During my active career my research has focused on two main areas. The first is on the effects of radiation on normal tissues, particularly the central nervous system and the various factors that determine the tolerance of the spinal cord. The spinal cord is probably the most critical normal tissue in radiation oncology, and my research has largely contributed to optimizing treatment of cancer close to the spine and preventing the development of debilitating complications. In addition to addressing important questions related to clinical radiation treatments, this research has elucidated the mechanisms and cellular basis of various radiation-induced changes in the CNS. My other research has aimed at overcoming radiation resistance mechanisms related to the tumor microenvironment, which are known to be mostly tumor hypoxia and proliferation. To study these aspects in a clinically relevant setting, my lab has developed a panel of patient-derived tumor xenografts that reflect the clinical heterogeneity of individual patients. The overall aim of this research has been to test new therapeutic modalities and develop assays to individualize treatment based on tumor biopsies and molecular imaging. Since my retirement in the Netherlands, I have had a part-time appointment in the UW Department of Human Oncology, where I consult and collaborate on various research projects and teach radiobiology courses for medical physics undergraduates and graduates and radiation oncology residents.
Education
PhD, University of Amsterdam, Radiation Biology (1979)
MS, Free University of Amsterdam, Zoology and Radiation Biology (1971)
Academic Appointments
Professor, Human Oncology (2011–pres.)
Professor and Head of Laboratory (1987-2011), Radiation Oncology, Radboud University Nijmegen (1987–2011)
Associate Professor, University of New Mexico–Albuquerque (and Los Alamos National Laboratory) (1984–1987)
Research Associate, Radiobiological Institute TNO, Rijswijk, Netherlands (1972–1984)
Selected Honors and Awards
ESTRO Lifetime Achievement Award (2013)
ESTRO Honorary Physicist Award (2011)
ICRU Gray Medal (2009)
Honorary member, Polish Society of Radiation Oncology (2004)
Bacq & Alexander Award, European Society of Radiation Biology (2002)
John S. Laughlin Visiting Professor, Memorial Sloan Kettering Cancer Center, New York (2002)
Emmanuel van der Schueren Award, Belgian Society of Radiation Oncology, Leuven, Belgium (2001)
Charles Botstein Memorial Visiting Professor, Albert Einstein College of Medicine and Montefiore Hospital, NY (1997) (1997)
ESTRO Gold Medal, 13th Annual meeting of the European Society of Therapeutic Radiology and Oncology (1994)
Research Focus
Effects of Radiation on Normal Tissue, Particularly the Central Nervous System, Tumor Microenvironment
Dr. Albert van der Kogel is a cancer biologist with expertise on the effects of radiation on normal tissues and radiation resistance mechanisms. He consults and collaborates on various research projects. He also teaches radiobiology courses for medical physics undergraduates, graduates and radiation oncology residents.
-
Are hybrid conferences the new standard? Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology
Baumann M, Bacchus C, Aznar MC, Coppes RP, Deutsch E, Georg D, Haustermans K, Hoskin P, Krause M, Lartigau EF, Lee WM, Löck S, Offersen BV, Overgaard J, Thwaites DI, Kogel vd, Heide vd, Valentini V
2023 Jul;184:109695. doi: 10.1016/j.radonc.2023.109695. Epub 2023 May 6.
-
The role of ESTRO guidelines in achieving consistency and quality in clinical radiation oncology practice Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology
Offersen BV, Aznar MC, Bacchus C, Coppes RP, Deutsch E, Georg D, Haustermans K, Hoskin P, Krause M, Lartigau EF, Lee WM, Löck S, Thwaites DI, Kogel vd, Heide vd, Valentini V, Overgaard J, Baumann M
2023 Feb;179:109446. doi: 10.1016/j.radonc.2022.109446. Epub 2022 Dec 23.
-
In Reply to Goyal and Madan International journal of radiation oncology, biology, physics
Medin P, Hrycushko B, Kogel vd
2022 May 1;113(1):232-233. doi: 10.1016/j.ijrobp.2022.01.042.
-
Personalised radiation therapy taking both the tumour and patient into consideration Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology
Overgaard J, Aznar MC, Bacchus C, Coppes RP, Deutsch E, Georg D, Haustermans K, Hoskin P, Krause M, Lartigau EF, Lee WM, Löck S, Offersen BV, Thwaites DI, Kogel vd, Heide vd, Valentini V, Baumann M
2022 Jan;166:A1-A5. doi: 10.1016/j.radonc.2022.01.010. Epub 2022 Jan 17.
-
Establishing the Gastroduodenal Maximum Tolerated Dose for Ablative 5-Fraction Stereotactic MR-guided Online Adaptive Radiation Therapy (SMART) in a Novel Swine Model International journal of radiation oncology, biology, physics
Mittauer KE, Bassetti MF, Meudt JJ, Graham M, Wood MW, Miller J, Lawless M, Frank JM, Ward R, Kogel VD, Shanmuganayagam D, Bayouth J
2021 Nov 1;111(3S):S141-S142. doi: 10.1016/j.ijrobp.2021.07.319.
-
More
PURPOSE/OBJECTIVE(S): Daily variations in abdominal anatomy introduce significant uncertainty in estimating the maximum tolerated dose (MTD). We investigated the gastroduodenal toxicity using stereotactic MR-guided online adaptive radiotherapy (SMART) to precisely control the dose to a novel swine model that closely matches human physiology and can be maintained at 80 kg. We hypothesized for a cumulative SMART course of 50 Gy in 5 fractions to the pancreas, the duodenum MTD is significantly > 37 Gy to 0.5 cc (D0.5cc).
MATERIALS/METHODS: A total of 18 WMSTM were included at 3 duodenal dose levels: 37 Gy (n = 12), 43.5 Gy (n = 3), and 50 Gy (n = 3) in 5 fractions. We defined MTD as: > 10% (≥2 of 12) WMSTM present with dose limiting toxicity (DLT). A DLT was defined as ≥ Grade 3 non-hematologic toxicity due to RT per NCI CTCAE v4.0. All WMSTM underwent SMART on a clinical MR-guided radiotherapy (MRgRT) LINAC, under ventilation at breath hold (BH). Plans were IMRT and prescribed 50 Gy/5 fractions to pancreatic head, while maintaining uniform dose level (37, 43.5, 50 Gy) to duodenal circumference at 3 cm inferior to pylorus sphincter, typical uniform irradiated range 3-9 cc based on duodenal diameter. Circumference approach was used to eliminate underestimation of cumulative D0.5cc. Duodenal hotspot was 1.03% of the dose level (37, 43.5, 50 Gy) to 2 cc, 1.05% to 0.1 cc. Gated BH delivery was performed with sagittal plane tracking of the duodenum at 4 frames per second. Toxicity monitoring occurred for 30 days post-RT. Fecal occult blood test (FOBT) was done during RT and toxicity monitoring. Endoscopy occurred 14 days post-RT. Gross pathology was obtained at time of necropsy, 30 days post-RT.
RESULTS: For the 37 Gy level, 1 of 12 WMSTM presented with DLT-pinpoint ulcer with surface damage on gross pathology (30 days post-RT). No DLT for 2 of 3 WMSTM at the 43.5 Gy level. One WMSTM at the 43.5 Gy level perforated on endoscopy, potentially due duodenal wall degradation and unknown RT-induced DLT on gross exam of emergency necropsy. Endoscopy was eliminated for the 50 Gy level. DLT was found for 2 of 3 WMSTM at the 50 Gy level upon gross pathology at necropsy, with each subject having 2-3 visible ulcerations at level of RT. One WMSTM at the 50 Gy level had duodenal pre-ulcer erosion at necropsy. All three WMSTM at 50 Gy level had visibly damaged papilla at necropsy. Another WMSTM at the 50 Gy level had common bile duct blockage, gall bladder obstruction, and jaundice at necropsy. FOBT were negative for all 18 WMSTM at 14 days post-RT.
CONCLUSION: A duodenum D0.5cc of 37 Gy in 5 fractions was found to be safe in a swine model for a 50 Gy SMART course to the pancreas, showing the duodenal MTD is > 37 Gy - an increase from the RTOG 1112 limit (i.e., 30 Gy). Our results translate to a recommended MTD in human subjects and can potentially allow for greater tumor dose escalation and survival outcomes in future pancreatic patients. We are currently pursuing an expansion cohort to evaluate the MTD at 43.5 Gy.
PMID:34700484 | DOI:10.1016/j.ijrobp.2021.07.319
View details for PubMedID 34700484
-
More
-
Brachial Plexus Tolerance to Single-Session SABR in a Pig Model International journal of radiation oncology, biology, physics
Hrycushko B, Kogel vd, Phillips L, Chhabra A, Folkert MR, Sayre JW, Vernino S, Hassan-Rezaeian N, Yamada Y, Timmerman R, Medin PM
2022 Feb 1;112(2):565-571. doi: 10.1016/j.ijrobp.2021.09.029. Epub 2021 Sep 28.
-
More
PURPOSE: The single-session dose tolerance of the spinal nerves has been observed to be similar to that of the spinal cord in pigs, counter to the perception that peripheral nerves are more tolerant to radiation. This pilot study aims to obtain a first impression of the single-session dose-response of the brachial plexus using pigs as a model.
METHODS AND MATERIALS: Ten Yucatan minipigs underwent computed tomography and magnetic resonance imaging for treatment planning, followed by single-session stereotactic ablative radiotherapy. A 2.5-cm length of the left-sided brachial plexus cords was irradiated. Pigs were distributed in 3 groups with prescription doses of 16 (n = 3), 19 (n = 4), and 22 Gy (n = 3). Neurologic status was assessed by observation for changes in gait and electrodiagnostic examination. Histopathologic examination was performed with light microscopy of paraffin-embedded sections stained with Luxol fast blue/periodic acid-Schiff and Masson's trichrome.
RESULTS: Seven of the 10 pigs developed motor deficit to the front limb of the irradiated side, with a latency from 5 to 8 weeks after irradiation. Probit analysis of the maximum nerve dose yields an estimated ED50 of 19.3 Gy for neurologic deficit, but the number of animals was insufficient to estimate 95% confidence intervals. No motor deficits were observed at a maximum dose of 17.6 Gy for any pig. Nerve conduction studies showed an absence of sensory response in all responders and absent or low motor response in most of the responders (71%). All symptomatic pigs showed histologic lesions to the left-sided plexus consistent with radiation-induced neuropathy.
CONCLUSIONS: The single-session ED50 for symptomatic plexopathy in Yucatan minipigs after irradiation of a 2.5-cm length of the brachial plexus cords was determined to be 19.3 Gy. The dose-response curve overlaps that of the spinal nerves and the spinal cord in the same animal model. The relationship between the brachial plexus tolerance in pigs and humans is unknown, and caution is warranted when extrapolating for clinical use.
PMID:34597718 | DOI:10.1016/j.ijrobp.2021.09.029
View details for PubMedID 34597718
-
More
-
In Silico Single-Fraction Stereotactic Ablative Radiation Therapy for the Treatment of Thoracic and Abdominal Oligometastatic Disease With Online Adaptive Magnetic Resonance Guidance Advances in radiation oncology
Lee S, Yadav P, Kogel vd, Bayouth J, Bassetti MF
2021 Jan 20;6(3):100652. doi: 10.1016/j.adro.2021.100652. eCollection 2021 May-Jun.
-
More
PURPOSE: Although SABR can improve oncologic outcomes for patients with oligometastatic disease, treatment of metastases near critical organs remains challenging. The purpose of this study is to determine the dosimetric feasibility of delivering magnetic resonance imaging (MRI)-guided adaptive SABR in a single fraction for abdominal and thoracic metastases.
METHODS AND MATERIALS: Previously delivered MRI-guided radiation therapy plans for 20 patients with oligometastatic disease in the thorax or abdomen, with 70% (14/20) of the lesions within 8 mm from dose-limiting organs at risk (OARs), were used to simulate the delivery of 24 Gy in a single fraction. Planning objectives included planning target volume (PTV) V95% >90%, optimized PTV (PTVopt) V95% >90%, and PTVopt D99% >20 Gy with no OAR dose violations, where PTVopt removed overlap with nearby planning organ at risk volume (PRV). Single-fraction plans were simulated on the first 5 daily setup breath-hold MRI scans, and the plans were reoptimized to consider variations in setup position and anatomy.
RESULTS: The mean PTV V95% for single-fraction SABR plans was lower compared with multifraction plans (mean 85.4% vs 92.6%, P = .02), but mean PTVopt V95% was not different (95.3% vs 98.2%, P = .62). After reoptimization of the single-fraction plan to the treatment day MRI, there was an increase in mean PTV V95% (85.0% vs 88.1%, P = .05), increase in mean PTVopt V95% (92.7% vs 96.3%, P = .02), increase in mean PTVopt D99% (19.7 Gy vs 23.8 Gy, P < .01), increase in mean frequency of meeting PTV D99% >20 Gy (52% vs 87%, P < .01), and increase in mean gross tumor volume minimum dose (17.5 Gy vs 19.3 Gy, P < .01). Reoptimization decreased mean frequency of OAR dose constraint violation (48% vs 0%, P < .01).
CONCLUSIONS: Single-fraction MRI-guided SABR is a dosimetrically feasible treatment for oligometastases that allows for on-table adaptation to avoid OAR dose constraint violations, but this method requires clinical validation.
