal V59.4 Gy (37.4% vs. 40.4%; P=0.014), lower small bowel median V59.4 Gy (0.11% vs. 0.37%; P=0.012), lower stomach median V59.4 Gy (0.01% vs. 0.1%; P=0.023), and lower median dose to 0.1 cc of the spinal cord {35.0 vs. 38.7 Gy [relative biological effectiveness (RBE)]; P=0.001}. Liver dose was higher in PBS plans for median V5 Gy (24.1% vs. 20.2%; P=0.032), V20 Gy (3.2% vs. 2.8%; P=0.010), and V25 Gy (2.6% vs. 2.2%; P=0.019). There was no difference in kidney dose between PBS and PS plans. Conclusions Proton therapy for locally advanced pancreatic cancer using PBS was not clearly associated with clinically meaningful reductions in normal tissue dose compared to PS. Some statistically significant improvements in PTV coverage were achieved using PBS. PBS may offer improved conformality for the treatment of irregular targets, and further evaluation of PBS and PS incorporating elective nodal irradiation should be considered.
Collapse
Affiliation(s)
- Michael Chuong
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Shahed N Badiyan
- University of Maryland Medical School of Medicine, Baltimore, MD, USA
| | - Man Yam
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Zuofeng Li
- University of Florida Proton Therapy Institute, Jacksonville, FL, USA
| | - Katja Langen
- University of Maryland Medical School of Medicine, Baltimore, MD, USA
| | - William Regine
- University of Maryland Medical School of Medicine, Baltimore, MD, USA
| | | | - James Snider
- University of Maryland Medical School of Medicine, Baltimore, MD, USA
| | - Minesh Mehta
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Soon Huh
- University of Florida Proton Therapy Institute, Jacksonville, FL, USA
| | - Michael Rutenberg
- University of Florida Proton Therapy Institute, Jacksonville, FL, USA
| | - Romaine C Nichols
- University of Florida Proton Therapy Institute, Jacksonville, FL, USA
| |
Collapse
|
55
|
Clinical evaluation of intensity-modulated radiotherapy for locally advanced pancreatic cancer. Radiat Oncol 2018; 13:118. [PMID: 29940984 PMCID: PMC6019294 DOI: 10.1186/s13014-018-1063-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 06/14/2018] [Indexed: 01/05/2023] Open
Abstract
Background The purpose was to retrospectively evaluate the effect of intensity-modulated radiotherapy (IMRT) on gastrointestinal (GI) toxicities and outcomes compared to three-dimensional conformal radiotherapy (3DCRT) for locally advanced pancreatic cancer (LAPC). Methods We included 107 consecutive patients who underwent CRT for LAPC from September 2001 to March 2015; 80 patients underwent 3DCRT and 27 patients underwent IMRT. They were compared for GI toxicities, locoregional progression free survival (LRPFS), distant metastasis free survival (DMS), and overall survival (OS). Results Median radiation dose and fractions for 3DCRT and IMRT were 54 Gy/30 fr. and 48 Gy/15 fr. The regimens of CRT consisted of weekly gemcitabine 250 mg/m2 (for 3DCRT) or 1000 mg/m2 (for IMRT). Acute GI toxicity ≥grade 2 occurred in 32 patients (40%) treated with 3DCRT compared with five patients (19%) treated with IMRT. Late GI toxicity of grade 3 occurred in 10 patients (12%) treated with 3DCRT and one patient (4%) treated with IMRT. Patients who underwent IMRT had superior 1-year LRPFS (73.1% vs. 63.2%, p = 0.035) and 1-year OS (92.3% vs. 68.2%, p = 0.037) as compared with those treated with 3DCRT. Multivariate analysis showed that in IMRT patients, higher dose (≥45 Gy) was an independent factor for better LRPFS and OS. Conclusions LAPC patients treated with hypofractionated full-dose gemcitabine IMRT had improved OS and LRPFS without increased GI toxicities when compared to those of patients treated with conventionally fractionated low dose gemcitabine 3DCRT. In IMRT patients, higher dose was an independent favorable prognostic factor for better LRPFS and OS, which suggests that dose escalation with IMRT for LAPC is a promising strategy.
Collapse
|
56
|
Hsieh M, Chang W, Yu H, Lu C, Chang C, Chow J, Chen S, Cheng Y, Wu S. Adjuvant radiotherapy and chemotherapy improve survival in patients with pancreatic adenocarcinoma receiving surgery: adjuvant chemotherapy alone is insufficient in the era of intensity modulation radiation therapy. Cancer Med 2018; 7:2328-2338. [PMID: 29665327 PMCID: PMC6010773 DOI: 10.1002/cam4.1479] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/24/2018] [Accepted: 03/02/2018] [Indexed: 12/13/2022] Open
Abstract
In the era of intensity modulation radiation therapy (IMRT), no prospective randomized trial has evaluated the efficacy of adjuvant therapies such as adjuvant concurrent chemoradiotherapy (CCRT), adjuvant sequential chemotherapy and radiotherapy (CT-RT), and adjuvant CT alone in resectable pancreatic adenocarcinoma (PA). Through propensity score matching, we designed a nationwide, population-based, head-to-head cohort study to determine the effects of dissimilar adjuvant treatments on resectable PA. We minimized the confounding of various adjuvant treatment outcomes among the following resectable PA groups of patients from the Taiwan Cancer Registry database: group 1, adjuvant CCRT; group 2, adjuvant sequential CT-RT; and group 3, adjuvant CT alone. All the studied techniques are IMRTs. The matching process yielded a final cohort of 588 patients (196, 196, and 196 patients in groups 1, 2, and 3, respectively). In both univariate and multivariate Cox regression analyses, adjusted hazard ratios (aHRs; 95% confidence interval [CI]) of death derived for the adjuvant CCRT and adjuvant sequential CT-RT cohorts compared with the adjuvant CT alone cohort were 0.398 (0.314-0.504) and 0.307 (0.235-0.402), respectively. A combination of adjuvant IMRT and CT for resectable PA treatment improves survival to a greater extent than does adjuvant CT alone.
Collapse
Affiliation(s)
- Mao‐Chih Hsieh
- Department of General SurgeryWan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
| | - Wei‐Wen Chang
- Department of General SurgeryWan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
| | - Hsin‐Hsien Yu
- Department of General SurgeryWan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
| | - Chang‐Yun Lu
- Department of General SurgeryWan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
| | - Chia‐Lun Chang
- Department of Hemato‐OncologyWan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
| | - Jyh‐Ming Chow
- Department of Hemato‐OncologyWan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
| | - Shee‐Uan Chen
- Department of Obstetrics and GynecologyNational Taiwan University HospitalTaipeiTaiwan
| | - Yunfeng Cheng
- Department of HematologyZhongshan Hospital Fudan UniversityShanghaiChina
- Department of HematologyZhongshan Hospital Qingpu BranchFudan UniversiyShanghaiChina
- Institute of Clinical ScienceZhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Institute of Clinical BioinformaticsFudan University Center for Clinical BioinformaticsShanghaiChina
| | - Szu‐Yuan Wu
- Department of Radiation OncologyWan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
- Department of Internal MedicineSchool of MedicineCollege of MedicineTaipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
57
|
De Blasio A, Vento R, Di Fiore R. Mcl-1 targeting could be an intriguing perspective to cure cancer. J Cell Physiol 2018; 233:8482-8498. [PMID: 29797573 DOI: 10.1002/jcp.26786] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 04/30/2018] [Indexed: 12/25/2022]
Abstract
The Bcl-2 family, which plays important roles in controlling cancer development, is divided into antiapoptotic and proapoptotic members. The change in the balance between these members governs the life and death of the cells. Mcl-1 is an antiapoptotic member of this family and its distribution in normal and cancerous tissues strongly differs from that of Bcl-2. In human cancers, where upregulation of antiapoptotic proteins is common, Mcl-1 expression is regulated independent of Bcl-2 and its inhibition promotes senescence, a major barrier to tumorigenesis. Cancer chemotherapy determines various kinds of responses, such as senescence and autophagy; however, the ideal response to chemotherapy is apoptosis. Mcl-1 is a potent oncogene that is regulated at the transcriptional, posttranscriptional, and posttranslational levels. Mcl-1 is a short-lived protein that, in the NH2 terminal region, contains sites for posttranslational regulation that can lead to proteasomal degradation. The USP9X Mcl-1 deubiquitinase regulates Mcl-1 and the levels of these two proteins are strongly correlated. Mcl-1 has three splicing variants (the antiapoptotic protein Mcl-1L and the proapoptotic proteins Mcl-1S and Mcl-1ES), each contributing toward apoptosis regulation. In cancers responsible for the most deaths in the world, the presence of Mcl-1 is associated with malignant cell growth and evasion of apoptosis. Mcl-1 is also one of the key regulators of cancer stem cells' self-renewal that contributes to tumor survival. A great number of indirect and selective Mcl-1 inhibitors have been produced and some of these have shown efficacy in several clinical trials. Thus, therapeutic manipulation of Mcl-1 can be a useful strategy to combat cancer.
Collapse
Affiliation(s)
- Anna De Blasio
- Laboratory of Biochemistry, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Polyclinic, Palermo, Italy.,Associazione Siciliana per la Lotta contro i Tumori (ASLOT), Palermo, Italy
| | - Renza Vento
- Laboratory of Biochemistry, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Polyclinic, Palermo, Italy.,Associazione Siciliana per la Lotta contro i Tumori (ASLOT), Palermo, Italy.,Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Riccardo Di Fiore
- Laboratory of Biochemistry, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Polyclinic, Palermo, Italy.,Associazione Siciliana per la Lotta contro i Tumori (ASLOT), Palermo, Italy.,Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
58
|
Kawashiro S, Yamada S, Okamoto M, Ohno T, Nakano T, Shinoto M, Shioyama Y, Nemoto K, Isozaki Y, Tsuji H, Kamada T. Multi-institutional Study of Carbon-ion Radiotherapy for Locally Advanced Pancreatic Cancer: Japan Carbon-ion Radiation Oncology Study Group (J-CROS) Study 1403 Pancreas. Int J Radiat Oncol Biol Phys 2018; 101:1212-1221. [PMID: 29907490 DOI: 10.1016/j.ijrobp.2018.04.057] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 03/09/2018] [Accepted: 04/23/2018] [Indexed: 01/31/2023]
Abstract
PURPOSE The aim of this multi-institutional study was to evaluate the efficacy and safety of carbon-ion radiotherapy (C-ion RT) for locally advanced pancreatic cancer (LAPC). METHODS AND MATERIALS Patients with LAPC treated with C-ion RT from April 2012 to December 2014 at 3 institutions were retrospectively analyzed. Patients with pathologically-confirmed invasive ductal adenocarcinoma of the pancreas were eligible. The prescribed dose was 52.8 Gy (relative biological effectiveness weighted absorbed dose; RBE) or 55.2 Gy (RBE) in 12 fractions. Overall survival (OS), distant metastasis-free survival (DMFS), local recurrence (LR), and toxicity were evaluated. RESULTS In total, 72 patients were included in this study. Tumors in the head of the pancreas were seen in 30 patients (42%), while those in the body or tail of the pancreas were seen in 42 patients (58%). Fifty-six patients (78%) received concurrent chemotherapy. The OS rates were 73% (95% confidence interval [CI], 62%-84%) at 1 year, and 46% (95% CI, 31%-61%) at 2 years with a median OS of 21.5 months (95% CI, 11.8-31.2 months). The 1- and 2-year DMFS rates were 41% (95% CI, 29%-52%) and 28% (95% CI, 16%-40%), respectively. The 1- and 2-year cumulative incidences of LR were 16% (95% CI, 9%-26%) and 24% (95% CI, 14%-36%), respectively. Nineteen patients (26%) experienced acute grade 3 or 4 hematological toxicities. Two patients (3%) had grade 3 anorexia. Late gastrointestinal (GI) grade 3 toxicity was observed in 1 patient (1%). No patients developed late grade 4 or 5 toxicity. CONCLUSIONS The first multi-institutional analysis of C-ion RT for LAPC indicated relatively favorable outcomes with limited toxicities, especially for tumors not in close proximity to GI tract.
