1
|
Martino A, Terracciano R, Milićević B, Milošević M, Simić V, Fallon BC, Carcamo-Bahena Y, Royal ALR, Carcamo-Bahena AA, Butler EB, Willson RC, Kojić M, Filgueira CS. An Insight into Perfusion Anisotropy within Solid Murine Lung Cancer Tumors. Pharmaceutics 2024; 16:1009. [PMID: 39204354 PMCID: PMC11360231 DOI: 10.3390/pharmaceutics16081009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
Blood vessels are essential for maintaining tumor growth, progression, and metastasis, yet the tumor vasculature is under a constant state of remodeling. Since the tumor vasculature is an attractive therapeutic target, there is a need to predict the dynamic changes in intratumoral fluid pressure and velocity that occur across the tumor microenvironment (TME). The goal of this study was to obtain insight into perfusion anisotropy within lung tumors. To achieve this goal, we used the perfusion marker Hoechst 33342 and vascular endothelial marker CD31 to stain tumor sections from C57BL/6 mice harboring Lewis lung carcinoma tumors on their flank. Vasculature, capillary diameter, and permeability distribution were extracted at different time points along the tumor growth curve. A computational model was generated by applying a unique modeling approach based on the smeared physical fields (Kojic Transport Model, KTM). KTM predicts spatial and temporal changes in intratumoral pressure and fluid velocity within the growing tumor. Anisotropic perfusion occurs within two domains: capillary and extracellular space. Anisotropy in tumor structure causes the nonuniform distribution of pressure and fluid velocity. These results provide insights regarding local vascular distribution for optimal drug dosing and delivery to better predict distribution and duration of retention within the TME.
Collapse
Affiliation(s)
- Antonio Martino
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.M.); (R.T.); (B.C.F.); (Y.C.-B.); (A.L.R.R.); (A.A.C.-B.); (M.K.)
- Department of Materials Science and Engineering, University of Houston, Houston, TX 77024, USA
| | - Rossana Terracciano
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.M.); (R.T.); (B.C.F.); (Y.C.-B.); (A.L.R.R.); (A.A.C.-B.); (M.K.)
- Department of Electronics and Telecommunications, Politecnico di Torino, 10129 Torino, Italy
| | - Bogdan Milićević
- Bioengineering Research and Development Center (BioIRC), 34000 Kragujevac, Serbia; (B.M.); (M.M.); (V.S.)
- Faculty of Engineering, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Miljan Milošević
- Bioengineering Research and Development Center (BioIRC), 34000 Kragujevac, Serbia; (B.M.); (M.M.); (V.S.)
- Institute for Information Technologies, University of Kragujevac, 34000 Kragujevac, Serbia
- Faculty of Information Technology, Belgrade Metropolitan University, 11000 Belgrade, Serbia
| | - Vladimir Simić
- Bioengineering Research and Development Center (BioIRC), 34000 Kragujevac, Serbia; (B.M.); (M.M.); (V.S.)
- Institute for Information Technologies, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Blake C. Fallon
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.M.); (R.T.); (B.C.F.); (Y.C.-B.); (A.L.R.R.); (A.A.C.-B.); (M.K.)
| | - Yareli Carcamo-Bahena
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.M.); (R.T.); (B.C.F.); (Y.C.-B.); (A.L.R.R.); (A.A.C.-B.); (M.K.)
| | - Amber Lee R. Royal
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.M.); (R.T.); (B.C.F.); (Y.C.-B.); (A.L.R.R.); (A.A.C.-B.); (M.K.)
| | - Aileen A. Carcamo-Bahena
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.M.); (R.T.); (B.C.F.); (Y.C.-B.); (A.L.R.R.); (A.A.C.-B.); (M.K.)
| | - Edward Brian Butler
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA;
| | - Richard C. Willson
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77024, USA;
| | - Miloš Kojić
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.M.); (R.T.); (B.C.F.); (Y.C.-B.); (A.L.R.R.); (A.A.C.-B.); (M.K.)
- Bioengineering Research and Development Center (BioIRC), 34000 Kragujevac, Serbia; (B.M.); (M.M.); (V.S.)
- Serbian Academy of Sciences and Arts, 11000 Belgrade, Serbia
| | - Carly S. Filgueira
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.M.); (R.T.); (B.C.F.); (Y.C.-B.); (A.L.R.R.); (A.A.C.-B.); (M.K.)
- Department of Cardiovascular Surgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| |
Collapse
|
2
|
Wirthl B, Janko C, Lyer S, Schrefler BA, Alexiou C, Wall WA. An in silico model of the capturing of magnetic nanoparticles in tumour spheroids in the presence of flow. Biomed Microdevices 2023; 26:1. [PMID: 38008813 PMCID: PMC10678808 DOI: 10.1007/s10544-023-00685-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2023] [Indexed: 11/28/2023]
Abstract
One of the main challenges in improving the efficacy of conventional chemotherapeutic drugs is that they do not reach the cancer cells at sufficiently high doses while at the same time affecting healthy tissue and causing significant side effects and suffering in cancer patients. To overcome this deficiency, magnetic nanoparticles as transporter systems have emerged as a promising approach to achieve more specific tumour targeting. Drug-loaded magnetic nanoparticles can be directed to the target tissue by applying an external magnetic field. However, the magnetic forces exerted on the nanoparticles fall off rapidly with distance, making the tumour targeting challenging, even more so in the presence of flowing blood or interstitial fluid. We therefore present a computational model of the capturing of magnetic nanoparticles in a test setup: our model includes the flow around the tumour, the magnetic forces that guide the nanoparticles, and the transport within the tumour. We show how a model for the transport of magnetic nanoparticles in an external magnetic field can be integrated with a multiphase tumour model based on the theory of porous media. Our approach based on the underlying physical mechanisms can provide crucial insights into mechanisms that cannot be studied conclusively in experimental research alone. Such a computational model enables an efficient and systematic exploration of the nanoparticle design space, first in a controlled test setup and then in more complex in vivo scenarios. As an effective tool for minimising costly trial-and-error design methods, it expedites translation into clinical practice to improve therapeutic outcomes and limit adverse effects for cancer patients.
Collapse
Affiliation(s)
- Barbara Wirthl
- Institute for Computational Mechanics, Technical University of Munich, TUM School of Engineering and Design, Department of Engineering Physics & Computation, Garching bei München, Germany.
| | - Christina Janko
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Stefan Lyer
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Professorship for AI-Guided Nanomaterials within the framework of the Hightech Agenda (HTA) of the Free State of Bavaria, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Bernhard A Schrefler
- Department of Civil, Environmental and Architectural Engineering, University of Padua, Padua, Italy
- Institute for Advanced Study, Technical University of Munich, Garching bei München, Germany
| | - Christoph Alexiou
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Wolfgang A Wall
- Institute for Computational Mechanics, Technical University of Munich, TUM School of Engineering and Design, Department of Engineering Physics & Computation, Garching bei München, Germany
| |
Collapse
|
3
|
Richfield O, Piotrowski-Daspit AS, Shin K, Saltzman WM. Rational nanoparticle design: Optimization using insights from experiments and mathematical models. J Control Release 2023; 360:772-783. [PMID: 37442201 PMCID: PMC10529591 DOI: 10.1016/j.jconrel.2023.07.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/22/2023] [Accepted: 07/08/2023] [Indexed: 07/15/2023]
Abstract
Polymeric nanoparticles are highly tunable drug delivery systems that show promise in targeting therapeutics to specific sites within the body. Rational nanoparticle design can make use of mathematical models to organize and extend experimental data, allowing for optimization of nanoparticles for particular drug delivery applications. While rational nanoparticle design is attractive from the standpoint of improving therapy and reducing unnecessary experiments, it has yet to be fully realized. The difficulty lies in the complexity of nanoparticle structure and behavior, which is added to the complexity of the physiological mechanisms involved in nanoparticle distribution throughout the body. In this review, we discuss the most important aspects of rational design of polymeric nanoparticles. Ultimately, we conclude that many experimental datasets are required to fully model polymeric nanoparticle behavior at multiple scales. Further, we suggest ways to consider the limitations and uncertainty of experimental data in creating nanoparticle design optimization schema, which we call quantitative nanoparticle design frameworks.
Collapse
Affiliation(s)
- Owen Richfield
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | | | - Kwangsoo Shin
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA; Department of Cellular & Molecular Physiology, Yale University, New Haven, CT 06511, USA; Department of Chemical & Environmental Engineering, Yale University, New Haven, CT 06511, USA; Department of Dermatology, Yale University, New Haven, CT 06511, USA.
| |
Collapse
|
4
|
Multiscale Modelling of Nanoparticle Distribution in a Realistic Tumour Geometry Following Local Injection. Cancers (Basel) 2022; 14:cancers14235729. [PMID: 36497210 PMCID: PMC9736186 DOI: 10.3390/cancers14235729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022] Open
Abstract
Radiosensitizers have proven to be an effective method of improving radiotherapy outcomes, with the distribution of particles being a crucial element to delivering optimal treatment outcomes due to the short range of effect of these particles. Here we present a computational model for the transport of nanoparticles within the tumour, whereby the fluid velocity and particle deposition are obtained and used as input into the convection-diffusion equation to calculate the spatio-temporal concentration of the nanoparticles. The effect of particle surface charge and injection locations on the distribution of nanoparticle concentration within the interstitial fluid and deposited onto cell surfaces is assessed. The computational results demonstrate that negatively charged particles can achieve a more uniform distribution throughout the tumour as compared to uncharged or positively charged particles, with particle volume within the fluid being 100% of tumour volume and deposited particle volume 44.5%. In addition, varying the injection location from the end to the middle of the tumour caused a reduction in particle volume of almost 20% for negatively charged particles. In conclusion, radiosensitizing particles should be negatively charged to maximise their spread and penetration within the tumour. Choosing an appropriate injection location can further improve the distribution of these particles.
