1
|
The Proper Administration Sequence of Radiotherapy and Anti-Vascular Agent-DMXAA Is Essential to Inhibit the Growth of Melanoma Tumors. Cancers (Basel) 2021; 13:cancers13163924. [PMID: 34439079 PMCID: PMC8394873 DOI: 10.3390/cancers13163924] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/30/2021] [Accepted: 07/30/2021] [Indexed: 01/13/2023] Open
Abstract
Vascular disrupting agents (VDAs), such as DMXAA, effectively destroy tumor blood vessels and cause the formation of large areas of necrosis in the central parts of the tumors. However, the use of VDAs is associated with hypoxia activation and residues of rim cells on the edge of the tumor that are responsible for tumor regrowth. The aim of the study was to combine DMXAA with radiotherapy (brachytherapy) and find the appropriate administration sequence to obtain the maximum synergistic therapeutic effect. We show that the combination in which tumors were irradiated prior to VDAs administration is more effective in murine melanoma growth inhibition than in either of the agents individually or in reverse combination. For the first time, the significance of immune cells' activation in such a combination is demonstrated. The inhibition of tumor growth is linked to the reduction of tumor blood vessels, the increased infiltration of CD8+ cytotoxic T lymphocytes and NK cells and the polarization of macrophages to the cytotoxic M1 phenotype. The reverse combination of therapeutic agents showed no therapeutic effect and even abolished the effect of DMXAA. The combination of brachytherapy and vascular disrupting agent effectively inhibits the growth of melanoma tumors but requires careful planning of the sequence of administration of the agents.
Collapse
|
2
|
Zhao D, Huang X, Zhang Z, Ding J, Cui Y, Chen X. Engineered nanomedicines for tumor vasculature blockade therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1691. [PMID: 33480163 DOI: 10.1002/wnan.1691] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/29/2020] [Accepted: 11/30/2020] [Indexed: 12/21/2022]
Abstract
Tumor vasculature blockade therapy (TVBT), including angiogenesis inhibition, vascular disruption, and vascular infarction, provides a promising treatment modality for solid tumors. However, low selectivity, drug resistance, and possible severe side effects have limited the clinical transformation of TVBT. Engineered nanoparticles offer potential solutions, including prolonged circulation time, targeted transportation, and controlled release of TVBT agents. Moreover, engineered nanomedicines provide a promising combination platform of TVBT with chemotherapy, radiotherapy, photodynamic therapy, photothermal therapy, ultrasound therapy, and gene therapy. In this article, we offer a comprehensive summary of the current progress of engineered nanomedicines for TVBT and also discuss current deficiencies and future directions for TVBT development. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Duoyi Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Xu Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China.,Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Zhiyu Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Yan Cui
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| |
Collapse
|
3
|
Kim MY, Shin JY, Kim JO, Son KH, Kim YS, Jung CK, Kang JH. Anti-tumor efficacy of CKD-516 in combination with radiation in xenograft mouse model of lung squamous cell carcinoma. BMC Cancer 2020; 20:1057. [PMID: 33143663 PMCID: PMC7607852 DOI: 10.1186/s12885-020-07566-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 10/26/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Hypoxic tumors are known to be highly resistant to radiotherapy and cause poor prognosis in non-small cell lung cancer (NSCLC) patients. CKD-516, a novel vascular disrupting agent (VDA), mainly affects blood vessels in the central area of the tumor and blocks tubulin polymerization, thereby destroying the aberrant tumor vasculature with a rapid decrease in blood, resulting in rapid tumor cell death. Therefore, we evaluated the anti-tumor efficacy of CKD-516 in combination with irradiation (IR) and examined tumor necrosis, delayed tumor growth, and expression of proteins involved in hypoxia and angiogenesis in this study. METHODS A xenograft mouse model of lung squamous cell carcinoma was established, and the tumor was exposed to IR 5 days per week. CKD-516 was administered with two treatment schedules (day 1 or days 1 and 5) 1 h after IR. After treatment, tumor tissues were stained with hematoxylin and eosin, and pimonidazole. HIF-1α, Glut-1, VEGF, CD31, and Ki-67 expression levels were evaluated using immunohistochemical staining. RESULTS Short-term treatment with IR alone and CKD-516 + IR (d1) significantly reduced tumor volume (p = 0.006 and p = 0.048, respectively). Treatment with CKD-516 + IR (d1 and d1, 5) resulted in a marked reduction in the number of blood vessels (p < 0.005). More specifically, CKD-516 + IR (d1) caused the most extensive tumor necrosis, which resulted in a significantly large hypoxic area (p = 0.02) and decreased HIF-1α, Glut-1, VEGF, and Ki-67 expression. Long-term administration of CKD-516 + IR reduced tumor volume and delayed tumor growth. This combination also greatly reduced the number of blood vessels (p = 0.0006) and significantly enhanced tumor necrosis (p = 0.004). CKD-516 + IR significantly increased HIF-1α expression (p = 0.0047), but significantly reduced VEGF expression (p = 0.0046). CONCLUSIONS Taken together, our data show that when used in combination, CKD-516 and IR can significantly enhance anti-tumor efficacy compared to monotherapy in lung cancer xenograft mice.
Collapse
Affiliation(s)
- Min-Young Kim
- Laboratory of Medical Oncology, Cancer Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung-Young Shin
- Laboratory of Medical Oncology, Cancer Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jeong-Oh Kim
- Laboratory of Medical Oncology, Cancer Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyoung-Hwa Son
- Laboratory of Medical Oncology, Cancer Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yeon Sil Kim
- Department of Radiation Oncology, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chan Kwon Jung
- Department of Pathology, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jin-Hyoung Kang
- Laboratory of Medical Oncology, Cancer Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Medical Oncology, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591 Republic of Korea
| |
Collapse
|
4
|
Smolarczyk R, Czapla J, Jarosz-Biej M, Czerwinski K, Cichoń T. Vascular disrupting agents in cancer therapy. Eur J Pharmacol 2020; 891:173692. [PMID: 33130277 DOI: 10.1016/j.ejphar.2020.173692] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 10/15/2020] [Accepted: 10/26/2020] [Indexed: 12/14/2022]
Abstract
Tumor blood vessel formation is a key process for tumor expansion. Tumor vessels are abnormal and differ from normal vessels in architecture and components. Besides oxygen and nutrients supply, the tumor vessels system, due to its abnormality, is responsible for: hypoxia formation, and metastatic routes. Tumor blood vessels can be a target of anti-cancer therapies. There are two types of therapies that target tumor vessels. The first one is the inhibition of the angiogenesis process. However, the inhibition is often ineffective because of alternative angiogenesis mechanism activation. The second type is a specific targeting of existing tumor blood vessels by vascular disruptive agents (VDAs). There are three groups of VDAs: microtubule destabilizing drugs, flavonoids with anti-vascular functions, and tumor vascular targeted drugs based on endothelial cell receptors. However, VDAs possess some limitations. They may be cardiotoxic and their application in therapy may leave viable residual, so called, rim cells on the edge of the tumor. However, it seems that a well-designed combination of VDAs with other anti-cancer drugs may bring a significant therapeutic effect. In this article, we describe three groups of vascular disruptive agents with their advantages and disadvantages. We mention VDAs clinical trials. Finally, we present the current possibilities of VDAs combination with other anti-cancer drugs.
