1
|
Gordon AB, Sheeka A, Cleator S, Leff D, Lim A. Tumour volume analysis applied to imaging and histological examinations in breast cancer. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109578. [PMID: 39823861 DOI: 10.1016/j.ejso.2025.109578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 11/12/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025]
Abstract
PURPOSE Response Evaluation Criteria in Solid Tumours (RECIST) determines partial response (PR) and progressive disease (PD) as a 30 % reduction and 20 % increase in the longest diameter (LD), respectively. Tumour volume analysis (TVA) utilises three diameters to calculate response parameters. PATIENTS AND METHODS We conducted a pilot investigation of patients who underwent neoadjuvant breast cancer treatment and evaluation using RECIST with LD measurements and TVA with three diametric measurements, using the parameters PR (>30 % tumour regression), PD (>20 % tumour growth), and intermediate stable disease (SD). According to TVA, RECIST miscategorised 7 of 28 patients (25 %). We evaluated 145 patients who underwent baseline breast magnetic resonance imaging (MRI), neoadjuvant chemotherapy, presurgical MRI, and surgery and calculated LD and volume from all MRI examinations. RESULTS Of the 173 patients, 157 had measurable disease at baseline and treatment completion, and 32 were miscategorised (20.4 %). The number of patients with a PR increased from 123 to 150 after TVA. The sensitivity of RECIST-measured responses (95 % confidence interval: 97-100 %) was 100 % for TVA. This altered the staging, as 32 of 157 (20.4 %) patients were allocated to another response group, with fewer cases of SD: 26 patients moved from SD to PR and 6 patients from SD to PD. CONCLUSION Measuring a solid mass using LD is fundamentally flawed, as the lesser axes considerably affect the volume, leading to inaccurate response categorisation, with implications for patient management. TVA is a novel method that increases accuracy of tumour size measurement and response to therapy.
Collapse
Affiliation(s)
- Angus B Gordon
- Imperial College Healthcare Trust, Fulham Palace Road, London, W6 8RF, England, UK.
| | - Alexander Sheeka
- Imperial College Healthcare Trust, Fulham Palace Road, London, W6 8RF, England, UK.
| | - Suzy Cleator
- Imperial College Healthcare Trust, Fulham Palace Road, London, W6 8RF, England, UK.
| | - Daniel Leff
- Imperial College Healthcare Trust, Fulham Palace Road, London, W6 8RF, England, UK.
| | - Adrian Lim
- Imperial College Healthcare Trust, Fulham Palace Road, London, W6 8RF, England, UK.
| |
Collapse
|
2
|
Harel R, Kaisman-Elbaz T, Emch T, Elson P, Chao ST, Suh JH, Angelov L. A quantitative and comparative evaluation of stereotactic spine radiosurgery local control: proposing a consistent measurement methodology. Neurosurg Focus 2022; 53:E10. [DOI: 10.3171/2022.8.focus22363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022]
Abstract
OBJECTIVE
Stereotactic body radiotherapy (SBRT) is a precise and conformal treatment modality used in the management of metastatic spine tumors. Multiple studies have demonstrated its safety and efficacy for pain and tumor control. However, no uniform quantitative imaging methodology exists to evaluate response to treatment in these patients. This study presents radiographic local control rates post-SBRT, systematically compares measurements acquired according to WHO and Response Evaluation Criteria in Solid Tumors (RECIST) criteria, and explores the relationship to patient outcome.
METHODS
The authors performed a retrospective review of prospectively obtained data from a cohort of 59 consecutive patients (81 metastatic isocenters) treated with SBRT and followed with serial MRI scans. Measurements were performed by a neuroradiologist blinded to the patients’ clinical course. Local control status was determined according to both WHO and RECIST measurements, and agreement between the measuring methodologies was calculated and reported.
RESULTS
Eighty-one isocenters (111 vertebral bodies) were treated with SBRT. The mean treatment dose was 13.96 Gy and the median follow-up duration was 10.8 months, during which 408 MRI scans were evaluated with both WHO and RECIST criteria for each scan point. Imaging demonstrated a mean unidimensional size decrease of 0.2 cm (p = 0.14) and a mean area size decrease of 0.99 cm2 (p = 0.03). Although 88% of the case classifications were concordant and the agreement was significant, WHO criteria were found to be more sensitive to tumor size change. The local control rates according to WHO and RECIST were 95% and 98%, respectively.
CONCLUSIONS
Although WHO volumetric measurements are admittedly superior for tumor size measurement, RECIST is simpler, reproducible, and for the first time is shown here to be comparable to WHO criteria. Thus, the application of RECIST methodology appears to be a suitable standard for evaluating post-SBRT treatment response. Moreover, using comprehensive and consistent measuring approaches, this study substantiates the efficacy of SBRT in the treatment of spine metastases.
Collapse
Affiliation(s)
- Ran Harel
- Department of Neurosurgery, Sheba Medical Center Affiliated to Tel-Aviv University, Tel-Aviv, Israel
| | - Tehila Kaisman-Elbaz
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland
- Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland
| | - Todd Emch
- Imaging Institute, Cleveland Clinic, Cleveland
| | - Paul Elson
- Quantitative Health Sciences, Cleveland Clinic, Cleveland; and
| | - Samuel T Chao
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - John H Suh
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Lilyana Angelov
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland
- Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland
| |
Collapse
|
3
|
Berber T, Sakin A. Role of Consolidative Stereotactic Body Radiation Therapy in Oligoresistant/Oligoprogressive Pulmonary Parenchymal Metastases. Cancer Manag Res 2022; 14:2597-2607. [PMID: 36068821 PMCID: PMC9441141 DOI: 10.2147/cmar.s360766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 07/29/2022] [Indexed: 11/23/2022] Open
Abstract
Aim To extend the survival of patients by providing local control of metastases in oligoresistance/oligoprogressive disease. Methods We retrospectively evaluated the efficacy of stereotactic body radiotherapy (SBRT) applied to 30 lesions in the lungs of 19 patients who were considered inoperable by the tumor board upon the development of oligoresistance/oligoprogressive lung metastasis while undergoing chemotherapy between January 2016 and December 2017. Each patient had one to five metastases in their lungs. The median SBRT biologic effective dose at α/β of 10 (BED10) was 180.0 (IQR: 115.5–180.0) Gy. Results We obtained effective, low-toxicity results. The rates of local control were 89.4%, 84.2%, and 78.9% for the 1st, 2nd, and 3rd years, respectively. The median local control time was 4 (IQR: 3–6) months. The median overall survival (OS) was 36.3 (IQR: 29.7–42.9) months. The rates of OS for the 1st, 2nd, and 3rd years were 89.5%, 73.7%, and 61.4%, respectively. Despite the nonoccurrence of grade 4–5 toxicity in the lungs, six (31.6%) patients had grade 1–3 pulmonary pneumonia, one patient had a grade 4 skin ulceration, and two patients had increased chronic obstructive pulmonary disease in the follow-up period. Discussion In patients with oligometastatic lung tumors, SBRT is very effective in terms of progression-free survival and OS.
Collapse
Affiliation(s)
- Tanju Berber
- Department of Radiation Oncology, Okmeydani Training and Research Hospital, Istanbul, Turkey
- Correspondence: Tanju Berber, Department of Radiation Oncology, Okmeydani Training and Research Hospital, Istanbul, 34307, Turkey, Tel +0905324111202, Email
| | - Abdullah Sakin
- Department of Medical Oncology, Medipol University, Bahçelievler Medipol Hospital, Istanbul, Turkey
| |
Collapse
|
4
|
The Role of Imaging in Soft Tissue Sarcoma Diagnosis and Management. Surg Clin North Am 2022; 102:539-550. [DOI: 10.1016/j.suc.2022.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
5
|
Fehrenbach U, Xin S, Hartenstein A, Auer TA, Dräger F, Froböse K, Jann H, Mogl M, Amthauer H, Geisel D, Denecke T, Wiedenmann B, Penzkofer T. Automatized Hepatic Tumor Volume Analysis of Neuroendocrine Liver Metastases by Gd-EOB MRI-A Deep-Learning Model to Support Multidisciplinary Cancer Conference Decision-Making. Cancers (Basel) 2021; 13:2726. [PMID: 34072865 PMCID: PMC8199286 DOI: 10.3390/cancers13112726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/22/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Rapid quantification of liver metastasis for diagnosis and follow-up is an unmet medical need in patients with secondary liver malignancies. We present a 3D-quantification model of neuroendocrine liver metastases (NELM) using gadoxetic-acid (Gd-EOB)-enhanced MRI as a useful tool for multidisciplinary cancer conferences (MCC). METHODS Manual 3D-segmentations of NELM and livers (149 patients in 278 Gd-EOB MRI scans) were used to train a neural network (U-Net architecture). Clinical usefulness was evaluated in another 33 patients who were discussed in our MCC and received a Gd-EOB MRI both at baseline and follow-up examination (n = 66) over 12 months. Model measurements (NELM volume; hepatic tumor load (HTL)) with corresponding absolute (ΔabsNELM; ΔabsHTL) and relative changes (ΔrelNELM; ΔrelHTL) between baseline and follow-up were compared to MCC decisions (therapy success/failure). RESULTS Internal validation of the model's accuracy showed a high overlap for NELM and livers (Matthew's correlation coefficient (φ): 0.76/0.95, respectively) with higher φ in larger NELM volume (φ = 0.80 vs. 0.71; p = 0.003). External validation confirmed the high accuracy for NELM (φ = 0.86) and livers (φ = 0.96). MCC decisions were significantly differentiated by all response variables (ΔabsNELM; ΔabsHTL; ΔrelNELM; ΔrelHTL) (p < 0.001). ΔrelNELM and ΔrelHTL showed optimal discrimination between therapy success or failure (AUC: 1.000; p < 0.001). CONCLUSION The model shows high accuracy in 3D-quantification of NELM and HTL in Gd-EOB-MRI. The model's measurements correlated well with MCC's evaluation of therapeutic response.
Collapse
Affiliation(s)
- Uli Fehrenbach
- Department of Radiology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany; (A.H.); (T.A.A.); (F.D.); (K.F.); (D.G.); (T.P.)
| | - Siyi Xin
- Division of Gastroenterology, Medical Department, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; (S.X.); (H.J.); (B.W.)
| | - Alexander Hartenstein
- Department of Radiology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany; (A.H.); (T.A.A.); (F.D.); (K.F.); (D.G.); (T.P.)
- Bayer AG, 13353 Berlin, Germany
| | - Timo Alexander Auer
- Department of Radiology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany; (A.H.); (T.A.A.); (F.D.); (K.F.); (D.G.); (T.P.)
- Berlin Institute of Health, 10178 Berlin, Germany
| | - Franziska Dräger
- Department of Radiology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany; (A.H.); (T.A.A.); (F.D.); (K.F.); (D.G.); (T.P.)
| | - Konrad Froböse
- Department of Radiology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany; (A.H.); (T.A.A.); (F.D.); (K.F.); (D.G.); (T.P.)
| | - Henning Jann
- Division of Gastroenterology, Medical Department, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; (S.X.); (H.J.); (B.W.)
| | - Martina Mogl
- Department of Surgery Campus Charité Mitte/Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany;
| | - Holger Amthauer
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany;
| | - Dominik Geisel
- Department of Radiology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany; (A.H.); (T.A.A.); (F.D.); (K.F.); (D.G.); (T.P.)
| | - Timm Denecke
- Department of Diagnostic and Interventional Radiology, University Hospital Leipzig, 04103 Leipzig, Germany;
| | - Bertram Wiedenmann
- Division of Gastroenterology, Medical Department, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; (S.X.); (H.J.); (B.W.)
| | - Tobias Penzkofer
- Department of Radiology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany; (A.H.); (T.A.A.); (F.D.); (K.F.); (D.G.); (T.P.)
