1
|
Salavati H, Pullens P, Debbaut C, Ceelen W. Hydraulic conductivity of human cancer tissue: A hybrid study. Bioeng Transl Med 2024; 9:e10617. [PMID: 38435818 PMCID: PMC10905546 DOI: 10.1002/btm2.10617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/22/2023] [Accepted: 10/15/2023] [Indexed: 03/05/2024] Open
Abstract
Background Elevated tumor tissue interstitial fluid pressure (IFP) is an adverse biomechanical biomarker that predicts poor therapy response and an aggressive phenotype. Advances in functional imaging have opened the prospect of measuring IFP non-invasively. Image-based estimation of the IFP requires knowledge of the tissue hydraulic conductivity (K), a measure for the ease of bulk flow through the interstitium. However, data on the magnitude of K in human cancer tissue are not available. Methods We measured the hydraulic conductivity of tumor tissue using modified Ussing chambers in surgical resection specimens. The effect of the tumor microenvironment (TME) on K was investigated by quantifying the collagen content, cell density, and fibroblast density of the tested samples using quantitative immune histochemistry. Also, we developed a computational fluid dynamics (CFD) model to evaluate the role of K on interstitial fluid flow and drug transport in solid tumors. Results The results show that the hydraulic conductivity of human tumor tissues is very limited, ranging from approximately 10-15 to 10-14 m2/Pa∙s. Moreover, K values varied significantly between tumor types and between different samples from the same tumor. A significant inverse correlation was found between collagen fiber density and hydraulic conductivity values. However, no correlation was detected between K and cancer cell or fibroblast densities. The computational model demonstrated the impact of K on the interstitial fluid flow and the drug concentration profile: higher K values led to a lower IFP and deeper drug penetration. Conclusions Human tumor tissue is characterized by a very limited hydraulic conductivity, representing a barrier to effective drug transport. The results of this study can inform the development of realistic computational models, facilitate non-invasive IFP estimation, and contribute to stromal targeting anticancer therapies.
Collapse
Affiliation(s)
- Hooman Salavati
- Department of Human Structure and RepairGhent UniversityGhentBelgium
- IBiTech–BioMMedA, Ghent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
| | - Pim Pullens
- Department of RadiologyUniversity Hospital GhentGhentBelgium
- Ghent Institute of Functional and Metabolic Imaging (GIFMI)Ghent UniversityGhentBelgium
- IBiTech–Medisip, Ghent UniversityGhentBelgium
| | - Charlotte Debbaut
- IBiTech–BioMMedA, Ghent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
| | - Wim Ceelen
- Department of Human Structure and RepairGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
| |
Collapse
|
2
|
Salavati H, Debbaut C, Pullens P, Ceelen W. Interstitial fluid pressure as an emerging biomarker in solid tumors. Biochim Biophys Acta Rev Cancer 2022; 1877:188792. [PMID: 36084861 DOI: 10.1016/j.bbcan.2022.188792] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/12/2022] [Accepted: 08/30/2022] [Indexed: 11/28/2022]
Abstract
The physical microenvironment of cancer is characterized by elevated stiffness and tissue pressure, the main component of which is the interstitial fluid pressure (IFP). Elevated IFP is an established negative predictive and prognostic parameter, directly affecting malignant behavior and therapy response. As such, measurement of the IFP would allow to develop strategies aimed at engineering the physical microenvironment of cancer. Traditionally, IFP measurement required the use of invasive methods. Recent progress in dynamic and functional imaging methods such as dynamic contrast enhanced (DCE) magnetic resonance imaging and elastography, combined with numerical models and simulation, allows to comprehensively assess the biomechanical landscape of cancer, and may help to overcome physical barriers to drug delivery and immune cell infiltration. Here, we provide a comprehensive overview of the origin of elevated IFP, and its role in the malignant phenotype. Also, we review the methods used to measure IFP using invasive and imaging based methods, and highlight remaining obstacles and potential areas of progress in order to implement IFP measurement in clinical practice.
Collapse
Affiliation(s)
- Hooman Salavati
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium; IBitech- Biommeda, Department of Electronics and Information Systems, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Charlotte Debbaut
- IBitech- Biommeda, Department of Electronics and Information Systems, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Pim Pullens
- Department of Radiology, Ghent University Hospital, Ghent, Belgium; Ghent Institute of Functional and Metabolic Imaging (GIFMI), Ghent University, Ghent, Belgium; IBitech- Medisip, Ghent University, Ghent, Belgium
| | - Wim Ceelen
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| |
Collapse
|
3
|
Mohammadi M, Aghanajafi C, Soltani M, Raahemifar K. Numerical Investigation on the Anti-Angiogenic Therapy-Induced Normalization in Solid Tumors. Pharmaceutics 2022; 14:363. [PMID: 35214095 PMCID: PMC8877966 DOI: 10.3390/pharmaceutics14020363] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/29/2022] [Accepted: 01/31/2022] [Indexed: 01/27/2023] Open
Abstract
This study numerically analyzes the fluid flow and solute transport in a solid tumor to comprehensively examine the consequence of normalization induced by anti-angiogenic therapy on drug delivery. The current study leads to a more accurate model in comparison to previous research, as it incorporates a non-homogeneous real-human solid tumor including necrotic, semi-necrotic, and well-vascularized regions. Additionally, the model considers the effects of concurrently chemotherapeutic agents (three macromolecules of IgG, F(ab')2, and F(ab')) and different normalization intensities in various tumor sizes. Examining the long-term influence of normalization on the quality of drug uptake by necrotic area is another contribution of the present study. Results show that normalization decreases the interstitial fluid pressure (IFP) and spreads the pressure gradient and non-zero interstitial fluid velocity (IFV) into inner areas. Subsequently, wash-out of the drug from the tumor periphery is decreased. It is also demonstrated that normalization can improve the distribution of solute concentration in the interstitium. The efficiency of normalization is introduced as a function of the time course of perfusion, which depends on the tumor size, drug type, as well as normalization intensity, and consequently on the dominant mechanism of drug delivery. It is suggested to accompany anti-angiogenic therapy by F(ab') in large tumor size (Req=2.79 cm) to improve reservoir behavior benefit from normalization. However, IgG is proposed as the better option in the small tumor (Req=0.46 cm), in which normalization finds the opportunity of enhancing uniformity of IgG average exposure by 22%. This study could provide a perspective for preclinical and clinical trials on how to take advantage of normalization, as an adjuvant treatment, in improving drug delivery into a non-homogeneous solid tumor.
Collapse
Affiliation(s)
- Mahya Mohammadi
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19967-15433, Iran; (M.M.); (C.A.)
- Department of Applied Mathematics, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Cyrus Aghanajafi
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19967-15433, Iran; (M.M.); (C.A.)
| | - M. Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19967-15433, Iran; (M.M.); (C.A.)
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Advanced Bioengineering Initiative Center, Multidisciplinary International Complex, K. N. Toosi University of Technology, Tehran 14176-14411, Iran
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Kaamran Raahemifar
- Data Science and Artificial Intelligence Program, College of Information Sciences and Technology (IST), Penn State University, State College, PA 16801, USA;
- School of Optometry and Vision Science, Faculty of Science, University of Waterloo, 200 University Ave W, Waterloo, ON N2L 3G1, Canada
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue W, Waterloo, ON N2L 3G1, Canada
| |
Collapse
|
4
|
Li B, Xu D, Zhou J, Wang SC, Cai YX, Li H, Xu HB. Monitoring Bevacizumab-Induced Tumor Vascular Normalization by Intravoxel Incoherent Motion Diffusion-Weighted MRI. J Magn Reson Imaging 2021; 56:427-439. [PMID: 34873766 DOI: 10.1002/jmri.28012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/21/2021] [Accepted: 11/23/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Accurate monitoring of tumor blood vessel normalization progression is beneficial to accurate treatment of patients. At present, there is a lack of safe and noninvasive monitoring methods. PURPOSE To serial monitor the vascular normalization time window of tumor antiangiogenesis treatment through intravoxel incoherent motion diffusion-weighted imaging (IVIM-DWI) and histopathological methods. STUDY TYPE Exploratory animal study. POPULATION Sixty rat C6 glioma models were randomly and equally divided into the control groups (N = 30) and bevacizumab treatment groups (N = 30). Twenty-five for magnetic resonance imaging (MRI) and five for electron microscope testing in each group. FIELD STRENGTH/SEQUENCE T1-weighted imaging (T1WI), T2WI with a fast spin echo sequence and IVIM-DWI with a spin-echo echo-planar imaging sequence at 3 T. ASSESSMENT IVIM-DWI quantitative parameters (f, D, D*, and fD*) were obtained on days 0, 2, 4, 6, and 8 after bevacizumab treatment. After MRI, the microvessel density (MVD), pericyte coverage, and hypoxia-inducible factor-1α (HIF-1α) were assessed. Electron microscope observation was performed at each time point. STATISTICAL TESTS One-way analysis of variance and Student's t-tests were used to compare differences within and between groups. Spearman's correlation coefficient (r) assess the correlation between IVIM and pathological parameters. The intragroup correlation coefficient was determined to assess the repeatability of each IVIM parameter. RESULTS The IVIM-DWI perfusion parameters (f and fD*) of the treated group were higher than the control group on days 2 and 4. Compared to the control group, MVD decreased on days 2 and pericyte coverage increased on days 4 in the treatment group. Electron microscopy showed that the tight junctions of the treatment group were prolonged on days 2-4. In the control group, f had the highest correlation with MVD (r = 0.689). In the treated group, f had a good correlation with pericyte coverage (r = 0.557), HIF-1α had a moderately positive correlation with f (r = 0.480) and fD*(r = 0.447). DATA CONCLUSION The vascular normalization time window of bevacizumab treatment of glioma was days 2-4 after antiangiogenesis treatment, which could be monitored noninvasively by IVIM-DWI. EVIDENCE LEVEL 2 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Bo Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China.,The First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Dan Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jie Zhou
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shou-Chao Wang
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yu-Xiang Cai
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Huan Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hai-Bo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
5
|
Cellular and Extracellular Components in Tumor Microenvironment and Their Application in Early Diagnosis of Cancers. Anal Cell Pathol (Amst) 2020; 2020:6283796. [PMID: 32377504 PMCID: PMC7199555 DOI: 10.1155/2020/6283796] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/11/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022] Open
Abstract
Tumors are surrounded by complex environmental components, including blood and lymph vessels, fibroblasts, endothelial cells, immune cells, cytokines, extracellular vesicles, and extracellular matrix. All the stromal components together with the tumor cells form the tumor microenvironment (TME). In addition, extracellular physical and chemical factors, including extracellular pH, hypoxia, elevated interstitial fluid pressure, and fibrosis, are closely associated with tumor progression, metastasis, immunosuppression, and drug resistance. Cellular and extracellular components in TME contribute to nearly all procedures of carcinogenesis. By summarizing the recent work in this field, we make a comprehensive review on the role of cellular and extracellular components in the process of carcinogenesis and their potential application in early diagnosis of cancer. We hope that a systematic review of the diverse aspects of TME will help both research scientists and clinicians in this field.
Collapse
|
6
|
Targeting the invincible barrier for drug delivery in solid cancers: interstitial fluid pressure. Oncotarget 2018; 9:35723-35725. [PMID: 30515264 PMCID: PMC6254664 DOI: 10.18632/oncotarget.26267] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 10/06/2018] [Indexed: 12/17/2022] Open
Abstract
Although a number of new systemic therapeutic options in patients with advanced solid cancers have emerged due to the improved knowledge of molecular dysregulation in cancers, the durable, long-term, objective responses infrequently occur. This editorial article highlights the major limitation of current systemic therapy due to an inefficient drug delivery. While several mechanisms contributing to cancer drug resistance have been described, the common key barrier among solid cancers is the unique tumor microenvironment that causes the high interstitial fluid pressure (IFP). We discussed the mechanism causing an elevated IFP and how it interferes with drug delivery. To target the high IFP, we demonstrated the novel approach using gold nanoparticle carrying recombinant human tumor necrosis factor (TNF), a vascular disrupting agent, that preferentially and specifically targets tumors while the systemic toxicity is markedly reduced. The addition of cytotoxic agent by either directly conjugating to the gold nanoparticle or by systemic administration following gold nanoparticle carrying TNF resulted in significantly reduced tumor burden and increased survival in multiple mouse models with primary and metastatic endocrine cancer and pancreatic ductal carcinoma. A clinical trial in patients with advanced solid cancers is warranted based on the promising results in preclinical studies.
