1
|
Rezaeian M, Heidari H, Raahemifar K, Soltani M. Image-Based Modeling of Drug Delivery during Intraperitoneal Chemotherapy in a Heterogeneous Tumor Nodule. Cancers (Basel) 2023; 15:5069. [PMID: 37894436 PMCID: PMC10604968 DOI: 10.3390/cancers15205069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/12/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Intraperitoneal (IP) chemotherapy is a promising treatment approach for patients diagnosed with peritoneal carcinomatosis, allowing the direct delivery of therapeutic agents to the tumor site within the abdominal cavity. Nevertheless, limited drug penetration into the tumor remains a primary drawback of this method. The process of delivering drugs to the tumor entails numerous complications, primarily stemming from the specific pathophysiology of the tumor. Investigating drug delivery during IP chemotherapy and studying the parameters affecting it are challenging due to the limitations of experimental studies. In contrast, mathematical modeling, with its capabilities such as enabling single-parameter studies, and cost and time efficiency, emerges as a potent tool for this purpose. In this study, we developed a numerical model to investigate IP chemotherapy by incorporating an actual image of a tumor with heterogeneous vasculature. The tumor's geometry is reconstructed using image processing techniques. The model also incorporates drug binding and uptake by cancer cells. After 60 min of IP treatment with Doxorubicin, the area under the curve (AUC) of the average free drug concentration versus time curve, serving as an indicator of drug availability to the tumor, reached 295.18 mol·m-3·s-1. Additionally, the half-width parameter W1/2, which reflects drug penetration into the tumor, ranged from 0.11 to 0.14 mm. Furthermore, the treatment resulted in a fraction of killed cells reaching 20.4% by the end of the procedure. Analyzing the spatial distribution of interstitial fluid velocity, pressure, and drug concentration in the tumor revealed that the heterogeneous distribution of tumor vasculature influences the drug delivery process. Our findings underscore the significance of considering the specific vascular network of a tumor when modeling intraperitoneal chemotherapy. The proposed methodology holds promise for application in patient-specific studies.
Collapse
Affiliation(s)
- Mohsen Rezaeian
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19967-15433, Iran;
| | - Hamidreza Heidari
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ 07102, USA;
| | - 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, Waterloo, ON N2L 3G1, Canada
- Department of Chemical Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19967-15433, Iran;
- 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 19967-15433, Iran
- Computational Medicine Center, K. N. Toosi University of Technology, Tehran 19967-15433, Iran
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON N2L 3G1, Canada
| |
Collapse
|
2
|
Beddok A, Cottu P, Fourquet A, Kirova Y. [Radiotherapy and targeted therapy for the management of breast cancer: A review]. Cancer Radiother 2023; 27:447-454. [PMID: 37173174 DOI: 10.1016/j.canrad.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 02/19/2023] [Accepted: 02/23/2023] [Indexed: 05/15/2023]
Abstract
The purpose of this study was to review the current knowledge regarding combinations of the most commonly used targeted therapies or those under development for the management of breast cancer with radiation therapy. Several studies have shown that the combination of radiation therapy and tamoxifen increased the risk of radiation-induced lung toxicity; therefore, the two modalities are generally not given concurrently. The combination of HER2 inhibitors (trastuzumab, pertuzumab) and radiation therapy appeared to be safe. However, trastuzumab emtansine (T-DM1) should not be given concomitantly with brain radiation therapy because this combination may increase the risk of brain radionecrosis. The combination of radiation therapy with other new targeted therapies such as new selective estrogen receptor modulators (SERDs), lapatinib, cell cycle inhibitors, immune checkpoint inhibitors, or molecules acting on DNA damage repair seems feasible but has been mainly evaluated on retrospective or prospective studies with small numbers of patients. Moreover, there is a great heterogeneity between these studies regarding the dose and fractionation used in radiotherapy, the dosage of systemic treatments and the sequence of treatments used. Therefore, the combination of these new molecules with radiotherapy should be proposed sparingly, under close monitoring, pending the ongoing prospective studies cited in this review.
Collapse
Affiliation(s)
- A Beddok
- Laboratoire d'imagerie translationnelle en oncologie (Lito), Institut Curie, université PSL, université Paris Saclay, Inserm, 91898 Orsay, France; Département de radiothérapie oncologique, institut Curie, université PSL, Centre de protonthérapie, centre universitaire, 91898 Orsay, France.
| | - P Cottu
- Département d'oncologie médicale, institut Curie, Paris, France
| | - A Fourquet
- Département de radiothérapie oncologique, institut Curie, université PSL, Paris, France
| | - Y Kirova
- Département de radiothérapie oncologique, institut Curie, université PSL, Paris, France
| |
Collapse
|
3
|
Singh N, Tapader R, Chatterjee S, Pal A, Pal A. Subtilisin from Bacillus amyloliquefaciens induces apoptosis in breast cancer cells through ubiquitin-proteasome-mediated tubulin degradation. Int J Biol Macromol 2022; 220:852-865. [PMID: 35985398 DOI: 10.1016/j.ijbiomac.2022.08.086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/20/2022] [Accepted: 08/11/2022] [Indexed: 11/05/2022]
Abstract
To search for novel proteases from environmental isolates which can induce apoptosis in cancer cells, we have purified subtilisin from Bacillus amyloliquefaciens and studied its anti-cancer properties. Subtilisin induced apoptosis in colon (HT29) and breast (MCF7) cancer cells but showed no effect on mouse peritoneal macrophages and normal breast cells (MCF10A). Western blot analysis showed that Bax, Bcl-2 level remained unchanged but tubulin level decreased significantly. Subtilisin does not induce the intrinsic pathway of apoptosis, rather it induced tubulin degradation in MCF-7 cells, whereas in normal cells (MCF-10A) tubulin degradation was not observed. Subtilisin activates ubiquitination and proteasomal-mediated tubulin degradation which was completely restored in presence of proteasome inhibitor MG-132. We further observed PARKIN, one of the known E3-ligase, is overexpressed and interacts with tubulin in subtilisin treated cells. Knockdown of PARKIN effectively downregulates ubiquitination and inhibits degradation of tubulin. PARKIN activation and tubulin degradation lead to ER-stress which in turn activates caspase-7 and PARP cleavage, thus guiding the subtilisin treated cells towards apoptosis. To our knowledge this is the first report of subtilisin induced apoptosis in cancer cells by proteasomal degradation of tubulin.
Collapse
Affiliation(s)
- Nanda Singh
- Division of Pathophysiology, ICMR-National Institute of Cholera and Enteric Diseases (NICED), Kolkata 700010, India
| | - Rima Tapader
- Division of Pathophysiology, ICMR-National Institute of Cholera and Enteric Diseases (NICED), Kolkata 700010, India
| | - Shruti Chatterjee
- Division of Biotechnology and Phycology, CSIR-Central Salt & Marine Chemical Research Institute, Bhavnagar 364002, India
| | - Ananda Pal
- Division of Clinical Medicine, ICMR-National Institute of Cholera and Enteric Diseases (NICED), Kolkata 700010, India
| | - Amit Pal
- Division of Pathophysiology, ICMR-National Institute of Cholera and Enteric Diseases (NICED), Kolkata 700010, India.
| |
Collapse
|
4
|
Kozin SV. Vascular damage in tumors: a key player in stereotactic radiation therapy? Trends Cancer 2022; 8:806-819. [PMID: 35835699 DOI: 10.1016/j.trecan.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/23/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022]
Abstract
The use of stereotactic radiation therapy (SRT) for cancer treatment has grown in recent years, showing excellent results for some tumors. The greatly increased doses per fraction in SRT compared to conventional radiotherapy suggest a 'new biology' that determines treatment outcome. Proposed mechanisms include significant damage to tumor blood vessels and enhanced antitumor immune responses, which are also vasculature-dependent. These ideas are mostly based on the results of radiation studies in animal models because direct observations in humans are limited. However, even preclinical findings are somewhat incomplete and result in ambiguous conclusions. Current evidence of vasculature-related mechanisms of SRT is reviewed. Understanding them could result in better optimization of SRT alone or in combination with immune or other cancer therapies.
Collapse
Affiliation(s)
- Sergey V Kozin
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
5
|
A spatiotemporal multi-scale computational model for FDG PET imaging at different stages of tumor growth and angiogenesis. Sci Rep 2022; 12:10062. [PMID: 35710559 PMCID: PMC9203789 DOI: 10.1038/s41598-022-13345-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/09/2022] [Indexed: 01/07/2023] Open
Abstract
A deeper understanding of the tumor microenvironment (TME) and its role in metabolic activity at different stages of vascularized tumors can provide useful insights into cancer progression and better support clinical assessments. In this study, a robust and comprehensive multi-scale computational model for spatiotemporal transport of F-18 fluorodeoxyglucose (FDG) is developed to incorporate important aspects of the TME, spanning subcellular-, cellular-, and tissue-level scales. Our mathematical model includes biophysiological details, such as radiopharmaceutical transport within interstitial space via convection and diffusion mechanisms, radiopharmaceutical exchange between intracellular and extracellular matrices by glucose transporters, cellular uptake of radiopharmaceutical, as well as its intracellular phosphorylation by the enzyme. Further, to examine the effects of tumor size by varying microvascular densities (MVDs) on FDG dynamics, four different capillary networks are generated by angiogenesis modeling. Results demonstrate that as tumor grows, its MVD increases, and hence, the spatiotemporal distribution of total FDG uptake by tumor tissue changes towards a more homogenous distribution. In addition, spatiotemporal distributions in tumor with lower MVD have relatively smaller magnitudes, due to the lower diffusion rate of FDG as well as lower local intravenous FDG release. Since mean standardized uptake value (SUVmean) differs at various stages of microvascular networks with different tumor sizes, it may be meaningful to normalize the measured values by tumor size and the MVD prior to routine clinical reporting. Overall, the present framework has the potential for more accurate investigation of biological phenomena within TME towards personalized medicine.
Collapse
|
6
|
Abazari MA, Soltani M, Moradi Kashkooli F, Raahemifar K. Synthetic 18F-FDG PET Image Generation Using a Combination of Biomathematical Modeling and Machine Learning. Cancers (Basel) 2022; 14:2786. [PMID: 35681767 PMCID: PMC9179454 DOI: 10.3390/cancers14112786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/21/2022] [Accepted: 06/01/2022] [Indexed: 12/10/2022] Open
Abstract
No previous works have attempted to combine generative adversarial network (GAN) architectures and the biomathematical modeling of positron emission tomography (PET) radiotracer uptake in tumors to generate extra training samples. Here, we developed a novel computational model to produce synthetic 18F-fluorodeoxyglucose (18F-FDG) PET images of solid tumors in different stages of progression and angiogenesis. First, a comprehensive biomathematical model is employed for creating tumor-induced angiogenesis, intravascular and extravascular fluid flow, as well as modeling of the transport phenomena and reaction processes of 18F-FDG in a tumor microenvironment. Then, a deep convolutional GAN (DCGAN) model is employed for producing synthetic PET images using 170 input images of 18F-FDG uptake in each of 10 different tumor microvascular networks. The interstitial fluid parameters and spatiotemporal distribution of 18F-FDG uptake in tumor and healthy tissues have been compared against previously published numerical and experimental studies, indicating the accuracy of the model. The structural similarity index measure (SSIM) and peak signal-to-noise ratio (PSNR) of the generated PET sample and the experimental one are 0.72 and 28.53, respectively. Our results demonstrate that a combination of biomathematical modeling and GAN-based augmentation models provides a robust framework for the non-invasive and accurate generation of synthetic PET images of solid tumors in different stages.
