101
|
Nguyen QD, Challapalli A, Smith G, Fortt R, Aboagye EO. Imaging apoptosis with positron emission tomography: 'bench to bedside' development of the caspase-3/7 specific radiotracer [(18)F]ICMT-11. Eur J Cancer 2012; 48:432-40. [PMID: 22226480 DOI: 10.1016/j.ejca.2011.11.033] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 11/21/2011] [Indexed: 12/20/2022]
Abstract
The capacity to evade apoptosis has been defined as one of the hallmarks of cancer and, thus, effective anti-cancer therapy often induces apoptosis. A biomarker for imaging apoptosis could assist in monitoring the efficacy of a wide range of current and future therapeutics. Despite the potential, there are limited clinical examples of the use of positron emission tomography for imaging of apoptosis. [(18)F]ICMT-11 is a novel reagent designed to non-invasively image caspase-3 activation and, hence, drug-induced apoptosis. Radiochemistry development of [(18)F]ICMT-11 has been undertaken to improve specific radioactivity, reduce content of stable impurities, reduce synthesis time and enable automation for manufacture of multi-patient dose. Due to the promising mechanistic and safety profile of [(18)F]ICMT-11, the radiotracer is transitioning to clinical development and has been selected as a candidate radiotracer by the QuIC-ConCePT consortium for further evaluation in preclinical models and humans. A successful outcome will allow use of the radiotracer as qualified method for evaluating the pharmaceutical industry's next generation therapeutics.
Collapse
Affiliation(s)
- Quang-Dé Nguyen
- Department of Surgery and Cancer, Imperial College, London, UK
| | | | | | | | | |
Collapse
|
102
|
Kanai R, Rabkin SD, Yip S, Sgubin D, Zaupa CM, Hirose Y, Louis DN, Wakimoto H, Martuza RL. Oncolytic virus-mediated manipulation of DNA damage responses: synergy with chemotherapy in killing glioblastoma stem cells. J Natl Cancer Inst 2011; 104:42-55. [PMID: 22173583 DOI: 10.1093/jnci/djr509] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Although both the alkylating agent temozolomide (TMZ) and oncolytic viruses hold promise for treating glioblastoma, which remains uniformly lethal, the effectiveness of combining the two treatments and the mechanism of their interaction on cancer stem cells are unknown. METHODS We investigated the efficacy of combining TMZ and the oncolytic herpes simplex virus (oHSV) G47Δ in killing glioblastoma stem cells (GSCs), using Chou-Talalay combination index analysis, immunocytochemistry and fluorescence microscopy, and neutral comet assay. The role of treatment-induced DNA double-strand breaks, activation of DNA damage responses, and virus replication in the cytotoxic interaction between G47Δ and TMZ was examined with a panel of pharmacological inhibitors and short-hairpin RNA (shRNA)-mediated knockdown of DNA repair pathways. Comparisons of cell survival and virus replication were performed using a two-sided t test (unpaired). The survival of athymic mice (n = 6-8 mice per group) bearing GSC-derived glioblastoma tumors treated with the combination of G47Δ and TMZ was analyzed by the Kaplan-Meier method and evaluated with a two-sided log-rank test. RESULTS The combination of G47Δ and TMZ acted synergistically in killing GSCs but not neurons, with associated robust induction of DNA damage. Pharmacological and shRNA-mediated knockdown studies suggested that activated ataxia telangiectasia mutated (ATM) is a crucial mediator of synergy. Activated ATM relocalized to HSV DNA replication compartments where it likely enhanced oHSV replication and could not participate in repairing TMZ-induced DNA damage. Sensitivity to TMZ and synergy with G47Δ decreased with O(6)-methylguanine-DNA-methyltransferase (MGMT) expression and MSH6 knockdown. Combined G47Δ and TMZ treatment extended survival of mice bearing GSC-derived intracranial tumors, achieving long-term remission in four of eight mice (median survival = 228 days; G47Δ alone vs G47Δ + TMZ, hazard ratio of survival = 7.1, 95% confidence interval = 1.9 to 26.1, P = .003) at TMZ doses attainable in patients. CONCLUSIONS The combination of G47Δ and TMZ acts synergistically in killing GSCs through oHSV-mediated manipulation of DNA damage responses. This strategy is highly efficacious in representative preclinical models and warrants clinical translation.
