1
|
Gertsenshteyn I, Giurcanu M, Vaupel P, Halpern H. Biological validation of electron paramagnetic resonance (EPR) image oxygen thresholds in tissue. J Physiol 2021; 599:1759-1767. [PMID: 32506448 PMCID: PMC7719598 DOI: 10.1113/jp278816] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/26/2020] [Indexed: 12/17/2022] Open
Abstract
Measuring molecular oxygen levels in vivo has been the cornerstone of understanding the effects of hypoxia in normal tissues and malignant tumors. Here we discuss the advances in a variety of partial pressure of oxygen ( P O 2 ) measurements and imaging techniques and relevant oxygen thresholds. A focus on electron paramagnetic resonance (EPR) imaging shows the validation of treating hypoxic tumours with a threshold of P O 2 ≤ 10 Torr, and demonstrates utility for in vivo oxygen imaging, as well as its current and future role in cancer studies.
Collapse
Affiliation(s)
- Inna Gertsenshteyn
- Department of Radiology, University of Chicago, IL, USA
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
- Center for EPR Imaging In Vivo Physiology, University of Chicago, Chicago, IL, USA
| | - Mihai Giurcanu
- Department of Public Health Sciences, University of Chicago, IL, USA
| | - Peter Vaupel
- Department of Radiation Oncology, Medical Center, University of Freiburg, Germany
- German Cancer Consortium (DKTK), Partner site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Howard Halpern
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
- Center for EPR Imaging In Vivo Physiology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
2
|
Imaging Hypoxia. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00074-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
3
|
Xie G, Liu Y, Yao Q, Zheng R, Zhang L, Lin J, Guo Z, Du S, Ren C, Yuan Q, Yuan Y. Hypoxia-induced angiotensin II by the lactate-chymase-dependent mechanism mediates radioresistance of hypoxic tumor cells. Sci Rep 2017; 7:42396. [PMID: 28205588 PMCID: PMC5311966 DOI: 10.1038/srep42396] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 01/09/2017] [Indexed: 01/12/2023] Open
Abstract
The renin-angiotensin system (RAS) is a principal determinant of arterial blood pressure and fluid and electrolyte balance. RAS component dysregulation was recently found in some malignancies and correlated with poor patient outcomes. However, the exact mechanism of local RAS activation in tumors is still unclear. Here, we find that the local angiotensin II predominantly exists in the hypoxic regions of tumor formed by nasopharyngeal carcinoma CNE2 cells and breast cancer MDA-MB-231 cells, where these tumor cells autocrinely produce angiotensin II by a chymase-dependent rather than an angiotensin converting enzyme-dependent mechanism. We further demonstrate in nasopharyngeal carcinoma CNE2 and 5–8F cells that this chymase-dependent effect is mediated by increased levels of lactate, a by-product of glycolytic metabolism. Finally, we show that the enhanced angiotensin II plays an important role in the intracellular accumulation of HIF-1α of hypoxic nasopharyngeal carcinoma cells and mediates the radiation-resistant phenotype of these nasopharyngeal carcinoma cells. Thus, our findings reveal the critical role of hypoxia in producing local angiotensin II by a lactate-chymase-dependent mechanism and highlight the importance of local angiotensin II in regulating radioresistance of hypoxic tumor cells.
Collapse
Affiliation(s)
- Guozhu Xie
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Ying Liu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qiwei Yao
- Department of Radiation Oncology, Teaching Hospital of Fujian Provincial Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| | - Rong Zheng
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Lanfang Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jie Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zhaoze Guo
- Breast Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Shasha Du
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Chen Ren
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Quan Yuan
- Jules Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Yawei Yuan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Department of Radiation Oncology, Cancer Center of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| |
Collapse
|
4
|
Lewin J, Khamly KK, Young RJ, Mitchell C, Hicks RJ, Toner GC, Ngan SYK, Chander S, Powell GJ, Herschtal A, Te Marvelde L, Desai J, Choong PFM, Stacker SA, Achen MG, Ferris N, Fox S, Slavin J, Thomas DM. A phase Ib/II translational study of sunitinib with neoadjuvant radiotherapy in soft-tissue sarcoma. Br J Cancer 2014; 111:2254-61. [PMID: 25321190 PMCID: PMC4264446 DOI: 10.1038/bjc.2014.537] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 08/29/2014] [Accepted: 09/17/2014] [Indexed: 12/16/2022] Open
Abstract
Background: Preoperative radiotherapy (RT) is commonly used to treat localised soft-tissue sarcomas (STS). Hypoxia is an important determinant of radioresistance. Whether antiangiogenic therapy can ‘normalise' tumour vasculature, thereby improving oxygenation, remains unknown. Methods: Two cohorts were prospectively enrolled. Cohort A evaluated the implications of hypoxia in STS, using the hypoxic tracer 18F-azomycin arabinoside (FAZA-PET). In cohort B, sunitinib was added to preoperative RT in a dose-finding phase 1b/2 design. Results: In cohort A, 13 out of 23 tumours were hypoxic (FAZA-PET), correlating with metabolic activity (r2=0.85; P<0.001). Two-year progression-free (PFS) and overall (OS) survival were 61% (95% CI: 0.44–0.84) and 87% (95% CI: 0.74–1.00), respectively. Hypoxia was associated with radioresistance (P=0.012), higher local recurrence (Hazard ratio (HR): 10.2; P=0.02), PFS (HR: 8.4; P=0.02), and OS (HR: 41.4; P<0.04). In Cohort B, seven patients received sunitinib at dose level (DL): 0 (50 mg per day for 2 weeks before RT; 25 mg per day during RT) and two patients received DL: −1 (37.5 mg per day for entire period). Dose-limiting toxicities were observed in 4 out of 7 patients at DL 0 and 2 out of 2 patients at DL −1, resulting in premature study closure. Although there was no difference in PFS or OS, patients receiving sunitinib had higher local failure (HR: 8.1; P=0.004). Conclusion: In STS, hypoxia is associated with adverse outcomes. The combination of sunitinib with preoperative RT resulted in unacceptable toxicities, and higher local relapse rates.
Collapse
Affiliation(s)
- J Lewin
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - K K Khamly
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - R J Young
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - C Mitchell
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - R J Hicks
- 1] Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia [2] The University of Melbourne, St Vincent's Hospital Campus, Fitzroy, Victoria, Australia
| | - G C Toner
- 1] Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia [2] The University of Melbourne, St Vincent's Hospital Campus, Fitzroy, Victoria, Australia
| | - S Y K Ngan
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - S Chander
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - G J Powell
- 1] Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia [2] Department of Orthopaedics, St. Vincent's Hospital, Fitzroy, Victoria, Australia [3] Department of Surgery, The University of Melbourne, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - A Herschtal
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - L Te Marvelde
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - J Desai
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - P F M Choong
- 1] Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia [2] Department of Orthopaedics, St. Vincent's Hospital, Fitzroy, Victoria, Australia [3] Department of Surgery, The University of Melbourne, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - S A Stacker
- 1] Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia [2] Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - M G Achen
- 1] Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia [2] Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - N Ferris
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - S Fox
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - J Slavin
- The University of Melbourne, St Vincent's Hospital Campus, Fitzroy, Victoria, Australia
| | - D M Thomas
- 1] Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia [2] The University of Melbourne, St Vincent's Hospital Campus, Fitzroy, Victoria, Australia [3] The Kinghorn Cancer Centre, Garvan Institute of Medical Research, 370 Victoria Street, Darlinghurst, New South Wales 2010, Australia
| |
Collapse
|
5
|
Chawla SP, Cranmer LD, Van Tine BA, Reed DR, Okuno SH, Butrynski JE, Adkins DR, Hendifar AE, Kroll S, Ganjoo KN. Phase II study of the safety and antitumor activity of the hypoxia-activated prodrug TH-302 in combination with doxorubicin in patients with advanced soft tissue sarcoma. J Clin Oncol 2014; 32:3299-306. [PMID: 25185097 DOI: 10.1200/jco.2013.54.3660] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE TH-302, a prodrug of the cytotoxic alkylating agent bromo-isophosphoramide mustard, is preferentially activated in hypoxic conditions. This phase II study investigated TH-302 in combination with doxorubicin, followed by single-agent TH-302 maintenance therapy in patients with first-line advanced soft tissue sarcoma (STS) to assess progression-free survival (PFS), response rate, overall survival, safety, and tolerability. PATIENTS AND METHODS In this open-label phase II study, TH-302 300 mg/m(2) was administered intravenously on days 1 and 8 with doxorubicin 75 mg/m(2) on day 1 of each 21-day cycle. After six cycles, patients with stable and/or responding disease could receive maintenance monotherapy with TH-302. RESULTS Ninety-one patients initiated TH-302 plus doxorubicin induction treatment. The PFS rate at 6 months (primary efficacy measure) was 58% (95% CI, 46% to 68%). Median PFS was 6.5 months (95% CI, 5.8 to 7.7 months); median overall survival was 21.5 months (95% CI, 16.0 to 26.2 months). Best tumor responses were complete response (n = 2 [2%]) and partial response (n = 30 [34%]). During TH-302 maintenance (n = 48), five patients improved from stable disease to partial response, and one patient improved from partial to complete response. The most common adverse events during induction were fatigue, nausea, and skin and/or mucosal toxicities as well as anemia, thrombocytopenia, and neutropenia. These were less severe and less frequent during maintenance. There was no evidence of TH-302-related hepatic, renal, or cardiac toxicity. CONCLUSION PFS, overall survival, and tumor response compared favorably with historical outcomes achieved with other first-line chemotherapies for advanced STS. A phase III study of TH-302 is ongoing (NCT01440088).
