1
|
Ren H, Chen J, Wu Z, Li C. Longitudinal study of factors associated with the anti-cancer efficacy and liver function in HCC patients treated with TACE in combination with percutaneous ablation. Front Oncol 2025; 15:1566865. [PMID: 40308490 PMCID: PMC12040659 DOI: 10.3389/fonc.2025.1566865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a major cancer challenge worldwide. Combination therapy using transcatheter arterial chemoembolization (TACE) and percutaneous ablation offers potential for improved outcomes. Objective To evaluate the efficacy and liver function preservation in HCC patients treated with combined TACE and percutaneous ablation, identifying key prognostic factors. Methods This longitudinal study included 200 HCC patients. Factors analyzed included tumor characteristics, liver function tests, and serologic markers. Statistical analyses determined associations with treatment outcomes and survival. Results Smaller tumors (≤5.0 cm) and lower AFP levels (<200 ng/mL) were associated with higher treatment efficacy, with an objective response rate of 67.3% for lower AFP levels versus 42.3% for higher levels. Liver function was better preserved in patients with lower AFP levels (78.2% vs. 57.7%). Tumor size and liver stiffness significantly influenced survival and liver function outcomes. Conclusion The combination of TACE and percutaneous ablation enhances outcomes in HCC, guided by specific prognostic markers. This supports the need for personalized approaches in HCC treatment and further research into combination therapies.
Collapse
Affiliation(s)
| | | | | | - Chen Li
- Hong Hui Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
2
|
Bawazir WA, Ali TE, Alsolimani AK, Assiri MA, Shati AA, Alfaifi MY, Elbehairi SEI. Novel ethyl 2-hydrazineylidenethiazolidin-5-ylidene acetate clubbed with coumarinylthiazolyl pyrazole system as potential VEGFR-2 inhibitors and apoptosis inducer: synthesis, cytotoxic evaluation, cell cycle, autophagy, in silico ADMET and molecular docking studies. RSC Adv 2025; 15:4829-4846. [PMID: 39957832 PMCID: PMC11822769 DOI: 10.1039/d5ra00250h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 02/09/2025] [Indexed: 02/18/2025] Open
Abstract
Novel derivatives of ethyl 3-substituted-2-{4-oxo-2-(2-((3-(2-oxo-2H-chromen-3-yl)-1-(4-phenylthiazol-2-yl)-1H-pyrazol-4-yl)methylene)hydrazineyl)thiazol-5(4H)-ylidene}acetate (5a-h) were synthesized and assessed for their cytotoxic potential against the liver cancer cell lines Huh-7 and HepG-2. Among these, compounds 5d and 5g demonstrated notable antiproliferative effects, which were benchmarked against the standard drug doxorubicin. To further understand the mechanisms behind their antiproliferative activity, compounds 5d and 5g were investigated for their impact on the cell cycle and their ability to induce apoptosis. They were found to induce significant cellular cycle arrest at the G1 phase. Besides, they potentially enhanced the cellular late apoptosis and reduced the cellular viability. In consent with the apoptosis results, compounds 5d and 5g displayed significant potential autophagic induction against the studied cancer cell lines. Further, both compounds 5d and 5g showed strong interactions with the VEGFR-2 receptor when they were studied using molecular docking. The ADMET prediction indicated that these bioactive compounds have the potential to serve as effective to fight liver cancer.
Collapse
Affiliation(s)
- Wafa A Bawazir
- Chemistry Department, Faculty of Science, King Abdulaziz University Jeddah Saudi Arabia
| | - Tarik E Ali
- Central Labs, King Khalid University AlQuraa Abha Saudi Arabia
- Department of Chemistry, Faculty of Science, King Khalid University AlQuraa Abha Saudi Arabia
| | - Ayat K Alsolimani
- Department of Chemistry, Faculty of Science, King Khalid University AlQuraa Abha Saudi Arabia
| | - Mohammed A Assiri
- Department of Chemistry, Faculty of Science, King Khalid University AlQuraa Abha Saudi Arabia
| | - Ali A Shati
- Department of Biology, Faculty of Science, King Khalid University AlQuraa Abha Saudi Arabia
| | - Mohammad Y Alfaifi
- Department of Biology, Faculty of Science, King Khalid University AlQuraa Abha Saudi Arabia
| | - Serag E I Elbehairi
- Department of Biology, Faculty of Science, King Khalid University AlQuraa Abha Saudi Arabia
| |
Collapse
|
3
|
Nakano A, Kawada T, Morita A, Nakahara T. Repeated treatment with VEGF receptor inhibitors induces phenotypic changes in endothelial cells and pericytes in the rat retina. Microvasc Res 2025; 157:104756. [PMID: 39454823 DOI: 10.1016/j.mvr.2024.104756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
Abnormal ocular angiogenesis is a major cause of visual impairment and vision loss in neovascularization-related diseases. Currently, anti-vascular endothelial growth factor (VEGF) drugs are used to treat ocular neovascularization, but repeated injections are needed to maintain their therapeutic effects. However, repeated injection of anti-VEGF drugs may affect the retinal blood vessel phenotype and diminish therapeutic effects. In this study, we aimed to investigate the phenotypic changes in endothelial cells and pericytes caused by the repeated interruption of the VEGF receptor signaling pathway in neonatal rats. KRN633 (10 mg/kg), a VEGF receptor tyrosine kinase inhibitor, was subcutaneously administered on postnatal day (P)-7 and P8 (first round), P14 and P15 (second round), and P21 and P22 (third round). The rat eyes were collected on P7, P9, P14, P16, P21, P23, P28, and P35. Using retinal flat-mount specimens stained with specific markers for vascular endothelial cells, basement membranes, and pericytes, the arteriolar tortuosity, capillary area density, and distribution of pericytes were evaluated. Significant loss of capillaries was observed the day after the first round of KRN633 treatment, after which aggressive angiogenesis occurred, leading to the formation of tortuous arterioles. Rats that completed second and third rounds of KRN633 treatment showed more severe abnormalities in the retinal vasculature than those that only completed first round treatment. Repeated treatment with KRN633 decreased the anti-angiogenic effects but increased the immunoreactivity of α-smooth muscle actin in the pericytes on veins and capillaries. α-Smooth muscle actin expression was inversely correlated to anti-angiogenic effects. Overall, these results revealed that repeated interruption of VEGF receptor signaling pathway altered the phenotypes of endothelial cells and pericytes and induced anti-VEGF drug resistance. Therefore, careful follow-up is necessary when using anti-VEGF drugs to treat abnormal angiogenesis-associated ocular diseases.
Collapse
Affiliation(s)
- Ayuki Nakano
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Takaaki Kawada
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Akane Morita
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Tsutomu Nakahara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan.
| |
Collapse
|
4
|
Hu X, Ye K, Bo S, Xiao Z, Ma M, Pan J, Zhong X, Zhang D, Mo X, Yu X, Chen M, Luo L, Shi C. Monitoring imatinib decreasing pericyte coverage and HIF-1α level in a colorectal cancer model by an ultrahigh-field multiparametric MRI approach. J Transl Med 2024; 22:712. [PMID: 39085929 PMCID: PMC11293104 DOI: 10.1186/s12967-024-05497-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Excessive pericyte coverage promotes tumor growth, and a downregulation may solve this dilemma. Due to the double-edged sword role of vascular pericytes in tumor microenvironment (TME), indiscriminately decreasing pericyte coverage by imatinib causes poor treatment outcomes. Here, we optimized the use of imatinib in a colorectal cancer (CRC) model in high pericyte-coverage status, and revealed the value of multiparametric magnetic resonance imaging (mpMRI) at 9.4T in monitoring treatment-related changes in pericyte coverage and the TME. METHODS CRC xenograft models were evaluated by histological vascular characterizations and mpMRI. Mice with the highest pericyte coverage were treated with imatinib or saline; then, vascular characterizations, tumor apoptosis and HIF-1α level were analyzed histologically, and alterations in the expression of Bcl-2/bax pathway were assessed through qPCR. The effects of imatinib were monitored by dynamic contrast-enhanced (DCE)-, diffusion-weighted imaging (DWI)- and amide proton transfer chemical exchange saturation transfer (APT CEST)-MRI at 9.4T. RESULTS The DCE- parameters provided a good histologic match the tumor vascular characterizations. In the high pericyte coverage status, imatinib exhibited significant tumor growth inhibition, necrosis increase and pericyte coverage downregulation, and these changes were accompanied by increased vessel permeability, decreased microvessel density (MVD), increased tumor apoptosis and altered gene expression of apoptosis-related Bcl-2/bax pathway. Strategically, a 4-day imatinib effectively decreased pericyte coverage and HIF-1α level, and continuous treatment led to a less marked decrease in pericyte coverage and re-elevated HIF-1α level. Correlation analysis confirmed the feasibility of using mpMRI parameters to monitor imatinib treatment, with DCE-derived Ve and Ktrans being most correlated with pericyte coverage, Ve with vessel permeability, AUC with microvessel density (MVD), DWI-derived ADC with tumor apoptosis, and APT CEST-derived MTRasym at 1 µT with HIF-1α. CONCLUSIONS These results provided an optimized imatinib regimen to achieve decreasing pericyte coverage and HIF-1α level in the high pericyte-coverage CRC model, and offered an ultrahigh-field multiparametric MRI approach for monitoring pericyte coverage and dynamics response of the TME to treatment.
Collapse
Affiliation(s)
- Xinpeng Hu
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Kunlin Ye
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Shaowei Bo
- Department of Medical Imaging, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Xingang Middle Road No. 466, Guangzhou, 510317, China
| | - Zeyu Xiao
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
- Engineering Research Center of Medical Imaging Artificial Intelligence for Precision Diagnosis and Treatment, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Mengjie Ma
- Department of Radiology, Guangzhou First People's Hospital, Panfu Road No. 1, Guangzhou, 510080, China
| | - Jinghua Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Xing Zhong
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Dong Zhang
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Xukai Mo
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Xiaojun Yu
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Minfeng Chen
- College of Pharmacy, Jinan University, West Huangpu Avenue No.601, Guangzhou, 510632, China.
| | - Liangping Luo
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China.
- Engineering Research Center of Medical Imaging Artificial Intelligence for Precision Diagnosis and Treatment, West Huangpu Avenue No. 613, Guangzhou, 510630, China.
| | - Changzheng Shi
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China.
- Engineering Research Center of Medical Imaging Artificial Intelligence for Precision Diagnosis and Treatment, West Huangpu Avenue No. 613, Guangzhou, 510630, China.
| |
Collapse
|
5
|
Sogo Y, Toyoda E, Nagai T, Takahashi T, Takizawa D, Watanabe M, Sato M. Disease-Modifying Effects of Lenvatinib, a Multiple Receptor Tyrosine Kinase Inhibitor, on Posttraumatic Osteoarthritis of the Knee. Int J Mol Sci 2024; 25:6514. [PMID: 38928219 PMCID: PMC11203559 DOI: 10.3390/ijms25126514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Angiogenesis and vascular endothelial growth factor (VEGF) are involved in osteoarthritis (OA). We previously reported the inhibitory effect of bevacizumab in a rabbit model of OA. In the current study, we investigated the effects of lenvatinib, an angiogenesis inhibitor targeting the VEGF and fibroblast growth factor receptors, on synovitis, osteophyte formation, and cartilage degeneration in a rabbit OA model. Posttraumatic OA was induced by anterior cruciate ligament transection (ACLT) on one knee of each rabbit. Rabbits were placed into four groups according to the following lenvatinib doses: untreated control (n = 12), L0.3: 0.3 mg/kg/day (n = 15), L1.0: 1.0 mg/kg/day (n = 14), and L3.0: 3.0 mg/kg/day (n = 13) groups. We evaluated limb pain using the weight distribution ratio measured with an incapacitance tester, macroscopic osteophyte formation, and femoral condyle synovium and cartilage histology. For cartilage evaluation, the following distal sites of the femur were evaluated separately: femoral-tibial (FT), femoral-patellar (FP), and femoral corner (between FP and FT). The weight distribution ratio at 12 weeks after surgery was higher in the L0.3 and L1.0 groups than in the control group. Osteophyte formation and synovitis scores were significantly lower in the L0.3, L1.0, and L3.0 groups than in the control group. The Osteoarthritis Research Society International scores of the FT, corner, and FP sites in the L0.3 group were lower than in the control group. The cartilage thickness ratio at the FT and corner sites was significantly lower in the L0.3 group than in the control group. Krenn's grading system of cartilage synovitis showed that all lenvatinib-administered groups had significantly lower scores than the control group. MMP3 expression level in cartilage tissue was significantly lower in the L3.0 group compared with the other three groups. ADAMTS5 expression was lower in the L3.0 group compared with the control and L0.3 groups. Oral administration of lenvatinib inhibited synovitis, osteophyte formation, and cartilage degeneration and reduced pain in a rabbit ACLT model. Lenvatinib is an oral VEGF inhibitor that is easier to administer than other VEGF inhibitors and may have potential as a treatment of posttraumatic OA.
Collapse
Affiliation(s)
- Yasuyuki Sogo
- Department of Orthopaedic Surgery, Surgical Science, School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan; (Y.S.); (E.T.); (T.T.); (D.T.); (M.W.)
- Center for Musculoskeletal innovative Research and Advancement (C-MiRA), Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan
| | - Eriko Toyoda
- Department of Orthopaedic Surgery, Surgical Science, School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan; (Y.S.); (E.T.); (T.T.); (D.T.); (M.W.)
- Center for Musculoskeletal innovative Research and Advancement (C-MiRA), Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan
| | - Toshihiro Nagai
- Department of Orthopaedic Surgery, Tokai University Hachioji Hospital, 1838 Ishikawa-cho, Hachioji 192-0032, Kanagawa, Japan;
| | - Takumi Takahashi
- Department of Orthopaedic Surgery, Surgical Science, School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan; (Y.S.); (E.T.); (T.T.); (D.T.); (M.W.)
- Center for Musculoskeletal innovative Research and Advancement (C-MiRA), Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan
| | - Daichi Takizawa
- Department of Orthopaedic Surgery, Surgical Science, School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan; (Y.S.); (E.T.); (T.T.); (D.T.); (M.W.)
- Center for Musculoskeletal innovative Research and Advancement (C-MiRA), Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan
| | - Masahiko Watanabe
- Department of Orthopaedic Surgery, Surgical Science, School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan; (Y.S.); (E.T.); (T.T.); (D.T.); (M.W.)
- Center for Musculoskeletal innovative Research and Advancement (C-MiRA), Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan
| | - Masato Sato
- Department of Orthopaedic Surgery, Surgical Science, School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan; (Y.S.); (E.T.); (T.T.); (D.T.); (M.W.)
