1
|
Hu M, Tang B, Zhang D, Wang X, Zhao X. The inhibitory effects of nimotuzumab on CD276 expression and immune escape in head and neck squamous cell carcinoma: Insights into anticancer mechanisms. Int Immunopharmacol 2025; 147:114005. [PMID: 39778280 DOI: 10.1016/j.intimp.2024.114005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
CD276 has been identified as a novel immune checkpoint, and its overexpression is associated with immune evasion and poor prognosis in various tumors, including head and neck squamous cell carcinoma (HNSCC). Nimotuzumab, a humanized anti-epidermal growth factor receptor (EGFR) monoclonal antibody, has been approved for various solid tumors. However, it remains unclear whether its anticancer efficacy involves a reduction in CD276 expression. The purpose of this study was to investigate the regulatory effects and potential mechanisms of nimotuzumab on CD276 expression both in vitro and in vivo. In a coculture system, nimotuzumab showed inhibitory effects on TGF-β-induced upregulation of CD276 at both the transcriptional and protein levels in HNSCC cell lines. Mechanistic studies revealed that nimotuzumab primarily suppressed TGF-β-induced CD276 upregulation by blocking EGFR/MEK/ERK, which was further validated by MEK and ERK inhibitors. In xenograft and mice HNSCC models, nimotuzumab exerted antitumor effects accompanied by significantly reduced CD276 expression during tumor progression. Analysis of tumor-infiltrating lymphocytes (TILs) profiles indicated that nimotuzumab orchestrated the tumor immune microenvironment (TIME) by notably increasing the frequency of T lymphocytes, including cytotoxic T lymphocytes and helper T lymphocytes, as well as macrophage cells. However, no significant changes were observed in the populations of NK cells, DC cells, and neutrophils. These findings offer new insights into the anticancer mechanisms of nimotuzumab and its underlying synergy in combined treatments with immunotherapy for HNSCC.
Collapse
MESH Headings
- Humans
- Animals
- Squamous Cell Carcinoma of Head and Neck/drug therapy
- Squamous Cell Carcinoma of Head and Neck/immunology
- Squamous Cell Carcinoma of Head and Neck/pathology
- Head and Neck Neoplasms/drug therapy
- Head and Neck Neoplasms/immunology
- Head and Neck Neoplasms/pathology
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Cell Line, Tumor
- Mice
- Tumor Escape/drug effects
- Tumor Microenvironment/drug effects
- Xenograft Model Antitumor Assays
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/drug effects
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- ErbB Receptors/antagonists & inhibitors
- Female
- Transforming Growth Factor beta/metabolism
- Mice, Nude
Collapse
Affiliation(s)
- Minwan Hu
- Department of Clinical Pharmacology, Affiliated Beijing Luhe Hospital of Capital Medical University, Beijing 101149, PR China; School of Pharmaceutical Sciences, Capital Medical University, Beijing, PR China; Department of National Institute for Drug Clinical Trial, Affiliated Beijing Tongren Hospital of Capital Medical University, Beijing 100005, PR China
| | - Borui Tang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, PR China; Department of National Institute for Drug Clinical Trial, Affiliated Beijing Tongren Hospital of Capital Medical University, Beijing 100005, PR China
| | - Di Zhang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, PR China; Department of National Institute for Drug Clinical Trial, Affiliated Beijing Tongren Hospital of Capital Medical University, Beijing 100005, PR China
| | - Xuhong Wang
- Department of Clinical Pharmacology, Affiliated Beijing Luhe Hospital of Capital Medical University, Beijing 101149, PR China
| | - Xiuli Zhao
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, PR China; Department of National Institute for Drug Clinical Trial, Affiliated Beijing Tongren Hospital of Capital Medical University, Beijing 100005, PR China.
| |
Collapse
|
2
|
Wang X, Yao C, Quan L, Zhou J. A review on intrathecal administration of medications for leptomeningeal metastases in solid tumors. Front Pharmacol 2025; 16:1472945. [PMID: 39981182 PMCID: PMC11841460 DOI: 10.3389/fphar.2025.1472945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 01/08/2025] [Indexed: 02/22/2025] Open
Abstract
Leptomeningeal disease (LMD) is a particular mode of central metastasis in malignant tumors. It occurs when tumor cells infiltrate the subarachnoid space and cerebrospinal fluid (CSF), spreading throughout the central nervous system (CNS). LMD is a rare but devastating complication of malignant tumors. It can occur in various types of cancers, with lung and breast cancer being the most frequently associated. The treatment approach for LMD includes a combination of supportive care, surgery, chemotherapy, radiotherapy, targeted therapy, immunotherapy, and intrathecal (IT) therapy, among other modalities. Despite the challenges in determining the optimal treatment for LMD, IT therapy remains one of the primary therapeutic strategies. This therapy can directly circumvent the blood-brain barrier. Moreover, a low-dose medication can achieve a higher drug concentration in the CSF, resulting in better cytotoxic effects. Chemotherapy drugs such as methotrexate, cytarabine, and thiotepa have been widely studied as traditional IT therapies. In recent years, the advent of novel anti-tumor drugs has led to a growing number of agents being employed for IT administration in the treatment of malignant tumors with LMD. This article presents a comprehensive review of the current advancements in IT administration of chemotherapy, targeted, and immunotherapy drugs for the treatment of LMD in solid tumors. In addition, we also discuss the safety issues associated with IT therapy, summarize the advantages of IT administration of different types of anti-tumor drugs, and put forward some suggestions for reducing adverse reactions. It is hoped that future research will focus on exploring more potentially effective anti-tumor drugs for IT treatment, conducting in-depth pharmacokinetic studies, and developing long-acting and low-toxic IT administration regimens for the treatment of meningeal metastases.
Collapse
Affiliation(s)
- Xuemei Wang
- Department of Clinical Pharmacy, Shifang People’s Hospital, North Sichuan Medical College, Deyang, China
| | - Chi Yao
- Department of Hepatobiliary Surgery, Shifang People’s Hospital, North Sichuan Medical College, Deyang, China
| | - Li Quan
- Department of Clinical Pharmacy, Shifang People’s Hospital, North Sichuan Medical College, Deyang, China
| | - Junxiang Zhou
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Afffliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
3
|
Hu M, Tang B, Dai Y, Zhao X. Unveiling the regulatory mechanism of nimotuzumab on PD-L1 expression in head and neck squamous cell carcinoma patients: Implications for enhanced anticancer treatment strategies. Cell Signal 2024; 121:111290. [PMID: 38977231 DOI: 10.1016/j.cellsig.2024.111290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/15/2024] [Accepted: 07/04/2024] [Indexed: 07/10/2024]
Abstract
The overexpression of programmed death ligand 1 (PD-L1) is associated with resistance to anticancer therapies and poor prognosis in patients with head and neck squamous cell carcinoma (HNSCC). Nimotuzumab, a humanized anti-epidermal growth factor receptor (EGFR) mAb, has been widely used clinically for treating several solid tumors. However, whether its anticancer effect involves a reduction in PD-L1 expression remains unclear. The current study aimed to investigate the regulatory effects and underlying mechanism of nimotuzumab on PD-L1 expression in HNSCC both in vitro and in vivo. In vitro, nimotuzumab inhibited IFN-γ-induced PD-L1 upregulation at both the transcriptional and protein levels in the HNSCC cell lines. Subsequent mechanism research revealed that nimotuzumab suppressed IFN-γ-stimulated PD-L1 upregulation mainly by inhibiting phosphorylation of EGFR/MEK/ERK pathway, which was further validated by MEK and ERK inhibitors. In a HNSCC tumor-bearing model, nimotuzumab significantly decreased PD-L1 expression during tumor progression or chemotherapy, and this reduction was accompanied by increased sensitivity of the tumor to docetaxel and atezolizumab. Additionally, nimotuzumab reversed PD-L1 upregulation when combined with Taxol + Cisplatin (TP) induction chemotherapy regimens and improved the CD4+ and CD8+ T cells infiltration in HNSCC patients. These findings provide new insights into the anticancer mechanisms of nimotuzumab in HNSCC.
