1
|
Wu Y, Chen Z, Shi M, Qiu S, Zhang Y. Nimotuzumab and bevacizumab combined with temozolomide and radiotherapy in patients with newly diagnosed glioblastoma multiforme: a retrospective single-arm study. J Neurooncol 2025:10.1007/s11060-024-04932-8. [PMID: 39760795 DOI: 10.1007/s11060-024-04932-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 12/26/2024] [Indexed: 01/07/2025]
Abstract
PURPOSE Glioblastoma (GBM), the most common malignant tumor of the central nervous system (CNS) in adults, continues to result in poor survival rates despite standard treatment. Advancements in understanding GBM's molecular complexity have increased interest in targeted therapeutic approaches. This retrospective, single-center, single-arm study combined nimotuzumab and bevacizumab with radiotherapy (RT) and temozolomide (TMZ) for the treatment of newly diagnosed GBM. The objectives were to determine the efficacy of this treatment combination and the associated toxicity. METHODS A retrospective analysis of clinical data of GBM patients treated at our institution from September 2021 to May 2023 with postoperative combination therapy of nimotuzumab, bevacizumab, and TMZ concurrent with RT, as well as maintenance therapy with bevacizumab and TMZ. Follow-ups were performed every 3 to 6 months via hospital visits and telephone interviews. The primary endpoints were overall survival (OS) and progression-free survival (PFS). The secondary endpoint was the incidence of adverse events (AEs). RESULTS A total of 18 patients were included. The median follow-up time was 23 months. The one-year PFS rate was 77.8%, and the one-year OS rate was 94.4%. The median PFS was 18 months (95%CI, 15.9-20.1), and the median OS was 28 months (95%CI, 18.9-37.1). All AEs were controllable. CONCLUSION The combination of nimotuzumab and bevacizumab with TMZ and RT appears to demonstrate efficacy and safety in newly diagnosed GBM patients, providing a reference for clinical treatment. Further prospective studies are needed to confirm our results.
Collapse
Affiliation(s)
- Yaping Wu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, China
| | - Zhiying Chen
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, China
| | - Mingtao Shi
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, China
| | - Shuo Qiu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, China
| | - Yongchun Zhang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, China.
| |
Collapse
|
2
|
Sung JS, Jung J, Kim TH, Kwon S, Bae HE, Kang MJ, Jose J, Lee M, Pyun JC. Epidermal Growth Factor Receptor (EGFR) Inhibitors Screened from Autodisplayed Fv-Antibody Library. Bioconjug Chem 2024; 35:1324-1334. [PMID: 39197031 DOI: 10.1021/acs.bioconjchem.4c00256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
Inhibitors of the epithermal growth factor receptor (EGFR) were screened from an autodisplayed Fv-antibody library using an anti-EGF antibody. The Fv-antibody library was expressed on the outer membrane of Escherichia coli, which corresponds to the heavy chain VH region of immunoglobulin G. The library was constructed by randomizing the CDR3 region of expressed VH regions (11 amino acid residues) by site-directed mutagenesis. Using an anti-EGF antibody as a screening probe, amino acid sequences (CDR3 region) with antibody binding affinity were screened from the Fv-antibody library. These amino acid sequences were considered to have similar chemical properties to EGF, which can bind to EGFR. Two autodisplayed clones with Fv-antibodies against EGFR were screened from the Fv-antibody library, and the screened Fv-antibodies were expressed as soluble proteins. The binding affinity (KD) was estimated using an SPR biosensor, and the inhibitory activity of expressed Fv-antibodies was observed for PANC-1 pancreatic tumor cells and T98G glioblastoma cells using Western blot analysis of proteins in the EGFR-mediated signaling pathway. The viability of PANC-1 and T98G cells was observed to decrease via the inhibitory activity of expressed Fv-antibodies. Finally, interactions between Fv-antibodies and EGFR were analyzed by using molecular docking simulations.
Collapse
Affiliation(s)
- Jeong Soo Sung
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea
| | - Jaeyong Jung
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea
| | - Tae-Hun Kim
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea
| | - Soonil Kwon
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea
| | - Hyung Eun Bae
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea
| | - Min-Jung Kang
- Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Joachim Jose
- Institute of Pharmaceutical and Medical Chemistry, Universität Münster, Münster 48149, Germany
| | - Misu Lee
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Korea
- Institute for New Drug Development, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, South Korea
| | - Jae-Chul Pyun
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea
| |
Collapse
|
3
|
Chai X, Zhang Y, Zhang W, Feng K, Jiang Y, Zhu A, Chen X, Di L, Wang R. Tumor Metabolism: A New Field for the Treatment of Glioma. Bioconjug Chem 2024; 35:1116-1141. [PMID: 39013195 DOI: 10.1021/acs.bioconjchem.4c00287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
The clinical treatment of glioma remains relatively immature. Commonly used clinical treatments for gliomas are surgery combined with chemotherapy and radiotherapy, but there is a problem of drug resistance. In addition, immunotherapy and targeted therapies also suffer from the problem of immune evasion. The advent of metabolic therapy holds immense potential for advancing more efficacious and tolerable therapies against this aggressive disease. Metabolic therapy alters the metabolic processes of tumor cells at the molecular level to inhibit tumor growth and spread, and lead to better outcomes for patients with glioma that are insensitive to conventional treatments. Moreover, compared with conventional therapy, it has less impact on normal cells, less toxicity and side effects, and higher safety. The objective of this review is to examine the changes in metabolic characteristics throughout the development of glioma, enumerate the current methodologies employed for studying tumor metabolism, and highlight the metabolic reprogramming pathways of glioma along with their potential molecular mechanisms. Importantly, it seeks to elucidate potential metabolic targets for glioblastoma (GBM) therapy and summarize effective combination treatment strategies based on various studies.
Collapse
Affiliation(s)
- Xiaoqian Chai
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Yingjie Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Wen Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Kuanhan Feng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Yingyu Jiang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Anran Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Xiaojin Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Ruoning Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| |
Collapse
|
4
|
Liu K, Wang Y, Ma L, Yang S, Zhang X. Efficacy and safety of a treatment in patients with locoregionally advanced nasopharyngeal carcinoma (LANC) involving carotid artery invasion. Eur Arch Otorhinolaryngol 2022; 279:5791-5799. [PMID: 35918541 DOI: 10.1007/s00405-022-07446-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/10/2022] [Indexed: 01/04/2023]
Abstract
PURPOSE Locally advanced nasopharyngeal carcinoma (LANC) often invades the parapharyngeal space and internal carotid artery. Are patients with LANC invading carotid artery are at risk of massive neck hemorrhage after radiotherapy? METHODS This retrospective study included 130 LANC patients with carotid artery invasion admitted to our hospital between January 2012 and September 2019. All patients were treated with induction chemotherapy followed by concurrent chemoradiotherapy (CCRT) ± epidermal growth factor receptor (EGFR) inhibitor. Univariate and multivariate analysis of risk factors were conducted for the prognosis and the occurrence of massive neck hemorrhage of LANC patients with carotid artery invasion. OUTCOMES The 5-year progression-free survival (PFS), distant metastasis-free survival (DMFS), local nodal recurrence-free survival (LNRFS), local recurrence-free survival (LRFS), nodal recurrence-free survival (NRFS) and overall survival (OS) of the 130 patients were 75.2%, 76.8%, 90.0%, 93.9%, 95.8% and 87.2%, respectively. The incidence of fatal bleeding after radiotherapy was 2.3% (3/130). The primary site of the three cases were all the pharyngeal recess, with more than 270° carotid artery invasion, suffering nasopharyngeal necrosis after radiotherapy (2 of which were diabetics and 1 received re-radiation after recurrence). Univariate analysis showed that clinical stage was negatively correlated with DMFS and PFS (P < 0.05). The induction chemotherapy TP regimen, platinum-based concurrent chemotherapy and EGFR inhibitors (Nituzumab/Cetuximab) significantly improved PFS and DMFS (P < 0.05). Patients with hemoglobin levels > 110 g/L had significantly inferior PFS, DMFS and OS than those with hemoglobin levels ≤ 110 g/L (P < 0.05). Multivariate analysis showed that the EGFR inhibitor was an independent risk factor for PFS and DMFS, while the lowest hemoglobin level was an independent risk factor for OS. CONCLUSIONS In LANC patients whose carotid artery invasion was < 270°, induction chemotherapy (IC) followed by helical tomotherapy (HT) and concurrent chemoradiotherapy (CCRT) with EGFR (epidermal growth factor receptor) inhibitor had mild and tolerable side effects, better PFS and DMFS, with no massive hemorrhage. In patients whose primary tumor was pharyngeal recess with carotid artery invasion ≥ 270°, poorly controlled diabetes or re-radiotherapy led to a higher risk of massive hemorrhage after radiotherapy.
Collapse
Affiliation(s)
- Kun Liu
- College of Otolaryngology Head and Neck Surgery, National Clinical Research Center for Otolaryngologic Diseases, Key Lab of Hearing Science, Ministry of Education, Beijing Key Lab of Hearing Impairment for Prevention and Treatment, Medical School of Chinese PLA, 28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Yang Wang
- College of Otolaryngology Head and Neck Surgery, National Clinical Research Center for Otolaryngologic Diseases, Key Lab of Hearing Science, Ministry of Education, Beijing Key Lab of Hearing Impairment for Prevention and Treatment, Medical School of Chinese PLA, 28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Lin Ma
- Department of Radiotherapy, Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Shiming Yang
- College of Otolaryngology Head and Neck Surgery, National Clinical Research Center for Otolaryngologic Diseases, Key Lab of Hearing Science, Ministry of Education, Beijing Key Lab of Hearing Impairment for Prevention and Treatment, Medical School of Chinese PLA, 28 Fuxing Road, Beijing, 100853, People's Republic of China.
| | - Xinxin Zhang
- College of Otolaryngology Head and Neck Surgery, National Clinical Research Center for Otolaryngologic Diseases, Key Lab of Hearing Science, Ministry of Education, Beijing Key Lab of Hearing Impairment for Prevention and Treatment, Medical School of Chinese PLA, 28 Fuxing Road, Beijing, 100853, People's Republic of China.
| |
Collapse
|
5
|
Yuan Y, Chen J, Fang M, Guo Y, Sun X, Yu D, Guo Y, Xin Y. Nimotuzumab combined with chemoradiotherapy for the treatment of cervical cancer: A meta-analysis of randomized controlled trials. Front Oncol 2022; 12:994726. [PMID: 36263226 PMCID: PMC9573994 DOI: 10.3389/fonc.2022.994726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/20/2022] [Indexed: 12/03/2022] Open
Abstract
Objectives To assess the clinical efficacy and toxicity of nimotuzumab in combination with chemoradiotherapy or chemoradiotherapy alone in the treatment of cervical cancer. Methods The PubMed, Embase, Cochrane Library, Web of Science, China National Knowledge Infrastructure, China Biomedical Medicine, Wanfang, and VIP databases were systematically searched for relevant literature. Ultimately, six randomised controlled trials (n=393) were included in our meta-analysis. Results A total of 393 patients were included, of which 197 were in the nimotuzumab combined with chemoradiotherapy group and 196 were in the chemoradiotherapy group. The results of our meta-analysis showed that the complete remission rate (risk ratio [RR] = 1.34, 95% confidence interval [CI]: 1.08-1.65, P = 0.007), objective response rate (RR = 1.30, 95% CI: 1.16-1.44, P < 0.05), and three-year survival rate (RR = 1.27, 95% CI: 1.06-1.51, P = 0.008) in the nimotuzumab combined with chemoradiotherapy group were significantly improved compared with the chemoradiotherapy group. This difference was not statistically significant when comparing the incidence of adverse reactions (such as leukocytopenia, gastrointestinal reaction, radiocystitis, and radioproctitis) between the two groups. Conclusions Nimotuzumab in combination with chemoradiotherapy has some advantages over chemoradiotherapy alone in the treatment of cervical cancer and does not increase toxicity. Therefore, nimotuzumab has the potential to be an effective treatment for cervical cancer; however, further evidence from large-scale randomised controlled trials is needed.
Collapse
Affiliation(s)
- Yan Yuan
- Department of Radiation, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Jiuzhou Chen
- Department of Radiation, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Miao Fang
- Department of Radiation, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Yaru Guo
- Department of Radiation, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Xueqing Sun
- Department of Radiation, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Dehong Yu
- Department of Radiation, the Affiliated Pizhou Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yilong Guo
- Department of Radiation, the Affiliated Pizhou Hospital of Xuzhou Medical University, Xuzhou, China
- *Correspondence: Yilong Guo, ; Yong Xin,
| | - Yong Xin
- Department of Radiation, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Cancer Institute, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Yilong Guo, ; Yong Xin,
| |
Collapse
|
6
|
She L, Gong X, Su L, Liu C. Radiotherapy Plus Temozolomide With or Without Nimotuzumab Against the Newly Diagnosed EGFR-Positive Glioblastoma: A Retrospective Cohort Study. Oncologist 2022; 28:e45-e53. [PMID: 36181764 PMCID: PMC9847561 DOI: 10.1093/oncolo/oyac202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 09/08/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) has a poor prognosis, and patients with epidermal growth factor receptor (EGFR) amplification have an even worse prognosis. Nimotuzumab is an EGFR monoclonal antibody thought to play a significant role in the treatment of GBM. This paper presents a retrospective cohort study that evaluates the clinical efficacy and safety of nimotuzumab in GBM. MATERIALS AND METHODS A total of 56 newly diagnosed patients with EGFR-positive GBM were included in our study. The patients were divided into radiotherapy (RT) + temozolomide (TMZ) + nimotuzumab (39 patients) and RT + TMZ (17 patients) groups based on whether or not nimotuzumab was added during RT. Progression-free survival (PFS), overall survival (OS), and toxicities were assessed. RESULTS The median follow-up time was 27.9 months (95% confidence interval [CI], 25.1-30.8). The median PFS was 12.4 months (95% CI, 7.8-17.0) and 8.2 months (95% CI, 6.1-10.3) in the 2 groups, respectively, P = .052. The median OS was 27.3 months (95% CI, 19.0-35.6) and 16.7 months (95% CI, 11.1-22.2), respectively, P = .018. In patients with unmethylated O6-methylguanine-DNA methyltransferase (MGMT) promoter, the PFS and OS were significantly better in patients treated with nimotuzumab than in those without nimotuzumab (median PFS: 19.3 vs 6.7 months, P = .001; median OS: 20.2 vs 13.8 months, P = .026). During the treatment period, no statistically significant difference in toxicity was noted between the 2 groups. CONCLUSION Our retrospective cohort study suggests the efficacy of Nimotuzumab combined with concurrent RT with TMZ in patients with newly diagnosed EGFR-positive GBM, and specifically those with unmethylated MGMT promoter. Further prospective studies are warranted to validate our findings. Besides, nimotuzumab demonstrated good safety and tolerability.
