1
|
Liang Y, Zhang S, Wang D, Ji P, Zhang B, Wu P, Wang L, Liu Z, Wang J, Duan Y, Yuan L. Dual-Functional Nanodroplet for Tumor Vasculature Ultrasound Imaging and Tumor Immunosuppressive Microenvironment Remodeling. Adv Healthc Mater 2024:e2401274. [PMID: 39031111 DOI: 10.1002/adhm.202401274] [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: 04/26/2024] [Revised: 06/24/2024] [Indexed: 07/22/2024]
Abstract
Accurately evaluating tumor neoangiogenesis and conducting precise interventions toward an immune-favorable microenvironment are of significant clinical importance. In this study, a novel nanodroplet termed as the nanodroplet-based ultrasound contrast agent and therapeutic (NDsUCA/Tx) is designed for ultrasound imaging and precise interventions of tumor neoangiogenesis. Briefly, the NDsUCA/Tx shell is constructed from an engineered CMs containing the tumor antigen, vascular endothelial growth factor receptor 1 (VEGFR1) extracellular domain 2-3, and CD93 ligand multimerin 2. The core is composed of perfluorohexane and the immune adjuvant R848. After injection, NDsUCA/Tx is found to be enriched in the tumor vasculature with high expression of CD93. When triggered by ultrasound, the perfluorohexane in NDsUCA/Tx underwent acoustic droplet vaporization and generated an enhanced ultrasound signal. Some microbubbles exploded and the resultant debris (with tumor antigen and R848) together with the adsorbed VEGF are taken up by nearby cells. This cleared the local VEGF for vascular normalization, and also served as a vaccine to activate the immune response. Using a syngeneic mouse model, the satisfactory performance of NDsUCA/Tx in tumor vasculature imaging and immune activation is confirmed. Thus, a multifunctional NDsUCA/Tx is successfully developed for molecular imaging of tumor neoangiogenesis and precise remodeling of the tumor microenvironment.
Collapse
Affiliation(s)
- Yuan Liang
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, P. R. China
| | - Siyan Zhang
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, P. R. China
| | - Dingyi Wang
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, P. R. China
| | - Panpan Ji
- Department of Digestive Surgery Xijing Hospital, Air Force Medical University, Xi'an, 710032, P. R. China
| | - Bin Zhang
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, P. R. China
| | - Pengying Wu
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, P. R. China
| | - Lantian Wang
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, P. R. China
| | - Zhaoyou Liu
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, P. R. China
| | - Jia Wang
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, P. R. China
| | - Yunyou Duan
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, P. R. China
| | - Lijun Yuan
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, P. R. China
| |
Collapse
|
2
|
Malekan M, Haass NK, Rokni GR, Gholizadeh N, Ebrahimzadeh MA, Kazeminejad A. VEGF/VEGFR axis and its signaling in melanoma: Current knowledge toward therapeutic targeting agents and future perspectives. Life Sci 2024; 345:122563. [PMID: 38508233 DOI: 10.1016/j.lfs.2024.122563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/10/2024] [Accepted: 03/13/2024] [Indexed: 03/22/2024]
Abstract
Melanoma is responsible for most skin cancer-associated deaths globally. The progression of melanoma is influenced by a number of pathogenic processes. Understanding the VEGF/VEGFR axis, which includes VEGF-A, PlGF, VEGF-B, VEGF-C, and VEGF-D and their receptors, VEGFR-1, VEGFR-2, and VEGFR-3, is of great importance in melanoma due to its crucial role in angiogenesis. This axis generates multifactorial and complex cellular signaling, engaging the MAPK/ERK, PI3K/AKT, PKC, PLC-γ, and FAK signaling pathways. Melanoma cell growth and proliferation, migration and metastasis, survival, and acquired resistance to therapy are influenced by this axis. The VEGF/VEGFR axis was extensively examined for their potential as diagnostic/prognostic biomarkers in melanoma patients and results showed that VEGF overexpression can be associated with unfavorable prognosis, higher level of tumor invasion and poor response to therapy. MicroRNAs linking to the VEGF/VEGFR axis were identified and, in this review, divided into two categories according to their functions, some of them promote melanoma angiogenesis (promotive group) and some restrict melanoma angiogenesis (protective group). In addition, the approach of treating melanoma by targeting the VEGF/VEGFR axis has garnered significant interest among researchers. These agents can be divided into two main groups: anti-VEGF and VEGFR inhibitors. These therapeutic options may be a prominent step along with the modern targeting and immune therapies for better coverage of pathological processes leading to melanoma progression and therapy resistance.
Collapse
Affiliation(s)
- Mohammad Malekan
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | | | - Ghasem Rahmatpour Rokni
- Department of Dermatology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nasim Gholizadeh
- Department of Dermatology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Ali Ebrahimzadeh
- Pharmaceutical Sciences Research Center, School of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Armaghan Kazeminejad
- Department of Dermatology, Antimicrobial Resistance Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences,Sari, Iran
| |
Collapse
|
3
|
Mullen SA, Das D, Ziamiavaghi N, High R, Datta K, Teply BA. Association of plasma NRP2 and VEGF-C levels with prostate cancer disease severity. Prostate 2024; 84:277-284. [PMID: 37942701 PMCID: PMC10842186 DOI: 10.1002/pros.24648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/24/2023] [Accepted: 10/31/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Neuropilin 2 (NRP2) expression in tissue is an independent prognostic factor for aggressive prostate cancer. Since the NRP2 pathway activation is thought to occur in part through secondary resistance, quantification of NRP2 in initial tissue biopsy specimens collected at diagnosis may have limited utility in identifying patients at highest risk for morbidity and mortality. Given that metastatic tissue is only occasionally obtained for analysis, there is a need for development of a plasma biomarker indicative of NRP2 pathway activation to potentially inform prostate cancer prognosis. Therefore, we investigated if plasma levels of NRP2 or vascular endothelial growth factor C (VEGF-C), a known soluble ligand of NRP2, are prognostic for prostate cancer. We hypothesized that plasma NRP2 and VEGF-C would be associated with more advanced disease or relapsed disease. METHODS NRP2 and VEGF-C levels were quantified by enzyme-linked immunoassay in plasma samples obtained from 145 prostate cancer patients in an opportunistic biobank. These patients were either (1) newly diagnosed (N = 28), (2) in remission (N = 56), or (3) relapsed disease (N = 61). Plasma samples from 15 adult males without known malignancy served as a comparator cohort. Statistical analysis was performed to investigate the association of plasma NRP2/VEGF-C with patient outcomes, adjusting for age, race, prostate-specific antigen (PSA), Gleason score, and tumor stage at diagnosis. RESULTS Neither NRP2 nor VEGF-C levels were significantly different in cancer patients compared to noncancer controls. We observed no clear association between plasma NRP2 and disease severity. Increased plasma VEGF-C was significantly associated with disease remission and correlated with Stage I/II and intermediate-risk Gleason score. Neither NRP2 nor VEGF-C correlated with PSA level. CONCLUSIONS Although tissue NRP2 expression correlates with severe disease, this was not observed for plasma NRP2. Plasma NRP2 levels did not correlate with disease severity or relapse. VEGF-C was highest in patients in remission and with less severe disease. Future investigation is needed to identify noninvasive methods to assess tumor NRP2 status.
Collapse
Affiliation(s)
- Sarah A Mullen
- Department of Internal Medicine, Division of Hematology/Oncology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Dipanwita Das
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Negin Ziamiavaghi
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Robin High
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Kaustubh Datta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Benjamin A Teply
- Department of Internal Medicine, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
4
|
Poormolaie N, Mohammadi M, Mir A, Asadi M, Kararoudi AN, Vahedian V, Maroufi NF, Rashidi M. Xanthomicrol: Effective therapy for cancer treatment. Toxicol Rep 2023; 10:436-440. [PMID: 37102154 PMCID: PMC10123071 DOI: 10.1016/j.toxrep.2023.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/13/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
Cancer treatment is one of the main challenges of global health. For decades, researchers have been trying to find anti-cancer compounds with minimal side effects. In recent years, flavonoids, as a group of polyphenolic compounds, have attracted the attention of researchers due to their beneficial effects on health. Xanthomicrol is one of the flavonoids that has the ability to inhibit growth, proliferation, survival and cell invasion and ultimately tumor progression. Xanthomicrol, as active anti-cancer compounds, can be effective in the prevention and treatment of cancer. Therefore, the use of flavonoids can be suggested as a treatment along with other medicinal agents. It is obvious that additional investigations in cellular levels and animal models are still needed. In this review article, the effects of xanthomicrol on various cancers have been reviewed.
Collapse
|
5
|
Yin D, Zhang H, Yang C, Zhang W, Yang S. A More Biomimetic Cell Migration Assay with High Reliability and Its Applications. Pharmaceuticals (Basel) 2022; 15:ph15060695. [PMID: 35745614 PMCID: PMC9229299 DOI: 10.3390/ph15060695] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/15/2022] [Accepted: 04/26/2022] [Indexed: 02/04/2023] Open
Abstract
Cell migration refers to the directional movement of cells to the surrounding cell-free zone in response to chemical and mechanical stimuli. A cell migration assay is an essential device for studying pharmaceutical and medical problems. In this paper, we present a novel approach to a cell migration assay on a chip with two merits, namely (i) simultaneous creation of many cell samples on the same condition and (ii) cells migrating while being stressed in a fluidic environment. The first merit has addressed the problem of poor reproducibility in experimental studies for medical problems such as wound healing, and the second merit has made the cell migration device, which is an in vitro environment, more biomimetic. The two merits are attributed to a novel mechanical method to simultaneously create many cell-free zones and to the design of a microfluidic process to create shear stress in cells uniformly. Two applications were studied on our device to explore its effectiveness. The first application is regarding the combination chemotherapy of cisplatin and doxorubicin (Adriamycin) on cervical cancer cells (HeLa). The second application is regarding inhibiting the migration of endothelial cells (HUVEC) in the process of anti-angiogenesis.
Collapse
Affiliation(s)
- Di Yin
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China; (D.Y.); (H.Z.)
| | - Hongbo Zhang
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China; (D.Y.); (H.Z.)
| | - Chun Yang
- Department of Mechanical Engineering, College of Engineering, Saskatoon, SK S7N 5A9, Canada;
| | - Wenjun Zhang
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China; (D.Y.); (H.Z.)
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
- Correspondence: (W.Z.); (S.Y.)
| | - Shihmo Yang
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China; (D.Y.); (H.Z.)
- Biomedical Science and Technology Research Centre, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
- Correspondence: (W.Z.); (S.Y.)
| |
Collapse
|
6
|
Virumbrales-Muñoz M, Ayuso JM, Loken JR, Denecke KM, Rehman S, Skala MC, Abel EJ, Beebe DJ. Microphysiological model of the renal cell carcinoma to inform anti-angiogenic therapy. Biomaterials 2022; 283:121454. [PMID: 35299086 PMCID: PMC9254636 DOI: 10.1016/j.biomaterials.2022.121454] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 02/18/2022] [Accepted: 03/03/2022] [Indexed: 12/18/2022]
Abstract
Renal cell carcinomas are common genitourinary tumors characterized by high vascularization and strong reliance on glycolysis. Despite the many available therapies for renal cell carcinomas, first-line targeted therapies, such as cabozantinib, and durable reaponses are seen in only a small percentage of patients. Yet, little is known about the mechanisms that drive response (or lack thereof). This dearth of knowledge can be explained by the dynamic and complex microenvironment of renal carcinoma, which remains challenging to recapitulate in vitro. Here, we present a microphysiological model of renal cell carcinoma, including a tubular blood vessel model of induced pluripotent stem cell-derived endothelial cells and an adjacent 3D carcinoma model. Our model recapitulated hypoxia, glycolic metabolism, and sprouting angiogenesis. Using our model, we showed that cabozantinib altered cancer cell metabolism and decreased sprouting angiogenesis but did not restore barrier function. This microphysiological model could be helpful to elucidate, through multiple endpoints, the contributions of the relevant environmental components in eliciting a functional response or resistance to therapy in renal cell carcinoma.