Collapse
Affiliation(s)
- Mohammad Amin Abazari
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19967-15433, Iran; (M.A.A.); (F.M.K.)
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19967-15433, Iran; (M.A.A.); (F.M.K.)
- Faculty of Science, School of Optometry and Vision Science, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
- Advanced Bioengineering Initiative Center, Multidisciplinary International Complex, K. N. Toosi Univesity of Technology, Tehran 14176-14411, Iran
- Center for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Department of Electrical and Computer Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Farshad Moradi Kashkooli
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19967-15433, Iran; (M.A.A.); (F.M.K.)
| | - Kaamran Raahemifar
- Faculty of Science, School of Optometry and Vision Science, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
- Data Science and Artificial Intelligence Program, College of Information Sciences and Technology (IST), Penn State University, State College, PA 16801, USA
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| |
Collapse
|
7
|
Kashkooli FM, Rezaeian M, Soltani M. Drug delivery through nanoparticles in solid tumors: a mechanistic understanding. Nanomedicine (Lond) 2022; 17:695-716. [PMID: 35451315 DOI: 10.2217/nnm-2021-0126] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: In this study, the main goal was to apply a multi-scale computational model in evaluating nano-sized drug-delivery systems, following extracellular drug release, into solid tumors in order to predict treatment efficacy. Methods: The impact of several parameters related to tumor (size, shape, vessel-wall pore size, and necrotic core size) and therapeutic agents (size of nanoparticles, binding affinity of drug, drug release rate from nanoparticles) are examined in detail. Results: This study illustrates that achieving a higher treatment efficacy requires smaller nanoparticles (NPs) or a low binding affinity and drug release rate. Long-term analysis finds that a slow release rate in extracellular space does not always improve treatment efficacy compared with a rapid release rate; NP size as well as binding affinity of drug are also highly influential. Conclusions: The presented methodology can be used as a step forward towards optimization of patient-specific nanomedicine plans.
Collapse
Affiliation(s)
| | - Mohsen Rezaeian
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - M Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran.,Department of Electrical & Computer Engineering, University of Waterloo, Waterloo, Canada.,Centre for Biotechnology & Bioengineering (CBB), University of Waterloo, Waterloo, Canada.,Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Iran
| |
Collapse
|
8
|
Varon E, Blumrosen G, Shefi O. A predictive model for personalization of nanotechnology-based phototherapy in cancer treatment. Front Oncol 2022; 12:1037419. [PMID: 36911792 PMCID: PMC9999042 DOI: 10.3389/fonc.2022.1037419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 11/21/2022] [Indexed: 01/06/2023] Open
Abstract
A major challenge in radiation oncology is the prediction and optimization of clinical responses in a personalized manner. Recently, nanotechnology-based cancer treatments are being combined with photodynamic therapy (PDT) and photothermal therapy (PTT). Predictive models based on machine learning techniques can be used to optimize the clinical setup configuration, including such parameters as laser radiation intensity, treatment duration, and nanoparticle features. In this article we demonstrate a methodology that can be used to identify the optimal treatment parameters for PDT and PTT by collecting data from in vitro cytotoxicity assay of PDT/PTT-induced cell death using a single nanocomplex. We construct three machine learning prediction models, employing regression, interpolation, and low- degree analytical function fitting, to predict the laser radiation intensity and duration settings that maximize the treatment efficiency. To examine the accuracy of these prediction models, we construct a dedicated dataset for PDT, PTT, and a combined treatment; this dataset is based on cell death measurements after light radiation treatment and is divided into training and test sets. The preliminary results show that the performance of all three models is sufficient, with death rate errors of 0.09, 0.15, and 0.12 for the regression, interpolation, and analytical function fitting approaches, respectively. Nevertheless, due to its simple form, the analytical function method has an advantage in clinical application and can be used for further analysis of the sensitivity of performance to the treatment parameters. Overall, the results of this study form a baseline for a future personalized prediction model based on machine learning in the domain of combined nanotechnology- and phototherapy-based cancer treatment.
Collapse
Affiliation(s)
- Eli Varon
- Faculty of Engineering, Bar-Ilan University, Ramat Gan, Israel.,Bar-Ilan Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - Gaddi Blumrosen
- Department of Digital Medical Technologies, Holon Institute of Technology, Holon, Israel.,Department of Computer Science, Holon Institute of Technology, Holon, Israel
| | - Orit Shefi
- Faculty of Engineering, Bar-Ilan University, Ramat Gan, Israel.,Bar-Ilan Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel.,Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
9
|
Beddok A, Cottu P, Fourquet A, Kirova Y. Combination of Modern Radiotherapy and New Targeted Treatments for Breast Cancer Management. Cancers (Basel) 2021; 13:cancers13246358. [PMID: 34944978 PMCID: PMC8699586 DOI: 10.3390/cancers13246358] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Since the introduction of hormone therapy for the treatment of breast cancer (BC) three decades ago, many new targeted therapies have been developed. Some of them are currently used, such as HER2 inhibitors, while others are still under development, such as cell cycle (CDK) inhibitors, immune checkpoint (PD1/PDL1) inhibitors, or molecules acting on DNA damage (PARP) repair. Besides this, radiation therapy (RT) is commonly used either as adjuvant treatment for early BC after breast conservative surgery or in palliative intent for the treatment of metastatic sites. Our research has shown that the combinations of the most commonly used targeted treatments and RT were feasible with a few toxicities. Nevertheless, most of the knowledge on this subject is based on retrospective studies and a small number of patients and care should be taken in this setting until these results would be confirmed in prospective randomized studies. Abstract Background: The objective of the present study was to review the essential knowledge about the combinations of the most commonly used or under development targeted treatments and radiation therapy (RT). Methods: Preclinical and clinical studies investigating this combination were extensively reviewed. Results: Several studies showed that the combination of RT and tamoxifen increased the risk of radiation-induced pulmonary toxicity; therefore, both modalities should not be given concomitantly. The combination of HER2 inhibitors (trastuzumab, pertuzumab) and RT seems to be safe. However, trastuzumab emtansine (T-DM1) should not be administered concurrently with brain RT since this combination could increase the risk of brain radionecrosis. The combination of RT and other new target treatments such as selective estrogen receptor degradants, lapatinib, cell cycle inhibitors, immune checkpoint inhibitors, or molecules acting on DNA damage repair seems feasible but was essentially evaluated on retrospective or prospective studies with a small number of patients. Furthermore, there is considerable heterogeneity among these studies regarding the dose and fractionation of radiation, the dosage of drugs, and the sequence of treatments used. Conclusions: The combination of RT with most targeted therapies for BC appears to be well-tolerated, but these results need to be confirmed in prospective randomized studies.
Collapse
Affiliation(s)
- Arnaud Beddok
- Department of Radiation Oncology, Institut Curie, 75005 Paris, France; (A.F.); (Y.K.)
- Department of Radiation Oncology, Institut Curie, 91400 Orsay, France
- Laboratory of Translational Imaging in Oncology (LITO), UMR (U1288), Institut Curie, 91400 Orsay, France
- Correspondence: or ; Tel.: +33-144324504
| | - Paul Cottu
- Department of Medical Oncology, Institut Curie, 75005 Paris, France;
| | - Alain Fourquet
- Department of Radiation Oncology, Institut Curie, 75005 Paris, France; (A.F.); (Y.K.)
| | - Youlia Kirova
- Department of Radiation Oncology, Institut Curie, 75005 Paris, France; (A.F.); (Y.K.)
| |
Collapse
|
10
|
Moradi Kashkooli F, Soltani M. Evaluation of solid tumor response to sequential treatment cycles via a new computational hybrid approach. Sci Rep 2021; 11:21475. [PMID: 34728726 PMCID: PMC8563754 DOI: 10.1038/s41598-021-00989-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/21/2021] [Indexed: 12/22/2022] Open
Abstract
The development of an in silico approach that evaluates and identifies appropriate treatment protocols for individuals could help grow personalized treatment and increase cancer patient lifespans. With this motivation, the present study introduces a novel approach for sequential treatment cycles based on simultaneously examining drug delivery, tumor growth, and chemotherapy efficacy. This model incorporates the physical conditions of tumor geometry, including tumor, capillary network, and normal tissue assuming real circumstances, as well as the intravascular and interstitial fluid flow, drug concentration, chemotherapy efficacy, and tumor recurrence. Three treatment approaches-maximum tolerated dose (MTD), metronomic chemotherapy (MC), and chemo-switching (CS)-as well as different chemotherapy schedules are investigated on a real tumor geometry extracted from image. Additionally, a sensitivity analysis of effective parameters of drug is carried out to evaluate the potential of using different other drugs in cancer treatment. The main findings are: (i) CS, MC, and MTD have the best performance in reducing tumor cells, respectively; (ii) multiple doses raise the efficacy of drugs that have slower clearance, higher diffusivity, and lower to medium binding affinities; (iii) the suggested approach to eradicating tumors is to reduce their cells to a predetermined rate through chemotherapy and then apply adjunct therapy.
Collapse
Affiliation(s)
| | - M Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran.
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON, Canada.
- Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran.
- Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Iran.
| |
Collapse
|
11
|
Zwicker F, Hauswald H, Weber KJ, Debus JÜ, Huber PE. In Vivo Evaluation of Combined CK2 Inhibition and Irradiation in Human WiDr Tumours. In Vivo 2021; 35:111-117. [PMID: 33402456 DOI: 10.21873/invivo.12238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/17/2020] [Accepted: 10/21/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM Casein kinase 2 (CK2) which sustains multiple pro-survival functions in cellular DNA-damage response, is strictly regulated in normal cells but elevated in cancer. CK2 is considered as a potential therapeutic target, and its inhibition has been associated with radiosensitization in mammalian cells in vitro. Here, we investigated potential radiosensitization by CK2 inhibition in vivo. MATERIALS AND METHODS The effect of CK2 inhibition in vivo was investigated in human WiDr-xenograft tumours grown subcutaneously on BALB/c nu/nu mice with and without fractionated irradiation. CK2 inhibition was performed using the specific inhibitor tetra-bromobenzotriazole (TBB). Histological examinations included staining for apoptosis and double-strand breaks. RESULTS Both TBB treatment alone and radiation alone significantly reduced tumour growth, which was reflected by increased apoptosis rates. However, TBB treatment did not boost radiation-induced tumour growth suppression in combined treatment, although the apoptosis rate increased and repair of double-strand breaks was reduced. This was in stark contrast to previous data on in vitro radiosensitization. CONCLUSION The absence of radiosensitization by CK2 inhibition should be investigated in different tumour models.