Collapse
|
5
|
Khan I, Baig MH, Mahfooz S, Imran MA, Khan MI, Dong JJ, Cho JY, Hatiboglu MA. Nanomedicine for Glioblastoma: Progress and Future Prospects. Semin Cancer Biol 2022; 86:172-186. [PMID: 35760272 DOI: 10.1016/j.semcancer.2022.06.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 06/09/2022] [Accepted: 06/21/2022] [Indexed: 11/29/2022]
Abstract
Glioblastoma is the most aggressive form of brain tumor, accounting for the highest mortality and morbidity rates. Current treatment for patients with glioblastoma includes maximal safe tumor resection followed by radiation therapy with concomitant temozolomide (TMZ) chemotherapy. The addition of TMZ to the conformal radiation therapy has improved the median survival time only from 12 months to 16 months in patients with glioblastoma. Despite these aggressive treatment strategies, patients' prognosis remains poor. This therapeutic failure is primarily attributed to the blood-brain barrier (BBB) that restricts the transport of TMZ from reaching the tumor site. In recent years, nanomedicine has gained considerable attention among researchers and shown promising developments in clinical applications, including the diagnosis, prognosis, and treatment of glioblastoma tumors. This review sheds light on the morphological and physiological complexity of the BBB. It also explains the development of nanomedicine strategies to enhance the permeability of drug molecules across the BBB.
Collapse
Affiliation(s)
- Imran Khan
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, Istanbul, Turkey
| | - Mohammad Hassan Baig
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Gangnam-gu, Seoul, 120-752, Republic of Korea
| | - Sadaf Mahfooz
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, Istanbul, Turkey
| | - Mohammad Azhar Imran
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Gangnam-gu, Seoul, 120-752, Republic of Korea
| | - Mohd Imran Khan
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Gangnam-gu, Seoul, 120-752, Republic of Korea
| | - Jae-June Dong
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Gangnam-gu, Seoul, 120-752, Republic of Korea
| | - Jae Yong Cho
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Gangnam-gu, Seoul, 120-752, Republic of Korea.
| | - Mustafa Aziz Hatiboglu
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, Istanbul, Turkey; Department of Neurosurgery, Bezmialem Vakif University Medical School, Vatan Street, Fatih, Istanbul, Turkey.
| |
Collapse
|
6
|
Kipouros T, Chamseddine I, Kokkolaras M. Using Parallel Coordinates in Optimization of Nano-Particle Drug Delivery. J Biomech Eng 2022; 144:1120776. [PMID: 34590693 DOI: 10.1115/1.4052578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Indexed: 11/08/2022]
Abstract
Nanoparticle drug delivery better targets neoplastic lesions than free drugs and thus has emerged as a safer form of cancer therapy. Nanoparticle design variables are important determinants of efficacy as they influence the drug biodistribution and pharmacokinetics. Previously, we determined optimal designs through mechanistic modeling and optimization. However, the numerical nature of the tumor model and numerous candidate nanoparticle designs hinder hypothesis generation and treatment personalization. In this paper, we utilize the parallel coordinates technique to visualize high-dimensional optimal solutions and extract correlations between nanoparticle design and treatment outcomes. We found that at optimality, two major design variables are dependent, and thus the optimization problem can be reduced. In addition, we obtained an analytical relationship between optimal nanoparticle sizes and optimal distribution, which could facilitate the utilization of tumors models in preclinical studies. Our approach has simplified the results of the previously integrated modeling and optimization framework developed for nanotherapy and enhanced the interpretation and utilization of findings. Integrated mathematical frameworks are increasing in the medical field, and our method can be applied outside nanotherapy to facilitate the clinical translation of computational methods.
Collapse
Affiliation(s)
- Timoleon Kipouros
- Department of Engineering, University of Cambridge, Cambridge CB2 1PZ, UK
| | - Ibrahim Chamseddine
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Michael Kokkolaras
- Department of Mechanical Engineering, McGill University, Montreal, QC H3A 0C3, Canada
| |
Collapse
|
7
|
MacCuaig WM, Fouts BL, McNally MW, Grizzle WE, Chuong P, Samykutty A, Mukherjee P, Li M, Jasinski J, Behkam B, McNally LR. Active Targeting Significantly Outperforms Nanoparticle Size in Facilitating Tumor-Specific Uptake in Orthotopic Pancreatic Cancer. ACS APPLIED MATERIALS & INTERFACES 2021; 13:49614-49630. [PMID: 34653338 PMCID: PMC9783196 DOI: 10.1021/acsami.1c09379] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Nanoparticles are widely studied as theranostic vehicles for cancer; however, clinical translation has been limited due to poor tumor specificity. Features that maximize tumor uptake remain controversial, particularly when using clinically relevant models. We report a systematic study that assesses two major features for the impact on tumor specificity, i.e., active vs passive targeting and nanoparticle size, to evaluate relative influences in vivo. Active targeting via the V7 peptide is superior to passive targeting for uptake by pancreatic tumors, irrespective of nanoparticle size, observed through in vivo imaging. Size has a secondary effect on uptake for actively targeted nanoparticles in which 26 nm nanoparticles outperform larger 45 and 73 nm nanoparticles. Nanoparticle size had no significant effect on uptake for passively targeted nanoparticles. Results highlight the superiority of active targeting over nanoparticle size for tumor uptake. These findings suggest a framework for optimizing similar nonaggregate nanoparticles for theranostic treatment of recalcitrant cancers.
Collapse
Affiliation(s)
- William M. MacCuaig
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, 73104, USA
- Department of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Benjamin L. Fouts
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, 73104, USA
| | - Molly W McNally
- Department of Surgery, University of Oklahoma, Oklahoma City, OK, 73104, USA
- Department of Cancer Biology, Wake Forest University, Winston-Salem, NC 27157, USA
| | - William E. Grizzle
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Phillip Chuong
- Department of Surgery, University of Louisville, Louisville, KY 40202, USA
| | - Abhilash Samykutty
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, 73104, USA
- Department of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
- Department of Cancer Biology, Wake Forest University, Winston-Salem, NC 27157, USA
| | | | - Min Li
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, 73104, USA
| | - Jacek Jasinski
- Conn Center Materials Characterization, University of Louisville, Louisville, KY 40202, USA
| | - Bahareh Behkam
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Lacey R. McNally
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, 73104, USA
- Department of Surgery, University of Oklahoma, Oklahoma City, OK, 73104, USA
- Department of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
- Department of Cancer Biology, Wake Forest University, Winston-Salem, NC 27157, USA
| |
Collapse
|
8
|
Tang Y, Zhang J, He D, Miao W, Liu W, Li Y, Lu G, Wu F, Wang S. GANDA: A deep generative adversarial network conditionally generates intratumoral nanoparticles distribution pixels-to-pixels. J Control Release 2021; 336:336-343. [PMID: 34197860 DOI: 10.1016/j.jconrel.2021.06.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/19/2021] [Accepted: 06/25/2021] [Indexed: 12/22/2022]
Abstract
Intratumoral nanoparticles (NPs) distribution is critical for the success of nanomedicine in imaging and treatment, but computational models to describe the NPs distribution remain unavailable due to the complex tumor-nano interactions. Here, we develop a Generative Adversarial Network for Distribution Analysis (GANDA) to describe and conditionally generates the intratumoral quantum dots (QDs) distribution after i.v. injection. This deep generative model is trained automatically by 27,775 patches of tumor vessels and cell nuclei decomposed from whole-slide images of 4 T1 breast cancer sections. The GANDA model can conditionally generate images of intratumoral QDs distribution under the constraint of given tumor vessels and cell nuclei channels with the same spatial resolution (pixels-to-pixels), minimal loss (mean squared error, MSE = 1.871) and excellent reliability (intraclass correlation, ICC = 0.94). Quantitative analysis of QDs extravasation distance (ICC = 0.95) and subarea distribution (ICC = 0.99) is allowed on the generated images without knowing the real QDs distribution. We believe this deep generative model may provide opportunities to investigate how influencing factors affect NPs distribution in individual tumors and guide nanomedicine optimization for molecular imaging and personalized treatment.
Collapse
Affiliation(s)
- Yuxia Tang
- Department of Radiology, Jinling Hospital, Nanjing, Jiangsu 210000, Nanjing Medical University, China
| | - Jiulou Zhang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Doudou He
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenfang Miao
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei Liu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yang Li
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guangming Lu
- Department of Radiology, Jinling Hospital, Nanjing, Jiangsu 210000, Nanjing Medical University, China.
| | - Feiyun Wu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Shouju Wang
- Department of Radiology, Jinling Hospital, Nanjing, Jiangsu 210000, Nanjing Medical University, China; Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
9
|
Butner JD, Wang Z, Elganainy D, Al Feghali KA, Plodinec M, Calin GA, Dogra P, Nizzero S, Ruiz-Ramírez J, Martin GV, Tawbi HA, Chung C, Koay EJ, Welsh JW, Hong DS, Cristini V. A mathematical model for the quantification of a patient's sensitivity to checkpoint inhibitors and long-term tumour burden. Nat Biomed Eng 2021; 5:297-308. [PMID: 33398132 PMCID: PMC8669771 DOI: 10.1038/s41551-020-00662-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/14/2020] [Indexed: 02/06/2023]
Abstract
A large proportion of patients with cancer are unresponsive to treatment with immune checkpoint blockade and other immunotherapies. Here, we report a mathematical model of the time course of tumour responses to immune checkpoint inhibitors. The model takes into account intrinsic tumour growth rates, the rates of immune activation and of tumour-immune cell interactions, and the efficacy of immune-mediated tumour killing. For 124 patients, four cancer types and two immunotherapy agents, the model reliably described the immune responses and final tumour burden across all different cancers and drug combinations examined. In validation cohorts from four clinical trials of checkpoint inhibitors (with a total of 177 patients), the model accurately stratified the patients according to reduced or increased long-term tumour burden. We also provide model-derived quantitative measures of treatment sensitivity for specific drug-cancer combinations. The model can be used to predict responses to therapy and to quantify specific drug-cancer sensitivities in individual patients.