Collapse
Affiliation(s)
| | | | - Lin Su
- Department of Oncology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Chao Liu
- Corresponding author: Chao Liu, MD, Department of Oncology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People’s Republic of China. Tel: +86 158 741 63692;
| |
Collapse
|
7
|
Parenrengi MA, Suryaningtyas W, Al Fauzi A, Hafid Bajamal A, Kusumastuti K, Utomo B, Muslim Hidayat Thamrin A, Sulistiono B. Nimotuzumab as Additional Therapy for GLIOMA in Pediatric and Adolescent: A Systematic Review. Cancer Control 2022; 29:10732748211053927. [PMID: 35191733 PMCID: PMC8874160 DOI: 10.1177/10732748211053927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION Pediatric gliomas represent the most common brain tumor in children and its higher grades are associated with higher recurrence and low survival rate. All therapeutic modalities are reported to be insufficient to achieve satisfactory result, with follow-up treatment such as adjuvant radiotherapy and chemotherapy recommended to increase survival and hinder tumor progression. Nimotuzumab is a monoclonal antibody that acts as an inhibitor of epidermal growth factor receptor found on the surface of glioma cells and had been studied for its usage in pediatric gliomas in recent years. METHODS A systematic review is performed according to Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. A through literature search was conducted on PubMed, Scopus, Cochrane, and clinicaltrials.gov database. Articles were selected systematically based on the PRISMA protocol and reviewed completely. The relevant data were summarized and discussed. We measured overall survival, progression-free survival, and adverse Events (AE) for nimotuzumab usage as an adjunct therapy in pediatric glioma population. RESULT From 5 studies included for qualitative analysis, 151 patients are included with overall survival (OS) that vary from 3.2-22.8 mo, progression-free survival (PFS) from 1.7-21.6 mo, and relatively low serious adverse events (0-21) are recorded. Follow-up ranged from 2.4-66 mo with four studies reporting diffuse intrinsic pontine glioma (DIPG) patients and only one study reporting nimotuzumab usage in pediatric high-grade glioma (HGG) patients with better outcome in HGG patients than DIPG. CONCLUSION There are no significant differences in the PFS and OS of nimotuzumab as adjunct therapy for pediatric compared to result of standard therapy in majority of previous studies. There were also no differences in the AE of nimotuzumab for pediatric glioma between studies, and low event of serious adverse events indicating its safety. But still there is an evidence of possible benefit of nimotuzumab as adjuvant therapy in pediatric glioma. We recommend further studies with larger number of patients that may lead to possibly different results. There should also be more studies with better level of evidence to further validate the effect of nimozutumab on pediatric glioma.
Collapse
Affiliation(s)
- Muhammad A Parenrengi
- Department of Neurosurgery, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Wihasto Suryaningtyas
- Department of Neurosurgery, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Asra Al Fauzi
- Department of Neurosurgery, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Abdul Hafid Bajamal
- Department of Neurosurgery, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Kurnia Kusumastuti
- Department of Neurology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Budi Utomo
- Department of Public Health and Preventive Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Ahmad Muslim Hidayat Thamrin
- Department of Neurosurgery, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Bagus Sulistiono
- Department of Neurosurgery, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| |
Collapse
|
8
|
Sun L, Gai Y, Li Z, Zhang X, Li J, Ma Y, Li H, Barajas RJ, Zeng D. Development of Dual Receptor Enhanced Pre-Targeting Strategy-A Novel Promising Technology for Immuno-Positron Emission Tomography Imaging. ADVANCED THERAPEUTICS 2021; 4:2100110. [PMID: 35309962 PMCID: PMC8932640 DOI: 10.1002/adtp.202100110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Indexed: 11/06/2022]
Abstract
PET imaging has become an important diagnostic tool in the era of precise medicine. Various pre-targeting systems have been reported to address limitations associated with traditional immuno-PET. However, the application of these mono-receptor based pre-targeting (MRPT) strategies is limited to non-internalizable antibodies, and the tumor uptake is usually much lower than that in the corresponding immuno-PET. To circumvent these limitations, we develop the first Dual-Receptor Pre-Targeting (DRPT) system through entrapping the tumor-receptor-specific radioligand by the pre-administered antibody. Besides the similar ligation pathway happens in MRPT, incorporation of a tumor-receptor-specific peptide into the radioligand in DRPT enhances both concentration and retention of the radioligand on tumor, promoting its ligation with pre-administered mAb on cell-surface and/or internalized into tumor-cells. In this study, 64Cu based DRPT shows superior performance over corresponding MRPT and immuno-PET using internalizable antibodies. Besides, the compatibility of DRPT with short-lived and generator-produced 68Ga is demonstrated, leveraging its advantage in reducing radio-dose exposure. Furthermore, the feasibility of reducing the amount of the pre-administered antibody is confirmed, indicating the cost saving potential of DRPT. In summary, synergizing advantages of dual-receptor targeting and pre-targeting, we expect that this DRPT strategy can become a breakthrough technology in the field of antibody-based molecular imaging.
Collapse
Affiliation(s)
- Lingyi Sun
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA; Center of Radiochemistry Research, Knight Cardiovascular Institute, Oregon Health & Science University, Portland 97239, USA
| | - Yongkang Gai
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA
| | - Zhonghan Li
- Center of Radiochemistry Research, Knight Cardiovascular Institute, Oregon Health & Science University, Portland 97239, USA
| | - Xiaohui Zhang
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA
| | - Jianchun Li
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA
| | - Yongyong Ma
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA
| | - Huiqiang Li
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA
| | - Ramon J Barajas
- Department of Diagnostic Radiology, Oregon Health & Science University, Portland 97239, USA; Advanced Imaging Research Center, Oregon Health & Science University, Portland 97239, USA; Translational Oncology Research Program, Knight Cancer Institute, Oregon Health & Science University, Portland 97239, USA
| | - Dexing Zeng
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA; Center of Radiochemistry Research, Knight Cardiovascular Institute, Oregon Health & Science University, Portland 97239, USA; Department of Diagnostic Radiology, Oregon Health & Science University, Portland 97239, USA
| |
Collapse
|
9
|
Piper K, DePledge L, Karsy M, Cobbs C. Glioma Stem Cells as Immunotherapeutic Targets: Advancements and Challenges. Front Oncol 2021; 11:615704. [PMID: 33718170 PMCID: PMC7945033 DOI: 10.3389/fonc.2021.615704] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/07/2021] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma is the most common and lethal primary brain malignancy. Despite major investments in research into glioblastoma biology and drug development, treatment remains limited and survival has not substantially improved beyond 1-2 years. Cancer stem cells (CSC) or glioma stem cells (GSC) refer to a population of tumor originating cells capable of self-renewal and differentiation. While controversial and challenging to study, evidence suggests that GCSs may result in glioblastoma tumor recurrence and resistance to treatment. Multiple treatment strategies have been suggested at targeting GCSs, including immunotherapy, posttranscriptional regulation, modulation of the tumor microenvironment, and epigenetic modulation. In this review, we discuss recent advances in glioblastoma treatment specifically focused on targeting of GCSs as well as their potential integration into current clinical pathways and trials.
Collapse
Affiliation(s)
- Keenan Piper
- Ben & Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, United States.,Sidney Kimmel Medical College, Philadelphia, PA, United States
| | - Lisa DePledge
- Ben & Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, United States.,University of Washington School of Medicine, Spokane, WA, United States
| | - Michael Karsy
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, United States
| | - Charles Cobbs
- Ben & Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, United States
| |
Collapse
|
10
|
A Signature That May Be Predictive of Early Versus Late Recurrence After Radiation Treatment for Breast Cancer That May Inform the Biology of Early, Aggressive Recurrences. Int J Radiat Oncol Biol Phys 2020; 108:686-696. [DOI: 10.1016/j.ijrobp.2020.05.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/30/2020] [Accepted: 05/11/2020] [Indexed: 01/20/2023]
|
11
|
Xu Z, Shu H, Zhang F, Luo W, Li Y, Chu J, Zhao Q, Lv Y. Nimotuzumab Combined With Irradiation Enhances the Inhibition to the HPV16 E6-Promoted Growth of Cervical Squamous Cell Carcinoma. Front Oncol 2020; 10:1327. [PMID: 32850421 PMCID: PMC7419688 DOI: 10.3389/fonc.2020.01327] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 06/25/2020] [Indexed: 12/24/2022] Open
Abstract
Human papillomavirus (HPV) 16 E6 has been proved to increase the radiosensitivity and lead to the EGFR overexpression in cervical cancer cells. In this study, to investigate the inhibition of nimotuzumab-mediated EGFR blockade combined with radiotherapy, we established a C33A cervical squamous cell line overexpressed HPV16-E6 and a nude mouse model bearing these cell lines. The CCK-8 assay was used to detect the effects of various treatments on the proliferation of C33A cells. Flow cytometry was used to detect the rates of apoptosis and cell cycle arrest. Gene transcription and protein expression were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot, respectively. Immunohistochemical staining was used to evaluate protein expression in tumor tissue. We revealed that E6-overexpressing C33A cells grew faster and were more sensitive to radiotherapy than control cells in vitro and in vivo. The expression levels of EGFR, as well as those of downstream signaling molecules AKT and ERK 1/2, were significantly upregulated in C33A cells that overexpressed E6. We observed that nimotuzumab combined with radiotherapy could enhance the inhibition of C33A cell growth induced by E6, both in vitro and in vivo. We also observed enhanced effect after combination on G2/M cell cycle arrest and apoptosis in E6-overexpressing C33A cells. Furthermore, the combined therapy of nimotuzumab and radiation remarkably reduced the protein expression levels of EGFR, AKT, ERK 1/2 in vitro, and in vivo. In conclusion, HPV16 E6 expression is positively correlated with levels of EGFR, AKT, and ERK 1/2 protein expression. The combined treatment with nimotuzumab and radiotherapy to enhance radiosensitivity in E6-positive cervical squamous cell carcinoma was related to enhanced G2/M cell cycle arrest and caspase-related apoptosis.
Collapse
Affiliation(s)
- Zhonghua Xu
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hang Shu
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fan Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Weiwei Luo
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yan Li
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jinjin Chu
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qihong Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Yin Lv
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
12
|
Scriba LD, Bornstein SR, Santambrogio A, Mueller G, Huebner A, Hauer J, Schedl A, Wielockx B, Eisenhofer G, Andoniadou CL, Steenblock C. Cancer Stem Cells in Pheochromocytoma and Paraganglioma. Front Endocrinol (Lausanne) 2020; 11:79. [PMID: 32158431 PMCID: PMC7051940 DOI: 10.3389/fendo.2020.00079] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 02/06/2020] [Indexed: 12/17/2022] Open
Abstract
Pheochromocytoma (PCC) and paraganglioma (PGL) are rare neuroendocrine tumors associated with high cardiovascular morbidity and variable risk of malignancy. The current therapy of choice is surgical resection. Nevertheless, PCCs/PGLs are associated with a lifelong risk of tumor persistence or recurrence. A high rate of germline or somatic mutations in numerous genes has been found in these tumors. For some, the tumorigenic processes are initiated during embryogenesis. Such tumors carry gene mutations leading to pseudohypoxic phenotypes and show more immature characteristics than other chromaffin cell tumors; they are also often multifocal or metastatic and occur at an early age, often during childhood. Cancer stem cells (CSCs) are cells with an inherent ability of self-renewal, de-differentiation, and capacity to initiate and maintain malignant tumor growth. Targeting CSCs to inhibit cancer progression has become an attractive anti-cancer therapeutic strategy. Despite progress for this strategy for solid tumors such as neuroblastoma, brain, breast, and colon cancers, no substantial advance has been made employing similar strategies in PCCs/PGLs. In the current review, we discuss findings related to the identification of normal chromaffin stem cells and CSCs, pathways involved in regulating the development of CSCs, and the importance of the stem cell niche in development and maintenance of CSCs in PCCs/PGLs. Additionally, we examine the development and feasibility of novel CSC-targeted therapeutic strategies aimed at eradicating especially recurrent and metastatic tumors.
