1
|
Moghassemi S, Dadashzadeh A, Lucci CM, Amorim CA. Tumor-Infiltration Mimicking Model of Contaminated Ovarian Tissue as an Innovative Platform for Advanced Cancer Research. AAPS J 2024; 27:7. [PMID: 39586867 DOI: 10.1208/s12248-024-00997-5] [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: 09/10/2024] [Accepted: 11/09/2024] [Indexed: 11/27/2024] Open
Abstract
The development of advanced preclinical models is crucial for the evaluation and validation of novel therapeutic strategies in oncology. Three-dimensional (3D) microtumor models, which incorporate both cancer and stromal cells within biomimetic hydrogels, have emerged as powerful tools that more accurately replicate the complex tumor microenvironment compared to traditional two-dimensional (2D) cell culture systems. In this context, our study aims to develop 3D microtumor models by integrating cancer and stromal cells within an extracellular-matrix-mimetic hydrogel, as a physiologically accurate microtumor model that can serve as an innovative platform for advanced cancer research and drug screening. Microtumors composed of varying ratios of leukemia cells (HL-60) to healthy ovarian stromal cells (SCs) (1:1, 1:10, 1:100, or 1:1000) were encapsulated in PEGylated fibrin hydrogel and cultured for 5 days. The proliferation and dynamics of cancerous and healthy cell populations were evaluated using CD43/Ki67 immunofluorescence double staining. Our findings indicate that tumor development and malignancy progression can be influenced by adjusting cell culture ratios and incubation time. Notably, the HL-60:SCs ratio of 1:100 closely replicated leukemia cell invasion in ovarian tissue, demonstrating detectable malignancy on the third and fifth days without significant changes in total cell density dynamics. This 3D leukemia microtumor model offers superior physiological relevance compared to traditional 2D in vitro assays and shows promising potential for applications in cellular analysis and drug screening.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale Et Clinique, Université Catholique de Louvain, Avenue Hippocrate 54, Bte B1.55.03, 1200, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale Et Clinique, Université Catholique de Louvain, Avenue Hippocrate 54, Bte B1.55.03, 1200, Brussels, Belgium
| | - Carolina M Lucci
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Christiani A Amorim
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale Et Clinique, Université Catholique de Louvain, Avenue Hippocrate 54, Bte B1.55.03, 1200, Brussels, Belgium.
| |
Collapse
|
2
|
Moghassemi S, Dadashzadeh A, Amorim CA. A novel bioprinted microtumour model for studying acute-leukemia-cell- contaminated in ovarian tissue. Life Sci 2024; 355:122992. [PMID: 39154811 DOI: 10.1016/j.lfs.2024.122992] [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: 05/30/2024] [Revised: 07/26/2024] [Accepted: 08/13/2024] [Indexed: 08/20/2024]
Abstract
Microtumor models, combining cancer and stromal cells within 3D hydrogels, are vital for testing anticancer therapies. Bioprinting hydrogel scaffolds allows tailored in vitro models. We created a 3D microtumor model using a bioprinter, with varying ratios of ovarian stromal cells and leukemia cells (HL-60). PEGylated fibrinogen and alginate hydrogel were used. Cell dynamics and proliferation were assessed via immunofluorescence staining. Microtumors with different HL-60 ratios (1:1, 1:10, 1:100) were cultured for 5 days. Results showed tumor development modulation by cell ratios and culture time. A significant cell density increase occurred in 1:1 ratio microtumors, indicating rapid cancer cell proliferation. No HL-60 cells were found in 1:100 ratio microtumors by day 5. The 1:10 ratio closely mimicked leukemia invasion in ovarian tissue, showing detectable cancer cells by days 3 and 5 without altering total cell density dynamics significantly. This bioprinted leukemia microtumor model offers better physiological relevance than 2D assays, promising applications in cellular analysis and drug screening.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Christiani A Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
3
|
Wang P, Zhang Y, Xiang R, Yang J, Xu Y, Deng T, Zhou W, Wang C, Xiao X, Wang S. Foretinib Is Effective in Acute Myeloid Leukemia by Inhibiting FLT3 and Overcoming Secondary Mutations That Drive Resistance to Quizartinib and Gilteritinib. Cancer Res 2024; 84:905-918. [PMID: 38231480 PMCID: PMC10940854 DOI: 10.1158/0008-5472.can-23-1534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/15/2023] [Accepted: 01/09/2024] [Indexed: 01/18/2024]
Abstract
FLT3 internal tandem duplication (FLT3-ITD) mutations are one of the most prevalent somatic alterations associated with poor prognosis in patients with acute myeloid leukemia (AML). The clinically approved FLT3 kinase inhibitors gilteritinib and quizartinib improve the survival of patients with AML with FLT3-ITD mutations, but their long-term efficacy is limited by acquisition of secondary drug-resistant mutations. In this study, we conducted virtual screening of a library of 60,411 small molecules and identified foretinib as a potent FLT3 inhibitor. An integrated analysis of the BeatAML database showed that foretinib had a lower IC50 value than other existing FLT3 inhibitors in patients with FLT3-ITD AML. Foretinib directly bound to FLT3 and effectively inhibited FLT3 signaling. Foretinib potently inhibited proliferation and promoted apoptosis in human AML cell lines and primary AML cells with FLT3-ITD mutations. Foretinib also significantly extended the survival of mice bearing cell-derived and patient-derived FLT3-ITD xenografts, exhibiting stronger efficacy than clinically approved FLT3 inhibitors in treating FLT3-ITD AML. Moreover, foretinib showed potent activity against secondary mutations of FLT3-ITD that confer resistance to quizartinib and gilteritinib. These findings support the potential of foretinib for treating patients with AML with FLT3-ITD mutations, especially for those carrying secondary mutations after treatment failure with other FLT3 inhibitors. SIGNIFICANCE Foretinib exhibits superior efficacy to approved drugs in AML with FLT3-ITD mutations and retains activity in AML with secondary FLT3 mutations that mediate resistance to clinical FLT3 inhibitors.
Collapse
Affiliation(s)
- Peihong Wang
- Department of Hematology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, P.R. China
| | - Yvyin Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Rui-Jin Hospital, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Rufang Xiang
- Department of General Practice, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Jie Yang
- Department of Hematology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, P.R. China
| | - Yanli Xu
- Department of Hematology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, P.R. China
| | - Tingfen Deng
- Department of Hematology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, P.R. China
| | - Wei Zhou
- Department of Hematology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, P.R. China
| | - Caixia Wang
- Department of Hematology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, P.R. China
| | - Xinhua Xiao
- Department of Hematology and Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, P.R. China
| | - Shunqing Wang
- Department of Hematology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, P.R. China
| |
Collapse
|
4
|
Borriello R, Cerrito L, Gasbarrini A, Ponziani FR. Pharmacokinetic considerations for angiogenesis inhibitors used to treat hepatocellular carcinoma: an overview. Expert Opin Drug Metab Toxicol 2023; 19:785-794. [PMID: 37847538 DOI: 10.1080/17425255.2023.2272598] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 10/16/2023] [Indexed: 10/18/2023]
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is the fifth malignancy in terms of frequency and the fourth malignancy in terms of cancer-related death worldwide. Systemic therapy of advanced HCC has probably gone through the greatest wave of change in the last decade, with the introduction of several anti-angiogenic drugs and immune checkpoint inhibitors, able to significantly improve patients' prognosis. AREAS COVERED In this review, we summarize the pharmacokinetic characteristic of the antiangiogenic drugs currently approved for the treatment of HCC, from oral tyrosine kinase inhibitors (sorafenib, lenvatinib, regorafenib and cabozantinib) to monoclonal antibodies (bevacizumab and ramucirumab), focusing on the main aspects that differ among compounds from the same class, on factors that can exert an influence on pharmacokinetic parameters and the main issues that could limit their clinical use. EXPERT OPINION Anti-angiogenic drugs have different profiles in terms of bioavailability, metabolism, elimination and interindividual variability in their pharmacokinetics and effectiveness. More studies should be developed to address the intrinsic and extrinsic factors influencing pharmacokinetics parameters to improve the individual therapeutic response and, furthermore, to evaluate the benefit and the harm of systemic therapy for advanced HCC in selected patients with liver impairment.
Collapse
Affiliation(s)
- Raffaele Borriello
- Digestive Disease Center-CEMAD, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Lucia Cerrito
- Digestive Disease Center-CEMAD, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Digestive Disease Center-CEMAD, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesca Romana Ponziani
- Digestive Disease Center-CEMAD, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
5
|
Huang ZF, Zhu W, Wang C, Mo LD, Huang HL, Tong XG. Association of Kinase-Insert-Domain-Containing Receptor Polymorphisms with Glioma Susceptibility in a Chinese Population: A Hospital-Based Case-Control Study. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2023; 2023:8808422. [PMID: 37114147 PMCID: PMC10129418 DOI: 10.1155/2023/8808422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 12/11/2022] [Accepted: 03/20/2023] [Indexed: 04/29/2023]
Abstract
Background Gliomas are the most common malignant tumors of the central nervous system. However, the inherited genetic variation in gliomas is presently unclear. Therefore, this study investigated the association of the rs2071559 and rs2239702 gene polymorphisms with glioma susceptibility in Chinese patients. Methods In this study, a case-control approach was used to compare and analyze whether two genes, rs2071559 and rs2239702, were associated with the risk of glioma formation. Results The cases and controls were matched for sex, smoking status, and family history of cancer using single nucleotide polymorphisms. Specific rs2071559 and rs2239702 alleles were found much more frequently in the glioma group than in the control group (P < 0.001 and P = 0.014, respectively). Conclusions These findings suggest that specific rs2071559 and rs2239702 polymorphisms are associated with a higher risk of glioma development; the risk allele is C in rs2071559 or A in rs2239702. Moreover, the kinase-insert-domain-containing receptor may act as a suppressor of tumor progression.
Collapse
Affiliation(s)
- Zhi-Fa Huang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Wei Zhu
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China
| | - Chen Wang
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Li-Dong Mo
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Hui-Ling Huang
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Xiao-Guang Tong
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China
| |
Collapse
|
6
|
Cao Y, Sun C, Huo G, Wang H, Wu Y, Wang F, Liu S, Zhai S, Zhang X, Zhao H, Hu M, Gu W, Yang Y, Wang S, Liang C, Lyu J, Lu T, Wang Y, Xie L, Fan C. Novel hKDR mouse model depicts the antiangiogenesis and apoptosis-promoting effects of neutralizing antibodies targeting vascular endothelial growth factor receptor 2. Cancer Sci 2022; 114:115-128. [PMID: 36114822 PMCID: PMC9807522 DOI: 10.1111/cas.15594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/31/2022] [Accepted: 09/06/2022] [Indexed: 01/07/2023] Open
Abstract
Vascular endothelial growth factor receptor 2 (VEGFR2)/KDR plays a critical role in tumor growth, diffusion, and invasion. The amino acid sequence homology of KDR between mouse and human in the VEGF ligand-binding domain was low, thus the WT mice could not be used to evaluate Abs against human KDR, and the lack of a suitable mouse model hindered both basic research and drug developments. Using the CRISPR/Cas9 technique, we successfully inserted different fragments of the human KDR coding sequence into the chromosomal mouse Kdr exon 4 locus to obtain an hKDR humanized mouse that can be used to evaluate the marketed Ab ramucirumab. In addition, the humanized mAb VEGFR-HK19 was developed, and a series of comparative assays with ramucirumab as the benchmark revealed that VEGFR-HK19 has higher affinity and superior antiproliferation activity. Moreover, VEGFR-HK19 selectively inhibited tumor growth in the hKDR mouse model but not in WT mice. The most important binding epitopes of VEGFR2-HK19 are D257, L313, and T315, located in the VEGF binding region. Therefore, the VEGFR2-HK19 Ab inhibits tumor growth by blocking VEGF-induced angiogenesis, inflammation, and promoting apoptosis. To our best knowledge, this novel humanized KDR mouse fills the gaps both in an animal model and the suitable in vivo evaluation method for developing antiangiogenesis therapies in the future, and the newly established humanized Ab is expected to be a drug candidate possibly benefitting tumor patients.