Collapse
Affiliation(s)
- María Virumbrales-Muñoz
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, 1111 Highland Avenue, Madison, WI, 53705, USA; University of Wisconsin Carbone Cancer Center, Wisconsin Institutes for Medical Research, 1111 Highland Ave, Madison, WI, 53705, USA; Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Jose M Ayuso
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, 1111 Highland Avenue, Madison, WI, 53705, USA; University of Wisconsin Carbone Cancer Center, Wisconsin Institutes for Medical Research, 1111 Highland Ave, Madison, WI, 53705, USA; Department of Dermatology, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Jack R Loken
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Kathryn M Denecke
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Shujah Rehman
- Morgridge Institute for Research, 330 N Orchard Street, Madison, WI, 53715, USA
| | - Melissa C Skala
- University of Wisconsin Carbone Cancer Center, Wisconsin Institutes for Medical Research, 1111 Highland Ave, Madison, WI, 53705, USA; Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, 53705, USA; Morgridge Institute for Research, 330 N Orchard Street, Madison, WI, 53715, USA
| | - E Jason Abel
- Department of Urology University of Wisconsin School of Medicine and Public Health, Madison, 1111 Highland Ave, Madison, WI, 53705, USA
| | - David J Beebe
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, 1111 Highland Avenue, Madison, WI, 53705, USA; University of Wisconsin Carbone Cancer Center, Wisconsin Institutes for Medical Research, 1111 Highland Ave, Madison, WI, 53705, USA; Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, 53705, USA.
| |
Collapse
|
7
|
García-Caballero M, Torres-Vargas JA, Marrero AD, Martínez-Poveda B, Medina MÁ, Quesada AR. Angioprevention of Urologic Cancers by Plant-Derived Foods. Pharmaceutics 2022; 14:pharmaceutics14020256. [PMID: 35213989 PMCID: PMC8875200 DOI: 10.3390/pharmaceutics14020256] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 02/05/2023] Open
Abstract
The number of cancer cases worldwide keeps growing unstoppably, despite the undeniable advances achieved by basic research and clinical practice. Urologic tumors, including some as prevalent as prostate, bladder or kidney tumors, are no exceptions to this rule. Moreover, the fact that many of these tumors are detected in early stages lengthens the duration of their treatment, with a significant increase in health care costs. In this scenario, prevention offers the most cost-effective long-term strategy for the global control of these diseases. Although specialized diets are not the only way to decrease the chances to develop cancer, epidemiological evidence support the role of certain plant-derived foods in the prevention of urologic cancer. In many cases, these plants are rich in antiangiogenic phytochemicals, which could be responsible for their protective or angiopreventive properties. Angiogenesis inhibition may contribute to slow down the progression of the tumor at very different stages and, for this reason, angiopreventive strategies could be implemented at different levels of chemoprevention, depending on the targeted population. In this review, epidemiological evidence supporting the role of certain plant-derived foods in urologic cancer prevention are presented, with particular emphasis on their content in bioactive phytochemicals that could be used in the angioprevention of cancer.
Collapse
Affiliation(s)
- Melissa García-Caballero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
| | - José Antonio Torres-Vargas
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
| | - Ana Dácil Marrero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
| | - Beatriz Martínez-Poveda
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), E-28019 Madrid, Spain
| | - Miguel Ángel Medina
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
- CIBER de Enfermedades Raras (CIBERER), E-29071 Malaga, Spain
| | - Ana R. Quesada
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
- CIBER de Enfermedades Raras (CIBERER), E-29071 Malaga, Spain
- Correspondence:
| |
Collapse
|
8
|
Qian K, Fu D, Jiang B, Wang Y, Tian F, Song L, Li L. Mechanism of Hedyotis Diffusa in the Treatment of Cervical Cancer. Front Pharmacol 2022; 12:808144. [PMID: 34975504 PMCID: PMC8715005 DOI: 10.3389/fphar.2021.808144] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 11/15/2021] [Indexed: 12/27/2022] Open
Abstract
Cervical cancer is one of the most common malignant tumors among women in the world. In clinical practice, Hedyotis diffusa has pharmacological effects in treating cervical cancer, but its components are relatively complex, and the mechanism of Hedyotis diffusa in treating cervical cancer is still unclear. In this work, the potential active components and mechanism of Hedyotis diffusa in the treatment of cervical cancer were explored by means of network pharmacology. By constructing its active ingredient-target network, and enriching and analyzing the targets, we found the key targets and their effective components (beta-Sitosterol and Quercetin) that play a therapeutic role. Finally, we evaluated the prognostic value of the core target genes through survival analysis. Our work initially explored the therapeutic mechanism of cervical cancer, which lays a theoretical foundation for further exploring its pharmacological action and its clinical application.
Collapse
Affiliation(s)
- Kai Qian
- Department of Cardiology, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Medical College of YiChun University, Yichun, China
| | - Dan Fu
- Medical College of YiChun University, Yichun, China
| | - Baorui Jiang
- Medical College of YiChun University, Yichun, China
| | - Yue Wang
- Medical College of YiChun University, Yichun, China
| | - Fei Tian
- Medical College of YiChun University, Yichun, China
| | - Li Song
- Medical College of YiChun University, Yichun, China
| | - Lei Li
- Department of Cardiology, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Medical College of YiChun University, Yichun, China
| |
Collapse
|
9
|
Alves ADCS, Bruinsmann FA, Guterres SS, Pohlmann AR. Organic Nanocarriers for Bevacizumab Delivery: An Overview of Development, Characterization and Applications. Molecules 2021; 26:4127. [PMID: 34299401 PMCID: PMC8305806 DOI: 10.3390/molecules26144127] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/29/2021] [Accepted: 07/02/2021] [Indexed: 01/08/2023] Open
Abstract
Bevacizumab (BCZ) is a recombinant humanized monoclonal antibody against the vascular endothelial growth factor, which is involved in the angiogenesis process. Pathologic angiogenesis is observed in several diseases including ophthalmic disorders and cancer. The multiple administrations of BCZ can cause adverse effects. In this way, the development of controlled release systems for BCZ delivery can promote the modification of drug pharmacokinetics and, consequently, decrease the dose, toxicity, and cost due to improved efficacy. This review highlights BCZ formulated in organic nanoparticles providing an overview of the physicochemical characterization and in vitro and in vivo biological evaluations. Moreover, the main advantages and limitations of the different approaches are discussed. Despite difficulties in working with antibodies, those nanocarriers provided advantages in BCZ protection against degradation guaranteeing bioactivity maintenance.
Collapse
Affiliation(s)
- Aline de Cristo Soares Alves
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre 90610-000, Brazil; (F.A.B.); (S.S.G.)
| | | | | | - Adriana Raffin Pohlmann
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre 90610-000, Brazil; (F.A.B.); (S.S.G.)
| |
Collapse
|
10
|
Kim D, Amatya R, Hwang S, Lee S, Min KA, Shin MC. BSA-Silver Nanoparticles: A Potential Multimodal Therapeutics for Conventional and Photothermal Treatment of Skin Cancer. Pharmaceutics 2021; 13:pharmaceutics13040575. [PMID: 33920666 PMCID: PMC8073043 DOI: 10.3390/pharmaceutics13040575] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/02/2021] [Accepted: 04/14/2021] [Indexed: 12/22/2022] Open
Abstract
Silver nanoparticles (NPs) have attracted a considerable interest in the field of cancer research due to their potential utility in cancer therapy. In the present study, we developed bovine serum albumin (BSA)-coated silver NPs (BSA-Silver NPs) and characterized in vitro multimodal therapeutic activities of NPs for the treatment of skin cancer. BSA-Silver NPs were synthesized by a single-step reduction process, and the successful preparation was verified through a list of physical characterizations, including transmission electron microscopy (TEM), ultraviolet-visible (UV–VIS) light spectroscopy, dynamic light scattering (DLS), and Fourier transform infrared (FT-IR) spectroscopy. The synthesized BSA-Silver NPs showed marked cytocidal effects on B16F10 melanoma cells, which was likely caused by oxidative stress. BSA-Silver NPs also elicited significant anti-angiogenic effects on HUVEC (human umbilical vein endothelial cell) by inhibiting their proliferation, migration, and tube formation. Moreover, BSA-Silver NPs showed a considerable light-to-heat conversion ability, suggesting their utility as photothermal agents. Overall, our findings suggest that BSA-Silver NPs may be promising candidates for the multimodal therapy of skin cancer.
Collapse
Affiliation(s)
- Dasom Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju, Gyeongnam 52828, Korea; (D.K.); (R.A.)
| | - Reeju Amatya
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju, Gyeongnam 52828, Korea; (D.K.); (R.A.)
| | - Seungmi Hwang
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, 197 Injero, Gimhae, Gyeongnam 50834, Korea; (S.H.); (S.L.)
| | - Sumi Lee
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, 197 Injero, Gimhae, Gyeongnam 50834, Korea; (S.H.); (S.L.)
| | - Kyoung Ah Min
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, 197 Injero, Gimhae, Gyeongnam 50834, Korea; (S.H.); (S.L.)
- Correspondence: (K.A.M.); (M.C.S.); Tel.: +82-55-320-3459 (K.A.M.); +82-55-772-2421 (M.C.S.)
| | - Meong Cheol Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju, Gyeongnam 52828, Korea; (D.K.); (R.A.)
- Correspondence: (K.A.M.); (M.C.S.); Tel.: +82-55-320-3459 (K.A.M.); +82-55-772-2421 (M.C.S.)
| |
Collapse
|
11
|
Singh HD, Ma JX, Takahashi Y. Distinct roles of LRP5 and LRP6 in Wnt signaling regulation in the retina. Biochem Biophys Res Commun 2021; 545:8-13. [PMID: 33545636 DOI: 10.1016/j.bbrc.2021.01.068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 01/20/2021] [Indexed: 12/11/2022]
Abstract
Dysregulation of Wnt signaling is implicated in multiple ocular disorders. The roles of Wnt co-receptors LRP5 and LRP6 in Wnt signaling regulation remain elusive, as most retinal cells express both of the co-receptors. To address this question, LRP5 and LRP6 were individually knocked-out in a human retinal pigment epithelium cell line using the CRISPR-Cas9 technology. Wnt signaling activity induced by various Wnt ligands was measured using wild-type and the KO cell lines. The results identified three groups of Wnt ligands based on their co-receptor specificity: 1) activation of Wnt signaling only through LRP6, 2) through both LRP5 and LRP6 and 3) predominantly through LRP5. These results indicate that LRP5 and LRP6 have differential roles in Wnt signaling regulation.
Collapse
Affiliation(s)
- Harminder D Singh
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jian-Xing Ma
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Yusuke Takahashi
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
12
|
Khasimbi S, Ali F, Manda K, Sharma A, Chauhan G, Wakode S. Dihydropyrimidinones Scaffold as a Promising Nucleus for Synthetic Profile and Various Therapeutic Targets: A Review. Curr Org Synth 2020; 18:270-293. [PMID: 33290199 DOI: 10.2174/1570179417666201207215710] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND This review elaborates the updated synthetic and pharmacological approaches of a known group of dihydropyrimidinones/thiones from the multi-component reaction like Biginelli reaction, which was named Pietro Biginelli in 1891. This review consists of the reaction of an aromatic aldehyde, urea and ethyl acetoacetate leading to dihydropyrimidinone/thione. Currently, the scientific movement to develop economically viable green methods using compounds that are reusable, non-volatile, easily obtained, etc. Objective: This review covers the recent synthesis and pharmacological advancement of dihydropyrimidinones/ thiones moiety, along with covering the structure-activity relationship of the most potent compounds, which may prove to become better, more efficacious and safer agents. Thus, this review may help the researchers in drug designing and development of new Dihydropyrimidinones entities. CONCLUSION This review focuses on the wide application of dihydropyrimidinone/thione review reports the design, synthesis and pharmacological activities of nitrogen-sulphur containing dihydropyrimidinone moiety by using multi-component reaction. Dihydropyrimidinones (DHPM) pharmacophore is an important heterocyclic ring in medicinal chemistry. It is derived from multi-component reactions, "Biginelli reaction" and plays a critical role as anticancer, antioxidant, antimicrobial, anti-inflammatory, anti-HIV-1, antimalarial, anti-inflammatory, antihypertensive and anti-tubercular agents. Exhaustive research has led to its vast biological profile, with a wide range of therapeutic application.