Collapse
|
7
|
Bettega D, Calzolari P, Ciocca M, Facoetti A, Lafiandra M, Marchesini R, Molinelli S, Pignoli E, Vischioni B. Combining proton or photon irradiation with epothilone B. An
in vitro
study of cytotoxicity in human cancer cells. Biomed Phys Eng Express 2017. [DOI: 10.1088/2057-1976/aa818f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
8
|
Robinson SP, Boult JKR, Vasudev NS, Reynolds AR. Monitoring the Vascular Response and Resistance to Sunitinib in Renal Cell Carcinoma In Vivo with Susceptibility Contrast MRI. Cancer Res 2017; 77:4127-4134. [PMID: 28566330 PMCID: PMC6175052 DOI: 10.1158/0008-5472.can-17-0248] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/03/2017] [Accepted: 05/22/2017] [Indexed: 12/25/2022]
Abstract
Antiangiogenic therapy is efficacious in metastatic renal cell carcinoma (mRCC). However, the ability of antiangiogenic drugs to delay tumor progression and extend survival is limited, due to either innate or acquired drug resistance. Furthermore, there are currently no validated biomarkers that predict which mRCC patients will benefit from antiangiogenic therapy. Here, we exploit susceptibility contrast MRI (SC-MRI) using intravascular ultrasmall superparamagnetic iron oxide particles to quantify and evaluate tumor fractional blood volume (fBV) as a noninvasive imaging biomarker of response to the antiangiogenic drug sunitinib. We also interrogate the vascular phenotype of RCC xenografts exhibiting acquired resistance to sunitinib. SC-MRI of 786-0 xenografts prior to and 2 weeks after daily treatment with 40 mg/kg sunitinib revealed a 71% (P < 0.01) reduction in fBV in the absence of any change in tumor volume. This response was associated with significantly lower microvessel density (P < 0.01) and lower uptake of the perfusion marker Hoechst 33342 (P < 0.05). The average pretreatment tumor fBV was negatively correlated (R2 = 0.92, P < 0.0001) with sunitinib-induced changes in tumor fBV across the cohort. SC-MRI also revealed suppressed fBV in tumors that acquired resistance to sunitinib. In conclusion, SC-MRI enabled monitoring of the antiangiogenic response of 786-0 RCC xenografts to sunitinib, which revealed that pretreatment tumor fBV was found to be a predictive biomarker of subsequent reduction in tumor blood volume in response to sunitinib, and acquired resistance to sunitinib was not associated with a parallel increase in tumor blood volume. Cancer Res; 77(15); 4127-34. ©2017 AACR.
Collapse
Affiliation(s)
- Simon P Robinson
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom.
| | - Jessica K R Boult
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Naveen S Vasudev
- Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Andrew R Reynolds
- Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
- Early Clinical Development, Innovative Medicines and Early Development, AstraZeneca, Cambridge, United Kingdom
| |
Collapse
|
9
|
Yan J, Zhang D, Yu H, Ma L, Deng M, Tang Z, Zhang X. Patupilone-loaded poly(L-glutamic acid)-graft-methoxy-poly(ethylene glycol) micelle for oncotherapy. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2017; 28:394-414. [DOI: 10.1080/09205063.2016.1277827] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Jing Yan
- Department of Chemistry, Xiangtan University, Xiangtan, PR China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, PR China
| | - Dawei Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, PR China
| | - Haiyang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, PR China
| | - Lili Ma
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, PR China
| | - Mingxiao Deng
- College of Chemistry, Northeast Normal University, Changchun, PR China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, PR China
| | - Xuefei Zhang
- Department of Chemistry, Xiangtan University, Xiangtan, PR China
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, Key Laboratory of Polymeric Materials and Application Technology of Hunan Province, Xiangtan University, Xiangtan, PR China
| |
Collapse
|
10
|
Yang SH, Lin J, Lu F, Han ZH, Fu CX, Lv P, Liu H, Gao DM. Evaluation of antiangiogenic and antiproliferative effects of sorafenib by sequential histology and intravoxel incoherent motion diffusion-weighted imaging in an orthotopic hepatocellular carcinoma xenograft model. J Magn Reson Imaging 2016; 45:270-280. [PMID: 27299302 DOI: 10.1002/jmri.25344] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/27/2016] [Indexed: 12/11/2022] Open
Abstract
PURPOSE To investigate the effectiveness of intravoxel incoherent motion (IVIM) in the assessment of the therapeutic efficacy of sorafenib in an orthotopic hepatocellular carcinoma (HCC) xenograft model. MATERIALS AND METHODS Thirty-five HCC nude mouse models were established. IVIM was performed on a 1.5T MR scanner at baseline (n = 5) and serially at 7, 14, and 21 days after sorafenib treatment. The apparent diffusion coefficient (ADCtotal ), true diffusion coefficient (D), pseudodiffusion coefficient (D*), and perfusion fraction (f) at these timepoints were measured and compared between the treated (n = 15) and control group (n = 15). Differences in measurements among different timepoints were evaluated. Correlations between IVIM parameters and histologic features including necrotic fraction (NF) and microvessel density (MVD) were analyzed. RESULTS Compared to the control group, ADCtotal and D were significantly higher at each timepoint (P = 0.009), while f significantly decreased at 7 days (P = 0.009) and increased at 21 days (P = 0.028) in the treated group. Serial measurements in the treated group showed that both ADCtotal and D increased significantly at 7, 14, and 21 days compared to baseline (P < 0.05), while f significantly declined at 7 days (P = 0.016) and increased at 21 days (P = 0.009). Significant correlations were found between ADCtotal and NF (r = 0.811, P < 0.001), D and NF (r = 0.838, P < 0.001), and between f and NF (r = 0.528, P = 0.017) in the treated group. CONCLUSION IVIM may provide useful biomarkers for evaluating the therapeutic effects of sorafenib on HCC. LEVEL OF EVIDENCE 1 J. Magn. Reson. Imaging 2017;45:270-280.
Collapse
Affiliation(s)
- Shuo-Hui Yang
- Department of Radiology, Zhongshan Hospital, Shanghai Medical College, Fudan University, and Shanghai Institute of Medical Imaging, Shanghai, China
| | - Jiang Lin
- Department of Radiology, Zhongshan Hospital, Shanghai Medical College, Fudan University, and Shanghai Institute of Medical Imaging, Shanghai, China
| | - Fang Lu
- Department of Radiology, Shuguang Hosipital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhi-Hong Han
- Department of Pathology, Shuguang Hosipital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cai-Xia Fu
- Siemens Shenzhen Magnetic Resonance Ltd., Shenzhen, China
| | - Peng Lv
- Department of Radiology, Zhongshan Hospital, Shanghai Medical College, Fudan University, and Shanghai Institute of Medical Imaging, Shanghai, China
| | - Hao Liu
- Department of Radiology, Zhongshan Hospital, Shanghai Medical College, Fudan University, and Shanghai Institute of Medical Imaging, Shanghai, China
| | - Dong-Mei Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| |
Collapse
|
11
|
Bold G, Schnell C, Furet P, McSheehy P, Brüggen J, Mestan J, Manley PW, Drückes P, Burglin M, Dürler U, Loretan J, Reuter R, Wartmann M, Theuer A, Bauer-Probst B, Martiny-Baron G, Allegrini P, Goepfert A, Wood J, Littlewood-Evans A. A Novel Potent Oral Series of VEGFR2 Inhibitors Abrogate Tumor Growth by Inhibiting Angiogenesis. J Med Chem 2015; 59:132-46. [DOI: 10.1021/acs.jmedchem.5b01582] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Guido Bold
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Christian Schnell
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Pascal Furet
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Paul McSheehy
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Josef Brüggen
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Jürgen Mestan
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Paul W. Manley
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Peter Drückes
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Marion Burglin
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Ursula Dürler
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Jacqueline Loretan
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Robert Reuter
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Markus Wartmann
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Andreas Theuer
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Beatrice Bauer-Probst
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Georg Martiny-Baron
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Peter Allegrini
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Arnaud Goepfert
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Jeanette Wood
- Oncology Research, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | | |
Collapse
|
12
|
Malvicini M, Fiore E, Ghiaccio V, Piccioni F, Rizzo M, Olmedo Bonadeo L, García M, Rodríguez M, Bayo J, Peixoto E, Atorrasagasti C, Alaniz L, Aquino J, Matar P, Mazzolini G. Tumor Microenvironment Remodeling by 4-Methylumbelliferone Boosts the Antitumor Effect of Combined Immunotherapy in Murine Colorectal Carcinoma. Mol Ther 2015; 23:1444-55. [PMID: 26105158 DOI: 10.1038/mt.2015.112] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 06/08/2015] [Indexed: 01/19/2023] Open
Abstract
We have previously demonstrated that a low dose of cyclophosphamide (Cy) combined with gene therapy of interleukin-12 (AdIL-12) has a synergistic, although limited, antitumoral effect in mice with colorectal carcinoma. The main mechanism involved in the efficacy of Cy+AdIL-12 was the induction of a specific immune response mediated by cytotoxic T lymphocytes. Our current aims were to evaluate the effects of 4-methylumbelliferone (4Mu), a selective inhibitor of hyaluronan (HA) synthesis, on tumor microenvironment (TME) and to investigate how 4Mu affects the therapeutic efficacy of Cy+AdIL-12. The results showed that 4Mu significantly reduced the amount of tumoral HA leading to a significant decrease in tumor interstitial pressure (TIP). As a consequence, tumor perfusion was improved allowing an increased adenoviral transgene expression. In addition, treatment with 4Mu boosted the number of cytotoxic T lymphocytes that reach the tumor after adoptive transfer resulting in a potent inhibition of tumor growth. Importantly, we observed complete tumor regression in 75% of mice when 4Mu was administrated in combination with Cy+AdIL-12. The triple combination 4Mu+Cy+AdIL-12 also induced a shift toward antiangiogenic factors production in tumor milieu. Our results showed that TME remodeling is an interesting strategy to increase the efficacy of anticancer immunotherapies based on gene and/or cell therapy.