Collapse
Affiliation(s)
- Yuan Cao
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Chunyun Sun
- Beijing Engineering Research Center of Protein and AntibodySinocelltech LtdBeijingChina
| | - Guitao Huo
- National Center for Safety Evaluation of Drugs, Institute for Food and Drug Safety EvaluationNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Huiyu Wang
- Beijing Engineering Research Center of Protein and AntibodySinocelltech LtdBeijingChina
| | - Yong Wu
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Fei Wang
- Beijing Engineering Research Center of Protein and AntibodySinocelltech LtdBeijingChina
| | - Susu Liu
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Shijie Zhai
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Xiao Zhang
- Beijing Engineering Research Center of Protein and AntibodySinocelltech LtdBeijingChina
| | - Haoyang Zhao
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Meiling Hu
- Beijing Engineering Research Center of Protein and AntibodySinocelltech LtdBeijingChina
| | - Wenda Gu
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Yanwei Yang
- National Center for Safety Evaluation of Drugs, Institute for Food and Drug Safety EvaluationNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Sanlong Wang
- National Center for Safety Evaluation of Drugs, Institute for Food and Drug Safety EvaluationNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Chunnan Liang
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Jianjun Lyu
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Tiangong Lu
- School of Life SciencesBeijing University of Chinese MedicineBeijingChina
| | - Youchun Wang
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, Institute for Biological Product ControlNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Liangzhi Xie
- Beijing Engineering Research Center of Protein and AntibodySinocelltech LtdBeijingChina,Beijing Key Laboratory of Monoclonal Antibody Research and DevelopmentSino Biological Inc.BeijingChina,Cell Culture Engineering CenterChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Changfa Fan
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| |
Collapse
|
7
|
Chen H, Xiao M, He J, Zhang Y, Liang Y, Liu H, Zhang Z. Aptamer-Functionalized Carbon Nanotube Field-Effect Transistor Biosensors for Alzheimer's Disease Serum Biomarker Detection. ACS Sens 2022; 7:2075-2083. [PMID: 35816677 DOI: 10.1021/acssensors.2c00967] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Blood-biomarker-based tests are highly important for the early clinical diagnosis of Alzheimer's disease (AD) and the treatment and care of AD patients, but the complex serum environment and extremely low abundance of AD blood protein biomarkers present challenges. Nanomaterials are promising for constructing highly sensitive transistor-based biosensors due to their small size. However, such biosensors are difficult to fabricate on a large scale and suffer from the lack of combined optimization of reproducibility and sensitivity in complex physiological fluids. In this work, field-effect transistor (FET) biosensors based on highly uniform semiconducting carbon nanotube (CNT) thin films are mass produced to achieve highly sensitive and selective detection of the AD core blood biomarkers of β-amyloid (Aβ). The combination of the mass-produced CNT FET sensors and oligonucleotide aptamers as efficient bioreceptors enables reliable and reproducible sub-femtomolar detection in full human serum for Aβ42 and Aβ40 peptides and has outperformed other methods reported to date. The adsorption of biological substrates to the sensor was significantly reduced by multiple blocking steps, resulting in selectivity ratios of up to 730% (Aβ40) and 800% (Aβ42). The aptamer-functionalized CNT FET biosensor exhibits a large dynamic range (>104), rapid response time (several minutes), and low variation (<10%) and can be delivered as a low-cost and rapid clinical detection technology for the early diagnosis and mass screening of AD. This platform will help bring complex laboratory-based and expensive diagnostic tools to the point of care.
Collapse
Affiliation(s)
- Hong Chen
- Hunan Institute of Advanced Sensing and Information Technology, Xiangtan University, Xiangtan 411105, Hunan, China
| | - Mengmeng Xiao
- Key Laboratory for the Physics and Chemistry of Nanodevices and Center for Carbon-based Electronics, School of Electronics, Peking University, Beijing 100871, China.,Jihua Laboratory, Foshan 528200, Guangdong China
| | - Jianping He
- Hunan Institute of Advanced Sensing and Information Technology, Xiangtan University, Xiangtan 411105, Hunan, China
| | - Yang Zhang
- Hunan Institute of Advanced Sensing and Information Technology, Xiangtan University, Xiangtan 411105, Hunan, China
| | - Yuqi Liang
- Key Laboratory for the Physics and Chemistry of Nanodevices and Center for Carbon-based Electronics, School of Electronics, Peking University, Beijing 100871, China
| | - Haiyang Liu
- Key Laboratory for the Physics and Chemistry of Nanodevices and Center for Carbon-based Electronics, School of Electronics, Peking University, Beijing 100871, China
| | - Zhiyong Zhang
- Hunan Institute of Advanced Sensing and Information Technology, Xiangtan University, Xiangtan 411105, Hunan, China.,Key Laboratory for the Physics and Chemistry of Nanodevices and Center for Carbon-based Electronics, School of Electronics, Peking University, Beijing 100871, China.,Jihua Laboratory, Foshan 528200, Guangdong China
| |
Collapse
|
8
|
Xu RH, Zhang Y, Pan H, Feng J, Zhang T, Liu T, Qin Y, Qin S, Yin X, Liu B, Ba Y, Yang N, Voon PJ, Tanasanvimon S, Zhou C, Zhang WL, Shen L. Efficacy and safety of weekly paclitaxel with or without ramucirumab as second-line therapy for the treatment of advanced gastric or gastroesophageal junction adenocarcinoma (RAINBOW-Asia): a randomised, multicentre, double-blind, phase 3 trial. Lancet Gastroenterol Hepatol 2021; 6:1015-1024. [PMID: 34626550 DOI: 10.1016/s2468-1253(21)00313-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND In the global phase 3 RAINBOW study, ramucirumab plus paclitaxel significantly improved overall survival compared with placebo plus paclitaxel in patients with advanced gastric or gastro-oesophageal junction (GEJ) adenocarcinoma. RAINBOW-Asia, a bridging study with similar design to RAINBOW, aimed to evaluate the efficacy and safety of ramucirumab plus paclitaxel for advanced gastric or GEJ adenocarcinoma in Asian, predominantly Chinese, patients. METHODS RAINBOW-Asia was a randomised, double-blind, placebo-controlled, phase 3 trial done at 32 centres in China, Malaysia, the Philippines, and Thailand. Adult patients (≥18 years) with metastatic or locally advanced, unresectable gastric or GEJ adenocarcinoma who previously received fluoropyrimidine-platinum-based chemotherapy were randomly assigned with a centralised interactive web response system in a 2:1 ratio to receive ramucirumab 8 mg/kg or placebo intravenously on days 1 and 15 plus paclitaxel 80 mg/m2 intravenously on days 1, 8, and 15 of every 28-day cycle. Randomisation was stratified by Eastern Cooperative Oncology Group performance status and presence of peritoneal metastases. The co-primary endpoints were progression-free survival and overall survival. Efficacy analyses were done in the intention-to-treat population, and safety analysis included patients who received at least one dose of study treatment. This trial is registered with ClinicalTrials.gov, NCT02898077, and has been completed. FINDINGS Between March 2, 2017, and June 30, 2020, 440 patients were randomly assigned to receive ramucirumab plus paclitaxel (n=294) or placebo plus paclitaxel (n=146). Median progression-free survival was 4·14 months (95% CI 3·71-4·30) in the ramucirumab plus paclitaxel group compared with 3·15 months (2·83-4·14) in the placebo plus paclitaxel group (hazard ratio [HR] 0·765, 95% CI 0·613-0·955, p=0·0184). Median overall survival was 8·71 months (95% CI 7·98-9·49) in the ramucirumab plus paclitaxel group and 7·92 months (6·31-9·10) in the placebo plus paclitaxel group (HR 0·963, 95% CI 0·771-1·203, p=0·7426). The most common grade 3 or worse treatment-emergent adverse events were decreased neutrophil count (159 [54%] of 293 patients in the ramucirumab plus paclitaxel group vs 56 [39%] of 145 in the placebo plus paclitaxel group), decreased white blood cell count (127 [43%] vs 42 [29%]), anaemia (46 [16%] vs 24 [17%]), hypertension (21 [7%] vs nine [6%]), and febrile neutropenia (18 [6%] vs one [<1%]). INTERPRETATION These findings, along with the results from RAINBOW, support the use of ramucirumab plus paclitaxel as second-line therapy in a predominantly Chinese population with advanced gastric or GEJ adenocarcinoma. FUNDING Eli Lilly and Company, USA. TRANSLATION For the Chinese translation of the abstract see Supplementary Materials section.
Collapse
MESH Headings
- Adenocarcinoma/diagnosis
- Adenocarcinoma/drug therapy
- Administration, Intravenous
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antineoplastic Agents, Phytogenic/administration & dosage
- Antineoplastic Agents, Phytogenic/adverse effects
- Antineoplastic Agents, Phytogenic/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Case-Control Studies
- China/epidemiology
- Double-Blind Method
- Esophageal Neoplasms/diagnosis
- Esophageal Neoplasms/drug therapy
- Esophagogastric Junction/pathology
- Female
- Humans
- Malaysia/epidemiology
- Male
- Middle Aged
- Paclitaxel/administration & dosage
- Paclitaxel/adverse effects
- Paclitaxel/therapeutic use
- Philippines/epidemiology
- Placebos/administration & dosage
- Progression-Free Survival
- Safety
- Stomach Neoplasms/pathology
- Thailand/epidemiology
- Treatment Outcome
- Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
- Ramucirumab
Collapse
Affiliation(s)
- Rui-Hua Xu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yanqiao Zhang
- Department of Gastroenterology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jifeng Feng
- Department of Medical Oncology, Jiangsu Cancer Hospital, Nanjing, China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianshu Liu
- Department of Medical Oncology, Fudan University Zhongshan Hospital, Shanghai Medical College, Shanghai, China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shukui Qin
- Cancer Center of Nanjing Bayi Hospital, Nanjing Chinese Medicine University, Nanjing, China
| | - Xianli Yin
- Gastroenterology and Urology Department, Hunan Cancer Hospital & The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Baorui Liu
- The Comprehensive Cancer Center of Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Yi Ba
- Medical Oncology, Tianjin Cancer Hospital, Tianjin, China
| | - Nong Yang
- Gastroenterology and Urology Department, Hunan Cancer Hospital & The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Pei Jye Voon
- Radiotherapy and Oncology Department, Hospital Umum Sarawak, Kuching, Malaysia
| | - Suebpong Tanasanvimon
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Chan Zhou
- Lilly China Drug Development and Medical Affairs Center, Eli Lilly and Company, Shanghai, China
| | - Wan Li Zhang
- Lilly China Drug Development and Medical Affairs Center, Eli Lilly and Company, Shanghai, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
9
|
de Moraes Neto JE, Pereira F, Neves RL, de Barros NMT, Gil CD, Fernandes AG, Watanabe SES, Meyer CH, Farah ME, Rodrigues EB. Preclinical assessment of intravitreal ramucirumab: in vitro and in vivo safety profile. Int J Retina Vitreous 2020. [DOI: 10.1186/s40942-020-00243-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Abstract
Background
Anti-angiogenic drugs remain the mainstay therapy for several vascular retinal pathologies. The repurposing of approved anti-angiogenic drugs for use in ophthalmology can increase therapeutic options and reduce costs. The purpose of this study was to investigate the ocular safety profile of intravitreal (IVT) ramucirumab, an approved anti-vascular endothelial growth factor molecule for systemic treatment, using cell culture and animal models.
Methods
The cytotoxicity of ramucirumab at different concentrations was evaluated in human retinal pigment epithelial cells (ARPE-19) using the MTT assay. In addition, 250 or 500 µg of ramucirumab or vehicle was injected in the eye of 16 chinchilla rabbits. The eyes were evaluated by ophthalmoscopy, electroretinography, spectral-domain optical coherence tomography (SD-OCT) and by light and transmission electron microscopy.
Results
Electroretinography or SD-OCT did not detect functional or morphological alterations at 24 h or one week after injection. Light and transmission electron microscopy confirmed the absence of major signs of toxicity, although we found a statistically significant reduction in ganglion cell number between the controls and the eyes that received 500 µg of ramucirumab after 7 days. Compared to lower concentrations, 500 µg of ramucirumab caused reduction in cell viability and changes in morphology in ARPE-19 cells. Compared to the baseline, ocular and serum osmolarity showed no difference after IVT injection at all timepoints.
Conclusion
In conclusion, IVT injection of ramucirumab in rabbits is safe and does not cause functional damage to the retina. At the lower dose tested in vivo (250 µg), the morphology and ultrastructural anatomy were normal at 24 h and 1 week after the injection. However, the 500 µg dose can cause a decrease in ganglion cell number seven days after the injection.