Collapse
Affiliation(s)
- Minwan Hu
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, PR China; Department of National Institute for Drug Clinical Trial, Affiliated Beijing Tongren Hospital of Capital Medical University, Beijing 100005, PR China
| | - Borui Tang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, PR China; Department of National Institute for Drug Clinical Trial, Affiliated Beijing Tongren Hospital of Capital Medical University, Beijing 100005, PR China
| | - Yuyang Dai
- Department of National Institute for Drug Clinical Trial, Affiliated Beijing Tongren Hospital of Capital Medical University, Beijing 100005, PR China
| | - Xiuli Zhao
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, PR China; Department of National Institute for Drug Clinical Trial, Affiliated Beijing Tongren Hospital of Capital Medical University, Beijing 100005, PR China.
| |
Collapse
|
4
|
Sabatelle RC, Colson YL, Sachdeva U, Grinstaff MW. Drug Delivery Opportunities in Esophageal Cancer: Current Treatments and Future Prospects. Mol Pharm 2024; 21:3103-3120. [PMID: 38888089 PMCID: PMC11331583 DOI: 10.1021/acs.molpharmaceut.4c00246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
With one of the highest mortality rates of all malignancies, the 5-year survival rate for esophageal cancer is under 20%. Depending on the stage and extent of the disease, the current standard of care treatment paradigm includes chemotherapy or chemoradiotherapy followed by surgical esophagogastrectomy, with consideration for adjuvant immunotherapy for residual disease. This regimen has high morbidity, due to anatomic changes inherent in surgery, the acuity of surgical complications, and off-target effects of systemic chemotherapy and immunotherapy. We begin with a review of current treatments, then discuss new and emerging targets for therapies and advanced drug delivery systems. Recent and ongoing preclinical and early clinical studies are evaluating traditional tumor targets (e.g., human epidermal growth factor receptor 2), as well as promising new targets such as Yes-associated protein 1 or mammalian target of rapamycin to develop new treatments for this disease. Due the function and location of the esophagus, opportunities also exist to pair these treatments with a drug delivery strategy to increase tumor targeting, bioavailability, and intratumor concentrations, with the two most common delivery platforms being stents and nanoparticles. Finally, early results with antibody drug conjugates and chimeric antigenic receptor T cells show promise as upcoming therapies. This review discusses these innovations in therapeutics and drug delivery in the context of their successes and failures, with the goal of identifying those solutions that demonstrate the most promise to shift the paradigm in treating this deadly disease.
Collapse
Affiliation(s)
- Robert C. Sabatelle
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA, 02215, USA
| | - Yolonda L. Colson
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Uma Sachdeva
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Mark W. Grinstaff
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA, 02215, USA
| |
Collapse
|
5
|
Wu WJ, An PG, Zhong YW, Hu X, Wang L, Zhang J. Tislelizumab plus nimotuzumab is effective against recurrent or metastatic oral squamous cell carcinoma among patients with a performance status score ≥ 2: a retrospective study. Front Oncol 2024; 13:1273798. [PMID: 38293699 PMCID: PMC10824828 DOI: 10.3389/fonc.2023.1273798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 12/31/2023] [Indexed: 02/01/2024] Open
Abstract
Objectives The efficacy of treatments targeting recurrent or metastatic head and neck squamous cell carcinoma are unsatisfactory in practice for patients with a ECOG PS score ≥ 2. Thus, this study retrospectively evaluated the safety and efficacy of a programmed cell death 1 inhibitor (tislelizumab) combined with an epidermal growth factor receptor inhibitor (nimotuzumab) in treating patients with a PS score ≥ 2 who suffer from recurrent or metastatic oral squamous cell carcinoma (OSCC). Materials and methods Fifteen patients were treated with tislelizumab (200 mg IV Q3W) and nimotuzumab (200 mg IV Q3W). Programmed cell death-ligand 1 (PD-L1) expression in tumor biopsies was assessed with immunohistochemistry. Whole-exome sequencing was used to evaluate treatment efficacy based on PD-L1 expression and gene mutation. Results At a median follow-up of 9.6 months, median overall survival was 10.1 months, and median progression-free survival was 4.0 months. Overall response rate was 40%, with 6/15 patients achieving partial response. Eight patients exhibited nine adverse events, eight out of nine being grade 2 and the remaining being grade 3. Whole-exome sequencing showed that DYNC1I2, THSD7A, and FAT1 mutations were associated with patient prognosis. Conclusion Combination therapy involving tislelizumab plus nimotuzumab is a promising, low-toxicity treatment for recurrent or metastatic OSCC in patients with a PS score ≥ 2.
Collapse
Affiliation(s)
- Wen-Jie Wu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- National Center of Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Pu-Gen An
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- National Center of Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yi-Wei Zhong
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- National Center of Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xiao Hu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- National Center of Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Lin Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- National Center of Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Jie Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- National Center of Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
6
|
Cai Z, Chen D, Qiu W, Liang C, Huang Y, Zhou J, Zhan Z, Xiang Y, Guo X, Lv X. Concurrent chemoradiotherapy combined with nimotuzumab in stage III–IVa nasopharyngeal carcinoma: a retrospective analysis. J Cancer Res Clin Oncol 2022; 149:2327-2344. [PMID: 36289067 DOI: 10.1007/s00432-022-04355-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 09/07/2022] [Indexed: 11/24/2022]
Abstract
PURPOSE The efficacy and safety of nimotuzumab (NTZ) added to concurrent chemoradiotherapy (CCRT) were investigated in patients with stage III-IVa nasopharyngeal carcinoma (NPC). METHODS Patients with stage III-IVa NPC treated with CCRT, with or without NTZ, were screened between January 2015 and December 2017. We compared patients' overall survival (OS), progression-free survival (PFS), locoregional recurrence-free survival (LRFS), and distant metastasis-free survival (DMFS) between different therapeutic regimens. Propensity score matching (PSM) was applied to reduce the selection bias. Nomogram models were developed to predict the survival of CCRT with or without NTZ. RESULTS Four hundred and twenty-six patients were included after PSM, with 213 patients in each regimen. Compared with NPC patients receiving CCRT alone, patients who received NTZ plus CCRT treatment had significantly better OS (5 year OS, 76.1 vs. 72.3%, P = 0.004), PFS (5 year PFS, 73.2 vs. 69.0%, P = 0.002), and LRFS (5 year LRFS, 73.2 vs. 69.0%, P = 0.028). A multivariate Cox regression analysis demonstrated that, compared with receiving CCRT alone, NTZ plus CCRT was an independently positive factor for OS, PFS, and LRFS. No significant difference was observed in the major toxicities between the two treatments (all P > 0.05). In addition, the nomogram presented good accuracy for predicting the prognosis of NPC patients. CONCLUSION CCRT combined with NTZ presented favorable clinical outcomes for stage III-IVa NPC patients with good tolerance and similar toxicity compared to CCRT alone. A prospective, randomized clinical trial is essential to validate the current findings.