Collapse
Affiliation(s)
- Shohei Kawashiro
- Hospital of the National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan; Department of Radiation Oncology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Shigeru Yamada
- Hospital of the National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan.
| | | | - Tatsuya Ohno
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
| | - Takashi Nakano
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
| | - Makoto Shinoto
- Ion Beam Therapy Center, SAGA HIMAT Foundation, Tosu, Japan
| | | | - Kenji Nemoto
- Department of Radiation Oncology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Yuka Isozaki
- Hospital of the National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Hiroshi Tsuji
- Hospital of the National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Tadashi Kamada
- Hospital of the National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
59
|
Sundahl N, De Wolf K, Kruse V, Meireson A, Reynders D, Goetghebeur E, Van Gele M, Speeckaert R, Hennart B, Brochez L, Ost P. Phase 1 Dose Escalation Trial of Ipilimumab and Stereotactic Body Radiation Therapy in Metastatic Melanoma. Int J Radiat Oncol Biol Phys 2018; 100:906-915. [DOI: 10.1016/j.ijrobp.2017.11.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/11/2017] [Accepted: 11/17/2017] [Indexed: 12/31/2022]
|
60
|
Hall WA, Heerkens HD, Paulson ES, Meijer GJ, Kotte AN, Knechtges P, Parikh PJ, Bassetti MF, Lee P, Aitken KL, Palta M, Myrehaug S, Koay EJ, Portelance L, Ben-Josef E, Erickson BA. Pancreatic gross tumor volume contouring on computed tomography (CT) compared with magnetic resonance imaging (MRI): Results of an international contouring conference. Pract Radiat Oncol 2018; 8:107-115. [PMID: 29426692 DOI: 10.1016/j.prro.2017.11.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 10/25/2017] [Accepted: 11/21/2017] [Indexed: 12/19/2022]
Abstract
PURPOSE Accurate identification of the gross tumor volume (GTV) in pancreatic adenocarcinoma is challenging. We sought to understand differences in GTV delineation using pancreatic computed tomography (CT) compared with magnetic resonance imaging (MRI). METHODS AND MATERIALS Twelve attending radiation oncologists were convened for an international contouring symposium. All participants had a clinical and research interest in pancreatic adenocarcinoma. CT and MRI scans from 3 pancreatic cases were used for contouring. CT and MRI GTVs were analyzed and compared. Interobserver variability was compared using Dice's similarity coefficient (DSC), Hausdorff distances, and Jaccard indices. Mann-Whitney tests were used to check for significant differences. Consensus contours on CT and MRI scans and constructed count maps were used to visualize the agreement. Agreement regarding the optimal method to determine GTV definition using MRI was reached. RESULTS Six contour sets (3 from CT and 3 from MRI) were obtained and compared for each observer, totaling 72 contour sets. The mean volume of contours on CT was significantly larger at 57.48 mL compared with a mean of 45.76 mL on MRI, P = .011. The standard deviation obtained from the CT contours was significantly larger than the standard deviation from the MRI contours (P = .027). The mean DSC was 0.73 for the CT and 0.72 for the MRI (P = .889). The conformity index measurement was similar for CT and MRI (P = .58). Count maps were created to highlight differences in the contours from CT and MRI. CONCLUSIONS Using MRI as a primary image set to define a pancreatic adenocarcinoma GTV resulted in smaller contours compared with CT. No differences in DSC or the conformity index were seen between MRI and CT. A stepwise method is recommended as an approach to contour a pancreatic GTV using MRI.
Collapse
Affiliation(s)
- William A Hall
- Department of Radiation Oncology, Medical College of Wisconsin and Clement J. Zablocki VA Medical Center, Milwaukee, Wisconsin.
| | - Hanne D Heerkens
- UMC Utrecht, Department of Radiation Oncology, Utrecht, the Netherlands
| | - Eric S Paulson
- Department of Radiation Oncology, Medical College of Wisconsin and Clement J. Zablocki VA Medical Center, Milwaukee, Wisconsin
| | - Gert J Meijer
- UMC Utrecht, Department of Radiation Oncology, Utrecht, the Netherlands
| | - Alexis N Kotte
- UMC Utrecht, Department of Radiation Oncology, Utrecht, the Netherlands
| | - Paul Knechtges
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Parag J Parikh
- Department of Radiation Oncology, Washington University, St. Louis, Missouri
| | - Michael F Bassetti
- Department of Radiation Oncology, University of Wisconsin Madison, Madison, Wisconsin
| | - Percy Lee
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California
| | | | - Manisha Palta
- Department of Radiation Oncology, Duke University, Durham, North Carolina
| | - Sten Myrehaug
- Department of Radiation Oncology, Sunnybrook Hospital, Toronto, Canada
| | - Eugene J Koay
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | | | - Edgar Ben-Josef
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Beth A Erickson
- Department of Radiation Oncology, Medical College of Wisconsin and Clement J. Zablocki VA Medical Center, Milwaukee, Wisconsin
| |
Collapse
|
61
|
Shabason JE, Chen J, Apisarnthanarax S, Damjanov N, Giantonio B, Loaiza-Bonilla A, O'Dwyer PJ, O'Hara M, Reiss KA, Teitelbaum U, Wissel P, Drebin JA, Vollmer C, Kochman M, Mick R, Vergara N, Jhala N, Doucette A, Lukens JN, Plastaras JP, Metz JM, Ben-Josef E. A phase I dose escalation trial of nab-paclitaxel and fixed dose radiation in patients with unresectable or borderline resectable pancreatic cancer. Cancer Chemother Pharmacol 2018; 81:609-614. [PMID: 29362902 DOI: 10.1007/s00280-018-3519-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 01/09/2018] [Indexed: 11/24/2022]
Abstract
PURPOSE Patients with locally advanced pancreatic cancer typically have poor outcomes, with a median survival of approximately 16 months. Novel methods to improve outcomes are needed. Nab-paclitaxel (Abraxane) has shown efficacy in pancreatic cancer and is FDA-approved for metastatic disease in combination with gemcitabine. Nab-paclitaxel is also a promising radiosensitizer based on laboratory studies, but it has never been clinically tested with definitive radiotherapy for locally advanced pancreatic carcinoma. METHODS We performed a phase 1 study using a 3 + 3 dose escalation strategy to determine the safety and tolerability of dose-escalated nab-paclitaxel with fractionated radiotherapy for patients with unresectable or borderline resectable pancreatic cancer. Following induction chemotherapy with two cycles of nab-paclitaxel and gemcitabine, patients were treated with weekly nab-paclitaxel and daily radiotherapy to a dose of 52.5 Gy in 25 fractions. Final dose-limiting toxicity (DLT) determination was performed at day 65 after the start of radiotherapy. RESULTS Nine patients received nab-paclitaxel at a dose level of either 100 mg/m2 (n = 3) or 125 mg/m2 (n = 6). There were no observed grade 3 gastrointestinal toxicities. One DLT (grade 3 neuropathy) was observed in a patient who received 125 mg/m2 of nab-paclitaxel. Other grade 3 toxicities included fatigue (11%), anemia (11%) and neutropenia (11%). No grade 4 toxicities were observed. Following chemoradiotherapy, four patients (borderline resectable, n = 2 and unresectable, n = 2) underwent surgical resection, all with negative margins and with significant treatment effect with limited tumor viability. CONCLUSIONS The combination of fractionated radiation and weekly full dose nab-paclitaxel was safe and well-tolerated.
Collapse
Affiliation(s)
- Jacob E Shabason
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, PCAM-2 West, 3400 Civic Center Blvd., Philadelphia, PA, 19104, USA.
| | - Jerry Chen
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, PCAM-2 West, 3400 Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - Smith Apisarnthanarax
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA, USA
| | - Nevena Damjanov
- Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Bruce Giantonio
- Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Arturo Loaiza-Bonilla
- Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Peter J O'Dwyer
- Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Mark O'Hara
- Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kim A Reiss
- Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Ursina Teitelbaum
- Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Paul Wissel
- Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Jeffrey A Drebin
- Department of Surgery, Memorial Sloane Kettering Cancer Center, New York, NY, USA
| | - Charles Vollmer
- Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Michael Kochman
- Gastroenterology Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Rosemarie Mick
- Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Norge Vergara
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Nirag Jhala
- Department of Pathology and Laboratory Medicine, Temple University Hospital, Philadelphia, PA, USA
| | - Abigail Doucette
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, PCAM-2 West, 3400 Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - John N Lukens
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, PCAM-2 West, 3400 Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - John P Plastaras
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, PCAM-2 West, 3400 Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - James M Metz
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, PCAM-2 West, 3400 Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - Edgar Ben-Josef
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, PCAM-2 West, 3400 Civic Center Blvd., Philadelphia, PA, 19104, USA
| |
Collapse
|
62
|
Abstract
Pancreatic cancer is an aggressive malignancy with a poor long-term survival and only mild improvement in outcomes over the past 30 years. Local failure remains a problem and radiation can help improve control. The role of radiation therapy in has been controversial and is still evolving. This article reviews the trials of pancreatic cancer and radiation in adjuvant, neoadjuvant, and unresectable lesions. The article reviews the impact and outcomes of evolving radiation technology.
Collapse
|
63
|
O Kane GM, Knox JJ. Locally advanced pancreatic cancer: An emerging entity. Curr Probl Cancer 2017; 42:12-25. [PMID: 29153290 DOI: 10.1016/j.currproblcancer.2017.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 10/29/2017] [Accepted: 10/30/2017] [Indexed: 12/23/2022]
Abstract
Pancreatic adenocarcinoma (PDAC) remains a highly fatal disease that is increasing in incidence. PDAC can be classified according to resectability status with 3 nonmetastatic groups defined: resectable, borderline resectable, and locally advanced PDAC (LAPC). Delineating these subtypes is important with the optimal treatment approach dictated by high-quality CT imaging and multidisciplinary team discussion. Patients with LAPC are thought unresectable and are therefore rarely cured. In these patients, chemotherapy remains the mainstay of treatment. Aggressive approaches in this cohort are increasingly employed. Local therapies after induction chemotherapy including standard fractionation radiation, stereotactic body radiotherapy (SBRT), and irreversible electroporation (IRE) are being investigated in an attempt to improve long-term control. In some cases, responses to neoadjuvant therapy may facilitate surgical resection. Biomarkers that can select patients most likely to benefit from these options are urgently needed. This review aims to highlight the emerging treatment of patients with LAPC and to discuss current trials.
Collapse
|
64
|
Dose-escalated radiotherapy for unresectable or locally recurrent pancreatic cancer: Dose volume analysis, toxicity and outcome of 28 consecutive patients. PLoS One 2017; 12:e0186341. [PMID: 29023527 PMCID: PMC5638513 DOI: 10.1371/journal.pone.0186341] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 10/01/2017] [Indexed: 01/10/2023] Open
Abstract
PURPOSE The role of radiotherapy for unresectable pancreatic cancer is controversial. A benefit of additional radiotherapy is supported by some observations. A dose-effect relationship was recently found by dose escalation employing image guided and intensity modulated radiotherapy. METHODS We retrospectively evaluated 28 consecutive patients, all with history of extensive prior therapies for unresectable locally advanced/ recurrent pancreatic cancer (LAPC/LRPC). Treatment was delivered by helical tomotherapy after daily position verification with computed tomography. Dose to the planned target volume (PTV) was 51 Gy, while the dose to the macroscopic tumor was escalated by a simultaneous integrated boost to a median cumulative dose of 66 Gy (60-66 Gy). Concomitant chemotherapy consisted mainly of capecitabine (n = 23). RESULTS 10 of 28 patients presented acute toxicities > grade 2, one patient succumbed to gastrointestinal bleeding after treatment. No correlations of toxicities and dose volume histograms (DVH) of retrospectively delineated small bowel loops were observed, although average small bowel volume receiving ≥ 20 Gy was 374 ml. DVH analyses revealed a correlation of splenic parameters and acute toxicity: Vomiting, anorexia, dehydration, hematologic toxicity, fatigue, combined gastro-intestinal toxicity wit R-values between 0.392 and 0.561 (all p-values > 0.05). Only one patient developed late toxicities > grade 2. With an average follow-up time in surviving patients of 14 months median overall survival time was 19 months and median time to local recurrence 13 months. In 8 patients with available imaging of local recurrence: 5 in field recurrences, 2 marginal recurrences and one lymph node recurrence outside the high dose radiation field were observed. In univariate analysis only ΔCA-19-9 during radiotherapy was associated with local control (p = 0.029) and overall survival (p = 0.049). CONCLUSION Dose escalated normo-fractionated radiotherapy for LAPC/LRPC seems feasible and suitable to prolong local control and in consequence long-term survival. However, in-field local progression is still frequently observed and possibilities to increase the local effectiveness should be evaluated. Exposure of the spleen was predictive for acute toxicity and should be further investigated.