PMID:34195490 | PMC:PMC8233469 | DOI:10.1016/j.adro.2021.100652
View details for PubMedID 34195490
-
More
-
Radiation oncology in the new virtual and digital era Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology
Aznar MC, Bacchus C, Coppes RP, Deutsch E, Georg D, Haustermans K, Hoskin P, Krause M, Lartigau EF, Löck S, Offersen B, Overgaard J, Thwaites DI, Kogel vd, Heide vd, Valentini V, Baumann M
2021 Jan;154:A1-A4. doi: 10.1016/j.radonc.2020.12.031. Epub 2020 Dec 30.
-
More
PMID:33387585 | PMC:PMC7773315 | DOI:10.1016/j.radonc.2020.12.031
View details for PubMedID 33387585
-
More
-
Existence of a Dose-Length Effect in Spinal Nerves Receiving Single-Session Stereotactic Ablative Radiation Therapy International journal of radiation oncology, biology, physics
Hrycushko B, Kogel vd, Phillips L, Folkert M, Sayre JW, Vernino S, Hassan-Rezaeian N, Foster RD, Yamada Y, Timmerman R, Medin PM
2020 Apr 1;106(5):1010-1016. doi: 10.1016/j.ijrobp.2019.11.417. Epub 2020 Jan 15.
-
More
PURPOSE: The spinal nerves have been observed to have a similar single-session dose tolerance to that of the spinal cord in pigs. Small-animal studies have shown that spinal cord dose tolerance depends on the length irradiated. This work aims to determine whether a dose-length effect exists for spinal nerves.
METHODS AND MATERIALS: Twenty-seven Yucatan minipigs underwent computed tomography and magnetic resonance imaging for treatment planning, followed by single-session stereotactic ablative radiation therapy. A 0.5 cm length of the left-sided C6, C7, and C8 spinal nerves was targeted. The pigs were distributed into 6 groups with prescription doses of 16 Gy (n = 5), 18 Gy (n = 5), 20 Gy (n = 5), 22 Gy (n = 5), 24 Gy (n = 5), or 36 Gy (n = 2) and corresponding maximum doses of 16.7, 19.1, 21.3, 23.1, 25.5, and 38.6 Gy, respectively. Neurologic status was assessed with a serial electrodiagnostic examination and daily observation of gait for approximately 52 weeks. A histopathologic examination of paraffin-embedded sections with Luxol fast blue/periodic acid-Schiff's staining was also performed.
RESULTS: Marked gait change was observed in 8 of 27 irradiated pigs. The latency for responding pigs was 11 to 16 weeks after irradiation. The affected animals presented with a limp in the left front limb, and 62.5% of these pigs had electrodiagnostic evidence of denervation in the C6 and C7 innervated muscles. A probit analysis showed the dose associated with a 50% incidence of gait change is 23.9 Gy (95% confidence interval, 22.5-25.8 Gy), which is 20% higher than that reported in a companion study where a 1.5 cm length was irradiated. All symptomatic pigs had demyelination and fibrosis in the irradiated nerves, but the contralateral nerves and spinal cord were normal.
CONCLUSIONS: A dose-length effect was observed for single-session irradiation of the spinal nerves in a Yucatan minipig model.
PMID:31953062 | DOI:10.1016/j.ijrobp.2019.11.417
View details for PubMedID 31953062
-
More
-
Spinal Nerve Tolerance to Single-Session Stereotactic Ablative Radiation Therapy International journal of radiation oncology, biology, physics
Hrycushko B, Kogel vd, Phillips L, Folkert MR, Sayre JW, Vernino S, Hassan-Rezaeian N, Foster RD, Yamada Y, Timmerman R, Medin PM
2019 Jul 15;104(4):845-851. doi: 10.1016/j.ijrobp.2019.03.044. Epub 2019 Apr 4.
-
More
PURPOSE: This study was performed to determine the dose-related incidence of neuropathy from single-session irradiation of the C6-C8 spinal nerves using a pig model and to test the hypothesis that the spinal nerves and spinal cord have the same tolerance to full cross-sectional irradiation.
METHODS AND MATERIALS: Twenty-five Yucatan minipigs received study treatment. Each animal underwent computed tomography and magnetic resonance imaging for treatment planning, followed by single-session stereotactic ablative radiation therapy. A 1.5-cm length of the left-sided C6, C7, and C8 spinal nerves was targeted. Pigs were distributed into 5 groups with prescription doses of 16 (n = 7), 18 (5), 20 (5), 22 (5), or 24 (3) Gy with corresponding maximum nerve doses of 17.3, 19.5, 21.6, 24.1, and 26.2 Gy. The neurologic status of all animals was followed for approximately 52 weeks by serial electrodiagnostic examination and daily observation of gait. Histopathologic examination of paraffin-embedded sections with Luxol fast blue/periodic acid-Schiff staining was performed on bilateral spinal nerves and the spinal cord.
RESULTS: Marked gait change was observed in 15 of the 25 irradiated pigs. Affected animals presented with a limp in their left front limb, and electromyography demonstrated evidence of denervation in C6 and C7 innervated muscles. Probit analysis showed the ED50 for gait change after irradiation of the spinal nerves to be 19.7 Gy (95% confidence interval, 18.5-21.1). The latency for all responding pigs was 9 to 15 weeks after irradiation. All symptomatic pigs had demyelination and fibrosis in their irradiated nerves, whereas contralateral nerves and spinal cord were normal.
CONCLUSIONS: The ED50 for symptomatic neuropathy after full cross-sectional irradiation of the spinal nerves was found to be 19.7 Gy. The dose response of the C6-C8 spinal nerves is not significantly different from that of full cross-sectional irradiation of the spinal cord as observed in companion studies.
PMID:30953713 | DOI:10.1016/j.ijrobp.2019.03.044
View details for PubMedID 30953713
-
More
-
Three discipline collaborative radiation therapy (3DCRT) special debate: The United States should build additional proton therapy facilities Journal of applied clinical medical physics
Braunstein S, Wang L, Newhauser W, Tenenholz T, Rong Y, Kogel vd, Dominello M, Joiner MC, Burmeister J
2019 Feb;20(2):7-12. doi: 10.1002/acm2.12537. Epub 2019 Feb 8.
-
Corrigendum: Potential role of the glycolytic oscillator in acute hypoxia in tumors (2015 <em>Phys. Med. Biol.</em> 60 9215) Physics in medicine and biology
Fru LC, Adamson EB, Campos DD, Fain SB, Jacques SL, Kogel vd, Nickel KP, Song C, Kimple RJ, Kissick MW
2018 Jun 6. doi: 10.1088/1361-6560/aacaba. Online ahead of print.
-
More
At the time of publication, our group had performed short tandem repeat (STR) testing on the SCC22B cell line and believed that had been correctly identified. As part of a recent comprehensive process to confirm the identity of cell lines in use in our lab, we repeated STR testing on all cell lines. These results were compared to the ExPASy Cellosaurus database (http://web.expasy.org/cellosaurus/). One cell line used in this manuscript was a near perfect match for T24 (CVCL_0554), a bladder carcinoma cell line commonly found as a cellular contaminant. Although we are unable to test the exact cells used in this manuscript, we believe that the cells labeled as SCC22B are most likely to actually be T24. The authors believe that neither the results nor the conclusions have been significantly changed on the basis of the specific cell line utilized.
PMID:29873307 | DOI:10.1088/1361-6560/aacaba
View details for PubMedID 29873307
-
More
-
SU-E-T-06: A Mathematical Explanation to Tumor's Response to Perfusion and Hypoxic Fraction after Radiation Medical physics
Yan Y, Kissick M, Bussink J, Jacques S, Kogel vd, Campos D, Zhao D
2012 Jun;39(6Part9):3703. doi: 10.1118/1.4735060.
-
More
PURPOSE: To develop a dynamic model that explains oxygen dynamics between the microvascular perfusion and the hypoxic cell population inside a tumor.
METHODS: Bussink et al (Radiat Res 153(4), p.398 (2000)) observed fast oxygen dynamics, faster than cell-death. Based on a simplified three-compartment-model: the microvasculature, well-oxygenated, and hypoxic tumor cell populations. We applied a first-order differential model for the tumor's transient response as a function of oxygen content within the blood vessels. The sink terms in our model for each compartment are fast changing parameters because radiation rapidly changes the oxygen consumption of the tumor cell in a time scale which is much faster than the population changes of the tumor. Transportation balance condition is also applied for each compartment.
RESULTS: Our simulation results can explain the experimental data in Bussink et al's (Radiat Res 153(4), p.398 (2000)) paper. We provide an explanation for the relative complex behavior of the microvascular perfusion after radiation that emphasizes the role of dynamic metabolic changes in addition to population changes.
CONCLUSIONS: A newly developed dynamic model leads our understanding to the interrelationship between microvascular oxygen content within the blood vessels and the hypoxia state of the tumor to a deeper level, which has the potential to provide the theoretical foundation for the patient' specific adaptive radiotherapy.
PMID:28519053 | DOI:10.1118/1.4735060
View details for PubMedID 28519053
-
More
-
SU-E-J-196: In-Vivo Tumor Blood-Oxygen Content Measurement via Interstitial Optical Transmission Spectroscopy Medical physics
Campos D, Yan Y, Zhao D, Kissick M, Jacques S, Kogel vd, Kimple R
2012 Jun;39(6Part9):3698. doi: 10.1118/1.4735037.
-
More
PURPOSE: To demonstrate the application of in-vivo diffuse optical transmission spectroscopy in quantifying oxygen saturation in interstitial tissue, and to use this technique to examine reoxygenation dynamics in real-time as tumors responds to radiotherapy.
METHODS: Two 200 micron core fiber optics were threaded through two 21 gauge hypodermic needles: one coupled to an OceanOptics QE65000 spectrometer, and the other to an Ocean Optics HL-2000-HP 20W light source. These needles were fixed approximately 3 mm apart, and inserted into nude mice with human head- and-neck tumor xenografts. The oxygen saturation was then measured as a function of time after irradiation at intervals of 0.5, 1, 2, 6, 12, and 24, to measure the tumors' prompt oxygen saturation response to radiation.
RESULTS: Blood volume, deoxy and oxy-hemoglobin concentrations were measured through least-squares fitting of transmission spectra. Furthermore, various configurations of interstitial fiber optic probes were explored to optimize signal strength. Improvement of the optical coupling to the biological system and a concurrent increase in source intensity are the main two focuses for boosting signal strength.
CONCLUSIONS: This work has the potential to give an understanding of the time-scales of hypoxia and reoxygenation in vivo as tumors respond to radiation injury. This technique is of particular interest for hypofractionated therapies particularly treatments of only two or three treatments, where optimizing treatment timing can increase the tumorcidal effect of the remaining fractions.
PMID:28519036 | DOI:10.1118/1.4735037
View details for PubMedID 28519036
-
More
-
SU-E-J-197: A Novel Optical Interstitial Fiber Spectroscopic System for Real-Time Tissue Micro-Vascular Hemodynamics Monitoring Medical physics
Zhao D, Campos D, Yan Y, Kimple R, Jacques S, Kogel vd, Kissick M
2012 Jun;39(6Part9):3698. doi: 10.1118/1.4735038.
-
More
PURPOSE: To demonstrate a novel interstitial optical fiber spectroscopic system, based on diffuse optical spectroscopies with spectral fitting, for the simultaneous monitoring of tumor blood volume and oxygen tension. The technique provides real-time, minimally-invasive and quantification of tissue micro-vascular hemodynamics.
METHODS: An optical fiber prototype probe characterizesthe optical transport in tissue between two large Numerical Aperture (NA) fibers of 200μm core diameter (BFH37-200, ThorLabs) spaced 3-mm apart. Two 21-Ga medical needles are used to protect fiber ends and to facilitate tissue penetration with minimum local blunt trauma in nude mice with xenografts. A 20W white light source (HL-2000-HP, Ocean Optics) is coupled to one fiber with SMA adapter. The other fiber is used to collect light, which is coupled into the spectrometer (QE65000 with Spectrasuite Operating software and OmniDriver, Ocean Optics). The wavelength response of the probe depends on the wavelength dependence of the light source, and of the light signal collection that includes considerable scatter, modeled with Monte-Carlo techniques (S. Jacques 2010 J. of Innov. Opt. Health Sci. 2 123-9). Measured spectra of tissue are normalized by a measured spectrum of a white standard, yielding the transmission spectrum. A head-and-neck xenograft on the flank of a live mouse is used for development.
RESULTS: The optical fiber probe delivers and collects light at an arbitrary depth in the tumor. By spectral fitting of the measured transmission spectrum, an analysis of blood volume and oxygen tension is obtained from the fitting parameters in real time.
CONCLUSIONS: A newly developed optical fiber spectroscopic system with an optical fiber probe takes spectroscopic techniques to a much deeper level in a tumor, which has potential applications for real-time monitoring hypoxic cell population dynamics for an eventual adaptive therapy metric of particular use in hypofractionated radiotherapy.