Collapse
Affiliation(s)
- Sant P Chawla
- Sant P. Chawla, Sarcoma Oncology Center, Santa Monica; Andrew E. Hendifar, Cedars Sinai Medical Center, Los Angeles; Stew Kroll, Threshold Pharmaceuticals, South San Francisco; Kristen N. Ganjoo, Stanford University Medical Center, Stanford, CA; Lee D. Cranmer, University of Arizona Cancer Center, Tucson, AZ; Brian A. Van Tine and Douglas R. Adkins, Washington University in St Louis, St Louis, MO; Damon R. Reed, Moffitt Cancer Center and Research Institute, Tampa, FL; Scott H. Okuno, Mayo Clinic, Rochester, MN; and James E. Butrynski, Dana-Farber Cancer Institute, Boston, MA.
| | - Lee D Cranmer
- Sant P. Chawla, Sarcoma Oncology Center, Santa Monica; Andrew E. Hendifar, Cedars Sinai Medical Center, Los Angeles; Stew Kroll, Threshold Pharmaceuticals, South San Francisco; Kristen N. Ganjoo, Stanford University Medical Center, Stanford, CA; Lee D. Cranmer, University of Arizona Cancer Center, Tucson, AZ; Brian A. Van Tine and Douglas R. Adkins, Washington University in St Louis, St Louis, MO; Damon R. Reed, Moffitt Cancer Center and Research Institute, Tampa, FL; Scott H. Okuno, Mayo Clinic, Rochester, MN; and James E. Butrynski, Dana-Farber Cancer Institute, Boston, MA
| | - Brian A Van Tine
- Sant P. Chawla, Sarcoma Oncology Center, Santa Monica; Andrew E. Hendifar, Cedars Sinai Medical Center, Los Angeles; Stew Kroll, Threshold Pharmaceuticals, South San Francisco; Kristen N. Ganjoo, Stanford University Medical Center, Stanford, CA; Lee D. Cranmer, University of Arizona Cancer Center, Tucson, AZ; Brian A. Van Tine and Douglas R. Adkins, Washington University in St Louis, St Louis, MO; Damon R. Reed, Moffitt Cancer Center and Research Institute, Tampa, FL; Scott H. Okuno, Mayo Clinic, Rochester, MN; and James E. Butrynski, Dana-Farber Cancer Institute, Boston, MA
| | - Damon R Reed
- Sant P. Chawla, Sarcoma Oncology Center, Santa Monica; Andrew E. Hendifar, Cedars Sinai Medical Center, Los Angeles; Stew Kroll, Threshold Pharmaceuticals, South San Francisco; Kristen N. Ganjoo, Stanford University Medical Center, Stanford, CA; Lee D. Cranmer, University of Arizona Cancer Center, Tucson, AZ; Brian A. Van Tine and Douglas R. Adkins, Washington University in St Louis, St Louis, MO; Damon R. Reed, Moffitt Cancer Center and Research Institute, Tampa, FL; Scott H. Okuno, Mayo Clinic, Rochester, MN; and James E. Butrynski, Dana-Farber Cancer Institute, Boston, MA
| | - Scott H Okuno
- Sant P. Chawla, Sarcoma Oncology Center, Santa Monica; Andrew E. Hendifar, Cedars Sinai Medical Center, Los Angeles; Stew Kroll, Threshold Pharmaceuticals, South San Francisco; Kristen N. Ganjoo, Stanford University Medical Center, Stanford, CA; Lee D. Cranmer, University of Arizona Cancer Center, Tucson, AZ; Brian A. Van Tine and Douglas R. Adkins, Washington University in St Louis, St Louis, MO; Damon R. Reed, Moffitt Cancer Center and Research Institute, Tampa, FL; Scott H. Okuno, Mayo Clinic, Rochester, MN; and James E. Butrynski, Dana-Farber Cancer Institute, Boston, MA
| | - James E Butrynski
- Sant P. Chawla, Sarcoma Oncology Center, Santa Monica; Andrew E. Hendifar, Cedars Sinai Medical Center, Los Angeles; Stew Kroll, Threshold Pharmaceuticals, South San Francisco; Kristen N. Ganjoo, Stanford University Medical Center, Stanford, CA; Lee D. Cranmer, University of Arizona Cancer Center, Tucson, AZ; Brian A. Van Tine and Douglas R. Adkins, Washington University in St Louis, St Louis, MO; Damon R. Reed, Moffitt Cancer Center and Research Institute, Tampa, FL; Scott H. Okuno, Mayo Clinic, Rochester, MN; and James E. Butrynski, Dana-Farber Cancer Institute, Boston, MA
| | - Douglas R Adkins
- Sant P. Chawla, Sarcoma Oncology Center, Santa Monica; Andrew E. Hendifar, Cedars Sinai Medical Center, Los Angeles; Stew Kroll, Threshold Pharmaceuticals, South San Francisco; Kristen N. Ganjoo, Stanford University Medical Center, Stanford, CA; Lee D. Cranmer, University of Arizona Cancer Center, Tucson, AZ; Brian A. Van Tine and Douglas R. Adkins, Washington University in St Louis, St Louis, MO; Damon R. Reed, Moffitt Cancer Center and Research Institute, Tampa, FL; Scott H. Okuno, Mayo Clinic, Rochester, MN; and James E. Butrynski, Dana-Farber Cancer Institute, Boston, MA
| | - Andrew E Hendifar
- Sant P. Chawla, Sarcoma Oncology Center, Santa Monica; Andrew E. Hendifar, Cedars Sinai Medical Center, Los Angeles; Stew Kroll, Threshold Pharmaceuticals, South San Francisco; Kristen N. Ganjoo, Stanford University Medical Center, Stanford, CA; Lee D. Cranmer, University of Arizona Cancer Center, Tucson, AZ; Brian A. Van Tine and Douglas R. Adkins, Washington University in St Louis, St Louis, MO; Damon R. Reed, Moffitt Cancer Center and Research Institute, Tampa, FL; Scott H. Okuno, Mayo Clinic, Rochester, MN; and James E. Butrynski, Dana-Farber Cancer Institute, Boston, MA
| | - Stew Kroll
- Sant P. Chawla, Sarcoma Oncology Center, Santa Monica; Andrew E. Hendifar, Cedars Sinai Medical Center, Los Angeles; Stew Kroll, Threshold Pharmaceuticals, South San Francisco; Kristen N. Ganjoo, Stanford University Medical Center, Stanford, CA; Lee D. Cranmer, University of Arizona Cancer Center, Tucson, AZ; Brian A. Van Tine and Douglas R. Adkins, Washington University in St Louis, St Louis, MO; Damon R. Reed, Moffitt Cancer Center and Research Institute, Tampa, FL; Scott H. Okuno, Mayo Clinic, Rochester, MN; and James E. Butrynski, Dana-Farber Cancer Institute, Boston, MA
| | - Kristen N Ganjoo
- Sant P. Chawla, Sarcoma Oncology Center, Santa Monica; Andrew E. Hendifar, Cedars Sinai Medical Center, Los Angeles; Stew Kroll, Threshold Pharmaceuticals, South San Francisco; Kristen N. Ganjoo, Stanford University Medical Center, Stanford, CA; Lee D. Cranmer, University of Arizona Cancer Center, Tucson, AZ; Brian A. Van Tine and Douglas R. Adkins, Washington University in St Louis, St Louis, MO; Damon R. Reed, Moffitt Cancer Center and Research Institute, Tampa, FL; Scott H. Okuno, Mayo Clinic, Rochester, MN; and James E. Butrynski, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
6
|
Ganjoo KN, Cranmer LD, Butrynski JE, Rushing D, Adkins D, Okuno SH, Lorente G, Kroll S, Langmuir VK, Chawla SP. A phase I study of the safety and pharmacokinetics of the hypoxia-activated prodrug TH-302 in combination with doxorubicin in patients with advanced soft tissue sarcoma. Oncology 2011; 80:50-6. [PMID: 21625179 DOI: 10.1159/000327739] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2011] [Accepted: 02/23/2011] [Indexed: 11/19/2022]
Abstract
PURPOSE The purpose of this study was to determine the dose-limiting toxicities (DLT), maximum tolerated dose (MTD), safety, pharmacokinetics and preliminary activity of TH-302, a hypoxia-activated prodrug, in combination with doxorubicin in patients with advanced soft tissue sarcoma. PATIENTS AND METHODS TH-302 was administered intravenously on days 1 and 8 and doxorubicin 75 mg/m² on day 1 (2 h after TH-302) of every 3-week cycle. TH-302 starting dose was 240 mg/m² with a classic 3 + 3 dose escalation. Pharmacokinetics were assessed on days 1 and 8 of cycle 1. Tumor assessments were performed after every second cycle. RESULTS Sixteen patients enrolled. Prophylactic growth factor support was added due to grade 4 neutropenia. The MTD was 300 mg/m². DLTs at 340 mg/m² were neutropenia-associated infection and grade 4 thrombocytopenia. Common adverse events included fatigue, nausea and skin rash. There was no evidence of pharmacokinetic interaction between TH-302 and doxorubicin. Five of 15 (33%) evaluable patients had a partial response by RECIST (Response Evaluation Criteria in Solid Tumors) criteria. CONCLUSIONS The hematologic toxicity of doxorubicin is increased when combined with TH-302. This can be mitigated by prophylactic growth factor support. Toxicities were manageable and there was evidence of antitumor activity.
Collapse
|
7
|
Yang M, Gao H, Sun X, Yan Y, Quan Q, Zhang W, Mohamedali KA, Rosenblum MG, Niu G, Chen X. Multiplexed PET probes for imaging breast cancer early response to VEGF₁₂₁/rGel treatment. Mol Pharm 2011; 8:621-8. [PMID: 21280671 DOI: 10.1021/mp100446t] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In this study, we applied multiplexed positron emission tomography (PET) probes to monitor glucose metabolism, cellular proliferation, tumor hypoxia and angiogenesis during VEGF₁₂₁/rGel therapy of breast cancer. Two doses of 12 mg/kg VEGF₁₂₁/rGel, administered intraperitoneally, resulted in initial delay of tumor growth, but the growth resumed 4 days after tumor treatment was stopped. The average tumor growth rate expressed as V/V(0), were 1.11 ± 0.07, 1.21 ± 0.10, 1.58 ± 0.36 and 2.64 ± 0.72 at days 1, 3, 7 and 14, respectively. Meanwhile, the VEGF₁₂₁/rGel treatment group showed V/V₀ ratios of 1.04 ± 0.06, 1.05 ± 0.11, 1.09 ± 0.17 and 1.86 ± 0.36 at days 1, 3, 7 and 14, respectively. VEGF₁₂₁/rGel treatment led to significantly decreased uptake of ¹⁸F-FPPRGD2 at day 1 (24.0 ± 8.8%, p < 0.05) and day 3 (36.3 ± 9.2%, p < 0.01), relative to the baseline, which slowly recovered to the baseline at day 14. ¹⁸F-FMISO uptake was increased in the treated tumors at day 1 (23.9 ± 15.7%, p < 0.05) and day 3 (51.4 ± 29.4%, p < 0.01), as compared to the control group. At days 7 and 14, ¹⁸F-FMISO uptake restored to the baseline level. The relative reductions in FLT uptake in treated tumors were approximately 13.0 ± 4.5% at day 1 and 25.0 ± 4.4% (p < 0.01) at day 3. No significant change of ¹⁸F-FDG uptake was observed in VEGF₁₂₁/rGel treated tumors, compared with the control group. The imaging findings were supported by ex vivo analysis of related biomarkers. Overall, longitudinal imaging studies with 4 PET tracers demonstrated the feasibility and usefulness of multiplexed probes for quantitative measurement of antitumor effects of VEGF₁₂₁/rGel at the early stage of treatment. This preclinical study should be helpful in accelerating anticancer drug development and promoting the clinical translation of molecular imaging.
Collapse
Affiliation(s)
- Min Yang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Wang K, Yorke E, Nehmeh SA, Humm JL, Ling CC. Modeling acute and chronic hypoxia using serial images of 18F-FMISO PET. Med Phys 2009; 36:4400-8. [PMID: 19928070 PMCID: PMC2852451 DOI: 10.1118/1.3213092] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Revised: 07/24/2009] [Accepted: 07/29/2009] [Indexed: 11/07/2022] Open
Abstract
PURPOSE Two types of tumor hypoxia most likely exist in human cancers: Chronic hypoxia due to the paucity of blood capillaries and acute hypoxia due to temporary shutdoWn of microvasculatures or fluctuation in the red cell flux. In a recent hypoxia imaging study using 18F-FMISO PET, the authors observed variation in tracer uptake in two sequential images and hypothesized that variation in acute hypoxia may be the cause. In this study, they develop an iterative optimization method to delineate chronic and acute hypoxia based on the 18F-FMISO PET serial images. METHODS They assume that (1) chronic hypoxia is the same in the two scans and can be represented by a Gaussian distribution, while (2) acute hypoxia varies in the two scans and can be represented by Poisson distributions. For validation, they used Monte Carlo simulations to generate pairs of 18F-FMISO PET images with known proportion of chronic and acute hypoxia and then applied the optimization method to the simulated serial images, yielding excellent fit between the input and the fitted results. They then applied this method to the serial 18F-FMISO PET images of 14 patients with head and neck cancers. RESULTS The results show good fit of the chronic hypoxia to Gaussian distributions for 13 out of 14 patients (with R2>0.7). Similarly, acute hypoxia appears to be well described by the Poisson distribution (R2>0.7) with three exceptions. The model calculation yielded the amount of acute hypoxia, which differed among the patients, ranging from approximately 13% to 52%, with an average of approximately 34%. CONCLUSIONS This is the first effort to separate acute and chronic hypoxia from serial PET images of cancer patients.