Collapse
Affiliation(s)
- Mariana Malvicini
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina.,CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Esteban Fiore
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina
| | - Valentina Ghiaccio
- Dipartamento di Sanità Pubblica, Medicina Clinica e Moleculare, Università degli studi di Cagliari, Sardegna, Italia
| | - Flavia Piccioni
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina
| | - Miguel Rizzo
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina
| | - Lucila Olmedo Bonadeo
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina
| | - Mariana García
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina.,CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Marcelo Rodríguez
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina
| | - Juan Bayo
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina
| | - Estanislao Peixoto
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina
| | - Catalina Atorrasagasti
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina.,CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Laura Alaniz
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina.,CIT NOBA, Universidad Nacional del Noroeste, Junín, Buenos Aires, Argentina
| | - Jorge Aquino
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina.,CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Pablo Matar
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina.,Institute of Experimental Genetics, School of Medical Sciences, Universidad Nacional de Rosario, Rosario, Argentina
| | - Guillermo Mazzolini
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina.,CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| |
Collapse
|
13
|
Broggini-Tenzer A, Sharma A, Nytko KJ, Bender S, Vuong V, Orlowski K, Hug D, O'Reilly T, Pruschy M. Combined treatment strategies for microtubule stabilizing agent-resistant tumors. J Natl Cancer Inst 2015; 107:dju504. [PMID: 25694444 DOI: 10.1093/jnci/dju504] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Resistance to microtubule-stabilizing agents is a major hurdle for successful cancer therapy. We investigated combined treatment of microtubule-stabilizing agents (MSAs) with inhibitors of angiogenesis to overcome MSA resistance. METHODS Treatment regimens of clinically relevant MSAs (patupilone and paclitaxel) and antiangiogenic agents (everolimus and bevacizumab) were investigated in genetically defined MSA-resistant lung (A549EpoB40) and colon adenocarcinoma (SW480) tumor xenografts in nude mice (CD1-Foxn1<nu>, ICRnu; 5-14 per group). Tumor growth delays were calculated by Kaplan-Meier analysis with Holm-Sidak tests. All statistical tests were two-sided. RESULTS Inhibition of mTOR-kinase by everolimus only minimally reduced the proliferative activity of β tubulin-mutated lung adenocarcinoma cells alone and in combination with the MSA patupilone, but everolimus inhibited expression and secretion of vascular endothelial growth factor (VEGF) from these cells. mTOR-kinase inhibition strongly sensitized tumor xenografts derived from these otherwise MSA-resistant tumor cells to patupilone. Tumors treated with the combined modality of everolimus and patupilone had statistically significantly reduced tumor volume and stronger tumor growth delay (16.2 ± 1.01 days) than control- (7.7 ± 0.3 days, P = .004), patupilone- (10 ± 0.97 days, P = .009), and everolimus-treated (10.6 ± 1.4 days, P = .014) tumors. A combined treatment modality with bevacizumab also resensitized this MSA-refractory tumor model to patupilone. Treatment combination also strongly reduced microvessel density, corroborating the relevance of VEGF targeting for the known antivasculature-directed potency of MSA alone in MSA-sensitive tumor models. Resensitization to MSAs was also probed in P glycoprotein-overexpressing SW480-derived tumor xenografts. Different bevacizumab regimens also sensitized this otherwise-resistant tumor model to clinically relevant MSA paclitaxel. CONCLUSIONS A treatment combination of MSAs with antiangiogenic agents is potent to overcome tumor cell-linked MSA resistance and should be considered as strategy for MSA-refractory tumor entities.
Collapse
Affiliation(s)
- Angela Broggini-Tenzer
- Laboratory for Molecular Radiobiology, Radiation Oncology, University Hospital Zurich, Zurich, Switzerland (ABT, AS, KJN, SB, VV, KO, DH, TOR, MP); Department of Dermatology, University Hospital Zurich, Zurich, Switzerland (DH); Clinical Research Priority Program Tumor Oxygenation, University Hospital Zurich, Zurich, Switzerland (KJN, MP).Current affiliation: Novartis Pharma Switzerland (KO)
| | - Ashish Sharma
- Laboratory for Molecular Radiobiology, Radiation Oncology, University Hospital Zurich, Zurich, Switzerland (ABT, AS, KJN, SB, VV, KO, DH, TOR, MP); Department of Dermatology, University Hospital Zurich, Zurich, Switzerland (DH); Clinical Research Priority Program Tumor Oxygenation, University Hospital Zurich, Zurich, Switzerland (KJN, MP).Current affiliation: Novartis Pharma Switzerland (KO)
| | - Katarzyna J Nytko
- Laboratory for Molecular Radiobiology, Radiation Oncology, University Hospital Zurich, Zurich, Switzerland (ABT, AS, KJN, SB, VV, KO, DH, TOR, MP); Department of Dermatology, University Hospital Zurich, Zurich, Switzerland (DH); Clinical Research Priority Program Tumor Oxygenation, University Hospital Zurich, Zurich, Switzerland (KJN, MP).Current affiliation: Novartis Pharma Switzerland (KO)
| | - Sabine Bender
- Laboratory for Molecular Radiobiology, Radiation Oncology, University Hospital Zurich, Zurich, Switzerland (ABT, AS, KJN, SB, VV, KO, DH, TOR, MP); Department of Dermatology, University Hospital Zurich, Zurich, Switzerland (DH); Clinical Research Priority Program Tumor Oxygenation, University Hospital Zurich, Zurich, Switzerland (KJN, MP).Current affiliation: Novartis Pharma Switzerland (KO)
| | - Van Vuong
- Laboratory for Molecular Radiobiology, Radiation Oncology, University Hospital Zurich, Zurich, Switzerland (ABT, AS, KJN, SB, VV, KO, DH, TOR, MP); Department of Dermatology, University Hospital Zurich, Zurich, Switzerland (DH); Clinical Research Priority Program Tumor Oxygenation, University Hospital Zurich, Zurich, Switzerland (KJN, MP).Current affiliation: Novartis Pharma Switzerland (KO)
| | - Katrin Orlowski
- Laboratory for Molecular Radiobiology, Radiation Oncology, University Hospital Zurich, Zurich, Switzerland (ABT, AS, KJN, SB, VV, KO, DH, TOR, MP); Department of Dermatology, University Hospital Zurich, Zurich, Switzerland (DH); Clinical Research Priority Program Tumor Oxygenation, University Hospital Zurich, Zurich, Switzerland (KJN, MP).Current affiliation: Novartis Pharma Switzerland (KO)
| | - Daniel Hug
- Laboratory for Molecular Radiobiology, Radiation Oncology, University Hospital Zurich, Zurich, Switzerland (ABT, AS, KJN, SB, VV, KO, DH, TOR, MP); Department of Dermatology, University Hospital Zurich, Zurich, Switzerland (DH); Clinical Research Priority Program Tumor Oxygenation, University Hospital Zurich, Zurich, Switzerland (KJN, MP).Current affiliation: Novartis Pharma Switzerland (KO)
| | - Terence O'Reilly
- Laboratory for Molecular Radiobiology, Radiation Oncology, University Hospital Zurich, Zurich, Switzerland (ABT, AS, KJN, SB, VV, KO, DH, TOR, MP); Department of Dermatology, University Hospital Zurich, Zurich, Switzerland (DH); Clinical Research Priority Program Tumor Oxygenation, University Hospital Zurich, Zurich, Switzerland (KJN, MP).Current affiliation: Novartis Pharma Switzerland (KO)
| | - Martin Pruschy
- Laboratory for Molecular Radiobiology, Radiation Oncology, University Hospital Zurich, Zurich, Switzerland (ABT, AS, KJN, SB, VV, KO, DH, TOR, MP); Department of Dermatology, University Hospital Zurich, Zurich, Switzerland (DH); Clinical Research Priority Program Tumor Oxygenation, University Hospital Zurich, Zurich, Switzerland (KJN, MP).Current affiliation: Novartis Pharma Switzerland (KO).
| |
Collapse
|
14
|
Abstract
Epothilones A and B are naturally occurring microtubule stabilizers with nanomolar or even sub-nanomolar activity against human cancer cells in vitro and potent in vivo antitumor activity against multidrug-resistant tumors. Over the last decade, ten epothilonetype agents have entered clinical trials in humans; of these, the epothilone B lactam ixabepilone (BMS-247550; Ixempra®) was approved by the FDA for breast cancer treatment in 2007. Numerous synthetic and semisynthetic analogs of epothilones have been prepared and their in vitro and (in selected cases) in vivo biological activity has been determined, producing a wealth of SAR information on this compound family. This chapter will provide a brief summary of the in vitro and in vivo biological properties of epothilone B (Epo B). The major part of the discussion will then be organized around those epothilone analogs that have entered clinical development. For each analog the underlying synthetic chemistry and the most important preclinical features will be reviewed, together with the properties of some important related structures.
Collapse
Affiliation(s)
- Raphael Schiess
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zürich HCI H405, Vladimir-Prelog-Weg 4 CH-8093 Zürich Switzerland
| | - Karl-Heinz Altmann
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zürich HCI H405, Vladimir-Prelog-Weg 4 CH-8093 Zürich Switzerland
| |
Collapse
|
15
|
Emblem KE, Farrar CT, Gerstner ER, Batchelor TT, Borra RJH, Rosen BR, Sorensen AG, Jain RK. Vessel caliber--a potential MRI biomarker of tumour response in clinical trials. Nat Rev Clin Oncol 2014; 11:566-84. [PMID: 25113840 PMCID: PMC4445139 DOI: 10.1038/nrclinonc.2014.126] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
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.
Collapse
Affiliation(s)
- Kyrre E Emblem
- The Intervention Centre, Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway
| | - Christian T Farrar
- Department of Radiology and Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Elizabeth R Gerstner
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, 100 Blossom Street, Boston, MA 02114, USA
| | - Tracy T Batchelor
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, 100 Blossom Street, Boston, MA 02114, USA
| | - Ronald J H Borra
- Department of Radiology and Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Bruce R Rosen
- Department of Radiology and Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - A Gregory Sorensen
- Siemens Healthcare Health Services, 51 Valley Stream Parkway, Malvern, PA 19355, USA
| | - Rakesh K Jain
- Edwin L. Steele Laboratory of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, 100 Blossom Street, Boston, MA 02114, USA
| |
Collapse
|
16
|
Troprès I, Pannetier N, Grand S, Lemasson B, Moisan A, Péoc'h M, Rémy C, Barbier EL. Imaging the microvessel caliber and density: Principles and applications of microvascular MRI. Magn Reson Med 2014; 73:325-41. [DOI: 10.1002/mrm.25396] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 07/08/2014] [Accepted: 07/11/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Irène Troprès
- IRMaGe; Université Grenoble Alpes; Grenoble France
- UMS 3552; CNRS; Grenoble France
- US 017; INSERM; Grenoble France
- IRMaGe, Hôpital Michallon; Centre Hospitalier Universitaire de Grenoble; Grenoble France
- Université Joseph Fourier; Grenoble Institut des Neurosciences; Grenoble France. INSERM; U836 Grenoble France
| | - Nicolas Pannetier
- Université Joseph Fourier; Grenoble Institut des Neurosciences; Grenoble France
- INSERM; U836 Grenoble France
| | - Sylvie Grand
- Université Joseph Fourier; Grenoble Institut des Neurosciences; Grenoble France
- INSERM; U836 Grenoble France
- CLUNI, Hôpital Michallon; Centre Hospitalier Universitaire de Grenoble; Grenoble France
| | - Benjamin Lemasson
- Université Joseph Fourier; Grenoble Institut des Neurosciences; Grenoble France
- INSERM; U836 Grenoble France
| | - Anaïck Moisan
- Université Joseph Fourier; Grenoble Institut des Neurosciences; Grenoble France
- INSERM; U836 Grenoble France
| | - Michel Péoc'h
- Service d'anatomo-pathologie; Centre Hospitalier Universitaire de Saint Etienne; Saint-Etienne France
- EA 2521; Université Jean Monnet; Saint-Etienne France
| | - Chantal Rémy
- Université Joseph Fourier; Grenoble Institut des Neurosciences; Grenoble France
- INSERM; U836 Grenoble France
| | - Emmanuel L. Barbier
- Université Joseph Fourier; Grenoble Institut des Neurosciences; Grenoble France
- INSERM; U836 Grenoble France
| |
Collapse
|
17
|
Ariffin AB, Forde PF, Jahangeer S, Soden DM, Hinchion J. Releasing Pressure in Tumors: What Do We Know So Far and Where Do We Go from Here? A Review. Cancer Res 2014; 74:2655-62. [DOI: 10.1158/0008-5472.can-13-3696] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
18
|
Weidensteiner C, Allegrini PR, Sticker-Jantscheff M, Romanet V, Ferretti S, McSheehy PMJ. Tumour T1 changes in vivo are highly predictive of response to chemotherapy and reflect the number of viable tumour cells--a preclinical MR study in mice. BMC Cancer 2014; 14:88. [PMID: 24528602 PMCID: PMC3932835 DOI: 10.1186/1471-2407-14-88] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 02/11/2014] [Indexed: 11/10/2022] Open
Abstract
Background Effective chemotherapy rapidly reduces the spin–lattice relaxation of water protons (T1) in solid tumours and this change (ΔT1) often precedes and strongly correlates with the eventual change in tumour volume (TVol). To understand the biological nature of ΔT1, we have performed studies in vivo and ex vivo with the allosteric mTOR inhibitor, everolimus. Methods Mice bearing RIF-1 tumours were studied by magnetic resonance imaging (MRI) to determine TVol and T1, and MR spectroscopy (MRS) to determine levels of the proliferation marker choline and levels of lipid apoptosis markers, prior to and 5 days (endpoint) after daily treatment with vehicle or everolimus (10 mg/kg). At the endpoint, tumours were ablated and an entire section analysed for cellular and necrotic quantification and staining for the proliferation antigen Ki67 and cleaved-caspase-3 as a measure of apoptosis. The number of blood-vessels (BV) was evaluated by CD31 staining. Mice bearing B16/BL6 melanoma tumours were studied by MRI to determine T1 under similar everolimus treatment. At the endpoint, cell bioluminescence of the tumours was measured ex vivo. Results Everolimus blocked RIF-1 tumour growth and significantly reduced tumour T1 and total choline (Cho) levels, and increased polyunsaturated fatty-acids which are markers of apoptosis. Immunohistochemistry showed that everolimus reduced the %Ki67+ cells but did not affect caspase-3 apoptosis, necrosis, BV-number or cell density. The change in T1 (ΔT1) correlated strongly with the changes in TVol and Cho and %Ki67+. In B16/BL6 tumours, everolimus also decreased T1 and this correlated with cell bioluminescence; another marker of cell viability. Receiver-operating-characteristic curves (ROC) for everolimus on RIF-1 tumours showed that ΔT1 had very high levels of sensitivity and specificity (ROCAUC = 0.84) and this was confirmed for the cytotoxic patupilone in the same tumour model (ROCAUC = 0.97). Conclusion These studies suggest that ΔT1 is not a measure of cell density but reflects the decreased number of remaining viable and proliferating tumour cells due to perhaps cell and tissue destruction releasing proteins and/or metals that cause T1 relaxation. ΔT1 is a highly sensitive and specific predictor of response. This MRI method provides the opportunity to stratify a patient population during tumour therapy in the clinic.