- Center for Musculoskeletal innovative Research and Advancement (C-MiRA), Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan
| |
Collapse
|
6
|
Abd El-Karim SS, Syam YM, El Kerdawy AM, Abdel-Mohsen HT. Rational design and synthesis of novel quinazolinone N-acetohydrazides as type II multi-kinase inhibitors and potential anticancer agents. Bioorg Chem 2024; 142:106920. [PMID: 37898082 DOI: 10.1016/j.bioorg.2023.106920] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/10/2023] [Accepted: 10/14/2023] [Indexed: 10/30/2023]
Abstract
In the current investigation, a new class of quinazolinone N-acetohydrazides 9a-v was designed as type II multi-kinase inhibitors. The target quinazolinones were tailored so that the quinazolinone moiety would occupy the front pocket of the binding sites of VEGFR-2, FGFR-1 and BRAF kinases, meanwhile, the phenyl group at position 2 would act as a spacer which was functionalized at position 4 with an N-acetohydrazide linker that could achieve the key interactions with the essential gate area amino acids. The hydrazide moiety was linked to diverse aryl derivatives to occupy the hydrophobic back pocket of the DFG-out conformation of target kinases. The synthesized quinazolinone derivatives 9a-v demonstrated moderate to potent VEGFR-2 inhibitory activity with IC50 spanning from 0.29 to 5.17 µM. Further evaluation of the most potent derivatives on FGFR-1, BRAFWT and BRAFV600E showed that the quinazolinone N-acetohydrazides 9d, 9e, 9f, 9l and 9m have a potent multi-kinase inhibitory activity. Concurrently, 9b, 9d, 9e, 9k, 9l, 9o, 9q demonstrated potent growth inhibitory activity on NCI cancer cell lines with GI50 reaching 0.72 µM. In addition, compound 9e arrested the cell cycle progression in MDA-MB-231 cell line at the G2/M phase and showed the ability to induce apoptosis.
Collapse
Affiliation(s)
- Somaia S Abd El-Karim
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre, P. O. Box 12622 El-Bohouth Street, Dokki, Cairo, Egypt
| | - Yasmin M Syam
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre, P. O. Box 12622 El-Bohouth Street, Dokki, Cairo, Egypt
| | - Ahmed M El Kerdawy
- School of Pharmacy, College of Health and Science, University of Lincoln, Joseph Banks Laboratories, Green Lane, Lincoln, Lincolnshire, United Kingdom; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, P.O. Box 11562, Cairo, Egypt
| | - Heba T Abdel-Mohsen
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, P. O. Box 12622 El-Bohouth Street, Dokki, Cairo, Egypt.
| |
Collapse
|
7
|
Lee K, Lee S, Jung S, Chin HS. Analysis of ocular fluid in patients with ranibizumab-recalcitrant neovascular age-related macular degeneration who have serum anti-ranibizumab antibodies. Graefes Arch Clin Exp Ophthalmol 2023; 261:3581-3587. [PMID: 37318582 DOI: 10.1007/s00417-023-06146-6] [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: 01/10/2023] [Revised: 05/22/2023] [Accepted: 06/06/2023] [Indexed: 06/16/2023] Open
Abstract
PURPOSE To evaluate whether anti-drug antibodies (ADAs) are present in the ocular fluid of patients with ranibizumab-recalcitrant neovascular age-related macular degeneration (nAMD). METHODS Two serum ADA-positive ranibizumab-recalcitrant patients and two serum ADA-negative controls were recruited from patients with nAMD treated with ranibizumab monotherapy. Recalcitrance was defined as persistent fluid after ≥6 monthly ranibizumab injections. Serum and aqueous humor ADAs were detected by enzyme-linked immunosorbent assay and immunoprecipitation, respectively. RESULTS Two of 156 ranibizumab-treated patients were ADA-positive. The patients received six and 14 ranibizumab injections, respectively, up to 4 weeks prior to blood collection. The serum ADA concentration was estimated to be approximately 50,000 ng/mL. Neutralizing ADAs were confirmed in both samples. A specific band was detected by immunoprecipitation only in ADA-positive samples, consistent with the results of enzyme-linked immunosorbent assay. Based on an assessment of the degree of sensitivity of commercially available anti-ranibizumab antibodies, it was estimated that the immunoprecipitation method could detect ADA levels >30 ng. Nevertheless, ADAs were not detected in the aqueous humor of either the experimental or control group. CONCLUSION In the aqueous humor, ADAs are either not present or are present at a lower concentration than that which can be detected by immunoprecipitation. This presumably reflects the fact that blood ADA is the product of systemic circulation clearance through anterior elimination of intravitreal ranibizumab. Based on our results, ADAs do not return to the eye in sufficient quantities to interfere with the action of ranibizumab in the vitreous cavity.
Collapse
Affiliation(s)
- Kanghoon Lee
- Department of Ophthalmology, Inha University School of Medicine, 27 Inhang-ro, Jung-gu, 22332, Incheon, Republic of Korea
| | - Soyoung Lee
- Department of Ophthalmology, Inha University School of Medicine, 27 Inhang-ro, Jung-gu, 22332, Incheon, Republic of Korea
| | - Soyeon Jung
- Department of Ophthalmology, Inha University School of Medicine, 27 Inhang-ro, Jung-gu, 22332, Incheon, Republic of Korea
| | - Hee Seung Chin
- Department of Ophthalmology, Inha University School of Medicine, 27 Inhang-ro, Jung-gu, 22332, Incheon, Republic of Korea.
| |
Collapse
|
8
|
Li MY, Gao RP, Zhu Q, Chen Y, Tao BB, Zhu YC. Skeletal muscle-derived FSTL1 starting up angiogenesis by regulating endothelial junction via activating Src pathway can be upregulated by hydrogen sulfide. Am J Physiol Cell Physiol 2023; 325:C1252-C1266. [PMID: 37694287 DOI: 10.1152/ajpcell.00219.2023] [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: 05/18/2023] [Revised: 08/30/2023] [Accepted: 09/03/2023] [Indexed: 09/12/2023]
Abstract
Hydrogen sulfide (H2S) promotes microangiogenesis and revascularization after ischemia. Neovascularization starts with the destruction of intercellular junctions and is accompanied by various endothelial cell angiogenic behaviors. Follistatin-like 1 (FSTL1) is a cardiovascular-protective myokine that works against ischemic injury. The present study examined whether FSTL1 was involved in H2S-induced angiogenesis and explored the underlying molecular mechanism. We observed that H2S accelerated blood perfusion after ischemia in the mouse hindlimb ischemia model. Western blot analysis showed that H2S stabilized FSTL1 transcript and increased FSTL1 and Human antigen R (HuR) levels in skeletal muscle. RNA-interference HuR significantly inhibited the H2S-promoted increase in FSTL1 levels. Exogenous FSTL1 promoted the wound-healing migration of human umbilical vein endothelial cells (HUVECs) and increased monolayer endothelial barrier permeability. Immunostaining showed that FSTL1 increased interendothelial gap formation and decreased VE-Cadherin, Occludin, Connexin-43, and Claudin-5 expression. In addition, FSTL1 significantly increased the phosphorylation of Src and VEGFR2. However, the Src inhibitor, not the VEGFR2 inhibitor, could block FSTL1-induced effects in angiogenesis. In conclusion, we demonstrated that H2S could upregulate the expression of FSTL1 by increasing the HuR levels in skeletal muscle, and paracrine FSTL1 could initiate angiogenesis by opening intercellular junctions via the Src signaling pathway.NEW & NOTEWORTHY The myocyte-derived paracrine protein FSTL1 acts on vascular endothelial cells and initiates the process of angiogenesis by opening the intercellular junction via activating Src kinase. H2S can significantly upregulate FSTL1 protein levels in skeletal muscles by increasing HuR expression.
Collapse
Affiliation(s)
- Meng-Yao Li
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ru-Pan Gao
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qi Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ying Chen
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Bei-Bei Tao
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yi-Chun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Han N, Zhang L, Guo M, Yu L. Knockdown of Krüppel-Like Factor 9 Inhibits Aberrant Retinal Angiogenesis and Mitigates Proliferative Diabetic Retinopathy. Mol Biotechnol 2023; 65:612-623. [PMID: 36109428 DOI: 10.1007/s12033-022-00559-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 09/05/2022] [Indexed: 10/14/2022]
Abstract
Advanced proliferative diabetic retinopathy (PDR) characterized by aberrant retinal angiogenesis is a leading cause of retinal detachment and blindness. Krüppel-like factor 9 (KLF9), a member of the zinc-finger family of transcription factors, participates in the development of diabetic nephropathy and the promotion of angiogenesis of human umbilical vein endothelial cells. Therefore, we speculate that KLF9 may exert a crucial role in PDR. The current study revealed that KLF9 was highly expressed in the high glucose (HG)-treated human retinal microvascular endothelial cells (HRMECs) and the retinas of oxygen-induced retinopathy (OIR) rats. Knockdown of KLF9 inhibited the proliferation, migratory capability, invasiveness and tube formation of HG-treated HRMECs. Besides, knockdown of KLF9 decreased the expression of yes-associated protein 1 (YAP1) in HG-treated HRMECs. Dual-luciferase reporter assays confirmed that KLF9 transcriptionally upregulated YAP1 expression. Overexpression of YAP1 reversed the KLF9 silencing-induced repression of HRMEC proliferation and tube formation. Further in vivo evidence demonstrated that knockdown of KLF9 reduced the expression of Ki67, CD31 and vascular endothelial growth factor A (VEGFA) in the retinas of OIR rats. Collectively, KLF9 silencing might mitigate the progression of PDR by inhibiting angiogenesis via blocking YAP1 transcription.
Collapse
Affiliation(s)
- Ning Han
- Department of Ophthalmology, The Second Hospital of Jilin University, Nanguan District, No.218, Ziqiang Street, Changchun, Jilin, China
| | - Lihong Zhang
- Department of Ophthalmology, Songyuan Derun Tongxin Hospital, Songyuan, Jilin, China
| | - Mi Guo
- Department of Ophthalmology, Baotou Eye Hospital, Baotou, Inner Mongolia Autonomous Region, China
| | - Li Yu
- Department of Ophthalmology, The Second Hospital of Jilin University, Nanguan District, No.218, Ziqiang Street, Changchun, Jilin, China.
| |
Collapse
|
10
|
Zheng F, Wang J, Wang D, Yang Q. Clinical Application of Small Extracellular Vesicles in Gynecologic Malignancy Treatments. Cancers (Basel) 2023; 15:cancers15071984. [PMID: 37046644 PMCID: PMC10093031 DOI: 10.3390/cancers15071984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Small extracellular vesicles (sEVs) are the key mediators of intercellular communication. They have the potential for clinical use as diagnostic or therapeutic biomarkers and have been explored as vectors for drug delivery. Identification of reliable and noninvasive biomarkers, such as sEVs, is important for early diagnosis and precise treatment of gynecologic diseases to improve patient prognosis. Previous reviews have summarized routine sEVs isolation and identification methods; however, novel and unconventional methods have not been comprehensively described. This review summarizes a convenient method of isolating sEVs from body fluids and liquid biopsy-related sEV markers for early, minimally invasive diagnosis of gynecologic diseases. In addition, the characteristics of sEVs as drug carriers and in precision treatment and drug resistance are introduced, providing a strong foundation for identifying novel and potential therapeutic targets for sEV therapy. We propose potential directions for further research on the applications of sEVs in the diagnosis and treatment of gynecologic diseases.
Collapse
|
11
|
Shao M, Zhong MC, Wang Z, Ke Z, Zhong Z, Zhou J. Non-Invasive Dynamic Reperfusion of Microvessels In Vivo Controlled by Optical Tweezers. Front Bioeng Biotechnol 2022; 10:952537. [PMID: 35910027 PMCID: PMC9331193 DOI: 10.3389/fbioe.2022.952537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Distributive shock is considered to be a condition of microvascular hypoperfusion, which can be fatal in severe cases. However, traditional therapeutic methods to restore the macro blood flow are difficult to accurately control the blood perfusion of microvessels, and the currently developed manipulation techniques are inevitably incompatible with biological systems. In our approach, infrared optical tweezers are used to dynamically control the microvascular reperfusion within subdermal capillaries in the pinna of mice. Furthermore, we estimate the effect of different optical trap positions on reperfusion at branch and investigate the effect of the laser power on reperfusion. The results demonstrate the ability of optical tweezers to control microvascular reperfusion. This strategy allows near-noninvasive reperfusion of the microvascular hypoperfusion in vivo. Hence, our work is expected to provide unprecedented insights into the treatment of distributive shock.
Collapse
Affiliation(s)
- Meng Shao
- Anhui Province Key Laboratory of Measuring Theory and Precision Instrument, School of Instrument Science and Optoelectronics Engineering, Hefei University of Technology, Hefei, China
| | - Min-Cheng Zhong
- Anhui Province Key Laboratory of Measuring Theory and Precision Instrument, School of Instrument Science and Optoelectronics Engineering, Hefei University of Technology, Hefei, China
- *Correspondence: Min-Cheng Zhong, ; Jinhua Zhou,
| | - Zixin Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei, China
| | - Zeyu Ke
- School of Biomedical Engineering, Anhui Medical University, Hefei, China
| | - Zhensheng Zhong
- School of Biomedical Engineering, Anhui Medical University, Hefei, China
| | - Jinhua Zhou
- School of Biomedical Engineering, Anhui Medical University, Hefei, China
- *Correspondence: Min-Cheng Zhong, ; Jinhua Zhou,
| |
Collapse
|
12
|
Osude C, Lin L, Patel M, Eckburg A, Berei J, Kuckovic A, Dube N, Rastogi A, Gautam S, Smith TJ, Sreenivassappa SB, Puri N. Mediating EGFR-TKI Resistance by VEGF/VEGFR Autocrine Pathway in Non-Small Cell Lung Cancer. Cells 2022; 11:1694. [PMID: 35626731 PMCID: PMC9139342 DOI: 10.3390/cells11101694] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 01/06/2022] [Accepted: 05/17/2022] [Indexed: 02/01/2023] Open
Abstract
NSCLC treatment includes targeting of EGFR with tyrosine kinase inhibitors (TKIs) such as Erlotinib; however, resistance to TKIs is commonly acquired through T790M EGFR mutations or overexpression of vascular endothelial growth factor receptor-2 (VEGFR-2). We investigated the mechanisms of EGFR-TKI resistance in NSCLC cell lines with EGFR mutations or acquired resistance to Erlotinib. These studies showed upregulated gene and protein expression of VEGF, VEGFR-2, and a VEGF co-receptor neuropilin-1 (NP-1) in Erlotinib-resistant (1.4-5.3-fold) and EGFR double-mutant (L858R and T790M; 4.1-8.3-fold) NSCLC cells compared to parental and EGFR single-mutant (L858R) NSCLC cell lines, respectively. Immunofluorescence and FACS analysis revealed increased expression of VEGFR-2 and NP-1 in EGFR-TKI-resistant cell lines compared to TKI-sensitive cell lines. Cell proliferation assays showed that treatment with a VEGFR-2 inhibitor combined with Erlotinib lowered cell survival in EGFR double-mutant NSCLC cells to 9% compared to 72% after treatment with Erlotinib alone. Furthermore, Kaplan-Meier analysis revealed shorter median survival in late-stage NSCLC patients with high vs. low VEGFR-2 expression (14 mos vs. 21 mos). The results indicate that VEGFR-2 may play a key role in EGFR-TKI resistance and that combined treatment of Erlotinib with a VEGFR-2 inhibitor may serve as an effective therapy in NSCLC patients with EGFR mutations.
