Vessel caliber--a potential MRI biomarker of tumour response in clinical trials

Kyrre E Emblem, Christian T Farrar, Elizabeth R Gerstner, Tracy T Batchelor, Ronald J H Borra, Bruce R Rosen, A Gregory Sorensen, Rakesh K Jain, Kyrre E Emblem, Christian T Farrar, Elizabeth R Gerstner, Tracy T Batchelor, Ronald J H Borra, Bruce R Rosen, A Gregory Sorensen, Rakesh K Jain

Abstract

Our understanding of the importance of blood vessels and angiogenesis in cancer has increased considerably over the past decades, and the assessment of tumour vessel calibre and structure has become increasingly important for in vivo monitoring of therapeutic response. The preferred method for in vivo imaging of most solid cancers is MRI, and the concept of vessel-calibre MRI has evolved since its initial inception in the early 1990s. Almost a quarter of a century later, unlike traditional contrast-enhanced MRI techniques, vessel-calibre MRI remains widely inaccessible to the general clinical community. The narrow availability of the technique is, in part, attributable to limited awareness and a lack of imaging standardization. Thus, the role of vessel-calibre MRI in early phase clinical trials remains to be determined. By contrast, regulatory approvals of antiangiogenic agents that are not directly cytotoxic have created an urgent need for clinical trials incorporating advanced imaging analyses, going beyond traditional assessments of tumour volume. To this end, we review the field of vessel-calibre MRI and summarize the emerging evidence supporting the use of this technique to monitor response to anticancer therapy. We also discuss the potential use of this biomarker assessment in clinical imaging trials and highlight relevant avenues for future research.

Trial registration: ClinicalTrials.gov NCT00254943 NCT00662506 NCT00756106.

Figures

Figure 1
Figure 1
Vessel calibres in solid cancers. Conceptual illustration showing key factors for vessel-calibre growth and remodelling within a tumour, including vessel dilatation after stimulation by proangiogenic factors such as VEGF, FGFs, ANG-2 and chemokines, and compression of vessels as a result of solid stress induced by tumour growth. In normal tissues, red vessels indicate oxygen-rich feeding arteries and arterioles, blue vessels are veins carrying deoxygenated blood and the intermediate regions indicate a transient capillary stage. In cancerous tissues, slow blood flow is indicated by reduced colour intensity. Abbreviations: ANG-2, angiopoietin-2; FGFs, fibroblast growth factors; VEGF, vascular endothelial growth factor.
Figure 2
Figure 2
Vessel-calibre MRI. a | Example anatomical CE T1-weighted, FLAIR, macrovessel volume-fraction and microvessel volume-fraction images from MRI assessments of a patient with glioblastoma. Note the sensitivity to high blood-volume fractions in the macrovessel image (total blood volume) compared with the microvessel image. b | Macrovessel and microvessel DSC-MRI in the same patient with glioblastoma, with contrast agent passing through the tissue. Before estimation of perfusion and vessel-calibre MRI parameters, the macrovessel (GE) and microvessel (SE) blood-volume relaxation rate images shown here are computed according to the relative decrease in MRI signal intensity over the course of a sequence of GE MRI and SE MRI images, respectively (images 30–34 in this case), thereby adjusting for baseline intensity values and image sampling time (echo time). The so-called ‘first-pass effect’ denotes the initial and transient passage of a relatively tight bolus of the contrast agent following intravenous administration of the agent and is observed as a peak in the relative relaxation rate signal (peaks for normal tissue approximately at image 32). The contrast agent first-pass effect is particularly dominant in large vessels in the macrovessel images because of high contrast agent concentrations combined with high sensitivity to the magnetic susceptibility effect induced by the agent. After the initial first-pass contrast agent passage, repeated passages of what remains of the circulating and increasingly dispersed contrast agent bolus can be appreciated as smaller and dissipating signal oscillations. c | Macrovessel and microvessel volume fractions are estimated from the area under the GE and SE first-pass curves (top left), respectively. The concept of vessel-calibre MRI stems from the relationship between increased macrovessel volume fractions for increasing vessel calibres, and microvessel volume fractions, which are selectively sensitive to small (radius <10 µm) vessel calibres (bottom left). Mean vessel density maps (Box 1) are thus derived from the quotient of macrovessel and microvessel volume fractions, whereas the vessel-calibre index also accounts for water diffusion and the absolute blood volume fraction. Abbreviations: CE, contrast-enhanced; DSC-MRI, dynamic susceptibility-contrast MRI; FLAIR, fluid-attenuated inversion recovery; GE, gradient-echo; SE, spin-echo.