Collapse
|
19
|
Scherzinger-Laude K, Schönherr C, Lewrick F, Süss R, Francese G, Rössler J. Treatment of neuroblastoma and rhabdomyosarcoma using RGD-modified liposomal formulations of patupilone (EPO906). Int J Nanomedicine 2013; 8:2197-211. [PMID: 23818777 PMCID: PMC3693827 DOI: 10.2147/ijn.s44025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Background Patupilone (EPO906) is a microtubule stabilizer with a potent antitumor effect. Integrin αVβ3-binding (RGD) liposomes were loaded with EPO906, and their antitumor efficacy was evaluated in two pediatric tumor models, ie, neuroblastoma and rhabdomyosarcoma. Methods Integrin αVβ3 gene expression, RGD-liposome cellular association, and the effect of EPO906 and liposomal formulations of EPO906 on cell viability were assessed in vitro in human umbilical vein endothelial cells (HUVEC), in the RH-30 rhabdomyosarcoma cell line, and in the Kelly neuroblastoma cell line. In vivo, mice bearing neuroblastoma or rhabdomyosarcoma tumors were treated with EPO906, EPO906-liposomes, or EPO906-RGD-liposomes. Tumor growth, cumulative survival, and toxicity were monitored. Results Integrin αVβ3 was highly expressed in HUVEC and RH-30, but not in Kelly cells. Accordingly, RGD-liposomes were highly associated with HUVEC and RH-30 cells in vitro, but not with the Kelly cells. EPO906 and its liposomal formulations inhibited HUVEC, RH-30, and Kelly cell viability to the same extent. In vivo, EPO906 1.5 mg/kg and liposomal EPO906 potently inhibited tumor growth in both xenograft models without triggering major toxicity. At this dose, liposomal EPO906 did not enhance the antitumor effect of EPO906 in neuroblastoma, but tended to have an increased antitumor effect in rhabdomyosarcoma. Using a lower dose of EPO906-RGD-liposomes significantly enhanced cumulative survival in rhabdomyosarcoma compared with EPO906 alone. Conclusion EPO906 shows a strong antitumor effect in neuroblastoma and rhabdomyosarcoma, without triggering major side effects. Its liposomal encapsulation does not alter its activity, and enhances cumulative survival when EPO906-RGD-liposomes are used at low dose in rhabdomyosarcoma.
Collapse
Affiliation(s)
- Karine Scherzinger-Laude
- Clinic IV, Pediatric Hematology and Oncology, Center of Pediatrics and Adolescent Medicine, University Medical Hospital Freiburg, Germany
| | | | | | | | | | | |
Collapse
|
20
|
Furmanova-Hollenstein P, Broggini-Tenzer A, Eggel M, Millard AL, Pruschy M. The microtubule stabilizer patupilone counteracts ionizing radiation-induced matrix metalloproteinase activity and tumor cell invasion. Radiat Oncol 2013; 8:105. [PMID: 23631818 PMCID: PMC3661365 DOI: 10.1186/1748-717x-8-105] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 04/23/2013] [Indexed: 02/06/2023] Open
Abstract
Background Ionizing radiation (IR) in combination with microtubule stabilizing agents (MSA) is a promising combined treatment modality. Supra-additive treatment responses might result from direct tumor cell killing and cooperative indirect, tumor cell-mediated effects on the tumor microenvironment. Here we investigated deregulation of matrix metalloproteinase (MMP) activity, as an important component of the tumor microenvironment, by the combined treatment modality of IR with the clinically relevant MSA patupilone. Methods Expression, secretion and activity of MMPs and related tissue inhibitors of metalloproteinases (TIMPs) were determined in cell extracts and conditioned media derived from human fibrosarcoma HT1080 and human glioblastoma U251 tumor cells in response to treatment with IR and the MSA patupilone. Treatment-dependent changes of the invasive capacities of these tumor cell lines were analysed using a Transwell invasion assay. Control experiments were performed using TIMP-directed siRNA and TIMP-directed inhibitory antibodies. Results Enzymatic activity of secreted MMPs was determined after treatment with patupilone and irradiation in the human fibrosarcoma HT1080 and the human glioblastoma U251 tumor cell line. IR enhanced the activity of secreted MMPs up to 2-fold and cellular pretreatment with low dose patupilone (0.05-0.2 nM) counteracted specifically the IR-induced MMP activity. The cell invasive capacity of HT1080 and U251 cells was increased after irradiation with 2 Gy by 30% and 50%, respectively, and patupilone treatment completely abrogated IR-induced cell invasion. Patupilone did not alter the level of MMP expression, but interestingly, the protein level of secreted TIMP-1 and TIMP-2 was lower after combined treatment than after irradiation treatment alone. Furthermore, siRNA depletion of TIMP-1 or TIMP-2 prevented IR-mediated induction of MMP activity and cell invasion. Conclusions These results indicate that patupilone counteracts an IR-induced MMP activation process by the reduction of secreted TIMP-1 and TIMP-2 proteins, which are required for activation of MMPs. Since IR-induced MMP activity could contribute to tumor progression, treatment combination of IR with patupilone might be of great clinical benefit for tumor therapy.
Collapse
|
21
|
Orlowski K, Rohrer Bley C, Zimmermann M, Vuong V, Hug D, Soltermann A, Broggini-Tenzer A, Pruschy M. Dynamics of tumor hypoxia in response to patupilone and ionizing radiation. PLoS One 2012; 7:e51476. [PMID: 23251549 PMCID: PMC3519688 DOI: 10.1371/journal.pone.0051476] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 11/07/2012] [Indexed: 12/29/2022] Open
Abstract
Tumor hypoxia is one of the most important parameters that determines treatment sensitivity and is mainly due to insufficient tumor angiogenesis. However, the local oxygen concentration in a tumor can also be shifted in response to different treatment modalities such as cytotoxic agents or ionizing radiation. Thus, combined treatment modalities including microtubule stabilizing agents could create an additional challenge for an effective treatment response due to treatment-induced shifts in tumor oxygenation. Tumor hypoxia was probed over a prolonged observation period in response to treatment with different cytotoxic agents, using a non-invasive bioluminescent ODD-Luc reporter system, in which part of the oxygen-dependent degradation (ODD) domain of HIF-1α is fused to luciferase. As demonstrated in vitro, this system not only detects hypoxia at an ambient oxygen concentration of 1% O2, but also discriminates low oxygen concentrations in the range from 0.2 to 1% O2. Treatment of A549 lung adenocarcinoma-derived tumor xenografts with the microtubule stabilizing agent patupilone resulted in a prolonged increase in tumor hypoxia, which could be used as marker for its antitumoral treatment response, while irradiation did not induce detectable changes in tumor hypoxia. Furthermore, despite patupilone-induced hypoxia, the potency of ionizing radiation (IR) was not reduced as part of a concomitant or adjuvant combined treatment modality.
Collapse
Affiliation(s)
- Katrin Orlowski
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
- KFSP Tumor Oxygenation, University of Zurich, Switzerland
| | - Carla Rohrer Bley
- Division of Radiation Oncology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Martina Zimmermann
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Van Vuong
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Daniel Hug
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Alex Soltermann
- Department of Pathology, University Hospital Zurich, Zurich, Switzerland
| | | | - Martin Pruschy
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
- KFSP Tumor Oxygenation, University of Zurich, Switzerland
- * E-mail:
| |
Collapse
|
22
|
Kim E, Cebulla J, Ward BD, Rhie K, Zhang J, Pathak AP. Assessing breast cancer angiogenesis in vivo: which susceptibility contrast MRI biomarkers are relevant? Magn Reson Med 2012; 70:1106-16. [PMID: 23225578 DOI: 10.1002/mrm.24530] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 09/20/2012] [Accepted: 09/22/2012] [Indexed: 01/17/2023]
Abstract
PURPOSE There is an impending need for noninvasive biomarkers of breast cancer angiogenesis to evaluate the efficacy of new anti-angiogenic therapies in vivo. The purpose of this study was to systematically evaluate the sensitivity of in vivo steady-state susceptibility contrast-MRI biomarkers of angiogenesis in a human breast cancer model. METHODS Orthotopic MDA-MB-231 human breast cancer xenografts were imaged by steady-state susceptibility contrast-MRI at post-inoculation week 3 and post-inoculation week 5, followed by ex vivo whole tumor 3D micro-CT angiography. "Absolute" (i.e., measures of vascular morphology in appropriate units) and "relative" (i.e., proportional to measures of vascular morphology) MRI biomarkers of tumor blood volume, vessel size, and vessel density were computed and their ability to predict the corresponding micro-CT analogs assessed using cross-validation analysis. RESULTS All MRI biomarkers significantly correlated with their micro-CT analogs and were sensitive to the micro-CT-measured decreases in tumor blood volume and vessel density from post-inoculation week 3 to post-inoculation week 5. However, cross-validation analysis revealed there was no significant difference between the predictive accuracy of "absolute" and "relative" biomarkers. CONCLUSION As "relative" biomarkers are more easily computed from steady-state susceptibility contrast-MRI (i.e., without additional MRI measurements) than "absolute" biomarkers, it makes them promising candidates for assessing breast cancer angiogenesis in vivo.