PMID:28519022 | DOI:10.1118/1.4735038
View details for PubMedID 28519022
-
More
-
Obituary and Tribute to John "Jack" Francis Fowler, PhD, DSc (1925-2016) International journal of radiation oncology, biology, physics
Harari PM, Ritter MA, Kogel vd
2017 Apr 1;97(5):886-888. doi: 10.1016/j.ijrobp.2017.01.015. Epub 2017 Jan 10.
-
Radiation Promptly Alters Cancer Live Cell Metabolic Fluxes: An In Vitro Demonstration Radiation research
Campos D, Peeters W, Nickel K, Burkel B, Bussink J, Kimple RJ, Kogel vd, Eliceiri KW, Kissick MW
2016 May;185(5):496-504. doi: 10.1667/RR14093.1. Epub 2016 Apr 29.
-
More
Quantitative data is presented that shows significant changes in cellular metabolism in a head and neck cancer cell line 30 min after irradiation. A head and neck cancer cell line (UM-SCC-22B) and a comparable normal cell line, normal oral keratinocyte (NOK) were each separately exposed to 10 Gy and treated with a control drug for disrupting metabolism (potassium cyanide; KCN). The metabolic changes were measured live by fluorescence lifetime imaging of the intrinsically fluorescent intermediate metabolite nicotinamide adenosine dinucleotide (NADH) fluorescence; this method is sensitive to the ratio of bound to free NADH. The results indicated a prompt shift in metabolic signature in the cancer cell line, but not in the normal cell line. Control KCN treatment demonstrated expected metabolic fluxes due to mitochondrial disruption. The detected radiation shift in the cancer cells was blunted in the presence of both a radical scavenger and a HIF-1α inhibitor. The HIF-1α abundance as detected by immunohistochemical staining also increased substantially for these cancer cells, but not for the normal cells. This type of live-cell metabolic monitoring could be helpful for future real-time studies and in designing adaptive radiotherapy approaches.
PMID:27128739 | PMC:PMC4882764 | DOI:10.1667/RR14093.1
View details for PubMedID 27128739
-
More
-
Potential role of the glycolytic oscillator in acute hypoxia in tumors Physics in medicine and biology
Fru LC, Adamson EB, Campos DD, Fain SB, Jacques SL, Kogel vd, Nickel KP, Song C, Kimple RJ, Kissick MW
2015 Dec 21;60(24):9215-25. doi: 10.1088/0031-9155/60/24/9215. Epub 2015 Nov 18.
-
More
Tumor acute hypoxia has a dynamic component that is also, at least partially, coherent. Using blood oxygen level dependent magnetic resonance imaging, we observed coherent oscillations in hemoglobin saturation dynamics in cell line xenograft models of head and neck squamous cell carcinoma. We posit a well-established biochemical nonlinear oscillatory mechanism called the glycolytic oscillator as a potential cause of the coherent oscillations in tumors. These data suggest that metabolic changes within individual tumor cells may affect the local tumor microenvironment including oxygen availability and therefore radiosensitivity. These individual cells can synchronize the oscillations in patches of similar intermediate glucose levels. These alterations have potentially important implications for radiation therapy and are a potential target for optimizing the cancer response to radiation.
PMID:26576743 | PMC:PMC4833657 | DOI:10.1088/0031-9155/60/24/9215
View details for PubMedID 26576743
-
More
-
Antitumor Effects of MEHD7945A, a Dual-Specific Antibody against EGFR and HER3, in Combination with Radiation in Lung and Head and Neck Cancers Molecular cancer therapeutics
Li C, Huang S, Armstrong EA, Francis DM, Werner LR, Sliwkowski MX, Kogel vd, Harari PM
2015 Sep;14(9):2049-59. doi: 10.1158/1535-7163.MCT-15-0155. Epub 2015 Jul 3.
-
More
Human epidermal growth factor receptor family members (EGFR, HER2, HER3, and HER4) play important roles in tumorigenesis and response to cancer therapeutics. In this study, we evaluated the capacity of the dual-target antibody MEHD7945A that simultaneously targets EGFR and HER3 to modulate radiation response in lung and head and neck cancer models. Antitumor effects of MEHD7945A in combination with radiation were evaluated in cell culture and tumor xenograft models. Mechanisms that may contribute to increased radiation killing by MEHD7945A, including DNA damage and inhibition of EGFR-HER signaling pathways, were analyzed. Immunohistochemical analysis of tumor xenografts was conducted to evaluate the effect of MEHD7945A in combination with radiation on tumor growth and microenvironment. MEHD7945A inhibited basal and radiation-induced EGFR and HER3 activation resulting in the inhibition of tumor cell growth and enhanced radiosensitivity. MEHD7945A was more effective in augmenting radiation response than treatment with individual anti-EGFR or anti-HER3 antibodies. An increase in DNA double-strand breaks associated γ-H2AX was observed in cells receiving combined treatment with MEHD7945A and radiation. Immunohistochemical staining evaluation in human tumor xenografts showed that MEHD7945A combined with radiation significantly reduced the expression of markers of tumor proliferation and tumor vasculature. These findings reveal the capacity of MEHD7945A to augment radiation response in lung and head and neck cancers. The dual EGFR/HER3-targeting action of MEHD7945A merits further investigation and clinical trial evaluation as a radiation sensitizer in cancer therapy.
PMID:26141946 | DOI:10.1158/1535-7163.MCT-15-0155
View details for PubMedID 26141946
-
More
-
Hypoxia, metabolism, and growth factor signaling in head and neck squamous cell carcinoma: correlation between primary and xenograft tumors Head & neck
Stegeman H, Rademakers SE, Span PN, Takes RP, Kogel vd, Kaanders AM, Bussink J
2014 Sep;36(9):1288-95. doi: 10.1002/hed.23446. Epub 2013 Nov 6.
-
More
BACKGROUND: Hypoxia, metabolism, and growth factor signaling are important prognostic features in most solid tumors. The purpose of this study was to determine whether head and neck squamous cell carcinoma (HNSCC) xenografts show similar biological and molecular characteristics as the primary tumor they originate from.
METHODS: Eighteen HNSCC primary tumor-xenograft pairs were immunofluorescently stained for pimonidazole (hypoxia), carbonic anhydrase IX (CAIX), glucose transporter-1 (GLUT-1), monocarboxylate transporter-1 (MCT-1), monocarboxylate transporter-4 (MCT-4), epidermal growth factor receptor (EGFR), and phosphorylated protein kinase B (pAKT).
RESULTS: Although no correlation was found for the amount of hypoxia, significant correlations between primary tumors and xenografts were observed for both the percentage of cells positive for expression and the hypoxia-related expression pattern of CAIX, GLUT-1, and MCT-1. For EGFR and MCT-4, the intensity of expression was correlated. No correlation was observed for pAKT.
CONCLUSION: Xenografts did not always resemble the primary tumor they originate from, but the xenografts did represent the variability in expression levels and patterns observed in the primary tumors.
PMID:24668936 | DOI:10.1002/hed.23446
View details for PubMedID 24668936
-
More
-
Systematic analysis of 18F-FDG PET and metabolism, proliferation and hypoxia markers for classification of head and neck tumors BMC cancer
Hoeben AW, Starmans HW, Leijenaar TH, Dubois LJ, Kogel vd, Kaanders AM, Boutros PC, Lambin P, Bussink J
2014 Feb 26;14:130. doi: 10.1186/1471-2407-14-130.
-
More
BACKGROUND: Quantification of molecular cell processes is important for prognostication and treatment individualization of head and neck cancer (HNC). However, individual tumor comparison can show discord in upregulation similarities when analyzing multiple biological mechanisms. Elaborate tumor characterization, integrating multiple pathways reflecting intrinsic and microenvironmental properties, may be beneficial to group most uniform tumors for treatment modification schemes. The goal of this study was to systematically analyze if immunohistochemical (IHC) assessment of molecular markers, involved in treatment resistance, and 18F-FDG PET parameters could accurately distinguish separate HNC tumors.
METHODS: Several imaging parameters and texture features for 18F-FDG small-animal PET and immunohistochemical markers related to metabolism, hypoxia, proliferation and tumor blood perfusion were assessed within groups of BALB/c nu/nu mice xenografted with 14 human HNC models. Classification methods were used to predict tumor line based on sets of parameters.
RESULTS: We found that 18F-FDG PET could not differentiate between the tumor lines. On the contrary, combined IHC parameters could accurately allocate individual tumors to the correct model. From 9 analyzed IHC parameters, a cluster of 6 random parameters already classified 70.3% correctly. Combining all PET/IHC characteristics resulted in the highest tumor line classification accuracy (81.0%; cross validation 82.0%), which was just 2.2% higher (p = 5.2×10-32) than the performance of the IHC parameter/feature based model.
CONCLUSIONS: With a select set of IHC markers representing cellular processes of metabolism, proliferation, hypoxia and perfusion, one can reliably distinguish between HNC tumor lines. Addition of 18F-FDG PET improves classification accuracy of IHC to a significant yet minor degree. These results may form a basis for development of tumor characterization models for treatment allocation purposes.
PMID:24571588 | PMC:PMC3940254 | DOI:10.1186/1471-2407-14-130
View details for PubMedID 24571588
-
More
-
Prognostic value of the proliferation marker Ki-67 in laryngeal carcinoma: results of the accelerated radiotherapy with carbogen breathing and nicotinamide phase III randomized trial Head & neck
Rademakers SE, Hoogsteen IJ, Rijken PF, Terhaard CH, Doornaert PA, Langendijk JA, Ende vd, Kogel vd, Bussink J, Kaanders JH
2015 Feb;37(2):171-6. doi: 10.1002/hed.23569. Epub 2014 Feb 12.
-
More
BACKGROUND: The prognostic and predictive value of the proliferation marker Ki-67 was investigated in a randomized trial comparing accelerated radiotherapy with carbogen breathing and nicotinamide (ARCON) to accelerated radiotherapy in laryngeal carcinoma.
METHODS: Labeling index of Ki-67 (Li Ki-67) in immunohistochemically stained biopsies and the colocalization with carbonic anhydrase IX (CAIX) were related to tumor control and patient survival.
RESULTS: On average, node-positive patients had a higher Li Ki-67 (median 14% vs 8%; p < .01). In patients with a high Li Ki-67, the 5-year regional control and metastases-free survival were 79% versus 96% (p < .01) and 71% versus 88% (p = .05) for accelerated radiotherapy and ARCON, respectively. The 5-year local control and disease-specific survival were not significantly different. Patients with low Ki-67 expression had an excellent outcome with accelerated radiotherapy alone.
CONCLUSION: Patients with laryngeal carcinomas with high proliferative activity are at increased risk of regional and distant metastases formation. This risk can be reduced by treatment with ARCON.
PMID:24347430 | DOI:10.1002/hed.23569
View details for PubMedID 24347430
-
More
-
Combining radiotherapy with MEK1/2, STAT5 or STAT6 inhibition reduces survival of head and neck cancer lines Molecular cancer
Stegeman H, Kaanders AM, Verheijen MG, Peeters JM, Wheeler DL, Iida M, Grénman R, Kogel vd, Span PN, Bussink J
2013 Nov 5;12(1):133. doi: 10.1186/1476-4598-12-133.
-
More
BACKGROUND: Kinases downstream of growth factor receptors have been implicated in radioresistance and are, therefore, attractive targets to improve radiotherapy outcome in head and neck squamous cell carcinoma (HNSCC) patients.
METHODS: An antibody-based array was used to quantify the expression levels of multiple phospho-kinases involved in growth factor signaling in nine untreated or irradiated HNSCC lines. Radiosensitivity was assessed with clonogenic cell survival assays and correlated with the expression levels of the phospho-kinases. Inhibitors of the kinases that were associated with radiosensitivity were tested for their ability to increase radiosensitivity in the 3 most radioresistant HNSCC lines.
RESULTS: The basal expression of phosphorylated Yes, Src and STAT5A, and the expression after radiotherapy of phosphorylated AKT, MSK1/2, Src, Lyn, Fyn, Hck, and STAT6, were correlated with radiosensitivity in the panel of HNSCC lines. In combination with radiotherapy, inhibitors of AKT, p38 and Src Family Kinases (SFK) were variably able to reduce survival, whereas MEK1/2, STAT5 and STAT6 inhibition reduced survival in all cell lines. The combined effect of radiotherapy and the kinase inhibitors on cell survival was mostly additive, although also supra-additive effects were observed for AKT, MEK1/2, p38 and STAT5 inhibition.
CONCLUSIONS: Kinases of the AKT, MAPK, STAT and SFK pathways correlated with radiosensitivity in a panel of HNSCC lines. Particularly inhibitors against MEK1/2, STAT5 and STAT6 were able to decrease survival in combination with radiotherapy. Hence, inhibitors against these kinases have the potential to improve radiotherapy outcome in HNSCC patients and further research is warranted to confirm this in vivo.
PMID:24192080 | PMC:PMC3842630 | DOI:10.1186/1476-4598-12-133
View details for PubMedID 24192080
-
More
-
Molecular radiation biology/oncology at its best: cutting edge research presented at the 13th International Wolfsberg Meeting on Molecular Radiation Biology/Oncology Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology
Baumann M, Bodis S, Dikomey E, Kogel vd, Overgaard J, Rodemann HP, Wouters B
2013 Sep;108(3):357-61. doi: 10.1016/j.radonc.2013.10.001.
-
On the importance of prompt oxygen changes for hypofractionated radiation treatments Physics in medicine and biology
Kissick M, Campos D, Kogel vd, Kimple R
2013 Oct 21;58(20):N279-85. doi: 10.1088/0031-9155/58/20/N279. Epub 2013 Sep 24.
-
More
This discussion is motivated by observations of prompt oxygen changes occurring prior to a significant number of cancer cells dying (permanently stopping their metabolic activity) from therapeutic agents like large doses of ionizing radiation. Such changes must be from changes in the vasculature that supplies the tissue or from the metabolic changes in the tissue itself. An adapted linear-quadratic treatment is used to estimate the cell survival variation magnitudes from repair and reoxygenation from a two-fraction treatment in which the second fraction would happen prior to significant cell death from the first fraction, in the large fraction limit. It is clear the effects of oxygen changes are likely to be the most significant factor for hypofractionation because of large radiation doses. It is a larger effect than repair. Optimal dose timing should be determined by the peak oxygen timing. A call is made to prioritize near real time measurements of oxygen dynamics in tumors undergoing hypofractionated treatments in order to make these treatments adaptable and patient-specific.