Collapse
|
10
|
Abstract
Ramucirumab (Cyramza®), a fully human anti-VEGFR-2 monoclonal antibody, has been approved as monotherapy for the treatment of patients with hepatocellular carcinoma (HCC) and α-fetoprotein levels ≥ 400 ng/mL who have been treated with sorafenib. Ramucirumab significantly prolonged overall survival (OS) and progression-free survival (PFS) relative to placebo in this population in the randomized, double-blind phase 3 REACH 2 trial. These benefits were seen in key prespecified subgroups based on demographic and disease characteristics. Ramucirumab had an acceptable tolerability profile and manageable safety profile in these patients, with the majority of treatment-related adverse events being mild or moderate in severity. The safety profile of ramucirumab was consistent with that expected for agents targeting the VEGF/VEGFR axis. Currently, ramucirumab is the only therapy specifically tested in patients with α-fetoprotein levels ≥ 400 ng/mL, which is associated with an aggressive disease and poor prognosis. Therefore, ramucirumab is an important treatment option for patients with HCC and α-fetoprotein levels ≥ 400 ng/mL who have been treated with sorafenib.
Collapse
Affiliation(s)
- Yahiya Y Syed
- Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|
11
|
Gao F, Yang C. Anti-VEGF/VEGFR2 Monoclonal Antibodies and their Combinations with PD-1/PD-L1 Inhibitors in Clinic. Curr Cancer Drug Targets 2020; 20:3-18. [PMID: 31729943 DOI: 10.2174/1568009619666191114110359] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/29/2019] [Accepted: 09/19/2019] [Indexed: 12/21/2022]
Abstract
The vascular endothelial growth factor (VEGF)/VEGF receptor 2 (VEGFR2) signaling pathway is one of the most important pathways responsible for tumor angiogenesis. Currently, two monoclonal antibodies, anti-VEGF-A antibody Bevacizumab and anti-VEGFR2 antibody Ramucizumab, have been approved for the treatment of solid tumors. At the same time, VEGF/VEGFR2 signaling is involved in the regulation of immune responses. It is reported that the inhibition of this pathway has the capability to promote vascular normalization, increase the intra-tumor infiltration of lymphocytes, and decrease the number and function of inhibitory immune cell phenotypes, including Myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs) and M2 macrophages. On this basis, a number of clinical studies have been performed to investigate the therapeutic potential of VEGF/VEGFR2-targeting antibodies plus programmed cell death protein 1 (PD-1)/ programmed cell death ligand 1 (PD-L1) inhibitors in various solid tumor types. In this context, VEGF/VEGFR2- targeting antibodies, Bevacizumab and Ramucizumab are briefly introduced, with a description of the differences between them, and the clinical studies involved in the combination of Bevacizumab/ Ramucizumab and PD-1/PD-L1 inhibitors are summarized. We hope this review article will provide some valuable clues for further clinical studies and usages.
Collapse
Affiliation(s)
- Feng Gao
- BuChang (Beijing) Pharmaceutical Co. Ltd, Hongda Industrial Park, Hongda North Road, Beijing 100176, China
| | - Chun Yang
- BuChang (Beijing) Pharmaceutical Co. Ltd, Hongda Industrial Park, Hongda North Road, Beijing 100176, China
| |
Collapse
|
12
|
Khan U, Shah MA. Ramucirumab for the treatment of gastric or gastro-esophageal junction cancer. Expert Opin Biol Ther 2019; 19:1135-1141. [PMID: 31452409 DOI: 10.1080/14712598.2019.1656715] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 08/13/2019] [Indexed: 01/13/2023]
Abstract
Introduction: Gastric cancer remains one of the most lethal malignancy, accounting for an estimated 783,000 deaths worldwide in 2018. Although there are several approved drugs for the treatment of gastric cancer, the survival of patients with advanced disease remains dismal. Ramucirumab, a vascular endothelial growth factor receptor-2 inhibitor, is an important new targeted drug approved for gastric and gastroesophageal adenocarcinoma (GEJ) in second-line setting. Areas covered: In this article, we have reviewed the role of ramucirumab in the management of gastric and GEJ adenocarcinoma. A comprehensive review of various clinical trials is presented that support the use of ramucirumab in gastric cancer. Expert opinion: In our opinion, ramucirumab should be considered as a standard of care option, either alone or with paclitaxel, after progression on first-line therapy for advanced or metastatic disease. The results of large, randomized phase III clinical trials show benefit of ramucirumab on median overall survival (OS). However, the benefit is limited, with only about two months OS benefit of using ramucirumab with paclitaxel compared to paclitaxel alone. Novel combination therapies, such as ramucirumab with other targeted agents and immune checkpoint inhibitors in ongoing clinical trials, may provide important information to further improve the patient outcomes.
Collapse
Affiliation(s)
- Uqba Khan
- Division of Hematology and Oncology, Weill Cornell Medicine/New York Presbyterian Hospital , New York , NY , USA
| | - Manish A Shah
- Division of Hematology and Oncology, Weill Cornell Medicine/New York Presbyterian Hospital , New York , NY , USA
| |
Collapse
|
13
|
Afshari F, Soleyman-Jahi S, Keshavarz-Fathi M, Roviello G, Rezaei N. The promising role of monoclonal antibodies for gastric cancer treatment. Immunotherapy 2019; 11:347-364. [PMID: 30678552 DOI: 10.2217/imt-2018-0093] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 12/03/2018] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer (GC) is the second leading cause of cancer-related death world-wide. Despite improvements in prevention, early detection and various therapeutic options, the prognosis is still poor. GC is often diagnosed at an advanced stage with survivals less than 1 year. Chemotherapy as the mainstay of treatment in advanced stage is not of notable advantages, underlining the need for novel more effective therapeutic options. Based on current knowledge of molecular and cellular mechanisms, a number of novel biologic approaches such as monoclonal antibodies have been recently introduced for cancer treatment that mainly affect the immune system or target signaling pathways playing role in cancer and metastasis development. In this review, various monoclonal antibodies for GC therapy were explained.
Collapse
Affiliation(s)
- Farzaneh Afshari
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Saeed Soleyman-Jahi
- Digestive Diseases Research Cores Center, Division of Gastroenterology, School of Medicine, Washington University, St. Louis, USA
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), St. Louis, USA
- Cancer Research Center, Cancer Institute of Iran, Tehran, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Giandomenico Roviello
- Medical Oncology Unit, Department of Oncology, San Donato Hospital, Via Nenni 20, Arezzo, Italy
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Arezzo, Italy
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Sheffield, UK
| |
Collapse
|
14
|
Avian leukosis virus subgroup J induces VEGF expression via NF-κB/PI3K-dependent IL-6 production. Oncotarget 2018; 7:80275-80287. [PMID: 27852059 PMCID: PMC5348319 DOI: 10.18632/oncotarget.13282] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 11/07/2016] [Indexed: 02/07/2023] Open
Abstract
Avian leukosis virus subgroup J (ALV-J) is an oncogenic virus causing hemangiomas and myeloid tumors in chickens. Interleukin-6 (IL-6) is a multifunctional pro-inflammatory interleukin involved in many types of cancer. We previously demonstrated that IL-6 expression was induced following ALV-J infection in chickens. The aim of this study is to characterize the mechanism by which ALV-J induces IL-6 expression, and the role of IL-6 in tumor development. Our results demonstrate that ALV-J infection increases IL-6 expression in chicken splenocytes, peripheral blood lymphocytes, and vascular endothelial cells. IL-6 production is induced by the ALV-J envelope protein gp85 and capsid protein p27 via PI3K- and NF-κB-mediated signaling. IL-6 in turn induced expression of vascular endothelial growth factor (VEGF)-A and its receptor, VEGFR-2, in vascular endothelial cells and embryonic vascular tissues. Suppression of IL-6 using siRNA inhibited the ALV-J induced VEGF-A and VEGFR-2 expression in vascular endothelial cells, indicating that the ALV-J-induced VEGF-A/VEGFR-2 expression is mediated by IL-6. As VEGF-A and VEGFR-2 are important factors in oncogenesis, our findings suggest that ALV-J hijacks IL-6 to promote tumorigenesis, and indicate that IL-6 could potentially serve as a therapeutic target in ALV-J infections.
Collapse
|
15
|
Huang J, Lu Z, Xiao Y, He B, Pan C, Zhou X, Xu N, Liu X. Inhibition of Siah2 Ubiquitin Ligase by Vitamin K3 Attenuates Chronic Myeloid Leukemia Chemo-Resistance in Hypoxic Microenvironment. Med Sci Monit 2018; 24:727-735. [PMID: 29400343 PMCID: PMC5810368 DOI: 10.12659/msm.908553] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND A hypoxic microenvironment is associated with resistance to tyrosine kinase inhibitors (TKIs) and a poor prognosis in chronic myeloid leukemia (CML). The E3 ubiquitin ligase Siah2 plays a vital role in the regulation of hypoxia response, as well as in leukemogenesis. However, the role of Siah2 in CML resistance is unclear, and it is unknown whether vitaminK3 (a Siah2 inhibitor) can improve the chemo-sensitivity of CML cells in a hypoxic microenvironment. MATERIAL AND METHODS The expression of Siah2 was detected in CML patients (CML-CP and CML-BC), K562 cells, and K562-imatinib-resistant cells (K562-R cells). We measured the expression of PHD3, HIF-1α, and VEGF in both cell lines under normoxia and hypoxic conditions, and the degree of leukemic sensitivity to imatinib and VitaminK3 were evaluated. RESULTS Siah2 was overexpressed in CML-BC patients (n=9) as compared to CML-CP patients (n=13). Similarly, K562-imatinib-resistant cells (K562-R cells) showed a significantly higher expression of Siah2 as compared to K562 cells in a hypoxic microenvironment. Compared to normoxia, under hypoxic conditions, both cell lines had lower PHD3, higher HIF-1α, and higher VEGF expression. Additionally, Vitamin K3 (an inhibitor of Siah2) reversed these changes and promoted a higher degree of leukemic sensitivity to imatinib. CONCLUSIONS Our findings indicate that the Siah2-PHD3- HIF-1α-VEGF axis is an important hypoxic signaling pathway in a leukemic microenvironment. An inhibitor of Siah2, combined with TKIs, might be a promising therapy for relapsing and refractory CML patients.
Collapse
Affiliation(s)
- Jixian Huang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Department of Hematology, Yuebei People's Hospital, Shantou University, Shaoguan, Guangdong, China (mainland)
| | - Ziyuan Lu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Yajuan Xiao
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Bolin He
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Chengyun Pan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Xuan Zhou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Na Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Xiaoli Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| |
Collapse
|
16
|
Lu Y, Xu Q, Zuo Y, Liu L, Liu S, Chen L, Wang K, Lei Y, Zhao X, Li Y. Isoprenaline/β2-AR activates Plexin-A1/VEGFR2 signals via VEGF secretion in gastric cancer cells to promote tumor angiogenesis. BMC Cancer 2017; 17:875. [PMID: 29262812 PMCID: PMC5738852 DOI: 10.1186/s12885-017-3894-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 12/08/2017] [Indexed: 11/10/2022] Open
Abstract
Background The role of stress signals in regulating gastric cancer initiation and progression is not quite clear. It is known that stress signals modulate multiple processes such as immune function, cell migration and angiogenesis. However, few studies have investigated the mechanisms of how stress signals contribute to gastric cancer angiogenesis. Methods Here, we used β2-adrenergic receptor (β2-AR) agonist isoprenaline to imitate a stress signal and demonstrated the molecular mechanism underlying stress’s influence on tumor angiogenesis. Results We found that isoprenaline stimulated vascular endothelial growth factor (VEGF) secretion in gastric cancer cells and plexin-A1 expression was induced by human recombinant VEGF165 in both gastric cancer cells and vascular endothelial cells. Furthermore, interfere with plexin-A1 expression in gastric cancer cells influence HUVEC tube formation, migration and tumor growth in vivo. Conclusions These findings suggest that isoprenaline stimulate VGEF secretion and subsequently up-regulate the expression of plexin-A1 and VEGFR2 in gastric cancer cells, which form a positive impetus to promote tumor angiogenesis. This study reveals a novel molecular mechanism that a stress signal like isoprenaline may enhance angiogenesis via activating plexin-A1/VEGFR2 signaling pathway in gastric cancer, which may be a potential target in development of an anti-angiogenic therapy for gastric cancer.