Collapse
Affiliation(s)
- Eugene Kim
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | |
Collapse
|
23
|
Meier V, Geigy C, Grosse N, McSheehy P, Rohrer Bley C. Use of epothilone B (patupilone) in refractory lymphoma and advanced solid tumors in dogs. J Vet Intern Med 2012. [PMID: 23205945 DOI: 10.1111/jvim.12019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND The epothilones are microtubule-stabilizing agents with promising antitumor effect in refractory and metastatic tumors in humans. The toxicity profile is considered more favorable than in taxanes. The safety of epothilone B (patupilone) has not been evaluated in tumor-bearing dogs. OBJECTIVES To evaluate the inhibition of proliferation in canine tumor cells after patupilone treatment. To assess toxicity profile and maximally tolerated dose of patupilone in dogs with refractory tumors. ANIMALS Twenty client-owned dogs with various malignancies. METHODS Prospective clinical study. The inhibition of proliferation was assessed with a proliferation assay in vitro in canine hemangiosarcoma and lymphoma cell lines. Dogs received patupilone IV once a week for 2 treatments (= 1 treatment cycle). Dose was escalated with 3 dogs per cohort and 20% increments. Adverse effects were graded according to the VCOG-CTCAE v1.0. RESULTS Both canine cell lines were sensitive to patupilone with approximately 50% decrease in proliferative activity at 0.2-1 nM. In vivo, dose-limiting adverse effects occurred at 3.3 mg/m(2); main adverse effects were diarrhea, anorexia, vomiting, and nausea. Neither neutropenia nor peripheral neuropathy was observed. Maximally tolerated dose for 2 patupilone administrations once weekly IV is 2.76 mg/m(2). Three per 11 dogs receiving more than 1 treatment cycle showed partial remission in the short period of observation. CONCLUSIONS AND CLINICAL IMPORTANCE Canine tumor cells show inhibition of proliferation to patupilone in vitro. Clinically, a dose of 2.76 mg/m(2) IV is well tolerated in dogs with spontaneously occurring tumors.
Collapse
Affiliation(s)
- V Meier
- Division of Radiation Oncology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.
| | | | | | | | | |
Collapse
|
24
|
Burrell JS, Bradley RS, Walker-Samuel S, Jamin Y, Baker LCJ, Boult JKR, Withers PJ, Halliday J, Waterton JC, Robinson SP. MRI measurements of vessel calibre in tumour xenografts: comparison with vascular corrosion casting. Microvasc Res 2012; 84:323-9. [PMID: 22921880 PMCID: PMC3657196 DOI: 10.1016/j.mvr.2012.08.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 08/06/2012] [Accepted: 08/09/2012] [Indexed: 02/08/2023]
Abstract
Vessel size index (Rv, μm) has been proposed as a quantitative magnetic resonance imaging (MRI) derived imaging biomarker in oncology, for the non-invasive assessment of tumour blood vessel architecture and vascular targeted therapies. Appropriate pre-clinical evaluation of Rv in animal tumour models will improve the interpretation and guide the introduction of the biomarker into clinical studies. The objective of this study was to compare Rv measured in vivo with vessel size measurements from high-resolution X-ray computed tomography (μCT) of vascular corrosion casts measured post mortem from the same tumours, with and without vascular targeted therapy. MRI measurements were first acquired from subcutaneous SW1222 colorectal xenografts in mice following treatment with 0 (n = 6), 30 (n = 6) or 200 mg/kg (n = 3) of the vascular disrupting agent ZD6126. The mice were then immediately infused with a low viscosity resin and, following polymerisation and maceration of surrounding tissues, the resulting tumour vascular casts were dissected and subsequently imaged using an optimised μCT imaging approach. Vessel diameters were not measurable by μCT in the 200 mg/kg group as the high dose of ZD6126 precluded delivery of the resin to the tumour vascular bed. The mean Rv for the three treatment groups was 24, 23 and 23.5 μm respectively; the corresponding μCT measurements from corrosion casts from the 0 and 30 mg/kg cohorts were 25 and 28 μm. The strong association between the in vivo MRI and post mortem μCT values supports the use of Rv as an imaging biomarker in clinical trials of investigational vascular targeted therapies.
Collapse
Affiliation(s)
- Jake S Burrell
- CR-UK & EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, 15 Cotswold Road Sutton, Surrey, SM2 5NG, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Chung SH, Yu H, Su MY, Cerussi AE, Tromberg BJ. Molecular imaging of water binding state and diffusion in breast cancer using diffuse optical spectroscopy and diffusion weighted MRI. JOURNAL OF BIOMEDICAL OPTICS 2012; 17:071304. [PMID: 22894465 PMCID: PMC3381027 DOI: 10.1117/1.jbo.17.7.071304] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Tissue water content and molecular microenvironment can provide important intrinsic contrast for cancer imaging. In this work, we examine the relationship between water optical spectroscopic features related to binding state and magnetic resonance imaging (MRI)-measured water diffusion dynamics. Broadband diffuse optical spectroscopic imaging (DOSI) and MR images were obtained from eight patients with locally-advanced infiltrating ductal carcinomas (tumor size=5.5 ± 3.2 cm). A DOSI-derived bound water index (BWI) was compared to the apparent diffusion coefficient (ADC) of diffusion weighted (DW) MRI. BWI and ADC were positively correlated (R=0.90, p-value=0.003) and BWI and ADC both decreased as the bulk water content increased (R=-0.81 and -0.89, respectively). BWI correlated inversely with tumor size (R=-0.85, p-value=0.008). Our results suggest underlying sensitivity differences between BWI and ADC to water in different tissue compartments (e.g., extracellular vs cellular). These data highlight the potential complementary role of DOSI and DW-MRI in providing detailed information on the molecular disposition of water in breast tumors. Because DOSI is a portable technology that can be used at the bedside, BWI may provide a low-cost measure of tissue water properties related to breast cancer biology.
Collapse
Affiliation(s)
- So Hyun Chung
- University of Pennsylvania, Department of Physics and Astronomy, 209 South 33rd Street, Philadelphia, Pennsylvania 19104-6396
- Address all correspondence to: So Hyun Chung, University of Pennsylvania, Department of Physics and Astronomy, 209 South 33rd Street, Philadelphia, Pennsylvania 19104-6396. Tel: +215-898-6833; Fax: +215-573-6391; E-mail:
| | - Hon Yu
- University of California, Irvine, Department of Radiological Sciences and Tu & Yuen Center for Functional Onco Imaging, 164 Irvine Hall, Irvine, California 92697
| | - Min-Ying Su
- University of California, Irvine, Department of Radiological Sciences and Tu & Yuen Center for Functional Onco Imaging, 164 Irvine Hall, Irvine, California 92697
| | - Albert E. Cerussi
- University of California, Irvine, Beckman Laser Institute and Medical Clinic, 1002 Health Sciences Road, Irvine, California 92612
| | - Bruce J. Tromberg
- University of California, Irvine, Beckman Laser Institute and Medical Clinic, 1002 Health Sciences Road, Irvine, California 92612
| |
Collapse
|
26
|
Oehler C, Frei K, Rushing EJ, McSheehy PM, Weber D, Allegrini PR, Weniger D, Lütolf UM, Knuth A, Yonekawa Y, Barath K, Broggini-Tenzer A, Pruschy M, Hofer S. Patupilone (Epothilone B) for Recurrent Glioblastoma: Clinical Outcome and Translational Analysis of a Single-Institution Phase I/II Trial. Oncology 2012; 83:1-9. [DOI: 10.1159/000339152] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 04/25/2012] [Indexed: 12/21/2022]
|
27
|
Rohrer Bley C, Furmanova P, Orlowski K, Grosse N, Broggini-Tenzer A, McSheehy PMJ, Pruschy M. Microtubule stabilising agents and ionising radiation: multiple exploitable mechanisms for combined treatment. Eur J Cancer 2012; 49:245-53. [PMID: 22683167 DOI: 10.1016/j.ejca.2012.05.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 05/07/2012] [Accepted: 05/08/2012] [Indexed: 12/18/2022]
Abstract
Combined radiochemotherapy treatment modalities are in use for many indications and therefore of high interest. Even though a combined modality in clinical use is often driven by pragmatic aspects, mechanistic preclinical-based concepts of interaction are of importance in order to translate and implement an optimal combination and scheduling of two modalities into the clinics. The use of microtubule stabilising agents is a promising strategy for anti-cancer therapy as a part of combined treatment modality with ionising radiation. Traditionally, microtubule targeting agents are classified as cytotoxic chemotherapeutics and are mostly used in a maximally tolerated dose regimen. Apart from direct cytotoxicity and similar to mechanisms of molecular targeting agents, microtubule stabilising agents interfere with multiple cellular processes, which can be exploited as part of combined treatment modalities. Recent preclinical investigations on the combination of ionising radiation and microtubule stabilising agents reveal new mechanistic interactions on the cellular and tumour level and elucidate the supra-additive tumour response observed particularly in vivo. The major focus on the mechanism of interaction was primarily based on radiosensitisation due to cell cycle arrest in the most radiosensitive G2/M-phase of the cell cycle. However, other mechanisms of interaction such as reoxygenation and direct as well as indirect endothelial damage have also been identified. In this review we summarise and allocate additive and synergistic effects induced by the combined treatment of clinically relevant microtubule stabilising agents and ionising radiation along a described radiobiological framework encompassing distinct mechanisms relevant for exploiting the combination of drugs and ionising radiation.
Collapse
Affiliation(s)
- Carla Rohrer Bley
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
28
|
Baumgart T, Klautke G, Kriesen S, Kuznetsov SA, Weiss DG, Fietkau R, Hildebrandt G, Manda K. Radiosensitizing effect of epothilone B on human epithelial cancer cells. Strahlenther Onkol 2012; 188:177-84. [PMID: 22234539 DOI: 10.1007/s00066-011-0029-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 10/20/2011] [Indexed: 01/09/2023]
Abstract
BACKGROUND A combined modality treatment employing radiation and chemotherapy plays a central role in the management of solid tumors. In our study, we examined the cytotoxic and radiosensitive effect of the microtubule stabilizer epothilone B on two human epithelial tumor cell lines in vitro and its influence on the microtubule assembly. METHODS Cancer cells were treated with epothilone B in proliferation assays and in combination with radiation in colony-forming assays. For the analysis of ionizing radiation-induced DNA damage and the influence of the drug on its repair a γH2AX foci assay was used. To determine the effect of epothilone B on the microtubule assembly in cells and on purified tubulin, immunofluorescence staining and tubulin polymerization assay, respectively, were conducted. RESULTS Epothilone B induced a concentration- and application-dependent antiproliferative effect on the cells, with IC(50) values in the low nanomolar range. Colony forming assays showed a synergistic radiosensitive effect on both cell lines which was dependent on incubation time and applied concentration of epothilone B. The γH2AX assays demonstrated that ionizing radiation combined with the drug resulted in a concentration-dependent increase in the number of double-strand breaks and suggested a reduction in DNA repair capacity. Epothilone B produced enhanced microtubule bundling and abnormal spindle formation as revealed by immunofluorescence microscopy and caused microtubule formation from purified tubulin. CONCLUSION The results of this study showed that epothilone B displays cytotoxic antitumor activity at low nanomolar concentrations and also enhances the radiation response in the tumor cells tested; this may be induced by a reduced DNA repair capacity triggered by epothilone B. It was also demonstrated that epothilone B in fact targets microtubules in a more effective manner than paclitaxel.
Collapse
Affiliation(s)
- T Baumgart
- Department of Radiotherapy and Radiation Oncology, University of Rostock, Südring 75, Rostock, Germany
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Kornfeld S, Goupille C, Vibet S, Chevalier S, Pinet A, Lebeau J, Tranquart F, Bougnoux P, Martel E, Maurin A, Richard S, Champeroux P, Mahéo K. Reducing endothelial NOS activation and interstitial fluid pressure with n-3 PUFA offset tumor chemoresistance. Carcinogenesis 2011; 33:260-7. [PMID: 22114075 DOI: 10.1093/carcin/bgr274] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The aim of this study was to determine how n-3 polyunsaturated fatty acid (PUFAs) counteracted tumor chemoresistance by restoring a functional vascularization. Rats with chemically induced mammary tumors were divided into two nutritional groups: a control group and a group fed with an n-3 PUFA-enriched diet. Both groups were treated with docetaxel. Functional vascular parameters (ultrasounds, interstitial fluid pressure) were determined for both nutritional groups before (W(0)) and during docetaxel treatment [every 2 h up to 1 week (W(+1)) for interstitial fluid pressure, at W(+1) for Evans blue extravasation and at W(+2) and W(+6) for ultrasounds]. In vitro n-3 PUFA-induced changes in endothelial cell migration, permeability and phosphorylation of endothelial nitric oxide synthase were evaluated using human umbilical vein endothelial cells. Whereas docetaxel stabilized tumor growth in the rat control group, it induced a 50% tumor regression in the n-3 PUFA group. Ultrasounds parameters were consistently lower in the n-3 PUFA group at all time points measured, down to ∼50% at W(+6). A single dose of docetaxel in the n-3 PUFA group markedly reduced interstitial fluid pressure from 2 h after injection up to W(+1) when Evans blue extravasation was increased by 3-fold. A decreased activation of endothelial nitric oxide synthase in tumors of the n-3 PUFA group, and in human umbilical vein endothelial cell cultured with n-3 PUFA, points toward a PUFA-induced disruption of nitric oxide signaling pathway. This normalization of tumor vasculature functions under n-3 PUFA diet indicates that such a supplementation, by improving drug delivery in mammary tumors, could be a complementary clinical strategy to decrease anticancer drug resistance.