Collapse
Affiliation(s)
- Shaik Khasimbi
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Sector-3, Pushp Vihar, New Delhi, India
| | - Faraat Ali
- Laboratory Services, Botswana Medicines Regulatory Authority, Gaborone, Botswana
| | - Kiran Manda
- Department of Pharmaceutical Chemistry, Andhra University South Campus, Andhra University, Visakhapatnam, Andhra Pradesh, 530003, India
| | - Anjali Sharma
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Sector-3, Pushp Vihar, New Delhi, India
| | - Garima Chauhan
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Sector-3, Pushp Vihar, New Delhi, India
| | - Sharad Wakode
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Sector-3, Pushp Vihar, New Delhi, India
| |
Collapse
|
13
|
Moon J, Choi SH, Lee MJ, Jo DH, Park UC, Yoon SO, Woo SJ, Oh JY. Ocular surface complications of local anticancer drugs for treatment of ocular tumors. Ocul Surf 2020; 19:16-30. [PMID: 33238207 DOI: 10.1016/j.jtos.2020.11.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/12/2020] [Accepted: 11/16/2020] [Indexed: 02/03/2023]
Abstract
Local chemotherapy is increasingly used, either in combination with surgery or as monotherapy, for management of ocular tumors. Yet many of the local chemotherapeutic agents used for ocular tumors are cytotoxic drugs that are frequently associated with toxicities in normal ocular tissues. Understanding and managing these side effects are important because they affect treatment tolerability, outcome and quality of vision. Herein, we review local anticancer drugs administered for the treatment of ocular tumors, with an emphasis on their toxicities to the ocular surface, adnexa and lacrimal drainage system. We provide the underlying mechanisms and management strategies for the ocular side effects. Recent innovations in anticancer immunotherapy and ocular drug delivery systems also are discussed as new potential therapeutic modalities for alleviation of side effects.
Collapse
Affiliation(s)
- Jayoon Moon
- Department of Ophthalmology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea; Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Se Hyun Choi
- Department of Ophthalmology, Hallym University Sacred Heart Hospital, 22, Gwanpyeong-ro 170 Beon-gil, Dongan-gu, Anyang-si, Gyeonggi-do, 14068, South Korea
| | - Min Joung Lee
- Department of Ophthalmology, Hallym University Sacred Heart Hospital, 22, Gwanpyeong-ro 170 Beon-gil, Dongan-gu, Anyang-si, Gyeonggi-do, 14068, South Korea
| | - Dong Hyun Jo
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Un Chul Park
- Department of Ophthalmology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Sun-Ok Yoon
- R & D Lab, Eutilex Co., Ltd, Gasan Digital 1-ro 25, Geumcheon-gu, Seoul, 08594, South Korea
| | - Se Joon Woo
- Department of Ophthalmology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea; Department of Ophthalmology, Seoul National University Bundang Hospital, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
| | - Joo Youn Oh
- Department of Ophthalmology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea; Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.
| |
Collapse
|
14
|
de Cristo Soares Alves A, Lavayen V, Figueiró F, Dallemole DR, de Fraga Dias A, Cé R, Battastini AMO, Guterres SS, Pohlmann AR. Chitosan-Coated Lipid-Core Nanocapsules Functionalized with Gold-III and Bevacizumab Induced In Vitro Cytotoxicity against C6 Cell Line and In Vivo Potent Antiangiogenic Activity. Pharm Res 2020; 37:91. [PMID: 32385723 DOI: 10.1007/s11095-020-02804-0] [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: 11/01/2019] [Accepted: 03/18/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE Bevacizumab (BCZ) is a recombinant monoclonal antibody that inhibits the biological activity of the vascular endothelial growth factor, which has an important role in angiogenesis for tumoral growth and progression. In this way, our objective was to develop chitosan-coated lipid-core nanocapsules functionalized with BCZ by an organometallic complex using gold-III. METHODS The formulation was produced and characterized in relation to physicochemical characteristics. Furthermore, the antitumoral and antiangiogenic activities were evaluated against C6 glioma cell line and chicken embryo chorioallantoic membrane (CAM), respectively. RESULTS Final formulation showed nanometric size, narrow polydispersity, positive zeta potential and gold clusters size lower than 2 nm. BCZ in aqueous solution (0.01-0.10 μmol L-1) did not show cytotoxic activity in vitro against C6 glioma cell line; although, MLNC-Au-BCZ showed cytotoxicity with a median inhibition concentration of 30 nmol L-1 of BCZ. Moreover, MLNC-Au-BCZ demonstrated cellular internalization dependent on incubation time and BCZ concentration. BCZ solution did not induce significant apoptosis as compared to MLNC-Au-BCZ within 24 h of treatment. CAM assay evidenced potent antiangiogenic activity for MLNC-Au-BCZ, representing a decrease of 5.6 times in BCZ dose comparing to BCZ solution. CONCLUSION MLNC-Au-BCZ is a promising product for the treatment of solid tumors.
Collapse
Affiliation(s)
- Aline de Cristo Soares Alves
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Av. Ipiranga, 2752, Porto Alegre, RS, 90610-000, Brazil.
| | - Vladimir Lavayen
- Departamento de Química Inorgânica, Instituto de Química, Universidade Federal do Rio Grande do Sul, Av. Bento Gonçalves, 9500, Porto Alegre, RS, 91501-970, Brazil
| | - Fabrício Figueiró
- Departamento de Bioquímica, Instituto de Ciências da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-003, Brazil.,Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-003, Brazil
| | - Danieli Rosane Dallemole
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Av. Ipiranga, 2752, Porto Alegre, RS, 90610-000, Brazil
| | - Amanda de Fraga Dias
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-003, Brazil
| | - Rodrigo Cé
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Av. Ipiranga, 2752, Porto Alegre, RS, 90610-000, Brazil
| | - Ana Maria Oliveira Battastini
- Departamento de Bioquímica, Instituto de Ciências da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-003, Brazil
| | - Silvia Stanisçuaski Guterres
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Av. Ipiranga, 2752, Porto Alegre, RS, 90610-000, Brazil
| | - Adriana Raffin Pohlmann
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Av. Ipiranga, 2752, Porto Alegre, RS, 90610-000, Brazil. .,Departamento de Química Orgânica, Instituto de Química, Universidade Federal do Rio Grande do Sul, Av. Bento Gonçalves, 9500, Porto Alegre, RS, 91501-970, Brazil.
| |
Collapse
|
15
|
Karsch-Bluman A, Benny O. Necrosis in the Tumor Microenvironment and Its Role in Cancer Recurrence. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1225:89-98. [PMID: 32030649 DOI: 10.1007/978-3-030-35727-6_6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer recurrence is one of the most imminent problems in the current world of medicine, and it is responsible for most of the cancer-related death rates worldwide. Long-term administration of anticancer cytotoxic drugs may act as a double-edged sword, as necrosis may lead to renewed cancer progression and treatment resistance. The lack of nutrients, coupled with the induced hypoxia, triggers cell death and secretion of signals that affect the tumor niche. Many efforts have been made to better understand the contribution of hypoxia and metabolic stress to cancer progression and resistance, but mostly with respect to inflammation. Here we provide an overview of the direct anticancer effects of necrotic signals, which are not necessarily mediated by inflammation and the role of DAMPs (damage-associated molecular patterns) on the formation of a pro-cancerous environment.
Collapse
Affiliation(s)
- Adi Karsch-Bluman
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ofra Benny
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
16
|
Lu L, Chen H, Hao D, Zhang X, Wang F. The functions and applications of A7R in anti-angiogenic therapy, imaging and drug delivery systems. Asian J Pharm Sci 2019; 14:595-608. [PMID: 32104486 PMCID: PMC7032227 DOI: 10.1016/j.ajps.2019.04.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 03/31/2019] [Accepted: 04/24/2019] [Indexed: 12/26/2022] Open
Abstract
Vascular endothelial growth factor receptor 2 (VEGFR-2) and neuropilin-1 (NRP-1) are two prominent antiangiogenic targets. They are highly expressed on vascular endothelial cells and some tumor cells. Therefore, targeting VEGFR-2 and NRP-1 may be a potential antiangiogenic and antitumor strategy. A7R, a peptide with sequence of Ala-Thr-Trp-Leu-Pro-Pro-Arg that was found by phage display of peptide libraries, can preferentially target VEGFR-2 and NRP-1 and destroy the binding between vascular endothelial growth factor 165 (VEGF165) and VEGFR-2 or NRP-1. This peptide is a new potent inhibitor of tumor angiogenesis and a targeting ligand for cancer therapy. This review describes the discovery, function and mechanism of the action of A7R, and further introduces the applications of A7R in antitumor angiogenic treatments, tumor angiogenesis imaging and targeted drug delivery systems. In this review, strategies to deliver different drugs by A7R-modified liposomes and nanoparticles are highlighted. A7R, a new dual targeting ligand of VEGFR-2 and NRP-1, is expected to have efficient therapeutic or targeting roles in tumor drug delivery.
Collapse
Affiliation(s)
- Lu Lu
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Hongyuan Chen
- Department of General Surgery, Shandong University Affiliated Shandong Provincial Hospital, Jinan 250021, China
| | - Dake Hao
- Department of Surgery, UC Davis Health Medical Center, Sacramento 95817, USA
| | - Xinke Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Fengshan Wang
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| |
Collapse
|
17
|
Zhou X, Ouyang S, Li J, Huang X, Ai X, Zeng Y, Lv Y, Cai M. The novel non-immunological role and underlying mechanisms of B7-H3 in tumorigenesis. J Cell Physiol 2019; 234:21785-21795. [PMID: 31222741 DOI: 10.1002/jcp.28936] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 12/12/2022]
Abstract
B7 homolog 3 (B7-H3) has been proven to be involved in tumorigenesis. An elucidation of its role and underlying mechanisms is essential to an understanding of tumorigenesis and the development of effective clinical applications. B7-H3 is abnormally overexpressed in many types of cancer and is generally associated with a poor clinical prognosis. B7-H3 inhibits the initiation of the "caspase cascade" by the Janus kinase/signal transducers and activators of transcription pathway to resist tumor cell apoptosis. B7-H3 accelerates malignant proliferation by attacking the checkpoint mechanism of the tumor cell cycle through the phosphatidylinositol 3-kinase and protein kinase B pathway. B7-H3 reprograms the metabolism of glucose and lipids and transforms the metabolic flux of tumor cells to promote tumorigenesis. B7-H3 induces abnormal angiogenesis by recruiting vascular endothelial growth factor and matrix metalloproteinase to tumor lesions. B7-H3 strongly promotes tumorigenesis through antiapoptotic, pro-proliferation, metabolism reprogramming, and pro-angiogenesis.