Collapse
Affiliation(s)
- Zhuochen Cai
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Dongni Chen
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510089, Guangdong, People's Republic of China
| | - Wenze Qiu
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, No 78, Hengzhigang Road, Yuexiu District, Guangzhou, 510095, Guangdong, People's Republic of China
| | - Chixiong Liang
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Yingying Huang
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Jiayu Zhou
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Zejiang Zhan
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Yanqun Xiang
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Xiang Guo
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China.
| | - Xing Lv
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China.
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Hu J, Chen Z, Lv J, Zheng Z, Bei Y, Chen X, Zheng L, Song W, Xu Y. The Application of Nimotuzumab Combined With Definitive Chemoradiotherapy Toward the Treatment of Locally Advanced Cervical Esophageal Carcinoma: A Retrospective Study. Front Oncol 2022; 12:905422. [PMID: 35898885 PMCID: PMC9310542 DOI: 10.3389/fonc.2022.905422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/07/2022] [Indexed: 11/25/2022] Open
Abstract
Objective To evaluate the safety and effectiveness of nimotuzumab in combination with chemoradiotherapy for locally advanced cervical esophageal squamous cell carcinoma. Methods Retrospective analysis was conducted from September 2012 to February 2017 among 50 locoregional-advanced cervical esophageal carcinoma (CEC) patients who received concurrent chemoradiotherapy (CRT) combined with or without nimotuzumab at Ningbo Medical Center Lihuili Hospital. Intensity-modulated radiotherapy (IMRT) was administrated on all patients. All patients were divided into two groups, of which 26 (Group A) received 200 mg (22 of 50) or 400 mg (4 of 50) of nimotuzumab per week with CRT and 24 (Group B) received definitive CRT. Results The median follow-up time was 23 months. The median overall survival (OS) and progression-free survival (PFS) were 40.6 and 21.1 months for all, respectively. The 1-, 2-, and 3-year OS rates on the whole were 79.6%, 62.1%, and 47.8%. There was no statistical difference in overall response rate and disease control rate between the two groups. Patients treated with nimotuzumab (group A) had better PFS than the definitive CRT group (group B) (P < 0.05). However, the median OS was 41.4 months in group A and 32.4 months in group B, respectively (P = 0.517). Multivariate analysis showed that PFS among those with lower Eastern Cooperative Oncology Group (ECOG) score (HR = 5.11; P < 0.01), stage II (HR = 9.52; P < 0.01) and the application of nimotuzumab combined with CRT (HR = 0.16; P < 0.01) was much longer. Furthermore, ECOG, stage, C-reactive protein (CRP) baseline, and histological grade can also be used as independent predictors of OS. Grade >3 adverse reactions were not observed. The most common adverse event related to nimotuzumab was mild fever and the occurrence rate was 19% (5 of 26). The incidence of anemia was 65.4% in group A and 87.5% in group B (P < 0.05). Conclusions For locoregional-advanced CEC, nimotuzumab combined with IMRT and concomitant chemotherapy was tolerated and effective. In addition, patients with a normal pretherapeutic serum CRP level (CRP < 10 mg/L) can achieve better OS.
Collapse
Affiliation(s)
- Jing Hu
- Department of Radiation Oncology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Zhe Chen
- Department of Thoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Jiaming Lv
- Department of Radiation Oncology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Zhen Zheng
- Department of Radiation Oncology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Yanping Bei
- Department of Radiation Oncology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Xue Chen
- Department of Radiation Oncology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Lu Zheng
- Department of Radiation Oncology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Wenjie Song
- Department of Thoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Yunbao Xu
- Department of Radiotherapy and Chemotherapy, Hwamei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| |
Collapse
|
10
|
Diaz H, Jiménez J, Hernández A, Valdés L, Martínez A, Porto L, Hernández R, Travieso N, Jova JH, Medel L, Troche M, Gorte A, Batista D, Valls AR, Cabrera L, Domeq M, Pérez L, Lorenzo-Luaces P, Sánchez L, Saavedra D, Ramos M, Crombet T. Nimotuzumab Increases the Recovery Rate of Severe and Critical COVID-19 Patients: Evaluation in the Real-World Scenario. Front Public Health 2022; 10:948520. [PMID: 35937253 PMCID: PMC9353117 DOI: 10.3389/fpubh.2022.948520] [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] [Received: 05/19/2022] [Accepted: 06/23/2022] [Indexed: 12/15/2022] Open
Abstract
EGFR signaling is an important regulator of SARS-CoV induced lung damage, inflammation and fibrosis. Nimotuzumab is a humanized anti-EGFR antibody registered for several cancer indications. An expanded access study was conducted to evaluate the safety and recovery rate of severe and critical patients with confirmed SARS-CoV-2 infection, treated with nimotuzumab in combination with the standard of care in the real-world scenario. The antibody was administered as an intravenous infusions every 72 h, up to 5 doses. In order to assess the impact of nimotuzumab, the recovery rate was compared with a paired retrospective cohort. Control patients received standard treatment according the national protocol but not nimotuzumab. Overall, 1,151 severe or critical patients received nimotuzumab in 21 hospitals of Cuba. Median age was 65 and 773 patients had at least one comorbidity. Nimotuzumab was very well-tolerated and mild or moderate adverse events were detected in 19 patients. 1,009 controls matching with the nimotuzumab patients, were selected using a "propensity score" method. The 14-day recovery rate of the nimotuzumab cohort was 72 vs. 42% in the control group. Controls had a higher mortality risk (RR 2.08, 95% CI: 1.79, 2.38) than the nimotuzumab treated patients. The attributable fraction was 0.52 (95% CI: 0.44%; 0.58), and indicates the proportion of deaths that were prevented with nimotuzumab. Our preliminary results suggest that nimotuzumab is a safe antibody that can reduce the mortality of severe and critical COVID-19 patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Loipa Medel
- Center of Molecular Immunology, Havana, Cuba
| | | | - Annia Gorte
- Center of Molecular Immunology, Havana, Cuba
| | | | | | | | | | | | | | | | | | - Mayra Ramos
- Center of Molecular Immunology, Havana, Cuba
| | | |
Collapse
|
11
|
Londres HD, Armada JJ, Martínez AH, Abdo Cuza AA, Sánchez YH, Rodríguez AG, Figueroa SS, Llanez Gregorich EM, Torres Lahera ML, Peire FG, González TM, González YZ, Añé Kouri AL, Palomo AG, Concepción MT, Pérez LM, Luaces-Alvarez PL, Iglesias DE, Hernández DS, Suzarte MR, Ramos TC. Blocking EGFR with nimotuzumab: a novel strategy for COVID-19 treatment. Immunotherapy 2022; 14:521-530. [PMID: 35306855 PMCID: PMC8936166 DOI: 10.2217/imt-2022-0027] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background: Lung injury and STAT1 deficit induce EGFR overexpression in SARS-CoV-2 infection. Patients & methods: A phase I/II trial was done to evaluate the safety and preliminary effect of nimotuzumab, an anti-EGFR antibody, in COVID-19 patients. Patients received from one to three infusions together with other drugs included in the national guideline. Results: 41 patients (31 severe and 10 moderate) received nimotuzumab. The median age was 62 years and the main comorbidities were hypertension, diabetes and cardiovascular disease. The antibody was very safe and the 14-day recovery rate was 82.9%. Inflammatory markers decreased over time. Patients did not show signs of fibrosis. Conclusion: Nimotuzumab is a safe antibody that might reduce inflammation and prevent fibrosis in severe and moderate COVID-19 patients. Clinical Trial Registration: RPCEC00000369 (rpcec.sld.cu). Background: After SARS-CoV-2 infection, many cells in the lung express a new receptor called EGFR. Overexpression of EGFR can worsen the pulmonary disease and provoke fibrosis. Patients & methods: The initial impact of using a drug that blocks EGFR, nimotuzumab, was evaluated in COVID-19 patients. Results: 41 patients received nimotuzumab by the intravenous route together with other medications. The median age was 62 years, and patients had many chronic conditions including hypertension, diabetes and cardiac problems. Treatment was well tolerated and 82.9% of the patients were discharged by day 14. Serial laboratory tests, x-rays and CT scan evaluations showed the improvement of the patients. Conclusion: Nimotuzumab is a safe drug that can be useful to treat COVID-19 patients.