Figure 3
Figure 3
Vessel-calibre MRI during antiangiogenic therapy. Selected MRI-derived images obtained over the course of a phase II trial of cediranib combined with CRT in adult patients with newly diagnosed glioblastomas. From top to bottom: contrast-enhanced T1-weighted MRI illustrating a permeable tumour vasculature; FLAIR MRI showing the tumour and vasogenic oedema; macrovessel blood-volume fractions; vessel-calibre maps; and volume renderings from 3D anatomical MRI with time-of-flight MRI angiography. Compared with pretreatment baseline images (day −1), cediranib therapy (days 1 and 8) decreased the signal enhancement in the tumour, indicative of reduced permeability of the tumour vasculature, by day 8 (yellow arrow), and oedema was subsequently reduced (yellow arrowhead; day 50). Treatment effects of antiangiogenic and adjuvant combination therapy were also observed after the end of CRT (up to day 400); however, the cancer eventually relapsed and progressed, although this outcome is not clear using standard contrast and FLAIR MRI protocols. Volume fractions and vessel calibres show reductions that are related to the responses observed by conventional contrast-enhanced and FLAIR MRI following therapy (white arrow; day 50), but the vessel-calibre response also reveals unique properties of tissue that do not match the spatial distribution of the volume-fraction map. In contrast to the major vessels identified by standard MRI angiography (bottom panels), the vessel-calibre map depicts changes in the tumour microvasculature that are well below the image resolution achievable using the conventional approach; note the increased vessel calibres in the relapsed tumour region (white arrowhead; day 400), which is not easily appreciated on conventional MRI nor on the volume-fraction map. Abbreviations: CRT, chemoradiation therapy; FLAIR, fluid-attenuated inversion recovery.
Figure 4
Figure 4
Advanced MRI assessments might inform optimal dose scheduling. On the basis of our experience with cediranib,,,, the initial responses, plateaus and reversals in tumour response that are observed using advanced MRI parameters occur at different timepoints after initiation of therapy. In this example model of tumour-vessel architecture, showing a typical vascular-normalization response after cediranib treatment and CRT in patients with newly diagnosed glioblastomas, the perfusion estimate (blood flow) peaks after approximately 1 week of therapy (day 8), whereas the maximum reduction in vessel calibres occurs 1 week later (day 15; note the reduced diameter of the example vessels). These effects are preceded by an early, substantial, and prolonged normalization of the abnormal arterio-venous ratio evident in the model of the baseline data (day 1; arteries/arteriole shown in red, with veins in blue), estimated using vessel architectural imaging. Consistent with the vessel-calibre response, the apparent change in abnormal ΔSO2 levels reaches its minimum at two weeks (day 15; purple colour). After the end of CRT, all of these parameters reverse, thereby indicating an end to the vascular-normalization window. Furthermore, in patients who were deemed unresponsive to therapy, limited evidence of such responses was seen. Collectively, advanced MRI protocols might facilitate the design of improved early-phase trials by informing the optimization of the dosing regimen for antiangiogenic agents and could also potentially support a personalized-medicine approach by enabling therapy to be tailored to individual patients based on parameters indicative of biological and, in particular, vascular responses. In addition, early identification of patients who are unlikely to respond to therapy could help decision making regarding whether therapy should be discontinued, particularly in patients who experience adverse events. Abbreviations: ΔSO2, relative oxygen saturation levels; CRT, chemoradiation therapy.

References

    1. Jain RK. Molecular regulation of vessel maturation. Nat. Med. 2003;9:685–693.
    1. Baluk P, Hashizume H, McDonald DM. Cellular abnormalities of blood vessels as targets in cancer. Curr. Opin. Genet. Dev. 2005;15:102–111.
    1. Fukumura D, Duda DG, Munn LL, Jain RK. Tumor microvasculature and microenvironment: novel insights through intravital imaging in pre-clinical models. Microcirculation. 2010;17:206–225.
    1. Stylianopoulos T, et al. Causes, consequences, and remedies for growth-induced solid stress in murine and human tumors. Proc. Natl Acad. Sci. USA. 2012;109:15101–15108.
    1. Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature. 2011;473:298–307.
    1. Vakoc BJ, et al. Three-dimensional microscopy of the tumor microenvironment in vivo using optical frequency domain imaging. Nat. Med. 2009;15:1219–1223.
    1. Nagy JA, Chang SH, Dvorak AM, Dvorak HF. Why are tumour blood vessels abnormal and why is it important to know? Br. J. Cancer. 2009;100:865–869.
    1. Jain RK. Normalizing tumor microenvironment to treat cancer: bench to bedside to biomarkers. J. Clin. Oncol. 2013;31:2205–2218.
    1. O’Connor JP, Jackson A, Parker GJ, Roberts C, Jayson GC. Dynamic contrast-enhanced MRI in clinical trials of antivascular therapies. Nat. Rev. Clin. Oncol. 2012;9:167–177.
    1. Jain RK, et al. Biomarkers of response and resistance to antiangiogenic therapy. Nat. Rev. Clin. Oncol. 2009;6:327–338.
    1. Wen PY, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J. Clin. Oncol. 2010;28:1963–1972.
    1. Lin NU, et al. Challenges relating to solid tumour brain metastases in clinical trials, part 1: patient population, response, and progression. A report from the RANO group. Lancet Oncol. 2013;14:e396–e406.
    1. Leach MO, et al. The assessment of antiangiogenic and antivascular therapies in early-stage clinical trials using magnetic resonance imaging: issues and recommendations. Br. J. Cancer. 2005;92:1599–1610.
    1. Michaelis LC, Ratain MJ. Measuring response in a post-RECIST world: from black and white to shades of grey. Nat. Rev. Cancer. 2006;6:409–414.