- Berlin Institute of Health, 10178 Berlin, Germany
| |
Collapse
|
6
|
Radiological and Clinical Efficacy of Intra-Arterial 90Y-DOTATATE in Patients with Unresectable, Progressive, Liver Dominant Neuroendocrine Neoplasms. J Clin Med 2021; 10:jcm10081794. [PMID: 33924160 PMCID: PMC8074370 DOI: 10.3390/jcm10081794] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/08/2021] [Accepted: 04/16/2021] [Indexed: 01/14/2023] Open
Abstract
This study was performed to determine if intra-arterial (i.a.) administration of 90Y DOTATATE can provide an effective and safe alternative to the accepted standard for i.v. of peptide receptor radionuclide therapy (PRRT) in liver-dominant metastases of gastrointestinal pancreatic neuroendocrine neoplasm (GEP-NEN). A single site, prospective, preliminary case series study included 39 patients with histologically proven liver-dominant NEN. PRRT in the form of 1.15GBq 90Y DOTATATE was given selectively into the liver via radiological catheterization of the hepatic artery, up to four times. The endpoint was radiological response (RECIST). Secondary endpoints assessed clinical well-being post-treatment, progression-free survival (PFS), overall survival (OS), and toxicity. Partial response (PR) was noted in 13% of subjects six weeks post-therapy, increasing to 24% at six months and dropping to 13% at 36 months. Disease progression (DP) was not seen at six weeks, was 5% at six months, and 47% at 36 months. Clinical response based on PS seen in 74% of patients at six weeks, 69% at six months, and 39% at 36 months had PFS and OS, respectively, of 22.7 months and 38.2 months. There was no difference in OS/PFS between those with RECIST PR and SD. One patient had significant toxicity (3%). Use of i.a. PRRT appears to be safe and effective in treating patients with liver-dominant NEN. In addition, the best OS (51 vs. 22 months) was seen when i.a. was used as an upfront treatment of bulky GEP-NEN liver metastases and not after i.v. 90Y DOTATATE. The use of i.a. 90Y DOTATATE PRRT appears to be safe and effective in treating patients with liver-dominant NEN.
Collapse
|
7
|
Yu SH, Choi SJ, Noh H, Lee IS, Park SH, Kim SJ. Comparison of CT Volumetry and RECIST to Predict the Treatment Response and Overall Survival in Gastric Cancer Liver Metastases. JOURNAL OF THE KOREAN SOCIETY OF RADIOLOGY 2021; 82:876-888. [PMID: 36238076 PMCID: PMC9514402 DOI: 10.3348/jksr.2020.0085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/30/2020] [Accepted: 09/08/2020] [Indexed: 11/24/2022]
Abstract
Purpose The aim of this study was to compare the diameter and volume of liver metastases on CT images in relation to overall survival and tumor response in patients with gastric cancer liver metastases (GCLM) treated with chemotherapy. Materials and Methods We recruited 43 patients with GCLM who underwent chemotherapy as a first-line treatment. We performed a three-dimensional quantification of the metastases for each patient. An independent survival analysis using the Response Evaluation Criteria in Solid Tumors (RECIST) was performed and compared to volumetric measurements. Overall survival was evaluated using Kaplan-Meier analysis and compared using Cox proportional hazard ratios following univariate analyses. Results When patients were classified as responders or non-responders based on volumetric criteria, the median overall survival was 23.6 months [95% confidence interval (CI), 8.63–38.57] and 7.6 months (95% CI, 3.78–11.42), respectively (p = 0.039). The volumetric analysis and RECIST of the non-progressing and progressing groups showed similar results based on the Kaplan-Meier method (p = 0.006) and the Cox proportional hazard model (p = 0.008). Conclusion Volumetric assessment of liver metastases could be an alternative predictor of overall survival for patients with GCLM treated with chemotherapy.
Collapse
Affiliation(s)
- Sung Hyun Yu
- Department of Radiology, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Seung Joon Choi
- Department of Radiology, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - HeeYeon Noh
- Department of Radiology, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - In seon Lee
- Department of Radiology, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - So Hyun Park
- Department of Radiology, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Se Jong Kim
- Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| |
Collapse
|
8
|
Ross BD, Chenevert TL, Meyer CR. Retrospective Registration in Molecular Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00080-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
9
|
Wang S, Hossack JA, Klibanov AL. From Anatomy to Functional and Molecular Biomarker Imaging and Therapy: Ultrasound Is Safe, Ultrafast, Portable, and Inexpensive. Invest Radiol 2020; 55:559-572. [PMID: 32776766 PMCID: PMC10290890 DOI: 10.1097/rli.0000000000000675] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ultrasound is the most widely used medical imaging modality worldwide. It is abundant, extremely safe, portable, and inexpensive. In this review, we consider some of the current development trends for ultrasound imaging, which build upon its current strength and the popularity it experiences among medical imaging professional users.Ultrasound has rapidly expanded beyond traditional radiology departments and cardiology practices. Computing power and data processing capabilities of commonly available electronics put ultrasound systems in a lab coat pocket or on a user's mobile phone. Taking advantage of new contributions and discoveries in ultrasound physics, signal processing algorithms, and electronics, the performance of ultrasound systems and transducers have progressed in terms of them becoming smaller, with higher imaging performance, and having lower cost. Ultrasound operates in real time, now at ultrafast speeds; kilohertz frame rates are already achieved by many systems.Ultrasound has progressed beyond anatomical imaging and monitoring blood flow in large vessels. With clinical approval of ultrasound contrast agents (gas-filled microbubbles) that are administered in the bloodstream, tissue perfusion studies are now routine. Through the use of modern ultrasound pulse sequences, individual microbubbles, with subpicogram mass, can be detected and observed in real time, many centimeters deep in the body. Ultrasound imaging has broken the wavelength barrier; by tracking positions of microbubbles within the vasculature, superresolution imaging has been made possible. Ultrasound can now trace the smallest vessels and capillaries, and obtain blood velocity data in those vessels.Molecular ultrasound imaging has now moved closer to clinic; the use of microbubbles with a specific affinity to endothelial biomarkers allows selective accumulation and retention of ultrasound contrast in the areas of ischemic injury, inflammation, or neoangiogenesis. This will aid in noninvasive molecular imaging and may provide additional help with real-time guidance of biopsy, surgery, and ablation procedures.The ultrasound field can be tightly focused inside the body, many centimeters deep, with millimeter precision, and ablate lesions by energy deposition, with thermal or mechanical bioeffects. Some of such treatments are already in clinical use, with more indications progressing through the clinical trial stage. In conjunction with intravascular microbubbles, focused ultrasound can be used for tissue-specific drug delivery; localized triggered release of sequestered drugs from particles in the bloodstream may take time to get to clinic. A combination of intravascular microbubbles with circulating drug and low-power ultrasound allows transient opening of vascular endothelial barriers, including blood-brain barrier; this approach has reached clinical trial stage. Therefore, the drugs that normally would not be getting to the target tissue in the brain will now have an opportunity to produce therapeutic efficacy.Overall, medical ultrasound is developing at a brisk rate, even in an environment where other imaging modalities are also advancing rapidly and may be considered more lucrative. With all the current advances that we discuss, and many more to come, ultrasound may help solve many problems that modern medicine is facing.
Collapse
|
10
|
Sulieman A, Mayhoub FH, Salah H, Al-Mohammed HI, Alkhorayef M, Moftah B, Al Rowaily M, Bradley DA. Occupational and ambient radiation exposures from Lu-177 DOTATATE during targeted therapy. Appl Radiat Isot 2020; 164:109240. [PMID: 32819499 DOI: 10.1016/j.apradiso.2020.109240] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/22/2020] [Accepted: 05/19/2020] [Indexed: 11/16/2022]
Abstract
Lutetium-177 (DOTATATE) (177Lu; T1/2 6.7 days), a labelled β- and Auger-electron emitter, is widely used in treatment of neuroendocrine tumours. During performance of the procedure, staff and other patients can potentially receive significant doses in interception of the gamma emissions [113 keV (6.4%) and 208 keV (11%)] that are associated with the particle decays. While radiation protection and safety assessment are required in seeking to ensure practices comply with international guidelines, only limited published studies are available. The objectives of present study are to evaluate patient and occupational exposures, measuring ambient doses and estimating the radiation risk. The results, obtained from studies carried out in Riyadh over an 11 month period, at King Faisal Specialist Hospital and Research Center, concerned a total of 33 177Lu therapy patients. Patient exposures were estimated using a calibrated Victoreen 451P survey meter (Fluke Biomedical), for separations of 30 cm, 100 cm and 300 cm, also behind a bed shield that was used during hospitalization of the therapy patients. Occupational and ambient doses were also measured through use of calibrated thermoluminescent dosimeters and an automatic TLD reader (Harshaw 6600). The mean and range of administered activity (in MBq)) was 7115.2 ± 917.2 (4329-7955). The ambient dose at corridors outside of therapy isolation rooms was 1.2 mSv over the 11 month period, that at the nursing station was below the limit of detection and annual occupational doses were below the annual dose limit of 20 mSv. Special concern needs to be paid to comforters (carers) and family members during the early stage of radioisotope administration.
Collapse
Affiliation(s)
- A Sulieman
- Prince Sattam Bin Abdulaziz University, College of Applied Medical Sciences, Radiology and Medical Imaging Department, P.O.Box 422, Alkharj, 11942, Saudi Arabia.
| | - Fareed H Mayhoub
- King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Hassan Salah
- INAYA Medical Collage, Nuclear Medicine Department, Riyadh, 13541, Saudi Arabia; College of Medical Radiologic Science, Sudan University of Science and Technology, P.O.Box 1908, Khartoum, 11111, Sudan
| | - H I Al-Mohammed
- Department of Radiological Sciences, College of Health and Rehabilitation Sciences, Princess Nourah Bint Abdulrahman University, P.O Box 84428, Riyadh, 11671, Saudi Arabia
| | - M Alkhorayef
- Department of Radiological Sciences, College of Applied Medical Sciences, King Saud University, P.O Box 10219, Riyadh, 11433, Saudi Arabia; Centre for Nuclear and Radiation Physics, Department of Physics, University of Surrey, Guildford, Surrey GU2 7XH, UK
| | - B Moftah
- King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - M Al Rowaily
- King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - D A Bradley
- Centre for Nuclear and Radiation Physics, Department of Physics, University of Surrey, Guildford, Surrey GU2 7XH, UK; Centre for Biomedical Physics, School of Healthcare and Medical Sciences, Sunway University, 47500, Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
11
|
Haveman LM, Ranft A, Vd Berg H, Smets A, Kruseova J, Ladenstein R, Brichard B, Paulussen M, Kuehne T, Juergens H, Klco-Brosius S, Dirksen U, Merks JHM. The relation of radiological tumor volume response to histological response and outcome in patients with localized Ewing Sarcoma. Cancer Med 2019; 8:1086-1094. [PMID: 30790456 PMCID: PMC6434194 DOI: 10.1002/cam4.2002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 01/08/2019] [Accepted: 01/10/2019] [Indexed: 12/19/2022] Open
Abstract
Background Magnetic resonance imaging (MRI) is the modality of choice for local staging and response evaluation of Ewing sarcoma (EwS). Aim of this study was to determine the relevance of tumor volume response (TVR) in relation to histological response (HisRes) and survival, in order to evaluate if early modification of chemotherapy might be indicated in patients with inadequate TVR. Methods Three dimensional (3D)‐tumor volume data at diagnosis, during early induction phase (1‐3 courses of chemotherapy; n = 195) and/or late induction phase (4‐6 courses; n = 175) from 241 localized patients were retrospectively analyzed. A distinction was made between adequate response (reduction ≥67%) and inadequate response (reduction <67% or progression). Correlations between TVR, HisRes, event free survival (EFS), and overall survival (OS) were analyzed using chi‐square tests, log‐rank tests, and the Cox‐regression model. Results Early adequate TVR, noted in 41% of patients, did not correlate with EFS (P = 0.92) or OS (P = 0.38). During late induction phase 62% of patients showed an adequate TVR. EFS for patients with late adequate TVR was better (78%) than for those with inadequate late TVR (61%) (P = 0.01); OS was 80% and 69% (P = 0.26), respectively. No correlation was found between TVR and HisRes. Multivariate analysis showed that poor HisRes, pelvic location and late inadequate TVR were associated with poor outcome. Conclusions Early inadequate TVR does not predict adverse outcome; therefore, changing the treatment to second line chemotherapy is not indicated in case of inadequate early TVR. Late adequate TVR and good HisRes correlate with better EFS; patients with late inadequate TVR might benefit from augmented therapy.