Collapse
Affiliation(s)
- Yanjie Lu
- Department of Pathology; Cancer Research Laboratory, Chengde Medical College, Shangerdaohezi Avenue, Chengde, Hebei, 067000, People's Republic of China
| | - Qian Xu
- Institute of Basic Medical Sciences, Chengde Medical College, Chengde, 067000, People's Republic of China
| | - Yanzhen Zuo
- Department of Pharmacology, Chengde Medical College, Chengde, 067000, People's Republic of China
| | - Lei Liu
- Department of Pathogenic Microorganism, Chengde Medical College, Chengde, 067000, People's Republic of China
| | - Shaochen Liu
- Department of Pathology, Chengde Medical College, Chengde, 067000, People's Republic of China
| | - Lei Chen
- Department of General Surgery, the 266th Hospital of Chinese People's Liberation Army, Puning Avenue, Chengde, Hebei, 067000, People's Republic of China
| | - Kang Wang
- Department of Pathology, Chengde Medical College, Chengde, 067000, People's Republic of China
| | - Yuntao Lei
- Department of Pathology, Chengde Medical College, Chengde, 067000, People's Republic of China
| | - Xiangyang Zhao
- Department of General Surgery, the 266th Hospital of Chinese People's Liberation Army, Puning Avenue, Chengde, Hebei, 067000, People's Republic of China.
| | - Yuhong Li
- Department of Pathology; Cancer Research Laboratory, Chengde Medical College, Shangerdaohezi Avenue, Chengde, Hebei, 067000, People's Republic of China.
| |
Collapse
|
17
|
O'Brien L, Westwood P, Gao L, Heathman M. Population pharmacokinetic meta-analysis of ramucirumab in cancer patients. Br J Clin Pharmacol 2017; 83:2741-2751. [PMID: 28833321 PMCID: PMC5698573 DOI: 10.1111/bcp.13403] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 07/21/2017] [Accepted: 08/07/2017] [Indexed: 12/29/2022] Open
Abstract
AIMS Ramucirumab is a human IgG1 monoclonal antibody that specifically binds vascular endothelial growth factor receptor-2 (VEGFR-2) and blocks binding of VEGF-A, VEGF-C and VEGF-D. The objective of the analysis was to characterize the clinical pharmacology profile of ramucirumab using a population pharmacokinetic approach. METHODS A total of 1639 patients with 6427 serum concentrations from 11 Phase 1b, 2 and 3 clinical trials in patients with various cancer indications were included in the analysis. Ramucirumab was administered as an intravenous infusion over 1 h at 8 mg kg-1 every 2 weeks or 10 mg kg-1 every 3 weeks. A series of pharmacostatistical models were developed to describe the concentration data. The best model was used to evaluate patient factors for their effect on ramucirumab pharmacokinetics. RESULTS The pharmacokinetics of ramucirumab were well characterized by a two-compartment model. Mean population estimates of clearance, volume of distribution and half-life for a typical 68-kg patient were 0.0148 l h-1 , 5.30 l and 13.4 days, respectively. A modest relationship was observed between body weight and ramucirumab disposition; clearance and central compartment volume increased with body weight. No other patient characteristics were shown to influence the disposition of ramucirumab in this patient population. CONCLUSIONS The final model adequately described the concentration-time profile of ramucirumab in patients with a range of cancer indications. The model confirmed that a weight-normalized dosing regimen is appropriate for ramucirumab therapy. Dose adjustment was not required for patients with mild to moderate renal impairment or mild hepatic impairment.
Collapse
MESH Headings
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/blood
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal, Humanized
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/blood
- Antineoplastic Agents, Immunological/pharmacokinetics
- Area Under Curve
- Clinical Trials as Topic
- Half-Life
- Humans
- Infusions, Intravenous
- Metabolic Clearance Rate
- Models, Biological
- Neoplasms/blood
- Neoplasms/drug therapy
- Treatment Outcome
- Ramucirumab
Collapse
Affiliation(s)
| | | | - Ling Gao
- Eli Lilly and CompanyBridgewaterNew JerseyUSA
| | | |
Collapse
|
18
|
Vennepureddy A, Singh P, Rastogi R, Atallah JP, Terjanian T. Evolution of ramucirumab in the treatment of cancer - A review of literature. J Oncol Pharm Pract 2017; 23:525-539. [PMID: 27306885 DOI: 10.1177/1078155216655474] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Ramucirumab is a recombinant human monoclonal antibody and is used in the treatment of advanced malignancies. Its mechanism of action is by inhibiting angiogenesis in tumor cells by targeting the vascular endothelial growth factor receptor 2. United States Food and Drug Administration (FDA) approved it initially in 2014 for the treatment of advanced gastric or gastro-esophageal junction adenocarcinoma and metastatic non-small cell lung carcinoma. It was approved by FDA in 2015 for the treatment of advanced colorectal cancer. This manuscript consolidates pre-clinical trials to phase I, II, and III trial data indicating the effects of ramucirumab on different cancer types, which led to its approval. By comparing these clinical trials alongside each other, we can more easily examine the studies that have already been completed, along with currently ongoing studies and potential further areas of interest for this newly approved treatment. This approach makes it convenient to compare dosages, overall survival, adverse events, as well as possible routes for combination therapy with ramucirumab. By compiling results for various oncological malignancies, we can differentiate between treatments that are effective and have the highest incidence of stable disease, and those that do not seem promising. Ramucirumab has been effective in the treatment of various carcinomas and this article outlines other tumors in which this treatment option may be successful.
Collapse
Affiliation(s)
- A Vennepureddy
- 1 Department of Internal Medicine, Staten Island University Hospital, NY, USA
| | - P Singh
- 1 Department of Internal Medicine, Staten Island University Hospital, NY, USA
| | - R Rastogi
- 1 Department of Internal Medicine, Staten Island University Hospital, NY, USA
| | - J P Atallah
- 2 Division of Hematology and Oncology, Staten Island University Hospital, NY, USA
| | - T Terjanian
- 2 Division of Hematology and Oncology, Staten Island University Hospital, NY, USA
| |
Collapse
|
19
|
Passaro D, Di Tullio A, Abarrategi A, Rouault-Pierre K, Foster K, Ariza-McNaughton L, Montaner B, Chakravarty P, Bhaw L, Diana G, Lassailly F, Gribben J, Bonnet D. Increased Vascular Permeability in the Bone Marrow Microenvironment Contributes to Disease Progression and Drug Response in Acute Myeloid Leukemia. Cancer Cell 2017; 32:324-341.e6. [PMID: 28870739 PMCID: PMC5598545 DOI: 10.1016/j.ccell.2017.08.001] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 04/25/2017] [Accepted: 08/01/2017] [Indexed: 12/11/2022]
Abstract
The biological and clinical behaviors of hematological malignancies can be influenced by the active crosstalk with an altered bone marrow (BM) microenvironment. In the present study, we provide a detailed picture of the BM vasculature in acute myeloid leukemia using intravital two-photon microscopy. We found several abnormalities in the vascular architecture and function in patient-derived xenografts (PDX), such as vascular leakiness and increased hypoxia. Transcriptomic analysis in endothelial cells identified nitric oxide (NO) as major mediator of this phenotype in PDX and in patient-derived biopsies. Moreover, induction chemotherapy failing to restore normal vasculature was associated with a poor prognosis. Inhibition of NO production reduced vascular permeability, preserved normal hematopoietic stem cell function, and improved treatment response in PDX.
Collapse
Affiliation(s)
- Diana Passaro
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Alessandro Di Tullio
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ander Abarrategi
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Kevin Rouault-Pierre
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Katie Foster
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Linda Ariza-McNaughton
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Beatriz Montaner
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Probir Chakravarty
- Bioinformatic Core Unit, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Leena Bhaw
- Advanced Sequencing Unit, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Giovanni Diana
- MRC Centre for Developmental Neurobiology, King's College London, London, UK
| | - François Lassailly
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - John Gribben
- Department of Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
20
|
Haghi A, Mohammadi S, Heshmati M, Ghavamzadeh A, Nikbakht M. Anti-Vascular Endothelial Growth Factor Effects of Sorafenib and Arsenic Trioxide in Acute Myeloid Leukemia Cell Lines. Asian Pac J Cancer Prev 2017; 18:1655-1661. [PMID: 28670885 PMCID: PMC6373800 DOI: 10.22034/apjcp.2017.18.6.1655] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML), is a clonal disorder caused by acquired somatic mutations and chromosomal
rearrangements. According to some evidence, progression of hematolymphoid malignancies depends on the induction
of new blood vessel formation under the influence of acute leukemia. Various factors are produced by cancer cells
under hypoxic conditions to increase vascular formation. Among these, vascular endothelial growth factor (VEGF)
plays a crucial role. Cytotoxicity and anticancer effects of arsenic trioxide (ATO) have been reported in many cancers.
Sorafenib, known as an angiogenic inhibitor, decreases leukemic cell survival. The aim of this study was to indicate
combination effects of ATO and sorafenib in two AML cell lines, KG-1 and U937. Effective doses was determined by
MTT assay for both single and combination treatments. Percentages of apoptotic cells were evaluated by Annexin V
FITC staining and mRNA levels of VEGF isoforms and receptor expression were investigated by Real-Time PCR. Our
data show that sorafenib (5μM and 7μM in KG-1 and U937 cell lines respectively), ATO (1.618μM and 1μM in KG-1
and U937 cell lines respectively), and also their combination significantly increased the percentage of apoptotic cells.
In addition the mRNA level of VEGF isoforms was downregulated in the U937 cell line while upregulated in KG-1
cells. Taken together, our results suggest that the VEGF autocrine loop may have an influence on AML development
and progression and could be consider as a therapeutic target. The combination of sorafenib as a VEGF inhibitor with
ATO synergistically inhibits cell proliferation and promotes apoptosis.
Collapse
Affiliation(s)
- Atousa Haghi
- Department of Molecular and Cellular Science, Faculty of Advanced Science and Technology, Islamic Azad University, Tehran-Iran.,Young Researchers and Elite Club, Pharmaceutical Science Branch, Islamic Azad University, Tehran-Iran.
| | | | | | | | | |
Collapse
|
21
|
Nokihara H, Yamamoto N, Yamada Y, Honda K, Asahina H, Tamura Y, Hozak RR, Gao L, Suzukawa K, Enatsu S, Tamura T. A phase 1 study of ramucirumab in Japanese patients with advanced solid tumors. Jpn J Clin Oncol 2017; 47:298-305. [PMID: 28158463 DOI: 10.1093/jjco/hyx008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Accepted: 01/15/2017] [Indexed: 12/16/2023] Open
Abstract
OBJECTIVE Ramucirumab is a recombinant human immunoglobulin G1 monoclonal antibody targeting the vascular endothelial growth factor receptor-2. The aim of this phase 1 study was to evaluate the safety and tolerability of ramucirumab monotherapy in Japanese patients with advanced solid tumors. METHODS Patients with solid tumors who had not responded to standard therapy or for whom no standard therapy was available received escalating doses of ramucirumab, administered once every 2 (Q2W) or 3 (Q3W) weeks. The primary objective was to establish the safety and pharmacokinetic profiles of ramucirumab. Secondary and exploratory objectives included assessment of immunogenicity and antitumor activity. ClinicalTrials.gov: NCT01005355. RESULTS Fifteen patients were treated with ramucirumab at a dose of 6 mg/kg Q2W (N = 3), 8 mg/kg Q2W (N = 6) or 10 mg/kg Q3W (N = 6). There were no dose-limiting toxicities and the maximum tolerated dose was not reached. The most common ramucirumab-related adverse events were headache, pyrexia, hypertension and increased aspartate aminotransferase. Following single-dose administration of ramucirumab, there appeared to be a dose-proportional increase in maximum observed drug concentration but not in area under the curve. Treatment-emergent anti-ramucirumab antibodies were not detected in any patient. CONCLUSIONS Ramucirumab monotherapy was well tolerated and feasible at the doses and schedules used in this study population of Japanese patients with advanced solid tumors.