Collapse
|
65
|
Deng Z, Yang W, Pang J, Bi X, Tuli R, Li D, Fan Z. Improved vessel-tissue contrast and image quality in 3D radial sampling-based 4D-MRI. J Appl Clin Med Phys 2017; 18:250-257. [PMID: 28980395 PMCID: PMC5689937 DOI: 10.1002/acm2.12194] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 07/21/2017] [Accepted: 08/27/2017] [Indexed: 12/26/2022] Open
Abstract
Purpose In radiation treatment planning for thoracic and abdominal tumors, 4D‐MRI has shown promise in respiratory motion characterization with improved soft‐tissue contrast compared to clinical standard, 4D computed tomography (4D‐CT). This study aimed to further improve vessel–tissue contrast and overall image quality in 3D radial sampling‐based 4D‐MRI using a slab‐selective (SS) excitation approach. Methods The technique was implemented in a 3D radial sampling with self‐gating‐based k‐space sorting sequence. The SS excitation approach was compared to a non‐selective (NS) approach in six cancer patients and two healthy volunteers at 3T. Improvements in vessel–tissue contrast ratio (CR) and vessel signal‐to‐noise ratio (SNR) were analyzed in five of the eight subjects. Image quality was visually assessed in all subjects on a 4‐point scale (0: poor; 3: excellent). Tumor (patients) and pancreas (healthy) motion trajectories were compared between the two imaging approaches. Results Compared with NS‐4D‐MRI, SS‐4D‐MRI significantly improved the overall vessel–tissue CR (2.60 ± 3.97 vs. 1.03 ± 1.44, P < 0.05), SNR (63.33 ± 38.45 vs. 35.74 ± 28.59, P < 0.05), and image quality score (2.6 ± 0.5 vs. 1.4 ± 0.5, P = 0.02). Motion trajectories from the two approaches exhibited strong correlation in the superior–inferior (0.96 ± 0.06), but weaker in the anterior–posterior (0.78 ± 0.24) and medial–lateral directions (0.46 ± 0.44). Conclusions The proposed 4D‐MRI with slab‐selectively excited 3D radial sampling allows for improved blood SNR, vessel–tissue CR, and image quality.
Collapse
Affiliation(s)
- Zixin Deng
- Department of Biomedical Sciences, Biomedical Imaging Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA.,Department of Bioengineering, University of California, Los Angeles, CA, USA
| | - Wensha Yang
- Department of Biomedical Sciences, Biomedical Imaging Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA.,Department of Radiation Oncology, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Jianing Pang
- Department of Biomedical Sciences, Biomedical Imaging Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA.,MR R&D, Siemens Healthineers, Chicago, IL, USA
| | - Xiaoming Bi
- MR R&D, Siemens Healthineers, Los Angeles, CA, USA
| | - Richard Tuli
- Department of Radiation Oncology, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Debiao Li
- Department of Biomedical Sciences, Biomedical Imaging Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA.,Department of Bioengineering, University of California, Los Angeles, CA, USA.,Department of Medicine, University of California, Los Angeles, CA, USA
| | - Zhaoyang Fan
- Department of Biomedical Sciences, Biomedical Imaging Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA.,Department of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
66
|
Rao AD, Feng Z, Shin EJ, He J, Waters KM, Coquia S, DeJong R, Rosati LM, Su L, Li D, Jackson J, Clark S, Schultz J, Hutchings D, Kim SH, Hruban RH, DeWeese TL, Wong J, Narang A, Herman JM, Ding K. A Novel Absorbable Radiopaque Hydrogel Spacer to Separate the Head of the Pancreas and Duodenum in Radiation Therapy for Pancreatic Cancer. Int J Radiat Oncol Biol Phys 2017; 99:1111-1120. [PMID: 28943075 DOI: 10.1016/j.ijrobp.2017.08.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 08/04/2017] [Accepted: 08/07/2017] [Indexed: 01/25/2023]
Abstract
PURPOSE We assessed the feasibility and theoretical dosimetric advantages of an injectable hydrogel to increase the space between the head of the pancreas (HOP) and duodenum in a human cadaveric model. METHODS AND MATERIALS Using 3 human cadaveric specimens, an absorbable radiopaque hydrogel was injected between the HOP and duodenum by way of open laparotomy in 1 case and endoscopic ultrasound (EUS) guidance in 2 cases. The cadavers were subsequently imaged using computed tomography and dissected for histologic confirmation of hydrogel placement. The duodenal dose reduction and planning target volume (PTV) coverage were characterized using pre- and postspacer injection stereotactic body radiation therapy (SBRT) plans for the 2 cadavers with EUS-guided placement, the delivery method that appeared the most clinically desirable. Modeling studies were performed using 60 SBRT plans consisting of 10 previously treated patients with unresectable pancreatic cancer, each with 6 different HOP-duodenum separation distances. The duodenal volume receiving 15 Gy (V15), 20 Gy (V20), and 33 Gy (V33) was assessed for each iteration. RESULTS In the 3 cadaveric studies, an average of 0.9 cm, 1.1 cm, and 0.9 cm HOP-duodenum separation was achieved. In the 2 EUS cases, the V20 decreased from 3.86 cm3 to 0.36 cm3 and 3.75 cm3 to 1.08 cm3 (treatment constraint <3 cm3), and the V15 decreased from 7.07 cm3 to 2.02 cm3 and 9.12 cm3 to 3.91 cm3 (treatment constraint <9 cm3). The PTV coverage improved or was comparable between the pre- and postinjection studies. Modeling studies demonstrated that a separation of 8 mm was sufficient to consistently reduce the V15, V20, and V33 to acceptable clinical constraints. CONCLUSIONS Currently, dose escalation has been limited owing to radiosensitive structures adjacent to the pancreas. We demonstrated the feasibility of hydrogel separation of the HOP and duodenum. Future studies will evaluate the safety and efficacy of this technique with the potential for more effective dose escalation using SBRT or intensity-modulated radiation therapy to improve the outcomes in patients with unresectable pancreatic cancer.
Collapse
Affiliation(s)
- Avani D Rao
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ziwei Feng
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Eun Ji Shin
- Department of Gastroenterology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jin He
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Kevin M Waters
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Stephanie Coquia
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Robert DeJong
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Lauren M Rosati
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Lin Su
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Dengwang Li
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Juan Jackson
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Stephen Clark
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jeffrey Schultz
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Danielle Hutchings
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Seong-Hun Kim
- Department of Gastroenterology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ralph H Hruban
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Theodore L DeWeese
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - John Wong
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Amol Narang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Joseph M Herman
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Kai Ding
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland.
| |
Collapse
|
67
|
Boimel PJ, Berman AT, Li J, Apisarnthanarax S, Both S, Lelionis K, Larson GL, Teitelbaum U, Lukens JN, Ben-Josef E, Metz JM, Plastaras JP. Proton beam reirradiation for locally recurrent pancreatic adenocarcinoma. J Gastrointest Oncol 2017; 8:665-674. [PMID: 28890817 DOI: 10.21037/jgo.2017.03.04] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Local recurrence following definitive treatment for pancreatic adenocarcinoma is common and can be associated with significant morbidity and mortality. Retreatment options for these patients are limited. Proton beam reirradiation (PRT) may limit dose and toxicity to previously irradiated normal tissues in patients without evidence of metastatic disease. METHODS Between 8/2010-2/2015, 15 patients with isolated, locally-recurrent pancreatic cancer were treated with PRT. Acute toxicity was graded using CTC v 4.0 and defined as occurring within 90 days. Kaplan-Meier survival analysis was performed from the start of PRT. A log-rank test was used to compare survival with or without concurrent chemotherapy. RESULTS Median follow-up was 15.7 months [2-48] from the start of PRT. The median clinical target volume (CTV) was 71 cc [15-200]. Ten (67%) patients received concurrent chemotherapy. Median PRT dose was 59.4 Gy (37.5-59.4 Gy). The median time interval from the prior treatment course was 26.7 months (7-461.3). There was a rate of 13% acute ≥ grade 3 toxicities attributed to PRT. The median overall survival (OS) was 16.7 months (95% CI, 4.7-36) and OS at 1 year was 67%. The "in-field" failure free survival at one year was 87%. The locoregional progression free survival (LPFS) and distant metastasis free survival (DMFS) at 1 year was 72% and 64% respectively. Concurrent chemotherapy was associated with a higher median survival. CONCLUSIONS PRT was well tolerated, resulted in prolonged clinical outcomes compared to historical controls, and should be considered as a treatment option with concurrent chemotherapy in selected patients with locally-recurrent pancreatic cancer.
Collapse
Affiliation(s)
- Pamela J Boimel
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, USA
| | - Abigail T Berman
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, USA
| | - Jonathan Li
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, USA
| | | | - Stefan Both
- Department of Radiation Oncology Memorial Sloan Kettering Cancer Center, New York, USA
| | - Kristi Lelionis
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, USA
| | | | - Ursina Teitelbaum
- Department of Hematology Oncology, University of Pennsylvania, Philadelphia, USA
| | - John N Lukens
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, USA
| | - Edgar Ben-Josef
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, USA
| | - James M Metz
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, USA
| | - John P Plastaras
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
68
|
Torgeson A, Lloyd S, Boothe D, Tao R, Whisenant J, Garrido-Laguna I, Cannon GM. Multiagent induction chemotherapy followed by chemoradiation is associated with improved survival in locally advanced pancreatic cancer. Cancer 2017. [DOI: 10.1002/cncr.30780] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Anna Torgeson
- University of Utah; Department of Radiation Oncology, University of Utah; Salt Lake City Utah
| | - Shane Lloyd
- University of Utah; Department of Radiation Oncology, University of Utah; Salt Lake City Utah
| | - Dustin Boothe
- University of Utah; Department of Radiation Oncology, University of Utah; Salt Lake City Utah
| | - Randa Tao
- University of Utah; Department of Radiation Oncology, University of Utah; Salt Lake City Utah
| | - Jonathan Whisenant
- Department of Internal Medicine; University of Utah; Salt Lake City Utah
| | | | - George M. Cannon
- Department of Radiation Oncology; Intermountain Medical Center; Murray Utah
| |
Collapse
|
69
|
Modelling duodenum radiotherapy toxicity using cohort dose-volume-histogram data. Radiother Oncol 2017; 123:431-437. [PMID: 28600084 PMCID: PMC5486774 DOI: 10.1016/j.radonc.2017.04.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 04/20/2017] [Accepted: 04/24/2017] [Indexed: 12/25/2022]
Abstract
Background and purpose Gastro-intestinal toxicity is dose-limiting in abdominal radiotherapy and correlated with duodenum dose-volume parameters. We aimed to derive updated NTCP model parameters using published data and prospective radiotherapy quality-assured cohort data. Material and methods A systematic search identified publications providing duodenum dose-volume histogram (DVH) statistics for clinical studies of conventionally-fractionated radiotherapy. Values for the Lyman-Kutcher-Burman (LKB) NTCP model were derived through sum-squared-error minimisation and using leave-one-out cross-validation. Data were corrected for fraction size and weighted according to patient numbers, and the model refined using individual patient DVH data for two further cohorts from prospective clinical trials. Results Six studies with published DVH data were utilised, and with individual patient data included outcomes for 531 patients in total (median follow-up 16 months). Observed gastro-intestinal toxicity rates ranged from 0% to 14% (median 8%). LKB parameter values for unconstrained fit to published data were: n = 0.070, m = 0.46, TD50(1) [Gy] = 183.8, while the values for the model incorporating the individual patient data were n = 0.193, m = 0.51, TD50(1) [Gy] = 299.1. Conclusions LKB parameters derived using published data are shown to be consistent to those previously obtained using individual patient data, supporting a small volume-effect and dependence on exposure to high threshold dose.