Collapse
Affiliation(s)
- Ryszard Smolarczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland.
| | - Justyna Czapla
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland.
| | - Magdalena Jarosz-Biej
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland.
| | - Kyle Czerwinski
- University of Manitoba, Faculty of Science. 66 Chancellors Cir, Winnipeg, Canada.
| | - Tomasz Cichoń
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland.
| |
Collapse
|
5
|
Combination of tumour-infarction therapy and chemotherapy via the co-delivery of doxorubicin and thrombin encapsulated in tumour-targeted nanoparticles. Nat Biomed Eng 2020; 4:732-742. [PMID: 32572197 DOI: 10.1038/s41551-020-0573-2] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 05/18/2020] [Indexed: 12/22/2022]
Abstract
Drugs that induce thrombosis in the tumour vasculature have not resulted in long-term tumour eradication owing to tumour regrowth from tissue in the surviving rim of the tumour, where tumour cells can derive nutrients from adjacent non-tumoral blood vessels and tissues. Here, we report the performance of a combination of tumour-infarction therapy and chemotherapy, delivered via chitosan-based nanoparticles decorated with a tumour-homing peptide targeting fibrin-fibronectin complexes overexpressed on tumour-vessel walls and in tumour stroma, and encapsulating the coagulation-inducing protease thrombin and the chemotherapeutic doxorubicin. Systemic administration of the nanoparticles into mice and rabbits bearing subcutaneous or orthotopic tumours resulted in higher tumour growth suppression and decreased tumour recurrence than nanoparticles delivering only thrombin or doxorubicin, with histological and haematological analyses indicating an absence of detectable toxicity. The co-administration of a cytotoxic payload and a protease to elicit vascular infarction in tumours with biodegradable tumour-targeted nanoparticles represents a promising strategy for improving the therapeutic index of coagulation-based tumour therapy.
Collapse
|
6
|
Zheng K, Kros JM, Li J, Zheng PP. DNA-nanorobot-guided thrombin-inducing tumor infarction: raising new potential clinical concerns. Drug Discov Today 2020; 25:951-955. [PMID: 32205200 DOI: 10.1016/j.drudis.2020.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 02/29/2020] [Accepted: 03/06/2020] [Indexed: 11/19/2022]
Abstract
DNA-nanorobot-guided thrombin-inducing tumor infarction (DNA NanorobotTh-ITI) is emerging as a powerful therapeutic strategy for treatment of solid cancers. The technology represents a major advance in the application of DNA nanotechnology for anticancer therapy. More importantly, the technology is being translated from preclinical studies to the clinic owing to its promising anticancer effects with fewer toxicities demonstrated in preclinical settings. However, despite these beneficial effects of the technology, it is important to point out that some important potential clinical concerns remain to be addressed. Here, we raise these clinical concerns along with these beneficial effects of the technology. Hopefully, these newly raised potential clinical concerns could drive forward research in this field to expedite its clinical translation.
Collapse
Affiliation(s)
- Kang Zheng
- Department of Orthopedics, Ningbo Medical Center Li Hui Li Hospital, Ningbo, Zhejiang, China
| | - Johan M Kros
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jin Li
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ping-Pin Zheng
- Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
7
|
Predicting Clinical Efficacy of Vascular Disrupting Agents in Rodent Models of Primary and Secondary Liver Cancers: An Overview with Imaging-Histopathology Correlation. Diagnostics (Basel) 2020; 10:diagnostics10020078. [PMID: 32024029 PMCID: PMC7168934 DOI: 10.3390/diagnostics10020078] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 01/24/2020] [Accepted: 01/28/2020] [Indexed: 12/16/2022] Open
Abstract
Vascular disrupting agents (VDAs) have entered clinical trials for over 15 years. As the leading VDA, combretastatin A4 phosphate (CA4P) has been evaluated in combination with chemotherapy and molecular targeting agents among patients with ovarian cancer, lung cancer and thyroid cancer, but still remains rarely explored in human liver cancers. To overcome tumor residues and regrowth after CA4P monotherapy, a novel dual targeting pan-anticancer theragnostic strategy, i.e., OncoCiDia, has been developed and shown promise previously in secondary liver tumor models. Animal model of primary liver cancer is time consuming to induce, but of value for more closely mimicking human liver cancers in terms of tumor angiogenesis, histopathological heterogeneity, cellular differentiation, tumor components, cancer progression and therapeutic response. Being increasingly adopted in VDA researches, multiparametric magnetic resonance imaging (MRI) provides imaging biomarkers to reflect in vivo tumor responses to drugs. In this article as a chapter of a doctoral thesis, we overview the construction and clinical relevance of primary and secondary liver cancer models in rodents. Target selection for CA4P therapy assisted by enhanced MRI using hepatobiliary contrast agents (CAs), and therapeutic efficacy evaluated by using MRI with a non-specific contrast agent, dynamic contrast enhanced (DCE) imaging, diffusion weighted imaging (DWI) are also described. We then summarize diverse responses among primary hepatocellular carcinomas (HCCs), secondary liver and pancreatic tumors to CA4P, which appeared to be related to tumor size, vascularity, and cellular differentiation. In general, imaging-histopathology correlation studies allow to conclude that CA4P tends to be more effective in secondary liver tumors and in more differentiated HCCs, but less effective in less differentiated HCCs and implanted pancreatic tumor. Notably, cirrhotic liver may be responsive to CA4P as well. All these could be instructive for future clinical trials of VDAs.