Collapse
Affiliation(s)
- Felix Zwicker
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; .,Clinical Cooperation Unit Molecular Radiation Oncology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Henrik Hauswald
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Klaus-Josef Weber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - JÜrgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Molecular Radiation Oncology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Peter E Huber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Molecular Radiation Oncology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| |
Collapse
|
12
|
Moradi Kashkooli F, Soltani M, Momeni MM. Computational modeling of drug delivery to solid tumors: A pilot study based on a real image. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
13
|
Huang D, Sun L, Huang L, Chen Y. Nanodrug Delivery Systems Modulate Tumor Vessels to Increase the Enhanced Permeability and Retention Effect. J Pers Med 2021; 11:124. [PMID: 33672813 PMCID: PMC7917988 DOI: 10.3390/jpm11020124] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 12/24/2022] Open
Abstract
The use of nanomedicine for antitumor therapy has been extensively investigated for a long time. Enhanced permeability and retention (EPR) effect-mediated drug delivery is currently regarded as an effective way to bring drugs to tumors, especially macromolecular drugs and drug-loaded pharmaceutical nanocarriers. However, a disordered vessel network, and occluded or embolized tumor blood vessels seriously limit the EPR effect. To augment the EPR effect and improve curative effects, in this review, we focused on the perspective of tumor blood vessels, and analyzed the relationship among abnormal angiogenesis, abnormal vascular structure, irregular blood flow, extensive permeability of tumor vessels, and the EPR effect. In this commentary, nanoparticles including liposomes, micelles, and polymers extravasate through the tumor vasculature, which are based on modulating tumor vessels, to increase the EPR effect, thereby increasing their therapeutic effect.
Collapse
Affiliation(s)
- Dong Huang
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; (D.H.); (L.S.)
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Lingna Sun
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; (D.H.); (L.S.)
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA;
| | - Yanzuo Chen
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; (D.H.); (L.S.)
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
14
|
Moradi Kashkooli F, Soltani M, Rezaeian M, Meaney C, Hamedi MH, Kohandel M. Effect of vascular normalization on drug delivery to different stages of tumor progression: In-silico analysis. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101989] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
15
|
Law JWF, Law LNS, Letchumanan V, Tan LTH, Wong SH, Chan KG, Ab Mutalib NS, Lee LH. Anticancer Drug Discovery from Microbial Sources: The Unique Mangrove Streptomycetes. Molecules 2020; 25:E5365. [PMID: 33212836 PMCID: PMC7698459 DOI: 10.3390/molecules25225365] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/08/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
Worldwide cancer incidence and mortality have always been a concern to the community. The cancer mortality rate has generally declined over the years; however, there is still an increased mortality rate in poorer countries that receives considerable attention from healthcare professionals. This suggested the importance of the prompt detection, effective treatment, and prevention strategies. The genus Streptomyces has been documented as a prolific producer of biologically active secondary metabolites. Streptomycetes from mangrove environments attract researchers' attention due to their ability to synthesize diverse, interesting bioactive metabolites. The present review highlights research on mangrove-derived streptomycetes and the production of anticancer-related compounds from these microorganisms. Research studies conducted between 2008 and 2019, specifically mentioning the isolation of streptomycetes from mangrove areas and described the successful purification of compound(s) or generation of crude extracts with cytotoxic activity against human cancer cell lines, were compiled in this review. It is anticipated that there will be an increase in prospects for mangrove-derived streptomycetes as one of the natural resources for the isolation of chemotherapeutic agents.
Collapse
Affiliation(s)
- Jodi Woan-Fei Law
- Novel Bacteria and Drug Discovery (NBDD) Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia; (J.W.-F.L.); (V.L.); (L.T.-H.T.)
| | - Lydia Ngiik-Shiew Law
- Monash Credentialed Pharmacy Clinical Educator, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville 3052, VIC, Australia;
| | - Vengadesh Letchumanan
- Novel Bacteria and Drug Discovery (NBDD) Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia; (J.W.-F.L.); (V.L.); (L.T.-H.T.)
| | - Loh Teng-Hern Tan
- Novel Bacteria and Drug Discovery (NBDD) Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia; (J.W.-F.L.); (V.L.); (L.T.-H.T.)
| | - Sunny Hei Wong
- Li Ka Shing Institute of Health Sciences, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong, China;
| | - Kok-Gan Chan
- Division of Genetics and Molecular Biology, Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia
- International Genome Centre, Jiangsu University, Zhenjiang 212013, China
| | - Nurul-Syakima Ab Mutalib
- UKM Medical Molecular Biology Institute (UMBI), UKM Medical Centre, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery (NBDD) Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia; (J.W.-F.L.); (V.L.); (L.T.-H.T.)
| |
Collapse
|
16
|
Moradi A, Pourseif MM, Jafari B, Parvizpour S, Omidi Y. Nanobody-based therapeutics against colorectal cancer: Precision therapies based on the personal mutanome profile and tumor neoantigens. Pharmacol Res 2020; 156:104790. [DOI: 10.1016/j.phrs.2020.104790] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/07/2020] [Accepted: 03/31/2020] [Indexed: 12/19/2022]
|
17
|
Moradi Kashkooli F, Soltani M, Hamedi MH. Drug delivery to solid tumors with heterogeneous microvascular networks: Novel insights from image-based numerical modeling. Eur J Pharm Sci 2020; 151:105399. [PMID: 32485347 DOI: 10.1016/j.ejps.2020.105399] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/27/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022]
Abstract
The present study examines chemotherapy by incorporating multi-scale mathematical modeling to predict drug delivery and its effects. This approach leads to a more-realistic physiological tumor model than is possible with previous approaches, as it obtains the capillary network geometry from an image, and also considers the tumor's necrotic core, drug binding, and cellular uptake. Modeling of the fluid flow and drug transport is then performed in the extracellular matrix. The results demonstrate a 10% drop in the fraction of killed cancer cells 69% rather than the 79% reported earlier for a tumor of similar geometry a more-accurate value. This study examines how tumor-related parameters including the necrotic core size and tumor size, and also drug-related parameters drug dosage, binding affinity of drug, and drug degradation can affect the delivery of the drug to solid tumors. Results indicate that concentration of drug are high in the tumor, low in normal tissue, and remarkably low in the necrotic core. Results also offer a treatment of tumors with smaller necrotic core. Tumor size, which implies the tumor progression, has a considerable impact on treatment outcomes, so to be more effective, treatment should be applied at a specific size of tumor. It is demonstrated that binding affinity of drugs to cell-surface receptors and drug dosage have significant impact on treatment efficacy, so they should be regulated based on a balanced quantification between maximum treatment efficacy and minimum side effects. On the other hand, considering the effects of drug degradation in the model has not significant effect on treatment efficacy. The findings of the present study provide insight into the mechanism of drug delivery to solid tumors based on analyzing the effective parameters and modeling how their behavior in the tumor microenvironment affects treatment efficacy.
Collapse
Affiliation(s)
- Farshad Moradi Kashkooli
- Department of Applied Mathematics, University of Waterloo, Waterloo, ON, Canada; Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran.
| | - M Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran; Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON, Canada; Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON, Canada; Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Iran; Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
18
|
Wang H, Yan B, Yue L, He M, Liu Y, Li H. The Diagnostic Value of 3D Power Doppler Ultrasound Combined With VOCAL in the Vascular Distribution of Breast Masses. Acad Radiol 2020; 27:198-203. [PMID: 31053481 DOI: 10.1016/j.acra.2019.02.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 01/08/2023]
Abstract
RATIONALE AND OBJECTIVES This study uses a three-dimensional energy Doppler technique combined with the Virtual Organ Computer-aided Analysis (VOCAL) method in order to determine the diagnostic threshold of blood flow index in breast tumors to provide a reference for evaluation and treatment options. MATERIALS AND METHODS We collected 322 solid lesions which had been operated. Each lesion met the definite pathological diagnosis; collected lesions included 262 cases of benign lesions and 60 cases of malignant lesions. All examinations were performed by using GE LOGIQ E9 with VOCAL software. Volume and four distinct vascular indices of gray mean (MG), power mean, ratio (R), and vascular flow index (VFI) were calculated by using the VOCAL software. Sampling and calculation were repeated three times and the mean value was calculated. RESULTS The average age and power of the malignant group were greater than those of the benign group, ie p < .01 which had significant differences. The gray mean of the malignant group was lower than that of the benign group, ie p > .05 which had no significant differences between benign and malignant groups. The ratio, vascular flow index and volume had significant differences, i.e. p < .01. The area under the receiver operating characteristic curve (AUC) were 0.864, 0.830, 0.800, 0.758, and 0.764 for age, power, ratio, vascular flow index, and volume, respectively. The research indicators were higher than 50% of the curve showing their diagnostic value. The cut-off points of age, power, ratio, vascular flow index, and volume were 37.5, 26.56, 0.031, 0.846, and 1.75, respectively. Their corresponding sensitivity were 93.3%, 75%, 81.7%, 68.3%, 63.3%, and the specificity were 68.7%, 81%, 70.2%, 75.6%, and 81.7%, respectively. Comparison of vascular indices combined with the Breast imaging reporting and data System (BI-RADS) score and simple BI-RADS method, the AUC of power + BI-RADS, ratio + BI-RADS, VFI + BI-RADS, and BI-RADS alone are 0.928, 0.903, 0.895, and 0.796, respectively, which were higher than 50% of the curve. Sensitivity was 81.7%, 80%, 88.3%, 86.7%, and specificity was 88.5%, 85.5%, 77.1%, 69.5%, respectively. The power + BI-RADS method has the highest AUC among these three methods. CONCLUSIONS Quantitative measurement of blood flow and blood vessel distribution in breast tumors by three-dimensional power Doppler ultrasound combined with the VOCAL method is more accurate and sensitive than the traditional two-dimensional ultrasound. And this method has potential promising applications in many current active research areas, such as the studies of random distribution of intratumoral blood vessels or the normalization of tumor blood vessels. Three-dimensional power Doppler ultrasound combined with the VOCAL method provides a new approach to achieving accurate judgments and the method evaluates the curative effect in breast cancer patients.
Collapse
|
19
|
Differential Effects of Ang-2/VEGF-A Inhibiting Antibodies in Combination with Radio- or Chemotherapy in Glioma. Cancers (Basel) 2019; 11:cancers11030314. [PMID: 30845704 PMCID: PMC6468722 DOI: 10.3390/cancers11030314] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/28/2019] [Accepted: 02/28/2019] [Indexed: 12/16/2022] Open
Abstract
Antiangiogenic strategies have not shown striking antitumor activities in the majority of glioma patients so far. It is unclear which antiangiogenic combination regimen with standard therapy is most effective. Therefore, we compared anti-VEGF-A, anti-Ang2, and bispecific anti-Ang-2/VEGF-A antibody treatments, alone and in combination with radio- or temozolomide (TMZ) chemotherapy, in a malignant glioma model using multiparameter two-photon in vivo microscopy in mice. We demonstrate that anti-Ang-2/VEGF-A lead to the strongest vascular changes, including vascular normalization, both as monotherapy and when combined with chemotherapy. The latter was accompanied by the most effective chemotherapy-induced death of cancer cells and diminished tumor growth. This was most probably due to a better tumor distribution of the drug, decreased tumor cell motility, and decreased formation of resistance-associated tumor microtubes. Remarkably, all these parameters where reverted when radiotherapy was chosen as combination partner for anti-Ang-2/VEGF-A. In contrast, the best combination partner for radiotherapy was anti-VEGF-A. In conclusion, while TMZ chemotherapy benefits most from combination with anti-Ang-2/VEGF-A, radiotherapy does from anti-VEGF-A. The findings imply that uninformed combination regimens of antiangiogenic and cytotoxic therapies should be avoided.