Collapse
Affiliation(s)
- Lianne M Haveman
- Emma Children's Hospital, Department of Pediatric Oncology, Academic Medical Center, Amsterdam, The Netherlands.,Prinses Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Andreas Ranft
- Department of Pediatric Hematology and Oncology, University of Essen, Essen, Germany.,Coordinating Center for Clinical Trials, Muenster, Germany
| | - Henk Vd Berg
- Emma Children's Hospital, Department of Pediatric Oncology, Academic Medical Center, Amsterdam, The Netherlands
| | - Anne Smets
- Department of Radiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Jarmila Kruseova
- Department of Pediatric Oncology, University Hospital Motol, Prague, Czech Republic
| | | | - Benedicte Brichard
- Saint Luc University Hospital, Department of Pediatric Hematology and Oncology, University of Louvain, Datteln, Belgium
| | - Michael Paulussen
- Witten/Herdecke University, Vestische Kinder- und Jugendklinik, Datteln, Germany
| | - Thomas Kuehne
- Department of Pediatric Oncology and Haematology, University Children Hospital, Basel, Switzerland
| | - Heribert Juergens
- Coordinating Center for Clinical Trials, Muenster, Germany.,Department of Pediatric Hematology and Oncology, University Children's Hospital, Muenster, Germany
| | - Stephanie Klco-Brosius
- Department of Pediatric Hematology and Oncology, University of Essen, Essen, Germany.,Coordinating Center for Clinical Trials, Muenster, Germany
| | - Uta Dirksen
- Department of Pediatric Hematology and Oncology, University of Essen, Essen, Germany.,Coordinating Center for Clinical Trials, Muenster, Germany
| | - Johannes H M Merks
- Emma Children's Hospital, Department of Pediatric Oncology, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Kurhanewicz J, Vigneron DB, Ardenkjaer-Larsen JH, Bankson JA, Brindle K, Cunningham CH, Gallagher FA, Keshari KR, Kjaer A, Laustsen C, Mankoff DA, Merritt ME, Nelson SJ, Pauly JM, Lee P, Ronen S, Tyler DJ, Rajan SS, Spielman DM, Wald L, Zhang X, Malloy CR, Rizi R. Hyperpolarized 13C MRI: Path to Clinical Translation in Oncology. Neoplasia 2019; 21:1-16. [PMID: 30472500 PMCID: PMC6260457 DOI: 10.1016/j.neo.2018.09.006] [Citation(s) in RCA: 314] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/27/2018] [Accepted: 09/27/2018] [Indexed: 12/22/2022]
Abstract
This white paper discusses prospects for advancing hyperpolarization technology to better understand cancer metabolism, identify current obstacles to HP (hyperpolarized) 13C magnetic resonance imaging's (MRI's) widespread clinical use, and provide recommendations for overcoming them. Since the publication of the first NIH white paper on hyperpolarized 13C MRI in 2011, preclinical studies involving [1-13C]pyruvate as well a number of other 13C labeled metabolic substrates have demonstrated this technology's capacity to provide unique metabolic information. A dose-ranging study of HP [1-13C]pyruvate in patients with prostate cancer established safety and feasibility of this technique. Additional studies are ongoing in prostate, brain, breast, liver, cervical, and ovarian cancer. Technology for generating and delivering hyperpolarized agents has evolved, and new MR data acquisition sequences and improved MRI hardware have been developed. It will be important to continue investigation and development of existing and new probes in animal models. Improved polarization technology, efficient radiofrequency coils, and reliable pulse sequences are all important objectives to enable exploration of the technology in healthy control subjects and patient populations. It will be critical to determine how HP 13C MRI might fill existing needs in current clinical research and practice, and complement existing metabolic imaging modalities. Financial sponsorship and integration of academia, industry, and government efforts will be important factors in translating the technology for clinical research in oncology. This white paper is intended to provide recommendations with this goal in mind.
Collapse
Affiliation(s)
- John Kurhanewicz
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
| | - Daniel B Vigneron
- Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, CA, USA
| | | | - James A Bankson
- Department of Imaging Physics, MD Anderson Medical Center, Houston, TX, USA
| | - Kevin Brindle
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | | | - Kayvan R Keshari
- Department of Radiology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Denmark
| | | | - David A Mankoff
- Department of Radiology, University of Pennsylvania, PA, USA
| | - Matthew E Merritt
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA
| | - Sarah J Nelson
- Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, CA, USA
| | - John M Pauly
- Department of Electric Engineering, Stanford University, USA
| | - Philips Lee
- Functional Metabolism Group, Singapore Biomedical Consortium, Agency for Science, Technology and Research, Singapore
| | - Sabrina Ronen
- Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, CA, USA
| | - Damian J Tyler
- Department of Biomedical Science, University of Oxford, Oxford, UK
| | - Sunder S Rajan
- Center for Devices and Radiological Health (CDRH), FDA, White Oak, MD, USA
| | - Daniel M Spielman
- Departments of Radiology and Electric Engineering, Stanford University, USA
| | - Lawrence Wald
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Xiaoliang Zhang
- Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, CA, USA
| | - Craig R Malloy
- Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rahim Rizi
- Department of Radiology, University of Pennsylvania, PA, USA
| |
Collapse
|
13
|
Clinical Response Rates From Interleukin-2 Therapy for Metastatic Melanoma Over 30 Years' Experience: A Meta-Analysis of 3312 Patients. J Immunother 2018; 40:21-30. [PMID: 27875387 DOI: 10.1097/cji.0000000000000149] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Interleukin-2 (IL-2), initially used in 1986, can induce clinical regression-complete responses (CR) and partial responses (PR) of metastatic malignant melanoma. IL-2 has been used alone or in combination, and in different dosage schedules, as an immunotherapeutic agent for melanoma treatment. This meta-analysis aimed to document and evaluate the spectrum of reported clinical response rates from the combined experience of almost 30 years of IL-2 clinical usage. Clinical trials using IL-2 for metastatic melanoma therapy that reported: dosage, combinations, study details, definitions and clinical CR, PR, and overall response (OR) rates were included. A meta-analysis was conducted using the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines. In total, 34 studies met inclusion criteria, with 41 separate treatment arms. For all IL-2 treatment modalities collectively, the CR rate was 4.0% [95% confidence interval (CI), 2.8-5.3], PR 12.5% (95% CI, 10.1-15.0), and OR 19.7% (95% CI, 15.9-23.5). CR pre-1994 was 2.7% versus 6.1% post-1994. High and intermediate-IL-2 dosage showed no CR difference, while low-dose IL-2 showed a nonstatistical trend toward an increased CR rate. The highest CR rate resulted from IL-2 combined with vaccine at 5.0%. The meta-analysis showed that IL-2 immunotherapy for advanced metastatic melanoma delivered a CR rate of 4% (range, 0-23%) across nearly 30 years of clinical studies, with gradual improvement over time. The significance is that, contrary to popular belief, the data demonstrated that CR rates were similar for intermediate versus high-IL-2 dosing.
Collapse
|
14
|
Rogowski W, Wachuła E, Lewczuk A, Kolasińska-Ćwikła A, Iżycka-Świeszewska E, Sulżyc-Bielicka V, Ćwikła JB. Baseline chromogranin A and its dynamics are prognostic markers in gastroenteropancreatic neuroendocrine tumors. Future Oncol 2017; 13:1069-1079. [DOI: 10.2217/fon-2016-0455] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: This study assessed whether absolute chromogranin A (CgA) values at various stages of treatment have prognostic value in patients with pancreatic and midgut neuroendocrine tumors, subjected to peptide receptor radionuclide therapy with 90Y-[DOTA0, D-Phe1, Tyr3]-octreotate. Patients & methods: CgA was determined before peptide receptor radionuclide therapy, 6 weeks, 6, 12, 18 and 24 months after the last dose of 90Y-[DOTA0, D-Phe1, Tyr3]-octreotate. The primary end point was overall survival. Results: Elevated baseline CgA concentrations and their relative increase within the first year of observation were unfavorable predictors of overall survival, but not progression. Conclusion: Even a single baseline measurement of CgA can be useful in establishing prognosis in this group, if this parameter exceeds its upper normal limit more than tenfold.
Collapse
Affiliation(s)
- Wojciech Rogowski
- Clinical Department of Chemotherapy, Hospital Ministry of the Interior & Administration & Warmia & Mazury Oncology Centre, Olsztyn, Poland
- Department of Medical Science, University of Varmia & Masuria, Olsztyn, Poland
| | - Ewa Wachuła
- Clinical Department of Chemotherapy, Hospital Ministry of the Interior & Administration & Warmia & Mazury Oncology Centre, Olsztyn, Poland
| | - Anna Lewczuk
- Department of Endocrinology, Medical University of Gdańsk, Gdańsk, Poland
| | - Agnieszka Kolasińska-Ćwikła
- Department of Clinical Oncology, Maria-Skłodowska-Curie Memorial Cancer Center & Institute of Oncology, Warsaw, Poland
| | | | | | - Jarosław B Ćwikła
- Department of Medical Science, University of Varmia & Masuria, Olsztyn, Poland
| |
Collapse
|
15
|
Lagmay JP, Krailo MD, Dang H, Kim A, Hawkins DS, Beaty O, Widemann BC, Zwerdling T, Bomgaars L, Langevin AM, Grier HE, Weigel B, Blaney SM, Gorlick R, Janeway KA. Outcome of Patients With Recurrent Osteosarcoma Enrolled in Seven Phase II Trials Through Children's Cancer Group, Pediatric Oncology Group, and Children's Oncology Group: Learning From the Past to Move Forward. J Clin Oncol 2016; 34:3031-8. [PMID: 27400942 DOI: 10.1200/jco.2015.65.5381] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
PURPOSE The use of radiographic response as the primary end point in phase II osteosarcoma trials may limit optimal detection of treatment response because of the calcified tumor matrix. We performed this study to determine if time to progression could be used as an end point for subsequent studies. PATIENTS AND METHODS We performed a retrospective analysis of outcome for patients with recurrent/refractory osteosarcoma enrolled in one of seven phase II trials conducted by the Children's Oncology Group and predecessor groups from 1997 to 2007. All trials used RECIST or WHO radiographic response criteria and the primary end point of response rate. The following potential prognostic factors-age, trial, number of prior chemotherapy regimens, sex, and race/ethnicity-were evaluated for their impact on event-free survival (EFS). We used data from a phase II study (AOST0221) of patients with osteosarcoma who were given inhaled granulocyte-macrophage colony-stimulating factor with first pulmonary recurrence who had an EFS as well as biologic end point to determine the historical disease control rate for patients with fully resected disease. RESULTS In each included trial, the drugs tested were determined to be inactive on the basis of radiographic response rates. The EFS for 96 patients with osteosarcoma and measurable disease was 12% at 4 months (95% CI, 6% to 19%). There was no significant difference in EFS across trials according to number of prior treatment regimens or patient age, sex, and ethnicity. The 12-month EFS for the 42 evaluable patients enrolled in AOST0221 was 20% (95% CI, 10% to 34%). CONCLUSION The EFS was uniformly poor for children with recurrent/refractory osteosarcoma in these single-arm phase II trials. We have now constructed baseline EFS outcomes that can be used as a comparison for future phase II trials for recurrent osteosarcoma.
Collapse
Affiliation(s)
- Joanne P Lagmay
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY.
| | - Mark D Krailo
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Ha Dang
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - AeRang Kim
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Douglas S Hawkins
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Orren Beaty
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Brigitte C Widemann
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Theodore Zwerdling
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Lisa Bomgaars
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Anne-Marie Langevin
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Holcombe E Grier
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Brenda Weigel
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Susan M Blaney
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Richard Gorlick
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Katherine A Janeway
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| |
Collapse
|
16
|
Rogowski W, Wachuła E, Lewczuk A, Buscombe JR, Seklecka N, Sankowski A, Ćwikła JB. Long-term efficacy of (90)Y-DOTATATE in patients with nonresectable pancreatic and small bowel neuroendocrine neoplasms. Future Oncol 2016; 12:1877-85. [PMID: 27156864 DOI: 10.2217/fon-2016-0031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
AIM To determine the efficacy of (90)Y [DOTA(0), D-Phe(1), Tyr(3)]-octreotate (DOTATATE) in 67 patients with pancreatic and small bowel neuroendocrine tumors (NETs). PATIENTS & METHODS The primary efficacy end point was overall survival (OS) and secondary end points were progression-free survival (PFS) and tumor response. RESULTS Median PFS in pancreatic and small bowel NETs was 25 and 28 months, respectively, and median OS was 42 and 38.5 months, respectively. No intergroup differences in median OS (p = 0.945) or PFS (p = 0.174) were found, also after adjustment for tumor origin, secretory status and grade, and patient's gender. CONCLUSION (90)Y-DOTATATE may have similar efficacy in pancreatic and small bowel NETs. Better WHO performance status at baseline seems to be associated with more favorable outcomes.