Collapse
Affiliation(s)
- Laura D. Scriba
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stefan R. Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Diabetes and Nutritional Sciences Division, King's College London, London, United Kingdom
| | - Alice Santambrogio
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Gregor Mueller
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Angela Huebner
- Children's Hospital, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Julia Hauer
- Department of Pediatrics, Pediatric Hematology and Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | | | - Ben Wielockx
- Institute of Clinical Chemistry, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Graeme Eisenhofer
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Institute of Clinical Chemistry, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Cynthia L. Andoniadou
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Charlotte Steenblock
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
13
|
Viada C, Vega AM, Robaina M, Frías A, Álvarez M, Santiesteban Y, Santiesteban Y, García L, Mestre B, Osorio MX, Pérez L, Macias A, Crombet T, Ramos M. Evaluation of Nimotuzumab for the treatment of head and neck cancer: Meta-analysis of controlled trials. BIONATURA 2020. [DOI: 10.21931/rb/2020.05.01.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Nimotuzumab, humanized monoclonal antibody, directed against the epidermal growth factor receptor: highly expressed protein in malignant tumors of epithelial origin. It has been registered for head and neck tumors since 2002. To determine the effectiveness of Nimotuzumab in head and neck cancer through the combined meta-analysis technique. A search was conducted in PubMed, in an indexed magazine with the words “Nimotuzumab”, “head and neck,” 48 articles published by Cuban and foreign authors were detected between April 1, 2005, and July 31, 2019, in which the results of clinical studies conducted with the monoclonal antibody Nimotuzumab are described. Seven clinical trials conducted in Cuba from 2005-2019 with Nimotuzumab are described; three Phase I / II (with 14, 10 and 10 patients respectively), a Phase II / III with 106 patients, a Phase II with 37 patients, two Phase IV (with 386 and 225 patients each) and a study promoted by the Researcher with 17 patients. From these studies, the three controlled trials were selected by the PRISMA flow chart. The meta-analysis consisted of the construction of the Forest Plot graph, the sensitivity analysis and the cumulative analysis. The meta-analysis shows favorable results for Nimotuzumab, without heterogeneity (I2 = 0%). The sensitivity analysis reveals that the test that differs most from the others is Phase II / III. The cumulative analysis indicates that after the second trial, there is already sufficient evidence.
Collapse
Affiliation(s)
- Carmen Viada
- Centro de Inmunología Molecular, CIM Calle 206 No. 1926 e/ 19 y 21, Atabey, Playa, CP 11600, La Habana, Cuba
| | - Aliz M. Vega
- Centro de Inmunología Molecular, CIM Calle 206 No. 1926 e/ 19 y 21, Atabey, Playa, CP 11600, La Habana, Cuba
| | - Mayte Robaina
- Centro Nacional Coordinador de Ensayos Clínicos, Calle 5ta A e/ 60 y 62, Playa, CP 11300, La Habana, Cuba
| | - Aliuska Frías
- Centro de Inmunología Molecular, CIM Calle 206 No. 1926 e/ 19 y 21, Atabey, Playa, CP 11600, La Habana, Cuba
| | - Mabel Álvarez
- Centro de Inmunología Molecular, CIM Calle 206 No. 1926 e/ 19 y 21, Atabey, Playa, CP 11600, La Habana, Cuba
| | - Yanela Santiesteban
- Centro de Inmunología Molecular, CIM Calle 206 No. 1926 e/ 19 y 21, Atabey, Playa, CP 11600, La Habana, Cuba
| | - Yuliannis Santiesteban
- Centro de Inmunología Molecular, CIM Calle 206 No. 1926 e/ 19 y 21, Atabey, Playa, CP 11600, La Habana, Cuba
| | - Lázara García
- Centro de Inmunología Molecular, CIM Calle 206 No. 1926 e/ 19 y 21, Atabey, Playa, CP 11600, La Habana, Cuba
| | - Braulio Mestre
- Instituto Nacional de Oncología y Radiobiología, Calle 29 e/ F y D, Vedado, Plaza de la Revolución, CP 10400, La Habana Cuba
| | - Marta xxx Osorio
- Instituto Nacional de Oncología y Radiobiología, Calle 29 e/ F y D, Vedado, Plaza de la Revolución, CP 10400, La Habana Cuba
| | - Leslie Pérez
- Centro de Inmunología Molecular, CIM Calle 206 No. 1926 e/ 19 y 21, Atabey, Playa, CP 11600, La Habana, Cuba
| | - Amparo Macias
- Centro de Inmunología Molecular, CIM Calle 206 No. 1926 e/ 19 y 21, Atabey, Playa, CP 11600, La Habana, Cuba
| | - Tania Crombet
- Centro de Inmunología Molecular, CIM Calle 206 No. 1926 e/ 19 y 21, Atabey, Playa, CP 11600, La Habana, Cuba
| | - Mayra Ramos
- Centro de Inmunología Molecular, CIM Calle 206 No. 1926 e/ 19 y 21, Atabey, Playa, CP 11600, La Habana, Cuba
| |
Collapse
|
14
|
Yu Y, Guan H, Jiang L, Li X, Xing L, Sun X. Nimotuzumab, an EGFR‑targeted antibody, promotes radiosensitivity of recurrent esophageal squamous cell carcinoma. Int J Oncol 2020; 56:945-956. [PMID: 32319582 DOI: 10.3892/ijo.2020.4981] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 12/12/2019] [Indexed: 11/06/2022] Open
Abstract
Local tumor recurrence is one of the main causes for the failure of esophageal cancer treatment following radiotherapy. Previous studies have demonstrated that epidermal growth factor receptor (EGFR)‑targeted therapy combined with radiotherapy is expected to become an effective means to control tumor recurrence. The aim of the present study was to investigate the effect and mechanism of nimotuzumab (an EGFR‑targeted antibody) in the treatment of recurrent esophageal carcinoma. The radiation responses of two esophageal squamous carcinoma cell lines, EC109 and TE‑1, with or without nimotuzumab, were first evaluated by CCK‑8 assay. Colony formation and apoptosis were used to measure anti‑proliferation effects. It was demonstrated that nimotuzumab arrested the cell cycle at the G2 phase in vitro. Western blotting and immunofluorescence analysis were used to determine signaling pathway changes. It was observed that nimotuzumab inhibited phosphorylation of EGFR in EC109 cells. Furthermore, recurrent tumor models were established and it was identified that the degree of tumor hypoxia was positively associated with EGFR overexpression. In EC109 cell xenografts, nimotuzumab combined with radiation led to a significant delay in recurrent tumor growth compared with that of radiation alone (P<0.001 for 0 Gy pre‑irradiation, P=0.005 for 20 Gy pre‑irradiation, P=0.005 for 10 Gy pre‑irradiation). These results suggest that nimotuzumab combined with radiation may be an effective means to control recurrent esophageal squamous cell carcinoma with EGFR overexpression.
Collapse
Affiliation(s)
- Yang Yu
- School of Medicine and Life Sciences, Shandong Academy of Medical Sciences, University of Jinan, Jinan, Shandong 250031, P.R. China
| | - Hui Guan
- Department of Radiation Oncology, The Fourth People's Hospital of Jinan, Jinan, Shandong 250031, P.R. China
| | - Liyang Jiang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Xiaolin Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Ligang Xing
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Xiaorong Sun
- Department of Nuclear Medicine, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
15
|
Du XJ, Li XM, Cai LB, Sun JC, Wang SY, Wang XC, Pang XL, Deng ML, Chen FF, Wang ZQ, Chen FR, Zhang HH, Wang HY, Piedra P, Chen ZP, Lin J, Wu SX. Efficacy and safety of nimotuzumab in addition to radiotherapy and temozolomide for cerebral glioblastoma: a phase II multicenter clinical trial. J Cancer 2019; 10:3214-3223. [PMID: 31289592 PMCID: PMC6603389 DOI: 10.7150/jca.30123] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 04/13/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Nimotuzumab is a humanized anti-epidermal growth factor receptor (EGFR) antibody that has shown preclinical and clinical anticancer activity in cerebral glioblastoma multiforme (GBM). We conducted a phase II, single-arm, multicenter clinical trial to evaluate the benefit of adding nimotuzumab to current standard chemo-radiotherapy for patients with GBM with positive EGFR expression. Methods: Newly diagnosed patients with histologically proven single supratentorial GBM and epidermal growth factor receptor (EGFR) positive expressions were recruited. All patients were treated with nimotuzumab, administered once a week intravenously for 6 weeks in addition to radiotherapy with concomitant and adjuvant temozolomide after surgery. The primary endpoints were overall survival (OS) and progression-free survival (PFS). Secondary objectives included objective response rate (ORR) and toxicity. Results: A total of 39 patients were enrolled and 36 patients were evaluated for efficacy. The ORR at the end of RT was 72.2%. Median OS and PFS were 24.5 and 11.9 months. The 1-year OS and PFS rates were 83.3% and 49.3%. The 2-year OS and PFS rates were 51.1% and 29.0%. O (6)-methylquanine DNA methyl-tranferase (MGMT) expression is known to affect the efficacy of chemotherapy and status of its expression is examined. No significant correlation between treatment outcomes and MGMT status was found. Most frequent treatment-related toxicities were mild to moderate and included constipation, anorexia, fatigue, nausea, vomiting, and leucopenia. Conclusions: Our study show that nimotuzumab in addition to standard treatment is well tolerable and has increased survival in newly diagnosed GBM patients with EGFR positive expression.
Collapse
Affiliation(s)
- Xiao-Jing Du
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng Road East, Guangzhou 510060, China
| | - Xian-Ming Li
- Department of Radiation Oncology, Shenzhen People's Hospital, No. 1017 Dongmen Road North, Luohu District, Shenzhen 518020, China
| | - Lin-Bo Cai
- Department of Radiation Oncology, Guangdong 999 Brain Hospital, No. 578 Shatai Road South, Guangzhou 510510, China
| | - Jian-Cong Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Guangzhou Medical University, No. 151 Yanjiang Road West, Guangzhou 510120, China
| | - Si-Yang Wang
- Department of Radiation Oncology, The 5th Affiliated Hospital of Sun Yat-sen University, No. 52 Meihua Road East, Zhuhai, 519000, China
| | - Xi-Cheng Wang
- Department of Radiation Oncology, The First Affiliated Hospital/School of Clinical Medicine of Guangdong Pharmaceutical University, No. 19 Nonglin Xia Road, Guangzhou 510080, China
| | - Xiao-Lin Pang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng Road East, Guangzhou 510060, China
| | - Mei-Ling Deng
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng Road East, Guangzhou 510060, China
| | - Fang-Fang Chen
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng Road East, Guangzhou 510060, China
| | - Zhi-Qiang Wang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng Road East, Guangzhou 510060, China
| | - Fu-Rong Chen
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng Road East, Guangzhou 510060, China
| | - Hong-Hong Zhang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng Road East, Guangzhou 510060, China
| | - Hui-Yun Wang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng Road East, Guangzhou 510060, China
| | - Patricia Piedra
- Center of Molecular Immunology, Avenue 15 and 216 St., Siboney, Playa, La Habana, Cuba. A.P 16040, La Habana 11600, Cuba
| | - Zhong-Ping Chen
- Department of Neuro-Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng Road East, Guangzhou 510060, China
| | - Jun Lin
- Department of Anesthesiology, Stony Brook University, School of Medicine, Health Sciences Tower, Level 4, Rm 060, Stony Brook, NY 11794-8480, United States
| | - Shao-Xiong Wu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng Road East, Guangzhou 510060, China
| |
Collapse
|
16
|
Fleischhack G, Massimino M, Warmuth-Metz M, Khuhlaeva E, Janssen G, Graf N, Rutkowski S, Beilken A, Schmid I, Biassoni V, Gorelishev SK, Kramm C, Reinhard H, Schlegel PG, Kortmann RD, Reuter D, Bach F, Iznaga-Escobar NE, Bode U. Nimotuzumab and radiotherapy for treatment of newly diagnosed diffuse intrinsic pontine glioma (DIPG): a phase III clinical study. J Neurooncol 2019; 143:107-113. [PMID: 30830679 DOI: 10.1007/s11060-019-03140-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 02/28/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Diffuse intrinsic pontine glioma (DIPG) is a devastating cancer of childhood and adolescence. METHODS The study included patients between 3 and 20 years with clinically and radiologically confirmed DIPG. Primary endpoint was 6-month progression-free survival (PFS) following administration of nimotuzumab in combination with external beam radiotherapy (RT). Nimotuzumab was administered intravenously at 150 mg/m2 weekly for 12 weeks. Radiotherapy at total dose of 54 Gy was delivered between week 3 and week 9. Response was evaluated based on clinical features and MRI findings according to RECIST criteria at week 12. Thereafter, patients continued to receive nimotuzumab every alternate week until disease progression/unmanageable toxicity. Adverse events (AE) were evaluated according to Common Terminology Criteria for Adverse Events (CTC-AE) Version 3.0 (CTC-AE3). RESULTS All 42 patients received at least one dose of nimotuzumab in outpatient settings. Two patients had partial response (4.8%), 27 had stable disease (64.3%), 10 had progressive disease (23.8%) and 3 patients (7.1%) could not be evaluated. The objective response rate (ORR) was 4.8%. Median PFS was 5.8 months and median overall survival (OS) was 9.4 months. Most common drug-related AEs were alopecia (14.3%), vomiting, headache and radiation skin injury (7.1% each). Therapy-related serious adverse events (SAEs) were intra-tumoral bleeding and acute respiratory failure, which were difficult to distinguish from effects of tumor progression. CONCLUSIONS Concomitant treatment with RT and nimotuzumab was feasible in an outpatient setting. The PFS and OS were comparable to results achieved with RT and intensive chemotherapy in hospitalized setting.