Collapse
|
20
|
Moradi Kashkooli F, Soltani M, Rezaeian M, Taatizadeh E, Hamedi MH. Image-based spatio-temporal model of drug delivery in a heterogeneous vasculature of a solid tumor - Computational approach. Microvasc Res 2019; 123:111-124. [PMID: 30711547 DOI: 10.1016/j.mvr.2019.01.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/29/2019] [Accepted: 01/29/2019] [Indexed: 12/31/2022]
Abstract
The solute transport distribution in a tumor is an important criterion in the evaluation of the cancer treatment efficacy. The fraction of killed cells after each treatment can quantify the therapeutic effect and plays as a helpful tool to evaluate the chemotherapy treatment schedules. In the present study, an image-based spatio-temporal computational model of a solid tumor is provided for calculation of interstitial fluid flow and solute transport. Current model incorporates heterogeneous microvasculature for angiogenesis instead of synthetic mathematical modeling. In this modeling process, a comprehensive model according to Convection-Diffusion-Reaction (CDR) equations is employed due to its high accuracy for simulating the binding and the uptake of the drug by tumor cells. Based on the velocity and the pressure distribution, transient distribution of the different drug concentrations (free, bound, and internalized) is calculated. Then, the fraction of killed cells is obtained according to the internalized concentration. Results indicate the dependence of the drug distribution on both time and space, as well as the microvasculature density. Free and bound drug concentration have the same trend over time, whereas, internalized and total drug concentration increases over time and reaches a constant value. The highest amount of concentration occurred in the tumor region due to the higher permeability of the blood vessels. Moreover, the fraction of killed cells is approximately 78.87% and 24.94% after treatment with doxorubicin for cancerous and normal tissues, respectively. In general, the presented methodology may be applied in the field of personalized medicine to optimize patient-specific treatments. Also, such image-based modeling of solid tumors can be used in laboratories that working on drug delivery and evaluating new drugs before using them for any in vivo or clinical studies.
Collapse
Affiliation(s)
| | - M Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran; Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Iran; Department of Electrical and Computer Engineering, University of Waterloo, ON, Canada; Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Ontario, Canada; Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohsen Rezaeian
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Erfan Taatizadeh
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | | |
Collapse
|
21
|
Chen X, Mims J, Huang X, Singh N, Motea E, Planchon SM, Beg M, Tsang AW, Porosnicu M, Kemp ML, Boothman DA, Furdui CM. Modulators of Redox Metabolism in Head and Neck Cancer. Antioxid Redox Signal 2018; 29:1660-1690. [PMID: 29113454 PMCID: PMC6207163 DOI: 10.1089/ars.2017.7423] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 11/04/2017] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Head and neck squamous cell cancer (HNSCC) is a complex disease characterized by high genetic and metabolic heterogeneity. Radiation therapy (RT) alone or combined with systemic chemotherapy is widely used for treatment of HNSCC as definitive treatment or as adjuvant treatment after surgery. Antibodies against epidermal growth factor receptor are used in definitive or palliative treatment. Recent Advances: Emerging targeted therapies against other proteins of interest as well as programmed cell death protein 1 and programmed death-ligand 1 immunotherapies are being explored in clinical trials. CRITICAL ISSUES The disease heterogeneity, invasiveness, and resistance to standard of care RT or chemoradiation therapy continue to constitute significant roadblocks for treatment and patients' quality of life (QOL) despite improvements in treatment modality and the emergence of new therapies over the past two decades. FUTURE DIRECTIONS As reviewed here, alterations in redox metabolism occur at all stages of HNSCC management, providing opportunities for improved prevention, early detection, response to therapies, and QOL. Bioinformatics and computational systems biology approaches are key to integrate redox effects with multiomics data from cells and clinical specimens and to identify redox modifiers or modifiable target proteins to achieve improved clinical outcomes. Antioxid. Redox Signal.
Collapse
Affiliation(s)
- Xiaofei Chen
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jade Mims
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Xiumei Huang
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Naveen Singh
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Edward Motea
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | | | - Muhammad Beg
- Department of Internal Medicine, Division of Hematology-Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Allen W. Tsang
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Mercedes Porosnicu
- Department of Internal Medicine, Section of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Melissa L. Kemp
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - David A. Boothman
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Cristina M. Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
22
|
Li W, Quan YY, Li Y, Lu L, Cui M. Monitoring of tumor vascular normalization: the key points from basic research to clinical application. Cancer Manag Res 2018; 10:4163-4172. [PMID: 30323672 PMCID: PMC6175544 DOI: 10.2147/cmar.s174712] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Tumor vascular normalization alleviates hypoxia in the tumor microenvironment, reduces the degree of malignancy, and increases the efficacy of traditional therapy. However, the time window for vascular normalization is narrow; therefore, how to determine the initial and final points of the time window accurately is a key factor in combination therapy. At present, the gold standard for detecting the normalization of tumor blood vessels is histological staining, including tumor perfusion, microvessel density (MVD), vascular morphology, and permeability. However, this detection method is almost unrepeatable in the same individual and does not dynamically monitor the trend of the time window; therefore, finding a relatively simple and specific monitoring index has important clinical significance. Imaging has long been used to assess changes in tumor blood vessels and tumor changes caused by the oxygen environment in clinical practice; some preclinical and clinical research studies demonstrate the feasibility to assess vascular changes, and some new methods were in preclinical research. In this review, we update the most recent insights of evaluating tumor vascular normalization.
Collapse
Affiliation(s)
- Wei Li
- Department of General Surgery, Zhuhai People's Hospital, Jinan University, Zhuhai, Guangdong, People's Republic of China,
| | - Ying-Yao Quan
- Department of Precision Medical Center, Zhuhai People's Hospital, Jinan University, Zhuhai, Guangdong, People's Republic of China
| | - Yong Li
- Department of Intervention, Zhuhai People's Hospital, Jinan University, Zhuhai, Guangdong, People's Republic of China,
| | - Ligong Lu
- Department of Intervention, Zhuhai People's Hospital, Jinan University, Zhuhai, Guangdong, People's Republic of China,
| | - Min Cui
- Department of General Surgery, Zhuhai People's Hospital, Jinan University, Zhuhai, Guangdong, People's Republic of China,
| |
Collapse
|
23
|
Melzig C, Golestaneh AF, Mier W, Schwager C, Das S, Schlegel J, Lasitschka F, Kauczor HU, Debus J, Haberkorn U, Abdollahi A. Combined external beam radiotherapy with carbon ions and tumor targeting endoradiotherapy. Oncotarget 2018; 9:29985-30004. [PMID: 30042828 PMCID: PMC6057461 DOI: 10.18632/oncotarget.25695] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 06/04/2018] [Indexed: 01/05/2023] Open
Abstract
External beam radiotherapy (EBRT) with carbon ions and endoradiotherapy using radiolabeled tumor targeting agents are emerging concepts in precision cancer therapy. We report on combination effects of these two promising strategies. Tumor targeting 131I-labelled anti-EGFR-antibody (Cetuximab) was used in the prototypic EGFR-expressing A431 human squamous cell carcinoma xenograft model. A 131I-labelled melanin-binding benzamide derivative was utilized targeting B16F10 melanoma in an orthotopic syngeneic C57bl6 model. Fractionated EBRT was performed using carbon ions in direct comparison with conventional photon irradiation. Tumor uptake of 131I-Cetuximab and 131I-Benzamide was enhanced by fractionated EBRT as determined by biodistribution studies. This effect was independent of radiation quality and significant for the small molecule 131I-Benzamide, i.e., >30% more uptake in irradiated vs. non-irradiated melanoma was found (p<0.05). Compared to each monotherapy, dual combination with 131I-Cetuximab and EBRT was most effective in inhibiting A431 tumor growth. A similar trend was seen for 131I-Benzamide and EBRT in B16F10 melanoma model. Addition of 131I-Benzamide endoradiotherapy to EBRT altered expression of genes related to DNA-repair, cell cycle and cell death. In contrast, immune-response related pathways such as type 1 interferon response genes (ISG15, MX1) were predominantly upregulated after combined 131I-Cetuximab and EBRT. The beneficial effects of combined 131I-Cetuximab and EBRT was further attributed to a reduced microvascular density (CD31) and decreased proliferation index (Ki-67). Fractionated EBRT could be favorably combined with endoradiotherapy. 131I-Benzamide endoradiotherapy accelerated EBRT induced cytotoxic effects. Activation of immune-response by carbon ions markedly enhanced anti-EGFR based endoradiotherapy suggesting further evaluation of this novel and promising radioimmunotherapy concept.
Collapse
Affiliation(s)
- Claudius Melzig
- German Cancer Consortium, Heidelberg, Germany.,Translational Radiation Oncology, National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany.,Division of Molecular and Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology, National Center for Radiation Research in Oncology, Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Azadeh Fahim Golestaneh
- German Cancer Consortium, Heidelberg, Germany.,Translational Radiation Oncology, National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany.,Division of Molecular and Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology, National Center for Radiation Research in Oncology, Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Walter Mier
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center, Heidelberg, Germany
| | - Christian Schwager
- German Cancer Consortium, Heidelberg, Germany.,Translational Radiation Oncology, National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany.,Division of Molecular and Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology, National Center for Radiation Research in Oncology, Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Samayita Das
- German Cancer Consortium, Heidelberg, Germany.,Translational Radiation Oncology, National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany.,Division of Molecular and Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology, National Center for Radiation Research in Oncology, Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Julian Schlegel
- German Cancer Consortium, Heidelberg, Germany.,Translational Radiation Oncology, National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany.,Division of Molecular and Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology, National Center for Radiation Research in Oncology, Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Felix Lasitschka
- Department of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Hans-Ulrich Kauczor
- Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jürgen Debus
- German Cancer Consortium, Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center, Heidelberg, Germany
| | - Amir Abdollahi
- German Cancer Consortium, Heidelberg, Germany.,Translational Radiation Oncology, National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany.,Division of Molecular and Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology, National Center for Radiation Research in Oncology, Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
24
|
Abstract
Evofosfamide, also formerly known as TH-302, is an investigational hypoxia-activated prodrug and is used to target cancerous cells under hypoxic conditions, which is a feature possessed by multiple solid tumors including pancreatic tumors. Gemcitabine, a cytotoxic agent, has for many years been the standard first-line treatment for metastatic pancreatic cancer in patients. In recent years, combination chemotherapeutic therapies have provided a new avenue for molecular targeting by increasing the probability of eliminating the cancer and minimizing the likelihood of resistance. We have evaluated multiple studies in an effort to shed light on an emerging prodrug, evofosfamide, which operates by selectively targeting the tumor hypoxic compartment. A web-based literature search was performed through PubMed and Google Scholar using the keywords 'evofosfamide', 'TH-302,' and 'pancreatic tumor.' Of the available results, 53 relevant studies were reviewed and summarized. Chemotherapeutic agents such as evofosfamide, which targets tumor hypoxia, are new agents against cancer cells. Current experience with these agents is limited as additional and longer prospective studies are needed to further evaluate the clinical efficacy and postmarketing safety profile.