Collapse
Affiliation(s)
| | | | | | - Kazunori Honda
- National Cancer Center Hospital, Tokyo, Japan
- Present address: Aichi Cancer Center Hospital, Nagoya, Japan
| | - Hajime Asahina
- National Cancer Center Hospital, Tokyo, Japan
- Present address: Hokkaido University Hospital, Sapporo, Japan
| | - Yosuke Tamura
- National Cancer Center Hospital, Tokyo, Japan
- Present address: Osaka Medical College Hospital, Osaka, Japan
| | | | - Ling Gao
- Eli Lilly and Company, Bridgewater, NJ, USA
| | | | | | - Tomohide Tamura
- National Cancer Center Hospital, Tokyo, Japan
- Present address: St Luke's International Hospital, Tokyo, Japan
| |
Collapse
|
22
|
Vahdat LT, Layman R, Yardley DA, Gradishar W, Salkeni MA, Joy A, Garcia AA, Ward P, Khatcheressian J, Sparano J, Rodriguez G, Tang S, Gao L, Dalal RP, Kauh J, Miller K. Randomized Phase II Study of Ramucirumab or Icrucumab in Combination with Capecitabine in Patients with Previously Treated Locally Advanced or Metastatic Breast Cancer. Oncologist 2017; 22:245-254. [PMID: 28220020 PMCID: PMC5344637 DOI: 10.1634/theoncologist.2016-0265] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/14/2016] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Icrucumab (ICR) and ramucirumab (RAM) bind vascular endothelial growth factor (VEGF) receptors 1 and 2 (VEGFR-1 and -2), respectively. This open-label, randomized phase II study evaluated their efficacy and safety in combination with capecitabine (CAP) in patients with previously treated unresectable, locally advanced or metastatic breast cancer. METHODS Patients were randomly assigned (1:1:1) to receive CAP (1,000 mg/m2 orally twice daily, days 1-14) alone or in combination with RAM (10 mg/kg intravenously [IV], days 1 and 8) (RAM + CAP) or ICR (12 mg/kg IV, days 1 and 8) (ICR + CAP) every 21 days. The primary endpoint was progression-free survival (PFS). Secondary endpoints included overall survival (OS), tumor response, safety, and pharmacokinetics. RESULTS Of 153 patients randomized, 150 received treatment. Median PFS (95% confidence interval) was 22.1 (12.1-36.1) weeks on RAM + CAP, 7.3 (6.3-13.0) weeks on ICR + CAP, and 19.0 (12.1-24.3) weeks on CAP (hazard ratios [HRs]: 0.691, p = .1315, RAM + CAP versus CAP; 1.480, p = .0851, ICR + CAP versus CAP). Median OS was 67.4 weeks on RAM + CAP, 62.1 weeks on ICR + CAP, and 71.6 weeks on CAP (HRs: 1.833, p = .0283, RAM + CAP versus CAP; 1.468, p = .1550, ICR + CAP versus CAP). There was no statistically significant difference in PFS or OS between either combination arm and CAP. Treatment-related adverse events more frequent (by ≥10%) on RAM + CAP than on CAP were constipation, decreased appetite, headache, epistaxis, and hypertension. Those more frequent (by ≥10%) on ICR + CAP than CAP were anemia, increased lacrimation, periorbital edema, nausea, vomiting, peripheral edema, facial edema, dehydration, and dyspnea. CONCLUSION Combining RAM or ICR with CAP did not improve PFS in the targeted study population. The Oncologist 2017;22:245-254 IMPLICATIONS FOR PRACTICE: Icrucumab and ramucirumab are recombinant human IgG1 monoclonal antibodies that bind vascular endothelial growth factor (VEGF) receptors 1 and 2 (VEGFR-1 and -2), respectively. VEGFR-1 activation on endothelial and tumor cell surfaces increases tumor vascularization and growth and supports tumor growth via multiple mechanisms, including contributions to angiogenesis and direct promotion of cancer cell proliferation. Strong preclinical and clinical evidence suggests key roles for VEGF and angiogenesis in breast cancer growth, invasion, and metastasis. This randomized phase II study evaluated the efficacy and safety of each antibody in combination with capecitabine in patients with previously treated unresectable, locally advanced or metastatic breast cancer.
Collapse
Affiliation(s)
- Linda T Vahdat
- Weill Cornell Breast Center, Weill Cornell Medicine, New York, New York, USA
| | | | | | - William Gradishar
- Northwestern University Feinburg School of Medicine, Chicago, Illinois, USA
| | - Mohamad A Salkeni
- Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, West Virginia, USA
| | - Anil Joy
- University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Agustin A Garcia
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Patrick Ward
- Oncology Hematology Care Incorporated, Cincinnati, Ohio, USA
| | | | - Joseph Sparano
- Weiler Division, Montefiore Medical Center, Bronx, New York, USA
| | | | - Shande Tang
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Ling Gao
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Rita P Dalal
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | - John Kauh
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Kathy Miller
- Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|
23
|
|
24
|
Frishman-Levy L, Izraeli S. Advances in understanding the pathogenesis of CNS acute lymphoblastic leukaemia and potential for therapy. Br J Haematol 2016; 176:157-167. [DOI: 10.1111/bjh.14411] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/15/2016] [Indexed: 12/16/2022]
Affiliation(s)
- Liron Frishman-Levy
- Childhood Leukaemia Research Section; Department of Paediatric Oncology and the Cancer Research Centre; Safra Children Hospital and Sheba Medical Centre; Tel Hashomer Ramat Gan Israel
- Department of Human Genetics and Biochemistry; Tel Aviv University; Tel Aviv Israel
| | - Shai Izraeli
- Childhood Leukaemia Research Section; Department of Paediatric Oncology and the Cancer Research Centre; Safra Children Hospital and Sheba Medical Centre; Tel Hashomer Ramat Gan Israel
- Department of Human Genetics and Biochemistry; Tel Aviv University; Tel Aviv Israel
| |
Collapse
|
25
|
Paulus V, Avrillon V, Pérol M. The safety and efficacy of ramucirumab in combination with docetaxel in the treatment of lung cancer. Expert Rev Anticancer Ther 2016; 16:1119-1129. [PMID: 27686510 DOI: 10.1080/14737140.2016.1241147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Angiogenesis is critical for tumor growth, proliferation and metastasis with the crucial role of Vascular Endothelial Growth factor (VEGF) pathway. Ramucirumab is a monoclonal antibody that specifically targets the extracellular domain of Vascular Endothelial Growth Factor Receptor 2. Areas covered: We performed a search on Medline to browse the current literature on Ramucirumab and anti-angiogenic agents, for the treatment of NSCLC. The REVEL study demonstrated a significant improvement of response rate, progression-free survival and overall survival by adding ramucirumab to docetaxel compared to docetaxel plus a placebo in second-line treatment of advanced non-small cell lung cancer, irrespective of histology, with an acceptable safety profile. This article has for objective to summarize efficacy and safety data of the use of ramucirumab in combination with docetaxel in second line in NSCLC. Expert commentary: REVEL constitutes the first significant advance in second-line setting for patients eligible to anti-angiogenic therapy. The landscape of post-platinum therapy in NSCLC is considerably evolving and the role of ramucirumab or other anti-angiogenic agents as nintedanib in this setting has to be discussed for each patient with other available treatment options, among which immune checkpoints inhibitors, as well as the best treatment sequence.
Collapse
Affiliation(s)
- Valérie Paulus
- a Department of Medical Oncology , Léon Bérard Cancer Center , Lyon , France
| | - Virginie Avrillon
- a Department of Medical Oncology , Léon Bérard Cancer Center , Lyon , France
| | - Maurice Pérol
- a Department of Medical Oncology , Léon Bérard Cancer Center , Lyon , France
| |
Collapse
|
26
|
Sanchez-Gastaldo A, Gonzalez-Exposito R, Garcia-Carbonero R. Ramucirumab Clinical Development: an Emerging Role in Gastrointestinal Tumors. Target Oncol 2016; 11:479-87. [PMID: 26887374 DOI: 10.1007/s11523-016-0419-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Ramucirumab (IMC-1121B, LY3009806) is a fully human G1 monoclonal antibody that specifically targets vascular endotelial growth factor receptor 2 (VEGFR-2) with a substantially greater binding affinity than that of its natural ligands. Early clinical trials in patients with advanced solid tumors demonstrated that biologically relevant blood target concentrations are achievable with tolerable doses, and also showed some preliminary evidence of clinical activity. Several pivotal phase III trials have now been concluded and have led regulatory agencies to grant marketing authorization to ramucirumab for use as second line therapy in patients with advanced gastric or gastroesophageal junction (GEJ) adenocarcinoma (as single agent or in combination with paclitaxel), in patients with advanced colorectal carcinoma (CRC) (in combination with infusional fluorouracil and irinotecan (FOLFIRI regimen)) and in patients with advanced non-small cell lung cancer (NSCLC) (in combination with docetaxel). In contrast, ramucirumab failed to significantly improve survival versus placebo as second line therapy in patients with advanced hepatocellular carcinoma (HCC). The aim of this review is to summarize the clinical development and emerging role of ramucirumab in gastrointestinal (GI) tumors, including relevant aspects of its mechanism of action, pharmacology, safety profile, and antitumor activity in gastric, HCC, and CRC carcinomas.
Collapse
Affiliation(s)
- Amparo Sanchez-Gastaldo
- Center affiliated to the Red Tematica de Investigacion Cooperativa en Cancer (RTICC), Instituto de Salud Carlos III, Spanish Ministry of Science and Innovation, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| | - Reyes Gonzalez-Exposito
- Center affiliated to the Red Tematica de Investigacion Cooperativa en Cancer (RTICC), Instituto de Salud Carlos III, Spanish Ministry of Science and Innovation, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| | - Rocío Garcia-Carbonero
- Center affiliated to the Red Tematica de Investigacion Cooperativa en Cancer (RTICC), Instituto de Salud Carlos III, Spanish Ministry of Science and Innovation, Hospital Universitario Doce de Octubre, Madrid, Spain.
- Oncology Department, Hospital Universitario Doce de Octubre, Avenida de Cordoba km 5.4, 28041, Madrid, Spain.
| |
Collapse
|
27
|
Falcon BL, Chintharlapalli S, Uhlik MT, Pytowski B. Antagonist antibodies to vascular endothelial growth factor receptor 2 (VEGFR-2) as anti-angiogenic agents. Pharmacol Ther 2016; 164:204-25. [PMID: 27288725 DOI: 10.1016/j.pharmthera.2016.06.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Interaction of numerous signaling pathways in endothelial and mesangial cells results in exquisite control of the process of physiological angiogenesis, with a central role played by vascular endothelial growth factor receptor 2 (VEGFR-2) and its cognate ligands. However, deregulated angiogenesis participates in numerous pathological processes. Excessive activation of VEGFR-2 has been found to mediate tissue-damaging vascular changes as well as the induction of blood vessel expansion to support the growth of solid tumors. Consequently, therapeutic intervention aimed at inhibiting the VEGFR-2 pathway has become a mainstay of treatment in cancer and retinal diseases. In this review, we introduce the concepts of physiological and pathological angiogenesis, the crucial role played by the VEGFR-2 pathway in these processes, and the various inhibitors of its activity that have entered the clinical practice. We primarily focus on the development of ramucirumab, the antagonist monoclonal antibody (mAb) that inhibits VEGFR-2 and has recently been approved for use in patients with gastric, colorectal, and lung cancers. We examine in-depth the pre-clinical studies using DC101, the mAb to mouse VEGFR-2, which provided a conceptual foundation for the role of VEGFR-2 in physiological and pathological angiogenesis. Finally, we discuss further clinical development of ramucirumab and the future of targeting the VEGF pathway for the treatment of cancer.
Collapse
|
28
|
Diaz-Serrano A, Riesco-Martinez MC, Garcia-Carbonero R. The safety and efficacy of ramucirumab for the treatment of metastatic colorectal cancer. Expert Rev Anticancer Ther 2016; 16:585-95. [PMID: 27144874 DOI: 10.1080/14737140.2016.1182430] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Ramucirumab (IMC-1121B, LY3009806) is a fully humanized monoclonal antibody that targets the extracellular domain of vascular endothelial growth factor receptor 2 (VEGFR2), the principal mediator of VEGF-A downstream effects in cancer angiogenesis. Ramucirumab has been recently approved for use in combination with FOLFIRI for the treatment of patients with metastatic colorectal cancer (mCRC) whose disease has progressed on a first line bevacizumab-, oxaliplatin- and fluoropyrimidine-containing regimen. This approval was based on the results of the RAISE phase III placebo-controlled trial. This study demonstrated that the addition of ramucirumab to irinotecan-based chemotherapy significantly improved progression-free and overall survival of patients with mCRC, with manageable toxicity. AREAS COVERED The aim of this drug profile is to briefly summarize the pharmacology, clinical efficacy, safety and tolerability of ramucirumab in the context of metastatic colorectal cancer, and to provide some perspective regarding the role of the drug in clinical practice. Expert commentary: Pending issues that shall be addressed in the upcoming years include the optimization of ramucirumab dosing schedule, assessment of its role with other chemotherapy regimens or in other treatment settings, comparative evaluation of this agent with other antiangiogenics, and identification of predictive biomarkers to improve the therapeutic index and cost-effectiveness of this drug.