Collapse
|
8
|
Paris JL, Villaverde G, Gómez-Graña S, Vallet-Regí M. Nanoparticles for multimodal antivascular therapeutics: Dual drug release, photothermal and photodynamic therapy. Acta Biomater 2020; 101:459-468. [PMID: 31706040 PMCID: PMC7616912 DOI: 10.1016/j.actbio.2019.11.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/24/2019] [Accepted: 11/01/2019] [Indexed: 12/13/2022]
Abstract
The poor delivery of nanoparticles to target cancer cells hinders their success in the clinical setting. In this work, an alternative target readily available for circulating nanoparticles has been selected to eliminate the need for nanoparticle penetration in the tissue: the tumor blood vessels. A tumor endothelium-targeted nanoparticle (employing an RGD-containing peptide) capable of co-delivering two anti-vascular drugs (one anti-angiogenic drug and one vascular disruption agent) is here presented. Furthermore, the nanodevice presents two additional anti-vascular capabilities upon activation by Near-Infrared light: provoking local hyperthermia (by gold nanorods in the system) and generating toxic reactive oxygen species (by the presence of a photosensitizer). RGD-targeting is shown to increase uptake by HUVEC cells, and while the nanoparticles are shown not to be toxic for these cells, upon Near-Infrared irradiation their almost complete killing is achieved. The combination of all four therapeutic modalities is then evaluated in an ex ovo fibrosarcoma xenograft model, which shows a significant reduction in the number of blood vessels irrigating the xenografts when the nanoparticles are present, as well as the destruction of the existing blood vessels upon irradiation. These results suggest that the combination of different anti-vascular therapeutic strategies in a single nanocarrier appears promising and should be further explored in the future. STATEMENT OF SIGNIFICANCE MVR2019: The combination of antivascular drugs with different mechanisms of action (such as antiangiogenic drugs and vascular disruption agents) has been recently proposed as a promising approach to maximize the therapeutic potential of anti-vascular therapeutics. Given the capacity of nanoparticles to co-deliver different drugs in optimizable ratios, nanomedicine appears to have a huge potential for the development of this kind of multimodal antivascular. To showcase this, an multimodal anti-vascular nanodevice for cancer therapy is here presented. This tumor endothelium-targeted nanosystem is capable of co-delivering two anti-vascular drugs (anti-angiogenic and vascular disruption agent) while also providing two additional therapeutic modalities that can be activated by Near-Infrared light: provoking local hyperthermia (photothermal therapy) and generating toxic reactive oxygen species (photodynamic therapy).
Collapse
Affiliation(s)
- Juan L Paris
- Dpto. Química en Ciencias Farmacéuticas (Unidad Docente de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| | - Gonzalo Villaverde
- Dpto. Química en Ciencias Farmacéuticas (Unidad Docente de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Sergio Gómez-Graña
- Dpto. Química en Ciencias Farmacéuticas (Unidad Docente de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - María Vallet-Regí
- Dpto. Química en Ciencias Farmacéuticas (Unidad Docente de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| |
Collapse
|
9
|
Schmitt F, Gold M, Rothemund M, Andronache I, Biersack B, Schobert R, Mueller T. New naphthopyran analogues of LY290181 as potential tumor vascular-disrupting agents. Eur J Med Chem 2019; 163:160-168. [PMID: 30503940 DOI: 10.1016/j.ejmech.2018.11.055] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 10/04/2018] [Accepted: 11/22/2018] [Indexed: 10/27/2022]
Abstract
A series of 19 analogues of the antiproliferative naphthopyran LY290181 were prepared for structure-activity relationship studies. We found the best activities for test compounds bearing small substituents at the meta position of the phenyl ring. The mode of action of LY290181 and eight new analogues was studied in detail. The compounds were highly anti-proliferative with IC50 values in the sub-nanomolar to triple-digit nanomolar range. The new analogues led to G2/M arrest due to interruption of the microtubule dynamics. In 518A2 melanoma cells they caused a mitotic catastrophe which eventually led to apoptosis. The naphthopyrans also induced a disruption of the vasculature in the chorioallantoic membrane (CAM) of fertilized chicken eggs as well as in xenograft tumors in mice. In a preliminary therapy trial, the difluoro derivative 2b retarded the growth of resistant xenograft tumors in mice.
Collapse
Affiliation(s)
- Florian Schmitt
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440, Bayreuth, Germany
| | - Madeleine Gold
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440, Bayreuth, Germany
| | - Matthias Rothemund
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440, Bayreuth, Germany
| | - Ion Andronache
- University of Bucharest, Research Center for Integrated Analysis and Territorial Management, 4-12, Regina Elisabeta Avenue, Bucharest, 3rd District, 030018, Romania
| | - Bernhard Biersack
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440, Bayreuth, Germany
| | - Rainer Schobert
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440, Bayreuth, Germany.
| | - Thomas Mueller
- Department of Internal Medicine IV, Oncology/Hematology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Straße 40, 06120, Halle, Germany
| |
Collapse
|
10
|
Liu YW, De Keyzer F, Feng YB, Chen F, Song SL, Swinnen J, Bormans G, Oyen R, Huang G, Ni YC. Intra-individual comparison of therapeutic responses to vascular disrupting agent CA4P between rodent primary and secondary liver cancers. World J Gastroenterol 2018; 24:2710-2721. [PMID: 29991876 PMCID: PMC6034151 DOI: 10.3748/wjg.v24.i25.2710] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/01/2018] [Accepted: 04/09/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To compare therapeutic responses of a vascular-disrupting-agent, combretastatin-A4-phosphate (CA4P), among hepatocellular carcinomas (HCCs) and implanted rhabdomyosarcoma (R1) in the same rats by magnetic-resonance-imaging (MRI), microangiography and histopathology.
METHODS Thirty-six HCCs were created by diethylnitrosamine gavage in 14 rats that were also intrahepatically implanted with one R1 per rat as monitored by T2-/T1-weighted images (T2WI/T1WI) on a 3.0T clinical MRI-scanner. Vascular response and tumoral necrosis were detected by dynamic contrast-enhanced (DCE-) and CE-MRI before, 1 h after and 12 h after CA4P iv at 10 mg/kg (treatment group n = 7) or phosphate-buffered saline at 1.0 mL/kg (control group n = 7). Tumor blood supply was calculated by a semiquantitative DCE parameter of area under the time signal intensity curve (AUC30). In vivo MRI findings were verified by postmortem techniques.