Collapse
|
25
|
|
26
|
Siemann DW, Chaplin DJ, Horsman MR. Realizing the Potential of Vascular Targeted Therapy: The Rationale for Combining Vascular Disrupting Agents and Anti-Angiogenic Agents to Treat Cancer. Cancer Invest 2017; 35:519-534. [DOI: 10.1080/07357907.2017.1364745] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- D. W. Siemann
- Department of Radiation Oncology, University of Florida, Gainesville, FL, USA
| | | | - M. R. Horsman
- Department of Experimental Clinical Oncology, Aarhus University, Denmark
| |
Collapse
|
27
|
Murray L, Longo J, Wan J, Chung C, Wang L, Dawson L, Milosevic M, Oza A, Brade A. Phase I dose escalation study of concurrent palliative radiation therapy with sorafenib in three anatomical cohorts (Thorax, Abdomen, Pelvis): The TAP study. Radiother Oncol 2017; 124:74-79. [PMID: 28668472 DOI: 10.1016/j.radonc.2017.06.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/28/2016] [Accepted: 06/05/2017] [Indexed: 01/29/2023]
Abstract
BACKGROUND AND PURPOSE To evaluate the tolerability and maximum tolerated dose (MTD) of sorafenib administered concurrently with palliative radiotherapy. MATERIAL AND METHODS In patients with incurable cancer, sorafenib was escalated independently in three cohorts based on irradiation site: thorax, abdomen or pelvis. Sorafenib was administered days 1-28 and radiotherapy (30Gy in 10 fractions) was delivered days 8-12 and 15-19. Dose-limiting toxicities (DLT) were acute grade 3+ toxicities attributable to radiotherapy. RESULTS For the thorax, abdomen and pelvis cohorts, 14, 16 and 4 patients were recruited, and Dose Levels 3, 3 and 2 were reached, respectively. Sorafenib-related systemic toxicity led to significant sorafenib interruption in 10 patients. There were 3 DLTs in total, one per cohort: grade 3 oesophagitis (thoracic), transaminase elevation (abdominal) and grade 5 bowel perforation (pelvic; patient with tumour invading bowel). Grade 2 radiation dermatitis developed in 12 patients. The trial was terminated early as slow accrual and sorafenib-related systemic toxicity prevented efficient evaluation of RT-related DLTs. CONCLUSIONS The MTD of sorafenib when used with 30Gy in 10 fractions was not established due to sorafenib-related systemic toxicity. Severe radiotherapy-related toxicities were also observed. These events suggest this concurrent combination does not warrant further study.
Collapse
Affiliation(s)
- Louise Murray
- Department of Radiation Oncology, University of Toronto, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Joseph Longo
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Canada
| | - Jonathan Wan
- Department of Radiation Oncology, University of Toronto, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Caroline Chung
- Department of Radiation Oncology, University of Toronto, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Lisa Wang
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Canada
| | - Laura Dawson
- Department of Radiation Oncology, University of Toronto, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Michael Milosevic
- Department of Radiation Oncology, University of Toronto, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Amit Oza
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Anthony Brade
- Department of Radiation Oncology, University of Toronto, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.
| |
Collapse
|
28
|
Phase I trial of radiation therapy and sorafenib in unresectable liver metastases. Radiother Oncol 2017; 123:234-239. [PMID: 28202227 DOI: 10.1016/j.radonc.2017.01.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 01/25/2017] [Accepted: 01/25/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND PURPOSE To determine maximum tolerated dose (MTD) and toxicities of sorafenib combined with stereotactic radiotherapy (SBRT) or whole liver radiotherapy (WLRT) in patients with liver metastases. MATERIAL AND METHODS Eligible patients had unresectable liver metastases. Sorafenib dose was escalated in 2 strata: I - SBRT: effective liver volume irradiated (Veff)<80% (30-60Gy in 6 fractions); II - WLRT: Veff>80% (21.6Gy in 6 fractions). Four weeks of sorafenib, with radiotherapy during weeks 2-3, was delivered at 3 escalating dose levels (200-400mg twice daily). Dose limiting toxicity was defined as any grade 3+ liver toxicity, or grade 4+ treatment-related toxicity. RESULTS Thirty-three patients were treated: 18 in stratum I (median dose 42Gy), 15 in stratum II. The MTD was not reached. Grade 3+ toxicity was seen in 33% of patients, at a median of 10days. Two deaths from non-classic liver toxicity occurred post WLRT in stratum II. The median overall survival was 22.3 and 5.7months for strata I and II respectively. CONCLUSIONS Sorafenib and 21.6Gy in 6 fraction WLRT resulted in unacceptably high rates of liver toxicity. Although sorafenib combined with SBRT was tolerable, the observed efficacy does not merit further clinical evaluation.
Collapse
|
29
|
Spatiotemporal distribution modeling of PET tracer uptake in solid tumors. Ann Nucl Med 2016; 31:109-124. [PMID: 27921285 DOI: 10.1007/s12149-016-1141-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 10/18/2016] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Distribution of PET tracer uptake is elaborately modeled via a general equation used for solute transport modeling. This model can be used to incorporate various transport parameters of a solid tumor such as hydraulic conductivity of the microvessel wall, transvascular permeability as well as interstitial space parameters. This is especially significant because tracer delivery and drug delivery to solid tumors are determined by similar underlying tumor transport phenomena, and quantifying the former can enable enhanced prediction of the latter. METHODS We focused on the commonly utilized FDG PET tracer. First, based on a mathematical model of angiogenesis, the capillary network of a solid tumor and normal tissues around it were generated. The coupling mathematical method, which simultaneously solves for blood flow in the capillary network as well as fluid flow in the interstitium, is used to calculate pressure and velocity distributions. Subsequently, a comprehensive spatiotemporal distribution model (SDM) is applied to accurately model distribution of PET tracer uptake, specifically FDG in this work, within solid tumors. RESULTS The different transport mechanisms, namely convention and diffusion from vessel to tissue and in tissue, are elaborately calculated across the domain of interest and effect of each parameter on tracer distribution is investigated. The results show the convection terms to have negligible effect on tracer transport and the SDM can be solved after eliminating these terms. CONCLUSION The proposed framework of spatiotemporal modeling for PET tracers can be utilized to comprehensively assess the impact of various parameters on the spatiotemporal distribution of PET tracers.
Collapse
|
30
|
Kim EH, Lee H, Jeong YK, Jung WG. Mechanisms of SU5416, an inhibitor of vascular endothelial growth factor receptor, as a radiosensitizer for colon cancer cells. Oncol Rep 2016; 36:763-70. [DOI: 10.3892/or.2016.4868] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/26/2016] [Indexed: 11/06/2022] Open
|
31
|
Esposito G, Gigli S, Seguella L, Nobile N, D'Alessandro A, Pesce M, Capoccia E, Steardo L, Cirillo C, Cuomo R, Sarnelli G. Rifaximin, a non-absorbable antibiotic, inhibits the release of pro-angiogenic mediators in colon cancer cells through a pregnane X receptor-dependent pathway. Int J Oncol 2016; 49:639-45. [PMID: 27279570 DOI: 10.3892/ijo.2016.3550] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 03/03/2016] [Indexed: 11/05/2022] Open
Abstract
Activation of intestinal human pregnane X receptor (PXR) has recently been proposed as a promising strategy for the chemoprevention of inflammation-induced colon cancer. The present study was aimed at evaluating the effect of rifaximin, a non-absorbable antibiotic, in inhibiting angiogenesis in a model of human colorectal epithelium and investigating the role of PXR in its mechanism of action. Caco-2 cells were treated with rifaximin (0.1, 1.0 and 10.0 µM) in the presence or absence of ketoconazole (10 µM) and assessed for cell proliferation, migration and expression of proliferating cell nuclear antigen (PCNA). The release of vascular endothelial growth factor (VEGF) and nitric oxide (NO), expression of Akt, mechanistic target of rapamycin (mTOR), p38 mitogen activated protein kinases (MAPK), nuclear factor κB (NF-κB) and metalloproteinase-2 and -9 (MMP-2 and -9) were also evaluated. Treatment with rifaximin 0.1, 1.0 and 10.0 µM caused significant and concentration-dependent reduction of cell proliferation, cell migration and PCNA expression in the Caco-2 cells vs. untreated cells. Treatment downregulated VEGF secretion, NO release, VEGFR-2 expression, MMP-2 and MMP-9 expression vs. untreated cells. Rifaximin treatment also resulted in a concentration-dependent decrease in the phosphorylation of Akt, mTOR, p38MAPK and inhibition of hypoxia-inducible factor 1-α (HIF-1α), p70S6K and NF-κB. Ketoconazole (PXR antagonist) treatment inhibited these effects. These findings demonstrated that rifaximin causes PXR-mediated inhibition of angiogenic factors in Caco-2 cell line and may be a promising anticancer tool.
Collapse
Affiliation(s)
- Giuseppe Esposito
- Department of Physiology and Pharmacology, 'Vittorio Erspamer', La Sapienza University of Rome, I-00185 Rome, Italy
| | - Stefano Gigli
- Department of Physiology and Pharmacology, 'Vittorio Erspamer', La Sapienza University of Rome, I-00185 Rome, Italy
| | - Luisa Seguella
- Department of Physiology and Pharmacology, 'Vittorio Erspamer', La Sapienza University of Rome, I-00185 Rome, Italy
| | - Nicola Nobile
- Department of Physiology and Pharmacology, 'Vittorio Erspamer', La Sapienza University of Rome, I-00185 Rome, Italy
| | - Alessandra D'Alessandro
- Department of Clinical Medicine and Surgery, 'Federico II' University of Naples, I-80131 Naples, Italy
| | - Marcella Pesce
- Department of Clinical Medicine and Surgery, 'Federico II' University of Naples, I-80131 Naples, Italy
| | - Elena Capoccia
- Department of Physiology and Pharmacology, 'Vittorio Erspamer', La Sapienza University of Rome, I-00185 Rome, Italy
| | - Luca Steardo
- Department of Physiology and Pharmacology, 'Vittorio Erspamer', La Sapienza University of Rome, I-00185 Rome, Italy
| | - Carla Cirillo
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, 3000 Leuven, Belgium
| | - Rosario Cuomo
- Department of Clinical Medicine and Surgery, 'Federico II' University of Naples, I-80131 Naples, Italy
| | - Giovanni Sarnelli
- Department of Clinical Medicine and Surgery, 'Federico II' University of Naples, I-80131 Naples, Italy
| |
Collapse
|
32
|
Glasgow MDK, Chougule MB. Recent Developments in Active Tumor Targeted Multifunctional Nanoparticles for Combination Chemotherapy in Cancer Treatment and Imaging. J Biomed Nanotechnol 2016; 11:1859-98. [PMID: 26554150 DOI: 10.1166/jbn.2015.2145] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nanotechnology and combination therapy are two major fields that show great promise in the treatment of cancer. The delivery of drugs via nanoparticles helps to improve drug's therapeutic effectiveness while reducing adverse side effects associated wifh high dosage by improving their pharmacokinetics. Taking advantage of molecular markers over-expressing on tumor tissues compared to normal cells, an "active" molecular marker targeted approach would be-beneficial for cancer therapy. These actively targeted nanoparticles would increase drug concentration at the tumor site, improving efficacy while further reducing chemo-resistance. The multidisciplinary approach may help to improve the overall efficacy in cancer therapy. This review article summarizes recent developments of targeted multifunctional nanoparticles in the delivery, of various drugs for a combinational chemotherapy approach to cancer treatment and imaging.