PMID:24061351 | PMC:PMC3856573 | DOI:10.1088/0031-9155/58/20/N279
View details for PubMedID 24061351
-
More
-
Paralysis following stereotactic spinal irradiation in pigs suggests a tolerance constraint for single-session irradiation of the spinal nerve Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology
Medin PM, Foster RD, Kogel vd, Meyer J, Sayre JW, Huang H, Öz OK
2013 Oct;109(1):107-11. doi: 10.1016/j.radonc.2013.08.025. Epub 2013 Sep 20.
-
More
BACKGROUND AND PURPOSE: Paralysis observed during a study of vertebral bone tolerance to single-session irradiation led to further study of the dose-related incidence of motor peripheral neuropathy.
MATERIALS AND METHODS: During a bone tolerance study, cervical spinal nerves of 15 minipigs received bilateral irradiation to levels C5-C8 distributed into three dose groups with mean maximum spinal nerve doses of 16.9 ± 0.3 Gy (n=5), 18.7 ± 0.5 Gy (n=5), and 24.3 ± 0.8 Gy (n=5). Changes developing in the gait of the group of pigs receiving a mean maximum dose of 24.3 Gy after 10-15 weeks led to the irradiation of two additional animals. They received mean maximum dose of 24.9 ± 0.2 Gy (n=2), targeted to the left spinal nerves of C5-C8. The followup period was one year. Histologic sections from spinal cords and available spinal nerves were evaluated. MR imaging was performed on pigs in the 24.9 Gy group.
RESULTS: No pig that received a maximum spinal nerve point dose ≤19.0 Gy experienced a change in gait while all pigs that received ≥24.1 Gy experienced paralysis. Extensive degeneration and fibrosis were observed in irradiated spinal nerves of the 24.9 Gy animals. All spinal cord sections were normal. Irradiated spinal nerve regions showed increased thickness and hypointensity on MR imaging.
CONCLUSION: The single-session tolerance dose of the cervical spinal nerves lies between 19.0 and 24.1 Gy for this model.
PMID:24060168 | PMC:PMC3840915 | DOI:10.1016/j.radonc.2013.08.025
View details for PubMedID 24060168
-
More
-
EGFR-inhibition enhances apoptosis in irradiated human head and neck xenograft tumors independent of effects on DNA repair Radiation research
Stegeman H, Span PN, Cockx SC, Peters PW, Rijken JW, Kogel vd, Kaanders AM, Bussink J
2013 Oct;180(4):414-21. doi: 10.1667/RR3349.2. Epub 2013 Sep 23.
-
More
Epidermal growth factor receptor (EGFR) inhibition using cetuximab improves the efficacy of radiotherapy in only a subgroup of head and neck squamous cell carcinoma (HNSCC) patients. Therefore, to improve patient selection a better understanding of tumor characteristics that affect treatment is necessary. Here, we investigated the effect of cetuximab on repair of radiation-induced DNA damage in a HNSCC xenograft model, which shows a synergistic effect to cetuximab and radiotherapy (SCCNij185) and a HNSCC model, which shows no additive effect of cetuximab to radiotherapy (SCCNij153). In both tumor models, clear increases were seen in the number of 53BP1 and Rad51 foci after irradiation. 53BP1 foci were present at comparable levels in hypoxic and normoxic tumor areas of the tumor xenografts, while the number of Rad51 foci was significantly higher in normoxic areas compared to hypoxic areas (P < 0.05). In both SCCNij185 and SCCNij153 xenografts an increased number of 53BP1 foci was observed in tumors treated with cetuximab and radiotherapy compared to radiotherapy alone. In SCCNij185 this increase was statistically significant in normoxic tumor areas (P = 0.04) and in SCCNij153 in both hypoxic and normoxic areas (P = 0.007 and P = 0.02, respectively). The number of Rad51 foci was not significantly different when cetuximab was added to radiotherapy compared to radiotherapy alone. Levels of pEGFR and pERK1/2 were decreased when cetuximab was added to radiotherapy in SCCNij185, but not in SCCNij153. Apoptosis was also only increased in SCCNij185 tumors at 4 days after cetuximab and radiotherapy treatment (P < 0.01). In conclusion, cetuximab inhibited DNA repair in both HNSCC models, but this effect was not predictive for the radiosensitizing effect of cetuximab in vivo. This lack of correlation may be related to differential effects of cetuximab and radiotherapy on ERK1/2 signaling and a decreased induction of apoptosis by cetuximab and radiotherapy in the resistant model.
PMID:24059677 | DOI:10.1667/RR3349.2
View details for PubMedID 24059677
-
More
-
Human epidermal growth factor receptor 3 (HER3) blockade with U3-1287/AMG888 enhances the efficacy of radiation therapy in lung and head and neck carcinoma Discovery medicine
Li C, Brand TM, Iida M, Huang S, Armstrong EA, Kogel vd, Wheeler DL
2013 Sep;16(87):79-92.
-
More
HER3 is a member of the epidermal growth factor receptor (EGFR) family of receptor tyrosine kinases. In the present study, we investigated the capacity of the HER3 blocking antibody, U3-1287/AMG888, to modulate the in vitro and in vivo radiation response of human squamous cell carcinomas of the lung and head and neck. We screened a battery of cell lines from these tumors for HER3 expression and demonstrated that all cell lines screened exhibited expression of HER3. Importantly, U3-1287/AMG888 treatment could block both basal HER3 activity and radiation induced HER3 activation. Proliferation assays indicated that HER3 blockade could decrease the proliferation of both HNSCC cell line SCC6 and NSCLC cell line H226. Further, we demonstrated that U3-1287/AMG888 can sensitize cells to radiation in clonogenic survival assays, in addition to increasing DNA damage as detected via λ-H2AX immunofluorescence. To determine if U3-1287/AMG888 could enhance radiation sensitivity in vivo we performed tumor growth delay experiments using SCC6, SCC1483, and H226 xenografts. The results of these experiments indicated that the combination of U3-1287/AMG888 and radiation could decrease tumor growth in studies using single or fractionated doses of radiation. Analysis of HER3 expression in tumor samples indicated that radiation treatment activated HER3 in vivo and that U3-1287/AMG888 could abrogate this activation. Immunohistochemistry analysis of SCC6 tumors treated with both U3-1287/AMG888 and a single dose of radiation demonstrated that various cell survival and proliferation markers could be reduced. Collectively our findings suggest that U3-1287/AMG888 in combination with radiation has an impact on cell and tumor growth by increasing DNA damage and cell death. These findings suggest that HER3 may play an important role in response to radiation therapy and blocking its activity in combination with radiation may be of therapeutic benefit in human tumors.
PMID:23998444 | PMC:PMC3901945
View details for PubMedID 23998444
-
More
-
Dasatinib Inhibits DNA Repair after Radiotherapy Specifically in pSFK-Expressing Tumor Areas in Head and Neck Xenograft Tumors Translational oncology
Stegeman H, Span PN, Rijken JW, Cockx SC, Wheeler DL, Iida M, Kogel vd, Kaanders AM, Bussink J
2013 Aug 1;6(4):413-9. doi: 10.1593/tlo.13259. Print 2013 Aug.
-
More
Src family kinases (SFKs) have been implicated in resistance to both radiation and epidermal growth factor receptor (EGFR) inhibition. Therefore, we investigated whether inhibition of SFK through dasatinib (DSB) can enhance the effect of radiotherapy in two in vivo human head and neck squamous cell carcinoma (HNSCC) models. Response to DSB and/or radiotherapy was assessed with tumor growth delay assays in two HNSCC xenograft models, SCCNij153 and SCCNij202. Effects on EGFR signaling were evaluated with Western blot analysis, and effects on DNA repair, hypoxia, and proliferation were investigated with immunohistochemistry. DSB and radiotherapy induced a significant growth delay in both HNSCC xenograft models, although to a lesser extent in SCCNij202. DSB did not inhibit phosphorylated protein kinase B (pAKT) or phosphorylated extracellular signal-regulated kinase 1/2 (pERK1/2) but did inhibit (phosphorylated) DNA-dependent protein kinase. Moreover, DSB reduced repair of radiation-induced DNA double-strand breaks as shown by an increase of p53-binding protein 1 (53BP1) staining 24 hours after radiation. This effect on DNA repair was only observed in the cell compartment where phosphorylated SFK (pSFK) was expressed: for SCCNij153 tumors in both normoxic and hypoxic areas and for SCCNij202 tumors only in hypoxic areas. No consistent effects of DSB on hypoxia or proliferation were observed. In conclusion, DSB enhances the effect of radiotherapy in vivo by inhibition of radiation-induced DNA repair and is a promising way to improve outcome in HNSCC patients.
PMID:23908684 | PMC:PMC3730016 | DOI:10.1593/tlo.13259
View details for PubMedID 23908684
-
More
-
The PERK/ATF4/LAMP3-arm of the unfolded protein response affects radioresistance by interfering with the DNA damage response Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology
Nagelkerke A, Bussink J, Kogel vd, Sweep GJ, Span PN
2013 Sep;108(3):415-21. doi: 10.1016/j.radonc.2013.06.037. Epub 2013 Jul 25.
-
More
BACKGROUND AND PURPOSE: Lysosome-associated membrane protein 3 (LAMP3) is induced by the PKR-like ER kinase (PERK)/activating transcription factor 4 (ATF4)-arm of the unfolded protein response (UPR) during hypoxia. LAMP3 has prognostic value in breast cancer patients treated with radiotherapy. Here, we specifically investigated the role of the PERK/ATF4/LAMP3-arm in the radiation response of breast cancer cells.
MATERIAL AND METHODS: Radiosensitivity of breast cancer cells was examined after siRNA-mediated knockdown of PERK, ATF4 and LAMP3. Activation of DNA damage repair proteins was evaluated by Western blotting and immunocytochemistry.
RESULTS: Knockdown of the PERK/ATF4/LAMP3-arm and chemical inhibition of PERK could radiosensitise MDA-MB-231 cells significantly. Western blot analysis of several DNA damage repair proteins showed that LAMP3 knockdowns had an attenuated DNA damage response after radiation compared to controls. γ-H2AX foci analysis revealed that LAMP3 knockdowns had a reduced number of positive cells after irradiation, indicating that their DNA damage repair signalling response is decreased. In addition, the effect of autophagy inhibition was examined and revealed a radiosensitising effect and the presence of residual γ-H2AX foci.
CONCLUSIONS: The PERK/ATF4/LAMP3-arm causes radioresistance of breast cancer cells by increasing DNA damage repair signalling. Inhibition of PERK and/or autophagy may sensitise tumours to radiotherapy.
PMID:23891100 | DOI:10.1016/j.radonc.2013.06.037
View details for PubMedID 23891100
-
More
-
Epidermal growth factor receptor expression in laryngeal cancer predicts the effect of hypoxia modification as an additive to accelerated radiotherapy in a randomised controlled trial European journal of cancer (Oxford, England : 1990)
Nijkamp MM, Span PN, Terhaard HJ, Doornaert AH, Langendijk JA, Ende vd, Jong Md, Kogel vd, Bussink J, Kaanders AM
2013 Oct;49(15):3202-9. doi: 10.1016/j.ejca.2013.06.024. Epub 2013 Jul 15.
-
More
Accelerated radiotherapy (AR) improves the poor prognosis associated with epidermal growth factor receptor (EGFR) overexpression frequently seen in head and neck carcinomas. Combining AR with carbogen and nicotinamide (ARCON) counteracts enhanced tumour cell proliferation- and hypoxia-related radioresistance. The purpose of this study was to investigate if EGFR expression levels are associated with response to ARCON in patients with carcinoma of the larynx. Patients (N=272) with advanced stage larynx carcinoma were randomised between AR alone and ARCON. Paraffin-embedded biopsies from these patients were processed for immunohistochemical staining of EGFR. EGFR fraction was quantitated by automated image analysis and related to clinical outcome. A large variation was observed in EGFR fraction between tumours with expression levels ranging from 0 to 0.93 (median fraction 0.4). No difference in 5-year locoregional control was found between low and high EGFR expressing tumours in the AR arm (69% versus 75%), which is in line with the established effect of AR in EGFR overexpressing tumours. There was, however, a significant association in the ARCON arm: patients with low EGFR levels had a better 5-year locoregional control (88% versus 72% p=0.02) and disease-specific survival (92% versus 77% p=0.01). ARCON improved locoregional control relative to AR only in patients with low EGFR expression (hazard ratio (HR) 0.34 p=0.009). In conclusion, only in tumours with a low EGFR fraction, adding hypoxia modification to AR has an additive beneficial effect on outcome. EGFR expression is a predictive biomarker for the selection of patients that will or will not respond to ARCON.