Collapse
Affiliation(s)
- Xiangqi Zhou
- Department of Radiotherapy of the First Affiliated Hospital, University of South China, Hengyang, China.,Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Shuhui Ouyang
- Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Jianjun Li
- Department of Urology of the Second Affiliated Hospital, University of South China, Hengyang, China
| | - Xin Huang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,Division of Gynecological Oncology, Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Xiaohong Ai
- Department of Radiotherapy of the First Affiliated Hospital, University of South China, Hengyang, China
| | - Yixin Zeng
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, China
| | - Yuncheng Lv
- Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Manbo Cai
- Department of Radiotherapy of the First Affiliated Hospital, University of South China, Hengyang, China
| |
Collapse
|
18
|
Nominato LF, Dias AC, Dias LC, Fantucci MZ, Mendes da Silva LEC, Murashima ADA, Rocha EM. Prevention of Corneal Neovascularization by Adenovirus Encoding Human Vascular Endothelial Growth Factor Soluble Receptor (s-VEGFR1) in Lacrimal Gland. Invest Ophthalmol Vis Sci 2019; 59:6036-6044. [PMID: 30574658 DOI: 10.1167/iovs.17-22322] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The aims of this study were (1) to determine the efficacy of adenovirus vector serotype 5 (Ad) encoding human soluble VEGF receptor 1 (s-VEGFR1) gene transfer to the lacrimal gland (LG); (2) to investigate whether expression of s-VEGFR1 prevents corneal neovascularization (CNV) induced by alkali burns; and (3) to evaluate the safety of the procedure. Methods AdVEGFR1 vectors (25 μL, 1 × 1010 pfu/mL) were injected in the right LGs of rats and were compared with AdNull vector (25 μL, 1 × 1010 pfu/mL) or 25 μL of saline (Control) before cornea alkali burns with 1 M NaOH. After 7 days, CNV was documented at the slit lamp. Tear secretion was measured with phenol red threads. The animals were tested for s-VEGFR1 mRNA and protein in the LG by quantitative (q)PCR and immunohistochemistry staining, respectively. qPCR was used to compare the mRNA levels of IL-1β, IL-6, and TNF-α in the LG and ipsilateral trigeminal ganglion (TG). Results Ad-VEGFR1 transfected 83% (10/12) of the rats. VEGFR1 was present in LG acinar cells. CNV was prevented in 9 of 12 animals in the Ad-VEGFR1 group, compared with the Ad-Null (3:10) and Control groups (1:10) (P = 0.0317). The tear secretion and cytokine mRNA levels in the LG and TG were similar in all three groups (P > 0.05). Conclusions Adenoviral vector gene transfer was safe for LG structure and function. The LG as the target tissue showed local expression of human s-VEGFR1, and CNV was prevented in most of the eyes exposed to alkali burns.
Collapse
Affiliation(s)
- Luis Fernando Nominato
- Department of Ophthalmology, Otorhinolaryngology and Head and Neck Surgery, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Ana Carolina Dias
- Department of Ophthalmology, Otorhinolaryngology and Head and Neck Surgery, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Lara Cristina Dias
- Department of Ophthalmology, Otorhinolaryngology and Head and Neck Surgery, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Marina Zilio Fantucci
- Department of Ophthalmology, Otorhinolaryngology and Head and Neck Surgery, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | | | - Adriana de Andrade Murashima
- Department of Ophthalmology, Otorhinolaryngology and Head and Neck Surgery, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Eduardo Melani Rocha
- Department of Ophthalmology, Otorhinolaryngology and Head and Neck Surgery, Ribeirão Preto Medical School, University of São Paulo, Brazil
| |
Collapse
|
19
|
Wang X, Xia HY, Qin HY, Kang XP, Hu HY, Zheng J, Jiang JY, Yao LA, Xu YW, Zhang T, Zhang XL. 20(S)-protopanaxadiol induces apoptosis in human umbilical vein endothelial cells by activating the PERK-eIF2alpha-ATF4 signaling pathway. J Cell Biochem 2018; 120:5085-5096. [PMID: 30259568 DOI: 10.1002/jcb.27785] [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] [Received: 07/20/2018] [Accepted: 09/06/2018] [Indexed: 01/19/2023]
Abstract
20(S)-protopanaxadiol (PPD)-type ginsenosides are generally believed to be the most pharmacologically active components of Panax ginseng. These compounds induce apoptotic cell death in various cancer cells, which suggests that they have anti-cancer activity. Anti-angiogenesis is a promising therapeutic approach for controlling angiogenesis-related diseases such as malignant tumors, age-related macular degeneration, and atherosclerosis. Studies showed that 20(S)-PPD at low concentrations induces endothelial cell growth, but in our present study, we found 20(S)-PPD at high concentrations inhibited cell growth and mediated apoptosis in human umbilical vein endothelial cells (HUVECs). The mechanism by which high concentrations of 20(S)-PPD mediate endothelial cell apoptosis remains elusive. The present current study investigated how 20(S)-PPD induces apoptosis in HUVECs for the first time. We found that caspase-9 and its downstream caspase, caspase-3, were cleaved into their active forms after 20(S)-PPD treatment. Treatment with 20(S)-PPD decreased the level of Bcl-2 expression but did not change the level of Bax expression. 20(S)-PPD induced endoplasmic reticulum stress in HUVECs and stimulated UPR signaling, initiated by protein kinase R-like endoplasmic reticulum kinase (PERK) activation. Total protein expression and ATF4 nuclear import were increased, and CEBP-homologous protein (CHOP) expression increased after treatment with 20(S)-PPD. Furthermore, siRNA-mediated knockdown of PERK or ATF4 inhibited the induction of CHOP expression and 20(s)-PPD-induced apoptosis. Collectively, our findings show that 20(S)-PPD inhibits HUVEC growth by inducing apoptosis and that ATF4 expression activated by the PERK-eIF2α signaling pathway is essential for this process. These findings suggest that high concentrations of 20(S)-PPD could be used to treat angiogenesis-related diseases.
Collapse
Affiliation(s)
- Xue Wang
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua-Ying Xia
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hong-You Qin
- Shanghai Shenyou Biological Technology Company, Shanghai, China
| | - Xiang-Ping Kang
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hai-Yan Hu
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Zheng
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jia-Ye Jiang
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ling-Ai Yao
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan-Wu Xu
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tong Zhang
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xue-Li Zhang
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
20
|
Dithiolethiones: a privileged pharmacophore for anticancer therapy and chemoprevention. Future Med Chem 2018; 10:1241-1260. [DOI: 10.4155/fmc-2017-0281] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dithiolethiones are five-membered sulfur-containing cyclic scaffolds that exhibit antioxidative, anti-inflammatory, antithrombic and chemotherapeutic activities. Dithiolethiones display the chemopreventive and cytoprotective effects by activating the antioxidant response element and mounting the transcription of cytoprotective phase II enzymatic machinery. In addition, several classes of dithiolethiones efficiently modulate the activities of proteins that play crucial roles in normal and cancer cells, including glutathione S-transferase, cyclooxygenases and master regulator NF-κB. The present paper summarizes synthetic aspects, pharmacological potentials and biological attributes of dithiolethiones and its derivatives. Additionally, this review concludes with a discussion on how the current state-of-the-art technologies may help in defining a structure–activity relationship of dithiolethiones, thereby facilitating the design and synthesis of potent drug candidates.
Collapse
|
21
|
Pan L, Yu G, Chen X, Li X. Nicotinic acid inhibits angiogenesis likely through cytoskeleton remodeling. Organogenesis 2017; 13:183-191. [PMID: 28933636 DOI: 10.1080/15476278.2017.1364829] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Angiogenesis is a physiological procedure during which the new blood vessels develop from the pre-existing vessels. Uncontrolled angiogenesis is related to various diseases including cancers. Clinical inhibition of undesired angiogenesis is still under investigation. We utilized nicotinic acid, a family member of the B-vitamin niacin (vitamin B3) that has been used in the prevention and treatment of atherosclerosis or other lipid-metabolic disorders, to treat human umbilical vein endothelial cells (HUVECs) and chick chorioallantoic membrane (CAM), and investigated its influence on angiogenesis in vitro and in vivo. We found that nicotinic acid could obviously inhibit HUVEC proliferation induced by vascular endothelial growth factor. Both the in vitro and in vivo assays showed that nicotinic acid could significantly inhibit the process of angiogenesis. To further investigate the mechanism underlying the effect of nicotinic acid on angiogenesis, we found that it might function via regulating the cytoskeleton arrangements, especially the rearranging the structures of F-actin and paxillin. In summary, we discovered that nicotinic acid could obviously inhibit the process of angiogenesis by changing the angiogenesis factor expression levels and inducing the cytoskeleton rearrangement of endothelial cells.
Collapse
Affiliation(s)
- Lemen Pan
- a Department of Vascular Surgery , The Second Affiliated Hospital of Suzhou University , Suzhou , Jiangsu Province , P.R. China.,b Department of Vascular Surgery , The First Affiliated Hospital of Wenzhou Medical University , Nanbaixiang, Ouhai District, Wenzhou , Zhejiang Province , P.R. China
| | - Guanfeng Yu
- b Department of Vascular Surgery , The First Affiliated Hospital of Wenzhou Medical University , Nanbaixiang, Ouhai District, Wenzhou , Zhejiang Province , P.R. China
| | - Xiangjian Chen
- b Department of Vascular Surgery , The First Affiliated Hospital of Wenzhou Medical University , Nanbaixiang, Ouhai District, Wenzhou , Zhejiang Province , P.R. China
| | - Xiaoqiang Li
- a Department of Vascular Surgery , The Second Affiliated Hospital of Suzhou University , Suzhou , Jiangsu Province , P.R. China
| |
Collapse
|
22
|
Antofine, a natural phenanthroindolizidine alkaloid, suppresses angiogenesis via regulation of AKT/mTOR and AMPK pathway in endothelial cells and endothelial progenitor cells derived from mouse embryonic stem cells. Food Chem Toxicol 2017; 107:201-207. [DOI: 10.1016/j.fct.2017.06.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/18/2017] [Accepted: 06/22/2017] [Indexed: 11/17/2022]
|
23
|
Hu W, Fang J, Nie J, Dai L, Zhang J, Chen X, Ma X, Tian G, Wu D, Han S, Han J, Wang Y, Long J. Efficacy and safety of extended use of platinum-based doublet chemotherapy plus endostatin in patients with advanced nonsmall cell lung cancer. Medicine (Baltimore) 2016; 95:e4183. [PMID: 27428214 PMCID: PMC4956808 DOI: 10.1097/md.0000000000004183] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The aim of this study was to investigate the efficacy and safety of the extended use of platinum-based doublet chemotherapy (PT-DC) plus endostatin in patients with advanced nonsmall cell lung cancer (NSCLC).We performed a retrospective analysis of 200 newly diagnosed advanced NSCLC patients who had received at least 1 cycle of endostatin plus PT-DC between September 2009 and November 2014. Of these patients, 155 received 4 or more cycles of therapy (the extended therapy group), while 45 received less than 4 cycles of therapy (the control group). Clinical tumor responses, progression-free survival (PFS), overall survival (OS), and toxicity profiles were recorded and retrospectively analyzed.In the extended therapy group, 67 patients (43.2%) achieved a best overall response rate of partial response (PR), while in the control group, 13 patients (28.9%) had a best overall response rate of PR. After a median follow-up of 15.9 months, the median PFS and OS were 8.0 and 23.1 months in the extended arm and 5.8 and 14.0 months in the control arm, respectively. There were statistically significant differences in median PFS and OS between these 2 arms. Hematologic and gastrointestinal toxicities occurred more frequently in the extended therapy group, but no statistically significant difference was detected in grade 3 to 4 toxicities overall between these 2 groups.In conclusion, extended treatment using endostatin combined with PT-DC can provide additional survival benefits and satisfactory toxicity profiles in previously untreated patients with NSCLC, which merits further evaluation in a larger prospective study.
Collapse
Affiliation(s)
| | - Jian Fang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Oncology II, Peking University Cancer Hospital and Institute, Beijing, China
- Correspondence: Jian Fang, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Oncology II, Peking University Cancer Hospital and Institute, No. 52 Fucheng Road, Haidian District, Beijing 100142, China (e-mail: )
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Gilcy GK, Kuttan G. Evaluation of Antiangiogenic Efficacy of Emilia sonchifolia (L.) DC on Tumor-Specific Neovessel Formation by Regulating MMPs, VEGF, and Proinflammatory Cytokines. Integr Cancer Ther 2016; 15:NP1-NP12. [PMID: 27146127 PMCID: PMC5739157 DOI: 10.1177/1534735416630807] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 12/12/2015] [Accepted: 01/07/2016] [Indexed: 10/28/2022] Open
Abstract
Formation of new blood vessels from preexisting vasculature is an indispensable process in tumor initiation, invasion, and metastasis. Novel therapeutic approaches target endothelial cells involved in the process of angiogenesis, due to their genetic stability relative to the rapidly mutating drug-resistant cancer cells. In the present study, we investigated the effect of an active fraction from Emilia sonchifolia, belonging to the family Asteraceae, a plant well known for its anti-inflammatory and antitumor effects, on the inhibition of tumor-specific angiogenesis. Administration of the active fraction from E sonchifolia (AFES; 5 mg/kg, body weight, intraperitoneally) containing the major compound γ-humulene significantly inhibited B16F10 melanoma-induced capillary formation in C57BL/6 mice. The level of serum vascular endothelial growth factor and serum proinflammatory cytokines such as interleukin-1β, interleukin-6, tumor necrosis factor-α, and granulocyte-macrophage colony-stimulating factor were also reduced significantly. At the same time, administration of AFES significantly enhanced the production of antiangiogenic factors such as tissue inhibitor of matrix metalloproteinase-1. Dose-dependent reduction can be seen in the budding and expansion of microvessels from rat thoracic aorta by AFES treatment. Inhibition of the activation of proenzyme to active enzyme of matrix metalloproteinase along with a successful reduction of proliferation, invasion, and migration of human umbilical vein endothelial cells demonstrated the antiangiogenic effect of AFES in vitro. To date, no study has examined the antiangiogenic activity of this plant with already well-known anti-inflammatory and antitumor effects. Results obtained in the present study by using both in vivo and in vitro angiogenic models altogether proved the inhibitory effect of AFES on tumor-specific neovessel formation.