    1. Morgan B. Opportunities and pitfalls of cancer imaging in clinical trials. Nat. Rev. Clin. Oncol. 2011;8:517–527.
    1. Folkman J. Angiogenesis: an organizing principle for drug discovery? Nat. Rev. Drug Discov. 2007;6:273–286.
    1. Ellis LM, Hicklin DJ. VEGF-targeted therapy: mechanisms of anti-tumour activity. Nat. Rev. Cancer. 2008;8:579–591.
    1. Bergers G, Hanahan D. Modes of resistance to anti-angiogenic therapy. Nat. Rev. Cancer. 2008;8:592–603.
    1. Ferrara N, Kerbel RS. Angiogenesis as a therapeutic target. Nature. 2005;438:967–974.
    1. Padera TP, et al. Pathology: cancer cells compress intratumour vessels. Nature. 2004;427:695.
    1. Jain RK. An indirect way to tame cancer. Sci. Am. 2014;310:46–53.
    1. Wilson WR, Hay MP. Targeting hypoxia in cancer therapy. Nat. Rev. Cancer. 2011;11:393–410.
    1. Dewhirst MW, Cao Y, Moeller B. Cycling hypoxia and free radicals regulate angiogenesis and radiotherapy response. Nat. Rev. Cancer. 2008;8:425–437.
    1. Yeo SG, Kim JS, Cho MJ, Kim KH, Kim JS. Interstitial fluid pressure as a prognostic factor in cervical cancer following radiation therapy. Clin. Cancer Res. 2009;15:6201–6207.
    1. Folkman J. Tumor angiogenesis: therapeutic implications. N. Engl. J. Med. 1971;285:1182–1186.
    1. Jain RK. Normalizing tumor vasculature with anti-angiogenic therapy: a new paradigm for combination therapy. Nat. Med. 2001;7:987–989.
    1. Winkler F, et al. Kinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiation: role of oxygenation, angiopoietin-1, and matrix metalloproteinases. Cancer Cell. 2004;6:553–563.
    1. Huang Y, Stylianopoulos T, Duda DG, Fukumura D, Jain RK. Benefits of vascular normalization are dose and time dependent—letter. Cancer Res. 2013;73:7144–7146.
    1. Jain RK, Duda DG, Clark JW, Loeffler JS. Lessons from phase III clinical trials on anti-VEGF therapy for cancer. Nat. Clin. Pract. Oncol. 2006;3:24–40.
    1. Van der Veldt AA, et al. Rapid decrease in delivery of chemotherapy to tumors after anti-VEGF therapy: implications for scheduling of anti-angiogenic drugs. Cancer Cell. 2012;21:82–91.
    1. Chauhan VP, et al. Normalization of tumour blood vessels improves the delivery of nanomedicines in a size-dependent manner. Nat. Nanotechnol. 2012;7:383–388.
    1. Arjaans M, et al. Bevacizumab-induced normalization of blood vessels in tumors hampers antibody uptake. Cancer Res. 2013;73:3347–3355.
    1. Gerlowski LE, Jain RK. Microvascular permeability of normal and neoplastic tissues. Microvasc. Res. 1986;31:288–305.
    1. Matsumura Y, Maeda H. A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res. 1986;46:6387–6392.
    1. Prabhakar U, et al. Challenges and key considerations of the enhanced permeability and retention effect for nanomedicine drug delivery in oncology. Cancer Res. 2013;73:2412–2417.
    1. Peer D, et al. Nanocarriers as an emerging platform for cancer therapy. Nat. Nanotechnol. 2007;2:751–760.
    1. Chauhan VP, Jain RK. Strategies for advancing cancer nanomedicine. Nat. Mater. 2013;12:958–962.
    1. Chauhan VP, et al. Angiotensin inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels. Nat. Commun. 2013;4:2516.
    1. Afaq A, Andreou A, Koh DM. Diffusion-weighted magnetic resonance imaging for tumour response assessment: why, when and how? Cancer Imaging. 2010;10(A):S179–S188.
    1. Patterson DM, Padhani AR, Collins DJ. Technology insight: water diffusion MRI—a potential new biomarker of response to cancer therapy. Nat. Clin. Pract. Oncol. 2008;5:220–233.
    1. van Osch MJ, Vonken EJ, Bakker CJ, Viergever MA. Correcting partial volume artifacts of the arterial input function in quantitative cerebral perfusion MRI. Magn. Reson. Med. 2001;45:477–485.
    1. Therasse P, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J. Natl Cancer Inst. 2000;92:205–216.
    1. Macdonald DR, Cascino TL, Schold SC, Jr, Cairncross JG. Response criteria for phase II studies of supratentorial malignant glioma. J. Clin. Oncol. 1990;8:1277–1280.
    1. Gehan EA, Tefft MC. Will there be resistance to the RECIST (Response Evaluation Criteria in Solid Tumors)? J. Natl Cancer Inst. 2000;92:179–181.
    1. Oxnard GR, et al. When progressive disease does not mean treatment failure: reconsidering the criteria for progression. J. Natl Cancer Inst. 2012;104:1534–1541.
    1. Therasse P, Eisenhauer EA, Verweij J. RECIST revisited: a review of validation studies on tumour assessment. Eur. J. Cancer. 2006;42:1031–1039.
    1. Twombly R. Criticism of tumor response criteria raises trial design questions. J. Natl Cancer Inst. 2006;98:232–234.
    1. Nishino M, et al. Personalized tumor response assessment in the era of molecular medicine: cancer-specific and therapy-specific response criteria to complement pitfalls of RECIST. AJR Am. J. Roentgenol. 2012;198:737–745.
    1. Llovet JM, et al. Design and endpoints of clinical trials in hepatocellular carcinoma. J. Natl Cancer Inst. 2008;100:698–711.