PMID:23867129 | DOI:10.1016/j.ejca.2013.06.024
View details for PubMedID 23867129
-
More
-
Evaluation and immunohistochemical qualification of carbogen-induced ΔR₂ as a noninvasive imaging biomarker of improved tumor oxygenation International journal of radiation oncology, biology, physics
Baker CJ, Boult KR, Jamin Y, Gilmour LD, Walker-Samuel S, Burrell JS, Ashcroft M, Howe FA, Griffiths JR, Raleigh JA, Kogel vd, Robinson SP
2013 Sep 1;87(1):160-7. doi: 10.1016/j.ijrobp.2013.04.051. Epub 2013 Jul 9.
-
More
PURPOSE: To evaluate and histologically qualify carbogen-induced ΔR2 as a noninvasive magnetic resonance imaging biomarker of improved tumor oxygenation using a double 2-nitroimidazole hypoxia marker approach.
METHODS AND MATERIALS: Multigradient echo images were acquired from mice bearing GH3 prolactinomas, preadministered with the hypoxia marker CCI-103F, to quantify tumor R2 during air breathing. With the mouse remaining positioned within the magnet bore, the gas supply was switched to carbogen (95% O2, 5% CO2), during which a second hypoxia marker, pimonidazole, was administered via an intraperitoneal line, and an additional set of identical multigradient echo images acquired to quantify any changes in tumor R2. Hypoxic fraction was quantified histologically using immunofluorescence detection of CCI-103F and pimonidazole adduct formation from the same whole tumor section. Carbogen-induced changes in tumor pO2 were further validated using the Oxylite fiberoptic probe.
RESULTS: Carbogen challenge significantly reduced mean tumor R2 from 116 ± 13 s(-1) to 97 ± 9 s(-1) (P<.05). This was associated with a significantly lower pimonidazole adduct area (2.3 ± 1%), compared with CCI-103F (6.3 ± 2%) (P<.05). A significant correlation was observed between ΔR2 and Δhypoxic fraction (r=0.55, P<.01). Mean tumor pO2 during carbogen breathing significantly increased from 6.3 ± 2.2 mm Hg to 36.0 ± 7.5 mm Hg (P<.01).
CONCLUSIONS: The combined use of intrinsic susceptibility magnetic resonance imaging with a double hypoxia marker approach corroborates carbogen-induced ΔR2 as a noninvasive imaging biomarker of increased tumor oxygenation.
PMID:23849692 | DOI:10.1016/j.ijrobp.2013.04.051
View details for PubMedID 23849692
-
More
-
Pattern of CAIX expression is prognostic for outcome and predicts response to ARCON in patients with laryngeal cancer treated in a phase III randomized trial Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology
Rademakers SE, Hoogsteen IJ, Rijken PF, Oosterwijk E, Terhaard CH, Doornaert PA, Langendijk JA, Ende vd, Takes R, Bree RD, Kogel vd, Bussink J, Kaanders JH
2013 Sep;108(3):517-22. doi: 10.1016/j.radonc.2013.04.022. Epub 2013 May 26.
-
More
BACKGROUND AND PURPOSE: In a phase III trial in patients with advanced stage laryngeal carcinoma comparing ARCON (accelerated radiotherapy with carbogen breathing and nicotinamide) to accelerated radiotherapy alone (AR) the prognostic and predictive value of CAIX, a hypoxia-associated protein, was investigated.
MATERIAL AND METHODS: 261 Paraffin embedded tumor biopsies and 79 fresh frozen biopsies from patients entered in the trial were immunohistochemically stained for CAIX. CAIX-fraction and CAIX expression pattern were related to tumor control and patient survival.
RESULTS: Low CAIX-fraction was prognostic for worse regional control and overall survival in patients treated with AR. Patients with a low CAIX-fraction treated with ARCON had better regional control and metastasis-free survival compared to AR (RC 97% vs 71%, p < 0.01 and MFS 92% vs 69%, p = 0.06). Patients with a perinecrotic CAIX staining pattern had a significantly worse local control, metastasis-free and overall survival compared to patients with a diffuse pattern (65% vs 84%, p = 0.01, 70% vs 96%, p < 0.01 and 42% vs 71%, p < 0.01 respectively), and this could not be improved with ARCON. After multivariate analysis CAIX pattern and N-stage emerged as significant predictors for metastasis-free survival and overall survival.
CONCLUSIONS: ARCON improves regional control and metastasis-free survival only in patients with low CAIX expression. The different patterns of CAIX expression suggest different mechanisms of upregulation and have important prognostic value.
PMID:23719582 | DOI:10.1016/j.radonc.2013.04.022
View details for PubMedID 23719582
-
More
-
PERK/eIF2α signaling protects therapy resistant hypoxic cells through induction of glutathione synthesis and protection against ROS Proceedings of the National Academy of Sciences of the United States of America
Rouschop KM, Dubois LJ, Keulers TG, Beucken vd, Lambin P, Bussink J, Kogel vd, Koritzinsky M, Wouters BG
2013 Mar 19;110(12):4622-7. doi: 10.1073/pnas.1210633110. Epub 2013 Mar 7.
-
More
Hypoxia is a common feature of tumors and an important contributor to malignancy and treatment resistance. The ability of tumor cells to survive hypoxic stress is mediated in part by hypoxia-inducible factor (HIF)-dependent transcriptional responses. More severe hypoxia activates endoplasmatic reticulum stress responses, including the double-stranded RNA-activated protein kinase (PKR)-like endoplasmic reticulum kinase (PERK)/eukaryotic initiation factor 2α (eIF2α)-dependent arm of the unfolded protein response (UPR). Although several studies implicate important roles for HIF and UPR in adaption to hypoxia, their importance for hypoxic cells responsible for therapy resistance in tumors is unknown. By using isogenic models, we find that HIF and eIF2α signaling contribute to the survival of hypoxic cells in vitro and in vivo. However, the eIF2α-dependent arm of the UPR is uniquely required for the survival of a subset of hypoxic cells that determine tumor radioresistance. We demonstrate that eIF2α signaling induces uptake of cysteine, glutathione synthesis, and protection against reactive oxygen species produced during periods of cycling hypoxia. Together these data imply that eIF2α signaling is a critical contributor to the tolerance of therapy-resistant cells that arise as a consequence of transient changes in oxygenation in solid tumors and thus a therapeutic target in curative treatments for solid cancers.
PMID:23471998 | PMC:PMC3607059 | DOI:10.1073/pnas.1210633110
View details for PubMedID 23471998
-
More
-
Predictive value of hypoxia, proliferation and tyrosine kinase receptors for EGFR-inhibition and radiotherapy sensitivity in head and neck cancer models Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology
Stegeman H, Kaanders JH, Kogel vd, Iida M, Wheeler DL, Span PN, Bussink J
2013 Mar;106(3):383-9. doi: 10.1016/j.radonc.2013.02.001. Epub 2013 Feb 28.
-
More
BACKGROUND AND PURPOSE: EGFR-inhibitor Cetuximab (C225) improves the efficacy of radiotherapy in only a subgroup of HNSCC patients. Identification of predictive tumor characteristics is essential to improve patient selection.
MATERIAL AND METHODS: Response to C225 and/or radiotherapy was assessed with tumor growth delay assays in 4 HNSCC xenograft models with varying EGFR-expression levels. Hypoxia and proliferation were quantified with immunohistochemistry and the expression of proteins involved in C225-resistance with Western blot.
RESULTS: EGFR-expression did not predict response to C225 and/or radiotherapy. Reduction of hypoxia by C225 was only observed in SCCNij202, which was highly sensitive to C225. Proliferation changes correlated with response to C225 and C225 combined with radiotherapy, as proliferation decreased after C225 treatment in C225-sensitive SCCNij202 and after combined treatment in SCCNij185, which showed a synergistic effect to combined C225-radiotherapy. Furthermore, C225-resistant SCCNij153 tumors expressed high levels of (activated) HER3 and MET.
CONCLUSIONS: EGFR-expression is needed for C225-response, but is not sufficient to predict response to C225 with or without radiotherapy. However, basal expression of additional growth factor receptors and effects on proliferation, but not hypoxia, correlated with response to combined C225-radiotherapy treatment and are potential clinically relevant predictive biomarkers.
PMID:23453541 | PMC:PMC3627829 | DOI:10.1016/j.radonc.2013.02.001
View details for PubMedID 23453541
-
More
-
Bioeffect modeling and equieffective dose concepts in radiation oncology--terminology, quantities and units Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology
Bentzen SM, Dörr W, Gahbauer R, Howell RW, Joiner MC, Jones B, Jones TL, Kogel vd, Wambersie A, Whitmore G
2012 Nov;105(2):266-8. doi: 10.1016/j.radonc.2012.10.006. Epub 2012 Nov 15.
-
More
The International Commission on Radiation Units and Measurements (ICRU) Report Committee on "Bioeffect Modeling and Biologically Equivalent Dose Concepts in Radiation Therapy" is currently developing a comprehensive and consistent framework for radiobiological effect modeling based on the equieffective dose, EQDX(α/β), a concept encompassing BED and EQD2 as special cases.
PMID:23157980 | DOI:10.1016/j.radonc.2012.10.006
View details for PubMedID 23157980
-
More
-
Activation of AKT by hypoxia: a potential target for hypoxic tumors of the head and neck BMC cancer
Stegeman H, Kaanders JH, Wheeler DL, Kogel vd, Verheijen MM, Waaijer SJ, Iida M, Grénman R, Span PN, Bussink J
2012 Oct 10;12:463. doi: 10.1186/1471-2407-12-463.
-
More
BACKGROUND: Only a minority of cancer patients benefits from the combination of EGFR-inhibition and radiotherapy in head and neck squamous cell carcinoma (HNSCC). A potential resistance mechanism is activation of EGFR and/or downstream pathways by stimuli in the microenvironment. The aim of this study was to find molecular targets induced by the microenvironment by determining the in vitro and in vivo expression of proteins of the EGFR-signaling network in 6 HNSCC lines. As hypoxia is an important microenvironmental parameter associated with poor outcome in solid tumors after radiotherapy, we investigated the relationship with hypoxia in vitro and in vivo.
METHODS: Six human HNSCC cell lines were both cultured as cell lines (in vitro) and grown as xenograft tumors (in vivo). Expression levels were determined via western blot analysis and localization of markers was assessed via immunofluorescent staining. To determine the effect of hypoxia and pAKT-inhibition on cell survival, cells were incubated at 0.5% O(2) and treated with MK-2206.
RESULTS: We observed strong in vitro-in vivo correlations for EGFR, pEGFR and HER2 (rs = 0.77, p = 0.10, rs = 0.89, p = 0.03) and rs = 0.93, p = 0.02, respectively), but not for pAKT, pERK1/2 or pSTAT3 (all r(s)<0.55 and p>0.30). In vivo, pAKT expression was present in hypoxic cells and pAKT and hypoxia were significantly correlated (rs = 0.51, p = 0.04). We confirmed in vitro that hypoxia induces activation of AKT. Further, pAKT-inhibition via MK-2206 caused a significant decrease in survival in hypoxic cells (p<0.01), but not in normoxic cells.
CONCLUSIONS: These data suggest that (p)EGFR and HER2 expression is mostly determined by intrinsic features of the tumor cell, while the activation of downstream kinases is highly influenced by the tumor microenvironment. We show that hypoxia induces activation of AKT both in vitro and in vivo, and that hypoxic cells can be specifically targeted by pAKT-inhibition. Targeting pAKT is thus a potential way to overcome therapy resistance induced by hypoxia and improve patient outcome.
PMID:23046567 | PMC:PMC3517352 | DOI:10.1186/1471-2407-12-463
View details for PubMedID 23046567
-
More
-
Spinal cord tolerance to single-session uniform irradiation in pigs: implications for a dose-volume effect Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology
Medin PM, Foster RD, Kogel vd, Sayre JW, McBride WH, Solberg TD
2013 Jan;106(1):101-5. doi: 10.1016/j.radonc.2012.08.007. Epub 2012 Sep 14.
-
More
BACKGROUND AND PURPOSE: This study was performed to test the hypothesis that spinal cord radiosensitivity is significantly modified by uniform versus laterally non-uniform dose distributions.
MATERIALS AND METHODS: A uniform dose distribution was delivered to a 4.5-7.0 cm length of cervical spinal cord in 22 mature Yucatan minipigs for comparison with a companion study in which a laterally non-uniform dose was given [1]. Pigs were allocated into four dose groups with mean maximum spinal cord doses of 17.5 ± 0.1 Gy (n=7), 19.5 ± 0.2 Gy (n=6), 22.0 ± 0.1 Gy (n=5), and 24.1 ± 0.2 Gy (n=4). The study endpoint was motor neurologic deficit determined by a change in gait within one year. Spinal cord sections were stained with a Luxol fast blue/periodic acid Schiff combination.
RESULTS: Dose-response curves for uniform versus non-uniform spinal cord irradiation were nearly identical with ED(50)'s (95% confidence interval) of 20.2 Gy (19.1-25.8) and 20.0 Gy (18.3-21.7), respectively. No neurologic change was observed for either dose distribution when the maximum spinal cord dose was ≤ 17.8 Gy while all animals experienced deficits at doses ≥ 21.8 Gy.
CONCLUSION: No dose-volume effect was observed in pigs for the dose distributions studied and the endpoint of motor neurologic deficit; however, partial spinal cord irradiation resulted in less debilitating neurologic morbidity and histopathology.