Collapse
|
29
|
Zheng Y, Sun Y, Yu X, Shao Y, Zhang P, Dai G, Fu J. Angiogenesis in Liquid Tumors: An In Vitro Assay for Leukemic-Cell-Induced Bone Marrow Angiogenesis. Adv Healthc Mater 2016; 5:1014-24. [PMID: 26924785 DOI: 10.1002/adhm.201501007] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 01/19/2016] [Indexed: 12/15/2022]
Abstract
The critical role of angiogenesis for solid tumor growth and metastatic spread has been well established. In contrast, even though increased vascularity has been commonly observed in bone marrows of patients with hematological malignancies (liquid tumors), the pathophysiology of leukemia-induced angiogenesis in the bone marrow remains elusive. This paper demonstrates the usage of a microengineered 3D biomimetic model to study leukemic-cell-induced bone marrow angiogenesis. Rational design of the 3D angiogenesis chip incorporating endothelial cells (ECs), leukemic cells, and bone marrow stromal fibroblasts provide an efficient biomimetic means to promote and visualize early angiogenic processes. Morphological features of angiogenesis induced by three different leukemic cell lines (U937, HL60, and K562) are investigated and compared. Quantitative measurements of angiogenic factors secreted from monocultures and cocultures of leukemic cells with bone marrow stromal fibroblasts suggest a synergistic relationship between ECs, leukemic cells, and bone marrow stromal fibroblasts for angiogenic induction, and also confirm the necessity of conducting functional angiogenic assays in proper 3D biomimetic cell culture systems like the one developed in this work.
Collapse
Affiliation(s)
- Yi Zheng
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yubing Sun
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Xinwei Yu
- Department of Applied Physics, University of Science and Technology of China, Anhui, Hefei, 230026, P. R. China
| | - Yue Shao
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ping Zhang
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Guohao Dai
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
30
|
Gambardella V, Tarazona N, Cejalvo JM, Roselló S, Cervantes A. Clinical pharmacokinetics and pharmacodynamics of ramucirumab in the treatment of colorectal cancer. Expert Opin Drug Metab Toxicol 2016; 12:449-56. [PMID: 26895445 DOI: 10.1517/17425255.2016.1156084] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Colorectal cancer is the third most common cancer worldwide. The prognosis of colorectal cancer patients still remains dismal and half of them will develop metastatic disease. Angiogenesis plays an essential role in colorectal tumorigenesis, and the VEGF pathway is one of the targets that has been validated up to now. The use of antiangiogenics along with chemotherapy has become an accepted standard for colorectal cancer. AREAS COVERED This review discusses the efficacy and safety profile of ramucirumab, a fully human immunoglobulin G1 monoclonal antibody against the vascular endothelial growth factor receptor-2 (VEGFR-2), for the treatment of second-line metastatic colorectal cancer upon progression to first-line chemotherapy including anti-angiogenics. EXPERT OPINION Ramucirumab in combination with chemotherapy represents a valid option in second-line treatment of advanced colorectal cancer patients, who progressed on previous bevacizumab-based combinations. This agent demonstrates a similar benefit in terms of overall survival to other angiogenesis inhibitors (bevacizumab and ziv-aflibercept) used in this setting.
Collapse
MESH Headings
- Angiogenesis Inhibitors/administration & dosage
- Angiogenesis Inhibitors/pharmacokinetics
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal, Humanized
- Clinical Trials, Phase I as Topic
- Clinical Trials, Phase II as Topic
- Clinical Trials, Phase III as Topic
- Colorectal Neoplasms/drug therapy
- Disease Models, Animal
- Drug Evaluation, Preclinical
- Humans
- Neovascularization, Pathologic/drug therapy
- Randomized Controlled Trials as Topic
- Ramucirumab
Collapse
Affiliation(s)
- Valentina Gambardella
- a Department of Medical Oncology , Biomedical Research Institute INCLIVA, University of Valencia , Valencia , Spain
| | - Noelia Tarazona
- a Department of Medical Oncology , Biomedical Research Institute INCLIVA, University of Valencia , Valencia , Spain
| | - Juan Miguel Cejalvo
- a Department of Medical Oncology , Biomedical Research Institute INCLIVA, University of Valencia , Valencia , Spain
| | - Susana Roselló
- a Department of Medical Oncology , Biomedical Research Institute INCLIVA, University of Valencia , Valencia , Spain
| | - Andrés Cervantes
- a Department of Medical Oncology , Biomedical Research Institute INCLIVA, University of Valencia , Valencia , Spain
| |
Collapse
|
31
|
Membrane-proximal TRAIL species are incapable of inducing short circuit apoptosis signaling: Implications for drug development and basic cytokine biology. Sci Rep 2016; 6:22661. [PMID: 26935795 PMCID: PMC4776141 DOI: 10.1038/srep22661] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/18/2016] [Indexed: 02/02/2023] Open
Abstract
TRAIL continues to garner substantial interest as a recombinant cancer therapeutic while the native cytokine itself serves important tumor surveillance functions when expressed in membrane-anchored form on activated immune effector cells. We have recently developed the genetically stabilized TRAIL platform TR3 in efforts to improve the limitations associated with currently available drug variants. While in the process of characterizing mesothelin-targeted TR3 variants using a single chain antibody (scFv) delivery format (SS-TR3), we discovered that the membrane-tethered cytokine had a substantially increased activity profile compared to non-targeted TR3. However, cell death proceeded exclusively via a bystander mechanism and protected the mesothelin-positive targets from apoptosis rather than leading to their elimination. Incorporation of a spacer-into the mesothelin surface antigen or the cancer drug itself-converted SS-TR3 into a cis-acting phenotype. Further experiments with membrane-anchored TR3 variants and the native cytokine confirmed our hypothesis that membrane-proximal TRAIL species lack the capacity to physically engage their cognate receptors coexpressed on the same cell membrane. Our findings not only provide an explanation for the “peaceful” coexistence of ligand and receptor of a representative member of the TNF superfamily but give us vital clues for the design of activity-enhanced TR3-based cancer therapeutics.
Collapse
|
32
|
Roviello G, Petrioli R, Marano L, Polom K, Marrelli D, Perrella A, Roviello F. Angiogenesis inhibitors in gastric and gastroesophageal junction cancer. Gastric Cancer 2016; 19:31-41. [PMID: 26329368 DOI: 10.1007/s10120-015-0537-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 08/15/2015] [Indexed: 02/07/2023]
Abstract
Despite significant improvements in systemic chemotherapy during the past two decades, the prognosis of patients with advanced gastric and gastroesophageal junction adenocarcinoma remains poor. Because of molecular heterogeneity, it is essential to classify tumors based on the underlying oncogenic pathways and to develop targeted therapies acting on individual tumors. Unfortunately, although a number of molecular targets have been studied, very few of these agents can be used in a clinical setting. In this review, we summarize the available data on anti-angiogenic agents in advanced/metastatic gastric cancer.
Collapse
Affiliation(s)
| | - Roberto Petrioli
- Medical Oncology Unit, University of Siena, Viale Bracci 11, 53100, Siena, Italy
| | - Luigi Marano
- Unit of General and Minimally Invasive Surgery, Department of Medical, Surgical and Neuroscience, University of Siena, Viale Bracci 11, Siena, 53100, Italy
| | - Karol Polom
- Unit of General and Minimally Invasive Surgery, Department of Medical, Surgical and Neuroscience, University of Siena, Viale Bracci 11, Siena, 53100, Italy
| | - Daniele Marrelli
- Section of Advanced Surgical Oncology, Department of Medical, Surgical and Neurosciences, University of Siena, Viale Bracci 11, Siena, 53100, Italy
| | - Armando Perrella
- Medical Oncology Unit, University of Siena, Viale Bracci 11, 53100, Siena, Italy
| | - Franco Roviello
- Unit of General and Minimally Invasive Surgery, Department of Medical, Surgical and Neuroscience, University of Siena, Viale Bracci 11, Siena, 53100, Italy
| |
Collapse
|
33
|
Roviello G, Polom K, Petrioli R, Marano L, Marrelli D, Paganini G, Savelli V, Generali D, De Franco L, Ravelli A, Roviello F. Monoclonal antibodies-based treatment in gastric cancer: current status and future perspectives. Tumour Biol 2016; 37:127-140. [PMID: 26566626 DOI: 10.1007/s13277-015-4408-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 11/09/2015] [Indexed: 12/11/2022] Open
Abstract
Gastric cancer (GC) is the second leading cause of cancer-related death, and despite having improved treatment modalities over the last decade, for most patients, only modest improvements have been seen in overall survival. Recent progress in understanding the molecular biology of GC and the related signaling pathways offers, from the clinical point of view, promising advances for selected groups of patients. In the past, targeted therapies have significantly impacted the treatment strategy of several common solid tumors such as breast, colorectal, and lung cancers. Unfortunately, translational and clinical research shows fewer encouraging targeted treatments with regards to the GC. To date, only two monoclonal antibodies (mAb), named trastuzumab and ramucirumab, are approved for the treatment of advanced GC, suggesting that in GC, maybe more than in other cancers, effective targeted therapy requires patient selection based on precise predictive molecular biomarkers. The aim of this review is to summarize the available data on the clinical advantages offered by the use of mAbs in the treatment of advanced/metastatic GC. Future perspective is also discussed.
Collapse
Affiliation(s)
- Giandomenico Roviello
- Section of pharmacology and University Center DIFF-Drug Innovation Forward Future, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25124, Brescia, Italy.
| | - Karol Polom
- Department of Medical, Surgical and Neuroscience; Unit of General and Minimally Invasive Surgery, University of Siena, Viale Bracci 11, 53100, Siena, Italy
| | - Roberto Petrioli
- Medical Oncology Unit, University of Siena, Viale Bracci 11, 53100, Siena, Italy
| | - Luigi Marano
- General, Minimally Invasive and Robotic Surgery, Department of Surgery, "San Matteo degli Infermi" Hospital, ASL Umbria 2, 06049, Spoleto, Italy
| | - Daniele Marrelli
- Department of Medical, Surgical and Neurosciences, Section of Advanced Surgical Oncology, University of Siena, Viale Bracci 11, 53100, Siena, Italy
| | - Giovanni Paganini
- Unit of General Medicine, Azienda Ospedaliera "C. Poma " Presidio ospedaliero di Pieve di Coriano, Mantova, Italy
| | - Vinno Savelli
- Department of Surgery and Bioengineering, Section of Surgery, University of Siena, Viale Bracci 11, 53100, Siena, Italy
| | - Daniele Generali
- Department of Medical, Surgery and Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - Lorenzo De Franco
- Department of Medical, Surgical and Neurosciences, Section of Advanced Surgical Oncology, University of Siena, Viale Bracci 11, 53100, Siena, Italy
| | - Andrea Ravelli
- Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy
| | - Franco Roviello
- Department of Medical, Surgical and Neuroscience; Unit of General and Minimally Invasive Surgery, University of Siena, Viale Bracci 11, 53100, Siena, Italy
| |
Collapse
|
34
|
Ma Q, Chen W, Chen W. Anti-tumor angiogenesis effect of a new compound: B-9-3 through interference with VEGFR2 signaling. Tumour Biol 2015; 37:6107-16. [DOI: 10.1007/s13277-015-4473-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 11/18/2015] [Indexed: 12/15/2022] Open
|
35
|
Xu WW, Li B, Lam AKY, Tsao SW, Law SYK, Chan KW, Yuan QJ, Cheung ALM. Targeting VEGFR1- and VEGFR2-expressing non-tumor cells is essential for esophageal cancer therapy. Oncotarget 2015; 6:1790-805. [PMID: 25595897 PMCID: PMC4359332 DOI: 10.18632/oncotarget.2781] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 11/19/2014] [Indexed: 11/25/2022] Open
Abstract
Increasing appreciation of tumor heterogeneity and the tumor-host interaction has stimulated interest in developing novel therapies that target both tumor cells and tumor microenvironment. Bone marrow derived cells (BMDCs) constitute important components of the tumor microenvironment. In this study, we aim to investigate the significance of VEGFR1- and VEGFR2-expressing non-tumor cells, including BMDCs, in esophageal cancer (EC) progression and in VEGFR1/VEGFR2-targeted therapies. Here we report that VEGFR1 or VEGFR2 blockade can significantly attenuate VEGF-induced Src and Erk signaling, as well as the proliferation and migration of VEGFR1⁺ and VEGFR2⁺ bone marrow cells and their pro-invasive effect on cancer cells. Importantly, our in vivo data show for the first time that systemic blockade of VEGFR1⁺ or VEGFR2⁺ non-tumor cells with neutralizing antibodies is sufficient to significantly suppress esophageal tumor growth, angiogenesis and metastasis in mice. Moreover, our tissue microarray study of human EC clinical specimens showed the clinicopathological significance of VEGFR1 and VEGFR2 in EC, which suggest that anti-VEGFR1/VEGFR2 therapies may be particularly beneficial for patients with aggressive EC. In conclusion, this study demonstrates the important contributions of VEGFR1⁺ and VEGFR2⁺ non-tumor cells in esophageal cancer progression, and substantiates the validity of these receptors as therapeutic targets for this deadly disease.
