1
|
Dewhirst MW, Oleson JR, Kirkpatrick J, Secomb TW. Accurate Three-Dimensional Thermal Dosimetry and Assessment of Physiologic Response Are Essential for Optimizing Thermoradiotherapy. Cancers (Basel) 2022; 14:1701. [PMID: 35406473 PMCID: PMC8997141 DOI: 10.3390/cancers14071701] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Numerous randomized trials have revealed that hyperthermia (HT) + radiotherapy or chemotherapy improves local tumor control, progression free and overall survival vs. radiotherapy or chemotherapy alone. Despite these successes, however, some individuals fail combination therapy; not every patient will obtain maximal benefit from HT. There are many potential reasons for failure. In this paper, we focus on how HT influences tumor hypoxia, since hypoxia negatively influences radiotherapy and chemotherapy response as well as immune surveillance. Pre-clinically, it is well established that reoxygenation of tumors in response to HT is related to the time and temperature of exposure. In most pre-clinical studies, reoxygenation occurs only during or shortly after a HT treatment. If this were the case clinically, then it would be challenging to take advantage of HT induced reoxygenation. An important question, therefore, is whether HT induced reoxygenation occurs in the clinic that is of radiobiological significance. In this review, we will discuss the influence of thermal history on reoxygenation in both human and canine cancers treated with thermoradiotherapy. Results of several clinical series show that reoxygenation is observed and persists for 24-48 h after HT. Further, reoxygenation is associated with treatment outcome in thermoradiotherapy trials as assessed by: (1) a doubling of pathologic complete response (pCR) in human soft tissue sarcomas, (2) a 14 mmHg increase in pO2 of locally advanced breast cancers achieving a clinical response vs. a 9 mmHg decrease in pO2 of locally advanced breast cancers that did not respond and (3) a significant correlation between extent of reoxygenation (as assessed by pO2 probes and hypoxia marker drug immunohistochemistry) and duration of local tumor control in canine soft tissue sarcomas. The persistence of reoxygenation out to 24-48 h post HT is distinctly different from most reported rodent studies. In these clinical series, comparison of thermal data with physiologic response shows that within the same tumor, temperatures at the higher end of the temperature distribution likely kill cells, resulting in reduced oxygen consumption rate, while lower temperatures in the same tumor improve perfusion. However, reoxygenation does not occur in all subjects, leading to significant uncertainty about the thermal-physiologic relationship. This uncertainty stems from limited knowledge about the spatiotemporal characteristics of temperature and physiologic response. We conclude with recommendations for future research with emphasis on retrieving co-registered thermal and physiologic data before and after HT in order to begin to unravel complex thermophysiologic interactions that appear to occur with thermoradiotherapy.
Collapse
Affiliation(s)
- Mark W Dewhirst
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - James R Oleson
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - John Kirkpatrick
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Timothy W Secomb
- Department of Physiology, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
2
|
Cirrik S, Ugurel E, Aksu AC, Oronsky B, Cabrales P, Yalcin O. Nitrite may serve as a combination partner and a biomarker for the anti-cancer activity of RRx-001. Biorheology 2019; 56:221-235. [DOI: 10.3233/bir-190213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
| | - Elif Ugurel
- Koc University School of Medicine, , , Turkey
| | | | | | | | | |
Collapse
|
3
|
Reymen BJT, van Gisbergen MW, Even AJG, Zegers CML, Das M, Vegt E, Wildberger JE, Mottaghy FM, Yaromina A, Dubois LJ, van Elmpt W, De Ruysscher D, Lambin P. Nitroglycerin as a radiosensitizer in non-small cell lung cancer: Results of a prospective imaging-based phase II trial. Clin Transl Radiat Oncol 2019; 21:49-55. [PMID: 32021913 PMCID: PMC6993056 DOI: 10.1016/j.ctro.2019.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023] Open
Abstract
Nitroglycerin didn’t improve overall survival of NSCLC patients. The toxicity of combining nitroglycerin with standard treatment was mild. Increased uptake of HX4 showed negative prognostic significance in NSCLC patients. Tumor perfusion after nitroglycerin treatment did not correlate with outcome.
Background Nitroglycerin is proposed as an agent to reduce tumour hypoxia by improving tumour perfusion. We investigated the potential of nitroglycerin as a radio-sensitizer in non-small cell lung cancer (NSCLC) and the potential of functional imaging for patient selection. Material and methods Trial NCT01210378 is a single arm phase II trial, designed to detect 15% improvement in 2-year overall survival (primary endpoint) in stage IB-IV NSCLC patients treated with radical (chemo-) radiotherapy and a Transiderm-Nitro 5 patch during radiotherapy. Patients underwent dynamic contrast-enhanced CTs (DCE-CT) and HX4 (hypoxia) PET/CTs before and after nitroglycerin. Secondary endpoints were progression-free survival, toxicity and the prognostic value of tumour perfusion/hypoxia at baseline and after nitroglycerin. Results The trial stopped after a futility analysis after 42 patients. At median follow-up of 41 months, two-year and median OS were 58% (95% CI: 44–78%) and 38 months (95% CI: 22–54 months), respectively. Nitroglycerin could not reduce tumour hypoxia. DCE-CT parameters did not correlate with OS, whereas hypoxic tumours had a worse OS (p = 0.029). Changes in high-uptake fraction of HX4 and tumour blood flow were negatively correlated (r = -0.650, p = 0.022). The heterogeneity in treatment modalities and patient characteristics combined with a small sample size made further subgroup analysis of survival results impossible. Toxicity related to nitroglyerin was limited to headache (17%) and hypotension (2.4%). Conclusion Nitroglycerin did not improve OS of NSCLC patients treated with (chemo-)radiotherapy. A general ability of nitroglycerin to reduce hypoxia was not shown.
Collapse
Key Words
- BF, blood flow
- BV, blood volume
- CI, confidence interval
- CoR, coefficient of repeatability
- DCE-CT, dynamic contrast-enhanced CT
- FHV, fraction of hypoxic volume hypoxic fraction of the GTV
- GTV, gross tumour volume
- GTVln, gross tumour volume of the lymph nodes
- GTVp, gross tumour volume of the primary tumour
- HX4
- HX4, 2-nitroimidazole [18F]-HX4 (flortanidazole, 3-[18F]fluoro-2-(4-((2-nitro-1Himidazol-1-yl)methyl)-1H-1,2,3-triazol-1-yl)-propan-1-ol)
- HX4-HF, HX4 hypoxic fraction
- HX4-HV, HX4 hypoxic volume
- Hypoxia
- INDAR, individualized accelerated radiotherapy
- IQR, interquartile range
- LRPFS, loco-regional progression free survival
- MFS, metastasis-free survival
- Mitochondria
- NO, nitric oxide
- NSCLC
- NSCLC, non-small cell lung cancer
- Nitroglycerin
- OS, overall survival
- PET, positron emission tomography
- Perfusion
- SUVmax, maximum standardised uptake value
- SUVmean, mean standardised uptake value
- TBR, tumour-to-blood ratio
- TTD, total tumour dose
Collapse
Affiliation(s)
- Bart J T Reymen
- Department of Radiation Oncology (MAASTRO), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Marike W van Gisbergen
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Aniek J G Even
- Department of Radiation Oncology (MAASTRO), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Catharina M L Zegers
- Department of Radiation Oncology (MAASTRO), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Institute of Data Science, Maastricht University, The Netherlands
| | - Marco Das
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Erik Vegt
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Joachim E Wildberger
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Felix M Mottaghy
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Nuclear Medicine, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Ludwig J Dubois
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Wouter van Elmpt
- Department of Radiation Oncology (MAASTRO), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Dirk De Ruysscher
- Department of Radiation Oncology (MAASTRO), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Philippe Lambin
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.,Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
4
|
Papaverine and its derivatives radiosensitize solid tumors by inhibiting mitochondrial metabolism. Proc Natl Acad Sci U S A 2018; 115:10756-10761. [PMID: 30201710 DOI: 10.1073/pnas.1808945115] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Tumor hypoxia reduces the effectiveness of radiation therapy by limiting the biologically effective dose. An acute increase in tumor oxygenation before radiation treatment should therefore significantly improve the tumor cell kill after radiation. Efforts to increase oxygen delivery to the tumor have not shown positive clinical results. Here we show that targeting mitochondrial respiration results in a significant reduction of the tumor cells' demand for oxygen, leading to increased tumor oxygenation and radiation response. We identified an activity of the FDA-approved drug papaverine as an inhibitor of mitochondrial complex I. We also provide genetic evidence that papaverine's complex I inhibition is directly responsible for increased oxygenation and enhanced radiation response. Furthermore, we describe derivatives of papaverine that have the potential to become clinical radiosensitizers with potentially fewer side effects. Importantly, this radiosensitizing strategy will not sensitize well-oxygenated normal tissue, thereby increasing the therapeutic index of radiotherapy.