PMID:22985780 | PMC:PMC3526692 | DOI:10.1016/j.radonc.2012.08.007
View details for PubMedID 22985780
-
More
-
(64)Cu-ATSM and (18)FDG PET uptake and (64)Cu-ATSM autoradiography in spontaneous canine tumors: comparison with pimonidazole hypoxia immunohistochemistry Radiation oncology (London, England)
Hansen AE, Kristensen AT, Jørgensen JT, McEvoy FJ, Busk M, Kogel vd, Bussink J, Engelholm SA, Kjær A
2012 Jun 15;7:89. doi: 10.1186/1748-717X-7-89.
-
More
BACKGROUND: The aim of this study was to compare (64)Cu-diacetyl-bis(N(4)-methylsemicarbazone) ((64)Cu-ATSM) and (18)FDG PET uptake characteristics and (64)Cu-ATSM autoradiography to pimonidazole immunohistochemistry in spontaneous canine sarcomas and carcinomas.
METHODS: Biopsies were collected from individual tumors between approximately 3 and 25 hours after the intravenous injection of (64)Cu-ATSM and pimonidazole. (64)Cu-ATSM autoradiography and pimonidazole immunostaining was performed on sectioned biopsies. Acquired (64)Cu-ATSM autoradiography and pimonidazole images were rescaled, aligned and their distribution patterns compared. (64)Cu-ATSM and (18)FDG PET/CT scans were performed in a concurrent study and uptake characteristics were obtained for tumors where available.
RESULTS: Maximum pimonidazole pixel value and mean pimonidazole labeled fraction was found to be strongly correlated to (18)FDG PET uptake levels, whereas more varying results were obtained for the comparison to (64)Cu-ATSM. In the case of the latter, uptake at scans performed 3 h post injection (pi) generally showed strong positive correlated to pimonidazole uptake.Comparison of distribution patterns of pimonidazole immunohistochemistry and (64)Cu-ATSM autoradiography yielded varying results. Significant positive correlations were mainly found in sections displaying a heterogeneous distribution of tracers.
CONCLUSIONS: Tumors with high levels of pimonidazole staining generally displayed high uptake of (18)FDG and (64)Cu-ATSM (3 h pi.). Similar regional distribution of (64)Cu-ATSM and pimonidazole was observed in most heterogeneous tumor regions. However, tumor and hypoxia level dependent differences may exist with regard to the hypoxia specificity of (64)Cu-ATSM in canine tumors.
PMID:22704363 | PMC:PMC3403947 | DOI:10.1186/1748-717X-7-89
View details for PubMedID 22704363
-
More
-
Vascular responses to radiotherapy and androgen-deprivation therapy in experimental prostate cancer Radiation oncology (London, England)
Røe K, Mikalsen LT, Kogel vd, Bussink J, Lyng H, Ree AH, Marignol L, Olsen DR
2012 May 23;7:75. doi: 10.1186/1748-717X-7-75.
-
More
BACKGROUND: Radiotherapy (RT) and androgen-deprivation therapy (ADT) are standard treatments for advanced prostate cancer (PC). Tumor vascularization is recognized as an important physiological feature likely to impact on both RT and ADT response, and this study therefore aimed to characterize the vascular responses to RT and ADT in experimental PC.
METHODS: Using mice implanted with CWR22 PC xenografts, vascular responses to RT and ADT by castration were visualized in vivo by DCE MRI, before contrast-enhancement curves were analyzed both semi-quantitatively and by pharmacokinetic modeling. Extracted image parameters were correlated to the results from ex vivo quantitative fluorescent immunohistochemical analysis (qIHC) of tumor vascularization (9 F1), perfusion (Hoechst 33342), and hypoxia (pimonidazole), performed on tissue sections made from tumors excised directly after DCE MRI.
RESULTS: Compared to untreated (Ctrl) tumors, an improved and highly functional vascularization was detected in androgen-deprived (AD) tumors, reflected by increases in DCE MRI parameters and by increased number of vessels (VN), vessel density (VD), and vessel area fraction (VF) from qIHC. Although total hypoxic fractions ( HF) did not change, estimated acute hypoxia scores (AHS)--the proportion of hypoxia staining within 50 μm from perfusion staining--were increased in AD tumors compared to in Ctrl tumors. Five to six months after ADT renewed castration-resistant (CR) tumor growth appeared with an even further enhanced tumor vascularization. Compared to the large vascular changes induced by ADT, RT induced minor vascular changes. Correlating DCE MRI and qIHC parameters unveiled the semi-quantitative parameters area under curve (AUC) from initial time-points to strongly correlate with VD and VF, whereas estimation of vessel size (VS) by DCE MRI required pharmacokinetic modeling. HF was not correlated to any DCE MRI parameter, however, AHS may be estimated after pharmacokinetic modeling. Interestingly, such modeling also detected tumor necrosis very strongly.
CONCLUSIONS: DCE MRI reliably allows non-invasive assessment of tumors' vascular function. The findings of increased tumor vascularization after ADT encourage further studies into whether these changes are beneficial for combined RT, or if treatment with anti-angiogenic therapy may be a strategy to improve the therapeutic efficacy of ADT in advanced PC.
PMID:22621752 | PMC:PMC3441216 | DOI:10.1186/1748-717X-7-75
View details for PubMedID 22621752
-
More
-
Accelerated radiotherapy with carbogen and nicotinamide for laryngeal cancer: results of a phase III randomized trial Journal of clinical oncology : official journal of the American Society of Clinical Oncology
Janssens GO, Rademakers SE, Terhaard CH, Doornaert PA, Bijl HP, Ende vd, Chin A, Marres HA, Bree Rd, Kogel vd, Hoogsteen IJ, Bussink J, Span PN, Kaanders JH
2012 May 20;30(15):1777-83. doi: 10.1200/JCO.2011.35.9315. Epub 2012 Apr 16.
-
More
PURPOSE: To report the results from a randomized trial comparing accelerated radiotherapy (AR) with accelerated radiotherapy plus carbogen inhalation and nicotinamide (ARCON) in laryngeal cancer.
PATIENTS AND METHODS: Patients with cT2-4 squamous cell laryngeal cancer were randomly assigned to AR (68 Gy within 36 to 38 days) or ARCON. To limit the risk of laryngeal necrosis, ARCON patients received 64 Gy on the laryngeal cartilage. The primary end point was local control. Secondary end points were regional control, larynx preservation, toxicity, disease-free survival, and overall survival. In a translational side study, the hypoxia marker pimonidazole was used to assess the oxygenation status in tumor biopsies.
RESULTS: From April 2001 to February 2008, 345 patients were accrued. After a median follow-up of 44 months, local tumor control rate at 5 years was 78% for AR versus 79% for ARCON (P = .80), with larynx preservation rates of 84% and 87%, respectively (P = .48). The 5-year regional control was significantly better with ARCON (93%) compared with AR (86%, P = .04). The improvement in regional control was specifically observed in patients with hypoxic tumors and not in patients with well-oxygenated tumors (100% v 55%, respectively; P = .01). AR and ARCON produced equal levels of toxicity.
CONCLUSION: Despite lack of benefit in local tumor control for advanced laryngeal cancers, a significant gain in regional control rate, with equal levels of toxicity, was observed in favor of ARCON. The poor regional control of patients with hypoxic tumors is specifically countered by ARCON treatment.
PMID:22508814 | DOI:10.1200/JCO.2011.35.9315
View details for PubMedID 22508814
-
More
-
Spinal cord tolerance to reirradiation with single-fraction radiosurgery: a swine model International journal of radiation oncology, biology, physics
Medin PM, Foster RD, Kogel vd, Sayre JW, McBride WH, Solberg TD
2012 Jul 1;83(3):1031-7. doi: 10.1016/j.ijrobp.2011.08.030. Epub 2011 Dec 22.
-
More
PURPOSE: This study was performed to determine swine spinal cord tolerance to single-fraction, partial-volume irradiation 1 year after receiving uniform irradiation to 30 Gy in 10 fractions.
METHODS AND MATERIALS: A 10-cm length of spinal cord (C3-T1) was uniformly irradiated to 30 Gy in 10 consecutive fractions and reirradiated 1 year later with a single radiosurgery dose centered within the previously irradiated segment. Radiosurgery was delivered to a cylindrical volume approximately 5 cm in length and 2 cm in diameter, which was positioned laterally to the cervical spinal cord, resulting in a dose distribution with the 90%, 50%, and 10% isodose lines traversing the ipsilateral, central, and contralateral spinal cord, respectively. Twenty-three pigs were stratified into six dose groups with mean maximum spinal cord doses of 14.9 ± 0.1 Gy (n = 2), 17.1 ± 0.3 Gy (n = 3), 19.0 ± 0.1 Gy (n = 5), 21.2 ± 0.1 Gy (n = 5), 23.4 ± 0.2 Gy (n = 5), and 25.4 ± 0.4 Gy (n = 3). The mean percentage of spinal cord volumes receiving ≥10 Gy for the same groups were 34% ± 1%, 40% ± 1%, 46% ± 3%, 52% ± 1%, 56 ± 3%, and 57% ± 1%. The study endpoint was motor neurologic deficit as determined by a change in gait during a 1- year follow-up period.
RESULTS: A steep dose-response curve was observed with a 50% incidence of paralysis (ED(50)) for the maximum point dose of 19.7 Gy (95% confidence interval, 17.4-21.4). With two exceptions, histology was unremarkable in animals with normal neurologic status, while all animals with motor deficits showed some degree of demyelination and focal white matter necrosis on the irradiated side, with relative sparing of gray matter. Histologic comparison with a companion study of de novo irradiated animals revealed that retreatment responders had more extensive tissue damage, including infarction of gray matter, only at prescription doses >20 Gy.
CONCLUSION: Pigs receiving spinal radiosurgery 1 year after receiving 30 Gy in 10 fractions were not at significantly higher risk of developing motor deficits than pigs that received radiosurgery alone.
PMID:22197239 | PMC:PMC3314093 | DOI:10.1016/j.ijrobp.2011.08.030
View details for PubMedID 22197239
-
More
-
Preclinical evaluation and validation of [18F]HX4, a promising hypoxia marker for PET imaging Proceedings of the National Academy of Sciences of the United States of America
Dubois LJ, Lieuwes NG, Janssen HM, Peeters JM, Windhorst AD, Walsh JC, Kolb HC, Ollers MC, Bussink J, van Dongen S, Kogel vd, Lambin P
2011 Aug 30;108(35):14620-5. doi: 10.1073/pnas.1102526108. Epub 2011 Aug 23.
-
More
Hypoxia has been shown to be an important microenvironmental parameter influencing tumor progression and treatment efficacy. Patient guidance for hypoxia-targeted therapy requires evaluation of tumor oxygenation, preferably in a noninvasive manner. The aim of this study was to evaluate and validate the uptake of [(18)F]HX4, a novel developed hypoxia marker for PET imaging. A heterogeneous accumulation of [(18)F]HX4 was found within rat rhabdomyosarcoma tumors that was significantly (P < 0.0001) higher compared with the surrounding tissues, with temporal increasing tumor-to-blood ratios reaching a plateau of 7.638 ± 0.926 and optimal imaging properties 4 h after injection. [(18)F]HX4 retention in normal tissues was found to be short-lived, homogeneous and characterized by a fast progressive temporal clearance. Heterogeneity in [(18)F]HX4 tumor uptake was analyzed based on 16 regions within the tumor according to the different orthogonal planes at the largest diameter. Validation of heterogeneous [(18)F]HX4 tumor uptake was shown by a strong and significant relationship (r = 0.722; P < 0.0001) with the hypoxic fraction as calculated by the percentage pimonidazole-positive pixels. Furthermore, a causal relationship with tumor oxygenation was established, because combination treatment of nicotinamide and carbogen resulted in a 40% reduction (P < 0.001) in [(18)F]HX4 tumor accumulation whereas treatment with 7% oxygen breathing resulted in a 30% increased uptake (P < 0.05). [(18)F]HX4 is therefore a promising candidate for noninvasive detection and evaluation of tumor hypoxia at a macroscopic level.
PMID:21873245 | PMC:PMC3167561 | DOI:10.1073/pnas.1102526108
View details for PubMedID 21873245
-
More
-
Characterization of positron emission tomography hypoxia tracer uptake and tissue oxygenation via electrochemical modeling Nuclear medicine and biology
Bowen SR, Kogel vd, Nordsmark M, Bentzen SM, Jeraj R
2011 Aug;38(6):771-80. doi: 10.1016/j.nucmedbio.2011.02.002. Epub 2011 May 5.
-
More
PURPOSE: Unique uptake and retention mechanisms of positron emission tomography (PET) hypoxia tracers make in vivo comparison between them challenging. Differences in imaged uptake of two common hypoxia radiotracers, [(61)Cu]Cu-ATSM and [(18)F]FMISO, were characterized via computational modeling to address these challenges.
MATERIALS AND METHODS: An electrochemical formalism describing bioreductive retention mechanisms of these tracers under steady-state conditions was adopted to relate time-averaged activity concentration to tissue partial oxygen tension (PO(2)), a common metric of hypoxia. Chemical equilibrium constants of product concentration to reactant concentration ratios were determined from free energy changes and reduction potentials of pertinent reactions reported in the literature. Resulting transformation functions between tracer uptake and PO(2) were compared against measured values in preclinical models. Additionally, calculated PO(2) distributions from imaged Cu-ATSM tracer activity concentrations of 12 head and neck squamous cell carcinoma (HNSCC) patients were validated against microelectrode PO(2) measurements in 69 HNSCC patients.