Collapse
Affiliation(s)
- G Fleischhack
- Paediatric Haematology and Oncology, Paediatrics III, University Hospital of Essen, 45122, Essen, Germany.
- Department of Paediatric Haematology/Oncology, Children Medical Hospital, University of Bonn, 53113, Bonn, Germany.
| | - M Massimino
- Paediatric Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133, Milano, Italy
| | - M Warmuth-Metz
- Department of Neuroradiology, University of Wuerzburg, 97080, Würzburg, Germany
| | - E Khuhlaeva
- Paediatric Neurosurgical Department, Burdenko Neurosurgical Institute, Moscow, 125047, Russia
| | - G Janssen
- Department of Paediatric Haematology/Oncology, Children's Medical Hospital, University of Duesseldorf, 40225, Düsseldorf, Germany
| | - N Graf
- Department of Paediatric Haematology/Oncology, Saarland University, 66421, Homburg/Saar, Germany
| | - S Rutkowski
- Department of Paediatric Hematology/Oncology, University of Wuerzburg, University Children's Hospital, 97080, Wuerzburg, Germany
- Department of Paediatric Haematology and Oncology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - A Beilken
- Department of Paediatric Haematology/Oncology, Medical School, Children's Medical Hospital, 30625, Hannover, Germany
| | - I Schmid
- Department of Paediatric Haematology/Oncology, Children's Medical Hospital, University of Munich, 80337, Munich, Germany
| | - V Biassoni
- Paediatric Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133, Milano, Italy
| | - S K Gorelishev
- Paediatric Neurosurgical Department, Burdenko Neurosurgical Institute, Moscow, 125047, Russia
| | - C Kramm
- Department of Paediatric Haematology/Oncology, Children's Medical Hospital, University of Duesseldorf, 40225, Düsseldorf, Germany
- Division of Paediatric Haematology and Oncology, Department of Child and Adolescent Health, University Medical Center Göttingen, 37075, Göttingen, Germany
| | - H Reinhard
- Department of Paediatric Haematology/Oncology, Saarland University, 66421, Homburg/Saar, Germany
- Paediatric Haematology and Oncology, Asklepios Hospital, 53757, Sankt Augustin, Germany
| | - P G Schlegel
- Department of Paediatric Hematology/Oncology, University of Wuerzburg, University Children's Hospital, 97080, Wuerzburg, Germany
| | - R-D Kortmann
- Department of RT and Radiooncology, University of Leipzig, 04103, Leipzig, Germany
| | - D Reuter
- Oncoscience GmbH, 22869, Schenefeld, Germany
| | - F Bach
- Oncoscience GmbH, 22869, Schenefeld, Germany
| | | | - U Bode
- Department of Paediatric Haematology/Oncology, Children Medical Hospital, University of Bonn, 53113, Bonn, Germany
| |
Collapse
|
17
|
Yao Z, Peng P, Xu D, Zhou X, Pan Z, Li Z, Yao J, Chen J. EGFR inhibitor C225 Increases the Radio-Sensitivity of Human Breast Cancer Cells. Asian Pac J Cancer Prev 2019; 20:311-319. [PMID: 30678455 PMCID: PMC6485551 DOI: 10.31557/apjcp.2019.20.1.311] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/06/2019] [Indexed: 01/08/2023] Open
Abstract
Objective: This study was undertaken to investigate the effect of C225 on the radio-sensitivity of MDA-MB-231 cells line and to disclosure underlying mechanism. Methods: CCK8 assay was used to measure the proliferation inhibition of C225 on MDA-MB-231 cells. The combined effects of C225 plus radiation on the proliferation of MDA-MB-231 cells were also evaluated by CCK-8 assay. The clonogenic assay was performed to evaluate the cell surviving fractions and to determine the radio-sensitizing effect of C225 on MDA-MB-231 cells. The apoptosis and cell cycle distribution were analyzed by flow cytometry. Western blot analysis was used to detect the expression of p-EGFR, p-Akt, p-P38, and caspase-3. Results: C225 had an inhibiting effect on the proliferation of cells in a concentration-dependent manner. The cloning formation capacity was decreased in C225 plus radiation group. C225 increased radio-sensitivity of cells and led to cell cycle arrest in G0/G1 phase markedly. Cells treated with C225 and radiation predominantly exhibited G0/G1 phase arrest and significant decreased in the fraction of cells in the S phase. Moreover, C225 and radiation significantly increased the apoptosis rate of cells. Decreased cell proliferation was further supported by the down-regulation of p-EGFR and its downstream singling pathway proteins such as p-Akt and p-P38. The up-regulation of the Caspase-3 expression in C225 plus radiation group revealed that C225 could increase radiation-inducing cell apoptosis. Conclusion: C225 could increase the radio-sensitivity of cells, which may be due to the anti-proliferative synergistic effect between C225 and radiation as well as the down-regulation of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Zhifeng Yao
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
- Department of Radiation Oncology, The Second Affliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Convection-enhanced delivery of cetuximab conjugated iron-oxide nanoparticles for treatment of spontaneous canine intracranial gliomas. J Neurooncol 2018; 137:653-663. [PMID: 29350351 DOI: 10.1007/s11060-018-2764-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 01/13/2018] [Indexed: 01/04/2023]
Abstract
Cetuximab conjugated iron-oxide nanoparticles (cetuximab-IONPs) have shown both in-vitro and in-vivo anti-tumor efficacy against gliomas. The purpose of this pilot study was to evaluate the safety and potential efficacy of cetuximab-IONPs for treatment of spontaneously occurring intracranial gliomas in canines after convection-enhanced delivery (CED). The use of CED allowed for direct infusion of the cetuximab-IONPs both intratumorally and peritumorally avoiding the blood brain barrier (BBB) and limiting systemic effects. A total of eight dogs participated in the study and only two developed mild post-operative complications, which resolved with medical therapy. All canines underwent a single CED treatment of the cetuximab-IONPs over 3 days and did not receive any further adjuvant treatments. Volumetric analysis showed a median reduction in tumor size of 54.9% by MRI at 1-month (4-6 weeks) follow-up. Five dogs were euthanized due to recurrence of neurological signs other than seizures, two due to recurrent seizures, and one dog died in his sleep. Median survival time after surgery was 248 days (mean 367 days).
Collapse
|
19
|
Clinically relevant radioresistant cell line: a simple model to understand cancer radioresistance. Med Mol Morphol 2017; 50:195-204. [PMID: 29067564 DOI: 10.1007/s00795-017-0171-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/18/2017] [Indexed: 12/20/2022]
Abstract
Radiotherapy (RT) is one of the major modalities for the treatment of human cancers and has been established as an excellent local treatment for malignant tumors. Conventional fractionated RT consists of 2-Gy X-rays, fractionated once a day, 5 days a week for 5-7 weeks in total 60 Gy. The efficacy of RT depends on the existence of radioresistant cells, which remains one of the most critical obstacles in RT and radio-chemotherapy. To improve the efficacy of RT, understanding the characteristics of radioresistant cells is one of the important subjects in radiation biology. Several studies have been reported to find out molecules implicated in radioresistance. However, it is noteworthy that cellular radioresistance has been mainly studied among cells with different genetic backgrounds and different origins. Therefore, making a system to compare between radioresistant and sensitive cells with the isogenic background is required. In this review, some aspects of cellular radioresistance mainly focusing on clinically relevant radioresistant (CRR) cell lines that can continue to proliferate even under exposure to 2-Gy X-rays, once a day, for more than 30 days, which is consistent with the conventional fractionated RT are discussed.
Collapse
|
20
|
Heckler M, Osterberg N, Guenzle J, Thiede-Stan NK, Reichardt W, Weidensteiner C, Saavedra JE, Weyerbrock A. The nitric oxide donor JS-K sensitizes U87 glioma cells to repetitive irradiation. Tumour Biol 2017; 39:1010428317703922. [PMID: 28653883 DOI: 10.1177/1010428317703922] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
As a potent radiosensitizer nitric oxide (NO) may be a putative adjuvant in the treatment of malignant gliomas which are known for their radio- and chemoresistance. The NO donor prodrug JS-K (O2-(2.4-dinitrophenyl) 1-[(4-ethoxycarbonyl) piperazin-1-yl] diazen-1-ium-1,2-diolate) allows cell-type specific intracellular NO release via enzymatic activation by glutathione-S-transferases overexpressed in glioblastoma multiforme. The cytotoxic and radiosensitizing efficacy of JS-K was assessed in U87 glioma cells in vitro focusing on cell proliferation, induction of DNA damage, and cell death. In vivo efficacy of JS-K and repetitive irradiation were investigated in an orthotopic U87 xenograft model in mice. For the first time, we could show that JS-K acts as a potent cytotoxic and radiosensitizing agent in U87 cells in vitro. This dose- and time-dependent effect is due to an enhanced induction of DNA double-strand breaks leading to mitotic catastrophe as the dominant form of cell death. However, this potent cytotoxic and radiosensitizing effect could not be confirmed in an intracranial U87 xenograft model, possibly due to insufficient delivery into the brain. Although NO donor treatment was well tolerated, neither a retardation of tumor growth nor an extended survival could be observed after JS-K and/or radiotherapy.
Collapse
Affiliation(s)
- Max Heckler
- 1 Department of Neurosurgery, University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nadja Osterberg
- 1 Department of Neurosurgery, University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jessica Guenzle
- 1 Department of Neurosurgery, University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,2 Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Nina Kristin Thiede-Stan
- 1 Department of Neurosurgery, University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wilfried Reichardt
- 3 German Cancer Consortium (DKTK), Heidelberg, Germany.,4 German Cancer Research Center (DKFZ), Heidelberg, Germany.,5 Department of Radiology-Medical Physics, University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Claudia Weidensteiner
- 5 Department of Radiology-Medical Physics, University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Joseph E Saavedra
- 6 Cancer and Inflammation Program, National Cancer Institute (NCI) at Frederick, Frederick, MD, USA
| | - Astrid Weyerbrock
- 1 Department of Neurosurgery, University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
21
|
Nimotuzumab increases the anti-tumor effect of photodynamic therapy in an oral tumor model. Oncotarget 2016; 6:13487-505. [PMID: 25918252 PMCID: PMC4537029 DOI: 10.18632/oncotarget.3622] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 04/08/2015] [Indexed: 12/23/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) represents 90% of all oral cancers and is characterized with poor prognosis and low survival rate. Epidermal growth factor receptor (EGFR) is highly expressed in oral cancer and is a target for cancer therapy and prevention. In this present work, we evaluate the efficacy of photodynamic therapy (PDT) in combination with an EGFR inhibitor, nimotuzumab in oral cancer cell lines and OSCC xenograft tumor model. PDT is a promising and minimally invasive treatment modality that involves the interaction of a photosensitizer, molecular oxygen and light to destroy tumors. We demonstrated that EGFR inhibitors nimotuzumab and cetuximab exhibits anti-angiogenic properties by inhibiting the migration and invasion of oral cancer cell lines and human endothelial cells. The EGFR inhibitors also significantly reduced tube formation of endothelial cells. Chlorin e6-PDT in combination with nimotuzumab and cetuximab reduced cell proliferation in different oral cancer and endothelial cells. Furthermore, our in vivo studies showed that the combination therapy of PDT and nimotuzumab synergistically delayed tumor growth when compared with control and PDT treated tumors. Downregulation of EGFR, Ki-67 and CD31 was observed in the tumors treated with combination therapy. Analysis of the liver and kidney function markers showed no treatment related toxicity. In conclusion, PDT outcome of oral cancer can be improved when combined with EGFR inhibitor nimotuzumab.
Collapse
|
22
|
Koukourakis MI, Mitrakas AG, Giatromanolaki A. Therapeutic interactions of autophagy with radiation and temozolomide in glioblastoma: evidence and issues to resolve. Br J Cancer 2016; 114:485-96. [PMID: 26889975 PMCID: PMC4782209 DOI: 10.1038/bjc.2016.19] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/21/2015] [Accepted: 12/31/2015] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma is a unique model of non-metastasising disease that kills the vast majority of patients through local growth, despite surgery and local irradiation. Glioblastoma cells are resistant to apoptotic stimuli, and their death occurs through autophagy. This review aims to critically present our knowledge regarding the autophagic response of glioblastoma cells to radiation and temozolomide (TMZ) and to delineate eventual research directions to follow, in the quest of improving the curability of this incurable, as yet, disease. Radiation and TMZ interfere with the autophagic machinery, but whether cell response is driven to autophagy flux acceleration or blockage is disputable and may depend on both cell individuality and radiotherapy fractionation or TMZ schedules. Potent agents that block autophagy at an early phase of initiation or at a late phase of autolysosomal fusion are available aside to agents that induce functional autophagy, or even demethylating agents that may unblock the function of autophagy-initiating genes in a subset of tumours. All these create a maze, which if properly investigated can open new insights for the application of novel radio- and chemosensitising policies, exploiting the autophagic pathways that glioblastomas use to escape death.