Collapse
|
5
|
Tahara Y, Yoshikawa T, Sato H, Mori Y, Zahangir MH, Kishimura A, Mori T, Katayama Y. Encapsulation of a nitric oxide donor into a liposome to boost the enhanced permeation and retention (EPR) effect. MEDCHEMCOMM 2017; 8:415-421. [PMID: 30108759 PMCID: PMC6072363 DOI: 10.1039/c6md00614k] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 12/12/2016] [Indexed: 02/05/2023]
Abstract
We propose a method to improve the enhanced permeability and retention (EPR) effect of nanomedicines based on tumor-specific vasodilation using a nitric oxide (NO) donor-containing liposome. NONOate, a typical NO donor, was incorporated into a PEGylated liposome to retard the protonation-induced release of NO from NONOate by the protecting lipid bilayer membrane. The NONOate-containing liposome (NONOate-LP) showed similar blood retention to an empty PEGylated liposome but almost twice the amount accumulated within the tumor. This improvement in the EPR effect is thought to have been caused by specific vasodilation in the tumor tissue by NO released from the NONOate-LP accumulated in the tumor. The improved EPR effect by NONOate-LP will be useful for the accumulation of co-administered nanomedicines.
Collapse
Affiliation(s)
- Yu Tahara
- Graduate School of Systems Life Sciences , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan . ; Tel: +81 092 802 2850
| | - Takuma Yoshikawa
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan
| | - Hikari Sato
- Graduate School of Systems Life Sciences , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan . ; Tel: +81 092 802 2850
| | - Yukina Mori
- Graduate School of Systems Life Sciences , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan . ; Tel: +81 092 802 2850
| | - Md Hosain Zahangir
- Graduate School of Systems Life Sciences , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan . ; Tel: +81 092 802 2850
| | - Akihiro Kishimura
- Graduate School of Systems Life Sciences , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan . ; Tel: +81 092 802 2850
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan
- International Research Center for Molecular Systems , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan
| | - Takeshi Mori
- Graduate School of Systems Life Sciences , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan . ; Tel: +81 092 802 2850
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan
| | - Yoshiki Katayama
- Graduate School of Systems Life Sciences , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan . ; Tel: +81 092 802 2850
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan
- International Research Center for Molecular Systems , Kyushu University , 744 Motooka , Nishi-ku , Fukuoka 819-0395 , Japan
- Center for Advanced Medical Innovation , Kyushu University , 744 Motooka , Nishi-Ku , Fukuoka 819-0395 , Japan
- Department of Biomedical Engineering , Chung Yuan Christian University , 200 Chung Pei Rd. , Chung Li , Taiwan , 32023 ROC
| |
Collapse
|
6
|
Rapozzi V, Varchi G, Della Pietra E, Ferroni C, Xodo LE. A photodynamic bifunctional conjugate for prostate cancer: an in vitro mechanistic study. Invest New Drugs 2016; 35:115-123. [PMID: 27726093 DOI: 10.1007/s10637-016-0396-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/28/2016] [Indexed: 01/12/2023]
Abstract
Photodynamic therapy (PDT) has drawn considerable attention for its efficacy against certain types of cancers. It shows however limits in the case of deep cancers, favoring tumor recurrence under suboptimal conditions. More insight into the molecular mechanisms of PDT-induced cytotoxicity and cytoprotection is essential to extend and strengthen this therapeutic modality. As PDT induces iNOS/NO in both tumor and microenvironment, we examined the role of nitric oxide (NO) in cytotoxicity and cytoprotection. Our findings show that NO mediates its cellular effects by acting on the NF-κB/YY1/RKIP loop, which controls cell growth and apoptosis. The cytoprotective effect of PDT-induced NO is observed at low NO levels, which activate the pro-survival/anti-apoptotic NF-κB and YY1, while inhibiting the anti-survival/pro-apoptotic and metastasis suppressor RKIP. In contrast, high PDT-induced NO levels inhibit NF-κB and YY1 and induce RKIP, resulting in significant anti-tumor activity. These findings reveal a critical role played by NO in PDT and suggest that the use of bifunctional PDT agents composed of a photosensitizer and a NO-donor could enhance the photo-treatment effect. A successful application of NO in anticancer therapy requires control of its concentration in the target tissue. To address this issue we propose as PDT agent, a bimolecular conjugate called DR2, composed of a photosensitizer (Pheophorbide a) and a non-steroidal anti-androgen molecule capable of releasing NO under the exclusive control of light. The mechanism of action of DR2 in prostate cancer cells is reported and discussed.
Collapse
Affiliation(s)
- Valentina Rapozzi
- Department of Medical and Biological Sciences, University of Udine, Piazzale Kolbe 4, 33100, Udine, Italy.