Collapse
Affiliation(s)
| | - Girija Kuttan
- Amala Cancer Research Centre, Thrissur, Kerala, India
| |
Collapse
|
25
|
Wu FTH, Lee CR, Bogdanovic E, Prodeus A, Gariépy J, Kerbel RS. Vasculotide reduces endothelial permeability and tumor cell extravasation in the absence of binding to or agonistic activation of Tie2. EMBO Mol Med 2016; 7:770-87. [PMID: 25851538 PMCID: PMC4459817 DOI: 10.15252/emmm.201404193] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Angiopoietin-1 (Ang1) activation of Tie2 receptors on endothelial cells (ECs) reduces adhesion by tumor cells (TCs) and limits junctional permeability to TC diapedesis. We hypothesized that systemic therapy with Vasculotide (VT)—a purported Ang1 mimetic, Tie2 agonist—can reduce the extravasation of potentially metastatic circulating TCs by similarly stabilizing the host vasculature. In vitro, VT and Ang1 treatments impeded endothelial hypermeability and the transendothelial migration of MDA-MB-231•LM2-4 (breast), HT29 (colon), or SN12 (renal) cancer cells to varying degrees. In mice, VT treatment inhibited the transit of TCs through the pulmonary endothelium, but not the hepatic or lymphatic endothelium. In the in vivo LM2-4 model, VT monotherapy had no effect on primary tumors, but significantly delayed distant metastatic dissemination to the lungs. In the post-surgical adjuvant treatment setting, VT therapeutically complemented sunitinib therapy, an anti-angiogenic tyrosine kinase inhibitor which limited the local growth of residual disease. Unexpectedly, detailed investigations into the putative mechanism of action of VT revealed no evidence of Tie2 agonism or Tie2 binding; alternative mechanisms have yet to be determined.
Collapse
Affiliation(s)
- Florence T H Wu
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Christina R Lee
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Elena Bogdanovic
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Aaron Prodeus
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Jean Gariépy
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Robert S Kerbel
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|
26
|
Olfert IM. Physiological Capillary Regression is not Dependent on Reducing VEGF Expression. Microcirculation 2016; 23:146-56. [PMID: 26660949 PMCID: PMC4744091 DOI: 10.1111/micc.12263] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 12/03/2015] [Indexed: 01/04/2023]
Abstract
Investigations into physiologically controlled capillary regression report the provocative finding that microvessel regression occurs in the face of persistent elevation of skeletal muscle VEGF expression. TSP-1, a negative angiogenic regulator, is increasingly being observed to temporally correlate with capillary regression, suggesting that increased TSP-1 (and not reduction in VEGF per se) is needed to initiate, and likely regulate, capillary regression. Based on evidence being gleaned from physiologically mediated regression of capillaries, it needs to be recognized that capillary regression (and perhaps capillary rarefaction with disease) is not simply the reversal of factors used to stimulate angiogenesis. Rather, the conceptual understanding that angiogenesis and capillary regression each have specific and unique requirements that are biologically constrained to opposite sides of the balance between positive and negative angioregulatory factors may shed light on why anti-VEGF therapies have not lived up to the promise in reversing angiogenesis and providing the cure that many had hoped toward fighting cancer. Emerging evidence from physiological controlled angiogenesis suggest that cases involving excessive or uncontrolled capillary expansion may be best treated by therapies designed to increase expression of negative angiogenic regulators, whereas those involving capillary rarefaction may benefit from inhibiting negative regulators (like TSP-1).
Collapse
Affiliation(s)
- I Mark Olfert
- Division of Exercise Physiology, Center for Cardiovascular and Respiratory Sciences, Mary Babb Randolph Cancer Center, West Virginia Clinical and Translational Science Institute, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| |
Collapse
|
27
|
Hu K, Babapoor-Farrokhran S, Rodrigues M, Deshpande M, Puchner B, Kashiwabuchi F, Hassan SJ, Asnaghi L, Handa JT, Merbs S, Eberhart CG, Semenza GL, Montaner S, Sodhi A. Hypoxia-inducible factor 1 upregulation of both VEGF and ANGPTL4 is required to promote the angiogenic phenotype in uveal melanoma. Oncotarget 2016; 7:7816-28. [PMID: 26761211 PMCID: PMC4884956 DOI: 10.18632/oncotarget.6868] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 01/04/2016] [Indexed: 12/25/2022] Open
Abstract
PURPOSE Expression of the hypoxia-inducible factor (HIF)-1-regulated gene product, vascular endothelial growth factor (VEGF), correlates with tumor vascularity in patients with uveal melanoma (UM). While the relationship between HIF-1 and VEGF in cancer is well-studied, their relative contribution to the angiogenic phenotype in UM has not previously been interrogated. Here we evaluate the contribution of HIF-1, VEGF, and a second HIF-1-regulated gene product, angiopoietin-like 4 (ANGPTL4), to angiogenesis in UM. EXPERIMENTAL DESIGN UM cells were examined for expression of HIF-1α, VEGF, and ANGPTL4. Their contribution to the angiogenic potential of UM cells was assessed using the endothelial cell tubule formation and directed in vivo angiogenesis assays. These results were corroborated in tissue from UM animal models and in tissue from patients with UM. RESULTS Inhibition of VEGF partially reduced tubule formation promoted by conditioned medium from UM cells. Inhibition of ANGPTL4, which was highly expressed in hypoxic UM cells, a UM orthotopic transplant model, a UM tumor array, and vitreous samples from UM patients, inhibited the angiogenic potential of UM cells in vitro and in vivo; this effect was additive to VEGF inhibition. CONCLUSIONS Targeting both ANGPTL4 and VEGF may be required for the effective inhibition of angiogenesis in UM.
Collapse
Affiliation(s)
- Ke Hu
- 1 Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA,2 The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | - Murilo Rodrigues
- 1 Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Monika Deshpande
- 1 Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Brooks Puchner
- 1 Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Syed Junaid Hassan
- 1 Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Laura Asnaghi
- 3 Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - James T. Handa
- 1 Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Shannath Merbs
- 1 Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Charles G. Eberhart
- 1 Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA,3 Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Gregg L. Semenza
- 4 Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, Biological Chemistry, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Silvia Montaner
- 5 Department of Oncology and Diagnostic Sciences, Greenebaum Cancer Center, University of Maryland, Baltimore, MD, USA
| | - Akrit Sodhi
- 1 Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
28
|
Role of miR-497 in VEGF-A-mediated cancer cell growth and invasion in non-small cell lung cancer. Int J Biochem Cell Biol 2016; 70:118-25. [DOI: 10.1016/j.biocel.2015.10.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/12/2015] [Accepted: 10/13/2015] [Indexed: 12/16/2022]
|
29
|
Li L, Wang Z. PDGF-BB, NGF and BDNF enhance pulp-like tissue regeneration via cell homing. RSC Adv 2016. [DOI: 10.1039/c6ra20290j] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In this study, we investigated the cytobiological effects of PDGF-BB, NGF and BDNF on the culture of BMSCs from rats and explored a viable approach for regenerating ectopic dental pulp-like tissue via cell homing.
Collapse
Affiliation(s)
- Lin Li
- Department of Endodontics
- Department of Oral Implantology
- School & Hospital of Stomatology
- Tongji University
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration
| | - Zuolin Wang
- Department of Endodontics
- Department of Oral Implantology
- School & Hospital of Stomatology
- Tongji University
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration
| |
Collapse
|
30
|
Liu F, Wang J, Fu Q, Zhang X, Wang Y, Liu J, Huang J, Lv X. VEGF-activated miR-144 regulates autophagic survival of prostate cancer cells against Cisplatin. Tumour Biol 2015; 37:10.1007/s13277-015-4383-1. [PMID: 26566625 DOI: 10.1007/s13277-015-4383-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 11/04/2015] [Indexed: 12/24/2022] Open
Abstract
Cisplatin is a commonly used chemotherapy drug for prostate cancer (PC). However, some PCs are resistant to cisplatin treatment, while the molecular mechanisms underlying the resistance of PCs to cisplatin are not completely understood. In this study, we found that cisplatin dose-dependently activated Beclin-1 in two PC cell lines, PC3 and LNCap. Autophagy suppression significantly increased the cisplatin-induced cell death of these PC cells in a CCK-8 assay. Moreover, microRNA (miR)-144 levels were significantly downregulated in cisplatin-treated PC cells, in a VEGF-dependent manner. Bioinformatics analysis showed that miR-144 targeted the 3'-UTR of Beclin-1 mRNA to inhibit its translation, which was confirmed by luciferase reporter assay. In PC patients after cisplatin treatment, low miR-144 levels appeared to predict poor outcome of patients' survival. Together, these data suggest that cisplatin may induce VEGF to suppress miR-144 levels in PC cells, which subsequently upregulates Beclin-1 to increase autophagic cell survival against cisplatin-induced cell death. Upregulation of miR-144 or suppression of cell autophagy may improve the outcome of cisplatin therapy in PC.