RESULTS On CE-T1WIs, unlike the negative response in all tumors of control animals, in treatment group CA4P caused rapid extensive vascular shutdown in all R1-tumors, but mildly or spottily in HCCs at 1 h. Consequently, tumor necrosis occurred massively in R1-tumors but patchily in HCCs at 12 h. AUC30 revealed vascular closure (66%) in R1-tumors at 1 h (P < 0.05), followed by further perfusion decrease at 12 h (P < 0.01), while less significant vascular clogging occurred in HCCs. Histomorphologically, CA4P induced more extensive necrosis in R1-tumors (92.6%) than in HCCs (50.2%) (P < 0.01); tumor vascularity heterogeneously scored +~+++ in HCCs but homogeneously scored ++ in R1-tumors.
CONCLUSION This study suggests superior performance of CA4P in metastatic over primary liver cancers, which could guide future clinical applications of vascular-disrupting-agents.
Collapse
MESH Headings
- Angiography
- Animals
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/therapeutic use
- Carcinoma, Hepatocellular/blood supply
- Carcinoma, Hepatocellular/chemically induced
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/pathology
- Contrast Media/administration & dosage
- Diethylnitrosamine/toxicity
- Humans
- Liver/diagnostic imaging
- Liver/pathology
- Liver Neoplasms/blood supply
- Liver Neoplasms/chemically induced
- Liver Neoplasms/drug therapy
- Liver Neoplasms/pathology
- Liver Neoplasms, Experimental/chemically induced
- Liver Neoplasms, Experimental/diagnostic imaging
- Liver Neoplasms, Experimental/drug therapy
- Liver Neoplasms, Experimental/pathology
- Magnetic Resonance Imaging/methods
- Male
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/pathology
- Rats
- Rhabdomyosarcoma/blood supply
- Rhabdomyosarcoma/drug therapy
- Rhabdomyosarcoma/pathology
- Rhabdomyosarcoma/secondary
- Stilbenes/pharmacology
- Stilbenes/therapeutic use
- Treatment Outcome
- Tumor Microenvironment/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Ye-Wei Liu
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
- Institute of Clinical Nuclear Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200025, China
| | | | - Yuan-Bo Feng
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Feng Chen
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Shao-Li Song
- Institute of Clinical Nuclear Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Johan Swinnen
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Guy Bormans
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Raymond Oyen
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Gang Huang
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
- Institute of Clinical Nuclear Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200025, China
| | - Yi-Cheng Ni
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| |
Collapse
|
11
|
El Bairi K, Amrani M, Afqir S. Starvation tactics using natural compounds for advanced cancers: pharmacodynamics, clinical efficacy, and predictive biomarkers. Cancer Med 2018; 7:2221-2246. [PMID: 29732738 PMCID: PMC6010871 DOI: 10.1002/cam4.1467] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/21/2018] [Accepted: 02/28/2018] [Indexed: 02/05/2023] Open
Abstract
The high mortality associated with oncological diseases is mostly due to tumors in advanced stages, and their management is a major challenge in modern oncology. Angiogenesis is a defined hallmark of cancer and predisposes to metastatic invasion and dissemination and is therefore an important druggable target for cancer drug discovery. Recently, because of drug resistance and poor prognosis, new anticancer drugs from natural sources targeting tumor vessels have attracted more attention and have been used in several randomized and controlled clinical trials as therapeutic options. Here, we outline and discuss potential natural compounds as salvage treatment for advanced cancers from recent and ongoing clinical trials and real-world studies. We also discuss predictive biomarkers for patients' selection to optimize the use of these potential anticancer drugs.
Collapse
Affiliation(s)
- Khalid El Bairi
- Faculty of Medicine and PharmacyMohamed Ist UniversityOujdaMorocco
| | - Mariam Amrani
- Equipe de Recherche en Virologie et Onco‐biologieFaculty of MedicinePathology DepartmentNational Institute of OncologyUniversité Mohamed VRabatMorocco
| | - Said Afqir
- Department of Medical OncologyMohamed VI University HospitalOujdaMorocco
| |
Collapse
|
12
|
The vascular disrupting agent combretastatin A-4 phosphate causes prolonged elevation of proteins involved in heme flux and function in resistant tumor cells. Oncotarget 2017; 9:4090-4101. [PMID: 29423106 PMCID: PMC5790523 DOI: 10.18632/oncotarget.23734] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/15/2017] [Indexed: 01/24/2023] Open
Abstract
Vascular disrupting agents (VDAs) represent a promising class of anti-cancer drugs for solid tumor treatment. Here, we aim to better understand the mechanisms underlying tumor reccurrence and treatment resistance following the administration of a VDA, combretastatin A-4 phosphate (CA4P). Firstly, we used photoacoustic tomography to noninvasively map the effect of CA4P on blood oxygen levels throughout subcutaneous non-small cell lung cancer (NSCLC) tumors in mice. We found that the oxygenation of peripheral tumor vessels was significantly decreased at 1 and 3 hours post-CA4P treatment. The oxygenation of the tumor core reduced significantly at 1 and 3 hours, and reached anoxia after 24 hours. Secondly, we examined the effect of CA4P on the levels of proteins involved in heme flux and function, which are elevated in lung tumors. Using immunohistochemistry, we found that CA4P substantially enhanced the levels of enzymes involved in heme biosynthesis, uptake, and degradation, as well as oxygen-utilizing hemoproteins. Furthermore, measurements of markers of mitochondrial function suggest that CA4P did not diminish mitochondrial function in resistant tumor cells. These results suggest that elevated levels of heme flux and function contribute to tumor regrowth and treatment resistance post-VDA administration.
Collapse
|
13
|
Siemann DW, Chaplin DJ, Horsman MR. Realizing the Potential of Vascular Targeted Therapy: The Rationale for Combining Vascular Disrupting Agents and Anti-Angiogenic Agents to Treat Cancer. Cancer Invest 2017; 35:519-534. [DOI: 10.1080/07357907.2017.1364745] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- D. W. Siemann
- Department of Radiation Oncology, University of Florida, Gainesville, FL, USA
| | | | - M. R. Horsman
- Department of Experimental Clinical Oncology, Aarhus University, Denmark
| |
Collapse
|
14
|
Seidi K, Jahanban-Esfahlan R, Zarghami N. Tumor rim cells: From resistance to vascular targeting agents to complete tumor ablation. Tumour Biol 2017; 39:1010428317691001. [DOI: 10.1177/1010428317691001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Current vascular targeting strategies pursue two main goals: anti-angiogenesis agents aim to halt sprouting and the formation of new blood vessels, while vascular disrupting agents along with coaguligands seek to compromise blood circulation in the vessels. The ultimate goal of such therapies is to deprive tumor cells out of oxygen and nutrients long enough to succumb cancer cells to death. Most of vascular targeting agents presented promising therapeutic potential, but the final goal which is cure is rarely achieved. Nevertheless, in both preclinical and clinical settings, tumors tend to grow back, featuring a highly invasive, metastatic, and extremely resistant form. This review highlights the critical significance of tumor rim cells as the main factor, determining therapy success with vascular targeting agents. We present an overview of different single and combination treatments with vascular targeting agents that enable efficient targeting of tumor rim cells and long-lasting tumor cure. Understanding the nature of tumor rim cells, how they establish, how they manage to survive of vascular targeting agents, and how they contribute in tumor refractoriness, may open new avenues to the development of beneficial strategies, capable to eliminate residual rim cells, and enable tumor ablation once and forever.