| | - Greta Varchi
- Institute of Organic Synthesis and Photoreactivity, Italian National Research Council, Via P. Gobetti 101, 40129, Bologna, Italy
| | - Emilia Della Pietra
- Department of Medical and Biological Sciences, University of Udine, Piazzale Kolbe 4, 33100, Udine, Italy
| | - Claudia Ferroni
- Institute of Organic Synthesis and Photoreactivity, Italian National Research Council, Via P. Gobetti 101, 40129, Bologna, Italy
| | - Luigi E Xodo
- Department of Medical and Biological Sciences, University of Udine, Piazzale Kolbe 4, 33100, Udine, Italy
| |
Collapse
|
7
|
NO to cancer: The complex and multifaceted role of nitric oxide and the epigenetic nitric oxide donor, RRx-001. Redox Biol 2015; 6:1-8. [PMID: 26164533 PMCID: PMC4529402 DOI: 10.1016/j.redox.2015.07.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 06/30/2015] [Accepted: 07/01/2015] [Indexed: 01/18/2023] Open
Abstract
The endogenous mediator of vasodilation, nitric oxide (NO), has been shown to be a potent radiosensitizer. However, the underlying mode of action for its role as a radiosensitizer – while not entirely understood – is believed to arise from increased tumor blood flow, effects on cellular respiration, on cell signaling, and on the production of reactive oxygen and nitrogen species (RONS), that can act as radiosensitizers in their own right. NO activity is surprisingly long-lived and more potent in comparison to oxygen. Reports of the effects of NO with radiation have often been contradictory leading to confusion about the true radiosensitizing nature of NO. Whether increasing or decreasing tumor blood flow, acting as radiosensitizer or radioprotector, the effects of NO have been controversial. Key to understanding the role of NO as a radiosensitizer is to recognize the importance of biological context. With a very short half-life and potent activity, the local effects of NO need to be carefully considered and understood when using NO as a radiosensitizer. The systemic effects of NO donors can cause extensive side effects, and also affect the local tumor microenvironment, both directly and indirectly. To minimize systemic effects and maximize effects on tumors, agents that deliver NO on demand selectively to tumors using hypoxia as a trigger may be of greater interest as radiosensitizers. Herein we discuss the multiple effects of NO and focus on the clinical molecule RRx-001, a hypoxia-activated NO donor currently being investigated as a radiosensitizer in the clinic. . NO radiosensitizes by reaction with DNA radicals, by its metabolites and by impact on the vasculature. Understanding the local and context-specific activity of NO is key for radiosensitizer development RRx-001 induces NO production under hypoxia with promising radiosensitizing activity.
Collapse
|
8
|
Jones LW, Fels DR, West M, Allen JD, Broadwater G, Barry WT, Wilke LG, Masko E, Douglas PS, Dash RC, Povsic TJ, Peppercorn J, Marcom PK, Blackwell KL, Kimmick G, Turkington TG, Dewhirst MW. Modulation of circulating angiogenic factors and tumor biology by aerobic training in breast cancer patients receiving neoadjuvant chemotherapy. Cancer Prev Res (Phila) 2013; 6:925-37. [PMID: 23842792 DOI: 10.1158/1940-6207.capr-12-0416] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Aerobic exercise training (AET) is an effective adjunct therapy to attenuate the adverse side-effects of adjuvant chemotherapy in women with early breast cancer. Whether AET interacts with the antitumor efficacy of chemotherapy has received scant attention. We carried out a pilot study to explore the effects of AET in combination with neoadjuvant doxorubicin-cyclophosphamide (AC+AET), relative to AC alone, on: (i) host physiology [exercise capacity (VO2 peak), brachial artery flow-mediated dilation (BA-FMD)], (ii) host-related circulating factors [circulating endothelial progenitor cells (CEP) cytokines and angiogenic factors (CAF)], and (iii) tumor phenotype [tumor blood flow ((15)O-water PET), tissue markers (hypoxia and proliferation), and gene expression] in 20 women with operable breast cancer. AET consisted of three supervised cycle ergometry sessions/week at 60% to 100% of VO2 peak, 30 to 45 min/session, for 12 weeks. There was significant time × group interactions for VO2 peak and BA-FMD, favoring the AC+AET group (P < 0.001 and P = 0.07, respectively). These changes were accompanied by significant time × group interactions in CEPs and select CAFs [placenta growth factor, interleukin (IL)-1β, and IL-2], also favoring the AC+AET group (P < 0.05). (15)O-water positron emission tomography (PET) imaging revealed a 38% decrease in tumor blood flow in the AC+AET group. There were no differences in any tumor tissue markers (P > 0.05). Whole-genome microarray tumor analysis revealed significant differential modulation of 57 pathways (P < 0.01), including many that converge on NF-κB. Data from this exploratory study provide initial evidence that AET can modulate several host- and tumor-related pathways during standard chemotherapy. The biologic and clinical implications remain to be determined.
Collapse
Affiliation(s)
- Lee W Jones
- Duke Cancer Institute, Duke University Medical Center, Box 3085, Durham, NC 27710, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Giles NM, Kumari S, Gang BP, Yuen CWW, Billaud EMF, Giles GI. The molecular design of S-nitrosothiols as photodynamic agents for controlled nitric oxide release. Chem Biol Drug Des 2012; 80:471-8. [PMID: 22642531 DOI: 10.1111/j.1747-0285.2012.01420.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nitric oxide is a small messenger molecule utilized by nature in cell signalling and the non-specific immune response. At present, nitric oxide releasing prodrugs cannot be efficiently targeted towards a specific body compartment, which restricts their therapeutic applications. To address this limitation, we have designed two photolabile nitric oxide releasing prodrugs, tert-butyl S-nitrosothiol and tert-dodecane S-nitrosothiol, which are based on the S-nitrosothiol functionality. By modulating the prodrugs' hydrophobicity, we postulated that we could increase their stability within the cell by preventing their interaction with hydrophilic thiols and metal ions; processes that are known to inactivate this prodrug class. Our data demonstrate that these prodrugs have improved nitric oxide release kinetics compared to currently available S-nitrosothiols, as they are highly stable in vitro in the absence of irradiation (t(1/2) > 3 h), while their rate of decomposition can be regulated by controlling the intensity or duration of the photostimulus. Nitric oxide release can readily be achieved using non-laser based light sources, which enabled us to characterize photoactivation as a trigger mechanism for nitric oxide release in A549 lung carcinoma cells. Here we confirmed that irradiation induced highly significant increases in cytotoxicity within a therapeutic drug range (1-100 μm), and the utility of this photoactivation switch opens up avenues for exploring the applications of these prodrugs for chemical biology studies and chemotherapy.
Collapse
Affiliation(s)
- Niroshini M Giles
- Department of Pharmacology and Toxicology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | | | | | | | | | | |
Collapse
|
10
|
Beyond antiangiogenesis: vascular modulation as an anticancer therapy-a review. Transl Oncol 2012; 5:133-40. [PMID: 22741032 DOI: 10.1593/tlo.12118] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 02/16/2012] [Accepted: 03/14/2012] [Indexed: 12/19/2022] Open
Abstract
This review attempts to move beyond the traditional borders of antiangiogenesis and toward the dynamic, evolving strategies of vascular modulation. This repositioning entails a two-fold paradigm shift: conceptually, to a view of antiangiogenesis as only one part of a larger story, and therapeutically, to approaches which attempt to modulate tumor blood flow instead of simply inhibiting it. Three vascular modulation strategies-provascular, antivascular, and redistributive-are presented with representative compounds. These vascular modulation strategies are described in specific measurable characteristics (blood vessel maturity and type, effect on blood flow, microenvironmental target, molecular target, angiogenic biomarker, and imaging biomarkers) that will help define the tumor types that are more susceptible to a particular vascular modulation strategy thereby guiding therapeutic agent selection and enabling a personalized medicine approach.
Collapse
|
11
|
Is Nitric Oxide (NO) the Last Word in Radiosensitization? A Review. Transl Oncol 2012; 5:66-71. [PMID: 22496921 DOI: 10.1593/tlo.11307] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 12/03/2011] [Accepted: 12/07/2011] [Indexed: 12/20/2022] Open
Abstract
As a short-lived radical that diffuses across membranes, rather than interacting with membrane-bound receptors, nitric oxide (NO) represents a significant departure from synthetically derived radiosensitizers. An endogenous compound, NO may equal or surpass its molecular cousin, oxygen, as a hypoxic radiosensitizer, through pleiotropic phenotypic effects on tumor perfusion, cell signaling, mitochondrial respiration, the fixation of radiation-induced damage, and the radioprotection of normal tissue. However, unlike oxygen, in the context of radiosensitization, the clinical role and utility of NO are poorly understood, with often contradictory and controversial reported effects: whether NO functions as a radiosensitizer may ultimately be contextual to the tumor microenvironment. This may make NO manipulation an ideal candidate for a personalized radiosensitization approach tailored to specific patient and tumor types/microenvironmental characteristics. Effective delivery of NO both systemically and directly to the tumor may be critical to the success of this approach. Compounds that release NO or NO precursors have the potential to drive innovation and result in a new fertile branch of the radiosensitizer tree.