Collapse
|
33
|
Chen L, Zhong X, Yi X, Huang M, Ning P, Liu T, Ge C, Chai Z, Liu Z, Yang K. Radionuclide 131I labeled reduced graphene oxide for nuclear imaging guided combined radio- and photothermal therapy of cancer. Biomaterials 2015; 66:21-8. [DOI: 10.1016/j.biomaterials.2015.06.043] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 06/22/2015] [Accepted: 06/23/2015] [Indexed: 01/07/2023]
|
34
|
Kim EH, Kim MS, Jeong YK, Cho I, You SH, Cho SH, Lee H, Jung WG, Kim HD, Kim J. Mechanisms for SU5416 as a radiosensitizer of endothelial cells. Int J Oncol 2015; 47:1440-50. [PMID: 26314590 DOI: 10.3892/ijo.2015.3127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 06/22/2015] [Indexed: 11/06/2022] Open
Abstract
Endothelial cells (ECs), that comprise the tumor vasculature, are critical targets for anticancer radiotherapy. The aim of this work was to study the mechanism by which SU5416, a known anti-angiogenesis inhibitor, modifies the radiation responses of human vascular ECs. Two human endothelial cell lines (HUVEC and 2H11) were treated with SU5416 alone, radiation alone, or a combination of both. In vitro tests were performed using colony forming assays, FACS analysis, western blotting, immunohistochemistry, migration assay, invasion assays and endothelial tube formation assays. The combination of radiation and SU5416 significantly inhibited cell survival, the repair of radiation-induced DNA damage, and induced apoptosis. It also caused cell cycle arrest, inhibited cell migration and invasion, and suppressed angiogenesis. In this study, our results first provide a scientific rationale to combine SU5416 with radiotherapy to target ECs and suggest its clinical application in combination cancer treatment with radiotherapy.
Collapse
Affiliation(s)
- Eun Ho Kim
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Mi-Sook Kim
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Youn Kyoung Jeong
- Research Center for Radiotherapy, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Ilsung Cho
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Seung Hoon You
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Sung Ho Cho
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Hanna Lee
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Won-Gyun Jung
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Hag Dong Kim
- Laboratory of Biochemistry, Division of Life Sciences, Korea University, Seoul 136-701, Republic of Korea
| | - Joon Kim
- Laboratory of Biochemistry, Division of Life Sciences, Korea University, Seoul 136-701, Republic of Korea
| |
Collapse
|
35
|
Effect of fluid friction on interstitial fluid flow coupled with blood flow through solid tumor microvascular network. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2015; 2015:673426. [PMID: 25960764 PMCID: PMC4417563 DOI: 10.1155/2015/673426] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 03/29/2015] [Indexed: 12/31/2022]
Abstract
A solid tumor is investigated as porous media for fluid flow simulation. Most of the studies use Darcy model for porous media. In Darcy model, the fluid friction is neglected and a few simplified assumptions are implemented. In this study, the effect of these assumptions is studied by considering Brinkman model. A multiscale mathematical method which calculates fluid flow to a solid tumor is used in this study to investigate how neglecting fluid friction affects the solid tumor simulation. The mathematical method involves processes such as blood flow through vessels and solute and fluid diffusion, convective transport in extracellular matrix, and extravasation from blood vessels. The sprouting angiogenesis model is used for generating capillary network and then fluid flow governing equations are implemented to calculate blood flow through the tumor-induced capillary network. Finally, the two models of porous media are used for modeling fluid flow in normal and tumor tissues in three different shapes of tumors. Simulations of interstitial fluid transport in a solid tumor demonstrate that the simplifications used in Darcy model affect the interstitial velocity and Brinkman model predicts a lower value for interstitial velocity than the values that Darcy model predicts.
Collapse
|
36
|
Sefidgar M, Soltani M, Raahemifar K, Sadeghi M, Bazmara H, Bazargan M, Mousavi Naeenian M. Numerical modeling of drug delivery in a dynamic solid tumor microvasculature. Microvasc Res 2015; 99:43-56. [PMID: 25724978 DOI: 10.1016/j.mvr.2015.02.007] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 12/24/2014] [Accepted: 02/11/2015] [Indexed: 01/01/2023]
Abstract
The complicated capillary network induced by angiogenesis is one of the main reasons of unsuccessful cancer therapy. A multi-scale mathematical method which simulates drug transport to a solid tumor is used in this study to investigate how capillary network structure affects drug delivery. The mathematical method involves processes such as blood flow through vessels, solute and fluid diffusion, convective transport in extracellular matrix, and extravasation from blood vessels. The effect of heterogeneous dynamic network on interstitial fluid flow and drug delivery is investigated by this multi-scale method. The sprouting angiogenesis model is used for generating capillary network and then fluid flow governing equations are implemented to calculate blood flow through the tumor-induced capillary network and fluid flow in normal and tumor tissues. Finally, convection-diffusion equation is used to simulate drug delivery. Three approaches are used to simulate drug transport based on the developed mathematical method: without a vascular network, using a static vascular network, and a dynamic vascular network. The avascular approach predicts more uniform and higher drug concentration than vascular approaches since the simplified assumptions are implemented in this method. The dynamic network which uses more realistic assumptions predicts more irregular blood vessels, high interstitial pressure, and more heterogeneity in drug distribution than other two approaches.
Collapse
Affiliation(s)
- M Sefidgar
- Department of Mechanical Engineering, K.N.T. University of Technology, Tehran, Iran.
| | - M Soltani
- Department of Mechanical Engineering, K.N.T. University of Technology, Tehran, Iran; Division of Nuclear Medicine, Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, MD, USA.
| | - K Raahemifar
- Electrical & Computer Department of Ryerson University, Toronto, Ontario, Canada.
| | - M Sadeghi
- Digital Health Hub, Simon Fraser University, Surrey, BC, Canada.
| | - H Bazmara
- Department of Mechanical Engineering, K.N.T. University of Technology, Tehran, Iran.
| | - M Bazargan
- Department of Mechanical Engineering, K.N.T. University of Technology, Tehran, Iran.
| | - M Mousavi Naeenian
- Department of Mechanical Engineering, K.N.T. University of Technology, Tehran, Iran.
| |
Collapse
|
37
|
Mehmood RK. Review of Cisplatin and oxaliplatin in current immunogenic and monoclonal antibody treatments. Oncol Rev 2014; 8:256. [PMID: 25992242 PMCID: PMC4419649 DOI: 10.4081/oncol.2014.256] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 08/21/2014] [Indexed: 02/06/2023] Open
Abstract
Platinum-based chemotherapy agents initially transformed cancer treatment. However their effectiveness peaked as combined regimes showed little additional benefit in trials. New research frontiers developed with the discovery that conventional chemotherapy can induce immunological cell death by recruiting high mobility group box 1 protein through T-cell immunity. Simultaneously monoclonal antibody agents (not effective as monotherapies) showed good results in combination with conventional chemotherapy. Some of these combinations are currently in use and researchers hope to develop regimes which can offer substantial benefits. Several resistance mechanisms against platinum compounds are known, but more knowledge is still needed to gain a full understanding. It seems reasonable therefore to revisit the pharmacology of these agents, which may also lead to identify rational combinations with monoclonal agents providing regimes with less toxicity and better efficacy. This article reviews the pharmacology of cisplatin and oxaliplatin and explores their possible association with monoclonal antibody treatments.
Collapse
Affiliation(s)
- Rao Khalid Mehmood
- Department of Colorectal and General Surgery, University Board Hospital , Rhyl, North Wales, UK
| |
Collapse
|
38
|
van Noort V, Schölch S, Iskar M, Zeller G, Ostertag K, Schweitzer C, Werner K, Weitz J, Koch M, Bork P. Novel drug candidates for the treatment of metastatic colorectal cancer through global inverse gene-expression profiling. Cancer Res 2014; 74:5690-9. [PMID: 25038229 DOI: 10.1158/0008-5472.can-13-3540] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Drug-induced gene-expression profiles that invert disease profiles have recently been illustrated to be a starting point for drug repositioning. In this study, we validate this approach and focus on prediction of novel drugs for colorectal cancer, for which there is a pressing need to find novel antimetastatic compounds. We computationally predicted three novel and still unknown compounds against colorectal cancer: citalopram (an antidepressant), troglitazone (an antidiabetic), and enilconazole (a fungicide). We verified the compounds by in vitro assays of clonogenic survival, proliferation, and migration and in a subcutaneous mouse model. We found evidence that the mode of action of these compounds may be through inhibition of TGFβ signaling. Furthermore, one compound, citalopram, reduced tumor size as well as the number of circulating tumor cells and metastases in an orthotopic mouse model of colorectal cancer. This study proposes citalopram as a potential therapeutic option for patients with colorectal cancer, illustrating the potential of systems pharmacology.
Collapse
Affiliation(s)
- Vera van Noort
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Meyerhofstrasse, Heidelberg, Germany. Centre of Microbial and Plant Genetics, KU Leuven, Kasteelpark Arenberg, Leuven, Belgium
| | - Sebastian Schölch
- Department of Gastrointestinal, Thoracic and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Murat Iskar
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Meyerhofstrasse, Heidelberg, Germany
| | - Georg Zeller
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Meyerhofstrasse, Heidelberg, Germany
| | - Kristina Ostertag
- Department of General, Gastrointestinal and Transplant Surgery, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Christine Schweitzer
- Department of Gastrointestinal, Thoracic and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Kristin Werner
- Department of Gastrointestinal, Thoracic and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jürgen Weitz
- Department of Gastrointestinal, Thoracic and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Moritz Koch
- Department of Gastrointestinal, Thoracic and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany. Moritz.Koch@uniklinikum
| | - Peer Bork
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Meyerhofstrasse, Heidelberg, Germany. Max-Delbrück-Centre (MDC) for Molecular Medicine, Berlin, Germany. Moritz.Koch@uniklinikum
| |
Collapse
|
39
|
Mehmood RK, Parker J, Ahmed S, Qasem E, Mohammed AA, Zeeshan M, Jehangir E. Review of Cisplatin and Oxaliplatin in Current Immunogenic and Monoclonal Antibodies Perspective. World J Oncol 2014; 5:97-108. [PMID: 29147386 PMCID: PMC5649811 DOI: 10.14740/wjon830w] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2014] [Indexed: 12/25/2022] Open
Abstract
Platinum-based chemotherapy made a paradigm shift in the treatment of different cancers initially; however, the success of these agents may have reached the peak as researchers have tried different combination regimes in different trials without having major differences in the end results. New frontiers of research were opened up firstly with this discovery that conventional chemo-radiation therapy can induce immunological cell death by recruiting high-mobility group box 1 (HMGB1) protein which triggers the T cell immunity and secondly monoclonal antibodies agents which were regrettably not effective as “monotherapy”; however, the combination with conventional chemotherapy had demonstrated good results. Different monoclonal antibodies and conventional chemotherapeutic combination regimes are currently in use and researchers are trying different other combinations as well to glean the maximum benefits from them. Several strategies conferring resistance to platinum compounds have been identified, but there is still significant research required to achieve full understanding of these resistance mechanisms to overcome the ineffectiveness or toxicities of platinum compounds. It seems reasonable in the current perspective when conventional chemotherapeutic agents exhibited immunogenic cell death and they are currently in use with monoclonal antibodies to revisit the platinum agent’s pharmacology. This may discover new basis for combination chemotherapy with monoclonal antibodies which may improve the current cancer treatments by opening new vistas for newer combination regimes with less toxicity and better efficacy. In this article we review the pharmacologies of both cisplatin and oxaliplatin in the drug development perspectives and explore the possible association of these drugs with monoclonal antibodies.