Collapse
|
70
|
Comito T, Cozzi L, Clerici E, Franzese C, Tozzi A, Iftode C, Navarria P, D’Agostino G, Rimassa L, Carnaghi C, Personeni N, Tronconi MC, De Rose F, Franceschini D, Ascolese AM, Fogliata A, Tomatis S, Santoro A, Zerbi A, Scorsetti M. Can Stereotactic Body Radiation Therapy Be a Viable and Efficient Therapeutic Option for Unresectable Locally Advanced Pancreatic Adenocarcinoma? Results of a Phase 2 Study. Technol Cancer Res Treat 2017; 16:295-301. [PMID: 27311310 PMCID: PMC5616043 DOI: 10.1177/1533034616650778] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/14/2016] [Accepted: 04/22/2016] [Indexed: 12/11/2022] Open
Abstract
PURPOSE To assess the efficacy of stereotactic body radiotherapy in patients with unresectable locally advanced pancreatic cancer. MATERIALS AND METHODS All patients received a prescription dose of 45 Gy in 6 fractions. Primary end point was freedom from local progression. Secondary end points were overall survival, progression-free survival, and toxicity. Actuarial survival analysis and univariate or multivariate analysis were investigated. RESULTS Forty-five patients were enrolled in a phase 2 trial. Median follow-up was 13.5 months. Freedom from local progression was 90% at 2 years. On univariate ( P < .03) and multivariate analyses ( P < .001), lesion size was statistically significant for freedom from local progression. Median progression-free survival and overall survival were 8 and 13 months, respectively. On multivariate analysis, tumor size ( P < .001) and freedom from local progression ( P < .002) were significantly correlated with overall survival. Thirty-two (71%) patients with locally advanced pancreatic cancer received chemotherapy before stereotactic body radiotherapy. Median overall survival from diagnosis was 19 months. Multivariate analysis showed that freedom from local progression ( P < .035), tumor diameter ( P < .002), and computed tomography before stereotactic body radiotherapy ( P < .001) were significantly correlated with overall survival from diagnosis. CONCLUSION Stereotactic body radiotherapy is a safe and effective treatment for patients with locally advanced pancreatic cancer with no G3 toxicity or greater and could be a promising therapeutic option in multimodality treatment regimen.
Collapse
Affiliation(s)
| | - L. Cozzi
- Radiotherapy, Istituto Clinico Humanitas, Milano, Italy
| | - E. Clerici
- Radiotherapy, Istituto Clinico Humanitas, Milano, Italy
| | - C. Franzese
- Radiotherapy, Istituto Clinico Humanitas, Milano, Italy
| | - A. Tozzi
- Radiotherapy, Istituto Clinico Humanitas, Milano, Italy
| | - C. Iftode
- Radiotherapy, Istituto Clinico Humanitas, Milano, Italy
| | - P. Navarria
- Radiotherapy, Istituto Clinico Humanitas, Milano, Italy
| | - G. D’Agostino
- Radiotherapy, Istituto Clinico Humanitas, Milano, Italy
| | - L. Rimassa
- Oncology and Hematology, Istituto Clinico Humanitas, Milano, Italy
| | - C. Carnaghi
- Oncology and Hematology, Istituto Clinico Humanitas, Milano, Italy
| | - N. Personeni
- Oncology and Hematology, Istituto Clinico Humanitas, Milano, Italy
| | - M. C. Tronconi
- Oncology and Hematology, Istituto Clinico Humanitas, Milano, Italy
| | - F. De Rose
- Radiotherapy, Istituto Clinico Humanitas, Milano, Italy
| | | | | | - A. Fogliata
- Radiotherapy, Istituto Clinico Humanitas, Milano, Italy
| | - S. Tomatis
- Radiotherapy, Istituto Clinico Humanitas, Milano, Italy
| | - A. Santoro
- Oncology and Hematology, Istituto Clinico Humanitas, Milano, Italy
| | - A. Zerbi
- Pancreatic Surgery, Istituto Clinico Humanitas, Milano, Italy
| | - M. Scorsetti
- Radiotherapy, Istituto Clinico Humanitas, Milano, Italy
| |
Collapse
|
71
|
Chung SY, Chang JS, Lee BM, Kim KH, Lee KJ, Seong J. Dose escalation in locally advanced pancreatic cancer patients receiving chemoradiotherapy. Radiother Oncol 2017; 123:438-445. [PMID: 28464997 DOI: 10.1016/j.radonc.2017.04.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 03/30/2017] [Accepted: 04/05/2017] [Indexed: 12/28/2022]
Abstract
PURPOSE To investigate whether radiotherapy (RT) dose escalation would improve treatment outcomes without increasing severe toxicity in locally advanced pancreatic cancer patients. METHODS From 2005 to 2015, 497 locally advanced pancreatic cancer patients who received neoadjuvant or definitive chemoradiotherapy (CCRT) were included. Patients were divided according to the total dose (TD). Overall survival (OS), progression-free survival (PFS), local failure-free rate (LFFR), distant failure-free rate (DFFR), and toxicity rates were compared between <61Gy (n=345) and ≥61Gy groups (n=152). Additionally, propensity score matching was performed. RESULTS At a median follow-up of 19.3months (range, 4.8-128.5months), the 1-year OS, PFS, LFFR, and DFFR were significantly higher in the ≥61Gy group. After multivariate analysis, a TD of ≥61Gy remained a significant favorable factor for OS (p=0.019), PFS (p=0.001), LFFR (p=0.004), and DFFR (p=0.008). After propensity score matching, the ≥61Gy group still showed higher OS, PFS, and LFFR, but not DFFR (p=0.205). The acute and late toxicity rates showed no significant difference between the two groups. CONCLUSION Patients who received a higher RT dose showed not only improved PFS and LFFR, but also improved OS without an increase in severe toxicity. Dose-escalated CCRT can be a favorable treatment option in locally advanced pancreatic cancer patients.
Collapse
Affiliation(s)
- Seung Yeun Chung
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jee Suk Chang
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byung Min Lee
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyung Hwan Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyong Joo Lee
- Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jinsil Seong
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
72
|
Colbert LE, Moningi S, Chadha A, Amer A, Lee Y, Wolff RA, Varadhachary G, Fleming J, Katz M, Das P, Krishnan S, Koay EJ, Park P, Crane CH, Taniguchi CM. Dose escalation with an IMRT technique in 15 to 28 fractions is better tolerated than standard doses of 3DCRT for LAPC. Adv Radiat Oncol 2017; 2:403-415. [PMID: 29114609 PMCID: PMC5605283 DOI: 10.1016/j.adro.2017.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 02/16/2017] [Accepted: 02/21/2017] [Indexed: 01/20/2023] Open
Abstract
Purpose To review acute and late toxicities after chemoradiation for locally advanced pancreatic ductal adenocarcinoma in patients who were treated with escalated dose radiation (EDR). Methods and materials Maximum Common Terminology Criteria for Adverse Events Version 4.0 acute toxicities (AT) during radiation and within 60 days after radiation were recorded for both acute gastrointestinal toxicity and overall toxicity (OT). Late toxicities were also recorded. EDR was generally delivered with daily image guidance and breath-hold techniques using intensity modulated radiation therapy (IMRT) planning. These were compared with patients who received standard dose radiation (SDR) delivered as 50.4 Gy in 28 fractions using 3-dimensional chemoradiation therapy planning. Results A total of 59 of 154 patients (39%) received EDR with biologically equivalent doses >70 Gy. The most frequent schedules were 63 Gy in 28 fractions (19 of 154 patients), 67.5 Gy in 15 fractions (10 of 154 patients), and 70 Gy in 28 fractions (15 of 154 patients). No grade 4 or grade 5 OT or late toxicities were reported. Rates of grade 3 acute gastrointestinal toxicity were significantly lower in patients who received EDR compared with SDR (1% vs 14%; P < .001). Similarly, rates of grade 3 OT were also lower for EDR compared with SDR (4% vs 16%; P = .004). The proportion of patients who experienced no AT was higher in the EDR group than the SDR group (36% vs 15%; P = .001). For EDR patients treated with IMRT, a lower risk of AT was associated with a later treatment year (P = .007), nonpancreatic head tumor location (P = .01), breath-hold (P = .002), 4-dimensional computed tomography (P = .003), computed tomography on rails (P = .002), and lower stomach V40 (P = .03). With a median time of 12 months (range, 1-79 months) from the start of radiation therapy to the last known follow-up in the EDR group, 51 of 59 patients (86%) had no late toxicity. Six of 59 EDR patients (10%) had either strictures or gastrointestinal bleeding that required intervention. No significant predictors of late toxicity were identified. Conclusion Overall acute and late toxicity rates were low with EDR using an IMRT technique with image guidance and respiratory gating.
Collapse
Affiliation(s)
- Lauren E Colbert
- Department of Radiation Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Shalini Moningi
- Department of Radiation Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Awalpreet Chadha
- Department of Radiation Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Ahmed Amer
- Department of Radiation Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Yeonju Lee
- Department of Radiation Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Robert A Wolff
- Department of Medical Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Gauri Varadhachary
- Department of Medical Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Jason Fleming
- Department of Surgical Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Matthew Katz
- Department of Surgical Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Prajnan Das
- Department of Radiation Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Sunil Krishnan
- Department of Radiation Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Eugene J Koay
- Department of Radiation Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Peter Park
- Department of Radiation Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Christopher H Crane
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Cullen M Taniguchi
- Department of Radiation Oncology, UT MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
73
|
Huguet F, Hajj C, Winston CB, Shi W, Zhang Z, Wu AJ, O’Reilly EM, Reidy DL, Allen P, Goodman KA. Chemotherapy and intensity-modulated radiation therapy for locally advanced pancreatic cancer achieves a high rate of R0 resection. Acta Oncol 2017; 56:384-390. [PMID: 27796165 DOI: 10.1080/0284186x.2016.1245862] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND To assess local control, survival and conversion to resectability among locally advanced pancreatic cancer (LAPC) patients treated with induction chemotherapy (ICT) followed by chemoradiotherapy treatment using intensity-modulated radiation therapy (IMRT). MATERIAL AND METHODS Between 2007 and 2012, 134 LAPC patients were treated with ICT followed by IMRT. After chemoradiotherapy, 40 patients received maintenance chemotherapy. RESULTS With a median follow-up of 20 months, median overall survival (OS) was 23 months. One- and two-year OS was 85% and 47%, respectively. On multivariate analysis, progression of disease after IMRT was associated with worse OS. Cumulative incidence of local failure was 10% at one year and 36% at two years. Twenty-six patients (19%) underwent resection after chemoradiotherapy including 22 patients (85%) with negative margins. On multivariate analysis, response to IMRT was associated with surgery (p = .01). Acute grade 3-4 hematologic and non-hematologic toxicity rates were 26% and 4.5%, respectively. CONCLUSION IMRT is safe in patients with LAPC. Patients with non-progressive LAPC after ICT and who received IMRT had high rates of local control and prolonged survival.
Collapse
Affiliation(s)
- Florence Huguet
- Department of Radiation Oncology, Tenon Hospital, Hôpitaux Universitaires Est Parisien, University Pierre and Marie Curie Paris VI, Paris, France
- Department of Radiation Oncology at Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Carla Hajj
- Department of Radiation Oncology at Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Corrine B. Winston
- Department of Radiology at Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Weiji Shi
- Department of Biostatistics at Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Zhigang Zhang
- Department of Biostatistics at Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Abraham J. Wu
- Department of Radiation Oncology at Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Eileen M. O’Reilly
- Department of Medical Oncology at Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Diane L. Reidy
- Department of Medical Oncology at Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Peter Allen
- Department of Surgery at Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Karyn A. Goodman
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
74
|
Chiuzan C, Shtaynberger J, Manji GA, Duong JK, Schwartz GK, Ivanova A, Lee SM. Dose-finding designs for trials of molecularly targeted agents and immunotherapies. J Biopharm Stat 2017; 27:477-494. [PMID: 28166468 DOI: 10.1080/10543406.2017.1289952] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Recently, there has been a surge of early phase trials of molecularly targeted agents (MTAs) and immunotherapies. These new therapies have different toxicity profiles compared to cytotoxic therapies. MTAs can benefit from new trial designs that allow inclusion of low-grade toxicities, late-onset toxicities, addition of an efficacy endpoint, and flexibility in the specification of a target toxicity probability. To study the degree of adoption of these methods, we conducted a Web of Science search of articles published between 2008 and 2014 that describe phase 1 oncology trials. Trials were categorized based on the dose-finding design used and the type of drug studied. Out of 1,712 dose-finding trials that met our criteria, 1,591 (92.9%) utilized a rule-based design, and 92 (5.4%; range 2.3% in 2009 to 9.7% in 2014) utilized a model-based or novel design. Over half of the trials tested an MTA or immunotherapy. Among the MTA and immunotherapy trials, 5.8% used model-based methods, compared to 3.9% and 8.3% of the chemotherapy or radiotherapy trials, respectively. While the percentage of trials using novel dose-finding designs has tripled since 2007, the adoption of these designs continues to remain low.