Collapse
Affiliation(s)
- Wojciech Rogowski
- Clinical Department of Chemotherapy, Hospital Ministry of the Interior & Administration & Warmia & Mazury Oncology Centre, Olsztyn, Poland
| | - Ewa Wachuła
- Clinical Department of Chemotherapy, Hospital Ministry of the Interior & Administration & Warmia & Mazury Oncology Centre, Olsztyn, Poland
| | - Anna Lewczuk
- Department of Endocrinology, Medical University of Gdansk, Gdansk, Poland
| | - John R Buscombe
- Department of Nuclear Medicine & PET, Addenbrooke's Hospital, Cambridge, UK
| | - Nina Seklecka
- Department of Radiology & Diagnostic Imaging, Central Clinical Hospital of the Ministry of Interior, Warsaw, Poland
| | - Artur Sankowski
- Department of Radiology & Diagnostic Imaging, Central Clinical Hospital of the Ministry of Interior, Warsaw, Poland
| | - Jarosław B Ćwikła
- Faculty of Medical Science, University of Varmia & Masuria, Olsztyn, Poland
| |
Collapse
|
17
|
Wu XY, Huang XE. Clinical Application of Serum Tumor Abnormal Protein (TAP) in Colorectal Cancer Patients. Asian Pac J Cancer Prev 2016; 16:3425-8. [PMID: 25921156 DOI: 10.7314/apjcp.2015.16.8.3425] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE To explore the association of serum tumor abnormal protein (TAP) with other serological biomarkers e.g. carcinoembryonic antigen (CEA), carbohydrate antigen 125 (CA125), carbohydrate antigen 19-9 (CA19-9) and its clinical application in colorectal cancer (CRC) patients. METHODS Patients (N=98) were enrolled into this study with histologically or cytologically confirmed CRC. Using a test kit, the level of TAP was determined, while chemiluminescence was used to measure the levels of some other common serological biomarkers e.g. CEA, CA125 and CA19-9. RESULTS The area of TAP condensed particulate matter decreased after chemotherapy compared with before chemotherapy when CT or MRI scans showed disease control. In contrast, it increased with disease progression (P<0.05). Furthermore, a statistically significant difference was confirmed in monitoring of TAP and common serological biomarkers e.g. CEA and CA19-9 (p<0.05). CONCLUSIONS Detecting TAP in CRC patients has high sensitivity and specificity and can be used as a new independent indicator for clinically monitoring CRC patients in the course of chemotherapy.
Collapse
Affiliation(s)
- Xue-Yan Wu
- Department of Chemotherapy, the Affiliated Jiangsu Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, China E-mail :
| | | |
Collapse
|
18
|
Quantitative Evaluation of Local Control and Wound Healing Following Surgery and Stereotactic Spine Radiosurgery for Spine Tumors. World Neurosurg 2015; 87:48-54. [PMID: 26548834 DOI: 10.1016/j.wneu.2015.10.075] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/21/2015] [Accepted: 10/22/2015] [Indexed: 11/21/2022]
Abstract
OBJECTIVE The present study evaluated the optimal measuring criteria to assess spinal tumor response to surgery followed by stereotactic spine radiosurgery (SRS) and reports the local control and wound complication rates following combined multimodality treatment. METHODS AND MATERIALS Prospectively collected patient information was retrospectively reviewed to identify patients treated with spine surgery followed by SRS. Tumor sizes and volumetric assessment were formally measured. Local control status was defined according to World Health Organization (WHO, bidimensional), RECIST (unidimensional), or volumetric size change. Statistical comparative assessments of tumor measurements were performed. RESULTS Twenty-two patients were eligible for evaluation after having undergone surgery followed by single-fraction SRS within a 2-month period. Seventeen had follow-up magnetic resonance imaging (MRI) with a mean patient follow-up of 12.59 months (range 3-36 months). None developed wound complication after radiation therapy (95% lower confidence bound 13%). Two patients had clinical recurrence while 15 of 17 achieved local control (88.3%). A test of marginal homogeneity for RECIST versus WHO was not statistically significant, P = 1.0 suggesting similar response classifications with both systems. Spearman correlations among 1) volumetric assessment, 2) bidimensional size, and 3) unidimensional size were significant for all groups (P < 0.05). CONCLUSION High local control rates can be achieved with surgery followed by SRS. Further, adjuvant SRS following spine tumor surgery delivers less radiation to the wound than conventional radiation and thus potentially reduces wound complications. Unidimensional, bidimensional, and volumetric tumor assessments demonstrate similar results. Hence the use of the simpler RECIST criteria is suitable and appropriate for evaluating the response to treatment after spine radiosurgery.
Collapse
|
19
|
Akshintala S, Marcus L, Warren KE, Murphy RF, Sissung TM, Srivastava A, Goodspeed WJ, Goodwin A, Brewer CC, Zalewski C, King KA, Kim A, Figg WD, Widemann BC. Phase 1 trial and pharmacokinetic study of the oral platinum analog satraplatin in children and young adults with refractory solid tumors including brain tumors. Pediatr Blood Cancer 2015; 62:603-10. [PMID: 25556988 PMCID: PMC4339515 DOI: 10.1002/pbc.25344] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 10/08/2014] [Indexed: 11/11/2022]
Abstract
BACKGROUND Based on pre-clinical and clinical activity in adult refractory tumors, and absence of significant neuro-, nephro-, or oto-toxicity, we conducted a pediatric phase 1 trial to determine the toxicities, maximum tolerated dose (MTD), and pharmacokinetics of satraplatin, an oral platinum analogue, in children and young adults with refractory solid tumors. PROCEDURE Satraplatin was administered orally once daily on days 1-5 of a 28-day cycle at dose level (DL) 1 (60 mg/m(2) /dose), and DL2 (80 mg/m(2) /dose). Toxicities, responses, satraplatin pharmacokinetics, and pharmacogenomic expression of specific DNA repair genes were evaluated. RESULTS Nine patients received 1-15 cycles (median = 2). The MTD was exceeded at DL2 with delayed prolonged myelosuppression as dose-limiting toxicity (DLT) in 2/4 patients. At DL1, 0/5 patients had DLTs. Common non-DLTs included myelosuppression, gastrointestinal toxicities, fatigue, headache, liver enzyme elevation, and electrolyte abnormalities. No significant neuro-, nephro-, or oto-toxicity was observed. No objective responses were observed but 2 patients experienced prolonged disease stabilization (---6-15 cycles). Satraplatin exposure (day 1 plasma ultrafiltrate area under the curve) was similar at DL1 and DL2. A strong correlation between estimated creatinine clearance and satraplatin pharmacokinetic parameters (clearance, area under the curve, and peak concentration) was observed. CONCLUSIONS The MTD of oral satraplatin in children with solid tumors was 60 mg/m(2) /dose daily ×5 days every 28 days, which is lower than the adult recommended dose of 80-120 mg/m(2) /dose. The toxicity profile was similar to adults and delayed myelosuppression was the DLT. No significant neuro-, nephro- or oto-toxicities were observed.
Collapse
Affiliation(s)
- Srivandana Akshintala
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| | - Leigh Marcus
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD,Center for Cancer and Blood Disorders, Children’s National Medical Center, Washington DC
| | - Katherine E. Warren
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| | - Robert F. Murphy
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| | - Tristan M. Sissung
- Clinical Pharmacology Program, Office of the Clinical Director, CCR, NCI, NIH, Bethesda, MD
| | - Anjali Srivastava
- Clinical Pharmacology Program, Office of the Clinical Director, CCR, NCI, NIH, Bethesda, MD
| | - Wendy J. Goodspeed
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| | - Anne Goodwin
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| | - Carmen C. Brewer
- National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD
| | - Christopher Zalewski
- National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD
| | - Kelly A. King
- National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD
| | - AeRang Kim
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD,Center for Cancer and Blood Disorders, Children’s National Medical Center, Washington DC
| | - William D. Figg
- Clinical Pharmacology Program, Office of the Clinical Director, CCR, NCI, NIH, Bethesda, MD
| | - Brigitte C. Widemann
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| |
Collapse
|
20
|
Demlova R, Melicharkova K, Rehak Z, Kren L, Oslejskova H, Sterba J. Successful use of metronomic vinblastine and fluorothymidine pet imaging for the management of intramedullary spinal cord anaplastic oligoastrocytoma in a child. ACTA ACUST UNITED AC 2014; 21:e790-3. [PMID: 25489269 DOI: 10.3747/co.21.2147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Children with high-grade glioma still have a poor prognosis despite the use of multimodal therapy including surgery, radiotherapy, and chemotherapy. New therapeutic strategies and methods evaluating such therapies are needed. OBSERVATION Here we describe a child with anaplastic oligodendroglioma of the spinal cord who was unable to tolerate standard chemoradiotherapy and who had still-vital residual tumour during therapy. A good response was obtained with low-dose metronomic treatment containing vinblastine. The treatment was guided according to gradual response assessed using various positron-emission tomography tracers. CONCLUSIONS Metronomic treatment guided by positron-emission tomography could be a reasonable option in some high-risk pediatric tumours.
Collapse
Affiliation(s)
- R Demlova
- Department of Pharmacology, Masaryk University School of Medicine, Brno, Czech Republic. ; Regional Center for Applied Molecular Oncology, Brno, Czech Republic
| | - K Melicharkova
- Department of Pediatric Oncology, University Hospital Brno, Brno, Czech Republic
| | - Z Rehak
- Department of Nuclear Medicine, Masaryk Memorial Cancer Institute, Brno, Czech Republic. ; Regional Center for Applied Molecular Oncology, Brno, Czech Republic
| | - L Kren
- Department of Pathology, University Hospital Brno, Brno, Czech Republic
| | - H Oslejskova
- Department of Pediatric Neurology, University Hospital Brno, Brno, Czech Republic
| | - J Sterba
- Regional Center for Applied Molecular Oncology, Brno, Czech Republic. ; Department of Pediatric Oncology, University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
21
|
Contemporary phase III clinical trial endpoints in advanced ovarian cancer: assessing the pros and cons of objective response rate, progression-free survival, and overall survival. Gynecol Oncol 2014; 136:121-9. [PMID: 25455732 DOI: 10.1016/j.ygyno.2014.10.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 10/06/2014] [Accepted: 10/13/2014] [Indexed: 11/23/2022]
Abstract
Among gynecologic cancers, ovarian cancer provides the greatest challenge because 75% to 80% of patients present with stage III/IV disease. Over the last 40 years, a series of large trials conducted by the Gynecologic Oncology Group and other cooperative groups has produced striking improvements in patient outcome; but the majority still dies of their disease. Further research in both the laboratory and the clinic is essential to continued improvement in patient management. Clinical trials, however, have become a major challenge because of issues with trial endpoints. Historically, overall survival (OS) has been regarded as the "gold standard" of endpoints. Lack of effective treatment for patients who progressed on or recurred after front-line therapy allowed trials to avoid obfuscation of OS by post-progression therapy. More recently, studies have identified over 20 agents active against ovarian cancer. Reasonable evidence shows that effective post-progression therapy with multiple lines of active agents can render the survival endpoint uninterpretable. Two other endpoints avoid this problem. The objective response rate, assessed by the Response Evaluation Criteria in Solid Tumors (RECIST), is an accepted endpoint for accelerated approval in ovarian cancer. More importantly, progression-free survival (PFS), measured from study entry to progression of disease, avoids post-progression therapy completely. Without effective post-progression therapy (prior to 1990), data show that PFS is a surrogate for OS. Recent experience with 4 large trials of bevacizumab shows that PFS can be accurately assessed if progression is clearly defined and if timing of assessments is consistent in all study arms. Acceptance of PFS as the optimal endpoint for ovarian cancer trials by investigators and regulatory agencies is crucial to further advances in management because effective post-progression therapy has rendered differences in OS virtually impossible to assess reliably.
Collapse
|
22
|
Emblem KE, Farrar CT, Gerstner ER, Batchelor TT, Borra RJH, Rosen BR, Sorensen AG, Jain RK. Vessel caliber--a potential MRI biomarker of tumour response in clinical trials. Nat Rev Clin Oncol 2014; 11:566-84. [PMID: 25113840 PMCID: PMC4445139 DOI: 10.1038/nrclinonc.2014.126] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Our understanding of the importance of blood vessels and angiogenesis in cancer has increased considerably over the past decades, and the assessment of tumour vessel calibre and structure has become increasingly important for in vivo monitoring of therapeutic response. The preferred method for in vivo imaging of most solid cancers is MRI, and the concept of vessel-calibre MRI has evolved since its initial inception in the early 1990s. Almost a quarter of a century later, unlike traditional contrast-enhanced MRI techniques, vessel-calibre MRI remains widely inaccessible to the general clinical community. The narrow availability of the technique is, in part, attributable to limited awareness and a lack of imaging standardization. Thus, the role of vessel-calibre MRI in early phase clinical trials remains to be determined. By contrast, regulatory approvals of antiangiogenic agents that are not directly cytotoxic have created an urgent need for clinical trials incorporating advanced imaging analyses, going beyond traditional assessments of tumour volume. To this end, we review the field of vessel-calibre MRI and summarize the emerging evidence supporting the use of this technique to monitor response to anticancer therapy. We also discuss the potential use of this biomarker assessment in clinical imaging trials and highlight relevant avenues for future research.