Collapse
Affiliation(s)
- Wen Wen Xu
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.,The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), China
| | - Bin Li
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.,The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), China.,Centre for Cancer Research, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Alfred K Y Lam
- Department of Pathology, Griffith Medical School and Griffith Health Institute, Gold Coast Campus, Gold Coast, QLD 4222, Australia
| | - Sai Wah Tsao
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.,Centre for Cancer Research, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Simon Y K Law
- Centre for Cancer Research, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.,Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kwok Wah Chan
- The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), China.,Centre for Cancer Research, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Qiu Ju Yuan
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Annie L M Cheung
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.,The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), China.,Centre for Cancer Research, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| |
Collapse
|
36
|
Larkins E, Scepura B, Blumenthal GM, Bloomquist E, Tang S, Biable M, Kluetz P, Keegan P, Pazdur R. U.S. Food and Drug Administration Approval Summary: Ramucirumab for the Treatment of Metastatic Non-Small Cell Lung Cancer Following Disease Progression On or After Platinum-Based Chemotherapy. Oncologist 2015; 20:1320-5. [PMID: 26446239 PMCID: PMC4718430 DOI: 10.1634/theoncologist.2015-0221] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/18/2015] [Indexed: 11/17/2022] Open
Abstract
UNLABELLED On December 12, 2014, the U.S. Food and Drug Administration (FDA) approved ramucirumab for use in combination with docetaxel for the treatment of patients with metastatic non-small cell lung cancer (NSCLC) with disease progression on or after platinum-based chemotherapy. Patients with epidermal growth factor receptor or anaplastic lymphoma kinase genomic tumor aberrations should have disease progression on FDA-approved therapy for these aberrations prior to receiving ramucirumab. This approval was based on an improvement in overall survival (OS) with an acceptable toxicity profile in a randomized, multicenter, double-blinded, placebo-controlled trial of 1,253 patients with metastatic NSCLC previously treated with a platinum-based combination therapy. Patients were randomized 1:1 to receive either ramucirumab in combination with docetaxel or placebo in combination with docetaxel. The primary endpoint was OS. Patients who received ramucirumab in combination with docetaxel had improved OS (hazard ratio [HR]: 0.86; 95% confidence interval [CI]: 0.75, 0.98). Median OS was 10.5 months on the ramucirumab plus docetaxel arm versus 9.1 months on the placebo plus docetaxel arm. The most frequent (≥ 30%) adverse reactions in ramucirumab-treated patients were fatigue, neutropenia, and diarrhea. The most frequent (≥ 5%) grade 3 and 4 adverse reactions in the ramucirumab arm were fatigue, neutropenia, febrile neutropenia, leukopenia, and hypertension. IMPLICATIONS FOR PRACTICE This report presents key information on the U.S. Food and Drug Administration approval of ramucirumab, a monoclonal antibody targeting vascular endothelial growth factor receptor-2, given in combination with docetaxel for the treatment of patients with metastatic non-small cell lung cancer whose disease has progressed on or after platinum-based chemotherapy. This report specifically addresses the issues of safety in patients with squamous cell tumors, effect of treatment in elderly patients, and uncertainties regarding effects in patients with tumors harboring epidermal growth factor receptor or anaplastic lymphoma kinase genomic tumor aberrations.
Collapse
Affiliation(s)
- Erin Larkins
- Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Barbara Scepura
- Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Gideon M Blumenthal
- Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Erik Bloomquist
- Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Shenghui Tang
- Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Missiratch Biable
- Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Paul Kluetz
- Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Patricia Keegan
- Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Richard Pazdur
- Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
37
|
ElHalawani H, Abdel-Rahman O. Critical evaluation of ramucirumab in the treatment of advanced gastric and gastroesophageal cancers. Ther Clin Risk Manag 2015; 11:1123-32. [PMID: 26251608 PMCID: PMC4524527 DOI: 10.2147/tcrm.s71045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Gastric (GC) and gastroesophageal junction (GEJ) cancers are two global health problems with a relatively high mortality, particularly in the advanced stage. Inhibition of angiogenesis is now contemplated as a classic treatment preference for myriad tumor types encompassing renal cell carcinoma, non-small cell lung cancer, colorectal cancer, glioblastoma, and ovarian cancer, among others. Bevacizumab and ramucirumab have been widely investigated in GC and GEJ cancer, with some controversy about their therapeutic role. Ramucirumab is a monoclonal antibody for vascular endothelial growth factor receptor-2, with demonstrated activity both as a monotherapy and as a part of combination strategy in the management of advanced GC/GEJ cancer. In this review article, we present a critical evaluation of the preclinical and clinical data underlying the use of this drug in this indication. Moreover, we provide a spotlight on the future perspectives in systemic therapy for advanced GC/GEJ cancer.
Collapse
Affiliation(s)
- Hesham ElHalawani
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Omar Abdel-Rahman
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
38
|
Lee WS, Pyun BJ, Kim SW, Shim SR, Nam JR, Yoo JY, Jin Y, Jin J, Kwon YG, Yun CO, Nam DH, Oh K, Lee DS, Lee SH, Yoo JS. TTAC-0001, a human monoclonal antibody targeting VEGFR-2/KDR, blocks tumor angiogenesis. MAbs 2015; 7:957-68. [PMID: 25942475 DOI: 10.1080/19420862.2015.1045168] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Angiogenesis is one of the most important processes for cancer cell survival, tumor growth and metastasis. Vascular endothelial growth factor (VEGF) and its receptor, particularly VEGF receptor-2 (VEGFR-2, or kinase insert domain-containing receptor, KDR), play critical roles in tumor-associated angiogenesis. We developed TTAC-0001, a human monoclonal antibody against VEGFR-2/KDR from a fully human naïve single-chain variable fragment phage library. TTAC-0001 was selected as a lead candidate based on its affinity, ligand binding inhibition and inhibition of VEGFR-2 signal in human umbilical vein endothelial cells (HUVEC). TTAC-0001 inhibited binding of VEGF-C and VEGF-D to VEGFR-2 in addition to VEGF-A. It binds on the N-terminal regions of domain 2 and domain 3 of VEGFR-2. It could inhibit the phosphorylation of VEGFR-2/KDR and ERK induced by VEGF in HUVEC. TTAC-0001 also inhibited VEGF-mediated endothelial cell proliferation, migration and tube formation in vitro, as well as ex vivo vessel sprouting from rat aortic rings and neovascularization in mouse matrigel model in vivo. Our data indicates that TTAC-0001 blocks the binding of VEGFs to VEGFR-2/KDR and inhibits VEGFR-induced signaling pathways and angiogenesis. Therefore, these data strongly support the further development of TTAC-0001 as an anti-cancer agent in the clinic.
Collapse
Affiliation(s)
- Weon Sup Lee
- a PharmAbcine, Inc. , #402; DaejeonBioventure Town; Jeonmin-dong; Yusung-gu; Daejeon , Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Fontana E, Sclafani F, Cunningham D. Anti-angiogenic therapies for advanced esophago-gastric cancer. Indian J Med Paediatr Oncol 2014; 35:253-62. [PMID: 25538401 PMCID: PMC4264270 DOI: 10.4103/0971-5851.144985] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Neo-vascularization is a vital process for tumor growth and development which involves the interaction between tumor cells and stromal endothelial cells through several growth factors and membranous receptors which ultimately activate pro-angiogenic intracellular signaling pathways. Inhibition of angiogenesis has become a standard treatment option for several tumor types including colorectal cancer, glioblastoma and ovarian cancer. In gastric cancer, the therapeutic role of anti-angiogenic agents is more controversial. Bevacizumab and ramucirumab, two monoclonal antibodies, which target vascular endothelial growth factor-A and vascular endothelial growth factor receptor-2, respectively, have been demonstrated antitumor activity in patients with tumors of the stomach or esophagogastric junction. However, especially for bevacizumab, this antitumor activity has not consistently translated into a survival advantage over standard treatment in randomized trials. In this article, we provide an overview of the role of angiogenesis in gastric cancer and discuss the results of clinical trials that investigated safety and effectiveness of antiangiogenic therapies in this disease. A review of the literature has been done using PubMed, ClinicalTrials.gov website and the ASCO Annual Meeting Library.
Collapse
Affiliation(s)
- Elisa Fontana
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Francesco Sclafani
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - David Cunningham
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| |
Collapse
|
40
|
Aprile G, Rijavec E, Fontanella C, Rihawi K, Grossi F. Ramucirumab: preclinical research and clinical development. Onco Targets Ther 2014; 7:1997-2006. [PMID: 25378934 PMCID: PMC4218907 DOI: 10.2147/ott.s61132] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Ramucirumab (IMC-1121B, LY3009806), a fully humanized monoclonal antibody directed against the extracellular domain of vascular endothelial growth factor receptor 2 (VEGFR-2), is a new therapeutic option that selectively inhibits the human VEGFR-2 with a much greater affinity than its natural ligands. Based on the promising results of both preclinical and early clinical studies, ramucirumab has been tested in different tumor types either alone or in combination with chemotherapy. While it has recently been granted its first US Food and Drug Administration approval for use as a single agent in patients with advanced or metastatic gastric cancer or gastroesophageal junction carcinoma, its role for metastatic breast cancer or advanced non-small-cell lung cancer is still debated. The aims of this review are to recall and discuss the most significant preclinical and clinical studies that led to the development of ramucirumab and to present the results of the randomized clinical trials that have tested its efficacy in different malignancies, including gastric and lung cancer.
Collapse
Affiliation(s)
- Giuseppe Aprile
- Department of Oncology, University Hospital of Udine, Udine, Italy
| | - Erika Rijavec
- Lung Cancer Unit, National Cancer Institut “San Martino”, Genoa, Italy
| | | | - Karim Rihawi
- Department of Oncology, University Hospital of Udine, Udine, Italy
| | - Francesco Grossi
- Lung Cancer Unit, National Cancer Institut “San Martino”, Genoa, Italy
| |
Collapse
|
41
|
Nixon AE, Sexton DJ, Ladner RC. Drugs derived from phage display: from candidate identification to clinical practice. MAbs 2014; 6:73-85. [PMID: 24262785 DOI: 10.4161/mabs.27240] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Phage display, one of today’s fundamental drug discovery technologies, allows identification of a broad range of biological drugs, including peptides, antibodies and other proteins, with the ability to tailor critical characteristics such as potency, specificity and cross-species binding. Further, unlike in vivo technologies, generating phage display-derived antibodies is not restricted by immunological tolerance. Although more than 20 phage display-derived antibody and peptides are currently in late-stage clinical trials or approved, there is little literature addressing the specific challenges and successes in the clinical development of phage-derived drugs. This review uses case studies, from candidate identification through clinical development, to illustrate the utility of phage display as a drug discovery tool, and offers a perspective for future developments of phage display technology.
Collapse
|
42
|
Aoyagi K, Kouhuji K, Kizaki J, Isobe T, Hashimoto K, Shirouzu K. Molecular targeting to treat gastric cancer. World J Gastroenterol 2014; 20:13741-55. [PMID: 25320512 PMCID: PMC4194558 DOI: 10.3748/wjg.v20.i38.13741] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 01/13/2014] [Accepted: 05/23/2014] [Indexed: 02/06/2023] Open
Abstract
Trastuzumab that targets human epidermal growth factor receptor 2 (HER2) protein is the only approved molecular targeting agent for treating gastric cancer in Japan and the outcomes have been favorable. However, trastuzumab is effective for only 10% to 20% of the population with gastric cancer that expresses HER2 protein. Molecular targeting therapy with bevacizumab against vascular endothelial growth factors (VEGF) and with cetuximab and panitumumab against the epidermal growth factors pathway that have been approved for treating colorectal cancer are not considered effective for treating gastric cancer according to several clinical trials. However, ramucirumab that targets VEGF receptor-2 prolonged overall survival in a large phase III clinical trial and it might be an effective molecular targeting therapy for gastric cancer. The significance of molecular targeting therapy for gastric cancer remains controversial. A large-scale randomized clinical trial of novel molecular targeting agents with which to treat gastric cancer is needed.