RESULTS: Both Cu-ASTM- and FMISO-modeled PO(2) transformation functions were in agreement with preclinical measured values within single-deviation confidence intervals. High correlation (r(2)=0.94, P<.05) was achieved between modeled PO(2) distributions and measured distributions in the patient populations. On average, microelectrode hypoxia thresholds (2.5 and 5.0 mmHg) corresponded to higher Cu-ATSM uptake [2.5 and 2.0 standardized uptake value (SUV)] and lower FMISO uptake (2.0 and 1.4 SUV). Uncertainties in the models were dominated by variations in the estimated specific activity and intracellular acidity.
CONCLUSIONS: Results indicated that the high dynamic range of Cu-ATSM uptake was representative of a narrow range of low oxygen tension whose values were dependent on microenvironment acidity, while FMISO uptake was representative of a wide range of PO(2) values that were independent of acidity. The models shed light on possible causes of these discrepancies, particularly as it pertains to image contrast, and may prove to be a useful methodology in quantifying relationships between other hypoxia tracers. Comprehensive and robust assessment of tumor hypoxia prior to as well as in response to therapy may be best provided by imaging of multiple hypoxia markers that provide complementary rather than interchangeable information.
PMID:21843774 | PMC:PMC3157049 | DOI:10.1016/j.nucmedbio.2011.02.002
View details for PubMedID 21843774
-
More
-
Spatial relationship of phosphorylated epidermal growth factor receptor and activated AKT in head and neck squamous cell carcinoma Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology
Nijkamp MM, Hoogsteen IJ, Span PN, Takes RP, Lok J, Rijken PF, Kogel vd, Bussink J, Kaanders AM
2011 Oct;101(1):165-70. doi: 10.1016/j.radonc.2011.06.022. Epub 2011 Jul 19.
-
More
BACKGROUND: Overexpression of EGFR correlates with decreased survival after radiotherapy in head and neck squamous cell carcinoma (HNSCC). However, the contribution of the activated form, pEGFR, and its downstream signaling (PI3-K/AKT) pathway is not clear yet.
METHODS: Fifty-eight patients with HNSCC were included in the study. pEGFR, pAKT, hypoxia, and vessels were visualized using immunohistochemistry. Fractions (defined as the tumor area positive for the respective markers relative to the total tumor area) were calculated by automated image analysis and related to clinical outcome.
RESULTS: Both pEGFR (median 0.6%, range 0-34%) and pAKT (median 1.8%, range 0-16%) expression differed between tumors. Also, a large variation in hypoxia was found (median pimonidazole fraction 3.9% 0-20%). A significant correlation between pEGFR and pAKT (r(s) 0.44, p=0.004) was seen, however, analysis revealed that this was not always based on spatial coexpression. Low pAKT expression was associated with increased risk of regional recurrence (p<0.05, log-rank) and distant metastasis (p=0.04).
CONCLUSION: The correlation between expression of pEGFR and pAKT is indicative of activation of the PI3-K/AKT pathway through phosphorylation of EGFR. Since not all tumors show coexpression to the same extent, other factors must be involved in the activation of this pathway as well.
PMID:21775008 | DOI:10.1016/j.radonc.2011.06.022
View details for PubMedID 21775008
-
More
-
Expression of E-cadherin and vimentin correlates with metastasis formation in head and neck squamous cell carcinoma patients Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology
Nijkamp MM, Span PN, Hoogsteen IJ, Kogel vd, Kaanders AM, Bussink J
2011 Jun;99(3):344-8. doi: 10.1016/j.radonc.2011.05.066. Epub 2011 Jun 22.
-
More
PURPOSE: E-cadherin is a transmembrane glycoprotein, involved in cell-cell adhesion and epithelial-mesenchymal transition (EMT). Vimentin is highly expressed in mesenchymal cells and is positively correlated with increased metastasis. Here we set out to determine the expression of E-cadherin and vimentin in head and neck squamous cell carcinomas (HNSCC).
PATIENTS AND METHODS: Twenty-six patients with primary stage II-IV HNSCC were included. E-cadherin and vimentin were visualised using immunohistochemistry, semi-automatically analysed for expression patterns and correlated with the clinical behaviour of these tumours.
RESULTS: A large variation in E-cadherin and vimentin expression was observed between tumours (median 17% range 0-51% respectively median 0% range 0-20%). Tumours with low E-cadherin expression showed a significantly higher incidence of metastasis formation compared to tumours with high expression (81% versus 19%, p=0.004). Enhanced expression of vimentin was associated with a trend towards a higher metastatic risk (33% versus 77%) compared to tumours without expression of vimentin. All patients with low E-cadherin and high vimentin expression (an EMT-phenotype) developed distant metastases versus only 44% of the other patients (p=0.008).
CONCLUSION: Loss of E-cadherin and gain of vimentin may be associated with enhanced migration of tumour cells, leading to higher metastatic risk of HNSCC patients.
PMID:21684617 | DOI:10.1016/j.radonc.2011.05.066
View details for PubMedID 21684617
-
More
-
Deregulation of cap-dependent mRNA translation increases tumour radiosensitivity through reduction of the hypoxic fraction Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology
Rouschop MA, Dubois L, Schaaf BE, Beucken vd, Lieuwes N, Keulers GH, Savelkouls GM, Bussink J, Kogel vd, Koritzinsky M, Wouters BG
2011 Jun;99(3):385-91. doi: 10.1016/j.radonc.2011.05.047. Epub 2011 Jun 12.
-
More
BACKGROUND AND PURPOSE: Tumour hypoxia is an important limiting factor in the successful treatment of cancer. Adaptation to hypoxia includes inhibition of mTOR, causing scavenging of eukaryotic initiation factor 4E (eIF4E), the rate-limiting factor for cap-dependent translation. The aim of this study was to determine the effect of preventing mTOR-dependent translation inhibition on hypoxic cell survival and tumour sensitivity towards irradiation.
MATERIAL AND METHODS: The effect of eIF4E-overexpression on cell proliferation, hypoxia-tolerance, and radiation sensitivity was assessed using isogenic, inducible U373 and HCT116 cells.
RESULTS: We found that eIF4E-overexpression significantly enhanced proliferation of cells under normal conditions, but not during hypoxia, caused by increased cell death during hypoxia. Furthermore, eIF4E-overexpression stimulated overall rates of tumour growth, but resulted in selective loss of hypoxic cells in established tumours and increased levels of necrosis. This markedly increased overall tumour sensitivity to irradiation.
CONCLUSIONS: Our results demonstrate that hypoxia induced inhibition of translational control through regulation of eIF4E is an important mediator of hypoxia tolerance and radioresistance of tumours. These data also demonstrate that deregulation of metabolic pathways such as mTOR can influence the proliferation and survival of tumour cells experiencing metabolic stress in opposite ways of nutrient replete cells.
PMID:21665307 | DOI:10.1016/j.radonc.2011.05.047
View details for PubMedID 21665307
-
More
-
Metabolic markers in relation to hypoxia; staining patterns and colocalization of pimonidazole, HIF-1α, CAIX, LDH-5, GLUT-1, MCT1 and MCT4 BMC cancer
Rademakers SE, Lok J, Kogel vd, Bussink J, Kaanders AM
2011 May 12;11:167. doi: 10.1186/1471-2407-11-167.
-
More
BACKGROUND: The cellular response of malignant tumors to hypoxia is diverse. Several important endogenous metabolic markers are upregulated under hypoxic conditions. We examined the staining patterns and co-expression of HIF-1α, CAIX, LDH-5, GLUT-1, MCT1 and MCT4 with the exogenous hypoxic cell marker pimonidazole and the association of marker expression with clinicopathological characteristics.
METHODS: 20 biopsies of advanced head and neck carcinomas were immunohistochemically stained and analyzed. All patients were given the hypoxia marker pimonidazole intravenously 2 h prior to biopsy taking. The tumor area positive for each marker, the colocalization of the different markers and the distribution of the markers in relation to the blood vessels were assessed by semiautomatic quantitative analysis.
RESULTS: MCT1 staining was present in hypoxic (pimonidazole stained) as well as non-hypoxic areas in almost equal amounts. MCT1 expression showed a significant overall correlation (r = 0.75, p < 0.001) and strong spatial relationship with CAIX. LDH-5 showed the strongest correlation with pimonidazole (r = 0.66, p = 0.002). MCT4 and GLUT-1 demonstrated a typical diffusion-limited hypoxic pattern and showed a high degree of colocalization. Both MCT4 and CAIX showed a higher expression in the primary tumor in node positive patients (p = 0.09 both).
CONCLUSIONS: Colocalization and staining patterns of metabolic and hypoxia-related proteins provides valuable additional information over single protein analyses and can improve the understanding of their functions and environmental influences.
PMID:21569415 | PMC:PMC3115911 | DOI:10.1186/1471-2407-11-167
View details for PubMedID 21569415
-
More
-
Spinal cord tolerance to single-fraction partial-volume irradiation: a swine model International journal of radiation oncology, biology, physics
Medin PM, Foster RD, Kogel vd, Sayre JW, McBride WH, Solberg TD
2011 Jan 1;79(1):226-32. doi: 10.1016/j.ijrobp.2010.07.1979. Epub 2010 Oct 8.
-
More
PURPOSE: To determine the spinal cord tolerance to single-fraction, partial-volume irradiation in swine.
METHODS AND MATERIALS: A 5-cm-long cervical segment was irradiated in 38-47-week-old Yucatan minipigs using a dedicated, image-guided radiosurgery linear accelerator. The radiation was delivered to a cylindrical volume approximately 5 cm in length and 2 cm in diameter that was positioned lateral to the cervical spinal cord, resulting in a dose distribution with the 90%, 50%, and 10% isodose lines traversing the ipsilateral, central, and contralateral spinal cord, respectively. The dose was prescribed to the 90% isodose line. A total of 26 pigs were stratified into eight dose groups of 12-47 Gy. The mean maximum spinal cord dose was 16.9 ± 0.1, 18.9 ± 0.1, 21.0 ± 0.1, 23.0 ± 0.2, and 25.3 ± 0.3 Gy in the 16-, 18-, 20-, 22-, and 24-Gy dose groups, respectively. The mean percentage of spinal cord volumes receiving ≥ 10 Gy for the same groups were 43% ± 3%, 48% ± 4%, 51% ± 2%, 57% ± 2%, and 59% ± 4%. The study endpoint was motor neurologic deficit determined by a change in gait during a 1-year follow-up period.
RESULTS: A steep dose-response curve was observed with a median effective dose for the maximum dose point of 20.0 Gy (95% confidence interval, 18.3-21.7). Excellent agreement was observed between the occurrence of neurologic change and the presence of histologic change. All the minipigs with motor deficits showed some degree of demyelination and focal white matter necrosis on the irradiated side, with relative sparing of the gray matter. The histologic findings were unremarkable in the minipigs with normal neurologic status.
CONCLUSIONS: Our results have indicated that for a dose distribution with a steep lateral gradient, the pigs had a lower median effective dose for paralysis than has been observed in rats and more closely resembles that for rats, mice, and guinea pigs receiving uniform spinal cord irradiation.
PMID:20934278 | PMC:PMC3005987 | DOI:10.1016/j.ijrobp.2010.07.1979
View details for PubMedID 20934278
-
More
-
Longitudinal magnetic resonance imaging-based assessment of vascular changes and radiation response in androgen-sensitive prostate carcinoma xenografts under androgen-exposed and androgen-deprived conditions Neoplasia (New York, N.Y.)
Røe K, Seierstad T, Kristian A, Mikalsen TG, Mælandsmo GM, Kogel vd, Ree AH, Olsen DR
2010 Oct;12(10):818-25. doi: 10.1593/neo.10484.
-
More
Prostate cancer (PCa) patients receive androgen-deprivation therapy (ADT) to reduce tumor burden. However, complete eradication of PCa is unusual, and recurrent disease is evident within approximately 2 years in high-risk patients. Clinical evidence suggests that combining ADT with radiotherapy improves local control and disease-free survival in these patients compared with radiotherapy alone. We investigated whether vascularization of androgen-sensitive PCa xenografts changed after ADT and whether such therapy affected radiation response. CWR22 xenografts received combinations of ADT by castration (CWR22-cas) and 15 Gy of single-dose irradiation. At a shortest tumor diameter of 8 mm, vascularization was visualized by dynamic contrast-enhanced magnetic resonance imaging before radiation and 1 and 9 days after radiation. Voxel-wise quantitative modeling of contrast enhancement curves extracted the hemodynamic parameter K(trans), reflecting a combination of permeability, density, and blood flow. Tumor volumes and prostate-specific antigen (PSA) were monitored during the experiment. The results showed that K(trans) of CWR22-cas tumors 36±4 days after ADT was 47.1% higher than K(trans) of CWR22 tumors (P = .01). CWR22-cas tumors showed no significant changes in K(trans) after radiation, whereas K(trans) of CWR22 tumors at day 1 decreased compared with pretreatment values (P = .04) before a continuous increase from day 1 to day 9 followed (P = .01). Total PSA in blood correlated positively to tumor volume (r = 0.59, P < .01). In conclusion, androgen-exposed xenografts demonstrated radiation-induced reductions in vascularization and tumor volumes, whereas androgen-deprived xenografts showed increased vascularization and growth inhibition, but no significant additive effect of radiation.