Collapse
Affiliation(s)
- Kelin Wang
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, 1250 1st Avenue, New York, New York 10065, USA
| | | | | | | | | |
Collapse
|
9
|
Yoshii Y, Furukawa T, Yoshii H, Mori T, Kiyono Y, Waki A, Kobayashi M, Tsujikawa T, Kudo T, Okazawa H, Yonekura Y, Fujibayashi Y. Cytosolic acetyl-CoA synthetase affected tumor cell survival under hypoxia: the possible function in tumor acetyl-CoA/acetate metabolism. Cancer Sci 2009; 100:821-7. [PMID: 19445015 PMCID: PMC11158093 DOI: 10.1111/j.1349-7006.2009.01099.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Understanding tumor-specific metabolism under hypoxia is important to find novel targets for antitumor drug design. Here we found that tumor cells expressed higher levels of cytosolic acetyl-CoA synthetase (ACSS2) under hypoxia than normoxia. Knockdown of ACSS2 by RNA interference (RNAi) in tumor cells enhanced tumor cell death under long-term hypoxia in vitro. Our data also demonstrated that the ACSS2 suppression slowed tumor growth in vivo. These findings showed that ACSS2 plays a significant role in tumor cell survival under hypoxia and that ACSS2 would be a potential target for tumor treatment. Furthermore, we found that tumor cells excreted acetate and the quantity increased under hypoxia: the pattern of acetate excretion followed the expression pattern of ACSS2. Additionally, the ACSS2 knockdown led to a corresponding reduction in the acetate excretion in tumor cells. These results mean that ACSS2 can conduct the reverse reaction from acetyl-CoA to acetate in tumor cells, which indicates that ACSS2 is a bi-directional enzyme in tumor cells and that ACSS2 might play a buffering role in tumor acetyl-CoA/acetate metabolism.
Collapse
Affiliation(s)
- Yukie Yoshii
- Biomedical Imaging Research Center, University of Fukui, 23-3, Shimoaizuki, Eiheiji, Fukui 910-1193, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Lee N, Nehmeh S, Schöder H, Fury M, Chan K, Ling CC, Humm J. Prospective trial incorporating pre-/mid-treatment [18F]-misonidazole positron emission tomography for head-and-neck cancer patients undergoing concurrent chemoradiotherapy. Int J Radiat Oncol Biol Phys 2009; 75:101-8. [PMID: 19203843 DOI: 10.1016/j.ijrobp.2008.10.049] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2008] [Revised: 10/10/2008] [Accepted: 10/24/2008] [Indexed: 01/06/2023]
Abstract
PURPOSE To report the results from a prospective study of a series of locoregionally advanced head-and-neck cancer patients treated with platinum-based chemotherapy and intensity-modulated radiotherapy and to discuss the findings of their pre-/mid-treatment [(18)F]-misonidazole ((18)F-FMISO) positron emission tomography (PET) scans. METHODS AND MATERIALS A total of 28 patients agreed to participate in this study. Of these 28 patients, 20 (90% with an oropharyngeal primary cancer) were able to undergo the requirements of the protocol. Each patient underwent four PET scans: one pretreatment fluorodeoxyglucose PET/computed tomography scan, two pretreatment (18)F-FMISO PET/computed tomography scans, and a third (18)F-FMISO PET (mid-treatment) scan performed 4 weeks after the start of chemoradiotherapy. The (18)F-FMISO PET scans were acquired 2-3 h after tracer administration. Patients were treated with 2-3 cycles of platinum-based chemotherapy concurrent with definitive intensity-modulated radiotherapy. RESULTS A heterogeneous distribution of (18)F-FMISO was noted in the primary and/or nodal disease in 90% of the patients. Two patients had persistent detectable hypoxia on their third mid-treatment (18)F-FMISO PET scan. One patient experienced regional/distant failure but had no detectable residual hypoxia on the mid-treatment (18)F-FMISO PET scan. CONCLUSION Excellent locoregional control was observed in this series of head-and-neck cancer patients treated with concurrent platinum-based chemotherapy and intensity-modulated radiotherapy despite evidence of detectable hypoxia on the pretreatment (18)F-FMISO PET/computed tomography scans of 18 of 20 patients. In this prospective study, neither the presence nor the absence of hypoxia, as defined by positive (18)F-FMISO findings on the mid-treatment PET scan, correlated with patient outcome. The results of this study have confirmed similar results reported previously.
Collapse
Affiliation(s)
- Nancy Lee
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| | | | | | | | | | | | | |
Collapse
|
11
|
Vaupel P. Pathophysiology of Solid Tumors. THE IMPACT OF TUMOR BIOLOGY ON CANCER TREATMENT AND MULTIDISCIPLINARY STRATEGIES 2009. [DOI: 10.1007/978-3-540-74386-6_4] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
12
|
He F, Deng X, Wen B, Liu Y, Sun X, Xing L, Minami A, Huang Y, Chen Q, Zanzonico PB, Ling CC, Li GC. Noninvasive molecular imaging of hypoxia in human xenografts: comparing hypoxia-induced gene expression with endogenous and exogenous hypoxia markers. Cancer Res 2008; 68:8597-606. [PMID: 18922936 DOI: 10.1158/0008-5472.can-08-0677] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Tumor hypoxia is important in the development and treatment of human cancers. We have developed a novel xenograft model for studying and imaging of hypoxia-induced gene expression. A hypoxia-inducible dual reporter herpes simplex virus type 1 thymidine kinase and enhanced green fluorescence protein (HSV1-TKeGFP), under the control of hypoxia response element (9HRE), was stably transfected into human colorectal HT29 cancer cells. Selected clones were further enriched by repeated live cell sorting gated for hypoxia-induced eGFP expression. Fluorescent microscopy, fluorescence-activated cell sorting, and radioactive substrate trapping assays showed strong hypoxia-induced expression of eGFP and HSV1-tk enzyme in the HT29-9HRE cells in vitro. Sequential micropositron emission tomography (PET) imaging of tumor-bearing animals, using the hypoxic cell tracer (18)F-FMISO and the reporter substrate (124)I-FIAU, yielded similar tumor hypoxia images for the HT29-9HRE xenograft but not in the parental HT29 tumor. Using autoradiography and IHC, detailed spatial distributions in tumor sections were obtained and compared for the following hypoxia-associated biomarkers in the HT29-9HRE xenograft: (124)I-FIAU, (18)F-FMISO, Hoechst (perfusion), lectin-TRITC (functional blood vessels), eGFP, pimonidazole, EF5, and CA9. Intratumoral distributions of (124)I-FIAU and (18)F-FMISO were similar, and eGFP, pimonidazole, EF5, and CA9 colocalized in the same areas but not in well-perfused regions that were positive for Hoechst and lectin-TRITC. In enabling the detection of hypoxia-induced molecular events and mapping their distribution in vivo with serial noninvasive positron emission tomography imaging, and multiple variable analysis with immunohistochemistry and fluorescence microscopy, this human xenograft model provides a valuable tool for studying tumor hypoxia and in validating existing and future exogenous markers for tumor hypoxia.
Collapse
Affiliation(s)
- Fuqiu He
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
The importance of hypoxia in disease pathogenesis and prognosis is gathering increasing clinical significance and having a greater impact on patient management and outcome. Previous efforts to evaluate hypoxia have included the invasive assessment of hypoxia with immunohistologic and histographic oxygen probes. The emergence of new radiotracers has allowed noninvasive assessment of hypoxia, with the most extensively investigated and validated positron emission tomography radiotracer of hypoxia to date being (18)F-fluoromisonodazole ((18)F-FMISO). This review discusses the relevance and biology of hypoxia in cells and organ systems, and reviews the laboratory and clinical applications of (18)F-FMISO in oncology and noncancer disease states.
Collapse
Affiliation(s)
- Sze Ting Lee
- Centre for PET, Austin Health, Heidelberg, Victoria, Australia
| | | |
Collapse
|
14
|
Reproducibility of intratumor distribution of (18)F-fluoromisonidazole in head and neck cancer. Int J Radiat Oncol Biol Phys 2008; 70:235-42. [PMID: 18086391 DOI: 10.1016/j.ijrobp.2007.08.036] [Citation(s) in RCA: 164] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2007] [Revised: 08/08/2007] [Accepted: 08/09/2007] [Indexed: 12/12/2022]
Abstract
PURPOSE Hypoxia is one of the main causes of the failure to achieve local control using radiotherapy. This is due to the increased radioresistance of hypoxic cells. (18)F-fluoromisonidazole ((18)F-FMISO) positron emission tomography (PET) is a noninvasive imaging technique that can assist in the identification of intratumor regions of hypoxia. The aim of this study was to evaluate the reproducibility of (18)F-FMISO intratumor distribution using two pretreatment PET scans. METHODS AND MATERIALS We enrolled 20 head and neck cancer patients in this study. Of these, 6 were excluded from the analysis for technical reasons. All patients underwent an (18)F-fluorodeoxyglucose study, followed by two (18)F-FMISO studies 3 days apart. The hypoxic volumes were delineated according to a tumor/blood ratio >or=1.2. The (18)F-FMISO tracer distributions from the two (18)F-FMISO studies were co-registered on a voxel-by-voxel basis using the computed tomography images from the PET/computed tomography examinations. A correlation between the (18)F-FMISO intensities of the corresponding spatial voxels was derived. RESULTS A voxel-by-voxel analysis of the (18)F-FMISO distributions in the entire tumor volume showed a strong correlation in 71% of the patients. Restraining the correlation to putatively hypoxic zones reduced the number of patients exhibiting a strong correlation to 46%. CONCLUSION Variability in spatial uptake can occur between repeat (18)F-FMISO PET scans in patients with head and neck cancer. Blood data for one patient was not available. Of 13 patients, 6 had well-correlated intratumor distributions of (18)F-FMISO-suggestive of chronic hypoxia. More work is required to identify the underlying causes of changes in intratumor distribution before single-time-point (18)F-FMISO PET images can be used as the basis of hypoxia-targeting intensity-modulated radiotherapy.