    1. Rustin GJ, et al. Re: New guidelines to evaluate the response to treatment in solid tumors (ovarian cancer) J. Natl Cancer Inst. 2004;96:487–488.
    1. Hoos A, et al. Improved endpoints for cancer immunotherapy trials. J. Natl Cancer Inst. 2010;102:1388–1397.
    1. Scher HI, Morris MJ, Kelly WK, Schwartz LH, Heller G. Prostate cancer clinical trial end points: “RECIST” ing a step backwards. Clin. Cancer Res. 2005;11:5223–5232.
    1. Faivre SJ, Bouattour M, Dreyer C, Raymond E. Sunitinib in hepatocellular carcinoma: redefining appropriate dosing, schedule, and activity end points. J. Clin. Oncol. 2009;27:e248–e250.
    1. Sorensen AG, et al. Comparison of diameter and perimeter methods for tumor volume calculation. J. Clin. Oncol. 2001;19:551–557.
    1. Goldberg SN, et al. Image-guided tumor ablation: standardization of terminology and reporting criteria. Radiology. 2005;235:728–739.
    1. Reuter M, et al. Impact of MRI head placement on glioma response assessment. J. Neurooncol. 2014;118:123–129.
    1. Brandsma D, Stalpers L, Taal W, Sminia P, van den Bent MJ. Clinical features, mechanisms, and management of pseudoprogression in malignant gliomas. Lancet Oncol. 2008;9:453–461.
    1. Wong CS, van der Kogel AJ. Mechanisms of radiation injury to the central nervous system: implications for neuroprotection. Mol. Interv. 2004;4:273–284.
    1. Waldman AD, et al. Quantitative imaging biomarkers in neuro-oncology. Nat. Rev. Clin. Oncol. 2009;6:445–454.
    1. Gore JC, Manning HC, Quarles CC, Waddell KW, Yankeelov TE. Magnetic resonance in the era of molecular imaging of cancer. Magn. Reson. Imaging. 2011;29:587–600.
    1. van den Bent MJ, Vogelbaum MA, Wen PY, Macdonald DR, Chang SM. End point assessment in gliomas: novel treatments limit usefulness of classical Macdonald’s Criteria. J. Clin. Oncol. 2009;27:2905–2908.
    1. Burrell JS, et al. MRI measurements of vessel calibre in tumour xenografts: comparison with vascular corrosion casting. Microvasc. Res. 2012;84:323–329.
    1. Zaharchuk G. Theoretical basis of hemodynamic MR imaging techniques to measure cerebral blood volume, cerebral blood flow, and permeability. AJNR Am. J. Neuroradiol. 2007;28:1850–1858.
    1. Ashton E, Riek J. Advanced MR techniques in multicenter clinical trials. J. Magn. Reson. Imaging. 2013;37:761–769.
    1. Meier P, Zierler KL. On the theory of the indicator-dilution method for measurement of blood flow and volume. J. Appl. Physiol. 1954;6:731–744.
    1. Neeman M, Dafni H. Structural, functional, and molecular MR imaging of the microvasculature. Annu. Rev. Biomed. Eng. 2003;5:29–56.
    1. Fisel CR, et al. MR contrast due to microscopically heterogeneous magnetic susceptibility: numerical simulations and applications to cerebral physiology. Magn. Reson. Med. 1991;17:336–347.
    1. Rosen BR, et al. Contrast agents and cerebral hemodynamics. Magn. Reson. Med. 1991;19:285–292.
    1. Rosen BR, et al. Susceptibility contrast imaging of cerebral blood volume: human experience. Magn. Reson. Med. 1991;22:293–299.
    1. Ogawa S, Lee TM, Kay AR, Tank DW. Brain magnetic resonance imaging with contrast dependent on blood oxygenation. Proc. Natl Acad. Sci. USA. 1990;87:9868–9872.
    1. Bandettini PA, Wong EC, Jesmanowicz A, Hinks RS, Hyde JS. Spin-echo and gradient-echo EPI of human brain activation using BOLD contrast: a comparative study at 1.5 T. NMR Biomed. 1994;7:12–20.
    1. Hoppel BE, et al. Measurement of regional blood oxygenation and cerebral hemodynamics. Magn. Reson. Med. 1993;30:715–723.
    1. Kennan RP, Zhong J, Gore JC. Intravascular susceptibility contrast mechanisms in tissues. Magn. Reson. Med. 1994;31:9–21.
    1. Weisskoff RM, Zuo CS, Boxerman JL, Rosen BR. Microscopic susceptibility variation and transverse relaxation: theory and experiment. Magn. Reson. Med. 1994;31:601–610.
    1. Boxerman JL, Hamberg LM, Rosen BR, Weisskoff RM. MR contrast due to intravascular magnetic susceptibility perturbations. Magn. Reson. Med. 1995;34:555–566.
    1. Kiselev VG, Posse S. Analytical model of susceptibility-induced MR signal dephasing: effect of diffusion in a microvascular network. Magn. Reson. Med. 1999;41:499–509.
    1. Dennie J, et al. NMR imaging of changes in vascular morphology due to tumor angiogenesis. Magn. Reson. Med. 1998;40:793–799.
    1. Donahue KM, et al. Utility of simultaneously acquired gradient-echo and spin-echo cerebral blood volume and morphology maps in brain tumor patients. Magn. Reson. Med. 2000;43:845–853.