Collapse
Affiliation(s)
- Joseph D Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Dalia Elganainy
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Karine A Al Feghali
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marija Plodinec
- Biozentrum and the Swiss Nanoscience Institute, University of Basel, Basel, Switzerland
| | - George A Calin
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Sara Nizzero
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Javier Ruiz-Ramírez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Geoffrey V Martin
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hussein A Tawbi
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caroline Chung
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eugene J Koay
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James W Welsh
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
10
|
Manzin A, Ferrero R, Vicentini M. From Micromagnetic to In Silico Modeling of Magnetic Nanodisks for Hyperthermia Applications. ADVANCED THEORY AND SIMULATIONS 2021. [DOI: 10.1002/adts.202100013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Alessandra Manzin
- Istituto Nazionale di Ricerca Metrologica (INRIM) Strada delle Cacce 91 Torino 10135 Italy
| | - Riccardo Ferrero
- Istituto Nazionale di Ricerca Metrologica (INRIM) Strada delle Cacce 91 Torino 10135 Italy
| | - Marta Vicentini
- Istituto Nazionale di Ricerca Metrologica (INRIM) Strada delle Cacce 91 Torino 10135 Italy
- Politecnico di Torino Corso Duca degli Abruzzi 24 Torino 10129 Italy
| |
Collapse
|
11
|
Fuentes D, Thompson E, Jacobsen M, Crouch AC, Layman RR, Riviere B, Cressman E. Imaging-based characterization of convective tissue properties. Int J Hyperthermia 2021; 37:155-163. [PMID: 33426993 PMCID: PMC7983068 DOI: 10.1080/02656736.2020.1845403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Convective transport is an important phenomenon for nanomedicine delivery. We present an imaging-based approach to recover tissue properties that are significant in the accumulation of nanoparticles delivered via systemic methods. The classical pharmacokinetic analysis develops governing equations for the particle transport from a first principle mass balance. Fundamentally, the governing equations for compartmental mass balance represent a spatially invariant mass transport between compartments and do not capture spatially variant convection phenomena. Further, the parameters recovered from this approach do not necessarily have direct meaning with respect to the governing equations for convective transport. In our approach, a framework is presented for directly measuring permeability in the sense of Darcy flow through porous tissue. Measurements from our approach are compared to an extended Tofts model as a control. We demonstrate that a pixel-wise iterative clustering algorithm may be applied to reduce the parameter space of the measurements. We show that measurements obtained from our approach are correlated with measurements obtained from the extended Tofts model control. These correlations demonstrate that the proposed approach contains similar information to an established compartmental model and may be useful in providing an alternative theoretical framework for parameterizing mathematical models for treatment planning and diagnostic studies involving nanomedicine where convection dominated effects are important.
Collapse
Affiliation(s)
- D Fuentes
- Departments of Imaging Physics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.,Department of Computational and Applied Mathematics, Rice University, Houston, TX, USA
| | - E Thompson
- Departments of Imaging Physics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - M Jacobsen
- Departments of Imaging Physics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - A Colleen Crouch
- Interventional Radiology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - R R Layman
- Departments of Imaging Physics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - B Riviere
- Department of Computational and Applied Mathematics, Rice University, Houston, TX, USA
| | - E Cressman
- Interventional Radiology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
12
|
Sahai N, Gogoi M, Ahmad N. Mathematical Modeling and Simulations for Developing Nanoparticle-Based Cancer Drug Delivery Systems: A Review. CURRENT PATHOBIOLOGY REPORTS 2021. [DOI: 10.1007/s40139-020-00219-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
13
|
Chamseddine IM, Kokkolaras M. A Dual Nanoparticle Delivery Strategy for Enhancing Drug Distribution in Cancerous Tissue. J Biomech Eng 2020; 142:124501. [PMID: 32601692 DOI: 10.1115/1.4047657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Indexed: 11/08/2022]
Abstract
Nanoparticle-mediated drug delivery may be a promising alternative to traditional chemotherapy of high systemic toxicity. Tumor tissue architecture poses a challenge to delivery of nanoparticles. Small and spherical nanoparticles have poor adherence to the tumor vasculature, while larger and more eccentric ones create high heterogeneity in tissue-to-drug exposure. In previous work, we quantified these tradeoffs using numerical optimization. In this study, we demonstrate that simultaneous delivery of multiple nanoparticle designs can enhance drug distribution in the cancerous tissue without compromising nanoparticle tumoral accumulation. We formulate and solve optimization problems to find the optimal constituent of the heterogeneous injection in terms of nanoparticle design diversity that increases drug distribution by 14%.
Collapse
Affiliation(s)
- Ibrahim M Chamseddine
- Systems Optimization Lab, Department of Mechanical Engineering, McGill University, Montreal, QC H3A 0G4, Canada
| | - Michael Kokkolaras
- Systems Optimization Lab, Department of Mechanical Engineering, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
14
|
Frieboes HB, Raghavan S, Godin B. Modeling of Nanotherapy Response as a Function of the Tumor Microenvironment: Focus on Liver Metastasis. Front Bioeng Biotechnol 2020; 8:1011. [PMID: 32974325 PMCID: PMC7466654 DOI: 10.3389/fbioe.2020.01011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022] Open
Abstract
The tumor microenvironment (TME) presents a challenging barrier for effective nanotherapy-mediated drug delivery to solid tumors. In particular for tumors less vascularized than the surrounding normal tissue, as in liver metastases, the structure of the organ itself conjures with cancer-specific behavior to impair drug transport and uptake by cancer cells. Cells and elements in the TME of hypovascularized tumors play a key role in the process of delivery and retention of anti-cancer therapeutics by nanocarriers. This brief review describes the drug transport challenges and how they are being addressed with advanced in vitro 3D tissue models as well as with in silico mathematical modeling. This modeling complements network-oriented techniques, which seek to interpret intra-cellular relevant pathways and signal transduction within cells and with their surrounding microenvironment. With a concerted effort integrating experimental observations with computational analyses spanning from the molecular- to the tissue-scale, the goal of effective nanotherapy customized to patient tumor-specific conditions may be finally realized.
Collapse
Affiliation(s)
- Hermann B. Frieboes
- Department of Bioengineering, University of Louisville, Louisville, KY, United States
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
- Center for Predictive Medicine, University of Louisville, Louisville, KY, United States
| | - Shreya Raghavan
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, United States
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States
- Department of Obstetrics and Gynecology, Houston Methodist Hospital, Houston, TX, United States
- Developmental Therapeutics Program, Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
15
|
Hassanzadeh P. Towards the quantum-enabled technologies for development of drugs or delivery systems. J Control Release 2020; 324:260-279. [DOI: 10.1016/j.jconrel.2020.04.050] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 12/20/2022]
|
16
|
Drug delivery: Experiments, mathematical modelling and machine learning. Comput Biol Med 2020; 123:103820. [PMID: 32658778 DOI: 10.1016/j.compbiomed.2020.103820] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/22/2020] [Accepted: 05/10/2020] [Indexed: 01/28/2023]
Abstract
We address the problem of determining from laboratory experiments the data necessary for a proper modeling of drug delivery and efficacy in anticancer therapy. There is an inherent difficulty in extracting the necessary parameters, because the experiments often yield an insufficient quantity of information. To overcome this difficulty, we propose to combine real experiments, numerical simulation, and Machine Learning (ML) based on Artificial Neural Networks (ANN), aiming at a reliable identification of the physical model factors, e.g. the killing action of the drug. To this purpose, we exploit the employed mathematical-numerical model for tumor growth and drug delivery, together with the ANN - ML procedure, to integrate the results of the experimental tests and feed back the model itself, thus obtaining a reliable predictive tool. The procedure represents a hybrid data-driven, physics-informed approach to machine learning. The physical and mathematical model employed for the numerical simulations is without extracellular matrix (ECM) and healthy cells because of the experimental conditions we reproduce.
Collapse
|
17
|
Chamseddine IM, Frieboes HB, Kokkolaras M. Multi-objective optimization of tumor response to drug release from vasculature-bound nanoparticles. Sci Rep 2020; 10:8294. [PMID: 32427977 PMCID: PMC7237449 DOI: 10.1038/s41598-020-65162-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/26/2020] [Indexed: 12/31/2022] Open
Abstract
The pharmacokinetics of nanoparticle-borne drugs targeting tumors depends critically on nanoparticle design. Empirical approaches to evaluate such designs in order to maximize treatment efficacy are time- and cost-intensive. We have recently proposed the use of computational modeling of nanoparticle-mediated drug delivery targeting tumor vasculature coupled with numerical optimization to pursue optimal nanoparticle targeting and tumor uptake. Here, we build upon these studies to evaluate the effect of tumor size on optimal nanoparticle design by considering a cohort of heterogeneously-sized tumor lesions, as would be clinically expected. The results indicate that smaller nanoparticles yield higher tumor targeting and lesion regression for larger-sized tumors. We then augment the nanoparticle design optimization problem by considering drug diffusivity, which yields a two-fold tumor size decrease compared to optimizing nanoparticles without this consideration. We quantify the tradeoff between tumor targeting and size decrease using bi-objective optimization, and generate five Pareto-optimal nanoparticle designs. The results provide a spectrum of treatment outcomes - considering tumor targeting vs. antitumor effect - with the goal to enable therapy customization based on clinical need. This approach could be extended to other nanoparticle-based cancer therapies, and support the development of personalized nanomedicine in the longer term.
Collapse
Affiliation(s)
- Ibrahim M Chamseddine
- Deparment of Integrated Mathematical Oncology, Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Hermann B Frieboes
- Department of Bioengineering, University of Louisville, Louisville, KY, USA.
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.
- Center for Predictive Medicine, University of Louisville, Louisville, KY, USA.
| | - Michael Kokkolaras
- Department of Mechanical Engineering, McGill University, Montreal, Quebec, Canada.
- GERAD - Group for Research in Decision Analysis, Montreal, Quebec, Canada.
| |
Collapse
|
18
|
Butner JD, Elganainy D, Wang CX, Wang Z, Chen SH, Esnaola NF, Pasqualini R, Arap W, Hong DS, Welsh J, Koay EJ, Cristini V. Mathematical prediction of clinical outcomes in advanced cancer patients treated with checkpoint inhibitor immunotherapy. SCIENCE ADVANCES 2020; 6:eaay6298. [PMID: 32426472 PMCID: PMC7190324 DOI: 10.1126/sciadv.aay6298] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 01/21/2020] [Indexed: 05/20/2023]
Abstract
We present a mechanistic mathematical model of immune checkpoint inhibitor therapy to address the oncological need for early, broadly applicable readouts (biomarkers) of patient response to immunotherapy. The model is built upon the complex biological and physical interactions between the immune system and cancer, and is informed using only standard-of-care CT. We have retrospectively applied the model to 245 patients from multiple clinical trials treated with anti-CTLA-4 or anti-PD-1/PD-L1 antibodies. We found that model parameters distinctly identified patients with common (n = 18) and rare (n = 10) malignancy types who benefited and did not benefit from these monotherapies with accuracy as high as 88% at first restaging (median 53 days). Further, the parameters successfully differentiated pseudo-progression from true progression, providing previously unidentified insights into the unique biophysical characteristics of pseudo-progression. Our mathematical model offers a clinically relevant tool for personalized oncology and for engineering immunotherapy regimens.