Collapse
|
15
|
Abstract
Data from 125 studies describing the pretreatment oxygenation status as measured in the clinical setting using the computerized Eppendorf pO2 histography system have been compiled in this article. Tumor oxygenation is heterogeneous and severely compromised as compared to normal tissue. Hypoxia results from inadequate perfusion and diffusion within tumors and from a reduced O2 transport capacity in anemic patients. The development of tumor hypoxia is independent of a series of relevant tumor characteristics (e.g., clinical size, stage, histology, and grade) and various patient demographics. Overall median pO2 in cancers of the uterine cervix, head and neck, and breast is 10 mm Hg with the overall hypoxic fraction (pO2 <or= 2.5 mm Hg) being approx. 25%. Metastatic lesions do not substantially deviate from the oxygenation status of (their) primary tumors. Whereas normal tissue oxygenation is independent of the hemoglobin level over the range of 8-15 g/dL, hypoxia is more pronounced in anemic patients and above this range in some cancers. Identification of tumor hypoxia may allow an assessment of a tumor's potential to develop an aggressive phenotype or acquired treatment resistance, both of which lead to poor prognosis. Detection of hypoxia in the clinical setting may therefore be helpful in selecting high-risk patients for individual and/or more intensive treatment schedules.
Collapse
Affiliation(s)
- Peter Vaupel
- Institute of Physiology and Pathophysiology, University of Mainz, Mainz, Germany.
| | | | | |
Collapse
|
16
|
Procissi D, Claus F, Burgman P, Koziorowski J, Chapman JD, Thakur SB, Matei C, Ling CC, Koutcher JA. In vivo19F Magnetic Resonance Spectroscopy and Chemical Shift Imaging of Tri-Fluoro-Nitroimidazole as a Potential Hypoxia Reporter in Solid Tumors. Clin Cancer Res 2007; 13:3738-47. [PMID: 17575240 DOI: 10.1158/1078-0432.ccr-06-1563] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE 2-Nitro-alpha-[(2,2,2-trifluoroethoxy)methyl]-imidazole-1-ethanol (TF-MISO) was investigated as a potential noninvasive marker of tissue oxygen levels in tumors using (19)F magnetic resonance spectroscopy (MRS) and (19)F chemical shift imaging. EXPERIMENTAL DESIGNS In vitro data were obtained using high-performance liquid chromatography on tumor cells incubated under varying oxygen conditions to determine the oxygen-binding characteristics. In vivo data were obtained using a well-characterized hypoxic murine breast tumor (MCa), in addition to studies on a rat prostate tumor model (R3327-AT) implanted in nude mice. Detection of intratumor (19)F signal from TF-MISO was done using MRS for up to 10 h following a 75 mg/kg i.v. injection. Localized distribution of the compound in the implanted MCa tumor has been imaged using slice-selective two-dimensional chemical shift imaging 6 h after injection. RESULTS The in vitro results showed that TF-MISO preferentially accumulates in cells incubated under anoxic conditions. The in vivo (19)F MR spectral features (line width and chemical shift) were recorded as a function of time after injection, and the results indicate that the fluorine atoms are indeed sensitive to changes in the local environment while still providing a detectable MR signal. Ex vivo spectra were collected and established the visibility of the (19)F signal under conditions of maximum hypoxia. Late time point (>6 h) tumor tissue concentrations, as obtained from (19)F MRS, suggest that TF-MISO is reduced and retained in hypoxic tumor. The feasibility of obtaining TF-MISO tumor distribution maps in a reasonable time frame was established. CONCLUSIONS Based on the results presented herein, it is suggested that TF-MISO has the potential to be a valid magnetic resonance hypoxia imaging reporter for both preclinical hypoxia studies and hypoxia-directed clinical therapy.
Collapse
Affiliation(s)
- Daniel Procissi
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
This article has been retracted from Cancer Imaging. The correct citation is Padhani AR, Krohn KA, Lewis JS, Alber M. Imaging oxygenation of human tumours.
Collapse
|
18
|
Abstract
Tumour hypoxia represents a significant challenge to the curability of human tumours leading to treatment resistance and enhanced tumour progression. Tumour hypoxia can be detected by non-invasive and invasive techniques but the inter-relationships between these remains largely undefined. [18F]Fluoromisonidazole-3-fluoro-1-(2
′-nitro-1
′-imidazolyl)-2-propanol ([18F]MISO) and Cu-diacetyl-bis(N4-methylthiosemicarbazone (Cu-ATSM)-positron emission tomography (PET), and blood oxygen level-dependent (BOLD)-magnetic resonance imaging (MRI) are the lead contenders for human application based on their non-invasive nature, ease of use and robustness, measurement of hypoxia status, validity, ability to
demonstrate heterogeneity and general availability; PET techniques are the primary focus of this review.
Collapse
Affiliation(s)
- Anwar Padhani
- Paul Strickland Scanner Centre, Mount Vernon Hospital, Rickmansworth Road, Northwood, Middlesex, HA6 2RN, UK.
| |
Collapse
|
19
|
Padhani AR, Krohn KA, Lewis JS, Alber M. Imaging oxygenation of human tumours. Eur Radiol 2006; 17:861-72. [PMID: 17043737 PMCID: PMC1820761 DOI: 10.1007/s00330-006-0431-y] [Citation(s) in RCA: 228] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2006] [Revised: 07/18/2006] [Accepted: 08/07/2006] [Indexed: 12/16/2022]
Abstract
Tumour hypoxia represents a significant challenge to the curability of human tumours leading to treatment resistance and enhanced tumour progression. Tumour hypoxia can be detected by non-invasive and invasive techniques but the inter-relationships between these remains largely undefined. 18F-MISO and Cu-ATSM-PET, and BOLD-MRI are the lead contenders for human application based on their non-invasive nature, ease of use and robustness, measurement of hypoxia status, validity, ability to demonstrate heterogeneity and general availability, these techniques are the primary focus of this review. We discuss where developments are required for hypoxia imaging to become clinically useful and explore potential new uses for hypoxia imaging techniques including biological conformal radiotherapy.
Collapse
Affiliation(s)
- Anwar R Padhani
- Paul Strickland Scanner Centre, Mount Vernon Hospital, Rickmansworth Road, Northwood, Middlesex, HA6 2RN, UK.
| | | | | | | |
Collapse
|
20
|
Toma-Daşu I, Daşu A, Karlsson M. Theoretical simulation of tumour hypoxia measurements. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2006; 578:369-74. [PMID: 16927718 DOI: 10.1007/0-387-29540-2_57] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Affiliation(s)
- Iuliana Toma-Daşu
- Department of Radiation Sciences, Umeå University, 901 87 Umeå, Sweden
| | | | | |
Collapse
|
21
|
Dressman HK, Hans C, Bild A, Olson JA, Rosen E, Marcom PK, Liotcheva VB, Jones EL, Vujaskovic Z, Marks J, Dewhirst MW, West M, Nevins JR, Blackwell K. Gene expression profiles of multiple breast cancer phenotypes and response to neoadjuvant chemotherapy. Clin Cancer Res 2006; 12:819-26. [PMID: 16467094 DOI: 10.1158/1078-0432.ccr-05-1447] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Breast cancer is a heterogeneous disease, and markers for disease subtypes and therapy response remain poorly defined. For that reason, we employed a prospective neoadjuvant study in locally advanced breast cancer to identify molecular signatures of gene expression correlating with known prognostic clinical phenotypes, such as inflammatory breast cancer or the presence of hypoxia. In addition, we defined molecular signatures that correlate with response to neoadjuvant chemotherapy. EXPERIMENTAL DESIGN Tissue was collected under ultrasound guidance from patients with stage IIB/III breast cancer before four cycles of neoadjuvant liposomal doxorubicin paclitaxel chemotherapy combined with local whole breast hyperthermia. Gene expression analysis was done using Affymetrix U133 Plus 2.0 GeneChip arrays. RESULTS Gene expression patterns were identified that defined the phenotypes of inflammatory breast cancer as well as tumor hypoxia. In addition, molecular signatures were identified that predicted the persistence of malignancy in the axillary lymph nodes after neoadjuvant chemotherapy. This persistent lymph node signature significantly correlated with disease-free survival in two separate large populations of breast cancer patients. CONCLUSIONS Gene expression signatures have the capacity to identify clinically significant features of breast cancer and can predict which individual patients are likely to be resistant to neoadjuvant therapy, thus providing the opportunity to guide treatment decisions.
Collapse
Affiliation(s)
- Holly K Dressman
- Duke Institute for Genome Sciences and Policy, Duke University Medical Center, Duke University, Durham, North Carolina 27710, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Toma-Daşu I, Daşu A, Karlsson M. Conversion of polarographic electrode measurements—a computer based approach. Phys Med Biol 2005; 50:4581-91. [PMID: 16177491 DOI: 10.1088/0031-9155/50/19/011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The polarographic measurement of tissue oxygenation is one of the most widely used methods in clinical practice for the quantification of tumour hypoxia. However, due to the particular features of the electrode measuring process, the results of the measurements do not accurately reflect the tumour oxygenation. This study aimed to find a correlation between the electrode measurements and the tumour oxygenation in an attempt to improve the accuracy of the predictions regarding the response to treatment based on electrode measurements. A previously developed computer model that allows the simulation of tumour tissue and electrode measurements was used. The oxygenation of a large number of tumours with biologically relevant distributions of blood vessels was theoretically calculated. Simulations of electrode measurements allowed the comparison between the real tissue oxygenation and the results obtained with the electrode. A semi-empirical relationship between the hypoxic fraction measured by the electrode and the real hypoxic fraction in the tissue has been found. The impact of the correction of the electrode measurements in terms of predictions for tumour control probability was estimated for a few clinical examples. The range of possible true values corresponding to one measurement has also proven useful for explaining the apparently unexpected response to the treatment of some patients. The corrected hypoxic fraction which is believed to be closer to the real value of tissue hypoxia predicts much smaller control probabilities than the raw electrode measurements. This could provide an explanation for the apparently unexpected failure to respond to the treatment of some of the patients with apparently favourable tumour oxygenation. This also means that the electrode measurements cannot be used directly for the quantitative modelling of tumour response to the treatment. The conversion method proposed in this paper might however strengthen the statistical power of the correlations between the electrode measurements and the treatment outcome.
Collapse
Affiliation(s)
- Iuliana Toma-Daşu
- Department of Radiation Sciences, Umeå University, 901 87 Umeå, Sweden.
| | | | | |
Collapse
|
23
|
Detwiller KY, Fernando NT, Segal NH, Ryeom SW, D'Amore PA, Yoon SS. Analysis of hypoxia-related gene expression in sarcomas and effect of hypoxia on RNA interference of vascular endothelial cell growth factor A. Cancer Res 2005; 65:5881-9. [PMID: 15994966 DOI: 10.1158/0008-5472.can-04-4078] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Vascular endothelial cell growth factor A (VEGF-A) and hypoxia play important roles in tumor angiogenesis. VEGF-A gene expression is up-regulated in tumors under hypoxic conditions, yet it is unclear how such up-regulation will affect the efficacy of RNA interference strategies targeting VEGF-A. Four potential short interfering RNA (siRNA) sequences for the VEGF-A gene were cloned into expression plasmids and transfected into HT1080 human fibrosarcoma cells. Stable transfection of these plasmids decreased VEGF-A mRNA levels and protein secretion by up to 99%. Our analysis of >100 hypoxia-related genes using oligonucleotide microarrays of 38 human sarcoma samples and 14 normal tissues identified distinctly different patterns of expression between sarcomas and normal tissues as assessed by hierarchical clustering analysis. Numerous hypoxia-related genes were significantly up-regulated in sarcomas including hypoxia-inducible factor 1alpha (HIF-1alpha). Exposure of wild-type HT1080 cells to 1% hypoxia resulted in HIF-1alpha up-regulation and a 74% increase in VEGF-A secretion as compared with secretion under normoxic conditions. Surprisingly, stable cell lines expressing VEGF-A siRNAs silenced VEGF-A expression equally well in hypoxia and normoxia. S.c. injection of cells with VEGF-A siRNAs into athymic nude mice led to slower-growing tumors, decreased blood vessel density, and greater apoptosis when compared with controls. Immunofluorescence analysis of tumor sections revealed areas of HIF-1alpha nuclear expression, suggesting areas of hypoxia, in both control tumors and VEGF-suppressed tumors. We conclude that hypoxia plays an important role in human sarcomas but hypoxic up-regulation of VEGF-A expression does not attenuate the efficacy of VEGF-A RNA interference.