Collapse
Affiliation(s)
- Sophie Kornfeld
- INSERM U921 Nutrition, Croissance et Cancer Université François Rabelais, Tours, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Rohrer Bley C, Orlowski K, Furmanova P, McSheehy PM, Pruschy M. Regulation of VEGF-expression by patupilone and ionizing radiation in lung adenocarcinoma cells. Lung Cancer 2011; 73:294-301. [DOI: 10.1016/j.lungcan.2011.01.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 12/23/2010] [Accepted: 01/18/2011] [Indexed: 10/18/2022]
|
31
|
Walker-Samuel S, Boult JKR, McPhail LD, Box G, Eccles SA, Robinson SP. Non-invasive in vivo imaging of vessel calibre in orthotopic prostate tumour xenografts. Int J Cancer 2011; 130:1284-93. [PMID: 21469141 DOI: 10.1002/ijc.26112] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Accepted: 03/25/2011] [Indexed: 12/16/2022]
Abstract
Susceptibility contrast magnetic resonance imaging (MRI), utilising ultrasmall superparamagnetic iron oxide (USPIO) particles, was evaluated for the quantitation of vessel size index (Rv, μm), a weighted average measure of tumour blood vessel calibre, and fractional tumour blood volume (fBV, %), in orthotopically propagated murine PC3 prostate tumour xenografts. Tumour vascular architecture was assessed in vivo by MRI prior to and 24 hr after treatment with 200 mg/kg of the vascular disrupting agent ZD6126. A Bayesian hierarchical model (BHM) was used to reduce the uncertainty associated with quantitation of Rv and fBV. Quantitative histological analyses of the uptake of Hoechst 33342 for perfused vasculature, and haematoxylin and eosin staining for necrosis, were also performed to qualify the MRI data. A relatively large median Rv of 40.3 μm (90% confidence interval (CI90) = 37.4, 44.0 μm) and a high fBV of 5.4% (CI90 = 5.3, 5.5%) were determined in control tumours, which agreed with histologically determined vessel size index. Treatment with ZD6126 significantly (p < 0.01) reduced tumour Rv (34.2 μm, CI90 = 31.2, 38.0 μm) and fBV (3.9%, CI90 = 3.8, 4.1%), which were validated against histologically determined significant reductions in perfusion and vessel size, and increased necrosis. Together these data (i) highlight the use of a BHM to optimise the inferential power available from susceptibility contrast MRI data, (ii) provide strong evaluation and qualification of R(v) and fBV as non-invasive imaging biomarkers of tumour vascular morphology, (iii) reveal the presence of a different vascular phenotype and (iv) demonstrate that ZD6126 exhibits good anti-vascular activity against orthotopic prostate tumours.
Collapse
Affiliation(s)
- Simon Walker-Samuel
- Cancer Research UK and EPSRC Cancer Imaging Centre, The Institute of Cancer Research and Royal Marsden NHS Trust, Sutton, Surrey, United Kingdom
| | | | | | | | | | | |
Collapse
|
32
|
Oehler C, von Bueren AO, Furmanova P, Broggini-Tenzer A, Orlowski K, Rutkowski S, Frei K, Grotzer MA, Pruschy M. The microtubule stabilizer patupilone (epothilone B) is a potent radiosensitizer in medulloblastoma cells. Neuro Oncol 2011; 13:1000-10. [PMID: 21743064 DOI: 10.1093/neuonc/nor069] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Concurrent radiochemotherapy for medulloblastoma includes the microtubule disrupting agent vincristine; however, vincristine alone or as part of a combined treatment regimen is highly toxic. A major goal is therefore to replace vincristine with novel potent chemotherapeutic agents-in particular, with microtubule stabilizing and destabilizing compounds-with a larger therapeutic window. Here, we investigated the antiproliferative, cytotoxic and radiosensitizing effect of patupilone (epothilone B [EPO906]), a novel, non-taxane-related and nonneurotoxic microtubule-stabilizing agent in human medulloblastoma cell lines. The antiproliferative and cytotoxic effects of patupilone alone and in combination with ionizing radiation was determined in the 3 representative human medulloblastoma cell lines D341Med, D425Med, and DAOY. Patupilone alone effectively reduced the proliferative activity and clonogenicity of all medulloblastoma cell lines tested at picomolar concentrations (50-200 pM) and resulted in an at least additive anticlonogenic effect in combination with clinically relevant doses of ionizing radiation (2 or 5 Gy). Cell-cycle analysis revealed a sequential G2-M arrest and sub-G1 accumulation in a dose- and treatment-dependent manner after exposure to patupilone. In tumor xenografts derived from D425Med cells, a minimal treatment regimen with patupilone and fractionated irradiation (1 × 2 mg/kg plus 3 × 3 Gy) resulted in an extended tumor growth delay for the 2 single treatment modalities alone and a supra-additive treatment response for the combined treatment modality, with complete tumor regressions. These results demonstrate the potent efficacy of patupilone against medulloblastoma cell lines and indicate that patupilone represents a promising candidate to replace vincristine as part of a combined treatment strategy with ionizing radiation.
Collapse
Affiliation(s)
- Christoph Oehler
- Department of Radiation Oncology, University Hospital Zurich, CH-8091 Zürich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Epothilones are a new group of microtubule-stabilizing agents that have demonstrated antitumor activity in taxane-resistant models. Taxanes remain some of the most active cytotoxic agents in current cancer therapy. Primary or acquired resistance to taxanes in tumor cells partly prevents their long-term efficacy. Certain side effects, such as myelosupression or irreversible neuropathy, can also limit prolonged taxane administration. Epothilone B (EPO906; patupilone), a natural compound, and its semisynthetic derivative, ixabepilone (BMS-247550), differ in their pharmacokinetic and toxicity profiles. Ovarian cancer patients frequently relapse after first-line treatment based on platinum–taxane doublets. Therefore, epothilones might represent a therapeutic alternative in this setting. Patupilone and ixabepilone have undergone parallel clinical development, but their future role in ovarian cancer therapeutics remains ill defined.
Collapse
Affiliation(s)
- Ivan Diaz-Padilla
- Princess Margaret Hospital, Bras Family Drug Development Program, Division of Medical Oncology, 610 University Avenue, Room 5-700, Toronto, ON M5G 2M9, Canada
| | | |
Collapse
|
34
|
Farrar CT, Kamoun WS, Ley CD, Kim YR, Catana C, Kwon SJ, Rosen BR, Jain RK, Sorensen AG. Sensitivity of MRI tumor biomarkers to VEGFR inhibitor therapy in an orthotopic mouse glioma model. PLoS One 2011; 6:e17228. [PMID: 21390238 PMCID: PMC3048404 DOI: 10.1371/journal.pone.0017228] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Accepted: 01/26/2011] [Indexed: 01/22/2023] Open
Abstract
MRI biomarkers of tumor edema, vascular permeability, blood volume, and average vessel caliber are increasingly being employed to assess the efficacy of tumor therapies. However, the dependence of these biomarkers on a number of physiological factors can compromise their sensitivity and complicate the assessment of therapeutic efficacy. Here we examine the response of these MRI tumor biomarkers to cediranib, a potent vascular endothelial growth factor receptor (VEGFR) inhibitor, in an orthotopic mouse glioma model. A significant increase in the tumor volume and relative vessel caliber index (rVCI) and a slight decrease in the water apparent diffusion coefficient (ADC) were observed for both control and cediranib treated animals. This contrasts with a clinical study that observed a significant decrease in tumor rVCI, ADC and volume with cediranib therapy. While the lack of a difference between control and cediranib treated animals in these biomarker responses might suggest that cediranib has no therapeutic benefit, cediranib treated mice had a significantly increased survival. The increased survival benefit of cediranib treated animals is consistent with the significant decrease observed for cediranib treated animals in the relative cerebral blood volume (rCBV), relative microvascular blood volume (rMBV), transverse relaxation time (T2), blood vessel permeability (Ktrans), and extravascular-extracellular space (νe). The differential response of pre-clinical and clinical tumors to cediranib therapy, along with the lack of a positive response for some biomarkers, indicates the importance of evaluating the whole spectrum of different tumor biomarkers to properly assess the therapeutic response and identify and interpret the therapy-induced changes in the tumor physiology.
Collapse
Affiliation(s)
- Christian T Farrar
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Just N. Histogram analysis of the microvasculature of intracerebral human and murine glioma xenografts. Magn Reson Med 2010; 65:778-89. [PMID: 21337410 DOI: 10.1002/mrm.22675] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 08/26/2010] [Accepted: 09/14/2010] [Indexed: 02/04/2023]
Abstract
The purpose of this study is to examine the usefulness of histogram analysis combined with vessel size index (VSI) magnetic resonance imaging for the specific characterization of brain tumor microvasculature in a panel of six volume-matched glioma xenografts. Using a simple descriptive histogram analysis, significant differences of the mean tumoral VSI (P=0.0035 for 9L, P=0.008 for glioma mix, P=0.05 for C6), the 75th VSI percentile (P=0.003-0.075) as well as the 25th and median blood volume (BV) percentiles were found in murine gliomas compared to their contralateral healthy brain. Using a segmented histogram analysis, dilatation of already existing vessels in murine gliomas and development of new small caliber vessels in human glioblastomas were suggested. Most gliomas showed a higher proportion of pixels with BV below 1% (glioma mix [21% vs 1%], Glioblastoma 2 (GBM2) [9% vs 3.7%]) and a smaller proportion of pixels with BV in the range 1.7-6.3% (65 vs 90% for glioma mix, 80 vs 85% in GBM2) relative to their contralateral part. In glioblastomas, VSI and BV distributions were similar to normal brain distributions and in agreement with immunohistochemical findings. The histogram analysis of VSI and BV heterogeneity in experimental brain tumors allowed detection of microregional differences in gliomas from different origins.
Collapse
Affiliation(s)
- Nathalie Just
- Laboratory for functional and metabolic imaging (LIFMET), EPFL, UNIL, Centre d'Imagerie Biomédicale, Department of Radiology, Lausanne, Switzerland.
| |
Collapse
|
36
|
Nielsen T, Murata R, Maxwell RJ, Stødkilde-Jørgensen H, Ostergaard L, Ley CD, Kristjansen PEG, Horsman MR. Non-invasive imaging of combretastatin activity in two tumor models: Association with invasive estimates. Acta Oncol 2010; 49:906-13. [PMID: 20831477 DOI: 10.3109/0284186x.2010.499135] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
INTRODUCTION The efficacy of the vascular disrupting agent combretastatin A-4 phosphate (CA4P) depends on several factors including tumor size, nitric oxide level, interstitial fluid pressure, and vascular permeability. These factors vary among tumor types. The aim of this study was to investigate all these factors in two tumor models that respond differently to CA4P. MATERIAL AND METHODS Mice bearing C3H mammary carcinomas or KHT sarcomas (200 to 800 mm(3)) were intraperitoneally injected with CA4P (100 mg/kg). Tumor size and the effect of a nitric oxide inhibitor nitro-L-arginine (NLA) administered intravenously were evaluated by necrotic fraction histologically assessed at 24 hours. Interstitial fluid pressure (IFP) was measured using the wick-in-needle technique, and vascular characteristics were assessed with dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI). RESULTS Initial necrotic fraction was about 10% in both tumor models at 200 mm(3), but only increased significantly with tumor size in the C3H mammary carcinoma. In this tumor, CA4P significantly induced further necrosis by about 15% at all sizes, but in the KHT tumor, the induced necrotic fraction depended on tumor size. For both tumor types, NLA with CA4P significantly increased necrotic fraction above that for each drug alone. CA4P significantly decreased IFP in all tumors except in the 800 mm(3) C3H tumor, which had an initially non-significant lower value. Interstitial volume estimated by DCE-MRI increased in all groups, except the 800 mm(3) C3H tumors. DCE-MRI vascular parameters showed different initial characteristics and general significant reductions following CA4P treatment. CONCLUSIONS Both tumor models showed differences in all factors before treatment, and in their response to CA4P. Perfusion and permeability as estimated by DCE-MRI play a central role in the CA4P response, and interstitial volume and IFP seemed related. These factors may be of clinical value in the planning of CA4P treatments.