Collapse
|
12
|
Sastyarina Y, Firdaus ARR, Nafisah Z, Hardianto A, Yusuf M, Ramli M, Mutalib A, Soedjanaatmadja UM. Modeling and Molecular Dynamic Simulation of F(ab') 2 Fragment of Nimotuzumab for Lung Cancer Diagnostics. Bioinform Biol Insights 2022; 15:11779322211002174. [PMID: 35173422 PMCID: PMC8842462 DOI: 10.1177/11779322211002174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 02/22/2021] [Indexed: 01/03/2023] Open
Abstract
Lung cancer is one of the leading causes of cancer-related deaths in the world among both men and women. Several studies in the literature report that overexpression and mutation of the epidermal growth factor receptor (EGFR) are implicated in the pathogenesis of some lung cancers. Nimotuzumab is a humanized monoclonal antibody (mAb) that inhibits EGF binding because it binds to the extracellular domain of the EGFR. Nimotuzumab requires bivalent binding for stable attachment to cellular surface, which leads to nimotuzumab selectively binding to cells that express mAbs of moderate to high EGFR levels, and this could explain its low toxicity. This property has an advantage for development of nimotuzumab as a therapeutic and diagnostic agent. Monoclonal antibodies are large in size (150 kDa), thus penetrating slowly and residing in the blood for extended periods of time (from days to weeks); their use in imaging studies can result in low signal-to-background ratios and poor image quality. A reduction in the size of the immunoglobulin molecule has also been proposed as a means for increasing tumor penetration by mAbs. Nevertheless, it is known that the penetration of mAb into tumor cell is slow, due to its high molecular weight. Therefore, mAb is not very attractive to be used for imaging diagnostic purpose because of its kinetics and potential to elicit antibody response. The objective of this research was to study the homology modeling of a simpler functional molecule based on nimotuzumab, which consists of 2 antigen-binding fragments (Fab), namely, F(ab′)2, using MODELER. The crystal structure of Fab of nimotuzumab from protein data bank was used as a template to construct the model of F(ab′)2. Molecular dynamic simulation was performed to evaluate the stability of F(ab′)2 and conformational changes of F(ab′)2 in simulation. The result showed the dynamic behavior of antigen-binding site region of F(ab′)2 throughout simulation. This result is expected to be useful in the further development of F(ab′)2 fragment nimotuzumab as a lung cancer diagnostic.
Collapse
Affiliation(s)
| | - Ade Rizqi Ridwan Firdaus
- Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Padjadjaran, Bandung, Indonesia
| | - Zuhrotun Nafisah
- Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Padjadjaran, Bandung, Indonesia
| | - Ari Hardianto
- Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Padjadjaran, Bandung, Indonesia
| | - Muhammad Yusuf
- Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Padjadjaran, Bandung, Indonesia
| | - Martalena Ramli
- Center for Radioisotopes and Radiopharmaceutical Tehnology, National Nuclear Agency (BATAN) Puspitek-Serpong, Tangerang, Indonesia
| | - Abdul Mutalib
- Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Padjadjaran, Bandung, Indonesia
| | - Ukun Ms Soedjanaatmadja
- Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
13
|
Lu J, Ding J, Liu Z, Chen T. Retrospective analysis of the preparation and application of immunotherapy in cancer treatment (Review). Int J Oncol 2022; 60:12. [PMID: 34981814 PMCID: PMC8759346 DOI: 10.3892/ijo.2022.5302] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/20/2021] [Indexed: 12/11/2022] Open
Abstract
Monoclonal antibody technology plays a vital role in biomedical and immunotherapy, which greatly promotes the study of the structure and function of genes and proteins. To date, monoclonal antibodies have gone through four stages: murine monoclonal antibody, chimeric monoclonal antibody, humanised monoclonal antibody and fully human monoclonal antibody; thousands of monoclonal antibodies have been used in the fields of biology and medicine, playing a special role in the pathogenesis, diagnosis and treatment of disease. In this review, we compare the advantages and disadvantages of hybridoma technology, phage display technology, ribosome display technology, transgenic mouse technology, single B cell monoclonal antibody generation technologies, and forecast the promising applications of these technologies in clinical medicine, disease diagnosis and tumour treatment.
Collapse
Affiliation(s)
- Jiachen Lu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jianing Ding
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhaoxia Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Tingtao Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
14
|
Abdo Cuza AA, Ávila JP, Martínez RM, González JJ, Aspuro GP, Gutiérrez Martínez JA, Suzarte MR, Hernández DS, Añé-Kouri AL, Ramos TC. Nimotuzumab for COVID-19: case series. Immunotherapy 2021; 14:185-193. [PMID: 34806405 PMCID: PMC8628863 DOI: 10.2217/imt-2021-0269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/08/2021] [Indexed: 12/15/2022] Open
Abstract
Background: In COVID-19, EGFR production is upregulated in the alveolar epithelial cells. EGFR overexpression further activates STAT-3 and increases lung pathology. The EGFR pathway is also one of the major nodes in pulmonary fibrosis. Methods: Nimotuzumab, a humanized anti-EGFR antibody, was used to treat three patients with severe or moderate COVID-19. The antibody was administered in combination with other drugs included in the national COVID-19 protocol. Results: Nimotuzumab was well tolerated. IL-6 decreased from the first antibody infusion. Clinical symptoms significantly improved after nimotuzumab administration, and the CT scans at discharge showed major resolution of the lung lesions and no signs of fibrosis. Conclusion: Safe anti-EGFR antibodies like nimotuzumab may modulate COVID-19-associated hyperinflammation and prevent fibrosis. Clinical Trial Registration: RPCEC00000369 (RPCEC rpcec.sld.cu).