    1. Provenzale JM, Mukundan S, Barboriak DP. Diffusion-weighted and perfusion MR imaging for brain tumor characterization and assessment of treatment response. Radiology. 2006;239:632–649.
    1. Covarrubias DJ, Rosen BR, Lev MH. Dynamic magnetic resonance perfusion imaging of brain tumors. Oncologist. 2004;9:528–537.
    1. Oostendorp M, Post MJ, Backes WH. Vessel growth and function: depiction with contrast-enhanced MR imaging. Radiology. 2009;251:317–335.
    1. Jensen JH, Chandra R. MR imaging of microvasculature. Magn. Reson. Med. 2000;44:224–230.
    1. Tropres I, et al. Vessel size imaging. Magn. Reson. Med. 2001;45:397–408.
    1. Kiselev VG, Strecker R, Ziyeh S, Speck O, Hennig J. Vessel size imaging in humans. Magn. Reson. Med. 2005;53:553–563.
    1. Remmele S, et al. Concurrent MR blood volume and vessel size estimation in tumors by robust and simultaneous DeltaR2 and DeltaR2* quantification. Magn. Reson. Med. 2011;66:144–153.
    1. Packard SD, et al. Functional response of tumor vasculature to PaCO2: determination of total and microvascular blood volume by, MRI. Neoplasia. 2003;5:330–338.
    1. Quarles CC, Schmainda KM. Assessment of the morphological and functional effects of the anti-angiogenic agent SU11657 on 9L gliosarcoma vasculature using dynamic susceptibility contrast MRI. Magn. Reson. Med. 2007;57:680–687.
    1. Caravan P, Ellison JJ, McMurry TJ, Lauffer RB. Gadolinium(III) chelates as MRI contrast agents: structure, dynamics, and applications. Chem. Rev. 1999;99:2293–2352.
    1. Schmiedeskamp H, et al. Simultaneous perfusion and permeability measurements using combined spin- and gradient-echo MRI. J. Cereb. Blood Flow Metab. 2013;33:732–743.
    1. Kim SG, et al. Cerebral blood volume MRI with intravascular superparamagnetic iron oxide nanoparticles. NMR Biomed. 2013;26:949–962.
    1. Farrar CT, et al. In vivo validation of MRI vessel caliber index measurement methods with intravital optical microscopy in a U87 mouse brain tumor model. Neuro Oncol. 2010;12:341–350.
    1. Weinstein JS, et al. Superparamagnetic iron oxide nanoparticles: diagnostic magnetic resonance imaging and potential therapeutic applications in neurooncology and central nervous system inflammatory pathologies, a review. J. Cereb. Blood Flow Metab. 2010;30:15–35.
    1. Christen T, et al. MR vascular fingerprinting: a new approach to compute cerebral blood volume, mean vessel radius, and oxygenation maps in the human brain. Neuroimage. 2014;89:262–270.
    1. Jochimsen TH, Moller HE. Increasing specificity in functional magnetic resonance imaging by estimation of vessel size based on changes in blood oxygenation. Neuroimage. 2008;40:228–236.
    1. Pannetier NA, Sohlin M, Christen T, Schad L, Schuff N. Numerical modeling of susceptibility-related MR signal dephasing with vessel size measurement: phantom validation at 3T. Magn. Reson. Med. .
    1. He X, Yablonskiy DA. Quantitative BOLD: mapping of human cerebral deoxygenated blood volume and oxygen extraction fraction: default state. Magn. Reson. Med. 2007;57:115–126.
    1. Hsu YY, Yang WS, Lim KE, Liu HL. Vessel size imaging using dual contrast agent injections. J. Magn. Reson. Imaging. 2009;30:1078–1084.
    1. Kiselev VG. Transverse relaxation effect of MRI contrast agents: a crucial issue for quantitative measurements of cerebral perfusion. J. Magn. Reson. Imaging. 2005;22:693–696.
    1. Emblem KE, et al. Vessel architectural imaging identifies cancer patient responders to anti-angiogenic therapy. Nat. Med. 2013;19:1178–1183.
    1. Kellner E, et al. Arterial input function measurements for bolus tracking perfusion imaging in the brain. Magn. Reson. Med. 2013;69:771–780.
    1. Germuska MA, Meakin JA, Bulte DP. The influence of noise on BOLD-mediated vessel size imaging analysis methods. J. Cereb. Blood Flow Metab. 2013;33:1857–1863.
    1. Lemasson B, et al. Assessment of multiparametric MRI in a human glioma model to monitor cytotoxic and anti-angiogenic drug effects. NMR Biomed. 2011;24:473–482.
    1. Fredrickson J, et al. Clinical translation of VSI using ferumoxytol: feasibility in a phase I oncology clinical trial population [abstract] Proc. Int. Soc. Mag. Reson. Med. (ISMRM) Annual Meeting. 2012:a1987.
    1. Persigehl T, et al. Tumor blood volume determination by using susceptibility-corrected DeltaR2* multiecho MR. Radiology. 2010;255:781–789.
    1. Wang Y-X. Superparamagnetic iron oxide based MRI contrast agents: current status of clinical application. Quant. Imaging Med. Surg. 2011;1:35–40.
    1. Pannetier N, et al. Vessel size index measurements in a rat model of glioma: comparison of the dynamic (Gd) and steady-state (iron-oxide) susceptibility contrast MRI approaches. NMR Biomed. 2012;25:218–226.