Collapse
Affiliation(s)
- Michael I Koukourakis
- Department of Radiotherapy/Oncology, Democritus University of Thrace, PO Box 12, Alexandroupolis 68100, Greece
| | - Achilleas G Mitrakas
- Department of Radiotherapy/Oncology, Democritus University of Thrace, PO Box 12, Alexandroupolis 68100, Greece
| | | |
Collapse
|
23
|
Zhang X, Du L, Zhao F, Wang Q, Yang S, Ma L. A Phase II Clinical Trial of Concurrent Helical Tomotherapy plus Cetuximab Followed by Adjuvant Chemotherapy with Cisplatin and Docetaxel for Locally Advanced Nasopharyngeal Carcinoma. Int J Biol Sci 2016; 12:446-53. [PMID: 27019628 PMCID: PMC4807163 DOI: 10.7150/ijbs.12937] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 12/17/2015] [Indexed: 01/30/2023] Open
Abstract
PURPOSE The present clinical trial was designed to evaluate the efficacy and safety of concurrent helical tomotherapy (HT) with cetuximab followed by adjuvant chemotherapy with docetaxel and cisplatin (TP) in the treatment of patients with locoregionally advanced nasopharyngeal carcinoma. MATERIALS AND METHODS This phase II clinical trial included 43 patients with Stage III/IV LANC (33 Stage III and 10 Stage IV). The treatment consisted of concurrent HT with cetuximab (400 mg/m(2) loading dose and weekly 250mg/m(2)), followed by four cycles of chemotherapy [docetaxel (70 mg/m(2) on Day 1) and cisplatin (40 mg/m(2) on Days 1 and 2 every 3 weeks). Side effects were evaluated with CTCAE criteria (Common Terminology Criteria for Adverse Events 3.0). RESULTS The median follow-up duration was 48.0 months [95% confidence interval (CI) 41.7-58.0 months], the 2-year locoregional failure-free rate (LFFR), progression-free survival (PFS), distant failure-free rate (DFFR) and overall survival (OS) were 95.2%, 79.1%, 88.1% and 93.0% respectively; the 3-year LFFR, DFFR, PFS and OS were 92.7%, 85.6%, 72.0% and 85.7% respectively. The most common grade 3 toxicities were oropharyngeal mucositis (81.4%) and RT-related dermatitis (7.0%). No patients had more than grade 3 radiation related toxicities and no patients required nasogastric feeding. One patient experienced grade 3 osteonecrosis at 18 months after treatment. CONCLUSIONS Concurrent HT with cetuximab followed by adjuvant chemotherapy with TP is an effective strategy for the treatment of LANC with encouraging survival rates and minimal side effects.
Collapse
Affiliation(s)
- Xinxin Zhang
- 1. Department of Otolaryngology, Head and Neck Surgery, Chinese People Liberation Army (PLA) General Hospital, Beijing, China
| | - Lei Du
- 2. Department of Radiation Oncology, Chinese People Liberation Army (PLA) General Hospital, Beijing, China
| | - Feifang Zhao
- 1. Department of Otolaryngology, Head and Neck Surgery, Chinese People Liberation Army (PLA) General Hospital, Beijing, China
| | - Qiuju Wang
- 1. Department of Otolaryngology, Head and Neck Surgery, Chinese People Liberation Army (PLA) General Hospital, Beijing, China
| | - Shiming Yang
- 1. Department of Otolaryngology, Head and Neck Surgery, Chinese People Liberation Army (PLA) General Hospital, Beijing, China
| | - Lin Ma
- 2. Department of Radiation Oncology, Chinese People Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
24
|
Kaluzova M, Bouras A, Machaidze R, Hadjipanayis CG. Targeted therapy of glioblastoma stem-like cells and tumor non-stem cells using cetuximab-conjugated iron-oxide nanoparticles. Oncotarget 2016; 6:8788-806. [PMID: 25871395 PMCID: PMC4496184 DOI: 10.18632/oncotarget.3554] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 02/10/2015] [Indexed: 01/21/2023] Open
Abstract
Malignant gliomas remain aggressive and lethal primary brain tumors in adults. The epidermal growth factor receptor (EGFR) is frequently overexpressed in the most common malignant glioma, glioblastoma (GBM), and represents an important therapeutic target. GBM stem-like cells (GSCs) present in tumors are felt to be highly tumorigenic and responsible for tumor recurrence. Multifunctional magnetic iron-oxide nanoparticles (IONPs) can be directly imaged by magnetic resonance imaging (MRI) and designed to therapeutically target cancer cells. The targeting effects of IONPs conjugated to the EGFR inhibitor, cetuximab (cetuximab-IONPs), were determined with EGFR- and EGFRvIII-expressing human GBM neurospheres and GSCs. Transmission electron microscopy revealed cetuximab-IONP GBM cell binding and internalization. Fluorescence microscopy and Prussian blue staining showed increased uptake of cetuximab-IONPs by EGFR- as well as EGFRvIII-expressing GSCs and neurospheres in comparison to cetuximab or free IONPs. Treatment with cetuximab-IONPs resulted in a significant antitumor effect that was greater than with cetuximab alone due to more efficient, CD133-independent cellular targeting and uptake, EGFR signaling alterations, EGFR internalization, and apoptosis induction in EGFR-expressing GSCs and neurospheres. A significant increase in survival was found after cetuximab-IONP convection-enhanced delivery treatment of 3 intracranial rodent GBM models employing human EGFR-expressing GBM xenografts.
Collapse
Affiliation(s)
- Milota Kaluzova
- Brain Tumor Nanotechnology Laboratory, Department of Neurosurgery, Emory University School of Medicine, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Alexandros Bouras
- Brain Tumor Nanotechnology Laboratory, Department of Neurosurgery, Emory University School of Medicine, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Revaz Machaidze
- Brain Tumor Nanotechnology Laboratory, Department of Neurosurgery, Emory University School of Medicine, Winship Cancer Institute of Emory University, Atlanta, GA, USA.,Present address: Brigham and Women's Hospital, Neurosurgery, Boston, MA, USA
| | - Costas G Hadjipanayis
- Brain Tumor Nanotechnology Laboratory, Department of Neurosurgery, Emory University School of Medicine, Winship Cancer Institute of Emory University, Atlanta, GA, USA.,Winship Brain Tumor Center of Emory University, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| |
Collapse
|
25
|
Nitta Y, Shimizu S, Shishido-Hara Y, Suzuki K, Shiokawa Y, Nagane M. Nimotuzumab enhances temozolomide-induced growth suppression of glioma cells expressing mutant EGFR in vivo. Cancer Med 2016; 5:486-99. [PMID: 26778701 PMCID: PMC4799951 DOI: 10.1002/cam4.614] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/14/2015] [Accepted: 11/19/2015] [Indexed: 11/08/2022] Open
Abstract
A mutant form of epidermal growth factor receptor (EGFR), EGFRvIII, is common in glioblastoma (GBM) and confers enhanced tumorigenic activity and drug resistance. Nimotuzumab, an anti-EGFR antibody, has shown preclinical and clinical activity to GBM, but its specific activity against EGFRvIII has not been fully investigated. Human glioma U87MG or LNZ308 cells overexpressing either wild-type (wt) EGFR or EGFRvIII were treated with nimotuzumab, temozolomide, or both. Expression and phosphorylation status of molecules were determined by Western blot analysis. Methylation status of promoter region of O(6) -methylguanine-DNA methyltransferase (MGMT) was detected by methylation-specific PCR. Antitumor activity was tested using nude mice bearing either subcutaneous or intracerebral xenografts along with analyses of EGFR phosphorylation status, proliferation, apoptosis, and vessel density. Nimotuzumab treatment resulted in reduction of EGFRvIII tyrosine phosphorylation with a decrease in Akt phosphorylation that was greater than that of wtEGFR. Correspondingly, antitumor effects, growth suppression and survival elongation, were more significant in mice bearing either subcutaneous or intracerebral tumor expressing EGFRvIII than in those expressing wtEGFR. These effects were markedly increased when temozolomide was combined with nimotuzumab. The post-treatment recurrent brain tumors exhibited a decrease in expression of the mismatch repair (MMR) proteins, MSH6 and MLH1, but their methylated MGMT status did not changed. Nimotuzumab has in vivo antitumor activity against GBM, especially those expressing EGFRvIII, when combined with temozolomide. This could provide a basis for preselection of patients with GBM by EGFR status who might benefit from the nimotuzumab and temozolomide combination therapy.
Collapse
Affiliation(s)
- Yusuke Nitta
- Department of Neurosurgery, Kugayama Hospital, 2-14-20 Kitakarasuyama, Setagaya, Tokyo, 157-0061, Japan.,Department of Neurosurgery, Kyorin University Faculty of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611, Japan
| | - Saki Shimizu
- Department of Neurosurgery, Kyorin University Faculty of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611, Japan
| | - Yukiko Shishido-Hara
- Department of Pathology, Kyorin University Faculty of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611, Japan.,Department of Anatomic Pathology, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku, Tokyo, 160-0023, Japan
| | - Kaori Suzuki
- Department of Neurosurgery, Kyorin University Faculty of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611, Japan
| | - Yoshiaki Shiokawa
- Department of Neurosurgery, Kyorin University Faculty of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611, Japan
| | - Motoo Nagane
- Department of Neurosurgery, Kyorin University Faculty of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611, Japan
| |
Collapse
|
26
|
Tabriz AG, Hermida MA, Leslie NR, Shu W. Three-dimensional bioprinting of complex cell laden alginate hydrogel structures. Biofabrication 2015; 7:045012. [PMID: 26689257 DOI: 10.1088/1758-5090/7/4/045012] [Citation(s) in RCA: 226] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Different bioprinting techniques have been used to produce cell-laden alginate hydrogel structures, however these approaches have been limited to 2D or simple three-dimension (3D) structures. In this study, a new extrusion based bioprinting technique was developed to produce more complex alginate hydrogel structures. This was achieved by dividing the alginate hydrogel cross-linking process into three stages: primary calcium ion cross-linking for printability of the gel, secondary calcium cross-linking for rigidity of the alginate hydrogel immediately after printing and tertiary barium ion cross-linking for long-term stability of the alginate hydrogel in culture medium. Simple 3D structures including tubes were first printed to ensure the feasibility of the bioprinting technique and then complex 3D structures such as branched vascular structures were successfully printed. The static stiffness of the alginate hydrogel after printing was 20.18 ± 1.62 KPa which was rigid enough to sustain the integrity of the complex 3D alginate hydrogel structure during the printing. The addition of 60 mM barium chloride was found to significantly extend the stability of the cross-linked alginate hydrogel from 3 d to beyond 11 d without compromising the cellular viability. The results based on cell bioprinting suggested that viability of U87-MG cells was 93 ± 0.9% immediately after bioprinting and cell viability maintained above 88% ± 4.3% in the alginate hydrogel over the period of 11 d.
Collapse
Affiliation(s)
- Atabak Ghanizadeh Tabriz
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, UK. Institute of Mechanical, Process and Energy Engineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, UK
| | | | | | | |
Collapse
|
27
|
Bouras A, Kaluzova M, Hadjipanayis CG. Radiosensitivity enhancement of radioresistant glioblastoma by epidermal growth factor receptor antibody-conjugated iron-oxide nanoparticles. J Neurooncol 2015; 124:13-22. [PMID: 25981803 DOI: 10.1007/s11060-015-1807-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/08/2015] [Indexed: 01/04/2023]
Abstract
The epidermal growth factor receptor deletion variant EGFRvIII is known to be expressed in a subset of patients with glioblastoma (GBM) tumors that enhances tumorigenicity and also accounts for radiation and chemotherapy resistance. Targeting the EGFRvIII deletion mutant may lead to improved GBM therapy and better patient prognosis. Multifunctional magnetic nanoparticles serve as a potential clinical tool that can provide cancer cell targeted drug delivery, imaging, and therapy. Our previous studies have shown that an EGFRvIII-specific antibody and cetuximab (an EGFR- and EGFRvIII-specific antibody), when bioconjugated to IONPs (EGFRvIII-IONPs or cetuximab-IONPs respectively), can simultaneously provide sensitive cancer cell detection by magnetic resonance imaging (MRI) and targeted therapy of experimental GBM. In this study, we investigated whether cetuximab-IONPs can additionally allow for the radiosensitivity enhancement of GBM. Cetuximab-IONPs were used in combination with single (10 Gy × 1) or multiple fractions (10 Gy × 2) of ionizing radiation (IR) for radiosensitization of EGFRvIII-overexpressing human GBM cells in vitro and in vivo after convection-enhanced delivery (CED). A significant GBM antitumor effect was observed in vitro after treatment with cetuximab-IONPs and subsequent single or fractionated IR. A significant increase in overall survival of nude mice implanted with human GBM xenografts was found after treatment by cetuximab-IONP CED and subsequent fractionated IR. Increased DNA double strands breaks (DSBs), as well as increased reactive oxygen species (ROS) formation, were felt to represent the mediators of the observed radiosensitization effect with the combination therapy of IR and cetuximab-IONPs treatment.