Collapse
Affiliation(s)
- Anselmo A Abdo Cuza
- Intensive Care Unit. Medical & Surgical Research Center (CIMEQ), Havana, Cuba
| | - Jonathan Pi Ávila
- Intensive Care Unit. Medical & Surgical Research Center (CIMEQ), Havana, Cuba
| | | | | | | | | | - Mayra Ramos Suzarte
- Clinical Research Direction. Center of Molecular Immunology (CIM), Havana, Cuba
| | | | - Ana L Añé-Kouri
- Clinical Research Direction. Center of Molecular Immunology (CIM), Havana, Cuba
| | - Tania Crombet Ramos
- Clinical Research Direction. Center of Molecular Immunology (CIM), Havana, Cuba
| |
Collapse
|
15
|
Sabanathan D, Lund ME, Campbell DH, Walsh BJ, Gurney H. Radioimmunotherapy for solid tumors: spotlight on Glypican-1 as a radioimmunotherapy target. Ther Adv Med Oncol 2021; 13:17588359211022918. [PMID: 34646364 PMCID: PMC8504276 DOI: 10.1177/17588359211022918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/17/2021] [Indexed: 12/24/2022] Open
Abstract
Radioimmunotherapy (i.e., the use of radiolabeled tumor targeting antibodies) is an emerging approach for the diagnosis, therapy, and monitoring of solid tumors. Often using paired agents, each targeting the same tumor molecule, but labelled with an imaging or therapeutic isotope, radioimmunotherapy has achieved promising clinical results in relatively radio-resistant solid tumors such as prostate. Several approaches to optimize therapeutic efficacy, such as dose fractionation and personalized dosimetry, have seen clinical success. The clinical use and optimization of a radioimmunotherapy approach is, in part, influenced by the targeted tumor antigen, several of which have been proposed for different solid tumors. Glypican-1 (GPC-1) is a heparan sulfate proteoglycan that is expressed in a variety of solid tumors, but whose expression is restricted in normal adult tissue. Here, we discuss the preclinical and clinical evidence for the potential of GPC-1 as a radioimmunotherapy target. We describe the current treatment paradigm for several solid tumors expressing GPC-1 and suggest the potential clinical utility of a GPC-1 directed radioimmunotherapy for these tumors.
Collapse
Affiliation(s)
- Dhanusha Sabanathan
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | | | | | | | - Howard Gurney
- Faculty of Medicine, Health and Human Sciences, Macquarie University, 2 Technology Place, Sydney, NSW 2109, Australia
| |
Collapse
|
16
|
Qiu B, Wang D, Li Q, Wu Y, Guo S, Jiang X, Fang J, Guo J, Liu F, Chu C, Wang B, Chen L, Zhang J, Liu Y, Hu Y, Liu H. Concurrent Chemoradiation Therapy With or Without Nimotuzumab in Locally Advanced Squamous Cell Lung Cancer: A Phase 2 Randomized Trial. Int J Radiat Oncol Biol Phys 2021; 111:917-925. [PMID: 34229051 DOI: 10.1016/j.ijrobp.2021.06.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 06/04/2021] [Accepted: 06/21/2021] [Indexed: 12/28/2022]
Abstract
PURPOSE The study aimed to evaluate the efficacy and safety of concurrent chemoradiation therapy (CCRT) combined with nimotuzumab in patients with unresectable stage III squamous cell lung cancer (SqCLC). METHODS AND MATERIALS A prospective, single-center, open-label, randomized phase 2 trial was performed in patients with unresectable stage III SqCLC. Patients were randomized to receive 65 Gy thoracic radiation over 5 weeks concurrent with docetaxel and cisplatin or the same CCRT regimen combined with 200 mg of nimotuzumab (NIMO-CCRT), administered weekly by intravenous infusion. The primary endpoint was overall survival. The secondary endpoints were progression-free survival, objective response rate, failure patterns, and treatment-related toxicity. RESULTS From August 2015 to June 2020, 126 patients with SqCLC were randomized. Four patients withdrew consent before the start of treatment, and 122 patients were included for analysis, including 57 in the NIMO-CCRT group and 65 in the CCRT group. The median OS was 24.9 months in the NIMO-CCRT group and 23.5 months in the CCRT group (P = .655). The median PFS was 12.1 months in the NIMO-CCRT group and 13.7 months in the CCRT group (P = .968). The NIMO-CCRT group had a significantly lower risk of brain metastasis, with adjusted subdistribution hazard ratio of 0.099 (95% confidence interval, 0.012-0.81; P = .031). The incidence of grade ≥3 pneumonitis (P = .894) and esophagitis (P = .974) was similar between the 2 arms. There was no grade 2 or higher skin toxicity in NIMO-CCRT group. CONCLUSIONS The coincident application of nimotuzumab with CCRT was well tolerated for locally advanced SCCL. The NIMO-CCRT group had an OS and PFS similar to that in the CCRT group, but a lower risk of brain metastasis. Further investigations are warranted.
Collapse
Affiliation(s)
- Bo Qiu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China; GuangDong Association Study of Thoracic Oncology, Guangzhou, Guangdong, China
| | - DaQuan 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, Guangzhou, Guangdong, China; GuangDong Association Study of Thoracic Oncology, Guangzhou, Guangdong, China
| | - QiWen Li
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China; GuangDong Association Study of Thoracic Oncology, Guangzhou, Guangdong, China
| | - YingJia 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, Guangzhou, Guangdong, China; GuangDong Association Study of Thoracic Oncology, Guangzhou, Guangdong, China
| | - SuPing Guo
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China; GuangDong Association Study of Thoracic Oncology, Guangzhou, Guangdong, China
| | - XiaoBo Jiang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - JianLan Fang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - JinYu Guo
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - FangJie Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China; GuangDong Association Study of Thoracic Oncology, Guangzhou, Guangdong, China
| | - Chu Chu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China; GuangDong Association Study of Thoracic Oncology, Guangzhou, Guangdong, China
| | - Bin 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, Guangzhou, Guangdong, China
| | - Li 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, Guangzhou, Guangdong, China
| | - Jun 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, Guangzhou, Guangdong, China
| | - YiMei Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - YongHong Hu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Hui Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China; GuangDong Association Study of Thoracic Oncology, Guangzhou, Guangdong, China.
| |
Collapse
|
17
|
Wang Y, Zhang M, Gong Y, Wu Q, Zhang L, Jiao S. Bioinformatic Analysis of Hepatocellular Carcinoma Cell Lines to the Efficacy of Nimotuzumab. Int J Gen Med 2021; 14:2611-2621. [PMID: 34168487 PMCID: PMC8217909 DOI: 10.2147/ijgm.s312770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/28/2021] [Indexed: 11/23/2022] Open
Abstract
Introduction Hepatocellular carcinoma (HCC) continues to be a cancer with rising incidence, high mortality, and recurrence rate. The therapeutic effects on HCC are not satisfactory currently. Epidermal growth factor receptor (EGFR) is an important factor, while anti-EGFR agencies have not shown ideal results in HCC. Materials and Methods We tested efficacy of nimotuzumab and EGFR expression on cell surface in six HCC cell lines (Hep 3B2.1–7, Li-7, PLC/PRF/5, SK-HEP-1, SNU-182, and SNU-387). Then, we analyzed RNA sequences of every cell line and performed a bioinformatic analysis. Differentially expressed genes (DEGs) were analyzed. The data, TCGA-LIHC from The Cancer Genome Atlas (TCGA) and GSE102079 from Gene Expression Omnibus (GEO), were used to analyse DEGs of Hoshida subclass. Results Hep 3B2.1–7 and PLC/PRF/5 were sensitive to nimotuzumab whereas Li-7, SK-HEP-1, SNU-182, and SNU-387 cell lines were resistant. Then, we compared the DEGs between sensitive and resistant group cell lines. We enriched DEGs in GO and KEGG and performed GSEA in each group. Genes in two groups did not show obvious different expressions in EGFR pathways, while Hoshida subclass of HCC seemed to associate with the efficacy of nimotuzumab in that S2 and S3 showed better therapeutic effect than S1. Therefore, we analyzed genes in human tumor samples which were from TCGA-LIHC and GSE102079. We found that COL1A1, COL1A2, COL3A1, and MMP9 were the focus DEGs of S1 and S2 & S3 related to EGFR. Conclusion The efficacy of nimotuzumab in HCC did not show direct relevance with EGFR protein expression and EGFR-related pathway. However, efficacy could associate with Hoshida subclass of HCC. Three ECM genes (COL1A1, COL1A2, COL3A1) and MMP9 were paid attention, as they might play important roles in the curative effect of nimotuzumab in HCC.