Collapse
|
12
|
Six degrees of separation: the oxygen effect in the development of radiosensitizers. Transl Oncol 2011; 4:189-98. [PMID: 21804913 DOI: 10.1593/tlo.11166] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 05/06/2011] [Accepted: 05/11/2011] [Indexed: 11/18/2022] Open
Abstract
The popular theory six degrees of separation is used in this review as an analogy to relate all radiosensitization to oxygen. As the prime mover of all radiosensitizers, the pervasive influence of oxygen has consciously or unconsciously influenced the direction of research and development and provided the benchmark against which all other compounds and approaches are measured. It is the aim of this review to develop the six degrees of separation from oxygen analogy as a unifying framework for conceptually organizing the field and for giving context to its varied subspecializations and theories. Under such a framework, it would become possible for one area to consider questions and problems found in other areas of radiosensitization, using a common analogy, that would allow for further development and unification of this multifaceted discipline. In this review, approaches to the development of radiosensitizers and the current state of research in this field are discussed, including promising new agents in various stages of clinical development.
Collapse
|
13
|
Sonveaux P, Jordan BF, Gallez B, Feron O. Nitric oxide delivery to cancer: Why and how? Eur J Cancer 2009; 45:1352-69. [DOI: 10.1016/j.ejca.2008.12.018] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Accepted: 12/12/2008] [Indexed: 02/07/2023]
|
14
|
Thrombospondin 1 and vasoactive agents indirectly alter tumor blood flow. Neoplasia 2008; 10:886-96. [PMID: 18670646 DOI: 10.1593/neo.08264] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2008] [Revised: 05/20/2008] [Accepted: 05/21/2008] [Indexed: 12/21/2022] Open
Abstract
Nitric oxide (NO) plays important physiological roles in the vasculature to regulate angiogenesis, blood flow, and hemostasis. In solid tumors, NO is generally acknowledged to mediate angiogenic responses to several growth factors. This contrasts with conflicting evidence that NO can acutely increase tumor perfusion through local vasodilation or diminish perfusion by preferential relaxation of peripheral vascular beds outside the tumor. Because thrombospondin 1 (TSP1) is an important physiological antagonist of NO in vascular cells, we examined whether, in addition to inhibiting tumor angiogenesis, TSP1 can acutely regulate tumor blood flow. We assessed this activity of TSP1 in the context of perfusion responses to NO as a vasodilator and epinephrine as a vasoconstrictor. Nitric oxide treatment of wild type and TSP1 null mice decreased perfusion of a syngeneic melanoma, whereas epinephrine transiently increased tumor perfusion. Acute vasoactive responses were also independent of the level of tumor-expressed TSP1 in a melanoma xenograft, but recovery of basal perfusion was modulated by TSP1 expression. In contrast, overexpression of truncated TSP1 lacking part of its CD47 binding domain lacked this modulating activity. These data indicate that TSP1 primarily regulates long-term vascular responses in tumors, in part, because the tumor vasculature has a limited capacity to acutely respond to vasoactive agents.
Collapse
|
15
|
iNOS as a therapeutic target for treatment of human tumors. Nitric Oxide 2008; 19:217-24. [PMID: 18515106 DOI: 10.1016/j.niox.2008.05.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2008] [Revised: 05/02/2008] [Accepted: 05/05/2008] [Indexed: 01/25/2023]
Abstract
Nitric oxide synthase (NOS) has been shown to be overexpressed in a number of human tumors compared to normal tissues and therefore potentially represents an exploitable target in future anticancer therapies. To achieve this, there will be a need to profile tumors to identify those expressing high levels of NOS; alternatively, endogenous (low) levels of NOS could be modulated by induction or through gene therapy approaches. NOS consists of a reductase domain which shares a high degree of sequence homology with P450 reductase and this domain supplies reducing equivalents to a haem containing oxygenase domain that is responsible for the production of nitric oxide. Thus, there are a number of routes of exploitation. Firstly, to take advantage of the reductase domain to activate bioreductive drugs as has been exemplified with tirapazamine and now extended to AQ4N (1,4-bis{2-(dimethylamino-N-oxide)ethylamino}5,8-dihydroxy-anthracene-9,10-dione). Secondly, to take advantage of nitric oxide production for its ability to increase the sensitivity of resistant hypoxic cells to radiation. Lastly, to utilize inhibition of HIF-1 to amplify NO based therapies. In this review we provide examples/evidence of how these objectives can be achieved.
Collapse
|
16
|
Sonveaux P. Provascular strategy: Targeting functional adaptations of mature blood vessels in tumors to selectively influence the tumor vascular reactivity and improve cancer treatment. Radiother Oncol 2008; 86:300-13. [PMID: 18313779 DOI: 10.1016/j.radonc.2008.01.024] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Revised: 01/30/2008] [Accepted: 01/30/2008] [Indexed: 12/22/2022]
|
17
|
Wardman P, Rothkamm K, Folkes LK, Woodcock M, Johnston PJ. Radiosensitization by nitric oxide at low radiation doses. Radiat Res 2007; 167:475-84. [PMID: 17388699 DOI: 10.1667/rr0827.1] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2006] [Accepted: 11/27/2006] [Indexed: 11/03/2022]
Abstract
Nitric oxide was shown to radiosensitize anoxic V79 and CHO hamster cells and MCF7 and UT-SCC-14 human cells, measuring clonogenic survival and/or DNA damage in vitro at low radiation doses (0.1-5 Gy). Radiosensitization was easily detected after 2 Gy in anoxic V79 cells exposed to 40 ppm ( approximately 70 nM) nitric oxide, indicating that nitric oxide is a significantly more efficient radiosensitizer than oxygen. The yield of double-strand breaks (as gamma-H2AX foci) in V79 and MCF7 cells was doubled by irradiation in 1% v/v nitric oxide/N(2), and there was a longer repair time in cells irradiated in nitric oxide than in air or anoxia; single-strand breaks ("comet" assay) also appeared to be enhanced. Potent radiosensitization by nitric oxide is consistent with near diffusion-controlled reaction of nitric oxide with purine and pyrimidine radicals observed by pulse radiolysis, with nitric oxide reacting two to three times faster than oxygen with the 5-hydroxy-uracil-6-yl radical. Stable NO/base adducts were formed with uracil radicals. Effects on the radiosensitivity of cells exposed to as low as 40 ppm v/v nitric oxide after doses of 1-2 Gy suggest that variations in radiosensitivity in individual patients after radiotherapy might include a component reflecting differing levels of nitric oxide in tumors.