Collapse
Affiliation(s)
- Rao Khalid Mehmood
- Betsi Cadwaladr University Health Board, Department of Surgery, Ysbyty Glan Clwyd, Rhyl, North Wales, LL18 5UJ, UK
| | - Jody Parker
- Betsi Cadwaladr University Health Board, Department of Surgery, Ysbyty Glan Clwyd, Rhyl, North Wales, LL18 5UJ, UK
| | - Shakil Ahmed
- The Royal Liverpool and Broadgreen University Hospitals NHS Trust, Prescot Street, Liverpool, L7 8XP, UK
| | - Eyas Qasem
- Betsi Cadwaladr University Health Board, Department of Surgery, Ysbyty Glan Clwyd, Rhyl, North Wales, LL18 5UJ, UK
| | - Ahmed A Mohammed
- Betsi Cadwaladr University Health Board, Department of Surgery, Ysbyty Glan Clwyd, Rhyl, North Wales, LL18 5UJ, UK
| | - Muhammed Zeeshan
- Acute University Hospitals NHS Trust, Cumberland Infirmary Carlisle, Newtown Rd, Carlisle, Cumbria, CA2 7HY, UK
| | - Ernest Jehangir
- Acute University Hospitals NHS Trust, Cumberland Infirmary Carlisle, Newtown Rd, Carlisle, Cumbria, CA2 7HY, UK
| |
Collapse
|
40
|
Effect of tumor shape, size, and tissue transport properties on drug delivery to solid tumors. J Biol Eng 2014; 8:12. [PMID: 24987457 PMCID: PMC4076317 DOI: 10.1186/1754-1611-8-12] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 05/30/2014] [Indexed: 12/21/2022] Open
Abstract
Background The computational methods provide condition for investigation related to the process of drug delivery, such as convection and diffusion of drug in extracellular matrices, drug extravasation from microvessels or to lymphatic vessels. The information of this process clarifies the mechanisms of drug delivery from the injection site to absorption by a solid tumor. In this study, an advanced numerical method is used to solve fluid flow and solute transport equations simultaneously to investigate the effect of tumor shape and size on drug delivery to solid tumor. Methods The advanced mathematical model used in our previous work is further developed by adding solute transport equation to the governing equations. After applying appropriate boundary and initial conditions on tumor and surrounding tissue geometry, the element-based finite volume method is used for solving governing equations of drug delivery in solid tumor. Also, the effects of size and shape of tumor and some of tissue transport parameters such as effective pressure and hydraulic conductivity on interstitial fluid flow and drug delivery are investigated. Results Sensitivity analysis shows that drug delivery in prolate shape is significantly better than other tumor shapes. Considering size effect, increasing tumor size decreases drug concentration in interstitial fluid. This study shows that dependency of drug concentration in interstitial fluid to osmotic and intravascular pressure is negligible. Conclusions This study shows that among diffusion and convection mechanisms of drug transport, diffusion is dominant in most different tumor shapes and sizes. In tumors in which the convection has considerable effect, the drug concentration is larger than that of other tumors at the same time post injection.
Collapse
|
41
|
Nicolay NH, Sommer E, Perez RL, Wirkner U, Bostel T, Ho AD, Lahn M, Debus J, Saffrich R, Huber PE. Mesenchymal stem cells are sensitive to treatment with kinase inhibitors and ionizing radiation. Strahlenther Onkol 2014; 190:1037-45. [DOI: 10.1007/s00066-014-0686-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 04/28/2014] [Indexed: 10/25/2022]
|
42
|
Farajpour Z, Rahbarizadeh F, Kazemi B, Ahmadvand D. A nanobody directed to a functional epitope on VEGF, as a novel strategy for cancer treatment. Biochem Biophys Res Commun 2014; 446:132-6. [PMID: 24569074 DOI: 10.1016/j.bbrc.2014.02.069] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 02/15/2014] [Indexed: 11/16/2022]
Abstract
Compelling evidence suggests that vascular endothelial growth factor (VEGF), due to its essential role in angiogenesis, is a critical target for cancer treatment. Neutralizing monoclonal antibodies against VEGF are important class of drugs used in cancer therapy. However, the cost of production, large size, and immunogenicity are main drawbacks of conventional monoclonal therapy. Nanobodies are the smallest antigen-binding antibody fragments, which occur naturally in camelidae. Because of their remarkable features, we decided to use an immune library of nanobody to direct phage display to recognition of novel functional epitopes on VEGF. Four rounds of selection were performed and six phage-displayed nanobodies were obtained from an immune phage library. The most reactive clone in whole-cell ELISA experiments, was purified and assessed in proliferation inhibition assay. Purified ZFR-5 not only blocked interaction of VEGF with its receptor in cell ELISA experiments, but also was able to significantly inhibit proliferation response of human umbilical vein endothelial cells to VEGF in a dose-dependent manner. Taken together, our study demonstrates that by using whole-cell ELISA experiments, nanobodies against antigenic regions included in interaction of VEGF with its receptors can be directed. Because of unique and intrinsic properties of a nanobody and the ability of selected nanobody for blocking the epitope that is important for biological function of VEGF, it represents novel potential drug candidate.
Collapse
Affiliation(s)
- Zahra Farajpour
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Bahram Kazemi
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davoud Ahmadvand
- School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
43
|
Weiss A, van Beijnum JR, Bonvin D, Jichlinski P, Dyson PJ, Griffioen AW, Nowak-Sliwinska P. Low-dose angiostatic tyrosine kinase inhibitors improve photodynamic therapy for cancer: lack of vascular normalization. J Cell Mol Med 2014; 18:480-91. [PMID: 24450440 PMCID: PMC3955154 DOI: 10.1111/jcmm.12199] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 10/25/2013] [Indexed: 12/30/2022] Open
Abstract
Photodynamic therapy (PDT) is an effective clinical treatment for a number of different cancers. PDT can induce hypoxia and inflammation, pro-angiogenic side effects, which may counteract its angio-occlusive mechanism. The combination of PDT with anti-angiogenic drugs offers a possibility for improved anti-tumour outcome. We used two tumour models to test the effects of the clinically approved angiostatic tyrosine kinase inhibitors sunitinib, sorafenib and axitinib in combination with PDT, and compared these results with the effects of bevacizumab, the anti-VEGF antibody, for the improvement of PDT. Best results were obtained from the combination of PDT and low-dose axitinib or sorafenib. Molecular analysis by PCR revealed that PDT in combination with axitinib suppressed VEGFR-2 expression in tumour vasculature. Treatment with bevacizumab, although effective as monotherapy, did not improve PDT outcome. In order to test for tumour vessel normalization effects, axitinib was also applied prior to PDT. The absence of improved PDT outcome in these experiments, as well as the lack of increased oxygenation in axitinib-treated tumours, suggests that vascular normalization did not occur. The current data imply that there is a future for certain anti-angiogenic agents to further improve the efficacy of photodynamic anti-cancer therapy.
Collapse
Affiliation(s)
- Andrea Weiss
- Medical Photonics Group, Institute of Bioengineering, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland; Angiogenesis Laboratory, Department of Medical Oncology, VU Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
44
|
Nagaraja TN, Aryal MP, Brown SL, Bagher-Ebadian H, Mikkelsen T, Yang JJ, Panda S, Keenan KA, Cabral G, Ewing JR. Cilengitide-induced temporal variations in transvascular transfer parameters of tumor vasculature in a rat glioma model: identifying potential MRI biomarkers of acute effects. PLoS One 2013; 8:e84493. [PMID: 24376814 PMCID: PMC3871527 DOI: 10.1371/journal.pone.0084493] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 11/14/2013] [Indexed: 11/30/2022] Open
Abstract
Increased efficacy of radiotherapy (RT) 4-8 h after Cilengitide treatment has been reported. We hypothesized that the effects of Cilengitide on tumor transvascular transfer parameters might underlie, and thus predict, this potentiation. Athymic rats with orthotopic U251 glioma were studied at ~21 days after implantation using dynamic contrast-enhanced (DCE)-MRI. Vascular parameters, viz: plasma volume fraction (vp), forward volume transfer constant (Ktrans) and interstitial volume fraction (ve) of a contrast agent, were determined in tumor vasculature once before, and again in cohorts 2, 4, 8, 12 and 24 h after Cilengitide administration (4 mg/kg; N = 31; 6-7 per cohort). Perfusion-fixed brain sections were stained for von Willebrand factor to visualize vascular segments. A comparison of pre- and post-treatment parameters showed that the differences between MR indices before and after Cilengitide treatment pivoted around the 8 h time point, with 2 and 4 h groups showing increases, 12 and 24 h groups showing decreases, and values at the 8 h time point close to the baseline. The vascular parameter differences between group of 2 and 4 h and group of 12 and 24 h were significant for Ktrans (p = 0.0001 and ve (p = 0,0271). Vascular staining showed little variation with time after Cilengitide. The vascular normalization occurring 8 h after Cilengitide treatment coincided with similar previous reports of increased treatment efficacy when RT followed Cilengitide by 8 h. Pharmacological normalization of vasculature has the potential to increase sensitivity to RT. Evaluating acute temporal responses of tumor vasculature to putative anti-angiogenic drugs may help in optimizing their combination with other treatment modalities.