Collapse
Affiliation(s)
- Kara Y Detwiller
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | | | | | | | | | | |
Collapse
|
24
|
Toma-Daşu I, Daşu A, Karlsson M. The relationship between temporal variation of hypoxia, polarographic measurements and predictions of tumour response to radiation. Phys Med Biol 2005; 49:4463-75. [PMID: 15552411 DOI: 10.1088/0031-9155/49/19/002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The polarographic oxygen sensor is one of the most used devices for in vivo measurements of oxygen and many other measurement techniques for measuring tumour hypoxia are correlated with electrode measurements. Little is known however about the relationship between electrode measurements and the real tissue oxygenation. This paper investigates the influence of the temporal change of the hypoxic pattern on the electrode measurements and the tumour response. Electrode measurements and tumour response were simulated using a computer program that allows both the calculation of the tissue oxygenation with respect to the two types of hypoxia that might arise in tumours and the virtual insertion of the electrode into the tissue. It was therefore possible to control the amount of each type of hypoxia in order to investigate their influence on the measurement results. Tissues with several vascular architectures ranging from well oxygenated to poorly oxygenated were taken into consideration as might be seen in practice. The influence of the electrode measurements on the treatment outcome was estimated by calculating the tumour control probability for the tumours characterized either by the real or by the measured tumour oxygenation. We have simulated electrode oxygen measurements in different types of tissues, covering a wide range of tumour oxygenations. The results of the simulations showed that the measured distribution depends on the details of the vascular network and not on the type of hypoxia. We have also simulated the effects of the temporal change of the acute hypoxic pattern due to the opening and the closure of different blood vessels during a full fractionated treatment. The results of this simulation suggested that the temporal variation of the hypoxic pattern does not lead to significantly different results for the electrode measurements or the predicted tumour control probabilities. In conclusion, it was found that the averaging effect of the electrode leads to a systematic deviation between the actual oxygen distribution and the measured distribution. However, as the electrode reflects the general trends of the tissue oxygenation it has the potential of being used for the general characterization of tumour hypoxia even if the actual type of hypoxia measured by the electrode cannot be determined. Indeed, the change in time of the acute hypoxic region does not compensate for the lack of oxygenation at a specific moment and therefore does not influence the polarographic oxygen measurements.
Collapse
Affiliation(s)
- Iuliana Toma-Daşu
- Department of Radiation Sciences, Umeå University, S-901 85 Umeå, Sweden.
| | | | | |
Collapse
|
25
|
Griffin RJ, Williams BW, Park HJ, Song CW. Preferential action of arsenic trioxide in solid-tumor microenvironment enhances radiation therapy. Int J Radiat Oncol Biol Phys 2005; 61:1516-22. [PMID: 15817358 DOI: 10.1016/j.ijrobp.2004.12.058] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2004] [Revised: 12/07/2004] [Accepted: 12/16/2004] [Indexed: 12/09/2022]
Abstract
PURPOSE To investigate the effect of arsenic trioxide, Trisenox (TNX), on primary cultures of endothelial cells and tumor tissue under varying pH and pO(2) environments and the effects of combined TNX and radiation therapy on experimental tumors. METHODS AND MATERIALS Human dermal microvascular endothelial cells were cultured in vitro and exposed to TNX under various combinations of aerobic, hypoxic, neutral, or acidic conditions, and levels of activated JNK MAP kinase were assessed by Western blotting. FSaII fibrosarcoma cells grown in the hind limb of female C3H mice were used to study the effect of TNX on tumor blood perfusion and oxygenation. The tumor-growth delay after a single or fractionated irradiation with or without TNX treatment was assessed. RESULTS A single intraperitoneal injection of 8 mg/kg TNX reduced the blood perfusion in FSaII tumors by 53% at 2 hours after injection. To increase the oxygenation of the tumor vasculature during TNX treatment, some animals were allowed to breathe carbogen (95% O(2)/5% CO(2)). Carbogen breathing alone for 2 hours reduced tumor perfusion by 33%. When carbogen breathing was begun immediately after TNX injection, no further reduction occurred in tumor blood perfusion at 2 hours after injection. In vitro, TNX exposure increased activity JNK MAP kinase preferentially in endothelial cells cultured in an acidic or hypoxic environment. In vivo, the median oxygenation in FSaII tumors measured at 3 or 5 days after TNX injection was found to be significantly elevated compared with control tumors. Subsequently, radiation-induced tumor-growth delay was synergistically increased when radiation and TNX injection were fractionated at 3-day or 5-day intervals. CONCLUSIONS Trisenox has novel vascular-damaging properties, preferentially against endothelium in regions of low pH or pO(2), which leads to tumor cell death and enhancement of the response of tumors to radiotherapy.
Collapse
Affiliation(s)
- Robert J Griffin
- Department of Therapeutic Radiology, University of Minnesota Medical School, Minneapolis, USA.
| | | | | | | |
Collapse
|
26
|
Abstract
There is a clear need in cancer treatment for a noninvasive imaging assay that evaluates the oxygenation status and heterogeneity of hypoxia and angiogenesis in individual patients. Such an assay could be used to select alternative treatments and to monitor the effects of treatment. Of the several methods available, each imaging procedure has at least one disadvantage. The limited quantitative potential of single-photon emission CT and MR imaging always limits tracer imaging based on these detection systems. PET imaging with FMISO and Cu-ATSM is ready for coordinated multicenter trials, however, that should move aggressively forward to resolve the debate over the importance of hypoxia in limiting response to cancer therapy. Advances in radiation treatment planning, such as intensity-modulated radiotherapy, provide the ability to customize radiation delivery based on physical conformity. With incorporation of regional biologic information, such as hypoxia and proliferating vascular density in treatment planning, imaging can create a biologic profile of the tumor to direct radiation therapy. Presence of widespread hypoxia in the tumor benefits from a systemic hypoxic cell cytotoxin. Angiogenesis is also an important therapeutic target. Imaging hypoxia and angiogenesis complements the efforts in development of antiangiogenesis and hypoxia-targeted drugs. The complementary use of hypoxia and angiogenesis imaging methods should provide the impetus for development and clinical evaluation of novel drugs targeted at angiogenesis and hypoxia. Hypoxia imaging brings in information different from that of FDG-PET but it will play an important niche role in oncologic imaging in the near future. FMISO, radioiodinated azamycin arabinosides, and Cu-ATSM are all being evaluated in patients. The Cu-ATSM images show the best contrast early after injection but these images are confounded by blood flow and their mechanism of localization is one step removed from the intracellular O2 concentration. FMISO has been criticized as inadequate because of its clearance characteristics, but its uptake after 2 hours is probably the most purely reflective of regional PO2 at the time the radiopharmaceutical is used. The FMISO images show less contrast than those of Cu-ATSM because of the lipophilicity and slower clearance of FMISO but attempts to increase the rate of clearance led to tracers whose distribution is contaminated by blood flow effects. For single-photon emission CT the only option is radioiodinated azamycin arabinosides, because the technetium agents are not yet ready for clinical evaluation. Rather than develop new and improved hypoxia agents, or even quibbling about the pros and cons of alternative agents, the nuclear medicine community needs to convince the oncology community that imaging hypoxia is an important procedure that can lead to improved treatment outcome.
Collapse
Affiliation(s)
- Joseph G Rajendran
- Division of Nuclear Medicine, Department of Radiology, Box 356113, University of Washington, Seattle, WA 98195, USA.
| | | |
Collapse
|
27
|
Rajendran JG, Mankoff DA, O'Sullivan F, Peterson LM, Schwartz DL, Conrad EU, Spence AM, Muzi M, Farwell DG, Krohn KA. Hypoxia and glucose metabolism in malignant tumors: evaluation by [18F]fluoromisonidazole and [18F]fluorodeoxyglucose positron emission tomography imaging. Clin Cancer Res 2004; 10:2245-52. [PMID: 15073099 DOI: 10.1158/1078-0432.ccr-0688-3] [Citation(s) in RCA: 288] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE The aim of this study is to compare glucose metabolism and hypoxia in four different tumor types using positron emission tomography (PET). (18)F-labeled fluorodeoxyglucose (FDG) evaluates energy metabolism, whereas the uptake of (18)F-labeled fluoromisonidazole (FMISO) is proportional to tissue hypoxia. Although acute hypoxia results in accelerated glycolysis, cellular metabolism is slowed in chronic hypoxia, prompting us to look for discordance between FMISO and FDG uptake. EXPERIMENTAL DESIGN Forty-nine patients (26 with head and neck cancer, 11 with soft tissue sarcoma, 7 with breast cancer, and 5 with glioblastoma multiforme) who had both FMISO and FDG PET scans as part of research protocols through February 2003 were included in this study. The maximum standardized uptake value was used to depict FDG uptake, and hypoxic volume and maximum tissue:blood ratio were used to quantify hypoxia. Pixel-by-pixel correlation of radiotracer uptake was performed on coregistered images for each corresponding tumor plane. RESULTS Hypoxia was detected in all four patient groups. The mean correlation coefficients between FMISO and FDG uptake were 0.62 for head and neck cancer, 0.47 for breast cancer, 0.38 for glioblastoma multiforme, and 0.32 for soft tissue sarcoma. The correlation between the overall tumor maximum standardized uptake value for FDG and hypoxic volume was small (Spearman r = 0.24), with highly significant differences among the different tumor types (P < 0.005). CONCLUSIONS Hypoxia is a general factor affecting glucose metabolism; however, some hypoxic tumors can have modest glucose metabolism, whereas some highly metabolic tumors are not hypoxic, showing discordance in tracer uptake that can be tumor type specific.