Collapse
Affiliation(s)
- Feng Liu
- Department of Urology, Shanghai Fengxian District Central Hospital, Shanghai, 201499, China
| | - Jihong Wang
- Department of Urology, Shanghai Jiaotong University Affiliated the Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| | - Qiang Fu
- Department of Urology, Shanghai Jiaotong University Affiliated the Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China
| | - Xinru Zhang
- Department of Urology, Shanghai Jiaotong University Affiliated the Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China
| | - Ying Wang
- Department of Urology, Shanghai the Fifth People's Hospital, Shanghai, China
| | - Jialin Liu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jianwen Huang
- Department of Urology, Shanghai Jiaotong University Affiliated the Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China
| | - Xiangguo Lv
- Department of Urology, Shanghai Jiaotong University Affiliated the Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China
| |
Collapse
|
31
|
Bear HD, Tang G, Rastogi P, Geyer CE, Liu Q, Robidoux A, Baez-Diaz L, Brufsky AM, Mehta RS, Fehrenbacher L, Young JA, Senecal FM, Gaur R, Margolese RG, Adams PT, Gross HM, Costantino JP, Paik S, Swain SM, Mamounas EP, Wolmark N. Neoadjuvant plus adjuvant bevacizumab in early breast cancer (NSABP B-40 [NRG Oncology]): secondary outcomes of a phase 3, randomised controlled trial. Lancet Oncol 2015; 16:1037-1048. [PMID: 26272770 PMCID: PMC4624323 DOI: 10.1016/s1470-2045(15)00041-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/03/2015] [Accepted: 06/05/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND NSABP B-40 was a 3 × 2 factorial trial testing whether adding capecitabine or gemcitabine to docetaxel followed by doxorubicin plus cyclophosphamide neoadjuvant chemotherapy would improve outcomes in women with operable, HER2-negative breast cancer and whether adding neoadjuvant plus adjuvant bevacizumab to neoadjuvant chemotherapy regimens would also improve outcomes. As reported previously, addition of neoadjuvant bevacizumab increased the proportion of patients achieving a pathological complete response, which was the primary endpoint. We present secondary patient outcomes, including disease-free survival, a specified endpoint by protocol, and data for distant recurrence-free interval, and overall survival, which were not prespecified endpoints but were collected prospectively. METHODS In this randomised controlled trial (NSABP B-40), we enrolled women aged 18 years or older, with operable, HER2-non-amplified invasive adenocarcinoma of the breast, 2 cm or greater in diameter by palpation, clinical stage T1c-3, cN0, cN1, or cN2a, without metastatic disease and diagnosed by core needle biopsy. Patients received one of three docetaxel-based neoadjuvant regimens for four cycles: docetaxel alone (100 mg/m(2)) with addition of capecitabine (825 mg/m(2) oral twice daily days 1-14, 75 mg/m(2) docetaxel) or with addition of gemcitabine (1000 mg/m(2) days 1 and 8 intravenously, 75 mg/m(2) docetaxel), all followed by neoadjuvant doxorubicin and cyclophosphamide (60 mg/m(2) and 600 mg/m(2) intravenously) every 3 weeks for four cycles. Those randomly assigned to bevacizumab groups were to receive bevacizumab (15 mg/kg, every 3 weeks for six cycles) with neoadjuvant chemotherapy and postoperatively for ten doses. Randomisation was done (1:1:1:1:1:1) via a biased-coin minimisation procedure to balance the characteristics with respect to clinical nodal status, clinical tumour size, hormone receptor status, and age. Intent-to-treat analyses were done for disease-free survival and overall survival. This study is registered with ClinicalTrials.gov, number NCT00408408. FINDINGS Between Jan 5, 2007, and June 30, 2010, 1206 patients were enrolled in the study. Follow-up data were collected from Oct 31, 2007 to March 27, 2014, and were available for overall survival in 1186 patients, disease-free survival in 1184, and distant recurrence-free interval in 1181. Neither capecitabine nor gemcitabine increased disease-free survival or overall survival. Median follow-up was 4·7 years (IQR 4·0-5·2). The addition of bevacizumab significantly increased overall survival (hazard ratio 0·65 [95% CI 0·49-0·88]; p=0·004) but did not significantly increase disease-free survival (0·80 [0·63-1·01]; p=0·06). Four deaths occurred on treatment due to vascular disorder (docetaxel plus capecitabine followed by doxorubicin plus cyclophosphamide group), sudden death (docetaxel plus capecitabine followed by doxorubicin plus cyclophosphamide group), infective endocarditis (docetaxel plus bevacizumab followed by doxorubicin plus cyclophosphamide and bevacizumab group), and visceral arterial ischaemia (docetaxel followed by doxorubicin plus cyclophosphamide group). The most common grade 3-4 adverse events in the bevacizumab group were neutropenia (grade 3, 99 [17%]; grade 4, 37 [6%]), hand-foot syndrome (grade 3, 63 [11%]), and hypertension (grade 3, 60 [10%]; grade 4, two [<1%]) and in the non-bevacizumab group were neutropenia (grade 3, 98 [16%]; grade 4, 36 [6%]), fatigue (grade 3, 53 [9%]), and hand-foot syndrome (grade 3, 43 [7%]). INTERPRETATION The addition of gemcitabine or capecitabine to neoadjuvant docetaxel plus doxorubicin plus cyclophosphamide does not seem to provide any benefit to patients with operable breast cancer, and should not change clinical practice in the short term. The improved overall survival with bevacizumab contradicts the findings of other studies of bevacizumab in breast cancer and may indicate the need for additional investigation of this agent. FUNDING National Institutes of Health, Genentech, Roche Laboratories, Lilly Research Laboratories, and Precision Therapeutics.
Collapse
Affiliation(s)
- Harry D Bear
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Virginia Commonwealth University School of Medicine and the Massey Cancer Center, Richmond, VA, USA.
| | - Gong Tang
- NRG Oncology and the University of Pittsburgh, Pittsburgh, PA, USA
| | - Priya Rastogi
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Charles E Geyer
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Virginia Commonwealth University School of Medicine and the Massey Cancer Center, Richmond, VA, USA
| | - Qing Liu
- NRG Oncology and the University of Pittsburgh, Pittsburgh, PA, USA
| | - André Robidoux
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | - Luis Baez-Diaz
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Minority-Based CCOP, San Juan, Puerto Rico
| | - Adam M Brufsky
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; University of Pittsburgh Cancer Institute, Magee Womens Hospital, Pittsburgh, PA, USA
| | - Rita S Mehta
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; University of California at Irvine, Division of Hematology/Oncology, Chao Family Comprehensive Cancer Center, Orange, CA, USA
| | - Louis Fehrenbacher
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Kaiser Permanente Oncology Clinical Trials, Northern California, Vallejo, CA, USA
| | - James A Young
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Colorado Cancer Research Program, Colorado Springs, CO, USA
| | - Francis M Senecal
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Northwest Medical Specialties, Tacoma, WA, USA
| | - Rakesh Gaur
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; CCOP, Kansas City, MO, USA
| | - Richard G Margolese
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Jewish General Hospital, McGill University, Montréal, QC, Canada
| | - Paul T Adams
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Genesys Regional Medical Center, Grand Blanc, MI, USA
| | - Howard M Gross
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; CCOP, Dayton, Dayton, OH, USA
| | | | - Soonmyung Paik
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Severance Biomedical Science Institute and Department of Medical Oncology, Yonsei University College of Medicine, Seoul, Korea
| | - Sandra M Swain
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Washington Cancer Institute, MedStar Washington Hospital Center, Washington, DC, USA
| | - Eleftherios P Mamounas
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; UF Health Cancer Center at Orlando Health, Orlando, FL, USA
| | - Norman Wolmark
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Allegheny Cancer Center at Allegheny General Hospital, Pittsburgh, PA, USA
| |
Collapse
|
32
|
Saghiri MA, Asatourian A, Sorenson CM, Sheibani N. Role of angiogenesis in endodontics: contributions of stem cells and proangiogenic and antiangiogenic factors to dental pulp regeneration. J Endod 2015; 41:797-803. [PMID: 25649306 PMCID: PMC5223201 DOI: 10.1016/j.joen.2014.12.019] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 12/15/2014] [Accepted: 12/16/2014] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Dental pulp regeneration is a part of regenerative endodontics, which includes isolation, propagation, and re-transplantation of stem cells inside the prepared root canal space. The formation of new blood vessels through angiogenesis is mandatory to increase the survival rate of re-transplanted tissues. Angiogenesis is defined as the formation of new blood vessels from preexisting capillaries, which has great importance in pulp regeneration and homeostasis. Here the contribution of human dental pulp stem cells and proangiogenic and antiangiogenic factors to angiogenesis process and regeneration of dental pulp is reviewed. METHODS A search was performed on the role of angiogenesis in dental pulp regeneration from January 2005 through April 2014. The recent aspects of the relationship between angiogenesis, human dental pulp stem cells, and proangiogenic and antiangiogenic factors in regeneration of dental pulp were assessed. RESULTS Many studies have indicated an intimate relationship between angiogenesis and dental pulp regeneration. The contribution of stem cells and mechanical and chemical factors to dental pulp regeneration has been previously discussed. CONCLUSIONS Angiogenesis is an indispensable process during dental pulp regeneration. The survival of inflamed vital pulp and engineered transplanted pulp tissue are closely linked to the process of angiogenesis at sites of application. However, the detailed regulatory mechanisms involved in initiation and progression of angiogenesis in pulp tissue require investigation.
Collapse
Affiliation(s)
- Mohammad Ali Saghiri
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; Department of Biomedical Engineering, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.
| | | | - Christine M Sorenson
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; Department of Biomedical Engineering, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
33
|
Ye H, Karim AA, Loh XJ. Current treatment options and drug delivery systems as potential therapeutic agents for ovarian cancer: A review. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2014; 45:609-19. [DOI: 10.1016/j.msec.2014.06.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 06/09/2014] [Indexed: 12/26/2022]
|
34
|
Inhibition of metastasis of oral squamous cell carcinoma by anti-PLGF treatment. Tumour Biol 2014; 36:2695-701. [DOI: 10.1007/s13277-014-2892-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 11/21/2014] [Indexed: 12/11/2022] Open
|
35
|
Chen HC, Qiu JT, Yang FL, Liu YC, Chen MC, Tsai RY, Yang HW, Lin CY, Lin CC, Wu TS, Tu YM, Xiao MC, Ho CH, Huang CC, Lai CS, Hua MY. Magnetic-Composite-Modified Polycrystalline Silicon Nanowire Field-Effect Transistor for Vascular Endothelial Growth Factor Detection and Cancer Diagnosis. Anal Chem 2014; 86:9443-50. [DOI: 10.1021/ac5001898] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Hsiao-Chien Chen
- Department
of Chemical and Materials Engineering and Biosensor Group, Biomedical Engineering Research Center, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
- Department
of Biochemistry, School of Medicine, Taipei Medical University, Taipei 11031, Taiwan, Republic of China
| | - Jian-Tai Qiu
- Department
of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
- Department
of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan, Republic of China
| | - Fu-Liang Yang
- National Nano Device Laboratories, Hsinchu Science Park, Hsinchu 30078, Taiwan, Republic of China
| | - Yin-Chih Liu
- Department
of Chemical and Materials Engineering and Biosensor Group, Biomedical Engineering Research Center, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
| | - Min-Cheng Chen
- National Nano Device Laboratories, Hsinchu Science Park, Hsinchu 30078, Taiwan, Republic of China
| | - Rung-Ywan Tsai
- Electronics
and Optoelectronics Research Laboratories, Industrial Technology Research Institute, Hsinchu 31040, Taiwan, Republic of China
| | - Hung-Wei Yang
- Department
of Chemical and Materials Engineering and Biosensor Group, Biomedical Engineering Research Center, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
| | - Chia-Yi Lin
- National Nano Device Laboratories, Hsinchu Science Park, Hsinchu 30078, Taiwan, Republic of China
| | - Chu-Chi Lin
- Department
of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
- Department
of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan, Republic of China
| | - Tzong-Shoon Wu
- Department
of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
- Department
of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan, Republic of China
| | - Yi-Ming Tu
- Department
of Chemical and Materials Engineering and Biosensor Group, Biomedical Engineering Research Center, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
| | - Min-Cong Xiao
- Department
of Chemical and Materials Engineering and Biosensor Group, Biomedical Engineering Research Center, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
| | - Chia-Hua Ho
- National Nano Device Laboratories, Hsinchu Science Park, Hsinchu 30078, Taiwan, Republic of China
| | - Chien-Chao Huang
- National Nano Device Laboratories, Hsinchu Science Park, Hsinchu 30078, Taiwan, Republic of China
| | - Chao-Sung Lai
- Department
of Electronic Engineering and Biosensor Group,
Biomedical Engineering Research Center, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
| | - Mu-Yi Hua
- Department
of Chemical and Materials Engineering and Biosensor Group, Biomedical Engineering Research Center, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
| |
Collapse
|
36
|
Gong J, Zhu S, Zhang Y, Wang J. Interplay of VEGFa and MMP2 regulates invasion of glioblastoma. Tumour Biol 2014; 35:11879-85. [DOI: 10.1007/s13277-014-2438-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 08/04/2014] [Indexed: 12/29/2022] Open
|
37
|
Chen B, Zhang C, Dong P, Guo Y, Mu N. Molecular regulation of cervical cancer growth and invasion by VEGFa. Tumour Biol 2014; 35:11587-93. [PMID: 25135429 DOI: 10.1007/s13277-014-2463-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 08/06/2014] [Indexed: 12/25/2022] Open
Abstract
Although antivascular endothelial growth factor a (VEGFa) treatment has been well applied in cervical cancer therapy, the underlying molecular basis has not been precisely identified. Here, we examined the levels of VEGFa on the tumor growth and invasion in four commonly used human cervical cancer cell lines. We found that overexpression of VEGFa in these lines increased the tumor growth and invasiveness, while inhibition of VEGFa decreased the tumor growth and invasiveness. To figure out the involved signaling pathways, we applied specific inhibitors for ERK/MAPK, JNK, and PI3K/Akt signaling pathways, respectively, to VEGFa-overexpressing cervical cancer lines and found that only inhibition of PI3K/Akt signal transduction abolished VEGFa-induced increases in cell growth and invasiveness. Inhibition of Akt downstream mTor signaling similarly inhibited cell growth and invasion in VEGFa-overexpressing cervical cancer cells, suggesting that VEGFa may activate PI3K/Akt, and subsequently its downstream mTor signaling pathway, to promote cervical cancer cell growth and invasion. Furthermore, the effects of VEGFa-induced activation of mTor signaling cascades appeared to promote cancer cell growth through cyclinD1 and CDK4 activation and promote cancer cell invasion through MMP2 and MMP3. Taken together, our data suggest that anti-VEGFa treatment in cervical cancer may inhibit both tumor cell growth and invasion through PI3k/Akt/mTor signaling pathway.