Collapse
Affiliation(s)
- Kyrre E Emblem
- The Intervention Centre, Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway
| | - Christian T Farrar
- Department of Radiology and Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Elizabeth R Gerstner
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, 100 Blossom Street, Boston, MA 02114, USA
| | - Tracy T Batchelor
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, 100 Blossom Street, Boston, MA 02114, USA
| | - Ronald J H Borra
- Department of Radiology and Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Bruce R Rosen
- Department of Radiology and Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - A Gregory Sorensen
- Siemens Healthcare Health Services, 51 Valley Stream Parkway, Malvern, PA 19355, USA
| | - Rakesh K Jain
- Edwin L. Steele Laboratory of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, 100 Blossom Street, Boston, MA 02114, USA
| |
Collapse
|
23
|
Ruan M, Shen Y, Chen L, Li M. RECIST 1.1 and serum thyroglobulin measurements in the evaluation of responses to sorafenib in patients with radioactive iodine-refractory differentiated thyroid carcinoma. Oncol Lett 2013; 6:480-486. [PMID: 24137351 PMCID: PMC3789091 DOI: 10.3892/ol.2013.1424] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 06/02/2013] [Indexed: 12/15/2022] Open
Abstract
The present study was designed to investigate the association between response evaluation criteria in solid tumors (RECIST) 1.1 and 1.0, and to explore the utility of thyroglobulin (Tg) measurements in assessing tumor responses to sorafenib in patients with radioactive iodine (RAI)-refractory differentiated thyroid carcinoma (DTC). In total, 23 patients with RAI-refractory DTC were enrolled. A comparison of RECIST 1.1 and 1.0 was performed in all patients with measurable disease. Following the exclusion of patients who were positive for anti-Tg antibody, the correlation between RECIST 1.1 and Tg was investigated in patients with measurable disease, and the concordance of the change in Tg between these patients and the patients with non-measurable disease only was analyzed over time. Tumor responses, assessed by RECIST 1.1 and 1.0, were concordant in 96% of the 23 records. However, the number of target lesions, according to RECIST 1.1, was significantly lower than when using RECIST 1.0. Progressive disease (PD) was identified in one of the five patients who underwent fluorodeoxyglucose-positron emission tomography (FDG-PET)/computed tomography (CT) scanning. A correlation between the Tg levels and the sum of the diameters of the target lesions was verified, with the percentage decrease in Tg levels significantly greater than that in the radiograph, demonstrating shrinkage. Furthermore, the percentage change in Tg levels was consistent between the patients with measurable disease and the subjects with non-measurable disease only. In conclusion, in patients with RAI-refractory DTC, RECIST 1.1 is highly concordant with RECIST 1.0 in the assessment of responses to sorafenib treatment, with the advantage of simplified procedures and the complementary use of FDG-PET. Tg measurements, in concordance with RECIST 1.1, are valuable in the evaluation of tumor responses.
Collapse
Affiliation(s)
- Maomei Ruan
- Department of Nuclear Medicine, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233
| | | | | | | |
Collapse
|
24
|
Li CG, Huang XE, Xu L, Li Y, Lu YY. Clinical application of serum tumor associated material (TAM) from non-small cell lung cancer patients. Asian Pac J Cancer Prev 2012; 13:301-4. [PMID: 22502689 DOI: 10.7314/apjcp.2012.13.1.301] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE To explore the associations of serum tumor associated material (TAM) with other common tumor markers like carcinoembryonic antigen (CEA), carbohydrate antigen 125 (CA125), carbohydrate antigen19-9 (CA19-9) and its clinical application in non-small cell lung cancer (NSCLC) patients. METHODS A total of 87 patients were enrolled into this study, all with histologically or cytologically confirmed NSCLC. With the method of chemical colorimetry, the level of TAM was determined and compared, while chemiluminescence was used to measure the levels of common tumor markers. RESULTS The level of TAM decreased after chemotherapy compared with before chemotherapy when CT or MRI scans showed disease control. Furthermore, it increased when disease progressed and there was no statistically significant difference in monitoring of TAM and common tumor markers (P>0.05). CONCLUSIONS Detecting TAM in NSCLC patients has a higher sensitivity and specificity, so it can be used as an indicator for clinical monitoring of lung cancer chemotherapy.
Collapse
Affiliation(s)
- Cheng-Guang Li
- Department of Chemotherapy, The Affiliated Jiangsu Cancer Hospital of Nanjing Medical University, Nanjing, China
| | | | | | | | | |
Collapse
|
25
|
Doi T, Hamaguchi T, Shirao K, Chin K, Hatake K, Noguchi K, Otsuki T, Mehta A, Ohtsu A. Evaluation of safety, pharmacokinetics, and efficacy of vorinostat, a histone deacetylase inhibitor, in the treatment of gastrointestinal (GI) cancer in a phase I clinical trial. Int J Clin Oncol 2012; 18:87-95. [PMID: 22234637 DOI: 10.1007/s10147-011-0348-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 10/29/2011] [Indexed: 01/08/2023]
Abstract
BACKGROUND Control of epigenetic changes using histone deacetylase inhibitors (HDACi) is thought to be a promising target in therapy of gastrointestinal (GI) cancer. In this study, we evaluated the safety, pharmacokinetics, and efficacy of two dosing regimens of vorinostat, an oral HDACi, in patients with GI tumors. METHODS Patients received either vorinostat 300 mg bid for 3 consecutive days followed by 4 rest days per cycle (n = 10) or vorinostat 400 mg qd for 21 consecutive days per cycle (n = 6). Pharmacokinetic parameters were assessed for the first treatment cycle. Efficacy was determined through evaluation of tumors and assessment of treatment response. RESULTS The median treatment duration of 300 mg bid was 52.0 days and of 400 mg qd was 51.5 days. The most common drug-related adverse events were anorexia, nausea, fatigue, and hyperglycemia. Two patients taking 400 mg qd had dose-limiting toxicities (DLTs) of thrombocytopenia. No patients taking 300 mg bid experienced DLT. Five patients taking 300 mg bid and 2 patients taking 400 mg qd maintained stable disease for >8 weeks, with the maximum duration of 245 days. Mean drug exposure (±SD) was generally higher with 400 mg qd (area under the curve [AUC(0-∞)] of 7.75 ± 2.79 μM h on Day 1 post-dose) compared with 300 mg bid (AUC(0-∞) of 3.94 ± 1.56 μM h on Day 1 post-dose). CONCLUSIONS Vorinostat 300 mg bid for 3 consecutive days followed by 4 days of rest was better tolerated in patients with GI cancer than a higher once daily dose. Additionally, there were patients in both groups who achieved stable disease, most maintaining it for longer than 8 weeks, suggesting vorinostat as a possible active agent in the treatment of GI cancer.
Collapse
|
26
|
Chai CY, Deneve JL, Beasley GM, Marzban SS, Chen YA, Rawal B, Grobmyer SR, Hochwald SN, Tyler DS, Zager JS. A multi-institutional experience of repeat regional chemotherapy for recurrent melanoma of extremities. Ann Surg Oncol 2011; 19:1637-43. [PMID: 22143576 DOI: 10.1245/s10434-011-2151-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Indexed: 11/18/2022]
Abstract
BACKGROUND Hyperthermic isolated limb perfusion (HILP) or isolated limb infusion (ILI) are well-accepted regional chemotherapy techniques for in-transit melanoma of extremity. The role and efficacy of repeat regional chemotherapy for recurrence and which salvage procedure is better remains debatable. We aimed to compare toxicities and clinical outcomes by procedure types and the sequence. METHODS Data from 44 patients, who underwent repeat HILPs or ILIs from 3 institutions beginning 1997 to 2010, were retrospectively reviewed. Regional toxicity assessed by Wieberdink grade, systemic toxicity assessed by serum creatine phosphokinase level, length of hospital stay (LOS), response rates at 3 months after the procedure, and time to in-field progression (TTP) were analyzed. RESULTS Of 44 patients, 46% were men and 54% women with a median age of 66 (range 29-85) years at diagnosis. The median follow-up was 21.4 (range 4-153) months. Of 70 ILIs and 28 HILPs, the following groups were identified: group A, ILI → ILI (n = 25); group B, ILI → HILP (n = 10); group C, HILP → ILI (n = 12); and group D, HILP → HILP (n = 3). The comparison of Wieberdink grade, serum creatine phosphokinase level, LOS, and response rate between procedures (HILP vs. ILI), between sequence (initial vs. repeat), and among their interactions showed no statistically significant differences. TTP after initial procedure did not differ between HILP and ILI (P = 0.08), and no survival difference was seen (P = 0.65) when TTP after repeat procedure was compared. CONCLUSIONS Most patients tolerated repeat regional chemotherapy without increased toxicity or LOS. No statistical difference in clinical outcomes was noted when comparing repeat procedures, even though repeat HILPs showed higher complete response compared to repeat ILIs.
Collapse
Affiliation(s)
- Christy Y Chai
- Department of Surgery, San Antonio Military Medical Center, San Antonio, TX, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Turaga KK, Beasley GM, Kane JM, Delman KA, Grobmyer SR, Gonzalez RJ, Letson GD, Cheong D, Tyler DS, Zager JS. Limb preservation with isolated limb infusion for locally advanced nonmelanoma cutaneous and soft-tissue malignant neoplasms. ACTA ACUST UNITED AC 2011; 146:870-5. [PMID: 21768436 DOI: 10.1001/archsurg.2011.139] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE To demonstrate the efficacy of isolated limb infusion (ILI) in limb preservation for patients with locally advanced soft-tissue sarcomas and nonmelanoma cutaneous malignant neoplasms. BACKGROUND Locally advanced nonmelanoma cutaneous and soft-tissue malignant neoplasms, including soft-tissue sarcomas of the extremities, can pose significant treatment challenges. We report our experience, including responses and limb preservation rates, using ILI in cutaneous and soft-tissue malignant neoplasms. METHODS We identified 22 patients with cutaneous and soft-tissue malignant neoplasms who underwent 26 ILIs with melphalan and dactinomycin from January 1, 2004, through December 31, 2009, from 5 institutions. Outcome measures included limb preservation and in-field response rates. Regional toxic effects were measured using the Wieberdink scale and serum creatinine phosphokinase levels. RESULTS The median age was 70 years (range, 19-92 years), and 12 patients (55%) were women. Fourteen patients (64%) had sarcomas, 7 (32%) had Merkel cell carcinoma, and 1 (5%) had squamous cell carcinoma. The median length of stay was 5.5 days (interquartile range, 4-8 days). Twenty-five of the 26 ILIs (96%) resulted in Wieberdink grade III or less toxicity, and 1 patient (4%) developed grade IV toxicity. The median serum creatinine phosphokinase level was 127 U/L for upper extremity ILIs and 93 U/L for lower extremity ILIs. Nineteen of 22 patients (86%) underwent successful limb preservation. The 3-month in-field response rate was 79% (21% complete and 58% partial), and the median follow-up was 8.6 months (range, 1-63 months). Five patients underwent resection of disease after an ILI, of whom 80% are disease free at a median of 8.6 months. CONCLUSIONS Isolated limb infusion provides an attractive alternative therapy for regional disease control and limb preservation in patients with limb-threatening cutaneous and soft-tissue malignant neoplasms. Short-term response rates appear encouraging, yet durability of response is unknown.
Collapse
Affiliation(s)
- Kiran K Turaga
- Division of Surgical Oncology, Medical College of Wisconsin Clinical Cancer Center, Milwaukee, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Shimada Y, Niho S, Ishii G, Hishida T, Yoshida J, Nishimura M, Yoh K, Goto K, Ohmatsu H, Ohe Y, Nagai K. Clinical features of unresectable high-grade lung neuroendocrine carcinoma diagnosed using biopsy specimens. Lung Cancer 2011; 75:368-73. [PMID: 21920624 DOI: 10.1016/j.lungcan.2011.08.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 08/19/2011] [Accepted: 08/20/2011] [Indexed: 01/30/2023]
Abstract
BACKGROUND The overall clinicopathological features or the optimal therapy for large cell neuroendocrine carcinoma (LCNEC) have yet to be defined, because LCNEC has not been studied in the same depth as had small cell lung carcinoma (SCLC) in both clinical and biological standpoints. The aim of this study was to elucidate the clinical features of high-grade neuroendocrine carcinoma (HGNEC)-probable LCNEC diagnosed by biopsy, and compare therapeutic efficacy with patients with SCLC. METHODS We retrospectively examined the chart of total of 25 patients who underwent chemotherapy or chemoradiotherapy as initial therapy for a histologic diagnosis of HGNEC-probable LCNEC, using biopsy samples and compared their data with those of 180 patients with SCLC. We analyzed their responses to chemotherapy and/or radiation therapy and survival outcomes. RESULTS In 25 patients with HGNEC-probable LCNEC, 18 patients initially received chemotherapy (17 (94%) of whom received platinum-based chemotherapy) with an overall response rate (ORR) of 61%. The remaining 7 patients received chemoradiotherapy with an ORR of 86%, and 12 of the 25 patients who received second-line chemotherapy had an ORR of 17%. A total of 101 patients with SCLC who initially received chemotherapy had an ORR of 63%, and 79 patients who initially received chemoradiotherapy had an ORR of 98%, and 102 of the 180 patients who received second-line chemotherapy had an ORR of 45%. The 1-year overall survival rate for patients with stage IV HGNEC-probable LCNEC (n=13) and those with ED-SCLC (n=80) was 34% and 49%, respectively (p=0.84). CONCLUSION The overall response rate to initial treatment and the survival outcomes of HGNEC-probable LCNEC were comparable to those of SCLC, but the effectiveness of second-line chemotherapy appeared to differ between the 2 groups.