Collapse
Affiliation(s)
- Khaled Seidi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
15
|
Liu Z, Wu H, Wei Z, Wang X, Shen P, Wang S, Wang A, Chen W, Lu Y. TRPM8: a potential target for cancer treatment. J Cancer Res Clin Oncol 2016; 142:1871-81. [PMID: 26803314 DOI: 10.1007/s00432-015-2112-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 12/28/2015] [Indexed: 01/09/2023]
Abstract
Transient receptor potential (TRP) cation channel superfamily plays critical roles in variety of processes, including temperature perception, pain transduction, vasorelaxation, male fertility, and tumorigenesis. One of seven families within the TRP superfamily of ion channels, the melastatin, or TRPM family comprises a group of eight structurally and functionally diverse channels. Of all the members of TRPM subfamily, TRPM8 is the most notable one. A lot of literatures have demonstrated that transient receptor potential melastatin 8 (TRPM8) could perform a myriad of functions in vertebrates and invertebrates alike. In addition to its well-known function in cold sensation, TRPM8 has an emerging role in a variety of biological systems, including thermoregulation, cancer, bladder function, and asthma. Recent studies have shown that TRPM8 is necessary to the initiation and progression of tumors, and the aberrant expression of TRPM8 was found in varieties of tumors, such as prostate tumor, melanoma, breast adenocarcinoma, bladder cancer, and colorectal cancer, making it a novel molecular target potentially useful in the diagnosis and treatment of cancer. This review outlines our current understanding on the role of TRPM8 in occurrence and development of different kinds of tumor and also includes discussion about the regulation of TRPM8 during carcinogenesis as well as therapeutic potential of targeting TRPM8 in tumor, which may be utilized for a potential pharmacological use as a target for anti-cancer therapy.
Collapse
Affiliation(s)
- Zhaoguo Liu
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Hongyan Wu
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, People's Republic of China
- Department of Pharmacy, Yancheng Health Vocational and Technical College, Yancheng, 224005, Jiangsu Province, China
| | - Zhonghong Wei
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Xu Wang
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Peiliang Shen
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Siliang Wang
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Aiyun Wang
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Wenxing Chen
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Yin Lu
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, People's Republic of China.
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| |
Collapse
|
16
|
Wang Z, Qi H, Shen Q, Lu G, Li M, Bao K, Wu Y, Zhang W. 4,5-Diaryl-3H-1,2-dithiole-3-thiones and related compounds as combretastatin A-4/oltipraz hybrids: Synthesis, molecular modelling and evaluation as antiproliferative agents and inhibitors of tubulin. Eur J Med Chem 2016; 122:520-529. [PMID: 27428395 DOI: 10.1016/j.ejmech.2016.06.055] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 06/29/2016] [Accepted: 06/29/2016] [Indexed: 11/30/2022]
Abstract
A new series of 4,5-diaryl-3H-1,2-dithiole-3-thiones and related compounds were designed and synthesised as combretastatin A-4/oltipraz hybrids. We evaluated the antiproliferative activities, inhibition of tubulin polymerization, and cell-cycle effects of these compounds. Several compounds in this series, such as 4d and 5c, displayed significant activity against SGC-7901, KB and HT-1080 cell lines, as determined using MTT assays. The most active compound, 4d, markedly inhibited tubulin polymerization, with an IC50 value of 4.44 μM being observed. In mechanistic studies, 4d caused cell arrest in G2/M phase, induced apoptotic cell death, and disrupted microtubule formation. Molecular docking studies revealed that 4d interacts and binds efficiently with the tubulin protein.
Collapse
Affiliation(s)
- Zhiwei Wang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China; Department of Pharmaceutical Chemistry, Jinzhou Medical University, No.40, Section 3, Songpo Road, Linghe District, Jinzhou, 121001, China
| | - Huan Qi
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Qirong Shen
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Guodong Lu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Mingyang Li
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Kai Bao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China; Gordon Center for Medical Imaging, Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02214, USA
| | - Yingliang Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| | - Weige Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| |
Collapse
|
17
|
Pérez-Pérez MJ, Priego EM, Bueno O, Martins MS, Canela MD, Liekens S. Blocking Blood Flow to Solid Tumors by Destabilizing Tubulin: An Approach to Targeting Tumor Growth. J Med Chem 2016; 59:8685-8711. [DOI: 10.1021/acs.jmedchem.6b00463] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
| | - Eva-María Priego
- Instituto de Química Médica (IQM-CSIC), Juan de la Cierva 3, E-28006 Madrid, Spain
| | - Oskía Bueno
- Instituto de Química Médica (IQM-CSIC), Juan de la Cierva 3, E-28006 Madrid, Spain
| | | | - María-Dolores Canela
- Instituto de Química Médica (IQM-CSIC), Juan de la Cierva 3, E-28006 Madrid, Spain
| | - Sandra Liekens
- Rega
Institute for Medical Research, KU Leuven, B-3000 Leuven, Belgium
| |
Collapse
|
18
|
Monk BJ, Sill MW, Walker JL, Darus CJ, Sutton G, Tewari KS, Martin LP, Schilder JM, Coleman RL, Balkissoon J, Aghajanian C. Randomized Phase II Evaluation of Bevacizumab Versus Bevacizumab Plus Fosbretabulin in Recurrent Ovarian, Tubal, or Peritoneal Carcinoma: An NRG Oncology/Gynecologic Oncology Group Study. J Clin Oncol 2016; 34:2279-86. [PMID: 27217446 DOI: 10.1200/jco.2015.65.8153] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The vascular disrupting agent fosbretabulin tromethamine selectively targets pre-existing tumor vasculature, which causes vascular shutdown and leads to cancer cell death and necrosis. Antiangiogenesis agents such as bevacizumab, a humanized antivascular endothelial growth factor monoclonal antibody, might prevent revascularization during and after treatment with a vascular disrupting agent. PATIENTS AND METHODS Patients with recurrent or persistent epithelial ovarian, tubal, or peritoneal carcinoma, measurable or detectable disease, and three or fewer prior regimens were randomly assigned to bevacizumab (15 mg/kg intravenously once every 3 weeks) or the combination of bevacizumab (15 mg/kg) plus fosbretabulin (60 mg/m(2)) intravenously once every 3 weeks until disease progression or toxicity. Randomization was stratified by disease status (measurable v nonmeasurable), prior bevacizumab, and platinum-free interval. The primary end point was progression-free survival (PFS). The study was designed with 80% power for a one-sided alternative at a 10% level of significance to detect a reduction in the hazard by 37.5%. RESULTS The study enrolled 107 patients. Median PFS was 4.8 months for bevacizumab and 7.3 months for bevacizumab plus fosbretabulin (hazard ratio, 0.69; 90% two-sided CI, 0.47 to 1.00; one-sided P = .05). The proportion responding (overall response rate) to bevacizumab was 28.2% among 39 patients with measurable disease and 35.7% among 42 patients treated with the combination. The relative probability of responding was 1.27 (90% CI, 0.74 to 2.17; one-sided P = .24). Adverse events greater than grade 3 were more common in the combination regimen than in bevacizumab only for hypertension (35% v 20%). There was one grade 3 thromboembolic event in the combination arm and one intestinal fistula in the bevacizumab only arm. CONCLUSION On the basis of the PFS, overall response rate, and tolerability of these two antivascular therapies, further evaluation is warranted for this chemotherapy-free regimen. Fosbretabulin in combination with bevacizumab increases the risk of hypertension.