    1. Schmiedeskamp H, Straka M, Bammer R. Compensation of slice profile mismatch in combined spin- and gradient-echo echo-planar imaging pulse sequences. Magn. Reson. Med. 2012;67:378–388.
    1. Paulson ES, Schmainda KM. Comparison of dynamic susceptibility-weighted contrast-enhanced MR methods: recommendations for measuring relative cerebral blood volume in brain tumors. Radiology. 2008;249:601–613.
    1. Li SP, et al. Primary human breast adenocarcinoma: imaging and histologic correlates of intrinsic susceptibility-weighted MR imaging before and during chemotherapy. Radiology. 2010;257:643–652.
    1. Benner T, Heiland S, Erb G, Forsting M, Sartor K. Accuracy of gamma-variate fits to concentration-time curves from dynamic susceptibility-contrast enhanced MRI: influence of time resolution, maximal signal drop and signal-to-noise. Magn. Reson. Imaging. 1997;15:307–317.
    1. Li X, Tian J, Millard RK. Erroneous and inappropriate use of gamma fits to tracer-dilution curves in magnetic resonance imaging and nuclear medicine. Magn. Reson. Imaging. 2003;21:1095–1096.
    1. Ito H, Kanno I, Ibaraki M, Hatazawa J, Miura S. Changes in human cerebral blood flow and cerebral blood volume during hypercapnia and hypocapnia measured by positron emission tomography. J. Cereb. Blood Flow Metab. 2003;23:665–670.
    1. Ito H, Ibaraki M, Kanno I, Fukuda H, Miura S. Changes in the arterial fraction of human cerebral blood volume during hypercapnia and hypocapnia measured by positron emission tomography. J. Cereb. Blood Flow Metab. 2005;25:852–857.
    1. Pathak AP, Ward BD, Schmainda KM. A novel technique for modeling susceptibility-based contrast mechanisms for arbitrary microvascular geometries: the finite perturber method. Neuroimage. 2008;40:1130–1143.
    1. Tropres I, Lamalle L, Farion R, Segebarth C, Remy C. Vessel size imaging using low intravascular contrast agent concentrations. MAGMA. 2004;17:313–316.
    1. Valable S, et al. Assessment of blood volume, vessel size, and the expression of angiogenic factors in two rat glioma models: a longitudinal in vivo and ex vivo study. NMR Biomed. 2008;21:1043–1056.
    1. Kim E, et al. Assessing breast cancer angiogenesis in vivo: which susceptibility contrast MRI biomarkers are relevant? Magn. Reson. Med. 2013;70:1106–1116.
    1. Persigehl T, et al. Vessel size imaging (VSI) by robust magnetic resonance (MR) relaxometry: MR-VSI of solid tumors in correlation with immunohistology and intravital microscopy. Mol. Imaging. 2013;12:1–11.
    1. Ungersma SE, et al. Vessel imaging with viable tumor analysis for quantification of tumor angiogenesis. Magn. Reson. Med. 2010;63:1637–1647.
    1. Bauerle T, Merz M, Komljenovic D, Zwick S, Semmler W. Drug-induced vessel remodeling in bone metastases as assessed by dynamic contrast enhanced magnetic resonance imaging and vessel size imaging: a longitudinal in vivo study. Clin. Cancer Res. 2010;16:3215–3225.
    1. Tofts PS, Collins DJ. Multicentre imaging measurements for oncology and in the brain. Br. J. Radiol. 2011;84(2):S213–S226.
    1. Larsen OA, Lassen NA. Cerebral hematocrit in normal man. J. Appl. Physiol. 1964;19:571–574.
    1. Ostergaard L, Weisskoff RM, Chesler DA, Gyldensted C, Rosen BR. High resolution measurement of cerebral blood flow using intravascular tracer bolus passages. Part I: mathematical approach and statistical analysis. Magn. Reson. Med. 1996;36:715–725.
    1. Schmainda KM, et al. Characterization of a first-pass gradient-echo spin-echo method to predict brain tumor grade and angiogenesis. AJNR Am. J. Neuroradiol. 2004;25:1524–1532.
    1. Lamalle L, et al. VSI and BV MRI of human brain tumours [abstract] Proc. 11th Annu. Meeting Int. Soc. Mag. Reson. Med. 2003:a1271.
    1. Kiselev VG, et al. quantitative vessel size imaging in humans [abstract] Proc. 11th Annu. Meeting Int. Soc. Mag. Reson. Med. 2003:a2192.
    1. Breyer T, et al. Clinical evaluation of vessel size imaging in 31 cases of human glial brain tumor [abstract] Proc. 15th Annu. Meeting Int. Soc. Mag. Reson. Med. 2007:a836.
    1. Xu C, Kiselev VG, Moller HE, Fiebach JB. Dynamic hysteresis between gradient echo and spin echo attenuations in dynamic susceptibility contrast imaging. Magn. Reson. Med. 2013;69:981–991.
    1. Pectasides M, et al. Evaluation of vessel size heterogeneity in brain tumors with dynamic contrast-enhanced dual echo perfusion weighted imaging [abstract] Proc. 16th Annu. Meeting Int. Soc. Mag. Reson. Med. 2004:a152.