Collapse
|
28
|
Salmon HW, Siemann DW. Utility of 19F MRS detection of the hypoxic cell marker EF5 to assess cellular hypoxia in solid tumors. Radiother Oncol 2004; 73:359-66. [PMID: 15588883 DOI: 10.1016/j.radonc.2004.07.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2004] [Revised: 05/25/2004] [Accepted: 07/15/2004] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND PURPOSE The present studies were undertaken to determine whether 19F MRS could be used to quantify the binding of the pentafluorinated derivative of etanidazole (EF5) in hypoxic cells of solid tumors. MATERIALS AND METHODS A 4.7 T imaging magnet was used for the in situ and in vitro evaluation of EF5 signals. In order to develop a better understanding of these NMR measurements the characteristics of parent, reduced unbound, and reduced bound EF5 signals were examined in vitro using a 12 T spectrometer. RESULTS In situ data acquired using a 4.7 T imaging magnet, showed retention of EF5 signals in KHT sarcomas that was absent in muscles for 6 h after EF5 injection. In vitro studies showed no difference in the NMR detectable signal of parent and reduced unbound EF5. T2 values determined using parent EF5 samples revealed a T2 time of 675 ms. In contrast, EF5 bound to KHT tumor cells gave rise to signals of low intensity, broad line widths, and T2 relaxation times of less than 30 ms. When the same samples were analyzed using the 4.7 T imaging magnet, the CF3 and CF2 fluorine peaks were readily identifiable in the parent EF5 sample but no fluorine signal could be detected from EF5 bound to KHT tumor cells. CONCLUSION The inability to resolve bound EF5 metabolites even at high field strengths (12 T), coupled with the short T2 relaxation times of the bound EF5, and the limits of detection of the in situ applied imaging magnet (4.7 T), meant that hypoxic cells could not be quantified in tumors using the 19F MRS technique. In situ 19F MRS measurements of EF5 signals (parent/reduced unbound) may reflect conditions of tumor physiology and thus indicate the extent of tumor hypoxia but they are not capable of resolving the cellular oxygenation status of the tumor cells.
Collapse
Affiliation(s)
- Howard W Salmon
- Department of Nuclear and Radiological Engineering, University of Florida, Gainesville, FL 32610, USA
| | | |
Collapse
|
29
|
Robinson SP, Griffiths JR. Current issues in the utility of 19F nuclear magnetic resonance methodologies for the assessment of tumour hypoxia. Philos Trans R Soc Lond B Biol Sci 2004; 359:987-96. [PMID: 15306411 PMCID: PMC1693379 DOI: 10.1098/rstb.2003.1376] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
It is now well established that uncontrolled proliferation of tumour cells together with the chaotic and poorly regulated blood supply of solid tumours result in tissue hypoxia, and that hypoxic regions of tumours are resistant to radiotherapy and chemotherapy. The development and application of non-invasive methods to rapidly determine the degree and extent of tumour hypoxia in an individual tumour would clearly enhance cancer treatment strategies. This review describes the current status of two (19)F nuclear magnetic resonance (NMR) methodologies that have been exploited to investigate tumour hypoxia, namely: (i) (19)F NMR oximetry following administration of perfluorocarbons, from which tumour p(O)(2) measurements can be made; and (ii) (19)F NMR measurements of the tumour retention of fluorinated 2-nitroimidazoles.
Collapse
Affiliation(s)
- Simon P Robinson
- Department of Basic Medical Sciences, St George's Hospital Medical School, Cranmer Terrace, London SW17 0RE, UK.
| | | |
Collapse
|
30
|
Kalliomäki T, Hill RP. Effects of tumour acidification with glucose+MIBG on the spontaneous metastatic potential of two murine cell lines. Br J Cancer 2004; 90:1842-9. [PMID: 15150590 PMCID: PMC2409743 DOI: 10.1038/sj.bjc.6601766] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
In addition to hypoxia, acidic extracellular pH (pHe) is recognised as one of the microenvironmental characteristics of solid tumours. A number of studies have examined ways to increase tumour acidity in order to improve tumour-specific targeting of certain drugs and the effectiveness of hyperthermia. However, previous data have shown that exposure of murine tumour cells to acid conditions in culture can enhance their metastatic potential when injected subsequently into mice, raising the concern that deliberate tumour acidification might increase the probability of metastasis. In this study, we examined the effects of in vivo tumour acidification and hypoxia on the spontaneous metastatic potential of the murine KHT-C fibrosarcoma and B16F1 melanoma cell lines. A tumour-specific increase in extracellular acidity, demonstrated by measurements with pH electrodes, was achieved by daily intraperitoneal injections of meta-iodo-benzylguanidine (MIBG) and/or glucose. This method of tumour acidification during tumour growth did not significantly enhance the spontaneous metastatic potential of the two murine cell lines.
Collapse
Affiliation(s)
- T Kalliomäki
- Experimental Therapeutics Division, Ontario Cancer Institute/Princess Margaret Hospital, Toronto, Ontario, Canada M5G 2M9
- Department of Medical Biophysics
| | - R P Hill
- Experimental Therapeutics Division, Ontario Cancer Institute/Princess Margaret Hospital, Toronto, Ontario, Canada M5G 2M9
- Department of Medical Biophysics
- Department of Radiation Oncology, University of Toronto, 610 University Avenue, Toronto, Ontario, Canada M5G 2M9
- Experimental Therapeutics Division, Ontario Cancer Institute/Princess Margaret Hospital, 610 University Avenue, Toronto, Ontario, Canada, M5G 2M9. E-mail:
| |
Collapse
|
31
|
Rodrigues LM, Howe FA, Griffiths JR, Robinson SP. Tumor R2* is a prognostic indicator of acute radiotherapeutic response in rodent tumors. J Magn Reson Imaging 2004; 19:482-8. [PMID: 15065173 DOI: 10.1002/jmri.20024] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
PURPOSE To test the prognostic potential of tumor R2* with respect to radiotherapeutic outcome. Blood oxygenation level dependent (BOLD) MRI images are sensitive to changes in deoxyhemoglobin concentration through the transverse MRI relaxation rate R2* of tissue water, hence the quantitative measurement of tumor R2* may be related to tissue oxygenation. METHODS AND MATERIALS Tumor growth inhibition in response to radiation was established for both GH3 prolactinomas and RIF-1 fibrosarcomas with animals breathing either air or carbogen during radiation. In a separate cohort, the baseline R2* and carbogen (95% O2, 5% CO2)-induced DeltaR2* of rat GH3 prolactinomas and murine RIF-1 fibrosarcomas were quantified using multigradient echo (MGRE) MRI prior to radiotherapy, and correlated with subsequent tumor growth inhibition in response to ionizing radiation, while the animals breathed air. RESULTS A radiation dose of 15 Gy caused pronounced growth delay in both tumor models and transient regression of the GH3 prolactinomas. When the animals breathed carbogen during radiation, the growth delay/regression was enhanced only in the GH3 prolactinomas. The GH3 prolactinomas, which exhibit a relatively fast baseline R2* and large DeltaR2* in response to carbogen breathing prior to radiotherapy, showed a substantial reduction in normalized tumor volume to 66 +/- 3% with air breathing and 36 +/- 5% with carbogen seven days after 15 Gy irradiation. In contrast, the effect of 15 Gy on the RIF-1 fibrosarcomas, which give a relatively slow baseline R2* and negligible DeltaR2* response to carbogen prior to treatment, showed a much smaller growth inhibition (143 +/- 3% with air, 133 +/- 12% with carbogen). CONCLUSION Quantitation of tumor R2* and carbogen-induced DeltaR2* by MGRE MRI provides completely noninvasive prognostic indicators of a potential acute radiotherapeutic response.
Collapse
Affiliation(s)
- Loreta M Rodrigues
- Cancer Research UK Biomedical Magnetic Resonance Research Group, Department of Basic Medical Sciences, St. George's Hospital Medical School, London, United Kingdom
| | | | | | | |
Collapse
|
32
|
Abstract
Low tissue oxygen concentration has been shown to be important in the response of human tumors to radiation therapy, chemotherapy and other treatment modalities. Hypoxia is also known to be a prognostic indicator, as hypoxic human tumors are more biologically aggressive and are more likely to recur locally and metastasize. Herein, we discuss and summarize the various methods under investigation to directly or indirectly measure tissue oxygen in vivo. Secondly, we consider the advantages and disadvantages of each of these techniques. These considerations are made in light of our specific hypotheses that hypoxia should be measured as a continuum, not a binary measurement and that moderate, not severe hypoxia is of great biological consequence.
Collapse
Affiliation(s)
- Sydney M Evans
- School of Medicine, Department of Radiation Oncology, 195 John Morgan Building, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | |
Collapse
|
33
|
Rajendran JG, Wilson DC, Conrad EU, Peterson LM, Bruckner JD, Rasey JS, Chin LK, Hofstrand PD, Grierson JR, Eary JF, Krohn KA. [(18)F]FMISO and [(18)F]FDG PET imaging in soft tissue sarcomas: correlation of hypoxia, metabolism and VEGF expression. Eur J Nucl Med Mol Imaging 2003; 30:695-704. [PMID: 12632200 DOI: 10.1007/s00259-002-1096-7] [Citation(s) in RCA: 204] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2002] [Accepted: 11/18/2002] [Indexed: 01/06/2023]
Abstract
Hypoxia imparts resistance to radiotherapy and chemotherapy and also promotes a variety of changes in tumor biology through inducible promoters. The purpose of this study was to evaluate the use of positron emission tomography (PET) imaging with fluorine-18 fluoromisonidazole (FMISO) in soft tissue sarcomas (STS) as a measure of hypoxia and to compare the results with those obtained using [(18)F]fluorodeoxyglucose (FDG) and other known biologic correlates. FDG evaluates energy metabolism in tumors while FMISO uptake is proportional to tissue hypoxia. FMISO uptake was compared with FDG uptake. Vascular endothelial growth factor (VEGF) expression was also compared with FMISO uptake. Nineteen patients with STS underwent PET scanning with quantitative determination of FMISO and FDG uptake prior to therapy (neo-adjuvant chemotherapy or surgery alone). Ten patients receiving neo-adjuvant chemotherapy were also imaged after chemotherapy but prior to surgical resection. Standardized uptake value (SUV) was used to describe FDG uptake; regional tissue to blood ratio (>or=1.2 was considered significant) was used for FMISO uptake. Significant hypoxia was found in 76% of tumors imaged prior to therapy. No correlation was identified between pretherapy hypoxic volume (HV) and tumor grade ( r=0.15) or tumor volume ( r=0.03). The correlation of HV with VEGF expression was 0.39. Individual tumors showed marked heterogeneity in regional VEGF expression. The mean pixel-by-pixel correlation between FMISO and FDG uptake was 0.49 (range 0.09-0.79) pretreatment and 0.32 (range -0.46-0.72) after treatment. Most tumors showed evidence of reduced uptake of both FMISO and FDG following chemotherapy. FMISO PET demonstrates areas of significant and heterogeneous hypoxia in soft tissue sarcomas. The significant discrepancy between FDG and FMISO uptake seen in this study indicates that regional hypoxia and glucose metabolism do not always correlate. Similarly, we did not find any relationship between the hypoxic volume and the tumor volume or VEGF expression. Identification of hypoxia and development of a more complete biologic profile of STS will serve to guide more rational, individualized cancer treatment approaches.