Collapse
Affiliation(s)
- Tavarekere N. Nagaraja
- Department of Anesthesiology, Henry Ford Hospital, Detroit, Michigan, United States of America
- * E-mail:
| | - Madhava P. Aryal
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, United States of America
- Department of Physics, Oakland University, Rochester, Michigan, United States of America
| | - Stephen L. Brown
- Department of Radiation Oncology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Hassan Bagher-Ebadian
- Department of Diagnostic Radiology, Henry Ford Hospital, Detroit, Michigan, United States of America
- Department of Physics, Oakland University, Rochester, Michigan, United States of America
| | - Tom Mikkelsen
- Department of Neurosurgery, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - James J. Yang
- Public Health Sciences, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Swayamprava Panda
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Kelly A. Keenan
- Department of Anesthesiology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Glauber Cabral
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - James R. Ewing
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, United States of America
- Department of Physics, Oakland University, Rochester, Michigan, United States of America
- Department of Neurology, Wayne State University, Detroit, Michigan, United States of America
| |
Collapse
|
45
|
Moserle L, Jiménez-Valerio G, Casanovas O. Antiangiogenic Therapies: Going beyond Their Limits. Cancer Discov 2013; 4:31-41. [DOI: 10.1158/2159-8290.cd-13-0199] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
46
|
Müller C, Schibli R. Prospects in folate receptor-targeted radionuclide therapy. Front Oncol 2013; 3:249. [PMID: 24069581 PMCID: PMC3781362 DOI: 10.3389/fonc.2013.00249] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 09/09/2013] [Indexed: 11/18/2022] Open
Abstract
Targeted radionuclide therapy is based on systemic application of particle-emitting radiopharmaceuticals which are directed toward a specific tumor-associated target. Accumulation of the radiopharmaceutical in targeted cancer cells results in high doses of absorbed radiation energy whereas toxicity to non-targeted healthy tissue is limited. This strategy has found widespread application in the palliative treatment of neuroendocrine tumors using somatostatin-based radiopeptides. The folate receptor (FR) has been identified as a target associated with a variety of frequent tumor types (e.g., ovarian, lung, brain, renal, and colorectal cancer). In healthy organs and tissue FR-expression is restricted to only a few sites such as for instance the kidneys. This demonstrates why FR-targeting is an attractive strategy for the development of new therapy concepts. Due to its high FR-binding affinity (KD < 10−9 M) the vitamin folic acid has emerged as an almost ideal targeting agent. Therefore, a variety of folic acid radioconjugates for nuclear imaging have been developed. However, in spite of the large number of cancer patients who could benefit of a folate-based radionuclide therapy, a therapeutic concept with folate radioconjugates has not yet been envisaged for clinical application. The reason is the generally high accumulation of folate radioconjugates in the kidneys where emission of particle-radiation may result in damage to the renal tissue. Therefore, the design of more sophisticated folate radioconjugates providing improved tissue distribution profiles are needed. This review article summarizes recent developments with regard to a therapeutic application of folate radioconjugates. A new construct of a folate radioconjugate and an application protocol which makes use of a pharmacological interaction allowed the first preclinical therapy experiments with radiofolates. These results raise hope for future application of such new concepts also in the clinic.
Collapse
Affiliation(s)
- Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute , Villigen-PSI , Switzerland
| | | |
Collapse
|
47
|
Reber J, Haller S, Leamon CP, Müller C. 177Lu-EC0800 combined with the antifolate pemetrexed: preclinical pilot study of folate receptor targeted radionuclide tumor therapy. Mol Cancer Ther 2013; 12:2436-45. [PMID: 24030631 DOI: 10.1158/1535-7163.mct-13-0422-t] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Targeted radionuclide therapy has shown impressive results for the palliative treatment of several types of cancer diseases. The folate receptor has been identified as specifically associated with a variety of frequent tumor types. Therefore, it is an attractive target for the development of new radionuclide therapies using folate-based radioconjugates. Previously, we found that pemetrexed (PMX) has a favorable effect in reducing undesired renal uptake of radiofolates. Moreover, PMX also acts as a chemotherapeutic and radiosensitizing agent on tumors. Thus, the aim of our study was to investigate the combined application of PMX and the therapeutic radiofolate (177)Lu-EC0800. Determination of the combination index (CI) revealed a synergistic inhibitory effect of (177)Lu-EC0800 and PMX on the viability of folate receptor-positive cervical (KB) and ovarian (IGROV-1) cancer cells in vitro (CI < 0.8). In an in vivo study, tumor-bearing mice were treated with (177)Lu-EC0800 (20 MBq) and a subtherapeutic (0.4 mg) or therapeutic amount (1.6 mg) of PMX. Application of (177)Lu-EC0800 with PMXther resulted in a two- to four-fold enhanced tumor growth delay and a prolonged survival of KB and IGROV-1 tumor-bearing mice, as compared to the combination with PMXsubther or untreated control mice. PMXsubther protected the kidneys from undesired side effects of (177)Lu-EC0800 (20 MBq) by reducing the absorbed radiation dose. Intact kidney function was shown by determination of plasma parameters and quantitative single-photon emission computed tomography using (99m)Tc-DMSA. Our results confirmed the anticipated dual role of PMX. Its unique features resulted in an improved antitumor effect of folate-based radionuclide therapy and prevented undesired radio-nephrotoxicity.
Collapse
Affiliation(s)
- Josefine Reber
- Corresponding Author: Cristina Müller, Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland.
| | | | | | | |
Collapse
|
48
|
Sunitinib combined with pemetrexed and carboplatin in patients with advanced solid malignancies--results of a phase I dose-escalation study. Invest New Drugs 2013; 31:1487-98. [PMID: 23963796 PMCID: PMC3825543 DOI: 10.1007/s10637-013-0010-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 08/02/2013] [Indexed: 01/14/2023]
Abstract
Objectives The maximum tolerated dose (MTD) and overall safety of sunitinib plus pemetrexed and carboplatin was determined in patients with advanced solid malignancies. Methods In this phase I dose-escalation study, patients received oral sunitinib on a continuous daily dosing (CDD) schedule (37.5 mg/day) or Schedule 2/1 (2 weeks on treatment, 1 week off treatment; 37.5 or 50 mg/day). Pemetrexed (400–500 mg/m2 IV) and carboplatin (AUC = 5 mg·min/ml IV) were administered q3w. At the MTD for the chosen schedule, a cohort of patients with non-small cell lung cancer (NSCLC) or mesothelioma was further evaluated. Results Twenty-one patients were enrolled on Schedule 2/1 (expansion cohort included) and 3 patients on the CDD schedule. The MTD on Schedule 2/1 was sunitinib 37.5 mg/day with pemetrexed 500 mg/m2 and carboplatin AUC = 5 mg·min/ml; MTD on the CDD schedule was not established. Dose-limiting toxicities included grade 3/4 neutropenia, grade 3 thrombocytopenia, and grade 3 hand–foot syndrome. The most common grade 3/4 drug-related non-hematologic adverse events at Schedule 2/1 MTD were fatigue/asthenia and diarrhea (both n = 4). Grade 3/4 hematologic abnormalities included neutropenia (83 %) and leukopenia (83 %). Pharmacokinetic data revealed no clinically significant drug–drug interactions. Best response at the Schedule 2/1 MTD was stable disease ≥8 weeks in 3/5 evaluable patients (60 %). Conclusions With this combination, in patients with advanced solid malignancies, sunitinib MTD on Schedule 2/1 was 37.5 mg/day. Sunitinib plus pemetrexed and carboplatin were tolerable at the MTD, although sunitinib dose delays and reductions were often required due to myelosuppression.
Collapse
|
49
|
Soltani M, Chen P. Numerical Modeling of Interstitial Fluid Flow Coupled with Blood Flow through a Remodeled Solid Tumor Microvascular Network. PLoS One 2013; 8:e67025. [PMID: 23840579 PMCID: PMC3694139 DOI: 10.1371/journal.pone.0067025] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 05/14/2013] [Indexed: 11/26/2022] Open
Abstract
Modeling of interstitial fluid flow involves processes such as fluid diffusion, convective transport in extracellular matrix, and extravasation from blood vessels. To date, majority of microvascular flow modeling has been done at different levels and scales mostly on simple tumor shapes with their capillaries. However, with our proposed numerical model, more complex and realistic tumor shapes and capillary networks can be studied. Both blood flow through a capillary network, which is induced by a solid tumor, and fluid flow in tumor’s surrounding tissue are formulated. First, governing equations of angiogenesis are implemented to specify the different domains for the network and interstitium. Then, governing equations for flow modeling are introduced for different domains. The conservation laws for mass and momentum (including continuity equation, Darcy’s law for tissue, and simplified Navier–Stokes equation for blood flow through capillaries) are used for simulating interstitial and intravascular flows and Starling’s law is used for closing this system of equations and coupling the intravascular and extravascular flows. This is the first study of flow modeling in solid tumors to naturalistically couple intravascular and extravascular flow through a network. This network is generated by sprouting angiogenesis and consisting of one parent vessel connected to the network while taking into account the non-continuous behavior of blood, adaptability of capillary diameter to hemodynamics and metabolic stimuli, non-Newtonian blood flow, and phase separation of blood flow in capillary bifurcation. The incorporation of the outlined components beyond the previous models provides a more realistic prediction of interstitial fluid flow pattern in solid tumors and surrounding tissues. Results predict higher interstitial pressure, almost two times, for realistic model compared to the simplified model.
Collapse
Affiliation(s)
- M. Soltani
- Waterloo Institute for Nanotechnology, Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario, Canada
- * E-mail:
| | - P. Chen
- Waterloo Institute for Nanotechnology, Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
50
|
Zwicker F, Kirsner A, Peschke P, Roeder F, Debus J, Huber PE, Weber KJ. Dichloroacetate induces tumor-specific radiosensitivity in vitro but attenuates radiation-induced tumor growth delay in vivo. Strahlenther Onkol 2013; 189:684-92. [PMID: 23793865 DOI: 10.1007/s00066-013-0354-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 03/14/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Inhibition of pyruvate dehydrogenase kinase (PDK) by dichloroacetate (DCA) can shift tumor cell metabolism from anaerobic glycolysis to glucose oxidation, with activation of mitochondrial activity and chemotherapy-dependent apoptosis. In radiotherapy, DCA could thus potentially enhance the frequently moderate apoptotic response of cancer cells that results from their mitochondrial dysfunction. The aim of this study was to investigate tumor-specific radiosensitization by DCA in vitro and in a human tumor xenograft mouse model in vivo. MATERIALS AND METHODS The interaction of DCA with photon beam radiation was investigated in the human tumor cell lines WIDR (colorectal) and LN18 (glioma), as well as in the human normal tissue cell lines HUVEC (endothelial), MRC5 (lung fibroblasts) and TK6 (lymphoblastoid). Apoptosis induction in vitro was assessed by DAPI staining and sub-G1 flow cytometry; cell survival was quantified by clonogenic assay. The effect of DCA in vivo was investigated in WIDR xenograft tumors growing subcutaneously on BALB/c-nu/nu mice, with and without fractionated irradiation. Histological examination included TUNEL and Ki67 staining for apoptosis and proliferation, respectively, as well as pinomidazole labeling for hypoxia. RESULTS DCA treatment led to decreased clonogenic survival and increased specific apoptosis rates in tumor cell lines (LN18, WIDR) but not in normal tissue cells (HUVEC, MRC5, TK6). However, this significant tumor-specific radiosensitization by DCA in vitro was not reflected by the situation in vivo: The growth suppression of WIDR xenograft tumors after irradiation was reduced upon additional DCA treatment (reflected by Ki67 expression levels), although early tumor cell apoptosis rates were significantly increased by DCA. This apparently paradoxical effect was accompanied by a marked DCA-dependent induction of hypoxia in tumor-tissue. CONCLUSION DCA induced tumor-specific radiosensitization in vitro but not in vivo. DCA also induced development of hypoxia in tumor tissue in vivo. Further investigations relating to the interplay between tumor cell metabolism and tumor microenvironment are necessary to explain the limited success of metabolic targeting in radiotherapy.
Collapse
Affiliation(s)
- F Zwicker
- Department of Radiation Oncology, University Hospital Center Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.
| | | | | | | | | | | | | |
Collapse
|