Collapse
|
43
|
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
|
44
|
Xiang Y, Li Q, Huang D, Tang X, Wang L, Shi Y, Zhang W, Yang T, Xiao C, Wang J. Preparation and antitumor effect of a toxin-linked conjugate targeting vascular endothelial growth factor receptor and urokinase plasminogen activator. Exp Biol Med (Maywood) 2014; 240:160-8. [PMID: 25125500 DOI: 10.1177/1535370214547154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The aberrant signaling activation of vascular endothelial growth factor receptor (VEGFR) and urokinase plasminogen activator (uPA) is a common characteristic of many tumors, including lung cancer. Accordingly, VEGFR and uPA have emerged as attractive targets for tumor. KDR (Flk-1/VEGFR-2), a member of the VEGFR family, has been recognized as an important target for antiangiogenesis in tumor. In this study, a recombinant immunotoxin was produced to specifically target KDR-expressing tumor vascular endothelial cells and uPA-expressing tumor cells and mediate antitumor angiogenesis and antitumor effect. Based on its potent inhibitory effect on protein synthesis, Luffin-beta (Lβ) ribosome-inactivating protein was selected as part of a recombinant fusion protein, a single-chain variable fragment against KDR (KDRscFv)-uPA cleavage site (uPAcs)-Lβ-KDEL (named as KPLK). The KDRscFv-uPAcs-Lβ-KDEL (KPLK) contained a single-chain variable fragment (scFv) against KDR, uPAcs, Lβ, and the retention signal for endoplasmic reticulum proteins KDEL (Lys-Asp-Glu-Leu). The KPLK-expressing vector was expressed in Escherichia coli, and the KPLK protein was isolated with nickel affinity chromatography and gel filtration chromatography. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis test demonstrated KPLK was effectively expressed. Result of in vitro cell viability assay on non-small cell lung cancer (NSCLC) H460 cell line (uPA-positive cell) revealed that KPLK significantly inhibited cell proliferation, induced apoptosis, and accumulated cells in S and G2/M phases, but the normal cell line (human submandibular gland cell) was unaffected. These effects were enhanced when uPA was added to digest KPLK to release Lβ. For in vivo assay of KPLK, subcutaneous xenograft tumor model of nude mice were established with H460 cells. Growth of solid tumors was significantly inhibited in animals treated with KPLK up to 21 days, tumor weights were decreased, and the expression of angiogenesis marker CD31 was downregulated; meanwhile, the apoptosis-related protein casspase-3 was upregulated. These results suggested that the recombinant KPLK may have therapeutic applications on tumors, especially uPA-overexpressing ones.
Collapse
Affiliation(s)
- Ying Xiang
- Department of Biotherapy and Hemo-oncology, Chongqing Cancer Institute, Chongqing 400030, China
| | - Qiying Li
- Department of Biotherapy and Hemo-oncology, Chongqing Cancer Institute, Chongqing 400030, China
| | - Dehong Huang
- Department of Biotherapy and Hemo-oncology, Chongqing Cancer Institute, Chongqing 400030, China
| | - Xianjun Tang
- Department of Biotherapy and Hemo-oncology, Chongqing Cancer Institute, Chongqing 400030, China
| | - Li Wang
- Department of Biotherapy and Hemo-oncology, Chongqing Cancer Institute, Chongqing 400030, China
| | - Yang Shi
- Department of Biotherapy and Hemo-oncology, Chongqing Cancer Institute, Chongqing 400030, China
| | - Wenjun Zhang
- Department of Biotherapy and Hemo-oncology, Chongqing Cancer Institute, Chongqing 400030, China
| | - Tao Yang
- Department of Biotherapy and Hemo-oncology, Chongqing Cancer Institute, Chongqing 400030, China
| | - Chunyan Xiao
- Department of Biotherapy and Hemo-oncology, Chongqing Cancer Institute, Chongqing 400030, China
| | - Jianghong Wang
- Center of Endoscopy Examination & Therapy, Chongqing Cancer Institute, Chongqing 400030, China
| |
Collapse
|
45
|
Aprile G, Bonotto M, Ongaro E, Pozzo C, Giuliani F. Critical appraisal of ramucirumab (IMC-1121B) for cancer treatment: from benchside to clinical use. Drugs 2013; 73:2003-15. [PMID: 24277700 DOI: 10.1007/s40265-013-0154-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Although antiangiogenic treatments have produced milestone advances in the treatment of several diseases, and have significantly extended the median survival of cancer patients, these agents share some weaknesses, including a limited impact on the overall cure rate, a fleeting effect because of redundant pathways or early appearance of resistance mechanisms, and the lack of predictive factors for treatment selection. Recent data suggest that antibodies targeting the vascular endothelial growth factor axis exert their activity through the inhibition of vascular endothelial growth factor receptor-2 phosphorylation, which has a pivotal role in the neoangiogenic process. Ramucirumab, a fully humanized monoclonal antibody specifically directed against the extracellular domain of the receptor, administered intravenously every 2 or 3 weeks, is emerging as a novel antiangiogenic opportunity. Starting with preclinical data and early clinical results, this concise review focuses on the development of the novel compound across multiple cancers (including gastrointestinal malignancies, breast cancer, lung carcinoma, and genitourinary tumors), and presents available data from randomized phase II and phase III trials. REGARD was the first phase III study to report on the efficacy of single-agent ramucirumab in patients with advanced cancer. Many other ongoing phase III trials are testing the efficacy of this interesting antiangiogenic compound as a single agent or in combination with chemotherapy in different cancer types.
Collapse
Affiliation(s)
- Giuseppe Aprile
- Department of Medical Oncology, University and General Hospital, Piazzale S Maria della Misericordia, 33100, Udine, Italy,
| | | | | | | | | |
Collapse
|
46
|
Tang YT, Jiang F, Guo L, Si MY, Jiao XY. Expression and significance of vascular endothelial growth factor A and C in leukemia central nervous system metastasis. Leuk Res 2013; 37:359-66. [PMID: 23137522 DOI: 10.1016/j.leukres.2012.10.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 10/03/2012] [Accepted: 10/12/2012] [Indexed: 02/05/2023]
Abstract
Metastasis to the central nervous system (CNS) is an obstacle for leukemia treatment, the mechanisms of which remain to be elucidated. VEGF-A and VEGF-C are suspected to participate in this process. Paired of cerebrospinal fluid (CSF) and serum samples were collected from leukemia and control cases. Levels of VEGF-A and VEGF-C in both CSF (VEGF-ACSF, VEGF-CCSF) and serum (VEGF-ASerum, VEGF-CSerum) were detected by ELISA. Our data show that higher levels of VEGF-ACSF are closely related to CNS leukemia (CNSL), and VEGF-ACSF may be a better predictor than the other risk factors elucidating the pathogenesis and development of CNSL.
Collapse
Affiliation(s)
- Yue-Ting Tang
- Department of Hematology Laboratory, First Affiliated Hospital of Shantou University Medical College, Guangdong, China
| | | | | | | | | |
Collapse
|
47
|
Fang B, Jiang L, Zhang M, Ren FZ. A novel cell-penetrating peptide TAT-A1 delivers siRNA into tumor cells selectively. Biochimie 2013; 95:251-7. [DOI: 10.1016/j.biochi.2012.09.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 09/17/2012] [Indexed: 12/20/2022]
|
48
|
Ozawa T, Piao X, Kobayashi E, Zhou Y, Sakurai H, Andoh T, Jin A, Kishi H, Muraguchi A. A novel rabbit immunospot array assay on a chip allows for the rapid generation of rabbit monoclonal antibodies with high affinity. PLoS One 2012; 7:e52383. [PMID: 23300658 PMCID: PMC3530603 DOI: 10.1371/journal.pone.0052383] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 11/12/2012] [Indexed: 12/25/2022] Open
Abstract
Antigen-specific rabbit monoclonal antibodies (RaMoAbs) are useful due to their high specificity and high affinity, and the establishment of a comprehensive and rapid RaMoAb generation system has been highly anticipated. Here, we present a novel system using immunospot array assay on a chip (ISAAC) technology in which we detect and retrieve antigen-specific antibody-secreting cells from the peripheral blood lymphocytes of antigen-immunized rabbits and produce antigen-specific RaMoAbs with 10–12 M affinity within a time period of only 7 days. We have used this system to efficiently generate RaMoAbs that are specific to a phosphorylated signal-transducing molecule. Our system provides a new method for the comprehensive and rapid production of RaMoAbs, which may contribute to laboratory research and clinical applications.
Collapse
Affiliation(s)
- Tatsuhiko Ozawa
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Xiuhong Piao
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Eiji Kobayashi
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Yue Zhou
- Department of Cancer Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Hiroaki Sakurai
- Department of Cancer Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Tsugunobu Andoh
- Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Aishun Jin
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
- Department of Immunology, College of Basic Medical Sciences, Harbin Medical University, Nangang District, Harbin, China
| | - Hiroyuki Kishi
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
- * E-mail:
| | - Atsushi Muraguchi
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| |
Collapse
|
49
|
Human ESC-derived hemogenic endothelial cells undergo distinct waves of endothelial to hematopoietic transition. Blood 2012; 121:770-80. [PMID: 23169780 DOI: 10.1182/blood-2012-07-444208] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Several studies have demonstrated that hematopoietic cells originate from endotheliumin early development; however, the phenotypic progression of progenitor cells during human embryonic hemogenesis is not well described. Here, we define the developmental hierarchy among intermediate populations of hematopoietic progenitor cells (HPCs) derived from human embryonic stem cells (hESCs). We genetically modified hESCs to specifically demarcate acquisition of vascular (VE-cadherin) and hematopoietic (CD41a) cell fate and used this dual-reporting transgenic hESC line to observe endothelial to hematopoietic transition by real-time confocal microscopy. Live imaging and clonal analyses revealed a temporal bias in commitment of HPCs that recapitulates discrete waves of lineage differentiation noted during mammalian hemogenesis. Specifically, HPCs isolated at later time points showed reduced capacity to form erythroid/ megakaryocytic cells and exhibited a tendency toward myeloid fate that was enabled by expression of the Notch ligand Dll4 on hESC-derived vascular feeder cells. These data provide a framework for defining HPC lineage potential, elucidate a molecular contribution from the vascular niche in promoting hematopoietic lineage progression, and distinguish unique subpopulations of hemogenic endothelium during hESC differentiation. KEY POINTS Live imaging of endothelial to hematopoietic conversion identifies distinct subpopulations of hESC-derived hemogenic endothelium. Expression of the Notch ligand DII4 on vascular ECs drives induction of myeloid fate from hESC-derived hematopoietic progenitors.
Collapse
|
50
|
Sun W. Angiogenesis in metastatic colorectal cancer and the benefits of targeted therapy. J Hematol Oncol 2012; 5:63. [PMID: 23057939 PMCID: PMC3537532 DOI: 10.1186/1756-8722-5-63] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 09/26/2012] [Indexed: 02/08/2023] Open
Abstract
The diverse pathways and molecules involved in angiogenesis, the formation of new blood vessels, have been targeted for the treatment of colorectal and other cancers. Vascular endothelial growth factor (VEGF)-A binding to VEGF receptor (VEGFR)-2 is believed to be the key signaling pathway mediating angiogenesis. Other VEGF pathways involved in angiogenesis include VEGF-A, VEGF-B, and placental growth factor binding to VEGFR-1, and VEGF-C and VEGF-D binding to VEGFR-2 and VEGFR-3. VEGF signaling also intersects with other pathways, including angiopoietin/Tie, Notch, hypoxia-inducible factor, and integrin pathways. The roles of these pathways in tumor angiogenesis and in various human cancers will be explored in this article. In addition, preclinical and clinical data on bevacizumab, aflibercept (known as ziv-aflibercept in the US), and investigational antiangiogenic agents in development for the treatment of colorectal and other cancers will be reviewed.
Collapse
Affiliation(s)
- Weijing Sun
- University of Pittsburgh School of Medicine, UPMC Cancer Pavilion, 5150 Centre Avenue, Fifth Floor, Pittsburgh, PA 15232, USA.
| |
Collapse
|