Collapse
Affiliation(s)
- Thomas Nielsen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Anti-Angiogenic/Vascular Effects of the mTOR Inhibitor Everolimus Are Not Detectable by FDG/FLT-PET. Transl Oncol 2010; 3:264-75. [PMID: 20689768 DOI: 10.1593/tlo.10127] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 04/20/2010] [Accepted: 04/21/2010] [Indexed: 01/27/2023] Open
Abstract
Noninvasive functional imaging of tumors can provide valuable early-response biomarkers, in particular, for targeted chemotherapy. Using various experimental tumor models, we have investigated the ability of positron emission tomography (PET) measurements of 2-deoxy-2-[(18)F]fluoro-glucose (FDG) and 3'-deoxy-3'-[(18)F]fluorothymidine (FLT) to detect response to the allosteric mammalian target of rapamycin (mTOR) inhibitor everolimus. Tumor models were declared sensitive (murine melanoma B16/BL6 and human lung H596) or relatively insensitive (human colon HCT116 and cervical KB31), according to the IC(50) values (concentration inhibiting cell growth by 50%) for inhibition of proliferation in vitro (<10 nM and >1 microM, respectively). Everolimus strongly inhibited growth of the sensitive models in vivo but also significantly inhibited growth of the insensitive models, an effect attributable to its known anti-angiogenic/vascular properties. However, although tumor FDG and FLT uptake was significantly reduced in the sensitive models, it was not affected in the insensitive models, suggesting that endothelial-directed effects could not be detected by these PET tracers. Consistent with this hypothesis, in a well-vascularized orthotopic rat mammary tumor model, other antiangiogenic agents also failed to affect FDG uptake, despite inhibiting tumor growth. In contrast, the cytotoxic patupilone, a microtubule stabilizer, blocked tumor growth, and markedly reduced FDG uptake. These results suggest that FDG/FLT-PET may not be a suitable method for early markers of response to antiangiogenic agents and mTOR inhibitors in which anti-angiogenic/vascular effects predominate because the method could provide false-negative responses. These conclusions warrant clinical testing.
Collapse
|
38
|
Everolimus and PTK/ZK show synergistic growth inhibition in the orthotopic BL16/BL6 murine melanoma model. Cancer Chemother Pharmacol 2010; 67:193-200. [PMID: 20512579 DOI: 10.1007/s00280-010-1307-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Accepted: 03/05/2010] [Indexed: 10/19/2022]
Abstract
PURPOSE Everolimus (RAD001, Afinitor) is an mTORC1 pathway inhibitor, and vatalanib (PTK/ZK) is a pan VEGF-R tyrosine kinase inhibitor (TKI). These two drugs have been shown to have overlapping but also distinct anti-angiogenic effects. Consequently, we investigated the pharmacokinetics (PK) and pharmacodynamics (PD) of their combination in vivo. METHODS Murine melanoma B16/BL6 cells were grown orthotopically in BL6/C57 mice by injection into the derma of both ears to create a primary tumour which metastasized rapidly to the cervical lymph nodes. Mice were treated daily p.o. with PTK/ZK (100 mg/kg) or everolimus (1 mg/kg) or their combination, and anti-tumour efficacy (PD) assessed. In the same model, plasma PK of everolimus was measured following single doses of the monotherapy or combination schedules. RESULTS Two independent experiments showed that combination of everolimus and PTK/ZK caused at least additive increases in anti-tumour activity compared to either monotherapy, without increases in toxicity. Pooling the data to improve the statistical power demonstrated the interactions to be synergistic. PK modelling showed that although PTK/ZK increased everolimus plasma concentrations by about twofold, this PK drug-drug interaction could not account for the increased anti-tumour effect of the combination. Modelling of the PTK/ZK dose-response curve in this model suggested that any effect of everolimus on the PK of PTK/ZK was unlikely to affect efficacy. Measurement of changes in tumour and plasma VEGF levels at the endpoint of therapy confirmed earlier observations of differential effects of these two agents. CONCLUSIONS The combination of everolimus and PTK/ZK hold promise for the treatment of human cancers.
Collapse
|
39
|
de Bazelaire C, Pluvinage A, Chapelier M, Hamy AS, Albiter M, Farges C, Bourrier P, Zagdanski AM, Espié M, de Kerviler E, Frija J. [Diffusion-weighted MR imaging of the breast]. JOURNAL DE RADIOLOGIE 2010; 91:394-407. [PMID: 20508574 DOI: 10.1016/s0221-0363(10)70055-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Diffusion-weighted imaging is helpful to further characterize lesions that remain indeterminate after morphological and dynamic MR evaluation. Suspicious lesions are hyperintense on diffusion-weighted images with corresponding low ADC values, indicating restricted diffusion and hypercellularity. Benign lesions and tumors responding to treatment usually have no diffusion restriction. ADC maps are useful for T2W hyperintense lesions that could mask the presence of restricted diffusion. Image fusion is sometimes needed to accurately localize enhancing lesions on ADC maps. For indeterminate lesions, a hypocellular appearance suggests a lower ACR category whereas the presence of restricted diffusion suggests a higher category.
Collapse
Affiliation(s)
- C de Bazelaire
- Service de radiologie, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, 75475 Paris cedex 10, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ott PA, Hamilton A, Jones A, Haas N, Shore T, Liddell S, Christos PJ, Doyle LA, Millward M, Muggia FM, Pavlick AC. A phase II trial of the epothilone B analog ixabepilone (BMS-247550) in patients with metastatic melanoma. PLoS One 2010; 5:e8714. [PMID: 20098694 PMCID: PMC2808339 DOI: 10.1371/journal.pone.0008714] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Accepted: 12/03/2009] [Indexed: 12/02/2022] Open
Abstract
Background Ixabepilone (BMS-247550), an epothilone B analog, is a microtubule stabilizing agent which has shown activity in several different tumor types and preclinical models in melanoma. In an open label, one-arm, multi-center phase II trial the efficacy and toxicity of this epothilone was investigated in two different cohorts: chemotherapy-naïve (previously untreated) and previously treated patients with metastatic melanoma. Methodology/Principal Findings Eligible patients had histologically-confirmed stage IV melanoma, with an ECOG performance status of 0 to 2. Ixabepilone was administered at a dose of 20 mg/m2 on days 1, 8, and 15 during each 28-day cycle. The primary endpoint was response rate (RR); secondary endpoints were time to progression (TTP) and toxicity. Twenty-four patients were enrolled and 23 were evaluable for response. Initial serum lactate dehydrogenase (LDH) levels were elevated in 6/11 (55%) of the previously treated and in 5/13 (38%) of the previously untreated patients. No complete or partial responses were seen in either cohort. One patient in the previously treated group developed neutropenia and fatal septic shock. Seventeen patients (8 in the previously untreated group and 9 in the previously treated group) progressed after 2 cycles, whereas six patients (3 in each group) had stable disease after 2–6 cycles. Median TTP was 1.74 months in the previously untreated group (95% CI = 1.51 months, upper limit not estimated) and 1.54 months in the previously treated group (95% CI = 1.15 months, 2.72 months). Grade 3 and/or 4 toxicities occurred in 5/11 (45%) of previously untreated and in 5/13 (38%) of previously treated patients and included neutropenia, peripheral neuropathy, fatigue, diarrhea, and dyspnea. Conclusions/Significance Ixabepilone has no meaningful activity in either chemotherapy-naïve (previously untreated) or previously treated patients with metastatic melanoma. Further investigation with ixabepilone as single agent in the treatment of melanoma is not warranted. Trial registration Clinical Trials.gov NCT00036764
Collapse
Affiliation(s)
- Patrick A. Ott
- Department of Medical Oncology, New York University Cancer Institute, New York, New York, United States of America
| | - Anne Hamilton
- Royal Prince Alfred Hospital, Sydney Cancer Centre, Sydney, Australia
- Sydney Melanoma Unit and University of Sydney, Sydney, Australia
| | - Amanda Jones
- Department of Medical Oncology, New York University Cancer Institute, New York, New York, United States of America
| | - Naomi Haas
- Division of Hematology/Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Tsiporah Shore
- Division of Hematology/Oncology, Weill Cornell Medical College and New York Presbyterian Hospital, New York, New York, United States of America
| | - Sandra Liddell
- Royal Prince Alfred Hospital, Sydney Cancer Centre, Sydney, Australia
| | - Paul J. Christos
- Division of Biostatistics and Epidemiology, Weill Cornell Medical College, New York, New York, United States of America
| | - L. Austin Doyle
- Investigational Drug Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Michael Millward
- Department of Medical Oncology, Charles Gairdner Hospital and University of Western Australia, Perth, Australia
| | - Franco M. Muggia
- Department of Medical Oncology, New York University Cancer Institute, New York, New York, United States of America
| | - Anna C. Pavlick
- Department of Medical Oncology, New York University Cancer Institute, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
41
|
McSheehy PMJ, Weidensteiner C, Cannet C, Ferretti S, Laurent D, Ruetz S, Stumm M, Allegrini PR. Quantified tumor t1 is a generic early-response imaging biomarker for chemotherapy reflecting cell viability. Clin Cancer Res 2009; 16:212-25. [PMID: 20008843 DOI: 10.1158/1078-0432.ccr-09-0686] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Identification of a generic response biomarker by comparison of chemotherapeutics with different action mechanisms on several noninvasive biomarkers in experimental tumor models. EXPERIMENTAL DESIGN The spin-lattice relaxation time of water protons (T(1)) was quantified using an inversion recovery-TrueFISP magnetic resonance imaging method in eight different experimental tumor models before and after treatment at several different time points with five different chemotherapeutics. Effects on T(1) were compared with other minimally invasive biomarkers including vascular parameters, apparent diffusion coefficient, and interstitial fluid pressure, and were correlated with efficacy at the endpoint and histologic parameters. RESULTS In all cases, successful chemotherapy significantly lowered tumor T(1) compared with vehicle and the fractional change in T(1) (DeltaT(1)) correlated with the eventual change in tumor size (range: r(2) = 0.21, P < 0.05 to r(2) = 0.73, P < 0.0001), except for models specifically resistant to that drug. In RIF-1 tumors, interstitial fluid pressure was decreased, but apparent diffusion coefficient and permeability increased in response to the microtubule stabilizer patupilone and 5-fluorouracil. Although DeltaT(1) was small (maximum of -20%), the variability was very low (5%) compared with other magnetic resonance imaging methods (24-48%). Analyses ex vivo showed unchanged necrosis, increased apoptosis, and decreased %Ki67 and total choline, but only Ki67 and choline correlated with DeltaT(1). Correlation of Ki67 and DeltaT(1) were observed in other models using patupilone, paclitaxel, a VEGF-R inhibitor, and the mammalian target of rapamycin inhibitor everolimus. CONCLUSIONS These results suggest that a decrease in tumor T(1) reflects hypocellularity and is a generic marker of response. The speed and robustness of the method should facilitate its use in clinical trials.