Collapse
Affiliation(s)
- Yoshihisa Shimada
- Division of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Walter B, Schrettenbrunner I, Vogelhuber M, Grassinger J, Bross K, Wilke J, Suedhoff T, Berand A, Wieland WF, Rogenhofer S, Andreesen R, Reichle A. Pioglitazone, etoricoxib, interferon-α, and metronomic capecitabine for metastatic renal cell carcinoma: final results of a prospective phase II trial. Med Oncol 2011; 29:799-805. [DOI: 10.1007/s12032-011-9982-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Accepted: 05/09/2011] [Indexed: 11/30/2022]
|
30
|
Grignani G, Palmerini E, Dileo P, Asaftei SD, D'Ambrosio L, Pignochino Y, Mercuri M, Picci P, Fagioli F, Casali PG, Ferrari S, Aglietta M. A phase II trial of sorafenib in relapsed and unresectable high-grade osteosarcoma after failure of standard multimodal therapy: an Italian Sarcoma Group study. Ann Oncol 2011; 23:508-16. [PMID: 21527590 DOI: 10.1093/annonc/mdr151] [Citation(s) in RCA: 280] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
PURPOSE After standard multimodal therapy, the prognosis of relapsed and unresectable high-grade osteosarcoma is dismal and unchanged over the last decades. Recently, mitogen-activated protein kinases were shown to be activated in osteosarcoma specimens, suggesting, therefore, they are suitable targets for the multikinase inhibitor sorafenib. Thus, we explored sorafenib activity in patients with relapsed and unresectable osteosarcoma. EXPERIMENTAL DESIGN Patients > 14 years, progressing after standard treatment, were eligible to receive 400 mg of sorafenib twice daily until progression or unacceptable toxicity. The primary end point was progression-free survival (PFS) at 4 months. Secondary objectives were PFS, overall survival (OS), clinical benefit rate (CBR), defined as no progression at 6 months and safety. This nonrandomized phase II study used a Simon two-stage design. PFS and OS at 95% confidence intervals (95% CIs) were calculated by the Kaplan-Meier method. All tests were two sided. RESULTS Thirty-five patients were enrolled. PFS at 4 months was 46% (95% CI 28% to 63%). Median PFS and OS were 4 (95% CI 2-5) and 7 (95% CI 7-8) months, respectively. The CBR was 29% (95% CI 13% to 44%). We observed 3 (8%) partial responses (PRs), 2 (6%) minor responses (< 30% tumor shrinkage) and 12 (34%) stable diseases (SDs). For six patients (17%), PR/SD lasted ≥ 6 months. Noteworthy, tumor density reduction and [(18)F]2-fluoro-2-deoxy-d-glucose-positron emission tomography responses were observed among SD patients. Sorafenib was reduced or briefly interrupted in 16 (46%) patients and permanently discontinued in one (3%) case due to toxicity. CONCLUSIONS Sorafenib demonstrated activity as a second- or third-line treatment in terms of PFS at 4 months with some unprecedented long-lasting responses. Sorafenib, the first targeted therapy showing activity in osteosarcoma patients, deserves further investigations.
Collapse
Affiliation(s)
- G Grignani
- Medical Oncology Unit, Institute for Cancer Research and Treatment, Candiolo, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Luboldt W, Hartmann H, Wiedemann B, Zöphel K, Luboldt HJ. Gastroenteropancreatic Neuroendocrine Tumors: Standardizing Therapy Monitoring with 68Ga-DOTATOC PET/CT Using the Example of Somatostatin Receptor Radionuclide Therapy. Mol Imaging 2010; 9:7290.2010.00035. [DOI: 10.2310/7290.2010.00035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
| | - Holger Hartmann
- From the Multiorgan Screening Foundation, Frankfurt, Germany
| | | | - Klaus Zöphel
- From the Multiorgan Screening Foundation, Frankfurt, Germany
| | | |
Collapse
|
32
|
Holloway RW, Grendys EC, Lefebvre P, Vekeman F, McMeekin S. Tolerability, efficacy, and safety of pegylated liposomal Doxorubicin in combination with Carboplatin versus gemcitabine-Carboplatin for the treatment of platinum-sensitive recurrent ovarian cancer: a systematic review. Oncologist 2010; 15:1073-82. [PMID: 20930103 PMCID: PMC3227899 DOI: 10.1634/theoncologist.2009-0331] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 07/26/2010] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE To compare the tolerability, efficacy, and safety profiles of pegylated liposomal doxorubicin in combination with carboplatin (PLD-Carbo) with those of gemcitabine-carboplatin (Gem-Carbo) for the treatment of patients with platinum-sensitive recurrent ovarian cancer (PSROC) by reviewing the published literature. METHODS Using the PubMed database, a systematic review of peer-reviewed literature published between January 2000 and September 2009 was undertaken to identify studies related to the treatment of patients with PSROC with PLD-Carbo or Gem-Carbo. Studies reporting either response rate, progression-free survival (PFS), and/or overall survival (OS) were included. Treatment regimens, efficacy endpoints, and safety profiles were compared between the two combination therapies. RESULTS Ten studies evaluating 608 patients (PLD-Carbo: 5 studies, 278 patients; Gem-Carbo: 5 studies, 330 patients) were identified. The mean planned doses were: PLD, 34.8 mg/m(2) and Gem, 993 mg/m(2). The dose intensity reported in Gem trials was lower (75% of the planned dose) than the dose intensity reported in PLD trials (93.7% of the planned dose), suggesting better tolerability for the PLD-Carbo regimen. Among patients receiving PLD-Carbo, 60.2% achieved a response (complete, 27.0%; partial, 33.2%), versus 51.4% of patients treated with Gem-Carbo (complete, 19.2%; partial, 32.2%). The median PFS times were 10.6 months and 8.9 months in the PLD-Carbo and the Gem-Carbo populations, respectively. The median OS was longer for the PLD-Carbo regimen (27.1 months) than for the Gem-Carbo regimen (19.7 months). The hematological safety profiles were comparable in the two groups, although grade III or IV anemia (PLD-Carbo, 13.6%; Gem-Carbo, 24.5%) and neutropenia (PLD-Carbo, 45.5%; Gem-Carbo, 62.9%) were more common in patients receiving Gem-Carbo. CONCLUSION Results from this systematic analysis of peer-reviewed literature suggest that PLD-Carbo therapy is a rational alternative to Gem-Carbo for the treatment of patients with PSROC.
Collapse
|
33
|
Kudo M, Kubo S, Takayasu K, Sakamoto M, Tanaka M, Ikai I, Furuse J, Nakamura K, Makuuchi M. Response Evaluation Criteria in Cancer of the Liver (RECICL) proposed by the Liver Cancer Study Group of Japan (2009 Revised Version). Hepatol Res 2010; 40:686-92. [PMID: 20633194 DOI: 10.1111/j.1872-034x.2010.00674.x] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The World Health Organization (WHO) criteria and Response Evaluation Criteria in Solid Tumors (RECIST) are inappropriate to assess the direct effects of treatment on the hepatocellular carcinoma (HCC) by locoreginal therapies such as radiofrequency ablation (RFA) and transcatheter arterial chemoembolization (TACE). Therefore, establishment of response evaluation criteria solely devoted for HCC is needed urgently in the clinical practice as well as in the clinical trials of HCC treatment, such as molecular targeted therapies, which cause necrosis of the tumor. Response Evaluation Criteria in Cancer of the Liver (RECICL) was revised in 2009 by Liver Cancer Study Group of Japan based on the 2004 version of RECICL, which was commonly used in Japan. Major revised points of the RECICL 2009 is to provide TE4a (Complete response with enough ablative margin) and TE4b (complete response without enough ablative margin) for local ablation therapy. Second revised point is that setting the timing at which the overall treatment effects are assessed. Third point is that emergence of new lesion in the liver is regarded as progressive disease, different from 2004 version. Finally, 3 tumor markers including alpha-fetoprotein (AFP) and AFP-L3 and des-gamma-carboxy protein (DCP) were also added for the overall treatment response. We hope this new treatment response criteria, RECICL, proposed by Liver Cancer Study Group of Japan will benefit the HCC treatment response evaluation in the setting of the daily clinical practice and clinical trials as well not only in Japan, but also internationally.
Collapse
Affiliation(s)
- Masatoshi Kudo
- The Liver Cancer Study Group of Japan, Department of Gastroenterology and Hepatology, Kinki University School of Medicine, Osaka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Molnár TF, Balikó Z, Sárosi V, Horváth PO. Survival after surgery following chemotherapy for non-small-cell lung cancer. Asian Cardiovasc Thorac Ann 2010; 18:141-6. [PMID: 20304848 DOI: 10.1177/0218492310361271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A retrospective study was carried out on 74 patients with advanced non-small-cell lung cancer (52 in stage IIIA, 22 in stage IIIB) who received platinum-based induction chemotherapy in doublets and triplets, followed by tumor resection. Thirty-day postoperative mortality was 5.4% (4 patients); 5 patients in stage IIIB and 17 in stage IIIA did not respond, but the other 47 (63.5%) were downstaged to < IIIA (26 were downstaged to stage I, 20 to stage II, and 1 had complete remission). There was no change in T factor in 22 (30%) patients, nor in N factor in 21 (28%). The actuarial 5-year survival rate for patients in postoperative stages IIIA and IIIB was 0.496; survival was significantly longer in patients who responded to therapy. Parallel improvement in both T and N status predicted worse survival than a multistage regression in any single factor. N status was found to be a stronger survival indicator than T status. Cell type did not influence the response rate or outcome. Induction chemotherapy significantly improved survival in patients who responded, despite a poor prognosis.
Collapse
Affiliation(s)
- Tamás F Molnár
- Department of Pneumonology, University of Pécs, Hungary.
| | | | | | | |
Collapse
|
35
|
Igawa S, Watanabe R, Ito I, Murakami H, Takahashi T, Nakamura Y, Tsuya A, Kaira K, Naito T, Endo M, Yamamoto N, Kameya T. Comparison of chemotherapy for unresectable pulmonary high-grade non-small cell neuroendocrine carcinoma and small-cell lung cancer. Lung Cancer 2010; 68:438-45. [DOI: 10.1016/j.lungcan.2009.07.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Revised: 06/26/2009] [Accepted: 07/03/2009] [Indexed: 10/20/2022]
|
36
|
Lam ET, Ringel MD, Kloos RT, Prior TW, Knopp MV, Liang J, Sammet S, Hall NC, Wakely PE, Vasko VV, Saji M, Snyder PJ, Wei L, Arbogast D, Collamore M, Wright JJ, Moley JF, Villalona-Calero MA, Shah MH. Phase II clinical trial of sorafenib in metastatic medullary thyroid cancer. J Clin Oncol 2010; 28:2323-30. [PMID: 20368568 PMCID: PMC2881718 DOI: 10.1200/jco.2009.25.0068] [Citation(s) in RCA: 273] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Accepted: 02/01/2010] [Indexed: 01/16/2023] Open
Abstract
PURPOSE Mutations in the RET proto-oncogene and vascular endothelial growth factor receptor (VEGFR) activity are critical in the pathogenesis of medullary thyroid cancer (MTC). Sorafenib, a multikinase inhibitor targeting Ret and VEGFR, showed antitumor activity in preclinical studies of MTC. PATIENTS AND METHODS In this phase II trial of sorafenib in patients with advanced MTC, the primary end point was objective response. Secondary end points included toxicity assessment and response correlation with tumor markers, functional imaging, and RET mutations. Using a two-stage design, 16 or 25 patients were to be enrolled onto arms A (hereditary) and B (sporadic). Patients received sorafenib 400 mg orally twice daily. RESULTS Of 16 patients treated in arm B, one achieved partial response (PR; 6.3%; 95% CI, 0.2% to 30.2%), 14 had stable disease (SD; 87.5%; 95% CI, 61.7% to 99.5%), and one was nonevaluable. In a post hoc analysis of 10 arm B patients with progressive disease (PD) before study, one patient had PR of 21+ months, four patients had SD >or= 15 months, four patients had SD CONCLUSION Sorafenib is reasonably well tolerated, with suggestion of clinical benefit for patients with sporadic MTC. Caution should be taken because of the rare but fatal toxicity potentially associated with sorafenib.