Collapse
Affiliation(s)
- Ryuichi Kanai
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114 , USA
| | | | | | | | | | | | | | | | | |
Collapse
|
103
|
Krajewski KM, Giardino AA, Zukotynski K, Van den Abbeele AD, Pedrosa I. Imaging in renal cell carcinoma. Hematol Oncol Clin North Am 2011; 25:687-715. [PMID: 21763963 DOI: 10.1016/j.hoc.2011.04.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Imaging plays a central role in the detection, diagnosis, staging, and follow-up of renal cell carcinoma (RCC). Most renal masses are incidentally detected with modern, high-resolution imaging techniques and a variety of management options exist for the renal masses encountered today. This article discusses the role of multiple imaging modalities in the diagnosis of RCC and the imaging features of specific pathologic subtypes and staging techniques. Future directions in RCC imaging are presented, including dynamic contrast-enhanced and unenhanced techniques, as well as the development of novel tracers for positron emission tomography.
Collapse
Affiliation(s)
- Katherine M Krajewski
- Department of Imaging, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA.
| | | | | | | | | |
Collapse
|
104
|
Beier D, Schulz JB, Beier CP. Chemoresistance of glioblastoma cancer stem cells--much more complex than expected. Mol Cancer 2011; 10:128. [PMID: 21988793 PMCID: PMC3207925 DOI: 10.1186/1476-4598-10-128] [Citation(s) in RCA: 239] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2011] [Accepted: 10/11/2011] [Indexed: 12/14/2022] Open
Abstract
Glioblastomas (GBM) are a paradigm for the investigation of cancer stem cells (CSC) in solid malignancies. The susceptibility of GBM CSC to standard chemotherapeutic drugs is controversial as the existing literature presents conflicting experimental data. Here, we summarize the experimental evidence on the resistance of GBM CSC to alkylating chemotherapeutic agents, with a special focus on temozolomide (TMZ). The data suggests that CSC are neither resistant nor susceptible to chemotherapy per se. Detoxifying proteins such as O6-methylguanine-DNA-methyltransferase (MGMT) confer a strong intrinsic resistance to CSC in all studies. Extrinsic factors may also contribute to the resistance of CSC to TMZ. These may include TMZ concentrations in the brain parenchyma, TMZ dosing schemes, hypoxic microenvironments, niche factors, and the re-acquisition of stem cell properties by non-stem cells. Thus, the interaction of CSC and chemotherapy is more complex than may be expected and it is necessary to consider these factors in order to overcome chemoresistance in the patient.
Collapse
Affiliation(s)
- Dagmar Beier
- Department of Neurology, RWTH Aachen, Medical School, Pauwelsstrasse 30, 52074 Aachen, Germany
| | | | | |
Collapse
|
105
|
Non-invasive imaging in experimental medicine for drug development. Curr Opin Pharmacol 2011; 11:501-7. [DOI: 10.1016/j.coph.2011.04.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 04/17/2011] [Indexed: 01/01/2023]
|
106
|
Jakubowicz-Gil J, Langner E, Rzeski W. Kinetic studies of the effects of Temodal and quercetin on astrocytoma cells. Pharmacol Rep 2011; 63:403-16. [PMID: 21602595 DOI: 10.1016/s1734-1140(11)70506-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2010] [Revised: 10/08/2010] [Indexed: 01/24/2023]
Abstract
The aim of the present study was to investigate the kinetics of the effects exerted by Temodal and quercetin on the survival of the human astrocytoma MOGGCCM cell line. Our results indicate that quercetin was toxic and induced necrosis, whereas Temodal induced autophagy-mediated cell death most effectively. The amount of cell death directly correlated with drug concentration and length of exposure. During combined administration of both drugs, Temodal attenuated the cytotoxic effects of quercetin. Combinations of both drugs were effective in inducing programmed cell death, but the type of cell death was concentration-dependent. Co-administration of Temodal (100 μM) with a low quercetin concentration (5 μM) resulted in a very significant induction of autophagy; however, after treatment with quercetin at a higher concentration (30 μM), apoptosis became the primary mechanism of cell death. The sequence of drug administration was also important. The highest number of dead cells was observed after simultaneous administration of both drugs or after pre-incubation with Temodal followed by treatment with quercetin. Apoptosis was identified through activation of the mitochondrial pathway including cleavage of caspase-3 and release of cytochrome c. Autophagy was identified through increased levels of LC3II. Our results indicate that Temodal and quercetin are synergistic inducers of programmed cell death, better together than applied separately. This drug combination appears to be a potent and promising therapeutic relevant to the treatment of gliomas.