Collapse
Affiliation(s)
- Chike Osude
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, USA; (C.O.); (L.L.); (M.P.); (A.E.); (J.B.); (A.K.); (N.D.); (A.R.); (S.G.)
| | - Leo Lin
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, USA; (C.O.); (L.L.); (M.P.); (A.E.); (J.B.); (A.K.); (N.D.); (A.R.); (S.G.)
| | - Meet Patel
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, USA; (C.O.); (L.L.); (M.P.); (A.E.); (J.B.); (A.K.); (N.D.); (A.R.); (S.G.)
| | - Adam Eckburg
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, USA; (C.O.); (L.L.); (M.P.); (A.E.); (J.B.); (A.K.); (N.D.); (A.R.); (S.G.)
| | - Joseph Berei
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, USA; (C.O.); (L.L.); (M.P.); (A.E.); (J.B.); (A.K.); (N.D.); (A.R.); (S.G.)
| | - Adijan Kuckovic
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, USA; (C.O.); (L.L.); (M.P.); (A.E.); (J.B.); (A.K.); (N.D.); (A.R.); (S.G.)
| | - Namrata Dube
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, USA; (C.O.); (L.L.); (M.P.); (A.E.); (J.B.); (A.K.); (N.D.); (A.R.); (S.G.)
| | - Aayush Rastogi
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, USA; (C.O.); (L.L.); (M.P.); (A.E.); (J.B.); (A.K.); (N.D.); (A.R.); (S.G.)
| | - Shruti Gautam
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, USA; (C.O.); (L.L.); (M.P.); (A.E.); (J.B.); (A.K.); (N.D.); (A.R.); (S.G.)
| | - Thomas J. Smith
- College of Education, Northern Illinois University, Dekalb, IL 60115, USA;
| | | | - Neelu Puri
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, USA; (C.O.); (L.L.); (M.P.); (A.E.); (J.B.); (A.K.); (N.D.); (A.R.); (S.G.)
| |
Collapse
|
13
|
Peptide Inhibitors of Vascular Endothelial Growth Factor A: Current Situation and Perspectives. Pharmaceutics 2021; 13:pharmaceutics13091337. [PMID: 34575413 PMCID: PMC8467741 DOI: 10.3390/pharmaceutics13091337] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/18/2021] [Accepted: 08/24/2021] [Indexed: 12/11/2022] Open
Abstract
Vascular endothelial growth factors (VEGFs) are the family of extracellular signaling proteins involved in the processes of angiogenesis. VEGFA overexpression and altered regulation of VEGFA signaling pathways lead to pathological angiogenesis, which contributes to the progression of various diseases, such as age-related macular degeneration and cancer. Monoclonal antibodies and decoy receptors have been extensively used in the anti-angiogenic therapies for the neutralization of VEGFA. However, multiple side effects, solubility and aggregation issues, and the involvement of compensatory VEGFA-independent pro-angiogenic mechanisms limit the use of the existing VEGFA inhibitors. Short chemically synthesized VEGFA binding peptides are a promising alternative to these full-length proteins. In this review, we summarize anti-VEGFA peptides identified so far and discuss the molecular basis of their inhibitory activity to highlight their pharmacological potential as anti-angiogenic drugs.
Collapse
|
14
|
Huang M, Chen M, Qi M, Ye G, Pan J, Shi C, Yang Y, Zhao L, Mo X, Zhang Y, Li Y, Zhong J, Lu W, Li X, Zhang J, Lin J, Luo L, Liu T, Tang PM, Hong A, Cao Y, Ye W, Zhang D. Perivascular cell-derived extracellular vesicles stimulate colorectal cancer revascularization after withdrawal of antiangiogenic drugs. J Extracell Vesicles 2021; 10:e12096. [PMID: 34035882 PMCID: PMC8138700 DOI: 10.1002/jev2.12096] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 04/02/2021] [Accepted: 05/04/2021] [Indexed: 12/22/2022] Open
Abstract
Antiangiogenic tyrosine kinase inhibitors (AA-TKIs) have become a promising therapeutic strategy for colorectal cancer (CRC). In clinical practice, a significant proportion of cancer patients temporarily discontinue AA-TKI treatment due to recurrent toxicities, economic burden or acquired resistance. However, AA-TKI therapy withdrawal-induced tumour revascularization frequently occurs, hampering the clinical application of AA-TKIs. Here, this study demonstrates that tumour perivascular cells mediate tumour revascularization after withdrawal of AA-TKI therapy. Pharmacological inhibition and genetic ablation of perivascular cells largely attenuate the rebound effect of CRC vascularization in the AA-TKI cessation experimental settings. Mechanistically, tumour perivascular cell-derived extracellular vehicles (TPC-EVs) contain Gas6 that instigates the recruitment of endothelial progenitor cells (EPCs) for tumour revascularization via activating the Axl pathway. Gas6 silence and an Axl inhibitor markedly inhibit tumour revascularization by impairing EPC recruitment. Consequently, combination therapy of regorafenib with the Axl inhibitor improves overall survival in mice metastatic CRC model by inhibiting tumour growth. Together, these data shed new mechanistic insights into perivascular cells in off-AA-TKI-induced tumour revascularization and indicate that blocking the Axl signalling may provide an attractive anticancer approach for sustaining long-lasting angiostatic effects to improve the therapeutic outcomes of antiangiogenic drugs in CRC.
Collapse
Affiliation(s)
- Maohua Huang
- College of PharmacyJinan UniversityGuangzhouChina
| | - Minfeng Chen
- College of PharmacyJinan UniversityGuangzhouChina
| | - Ming Qi
- College of PharmacyJinan UniversityGuangzhouChina
| | - Geni Ye
- College of PharmacyJinan UniversityGuangzhouChina
| | - Jinghua Pan
- Department of General Surgerythe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Changzheng Shi
- Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translationthe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Yunlong Yang
- Department of Cellular and Genetic MedicineSchool of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Luyu Zhao
- Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translationthe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Xukai Mo
- Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translationthe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Yiran Zhang
- Department of General Surgerythe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Yong Li
- College of PharmacyJinan UniversityGuangzhouChina
| | | | - Weijin Lu
- College of PharmacyJinan UniversityGuangzhouChina
| | - Xiaobo Li
- College of PharmacyJinan UniversityGuangzhouChina
| | - Jiayan Zhang
- College of PharmacyJinan UniversityGuangzhouChina
| | - Jinrong Lin
- Department of Obstetricsthe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Liangping Luo
- Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translationthe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | | | - Patrick Ming‐Kuen Tang
- Department of Anatomical and Cellular PathologyPrince of Wales HospitalThe Chinese University of Hong KongSha TinHong Kong
| | - An Hong
- Department of Cell BiologyJinan UniversityGuangzhouChina
| | - Yihai Cao
- Department of MicrobiologyTumor and Cell BiologyKarolinska InstituteStockholmSweden
| | - Wencai Ye
- College of PharmacyJinan UniversityGuangzhouChina
| | | |
Collapse
|
15
|
Xiao H, Wang K, Li D, Wang K, Yu M. Evaluation of FGFR1 as a diagnostic biomarker for ovarian cancer using TCGA and GEO datasets. PeerJ 2021; 9:e10817. [PMID: 33604191 PMCID: PMC7866899 DOI: 10.7717/peerj.10817] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 12/30/2020] [Indexed: 12/30/2022] Open
Abstract
Background Malignant ovarian cancer is associated with the highest mortality of all gynecological tumors. Designing therapeutic targets that are specific to OC tissue is important for optimizing OC therapies. This study aims to identify different expression patterns of genes related to FGFR1 and the usefulness of FGFR1 as diagnostic biomarker for OC. Methods We collected data from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases. In the TCGA cohort we analyzed clinical information according to patient characteristics, including age, stage, grade, longest dimension of the tumor and the presence of a residual tumor. GEO data served as a validation set. We obtained data on differentially expressed genes (DEGs) from the two microarray datasets. We then used gene set enrichment analysis (GSEA) to analyze the DEG data in order to identify enriched pathways related to FGFR1. Results Differential expression analysis revealed that FGFR1 was significantly downregulated in OC specimens. 303 patients were included in the TCGA cohort. The GEO dataset confirmed these findings using information on 75 Asian patients. The GSE105437 and GSE12470 database highlighted the significant diagnostic value of FGFR1 in identifying OC (AUC = 1, p = 0.0009 and AUC = 0.8256, p = 0.0015 respectively). Conclusions Our study examined existing TCGA and GEO datasets for novel factors associated with OC and identified FGFR1 as a potential diagnostic factor. Further investigation is warranted to characterize the role played by FGFR1 in OC.
Collapse
Affiliation(s)
- Huiting Xiao
- Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Kun Wang
- Department of Urologic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Dan Li
- Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Ke Wang
- Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Min Yu
- Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
16
|
Prognostic and Therapeutic Utility of Variably Expressed Cell Surface Receptors in Osteosarcoma. Sarcoma 2021; 2021:8324348. [PMID: 33603563 PMCID: PMC7872755 DOI: 10.1155/2021/8324348] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/17/2020] [Accepted: 01/07/2021] [Indexed: 01/05/2023] Open
Abstract
Background Six cell surface receptors, human epidermal growth factor receptor-2 (Her-2), platelet-derived growth factor receptor-β (PDGFR-β), insulin-like growth factor-1 receptor (IGF-1R), insulin receptor (IR), c-Met, and vascular endothelial growth factor receptor-3 (VEGFR-3), previously demonstrated variable expression across varying patient-derived and standard osteosarcoma (OS) cell lines. The current study sought to validate previous expression patterns and evaluate whether these receptors offer prognostic and/or therapeutic value. Methods Patient-derived OS cell lines (n = 52) were labeled with antibodies to Her-2, PDGFR-β, IGF-1R, IR, c-Met, and VEGFR-3. Expression was characterized using flow cytometry. The difference in geometric mean fluorescent intensity (geoMFIdiff = geoMFIpositive - geoMFInegative) was calculated for each receptor across all cell lines. Receptor expression was categorized as low (Q1), intermediate (Q2, Q3), or high (Q4). The event-free survival (EFS) and overall survival for the six cell surface receptors were estimated by the Kaplan-Meier method. Differences in hazard for EFS event and overall survival event for patients in each of the three expression levels in each of the six cell surface receptors were assessed using the log-rank test. Results All 6 receptors were variably expressed in the majority of cell lines. IR and PDGFR-β expressions were found to be significant predictors for EFS amongst patients with nonmetastatic disease (p=0.02 and 0.01, respectively). The hazard ratio for EFS was significantly higher between high IR and intermediate IR expression (HR = 2.66, p=0.02), as well as between high PDGFR-β and intermediate PDGFR-β expression (HR = 5.68, p=0.002). Her-2, c-Met, IGF-1R, and VEGFR-3 were not found to be significant predictors for either EFS or overall survival. Conclusion The six cell surface receptors demonstrated variable expression across the majority of patient-derived OS cell lines tested. Limited prognostic value was offered by IR and PDGFR-β expression within nonmetastatic patients. The remaining receptors do not provide clear prognostic utility. Nevertheless, their consistent, albeit variable, surface expression across a large panel of patient-derived OS cell lines maintains their potential use as future therapeutic targets.
Collapse
|
17
|
Ruzzo A, Graziano F, Bagaloni I, Di Bartolomeo M, Prisciandaro M, Aprile G, Ongaro E, Vincenzi B, Perrone G, Santini D, Fornaro L, Vivaldi C, Tomasello G, Loupakis F, Lonardi S, Fassan M, Valmasoni M, Sarti D, Lorenzini P, Catalano V, Bisonni R, Del Prete M, Collina G, Magnani M. Glycolytic competence in gastric adenocarcinomas negatively impacts survival outcomes of patients treated with salvage paclitaxel-ramucirumab. Gastric Cancer 2020; 23:1064-1074. [PMID: 32372141 PMCID: PMC7567716 DOI: 10.1007/s10120-020-01078-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/23/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION For energy production, cancer cells maintain a high rate of glycolysis instead of oxidative phosphorylation converting glucose into lactic acid. This metabolic shift is useful to survive in unfavorable microenvironments. We investigated whether a positive glycolytic profile (PGP) in gastric adenocarcinomas may be associated with unfavorable outcomes under an anticancer systemic therapy, including the anti-angiogenic ramucirumab. MATERIALS AND METHODS Normal mucosa (NM) and primary tumor (PT) of 40 metastatic gastric adenocarcinomas patients who received second-line paclitaxel-ramucirumab (PR) were analyzed for mRNA expression of the following genes: HK-1, HK-2, PKM-2, LDH-A, and GLUT-1. Patients were categorized with PGP when at least a doubling of mRNA expression (PT vs. NM) in all glycolytic core enzymes (HK-1 or HK-2, PKM-2, LDH-A) was observed. PGP was also related to TP53 mutational status. RESULTS Mean LDH-A, HK-2, PKM-2 mRNA expression levels were significantly higher in PT compared with NM. 18 patients were classified as PGP, which was associated with significantly worse progression-free and overall survival times. No significant association was observed between PGP and clinical-pathologic features, including TP53 positive mutational status, in 28 samples. CONCLUSIONS Glycolytic proficiency may negatively affect survival outcomes of metastatic gastric cancer patients treated with PR systemic therapy. TP53 mutational status alone does not seem to explain such a metabolic shift.