Collapse
Affiliation(s)
- Peter Wardman
- University of Oxford, Gray Cancer Institute, Mount Vernon Hospital, Northwood, Middlesex, United Kingdom.
| | | | | | | | | |
Collapse
|
18
|
Wardman P. Chemical radiosensitizers for use in radiotherapy. Clin Oncol (R Coll Radiol) 2007; 19:397-417. [PMID: 17478086 DOI: 10.1016/j.clon.2007.03.010] [Citation(s) in RCA: 327] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Accepted: 03/13/2007] [Indexed: 12/21/2022]
Abstract
Radiosensitizers are intended to enhance tumour cell killing while having much less effect on normal tissues. Some drugs target different physiological characteristics of the tumour, particularly hypoxia associated with radioresistance. Oxygen is the definitive hypoxic cell radiosensitizer, the large differential radiosensitivity of oxic vs hypoxic cells being an attractive factor. The combination of nicotinamide to reduce acute hypoxia with normobaric carbogen breathing is showing clinical promise. 'Electron-affinic' chemicals that react with DNA free radicals have the potential for universal activity to combat hypoxia-associated radioresistance; a nitroimidazole, nimorazole, is clinically effective at tolerable doses. Hypoxia-specific cytotoxins, such as tirapazamine, are valuable adjuncts to radiotherapy. Nitric oxide is a potent hypoxic cell radiosensitizer; variations in endogenous levels might have prognostic significance, and routes to deliver nitric oxide specifically to tumours are being developed. In principle, many drugs can be delivered selectively to hypoxic tumours using either reductase enzymes or radiation-produced free radicals to activate drug release from electron-affinic prodrugs. A redox-active agent based on a gadolinium chelate is being evaluated clinically. Pyrimidines substituted with bromine or iodine are incorporated into DNA and enhance free radical damage; fluoropyrimidines act by different mechanisms. A wide variety of drugs that influence the nature or repair of DNA damage are being evaluated in conjunction with radiation; it is often difficult to define the mechanisms underlying chemoradiation regimens. Drugs being evaluated include topoisomerase inhibitors (e.g. camptothecin, topotecan), and the hypoxia-activated anthraquinone AQ4N; alkylating agents include temozolomide. Drugs involved in DNA repair pathways being investigated include the potent poly(ADP ribose)polymerase inhibitor, AG14,361. Proteins involved in cell signalling, such as the Ras family, are attractive targets linked to radioresistance, as are epidermal growth factor receptors and linked kinases (drugs including vandetanib [ZD6,474], cetuximab and gefitinib), and cyclooxygenase-2 (celecoxib). The suppression of radioprotective thiols seems to offer more potential with alkylating agents than with radiotherapy, although it remains a strategy worthy of exploration.
Collapse
Affiliation(s)
- P Wardman
- University of Oxford, Gray Cancer Institute, PO Box 100, Mount Vernon Hospital, Northwood HA6 2JR, UK.
| |
Collapse
|
19
|
Dewhirst MW, Navia IC, Brizel DM, Willett C, Secomb TW. Multiple Etiologies of Tumor Hypoxia Require Multifaceted Solutions: Fig. 1. Clin Cancer Res 2007; 13:375-7. [PMID: 17255256 DOI: 10.1158/1078-0432.ccr-06-2629] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Mark W Dewhirst
- Department of Radiation Oncology and Biomedical Engineering, Duke University, Durham, NC 27710, USA.
| | | | | | | | | |
Collapse
|
20
|
Uğar-Cankal D, Ozmeric N. A multifaceted molecule, nitric oxide in oral and periodontal diseases. Clin Chim Acta 2006; 366:90-100. [PMID: 16387291 DOI: 10.1016/j.cca.2005.10.018] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2005] [Revised: 10/19/2005] [Accepted: 10/20/2005] [Indexed: 11/28/2022]
Abstract
Nitric oxide (NO) is a molecule with multiple effects on different tissues. NO takes important roles in vasodilatation, bacterial challenge and cytokine stimulation, regulation of mineralized tissue function, neurotransmission, and platelet aggregation, etc. However, under pathological conditions, NO has damaging effects. NO is synthesized by NO synthases (NOS) and inducible isoform of NOS (iNOS) is closely related to the pathophysiological characteristics of inflammatory diseases such as periodontal diseases. The expression of iNOS has been investigated in salivary gland-related diseases, temporomandibular joint disorders and oral cancer as well. The beneficial and damaging effects of NO in diseases related with periodontal, dental and maxillofacial area are discussed in this review. The biological pathways involved with NO and NO inhibitors may be good drug targets to have a role in the future management of patients with diseases in orofacial region.
Collapse
Affiliation(s)
- Dilek Uğar-Cankal
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Gazi University, Biskek caddesi 84.sokak 06510 Emek, Ankara, Turkey.
| | | |
Collapse
|
21
|
Sonveaux P, Kaz AM, Snyder SA, Richardson RA, Cárdenas-Navia LI, Braun RD, Pawloski JR, Tozer GM, Bonaventura J, McMahon TJ, Stamler JS, Dewhirst MW. Oxygen regulation of tumor perfusion by S-nitrosohemoglobin reveals a pressor activity of nitric oxide. Circ Res 2005; 96:1119-26. [PMID: 15879309 DOI: 10.1161/01.res.0000168740.04986.a7] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In erythrocytes, S-nitrosohemoglobin (SNO-Hb) arises from S-nitrosylation of oxygenated hemoglobin (Hb). It has been shown that SNO-Hb behaves as a nitric oxide (NO) donor at low oxygen tensions. This property, in combination with oxygen transport capacity, suggests that SNO-Hb may have unique potential to reoxygenate hypoxic tissues. The present study was designed to test the idea that the allosteric properties of SNO-Hb could be manipulated to enhance oxygen delivery in a hypoxic tumor. Using Laser Doppler flowmetry, we showed that SNO-Hb infusion to animals breathing 21% O2 reduced tumor perfusion without affecting blood pressure and heart rate. Raising the pO2 (100% O2) slowed the release of NO bioactivity from SNO-Hb (ie, prolonged the plasma half-life of the SNO in Hb), preserved tumor perfusion, and raised the blood pressure. In contrast, native Hb reduced both tumor perfusion and heart rate independently of the oxygen concentration of the inhaled gas, and did not elicit hypertensive effects. Window chamber (to image tumor arteriolar reactivity in vivo) and hemodynamic measurements indicated that the preservation of tissue perfusion by micromolar concentrations of SNO-Hb is a composite effect created by reduced peripheral vascular resistance and direct inhibition of the baroreceptor reflex, leading to increased blood pressure. Overall, these results indicate that the properties of SNO-Hb are attributable to allosteric control of NO release by oxygen in central as well as peripheral issues.
Collapse
Affiliation(s)
- Pierre Sonveaux
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Shan S, Robson ND, Cao Y, Qiao T, Li CY, Kontos CD, Garcia-Blanco M, Dewhirst MW. Responses of vascular endothelial cells to angiogenic signaling are important for tumor cell survival. FASEB J 2003; 18:326-8. [PMID: 14688196 DOI: 10.1096/fj.03-0765fje] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Neoplastic cells overexpress several angiogenic cytokines, which stimulate neovascularization. Whether the responses of the host endothelial cells to these signaling molecules affect tumor cells during early tumorigenesis has not been investigated. We investigated pre-angiogenic tumor cell survival and angiogenesis initiation by two murine tumor lines (4T1 mammary carcinoma and B16 melanoma), which constitutively expressed GFP, in dorsal skin-fold window chambers of mice treated with extracellular domain of Tie-2 (ExTek) or bFGF. ExTek reduced tumor cell survival, retarded tumor growth, and inhibited angiogenesis onset compared with controls. bFGF increased tumor cell survival and promoted earlier angiogenesis and tumor growth. Neither bFGF nor ExTek affected cell proliferation in vitro. RT-PCR showed mRNA expression of bFGF receptor 2 (FGFR2) IIIb, which does not bind bFGF efficiently, by 4T1 cells and B16 cells express FGFR1 but not FGFR2. B16 cells expressed angiopoietin (Ang) 2, but neither cell line expresses Ang1. Both tumor lines express VEGF. These findings suggested that effects of bFGF and ExTek on tumor cell survival and angiogenesis were not due to direct action but were instead a result of paracrine factors secreted by endothelial cells. These subsequent signals from endothelial cells promote early survival and proliferation of disseminated tumor cells before onset of angiogenesis.