Collapse
Affiliation(s)
- Bradley J Monk
- Bradley J. Monk, University of Arizona Cancer Center at Dignity Health St Joseph's Hospital and Medical Center, Phoenix, AZ; Michael W. Sill, Roswell Park Cancer Institute, Buffalo; Carol Aghajanian, Memorial Sloan Kettering Cancer Center, New York, NY; Joan L. Walker, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Christopher J. Darus, Maine Medical Center, Portland, ME; Gregory Sutton, Washington University School of Medicine, St. Louis, MO; Krishnansu S. Tewari, University of California Irvine Medical Center, Orange; Jai Balkissoon, Pharmaceutical Product Development International, South San Francisco, CA; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; Jeanne M. Schilder, Indiana University Medical Center, Indianapolis, IN; and Robert L. Coleman, The University of Texas MD Anderson Cancer Center, Houston, TX.
| | - Michael W Sill
- Bradley J. Monk, University of Arizona Cancer Center at Dignity Health St Joseph's Hospital and Medical Center, Phoenix, AZ; Michael W. Sill, Roswell Park Cancer Institute, Buffalo; Carol Aghajanian, Memorial Sloan Kettering Cancer Center, New York, NY; Joan L. Walker, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Christopher J. Darus, Maine Medical Center, Portland, ME; Gregory Sutton, Washington University School of Medicine, St. Louis, MO; Krishnansu S. Tewari, University of California Irvine Medical Center, Orange; Jai Balkissoon, Pharmaceutical Product Development International, South San Francisco, CA; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; Jeanne M. Schilder, Indiana University Medical Center, Indianapolis, IN; and Robert L. Coleman, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Joan L Walker
- Bradley J. Monk, University of Arizona Cancer Center at Dignity Health St Joseph's Hospital and Medical Center, Phoenix, AZ; Michael W. Sill, Roswell Park Cancer Institute, Buffalo; Carol Aghajanian, Memorial Sloan Kettering Cancer Center, New York, NY; Joan L. Walker, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Christopher J. Darus, Maine Medical Center, Portland, ME; Gregory Sutton, Washington University School of Medicine, St. Louis, MO; Krishnansu S. Tewari, University of California Irvine Medical Center, Orange; Jai Balkissoon, Pharmaceutical Product Development International, South San Francisco, CA; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; Jeanne M. Schilder, Indiana University Medical Center, Indianapolis, IN; and Robert L. Coleman, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Christopher J Darus
- Bradley J. Monk, University of Arizona Cancer Center at Dignity Health St Joseph's Hospital and Medical Center, Phoenix, AZ; Michael W. Sill, Roswell Park Cancer Institute, Buffalo; Carol Aghajanian, Memorial Sloan Kettering Cancer Center, New York, NY; Joan L. Walker, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Christopher J. Darus, Maine Medical Center, Portland, ME; Gregory Sutton, Washington University School of Medicine, St. Louis, MO; Krishnansu S. Tewari, University of California Irvine Medical Center, Orange; Jai Balkissoon, Pharmaceutical Product Development International, South San Francisco, CA; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; Jeanne M. Schilder, Indiana University Medical Center, Indianapolis, IN; and Robert L. Coleman, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gregory Sutton
- Bradley J. Monk, University of Arizona Cancer Center at Dignity Health St Joseph's Hospital and Medical Center, Phoenix, AZ; Michael W. Sill, Roswell Park Cancer Institute, Buffalo; Carol Aghajanian, Memorial Sloan Kettering Cancer Center, New York, NY; Joan L. Walker, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Christopher J. Darus, Maine Medical Center, Portland, ME; Gregory Sutton, Washington University School of Medicine, St. Louis, MO; Krishnansu S. Tewari, University of California Irvine Medical Center, Orange; Jai Balkissoon, Pharmaceutical Product Development International, South San Francisco, CA; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; Jeanne M. Schilder, Indiana University Medical Center, Indianapolis, IN; and Robert L. Coleman, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Krishnansu S Tewari
- Bradley J. Monk, University of Arizona Cancer Center at Dignity Health St Joseph's Hospital and Medical Center, Phoenix, AZ; Michael W. Sill, Roswell Park Cancer Institute, Buffalo; Carol Aghajanian, Memorial Sloan Kettering Cancer Center, New York, NY; Joan L. Walker, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Christopher J. Darus, Maine Medical Center, Portland, ME; Gregory Sutton, Washington University School of Medicine, St. Louis, MO; Krishnansu S. Tewari, University of California Irvine Medical Center, Orange; Jai Balkissoon, Pharmaceutical Product Development International, South San Francisco, CA; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; Jeanne M. Schilder, Indiana University Medical Center, Indianapolis, IN; and Robert L. Coleman, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lainie P Martin
- Bradley J. Monk, University of Arizona Cancer Center at Dignity Health St Joseph's Hospital and Medical Center, Phoenix, AZ; Michael W. Sill, Roswell Park Cancer Institute, Buffalo; Carol Aghajanian, Memorial Sloan Kettering Cancer Center, New York, NY; Joan L. Walker, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Christopher J. Darus, Maine Medical Center, Portland, ME; Gregory Sutton, Washington University School of Medicine, St. Louis, MO; Krishnansu S. Tewari, University of California Irvine Medical Center, Orange; Jai Balkissoon, Pharmaceutical Product Development International, South San Francisco, CA; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; Jeanne M. Schilder, Indiana University Medical Center, Indianapolis, IN; and Robert L. Coleman, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jeanne M Schilder