Collapse
Affiliation(s)
- Alexandros Bouras
- Brain Tumor Nanotechnology Laboratory, Department of Neurosurgery, Emory University School of Medicine, Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | | | | |
Collapse
|
28
|
Wichmann H, Güttler A, Bache M, Taubert H, Rot S, Kessler J, Eckert AW, Kappler M, Vordermark D. Targeting of EGFR and HER2 with therapeutic antibodies and siRNA: a comparative study in glioblastoma cells. Strahlenther Onkol 2014; 191:180-91. [PMID: 25159136 DOI: 10.1007/s00066-014-0743-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/16/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND The epidermal growth factor receptors, EGFR (HER1) and HER2, have proven prognostic relevance in a variety of human malignancies and both are functionally involved in the molecular pathogenesis of malignant gliomas. MATERIAL AND METHODS We selectively inhibited EGFR and HER2 in glioblastoma cell lines via EGFR- and HER2-specific siRNAs and through the binding of the therapeutic antibodies cetuximab and trastuzumab. The expression of EGFR and HER2 was verified by real-time PCR and western blot analyses. We examined the growth rate, cell cycle distribution, cell migration, clonogenic survival, and radiosensitivity of U251MG and LN-229 glioblastoma cell lines to determine the physiological and cell biological effects of EGFR and HER2 targeting. RESULTS EGFR and HER2 targeting using the therapeutic antibodies cetuximab and trastuzumab had no effect on cellular growth rate, cell cycle distribution, cell migration, clonogenic survival, and radiosensitivity in the cell lines U251 and LN-229. In contrast, siRNA knock-down of EGFR and HER2, reduced the growth rate by 40-65 %. The knock-down of EGFR did not change the cell migration rate in the cell lines U251 and LN-229. However, knock-down of HER2 reduced the cell migration rate by 50 %. Radiobiological analysis revealed that EGFR knock-down induced no radiosensitization in U251MG and LN-229 cells. However, the knock-down of HER2 induced radiosensitization in U251MG cells. CONCLUSION The epidermal growth factor receptor HER2 is a promising anti-tumor target for the therapy of glioblastoma. HER2 targeting may represent a promising strategy to induce cell physiological and radiobiological anti-tumor effects in glioblastoma.
Collapse
Affiliation(s)
- Henri Wichmann
- Department of Radiotherapy, Martin-Luther-University Halle-Wittenberg, Halle, Germany,
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Mäbert K, Cojoc M, Peitzsch C, Kurth I, Souchelnytskyi S, Dubrovska A. Cancer biomarker discovery: current status and future perspectives. Int J Radiat Biol 2014; 90:659-77. [PMID: 24524284 DOI: 10.3109/09553002.2014.892229] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE Cancer is a multigene disease which arises as a result of mutational and epigenetic changes coupled with activation of complex signaling networks. The use of biomarkers for early cancer detection, staging and individualization of therapy might improve patient care. A few fundamental issues such as tumor heterogeneity, a highly dynamic nature of the intrinsic and extrinsic determinants of radio- and chemoresistance, along with the plasticity and diversity of cancer stem cells (CSC) make biomarker development a challenging task. In this review we outline the preclinical strategies of cancer biomarker discovery including genomic, proteomic, metabolomic and microRNomic profiling, comparative genome hybridization (CGH), single nucleotide polymorphism (SNP) analysis, high throughput screening (HTS) and next generation sequencing (NGS). Other promising approaches such as assessment of circulating tumor cells (CTC), analysis of CSC-specific markers and cell-free circulating tumor DNA (ctDNA) are also discussed. CONCLUSIONS The emergence of powerful proteomic and genomic technologies in conjunction with advanced bioinformatic tools allows the simultaneous analysis of thousands of biological molecules. These techniques yield the discovery of new tumor signatures, which are sensitive and specific enough for early cancer detection, for monitoring disease progression and for proper treatment selection, paving the way to individualized cancer treatment.
Collapse
Affiliation(s)
- Katrin Mäbert
- OncoRay-National Center for Radiation Research in Oncology, Medical Faculty Dresden Carl Gustav Carus , TU Dresden , Germany
| | | | | | | | | | | |
Collapse
|
30
|
Templeton AK, Miyamoto S, Babu A, Munshi A, Ramesh R. Cancer stem cells: progress and challenges in lung cancer. Stem Cell Investig 2014; 1:9. [PMID: 27358855 DOI: 10.3978/j.issn.2306-9759.2014.03.06] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 03/07/2014] [Indexed: 12/17/2022]
Abstract
The identification of a subpopulation of tumor cells with stem cell-like characteristics first in hematological malignancies and later in solid tumors has emerged into a novel field of cancer research. It has been proposed that this aberrant population of cells now called "cancer stem cells" (CSCs) drives tumor initiation, progression, metastasis, recurrence, and drug resistance. CSCs have been shown to have the capacity of self-renewal and multipotency. Adopting strategies from the field of stem cell research has aided in identification, localization, and targeting of CSCs in many tumors. Despite the huge progress in other solid tumors such as brain, breast, and colon cancers no substantial advancements have been made in lung cancer. This is most likely due to the current rudimentary understanding of lung stem cell hierarchy and heterogeneous nature of lung disease. In this review, we will discuss the most recent findings related to identification of normal lung stem cells and CSCs, pathways involved in regulating the development of CSCs, and the importance of the stem cell niche in development and maintenance of CSCs. Additionally, we will examine the development and feasibility of novel CSC-targeted therapeutic strategies aimed at eradicating lung CSCs.
Collapse
Affiliation(s)
- Amanda K Templeton
- 1 Department of Pathology, 2 Peggy and Charles Stephenson Cancer Center, 3 Department of Radiation Oncology, 4 Graduate Program in Biomedical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Shinya Miyamoto
- 1 Department of Pathology, 2 Peggy and Charles Stephenson Cancer Center, 3 Department of Radiation Oncology, 4 Graduate Program in Biomedical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Anish Babu
- 1 Department of Pathology, 2 Peggy and Charles Stephenson Cancer Center, 3 Department of Radiation Oncology, 4 Graduate Program in Biomedical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Anupama Munshi
- 1 Department of Pathology, 2 Peggy and Charles Stephenson Cancer Center, 3 Department of Radiation Oncology, 4 Graduate Program in Biomedical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Rajagopal Ramesh
- 1 Department of Pathology, 2 Peggy and Charles Stephenson Cancer Center, 3 Department of Radiation Oncology, 4 Graduate Program in Biomedical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
31
|
Song H, Pan B, Yi J, Chen L. Featured article: autophagic activation with nimotuzumab enhanced chemosensitivity and radiosensitivity of esophageal squamous cell carcinoma. Exp Biol Med (Maywood) 2014; 239:529-41. [PMID: 24625442 DOI: 10.1177/1535370214525315] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Chemotherapy and radiotherapy are two indispensible methods for esophageal squamous cell carcinoma (ESCC), especially for those recurring and metastatic ones, but therapeutic toxicity remains a major problem to overcome. In the present study, the potential therapeutic value of nimotuzumab (an antiepidermal growth factor receptor [EGFR] monoclonal antibody) in combination with chemotherapy and radiotherapy was evaluated on Eca109 and TE-1 ESCC cells, with high and low expression of EGFR, respectively. It was shown that nimotuzumab enhanced the sensitivity of Eca109 cells to other cytotoxic agents (paclitaxel and cis-platinum) and X-ray radiation, and the cytotoxicity was associated with increased autophagy. Conversely, the chemo- and radio-sensitivity of TE-1 cells showed no improvement with addition of nimotuzumab, but could be increased by combining with rapamycin, an autophagy inducer. Therefore, it was concluded that autophagic activation mediated by nimotuzumab could promote autophagic cell death and produce additive antitumor effects.
Collapse
Affiliation(s)
- Haizhu Song
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, P.R. China
| | | | | | | |
Collapse
|
32
|
Solomon MT, Miranda N, Jorrín E, Chon I, Marinello JJ, Alert J, Lorenzo-Luaces P, Crombet T. Nimotuzumab in combination with radiotherapy in high grade glioma patients: a single institution experience. Cancer Biol Ther 2014; 15:504-9. [PMID: 24521695 DOI: 10.4161/cbt.28021] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Nimotuzumab, a humanized antibody targeting epidermal growth factor receptor, has potent anti-proliferative, anti-angiogenic, and pro-apoptotic effects in vitro and in vivo. It also reduces the number of radio-resistant CD133(+) glioma stem cells. The antibody has been extensively evaluated in patients with advanced head and neck, glioma, lung, esophageal, pancreatic, and gastric cancer. In this single institution experience, 35 patients with anaplastic astrocytoma (AA) or glioblastoma multiforme (GBM) were treated with irradiation and 200 mg doses of nimotuzumab. The first 6 doses were administered weekly, together with radiotherapy, and then treatment continued every 21 days until 1 year. The median number of doses was 12, and the median cumulative dose was thus 2400 mg of nimotuzumab. The most frequent treatment-related toxicities were increase in liver function tests, fever, nausea, anorexia, asthenia, dizziness, and tremors. These adverse reactions were classified as mild and moderate. The median survival time was 12.4 mo or 27.0 mo for patients with GBM or AA patients, respectively, who received curative-intent radiotherapy in combination with the antibody. The survival time of a matched population treated at the same hospital with irradiation alone was decreased (median 8.0 and 12.2 mo for GBM and AA patients, respectively) compared with that of the patients who received nimotuzumab and curative-intent radiotherapy. We have thus confirmed that nimotuzumab is a very well-tolerated drug, lacking cumulative toxicity after maintenance doses. This study, in a poor prognosis population, validates the previous data of survival gain after combining nimotuzumab and radiotherapy, in newly diagnosed high-grade glioma patients.
Collapse
Affiliation(s)
| | | | | | - Ivonne Chon
- National Institute of Oncology and Radiobiology; Havana, Cuba
| | | | - José Alert
- National Institute of Oncology and Radiobiology; Havana, Cuba
| | | | | |
Collapse
|
33
|
Cho DY, Lin SZ, Yang WK, Lee HC, Hsu DM, Lin HL, Chen CC, Liu CL, Lee WY, Ho LH. Targeting cancer stem cells for treatment of glioblastoma multiforme. Cell Transplant 2014; 22:731-9. [PMID: 23594862 DOI: 10.3727/096368912x655136] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cancer stem cells (CSCs) in glioblastoma multiforme (GBM) are radioresistant and chemoresistant, which eventually results in tumor recurrence. Targeting CSCs for treatment is the most crucial issue. There are five methods for targeting the CSCs of GBM. One is to develop a new chemotherapeutic agent specific to CSCs. A second is to use a radiosensitizer to enhance the radiotherapy effect on CSCs. A third is to use immune cells to attack the CSCs. In a fourth method, an agent is used to promote CSCs to differentiate into normal cells. Finally, ongoing gene therapy may be helpful. New therapeutic agents for targeting a signal pathway, such as epidermal growth factor (EGF) and vascular epidermal growth factor (VEGF) or protein kinase inhibitors, have been used for GBM but for CSCs the effects still require further evaluation. Nonsteroidal anti-inflammatory drugs (NSAIDs) such as cyclooxygenase-2 (Cox-2) inhibitors have proven to be effective for increasing radiation sensitivity of CSCs in culture. Autologous dendritic cells (DCs) are one of the promising immunotherapeutic agents in clinical trials and may provide another innovative method for eradication of CSCs. Bone-morphogenetic protein 4 (BMP4) is an agent used to induce CSCs to differentiate into normal glial cells. Research on gene therapy by viral vector is also being carried out in clinical trials. Targeting CSCs by eliminating the GBM tumor may provide an innovative way to reduce tumor recurrence by providing a synergistic effect with conventional treatment. The combination of conventional surgery, chemotherapy, and radiotherapy with stem cell-orientated therapy may provide a new promising treatment for reducing GBM recurrence and improving the survival rate.
Collapse
Affiliation(s)
- Der-Yang Cho
- Department of Neurosurgery, Neuropsychiatry Center, China Medical University Hospital, Taichung, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Blockage of a miR-21/EGFR regulatory feedback loop augments anti-EGFR therapy in glioblastomas. Cancer Lett 2013; 342:139-49. [PMID: 24012640 DOI: 10.1016/j.canlet.2013.08.043] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 08/25/2013] [Accepted: 08/28/2013] [Indexed: 01/23/2023]
Abstract
Epidermal growth factor receptors (EGFR) expression is frequently amplified in human glioblastoma cells. Nimotuzumab, a monoclonal antibody (mAb) against EGFR, has been used globally in clinics as an anti-cancer agent. It is largely unknown whether the blockade of miR-21, a microRNA that is upregulated in glioma cells, could amplify the effects of nimotuzumab. Herein, we have demonstrated that miR-21 directly targets von Hippel-Lindau (VHL) and peroxisome-proliferator-activated receptor α (PPARα) and that miR-21 regulates EGFR/AKT signaling through VHL/β-catenin and the PPARα/AP-1 axis. Further, the expression of miR-21 is regulated by EGFR via the activation of β-catenin and AP-1. These data indicate that a feedback loop exists between miR-21 and EGFR. We also show that the combination of nimotuzumab and an inhibitor of miR-21 is superior to single-agent therapy. These results clarify a novel association between miR-21 and EGFR in the regulation of cancer cell progression.
Collapse
|
35
|
Nimotuzumab enhances the radiosensitivity of cancer cells in vitro by inhibiting radiation-induced DNA damage repair. PLoS One 2013; 8:e70727. [PMID: 23976954 PMCID: PMC3745376 DOI: 10.1371/journal.pone.0070727] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 06/18/2013] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Nimotuzumab is a humanized IgG1 monoclonal antibody specifically targeting EGFR. In this study, we aimed to investigate the molecular mechanisms of nimotuzumab in its effects of enhancing cancer cell radiosensitivity. PRINCIPAL FINDING Lung cancer A549 cells and breast cancer MCF-7 cells were pretreated with or without nimotuzumab for 24 h before radiation to perform the clonogenic survival assay and to analyze the cell apoptosis by flow ctyometry. γ-H2AX foci were detected by confocal microscopy to assess the effect of nimotuzumab on radiation induced DNA repair. EGFR activation was examined and the levels of DNA damage repair related proteins in A549 cells at different time point and at varying doses exposure after nimotuzumab and radiation treatment were examined by Western blot. Pretreatment with nimotuzumab reduced clonogenic survival after radiation, inhibited radiation-induced EGFR activation and increased the radiation-induced apoptosis in both A549 cells and MCF-7 cells. The foci of γ-H2AX 24 h after radiation significantly increased in nimotuzumab pretreated cells with different doses. The phosphorylation of AKT and DNA-PKcs were remarkably inhibited in the combination group at each dose point as well as time point. CONCLUSIONS Our results revealed that the possible mechanism of nimotuzumab enhancing the cancer radiosensitivity is that nimotuzumab inhibited the radiation-induced activation of DNA-PKcs through blocking the PI3K/AKT pathway, which ultimately affected the DNA DSBs repair.