Collapse
MESH Headings
- Angiogenesis Inducing Agents/chemistry
- Angiogenesis Inducing Agents/pharmacology
- Animals
- Breast Neoplasms/pathology
- Cell Division/drug effects
- Cell Line, Tumor
- Cell Survival/drug effects
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/pathology
- Fibroblast Growth Factor 2/pharmacology
- Melanoma, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Models, Biological
- Neovascularization, Pathologic
- Paracrine Communication/drug effects
- Peptide Fragments/chemistry
- Peptide Fragments/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor Protein-Tyrosine Kinases/genetics
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptor, Fibroblast Growth Factor, Type 1
- Receptor, Macrophage Colony-Stimulating Factor/chemistry
- Receptor, TIE-2/chemistry
- Receptors, Fibroblast Growth Factor/genetics
- Receptors, Fibroblast Growth Factor/metabolism
- Signal Transduction/drug effects
- Solubility
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Siqing Shan
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710-3455, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Jaszewski AR, Fann YC, Chen YR, Sato K, Corbett J, Mason RP. EPR spectroscopy studies on the structural transition of nitrosyl hemoglobin in the arterial-venous cycle of DEANO-treated rats as it relates to the proposed nitrosyl hemoglobin/nitrosothiol hemoglobin exchange. Free Radic Biol Med 2003; 35:444-51. [PMID: 12899946 DOI: 10.1016/s0891-5849(03)00324-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In vivo studies show a dynamic cycle in which alpha-nitrosylated hemoglobin is mainly in the relaxed state in arterial blood of rats treated with 2-(N,N-diethylamino)-diazenolate-2-oxide, but converts mainly to the tense state during the arterial-venous transit. A detailed analysis shows that different electron paramagnetic resonance spectra recorded for alpha-nitrosyl hemoglobin in arterial and venous blood at 77 K originate only from a different ratio between 5- and 6-coordinate heme without any change in the concentration of nitrosyl hemoglobin. In venous blood, the five- and six-coordination equilibrium of the alpha-nitrosyl heme is shifted in favor of the 5-coordinate state (58% venous vs. 20% arterial). These results are not consistent with the recently proposed exchange of nitrosyl heme with the beta-93 nitrosothiol group of hemoglobin during the arterial-venous cycle.
Collapse
Affiliation(s)
- Adrian R Jaszewski
- Laboratory of Pharmacology and Chemistry, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Baudelet C, Gallez B. Cluster analysis of BOLD fMRI time series in tumors to study the heterogeneity of hemodynamic response to treatment. Magn Reson Med 2003; 49:985-90. [PMID: 12768574 DOI: 10.1002/mrm.10468] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BOLD-contrast functional MRI (fMRI) has been used to assess the evolution of tumor oxygenation and blood flow after treatment. The aim of this study was to evaluate K-means-based cluster analysis as a exploratory, data-driven method. The advantage of this approach is that it can be used to extract information without the need for prior knowledge concerning the hemodynamic response function. Two data sets were acquired to illustrate different types of BOLD fMRI response inside tumors: the first set following a respiratory challenge with carbogen, and the second after pharmacological modulation of tumor blood flow using flunarizine. To improve the efficiency of the clustering, a power density spectrum analysis was first used to isolate voxels for which signal changes did not originate from noise or linear drift. The technique presented here can be used to assess hemodynamic response to treatment, and especially to display areas of the tumor with heterogeneous responses.
Collapse
Affiliation(s)
- Christine Baudelet
- Medicinal Chemistry and Radiopharmacy Unit, Université Catholique de Louvain, Brussels, Belgium
| | | |
Collapse
|
25
|
Abstract
Although previously regarded as a toxic pollutant gas, nitric oxide (NO) is a short-lived molecule that plays a key role in many physiological and pathological processes. It is produced in vivo from the amino acid L-arginine by a complex family of enzymes termed nitric oxide synthase (NOS). Since its discovery as a biological messenger in 1987, NO has been implicated in many disease processes, ranging from septic shock to cancer. It is a highly reactive free radical and causes concentration-dependent conformational changes in proteins, enzymes and DNA, predominantly by its reaction with transition metals and thiol residues. Although high concentrations of NO are cytotoxic, the levels produced in many human cancers possibly facilitate tumour growth and dissemination. The interest in this molecule by scientists and clinicians involved with the oral cavity and head and neck regions is fairly recent, and only a tiny minority of 50,000 papers currently cited on NO relate to diseases in this anatomical area. This review gives an overview of NO, outlining its basic chemistry, formation by NOS and its possible roles in the oral diseases studied to date. The implications for possible therapeutic manipulation of NO are also discussed.
Collapse
Affiliation(s)
- P A Brennan
- Department of Maxillofacial and Head and Neck Surgery, Queen Alexandra Hospital, Portsmouth PO6 3LY, UK.
| | | | | |
Collapse
|
26
|
Braun RD, Lanzen JL, Turnage JA, Rosner G, Dewhirst MW. Effects of the interaction between carbogen and nicotinamide on R3230 Ac tumor blood flow in Fischer 344 rats. Radiat Res 2001; 155:724-33. [PMID: 11302770 DOI: 10.1667/0033-7587(2001)155[0724:eotibc]2.0.co;2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Braun, R. D., Lanzen, J. L., Turnage, J. A., Rosner, G. and Dewhirst, M. W. Effects of the Interaction between Carbogen and Nicotinamide on R3230 Ac Tumor Blood Flow in Fischer 344 Rats. Radiat. Res. 155, 724-733 (2001). The purpose of this study was to determine whether there are interactions between carbogen breathing and various doses of nicotinamide at the level of the tumor arteriole that might contribute to the improvement in tumor blood flow and pO(2) that is often seen with this combination treatment. R3230 adenocarcinomas were implanted and grown to 4-5 mm in dorsal skin flap window chambers in F344 rats. Saline or 65, 200 or 500 mg/kg nicotinamide was injected i.p. while the rat breathed air through a face mask. After 20 min, either the breathing gas was switched to carbogen for 60 min or the animal remained on air. Measured end points included diameter of tumor arterioles, tumor perfusion, mean arterial blood pressure, and heart rate. None of the measured parameters were affected by injection of saline or nicotinamide, except at the highest nicotinamide dose (500 mg/kg). Mean arterial blood pressure showed a median decrease of 25% when 500 mg/kg nicotinamide was given. Diameter of tumor arterioles decreased significantly from 5-15 min after 500 mg/kg nicotinamide was given but was back to baseline by 20 min. Blood flow decreased significantly 5-20 min after administration of 500 mg/kg nicotinamide compared to the baseline prior to injection. Carbogen breathing resulted in a small increase in mean arterial blood pressure in all groups. There was a transient decrease in the diameter of tumor arterioles and blood flow during the first 5 min of carbogen breathing that was statistically significant in several groups. In the group injected with 500 mg/kg nicotinamide, the diameter of tumor arterioles increased by about 10% during the first 25 min of carbogen breathing, and blood flow increased by a median of 75% over the level prior to carbogen breathing up to 40 min after carbogen breathing. The increase in flow in this group was most likely caused by the concomitant arteriolar vasodilation. Thus there was direct evidence for an interaction between carbogen breathing and nicotinamide, but only at the dose of 500 mg/kg nicotinamide. Since this dose yields plasma levels of nicotinamide that are higher than can be tolerated clinically, it is uncertain whether these changes in arteriolar diameter and blood flow would occur in human tumors.