Collapse
Affiliation(s)
- Annamaria Ruzzo
- Department of Biomolecular Sciences (DiSB), University of Urbino "Carlo Bo", Via Arco d'Augusto, 2, 61032, Fano, PU, Italy.
| | - Francesco Graziano
- Department of Onco-Hematology, Division of Oncology, Azienda Ospedaliera "Ospedali Riuniti Marche Nord", 61122, Pesaro, Italy.
| | - Irene Bagaloni
- Department of Biomolecular Sciences (DiSB), University of Urbino "Carlo Bo", Via Arco d'Augusto, 2, 61032, Fano, PU, Italy
| | | | | | - Giuseppe Aprile
- Department of Medical Oncology, San Bortolo General Hospital, AULSS8 Berica, Vicenza, Italy
| | - Elena Ongaro
- Department of Oncology, University and General Hospital, Udine, Italy
| | | | | | | | | | | | | | - Fotios Loupakis
- Department of Oncology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Sara Lonardi
- Department of Oncology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Michele Valmasoni
- Clinica Chirurgica 3, Department of Surgical, Oncological and Gastroenterological Sciences (DISCOG), University of Padua, Padua, Italy
| | - Donatella Sarti
- Department of Onco-Hematology, Division of Oncology, Azienda Ospedaliera "Ospedali Riuniti Marche Nord", 61122, Pesaro, Italy
| | - Paola Lorenzini
- Department of Onco-Hematology, Division of Oncology, Azienda Ospedaliera "Ospedali Riuniti Marche Nord", 61122, Pesaro, Italy
| | - Vincenzo Catalano
- Department of Onco-Hematology, Division of Oncology, Azienda Ospedaliera "Ospedali Riuniti Marche Nord", 61122, Pesaro, Italy
| | | | | | - Guido Collina
- Area vasta 5, Ospedale "C. e G. Mazzoni" Ascoli Piceno, Ascoli Piceno, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences (DiSB), University of Urbino "Carlo Bo", Via Arco d'Augusto, 2, 61032, Fano, PU, Italy
| |
Collapse
|
18
|
Modi SJ, Kulkarni VM. Discovery of VEGFR-2 inhibitors exerting significant anticancer activity against CD44+ and CD133+ cancer stem cells (CSCs): Reversal of TGF-β induced epithelial-mesenchymal transition (EMT) in hepatocellular carcinoma. Eur J Med Chem 2020; 207:112851. [PMID: 33002846 DOI: 10.1016/j.ejmech.2020.112851] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/02/2020] [Accepted: 09/13/2020] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) is a malignancy characterized by neoangiogenesis, which is an augmented production of proangiogenic factors by the tumor and its adjacent infected cells. These dysregulated angiogenic factors are the therapeutic targets in anti-angiogenic drug development. The signaling pathway of vascular endothelial growth factor (VEGF)/VEGFR-2 is crucial for controlling the angiogenic responses in endothelial cells (ECs). In this study, we carried out a rational drug design approach wherein we have identified the novel orally bioavailable compound VS 8 as a potent VEGFR-2 inhibitor, which remarkably suppresses hVEGF and hVEGFR-2 expression in HUVECs and exhibits significant anti-angiogenic effects in CAM assay. Besides, VS 8 significantly induces apoptosis in HCC cell line (Hep G2). Later we examined its effectiveness against CD44+ and CD133+ CSCs. Here, VS 8 was found to be active against CSCs, and adequate for the cessation of the cell cycle at 'G0/G1' and 'S' phase in CD44+ and CD133+ CSCs respectively. Factually, transforming growth factor-β (TGF-β) stimulated epithelial-mesenchymal transition (EMT) induces invasion and migration of HCC cells, which results in the metastasis. Therefore, we studied the effect of VS 8 on EMT markers using flow cytometry, which suggested that VS 8 significantly upregulates E-cadherin (epithelial biomarker) and downregulates vimentin (mesenchymal biomarker). Further, VS 8 downregulates the expression of EMT-inducing transcription factors (EMT-TFs), i.e., SNAIL. Altogether, our findings indicate that VS 8 could be a promising drug candidate for cancer therapy.
Collapse
Affiliation(s)
- Siddharth J Modi
- Department of Pharmaceutical Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be University), Pune, 411038, Maharashtra, India
| | - Vithal M Kulkarni
- Department of Pharmaceutical Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be University), Pune, 411038, Maharashtra, India.
| |
Collapse
|
19
|
Liu X, Gao Q, Zhang Y, Li Y, Li B. In Vivo Optofluidic Switch for Controlling Blood Microflow. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001414. [PMID: 32714772 PMCID: PMC7375249 DOI: 10.1002/advs.202001414] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/12/2020] [Indexed: 05/13/2023]
Abstract
Control of blood microflow is crucial for the prevention and therapy of blood disorders, such as cardiovascular diseases and their complications. Conventional control strategies generally implant exogenous synthetic materials into blood vessels as labeling markers or actuating sources, which are invasive and incompatible with biological systems. Here, a label-free, noninvasive, and biocompatible device constructed from natural red blood cells (RBCs) for controlling blood microflow in vivo is reported. The RBCs, optically manipulated, arranged, and rotated using scanning optical tweezers, can function as an optofluidic switch for targeted switching, directional enrichment, dynamic redirecting, and rotary actuation of blood microflow inside zebrafish. The regulation precision of the switch is determined to be at the single-cell level, and the response time is measured as ≈200 ms using a streamline tracking method. This in vivo optofluidic switch may provide a biofriendly device for exploring blood microenvironments in a noncontact and noninvasive manner.
Collapse
Affiliation(s)
- Xiaoshuai Liu
- Institute of NanophotonicsJinan UniversityGuangzhou511‐443China
| | - Qing Gao
- Institute of NanophotonicsJinan UniversityGuangzhou511‐443China
| | - Yao Zhang
- Institute of NanophotonicsJinan UniversityGuangzhou511‐443China
| | - Yuchao Li
- Institute of NanophotonicsJinan UniversityGuangzhou511‐443China
| | - Baojun Li
- Institute of NanophotonicsJinan UniversityGuangzhou511‐443China
| |
Collapse
|
20
|
Moradi A, Pourseif MM, Jafari B, Parvizpour S, Omidi Y. Nanobody-based therapeutics against colorectal cancer: Precision therapies based on the personal mutanome profile and tumor neoantigens. Pharmacol Res 2020; 156:104790. [DOI: 10.1016/j.phrs.2020.104790] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/07/2020] [Accepted: 03/31/2020] [Indexed: 12/19/2022]
|
21
|
Drug resistance in papillary RCC: from putative mechanisms to clinical practicalities. Nat Rev Urol 2019; 16:655-673. [PMID: 31602010 DOI: 10.1038/s41585-019-0233-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2019] [Indexed: 11/08/2022]
Abstract
Papillary renal cell carcinoma (pRCC) is the second most common renal cell carcinoma (RCC) subtype and accounts for 10-15% of all RCCs. Despite clinical need, few pharmacogenomics studies in pRCC have been performed. Moreover, current research fails to adequately include pRCC laboratory models, such as the ACHN or Caki-2 pRCC cell lines. The molecular mechanisms involved in pRCC development and drug resistance are more diverse than in clear-cell RCC, in which inactivation of VHL occurs in the majority of tumours. Drug resistance to multiple therapies in pRCC occurs via genetic alteration (such as mutations resulting in abnormal receptor tyrosine kinase activation or RALBP1 inhibition), dysregulation of signalling pathways (such as GSK3β-EIF4EBP1, PI3K-AKT and the MAPK or interleukin signalling pathways), deregulation of cellular processes (such as resistance to apoptosis or epithelial-to-mesenchymal transition) and interactions between the cell and its environment (for example, through activation of matrix metalloproteinases). Improved understanding of resistance mechanisms will facilitate drug discovery and provide new effective therapies. Further studies on novel resistance biomarkers are needed to improve patient prognosis and stratification as well as drug development.
Collapse
|
22
|
Vascular Endothelial Growth Factor Receptor (VEGFR-2)/KDR Inhibitors: Medicinal Chemistry Perspective. MEDICINE IN DRUG DISCOVERY 2019. [DOI: 10.1016/j.medidd.2019.100009] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
23
|
Dimova I, Karthik S, Makanya A, Hlushchuk R, Semela D, Volarevic V, Djonov V. SDF-1/CXCR4 signalling is involved in blood vessel growth and remodelling by intussusception. J Cell Mol Med 2019; 23:3916-3926. [PMID: 30950188 PMCID: PMC6533523 DOI: 10.1111/jcmm.14269] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 02/15/2019] [Accepted: 02/26/2019] [Indexed: 12/14/2022] Open
Abstract
The precise mechanisms of SDF‐1 (CXCL12) in angiogenesis are not fully elucidated. Recently, we showed that Notch inhibition induces extensive intussusceptive angiogenesis by recruitment of mononuclear cells and it was associated with increased levels of SDF‐1 and CXCR4. In the current study, we demonstrated SDF‐1 expression in liver sinusoidal vessels of Notch1 knockout mice with regenerative hyperplasia by means of intussusception, but we did not detect any SDF‐1 expression in wild‐type mice with normal liver vessel structure. In addition, pharmacological inhibition of SDF‐1/CXCR4 signalling by AMD3100 perturbs intussusceptive vascular growth and abolishes mononuclear cell recruitment in the chicken area vasculosa. In contrast, treatment with recombinant SDF‐1 protein increased microvascular density by 34% through augmentation of pillar number compared to controls. The number of extravasating mononuclear cells was four times higher after SDF‐1 application and two times less after blocking this pathway. Bone marrow‐derived mononuclear cells (BMDC) were recruited to vessels in response to elevated expression of SDF‐1 in endothelial cells. They participated in formation and stabilization of pillars. The current study is the first report to implicate SDF‐1/CXCR4 signalling in intussusceptive angiogenesis and further highlights the stabilizing role of BMDC in the formation of pillars during vascular remodelling.
Collapse
Affiliation(s)
- Ivanka Dimova
- Institute of Anatomy, University of Bern, Bern, Switzerland.,Center of Molecular Medicine, Medical University Sofia, Sofia, Bulgaria
| | - Swapna Karthik
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Andrew Makanya
- Institute of Anatomy, University of Bern, Bern, Switzerland.,Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya
| | | | - David Semela
- Liver Biology Laboratory, Medical Research Center, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Vladislav Volarevic
- Institute of Anatomy, University of Bern, Bern, Switzerland.,Center of Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | | |
Collapse
|
24
|
Soluble E-cadherin promotes tumor angiogenesis and localizes to exosome surface. Nat Commun 2018; 9:2270. [PMID: 29891938 PMCID: PMC5995921 DOI: 10.1038/s41467-018-04695-7] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 05/18/2018] [Indexed: 02/08/2023] Open
Abstract
The limitations of current anti-angiogenic therapies necessitate other targets with complimentary mechanisms. Here, we show for the first time that soluble E-cadherin (sE-cad) (an 80-kDa soluble form), which is highly expressed in the malignant ascites of ovarian cancer patients, is a potent inducer of angiogenesis. In addition to ectodomain shedding, we provide further evidence that sE-cad is abundantly released in the form of exosomes. Mechanistically, sE-cad-positive exosomes heterodimerize with VE-cadherin on endothelial cells and transduce a novel sequential activation of β-catenin and NFκB signaling. In vivo and clinical data prove the relevance of sE-cad-positive exosomes for malignant ascites formation and widespread peritoneal dissemination. These data advance our understanding of the molecular regulation of angiogenesis in ovarian cancer and support the therapeutic potential of targeting sE-cad. The exosomal release of sE-cad, which represents a common route for externalization in ovarian cancer, could potentially be biomarkers for diagnosis and prognosis. A soluble form E-cadherin is highly expressed in ovarian cancer. Here, the authors show that soluble E-cadherin is released by ovarian cancer cells packaged in exosomes and promotes tumor angiogenesis through β-catenin and NFkB signaling activation.
Collapse
|
25
|
Correlation of c-MET Expression with PD-L1 Expression in Metastatic Clear Cell Renal Cell Carcinoma Treated by Sunitinib First-Line Therapy. Target Oncol 2018; 12:487-494. [PMID: 28550387 DOI: 10.1007/s11523-017-0498-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is highly metastatic. Cabozantinib, an anti-angiogenic tyrosine kinase inhibitor that targets c-MET, provided interesting results in metastatic ccRCC treatment. OBJECTIVE To understand better the role of c-MET in ccRCC, we assessed its status in a population of patients with metastatic ccRCC. PATIENTS AND METHODS For this purpose, tumor samples were analyzed for c-MET expression by immunohistochemistry (IHC), for c-MET copy number alterations by fluorescence in situ hybridization (FISH), and for c-MET mutations by next generation sequencing (NGS) in a retrospective cohort of 90 primary ccRCC of patients with metastases treated by first-line sunitinib. The expression of c-MET was correlated with pathological, immunohistochemical (VEGFA, CAIX, PD-L1), clinical, and molecular criteria (VHL status) by univariate and multivariate analyses and to clinical outcome using Kaplan-Meier curves compared by log-rank test. RESULTS Of ccRCC, 31.1% had low c-MET expression (absent to weak intensity by IHC) versus 68.9% with high expression (moderate to strong intensity). High expression of c-MET was associated with a gain in FISH analysis (p=0.0284) without amplification. No mutations were detected in NGS. Moreover, high c-MET expression was associated with lymph node metastases (p=0.004), sarcomatoid component (p=0.029), VEGFA (p=0.037), and PD-L1 (p=0.001) overexpression, the only factor that remained independently associated (p<0.001) after logistic regression. No difference was observed in clinical outcomes. CONCLUSION This study is the first to analyse c-MET status in metastatic ccRCC. The high expression of c-MET in the majority of ccRCC and its independent association with PD-L1 expression, may suggest a potential benefit from combining c-MET inhibitors and targeted immunotherapy.
Collapse
|
26
|
Jiang J, Xu K, Wang L, Xin W, Zhao G, Huang M, Li S, Luan X, Fang J. Pharmacology study of a chimeric decoy receptor trap fusion protein on retina neovascularization by dual blockage of VEGF and FGF-2. Eur J Pharm Sci 2018; 121:251-259. [PMID: 29715501 DOI: 10.1016/j.ejps.2018.04.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/08/2017] [Accepted: 04/27/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Clinical anti-vascular epithelial growth factor (VEGF) therapy trials faced a major challenge due to upregulated expression of other pro-angiogenic factors, such as fibroblast growth factor-2 (FGF-2). RC28, a novel recombinant dual decoy receptor IgG1 Fc-fusion protein, can block VEGFA and FGF-2 simultaneously. It is designed for the treatment of neovascular age-related macular degeneration and other pathological ocular neovascularization. The present study investigated the prevention efficacy of RC28 on choroidal neovascularization (CNV) in a monkey model and compared to the other mono VEGF antagonists; biodistribution and pharmacokinetics performance were also investigated. METHODS ELISA and endothelial cell proliferation, migration, and tubule formation assay evaluated the bioactivity of RC28 in vitro, and an initial comparison was made among the mono target antagonists, Bevacizumab (Avastin), Ranibizumab (Lucentis), Aflibercept (EYLEA), Conbercept (KH902), and Ranibizumab (Lucentis). Laser-induced CNV in monkeys, and both VEGF and FGF-2 serum levels were detected in animals before and after the CNV model were induced. RC28 prevention efficacy was compared to other VEGF antagonists on CNV with respect to the incidence of CNV and several ophthalmic examinations. Ocular and systemic levels of RC28 were analyzed by 89Zr-labeled RC28 after single intravitreal administration for the biodistribution and pharmacokinetic profiles. RESULTS RC28 is a unique fusion protein with high affinity to both VEGF and FGF-2, and beneficial to in vitro and in vivo bioactivity. The in vivo pharmacological studies demonstrated that the incidence of CNV formation was largely reduced in RC28 treatment groups with a low dosage as compared to other VEGF antagonist control groups. Furthermore, traces of RC28 were detected as dispersing from eyeballs to the liver after 20 days, and a prolonged half-time pharmacokinetic profile was exhibited.