Collapse
Affiliation(s)
- Joanna Jakubowicz-Gil
- Department of Comparative Anatomy and Anthropology, Maria Curie-Sklodowska University, Akademicka 19, PL 20-033 Lublin, Poland.
| | | | | |
Collapse
|
107
|
Determination of temozolomide in serum and brain tumor with micellar electrokinetic capillary chromatography. J Chromatogr B Analyt Technol Biomed Life Sci 2011; 879:2229-33. [DOI: 10.1016/j.jchromb.2011.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 05/25/2011] [Accepted: 06/06/2011] [Indexed: 11/17/2022]
|
108
|
Delivery of molecularly targeted therapy to malignant glioma, a disease of the whole brain. Expert Rev Mol Med 2011; 13:e17. [PMID: 21676290 DOI: 10.1017/s1462399411001888] [Citation(s) in RCA: 217] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Glioblastoma multiforme, because of its invasive nature, can be considered a disease of the entire brain. Despite recent advances in surgery, radiotherapy and chemotherapy, current treatment regimens have only a marginal impact on patient survival. A crucial challenge is to deliver drugs effectively to invasive glioma cells residing in a sanctuary within the central nervous system. The blood-brain barrier (BBB) restricts the delivery of many small and large molecules into the brain. Drug delivery to the brain is further restricted by active efflux transporters present at the BBB. Current clinical assessment of drug delivery and hence efficacy is based on the measured drug levels in the bulk tumour mass that is usually removed by surgery. Mounting evidence suggests that the inevitable relapse and lethality of glioblastoma multiforme is due to a failure to effectively treat invasive glioma cells. These invasive cells hide in areas of the brain that are shielded by an intact BBB, where they continue to grow and give rise to the recurrent tumour. Effective delivery of chemotherapeutics to the invasive glioma cells is therefore critical, and long-term efficacy will depend on the ability of a molecularly targeted agent to penetrate an intact and functional BBB throughout the entire brain. This review highlights the various aspects of the BBB, and also the brain-tumour-cell barrier (a barrier due to expression of efflux transporters in tumour cells), that together can significantly influence drug response. It then discusses the challenge of glioma as a disease of the whole brain, which lends emphasis to the need to deliver drugs effectively across the BBB to reach both the central tumour and the invasive glioma cells.
Collapse
|
109
|
Greenhalgh AD, Ogungbenro K, Rothwell NJ, Galea JP. Translational pharmacokinetics: challenges of an emerging approach to drug development in stroke. Expert Opin Drug Metab Toxicol 2011; 7:681-95. [PMID: 21521135 DOI: 10.1517/17425255.2011.570259] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
INTRODUCTION There is increasing recognition of the importance of translational pharmacokinetics in stroke research, lack of which has been cited as one of the main contributing factors to failure of Phase III trials. AREAS COVERED The article reviews the translational issues in administration, distribution and sampling in the pharmacokinetics of putative therapeutic drugs in stroke. In addition, the role of translational pharmacometrics in drug development is discussed. The review uses the anti-inflammatory agent, IL-1 receptor antagonist, as an example. The reader will gain an insight into the pitfalls that are commonplace in translating pharmacokinetics from the preclinical to the clinical scenario. The reader will also gain an understanding of the complexities of blood-central nervous system (CNS) barriers in relation to brain pharmacokinetics and the increasing use of translational pharmacometrics in stroke research. EXPERT OPINION The translation of preclinical to clinical pharmacokinetics is a discipline that is traditionally overlooked and is likely to be a key factor responsible for failure of clinical trials. With a clear comprehensive insight into the benefits and limitations of translational pharmacokinetics in stroke, translational pharmacokinetics can be safely used to enhance the efficacy of clinical trials in stroke and their likelihood of success.