Collapse
Affiliation(s)
- Yu Wang
- Department of Oncology, Oncology Faculty, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Meng Zhang
- Department of Research and Development, Beijing DCTY® Biotech Co.,Ltd, Beijing, People's Republic of China.,Department of Hepatobiliary Surgery, PLA Rocket Force Characteristic Medical Center, Beijing, People's Republic of China
| | - Yixin Gong
- Department of Research and Development, Beijing DCTY® Biotech Co.,Ltd, Beijing, People's Republic of China
| | - Qiyan Wu
- Department of Oncology, Oncology Faculty, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Lijun Zhang
- Department of Oncology, Oncology Faculty, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Shunchang Jiao
- Department of Oncology, Oncology Faculty, Chinese PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|
18
|
Addition of Nimotuzumab to Standard TPF Regimen in Locally Advanced Head and Neck Cancer: A Single Institutional Study. JOURNAL OF ONCOLOGY 2021; 2021:6641963. [PMID: 33953747 PMCID: PMC8068547 DOI: 10.1155/2021/6641963] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/23/2021] [Accepted: 04/01/2021] [Indexed: 11/20/2022]
Abstract
Background Induction docetaxel, cisplatin, and 5-fluorouracil (TPF) chemotherapy followed by definitive concurrent chemoradiation remains the standard of care in locally advanced squamous cell carcinoma of head and neck cancers despite which the survival remains low. So, we analyzed the efficacy and adverse effect profile of the addition of nimotuzumab to standard TPF induction chemotherapy. Methods. We included 20 patients with locally advanced squamous cell carcinoma of the head and neck. Patients were administered with induction chemotherapy with nimotuzumab plus docetaxel, cisplatin, and 5-fluorouracil (TPF + N) followed by definitive concurrent chemoradiation with carboplatin. Treatment responses were assessed by PET-CT following induction chemotherapy and concurrent chemoradiation. Response rates, survival, and adverse effects data were tabulated and analyzed using the Kaplan Meier method. Results At a minimum follow-up of two years, the median progression-free survival (PFS) and median overall survival (OS) were 16 months and 38 months, respectively. PFS and OS were not reached (NR) in patients who showed a complete radiological response (CR). Median PFS and OS in patients who had partial response were 17.6 and 34.5 months, respectively. All subsites of primary including oral cavity, hypopharynx, and oropharynx showed similar response rates and survival. Overall the treatment was well tolerated with predominantly grade 1/2 toxicities. Conclusions Patients with locally advanced head and neck cancer could possibly have a better response and survival with nimotuzumab added to the standard TPF regimen. A complete response may serve as a good surrogate for survival irrespective of the primary site of head and neck cancer.
Collapse
|
19
|
Zhang L, Song Y, Jiang N, Huang Y, Dong B, Li W, He Y, Chen Y, Liu H, Yu R. Efficacy and safety of anti-epidermal growth factor receptor agents for the treatment of oesophageal cancer: a systematic review and meta-analysis. BMJ Open 2021; 11:e046352. [PMID: 33753446 PMCID: PMC7986677 DOI: 10.1136/bmjopen-2020-046352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVES Despite remarkable advances in the treatment of oesophageal cancer (OC), the role of antiepidermal growth factor receptor (anti-EGFR) agents in treating OC remains controversial. Herein, a systematic review and meta-analysis were conducted to elucidate the efficacy and safety of anti-EGFR agents in patients with OC. DESIGN Meta-analysis of randomised controlled trials (RCTs) identified by searching the PubMed, Embase, Web of Science, ClinicalTrials.gov, Cochrane Library, Chinese Biology Medicine, China National Knowledge Infrastructure and Wanfang Data Knowledge Service Platform databases from inception to December 2019. We followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. SETTING RCTs from any country and healthcare setting. PARTICIPANTS Patients with OC. INTERVENTIONS Combination therapy with anti-EGFR agents and conventional treatments versus conventional treatments alone in patients with OC. PRIMARY AND SECONDARY OUTCOME MEASURES Overall survival (OS) and progression-free survival (PFS) were primary outcome measures, and objective response rate (ORR), disease control rate (DCR) and treatment toxicities were secondary outcome measures. RESULTS In total, 25 RCTs comprising 3406 patients with OC were included. Overall, anti-EGFR treatment significantly improved the OS (HR: 0.81, 95% CI 0.74 to 0.89, p<0.00001), ORR (relative risk (RR): 1.33, 95% CI 1.16 to 1.52, p<0.0001) and DCR (RR: 1.22, 95% CI 1.11 to 1.34, p<0.0001) but not PFS (HR: 0.91, 95% CI 0.76 to 1.08, p=0.26). Anti-EGFR treatment was significantly associated with higher incidences of myelosuppression, diarrhoea, acne-like rash and hypomagnesaemia. CONCLUSIONS Overall, anti-EGFR agents have positive effects on OS, the ORR and DCR in OC. However, considering the high incidence of adverse effects, such as myelosuppression, diarrhoea, acne-like rashes and hypomagnesaemia, careful monitoring of patients with OC is recommended during anti-EGFR treatment. TRIAL REGISTRATION NUMBER CRD42020169230.