Collapse
Affiliation(s)
- Joseph D. Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Dalia Elganainy
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Charles X. Wang
- MD/PhD Program, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shu-Hsia Chen
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, USA
- Cancer Center, Houston Methodist Research Institute, Houston, TX, USA
| | - Nestor F. Esnaola
- Cancer Center, Houston Methodist Research Institute, Houston, TX, USA
- Department of Surgical Oncology, Fox Chase Cancer Center—Temple Health, Philadelphia, PA, USA
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey at University Hospital, Newark, NJ, USA
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey at University Hospital, Newark, NJ, USA
- Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - David S. Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eugene J. Koay
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Corresponding author. (V.C.); (E.J.K.)
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
- MD/PhD Program, Drexel University College of Medicine, Philadelphia, PA 19102, USA
- Corresponding author. (V.C.); (E.J.K.)
| |
Collapse
|
19
|
Dogra P, Butner JD, Ruiz Ramírez J, Chuang YL, Noureddine A, Jeffrey Brinker C, Cristini V, Wang Z. A mathematical model to predict nanomedicine pharmacokinetics and tumor delivery. Comput Struct Biotechnol J 2020; 18:518-531. [PMID: 32206211 PMCID: PMC7078505 DOI: 10.1016/j.csbj.2020.02.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/14/2020] [Accepted: 02/22/2020] [Indexed: 02/07/2023] Open
Abstract
Towards clinical translation of cancer nanomedicine, it is important to systematically investigate the various parameters related to nanoparticle (NP) physicochemical properties, tumor characteristics, and inter-individual variability that affect the tumor delivery efficiency of therapeutic nanomaterials. Comprehensive investigation of these parameters using traditional experimental approaches is impractical due to the vast parameter space; mathematical models provide a more tractable approach to navigate through such a multidimensional space. To this end, we have developed a predictive mathematical model of whole-body NP pharmacokinetics and their tumor delivery in vivo, and have conducted local and global sensitivity analyses to identify the factors that result in low tumor delivery efficiency and high off-target accumulation of NPs. Our analyses reveal that NP degradation rate, tumor blood viscosity, NP size, tumor vascular fraction, and tumor vascular porosity are the key parameters in governing NP kinetics in the tumor interstitium. The impact of these parameters on tumor delivery efficiency of NPs is discussed, and optimal values for maximizing NP delivery are presented.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Joseph D. Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Javier Ruiz Ramírez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Yao-li Chuang
- Department of Mathematics, California State University, Northridge, CA 91330, USA
| | - Achraf Noureddine
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM 87106, USA
| | - C. Jeffrey Brinker
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM 87106, USA
- UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87102, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
- Corresponding author at: Mathematics in Medicine Program, The Houston Methodist Research Institute, HMRI R8-122, 6670 Bertner Ave, Houston, TX 77030, USA.
| |
Collapse
|
20
|
Ng TS, Garlin MA, Weissleder R, Miller MA. Improving nanotherapy delivery and action through image-guided systems pharmacology. Theranostics 2020; 10:968-997. [PMID: 31938046 PMCID: PMC6956809 DOI: 10.7150/thno.37215] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 08/04/2019] [Indexed: 12/12/2022] Open
Abstract
Despite recent advances in the translation of therapeutic nanoparticles (TNPs) into the clinic, the field continues to face challenges in predictably and selectively delivering nanomaterials for the treatment of solid cancers. The concept of enhanced permeability and retention (EPR) has been coined as a convenient but simplistic descriptor of high TNP accumulation in some tumors. However, in practice EPR represents a number of physiological variables rather than a single one (including dysfunctional vasculature, compromised lymphatics and recruited host cells, among other aspects of the tumor microenvironment) — each of which can be highly heterogenous within a given tumor, patient and across patients. Therefore, a clear need exists to dissect the specific biophysical factors underlying the EPR effect, to formulate better TNP designs, and to identify patients with high-EPR tumors who are likely to respond to TNP. The overall pharmacology of TNP is governed by an interconnected set of spatially defined and dynamic processes that benefit from a systems-level quantitative approach, and insights into the physiology have profited from the marriage between in vivo imaging and quantitative systems pharmacology (QSP) methodologies. In this article, we review recent developments pertinent to image-guided systems pharmacology of nanomedicines in oncology. We first discuss recent developments of quantitative imaging technologies that enable analysis of nanomaterial pharmacology at multiple spatiotemporal scales, and then examine reports that have adopted these imaging technologies to guide QSP approaches. In particular, we focus on studies that have integrated multi-scale imaging with computational modeling to derive insights about the EPR effect, as well as studies that have used modeling to guide the manipulation of the EPR effect and other aspects of the tumor microenvironment for improving TNP action. We anticipate that the synergistic combination of imaging with systems-level computational methods for effective clinical translation of TNPs will only grow in relevance as technologies increase in resolution, multiplexing capability, and in the ability to examine heterogeneous behaviors at the single-cell level.
Collapse
|
21
|
Tamura R, Miyoshi H, Yoshida K, Okano H, Toda M. Recent progress in the research of suicide gene therapy for malignant glioma. Neurosurg Rev 2019; 44:29-49. [PMID: 31781985 DOI: 10.1007/s10143-019-01203-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/14/2019] [Accepted: 10/28/2019] [Indexed: 12/15/2022]
Abstract
Malignant glioma, which is characterized by diffuse infiltration into the normal brain parenchyma, is the most aggressive primary brain tumor with dismal prognosis. Over the past 40 years, the median survival has only slightly improved. Therefore, new therapeutic modalities must be developed. In the 1990s, suicide gene therapy began attracting attention for the treatment of malignant glioma. Some clinical trials used a viral vector for suicide gene transduction; however, it was found that viral vectors cannot cover the large invaded area of glioma cells. Interest in this therapy was recently revived because some types of stem cells possess a tumor-tropic migratory capacity, which can be used as cellular delivery vehicles. Immortalized, clonal neural stem cell (NSC) line has been used for patients with recurrent high-grade glioma, which showed safety and efficacy. Embryonic and induced pluripotent stem cells may be considered as sources of NSC because NSC is difficult to harvest, and ethical issues have been raised. Mesenchymal stem cells are alternative candidates for cellular vehicle and are easily harvested from the bone marrow. In addition, a new type of nonlytic, amphotropic retroviral replicating vector encoding suicide gene has shown efficacy in patients with recurrent high-grade glioma in a clinical trial. This replicating viral capacity is another possible candidate as delivery vehicle to tackle gliomas. Herein, we review the concept of suicide gene therapy, as well as recent progress in preclinical and clinical studies in this field.
Collapse
Affiliation(s)
- Ryota Tamura
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroyuki Miyoshi
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazunari Yoshida
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Masahiro Toda
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
22
|
Pharmacokinetic/Pharmacodynamics Modeling of Drug-Loaded PLGA Nanoparticles Targeting Heterogeneously Vascularized Tumor Tissue. Pharm Res 2019; 36:185. [PMID: 31773287 DOI: 10.1007/s11095-019-2721-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/14/2019] [Indexed: 12/24/2022]
Abstract
PURPOSE Nanoparticle-mediated drug delivery and efficacy for cancer applications depends on systemic as well as local microenvironment characteristics. Here, a novel coupling of a nanoparticle (NP) kinetic model with a drug pharmacokinetic/pharmacodynamics model evaluates efficacy of cisplatin-loaded poly lactic-co-glycolic acid (PLGA) NPs in heterogeneously vascularized tumor tissue. METHODS Tumor lesions are modeled with various levels of vascular heterogeneity, as would be encountered with different types of tumors. The magnitude of the extracellular to cytosolic NP transport is varied to assess tumor-dependent cellular uptake. NP aggregation is simulated to evaluate its effects on drug distribution and tumor response. RESULTS Cisplatin-loaded PLGA NPs are most effective in decreasing tumor size in the case of high vascular-induced heterogeneity, a high NP cytosolic transfer coefficient, and no NP aggregation. Depending on the level of tissue heterogeneity, NP cytosolic transfer and drug half-life, NP aggregation yielding only extracellular drug release could be more effective than unaggregated NPs uptaken by cells and releasing drug both extra- and intra-cellularly. CONCLUSIONS Model-based customization of PLGA NP and drug design parameters, including cellular uptake and aggregation, tailored to patient tumor tissue characteristics such as proportion of viable tissue and vascular heterogeneity, could help optimize the NP-mediated tumor drug response.
Collapse
|
23
|
Loya J, Zhang C, Cox E, Achrol AS, Kesari S. Biological intratumoral therapy for the high-grade glioma part II: vector- and cell-based therapies and radioimmunotherapy. CNS Oncol 2019; 8:CNS40. [PMID: 31747784 PMCID: PMC6880300 DOI: 10.2217/cns-2019-0002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Management of high-grade gliomas (HGGs) remains a complex challenge with an overall poor prognosis despite aggressive multimodal treatment. New translational research has focused on maximizing tumor cell eradication through improved tumor cell targeting while minimizing collateral systemic side effects. In particular, biological intratumoral therapies have been the focus of novel translational research efforts due to their inherent potential to be both dynamically adaptive and target specific. This two part review will provide an overview of biological intratumoral therapies that have been evaluated in human clinical trials in HGGs, and summarize key advances and remaining challenges in the development of these therapies as a potential new paradigm in the management of HGGs. Part II discusses vector-based therapies, cell-based therapies and radioimmunotherapy.