Collapse
Affiliation(s)
- R D Braun
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | |
Collapse
|
27
|
Meyer RE, Braun RD, Rosner GL, Dewhirst MW. Local 42 degrees C hyperthermia improves vascular conductance of the R3230Ac rat mammary adenocarcinoma during sodium nitroprusside infusion. Radiat Res 2000; 154:196-201. [PMID: 10931692 DOI: 10.1667/0033-7587(2000)154[0196:lchivc]2.0.co;2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The effect of sodium nitroprusside-induced hypotension on the perfusion of the R3230 adenocarcinoma during local 42 degrees C hyperthermia was studied using a combination of intravital microscopy and laser Doppler flowmetry. Fischer 344 rats were implanted with dorsal skin flap window chambers containing the R3230Ac tumor and allocated to three treatment groups (34 degrees C with nitroprusside, 42 degrees C with nitroprusside, and 42 degrees C with 0.9% saline). After baseline observation at 34 degrees C, tumors were locally heated to 42 degrees C using a water bath and either 0.9% saline or nitroprusside sufficient to reduce blood pressure 20% below pretreatment baseline was infused. Nitroprusside at 34 degrees C decreased tumor vascular conductance 40% with no effect on the diameter of arterioles entering the tumor. The diameter of arterioles entering 42 degrees C heated tumors increased 35% independent of blood pressure change. Saline at 42 degrees C had no effect on tumor vascular conductance; however, nitroprusside at 42 degrees C increased tumor vascular conductance 55%. Local 42 degrees C tumor heating, combined with a moderate reduction in blood pressure with nitroprusside, overrides the vascular steal effect associated with reduced perfusion pressure alone and results in improved tumor perfusion. Observations of the effect of vasodilator substances on normothermic tumor perfusion cannot be extrapolated to situations where moderate hyperthermia is used.
Collapse
Affiliation(s)
- R E Meyer
- Department of Anatomy, Physiological Sciences, and Radiology, North Carolina State University, Raleigh, North Carolina 27606, USA
| | | | | | | |
Collapse
|
28
|
Brennan PA. The actions and interactions of nitric oxide in solid tumours. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2000; 26:434-7. [PMID: 11016461 DOI: 10.1053/ejso.1999.0917] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The small molecule nitric oxide (NO) has undergone an image change since its identification as a biological messenger in 1987. It is a free radical, with a diverse range of actions in both physiological and pathological processes. Whilst over 30000 research papers have been written to date on NO, its role in tumour biology remains incompletely understood and research in this field is still in its infancy. NO would appear to have both tumour promoting and inhibiting effects which are presumed to be dependent on its local concentration within the tumour. Recently the relationships of NO to the tumour suppressor gene p53 have been experimentally elucidated, demonstrating how mutations of p53 may adversely affect the host by enhancing NO production. This review summarizes the brief history of this molecule, outlines its roles in the common solid tumours and suggests areas for future research.
Collapse
Affiliation(s)
- P A Brennan
- Maxillofacial Department, Queen Alexandra Hospital, Portsmouth, UK
| |
Collapse
|
29
|
Thews O, Kelleher DK, Vaupel P. Disparate responses of tumour vessels to angiotensin II: tumour volume-dependent effects on perfusion and oxygenation. Br J Cancer 2000; 83:225-31. [PMID: 10901375 PMCID: PMC2363484 DOI: 10.1054/bjoc.2000.1229] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Perfusion and oxygenation of experimental tumours were studied during angiotensin II (AT II) administration whereby the rate of the continuous AT II infusion was chosen to increase the mean arterial blood pressure (MABP) by 50-70 mmHg. In subcutaneous DS-sarcomas the red blood cell (RBC) flux was assessed using the laser Doppler technique and the mean tumour oxygen partial pressure (pO2) was measured polarographically using O2-sensitive catheter and needle electrodes. Changes in RBC flux with increasing MABP depended mainly on tumour size. In small tumours, RBC flux decreased with rising MABP whereas in larger tumours RBC flux increased parallel to the MABP. As a result of these volume-dependent effects on tumour blood flow, the impact of AT II on tumour pO2 was also mainly tumour volume-related. In small tumours oxygenation decreased with increasing MABP during AT II infusion, whereas in large tumours a positive relationship between blood pressure and O2 status was found. This disparate behaviour might be the result of the co-existence of two functionally distinct populations of tumour vessels. In small tumours, perfusion decreases presumably due to vasoconstriction of pre-existing host vessels feeding the tumour. In larger malignancies, newly formed tumour vessels predominate and seem not to have this vasoresponsive capability (lack of smooth muscle cells and/or AT receptors), resulting in an improvement of perfusion which is not tumour-related per se, but is due to the increased perfusion pressure.
Collapse
Affiliation(s)
- O Thews
- Institute of Physiology and Pathophysiology, University of Mainz, Germany
| | | | | |
Collapse
|
30
|
Brennan PA, Downie IP, Langdon JD, Zaki GA. Emerging role of nitric oxide in cancer. Br J Oral Maxillofac Surg 1999; 37:370-3. [PMID: 10577750 DOI: 10.1054/bjom.1999.0201] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The small molecule nitric oxide has generated an exponential amount of research since its identification as a biological messenger in 1987. It is a free radical, the actions of which are diverse, and many are still poorly understood. An area of great interest is the role of nitric oxide in the growth and metastasis of solid tumours, where it seems to have a complex action including both inhibitory and tumour-promoting activity. Over 28,000 research papers have been written on nitric oxide, with only a minority concentrating on the head and neck. In this review, we give a brief history of this fascinating molecule, concentrating on its possible interest to oral and maxillofacial surgeons.
Collapse
|
31
|
Braun RD, Lanzen JL, Dewhirst MW. Fourier analysis of fluctuations of oxygen tension and blood flow in R3230Ac tumors and muscle in rats. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:H551-68. [PMID: 10444480 DOI: 10.1152/ajpheart.1999.277.2.h551] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor hypoxia is a major barrier to tumor radiation therapy. Typically tumor hypoxia occurs in two forms: chronic and acute. Although the existence of acute hypoxia has long been acknowledged, its temporal characteristics have never been directly measured and documented. In this study tumor PO(2), blood flow (BF), and arterial blood pressure (BP) were measured simultaneously in nine Fischer 344 rats bearing R3230Ac rat mammary adenocarcinomas in the subcutis of the left hindleg. We measured PO(2) at a single location for 36-125 min using recessed-tip oxygen microelectrodes. Simultaneously, we measured tumor BF at two sites within the tumor using laser-Doppler flowmetry (LDF). Similar recordings were made in the quadriceps muscle of seven non-tumor-bearing rats. The PO(2), tumor BF, and BP records were subjected to Fourier analysis. PO(2) and BF showed low-frequency fluctuations (<2 cycles/min) in both tumor and muscle, but the magnitude of the changes in tumor was greater. Tumor BF showed more activity at low frequencies than muscle BF, and the magnitude tended to be greater. No strong correlations were found between PO(2) and BF power spectra for either tumor or muscle or between the frequency patterns of BP and tumor PO(2) spectra. These results quantitatively demonstrate, for the first time, that BF and PO(2) fluctuate at very low frequencies in tumors. In addition to having biological significance for tumor therapy, these fluctuations may have the potential to alter tumor cell behavior via induction of hypoxia reoxygenation injury and/or altered gene expression.