Collapse
Affiliation(s)
- Jing Jiang
- Department of Pharmacology, Binzhou Medical University, Yantai 256603, Shandong, China.
| | - Ke Xu
- Department of Pharmacology and Toxicology, Beijing, Institute of Radiation Medicine, Beijing 100850, China
| | - Ling Wang
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Wei Xin
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Guorui Zhao
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Min Huang
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Shenjun Li
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | | | - Jianmin Fang
- School of Life Science and Technology, Tongji University, Shanghai 200092, China
| |
Collapse
|
27
|
Bezabeh B, Fleming R, Fazenbaker C, Zhong H, Coffman K, Yu XQ, Leow CC, Gibson N, Wilson S, Stover CK, Wu H, Gao C, Dimasi N. Insertion of scFv into the hinge domain of full-length IgG1 monoclonal antibody results in tetravalent bispecific molecule with robust properties. MAbs 2017; 9:240-256. [PMID: 27981887 DOI: 10.1080/19420862.2016.1270492] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
By simultaneous binding two disease mediators, bispecific antibodies offer the opportunity to broaden the utility of antibody-based therapies. Herein, we describe the design and characterization of Bs4Ab, an innovative and generic bispecific tetravalent antibody platform. The Bs4Ab format comprises a full-length IgG1 monoclonal antibody with a scFv inserted into the hinge domain. The Bs4Ab design demonstrates robust manufacturability as evidenced by MEDI3902, which is currently in clinical development. To further demonstrate the applicability of the Bs4Ab technology, we describe the molecular engineering, biochemical, biophysical, and in vivo characterization of a bispecific tetravalent Bs4Ab that, by simultaneously binding vascular endothelial growth factor and angiopoietin-2, inhibits their function. We also demonstrate that the Bs4Ab platform allows Fc-engineering similar to that achieved with IgG1 antibodies, such as mutations to extend half-life or modulate effector functions.
Collapse
Affiliation(s)
- Binyam Bezabeh
- a Antibody Discovery and Protein Engineering , Gaithersburg , MA , USA
| | - Ryan Fleming
- a Antibody Discovery and Protein Engineering , Gaithersburg , MA , USA
| | | | | | - Karen Coffman
- c Clinical Pharmacology and DMPK , Gaithersburg , MA , USA
| | - Xiang-Qing Yu
- c Clinical Pharmacology and DMPK , Gaithersburg , MA , USA
| | | | - Nerea Gibson
- a Antibody Discovery and Protein Engineering , Gaithersburg , MA , USA
| | - Susan Wilson
- a Antibody Discovery and Protein Engineering , Gaithersburg , MA , USA
| | | | - Herren Wu
- a Antibody Discovery and Protein Engineering , Gaithersburg , MA , USA
| | - Changshou Gao
- a Antibody Discovery and Protein Engineering , Gaithersburg , MA , USA
| | - Nazzareno Dimasi
- a Antibody Discovery and Protein Engineering , Gaithersburg , MA , USA
| |
Collapse
|
28
|
Razpotnik R, Novak N, Čurin Šerbec V, Rajcevic U. Targeting Malignant Brain Tumors with Antibodies. Front Immunol 2017; 8:1181. [PMID: 28993773 PMCID: PMC5622144 DOI: 10.3389/fimmu.2017.01181] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 09/06/2017] [Indexed: 12/31/2022] Open
Abstract
Antibodies have been shown to be a potent therapeutic tool. However, their use for targeting brain diseases, including neurodegenerative diseases and brain cancers, has been limited, particularly because the blood–brain barrier (BBB) makes brain tissue hard to access by conventional antibody-targeting strategies. In this review, we summarize new antibody therapeutic approaches to target brain tumors, especially malignant gliomas, as well as their potential drawbacks. Many different brain delivery platforms for antibodies have been studied such as liposomes, nanoparticle-based systems, cell-penetrating peptides (CPPs), and cell-based approaches. We have already shown the successful delivery of single-chain fragment variable (scFv) with CPP as a linker between two variable domains in the brain. Antibodies normally face poor penetration through the BBB, with some variants sufficiently passing the barrier on their own. A “Trojan horse” method allows passage of biomolecules, such as antibodies, through the BBB by receptor-mediated transcytosis (RMT). Such examples of therapeutic antibodies are the bispecific antibodies where one binding specificity recognizes and binds a BBB receptor, enabling RMT and where a second binding specificity recognizes an antigen as a therapeutic target. On the other hand, cell-based systems such as stem cells (SCs) are a promising delivery system because of their tumor tropism and ability to cross the BBB. Genetically engineered SCs can be used in gene therapy, where they express anti-tumor drugs, including antibodies. Different types and sources of SCs have been studied for the delivery of therapeutics to the brain; both mesenchymal stem cells (MSCs) and neural stem cells (NSCs) show great potential. Following the success in treatment of leukemias and lymphomas, the adoptive T-cell therapies, especially the chimeric antigen receptor-T cells (CAR-Ts), are making their way into glioma treatment as another type of cell-based therapy using the antibody to bind to the specific target(s). Finally, the current clinical trials are reviewed, showing the most recent progress of attractive approaches to deliver therapeutic antibodies across the BBB aiming at the specific antigen.
Collapse
Affiliation(s)
- Rok Razpotnik
- Department of Research and Development, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Neža Novak
- Department of Research and Development, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Vladka Čurin Šerbec
- Department of Research and Development, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Uros Rajcevic
- Department of Research and Development, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| |
Collapse
|
29
|
Mirabelli P, Mukwaya A, Lennikov A, Xeroudaki M, Peebo B, Schaupper M, Lagali N. Genome-wide expression differences in anti-Vegf and dexamethasone treatment of inflammatory angiogenesis in the rat cornea. Sci Rep 2017; 7:7616. [PMID: 28811496 PMCID: PMC5557983 DOI: 10.1038/s41598-017-07129-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 06/22/2017] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis as a pathological process in the eye can lead to blindness. In the cornea, suppression of angiogenesis by anti-VEGF treatment is only partially effective while steroids, although effective in treating inflammation and angiogenesis, have broad activity leading to undesirable side effects. In this study, genome-wide expression was investigated in a suture-induced corneal neovascularization model in rats, to investigate factors differentially targeted by dexamethasone and anti-Vegf. Topical treatment with either rat-specific anti-Vegf, dexamethasone, or normal goat IgG (sham) was given to sutured corneas for 48 hours, after which in vivo imaging, tissue processing for RNA microarray, and immunofluorescence were performed. Dexamethasone suppressed limbal vasodilation (P < 0.01) and genes in PI3K-Akt, focal adhesion, and chemokine signaling pathways more effectively than anti-Vegf. The most differentially expressed genes were confirmed by immunofluorescence, qRTPCR and Western blot. Strong suppression of Reg3g and the inflammatory chemokines Ccl2 and Cxcl5 and activation of classical complement pathway factors C1r, C1s, C2, and C3 occurred with dexamethasone treatment, effects absent with anti-Vegf treatment. The genome-wide results obtained in this study provide numerous potential targets for specific blockade of inflammation and angiogenesis in the cornea not addressed by anti-Vegf treatment, as possible alternatives to broad-acting immunosuppressive therapy.
Collapse
Affiliation(s)
- Pierfrancesco Mirabelli
- Department of Ophthalmology, Institute for Clinical and Experimental Medicine,Faculty of Health Sciences, Linkoping University, 58183, Linköping, Sweden
| | - Anthony Mukwaya
- Department of Ophthalmology, Institute for Clinical and Experimental Medicine,Faculty of Health Sciences, Linkoping University, 58183, Linköping, Sweden
| | - Anton Lennikov
- Department of Ophthalmology, Institute for Clinical and Experimental Medicine,Faculty of Health Sciences, Linkoping University, 58183, Linköping, Sweden
| | - Maria Xeroudaki
- Department of Ophthalmology, Institute for Clinical and Experimental Medicine,Faculty of Health Sciences, Linkoping University, 58183, Linköping, Sweden
| | - Beatrice Peebo
- Department of Ophthalmology, Institute for Clinical and Experimental Medicine,Faculty of Health Sciences, Linkoping University, 58183, Linköping, Sweden
| | - Mira Schaupper
- Department of Ophthalmology, Institute for Clinical and Experimental Medicine,Faculty of Health Sciences, Linkoping University, 58183, Linköping, Sweden
| | - Neil Lagali
- Department of Ophthalmology, Institute for Clinical and Experimental Medicine,Faculty of Health Sciences, Linkoping University, 58183, Linköping, Sweden.
| |
Collapse
|
30
|
Lodola F, Laforenza U, Cattaneo F, Ruffinatti FA, Poletto V, Massa M, Tancredi R, Zuccolo E, Khdar DA, Riccardi A, Biggiogera M, Rosti V, Guerra G, Moccia F. VEGF-induced intracellular Ca 2+ oscillations are down-regulated and do not stimulate angiogenesis in breast cancer-derived endothelial colony forming cells. Oncotarget 2017; 8:95223-95246. [PMID: 29221123 PMCID: PMC5707017 DOI: 10.18632/oncotarget.20255] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/12/2017] [Indexed: 01/08/2023] Open
Abstract
Endothelial colony forming cells (ECFCs) represent a population of truly endothelial precursors that promote the angiogenic switch in solid tumors, such as breast cancer (BC). The intracellular Ca2+ toolkit, which drives the pro-angiogenic response to VEGF, is remodelled in tumor-associated ECFCs such that they are seemingly insensitive to this growth factor. This feature could underlie the relative failure of anti-VEGF therapies in cancer patients. Herein, we investigated whether and how VEGF uses Ca2+ signalling to control angiogenesis in BC-derived ECFCs (BC-ECFCs). Although VEGFR-2 was normally expressed, VEGF failed to induce proliferation and in vitro tubulogenesis in BC-ECFCs. Likewise, VEGF did not trigger robust Ca2+ oscillations in these cells. Similar to normal cells, VEGF-induced intracellular Ca2+ oscillations were triggered by inositol-1,4,5-trisphosphate-dependent Ca2+ release from the endoplasmic reticulum (ER) and maintained by store-operated Ca2+ entry (SOCE). However, InsP3-dependent Ca2+ release was significantly lower in BC-ECFCs due to the down-regulation of ER Ca2+ levels, while there was no remarkable difference in the amplitude, pharmacological profile and molecular composition of SOCE. Thus, the attenuation of the pro-angiogenic Ca2+ response to VEGF was seemingly due to the reduction in ER Ca2+ concentration, which prevents VEGF from triggering robust intracellular Ca2+ oscillations. However, the pharmacological inhibition of SOCE prevented BC-ECFC proliferation and in vitro tubulogenesis. These findings demonstrate for the first time that BC-ECFCs are insensitive to VEGF, which might explain at cellular and molecular levels the failure of anti-VEGF therapies in BC patients, and hint at SOCE as a novel molecular target for this disease.
Collapse
Affiliation(s)
- Francesco Lodola
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia 27100, Italy.,Current address: Italian Institute of Technology, Center for Nano Science and Technology, Milano 20133, Italy
| | - Umberto Laforenza
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
| | - Fabio Cattaneo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples 80131, Italy
| | | | - Valentina Poletto
- Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, Foundation IRCCS Policlinico San Matteo, Pavia 27100, Italy
| | - Margherita Massa
- Laboratory of Immunology Transplantation, Foundation IRCCS Policlinico San Matteo, Pavia 27100, Italy
| | - Richard Tancredi
- Medical Oncology Unit, Foundation IRCCS Salvatore Maugeri, Pavia 27100, Italy
| | - Estella Zuccolo
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia 27100, Italy
| | - Dlzar Alì Khdar
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia 27100, Italy
| | - Alberto Riccardi
- Medical Oncology Unit, Foundation IRCCS Salvatore Maugeri, Pavia 27100, Italy.,Department of Internal Medicine, University of Pavia, Pavia 27100, Italy
| | - Marco Biggiogera
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia 27100, Italy
| | - Vittorio Rosti
- Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, Foundation IRCCS Policlinico San Matteo, Pavia 27100, Italy
| | - Germano Guerra
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso 86100, Italy
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia 27100, Italy
| |
Collapse
|
31
|
Hilar fat infiltration: A new prognostic factor in metastatic clear cell renal cell carcinoma with first-line sunitinib treatment. Urol Oncol 2017; 35:603.e7-603.e14. [PMID: 28619630 DOI: 10.1016/j.urolonc.2017.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 05/13/2017] [Accepted: 05/20/2017] [Indexed: 11/22/2022]
Abstract
INTRODUCTION The selection of patients with metastatic clear cell renal cell carcinoma (ccRCC) who may benefit from targeted tyrosine kinase inhibitors has been a challenge, even more so now with the advent of new therapies. Hilar fat infiltration (HFI) is a validated prognostic factor in nonmetastatic ccRCC (TNM 2009 staging system) but has never been studied in metastatic patients. We aimed to assess its phenotype and prognostic effect in patients with metastatic ccRCC treated with first-line sunitinib. MATERIALS AND METHODS In a multicentric study, we retrospectively included 90 patients and studied the corresponding ccRCC at the pathological, immunohistochemical, and molecular levels. Patient and tumor characteristics were compared using univariate and multivariate analysis. All the features were then studied by Cox models for prognostic effect. RESULTS HFI was found in 42 patients (46.7%), who had worse prognosis (Heng criteria) (P = 0.003), liver metastases (P = 0.036), and progressive diseases at first radiological evaluation (P = 0.024). The corresponding ccRCC was associated with poor pathological prognostic factors that are well known in nonmetastatic ccRCC. For these patients, median progression-free survival was 4 months vs. 13 months (P = 0.02), and median overall survival was 14 months vs. 29 months (P = 0.006). In a multivariate Cox model integrating all the variables, only poor prognosis, according to the Heng criteria and HFI, remained independently associated with both progression-free survival and overall survival. CONCLUSION HFI was demonstrated for the first time to be an independent poor prognostic factor. Its potential role in predicting resistance to antiangiogenic therapy warrants further investigation.