- Bradley J. Monk, University of Arizona Cancer Center at Dignity Health St Joseph's Hospital and Medical Center, Phoenix, AZ; Michael W. Sill, Roswell Park Cancer Institute, Buffalo; Carol Aghajanian, Memorial Sloan Kettering Cancer Center, New York, NY; Joan L. Walker, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Christopher J. Darus, Maine Medical Center, Portland, ME; Gregory Sutton, Washington University School of Medicine, St. Louis, MO; Krishnansu S. Tewari, University of California Irvine Medical Center, Orange; Jai Balkissoon, Pharmaceutical Product Development International, South San Francisco, CA; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; Jeanne M. Schilder, Indiana University Medical Center, Indianapolis, IN; and Robert L. Coleman, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Robert L Coleman
- Bradley J. Monk, University of Arizona Cancer Center at Dignity Health St Joseph's Hospital and Medical Center, Phoenix, AZ; Michael W. Sill, Roswell Park Cancer Institute, Buffalo; Carol Aghajanian, Memorial Sloan Kettering Cancer Center, New York, NY; Joan L. Walker, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Christopher J. Darus, Maine Medical Center, Portland, ME; Gregory Sutton, Washington University School of Medicine, St. Louis, MO; Krishnansu S. Tewari, University of California Irvine Medical Center, Orange; Jai Balkissoon, Pharmaceutical Product Development International, South San Francisco, CA; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; Jeanne M. Schilder, Indiana University Medical Center, Indianapolis, IN; and Robert L. Coleman, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jai Balkissoon
- Bradley J. Monk, University of Arizona Cancer Center at Dignity Health St Joseph's Hospital and Medical Center, Phoenix, AZ; Michael W. Sill, Roswell Park Cancer Institute, Buffalo; Carol Aghajanian, Memorial Sloan Kettering Cancer Center, New York, NY; Joan L. Walker, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Christopher J. Darus, Maine Medical Center, Portland, ME; Gregory Sutton, Washington University School of Medicine, St. Louis, MO; Krishnansu S. Tewari, University of California Irvine Medical Center, Orange; Jai Balkissoon, Pharmaceutical Product Development International, South San Francisco, CA; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; Jeanne M. Schilder, Indiana University Medical Center, Indianapolis, IN; and Robert L. Coleman, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Carol Aghajanian
- Bradley J. Monk, University of Arizona Cancer Center at Dignity Health St Joseph's Hospital and Medical Center, Phoenix, AZ; Michael W. Sill, Roswell Park Cancer Institute, Buffalo; Carol Aghajanian, Memorial Sloan Kettering Cancer Center, New York, NY; Joan L. Walker, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Christopher J. Darus, Maine Medical Center, Portland, ME; Gregory Sutton, Washington University School of Medicine, St. Louis, MO; Krishnansu S. Tewari, University of California Irvine Medical Center, Orange; Jai Balkissoon, Pharmaceutical Product Development International, South San Francisco, CA; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; Jeanne M. Schilder, Indiana University Medical Center, Indianapolis, IN; and Robert L. Coleman, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
19
|
Liang W, Ni Y, Chen F. Tumor resistance to vascular disrupting agents: mechanisms, imaging, and solutions. Oncotarget 2016; 7:15444-59. [PMID: 26812886 PMCID: PMC4941252 DOI: 10.18632/oncotarget.6999] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 01/14/2016] [Indexed: 01/04/2023] Open
Abstract
The emergence of vascular disrupting agents (VDAs) is a significant advance in the treatment of solid tumors. VDAs induce rapid and selective shutdown of tumor blood flow resulting in massive necrosis. However, a viable marginal tumor rim always remains after VDA treatment and is a major cause of recurrence. In this review, we discuss the mechanisms involved in the resistance of solid tumors to VDAs. Hypoxia, tumor-associated macrophages, and bone marrow-derived circulating endothelial progenitor cells all may contribute to resistance. Resistance can be monitored using magnetic resonance imaging markers. The various solutions proposed to manage tumor resistance to VDAs emphasize combining these agents with other approaches including antiangiogenic agents, chemotherapy, radiotherapy, radioimmunotherapy, and sequential dual-targeting internal radiotherapy.
Collapse
Affiliation(s)
- Wenjie Liang
- Department of Radiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yicheng Ni
- Radiology Section, University Hospitals, University of Leuven, Leuven, Belgium
| | - Feng Chen
- Department of Radiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
20
|
Antiangiogenesis and vascular disrupting agents in cancer: circumventing resistance and augmenting their therapeutic utility. Future Med Chem 2016; 8:443-62. [DOI: 10.4155/fmc.16.6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Angiogenesis is a process essential for tumor growth and metastasis. Inhibition of angiogenesis as an anticancer strategy has shown only moderately improved results and is beset with practical limitations, despite theoretical therapeutic advantages. Inevitably resistance develops, through redundancy of signaling pathways and selection for subclonal populations adapted for hypoxic conditions, with more invasive phenotypes. Antiangiogenic-targeted therapies may find improved efficacy in combination therapies; with others in this class, that directly or indirectly target separate pathways or different components of the same pathway, or with a separate class of tumor vasculature-disrupting agents. This review discusses the challenges and strategies for optimization of combination therapies including metronomic administration of drugs and the need for suitable prognostic and surrogate response biomarkers.