Collapse
Affiliation(s)
- Cody Chiuzan
- a Department of Biostatistics, Mailman School of Public Health , Columbia University , New York , New York , USA
| | - Jonathan Shtaynberger
- a Department of Biostatistics, Mailman School of Public Health , Columbia University , New York , New York , USA
| | - Gulam A Manji
- b Division of Hematology and Oncology, Department of Medicine , Columbia University , New York , New York , USA
| | - Jimmy K Duong
- a Department of Biostatistics, Mailman School of Public Health , Columbia University , New York , New York , USA
| | - Gary K Schwartz
- b Division of Hematology and Oncology, Department of Medicine , Columbia University , New York , New York , USA
| | - Anastasia Ivanova
- c Department of Biostatistics , UNC at Chapel Hill , Chapel Hill , North Carolina , USA
| | - Shing M Lee
- a Department of Biostatistics, Mailman School of Public Health , Columbia University , New York , New York , USA
| |
Collapse
|
75
|
Verginadis II, Kanade R, Bell B, Koduri S, Ben-Josef E, Koumenis C. A Novel Mouse Model to Study Image-Guided, Radiation-Induced Intestinal Injury and Preclinical Screening of Radioprotectors. Cancer Res 2016; 77:908-917. [PMID: 28011621 DOI: 10.1158/0008-5472.can-16-2724] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 11/14/2016] [Accepted: 11/23/2016] [Indexed: 11/16/2022]
Abstract
Radiation is an important treatment modality for gastrointestinal tumors, but intestinal injury is a common side effect. Here we describe a physiologically relevant model for studying the molecular determinants of radiation-induced intestinal damage and testing novel radioprotectors. The model employs a radiopaque marker implanted into the surface of the mouse jejunum, serving as a fiducial marker for precise radiation targeting. Mice were imaged with Cone-Beam CT (CBCT) and irradiated (IR) to the marked area using the Small Animal Radiation Research Platform (SARRP). IR-induced damage was acute but reversible and largely restricted to the area of the marker, leaving the surrounding tissues intact. Although whole gut irradiation with these doses caused lethal GI syndrome, focal (5 mm) radiation of the intestine did not cause any weight loss or lethality. However, fibrosis and collagen deposition 4 months post-IR indicated chronic intestinal damage. A separate cohort of mice was treated daily with curcumin, a clinically tested radioprotector, prior to and post-IR. Curcumin-treated mice showed significant decreases in both local and systemic inflammatory cytokine levels and in fibrosis, suggesting it is an effective radioprotector of the intestine. Our results indicate that this model, which emulates clinically relevant intestinal radiation-induced injury, can be used to assess radioprotectors prior to testing in the clinic. Cancer Res; 77(4); 908-17. ©2016 AACR.
Collapse
Affiliation(s)
- Ioannis I Verginadis
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rahul Kanade
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brett Bell
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sravya Koduri
- Department of Biology, Drexel University, Philadelphia, Pennsylvania
| | - Edgar Ben-Josef
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Constantinos Koumenis
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
76
|
Neoadjuvant Gemcitabine Chemotherapy followed by Concurrent IMRT Simultaneous Boost Achieves High R0 Resection in Borderline Resectable Pancreatic Cancer Patients. PLoS One 2016; 11:e0166606. [PMID: 27935952 PMCID: PMC5147831 DOI: 10.1371/journal.pone.0166606] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 11/01/2016] [Indexed: 12/17/2022] Open
Abstract
Background To study the feasibility of down stage the borderline resectable pancreatic cancer (BRPC) to resectable disease, we reported our institutional results using an intensity-modulated radiation therapy (IMRT) simultaneous integrated boost (SIB) dose escalation approach to improve R0 resectability. Methods We reviewed our past 7 years of experience of using neoadjuvant induction chemotherapy with Gemcitabine followed by concurrent chemoradiaiton for BRPC. During the concurrent, chemo was 5-FU and radiation were IMRT with SIB technique to target the key areas with dose escalation to 5600 in 28 fractions. The key areas were defined by PET positive area. This was followed by restaging imaging to rule out distant metastases before resection. Results 25 finished dose escalation protocol. 2 of the 25 cases developed distant metastases, 23 (92%) patients without distant metastases underwent pancreatectomy. Among the those received pancreatectomy, 22 (95%) achieved negative margin (R0). The gastrointestinal toxicity > grade 2 was 8% and there was no grade 4 toxicity. Conclusion Neoadjuvant Gemcitabine-based induction chemotherapy followed by 5-FU-based IMRT-SIB is a feasible option in improving the likelihood of R0 resection rate in BRPC without compromising the organs at risk for toxicity.
Collapse
|
77
|
Panje C, Andratschke N, Brunner TB, Niyazi M, Guckenberger M. Stereotactic body radiotherapy for renal cell cancer and pancreatic cancer : Literature review and practice recommendations of the DEGRO Working Group on Stereotactic Radiotherapy. Strahlenther Onkol 2016; 192:875-885. [PMID: 27778052 DOI: 10.1007/s00066-016-1053-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 09/19/2016] [Indexed: 01/05/2023]
Abstract
PURPOSE This report of the Working Group on Stereotactic Radiotherapy of the German Society of Radiation Oncology (DEGRO) aims to provide a literature review and practice recommendations for stereotactic body radiotherapy (SBRT) of primary renal cell cancer and primary pancreatic cancer. METHODS A literature search on SBRT for both renal cancer and pancreatic cancer was performed with focus on prospective trials and technical aspects for clinical implementation. RESULTS Data on renal and pancreatic SBRT are limited, but show promising rates of local control for both treatment sites. For pancreatic cancer, fractionated SBRT should be preferred to single-dose treatment to reduce the risk of gastrointestinal toxicity. Motion-compensation strategies and image guidance are paramount for safe SBRT delivery in both tumor entities. CONCLUSION SBRT for renal cancer and pancreatic cancer have been successfully evaluated in phase I and phase II trials. Pancreatic SBRT should be practiced carefully and only within prospective protocols due to the risk of severe gastrointestinal toxicity. SBRT for primary renal cell cancer appears a viable option for medically inoperable patients but future research needs to better define patient selection criteria and the detailed practice of SBRT.
Collapse
Affiliation(s)
- Cédric Panje
- Department of Radiation Oncology, Zurich University Hospital, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Nikolaus Andratschke
- Department of Radiation Oncology, Zurich University Hospital, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Thomas B Brunner
- Department of Radiation Oncology, Freiburg University Hospital, Freiburg, Germany
| | - Maximilian Niyazi
- Department of Radiation Oncology, University of Munich, Munich, Germany
| | - Matthias Guckenberger
- Department of Radiation Oncology, Zurich University Hospital, Rämistrasse 100, 8091, Zurich, Switzerland.
| |
Collapse
|
78
|
Abstract
Particle irradiation of cancerous disease has gained great traction in recent years. The ability for particle therapy centers to deliver radiation with a highly conformal dose distribution while maintaining minimal exit or excess dose delivered to normal tissue, coupled with various biological advantages particularly found with heavy-ion beams, enables treatment of diseases inapproachable with conventional radiotherapy. Here, we present a review of the current status of particle therapy with regard to cancers of the gastrointestinal tract, including esophagus, liver, pancreas, and recurrent rectal cancer.
Collapse
Affiliation(s)
- Makoto Shinoto
- Ion Beam Therapy Center, SAGA HIMAT Foundation, Saga, Japan
| | - Daniel K Ebner
- Research Center Hospital for Charged Particle Therapy, National Institute of Radiological Sciences, Chiba, Japan.,Brown University Alpert Medical School, Providence, RI, USA
| | - Shigeru Yamada
- Research Center Hospital for Charged Particle Therapy, National Institute of Radiological Sciences, Chiba, Japan.
| |
Collapse
|
79
|
Kausar T, Schreiber JS, Karnak D, Parsels LA, Parsels JD, Davis MA, Zhao L, Maybaum J, Lawrence TS, Morgan MA. Sensitization of Pancreatic Cancers to Gemcitabine Chemoradiation by WEE1 Kinase Inhibition Depends on Homologous Recombination Repair. Neoplasia 2016; 17:757-66. [PMID: 26585231 PMCID: PMC4656803 DOI: 10.1016/j.neo.2015.09.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 09/18/2015] [Accepted: 09/24/2015] [Indexed: 12/12/2022] Open
Abstract
To improve the efficacy of chemoradiation therapy for locally advanced pancreatic cancer and begin to establish patient selection criteria, we investigated the combination of the WEE1 inhibitor AZD1775 with gemcitabine-radiation in homologous recombination (HR) repair proficient and deficient pancreatic cancers. Sensitization to gemcitabine-radiation by AZD1775 was assessed in pancreatic cancer cells by clonogenic survival and in patient-derived xenografts by tumor growth. The contributions of HR repair inhibition and G2 checkpoint abrogation to sensitization were assessed by γH2AX, BRCA2 manipulation, and RAD51 focus formation and pHistone H3 flow cytometry, respectively. We found that AZD1775 sensitized to gemcitabine-radiation in BRCA2 wild-type but not BRCA2 mutant pancreatic cancer cells. In all cells, AZD1775 caused inhibition of CDK1 phosphorylation and G2 checkpoint abrogation. However, sensitization by AZD1775 was associated with persistent γH2AX and inhibition of RAD51 focus formation. In HR-proficient (BRCA2 wild-type) or -deficient (BRAC2 null) isogenic cells, AZD1775 sensitized to gemcitabine-radiation in BRCA2 wild-type, but not in BRCA2 null cells, despite significant G2 checkpoint abrogation. In patient-derived pancreatic tumor xenografts, AZD1775 significantly inhibited tumor growth and impaired RAD51 focus formation in response to gemcitabine-radiation. In conclusion, WEE1 inhibition by AZD1775 is an effective strategy for sensitizing pancreatic cancers to gemcitabine chemoradiation. Although this sensitization is accompanied by inhibition of CDK1 phosphorylation and G2 checkpoint abrogation, this mechanism is not sufficient for sensitization. Our findings demonstrate that sensitization to chemoradiation by WEE1 inhibition results from inhibition of HR repair and suggest that patient tumors without underlying HR defects would benefit most from this therapy.
Collapse
Affiliation(s)
- Tasneem Kausar
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Jason S Schreiber
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - David Karnak
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Leslie A Parsels
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Joshua D Parsels
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Mary A Davis
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Lili Zhao
- Biostatistics Unit, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109
| | - Jonathan Maybaum
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Meredith A Morgan
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI 48109.
| |
Collapse
|
80
|
Abstract
Currently, the use of radiation therapy for patients with pancreatic cancer is subject to discussion. In adjuvant setting, the standard treatment is 6 months of chemotherapy with gemcitabine and capecitabine. Chemoradiation (CRT) may improve the survival of patients with incompletely resected tumors (R1). This should be confirmed by a prospective trial. Neoadjuvant CRT is a promising treatment especially for patients with borderline resectable tumors. For patients with locally advanced tumors, there is no a standard. An induction chemotherapy followed by CRT for non-progressive patients reduces the rate of local relapse. Whereas in the first trials of CRT large fields were used, the treated volumes have been reduced to improve tolerance. Tumor movements induced by breathing should be taken in account. Intensity modulated radiation therapy allows a reduction of doses to the organs at risk. Whereas widely used, this technique is not recommended.
Collapse
|
81
|
Kishi T, Matsuo Y, Nakamura A, Nakamoto Y, Itasaka S, Mizowaki T, Togashi K, Hiraoka M. Comparative evaluation of respiratory-gated and ungated FDG-PET for target volume definition in radiotherapy treatment planning for pancreatic cancer. Radiother Oncol 2016; 120:217-21. [PMID: 27492203 DOI: 10.1016/j.radonc.2016.07.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 07/15/2016] [Accepted: 07/17/2016] [Indexed: 01/08/2023]
Abstract
OBJECTIVE The purpose of this study was to evaluate the usefulness of respiratory-gated positron emission tomography (4D-PET) in pancreatic cancer radiotherapy treatment planning (RTTP). MATERIALS AND METHODS Fourteen patients with 18F-fluorodeoxyglucose (FDG)-avid pancreatic tumours were evaluated between December 2013 and March 2015. Two sets of volumes were contoured for the pancreatic tumour of each patient. The biological target volume in three-dimensional RTTP (BTV3D) was contoured using conventional respiratory un-gated PET. The BTV3D was then expanded using population-based margins to generate a series of internal target volume 3D (ITV3D) values. The ITV 4D (ITV4D) was contoured using 4D-PET. Each of the five phases of 4D-PET was used for 4D contouring, and the ITV4D was constructed by summing the volumes defined on the five individual 4D-PET images. The relative volumes and normalized volumetric overlap were computed between ITV3D and ITV4D. RESULTS On average, the FDG-avid tumour volumes were 1.6 (range: 0.8-2.3) fold greater in the ITV4D than in the BTV3D. On average, the ITV3D values were 2.0 (range: 1.1-3.4) fold larger than the corresponding ITV4D values. CONCLUSION The ITV generated from 4D-PET can be used to improve the accuracy or reduce normal tissue irradiation compared with conventional un-gated PET-based ITV.