PMID:20927320 | PMC:PMC2950331 | DOI:10.1593/neo.10484
View details for PubMedID 20927320
-
More
-
Benign uterine uptake of FDG: a case report and review of literature The Netherlands journal of medicine
Vriens D, Geus-Oei Fd, Flucke UE, Kogel vd, Oyen JG, Vierhout ME, Meer vd
2010 Sep;68(9):379-80.
-
Effect of cetuximab and fractionated irradiation on tumour micro-environment Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology
Santiago A, Eicheler W, Bussink J, Rijken P, Yaromina A, Beuthien-Baumann B, Kogel vd, Baumann M, Krause M
2010 Nov;97(2):322-9. doi: 10.1016/j.radonc.2010.07.007.
-
More
BACKGROUND AND PURPOSE: Previous experiments have shown that application of the anti-EGFR monoclonal antibody C225 (cetuximab) improves local tumour control after irradiation in FaDu human squamous cell carcinoma (hSCC) due to the combined effect of decreased repopulation and improved reoxygenation. The present study investigates early changes of the pimonidazole hypoxic fraction of FaDu tumours and the expression and phosphorylation of the EGFR and its downstream signal transduction molecules after treatment with C225 alone or in combination with irradiation.
MATERIAL AND METHODS: FaDu tumour xenografts were irradiated with up to 3×3Gy with or without additional C225 treatment and excised at different time points. Tumour hypoxia was evaluated using pimonidazole. EGFR expression and phosphorylation and intratumoural distribution of C225 were assessed by immunofluorescence analysis. Western blots were performed to evaluate expression and phosphorylation of EGFR, ErbB2, AKT and MAPK (ERK1/2).
RESULTS: Hypoxia did not change during the 4days of treatment in the tumours treated with C225 alone or combined with irradiation. C225 treatment led to downregulation of the total EGFR in FaDu tumours, accompanied by a change of the spatial distribution of the receptor favouring the membranous expression. An induction of phosphorylation of the EGFR (tyr992, tyr1173) was observed with C225 alone or combined with irradiation. AKT phosphorylation was decreased, whereas MAPK phosphorylation remained unchanged. C225 membrane staining was homogeneously distributed over the whole tumour with no differences between hypoxic and non-hypoxic tumour cells.
CONCLUSION: Pimonidazole-hypoxia of FaDu tumours during the initial part of fractionated irradiation is not influenced by C225, indicating that external hypoxia markers may not be promising as biomarkers for tumour response to combined treatment. The downregulation of the total EGFR, but at the same time higher membrane staining, as well as the changes in downstream signal transduction molecules, warrants further investigation in other tumour models.
PMID:20667608 | DOI:10.1016/j.radonc.2010.07.007
View details for PubMedID 20667608
-
More
-
Augmentation of radiation response by motesanib, a multikinase inhibitor that targets vascular endothelial growth factor receptors Clinical cancer research : an official journal of the American Association for Cancer Research
Kruser TJ, Wheeler DL, Armstrong EA, Iida M, Kozak KR, Kogel vd, Bussink J, Coxon A, Polverino A, Harari PM
2010 Jul 15;16(14):3639-47. doi: 10.1158/1078-0432.CCR-09-3385. Epub 2010 May 27.
-
More
BACKGROUND: Motesanib is a potent inhibitor of vascular endothelial growth factor receptors (VEGFR) 1, 2, and 3, platelet-derived growth factor receptor, and Kit receptors. In this report we examine the interaction between motesanib and radiation in vitro and in head and neck squamous cell carcinoma (HNSCC) xenograft models.
EXPERIMENTAL DESIGN: In vitro assays were done to assess the impact of motesanib on VEGFR2 signaling pathways in human umbilical vein endothelial cells (HUVEC). HNSCC lines grown as tumor xenografts in athymic nude mice were utilized to assess the in vivo activity of motesanib alone and in combination with radiation.
RESULTS: Motesanib inhibited VEGF-stimulated HUVEC proliferation in vitro, as well as VEGFR2 kinase activity. Additionally, motesanib and fractionated radiation showed additive inhibitory effects on HUVEC proliferation. In vivo combination therapy with motesanib and radiation showed increased response compared with drug or radiation alone in UM-SCC1 (P < 0.002) and SCC-1483 xenografts (P = 0.001); however, the combination was not significantly more efficacious than radiation alone in UM-SCC6 xenografts. Xenografts treated with motesanib showed a reduction of vessel penetration into tumor parenchyma, compared with control tumors. Furthermore, triple immunohistochemical staining for vasculature, proliferation, and hypoxia showed well-defined spatial relationships among these parameters in HNSCC xenografts. Motesanib significantly enhanced intratumoral hypoxia in the presence and absence of fractionated radiation.
CONCLUSIONS: These studies identify a favorable interaction when combining radiation and motesanib in HNSCC models. The data presented suggest that motesanib reduces blood vessel penetration into tumors and thereby increases intratumoral hypoxia. These findings suggest that clinical investigations examining combinations of radiation and motesanib are warranted in HNSCC.
PMID:20507929 | PMC:PMC3216115 | DOI:10.1158/1078-0432.CCR-09-3385
View details for PubMedID 20507929
-
More
-
Assessment of Blood Hemodynamics by USPIO-Induced R(1) Changes in MRI of Murine Colon Carcinoma Applied magnetic resonance
Gambarota G, van Laarhoven M, Philippens M, Peeters JM, Rijken P, Kogel vd, Punt JA, Heerschap A
2010 Jun;38(3):349-360. doi: 10.1007/s00723-010-0124-1. Epub 2010 Mar 28.
-
More
The objective of this study is to assess whether ultrasmall superparamagnetic iron oxide (USPIO)-induced changes of the water proton longitudinal relaxation rate (R(1)) provide a means to assess blood hemodynamics of tumors. Two types of murine colon tumors (C26a and C38) were investigated prior to and following administration of USPIO blood-pool contrast agent with fast R(1) measurements. In a subpopulation of mice, R(1) was measured following administration of hydralazine, a well-known blood hemodynamic modifier. USPIO-induced R(1) increase in C38 tumors (DeltaR(1) = 0.072 +/- 0.0081 s(-1)) was significantly larger than in C26a tumors (DeltaR(1) = 0.032 +/- 0.0018 s(-1), N = 9, t test, P < 0.001). This was in agreement with the immunohistochemical data that showed higher values of relative vascular area (RVA) in C38 tumors than in C26a tumors (RVA = 0.059 +/- 0.015 vs. 0.020 +/- 0.011; P < 0.05). Following administration of hydralazine, a decrease in R(1) value was observed. This was consistent with the vasoconstriction induced by the steal effect mechanism. In conclusion, R(1) changes induced by USPIO are sensitive to tumor vascular morphology and to blood hemodynamics. Thus, R(1) measurements following USPIO administration can give novel insight into the effects of blood hemodynamic modifiers, non-invasively and with a high temporal resolution.
PMID:20502507 | PMC:PMC2866959 | DOI:10.1007/s00723-010-0124-1
View details for PubMedID 20502507
-
More
-
Histopathologic validation of 3'-deoxy-3'-18F-fluorothymidine PET in squamous cell carcinoma of the oral cavity Journal of nuclear medicine : official publication, Society of Nuclear Medicine
Troost GC, Bussink J, Slootweg PJ, Peeters JM, Merkx AW, Kogel vd, Oyen JG, Kaanders AM
2010 May;51(5):713-9. doi: 10.2967/jnumed.109.071910. Epub 2010 Apr 15.
-
More
Accelerated tumor cell repopulation is an important mechanism adversely affecting therapeutic outcome in head and neck cancer. The noninvasive assessment of the proliferative state of a tumor by PET may provide a selection tool for customized treatment. 3'-deoxy-3'-(18)F-fluorothymidine ((18)F-FLT) is a PET tracer that is phosphorylated by thymidine kinase 1 (TK-1) and, as such, reflects cellular proliferation. Before the use of (18)F-FLT PET for tumor characterization is accepted and introduced into clinical studies, validation against tumor histology is mandatory. The aim of this study was to validate (18)F-FLT PET in squamous cell carcinomas of the oral cavity using immunohistochemical staining for the proliferation marker iododeoxyuridine and for TK-1.
METHODS: Seventeen patients with primary squamous cell carcinomas of the oral cavity underwent an (18)F-FLT PET/CT scan before surgery, and iododeoxyuridine was administered 20 min before tumor resection. (18)F-FLT PET/CT scans were segmented, and PET/CT volumes and PET signal intensities were calculated (mean standardized uptake value [SUV(mean)] and maximum standardized uptake value [SUV(max)]). Multiple paraffin-embedded tumor sections were immunohistochemically stained for iododeoxyuridine and TK-1. For iododeoxyuridine, labeling indices and optical densities were calculated and correlated with SUV(mean) and SUV(max). TK-1 staining was visually and semiquantitatively assessed.
RESULTS: All primary tumors were identified with (18)F-FLT PET but with a large range in tracer uptake (mean SUV(max), 5.9; range, 2.2-15.2). Also, there was a large variability in iododeoxyuridine labeling indices (mean, 0.09; range, 0.01-0.29) and optical densities (mean, 28.2; range, 12.6-37.8). The iododeoxyuridine optical densities correlated significantly with SUV(mean) and SUV(max), but the labeling indices did not. In most tumors, TK-1 staining of varying intensity was present but correlated with neither iododeoxyuridine binding nor (18)F-FLT uptake.
CONCLUSION: The current study demonstrated only a weak correlation between (18)F-FLT uptake and iododeoxyuridine staining intensity in oral cavity tumors. This weak correlation may be explained by differences in biomarker characteristics, resolution, and quantification methods.
PMID:20395329 | DOI:10.2967/jnumed.109.071910
View details for PubMedID 20395329
-
More
-
Radiation dose-volume effects in the spinal cord International journal of radiation oncology, biology, physics
Kirkpatrick JP, Kogel vd, Schultheiss TE
2010 Mar 1;76(3 Suppl):S42-9. doi: 10.1016/j.ijrobp.2009.04.095.
-
More
Dose-volume data for myelopathy in humans treated with radiotherapy (RT) to the spine is reviewed, along with pertinent preclinical data. Using conventional fractionation of 1.8-2 Gy/fraction to the full-thickness cord, the estimated risk of myelopathy is <1% and <10% at 54 Gy and 61 Gy, respectively, with a calculated strong dependence on dose/fraction (alpha/beta = 0.87 Gy.) Reirradiation data in animals and humans suggest partial repair of RT-induced subclinical damage becoming evident about 6 months post-RT and increasing over the next 2 years. Reports of myelopathy from stereotactic radiosurgery to spinal lesions appear rare (<1%) when the maximum spinal cord dose is limited to the equivalent of 13 Gy in a single fraction or 20 Gy in three fractions. However, long-term data are insufficient to calculate a dose-volume relationship for myelopathy when the partial cord is treated with a hypofractionated regimen.
PMID:20171517 | DOI:10.1016/j.ijrobp.2009.04.095
View details for PubMedID 20171517
-
More
-
The unfolded protein response protects human tumor cells during hypoxia through regulation of the autophagy genes MAP1LC3B and ATG5 The Journal of clinical investigation
Rouschop MA, Beucken vd, Dubois L, Niessen H, Bussink J, Savelkouls K, Keulers T, Mujcic H, Landuyt W, Voncken JW, Lambin P, Kogel vd, Koritzinsky M, Wouters BG
2010 Jan;120(1):127-41. doi: 10.1172/JCI40027. Epub 2009 Dec 14.
-
More
Tumor hypoxia is a common microenvironmental factor that adversely influences tumor phenotype and treatment response. Cellular adaptation to hypoxia occurs through multiple mechanisms, including activation of the unfolded protein response (UPR). Recent reports have indicated that hypoxia activates a lysosomal degradation pathway known as autophagy, and here we show that the UPR enhances the capacity of hypoxic tumor cells to carry out autophagy, and that this promotes their survival. In several human cancer cell lines, hypoxia increased transcription of the essential autophagy genes microtubule-associated protein 1 light chain 3beta (MAP1LC3B) and autophagy-related gene 5 (ATG5) through the transcription factors ATF4 and CHOP, respectively, which are regulated by PKR-like ER kinase (PERK, also known as EIF2AK3). MAP1LC3B and ATG5 are not required for initiation of autophagy but mediate phagophore expansion and autophagosome formation. We observed that transcriptional induction of MAP1LC3B replenished MAP1LC3B protein that was turned over during extensive hypoxia-induced autophagy. Correspondingly, cells deficient in PERK signaling failed to transcriptionally induce MAP1LC3B and became rapidly depleted of MAP1LC3B protein during hypoxia. Consistent with these data, autophagy and MAP1LC3B induction occurred preferentially in hypoxic regions of human tumor xenografts. Furthermore, pharmacological inhibition of autophagy sensitized human tumor cells to hypoxia, reduced the fraction of viable hypoxic tumor cells, and sensitized xenografted human tumors to irradiation. Our data therefore demonstrate that the UPR is an important mediator of the hypoxic tumor microenvironment and that it contributes to resistance to treatment through its ability to facilitate autophagy.
PMID:20038797 | PMC:PMC2798689 | DOI:10.1172/JCI40027
View details for PubMedID 20038797
-
More
Contact Information
Albert van der Kogel, PhD
1111 Highland Avenue, 3137 WIMR,Madison, WI 53705