Collapse
|
32
|
Jamil MO, Jerome MS, Miley D, Selander KS, Robert F. A pilot study of zoledronic acid in the treatment of patients with advanced malignant pleural mesothelioma. LUNG CANCER-TARGETS AND THERAPY 2017; 8:39-44. [PMID: 28652837 PMCID: PMC5476579 DOI: 10.2147/lctt.s135802] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Purpose Malignant pleural mesothelioma (MPM) is a rare malignancy with a dismal median survival of <12 months with current therapy. Single and combination chemotherapy regimens have shown only modest clinical benefit. In preclinical studies, nitrogen-containing bisphosphonates (zoledronic acid) inhibit growth of mesothelioma cells by different mechanisms: inhibition of mevalonate pathway, inhibition of angiogenesis, activation of apoptosis through caspase activation, and alteration in activity of matrix metalloproteinases, thereby affecting invasiveness of cancer cells. Patients and methods We investigated the role of zoledronic acid in a pilot, single-arm trial of MPM patients with Eastern Cooperative Oncology Group (ECOG) performance status (PS) 0–2 who had progressed on prior treatments or had not received systemic therapy due to poor PS. Primary end point was composite response rate by modified response evaluation criteria in solid tumors and/or metabolic response by 2-deoxy-2-[fluorine-18]fluoro-d-glucose (18F-FDG) positron emission tomography criteria. Secondary end points were progression-free survival (PFS) and overall survival (OS). Exploratory end points include the effect of zoledronic acid therapy on vascular endothelial growth factor (VEGF), basic fibroblast growth factor, interleukin 8, transforming growth factor beta, mesothelin, and osteopontin levels. Results Eight male patients (median age of 62 years) with the following clinical characteristics were treated; ECOG PS was 0–2, 75% with epithelioid type, and 62% had prior chemotherapy Overall composite response rate was 12.5% and the clinical benefit rate (response + stable disease) was 37.5%. Median PFS was 2 months (0.5–21 months) and median OS was 7 months (0.8–28 months). No treatment-related toxicities were observed. Lower VEGF levels were predictive of favorable response and mesothelin levels correlated with disease course. Conclusion Zoledronic acid shows modest clinical activity without significant toxicity in patients with advanced MPM.
Collapse
Affiliation(s)
- Muhammad Omer Jamil
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mary S Jerome
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Deborah Miley
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Katri S Selander
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Francisco Robert
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
33
|
Bispecific Antibodies as a Development Platform for New Concepts and Treatment Strategies. Int J Mol Sci 2016; 18:ijms18010048. [PMID: 28036020 PMCID: PMC5297683 DOI: 10.3390/ijms18010048] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 12/16/2016] [Accepted: 12/21/2016] [Indexed: 12/11/2022] Open
Abstract
With the development of molecular cloning technology and the deep understanding of antibody engineering, there are diverse bispecific antibody formats from which to choose to pursue the optimal biological activity and clinical purpose. The single-chain-based bispecific antibodies usually bridge tumor cells with immune cells and form an immunological synapse because of their relatively small size. Bispecific antibodies in the IgG format include asymmetric bispecific antibodies and homodimerized bispecific antibodies, all of which have an extended blood half-life and their own crystalline fragment (Fc)-mediated functions. Besides retargeting effector cells to the site of cancer, new applications were established for bispecific antibodies. Bispecific antibodies that can simultaneously bind to cell surface antigens and payloads are a very ideal delivery system for therapeutic use. Bispecific antibodies that can inhibit two correlated signaling molecules at the same time can be developed to overcome inherent or acquired resistance and to be more efficient angiogenesis inhibitors. Bispecific antibodies can also be used to treat hemophilia A by mimicking the function of factor VIII. Bispecific antibodies also have broad application prospects in bone disorders and infections and diseases of the central nervous system. The latest developments of the formats and application of bispecific antibodies will be reviewed. Furthermore, the challenges and perspectives are summarized in this review.
Collapse
|
34
|
Abstract
Cabozantinib inhibits a variety of cellular receptors including VEGFR1-3, MET, AXL, RET, FLT3 and KIT. These signaling pathways have been shown to be important in genitourinary malignancies. Along its clinical development, it has shown most activity in advanced renal cell carcinoma; the METEOR study compared cabozantinib to everolimus and showed clinically and statistically significant improvements in both progression-free survival and overall survival. Herein, we review the development of cabozantinib in the genitourinary malignancies of renal cell carcinoma, prostate adenocarcinoma and urothelial carcinoma.
Collapse
Affiliation(s)
- Tian Zhang
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Se Eun Park
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Cierra Hong
- Duke University School of Medicine, Durham, NC 27710, USA
| | - Daniel J George
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA.,Division of Urology, Departments of Medicine & Surgery, Duke Cancer Institute, Duke University Medical Center, DUMC 103861, Durham, NC 27710, USA
| |
Collapse
|
35
|
Yang S, Zhao J, Sun X. Resistance to anti-VEGF therapy in neovascular age-related macular degeneration: a comprehensive review. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:1857-67. [PMID: 27330279 PMCID: PMC4898027 DOI: 10.2147/dddt.s97653] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
As a progressive chronic disease, age-related macular degeneration (AMD) is the leading cause of irreversible vision impairment worldwide. Experimental and clinical evidence has demonstrated that vascular endothelial growth factor (VEGF) plays a vital role in the formation of choroidal neovascularization. Intravitreal injections of anti-VEGF agents have been recommended as a first-line treatment for neovascular AMD. However, persistent fluid or recurrent exudation still occurs despite standardized anti-VEGF therapy. Patients suffering from refractory or recurrent neovascular AMD may develop mechanisms of resistance to anti-VEGF therapy, which results in a diminished therapeutic effect. Until now, there has been no consensus on the definitions of refractory neovascular AMD and recurrent neovascular AMD. This article aims at clarifying these concepts to evaluate the efficacy of switching drugs, which contributes to making clinical decision more scientifically. Furthermore, insight into the causes of resistance to anti-VEGF therapy would be helpful for developing possible therapeutic approaches, such as combination therapy and multi-target treatment that can overcome this resistance.
Collapse
Affiliation(s)
- Shiqi Yang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jingke Zhao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; Eye Research Institute of Shanghai Jiao Tong University, Shanghai, People's Republic of China; Shanghai Key Laboratory of Fundus Disease, Shanghai, People's Republic of China
| |
Collapse
|
36
|
Lee D, Kim D, Choi YB, Kang K, Sung ES, Ahn JH, Goo J, Yeom DH, Jang HS, Moon KD, Lee SH, You WK. Simultaneous blockade of VEGF and Dll4 by HD105, a bispecific antibody, inhibits tumor progression and angiogenesis. MAbs 2016; 8:892-904. [PMID: 27049350 DOI: 10.1080/19420862.2016.1171432] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Several angiogenesis inhibitors targeting the vascular endothelial growth factor (VEGF) signaling pathway have been approved for cancer treatment. However, VEGF inhibitors alone were shown to promote tumor invasion and metastasis by increasing intratumoral hypoxia in some preclinical and clinical studies. Emerging reports suggest that Delta-like ligand 4 (Dll4) is a promising target of angiogenesis inhibition to augment the effects of VEGF inhibitors. To evaluate the effects of simultaneous blockade against VEGF and Dll4, we developed a bispecific antibody, HD105, targeting VEGF and Dll4. The HD105 bispecific antibody, which is composed of an anti-VEGF antibody (bevacizumab-similar) backbone C-terminally linked with a Dll4-targeting single-chain variable fragment, showed potent binding affinities against VEGF (KD: 1.3 nM) and Dll4 (KD: 30 nM). In addition, the HD105 bispecific antibody competitively inhibited the binding of ligands to their receptors, i.e., VEGF to VEGFR2 (EC50: 2.84 ± 0.41 nM) and Dll4 to Notch1 (EC50: 1.14 ± 0.06 nM). Using in vitro cell-based assays, we found that HD105 effectively blocked both the VEGF/VEGFR2 and Dll4/Notch1 signaling pathways in endothelial cells, resulting in a conspicuous inhibition of endothelial cell proliferation and sprouting. HD105 also suppressed Dll4-induced Notch1-dependent activation of the luciferase gene. In vivo xenograft studies demonstrated that HD105 more efficiently inhibited the tumor progression of human A549 lung and SCH gastric cancers than an anti-VEGF antibody or anti-Dll4 antibody alone. In conclusion, HD105 may be a novel therapeutic bispecific antibody for cancer treatment.
Collapse
Affiliation(s)
- Dongheon Lee
- a Hanwha Chemical R&D Center , Biologics Business Unit , Gajeong-Ro, Yuseong-Gu , Daejeon , Republic of Korea
| | - Dongin Kim
- a Hanwha Chemical R&D Center , Biologics Business Unit , Gajeong-Ro, Yuseong-Gu , Daejeon , Republic of Korea
| | - Yu Bin Choi
- a Hanwha Chemical R&D Center , Biologics Business Unit , Gajeong-Ro, Yuseong-Gu , Daejeon , Republic of Korea
| | - Kyungjae Kang
- a Hanwha Chemical R&D Center , Biologics Business Unit , Gajeong-Ro, Yuseong-Gu , Daejeon , Republic of Korea
| | - Eun-Sil Sung
- a Hanwha Chemical R&D Center , Biologics Business Unit , Gajeong-Ro, Yuseong-Gu , Daejeon , Republic of Korea
| | - Jin-Hyung Ahn
- a Hanwha Chemical R&D Center , Biologics Business Unit , Gajeong-Ro, Yuseong-Gu , Daejeon , Republic of Korea
| | - Junseo Goo
- a Hanwha Chemical R&D Center , Biologics Business Unit , Gajeong-Ro, Yuseong-Gu , Daejeon , Republic of Korea
| | - Dong-Hoon Yeom
- a Hanwha Chemical R&D Center , Biologics Business Unit , Gajeong-Ro, Yuseong-Gu , Daejeon , Republic of Korea
| | - Hyun Sook Jang
- a Hanwha Chemical R&D Center , Biologics Business Unit , Gajeong-Ro, Yuseong-Gu , Daejeon , Republic of Korea
| | - Kyung Duk Moon
- a Hanwha Chemical R&D Center , Biologics Business Unit , Gajeong-Ro, Yuseong-Gu , Daejeon , Republic of Korea
| | - Sang Hoon Lee
- a Hanwha Chemical R&D Center , Biologics Business Unit , Gajeong-Ro, Yuseong-Gu , Daejeon , Republic of Korea
| | - Weon-Kyoo You
- a Hanwha Chemical R&D Center , Biologics Business Unit , Gajeong-Ro, Yuseong-Gu , Daejeon , Republic of Korea
| |
Collapse
|
37
|
Rusckowski M, Wang Y, Blankenberg FG, Levashova Z, Backer MV, Backer JM. Targeted scVEGF/(177)Lu radiopharmaceutical inhibits growth of metastases and can be effectively combined with chemotherapy. EJNMMI Res 2016; 6:4. [PMID: 26780081 PMCID: PMC4715132 DOI: 10.1186/s13550-016-0163-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 01/11/2016] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND scVEGF/(177)Lu is a novel radiopharmaceutical targeted by recombinant single-chain (sc) derivative of vascular endothelial growth factor (VEGF) that binds to and is internalized by vascular endothelial growth factor receptors (VEGFR). scVEGF/(177)Lu potential as adjuvant and neoadjuvant anti-angiogenic therapy was assessed in metastatic and orthotopic mouse models of triple-negative breast cancer. METHODS Metastatic lesions in Balb/c mice were established by intracardiac injection of luciferase-expressing 4T1luc mouse breast carcinoma cells. Mice with metastatic lesions received single intravenous (i.v.) injection of well-tolerated dose of scVEGF/(177)Lu (7.4 MBq/mouse) at day 8 after 4T1luc cell injection. Primary orthotopic breast tumors in immunodeficient mice were established by injecting luciferase-expressing MDA231luc human breast carcinoma cells into mammary fat pad. Tumor-bearing mice were treated with single injections of scVEGF/(177)Lu (7.4 MBq/mouse, i.v), or liposomal doxorubicin (Doxil, 1 mg doxorubicin per kg, i.v.), or with a combination of Doxil and scVEGF/(177)Lu given at the same doses, but two hours apart. "Cold" scVEGF-targeting conjugate was included in controls and in Doxil alone group. The effects of treatments were defined by bioluminescent imaging (BLI), computed tomography (CT), computed microtomography (microCT), measurements of primary tumor growth, and immunohistochemical analysis. RESULTS In metastatic model, adjuvant treatment with scVEGF/(177)Lu decreased overall metastatic burden and improved survival. In orthotopic primary tumor model, a combination of Doxil and scVEGF/(177)Lu was more efficient in tumor growth inhibition than each treatment alone. scVEGF/(177)Lu treatment decreased immunostaining for VEGFR-1, VEGFR-2, and pro-tumorigenic M2-type macrophage marker CD206. CONCLUSIONS Selective targeting of VEGFR with well-tolerated doses of scVEGF/(177)Lu is effective in metastatic and primary breast cancer models and can be combined with chemotherapy. As high level of VEGFR expression is a common feature in a variety of cancers, targeted delivery of (177)Lu for specific receptor-mediated uptake warrants further exploration.