Collapse
Affiliation(s)
- Takahiro Kishi
- Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, Japan
| | - Yukinori Matsuo
- Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, Japan
| | - Akira Nakamura
- Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, Japan
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Japan
| | - Satoshi Itasaka
- Department of Radiation Oncology, Kurashiki Central Hospital, Japan
| | - Takashi Mizowaki
- Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, Japan
| | - Kaori Togashi
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Japan
| | - Masahiro Hiraoka
- Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, Japan
| |
Collapse
|
82
|
CA19-9 Normalization During Pre-operative Treatment Predicts Longer Survival for Patients with Locally Progressed Pancreatic Cancer. J Gastrointest Surg 2016; 20:1331-42. [PMID: 27114246 PMCID: PMC4919020 DOI: 10.1007/s11605-016-3149-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/11/2016] [Indexed: 01/31/2023]
Abstract
BACKGROUND Compared to the widely adopted 2-4 months of pre-operative therapy for patients with borderline resectable (BR) or locally advanced (LA) pancreatic ductal adenocarcinoma (PDAC), our institution tends to administer a longer duration before considering surgical resection. Using this unique approach, the aim of this study was to determine pre-operative variables associated with survival. METHODS Records from patients with BR/LA PDAC who underwent attempt at surgical resection from 1992-2014 were reviewed. RESULTS After a median duration of 6 months of pre-operative treatment, 109 patients with BR/LA PDAC (BR 63, LA 46) were explored; 93 (85.3 %) underwent pancreatectomy. Those who received at least 6 months of pre-operative treatment had longer median overall survival (OS) than those who received less (52.8 vs. 32.1 months, P = 0.044). On multivariate analysis, pre-operative treatment duration was the strongest predictor of survival (hazard ratio (HR) 4.79, P = 0.043). However, OS was similar in those whose CA19-9 normalized regardless of whether they received more or less than 6 months of chemotherapy (71.4 vs. 101.8 months, P = 0.930). CONCLUSIONS Pre-operative CA19-9 decline can guide treatment duration in patients with BR/LA PDAC. We endorse 6 months of therapy except in those patients whose values normalize, where surgery can be considered after a shorter course.
Collapse
|
83
|
Abstract
The outcomes for treatment of pancreatic cancer have not improved dramatically in many decades. However, the recent promising results with combination chemotherapy regimens for metastatic disease increase optimism for future treatments. With greater control of overt or occult metastatic disease, there will likely be an expanding role for local treatment modalities, especially given that nearly a third of pancreatic cancer patients have locally destructive disease without distant metastatic disease at the time of death. Technical advances have allowed for the safe delivery of dose-escalated radiation therapy, which can then be combined with chemotherapy, targeted agents, immunotherapy, and nanoparticulate drug delivery techniques to produce novel and improved synergistic effects. Here we discuss recent advances and future directions for multimodality therapy in pancreatic cancer.
Collapse
|
84
|
Patient-reported outcomes of a multicenter phase 2 study investigating gemcitabine and stereotactic body radiation therapy in locally advanced pancreatic cancer. Pract Radiat Oncol 2016; 6:417-424. [PMID: 27552809 DOI: 10.1016/j.prro.2016.05.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 04/20/2016] [Accepted: 05/19/2016] [Indexed: 01/09/2023]
Abstract
PURPOSE We previously reported clinical outcomes and physician-reported toxicity of gemcitabine and hypofractionated stereotactic body radiation therapy (SBRT) in locally advanced pancreatic cancer (LAPC). Here we prospectively investigate the impact of gemcitabine and SBRT on patient-reported quality of life (QoL). METHODS AND MATERIALS Forty-nine LAPC patients received 33 Gy SBRT (6.6 Gy daily fractions) upfront or after ≤3 doses of gemcitabine (1000 mg/m2) followed by gemcitabine until progression. European Organization for Research and Treatment of Cancer QoL core cancer (QLQ-C30) and pancreatic cancer-specific (European Organization for Research and Treatment of Cancer QLQ-PAN26) questionnaires were administered to patients pre-SBRT and at 4 to 6 weeks (first follow-up [1FUP]) and 4 months (2FUP) post-SBRT. Changes in QoL scores were deemed clinically relevant if median changes were at least 5 points in magnitude. RESULTS Forty-three (88%) patients completed pre-SBRT questionnaires. Of these, 88% and 51% completed questionnaires at 1FUP and 2FUP, respectively. There was no change in global QoL from pre-SBRT to 1FUP (P = .17) or 2FUP (P > .99). Statistical and clinical improvements in pancreatic pain (P = .001) and body image (P = .007) were observed from pre-SBRT to 1FUP. Patients with 1FUP and 2FUP questionnaires reported statistically and clinically improved body image (P = .016) by 4 months. Although pancreatic pain initially demonstrated statistical and clinical improvement (P = .020), scores returned to enrollment levels by 2FUP (P = .486). A statistical and clinical decline in role functioning (P = .002) was observed in patients at 2FUP. CONCLUSIONS Global QoL scores are not reduced with gemcitabine and SBRT. In this exploratory analysis, patients experience clinically relevant short-term improvements in pancreatic cancer-specific symptoms. Previously demonstrated acceptable clinical outcomes combined with these favorable QoL data indicate that SBRT can be easily integrated with other systemic therapies and may be a potential standard of care option in patients with LAPC.
Collapse
|
85
|
Eskander MF, Bliss LA, Tseng JF. Pancreatic adenocarcinoma. Curr Probl Surg 2016; 53:107-54. [DOI: 10.1067/j.cpsurg.2016.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 01/04/2016] [Indexed: 12/17/2022]
|
86
|
Affiliation(s)
- Hee Chul Park
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
87
|
Induction Chemotherapy Followed by Concurrent Full-dose Gemcitabine and Intensity-modulated Radiation Therapy for Borderline Resectable and Locally Advanced Pancreatic Adenocarcinoma. Am J Clin Oncol 2016; 39:1-7. [DOI: 10.1097/coc.0000000000000003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
88
|
Carbon Ion Radiation Therapy With Concurrent Gemcitabine for Patients With Locally Advanced Pancreatic Cancer. Int J Radiat Oncol Biol Phys 2015; 95:498-504. [PMID: 26883565 DOI: 10.1016/j.ijrobp.2015.12.362] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 12/14/2015] [Accepted: 12/15/2015] [Indexed: 12/15/2022]
Abstract
PURPOSE To determine, in the setting of locally advanced pancreatic cancer, the maximum tolerated dose of carbon ion radiation therapy (C-ion RT) and gemcitabine dose delivered concurrently and to estimate local effect and survival. METHODS AND MATERIALS Eligibility included pathologic confirmation of pancreatic invasive ductal carcinomas and radiographically unresectable disease without metastasis. Concurrent gemcitabine was administered on days 1, 8, and 15, and the dose levels were escalated from 400 to 1000 mg/m(2) under the starting dose level (43.2 GyE) of C-ion RT. The dose levels of C-ion RT were escalated from 43.2 to 55.2 GyE at 12 fractions under the fixed recommended gemcitabine dose determined. RESULTS Seventy-six patients were enrolled. Among the 72 treated patients, dose-limiting toxicity was observed in 3 patients: grade 3 infection in 1 patient and grade 4 neutropenia in 2 patients. Only 1 patient experienced a late grade 3 gastric ulcer and bleeding 10 months after C-ion RT. The recommended dose of gemcitabine with C-ion RT was found to be 1000 mg/m(2). The dose of C-ion RT with the full dose of gemcitabine (1000 mg/m(2)) was safely increased to 55.2 GyE. The freedom from local progression rate was 83% at 2 years using the Response Evaluation Criteria in Solid Tumors. The 2-year overall survival rates in all patients and in the high-dose group with stage III (≥45.6 GyE) were 35% and 48%, respectively. CONCLUSIONS Carbon ion RT with concurrent full-dose gemcitabine was well tolerated and effective in patients with unresectable locally advanced pancreatic cancer.
Collapse
|
89
|
Krishnan S, Chadha AS, Suh Y, Chen HC, Rao A, Das P, Minsky BD, Mahmood U, Delclos ME, Sawakuchi GO, Beddar S, Katz MH, Fleming JB, Javle MM, Varadhachary GR, Wolff RA, Crane CH. Focal Radiation Therapy Dose Escalation Improves Overall Survival in Locally Advanced Pancreatic Cancer Patients Receiving Induction Chemotherapy and Consolidative Chemoradiation. Int J Radiat Oncol Biol Phys 2015; 94:755-65. [PMID: 26972648 DOI: 10.1016/j.ijrobp.2015.12.003] [Citation(s) in RCA: 242] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 11/16/2015] [Accepted: 12/04/2015] [Indexed: 12/18/2022]
Abstract
PURPOSE To review outcomes of locally advanced pancreatic cancer (LAPC) patients treated with dose-escalated intensity modulated radiation therapy (IMRT) with curative intent. METHODS AND MATERIALS A total of 200 patients with LAPC were treated with induction chemotherapy followed by chemoradiation between 2006 and 2014. Of these, 47 (24%) having tumors >1 cm from the luminal organs were selected for dose-escalated IMRT (biologically effective dose [BED] >70 Gy) using a simultaneous integrated boost technique, inspiration breath hold, and computed tomographic image guidance. Fractionation was optimized for coverage of gross tumor and luminal organ sparing. A 2- to 5-mm margin around the gross tumor volume was treated using a simultaneous integrated boost with a microscopic dose. Overall survival (OS), recurrence-free survival (RFS), local-regional and distant RFS, and time to local-regional and distant recurrence, calculated from start of chemoradiation, were the outcomes of interest. RESULTS Median radiation dose was 50.4 Gy (BED = 59.47 Gy) with a concurrent capecitabine-based (86%) regimen. Patients who received BED >70 Gy had a superior OS (17.8 vs 15.0 months, P=.03), which was preserved throughout the follow-up period, with estimated OS rates at 2 years of 36% versus 19% and at 3 years of 31% versus 9% along with improved local-regional RFS (10.2 vs 6.2 months, P=.05) as compared with those receiving BED ≤70 Gy. Degree of gross tumor volume coverage did not seem to affect outcomes. No additional toxicity was observed in the high-dose group. Higher dose (BED) was the only predictor of improved OS on multivariate analysis. CONCLUSION Radiation dose escalation during consolidative chemoradiation therapy after induction chemotherapy for LAPC patients improves OS and local-regional RFS.
Collapse
Affiliation(s)
- Sunil Krishnan
- Department of Radiation Oncology, The University of Texas, Houston, Texas.
| | - Awalpreet S Chadha
- Department of Radiation Oncology, The University of Texas, Houston, Texas
| | - Yelin Suh
- Department of Radiation Physics, The University of Texas, Houston, Texas
| | - Hsiang-Chun Chen
- Department of Biostatistics, MD Anderson Cancer Center, Houston, Texas
| | - Arvind Rao
- Department of Bioinformatics and Computational Biology, MD Anderson Cancer Center, Houston, Texas
| | - Prajnan Das
- Department of Radiation Oncology, The University of Texas, Houston, Texas
| | - Bruce D Minsky
- Department of Radiation Oncology, The University of Texas, Houston, Texas
| | - Usama Mahmood
- Department of Radiation Oncology, The University of Texas, Houston, Texas
| | - Marc E Delclos
- Department of Radiation Oncology, The University of Texas, Houston, Texas
| | - Gabriel O Sawakuchi
- Department of Radiation Physics, The University of Texas, Houston, Texas; Graduate School of Biomedical Sciences, The University of Texas, Houston, Texas
| | - Sam Beddar
- Department of Radiation Physics, The University of Texas, Houston, Texas
| | - Matthew H Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jason B Fleming
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Milind M Javle
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gauri R Varadhachary
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert A Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
90
|
Silvestris N, Longo V, Cellini F, Reni M, Bittoni A, Cataldo I, Partelli S, Falconi M, Scarpa A, Brunetti O, Lorusso V, Santini D, Morganti A, Valentini V, Cascinu S. Neoadjuvant multimodal treatment of pancreatic ductal adenocarcinoma. Crit Rev Oncol Hematol 2015; 98:309-24. [PMID: 26653573 DOI: 10.1016/j.critrevonc.2015.11.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 09/14/2015] [Accepted: 11/19/2015] [Indexed: 02/07/2023] Open
Abstract
Treatment of pancreatic ductal adenocarcinoma (PDAC) is increasingly multidisciplinary, with neoadjuvant strategies (chemotherapy, radiation, and surgery) administered in patients with resectable, borderline resectable, or locally advanced disease. The rational supporting this management is the achievement of both higher margin-negative resections and conversion rates into potentially resectable disease and in vivo assessment of novel therapeutics. International guidelines suggest an initial staging of the disease followed by a multidisciplinary approach, even considering the lack of a treatment approach to be considered as standard in this setting. This review will focus on both literature data supporting these guidelines and on new opportunities related to current more active chemotherapy regimens. An analysis of the pathological assessment of response to therapy and the potential role of target therapies and translational biomarkers and ongoing clinical trials of significance will be discussed.