Collapse
|
36
|
Diaz-Miqueli A, Martinez GS. Nimotuzumab as a radiosensitizing agent in the treatment of high grade glioma: challenges and opportunities. Onco Targets Ther 2013; 6:931-42. [PMID: 23926436 PMCID: PMC3729249 DOI: 10.2147/ott.s33532] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Nimotuzumab is a humanized monoclonal antibody that binds specifically to human epidermal growth factor receptor, blocking receptor activation. Evidence of its radiosensitizing capacity has been widely evaluated. This article integrates published research findings regarding the role of nimotuzumab in the treatment of high grade glioma in combination with radiotherapy or radiochemotherapy in adult and pediatric populations. First, the mechanisms of action of nimotuzumab and its current applications in clinical trials containing both radiation and chemoradiation therapies are reviewed. Second, a comprehensive explanation of potential mechanisms driving radiosensitization by nimotuzumab in experimental settings is given. Finally, future directions of epidermal growth factor receptor targeting with nimotuzumab in combination with radiation containing regimens, based on its favorable toxicity profile, are proposed. It is hoped that this review may provide further insight into the rational design of new approaches employing nimotuzumab as a useful alternative for the therapeutic management of high grade glioma.
Collapse
|
37
|
Pinel S, Mriouah J, Vandamme M, Chateau A, Plénat F, Guérin E, Taillandier L, Bernier-Chastagner V, Merlin JL, Chastagner P. Synergistic antitumor effect between gefitinib and fractionated irradiation in anaplastic oligodendrogliomas cannot be predicted by the Egfr signaling activity. PLoS One 2013; 8:e68333. [PMID: 23874590 PMCID: PMC3715478 DOI: 10.1371/journal.pone.0068333] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 05/28/2013] [Indexed: 11/18/2022] Open
Abstract
In high-grade gliomas, the identification of patients that could benefit from EGFR inhibitors remains a challenge, hindering the use of these agents. Using xenografts models, we evaluated the antitumor effect of the combined treatment "gefitinib + radiotherapy" and aimed to identify the profile of responsive tumors. Expression of phosphorylated proteins involved in the EGFR-dependent signaling pathways was analyzed in 10 glioma models. We focused on three models of anaplastic oligodendrogliomas (TCG2, TCG3 and TCG4) harboring high levels of phospho-EGFR, phospho-AKT and phospho-MEK1. They were treated with gefitinib (GEF 75 mg/kg/day x 5 days/week, for 2 weeks) and/or fractionated radiotherapy (RT: 5x2Gy/week for 2 weeks). Our results showed that GEF and/or RT induced significant tumor growth delays. However, only the TCG3 xenografts were highly responsive to the combination GEF+RT, with ∼50% of tumor cure. Phosphoproteins analysis five days after treatment onset demonstrated in TCG3 xenografts, but not in TCG2 model, that the EGFR-dependent pathways were inhibited after GEF treatment. Moreover, TCG3-bearing mice receiving GEF monotherapy exhibited a transient beneficial therapeutic response, rapidly followed by tumor regrowth, along with a major vascular remodeling. Taken together, our data evoked an "EGFR-addictive" behavior for TCG3 tumors. This study confirms that combination of gefitinib with fractionated irradiation could be a potent therapeutic strategy for anaplastic oligodendrogliomas harboring EGFR abnormalities but this treatment seems mainly beneficial for "EGFR-addictive" tumors. Unfortunately, neither the usual molecular markers (EGFR amplification, PTEN loss) nor the basal overexpression of phosphoproteins were useful to distinguish this responsive tumor. Evaluating the impact of TKIs on the EGFR-dependent pathways during the treatment might be more relevant, and requires further validation.
Collapse
Affiliation(s)
- Sophie Pinel
- Université de Lorraine, CRAN, UMR 7039, Campus Science, Vandoeuvre-les-Nancy, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Solomón MT, Selva JC, Figueredo J, Vaquer J, Toledo C, Quintanal N, Salva S, Domíngez R, Alert J, Marinello JJ, Catalá M, Griego MG, Martell JA, Luaces PL, Ballesteros J, de-Castro N, Bach F, Crombet T. Radiotherapy plus nimotuzumab or placebo in the treatment of high grade glioma patients: results from a randomized, double blind trial. BMC Cancer 2013; 13:299. [PMID: 23782513 PMCID: PMC3691625 DOI: 10.1186/1471-2407-13-299] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 06/14/2013] [Indexed: 01/23/2023] Open
Abstract
Background The prognosis of patients bearing high grade glioma remains dismal. Epidermal Growth Factor Receptor (EGFR) is well validated as a primary contributor of glioma initiation and progression. Nimotuzumab is a humanized monoclonal antibody that recognizes the EGFR extracellular domain and reaches Central Nervous System tumors, in nonclinical and clinical setting. While it has similar activity when compared to other anti-EGFR antibodies, it does not induce skin toxicity or hypomagnesemia. Methods A randomized, double blind, multicentric clinical trial was conducted in high grade glioma patients (41 anaplastic astrocytoma and 29 glioblastoma multiforme) that received radiotherapy plus nimotuzumab or placebo. Treatment and placebo groups were well-balanced for the most important prognostic variables. Patients received 6 weekly doses of 200 mg nimotuzumab or placebo together with irradiation as induction therapy. Maintenance treatment was given for 1 year with subsequent doses administered every 3 weeks. The objectives of this study were to assess the comparative overall survival, progression free survival, response rate, immunogenicity and safety. Results The median cumulative dose was 3200 mg of nimotuzumab given over a median number of 16 doses. The combination of nimotuzumab and RT was well-tolerated. The most prevalent related adverse reactions included nausea, fever, tremors, anorexia and hepatic test alteration. No anti-idiotypic response was detected, confirming the antibody low immunogenicity. The mean and median survival time for subjects treated with nimotuzumab was 31.06 and 17.76 vs. 21.07 and 12.63 months for the control group. Conclusions In this randomized trial, nimotuzumab showed an excellent safety profile and significant survival benefit in combination with irradiation. Trial registration Cuban National Register for clinical trials (No. 1745) (http://registroclinico.sld.cu/ensayos).
Collapse
|
39
|
Perez R, Crombet T, de Leon J, Moreno E. A view on EGFR-targeted therapies from the oncogene-addiction perspective. Front Pharmacol 2013; 4:53. [PMID: 23637683 PMCID: PMC3636470 DOI: 10.3389/fphar.2013.00053] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 04/04/2013] [Indexed: 12/28/2022] Open
Abstract
Tumor cell growth and survival can often be impaired by inactivating a single oncogen– a phenomenon that has been called as “oncogene addiction.” It is in such scenarios that molecular targeted therapies may succeed. among known oncogenes, the epidermal growth factor receptor (EGFR) has become the target of different cancer therapies. So far, however, the clinical benefit from EGFR-targeted therapies has been rather limited. a critical review of the large amount of clinical data obtained with anti-EGFR agents, carried out from the perspective of the oncogene addiction concept, may help to understand the causes of the unsatisfactory results. In this article we intend to do such an exercise taking as basis for the analysis a few case studies of anti-EGFR agents that are currently in the clinic. There, the “EGFR addiction” phenomenon becomes apparent in high-responder patients. We further discuss how the concept of oncogene addiction needs to be interpreted on the light of emerging experimental evidences and ideas; in particular, that EGFR addiction may reflect the interconnection of several cellular pathways. In this regard we set forth several hypotheses; namely, that requirement of higher glucose uptake by hypoxic tumor cells may reinforce EGFR addiction; and that chronic use of EGFR-targeted antibodies in EGFR-addicted tumors would induce stable disease by reversing the malignant phenotype of cancer stem cells and also by sustaining an anti-tumor T cell response. Finally, we discuss possible reasons for the failure of certain combinatorial therapies involving anti-EGFR agents, arguing that some of these agents might produce either a negative or a positive trans-modulation effect on other oncogenes. It becomes evident that we need operational definitions of EGFR addiction in order to determine which patient populations may benefit from treatment with anti-EGFR drugs, and to improve the design of these therapies.
Collapse
Affiliation(s)
- Rolando Perez
- Center of Molecular Immunology Havana, Cuba ; Biotech Pharmaceuticals Co. Ltd. Beijing, China
| | | | | | | |
Collapse
|
40
|
Zhao L, He LR, Xi M, Cai MY, Shen JX, Li QQ, Liao YJ, Qian D, Feng ZZ, Zeng YX, Xie D, Liu MZ. Nimotuzumab promotes radiosensitivity of EGFR-overexpression esophageal squamous cell carcinoma cells by upregulating IGFBP-3. J Transl Med 2012; 10:249. [PMID: 23232108 PMCID: PMC3540017 DOI: 10.1186/1479-5876-10-249] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Accepted: 12/07/2012] [Indexed: 11/21/2022] Open
Abstract
Background Epidermal growth factor receptor (EGFR) is suggested to predict the radiosensitivity and/or prognosis of human esophageal squamous cell carcinoma (ESCC). The objective of this study was to investigate the efficacy of Nimotuzumab (an anti-EGFR monoclonal antibody) on ESCC radiotherapy (RT) and underlying mechanisms. Methods Nimotuzumab was administrated to 2 ESCC cell lines KYSE30 and TE-1 treated with RT. Cell growth, colony formation and apoptosis were used to measure anti-proliferation effects. The method of RNA interference was used to investigate the role of insulin-like growth factor binding protein-3 (IGFBP-3) in ESCC cells radiosensitivity treated with Nimotuzumab. In vivo effect of Nimotuzumab on ESCC radiotherapy was done using a mouse xenograft model. Results Nimotuzumab enhanced radiation response of KYSE30 cells (with high EGFR expression) in vitro, as evidenced by increased radiation-inhibited cell growth and colony formation and radiation-mediated apoptosis. Mechanism study revealed that Nimotuzumab inhibited phosphorylated EGFR (p-EGFR) induced by EGF in KYSE30 cells. In addition, knockdown of IGFBP-3 by short hairpin RNA significantly reduced KYSE30 cells radiosensitivity (P<0.05), and even after the administration of Nimotuzumab, the RT response of IGFBP-3 silenced KYSE30 cells was not enhanced (P>0.05). In KYSE30 cell xenografts, Nimotuzumab combined with radiation led to significant tumor growth delay, compared with that of radiation alone (P=0.029), and also with IGFBP-3 up-regulation in tumor tissue. Conclusions Nimotuzumab could enhance the RT effect of ESCC cells with a functional active EGFR pathway. In particular, the increased ESCC radiosensitivity by Nimotuzumab might be dependent on the up-regulation of IGFBP-3 through EGFR-dependent pathway.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, No 651 Dongfeng Road East, Guangzhou 510060, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Bode U, Massimino M, Bach F, Zimmermann M, Khuhlaeva E, Westphal M, Fleischhack G. Nimotuzumab treatment of malignant gliomas. Expert Opin Biol Ther 2012; 12:1649-59. [PMID: 23043252 DOI: 10.1517/14712598.2012.733367] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION In spite of new alkylating medication and recently accumulated knowledge about genomics, the prognosis of malignant gliomas remains poor. The introduction of single substances interfering with tumour proliferation dynamics has been disappointing and the lessons learned indicate that a complicated network of proliferation needs time consuming, in-depth analysis in order to more specifically treat now distinguishable subgroups of a disease, which too long was thought of as a uniform entity. AREAS COVERED The clinical trials using the EGFR antibody nimotuzumab in the treatment of malignant gliomas are reviewed. Pending conformation in future studies the antibody might be part of the treatment of MGMT-negative, EGFR-amplified, not completely resected gliomas of adulthood and juvenile DIPG (pontine gliomas). Upcoming genomic results of the different tumour entities may suggest certain combination partners of the antibody. Recent studies of nimotuzumab indicate the reason for the lack of toxicity, which is the most attractive argument for its clinical use besides modest efficacy. EXPERT OPINION We await the final results on the use of the antibody together with vinorelbine and radiation therapy for the therapy of DIPG. Adult patients with MGMT-negative, EGFR amplified, not totally resected GBM may also profit from this combination therapy. TK-inhibitors combined with the antibody and irradiation may be an option for a therapeutic trial in paediatric patients.
Collapse
Affiliation(s)
- Udo Bode
- University of Bonn Medical School, Department Paed. Haematology/Oncology, Bonn, Germany.
| | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Brain tumors--particularly glioblastoma multiforme--pose an important public health problem in the United States. Despite surgical and medical advances, the prognosis for patients with malignant gliomas remains grim: current therapy is insufficient with nearly universal recurrence. A major reason for this failure is the difficulty of delivering therapeutic agents to the brain: better delivery approaches are needed to improve treatment. In this article, we summarize recent progress in drug delivery to the brain, with an emphasis on convection-enhanced delivery of nanocarriers. We examine the potential of new delivery methods to permit novel drug- and gene-based therapies that target brain cancer stem cells and discuss the use of nanomaterials for imaging of tumors and drug delivery.