Collapse
Affiliation(s)
- Baohuan Chen
- Department of Gynecology, Yantaishan Hospital, 91 Jiefang Road, Yantai, 264001, China
| | | | | | | | | |
Collapse
|
38
|
Maroof H, Salajegheh A, Smith RA, Lam AKY. Role of microRNA-34 family in cancer with particular reference to cancer angiogenesis. Exp Mol Pathol 2014; 97:298-304. [PMID: 25102298 DOI: 10.1016/j.yexmp.2014.08.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 08/01/2014] [Indexed: 02/07/2023]
Abstract
MicroRNA-34 is involved in pathogenesis in cancer by targeting different tumor-related genes. It could be a biomarker for predicting the prognosis of patients with cancer. In addition, miR-34 is involved in the tumor angiogenesis. Understanding the mechanism of the miR-34 in cancer and tumor angiogenesis will open horizons for development of anti-cancer and anti-angiogenesis drugs.
Collapse
Affiliation(s)
- Hamidreza Maroof
- Cancer Molecular Pathology, School of Medicine and Griffith Health Institute, Griffith University, Gold Coast, Queensland, Australia
| | - Ali Salajegheh
- Cancer Molecular Pathology, School of Medicine and Griffith Health Institute, Griffith University, Gold Coast, Queensland, Australia
| | - Robert Anthony Smith
- Cancer Molecular Pathology, School of Medicine and Griffith Health Institute, Griffith University, Gold Coast, Queensland, Australia
| | - Alfred King-Yin Lam
- Cancer Molecular Pathology, School of Medicine and Griffith Health Institute, Griffith University, Gold Coast, Queensland, Australia.
| |
Collapse
|
39
|
Xu L, Stevens J, Hilton MB, Seaman S, Conrads TP, Veenstra TD, Logsdon D, Morris H, Swing DA, Patel NL, Kalen J, Haines DC, Zudaire E, St Croix B. COX-2 inhibition potentiates antiangiogenic cancer therapy and prevents metastasis in preclinical models. Sci Transl Med 2014; 6:242ra84. [PMID: 24964992 PMCID: PMC6309995 DOI: 10.1126/scitranslmed.3008455] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Antiangiogenic agents that block vascular endothelial growth factor (VEGF) signaling are important components of current cancer treatment modalities but are limited by alternative ill-defined angiogenesis mechanisms that allow persistent tumor vascularization in the face of continued VEGF pathway blockade. We identified prostaglandin E2 (PGE2) as a soluble tumor-derived angiogenic factor associated with VEGF-independent angiogenesis. PGE2 production in preclinical breast and colon cancer models was tightly controlled by cyclooxygenase-2 (COX-2) expression, and COX-2 inhibition augmented VEGF pathway blockade to suppress angiogenesis and tumor growth, prevent metastasis, and increase overall survival. These results demonstrate the importance of the COX-2/PGE2 pathway in mediating resistance to VEGF pathway blockade and could aid in the rapid development of more efficacious anticancer therapies.
Collapse
Affiliation(s)
- Lihong Xu
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI) at Frederick, National Institutes of Health, Frederick, MD 21702, USA
| | - Janine Stevens
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI) at Frederick, National Institutes of Health, Frederick, MD 21702, USA
| | - Mary Beth Hilton
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI) at Frederick, National Institutes of Health, Frederick, MD 21702, USA. Basic Research Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Steven Seaman
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI) at Frederick, National Institutes of Health, Frederick, MD 21702, USA
| | - Thomas P Conrads
- Laboratory of Proteomics and Analytical Technologies, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Timothy D Veenstra
- Laboratory of Proteomics and Analytical Technologies, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Daniel Logsdon
- Basic Research Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Holly Morris
- Transgenic Core Facility, MCGP, NCI, Frederick, MD 21702, USA
| | - Deborah A Swing
- Transgenic Core Facility, MCGP, NCI, Frederick, MD 21702, USA
| | - Nimit L Patel
- Small Animal Imaging Program/LASP, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Joseph Kalen
- Small Animal Imaging Program/LASP, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Diana C Haines
- Pathology/Histotechnology Laboratory, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Enrique Zudaire
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI) at Frederick, National Institutes of Health, Frederick, MD 21702, USA
| | - Brad St Croix
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI) at Frederick, National Institutes of Health, Frederick, MD 21702, USA.
| |
Collapse
|
40
|
Zhou X, Qi Y. PLGF inhibition impairs metastasis of larynx carcinoma through MMP3 downregulation. Tumour Biol 2014; 35:9381-6. [PMID: 24946722 DOI: 10.1007/s13277-014-2232-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 06/11/2014] [Indexed: 12/25/2022] Open
Abstract
Cancer neovascularization plays a key role in the metastasis of larynx carcinoma. However, the molecular mechanism for the neovascularization control in larynx carcinoma is poorly understood. Since placental growth factor (PLGF) has been reported to be involved in pathological angiogenesis, and since matrix metalloproteinases (MMPs) are essential for extracellular matrix degradation during neovascularization, here we were prompted to examine whether PLGF and MMPs may play a coordinate role in the metastasis of larynx carcinoma. Our data showed that the expression of PLGF and MMP3 strongly correlated in the larynx carcinoma in the patients, and significant higher levels of PLGF and MMP3 were detected in the larynx carcinoma from the patients with metastasis of the primary cancer. Thus, we used a human larynx carcinoma cell line, Hep-2, to examine whether expression of PLGF and MMP3 may affect each other. We found that overexpression of PLGF in Hep-2 cells increased expression of MMP3, while inhibition of PLGF in Hep-2 cells decreased expression of MMP3. However, neither overexpression, nor inhibition of MMP3 in Hep-2 cells affected the expression level of PLGF. These data suggest that PLGF may function upstream of MMP3 in larynx carcinoma cells. We then analyzed how PLGF affected MMP3. Application of a specific ERK1/2 inhibitor to PLGF-overexpressing Hep-2 cells substantially abolished the effect of PLGF on MMP3 activation, suggesting that PLGF may increase expression of MMP3 via ERK/MAPK signaling pathway. Since anti-PLGF was recently applied in clinical trials to inhibit cancer-related angiogenesis, here our data further demonstrate that inhibition of cancer neovascularization by anti-PLGF is mediated not only by direct effect on endothelial growth and capillary permeability, but also by indirect effect via MMP3 on the extracellular matrix degradation in larynx carcinoma.
Collapse
Affiliation(s)
- Xu Zhou
- Otorhinolaryngology Department, Zhongshan Hospital, Fu Dan University, 180 Fenglin Road, 200032, ShangHai, China,
| | | |
Collapse
|
41
|
Zhuang H, Zhao X, Zhao L, Chang JY, Wang P. Progress of clinical research on targeted therapy combined with thoracic radiotherapy for non-small-cell lung cancer. Drug Des Devel Ther 2014; 8:667-75. [PMID: 24936128 PMCID: PMC4047835 DOI: 10.2147/dddt.s61977] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The combination of radiotherapy and targeted therapy is an important approach in the application of targeted therapy in clinical practice, and represents an important opportunity for the development of radiotherapy itself. Numerous agents, including epidermal growth factor receptor, monoclonal antibodies, tyrosine kinase inhibitors, and antiangiogenic therapies, have been used for targeted therapy. A number of studies of radiotherapy combined with targeted therapy in non-small-cell lung carcinoma have been completed or are ongoing. This paper briefly summarizes the drugs involved and the important related clinical research, and indicates that considerable progress has been made with the joint efforts of the two disciplines. Many issues, including drug selection, identification of populations most likely to benefit, timing of administration of medication, and side effects of treatment require further investigation. However, further fundamental research and accumulation of clinical data will provide a more comprehensive understanding of these therapies. Targeted therapy in combination with radiotherapy has a bright future.
Collapse
Affiliation(s)
- Hongqing Zhuang
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, and Tianjin Lung Cancer Center, Tianjin, People’s Republic of China
| | - Xianzhi Zhao
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, and Tianjin Lung Cancer Center, Tianjin, People’s Republic of China
| | - Lujun Zhao
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, and Tianjin Lung Cancer Center, Tianjin, People’s Republic of China
| | - Joe Y Chang
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ping Wang
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, and Tianjin Lung Cancer Center, Tianjin, People’s Republic of China
| |
Collapse
|
42
|
Mao D, Zhang Y, Lu H, Zhang H. Molecular basis underlying inhibition of metastasis of gastric cancer by anti-VEGFa treatment. Tumour Biol 2014; 35:8217-23. [PMID: 24850176 DOI: 10.1007/s13277-014-2095-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 05/13/2014] [Indexed: 12/25/2022] Open
Abstract
Neovascularization plays a substantial role in the invasiveness and metastasis of gastric cancer. However, the molecular mechanism underlying the control of neovascularization of gastric cancer remains undefined. Both vascular endothelial growth factor a (VEGFa) and matrix metalloproteinases 2 (MMP2) are essential for neovascularization by promoting endothelial mitogenesis and permeability and by promoting extracellular matrix degradation, respectively. Therefore, we were prompted to examine whether VEGFa and MMP2 may affect expression of each other to coordinate the neovascularization process. We found strong positive correlation of VEGFa and MMP2 levels in the gastric patients. Moreover, patients with metastasis of the original cancer had significantly higher levels of VEGFa and MMP2. Thus, we used a human gastric cancer cell line, SNU-5, to examine whether expression of VEGFa and MMP2 may affect each other. We found that overexpression of VEGFa in SNU-5 cells increased expression of MMP2, while inhibition of VEGFa in SNU-5 cells decreased expression of MMP2. On the other hand, overexpression of MMP2 in SNU-5 cells increased expression of VEGFa, while inhibition of MMP2 in SNU-5 cells decreased expression of VEGFa. These data suggest that expression of VEGFa and MMP2 may activate each other to reinforce a promoting effect on neovascularization. We then analyzed how VEGFa expression affects MMP2 level. Application of a specific Akt inhibitor to VEGFa-overexpressing SNU-5 cells substantially abolished the effect of VEGFa on MMP2 activation, suggesting that VEGFa may increase expression of MMP2 via PI3K/Akt signaling pathway. Since anti-VEGFa is a well-established therapy for many cancers, our data suggest that anti-VEGFa may not only inhibit neovascularization by prohibiting VEGFa-dependent endothelial mitogenesis and permeability increase but also by downregulating MMP2 to abolish the extracellular matrix degradation in gastric cancer.