Collapse
Affiliation(s)
- Elaine T Lam
- Departments of Internal Medicine, Center for Biostatistics, The Ohio State University, Columbus, OH, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Weekly paclitaxel for heavily treated advanced or recurrent gastric cancer refractory to fluorouracil, irinotecan, and cisplatin. Gastric Cancer 2010; 12:206-11. [PMID: 20047125 DOI: 10.1007/s10120-009-0524-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Accepted: 09/02/2009] [Indexed: 02/07/2023]
Abstract
BACKGROUND Although triweekly administration of paclitaxel is approved for gastric cancer in Japan, currently, the drug is often delivered with a weekly schedule because of the equivalent efficacy and lesser toxicity of this dosing schedule as compared with the triweekly administration schedule. Weekly administration of paclitaxel as second-line or first-line chemotherapy for gastric cancer has been reported to yield a response rate of about 20%. Because there has been no report of the efficacy of weekly paclitaxel in the third-line setting, this retrospective study investigated the efficacy and toxicities of weekly paclitaxel used in the third-line setting for the treatment of gastric cancer refractory to all three key drugs, fluorouracil, irinotecan, and cisplatin, used in clinical practice. METHODS In 85 patients with advanced or recurrent histologically confirmed gastric adenocarcinoma who had failed to respond to prior chemotherapy regimens containing fluorouracil, irinotecan, and cisplatin, paclitaxel (80 mg/m(2)) was administered weekly, three times, for 3 weeks out of 4. RESULTS The median number of courses was 3 (range, 1-38). The overall response rate was 23.2% (19/82) in the patients with measurable lesions, and ascites disappeared in 15 of 48 patients (31.3%). Progression-free survival was 105 days and the median survival time was 201 days from the initiation of paclitaxel administration. Grade 3 or 4 leukopenia, neutropenia, anemia, and thrombocytopenia were observed in 25 (29%), 25 (29%), 37 (44%), and 3 (4%) patients. Other, nonhematological, toxicities were nausea, vomiting, anorexia, sensory neuropathy, fatigue, and febrile neutropenia. CONCLUSION Weekly paclitaxel administration shows activity against advanced gastric cancer also in the third-line setting.
Collapse
|
38
|
Blattmann C, Oertel S, Schulz-Ertner D, Rieken S, Haufe S, Ewerbeck V, Unterberg A, Karapanagiotou-Schenkel I, Combs SE, Nikoghosyan A, Bischof M, Jäkel O, Huber P, Kulozik AE, Debus J. Non-randomized therapy trial to determine the safety and efficacy of heavy ion radiotherapy in patients with non-resectable osteosarcoma. BMC Cancer 2010; 10:96. [PMID: 20226028 PMCID: PMC2846886 DOI: 10.1186/1471-2407-10-96] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Accepted: 03/12/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Osteosarcoma is the most common primary malignant bone tumor in children and adolescents. For effective treatment, local control of the tumor is absolutely critical, because the chances of long term survival are <10% and might effectively approach zero if a complete surgical resection of the tumor is not possible. Up to date there is no curative treatment protocol for patients with non-resectable osteosarcomas, who are excluded from current osteosarcoma trials, e.g. EURAMOS1. Local photon radiotherapy has previously been used in small series and in an uncontrolled, highly individualized fashion, which, however, documented that high dose radiotherapy can, in principle, be used to achieve local control. Generally the radiation dose that is necessary for a curative approach can hardly be achieved with conventional photon radiotherapy in patients with non-resectable tumors that are usually located near radiosensitive critical organs such as the brain, the spine or the pelvis. In these cases particle Radiotherapy (proton therapy (PT)/heavy ion therapy (HIT) may offer a promising new alternative. Moreover, compared with photons, heavy ion beams provide a higher physical selectivity because of their finite depth coverage in tissue. They achieve a higher relative biological effectiveness. Phase I/II dose escalation studies of HIT in adults with non-resectable bone and soft tissue sarcomas have already shown favorable results. METHODS/DESIGN This is a monocenter, single-arm study for patients > or = 6 years of age with non-resectable osteosarcoma. Desired target dose is 60-66 Cobalt Gray Equivalent (Gy E) with 45 Gy PT (proton therapy) and a carbon ion boost of 15-21 GyE. Weekly fractionation of 5-6 x 3 Gy E is used. PT/HIT will be administered exclusively at the Ion Radiotherapy Center in Heidelberg. Furthermore, FDG-PET imaging characteristics of non-resectable osteosarcoma before and after PT/HIT will be investigated prospectively. Systemic disease before and after PT/HIT is targeted by standard chemotherapy protocols and is not part of this trial. DISCUSSION The primary objectives of this trial are the determination of feasibility and toxicity of HIT. Secondary objectives are tumor response, disease free survival and overall survival. The aim is to improve outcome for patients with non-resectable osteosarcoma. TRIAL REGISTRATION Registration number (ClinicalTrials.gov): NCT01005043.
Collapse
Affiliation(s)
- Claudia Blattmann
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Im Neuenheimer Feld 154, 69120 Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Igawa S, Murakami H, Takahashi T, Nakamura Y, Tsuya A, Naito T, Kaira K, Ono A, Shukuya T, Tamiya A, Endo M, Yamamoto N. Efficacy of chemotherapy with carboplatin and paclitaxel for unresectable thymic carcinoma. Lung Cancer 2010; 67:194-7. [DOI: 10.1016/j.lungcan.2009.03.031] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Revised: 03/27/2009] [Accepted: 03/30/2009] [Indexed: 11/29/2022]
|
40
|
Kudo M, Kubo S, Takayasu K, Sakamoto M, Tanaka M, Ikai I, Furuse J, Nakamura K, Makuuchi M. Response Evaluation Criteria in Cancer of the Liver (RECICL). ACTA ACUST UNITED AC 2010. [DOI: 10.2957/kanzo.51.261] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
41
|
Cwikla JB, Sankowski A, Seklecka N, Buscombe JR, Nasierowska-Guttmejer A, Jeziorski KG, Mikolajczak R, Pawlak D, Stepien K, Walecki J. Efficacy of radionuclide treatment DOTATATE Y-90 in patients with progressive metastatic gastroenteropancreatic neuroendocrine carcinomas (GEP-NETs): a phase II study. Ann Oncol 2009; 21:787-794. [PMID: 19833821 DOI: 10.1093/annonc/mdp372] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND To evaluate the clinical and radiological effectiveness of [DOTA(0), D-Phe(1), Tyr(3)]-octreotate (DOTATATE) Y-90 in patients with extensive progressive gastroenteropancreatic neuroendocrine carcinomas (GEP-NETs). MATERIALS AND METHODS Sixty patients with histologically proven GEP-NETs were treated with DOTATATE Y-90. Clinical responses were assessed 6 weeks after completing therapy and then after each of the 3- to 6-month intervals. The radiological response was classified according to RECIST criteria. RESULTS At 6 months after final treatment, radiological partial response (PR; at least a 30% decrease in the sum of the longest diameter of target lesions) was observed in 13 patients (23%), and the remaining patients had stable disease (SD; less than 30% decrease in the sum of the longest diameter of target lesions or less than 20% increase in the sum of the longest diameter of target lesions) (77%). Clinical PR at 6 months was in 43 patients (72%), nine patients had SD and progressive disease (PD) was noted in eight patients. Median progression-free survival (PFS) was 17 months, while the median overall survival (OS) was 22 months. In eight patients with early PD, the PFS was 4.5 and OS 9.5 months, while in those with SD or PR, PFS and OS were 19.5 and 23.5 months, respectively. After 12 months of follow-up, five patients had World Health Organization (WHO) grade 2 or 3 renal toxicity. Haematological toxicity (WHO grade 3 and 4) was noted during therapy in 10% of patients and persisted in 5%. CONCLUSIONS DOTATATE Y-90 therapy is effective and relatively safe in patients with GEP-NET. Standard doses of DOTATATE Y-90 result in a relatively low risk of myelotoxicity. However, due to ongoing risk of renal toxicity, careful monitoring of the kidney is recommended.
Collapse
Affiliation(s)
- J B Cwikla
- Department of Radiology and Diagnostic Imaging, Medical Centre for Postgraduate Education and Central Clinical Hospital of Ministry of Internal Affairs and Administration, Warsaw.
| | - A Sankowski
- Department of Radiology and Diagnostic Imaging, Central Clinical Hospital of Ministry of Internal Affairs and Administration, Warsaw, Poland
| | - N Seklecka
- Department of Radiology and Diagnostic Imaging, Central Clinical Hospital of Ministry of Internal Affairs and Administration, Warsaw, Poland
| | - J R Buscombe
- Department of Nuclear Medicine, Royal Free Hospital, London, UK
| | - A Nasierowska-Guttmejer
- Department of Pathology, Central Clinical Hospital of Ministry of Internal Affairs and Administration, Warsaw
| | - K G Jeziorski
- Department of Radiology and Diagnostic Imaging, Central Clinical Hospital of Ministry of Internal Affairs and Administration, Warsaw, Poland; Department of Upper Digestive Tract Cancer, Maria Skłodowska-Curie Memorial Cancer Centre and Institute of Oncology, Warsaw
| | - R Mikolajczak
- Institute of Atomic Energy, Radioisotope Centre POLATOM, Otwock-Swierk
| | - D Pawlak
- Institute of Atomic Energy, Radioisotope Centre POLATOM, Otwock-Swierk
| | - K Stepien
- Central Laboratory, Central Clinical Hospital of Ministry of Internal Affairs and Administration, Warsaw
| | - J Walecki
- Department of Radiology and Diagnostic Imaging, Medical Centre for Postgraduate Education and Central Clinical Hospital of Ministry of Internal Affairs and Administration, Warsaw; Department of Experimental Pharmacology, Polish Academy of Science Medical Research Center, Warsaw, Poland
| |
Collapse
|
42
|
Ramaswamy B, Bekaii-Saab T, Schaaf LJ, Lesinski GB, Lucas DM, Young DC, Ruppert AS, Byrd JC, Culler K, Wilkins D, Wright JJ, Grever MR, Shapiro CL. A dose-finding and pharmacodynamic study of bortezomib in combination with weekly paclitaxel in patients with advanced solid tumors. Cancer Chemother Pharmacol 2009; 66:151-8. [PMID: 19774377 DOI: 10.1007/s00280-009-1145-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Accepted: 09/12/2009] [Indexed: 01/04/2023]
Abstract
PURPOSE A phase I study to determine the maximum tolerated dose (MTD) of bortezomib (B) when combined with weekly paclitaxel in patients with advanced solid tumors. PATIENTS AND METHODS Eligible patients received escalating doses of intravenous (IV) bortezomib (0.6-2 mg/m(2)) on days 2 and 9 and IV paclitaxel at 100 mg/m(2) on days 1 and 8 of a 21-day cycle. Dose escalation was based on two end-points: not exceeding 80% 20S-proteasome inhibition (20-S PI) and the development of dose-limiting toxicity defined as grade 3 or greater non-hematologic or grade 4 hematologic toxicities. RESULTS Forty-five patients with advanced solid tumors and a median of 3 prior chemotherapy regimens (range 0-9), received 318 doses (median 5, range 1-34) of bortezomib and paclitaxel. Dose-related inhibition of 20-S PI was observed with a maximum inhibition of 70-80% at the MTD of 1.8 mg/m(2) of bortezomib. At the MTD (N = 9) the following toxicities were observed: grade 4 neutropenia without fever (n = 2) and cerebrovascular ischemia (n = 1); grade 3 neutropenia (n = 3), diarrhea (n = 2), nausea (n = 1), and fatigue (n = 1); grade 2 fatigue (n = 5), diarrhea (n = 4), and dyspnea (n = 2). There was one partial response in a patient with an eccrine porocarcinoma. Stabilization of disease was observed in 7 (16%) patients, 3 of whom had advanced pancreatic cancer. CONCLUSION Sequential paclitaxel and bortezomib in previously treated patients with advanced solid tumors resulted in acceptable toxicity and no evidence of interaction. The recommended phase II dose of bortezomib in combination with weekly paclitaxel was 1.8 mg/m(2).