Collapse
|
43
|
Abstract
Glioblastoma multiforme is a histopathologically heterogeneous disease with few treatment options. Therapy based on genomic alterations is rapidly gaining popularity because of the high response rate and high specificity. DNA copy number and exon-sequencing studies of glioblastoma multiforme samples have revealed recurrent genomic alterations in genes such as TP53, EGFR, and IDH1, but to date, this has not resulted in novel glioblastoma multiforme therapies. Identification of expression subtypes has resulted in new insights such as the association between genomic abnormalities and expression signatures. This review describes the types of genomic studies that have been performed and that are underway, the most prominent results, and the implications of genomic research for the development of clinical treatment modalities.
Collapse
|
44
|
Tinhofer I, Hristozova T, Stromberger C, Keilhoiz U, Budach V. Monitoring of circulating tumor cells and their expression of EGFR/phospho-EGFR during combined radiotherapy regimens in locally advanced squamous cell carcinoma of the head and neck. Int J Radiat Oncol Biol Phys 2012; 83:e685-90. [PMID: 22583603 DOI: 10.1016/j.ijrobp.2012.02.009] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 02/02/2012] [Accepted: 02/03/2012] [Indexed: 12/11/2022]
Abstract
PURPOSE The numbers of circulating tumor cells (CTCs) and their expression/activation of epidermal growth factor receptor (EGFR) during the course of combined chemo- or bioradiotherapy regimens as potential biomarkers of treatment efficacy in squamous cell carcinoma of the head and neck (SCCHN) were determined. METHODS AND MATERIALS Peripheral blood samples from SCCHN patients with locally advanced stage IVA/B disease who were treated with concurrent radiochemotherapy or induction chemotherapy followed by bioradiation with cetuximab were included in this study. Using flow cytometry, the absolute number of CTCs per defined blood volume as well as their expression of EGFR and its phosphorylated form (pEGFR) during the course of treatment were assessed. RESULTS Before treatment, we detected ≥1 CTC per 3.75 mL blood in 9 of 31 patients (29%). Basal expression of EGFR was detected in 100% and pEGFR in 55% of the CTC+ cases. The frequency of CTC detection was not influenced by induction chemotherapy. However, the number of CTC+ samples significantly increased after radiotherapy. This radiation-induced increase in CTC numbers was less pronounced when radiotherapy was combined with cetuximab compared to its combination with cisplatin/5-fluorouracil. The former treatment regimen was also more effective in reducing pEGFR expression in CTCs. CONCLUSIONS Definitive radiotherapy regimens of locally advanced SCCHN can increase the number of CTCs and might thus contribute to a systemic spread of tumor cells. Further studies are needed to evaluate the predictive value of the radiation-induced increase in CTC numbers and the persistent activation of the EGFR signalling pathway in individual CTC+ cases.
Collapse
Affiliation(s)
- Ingeborg Tinhofer
- Translational Radiooncology Laboratory, Department of Radiooncology and Radiotherapy, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany.
| | | | | | | | | |
Collapse
|
45
|
Hong J, Peng Y, Liao Y, Jiang W, Wei R, Huo L, Han Z, Duan C, Zhong M. Nimotuzumab prolongs survival in patients with malignant gliomas: A phase I/II clinical study of concomitant radiochemotherapy with or without nimotuzumab. Exp Ther Med 2012; 4:151-157. [PMID: 23060940 DOI: 10.3892/etm.2012.555] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Accepted: 04/13/2012] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to determine whether nimotuzumab enhances the effect of radiochemotherapy in malignant gliomas. Patients (n=41) with malignant gliomas were divided into 20 cases (treatment group) in which nimotuzumab plus radiochemotherapy were offered and 21 cases (control group) in which placebo and radiochemotherapy were administered to the patients. The response to treatment was evaluated according to the Response Evaluation Criteria in Solid Tumors, the Kaplan-Meier method was used to calculate the mean and median survival times and 1-year survival rate, and the log-rank test and the Chi-square test were used to analyze the difference in the survival and response rate between the treatment and control groups. The mean survival times of the treatment and control groups were 14.3 and 10.4 months and the median survival times of the treatment and control groups were 16.5 and 10.5 months, respectively. The 1-year survival rates of the treatment and control groups were 81.3 and 69.1%, respectively, with no significant difference (P>0.05). The objective response rates of the treatment and control groups were 70.0 and 52.4%, respectively, with no significant difference (P>0.05). In conclusion, there was a trend towards improved treatment efficacy of radiochemotherapy combined with nimotuzumab against malignant gliomas. This study demonstrated that the use of nimotuzumab combined with radiotherapy and concomitant temozolomide chemotherapy is effective for malignant gliomas.
Collapse
|
46
|
The overexpression of IGFBP-3 is involved in the chemosensitivity of esophageal squamous cell carcinoma cells to nimotuzumab combined with cisplatin. Tumour Biol 2012; 33:1115-23. [PMID: 22389159 DOI: 10.1007/s13277-012-0352-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 02/08/2012] [Indexed: 10/28/2022] Open
Abstract
Nimotuzumab is an antibody against epidermal growth factor receptor (EGFR). The objective of this study was to examine the capacity and specific underlying mechanisms of nimotuzumab to modulate cytotoxicity of cisplatin (DDP) in esophageal squamous cell carcinoma (ESCC) cell lines with different EGFR expression levels. Nimotuzumab was administrated to two ESCC cell lines KYSE30 and TE-1 treated with DDP. Cell growth, colony formation, and apoptosis were analyzed by MTT and flow cytometry assays. The method of RNA interference was used to investigate the role of insulin-like growth factor binding protein-3 (IGFBP-3) in ESCC cells chemosensitivity treated with nimotuzumab. Combination of nimotuzumab and DDP resulted in a DDP cytotoxicity increase in overexpressing EGFR cells (KYSE30) but not in low-expressing EGFR cells (TE-1). Meantime, DDP activated the EGFR pathway in the two cell lines in a ligand-independent fashion. Furthermore, DDP-induced EGFR activation was inhibited by nimotuzumab in KYSE30 cells, and this result was not observed in TE-1 cells. EGF reduced the expression of IGFBP-3 in KYSE30 cells; however, nimotuzumab could reverse the downregulation of IGFBP-3, and this result was also not observed in TE-1 cells. After IGFBP-3 was silenced by small interfering RNA, the potential of nimotuzumab to enhance DDP-mediated cytotoxicity was inhibited in KYSE30 cells. The results indicated that the increased ESCC chemosensitivity to DDP by nimotuzumab might be dependent on IGFBP-3 upregulation through EGFR-dependent pathway, which would facilitate preselection of ESCC patients for treatment of nimotuzumab combined with DDP.
Collapse
|
47
|
Wang YX, Gao JX, Wang XY, Zhang L, Liu CM. Antiproliferative effects of selective cyclooxygenase-2 inhibitor modulated by nimotuzumab in estrogen-dependent breast cancer cells. Tumour Biol 2012; 33:957-66. [PMID: 22252523 DOI: 10.1007/s13277-012-0324-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Accepted: 01/04/2012] [Indexed: 11/30/2022] Open
Abstract
Breast cancer is the most common malignancy in women, and many breast cancer patients fail conventional treatment strategies of chemotherapy, radiation, and antiestrogen therapy. Research into the molecular pathways and biomarkers involved in the development of breast cancer should yield information that will guide therapeutic decisions. Epidermal growth factor receptor (EGFR) and cyclooxygenase-2 (COX-2) are involved in the carcinogenesis of breast cancer and exist tight crosstalk with estrogen receptor (ER) pathway. Combination of EGFR and COX-2 inhibitors, therefore, could be an effective strategy for reducing cell growth in estrogen-dependent breast cancer. In order to verify the effects of EGFR and COX-2 inhibitors, breast cancer cells MCF-7 and SKBR-3 were characterized for receptors status and then treated with respective inhibitors (nimotuzumab and celecoxib) alone and in combination. Both cell lines were sensitive to celecoxib, but not to nimotuzumab. However, combination of two drugs demonstrated synergistic effects on cell killing. Moreover, association of two drugs resulted in SKBR-3 cells, a further G0/G1 phase arrest than one drug alone. Downregulation of p-EGFR, p-Akt, p-mTOR, and amplified in breast cancer 1 (AIB1) were observed in both cell lines, and upregulation of E-cadherin was only found in MCF-7, after treatment with single agent or in combination. These studies suggest that nimotuzumab and celecoxib exert synergistic antiproliferation effects in breast cancer, which partly correlates with ER status. Due to Akt/mTOR, EMT and AIB1 pathways participate in this process, therefore, E-cadherin and AIB1 may be considered as possible biomarkers to predict response in ER-positive breast cancer cells treated with EGFR and COX-2 inhibitors.
Collapse
Affiliation(s)
- Ying-Xue Wang
- Department of Endocrinology, School of Clinical Medicine, Binzhou Medical University, No.661, Yellow-River Second Street, 256603 Binzhou, China.
| | | | | | | | | |
Collapse
|
48
|
Monoclonal antibody therapy for malignant glioma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 746:121-41. [PMID: 22639164 DOI: 10.1007/978-1-4614-3146-6_10] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Monoclonal antibody (mAb) therapy is a rapidly evolving treatment immunotherapy modality for malignant gliomas. Many studies have provided evidence that the blood brain barrier-both at baseline and in the context of malignancy-is permissive for mAbs, thus providing a rationale for their use in treating intracranial malignancy. Furthermore, techniques such as convection enhanced delivery (CED) are being implemented to maximize exposure of tumor cells to mAb therapy. The mechanisms and designs of mAbs are widely varying, including unarmed immunoglobulins as well as immunoglobulins conjugated to radioisotopes, biological toxins, boronated dendrimers and immunoliposomes. The very structure of the immunoglobulin molecule has also been manipulated to generate a diverse armamentarium including single-chain Fv, bispecific T-cell engagers and chimeric antigen receptors. The targeted neutralization capacity of mAbs has been employed to modulate the immunologic milieu in hopes of optimizing other immunotherapy platforms. Many clinical trials have evaluated these mAb strategies to treat malignant gliomas, and the implementation of mAb therapy seems imminent and optimistic.
Collapse
|
49
|
Diaz A, Blanco R, Lemm M, Fichtner I, Leon K, Montero E. Preclinical Efficacy of Nimotuzumab, an Anti-Egfr Monoclonal Antibody as a Single Agent Therapy in Human GBM u87mg Xenografts. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/jct.2012.34035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
50
|
Cho J, Pastorino S, Zeng Q, Xu X, Johnson W, Vandenberg S, Verhaak R, Cherniack A, Watanabe H, Dutt A, Kwon J, Chao YS, Onofrio RC, Chiang D, Yuza Y, Kesari S, Meyerson M. Glioblastoma-derived epidermal growth factor receptor carboxyl-terminal deletion mutants are transforming and are sensitive to EGFR-directed therapies. Cancer Res 2011; 71:7587-96. [PMID: 22001862 PMCID: PMC3242822 DOI: 10.1158/0008-5472.can-11-0821] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Genomic alterations of the epidermal growth factor receptor (EGFR) gene play a crucial role in pathogenesis of glioblastoma multiforme (GBM). By systematic analysis of GBM genomic data, we have identified and characterized a novel exon 27 deletion mutation occurring within the EGFR carboxyl-terminus domain (CTD), in addition to identifying additional examples of previously reported deletion mutations in this region. We show that the GBM-derived EGFR CTD deletion mutants are able to induce cellular transformation in vitro and in vivo in the absence of ligand and receptor autophosphorylation. Treatment with the EGFR-targeted monoclonal antibody, cetuximab, or the small molecule EGFR inhibitor, erlotinib, effectively impaired tumorigenicity of oncogenic EGFR CTD deletion mutants. Cetuximab in particular prolonged the survival of intracranially xenografted mice with oncogenic EGFR CTD deletion mutants, compared with untreated control mice. Therefore, we propose that erlotinib and, especially, cetuximab treatment may be a promising therapeutic strategy in GBM patients harboring EGFR CTD deletion mutants.
Collapse
Affiliation(s)
- Jeonghee Cho
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Genomic Analysis Center, Samsung Cancer Reseacrh Institute, Samsung Medical Center, Seoul, 135-710, Republic of Korea
| | - Sandra Pastorino
- Department of Neurosciences, Moores Cancer Center, UC San Diego, La Jolla, CA, 92093, USA
| | - Qing Zeng
- Department of Radiology, Brigham and Women’s Hospital, Boston, MA, 02115, USA
| | - Xiaoyin Xu
- Department of Radiology, Brigham and Women’s Hospital, Boston, MA, 02115, USA
| | - William Johnson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | | | - Roel Verhaak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Andrew Cherniack
- Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Hideo Watanabe
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Amit Dutt
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Jihyun Kwon
- Genomic Analysis Center, Samsung Cancer Reseacrh Institute, Samsung Medical Center, Seoul, 135-710, Republic of Korea
| | - Ying S. Chao
- Department of Neurosciences, Moores Cancer Center, UC San Diego, La Jolla, CA, 92093, USA
| | | | - Derek Chiang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Yuki Yuza
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Santosh Kesari
- Department of Neurosciences, Moores Cancer Center, UC San Diego, La Jolla, CA, 92093, USA
| | - Matthew Meyerson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| |
Collapse
|