Collapse
Affiliation(s)
- Dong Mao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Liaoning Medical College, No. 5-2 People Road, Jinzhou, 121001, China,
| | | | | | | |
Collapse
|
43
|
Blosser W, Vakana E, Wyss LV, Swearingen ML, Stewart J, Stancato L, Tate CM. A method to assess target gene involvement in angiogenesis in vitro and in vivo using lentiviral vectors expressing shRNA. PLoS One 2014; 9:e96036. [PMID: 24759702 PMCID: PMC3997504 DOI: 10.1371/journal.pone.0096036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 04/03/2014] [Indexed: 02/07/2023] Open
Abstract
Current methods to study angiogenesis in cancer growth and development can be difficult and costly, requiring extensive use of in vivo methodologies. Here, we utilized an in vitro adipocyte derived stem cell and endothelial colony forming cell (ADSC/ECFC) co-culture system to investigate the effect of lentiviral-driven shRNA knockdown of target genes compared to a non-targeting shRNA control on cord formation using High Content Imaging. Cord formation was significantly reduced following knockdown of the VEGF receptor VEGFR2 in VEGF-driven cord formation and the FGF receptor FGFR1 in basic FGF (bFGF)-driven cord formation. In addition, cord formation was significantly reduced following knockdown of the transcription factor forkhead box protein O1 (FOXO1), a protein with known positive effects on angiogenesis and blood vessel stabilization in VEGF- and bFGF-driven cord formation. Lentiviral shRNA also demonstrated utility for stable knockdown of VEGFR2 and FOXO1 in ECFCs, allowing for interrogation of protein knockdown effects on in vivo neoangiogenesis in a Matrigel plug assay. In addition to interrogating the effect of gene knockdown in endothelial cells, we utilized lentiviral shRNA to knockdown specificity protein 1 (SP1), a transcription factor involved in the expression of VEGF, in U-87 MG tumor cells to demonstrate the ability to analyze angiogenesis in vitro in a tumor-driven transwell cord formation system and in tumor angiogenesis in vivo. A significant reduction in tumor-driven cord formation, VEGF secretion, and in vivo tumor angiogenesis was observed upon SP1 knockdown. Therefore, evaluation of target gene knockdown effects in the in vitro co-culture cord formation assay in the ADSC/ECFC co-culture, ECFCs alone, and in tumor cells translated directly to in vivo results, indicating the in vitro method as a robust, cost-effective and efficient in vitro surrogate assay to investigate target gene involvement in endothelial or tumor cell function in angiogenesis.
Collapse
Affiliation(s)
- Wayne Blosser
- Oncology Research, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Eliza Vakana
- Oncology Research, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Lisa V. Wyss
- Advanced Testing Laboratories, Cincinnati, Ohio, United States of America
| | - Michelle L. Swearingen
- Oncology Research, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Julie Stewart
- Oncology Research, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Louis Stancato
- Oncology Research, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Courtney M. Tate
- Oncology Research, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| |
Collapse
|
44
|
Lerario AM, Moraitis A, Hammer GD. Genetics and epigenetics of adrenocortical tumors. Mol Cell Endocrinol 2014; 386:67-84. [PMID: 24220673 PMCID: PMC3943605 DOI: 10.1016/j.mce.2013.10.028] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 10/24/2013] [Indexed: 02/08/2023]
Abstract
Adrenocortical tumors are common neoplasms. Most are benign, nonfunctional and clinically irrelevant. However, adrenocortical carcinoma is a rare disease with a dismal prognosis and no effective treatment apart from surgical resection. The molecular genetics of adrenocortical tumors remain poorly understood. For decades, molecular studies relied on a small number of samples and were directed to candidate-genes. This approach, based on the elucidation of the genetics of rare genetic syndromes in which adrenocortical tumors are a manifestation, has led to the discovery of major dysfunctional molecular pathways in adrenocortical tumors, such as the IGF pathway, the Wnt pathway and TP53. However, with the advent of high-throughput methodologies and the organization of international consortiums to obtain a larger number of samples and high-quality clinical data, this paradigm is rapidly changing. In the last decade, genome-wide expression profile studies, microRNA profiling and methylation profiling allowed the identification of subgroups of tumors with distinct genetic markers, molecular pathways activation patterns and clinical behavior. As a consequence, molecular classification of tumors has proven to be superior to traditional histological and clinical methods in prognosis prediction. In addition, this knowledge has also allowed the proposal of molecular-targeted approaches to provide better treatment options for advanced disease. This review aims to summarize the most relevant data on the rapidly evolving field of genetics of adrenal disorders.
Collapse
Affiliation(s)
- Antonio M Lerario
- Adrenal Disorders Unit - LIM/42, Department of Endocrinology and Metabolism, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HC-FMUSP), Sao Paulo, Brazil
| | - Andreas Moraitis
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine Endocrine Oncology Program, University of Michigan Comprehensive Cancer Center, 1500 E. Medical Center Drive, Ann Arbor, MI 48109-5902, USA
| | - Gary D Hammer
- Endocrine Oncology Program, Center for Organogenesis, University of Michigan Health System, 109 Zina Pitcher Place, 1528 BSRB, Ann Arbor, MI 48109-2200, USA.
| |
Collapse
|
45
|
Else T, Kim AC, Sabolch A, Raymond VM, Kandathil A, Caoili EM, Jolly S, Miller BS, Giordano TJ, Hammer GD. Adrenocortical carcinoma. Endocr Rev 2014; 35:282-326. [PMID: 24423978 PMCID: PMC3963263 DOI: 10.1210/er.2013-1029] [Citation(s) in RCA: 564] [Impact Index Per Article: 56.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Adrenocortical carcinoma (ACC) is a rare endocrine malignancy, often with an unfavorable prognosis. Here we summarize the knowledge about diagnosis, epidemiology, pathophysiology, and therapy of ACC. Over recent years, multidisciplinary clinics have formed and the first international treatment trials have been conducted. This review focuses on evidence gained from recent basic science and clinical research and provides perspectives from the experience of a large multidisciplinary clinic dedicated to the care of patients with ACC.
Collapse
Affiliation(s)
- Tobias Else
- MEND/Division of Metabolism, Endocrinology, and Diabetes (T.E., T.J.G., G.D.H.), Division of Molecular Medicine and Genetics (V.M.R.), Department of Internal Medicine; Departments of Radiation Oncology (A.S., J.S.), Pathology (T.J.G.), and Radiology (A.K., E.M.C.); and Division of Endocrine Surgery (B.S.M.), Section of General Surgery, (A.C.K.), Department of Surgery, University of Michigan Hospital and Health Systems, Ann Arbor, Michigan 48109
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kienast Y, Klein C, Scheuer W, Raemsch R, Lorenzon E, Bernicke D, Herting F, Yu S, The HH, Martarello L, Gassner C, Stubenrauch KG, Munro K, Augustin HG, Thomas M. Ang-2-VEGF-A CrossMab, a Novel Bispecific Human IgG1 Antibody Blocking VEGF-A and Ang-2 Functions Simultaneously, Mediates Potent Antitumor, Antiangiogenic, and Antimetastatic Efficacy. Clin Cancer Res 2013; 19:6730-40. [DOI: 10.1158/1078-0432.ccr-13-0081] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
47
|
Mountzios G, Pentheroudakis G, Carmeliet P. Bevacizumab and micrometastases: revisiting the preclinical and clinical rollercoaster. Pharmacol Ther 2013; 141:117-24. [PMID: 24076268 DOI: 10.1016/j.pharmthera.2013.09.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 09/03/2013] [Indexed: 01/07/2023]
Abstract
The use of bevacizumab, a monoclonal antibody against vascular endothelial growth factor (VEGF), in combination with standard therapeutic approaches, has offered clinical benefit for patients with advanced colorectal, breast, ovarian, renal, non small-cell lung cancer and glioblastoma. However, the strategy of administering bevacizumab until disease progression has been challenged by certain preclinical evidence, suggesting that prolonged exposure to anti-VEGF treatment may elicit an adaptive-evasive response, resulting in a more aggressive tumor phenotype. Moreover, the use of bevacizumab in adjuvant chemotherapeutic regimens has led to less promising results than expected. Despite our poor understanding of how bevacizumab acts in micrometastatic disease, numerous clinical trials (involving >20,000 cancer patients) are ongoing or are planned to test the therapeutic benefit in the adjuvant setting. The discrepancy of bevacizumab's efficiency in the two settings calls into question the validity of current strategies that use similar treatment regimens for early and advanced diseases. Herein, we review the mechanisms of bevacizumab activity in the macro- as compared to the micrometastatic environment and discuss possible alternative strategies in the adjuvant setting that might spur attention for future clinical trials. Rather than providing an encyclopedic survey of the literature, we highlight exemplary principles.
Collapse
Affiliation(s)
- Giannis Mountzios
- Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece.
| | - George Pentheroudakis
- Department of Medical Oncology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, University of Leuven, Leuven, Belgium
| |
Collapse
|
48
|
Animal Models to Test Adjuvant Treatment: An Experimental Model of Colon Cancer. CURRENT COLORECTAL CANCER REPORTS 2013. [DOI: 10.1007/s11888-013-0180-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
49
|
Wang Z, Wang N, Han S, Wang D, Mo S, Yu L, Huang H, Tsui K, Shen J, Chen J. Dietary compound isoliquiritigenin inhibits breast cancer neoangiogenesis via VEGF/VEGFR-2 signaling pathway. PLoS One 2013; 8:e68566. [PMID: 23861918 PMCID: PMC3702614 DOI: 10.1371/journal.pone.0068566] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 05/14/2013] [Indexed: 11/26/2022] Open
Abstract
Angiogenesis is crucial for cancer initiation, development and metastasis. Identifying natural botanicals targeting angiogenesis has been paid much attention for drug discovery in recent years, with the advantage of increased safety. Isoliquiritigenin (ISL) is a dietary chalcone-type flavonoid with various anti-cancer activities. However, little is known about the anti-angiogenic activity of isoliquiritigenin and its underlying mechanisms. Herein, we found that ISL significantly inhibited the VEGF-induced proliferation of human umbilical vein endothelial cells (HUVECs) at non-toxic concentration. A series of angiogenesis processes including tube formation, invasion and migration abilities of HUVECs were also interrupted by ISL in vitro. Furthermore, ISL suppressed sprout formation from VEGF-treated aortic rings in an ex-vivo model. Molecular mechanisms study demonstrated that ISL could significantly inhibit VEGF expression in breast cancer cells via promoting HIF-1α (Hypoxia inducible factor-1α) proteasome degradation and directly interacted with VEGFR-2 to block its kinase activity. In vivo studies further showed that ISL administration could inhibit breast cancer growth and neoangiogenesis accompanying with suppressed VEGF/VEGFR-2 signaling, elevated apoptosis ratio and little toxicity effects. Molecular docking simulation indicated that ISL could stably form hydrogen bonds and aromatic interactions within the ATP-binding region of VEGFR-2. Taken together, our study shed light on the potential application of ISL as a novel natural inhibitor for cancer angiogenesis via the VEGF/VEGFR-2 pathway. Future studies of ISL for chemoprevention or chemosensitization against breast cancer are thus warranted.
Collapse
Affiliation(s)
- Zhiyu Wang
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Neng Wang
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Shouwei Han
- Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, China
| | - Dongmei Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Suilin Mo
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Linzhong Yu
- School of Chinese Medicine, South Medical University, Guangzhou, China
| | - Hui Huang
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kamchuen Tsui
- The Hong Kong Associate of Chinese Medicine, Hong Kong, China
| | - Jiangang Shen
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jianping Chen
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- * E-mail:
| |
Collapse
|
50
|
Chang JH, Garg NK, Lunde E, Han KY, Jain S, Azar DT. Corneal neovascularization: an anti-VEGF therapy review. Surv Ophthalmol 2012; 57:415-29. [PMID: 22898649 DOI: 10.1016/j.survophthal.2012.01.007] [Citation(s) in RCA: 261] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 01/14/2012] [Accepted: 01/16/2012] [Indexed: 01/02/2023]
Abstract
Corneal neovascularization is a serious condition that can lead to a profound decline in vision. The abnormal vessels block light, cause corneal scarring, compromise visual acuity, and may lead to inflammation and edema. Corneal neovascularization occurs when the balance between angiogenic and antiangiogenic factors is tipped toward angiogenic molecules. Vascular endothelial growth factor (VEGF), one of the most important mediators of angiogenesis, is upregulated during neovascularization. In fact, anti-VEGF agents have efficacy in the treatment of neovascular age-related macular degeneration, diabetic retinopathy, macular edema, neovascular glaucoma, and other neovascular diseases. These same agents have great potential for the treatment of corneal neovascularization. We review some of the most promising anti-VEGF therapies, including bevacizumab, VEGF trap, siRNA, and tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago 60612, USA.
| | | | | | | | | | | |
Collapse
|