Collapse
Affiliation(s)
- R D Braun
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | |
Collapse
|
32
|
Dewhirst MW, Ong ET, Braun RD, Smith B, Klitzman B, Evans SM, Wilson D. Quantification of longitudinal tissue pO2 gradients in window chamber tumours: impact on tumour hypoxia. Br J Cancer 1999; 79:1717-22. [PMID: 10206282 PMCID: PMC2362789 DOI: 10.1038/sj.bjc.6690273] [Citation(s) in RCA: 140] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
We previously reported that the arteriolar input in window chamber tumours is limited in number and is constrained to enter the tumour from one surface, and that the pO2 of tumour arterioles is lower than in comparable arterioles of normal tissues. On average, the vascular pO2 in vessels of the upper surface of these tumours is lower than the pO2 of vessels on the fascial side, suggesting that there may be steep vascular longitudinal gradients (defined as the decline in vascular pO2 along the afferent path of blood flow) that contribute to vascular hypoxia on the upper surface of the tumours. However, we have not previously measured tissue pO2 on both surfaces of these chambers in the same tumour. In this report, we investigated the hypothesis that the anatomical constraint of arteriolar supply from one side of the tumour results in longitudinal gradients in pO2 sufficient in magnitude to create vascular hypoxia in tumours grown in dorsal flap window chambers. Fischer-344 rats had dorsal flap window chambers implanted in the skin fold with simultaneous transplantation of the R3230AC tumour. Tumours were studied at 9-11 days after transplantation, at a diameter of 3-4 mm; the tissue thickness was 200 microm. For magnetic resonance microscopic imaging, gadolinium DTPA bovine serum albumin (BSA-DTPA-Gd) complex was injected i.v., followed by fixation in 10% formalin and removal from the animal. The sample was imaged at 9.4 T, yielding voxel sizes of 40 microm. Intravital microscopy was used to visualize the position and number of arterioles entering window chamber tumour preparations. Phosphorescence life time imaging (PLI) was used to measure vascular pO2. Blue and green light excitations of the upper and lower surfaces of window chambers were made (penetration depth of light approximately 50 vs >200 microm respectively). Arteriolar input into window chamber tumours was limited to 1 or 2 vessels, and appeared to be constrained to the fascial surface upon which the tumour grows. PLI of the tumour surface indicated greater hypoxia with blue compared with green light excitation (P < 0.03 for 10th and 25th percentiles and for per cent pixels < 10 mmHg). In contrast, illumination of the fascial surface with blue light indicated less hypoxia compared with illumination of the tumour surface (P < 0.05 for 10th and 25th percentiles and for per cent pixels < 10 mmHg). There was no significant difference in pO2 distributions for blue and green light excitation from the fascial surface nor for green light excitation when viewed from either surface. The PLI data demonstrates that the upper surface of the tumour is more hypoxic because blue light excitation yields lower pO2 values than green light excitation. This is further verified in the subset of chambers in which blue light excitation of the fascial surface showed higher pO2 distributions compared with the tumour surface. These results suggest that there are steep longitudinal gradients in vascular pO2 in this tumour model that are created by the limited number and orientation of the arterioles. This contributes to tumour hypoxia. Arteriolar supply is often limited in other tumours as well, suggesting that this may represent another cause for tumour hypoxia. This report is the first direct demonstration that longitudinal oxygen gradients actually lead to hypoxia in tumours.
Collapse
Affiliation(s)
- M W Dewhirst
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Hahn SM, DeLuca AM, Coffin D, Krishna CM, Mitchell JB. In vivo radioprotection and effects on blood pressure of the stable free radical nitroxides. Int J Radiat Oncol Biol Phys 1998; 42:839-42. [PMID: 9845107 DOI: 10.1016/s0360-3016(98)00317-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE The purpose of this study was to screen several water soluble nitroxides for in vivo radioprotection, to evaluate their pharmacology, and to measure the effect of nitroxides on systemic blood pressure as a means of exploring the mechanism of in vivo radioprotection. METHODS AND MATERIALS A number of water soluble nitroxides were screened for in vivo radioprotection in C3H mice at a single radiation dose. Selected nitroxides were administered by the intraperitoneal route 10 minutes prior to a whole body radiation dose of 9 Gy. Electron paramagnetic resonance spectroscopy (EPR) was used to measure whole blood levels of nitroxides. The nitroxides were evaluated for effects on systemic blood pressure in C3H mice. RESULTS All of the nitroxides studied demonstrated radioprotection compared to saline-treated controls. The 6-membered piperidine ring nitroxides including Tempol were reduced to the inactive hydroxylamine rapidly over 10-20 minutes. The 5-membered ring nitroxides were reduced more slowly over time. The 5-membered ring 3-carbamoyl-PROXYL did not produce a substantial decrease in systemic blood pressure after intraperitoneal administration compared to the other nitroxides studied. 3-carbamoyl-PROXYL was further evaluated over a range of whole body radiation doses and was found to provide radioprotection. CONCLUSION All of the nitroxides studied provided radioprotection. In vivo radioprotection for all of the compounds except 3-carbamoyl-PROXYL may be at least partially explained by the induction of hypotension and bone marrow hypoxia. 3-carbamoyl-PROXYL provided in vivo radioprotection similar in magnitude to Tempol and had little effect on blood pressure compared to the other nitroxides. Other mechanisms for radioprotection, including scavenging of free radicals are likely. 3-carbamoyl-PROXYL should be evaluated further as a systemic radioprotector.
Collapse
Affiliation(s)
- S M Hahn
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, USA
| | | | | | | | | |
Collapse
|
34
|
Chaplin DJ, Hill SA, Bell KM, Tozer GM. Modification of tumor blood flow: current status and future directions. Semin Radiat Oncol 1998; 8:151-63. [PMID: 9634492 DOI: 10.1016/s1053-4296(98)80041-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Suboptimal drug distribution and hypoxia, which can contribute to treatment failure, are a direct consequence of the spatial and temporal heterogeneity in perfusion that occurs in solid tumors. Therefore, improvements in tumor blood flow have wide-ranging therapeutic importance. Paradoxically, controlled decreases in tumor blood flow can also be exploited and, if permanent, induce extensive tumor cell death on their own. We review the current knowledge of the factors controlling tumor blood flow with emphasis on the roles of the endogeneous vasodilator nitric oxide and the endogenous vasoconstrictor endothelin-1. The potential importance and application of approaches that irreversibly damage vascular function, so-called vascular targeting, are also discussed. Emphasis is given to the drug-based approaches to vascular targeting that are now entering clinical evaluation. There is no doubt that increased understanding of the processes that determine blood flow in tumors, coupled with the availability of techniques to monitor blood flow noninvasively in the clinic, will enable strategies for selectively modifying tumor blood flow to be transferred from the laboratory to the clinical setting.
Collapse
Affiliation(s)
- D J Chaplin
- Gray Laboratory Cancer Research Trust, Mount Vernon Hospital, Northwood, Middlesex, UK
| | | | | | | |
Collapse
|