    1. Batchelor TT, et al. AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell. 2007;11:83–95.
    1. Sorensen AG, et al. A “vascular normalization index” as potential mechanistic biomarker to predict survival after a single dose of cediranib in recurrent glioblastoma patients. Cancer Res. 2009;69:5296–5300.
    1. Batchelor TT, et al. Improved tumor oxygenation and survival in glioblastoma patients who show increased blood perfusion after cediranib and chemoradiation. Proc. Natl Acad. Sci. USA. 2013;110:19059–19064.
    1. Polaskova P, et al. Repeatability of MR-based vessel caliber estimates in brain tumor imaging [abstract] Proc. Am. Soc. Neuroradiol. Annu. Meeting. 2013:O-404.
    1. Lüdemann L, et al. Simultaneous quantification of perfusion and permeability in the prostate using dynamic contrast-enhanced magnetic resonance imaging with an inversion-prepared dual-contrast sequence. Ann. Biomed. Eng. 2009;37:749–762.
    1. Jin N, et al. GESFIDE-PROPELLER approach for simultaneous R2 and R2* measurements in the abdomen. Magn. Reson. Imaging. 2013;31:1760–1765.
    1. Yang X, et al. Evaluation of renal oxygenation in rat by using R2’ at 3-T magnetic resonance: initial observation. Acad. Radiol. 2008;15:912–918.
    1. Ferretti S, et al. Patupilone induced vascular disruption in orthotopic rodent tumor models detected by magnetic resonance imaging and interstitial fluid pressure. Clin. Cancer Res. 2005;11:7773–7784.
    1. Howe FA, McPhail LD, Griffiths JR, McIntyre DJ, Robinson SP. Vessel size index magnetic resonance imaging to monitor the effect of antivascular treatment in a rodent tumor model. Int. J. Radiat. Oncol. Biol. Phys. 2008;71:1470–1476.
    1. Kording F, et al. Simultaneous assessment of vessel size index, relative blood volume, and vessel permeability in a mouse brain tumor model using a combined spin echo gradient echo echo-planar imaging sequence and viable tumor analysis. J. Magn. Reson. Imaging. .
    1. Nielsen T, et al. Combretastatin A-4 phosphate affects tumor vessel volume and size distribution as assessed using MRI-based vessel size imaging. Clin. Cancer Res. 2012;18:6469–6477.
    1. Yuan F, et al. Vascular permeability and microcirculation of gliomas and mammary carcinomas transplanted in rat and mouse cranial windows. Cancer Res. 1994;54:4564–4568.
    1. Kamoun WS, et al. Edema control by cediranib, a vascular endothelial growth factor receptor-targeted kinase inhibitor, prolongs survival despite persistent brain tumor growth in mice. J. Clin. Oncol. 2009;27:2542–2552.
    1. Walker-Samuel S, et al. Non-invasive in vivo imaging of vessel calibre in orthotopic prostate tumour xenografts. Int. J. Cancer. 2012;130:1284–1293.
    1. Zwick S, et al. Assessment of vascular remodeling under antiangiogenic therapy using DCE-MRI and vessel size imaging. J. Magn. Reson. Imaging. 2009;29:1125–1133.
    1. Lemasson B, et al. Avastin alone or combined to Campto® reduces local blood oxygen saturation in an orthotopic human glioblastoma model (U87-MG) in nude rats [abstract] Proc. 17th Annu. Meeting Int. Soc. Mag. Reson. Med. 2009:a1013.
    1. Merz M, Komljenovic D, Zwick S, Semmler W, Bauerle T. Sorafenib tosylate and paclitaxel induce anti-angiogenic, anti-tumour and anti-resorptive effects in experimental breast cancer bone metastases. Eur. J. Cancer. 2011;47:277–286.
    1. Zwick S, et al. Dynamic contrast-enhanced MRI and vessel size imaging sensitively indicate antiangiogenic therapy effects on tumor xenografts in mice [abstract] Proc. 15th Annu. Meeting Int. Soc. Mag. Reson. Med. 2007:a564.
    1. Farrar CT, et al. Sensitivity of MRI tumor biomarkers to VEGFR inhibitor therapy in an orthotopic mouse glioma model. PLoS ONE. 2011;6:e17228.
    1. Woenne EC, et al. MMP inhibition blocks fibroblast-dependent skin cancer invasion, reduces vascularization and alters VEGF-A and PDGF-BB expression. Anticancer Res. 2010;30:703–711.
    1. Palmowski M, et al. Vessel fractions in tumor xenografts depicted by flow- or contrast-sensitive three-dimensional high-frequency Doppler ultrasound respond differently to antiangiogenic treatment. Cancer Res. 2008;68:7042–7049.
    1. Benjamin LE, Golijanin D, Itin A, Pode D, Keshet E. Selective ablation of immature blood vessels in established human tumors follows vascular endothelial growth factor withdrawal. J. Clin. Invest. 1999;103:159–165.
    1. Remmele S, Senegas J, Persigehl T, Bremer C, Ring J. Simultaneous blood volume and vessel size imaging technique for localized therapy response detection [abstract] Proc. 17th Annu. Meeting Int. Soc. Mag. Reson. Med. 2009:a4203.
    1. Opstad KS, Howe FA. Vessel size index MRI to monitor the effects of vascular disruption by ASA404 (vadimezan, 5,6-dimethylxanthenone-4-acetic acid) in orthotopic gliomas [abstract] Proc. 18th Annu. Meeting Int. Soc. Mag. Reson. Med. 2010:a4837.