Collapse
Affiliation(s)
- Joshua Loya
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Charlie Zhang
- Buffalo School of Medicine, State University of New York, Buffalo, NY 14202, USA
| | - Emily Cox
- Providence Medical Research Center, Spokane, WA 99204, USA
| | - Achal S Achrol
- John Wayne Cancer Institute, Pacific Neuroscience Institute, Santa Monica, CA 90404, USA
| | - Santosh Kesari
- John Wayne Cancer Institute, Pacific Neuroscience Institute, Santa Monica, CA 90404, USA
| |
Collapse
|
24
|
Chamseddine IM, Kokkolaras M. Nanoparticle Optimization for Enhanced Targeted Anticancer Drug Delivery. J Biomech Eng 2019; 140:2658265. [PMID: 29049542 DOI: 10.1115/1.4038202] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Indexed: 11/08/2022]
Abstract
Nanoparticle (NP)-based drug delivery is a promising method to increase the therapeutic index of anticancer agents with low median toxic dose. The delivery efficiency, corresponding to the fraction of the injected NPs that adhere to the tumor site, depends on NP size a and aspect ratio AR. Values for these variables are currently chosen empirically, which may not result in optimal targeted drug delivery. This study applies rigorous optimization to the design of NPs. A preliminary investigation revealed that delivery efficiency increases monotonically with a and AR. However, maximizing a and AR results in nonuniform drug distribution, which impairs tumor regression. Therefore, a multiobjective optimization (MO) problem is formulated to quantify the trade-off between NPs accumulation and distribution. The MO is solved using the derivative-free mesh adaptive direct search algorithm. Theoretically, the Pareto-optimal set consists of an infinite number of mathematically equivalent solutions to the MO problem. However, interesting design solutions can be identified subjectively, e.g., the ellipsoid with a major axis of 720 nm and an aspect ratio of 7.45, as the solution closest to the utopia point. The MO problem formulation is then extended to optimize NP biochemical properties: ligand-receptor binding affinity and ligand density. Optimizing physical and chemical properties simultaneously results in optimal designs with reduced NP sizes and thus enhanced cellular uptake. The presented study provides an insight into NP structures that have potential for producing desirable drug delivery.
Collapse
Affiliation(s)
- Ibrahim M Chamseddine
- Department of Mechanical Engineering, McGill University, Montreal, QC H3A 0C3, Canada e-mail:
| | - Michael Kokkolaras
- Department of Mechanical Engineering, McGill University, Montreal, QC H3A 0C3, Canada e-mail:
| |
Collapse
|
25
|
Shamsi M, Mohammadi A, Manshadi MK, Sanati-Nezhad A. Mathematical and computational modeling of nano-engineered drug delivery systems. J Control Release 2019; 307:150-165. [DOI: 10.1016/j.jconrel.2019.06.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/10/2019] [Accepted: 06/12/2019] [Indexed: 12/20/2022]
|
26
|
Chamseddine IM, Rejniak KA. Hybrid modeling frameworks of tumor development and treatment. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2019; 12:e1461. [PMID: 31313504 PMCID: PMC6898741 DOI: 10.1002/wsbm.1461] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/13/2019] [Accepted: 06/13/2019] [Indexed: 12/15/2022]
Abstract
Tumors are complex multicellular heterogeneous systems comprised of components that interact with and modify one another. Tumor development depends on multiple factors: intrinsic, such as genetic mutations, altered signaling pathways, or variable receptor expression; and extrinsic, such as differences in nutrient supply, crosstalk with stromal or immune cells, or variable composition of the surrounding extracellular matrix. Tumors are also characterized by high cellular heterogeneity and dynamically changing tumor microenvironments. The complexity increases when this multiscale, multicomponent system is perturbed by anticancer treatments. Modeling such complex systems and predicting how tumors will respond to therapies require mathematical models that can handle various types of information and combine diverse theoretical methods on multiple temporal and spatial scales, that is, hybrid models. In this update, we discuss the progress that has been achieved during the last 10 years in the area of the hybrid modeling of tumors. The classical definition of hybrid models refers to the coupling of discrete descriptions of cells with continuous descriptions of microenvironmental factors. To reflect on the direction that the modeling field has taken, we propose extending the definition of hybrid models to include of coupling two or more different mathematical frameworks. Thus, in addition to discussing recent advances in discrete/continuous modeling, we also discuss how these two mathematical descriptions can be coupled with theoretical frameworks of optimal control, optimization, fluid dynamics, game theory, and machine learning. All these methods will be illustrated with applications to tumor development and various anticancer treatments. This article is characterized under:Analytical and Computational Methods > Computational Methods Translational, Genomic, and Systems Medicine > Therapeutic Methods Models of Systems Properties and Processes > Organ, Tissue, and Physiological Models
Collapse
Affiliation(s)
- Ibrahim M. Chamseddine
- Department of Integrated Mathematical OncologyH. Lee Moffitt Cancer Center and Research InstituteTampaFlorida
| | - Katarzyna A. Rejniak
- Department of Integrated Mathematical OncologyH. Lee Moffitt Cancer Center and Research InstituteTampaFlorida
- Department of Oncologic Sciences, Morsani College of MedicineUniversity of South FloridaTampaFlorida
| |
Collapse
|
27
|
Abstract
Cancer continues to be among the leading healthcare problems worldwide, and efforts continue not just to find better drugs, but also better drug delivery methods. The need for delivering cytotoxic agents selectively to cancerous cells, for improved safety and efficacy, has triggered the application of nanotechnology in medicine. This effort has provided drug delivery systems that can potentially revolutionize cancer treatment. Nanocarriers, due to their capacity for targeted drug delivery, can shift the balance of cytotoxicity from healthy to cancerous cells. The field of cancer nanomedicine has made significant progress, but challenges remain that impede its clinical translation. Several biophysical barriers to the transport of nanocarriers to the tumor exist, and a much deeper understanding of nano-bio interactions is necessary to change the status quo. Mathematical modeling has been instrumental in improving our understanding of the physicochemical and physiological underpinnings of nanomaterial behavior in biological systems. Here, we present a comprehensive review of literature on mathematical modeling works that have been and are being employed towards a better understanding of nano-bio interactions for improved tumor delivery efficacy.
Collapse
|
28
|
Cova TFGG, Bento DJ, Nunes SCC. Computational Approaches in Theranostics: Mining and Predicting Cancer Data. Pharmaceutics 2019; 11:E119. [PMID: 30871264 PMCID: PMC6471740 DOI: 10.3390/pharmaceutics11030119] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 02/26/2019] [Accepted: 03/07/2019] [Indexed: 02/02/2023] Open
Abstract
The ability to understand the complexity of cancer-related data has been prompted by the applications of (1) computer and data sciences, including data mining, predictive analytics, machine learning, and artificial intelligence, and (2) advances in imaging technology and probe development. Computational modelling and simulation are systematic and cost-effective tools able to identify important temporal/spatial patterns (and relationships), characterize distinct molecular features of cancer states, and address other relevant aspects, including tumor detection and heterogeneity, progression and metastasis, and drug resistance. These approaches have provided invaluable insights for improving the experimental design of therapeutic delivery systems and for increasing the translational value of the results obtained from early and preclinical studies. The big question is: Could cancer theranostics be determined and controlled in silico? This review describes the recent progress in the development of computational models and methods used to facilitate research on the molecular basis of cancer and on the respective diagnosis and optimized treatment, with particular emphasis on the design and optimization of theranostic systems. The current role of computational approaches is providing innovative, incremental, and complementary data-driven solutions for the prediction, simplification, and characterization of cancer and intrinsic mechanisms, and to promote new data-intensive, accurate diagnostics and therapeutics.
Collapse
Affiliation(s)
- Tânia F G G Cova
- Coimbra Chemistry Centre, Department of Chemistry, Faculty of Sciences and Technology, University of Coimbra, 3004-535 Coimbra, Portugal.
| | - Daniel J Bento
- Coimbra Chemistry Centre, Department of Chemistry, Faculty of Sciences and Technology, University of Coimbra, 3004-535 Coimbra, Portugal.
| | - Sandra C C Nunes
- Coimbra Chemistry Centre, Department of Chemistry, Faculty of Sciences and Technology, University of Coimbra, 3004-535 Coimbra, Portugal.
| |
Collapse
|
29
|
Efficacy of Surface-Modified PLGA Nanoparticles as a Function of Cervical Cancer Type. Pharm Res 2019; 36:66. [PMID: 30868271 DOI: 10.1007/s11095-019-2602-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/04/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Hypovascularization of cervical tumors, coupled with intrinsic and acquired drug resistance, has contributed to marginal therapeutic outcomes by hindering chemotherapeutic transport and efficacy. Recently, the heterogeneous penetration and distribution of cell penetrating peptide (CPP, here MPG) and polyethylene glycol (PEG) modified poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) were evaluated as a function of tumor type and morphology in cervical cancer spheroids modeling hypovascularized tumor nodules. Building upon this work, this study investigates the efficacy imparted by surface-modified Doxorubicin-loaded NPs transported into hypovascularized tissue. METHODS NP efficacy was measured in HeLa, CaSki, and SiHa cells. NP internalization and association, and associated cell viability, were determined in monolayer and spheroid models. RESULTS MPG and PEG-NP co-treatment was most efficacious in HeLa cells, while PEG NPs were most efficacious in CaSki cells. NP surface-modifications were unable to improve efficacy, relative to unmodified NPs, in SiHa cells. CONCLUSIONS The results highlight the dependence of efficacy on tumor type and the associated microenvironment. The results further relate previous NP transport studies to efficacy, as a function of surface-modification and cell type. Longer-term, this information may help guide the design of NP-mediated strategies to maximize efficacy based on patient-specific cervical tumor origin and characteristics.
Collapse
|
30
|
He H, Yuan D, Wu Y, Cao Y. Pharmacokinetics and Pharmacodynamics Modeling and Simulation Systems to Support the Development and Regulation of Liposomal Drugs. Pharmaceutics 2019; 11:E110. [PMID: 30866479 PMCID: PMC6471205 DOI: 10.3390/pharmaceutics11030110] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/25/2019] [Accepted: 02/28/2019] [Indexed: 12/27/2022] Open
Abstract
Liposomal formulations have been developed to improve the therapeutic index of encapsulated drugs by altering the balance of on- and off-targeted distribution. The improved therapeutic efficacy of liposomal drugs is primarily attributed to enhanced distribution at the sites of action. The targeted distribution of liposomal drugs depends not only on the physicochemical properties of the liposomes, but also on multiple components of the biological system. Pharmacokinetic⁻pharmacodynamic (PK⁻PD) modeling has recently emerged as a useful tool with which to assess the impact of formulation- and system-specific factors on the targeted disposition and therapeutic efficacy of liposomal drugs. The use of PK⁻PD modeling to facilitate the development and regulatory reviews of generic versions of liposomal drugs recently drew the attention of the U.S. Food and Drug Administration. The present review summarizes the physiological factors that affect the targeted delivery of liposomal drugs, challenges that influence the development and regulation of liposomal drugs, and the application of PK⁻PD modeling and simulation systems to address these challenges.
Collapse
Affiliation(s)
- Hua He
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China.
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Dongfen Yuan
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Yun Wu
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, 332 Bonner Hall, Buffalo, NY 14260, USA.