Collapse
Affiliation(s)
- Lijuan Zhang
- School of Nursing, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Yanli Song
- School of Nursing, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Nan Jiang
- School of Nursing, Tianjin Medical University, Tianjin, China
| | - Yaqi Huang
- School of Nursing, Tianjin Medical University, Tianjin, China
| | - Bo Dong
- School of Nursing, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Wei Li
- School of Nursing, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Yanze He
- School of Nursing, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Yun Chen
- Department of Colorectal Surgery, Liaoning Cancer Institute and Hospital, Shenyang, China
| | - Haibin Liu
- Department of Critical Care Medicine, Liaoning Cancer Institute and Hospital, Shenyang, China
| | - Rui Yu
- Department of Science and Technology, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
20
|
Galluzzi L, Vacchelli E, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zucman-Rossi J, Zitvogel L, Kroemer G. Trial Watch: Monoclonal antibodies in cancer therapy. Oncoimmunology 2021; 1:28-37. [PMID: 22720209 DOI: 10.4161/onci.1.1.17938] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Since the advent of hybridoma technology, dating back to 1975, monoclonal antibodies have become an irreplaceable diagnostic and therapeutic tool for a wide array of human diseases. During the last 15 years, several monoclonal antibodies (mAbs) have been approved by FDA for cancer therapy. These mAbs are designed to (1) activate the immune system against tumor cells, (2) inhibit cancer cell-intrinsic signaling pathways, (3) bring toxins in the close proximity of cancer cells, or (4) interfere with the tumor-stroma interaction. More recently, major efforts have been made for the development of immunostimulatory mAbs that either enhance cancer-directed immune responses or limit tumor- (or therapy-) driven immunosuppression. Some of these antibodies, which are thought to facilitate tumor eradication by initiating or sustaining a tumor-specific immune response, have already entered clinical trials. In this Trial Watch, we will review and discuss the clinical progress of the most important mAbs that are have entered clinical trials after January 2008.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- INSERM, U848; Villejuif, France ; Institut Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Phase 2 Study of Nimotuzumab in Combination With Concurrent Chemoradiotherapy in Patients With Locally Advanced Non-Small-Cell Lung Cancer. Clin Lung Cancer 2020; 22:134-141. [PMID: 33518480 DOI: 10.1016/j.cllc.2020.12.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 11/23/2022]
Abstract
BACKGROUND We evaluated the tolerability and efficacy of nimotuzumab, a humanized IgG1 monoclonal anti-epidermal growth factor receptor antibody, with concurrent chemoradiotherapy in patients with unresectable locally advanced non-small-cell lung cancer. PATIENTS AND METHODS In this multicenter, single-arm, open-label, phase 2 trial conducted in Japan (JapicCTI-090825), patients received thoracic radiotherapy (60 Gy, 2 Gy per fraction, 6 weeks) and four 4-week cycles of chemotherapy (day 1, cisplatin 80 mg/m2; days 1 and 8, vinorelbine 20 mg/m2). Nimotuzumab 200 mg was administrated weekly for 16 weeks. The primary endpoint was treatment completion rate, defined as the percentage of patients completing 60 Gy of radiotherapy within 8 weeks, 2 cycles of chemotherapy, and at least 75% of the required nimotuzumab dose during the initial 2-cycle concurrent chemoradiotherapy period. RESULTS Of 40 patients enrolled, 39 received the study treatment, which was well tolerated, with a completion rate of 87.2%. Thirty-eight patients completed 60 Gy of radiotherapy within 8 weeks. Infusion reaction, grade 3 or higher rash, grade 3 or higher radiation pneumonitis, or grade 4 or higher nonhematologic toxicity were not observed. The objective response rate was 69.2%. The median progression-free survival (PFS) and 5-year PFS rate were 508 days and 29.0%, respectively. The 5-year PFS rate in patients with non-squamous cell carcinoma (n = 23) was 13.7% and in patients with squamous cell carcinoma (n = 16) was 50.0%. The 5-year overall survival rate was 58.4%. CONCLUSION Addition of nimotuzumab to the concurrent chemoradiotherapy regimen was well tolerated and showed potential for treating patients with locally advanced non-small-cell lung cancer, particularly squamous cell carcinoma.
Collapse
|
22
|
Suman S, Priya R, Kameswaran M. Induction of different cellular arrest and molecular responses in low EGFR expressing A549 and high EGFR expressing A431 tumor cells treated with various doses of 177Lu-Nimotuzumab. Int J Radiat Biol 2020; 96:1144-1156. [PMID: 32657634 DOI: 10.1080/09553002.2020.1793012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Radioimmunotherapy (RIT) is a major anti-cancer therapy in cancer management multimodalities. 177Lu-Nimotuzumab has been in the use for radioimmunotherapy of EGFR expressing tumors. This study focuses on understanding the differential cellular and molecular mechanisms of anti-tumor effects of 177Lu-Nimotuzumab on low EGFR expressing A549 and high EGFR expressing A431 tumor cells. MATERIALS AND METHODS Nimotuzumab labeled with 177Lu was characterized by SE-HPLC. Specificity of 177Lu-Nimotuzumab to EGFR expressed on A549 and A431 cells was confirmed by competitive assay using increasing amounts of unlabeled Nimotuzumab. Cellular responses of A549 (low EGFR) and A431 (high EGFR) in response to different doses of 177Lu-Nimotuzumab were determined by Viable count assay for cellular viability, cell-cycle analysis by DNA staining, apoptotic assay for cell death, and CFSE dilution assay for cellular proliferation capacity. The number of DNA DSBs formed was determined using γ-H2AX assay with PI staining. Transcription of genes involved in DNA damage response and repair (DRR) pathways was monitored by RT-qPCR. RESULTS 177Lu-Nimotuzumab characterized by SE-HPLC exhibited a radiochemical purity of 99.1 ± 0.6%. Cell binding competition studies with 177Lu-Nimotuzumab showed specific binding of 34.3 ± 1.7% with A431 cells and 18.4 ± 1.9% with A549 cells which decreased when co-incubated with unlabeled Nimotuzumab. Cytotoxicity and DNA damage (DNA DSBs) increased with an increase in the dose of 177Lu-Nimotuzumab. A549 displayed G2/M arrest while A431 showed G1 arrest. Apoptotic death was determined to be one of the modes of death of arrested A549 and A431 cells which increases with the increase in the dose of 177Lu-Nimotuzumab. Loss of proliferation capacity was higher in A431 showed by CFSE staining at different doses of 177Lu-Nimotuzumab. Transcription profile of most DRR genes in A431 and A549 showed a decrease in the transcription at 4 h followed by recovery at 16 h post-treatment. The degree of transcription of most DRR genes was similar, irrespective of 177Lu-Nimotuzumab dose. CONCLUSION 177Lu-Nimotuzumab induces different cellular arrest and molecular responses in low EGFR expressing A549 and high EGFR expressing A431 tumor cells. This study would enable the development of integrative novel treatment strategies for radioimmunotherapy in low and high EGFR expressing tumors by 177Lu-Nimotuzumab with therapeutic gains.
Collapse
Affiliation(s)
- ShishuKant Suman
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Rashmi Priya
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Mythili Kameswaran
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| |
Collapse
|
23
|
Crombet Ramos T, Mestre Fernández B, Mazorra Herrera Z, Iznaga Escobar NE. Nimotuzumab for Patients With Inoperable Cancer of the Head and Neck. Front Oncol 2020; 10:817. [PMID: 32537431 PMCID: PMC7266975 DOI: 10.3389/fonc.2020.00817] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/27/2020] [Indexed: 12/18/2022] Open
Abstract
EGFR activation induces cell proliferation, neoformation of blood vessels, survival, and metastasis of the cancer cells. Nimotuzumab is an engineered, intermediate affinity anti-EGFR antibody, that apart from other drugs in its class, is very safe and does not cause hypomagnesemia or grade 3–4 cutaneous rash. The antibody inhibits cell proliferation and angiogenesis, activates natural killer cells, stimulates dendritic cell maturation, and induces cytotoxic T cells. Nimotuzumab restores MHC-I expression on tumor cells, hindering one of the EGFR immune-escape ways. The antibody has been extensively studied in 7 clinical trials, concurrently with irradiation or irradiation plus chemotherapy in subjects with inoperable head and neck tumors. Nimotuzumab was safe and efficacious in unfit patients receiving irradiation alone and in subjects treated with cisplatin and radiotherapy. In patients with locally advanced squamous cell carcinomas of the head and neck, nimotuzumab in combination with low dose cisplatin and radiotherapy was superior to cisplatin and radiotherapy in progression free survival, disease free survival, and locoregional tumor control.