Collapse
Affiliation(s)
- J G Rajendran
- Department of Radiology, University of Washington Medical Center, Seattle, Washington 98195, USA,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Doll CM, Milosevic M, Pintilie M, Hill RP, Fyles AW. Estimating hypoxic status in human tumors: a simulation using Eppendorf oxygen probe data in cervical cancer patients. Int J Radiat Oncol Biol Phys 2003; 55:1239-46. [PMID: 12654433 DOI: 10.1016/s0360-3016(02)04474-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
PURPOSE To define the minimal number of pO(2) measurements, with 90% sensitivity and 90% specificity, needed to categorize cervical tumors as either hypoxic or oxic. METHODS AND MATERIALS Using Eppendorf oxygen probe data from our ongoing prospective trial, we simulated the measurement of tumor oxygenation with a smaller number of data points in 135 patients with cervical cancer. The hypoxic proportion, defined as the percentage of pO(2) values <5 mm Hg (HP5), was calculated for each tumor. Hypoxic tumors were defined as those with a median HP5 >50%, and tumors with normal oxygen levels as those with a median HP5 < or =50%. A small number of pO(2) measurements were randomly selected from the Eppendorf measurements in each tumor, or per Eppendorf track, and used to define the tumor as hypoxic or oxic. The sensitivity and specificity were calculated, considering the classification as given by the complete set of Eppendorf measurements as the reference standard. RESULTS The probability of falsely classifying the tumor decreased as the selected number of pO(2) measurements per tumor increased, and at 16 measurements was approximately 10%. Adding additional measurements per tumor beyond 24 improved the ability to classify the tumor accurately only slightly. The probability of falsely classifying the tumor decreased as the pO(2) measurements per track increased. At five measurements per track, the probability of falsely classifying the tumor was approximately 9%. CONCLUSION Approximately 20 measurements per tumor, or five measurements per track, using the Eppendorf pO(2) histograph, are sufficient to categorize cervical tumors as hypoxic or oxic. The results of this study will serve as a guide for research clinicians in the use of this and other systems in the assessment of tumor oxygenation in humans.
Collapse
Affiliation(s)
- Corinne M Doll
- Department of Radiation Oncology, Princess Margaret Hospital, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
35
|
Abstract
Over the past decade, there has been significant emphasis on the elucidation of clinicopathologic prognostic factors in STS. This has allowed for identification of the "high-risk" patient at presentation. Molecular factors may further refine the identification of high-risk patients. Setting-related prognostic factors are often amenable to change, and all STS patients should probably be referred for specialty consultation before treatment so that they can benefit from optimal diagnostic, therapeutic, and multidisciplinary approaches. The ongoing pursuit of prognostic issues should also recognize the dynamic nature of prognosis course in a patient's disease.
Collapse
Affiliation(s)
- Brian O'Sullivan
- Department of Radiation Oncology, Princess Margaret Hospital, University of Toronto, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada.
| | | |
Collapse
|
36
|
Papadopoulou MV, Ji M, Bloomer WD, Hollingshead MG. Enhancement of the antitumor effect of cyclophosphamide with the hypoxia-selective cytotoxin NLCQ-1 against murine tumors and human xenografts. JOURNAL OF EXPERIMENTAL THERAPEUTICS AND ONCOLOGY 2002; 2:298-305. [PMID: 12416033 DOI: 10.1046/j.1359-4117.2002.01054.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The antitumor effect of cyclophosphamide (CPM) was investigated against SCCVII murine tumors and PC-3 human xenografts in combination with the hypoxia-selective cytotoxin 4-[3-(2-nitro-1-imidazolyl)-propylamino]-7-chloroquinoline hydrochloride (NLCQ-1). The in vivo-in vitro and the tumor regrowth assays were used, respectively, as end points. In certain cases the hypoxia-selective cytotoxin tirapazamine (TPZ) was included for comparison purposes. In the SCCVII/C3H model, bone marrow toxicity studies were performed in parallel by using a modified CFU-GM assay. In the SCCVII/C3H model, when NLCQ-1 (10 mg/kg i.p.) was given 1 h before cyclophosphamide (CPM; 75-200 mg/kg i.p.), dose-modification factors (DMFs) of 1.9 and 1.0 were achieved for the antitumor effect and bone marrow toxicity, respectively. The corresponding DMF values obtained with TPZ (23 mg/kg) given 2.5 h (optimal time) before CPM were 1.3 and 1.0, respectively. Thus, therapeutic indices (T.I.) of 1.9 and 1.3 were achieved with NLCQ-1 and TPZ, respectively. In the PC-3/athymic nude mouse model, NLCQ-1 (10 mg/kg) given 90 min before CPM (36 mg/kg), qd x 4, increased tumor regrowth delay by 8.7 days compared to CPM alone, at 16-fold the original tumor size. The corresponding log cell kill was 0.86 and -0.03 for NLCQ-1 + CPM and CPM alone, respectively. In general, NLCQ-1 in combination with nontoxic but inactive CPM doses (36 or 54 mg/kg, qd x 4) elicited good antitumor activity without subsequent additive systemic toxicity, whereas NLCQ-1 had minimal effect in combination with the active but toxic (> 10% mean net weight loss) CPM dose of 80 mg/kg. These results suggest a potential use of NLCQ-1 in the clinic as an adjuvant to chemotherapy with CPM.
Collapse
Affiliation(s)
- Maria V Papadopoulou
- Radiation Medicine Institute, Evanston Northwestern Healthcare, Evanston, IL 60201, USA.
| | | | | | | |
Collapse
|
37
|
Koch CJ. Measurement of absolute oxygen levels in cells and tissues using oxygen sensors and 2-nitroimidazole EF5. Methods Enzymol 2002; 352:3-31. [PMID: 12125356 DOI: 10.1016/s0076-6879(02)52003-6] [Citation(s) in RCA: 157] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We have established basic methods, using quantitative measures of EF5 binding, to estimate the actual pO2 of cells and tissues. In situations where the tissue can be dissociated into single cells, or for cell cultures, we can measure the distribution of cellular binding rates using flow cytometry and these can be compared with cells treated under pO2S controlled by the spinner vial or thin-film methods in vitro. The flow cytometer is calibrated by staining V79 cells treated with EF5 under "standard" conditions. For intact tissues treated with EF5 in vivo, we need to correct for possible variations in drug exposure (AUC). Frozen sections are stained for EF5 binding and are analyzed by a sensitive (cooled) CCD camera with linear output vs fluorescence [figure: see text] input. The camera has very consistent sensitivity, but the entire optical system, including the camera, can be calibrated by an absolute fluorescence standard (dye in hemocytometer). This system can also be used to measure the fluorescence of the flow cytometer standards, providing a direct link between the two assays. We can measure the maximum binding rate using the tissue cube method, but need to assume an "average" oxygen dependence of binding for intact tissues. The best-fit approximation for existing data is an inverse relationship between binding and pO2, with binding decreasing 50-fold between 0.1 and 10% oxygen. Using these methods, we routinely estimate the minimum pO2 (maximum binding) in experimental rodent and human tumors. In normal tissue models, an excellent correlation is found between near-maximal binding (severe hypoxia) and apoptosis (heart infarct and ductus arteriosus). Some normal tissues (e.g., skeletal muscle) are refractory to both cellular disaggregation and cube calibration methods. To extend the tissue imaging measurements to a complete two- or three-dimensional analysis of the distribution of tissue pO2s requires a substantial additional investment of imaging methods, which are currently being implemented.
Collapse
Affiliation(s)
- Cameron J Koch
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
38
|
Vukovic V, Nicklee T, Hedley DW. Multiparameter fluorescence mapping of nonprotein sulfhydryl status in relation to blood vessels and hypoxia in cervical carcinoma xenografts. Int J Radiat Oncol Biol Phys 2002; 52:837-43. [PMID: 11849809 DOI: 10.1016/s0360-3016(01)02716-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
PURPOSE Aberrant architecture of the tumor vasculature and temporal fluctuations in blood flow can result in tumor hypoxia. The aim of this study was to classify tumor hypoxia based on distance to blood vessels, and to characterize its biologic significance by determining levels of nonprotein sulfhydryls (NPSH) in hypoxic regions located proximally and distally to tumor blood vessels. METHODS AND MATERIALS A dual fluorescence method was developed for the spatial colocalization of the vasculature and hypoxia in frozen sections from SiHa cervical carcinoma xenografts. A parallel section was stained with the sulfhydryl stain mercury orange. Composite fluorescence images were generated by imaging and tiling individual fields of view into 2D image arrays. Image arithmetic techniques were combined with feature-based image segmentation to characterize expression of NPSH as a function of the hypoxic tumor microenvironment. RESULTS NPSH levels were higher in hypoxic areas of the SiHa xenografts (15.1 +/- 0.5 vs. 13.5 +/- 0.5 integrated optical density [IOD], p < 0.03). When tumor hypoxia was classified by distance to the nearest visible blood vessel, significantly higher NPSH levels were found in hypoxic regions close to blood vessels than in regions at a distance from blood vessels. CONCLUSION The results of this study indicate differential expression of NPSH levels in regions of hypoxia that are proximal or distal to blood vessels in SiHa tumors.
Collapse
Affiliation(s)
- Vojislav Vukovic
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
39
|
Affiliation(s)
- Ryan P Smith
- Radiation Oncology, The Hospital of the University of Pennsylvania, USA
| |
Collapse
|
40
|
Evans SM, Hahn SM, Magarelli DP, Koch CJ. Hypoxic heterogeneity in human tumors: EF5 binding, vasculature, necrosis, and proliferation. Am J Clin Oncol 2001; 24:467-72. [PMID: 11586098 DOI: 10.1097/00000421-200110000-00011] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We evaluated the levels and distribution of hypoxia in 31 human tumors using fluorescent immunohistochemical detection of binding by the 2-nitroimidazole, EF5. Hypoxia was found to be a heterogeneous property of human tumors. Necrosis was usually found adjacent to the highest level of binding in an individual patient's tumor. However, hypoxia often occurred without necrosis. In the group of tumors studied, the most common relationship between blood vessels (PECAM/CD31) and EF5 staining was consistent with diffusion-limited hypoxia; acute hypoxia occurred infrequently. Within a given patient's tumor, there was an inverse correlation between regions of proliferation (Ki-67) and regions of hypoxia. Again, however, when these parameters were examined in a group of patients, the absence of proliferation did not predict the presence of hypoxia. The relationships between hypoxia and other biologic endpoints are complex, but, within a given tumor's spatial relationships, they are in accord with known physiologic principles. Thus, our data emphasize that the relationships between hypoxia and other biologic parameters vary between patients. Necrosis, proliferation, and blood vessel distribution cannot predict the level or presence of hypoxia in an individual patient's tumor.
Collapse
Affiliation(s)
- S M Evans
- University of Pennsylvania, School of Medicine, Department of Radiation Oncology, Philadelphia, Pennsylvania 19104, USA
| | | | | | | |
Collapse
|
41
|
Josse O, Labar D, Georges B, Grégoire V, Marchand-Brynaert J. Synthesis of [18F]-labeled EF3 [2-(2-nitroimidazol-1-yl)-N-(3,3,3-trifluoropropyl)-acetamide], a marker for PET detection of hypoxia. Bioorg Med Chem 2001; 9:665-75. [PMID: 11310602 DOI: 10.1016/s0968-0896(00)00279-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
[18F]-2-(2-Nitroimidazol-1-yl)-N-(3,3,3-trifluoropropyl)-acetamide ([18F]-EF3) has been prepared, in 65% chemical yield and 5% radiochemical yield, by coupling 2,3,5,6-tetrafluorophenyl 2-(2-nitroimidazol-1-yl) acetate 1 with [18F]-3,3,3-trifluoropropylamine 7. This original radiolabelled key-synthon was obtained in 40% overall chemical yield by oxidative [18F]-fluorodesulfurization of ethyl N-phthalimido-3-aminopropane dithioate 4, followed by deprotection with hydrazine of the resulting [18F]-N-phthalimido-3,3,3-trifluoropropylamine 5. All the process was performed within 90 min, from the [18F]-HF production in the cyclotron to the purification of the final target.