Collapse
|
21
|
Tsai RY, Cheng YC, Wong CS. (+)-Naloxone inhibits morphine-induced chemotaxis via prevention of heat shock protein 90 cleavage in microglia. J Formos Med Assoc 2015; 114:446-55. [PMID: 25649471 DOI: 10.1016/j.jfma.2014.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 10/30/2014] [Accepted: 12/26/2014] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND/PURPOSE Microglia have a crucial role in maintaining neuronal homeostasis in the central nervous system. Immune factors released from microglia have important roles in nociceptive signal transduction. Activation of microglia seems to be a shared mechanism in pathological pain and morphine tolerance because pharmacological attenuation of microglia activation provides satisfactory management in both situations. METHODS In the present study, we investigated the effect of 1nM (+)-naloxone, which is not an opioid receptor antagonist, on morphine-induced activation of microglia EOC13.31 cells. RESULTS Our results showed that 1μM morphine enhanced microglia activation and migration, decreased α-tubulin acetylation, and induced heat shock protein 90 (HSP90) fragmentation and histone deacetylase 6 (HDAC6) expression. Morphine-induced α-tubulin deacetylation and HSP90 fragmentation were HDAC6-dependent. Pretreatment with (+)-naloxone (1nM) inhibited morphine-evoked microglia activation and chemotaxis and prevented α-tubulin deacetylation and HSP90 fragmentation by inhibiting HDAC6 expression. CONCLUSION Based on the findings of the present study, we suggest that (+)-naloxone inhibits morphine-induced microglia activation by regulating HDAC6-dependent α-tubulin deacetylation and HSP90 fragmentation.
Collapse
Affiliation(s)
- Ru-Yin Tsai
- Department of Nursing, Da-Yeh University, Changhua, Taiwan; Department of Anesthesiology, Cathay General Hospital, Taipei, Taiwan
| | - Yu-Che Cheng
- Department of Medical Research, Cathay General Hospital, Taipei, Taiwan
| | - Chih-Shung Wong
- Department of Anesthesiology, Cathay General Hospital, Taipei, Taiwan; School of Medicine, Fu-Jen Catholic University, New Taipei, Taiwan; Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
22
|
Huang L, Hu C, DI Benedetto M, Varin R, Liu J, Jin J, Wang L, Vannier JP, Janin A, Lu H, Li H. Cross-drug resistance to sunitinib induced by doxorubicin in endothelial cells. Oncol Lett 2014; 9:1287-1292. [PMID: 25663899 PMCID: PMC4315062 DOI: 10.3892/ol.2014.2819] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 10/15/2014] [Indexed: 01/08/2023] Open
Abstract
Multiple drug resistance remains an unsolved problem in cancer therapy. A previous study has demonstrated that the chemotherapeutic drug doxorubicin (Dox) induced upregulation of P-glycoprotein in endothelial cells, resulting in a 20-fold increase in drug resistance and reduced efficiency of doxorubicin treatment in a mouse tumor model. In the present study, the cross-resistance and sensitivity of HMECd1 and HMECd2 established cell lines to anti-angiogenic drugs, particularly sunitinib, was explored. The results revealed that Dox treatment induced a significant increase in the breast cancer resistance protein (ABCG2) gene transcription and protein expression. This increase gave rise to a 4- to 5-fold increase in the half maximal inhibitory concentration of the HMECd1 and HMECd2 cells in response to sunitinib treatment in vitro. Functionally, the role of ABCG2 in the resistance to sunitinib was confirmed by the use of the ABCG2 inhibitors fumitremorgin C and diethylstilbestrol, which blocked cell resistance. The present study indicates that endothelial cells exhibit cross-resistance between cytotoxic drugs and anti-angiogenic drugs. This suggests that multiple drug resistance induced by chemotherapy in endothelial cells may affect the efficiency of anti-angiogenic drugs.
Collapse
Affiliation(s)
- Limin Huang
- Department of Oncology, People's Hospital of Guizhou Province, Guiyang, Guizhou 550000, P.R. China
| | - Chaoquan Hu
- Department of Surgery, Affiliated Hospital of Guiyang Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Mélanie DI Benedetto
- French Institute of Health and Medical Research, UMR-S 1165, University Institute of Hematology, Saint Louis Hospital, Paris 75010, France
| | - Rémi Varin
- Laboratory of MERCI (EA 3829), Faculty of Medicine and Pharmacy, University of Rouen, Rouen 76183, France
| | - Jielin Liu
- Department of Surgery, Affiliated Hospital of Guiyang Medical University, Guiyang, Guizhou 550004, P.R. China ; French Institute of Health and Medical Research, UMR-S 1165, University Institute of Hematology, Saint Louis Hospital, Paris 75010, France
| | - Jian Jin
- French Institute of Health and Medical Research, UMR-S 1165, University Institute of Hematology, Saint Louis Hospital, Paris 75010, France ; School of Medicine and Pharmaceutics, Jiangnan University, Wuxi, Jiangsu 214122, P.R. China
| | - Li Wang
- French Institute of Health and Medical Research, UMR-S 1165, University Institute of Hematology, Saint Louis Hospital, Paris 75010, France
| | - Jean-Pierre Vannier
- Laboratory of MERCI (EA 3829), Faculty of Medicine and Pharmacy, University of Rouen, Rouen 76183, France
| | - Anne Janin
- French Institute of Health and Medical Research, UMR-S 1165, University Institute of Hematology, Saint Louis Hospital, Paris 75010, France ; Laboratory of Pathology, Paris Diderot University, Sorbonne Paris Cité, UMR-S 1165, France ; Saint-Louis Hospital, Laboratory of Pathology, Paris 75010, France
| | - He Lu
- French Institute of Health and Medical Research, UMR-S 1165, University Institute of Hematology, Saint Louis Hospital, Paris 75010, France ; Laboratory of Pathology, Paris Diderot University, Sorbonne Paris Cité, UMR-S 1165, France
| | - Hong Li
- Laboratory of MERCI (EA 3829), Faculty of Medicine and Pharmacy, University of Rouen, Rouen 76183, France
| |
Collapse
|
23
|
Synthesis and biological evaluation of novel 5,7-diphenylimidazo[1,2-a]pyridine derivatives. Chem Res Chin Univ 2014. [DOI: 10.1007/s40242-014-4081-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
24
|
Colliez F, Neveu MA, Magat J, Cao Pham TT, Gallez B, Jordan BF. Qualification of a Noninvasive Magnetic Resonance Imaging Biomarker to Assess Tumor Oxygenation. Clin Cancer Res 2014; 20:5403-11. [DOI: 10.1158/1078-0432.ccr-13-3434] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|