Collapse
Affiliation(s)
- Bhuvaneswari Ramaswamy
- The Ohio State University, B401, Starling Loving Hall, 320 W 10th av, Columbus, OH 43210, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Blanch Alerany M, Calvo Campos M, Romera Villegas A, Pérez-Piqueras Gómez A, Riera Batalla S, Cairols Castellote MA. [Deep venous thrombosis as a marker of tumor recurrence]. Med Clin (Barc) 2009; 133:529-32. [PMID: 19656530 DOI: 10.1016/j.medcli.2009.04.054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Accepted: 04/23/2009] [Indexed: 10/20/2022]
Abstract
BACKGROUND AND OBJECTIVE There is a bidirectional association between neoplasia and venous thromboembolism (VTE). However, whether this association has an impact in tumor recurrence is unknown. The aim of the study is to assess the incidence of cancer recurrence after diagnosis of VTE and determine if VTE could be a marker for cancer recurrence. PATIENTS AND METHOD This was a retrospective cohort observational study. The observations were made in June 2008. The study included 132 patients diagnosed with a deep vein thrombosis (DVT) between January 2002 and December 2005 and had a cancer without activity in the last 12 months in their medical history. The cancer recurrence was assessed. The sample was divided according to the presence or absence of cancer recurrence and study variables were compared. RESULTS The incidence of cancer recurrence after diagnosis with DVT was 50,7%. This recurrence occurred in the first 4 months in 55,2% and more frequent and earlier in patients with breast tumors (p:00014), with carcinoma (p:0,02), in patients under palliative chemotherapy and in patients with previous presence of metastasis. (p:0,00). CONCLUSIONS In 50,7% of patients without neoplasic activity in the last year, DVT was the first sign of tumor recurrence. These patients should be referred to an oncology department as soon as possible in order to diagnose the tumor recurrence.
Collapse
Affiliation(s)
- Montserrat Blanch Alerany
- Servei d'Angiologia i Cirurgia Vascular, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, España.
| | | | | | | | | | | |
Collapse
|
44
|
Kloos RT, Ringel MD, Knopp MV, Hall NC, King M, Stevens R, Liang J, Wakely PE, Vasko VV, Saji M, Rittenberry J, Wei L, Arbogast D, Collamore M, Wright JJ, Grever M, Shah MH. Phase II trial of sorafenib in metastatic thyroid cancer. J Clin Oncol 2009; 27:1675-84. [PMID: 19255327 DOI: 10.1200/jco.2008.18.2717] [Citation(s) in RCA: 395] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Based on the pivotal role of Ras-Raf-MAP-ERK signaling and vascular endothelial growth factor (VEGF) in papillary thyroid cancer (PTC), we conducted a phase II clinical trial of sorafenib targeting RAF and VEGF receptor kinases in PTC. PATIENTS AND METHODS The primary end point was the objective response rate. Secondary end points included response correlation with serum thyroglobulin (Tg); functional imaging; tumor genotype; and signaling inhibition in tumor biopsies. Using a Simon minimax two-stage design, 16 or 25 chemotherapy-naïve metastatic PTC patients were to be enrolled in arm A (accessible tumor for biopsy). Arm B patients had other subtypes of thyroid carcinoma or prior chemotherapy, and did not require tumor biopsies. Patients received 400 mg orally twice per day of sorafenib. Response was assessed every 2 months using RECIST (Response Evaluation Criteria in Solid Tumors). RESULTS Of 41 PTC patients, six patients had a partial response (PR; 15%; 95% CI, 6 to 29) and 23 patients (56%; 95% CI, 40 to 72) had stable disease longer than 6 months. Median duration of PR was 7.5 months (range, 6 to 14). Median progression-free survival was 15 months (95% CI, 10 to 27.5). In 14 (78%) of 18 Tg-assessable PTC patients, Tg declined more than 25%. Common grade 3 adverse events included hand-foot skin reaction, musculoskeletal pain, and fatigue. BRAF mutation was detected in 17 (77%) of 22 PTCs analyzed. Four of 10 paired tumor biopsies from PTC patients showed a reduction in levels of vascular endothelial growth factor receptor phosphorylation, ERK phosphorylation, and in VEGF expression during sorafenib therapy. No PRs were noted among non-PTC patients. CONCLUSION Sorafenib is reasonably well-tolerated therapy with clinical and biologic antitumor activity in metastatic PTC.
Collapse
Affiliation(s)
- Richard T Kloos
- Ohio State University, Department of Internal Medicine, Molecular Virology, Immunology, and Genetics, Pathology, Radiology, Center for Biostatistics, Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Mantatzis M, Kakolyris S, Amarantidis K, Karayiannakis A, Prassopoulos P. Treatment response classification of liver metastatic disease evaluated on imaging. Are RECIST unidimensional measurements accurate? Eur Radiol 2009; 19:1809-16. [PMID: 19238395 DOI: 10.1007/s00330-009-1327-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2008] [Revised: 12/07/2008] [Accepted: 01/05/2009] [Indexed: 11/29/2022]
Abstract
The purpose of this study was to evaluate the accuracy of unidimensional measurements (response evaluation criteria in solid tumors, RECIST) compared with volumetric measurements in patients with liver metastases undergoing chemotherapy. Forty-four patients with newly diagnosed liver lesions underwent three MRI examinations at treatment initiation, during chemotherapy, and immediately post-treatment. Measurements based on RECIST guidelines and volume calculations were performed on the "target" lesions (TLs). The two methods were in agreement in 64/77 of patients and 253/301 of individual lesions classification in response categories ("good" agreement, Cohen kappa = 0.735 and 0.741, respectively). In 16.88% of the comparisons the two methods stratified patients to a different response category; 27.6% of TLs did not follow the response category of the patient in whom lesions were located. The actual volume of TLs differs from the calculated volume of a sphere with the same diameter. Our study supports the use of volumetric techniques that may overcome certain disadvantages of unidimensional measurements.
Collapse
Affiliation(s)
- Michael Mantatzis
- Department of Radiology, University Hospital of Alexandroupolis, Opsikiou 1, 681 00, Alexandroupolis, Greece.
| | | | | | | | | |
Collapse
|
46
|
Sørensen PD, Jakobsen EH, Langkjer ST, Bokmand S, Østergaard B, Olsen DA, Madsen JS, Brandslund I. Serum HER-2 concentrations for monitoring women with breast cancer in a routine oncology setting. Clin Chem Lab Med 2009; 47:1117-23. [DOI: 10.1515/cclm.2009.241] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
47
|
Fearmonti RM, Keyomarsi K, Hunt KK. Biomarkers in neoadjuvant trials. Cancer Treat Res 2009; 147:1-36. [PMID: 21461824 DOI: 10.1007/978-0-387-09463-2_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
48
|
Srirajaskanthan R, Toumpanakis C, Karpathakis A, Marelli L, Quigley AM, Dusmet M, Meyer T, Caplin ME. Surgical management and palliative treatment in bronchial neuroendocrine tumours: a clinical study of 45 patients. Lung Cancer 2008; 65:68-73. [PMID: 19070398 DOI: 10.1016/j.lungcan.2008.10.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2008] [Revised: 10/13/2008] [Accepted: 10/20/2008] [Indexed: 11/28/2022]
Abstract
Bronchial neuroendocrine tumours account for 1-2% of all lung cancers; they are thought to arise from the neuroendocrine cells located in the bronchial mucosa. The majority of the literature available comprises surgical series and there is a scarcity of data available for the management of patients with inoperable disease. We present a series of 45 patients referred to our institution from 1998 to 2006, with a mean follow-up of 54 months. Histological diagnosis from our department was available for 39 patients, with the remainder having had histological assessment performed previously. Typical carcinoid was present in 25 cases, atypical in 9 cases, large cell neuroendocrine carcinoma in 4 and 1 case of small cell lung carcinoma. All patients were staged at time of initial diagnosis with CT scan, in addition Octreoscans were performed when appropriate. Twenty-six of these 45 cases had unresectable disease, whilst the remainder were treated with surgical resection. Initial therapy with surgical resection was performed in 19 patients, 2 of whom had undergone neo-adjuvant chemotherapy. Recurrence occurred in 7 (36.8%), average duration of disease-free survival post-surgery was 61 months. Chemotherapy was first line therapy in five cases, four achieved disease stabilization and one case had progressive disease. Somatostatin analogues were used as first line therapy in six patients, for symptom control and anti-tumour effect. Peptide receptor radionuclide therapy, with Yttrium-90 DOTA-Octreotate, was given in two cases, both of whom achieved disease stabilization for 9-12 months respectively. There was a significant difference between Stage 4 and Stage 1 disease at presentation and survival. In conclusion curative surgical resection is treatment of choice, however, chemotherapy, somatostatin analogues and peptide receptor radionuclide therapy offers palliation improving both symptoms and mortality.
Collapse
Affiliation(s)
- R Srirajaskanthan
- Neuroendocrine Tumour Unit, Royal Free Hospital, London NW3 2QG, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Van den Abbeele AD, Ertuk M. FDG-PET to Measure Response to Targeted Therapy: The Example of Gastrointestinal Stromal Tumor and Imatinib Mesylate (Gleevec). PET Clin 2008; 3:77-87. [PMID: 27158147 DOI: 10.1016/j.cpet.2008.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Imaging has played a major role in the evaluation of molecular targeted therapies in patients with gastrointestinal stromal tumors. FDG-PET has been instrumental in the evaluation of the tyrosine kinase inhibitors used to treat this disease. Imaging findings on FDG-PET and CT have illustrated the need to re-evaluate traditional response assessment criteria. Because most PET studies are now performed on hybrid PET/CT scanners, there is an opportunity to test and validate existing and new PET tracers and to provide anatomic and functional information in one setting. The inclusion of imaging in the testing and implementation of targeted therapies is helping to define the concept of personalized medicine.
Collapse
Affiliation(s)
- Annick D Van den Abbeele
- Harvard Medical School, Boston, MA, USA; Department of Radiology, Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115, USA; Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Tumor Imaging Metrics Core, Dana-Farber/Harvard Cancer Center, Boston, MA, USA.
| | - Mehmet Ertuk
- Department of Radiology, Sisli Etfal Training and Research Hospital, Şişli 34360, Istanbul, Turkey
| |
Collapse
|
50
|
Park MS, Klotz E, Kim MJ, Song SY, Park SW, Cha SW, Lim JS, Seong J, Chung JB, Kim KW. Perfusion CT: noninvasive surrogate marker for stratification of pancreatic cancer response to concurrent chemo- and radiation therapy. Radiology 2008; 250:110-7. [PMID: 18984781 DOI: 10.1148/radiol.2493080226] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
PURPOSE To prospectively determine whether perfusion computed tomography (CT) parameters, such as volume transfer constant (K(trans)) between blood plasma and extracellular extravascular space (EES) and blood volume calculated from dynamic CT data, can be used to predict response of pancreatic cancer to concurrent chemotherapy and radiation therapy (CCRT). MATERIALS AND METHODS This prospective study was institutional review board approved, and written informed consent was obtained. Thirty patients with pancreatic cancer underwent perfusion CT with 64-detector row CT before gemcitabine-based CCRT. Two perfusion parameters (K(trans) and blood volume) measured before treatment were compared between patients who responded to treatment and those who did not, as determined with World Health Organization criteria from first and second posttherapeutic follow-up CT examinations, which were performed at 3- and 6-month follow-up. Statistical analysis was performed with the two-sample t test. A receiver operating characteristic curve was used to determine the best cutoff value of perfusion parameters for differentiation of responders from nonresponders. RESULTS Twenty of 30 patients examined at 3-month follow-up responded to therapy. Their pretreatment K(trans) value was significantly higher than that of nonresponders (50.8 mL/100 mL/min +/- 30.5 [standard deviation] vs 19.0 mL/100 mL/min +/- 10.8, P = .001). The best cutoff value for differentiating between responders and nonresponders was 31.8 mL/100 mL/min, which yielded 75.0% sensitivity and 90.0% specificity. Ten of 18 patients examined at 6-month follow-up responded to therapy. Their pretreatment K(trans) value was significantly higher than that of nonresponders (58.6 mL/100 mL/min +/- 43.2 vs 19.8 mL/100 mL/min +/- 10.9, P = .002). Responders also had higher blood volume values, but this difference was not significant. CONCLUSION Tumors with a high pretreatment K(trans) value tended to respond better to CCRT than did tumors with a low pretreatment K(trans) value. Perfusion CT may be used to predict tumor response to CCRT in patients with pancreatic cancer. This might aid in development of a tailored approach to therapy in these patients.
Collapse
Affiliation(s)
- Mi-Suk Park
- Department of Diagnostic Radiology, Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seodaemun-ku, Shinchon-dong 134, Seoul 120-752, South Korea.
| | | | | | | | | | | | | | | | | | | |
Collapse
|