Collapse
Affiliation(s)
- Paul M J McSheehy
- Oncology Research and Global Imaging Group, Novartis Institutes for Biomedical Research, Basel, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Tumor interstitial fluid pressure as an early-response marker for anticancer therapeutics. Neoplasia 2009; 11:874-81. [PMID: 19724681 DOI: 10.1593/neo.09554] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Revised: 06/02/2009] [Accepted: 06/03/2009] [Indexed: 12/21/2022] Open
Abstract
Solid tumors have a raised interstitial fluid pressure (IFP) due to high vessel permeability, low lymphatic drainage, poor perfusion, and high cell density around the blood vessels. To investigate tumor IFP as an early-response biomarker, we have tested the effect of seven anticancer chemotherapeutics including cytotoxics and targeted cytostatics in 13 experimental tumor models. IFP was recorded with the wick-in-needle method. Models were either ectopic or orthotopic and included mouse and rat syngeneic as well as human xenografts in nude mice. The mean basal IFP was between 4.4 and 15.2mm Hg; IFP was lowest in human tumor xenografts and highest in rat syngeneic models. Where measured, basal IFP correlated positively with relative tumor blood volume (rTBV) determined by dynamic contrast-enhanced magnetic resonance imaging. Most chemotherapeutics sooner (2 or 3 days) or later (6 or 7 days) lowered tumor IFP significantly, and the cytotoxic patupilone caused the greatest decrease in IFP. In rat mammary orthotopic BN472 tumors, significant drug-induced decreases in IFP and rTBV correlated positively with each other for both patupilone and the cytostatic vatalanib. In the two orthotopic models studied, early decreases in IFP were significantly (P < or = .005) correlated with late changes in tumor volume. Thus, drug-induced decreases in tumor IFP are an early marker of response to therapy, which could aid clinical development.
Collapse
|
43
|
Beaumont M, Lemasson B, Farion R, Segebarth C, Rémy C, Barbier EL. 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; 29:1714-26. [PMID: 19584891 PMCID: PMC3348120 DOI: 10.1038/jcbfm.2009.86] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This study aimed at combining an iron-based, steady-state, vessel size index magnetic resonance imaging (VSI MRI) approach, and a gadolinium (Gd)-based, dynamic contrast-enhanced MRI approach (DCE MRI) to characterize tumoral microvasculature. Rats bearing an orthotopic glioma (C6, n=14 and RG2, n=6) underwent DCE MRI and combined VSI and DCE MRI 4 h later, at 2.35 T. Gd-DOTA (200 mumol of Gd per kg) and ultrasmall superparamagnetic iron oxide (USPIO) (200 micromol of iron per kg) were used for DCE and VSI MRI, respectively. C6 and RG2 gliomas were equally permeable to Gd-DOTA but presented different blood volume fractions and VSI, in good agreement with histologic data. The presence of USPIO yielded reduced K(trans) values. The K(trans) values obtained with Gd-DOTA in the absence and in the presence of USPIO were well correlated for the C6 glioma but not for the RG2 glioma. It was also observed that, within the time frame of DCE MRI, USPIO remained intravascular in the C6 glioma whereas it extravasated in the RG2 glioma. In conclusion, VSI and DCE MRI can be combined provided that USPIO does not extravasate with the time frame of the DCE MRI experiment. The mechanisms at the origin of USPIO extravasation remain to be elucidated.
Collapse
|
44
|
Comparison of [18F]-Tracers in Various Experimental Tumor Models by PET Imaging and Identification of an Early Response Biomarker for the Novel Microtubule Stabilizer Patupilone. Mol Imaging Biol 2009; 11:308-21. [DOI: 10.1007/s11307-009-0216-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Revised: 10/06/2008] [Accepted: 10/24/2008] [Indexed: 01/29/2023]
|
45
|
Lane HA, Wood JM, McSheehy PMJ, Allegrini PR, Boulay A, Brueggen J, Littlewood-Evans A, Maira SM, Martiny-Baron G, Schnell CR, Sini P, O'Reilly T. mTOR inhibitor RAD001 (everolimus) has antiangiogenic/vascular properties distinct from a VEGFR tyrosine kinase inhibitor. Clin Cancer Res 2009; 15:1612-22. [PMID: 19223496 DOI: 10.1158/1078-0432.ccr-08-2057] [Citation(s) in RCA: 188] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Comparison of the antiangiogenic/vascular properties of the oral mammalian target of rapamycin (mTOR) inhibitor RAD001 (everolimus) and the vascular endothelial growth factor receptor (VEGFR) inhibitor vatalanib (PTK/ZK). EXPERIMENTAL DESIGN Antiproliferative activity against various tumor histotypes and downstream effects on the mTOR pathway were measured in vitro. In vivo, antitumor activity, plasma, and tumor RAD001 levels were measured. Activity in several different angiogenic/vascular assays in vitro and in vivo was assessed and compared with PTK/ZK. RESULTS RAD001 inhibited proliferation in vitro (IC50 values<1 nmol/L to >1 micromol/L), and in sensitive and insensitive tumor cells, pS6 kinase and 4E-BP1 were inhibited. Activity in vitro did not correlate with activity in vivo and significant responses were seen in tumors with IC50 values>10-fold higher than tumor RAD001 concentrations. In vitro, RAD001 inhibited the proliferation of VEGF-stimulated and fibroblast growth factor-stimulated human endothelial cells but not dermal fibroblasts and impaired VEGF release from both sensitive and insensitive tumor cells but did not inhibit migration of human endothelial cells. In vivo, in tumor models derived from either sensitive or insensitive cells, RAD001 reduced Tie-2 levels, the amount of mature and immature vessels, total plasma, and tumor VEGF. RAD001 did not affect blood vessel leakiness in normal vasculature acutely exposed to VEGF nor did it affect tumor vascular permeability (Ktrans) as measured by dynamic contrast-enhanced magnetic resonance imaging. However, the pan-VEGFR inhibitor PTK/ZK inhibited endothelial cell migration and vascular permeability but had less effect on mature vessels compared with RAD001. CONCLUSIONS VEGFR and mTOR inhibitors show similar but also distinct effects on tumor vascular biology, which has implications for their clinical activity alone or in combination.
Collapse
Affiliation(s)
- Heidi A Lane
- Oncology Research, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Bley CR, Jochum W, Orlowski K, Furmanova P, Vuong V, McSheehy PM, Pruschy M. Role of the Microenvironment for Radiosensitization by Patupilone. Clin Cancer Res 2009; 15:1335-42. [DOI: 10.1158/1078-0432.ccr-08-0969] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
47
|
Pathak AP. Magnetic resonance susceptibility based perfusion imaging of tumors using iron oxide nanoparticles. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2008; 1:84-97. [DOI: 10.1002/wnan.17] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Arvind P. Pathak
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University In Vivo Cellular Molecular Imaging Center (JHU ICMIC), Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
48
|
McSheehy P, Allegrini P, Ametaby S, Becquet M, Ebenhan T, Honer M, Ferretti S, Lane H, Schubiger P, Schnell C, Stumm M, Wood J. Minimally invasive biomarkers for therapy monitoring. ACTA ACUST UNITED AC 2008:153-88. [PMID: 18811057 DOI: 10.1007/2789_2008_093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
Abstract
Development of new drugs and optimal application of the drugs currently in use in clinical chemotherapy requires the application of biomarkers. Ideally, these biomarkers would stratify patients so that only those patients likely to respond to a particular therapy receive that therapy. However, that is not always feasible, and an alternative is to make use of early response biomarkers to determine the responding population. In this paper, a number of generic (i.e. not necessarily specific to the action mechanism of the compound) early-response biomarkers are discussed and compared in different models and with three compounds with quite different mechanisms of action: a VEGF-R inhibitor (PTK787), an mTOR inhibitor (RAD001) and a microtubule stabiliser (EPO906). The methods include noninvasive DCE-MRI and PET imaging for measuring tumour vascularity, metabolism and proliferation, as well as the minimally invasive WIN method for measuring tumour interstitial pressure (IFP). The data show that drug-induced changes in IFP (delta IFP) involve mechanism-dependent changes in the tumour vascular architecture, and that delta IFP may be considered a universal generic early-response marker of tumour response to therapy.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/metabolism
- Cell Line, Tumor
- Extracellular Fluid/physiology
- Humans
- Immunohistochemistry
- Magnetic Resonance Imaging
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Nude
- Neoplasm Transplantation
- Neoplasms, Experimental/diagnostic imaging
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/physiopathology
- Positron-Emission Tomography
- Pressure
- Rats
- Rats, Inbred BN
- Transplantation, Heterologous
Collapse
Affiliation(s)
- P McSheehy
- Oncology Research, Novartis Pharma AG, 4002 Basel, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Valable S, Lemasson B, Farion R, Beaumont M, Segebarth C, Remy C, Barbier EL. 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 IN BIOMEDICINE 2008; 21:1043-56. [PMID: 18615861 DOI: 10.1002/nbm.1278] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Assessment of angiogenesis may help to determine tumor grade and therapy follow-up. In vivo imaging methods for non-invasively monitoring microvasculature evolution are therefore of major interest for tumor management. MRI evaluation of blood volume fraction (BVf) and vessel size index (VSI) was applied to assess the evolution of tumor microvasculature in two rat models of glioma (C6 and RG2). The results show that repeated MRI of BVf and VSI - which involves repeated injection of an iron-based MR contrast agent - does not affect either the physiological status of the animals or the accuracy of the MR estimates of the microvascular parameters. The MR measurements were found to correlate well with those obtained from histology. They indicate that microvascular evolution differs significantly between the two glioma models, in good agreement with expression of angiogenic factors (vascular endothelial growth factor, angiopoietin-2) and with activities of matrix metalloproteinases, also assessed in this study. These MRI methods thus provide considerable potential for assessing the response of gliomas to anti-angiogenic and anti-vascular agents, in preclinical studies as well as in the clinic. Furthermore, as differences between the fate of tumor microvasculature may underlie differences in therapeutic response, there is a need for preclinical study of several tumor models.
Collapse
|
50
|
Schnell CR, Stauffer F, Allegrini PR, O'Reilly T, McSheehy PMJ, Dartois C, Stumm M, Cozens R, Littlewood-Evans A, García-Echeverría C, Maira SM. Effects of the dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235 on the tumor vasculature: implications for clinical imaging. Cancer Res 2008; 68:6598-607. [PMID: 18701483 DOI: 10.1158/0008-5472.can-08-1044] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Dysregulated angiogenesis and high tumor vasculature permeability, two vascular endothelial growth factor (VEGF)-mediated processes and hallmarks of human tumors, are in part phosphatidylinositol 3-kinase (PI3K) dependent. NVP-BEZ235, a dual PI3K/mammalian target of rapamycin (mTOR) inhibitor, was found to potently inhibit VEGF-induced cell proliferation and survival in vitro and VEGF-induced angiogenesis in vivo as shown with s.c. VEGF-impregnated agar chambers. Moreover, the compound strongly inhibited microvessel permeability both in normal tissue and in BN472 mammary carcinoma grown orthotopically in syngeneic rats. Similarly, tumor interstitial fluid pressure, a phenomenon that is also dependent of tumor permeability, was significantly reduced by NVP-BEZ235 in a dose-dependent manner on p.o. administration. Because RAD001, a specific mTOR allosteric inhibitor, was ineffective in the preceding experiments, we concluded that the effects observed for NVP-BEZ235 are in part driven by PI3K target modulation. Hence, tumor vasculature reduction was correlated with full blockade of endothelial nitric oxide (NO) synthase, a PI3K/Akt-dependent but mTORC1-independent effector involved in tumor permeability through NO production. In the BN472 tumor model, early reduction of permeability, as detected by K(trans) quantification using the dynamic contrast-enhanced magnetic resonance imaging contrasting agent P792 (Vistarem), was found to be a predictive marker for late-stage antitumor activity by NVP-BEZ235.
Collapse
Affiliation(s)
- Christian R Schnell
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|