Collapse
Affiliation(s)
- Andrew D Greenhalgh
- Manchester Academic Health Sciences Centre (MAHSC), Faculty of Life Sciences, AV Hill Building, Oxford Road, Manchester M13 9PT, UK
| | | | | | | |
Collapse
|
110
|
Daniel RA, Rozanska AL, Mulligan EA, Drew Y, Thomas HD, Castelbuono DJ, Hostomsky Z, Plummer ER, Tweddle DA, Boddy AV, Clifford SC, Curtin NJ. Central nervous system penetration and enhancement of temozolomide activity in childhood medulloblastoma models by poly(ADP-ribose) polymerase inhibitor AG-014699. Br J Cancer 2010; 103:1588-96. [PMID: 20978505 PMCID: PMC2990587 DOI: 10.1038/sj.bjc.6605946] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 09/10/2010] [Accepted: 09/18/2010] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Temozolomide shows activity against medulloblastoma, the most common malignant paediatric brain tumour. Poly(ADP-ribose) polymerase (PARP) inhibitors enhance temozolomide activity in extracranial adult and paediatric human malignancies. METHODS We assessed the effect of AG-014699, a clinically active PARP inhibitor, on temozolomide-induced growth inhibition in human medulloblastoma models. Pharmacokinetic, pharmacodynamic and toxicity assays were performed in tumour-bearing mice. RESULTS Sensitivity to temozolomide in vitro was consistent with methylguanine methyltransferase (MGMT) and DNA mismatch repair (MMR) status; MGMT(+) MMR(+) D384Med cells (temozolomide GI(50)=220 μM), representative of most primary medulloblastomas, were sensitised fourfold by AG-014699; MGMT⁻ MMR(+) D425Med cells were hypersensitive (GI(50)=9 μM) and not sensitised by AG-014699, whereas MGMT(+) MMR⁻ temozolomide-resistant D283Med cells (GI₅₀=807 μM) were sensitised 20-fold. In xenograft models, co-administration of AG-014699 produced an increase in temozolomide-induced tumour growth delay in D384Med xenografts. Consistent with the in vitro data, temozolomide caused complete tumour regressions of D425Med xenografts, whereas D283Med xenografts were relatively resistant. AG-014699 was not toxic, accumulated and reduced PARP activity ≥75% in xenograft and brain tissues. CONCLUSION We show for the first time central nervous system penetration and inhibition of brain PARP activity by AG-014699. Taken together with our in vitro chemosensitisation and toxicity data, these findings support further evaluation of the clinical potential of AG-014699-temozolomide combinations in intra-cranial malignancies.
Collapse
Affiliation(s)
- R A Daniel
- Northern Institute for Cancer Research, Newcastle University, Paul O’Gorman Building, Newcastle upon Tyne, NE2 4HH, UK
| | - A L Rozanska
- Northern Institute for Cancer Research, Newcastle University, Paul O’Gorman Building, Newcastle upon Tyne, NE2 4HH, UK
| | - E A Mulligan
- Northern Institute for Cancer Research, Newcastle University, Paul O’Gorman Building, Newcastle upon Tyne, NE2 4HH, UK
| | - Y Drew
- Northern Institute for Cancer Research, Newcastle University, Paul O’Gorman Building, Newcastle upon Tyne, NE2 4HH, UK
| | - H D Thomas
- Northern Institute for Cancer Research, Newcastle University, Paul O’Gorman Building, Newcastle upon Tyne, NE2 4HH, UK
| | - D J Castelbuono
- Northern Institute for Cancer Research, Newcastle University, Paul O’Gorman Building, Newcastle upon Tyne, NE2 4HH, UK
| | | | - E R Plummer
- Northern Institute for Cancer Research, Newcastle University, Paul O’Gorman Building, Newcastle upon Tyne, NE2 4HH, UK
| | - D A Tweddle
- Northern Institute for Cancer Research, Newcastle University, Paul O’Gorman Building, Newcastle upon Tyne, NE2 4HH, UK
| | - A V Boddy
- Northern Institute for Cancer Research, Newcastle University, Paul O’Gorman Building, Newcastle upon Tyne, NE2 4HH, UK
| | - S C Clifford
- Northern Institute for Cancer Research, Newcastle University, Paul O’Gorman Building, Newcastle upon Tyne, NE2 4HH, UK
| | - N J Curtin
- Northern Institute for Cancer Research, Newcastle University, Paul O’Gorman Building, Newcastle upon Tyne, NE2 4HH, UK
| |
Collapse
|
111
|
Schreiber S, Prox-Vagedes V, Elolf E, Brueggemann I, Gademann G, Galazky I, Bartels C. Bilateral posterior RION after concomitant radiochemotherapy with temozolomide in a patient with glioblastoma multiforme: a case report. BMC Cancer 2010; 10:520. [PMID: 20920315 PMCID: PMC2958936 DOI: 10.1186/1471-2407-10-520] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Accepted: 10/01/2010] [Indexed: 11/10/2022] Open
Abstract
Background Radiation induced optic neuropathy (RION) is a rare but severe consequence of radiation therapy that is associated with adjuvant chemotherapy, specifically therapy with vincristine or nitrosoureas. However, there is very little evidence regarding the occurrence of RION after concomitant radiochemotherapy with temozolomide. Case Presentation The case of a 63 year old woman with glioblastoma multiforme and concomitant radiochemotherapy with temozolomide is described. Due to a slight depressive episode the patient also took hypericum perforatum. Five months after cessation of fractionated radiation and adjuvant chemotherapy with temozolomide (cumulative dose of 11040 mg) the patient developed bilateral amaurosis due to RION. Tumor regrowth was excluded by magnetic resonance imaging. After the application of gadolinium a pathognomonic contrast enhancement of both prechiasmatic optic nerves could be observed. Conclusions In this patient, the occurrence of RION may have been the result of radiosensitization by temozolomide, which could have been strengthened by hypericin. Consequently, physicians should avoid a concomitant application of hypericum perforatum and radiochemotherapy.