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
31
|
Benchimol MJ, Bourne D, Moghimi SM, Simberg D. Pharmacokinetic analysis reveals limitations and opportunities for nanomedicine targeting of endothelial and extravascular compartments of tumours. J Drug Target 2019; 27:690-698. [PMID: 30614276 DOI: 10.1080/1061186x.2019.1566339] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Targeting of nanoparticles to tumours can potentially improve the specificity of imaging and treatments. We have developed a multicompartmental pharmacokinetic model in order to analyse some of the factors that control efficiency of targeting to intravascular (endothelium) and extravascular (tumour cells and stroma) compartments. We make the assumption that transport across tumour endothelium is an important step for subsequent nanoparticle accumulation in the tumour (area-under-the-curve, AUC) regardless of entry route (interendothelial and transendothelial routes) and study this through a multicompartmental simulation. Our model reveals that increasing endothelial targeting efficiency has a much stronger effect on the AUC than increasing extravascular targeting efficiency. Furthermore, our analysis reveals that both extravasation and intratumoral diffusion rates need to be increased in order to significantly increase the AUC of extravascular-targeted nanoparticles. Increasing the nanoparticle circulation half-life increases the AUC independently of extravasation and intratumoral diffusion. Targeting the extravascular compartment leads to a buildup in the first layer surrounding blood vessels at the expense of deeper layers (binding site barrier). This model explains some of the limitations of tumour targeting and provides important guidelines for the design of targeted nanomedicines.
Collapse
Affiliation(s)
| | - David Bourne
- b The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus , Aurora , CO , USA.,c Center for Translational Pharmacokinetics and Pharmacogenomics , The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus , Aurora , CO , USA
| | - Seyed Moein Moghimi
- d Colorado Center for Nanomedicine and Nanosafety , Aurora , CO , USA.,e School of Pharmacy, The Faculty of Medical Sciences, King George VI Building , Newcastle University , Newcastle upon Tyne , UK.,f Division of Stratified Medicine, Biomarkers & Therapeutics , Institute of Cellular Medicine, Newcastle University , Newcastle upon Tyne , UK
| | - Dmitri Simberg
- b The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus , Aurora , CO , USA.,d Colorado Center for Nanomedicine and Nanosafety , Aurora , CO , USA
| |
Collapse
|
32
|
Chamseddine IM, Frieboes HB, Kokkolaras M. Design Optimization of Tumor Vasculature-Bound Nanoparticles. Sci Rep 2018; 8:17768. [PMID: 30538267 PMCID: PMC6290012 DOI: 10.1038/s41598-018-35675-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/26/2018] [Indexed: 01/07/2023] Open
Abstract
Nanotherapy may constitute a promising approach to target tumors with anticancer drugs while minimizing systemic toxicity. Computational modeling can enable rapid evaluation of nanoparticle (NP) designs and numerical optimization. Here, an optimization study was performed using an existing tumor model to find NP size and ligand density that maximize tumoral NP accumulation while minimizing tumor size. Optimal NP avidity lies at lower bound of feasible values, suggesting reduced ligand density to prolong NP circulation. For the given set of tumor parameters, optimal NP diameters were 288 nm to maximize NP accumulation and 334 nm to minimize tumor diameter, leading to uniform NP distribution and adequate drug load. Results further show higher dependence of NP biodistribution on the NP design than on tumor morphological parameters. A parametric study with respect to drug potency was performed. The lower the potency of the drug, the bigger the difference is between the maximizer of NP accumulation and the minimizer of tumor size, indicating the existence of a specific drug potency that minimizes the differential between the two optimal solutions. This study shows the feasibility of applying optimization to NP designs to achieve efficacious cancer nanotherapy, and offers a first step towards a quantitative tool to support clinical decision making.
Collapse
Affiliation(s)
| | - Hermann B Frieboes
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Michael Kokkolaras
- Department of Mechanical Engineering, McGill University, Montreal, Quebec, Canada.
- GERAD - Group for Research in Decision Analysis, Montreal, Quebec, Canada.
| |
Collapse
|
33
|
Miller HA, Frieboes HB. Evaluation of Drug-Loaded Gold Nanoparticle Cytotoxicity as a Function of Tumor Vasculature-Induced Tissue Heterogeneity. Ann Biomed Eng 2018; 47:257-271. [PMID: 30298374 DOI: 10.1007/s10439-018-02146-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 10/01/2018] [Indexed: 01/10/2023]
Abstract
The inherent heterogeneity of tumor tissue presents a major challenge to nanoparticle-mediated drug delivery. This heterogeneity spans from the molecular (genomic, proteomic, metabolomic) to the cellular (cell types, adhesion, migration) and to the tissue (vasculature, extra-cellular matrix) scales. In particular, tumor vasculature forms abnormally, inducing proliferative, hypoxic, and necrotic tumor tissue regions. As the vasculature is the main conduit for nanotherapy transport into tumors, vasculature-induced tissue heterogeneity can cause local inadequate delivery and concentration, leading to subpar response. Further, hypoxic tissue, although viable, would be immune to the effects of cell-cycle specific drugs. In order to enable a more systematic evaluation of such effects, here we employ computational modeling to study the therapeutic response as a function of vasculature-induced tumor tissue heterogeneity. Using data with three-layered gold nanoparticles loaded with cisplatin, nanotherapy is simulated interacting with different levels of tissue heterogeneity, and the treatment response is measured in terms of tumor regression. The results quantify the influence that varying levels of tumor vascular density coupled with the drug strength have on nanoparticle uptake and washout, and the associated tissue response. The drug strength affects the proportion of proliferating, hypoxic, and necrotic tissue fractions, which in turn dynamically affect and are affected by the vascular density. Higher drug strengths may be able to achieve stronger tumor regression but only if the intra-tumoral vascular density is above a certain threshold that affords sufficient transport. This study establishes an initial step towards a more systematic methodology to assess the effect of vasculature-induced tumor tissue heterogeneity on the response to nanotherapy.
Collapse
Affiliation(s)
- Hunter A Miller
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Hermann B Frieboes
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA. .,Department of Bioengineering, University of Louisville, Lutz Hall 419, Louisville, KY, 40292, USA. .,James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
34
|
Gomez-Garcia MJ, Doiron AL, Steele RRM, Labouta HI, Vafadar B, Shepherd RD, Gates ID, Cramb DT, Childs SJ, Rinker KD. Nanoparticle localization in blood vessels: dependence on fluid shear stress, flow disturbances, and flow-induced changes in endothelial physiology. NANOSCALE 2018; 10:15249-15261. [PMID: 30066709 DOI: 10.1039/c8nr03440k] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Nanoparticles in the bloodstream are subjected to complex fluid forces as they move through the curves and branches of healthy or tumor vasculature. While nanoparticles are known to preferentially accumulate in angiogenic vessels, little is known about the flow conditions in these vessels and how these conditions may influence localization. Here, we report a methodology which combines confocal imaging of nanoparticle-injected transgenic zebrafish embryos, 3D modeling of the vasculature, particle mapping, and computational fluid dynamics, to quantitatively assess the effects of fluid forces on nanoparticle distribution in vivo. Six-fold lower accumulation was found in zebrafish arteries compared to the lower velocity veins. Nanoparticle localization varied inversely with shear stress. Highest accumulation was present in regions of disturbed flow found at branch points and curvatures in the vasculature. To further investigate cell-particle association under flow, human endothelial cells were exposed to nanoparticles under hemodynamic conditions typically found in human vessels. Physiological adaptations of endothelial cells to 20 hours of flow enhanced nanoparticle accumulation in regions of disturbed flow. Overall our results suggest that fluid shear stress magnitude, flow disturbances, and flow-induced changes in endothelial physiology modulate nanoparticle localization in angiogenic vessels.
Collapse
|
35
|
Hassanzadeh P, Atyabi F, Dinarvand R. Ignoring the modeling approaches: Towards the shadowy paths in nanomedicine. J Control Release 2018; 280:58-75. [DOI: 10.1016/j.jconrel.2018.04.042] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/22/2018] [Accepted: 04/23/2018] [Indexed: 12/30/2022]
|
36
|
Sims LB, Frieboes HB, Steinbach-Rankins JM. Nanoparticle-mediated drug delivery to treat infections in the female reproductive tract: evaluation of experimental systems and the potential for mathematical modeling. Int J Nanomedicine 2018; 13:2709-2727. [PMID: 29760551 PMCID: PMC5937491 DOI: 10.2147/ijn.s160044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
A variety of drug-delivery platforms have been employed to deliver therapeutic agents across cervicovaginal mucus (CVM) and the vaginal mucosa, offering the capability to increase the longevity and retention of active agents to treat infections of the female reproductive tract (FRT). Nanoparticles (NPs) have been shown to improve retention, diffusion, and cell-specific targeting via specific surface modifications, relative to other delivery platforms. In particular, polymeric NPs represent a promising option that has shown improved distribution through the CVM. These NPs are typically fabricated from nontoxic, non-inflammatory, US Food and Drug Administration-approved polymers that improve biocompatibility. This review summarizes recent experimental studies that have evaluated NP transport in the FRT, and highlights research areas that more thoroughly and efficiently inform polymeric NP design, including mathematical modeling. An overview of the in vitro, ex vivo, and in vivo NP studies conducted to date – whereby transport parameters are determined, extrapolated, and validated – is presented first. The impact of different NP design features on transport through the FRT is summarized, and gaps that exist due to the limitations of iterative experimentation alone are identified. The potential of mathematical modeling to complement the characterization and evaluation of diffusion and transport of delivery vehicles and active agents through the CVM and mucosa is discussed. Lastly, potential advancements combining experimental and mathematical knowledge are suggested to inform next-generation NP designs, such that infections in the FRT may be more effectively treated.