Collapse
Affiliation(s)
- Nicola Silvestris
- Medical Oncology Unit, National Cancer Research Centre "Giovanni Paolo II", Bari, Italy.
| | - Vito Longo
- Medical Oncology Unit, 'Mons R Dimiccoli' Hospital, Barletta, Italy
| | - Francesco Cellini
- Radiation Oncology Department, Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Michele Reni
- Medical Oncology Department, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Alessandro Bittoni
- Medical Oncology Clinic, AOU Ospedali Riuniti, Polytechnic University of the Marche Region, Ancona, Italy
| | - Ivana Cataldo
- ARC-NET Research Centre, University of Verona, Italy
| | - Stefano Partelli
- Pancreatic Unit, Department of Surgery, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Falconi
- Pancreatic Unit, Department of Surgery, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Aldo Scarpa
- ARC-NET Research Centre, University of Verona, Italy
| | - Oronzo Brunetti
- Medical Oncology Unit, National Cancer Research Centre "Giovanni Paolo II", Bari, Italy
| | - Vito Lorusso
- Medical Oncology Unit, National Cancer Research Centre "Giovanni Paolo II", Bari, Italy
| | - Daniele Santini
- Medical Oncology Unit, University Campus Biomedico, Roma, Italy
| | - Alessio Morganti
- Radiation Oncology Center, Dept. of Experimental, Diagnostic and Specialty Medicine - DIMES, University of Bologna, Italy
| | - Vincenzo Valentini
- Radiation Oncology Department, Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Stefano Cascinu
- Medical Oncology Clinic, AOU Ospedali Riuniti, Polytechnic University of the Marche Region, Ancona, Italy
| |
Collapse
|
91
|
Katz MHG, Landry J, Kindler HL. Current controversies in the stage-specific multidisciplinary management of pancreatic cancer. Am Soc Clin Oncol Educ Book 2015:e157-64. [PMID: 24857097 DOI: 10.14694/edbook_am.2014.34.e157] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Encouraging therapeutic approaches and treatment regimens for patients with both localized and metastatic pancreatic cancer have emerged over the last 5 years. However, these new strategies have brought important challenges and controversy. Clinical staging criteria are constantly evolving. No system has been uniformly adopted, limiting our understanding of the role of both pancreatectomy and neoadjuvant therapies for localized disease. The role of radiation therapy for the treatment of both resectable and unresectable pancreatic cancer remains unclear despite multiple prospective studies. Although two new systemic chemotherapy regimens have essentially transformed the care of many patients with metastatic cancer, criteria to guide their use in the general population have yet to be clearly established. Herein we provide an overview of these important controversies in the context of a broad update on the stage-specific management of patients with newly diagnosed pancreatic cancer.
Collapse
Affiliation(s)
- Matthew H G Katz
- From The University of Texas MD Anderson Cancer Center, Houston, TX; Winship Cancer Institute, Emory University, Atlanta, GA; Section of Hematology/Oncology, University of Chicago, Chicago, IL
| | - Jerome Landry
- From The University of Texas MD Anderson Cancer Center, Houston, TX; Winship Cancer Institute, Emory University, Atlanta, GA; Section of Hematology/Oncology, University of Chicago, Chicago, IL
| | - Hedy Lee Kindler
- From The University of Texas MD Anderson Cancer Center, Houston, TX; Winship Cancer Institute, Emory University, Atlanta, GA; Section of Hematology/Oncology, University of Chicago, Chicago, IL
| |
Collapse
|
92
|
Kim MM, Camelo-Piragua S, Schipper M, Tao Y, Normolle D, Junck L, Mammoser A, Betz BL, Cao Y, Kim CJ, Heth J, Sagher O, Lawrence TS, Tsien CI. Gemcitabine Plus Radiation Therapy for High-Grade Glioma: Long-Term Results of a Phase 1 Dose-Escalation Study. Int J Radiat Oncol Biol Phys 2015; 94:305-11. [PMID: 26853339 DOI: 10.1016/j.ijrobp.2015.10.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/15/2015] [Accepted: 10/20/2015] [Indexed: 01/25/2023]
Abstract
PURPOSE To evaluate the tolerability and efficacy of gemcitabine plus radiation therapy (RT) in this phase 1 study of patients with newly diagnosed malignant glioma (HGG). PATIENTS AND METHODS Between 2004 and 2012, 29 adults with HGG were enrolled. After any extent of resection, RT (60 Gy over 6 weeks) was given concurrent with escalating doses of weekly gemcitabine. Using a time-to-event continual reassessment method, 5 dose levels were evaluated starting at 500 mg/m(2) during the last 2 weeks of RT and advanced stepwise into earlier weeks. The primary objective was to determine the recommended phase 2 dose of gemcitabine plus RT. Secondary objectives included progression-free survival, overall survival (OS), and long-term toxicity. RESULTS Median follow-up was 38.1 months (range, 8.9-117.5 months); 24 patients were evaluable for toxicity. After 2005 when standard practice changed, patients with World Health Organization grade 4 tumors were no longer enrolled. Median progression-free survival for 22 patients with grade 3 tumors was 26.0 months (95% confidence interval [CI] 15.6-inestimable), and OS was 48.5 months (95% CI 26.8-inestimable). In 4 IDH mutated, 1p/19q codeleted patients, no failures occurred, with all but 1 alive at time of last follow-up. Seven with IDH mutated, non-codeleted tumors with ATRX loss had intermediate OS of 73.5 months (95% CI 32.8-inestimable). Six nonmutated, non-codeleted patients had a median OS of 26.5 months (95% CI 25.4-inestimable). The recommended phase 2 dose of gemcitabine plus RT was 750 mg/m(2)/wk given the last 4 weeks of RT. Dose reductions were most commonly due to grade 3 neutropenia; no grade 4 or 5 toxicities were seen. CONCLUSIONS Gemcitabine concurrent with RT is well-tolerated and yields promising outcomes, including in patients with adverse molecular features. It is a candidate for further study, particularly for poor-prognosis patient subgroups with HGG.
Collapse
Affiliation(s)
- Michelle M Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan.
| | | | - Matthew Schipper
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Yebin Tao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Daniel Normolle
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Larry Junck
- Department of Neurology, University of Michigan, Ann Arbor, Michigan
| | - Aaron Mammoser
- Department of Neurology, University of Michigan, Ann Arbor, Michigan
| | - Bryan L Betz
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Yue Cao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Department of Radiology, University of Michigan, Ann Arbor, Michigan; Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Christopher J Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Jason Heth
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan
| | - Oren Sagher
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Christina I Tsien
- Department of Radiation Oncology, Washington University, St. Louis, Missouri
| |
Collapse
|
93
|
Lischalk JW, Kole TP, Anjum HM, Obayomi-Davies O, Rashid A, Unger K. Four-dimensional computed tomography prediction of inter- and intrafractional upper gastrointestinal tumor motion during fractionated stereotactic body radiation therapy. Pract Radiat Oncol 2015; 6:176-182. [PMID: 26746816 DOI: 10.1016/j.prro.2015.10.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/25/2015] [Accepted: 10/08/2015] [Indexed: 12/25/2022]
Abstract
PURPOSE Respiratory-induced tumor motion of upper gastrointestinal (GI) tumors during radiation therapy is often assessed using a single 4-dimensional computed tomography (4D-CT) and presumed to be representative during fractionated treatment regimens. The purpose of this study was to examine the intra- and interfraction correlations of tumor motion between pretreatment 4D-CT and real-time fiducial-based motion tracking in patients treated with fractionated stereotactic body radiation therapy (SBRT) for upper GI malignancies. METHODS AND MATERIALS Fourteen patients with upper GI tumors underwent fractionated SBRT using the CyberKnife radiosurgical system with Synchrony respiratory motion management. Before treatment, each patient underwent a free-breathing 4D-CT scan and fiducial motion was tracked for each phase of the respiratory cycle. Real-time fiducial positions recorded during delivery of each SBRT fraction were extracted from the CyberKnife planning system. Displacements were compared between those predicted by 4D-CT and those recorded by Synchrony during treatment in the left-right (LR), anteroposterior (AP), and superoinferior (SI) directions. RESULTS The 4D-CT scans demonstrated little correlation with real-time mean fiducial displacement as determined by Pearson correlation with coefficients of 0.45, 0.52, and 0.63 in the SI, AP, and LR directions, respectively. Cohort-averaged maximum fiducial displacements based on 4D-CT and real-time tracking were measured to be 3.86 ± 1.40 mm versus 10.73 ± 7.03 mm, 2.29 ± 1.02 mm versus 4.44 ± 3.33 mm, and 1.45 ± 0.49 mm versus 2.67 ± 2.49 mm in the SI, AP, and LR directions, respectively. Mean fiducial displacements were greater than that predicted by the maximum displacements on the corresponding 4D-CT scan in 39%, 22%, and 25% of SBRT fractions in the SI, AP, and LR directions, respectively. CONCLUSIONS Comparison of 4D-CT with real-time fiducial tracking demonstrated significant inter- and intrafractional discrepancies, particularly in the SI direction, which could result in compromise of target coverage when planning with a single free-breathing 4D-CT.
Collapse
Affiliation(s)
- Jonathan W Lischalk
- Department of Radiation Medicine, Lombardi Comprehensive Cancer Center, Georgetown University Hospital, Washington, DC.
| | - Thomas P Kole
- Department of Radiation Medicine, Lombardi Comprehensive Cancer Center, Georgetown University Hospital, Washington, DC
| | - Hozaifa M Anjum
- Department of Radiation Medicine, Lombardi Comprehensive Cancer Center, Georgetown University Hospital, Washington, DC
| | - Olusola Obayomi-Davies
- Department of Radiation Medicine, Lombardi Comprehensive Cancer Center, Georgetown University Hospital, Washington, DC
| | - Abdul Rashid
- Department of Radiation Medicine, Lombardi Comprehensive Cancer Center, Georgetown University Hospital, Washington, DC
| | - Keith Unger
- Department of Radiation Medicine, Lombardi Comprehensive Cancer Center, Georgetown University Hospital, Washington, DC
| |
Collapse
|
94
|
Thompson RF, Mayekar SU, Zhai H, Both S, Apisarnthanarax S, Metz JM, Plastaras JP, Ben-Josef E. A dosimetric comparison of proton and photon therapy in unresectable cancers of the head of pancreas. Med Phys 2015; 41:081711. [PMID: 25086521 DOI: 10.1118/1.4887797] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Uncontrolled local growth is the cause of death in ∼ 30% of patients with unresectable pancreatic cancers. The addition of standard-dose radiotherapy to gemcitabine has been shown to confer a modest survival benefit in this population. Radiation dose escalation with three-dimensional planning is not feasible, but high-dose intensity-modulated radiation therapy (IMRT) has been shown to improve local control. Still, dose-escalation remains limited by gastrointestinal toxicity. In this study, the authors investigate the potential use of double scattering (DS) and pencil beam scanning (PBS) proton therapy in limiting dose to critical organs at risk. METHODS The authors compared DS, PBS, and IMRT plans in 13 patients with unresectable cancer of the pancreatic head, paying particular attention to duodenum, small intestine, stomach, liver, kidney, and cord constraints in addition to target volume coverage. All plans were calculated to 5500 cGy in 25 fractions with equivalent constraints and normalized to prescription dose. All statistics were by two-tailed paired t-test. RESULTS Both DS and PBS decreased stomach, duodenum, and small bowel dose in low-dose regions compared to IMRT (p < 0.01). However, protons yielded increased doses in the mid to high dose regions (e.g., 23.6-53.8 and 34.9-52.4 Gy for duodenum using DS and PBS, respectively; p < 0.05). Protons also increased generalized equivalent uniform dose to duodenum and stomach, however these differences were small (<5% and 10%, respectively; p < 0.01). Doses to other organs-at-risk were within institutional constraints and placed no obvious limitations on treatment planning. CONCLUSIONS Proton therapy does not appear to reduce OAR volumes receiving high dose. Protons are able to reduce the treated volume receiving low-intermediate doses, however the clinical significance of this remains to be determined in future investigations.