Collapse
Affiliation(s)
- Stefanie Schreiber
- Department of Neurology, Otto-von-Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany.
| | | | | | | | | | | | | |
Collapse
|
112
|
Andrasi M, Bustos R, Gaspar A, Gomez FA, Klekner A. Analysis and stability study of temozolomide using capillary electrophoresis. J Chromatogr B Analyt Technol Biomed Life Sci 2010; 878:1801-8. [DOI: 10.1016/j.jchromb.2010.05.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2009] [Revised: 05/04/2010] [Accepted: 05/04/2010] [Indexed: 11/25/2022]
|
113
|
Hühn E, Buchholz HG, Shazly G, Maus S, Thews O, Bausbacher N, Rösch F, Schreckenberger M, Langguth P. Predicting the in vivo release from a liposomal formulation by IVIVC and non-invasive positron emission tomography imaging. Eur J Pharm Sci 2010; 41:71-7. [PMID: 20566325 DOI: 10.1016/j.ejps.2010.05.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 05/12/2010] [Accepted: 05/30/2010] [Indexed: 11/26/2022]
Abstract
This study aimed to predict the in vivo performance from the in vitro release of a low-molecular weight model compound, [(18)F]-2-fluoro-2-deoxy-d-glucose ([(18)F]FDG), from liposomes and by means of positron emission tomography (PET). Liposomes composed of hydrogenated phosphatidylcholine (HPC) were prepared by a freeze-thaw method. Particle size distribution was measured by dynamic light scattering (DLS). In vitro release was examined with a dispersion method detecting the radioactivity of [(18)F]FDG. In vivo release of [(18)F]FDG, following i.p. injection of the liposomes in rats, was determined by using a Micro-PET scanner. Convolution was performed to predict the in vivo profiles from the in vitro data and to establish an in vitro-in vivo correlation (IVIVC). The in vivo predictions slightly underestimated the experimentally determined values. The magnitude of the prediction errors (13% and 19%) displayed a satisfactory IVIV relationship leaving yet room for further improvement. This study demonstrated for the first time the use of PET in attaining an IVIVC for a parenterally administered modified release dosage form. It is therefore possible to predict target tissue concentrations, e.g., in the brain, from in vitro release experiments. IVIVC using non-invasive PET imaging could thus be a valuable tool in drug formulation development, resulting in reduced animal testing.
Collapse
Affiliation(s)
- Eva Hühn
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Staudinger Weg 5, 55099 Mainz, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
114
|
van der Veldt AAM, Meijerink MR, van den Eertwegh AJM, Boven E. Targeted therapies in renal cell cancer: recent developments in imaging. Target Oncol 2010; 5:95-112. [PMID: 20625845 PMCID: PMC2929340 DOI: 10.1007/s11523-010-0146-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Accepted: 06/24/2010] [Indexed: 01/14/2023]
Abstract
Targeted therapy has significantly improved the perspectives of patients with metastatic renal cell cancer (mRCC). Frequently, these new molecules cause disease stabilization rather than substantial tumor regression. As treatment options expand with the growing number of targeted agents, there is an increasing need for surrogate markers to early assess tumor response. Here, we review the currently available imaging techniques and response evaluation criteria for the assessment of tumor response in mRCC patients. For computed tomography (CT), different criteria are discussed including the Response Evaluation Criteria in Solid Tumors (RECIST), the Choi criteria, the modified Choi criteria, and the size and attenuation CT (SACT) criteria. Functional imaging modalities are discussed, such as dynamic contrast-enhanced CT (DCE-CT), dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), dynamic contrast-enhanced ultrasonography (DCE-US), and positron emission tomography (PET).