Collapse
Affiliation(s)
- Mary Rusckowski
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | | | | | - Zoia Levashova
- Department of Radiology/MIPS, Stanford University, Palo Alto, CA, 94305, USA.,Current address: Igenica Biotherapeutics, Inc., Burlingame, CA, 94010, USA
| | | | | |
Collapse
|
38
|
Wang Y, Yan P, Liu Z, Yang X, Wang Y, Shen Z, Bai H, Wang J, Wang Z. MEK inhibitor can reverse the resistance to bevacizumab in A549 cells harboring Kirsten rat sarcoma oncogene homolog mutation. Thorac Cancer 2015; 7:279-87. [PMID: 27148412 PMCID: PMC4846615 DOI: 10.1111/1759-7714.12325] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 11/10/2015] [Indexed: 01/28/2023] Open
Abstract
Background Bevacizumab (BV) is broadly used to treat a number of cancers; however, BV resistance mechanisms and strategies to overcome this resistance are yet to be determined. Methods We established xenograft mice models harboring Kirsten rat sarcoma oncogene homolog (KRAS) mutations based on the A549 cell line, and tested the responses of xenograft tumors to a series of drugs in ex vivo and in vivo experiments. Changes in transcriptive level were analyzed by ribonucleic acid (RNA) sequencing and the expressions of connexins were determined by immunohistochemistry staining. Results A549 cell mutation type (KRAS G12S) was confirmed by sequencing. After treating the xenograft tumors with BV, the median interval time from BV administration to tumor volume more than 2.5‐fold of the original was 37 days, compared with 21 days in the control (P = 0.025). A549 cells showed resistantance to selumitinib (MEK inhibitor) but were sensitive to selumitinib plus BEZ235 (phosphoinositide 3‐kinase/mammalian target of rapamycin dual inhibitor). However, selumitinib could effectively reverse the resistance to BV in in vivo experiments. RNA sequencing showed that mouse genes, but not human genes, activated the mitogen‐activated protein kinase signaling pathway, accompanied by activation of the Wnt and Hedgehog pathways. Connexin43 (S261) was phosphorylated before and during BV treatment, and subsequently transitioned to negative phosphorylated‐connexin 43‐S261 after resistance to BV. Conclusion Combining an MEK inhibitor with BV was a potential strategy to reverse initial BV resistance. Phosphorylated‐connexin 43 might be associated with the response to BV.
Collapse
Affiliation(s)
- Yuan Wang
- The Department of Ophthalmology National Key Discipline of Pediatrics (Capital Medical University) Ministry of Education Beijing Children's Hospital Capital Medical University Beijing China
| | - Ping Yan
- The Department of Emergency Medicine Qingdao Municipal Hospital Qingdao China
| | - Zhentao Liu
- The Department of Thoracic Medical Oncology Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) Peking University Cancer Hospital & Beijing Institute for Cancer Research Beijing China
| | - Xiaodan Yang
- The Department of Thoracic Medical Oncology Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) Peking University Cancer Hospital & Beijing Institute for Cancer Research Beijing China
| | - Yaping Wang
- The University of Texas School of Public Health Houston Texas USA
| | - Zhirong Shen
- National Institute of Biological Sciences Zhongguancun Life Science Park Beijing China
| | - Hua Bai
- The Department of Medical Oncology Cancer Hospital Chinese Academy of Medical Sciences Beijing China
| | - Jie Wang
- The Department of Medical Oncology Cancer Hospital Chinese Academy of Medical Sciences Beijing China
| | - Zhijie Wang
- The Department of Thoracic Medical Oncology Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) Peking University Cancer Hospital & Beijing Institute for Cancer Research Beijing China
| |
Collapse
|
39
|
Awasthi N, Schwarz RE. Profile of nintedanib in the treatment of solid tumors: the evidence to date. Onco Targets Ther 2015; 8:3691-701. [PMID: 26677336 PMCID: PMC4677757 DOI: 10.2147/ott.s78805] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Angiogenesis is an essential process for tumor growth and metastasis, and remains a promising therapeutic target process in cancer treatment for several cancer types. Bevacizumab, a monoclonal antibody that targets vascular endothelial growth factor (VEGF), was the first antiangiogenic agent approved for cancer therapy. Novel antiangiogenic agents, such as sunitinib, sorafenib, pazopanib, or vandetanib that target additional proangiogenic signaling pathways beyond VEGF, have also been approved for the treatment of various malignant diseases. While most of these agents are approved in combination with cytotoxic chemotherapy for indications including metastatic colorectal cancer, non-small-cell lung cancer, breast cancer, renal cell carcinoma (RCC), and gastric cancer, some are used as approved monotherapy for advanced RCC, hepatocellular carcinoma and medullary thyroid cancer. Major challenges to the success of antiangiogenic therapy include associated toxicity risks, limitation of efficacy through the possible development of resistance and induction or promotion of metastatic progression. Nintedanib (formally known as BIBF 1120) is a triple angiokinase inhibitor of VEGF, fibroblast growth factor, platelet-derived growth factor signaling with lesser activity against RET, Flt-3, and Src. Through this unique targeting profile nintedanib has demonstrated significant antitumor activity in several tumor types in preclinical studies. Nintedanib has also shown promising clinical efficacy in combination with docetaxel and has been approved for treating patients with locally advanced and metastatic non-small-cell lung cancer in Europe. Nintedanib has also been found to be clinically promising in terms of efficacy and safety in several other solid tumors including ovarian cancer (Phase III), RCC (Phase II), and prostate cancer (Phase II). This review article provides a comprehensive summary of the preclinical and clinical efficacy of nintedanib in the treatment of solid tumors.
Collapse
Affiliation(s)
- Niranjan Awasthi
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, USA
| | - Roderich E Schwarz
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, USA ; Indiana University Health Goshen Center for Cancer Care, Goshen, IN, USA
| |
Collapse
|
40
|
Ortega HH, Veiga-Lopez A, Sreedharan S, del Luján Velázquez MM, Salvetti NR, Padmanabhan V. Developmental Programming: Does Prenatal Steroid Excess Disrupt the Ovarian VEGF System in Sheep? Biol Reprod 2015; 93:58. [PMID: 26178718 DOI: 10.1095/biolreprod.115.131607] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 07/10/2015] [Indexed: 01/10/2023] Open
Abstract
Prenatal testosterone (T), but not dihydrotestosterone (DHT), excess disrupts ovarian cyclicity and increases follicular recruitment and persistence. We hypothesized that the disruption in the vascular endothelial growth factor (VEGF) system contributes to the enhancement of follicular recruitment and persistence in prenatal T-treated sheep. The impact of T/DHT treatments from Days 30 to 90 of gestation on VEGFA, VEGFB, and their receptor (VEGFR-1 [FLT1], VEGFR-2 [KDR], and VEGFR-3 [FLT4]) protein expression was examined by immunohistochemistry on Fetal Days 90 and 140, 22 wk, 10 mo (postpubertal), and 21 mo (adult) of age. Arterial morphometry was performed in Fetal Day 140 and postpubertal ovaries. VEGFA and VEGFB expression were found in granulosa cells at all stages of follicular development with increased expression in antral follicles. VEGFA was present in theca interna, while VEGFB was present in theca interna/externa and stromal cells. All three receptors were expressed in the granulosa, theca, and stromal cells during all stages of follicular development. VEGFR-3 increased with follicular differentiation with the highest level seen in the granulosa cells of antral follicles. None of the members of the VEGF family or their receptor expression were altered by age or prenatal T/DHT treatments. At Fetal Day 140, area, wall thickness, and wall area of arteries from the ovarian hilum were larger in prenatal T- and DHT-treated females, suggestive of early androgenic programming of arterial differentiation. This may facilitate increased delivery of endocrine factors and thus indirectly contribute to the development of the multifollicular phenotype.
Collapse
Affiliation(s)
- Hugo Héctor Ortega
- Department of Morphological Sciences, Faculty of Veterinary Sciences, National University of Litoral, Esperanza, Santa Fe, Argentina Argentine National Research Council (CONICET), Esperanza, Santa Fe, Argentina
| | - Almudena Veiga-Lopez
- Department of Pediatrics and the Reproductive Sciences Program, University of Michigan, Ann Arbor, Michigan
| | - Shilpa Sreedharan
- Department of Pediatrics and the Reproductive Sciences Program, University of Michigan, Ann Arbor, Michigan
| | - Melisa María del Luján Velázquez
- Department of Morphological Sciences, Faculty of Veterinary Sciences, National University of Litoral, Esperanza, Santa Fe, Argentina Argentine National Research Council (CONICET), Esperanza, Santa Fe, Argentina
| | - Natalia Raquel Salvetti
- Department of Morphological Sciences, Faculty of Veterinary Sciences, National University of Litoral, Esperanza, Santa Fe, Argentina Argentine National Research Council (CONICET), Esperanza, Santa Fe, Argentina
| | - Vasantha Padmanabhan
- Department of Pediatrics and the Reproductive Sciences Program, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
41
|
Spaide RF. Optical Coherence Tomography Angiography Signs of Vascular Abnormalization With Antiangiogenic Therapy for Choroidal Neovascularization. Am J Ophthalmol 2015; 160:6-16. [PMID: 25887628 DOI: 10.1016/j.ajo.2015.04.012] [Citation(s) in RCA: 213] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/06/2015] [Accepted: 04/07/2015] [Indexed: 12/31/2022]
Abstract
PURPOSE To investigate the vascular appearance of choroidal neovascularization (CNV) treated with recurrent intravitreous anti-vascular endothelial growth factor (VEGF) injections, which have been proposed to cause transient vascular normalization along with decreased vascularity and leakage. DESIGN Retrospective case series with perspective on the topic. METHODS Patients with treated CNV secondary to age-related macular degeneration from a community-based retinal referral practice were evaluated with optical coherence tomography angiography employing split-spectrum amplitude decorrelation. The choroidal neovascular morphology of the 17 eyes of 14 consecutive patients was described. RESULTS The mean age of the patients, 8 men and 6 women, was 78.4 (standard deviation ± 9.3) years. The mean greatest linear dimension of the lesion was 3600 μm. The mean number of anti-VEGF injections was 47 (±21). The vascular diameter of the vessels in the CNV appeared large even in small lesions, with feeder vessels approaching the size of the major arcade vessels of the retina. The vessels had few branch points and many vascular anastomotic connections among larger vessels. There was a paucity of capillaries visualized within the lesions. CONCLUSIONS The findings of this study do not support the hypothesis of vascular normalization in eyes receiving recurrent periodic antiangiogenic treatment. The observed "abnormalization" of the vessels may be explained by periodic pruning of angiogenic vascular sprouts by VEGF withdrawal in the face of unimpeded arteriogenesis. As the eye is a readily accessible VEGF laboratory, features expressed therein may also apply to neovascularization elsewhere in the body, such as in tumors.
Collapse
|
42
|
Hatch AJ, Clarke JM, Nixon AB, Hurwitz HI. Identifying Blood-Based Protein Biomarkers for Antiangiogenic Agents in the Clinic: A Decade of Progress. Cancer J 2015; 21:322-6. [PMID: 26222085 DOI: 10.1097/ppo.0000000000000129] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Agents that inhibit tumor angiogenesis are widely used and have provided meaningful survival benefits to patients in multiple disease settings. However, these agents differ significantly in their mechanisms of action and potential toxicities, and there are currently no prospectively validated biomarkers to guide the selection of agents for individual patients. Blood-based protein biomarkers are well suited for trials investigating antiangiogenic agents for multiple reasons. Many elements of the molecular pathways that antiangiogenic agents target are present and detectable in the circulation, sample collection is minimally invasive, and samples can be collected throughout the course of treatment. Blood-based biomarkers for antiangiogenic therapies are urgently needed to guide the development of therapeutic strategies. This review provides a brief summary of the current blood-based protein biomarkers for antiangiogenic therapies.
Collapse
Affiliation(s)
- Ace J Hatch
- From Duke University Medical Center, Durham, NC
| | | | | | | |
Collapse
|
43
|
Choi IK, Shin H, Oh E, Yoo JY, Hwang JK, Shin K, Yu DC, Yun CO. Potent and long-term antiangiogenic efficacy mediated by FP3-expressing oncolytic adenovirus. Int J Cancer 2015; 137:2253-69. [PMID: 25944623 DOI: 10.1002/ijc.29592] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 04/20/2015] [Indexed: 02/05/2023]
Abstract
Various ways to inhibit vascular endothelial growth factor (VEGF), a key facilitator in tumor angiogenesis, are being developed to treat cancer. The soluble VEGF decoy receptor (FP3), due to its high affinity to VEGF, is a highly effective and promising strategy to disrupt VEGF signaling pathway. Despite potential advantage and potent therapeutic efficacy, its employment has been limited by very poor in vivo pharmacokinetic properties. To address this challenge, we designed a novel oncolytic adenovirus (Ad) expressing FP3 (RdB/FP3). To demonstrate the VEGF-specific nature of RdB/FP3, replication-incompetent Ad expressing FP3 (dE1/FP3) was also generated. dE1/FP3 was highly effective in reducing VEGF expression and functionally elicited an antiangiogeneic effect. Furthermore, RdB/FP3 exhibited a potent antitumor effect compared with RdB or recombinant FP3. Consistent with these data, RdB/FP3 was shown to greatly decrease VEGF expression level and vessel density and increase apoptosis in both tumor endothelial and tumor cells, verifying potent suppressive effects of RdB/FP3 on VEGF-mediated tumor angiogenesis in vivo. Importantly, the therapeutic mechanism of antitumor effect mediated by RdB/FP3 is associated with prolonged VEGF silencing efficacy and enhanced oncolysis via cancer cell-specific replication of oncolytic Ad. Taken together, RdB/FP3 provides a new promising therapeutic approach in the treatment of cancer and angiogenesis-related diseases.
Collapse
Affiliation(s)
- Il-Kyu Choi
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-Gu, Seoul, Korea
| | - Hyewon Shin
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-Gu, Seoul, Korea
| | - Eonju Oh
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-Gu, Seoul, Korea
| | - Ji Young Yoo
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-Gu, Seoul, Korea
| | - June Kyu Hwang
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-Gu, Seoul, Korea
| | - Kyungsub Shin
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-Gu, Seoul, Korea
| | - De-Chao Yu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-Gu, Seoul, Korea
| |
Collapse
|
44
|
Koutras A, Kotoula V, Fountzilas G. Prognostic and predictive role of vascular endothelial growth factor polymorphisms in breast cancer. Pharmacogenomics 2015; 16:79-94. [DOI: 10.2217/pgs.14.148] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Current evidence indicates that angiogenesis plays an important role in the pathogenesis of several malignancies, including breast cancer. The vascular endothelial growth factor (VEGF) pathway has been investigated extensively, due to its important role in angiogenesis. The major mediator of tumor angiogenesis is VEGF-A, frequently referred to as VEGF, which activates the VEGF receptor-2. The VEGF gene is located on chromosome 6 and constitutes a highly polymorphic gene. Numerous SNPs in the promoter, 5′- and 3′-untranslated regions (UTR) of VEGF gene have been recognized. This genetic variability possibly influences the production and function of VEGF. Subsequently, the VEGF SNPs may have an impact on breast cancer risk and disease outcome. Moreover, these SNPs may be of predictive value in patients receiving agents targeting the VEGF pathway. This review presents an update on the potential role of VEGF SNPs as prognostic and/or predictive markers in patients with breast cancer.