Collapse
Affiliation(s)
- Reid F Thompson
- University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Sonal U Mayekar
- Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Huifang Zhai
- University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Stefan Both
- University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | | | - James M Metz
- University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | | | - Edgar Ben-Josef
- University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
95
|
Mahipal A, Frakes J, Hoffe S, Kim R. Management of borderline resectable pancreatic cancer. World J Gastrointest Oncol 2015; 7:241-249. [PMID: 26483878 PMCID: PMC4606178 DOI: 10.4251/wjgo.v7.i10.241] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 07/07/2015] [Accepted: 08/11/2015] [Indexed: 02/05/2023] Open
Abstract
Pancreatic cancer is the fourth most common cause of cancer death in the United States. Surgery remains the only curative option; however only 20% of the patients have resectable disease at the time of initial presentation. The definition of borderline resectable pancreatic cancer is not uniform but generally denotes to regional vessel involvement that makes it unlikely to have negative surgical margins. The accurate staging of pancreatic cancer requires triple phase computed tomography or magnetic resonance imaging of the pancreas. Management of patients with borderline resectable pancreatic cancer remains unclear. The data for treatment of these patients is primarily derived from retrospective single institution experience. The prospective trials have been plagued by small numbers and poor accrual. Neoadjuvant therapy is recommended and typically consists of chemotherapy and radiation therapy. The chemotherapeutic regimens continue to evolve along with type and dose of radiation therapy. Gemcitabine or 5-fluorouracil based chemotherapeutic combinations are administered. The type and dose of radiation vary among different institutions. With neoadjuvant treatment, approximately 50% of the patients are able to undergo surgical resections with negative margins obtained in greater than 80% of the patients. Newer trials are attempting to standardize the definition of borderline resectable pancreatic cancer and treatment regimens. In this review, we outline the definition, imaging requirements and management of patients with borderline resectable pancreatic cancer.
Collapse
|
96
|
Prasad S, Cambridge L, Huguet F, Chou JF, Zhang Z, Wu AJ, O'Reilly EM, Allen PJ, Goodman KA. Intensity modulated radiation therapy reduces gastrointestinal toxicity in locally advanced pancreas cancer. Pract Radiat Oncol 2015; 6:78-85. [PMID: 26577010 DOI: 10.1016/j.prro.2015.09.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/08/2015] [Accepted: 09/18/2015] [Indexed: 11/18/2022]
Abstract
PURPOSE We compared gastrointestinal (GI) and hematologic toxicity in patients with locally advanced pancreas cancer (LAPC) undergoing definitive chemoradiation using intensity modulated radiation therapy (IMRT) or 3-dimensional conformal radiation therapy (3D-CRT) planning. METHODS AND MATERIALS We retrospectively studied 205 patients with LAPC undergoing IMRT (n = 134) and 3D-CRT (n = 71) between May 2003 and March 2012. Patient, tumor, and treatment characteristics and acute GI/hematology toxicity according to the Common Terminology Criteria for Adverse Events, version 3.0, were recorded. Multivariable logistic regression models were used to test association between acute grade 2+ GI and hematologic toxicity outcomes and predictors. Propensity score analysis for grade 2+ GI toxicity was performed to reduce bias for confounding variables: age, gender, radiation dose, field size, and chemotherapy type. RESULTS Median follow-up time for survivors was 22 months and was similar between groups. Median RT dose was significantly higher for IMRT versus 3D-CRT (5600 cGy vs 5040 cGy, P < .001); concurrent chemotherapy was mainly gemcitabine (56%) or 5-fluorouracil (38%). Grade 2+ GI toxicity occurred in 34% (n = 24) of 3D-CRT compared with 16% (n = 21) of IMRT patients. Using propensity score analysis, 3D-CRT had significantly higher grade 2+ GI toxicity (odds ratio, 1.26; 95% confidence interval, 1.08-1.45; P = .001). Grade 2+ hematologic toxicity was similar between IMRT and 3D-CRT groups, but was significantly greater in recipients of concurrent gemcitabine than in 5-fluorouracil (62% vs 29%, P < .0001). CONCLUSIONS IMRT is associated with significant lower grade 2+ GI toxicity versus 3D-CRT for patients undergoing definitive chemoradiation therapy for LAPC. Because IMRT is better tolerated at higher doses and may allow further dose escalation, potentially improving local control for this aggressive disease. Further prospective studies of dose-escalated chemoradiation using IMRT are warranted.
Collapse
Affiliation(s)
- Shreya Prasad
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lajhem Cambridge
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Florence Huguet
- Department of Radiation Oncology, Tenon Hospital, APHP, University Paris VI, Paris, France
| | - Joanne F Chou
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Zhigang Zhang
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Abraham J Wu
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Peter J Allen
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Karyn A Goodman
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
97
|
Wei Q, Yu W, Rosati LM, Herman JM. Advances of stereotactic body radiotherapy in pancreatic cancer. Chin J Cancer Res 2015; 27:349-57. [PMID: 26361404 DOI: 10.3978/j.issn.1000-9604.2015.04.12] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 04/22/2015] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer (PCA) is one of the most aggressive tumors with few effective treatment modalities. It is the 4th and 7th leading cause of cancer death in the United States and China, respectively. At the time of diagnosis, only 20% of cases present with a resectable tumor, and about 40% with a locally advanced tumor that is considered unresectable. Even resected patients still have a poor prognosis, with an incidence of local recurrence ranging from 20% to 60%. It is also reported that up to 30% of PCA patients die from locally obstructive disease with few or no distant metastases. These findings have highlighted the importance of local radiation therapy in the treatment of PCA. As the role of conventional chemoradiotherapy remains controversial, the dawn of the pancreas stereotactic body radiation therapy (SBRT) era represents a potential paradigm shift in the management of PCA. SBRT delivers a higher biological effective dose to the tumor with sharp dose escalation in a shorter treatment time course. Pancreas SBRT is a novel therapeutic option to achieve local tumor control with minimal toxicity. Herein, we review the advancement of SBRT for PCA patients with different stages of pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Qichun Wei
- 1 Department of Radiation Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China ; 2 Department of Radiation Oncology & Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Wei Yu
- 1 Department of Radiation Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China ; 2 Department of Radiation Oncology & Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lauren M Rosati
- 1 Department of Radiation Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China ; 2 Department of Radiation Oncology & Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Joseph M Herman
- 1 Department of Radiation Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China ; 2 Department of Radiation Oncology & Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
98
|
Buwenge M, Cellini F, Silvestris N, Cilla S, Deodato F, Macchia G, Mattiucci GC, Valentini V, Morganti AG. Robotic radiosurgery in pancreatic cancer: A systematic review. World J Gastroenterol 2015; 21:9420-9429. [PMID: 26309369 PMCID: PMC4541395 DOI: 10.3748/wjg.v21.i31.9420] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/21/2014] [Accepted: 04/03/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To present a systematic review of techniques and clinical results.
METHODS: A systematic review of published literature was performed. Only studies reporting patient outcome after radiosurgery (single fraction) delivered with robotic devices [i.e., robotic radiosurgery (RRS)] have been analyzed.
RESULTS: A total of 96 patients from 5 studies were included. The studies are characterized by small series and different methods in terms of dose, target definition, combination with chemotherapy and/or standard fractionated radiotherapy and evaluation modalities. Preliminary results are positive in terms of tumor response (ORR = 56%) and local control of the tumor (crude rate of local progressions: 19.5%). Results for median overall survival (11.4 mo) seem comparable with the ones of prolonged chemoradiation (range: 8.6-13.0 mo). However, gastrointestinal toxicity seems to be the main limitation of RRS, especially at the duodenal level.
CONCLUSION: RRS allows for local treatment in a shortened time (1 fraction) compared to traditional treatments (about 1 mo), providing the possibility for an easy integration with systemic therapies. Preliminary results did not show any outcome differences compared to standard chemoradiation. Thus, further efforts to reduce gastrointestinal toxicity are strongly needed.
Collapse
|
99
|
Boyle J, Czito B, Willett C, Palta M. Adjuvant radiation therapy for pancreatic cancer: a review of the old and the new. J Gastrointest Oncol 2015; 6:436-44. [PMID: 26261730 DOI: 10.3978/j.issn.2078-6891.2015.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/22/2015] [Indexed: 12/23/2022] Open
Abstract
Surgery represents the only potential curative treatment option for patients diagnosed with pancreatic adenocarcinoma. Despite aggressive surgical management for patients deemed to be resectable, rates of local recurrence and/or distant metastases remain high, resulting in poor long-term outcomes. In an effort to reduce recurrence rates and improve survival for patients having undergone resection, adjuvant therapies (ATs) including chemotherapy and chemoradiation therapy (CRT) have been explored. While adjuvant chemotherapy has been shown to consistently improve outcomes, the data regarding adjuvant radiation therapy (RT) is mixed. Although the ability of radiation to improve local control has been demonstrated, it has not always led to improved survival outcomes for patients. Early trials are flawed in their utilization of sub-optimal radiation techniques, limiting their generalizability. Recent and ongoing trials incorporate more optimized RT approaches and seek to clarify its role in treatment strategies. At the same time novel radiation techniques such as intensity modulated RT (IMRT) and stereotactic body RT (SBRT) are under active investigation. It is hoped that these efforts will lead to improved disease-related outcomes while reducing toxicity rates.
Collapse
Affiliation(s)
- John Boyle
- Department of Radiation Oncology, Duke University, Durham, NC 27710, USA
| | - Brian Czito
- Department of Radiation Oncology, Duke University, Durham, NC 27710, USA
| | | | - Manisha Palta
- Department of Radiation Oncology, Duke University, Durham, NC 27710, USA
| |
Collapse
|
100
|
Goji T, Kimura T, Miyamoto H, Takehara M, Kagemoto K, Okada Y, Okazaki J, Takaoka Y, Miyamoto Y, Mitsui Y, Matsumoto S, Sueuchi T, Tanaka K, Fujino Y, Takaoka T, Kitamura S, Okamoto K, Kimura M, Sogabe M, Muguruma N, Okahisa T, Sato Y, Sagawa T, Fujikawa K, Sato Y, Ikushima H, Takayama T. A phase I/II study of fixed-dose-rate gemcitabine and S-1 with concurrent radiotherapy for locally advanced pancreatic cancer. Cancer Chemother Pharmacol 2015. [PMID: 26220846 DOI: 10.1007/s00280-015-2835-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE This study was conducted to identify the maximum-tolerated dose (MTD) of fixed-dose-rate gemcitabine (FDR-gem) administered concurrently with S-1 and radical radiation for locally advanced pancreatic cancer (LAPC) and to provide efficacy and safety data. METHODS Patients with unrespectable pancreatic cancer confined to the pancreatic region were treated with FDR-gem (300-400 mg/m(2), 5 mg/m(2)/min) on days 1, 8, 22, and 29 and 60 mg/m(2) of S-1 orally on days 1-14, 22-35. A total radiation dose of 50.4 Gy (1.8 Gy/day, 28 fractions) was delivered concurrently. RESULTS Twenty-five patients were enrolled; all were evaluable for toxicity assessment. In phase I, eight patients were treated in sequential cohorts of three to five patients per dose level. The MTD was reached at level 2, and dose-limiting toxicities were neutropenia and thrombocytopenia. The recommended doses were 300 mg/m(2) of gemcitabine and 60 mg/m(2) of S-1 daily. The overall response rate was 25% and disease control rate (partial response plus stable disease) was 92%. The progression-free survival was 11.0 months. The median overall survival and 1-year survival rates were 16.0 months and 73%, respectively. CONCLUSION The combination of FDR-gem and S-1 with radiation is a feasible regimen that shows favorable antitumor activity with an acceptable safety profile in patients with LAPC.
Collapse
Affiliation(s)
- Takahiro Goji
- Department of Gastroenterology and Oncology, Institute of Health Biosciences, University of Tokushima Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|