Collapse
Affiliation(s)
- Astrid A M van der Veldt
- Department of Nuclear Medicine & PET Research, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
115
|
|
116
|
Shidahara M, Ito H, Otsuka T, Ikoma Y, Arakawa R, Kodaka F, Seki C, Takano H, Takahashi H, Turkheimer FE, Kimura Y, Kanno I, Suhara T. Measurement error analysis for the determination of dopamine D(2) receptor occupancy using the agonist radioligand [(11)C]MNPA. J Cereb Blood Flow Metab 2010; 30:187-95. [PMID: 19756020 PMCID: PMC2949103 DOI: 10.1038/jcbfm.2009.193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The purpose of this study is to investigate errors in quantitative analysis for estimating dopamine D(2) receptor occupancy of antipsychotics with agonist radioligand [(11)C]MNPA by numerical simulation, with particular attention to the validity of a quantitative approach based on the use of a reference region. Synthetic data were validated using clinical data combined with a bootstrap approach. Time-activity curves (TACs) of [(11)C]MNPA were simulated, and the reliability of binding potential (BP(ND)) and occupancy estimated by nonlinear least square (NLS) fitting and a simplified reference tissue model (SRTM) were investigated for various noise levels and scan durations. In the human positron emission tomography (PET) study with and without antipsychotic, risperidone, the uncertainty of BP(ND) and occupancy estimated by SRTM was investigated using resampled TACs based on bootstrap approach with weighted residual errors of fitting. For both NLS and SRTM, it was possible to have <3% of bias in occupancy estimates of [(11)C]MNPA by 60 mins. However, shortened scan duration degrades the quantification of very small binding potentials, especially in case of SRTM. Observations were replicated on the clinical data. Results showed that dopamine D(2) receptor occupancy by antipsychotics can be estimated precisely in region of interest analysis by SRTM with a longer than 60-min [(11)C]MNPA PET scan duration.
Collapse
Affiliation(s)
- Miho Shidahara
- Biophysics Group, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Portnow J, Badie B, Chen M, Liu A, Blanchard S, Synold TW. The neuropharmacokinetics of temozolomide in patients with resectable brain tumors: potential implications for the current approach to chemoradiation. Clin Cancer Res 2009; 15:7092-8. [PMID: 19861433 DOI: 10.1158/1078-0432.ccr-09-1349] [Citation(s) in RCA: 175] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Intracerebral microdialysis (ICMD) is an accepted method for monitoring changes in neurochemistry from acute brain injury. The goal of this pilot study was to determine the feasibility of using ICMD to examine the neuropharmacokinetics of temozolomide in brain interstitium following oral administration. EXPERIMENTAL DESIGN Patients with primary or metastatic brain tumors had a microdialysis catheter placed in peritumoral brain tissue at the time of surgical debulking. Computerized tomography scan confirmed the catheter location. Patients received a single oral dose of temozolomide (150 mg/m2) on the first postoperative day, serial plasma and ICMD samples were collected over 24 hours, and temozolomide concentrations were determined by tandem mass spectrometry. RESULTS Nine patients were enrolled. Dialysate and plasma samples were successfully collected from seven of the nine patients. The mean temozolomide areas under the concentration-time curve (AUC) in plasma and brain interstitium were 17.1 and 2.7 microg/mL x hour, with an average brain interstitium/plasma AUC ratio of 17.8%. The mean peak temozolomide concentration in the brain was 0.6 +/- 0.3 microg/mL, and the mean time to reach peak level in brain was 2.0 +/- 0.8 hours. CONCLUSIONS The use of ICMD to measure the neuropharmacokinetics of systemically administered chemotherapy is safe and feasible. Concentrations of temozolomide in brain interstitium obtained by ICMD are consistent with published data obtained in a preclinical ICMD model, as well as from clinical studies of cerebrospinal fluid. However, the delayed time required to achieve maximum temozolomide concentrations in brain suggests that current chemoradiation regimens may be improved by administering temozolomide 2 to 3 hours before radiation.
Collapse
Affiliation(s)
- Jana Portnow
- Department of Medical Oncology and Experimental Therapeutics, 1500 East Duarte Road, Duarte, CA 91010, USA.
| | | | | | | | | | | |
Collapse
|