Collapse
Affiliation(s)
- Angelos Koutras
- Division of Oncology, Department of Medicine, University Hospital, University of Patras Medical School, Patras, Rion 26504, Greece
| | - Vasiliki Kotoula
- Department of Pathology, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - George Fountzilas
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
- Department of Medical Oncology, ‘Papageorgiou’ Hospital, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| |
Collapse
|
45
|
Favia A, Desideri M, Gambara G, D'Alessio A, Ruas M, Esposito B, Del Bufalo D, Parrington J, Ziparo E, Palombi F, Galione A, Filippini A. VEGF-induced neoangiogenesis is mediated by NAADP and two-pore channel-2-dependent Ca2+ signaling. Proc Natl Acad Sci U S A 2014; 111:E4706-15. [PMID: 25331892 PMCID: PMC4226099 DOI: 10.1073/pnas.1406029111] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) and its receptors VEGFR1/VEGFR2 play major roles in controlling angiogenesis, including vascularization of solid tumors. Here we describe a specific Ca(2+) signaling pathway linked to the VEGFR2 receptor subtype, controlling the critical angiogenic responses of endothelial cells (ECs) to VEGF. Key steps of this pathway are the involvement of the potent Ca(2+) mobilizing messenger, nicotinic acid adenine-dinucleotide phosphate (NAADP), and the specific engagement of the two-pore channel TPC2 subtype on acidic intracellular Ca(2+) stores, resulting in Ca(2+) release and angiogenic responses. Targeting this intracellular pathway pharmacologically using the NAADP antagonist Ned-19 or genetically using Tpcn2(-/-) mice was found to inhibit angiogenic responses to VEGF in vitro and in vivo. In human umbilical vein endothelial cells (HUVECs) Ned-19 abolished VEGF-induced Ca(2+) release, impairing phosphorylation of ERK1/2, Akt, eNOS, JNK, cell proliferation, cell migration, and capillary-like tube formation. Interestingly, Tpcn2 shRNA treatment abolished VEGF-induced Ca(2+) release and capillary-like tube formation. Importantly, in vivo VEGF-induced vessel formation in matrigel plugs in mice was abolished by Ned-19 and, most notably, failed to occur in Tpcn2(-/-) mice, but was unaffected in Tpcn1(-/-) animals. These results demonstrate that a VEGFR2/NAADP/TPC2/Ca(2+) signaling pathway is critical for VEGF-induced angiogenesis in vitro and in vivo. Given that VEGF can elicit both pro- and antiangiogenic responses depending upon the balance of signal transduction pathways activated, targeting specific VEGFR2 downstream signaling pathways could modify this balance, potentially leading to more finely tailored therapeutic strategies.
Collapse
Affiliation(s)
- Annarita Favia
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Marianna Desideri
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, 00128 Rome, Italy
| | - Guido Gambara
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Alessio D'Alessio
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy; Institute of Histology and Embryology, Catholic University of the Sacred Heart, 00168 Rome, Italy; and
| | - Margarida Ruas
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom
| | - Bianca Esposito
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Donatella Del Bufalo
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, 00128 Rome, Italy
| | - John Parrington
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom
| | - Elio Ziparo
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Fioretta Palombi
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom
| | - Antonio Filippini
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy;
| |
Collapse
|
46
|
Ghavamipour F, Shahangian SS, Sajedi RH, Arab SS, Mansouri K, Aghamaali MR. Development of a highly-potent anti-angiogenic VEGF8-109 heterodimer by directed blocking of its VEGFR-2 binding site. FEBS J 2014; 281:4479-94. [PMID: 25132001 DOI: 10.1111/febs.12956] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 07/23/2014] [Accepted: 07/30/2014] [Indexed: 01/22/2023]
Abstract
Angiogenesis is a hallmark of various pathological conditions and is controlled by a variety of angiogenic factors. Blockade of vascular endothelial growth factor (VEGF) as the most pivotal stimulator of angiogenesis offers a promising therapeutic approach for some diseases, typically cancer. In the present study, a heterodimeric antagonistic VEGF was precisely designed based on structural information of recently-crystallized VEGF/VEGF receptor-2 (VEGFR-2/fetal liver kinase 1/kinase domain region) complex. Directed blocking of kinase domain region occurs via substitution of a VEGF receptor binding site by two peptide segments in one pole, whereas the binding domain of the other pole of VEGF was intact. Candidate peptides for substitution were selected considering to some sequence and structural criteria. A reliable model of modified VEGF was built, refined using molecular dynamics simulation and docked with VEGFR-2. Docking analysis revealed that binding affinity of mutant VEGF was notably diminished, corroborating our design. Heterodimeric VEGF was expressed, refolded and highly purified by two-step affinity chromatography. Dimerization of this antagonist was confirmed using some analytical techniques. Spectroscopic studies assured us to obtain the heterodimeric form of VEGF. Some angiogenic in vitro assays such endothelial cell proliferation and tube formation indicated that this antagonist is not only strongly capable of inhibiting angiogenesis (half maximal inhibitory concentration of 33 and 24 ng · mL(-1) , respectively), but also showed the highest inhibitory effect compared to all other heterodimeric VEGF variants. The high anti-angiogenic potency of this VEGF antagonist may allow its future use as an anti-tumor agent.
Collapse
Affiliation(s)
- Fahimeh Ghavamipour
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | | | | | | | | | | |
Collapse
|
47
|
Penson RT, Moore KM, Fleming GF, Braly P, Schimp V, Nguyen H, Matulonis UA, Banerjee S, Haluska P, Gore M, Bodurka DC, Hozak RR, Joshi A, Xu Y, Schwartz JD, McGuire WP. A phase II study of ramucirumab (IMC-1121B) in the treatment of persistent or recurrent epithelial ovarian, fallopian tube or primary peritoneal carcinoma. Gynecol Oncol 2014; 134:478-85. [PMID: 25016924 PMCID: PMC5166425 DOI: 10.1016/j.ygyno.2014.06.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 06/25/2014] [Accepted: 06/28/2014] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Vascular endothelial growth factor (VEGF) receptor-mediated signaling contributes to ovarian cancer pathogenesis. Elevated VEGF expression is associated with poor clinical outcomes. We investigated ramucirumab, a fully human anti-VEGFR-2 antibody, in patients with persistent or recurrent epithelial ovarian, fallopian tube, or primary peritoneal carcinoma. Primary endpoints were progression-free survival at 6 months (PFS-6) and confirmed objective response rate (ORR). METHODS Women who received ≥ 1 platinum-based chemotherapeutic regimen and had a platinum-free interval of <12 months with measurable disease were eligible. Patients received 8 mg/kg ramucirumab intravenously every 2 weeks. RESULTS Sixty patients were treated; one patient remained on study as of September 2013. The median age was 62 years (range: 27-80), and median number of prior regimens was 3. Forty-five (75%) patients had platinum refractory/resistant disease. Thirty-nine patients (65.0%) had serous tumors. PFS-6 was 25.0% (n=15/60, 95% CI: 14.7-37.9%). Best overall response was: partial response 5.0% (n=3/60), stable disease 56.7% (n=34/60), and progressive disease 33.3% (n=20/60). The most common treatment-emergent adverse events possibly related to study drug were headache (65.0%; 10.0% Grade ≥ 3), fatigue (56.7%; 3.3% Grade ≥ 3), diarrhea (28.3%; 1.7% Grade ≥ 3), hypertension (25.0%; 3.3% Grade ≥ 3), and nausea (20.0%; no Grade ≥ 3). Two patients experienced intestinal perforations (3.3% Grade ≥ 3). Pharmacodynamic analyses revealed changes in several circulating VEGF proteins following initial ramucirumab infusion, including increased VEGF-A, PlGF and decreased sVEGFR-2. CONCLUSIONS Although antitumor activity was observed, the predetermined efficacy endpoints were not met.
Collapse
Affiliation(s)
- Richard T Penson
- Massachusetts General Hospital for the Dana-Farber/Harvard Cancer Center, Boston, MA, USA
| | - Kathleen M Moore
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Patricia Braly
- Hematology and Oncology Specialists, LLC, Marrero, LA, USA
| | | | - Hoa Nguyen
- Gynecologic Oncology Associates, Inc., Hollywood, USA
| | | | | | | | | | - Diane C Bodurka
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Yihuan Xu
- ImClone Systems, a wholly-owned subsidiary of Eli Lilly and Company, Bridgewater, NJ, USA
| | - Jonathan D Schwartz
- ImClone Systems, a wholly-owned subsidiary of Eli Lilly and Company, Bridgewater, NJ, USA
| | | |
Collapse
|
48
|
Serum protein profiling reveals baseline and pharmacodynamic biomarker signatures associated with clinical outcome in mCRC patients treated with chemotherapy ± cediranib. Br J Cancer 2014; 111:1590-604. [PMID: 25121956 PMCID: PMC4200086 DOI: 10.1038/bjc.2014.436] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 06/10/2014] [Accepted: 07/09/2014] [Indexed: 02/07/2023] Open
Abstract
Background: This study evaluated soluble serum proteins as biomarkers to subset patients with metastatic colorectal cancer (mCRC) treated with chemotherapy±cediranib, a vascular endothelial growth factor (VEGF) signalling inhibitor (VEGFi). Exploring biomarkers at pre- and on-treatment may identify patient subgroups showing clinical benefit on cediranib combination. Methods: Two hundred and seven serum proteins were analysed in 588 mCRC patients at pre- and on-treatment with chemotherapy (FOLFOX/CAPOX)±cediranib 20 mg. Patients were enrolled in the phase III trial HORIZON II. We correlated baseline biomarker signatures and pharmacodynamic (PD) biomarkers with PFS and OS. Results: We identified a baseline signature (BS) of 47 biomarkers that included VEGFA, VEGFD, VEGFR2, VEGFR3 and TIE-2, which defined two distinct subgroups of patients. Patients treated with chemotherapy plus cediranib who had ‘high' BS had shorter PFS (HR=1.82, P=0.003) than patients with ‘low' BS. This BS did not correlate with PFS of the patients treated with chemotherapy plus placebo. In addition, we identified a profile of 16 PD proteins on treatment associated with PFS (HR=0.58, P<0.001) and OS (HR=0.52, P<0.001) in patients treated with chemotherapy plus cediranib. This PD profile did not correlate with PFS and OS in patients treated with chemotherapy plus placebo. Conclusions: Serum proteins may represent relevant biomarkers to predict the outcome of patients treated with VEGFi-based therapies. We report a BS and PD biomarkers that may identify mCRC patients showing increased benefit of combining cediranib with chemotherapy. These exploratory findings need to be validated in future prospective studies.
Collapse
|
49
|
Pastushenko I, Vermeulen PB, Van den Eynden GG, Rutten A, Carapeto FJ, Dirix LY, Van Laere S. Mechanisms of tumour vascularization in cutaneous malignant melanoma: clinical implications. Br J Dermatol 2014; 171:220-33. [PMID: 24641095 DOI: 10.1111/bjd.12973] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2014] [Indexed: 01/02/2023]
Abstract
Malignant melanoma represents < 10% of all skin cancers but is responsible for the majority of skin-cancer-related deaths. Metastatic melanoma has historically been considered as one of the most therapeutically challenging malignancies. Fortunately, for the first time after decades of basic research and clinical investigation, new drugs have produced major clinical responses. Angiogenesis has been considered an important target for cancer treatment. Initial efforts have focused primarily on targeting endothelial and tumour-related vascular endothelial growth factor signalling. Here, we review different mechanisms of tumour vascularization described in melanoma and discuss the potential clinical implications.
Collapse
Affiliation(s)
- I Pastushenko
- Department of Dermatology, Hospital Clínico Universitario 'Lozano Blesa', Zaragoza, 50009, Spain
| | | | | | | | | | | | | |
Collapse
|
50
|
Mross K, Büchert M, Frost A, Medinger M, Stopfer P, Studeny M, Kaiser R. Vascular effects, efficacy and safety of nintedanib in patients with advanced, refractory colorectal cancer: a prospective phase I subanalysis. BMC Cancer 2014; 14:510. [PMID: 25012508 PMCID: PMC4105047 DOI: 10.1186/1471-2407-14-510] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 07/04/2014] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Nintedanib is a potent, oral angiokinase inhibitor that targets VEGF, PDGF and FGF signalling, as well as RET and Flt3. The maximum tolerated dose of nintedanib was evaluated in a phase I study of treatment-refractory patients with advanced solid tumours. In this preplanned subanalysis, the effect of nintedanib on the tumour vasculature, along with efficacy and safety, was assessed in 30 patients with colorectal cancer (CRC). METHODS Patients with advanced CRC who had failed conventional treatment, or for whom no therapy of proven efficacy existed, were treated with nintedanib ranging from 50-450 mg once-daily (n = 14) or 150-250 mg twice-daily (n = 16) for 28 days. After a 1-week rest, further courses were permitted in the absence of progression or undue toxicity. The primary objective was the effect on the tumour vasculature using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and expressed as the initial area under the DCE-MRI contrast agent concentration-time curve after 60 seconds (iAUC60) or the volume transfer constant between blood plasma and extravascular extracellular space (Ktrans). RESULTS Patients received a median of 4.0 courses (range: 1-13). Among 21 evaluable patients, 14 (67%) had a ≥40% reduction from baseline in Ktrans and 13 (62%) had a ≥40% decrease from baseline in iAUC60, representing clinically relevant effects on tumour blood flow and permeability, respectively. A ≥40% reduction from baseline in Ktrans was positively associated with non-progressive tumour status (Fisher's exact: p = 0.0032). One patient achieved a partial response at 250 mg twice-daily and 24 (80%) achieved stable disease lasting ≥8 weeks. Time to tumour progression (TTP) at 4 months was 26% and median TTP was 72.5 days (95% confidence interval: 65-114). Common drug-related adverse events (AEs) included nausea (67%), vomiting (53%) and diarrhoea (40%); three patients experienced drug-related AEs ≥ grade 3. Four patients treated with nintedanib once-daily had an alanine aminotransferase and/or aspartate aminotransferase increase ≥ grade 3. No increases > grade 2 were seen in the twice-daily group. CONCLUSIONS Nintedanib modulates tumour blood flow and permeability in patients with advanced, refractory CRC, while achieving antitumour activity and maintaining an acceptable safety profile.
Collapse
Affiliation(s)
- Klaus Mross
- Tumor Biology Center, Department of Medical Oncology, Breisacherstrasse 117, D-79106 Freiburg in Breisgau, Germany
| | - Martin Büchert
- Magnetic Resonance Development and Application Center, Department of Radiology, University Medical Center Freiburg, Freiburg, Germany
| | - Annette Frost
- Tumor Biology Center, Department of Medical Oncology, Breisacherstrasse 117, D-79106 Freiburg in Breisgau, Germany
| | - Michael Medinger
- Tumor Biology Center, Department of Medical Oncology, Breisacherstrasse 117, D-79106 Freiburg in Breisgau, Germany
| | - Peter Stopfer
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Matus Studeny
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Rolf Kaiser
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| |
Collapse
|