Collapse
Affiliation(s)
- O Josse
- Unité de Chimie Organique et Médicinale, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | | | | | | | | |
Collapse
|
42
|
Fyles AW, Milosevic M, Pintilie M, Syed A, Hill RP. Anemia, hypoxia and transfusion in patients with cervix cancer: a review. Radiother Oncol 2000; 57:13-9. [PMID: 11033184 DOI: 10.1016/s0167-8140(00)00245-0] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND PURPOSE With the recent development of hemopoietic growth factors and alternatives to transfusion, there has been a renewed interest in the relationships between anemia, tumor hypoxia and treatment outcome in a number of human malignancies. This review is intended to provoke a reconsideration of these issues and their effect on clinical trials, aimed at improving treatment outcome in patients with cervix cancer. MATERIALS AND METHODS Using data from the literature and from our own prospective series of tumor oxygenation in cervix cancer, we modeled the impact of anemia on tumor blood flow and hypoxia in animal models and human tumors, examined the relationship between anemia and hypoxia and treatment outcome in patients, and reviewed the impact of transfusion on tumor hypoxia and treatment outcome in cervix cancer. RESULTS Anemia may result in a significant reduction in oxygen delivery to tumors, but compensatory mechanisms reduce the impact on tumor oxygenation. Anemia is associated with inferior treatment outcome in cervix cancer, but hemoglobin levels prior to and during treatment are strongly correlated with tumor size, and this may explain the prognostic impact of anemia in older studies. Transfusion and erythropoietin ameliorate hypoxia in only a proportion of anemic patients. Critical analysis of the published data from the Princess Margaret Hospital randomized trial of transfusion in cervix cancer reveals that, when analyzed by intention-to-treat, transfusion did not result in a benefit to patients. CONCLUSIONS This review suggests that the relationships among anemia, hypoxia, transfusion and treatment outcome are complex. Further study of anemia as an independent prognostic factor is required and randomized studies of transfusion alternatives, such as erythropoietin, must be of sufficient size to detect small treatment effects.
Collapse
Affiliation(s)
- A W Fyles
- Department of Radiation Oncology, Princess Margaret Hospital/University Health Network, 610 University Ave., Ontario M5G 2M9, Toronto, Canada
| | | | | | | | | |
Collapse
|
43
|
Siemann DW, Warrington KH, Horsman MR. Targeting tumor blood vessels: an adjuvant strategy for radiation therapy. Radiother Oncol 2000; 57:5-12. [PMID: 11033183 DOI: 10.1016/s0167-8140(00)00243-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND PURPOSE The neovascularization of tumor cells is a prerequisite if a clinically relevant tumor size is to be reached. A continuously expanding vessel network supplying nutritional requirements and removing waste products is essential for continued tumor development, growth and survival. RESULTS In many tumors, the growing endothelium is unable to fully support the demands of the neoplastic cell population. As a consequence of the inadequacies of the resulting aberrant vasculature, microenvironmental conditions develop in tumors which are not only detrimental to the response of tumors to conventional anticancer treatments, but may lead to or predispose cells to genetic modifications resulting in more aggressive phenotypes and higher metastatic potential. Yet the utter dependence of the tumor on its induced vessel formation for growth, survival and spread has also created a great deal of enthusiasm for developing therapeutic approaches to specifically targeting the tumor microcirculation. CONCLUSIONS The application of such strategies as adjuvants to conventional radiation treatments offers unique opportunities to develop more effective cancer therapies.
Collapse
Affiliation(s)
- D W Siemann
- Department of Radiation Oncology, Shands Cancer Center, University of Florida, Box 100385, Gainesville, FL 32610, USA
| | | | | |
Collapse
|
44
|
Conover DL, Fenton BM, Foster TH, Hull EL. An evaluation of near infrared spectroscopy and cryospectrophotometry estimates of haemoglobin oxygen saturation in a rodent mammary tumour model. Phys Med Biol 2000; 45:2685-700. [PMID: 11008965 DOI: 10.1088/0031-9155/45/9/318] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Haemoglobin oxygen saturation in subcutaneous rat mammary tumours was measured using near infrared spectroscopy (NIRS) in vivo and in rapidly frozen sections from the same tumours using cryospectrophotometry, which reports oxygen saturation in individual blood vessels to depths of 4 mm from the tissue surface. Measurements were performed on tumours while animals breathed either room air or carbogen. In five of nine tumours, the average saturation calculated from cryospectrophotometric measurements agreed with that determined from NIRS to within 13%, and in four of these five tumours agreement was 5% or better. In the remaining four of nine tumours, where agreement was poor, the volume-averaged saturations estimated from NIRS were consistently higher than those calculated from cryospectrophotometry. Monte Carlo simulations demonstrated that the depth of tissue probed by NIRS was significantly greater than that sampled by cryospectrophotometry. Analysis of the frequency of severely hypoxic vessels showed that when NIRS reported a saturation of approximately 70% or higher, the fraction of tumour vessels with saturations less than 10% was limited to 0.06 or less. Sensitivity and specificity analysis suggests that NIRS and NIRS imaging may identify clinically relevant hypoxia, even when its spatial extent is below the resolution limit of the NIRS technique.
Collapse
Affiliation(s)
- D L Conover
- Department of Radiology, University of Rochester Medical Center, NY 14642, USA
| | | | | | | |
Collapse
|
45
|
Vaupel P, Hoeckel M. Predictive power of the tumor oxygenation status. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2000; 471:533-9. [PMID: 10659187 DOI: 10.1007/978-1-4615-4717-4_63] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
We have previously demonstrated with a prospective study in primary cancer of the uterine cervix (clinical size > 2 cm) that tumor hypoxia not only indicates decreased radiocurability but is generally associated with malignant progression of the disease. This finding also holds true for other tumor entities (soft tissue sarcomas, squamous cell carcinomas of the head and neck region), for lymph node metastases of head and neck lesions, and for locoregional recurrences of cervical cancers. All data available so far support our thesis that in cervical cancers (and in other solid tumors as well), tumor hypoxia and clinical aggressiveness in terms of resistance to therapy and tumor dissemination, are interrelated.
Collapse
Affiliation(s)
- P Vaupel
- Institute of Physiology and Pathophysiology, University of Mainz, Germany
| | | |
Collapse
|
46
|
Karkan DM, van Breemen C, Skarsgard PL, Lagaud GJ, Hardwick DF. A link between vasomotion and spontaneous oscillations of oxygen in rat brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2000; 471:111-6. [PMID: 10659137 DOI: 10.1007/978-1-4615-4717-4_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Affiliation(s)
- D M Karkan
- Department of Pharmacology and Therapeutics, University of British Columbia
| | | | | | | | | |
Collapse
|
47
|
Hustinx R, Eck SL, Alavi A. Potential Applications of PET Imaging in Developing Novel Cancer Therapies. J Vasc Interv Radiol 2000. [DOI: 10.1016/s1051-0443(00)70185-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
48
|
Becker A, Stadler P, Lavey RS, Hänsgen G, Kuhnt T, Lautenschläger C, Feldmann HJ, Molls M, Dunst J. Severe anemia is associated with poor tumor oxygenation in head and neck squamous cell carcinomas. Int J Radiat Oncol Biol Phys 2000; 46:459-66. [PMID: 10661354 DOI: 10.1016/s0360-3016(99)00384-3] [Citation(s) in RCA: 161] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE To investigate the relationship between tumor oxygenation and the blood hemoglobin (Hb) concentration in patients with squamous cell carcinoma of the head and neck (SCCHN). METHODS AND MATERIALS A total of 133 patients with SCCHN underwent pretreatment polarographic pO2 measurements of their tumors. In 66 patients measurements were also made in sternocleidomastoid muscles. The patients were divided into three groups according to their Hb concentration-severe anemia (Hb < 11.0 g/dl), mild anemia (female: Hb 11.0-11.9 g/dl; male: Hb 11.0-12.9 g/dl), and normal Hb concentration (female: Hb > or =12.0 g/dl; male: > or =13.0 g/dl). RESULTS No significant difference in tumor oxygenation could be detected between mildly anemic patients and patients with a normal Hb level. However, the tumor oxygenation in the severely anemic group was significantly below that of each of the other two groups (p < 0.0001). There was no significant difference between the Hb groups in oxygenation of sternocleidomastoid muscles. In a multivariate analysis including Hb group, tumor volume, smoking habits, gender, T-stage, N-stage, and histologic grade a Hb level < 11 g/dl was found to be the strongest predictor for a poor tumor oxygenation. Smoking also had a marginal influence on median pO2. CONCLUSION Our data suggest that a low Hb concentration and cigarette smoking contribute to inadequate oxygenation of SCCHN and thus for increased radioresistance. Consequently, Hb correction and abstinence from smoking may significantly improve tumor oxygenation.
Collapse
Affiliation(s)
- A Becker
- Department of Radiooncology, Martin Luther University, Halle-Wittenberg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
|
50
|
Vujaskovic Z, Poulson JM, Gaskin AA, Thrall DE, Page RL, Charles HC, MacFall JR, Brizel DM, Meyer RE, Prescott DM, Samulski TV, Dewhirst MW. Temperature-dependent changes in physiologic parameters of spontaneous canine soft tissue sarcomas after combined radiotherapy and hyperthermia treatment. Int J Radiat Oncol Biol Phys 2000; 46:179-85. [PMID: 10656391 DOI: 10.1016/s0360-3016(99)00362-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE The objectives of this study were to evaluate effects of hyperthermia on tumor oxygenation, extracellular pH (pHe), and blood flow in 13 dogs with spontaneous soft tissue sarcomas prior to and after local hyperthermia. METHODS AND MATERIALS Tumor pO2 was measured using an Eppendorf polarographic device, pHe using interstitial electrodes, and blood flow using contrast-enhanced magnetic resonance imaging (MRI). RESULTS There was an overall improvement in tumor oxygenation observed as an increase in median pO2 and decrease in hypoxic fraction (% of pO2 measurements <5 mm Hg) at 24-h post hyperthermia. These changes were most pronounced when the median temperature (T50) during hyperthermia treatment was less than 44 degrees C. Tumors with T50 > 44 degrees C were characterized by a decrease in median PO2 and an increase in hypoxic fraction. Similar thermal dose-related changes were observed in tumor perfusion. Perfusion was significantly higher after hyperthermia. Increases in perfusion were most evident in tumors with T50 < 44 degrees C. With T50 > 44 degrees C, there was no change in perfusion after hyperthermia. On average, pHe values declined in all animals after hyperthermia, with the greatest reduction seen for larger T50 values. CONCLUSION This study suggests that hyperthermia has biphasic effects on tumor physiologic parameters. Lower temperatures tend to favor improved perfusion and oxygenation, whereas higher temperatures are more likely to cause vascular damage, thus leading to greater hypoxia. While it has long been recognized that such effects occur in rodent tumors, this is the first report to tie such changes to temperatures achieved during hyperthermia in the clinical setting. Furthermore, it suggests that the thermal threshold for vascular damage is higher in spontaneous tumors than in more rapidly growing rodent tumors.
Collapse
Affiliation(s)
- Z Vujaskovic
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|