    1. Ullrich RT, et al. In-vivo visualization of tumor microvessel density and response to anti-angiogenic treatment by high resolution MRI in mice. PLoS ONE. 2011;6:e19592.
    1. Boult JK, et al. False-negative MRI biomarkers of tumour response to targeted cancer therapeutics. Br J. Cancer. 2012;106:1960–1966.
    1. Boult JK, Terkelsen J, Walker-Samuel S, Bradley DP, Robinson SP. A multi-parametric imaging investigation of the response of C6 glioma xenografts to MLN0518 (tandutinib) treatment. PLoS ONE. 2013;8:e63024.
    1. US National Library of Medicine. . 2014 [online], .
    1. US National Library of Medicine. . 2014 [online], .
    1. US National Library of Medicine. . 2014 [online], .
    1. Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science. 2005;307:58–62.
    1. US National Library of Medicine. . 2014 [online], .
    1. Batchelor TT, et al. Phase III randomized trial comparing the efficacy of cediranib as monotherapy, and in combination with lomustine, versus lomustine alone in patients with recurrent glioblastoma. J. Clin. Oncol. 2013;31:3212–3218.
    1. Gilbert MR, et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N. Engl. J. Med. 2014;370:699–708.
    1. Chinot OL, et al. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N. Engl. J. Med. 2014;370:709–722.
    1. Weller M, Yung WK. Angiogenesis inhibition for glioblastoma at the edge: beyond AVAGlio and RTOG 0825. Neuro Oncol. 2013;15:971.
    1. NCI Cancer Imaging Program. Imaging Guidelines for Clinical Trials. 2014 [online], .
    1. Caseiras GB, et al. Relative cerebral blood volume measurements of low-grade gliomas predict patient outcome in a multi-institution setting. Eur. J. Radiol. 2010;73:215–220.
    1. Ng CS, et al. Reproducibility of perfusion parameters in dynamic contrast-enhanced MRI of lung and liver tumors: effect on estimates of patient sample size in clinical trials and on individual patient responses. AJR Am. J. Roentgenol. 2010;194:W134–W140.
    1. Gunter JL, et al. Measurement of MRI scanner performance with the ADNI phantom. Med. Phys. 2009;36:2193–2205.
    1. Rosen BR, Belliveau JW, Vevea JM, Brady TJ. Perfusion imaging with NMR contrast agents. Magn. Reson. Med. 1990;14:249–265.
    1. NCI Cancer Imaging Program. Imaging Response Criteria. 2014 [online], .
    1. Gaustad JV, Pozdniakova V, Hompland T, Simonsen TG, Rofstad EK. Magnetic resonance imaging identifies early effects of sunitinib treatment in human melanoma xenografts. J. Exp. Clin. Cancer Res. 2013;32:93.
    1. Yang X, Knopp MV. Quantifying tumor vascular heterogeneity with dynamic contrast-enhanced magnetic resonance imaging: a review. J. Biomed. Biotechnol. 2011;2011:732–848.
    1. Christen T, Bolar DS, Zaharchuk G. Imaging brain oxygenation with MRI using blood oxygenation approaches: methods, validation, and clinical applications. AJNR Am. J. Neuroradiol. 2013;34:1113–1123.
    1. Jespersen SN, Ostergaard L. The roles of cerebral blood flow, capillary transit time heterogeneity, and oxygen tension in brain oxygenation and metabolism. J. Cereb. Blood Flow Metab. 2012;32:264–277.
    1. Sorensen AG, et al. Increased survival of glioblastoma patients who respond to antiangiogenic therapy with elevated blood perfusion. Cancer Res. 2012;72:402–407.
    1. Badruddoja MA, et al. Antiangiogenic effects of dexamethasone in 9L gliosarcoma assessed by MRI cerebral blood volume maps. Neuro Oncol. 2003;5:235–243.
    1. Tropres I, et al. In vivo assessment of tumoral angiogenesis. Magn. Reson. Med. 2004;51:533–541.
    1. Beaumont M, et al. Characterization of tumor angiogenesis in rat brain using iron-based vessel size index MRI in combination with gadolinium-based dynamic contrast-enhanced MRI. J. Cereb. Blood Flow Metab. 2009;29:1714–1726.
    1. Douma K, et al. Evaluation of magnetic resonance vessel size imaging by two-photon laser scanning microscopy. Magn. Reson. Med. 2010;63:930–939.
    1. Lemasson B, et al. In vivo imaging of vessel diameter, size, and density: a comparative study between MRI and histology. Magn. Reson. Med. 2013;69:18–26.
    1. Zhang Y, Jiang J, Zhang S, Xiong W, Zhu W. MR Vessel size imaging of brain tumors [abstract] Proc. 98th Annu. Meeting Radiol. Soc. N. Am. 2012 LL-NRS-TU5A.
    1. Viel T, et al. Non-invasive imaging of glioma vessel size and densities in correlation with tumour cell proliferation by small animal PET and MRI. Eur. J. Nucl. Med. Mol. Imaging. 2013;40:1595–1606.
    1. Sampath D, et al. Multimodal microvascular imaging reveals that selective inhibition of class I PI3K is sufficient to induce an antivascular response. Neoplasia. 2013;15:694–711.

Source: PubMed

3
Suscribir