Collapse
Affiliation(s)
- Lee B Sims
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Hermann B Frieboes
- Department of Bioengineering, University of Louisville, Louisville, KY, USA.,James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Jill M Steinbach-Rankins
- Department of Bioengineering, University of Louisville, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA.,Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA.,Center for Predictive Medicine, University of Louisville, Louisville, KY, USA
| |
Collapse
|
37
|
Ozdemir-Kaynak E, Qutub AA, Yesil-Celiktas O. Advances in Glioblastoma Multiforme Treatment: New Models for Nanoparticle Therapy. Front Physiol 2018; 9:170. [PMID: 29615917 PMCID: PMC5868458 DOI: 10.3389/fphys.2018.00170] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 02/20/2018] [Indexed: 11/30/2022] Open
Abstract
The most lethal form of brain cancer, glioblastoma multiforme, is characterized by rapid growth and invasion facilitated by cell migration and degradation of the extracellular matrix. Despite technological advances in surgery and radio-chemotherapy, glioblastoma remains largely resistant to treatment. New approaches to study glioblastoma and to design optimized therapies are greatly needed. One such approach harnesses computational modeling to support the design and delivery of glioblastoma treatment. In this paper, we critically summarize current glioblastoma therapy, with a focus on emerging nanomedicine and therapies that capitalize on cell-specific signaling in glioblastoma. We follow this summary by discussing computational modeling approaches focused on optimizing these emerging nanotherapeutics for brain cancer. We conclude by illustrating how mathematical analysis can be used to compare the delivery of a high potential anticancer molecule, delphinidin, in both free and nanoparticle loaded forms across the blood-brain barrier for glioblastoma.
Collapse
Affiliation(s)
- Elif Ozdemir-Kaynak
- Department of Bioengineering, Faculty of Engineering, Ege University, Bornova-Izmir, Turkey
| | - Amina A Qutub
- Department of Bioengineering, Rice University, Houston, TX, United States
| | - Ozlem Yesil-Celiktas
- Department of Bioengineering, Faculty of Engineering, Ege University, Bornova-Izmir, Turkey.,Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, United States
| |
Collapse
|
38
|
Konstorum A, Lowengrub JS. Activation of the HGF/c-Met axis in the tumor microenvironment: A multispecies model. J Theor Biol 2017; 439:86-99. [PMID: 29203124 DOI: 10.1016/j.jtbi.2017.11.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/24/2017] [Accepted: 11/30/2017] [Indexed: 02/06/2023]
Abstract
The tumor microenvironment is an integral component in promoting tumor development. Cancer-associated fibroblasts (CAFs), which reside in the tumor stroma, produce Hepatocyte Growth Factor (HGF), an important trigger for invasive and metastatic tumor behavior. HGF contributes to a pro-tumorigenic environment by activating its cognate receptor, c-Met, on tumor cells. Tumor cells, in turn, secrete growth factors that upregulate HGF production in CAFs, thereby establishing a dynamic tumor-host signaling program. Using a spatiotemporal multispecies model of tumor growth, we investigate how the development and spread of a tumor is impacted by the initiation of a dynamic interaction between tumor-derived growth factors and CAF-derived HGF. We show that establishment of such an interaction results in increased tumor growth and morphological instability, the latter due in part to increased cell species heterogeneity at the tumor-host boundary. Invasive behavior is further increased if the tumor lowers responsiveness to paracrine pro-differentiation signals, which is a hallmark of neoplastic development. By modeling anti-HGF and anti-c-Met therapy, we show how disruption of the HGF/c-Met axis can reduce tumor invasiveness and growth, thereby providing theoretical evidence that targeting tumor-microenvironment interactions is a promising avenue for therapeutic development.
Collapse
Affiliation(s)
- Anna Konstorum
- Center for Quantitative Medicine, UConn Health, Farmington, CT, USA.
| | - John S Lowengrub
- Department of Mathematics, University of California, Irvine, CA, USA; Center for Complex Biological Systems, University of California, Irvine, CA, USA; Department of Biomedical Engineering, University of California, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, USA.
| |
Collapse
|
39
|
Abstract
In vivo imaging, which enables us to peer deeply within living subjects, is producing tremendous opportunities both for clinical diagnostics and as a research tool. Contrast material is often required to clearly visualize the functional architecture of physiological structures. Recent advances in nanomaterials are becoming pivotal to generate the high-resolution, high-contrast images needed for accurate, precision diagnostics. Nanomaterials are playing major roles in imaging by delivering large imaging payloads, yielding improved sensitivity, multiplexing capacity, and modularity of design. Indeed, for several imaging modalities, nanomaterials are now not simply ancillary contrast entities, but are instead the original and sole source of image signal that make possible the modality's existence. We address the physicochemical makeup/design of nanomaterials through the lens of the physical properties that produce contrast signal for the cognate imaging modality-we stratify nanomaterials on the basis of their (i) magnetic, (ii) optical, (iii) acoustic, and/or (iv) nuclear properties. We evaluate them for their ability to provide relevant information under preclinical and clinical circumstances, their in vivo safety profiles (which are being incorporated into their chemical design), their modularity in being fused to create multimodal nanomaterials (spanning multiple different physical imaging modalities and therapeutic/theranostic capabilities), their key properties, and critically their likelihood to be clinically translated.
Collapse
Affiliation(s)
- Bryan Ronain Smith
- Stanford University , 3155 Porter Drive, #1214, Palo Alto, California 94304-5483, United States
| | - Sanjiv Sam Gambhir
- The James H. Clark Center , 318 Campus Drive, First Floor, E-150A, Stanford, California 94305-5427, United States
| |
Collapse
|
40
|
Abstract
Recently, there has been an emerging interest in controlling 3D structures and designing novel 3D shapes for drug carriers at nano- and micro-scales. Certain 3D shapes and structures of drug particles enable transportation of the drugs to desired areas of the body, allow drugs to target specific cells and tissues, and influence release kinetics. Advanced nano- and micro-manufacturing methods including 3D printing, photolithography-based processes, microfluidics and DNA origami have been developed to generate defined 3D shapes and structures for drug carriers. This paper reviews the importance of 3D structures and shapes on controlled drug delivery, and the current state-of-the-art technologies that allow the creation of novel 3D drug carriers at nano- and micro-scales.
Collapse
|
41
|
Decuzzi P. Facilitating the Clinical Integration of Nanomedicines: The Roles of Theoretical and Computational Scientists. ACS NANO 2016; 10:8133-8. [PMID: 27604416 DOI: 10.1021/acsnano.6b05536] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Since the launch of multiple research initiatives on nanotechnology applied to medicine in the early 2000s, a plethora of nanomedicines have been developed that exhibit great therapeutic efficacy in preclinical models but yet minimal impact in daily clinical practice. The successful and complete clinical fruition of nanomedicines requires addressing three major technical challenges: improving loading efficacy and on-command release, modulating recognition and sequestration by immune cells, and maximizing accumulation at biological targets. In this Perspective, I describe how theoretical and computational models can help address each of these challenges. This armamentarium represents an ideal tool for maximizing the therapeutic efficacy of nanomedicines, thus facilitating their integration into daily clinical operations.
Collapse
Affiliation(s)
- Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia Via Morego 30, Genoa 16163, Italy
| |
Collapse
|
42
|
Generalized plasma skimming model for cells and drug carriers in the microvasculature. Biomech Model Mechanobiol 2016; 16:497-507. [PMID: 27655421 DOI: 10.1007/s10237-016-0832-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 09/09/2016] [Indexed: 02/06/2023]
Abstract
In microvascular transport, where both blood and drug carriers are involved, plasma skimming has a key role on changing hematocrit level and drug carrier concentration in capillary beds after continuous vessel bifurcation in the microvasculature. While there have been numerous studies on modeling the plasma skimming of blood, previous works lacked in consideration of its interaction with drug carriers. In this paper, a generalized plasma skimming model is suggested to predict the redistributions of both the cells and drug carriers at each bifurcation. In order to examine its applicability, this new model was applied on a single bifurcation system to predict the redistribution of red blood cells and drug carriers. Furthermore, this model was tested at microvascular network level under different plasma skimming conditions for predicting the concentration of drug carriers. Based on these results, the applicability of this generalized plasma skimming model is fully discussed and future works along with the model's limitations are summarized.
Collapse
|
43
|
Thomann S, Baek S, Ryschich E. Impact of wall shear stress and ligand avidity on binding of anti-CD146-coated nanoparticles to murine tumor endothelium under flow. Oncotarget 2016; 6:39960-8. [PMID: 26503468 PMCID: PMC4741872 DOI: 10.18632/oncotarget.5662] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 10/02/2015] [Indexed: 12/18/2022] Open
Abstract
The endothelial phenotype of tumor blood vessels differs from the liver and forms an important base for endothelium-specific targeting by antibody-coated nanoparticles. Although differences of shear stress and ligand avidity can modulate the nanoparticle binding to endothelium, these mechanisms are still poorly studied. This study analyzed the binding of antibody-coated nanoparticles to tumor and liver endothelium under controlled flow conditions and verified this binding in tumor models in vivo. Binding of anti-CD146-coated nanoparticles, but not of antibody was significantly reduced under increased wall shear stress and the degree of nanoparticle binding correlated with the avidity of the coating. The intravascular wall shear stress favors nanoparticle binding at the site of higher avidity of endothelial epitope which additionally promotes the selectivity to tumor endothelium. After intravenous application in vivo, pegylated self-coated nanoparticles showed specific binding to tumor endothelium, whereas the nanoparticle binding to the liver endothelium was very low. This study provides a rationale that selective binding of mAb-coated nanoparticles to tumor endothelium is achieved by two factors: higher expression of endothelial epitope and higher nanoparticle shearing from liver endothelium. The combination of endothelial marker targeting and the use of shear stress-controlled nanoparticle capture can be used for selective intratumoral drug delivery.
Collapse
Affiliation(s)
- Stefan Thomann
- Department of Surgery, University of Heidelberg, Heidelberg, Germany.,Department of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Sunhwa Baek
- Department of Surgery, University of Heidelberg, Heidelberg, Germany
| | - Eduard Ryschich
- Department of Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
44
|
Ben Amar M, Bianca C. Towards a unified approach in the modeling of fibrosis: A review with research perspectives. Phys Life Rev 2016; 17:61-85. [DOI: 10.1016/j.plrev.2016.03.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 03/29/2016] [Indexed: 12/12/2022]
|
45
|
Wang Z, Kerketta R, Chuang YL, Dogra P, Butner JD, Brocato TA, Day A, Xu R, Shen H, Simbawa E, AL-Fhaid AS, Mahmoud SR, Curley SA, Ferrari M, Koay EJ, Cristini V. Theory and Experimental Validation of a Spatio-temporal Model of Chemotherapy Transport to Enhance Tumor Cell Kill. PLoS Comput Biol 2016; 12:e1004969. [PMID: 27286441 PMCID: PMC4902302 DOI: 10.1371/journal.pcbi.1004969] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 05/09/2016] [Indexed: 12/14/2022] Open
Abstract
It has been hypothesized that continuously releasing drug molecules into the tumor over an extended period of time may significantly improve the chemotherapeutic efficacy by overcoming physical transport limitations of conventional bolus drug treatment. In this paper, we present a generalized space- and time-dependent mathematical model of drug transport and drug-cell interactions to quantitatively formulate this hypothesis. Model parameters describe: perfusion and tissue architecture (blood volume fraction and blood vessel radius); diffusion penetration distance of drug (i.e., a function of tissue compactness and drug uptake rates by tumor cells); and cell death rates (as function of history of drug uptake). We performed preliminary testing and validation of the mathematical model using in vivo experiments with different drug delivery methods on a breast cancer mouse model. Experimental data demonstrated a 3-fold increase in response using nano-vectored drug vs. free drug delivery, in excellent quantitative agreement with the model predictions. Our model results implicate that therapeutically targeting blood volume fraction, e.g., through vascular normalization, would achieve a better outcome due to enhanced drug delivery.
Collapse
Affiliation(s)
- Zhihui Wang
- Department of NanoMedicine and Biomedical Engineering, University of Texas Medical School at Houston, Houston, Texas, United States of America
- Brown Foundation Institute of Molecular Medicine, University of Texas Medical School at Houston, Houston, Texas, United States of America
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Romica Kerketta
- Department of Pathology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Yao-Li Chuang
- Department of Mathematics, California State University, Northridge, California, United States of America
| | - Prashant Dogra
- Department of Pathology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Joseph D. Butner
- Department of Chemical and Biological Engineering and Center for Biomedical Engineering, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Terisse A. Brocato
- Department of Chemical and Biological Engineering and Center for Biomedical Engineering, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Armin Day
- Department of Pathology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Rong Xu
- Department of Nanomedicine, Methodist Hospital Research Institute, Houston, Texas, United States of America
| | - Haifa Shen
- Department of Nanomedicine, Methodist Hospital Research Institute, Houston, Texas, United States of America
| | - Eman Simbawa
- Department of Mathematics, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - A. S. AL-Fhaid
- Department of Mathematics, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - S. R. Mahmoud
- Department of Mathematics, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Steven A. Curley
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States of America
| | - Mauro Ferrari
- Department of Nanomedicine, Methodist Hospital Research Institute, Houston, Texas, United States of America
| | - Eugene J. Koay
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail: (EJK); (VC)
| | - Vittorio Cristini
- Department of NanoMedicine and Biomedical Engineering, University of Texas Medical School at Houston, Houston, Texas, United States of America
- Brown Foundation Institute of Molecular Medicine, University of Texas Medical School at Houston, Houston, Texas, United States of America
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of Mathematics, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- * E-mail: (EJK); (VC)
| |
Collapse
|
46
|
Mathematical Based Calculation of Drug Penetration Depth in Solid Tumors. BIOMED RESEARCH INTERNATIONAL 2016; 2016:8437247. [PMID: 27376087 PMCID: PMC4916326 DOI: 10.1155/2016/8437247] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 05/17/2016] [Indexed: 01/19/2023]
Abstract
Cancer is a class of diseases characterized by out-of-control cells' growth which affect cells and make them damaged. Many treatment options for cancer exist. Chemotherapy as an important treatment option is the use of drugs to treat cancer. The anticancer drug travels to the tumor and then diffuses in it through capillaries. The diffusion of drugs in the solid tumor is limited by penetration depth which is different in case of different drugs and cancers. The computation of this depth is important as it helps physicians to investigate about treatment of infected tissue. Although many efforts have been made on studying and measuring drug penetration depth, less works have been done on computing this length from a mathematical point of view. In this paper, first we propose phase lagging model for diffusion of drug in the tumor. Then, using this model on one side and considering the classic diffusion on the other side, we compute the drug penetration depth in the solid tumor. This computed value of drug penetration depth is corroborated by comparison with the values measured by experiments.
Collapse
|
47
|
Kirui DK, Ferrari M. Intravital Microscopy Imaging Approaches for Image-Guided Drug Delivery Systems. Curr Drug Targets 2016; 16:528-41. [PMID: 25901526 DOI: 10.2174/1389450116666150330114030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 12/10/2014] [Accepted: 03/13/2015] [Indexed: 12/31/2022]
Abstract
Rapid technical advances in the field of non-linear microscopy have made intravital microscopy a vital pre-clinical tool for research and development of imaging-guided drug delivery systems. The ability to dynamically monitor the fate of macromolecules in live animals provides invaluable information regarding properties of drug carriers (size, charge, and surface coating), physiological, and pathological processes that exist between point-of-injection and the projected of site of delivery, all of which influence delivery and effectiveness of drug delivery systems. In this Review, we highlight how integrating intravital microscopy imaging with experimental designs (in vitro analyses and mathematical modeling) can provide unique information critical in the design of novel disease-relevant drug delivery platforms with improved diagnostic and therapeutic indexes. The Review will provide the reader an overview of the various applications for which intravital microscopy has been used to monitor the delivery of diagnostic and therapeutic agents and discuss some of their potential clinical applications.
Collapse
Affiliation(s)
| | - Mauro Ferrari
- Houston Methodist Research Institute, Department of NanoMedicine, 6670 Bertner Avenue, MS R8-460, Houston, TX 77030, USA.
| |
Collapse
|
48
|
Sims LB, Curtis LT, Frieboes HB, Steinbach-Rankins JM. Enhanced uptake and transport of PLGA-modified nanoparticles in cervical cancer. J Nanobiotechnology 2016; 14:33. [PMID: 27102372 PMCID: PMC4840861 DOI: 10.1186/s12951-016-0185-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/12/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Uncoordinated cellular proliferation and dysregulated angiogenesis in solid tumors are coupled with inadequate tissue, blood, and lymphatic vascularization. Consequently, tumors are often characterized by hypoxic regions with limited access to vascular-borne substances. In particular, systemically administered nanoparticles (NPs) targeting tumor cells and relying on vascular access to reach tumor tissue can suffer from limited therapeutic efficacy due to inhomogeneous intra-tumor distribution and insufficient cellular internalization of NPs. To circumvent these challenges, NP surfaces can be modified to facilitate tumor interstitial transport and cellular uptake. RESULTS We create poly(lactic-co-glycolic) acid NPs modified with MPG, polyethylene glycol (PEG), MPG/PEG, and Vimentin (VIM), and evaluate their cellular uptake in 2D (monolayer) cell culture of human cervical carcinoma (HeLa). We compare NP performance by evaluating uptake by non-cancerous vaginal (VK2) cells. We further assess NP interstitial transport in hypo-vascularized lesions by evaluating the effect of the various modifications on NP penetration in 3D cell culture of the HeLa cells. Results show that after 24 h incubation with HeLa cells in monolayer, MPG, MPG/PEG, PEG, and VIM NPs were internalized at 66×, 24×, 30×, and 15× that of unmodified NPs, respectively. In contrast, incubation with VK2 cells in monolayer showed that MPG , MPG/PEG , PEG , and VIM NPs internalized at 6.3×, 4.3×, 12.4×, and 3.0× that of unmodified NPs, respectively. Uptake was significantly enhanced in tumorigenic vs. normal cells, with internalization of MPG NPs by HeLa cells being twice that of PEG NPs by VK2 cells. After 24 h incubation in HeLa 3D cell culture, MPG and MPG/PEGNPs were internalized 2× and 3× compared to PEG and VIM NPs, respectively. Whereas MPG NPs were internalized mostly in the cell culture periphery (1.2×, 1.4×, and 2.7× that of PEG, MPG/PEG, and VIM NPs, respectively), PEG NPs at 250 μm penetrated 2× farther into the tissue culture than MPG NPs. For all NP types, cellular internalization was severely hindered in 3D compared to monolayer. CONCLUSIONS Although MPG surface modification enhances internalization and uptake in hypo-vascularized cervical tissue culture, coating with PEG reduces this internalization while enhancing penetration. A delivery strategy combining NPs with either modification may balance cellular internalization vs. tissue penetration in hypo-vascularized cervical cancer lesions.
Collapse
Affiliation(s)
- Lee B Sims
- Department of Bioengineering, University of Louisville, 505 S. Hancock, CTRB 623, Louisville, KY, 40208, USA
| | - Louis T Curtis
- Department of Bioengineering, University of Louisville, 505 S. Hancock, CTRB 623, Louisville, KY, 40208, USA
| | - Hermann B Frieboes
- Department of Bioengineering, University of Louisville, 505 S. Hancock, CTRB 623, Louisville, KY, 40208, USA.,James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Jill M Steinbach-Rankins
- Department of Bioengineering, University of Louisville, 505 S. Hancock, CTRB 623, Louisville, KY, 40208, USA. .,Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA. .,Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA. .,Center for Predictive Medicine, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
49
|
Hoshyar N, Gray S, Han H, Bao G. The effect of nanoparticle size on in vivo pharmacokinetics and cellular interaction. Nanomedicine (Lond) 2016; 11:673-92. [PMID: 27003448 DOI: 10.2217/nnm.16.5] [Citation(s) in RCA: 1160] [Impact Index Per Article: 128.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Nanoparticle-based technologies offer exciting new approaches to disease diagnostics and therapeutics. To take advantage of unique properties of nanoscale materials and structures, the size, shape and/or surface chemistry of nanoparticles need to be optimized, allowing their functionalities to be tailored for different biomedical applications. Here we review the effects of nanoparticle size on cellular interaction and in vivo pharmacokinetics, including cellular uptake, biodistribution and circulation half-life of nanoparticles. Important features of nanoparticle probes for molecular imaging and modeling of nanoparticle size effects are also discussed.
Collapse
Affiliation(s)
- Nazanin Hoshyar
- Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA
| | - Samantha Gray
- Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA
| | - Hongbin Han
- Department of Radiology, Peking University Third Hospital, Beijing 100191, China
| | - Gang Bao
- Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA.,Department of Bioengineering, Rice University, Houston, TX 77030, USA
| |
Collapse
|
50
|
Anselmo AC, Mitragotri S. A chemical engineering perspective of nanoparticle-based targeted drug delivery: A unit process approach. AIChE J 2016. [DOI: 10.1002/aic.15189] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Aaron C. Anselmo
- David H. Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139
| | - Samir Mitragotri
- Dept. of Chemical Engineering, Center for Bioengineering; University of California; Santa Barbara CA 93106 and Editor-in-Chief, Bioengineering & Translational Medicine
| |
Collapse
|