Collapse
|
24
|
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
|
25
|
Bai SX, Zhang RR, Chen WH, Dong HM, Wang G, Li XK, Wang W. Clinical efficacy and safety of nimotuzumab plus chemotherapy in patients with advanced colorectal cancer: a retrospective analysis. J Int Med Res 2020; 48:300060519895858. [PMID: 31948326 PMCID: PMC7113702 DOI: 10.1177/0300060519895858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective To compare the clinical efficacy and safety of nimotuzumab combined with chemotherapy versus chemotherapy alone as first-line treatment for advanced colorectal cancer (ACRC). Method We retrospectively enrolled patients with ACRC treated with nimotuzumab plus chemotherapy (n = 40) or chemotherapy alone (n = 44). Responses were evaluated according to the Response Evaluation Criteria in Solid Tumors and adverse events according to the Common Terminology Criteria for Adverse Events 3.0. Results The objective overall response rate and disease control rate were higher in the combined-treatment group (55.0% vs 36.4% and 85.0% vs 75.0%, respectively), but the differences were not significant. There was no significant difference in median progression-free survival or median survival time between the combined-treatment and chemotherapy-alone groups (9.89 vs 7.86 months and 22.32 vs 18.10 months, respectively). There was no significant difference in adverse events between the two groups. Conclusion Nimotuzumab combined with chemotherapy had similar efficacy and safety to chemotherapy alone in patients with ACRC. The efficacy and safety of the combined treatment should be further studied in a randomized multicenter trial with a larger number of patients with ACRC.
Collapse
Affiliation(s)
- Sai-Xi Bai
- Department of Abdominal Oncology, Guizhou Cancer Hospital, Guiyang, China
| | - Ruo-Rong Zhang
- Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Wang-Hua Chen
- Department of Oncology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Hong-Min Dong
- Department of Oncology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Gang Wang
- Department of Oncology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xiao-Kai Li
- Department of Oncology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Wenling Wang
- Department of Oncology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
26
|
Zhi-Qiang W, Qi M, Ji-Bin L, Rui Y, You-Ping L, Rui S, Guang-Yuan H, Ming-Yuan C, Yi-Jun H. The long-term survival of patients with III-IVb stage nasopharyngeal carcinoma treated with IMRT with or without Nimotuzumab: a propensity score-matched analysis. BMC Cancer 2019; 19:1122. [PMID: 31744469 PMCID: PMC6862826 DOI: 10.1186/s12885-019-6156-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 09/13/2019] [Indexed: 11/10/2022] Open
Abstract
Background To assess the efficacy of Nimotuzumab in combination with first-line chemoradiotherapy treatment in Chinese patients with primary III-IVb stage nasopharyngeal carcinoma. Methods Patients with primary locoregionally advanced nasopharyngeal carcinoma who were treated with intensity-modulated radiotherapy (IMRT) and concurrent cisplatin-based chemotherapy between January 2008 and December 2013 at a single institution were retrospectively reviewed. Group A received at least 6 doses of Nimotuzumab, while Group B did not receive Nimotuzumab. A propensity score matching method was used to match patients from each group in a 1:3 ratio. Results In total, 730 eligible patients were propensity matched, with 184 patients in Group A and 546 patients in Group B. Significant differences were not observed in the patient and tumor characteristics between Group A and Group B. At a median follow-up of 74.78 months (range 3.53–117.83 months), locoregional recurrence, distant failure and death were observed in 10.68, 11.10 and 16.03% of all patients, respectively. The estimated 5-year locoregional relapse–free survival, distant metastasis–free survival, progression-free survival and overall survival in the Group A versus Group B were 85.34% versus 89.79% (P = 0.156), 93.09% versus 85.61% (P = 0.012), 79.96% versus 77.99% (P = 0.117) and 88.91% versus 78.30% (P = 0.006), respectively. Conclusions This nimotuzumab-containing regimen resulted in improved long-term survival of III-IVb stage NPC patients and warrants further prospective evaluation.
Collapse
Affiliation(s)
- Wang Zhi-Qiang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Mei Qi
- Department of Oncology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Techology, Wuhan, China
| | - Li Ji-Bin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Clinical Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - You Rui
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Liu You-Ping
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Sun Rui
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Hu Guang-Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Techology, Wuhan, China
| | - Chen Ming-Yuan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China. .,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.
| | - Hua Yi-Jun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China. .,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.
| |
Collapse
|
27
|
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
|
28
|
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
|
29
|
Kareemaghay S, Tavassoli M. Clinical immunotherapeutic approaches for the treatment of head and neck cancer. Int J Oral Maxillofac Surg 2018; 48:419-436. [PMID: 30401512 DOI: 10.1016/j.ijom.2018.10.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 09/08/2018] [Accepted: 10/19/2018] [Indexed: 12/12/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common malignancy worldwide, accounting for more than 550,000 cases and 380,000 deaths annually. The primary risk factors associated with HNSCC are tobacco use and alcohol consumption; nevertheless genetic predisposition and oncogenic viruses also play important roles in the development of these malignancies. The current treatments for HNSCC patients include surgery, chemotherapy, radiotherapy, and cetuximab, and combinations of these. However, these treatments are associated with significant toxicity, and many patients are either refractory to the treatment or relapse after a short period. Despite improvements in the treatment of patients with HNSCC, the clinical outcomes of those who have been treated with standard therapies have remained unchanged for over three decades and the 5-year overall survival rate in these patients remains around 40-50%. Therefore, more specific and less toxic therapies are needed in order to improve patient outcomes. The tumour microenvironment of HNSCC is immunosuppressive; therefore immunotherapy strategies that can overcome the immunosuppressive environment and produce long-term tumour immunosurveillance will have a significant therapeutic impact in these patients. This review focuses on the current immunological treatment options under investigation or available for clinical use in patients with HNSCC.
Collapse
Affiliation(s)
- S Kareemaghay
- Department of Molecular Oncology, King's College London, London, UK
| | - M Tavassoli
- Department of Molecular Oncology, King's College London, London, UK.
| |
Collapse
|
30
|
Si X, Wu S, Wang H, Zhang X, Wang M, Zeng X, Zhang L. Nimotuzumab combined with chemotherapy as first-line treatment for advanced lung squamous cell carcinoma. Thorac Cancer 2018; 9:1056-1061. [PMID: 29920955 PMCID: PMC6068447 DOI: 10.1111/1759-7714.12789] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 05/12/2018] [Accepted: 05/23/2018] [Indexed: 12/01/2022] Open
Abstract
Background This study was conducted to evaluate the efficacy and safety of nimotuzumab combined with chemotherapy as first‐line therapy in advanced lung squamous cell carcinoma (LSCC), and to explore predictive biomarkers of the efficacy of nimotuzumab. Methods A retrospective study was conducted of patients with advanced LSCC administered nimotuzumab combined with chemotherapy as first‐line therapy from June 2012 to December 2016 at the Department of Respiratory Medicine, Peking Union Medical College Hospital. The associations between EGFR expression, EGFR gene copy numbers, and clinical efficacy were detected by immunohistochemistry and fluorescence in situ hybridization (FISH). Results Twenty‐six patients were enrolled, including 22 men and 4 women. The objective response rate was 50% and the disease control rate was 100%. The median progression‐free survival (PFS) and overall survival were 6.7 and 16.3 months, respectively. Patients whose samples were tested via FISH and showed positive EGFR expression had a trend of longer median PFS (10.0 months; P = 0.10). Adverse effects included 15 cases (57.7%) of bone marrow suppression, 15 (57.7%) of sensory neuropathy, 14 (53.8%) of alopecia, nine (34.6%) of nausea/vomiting and one case (3.8%) of elevated creatinine level. All adverse effects were attributed to chemotherapy. Conclusion Nimotuzumab combined with chemotherapy might be a possible option as first‐line therapy in patients with advanced LSCC. EGFR gene copy number examined by FISH might be a possible predictive biomarker.
Collapse
Affiliation(s)
- Xiaoyan Si
- Department of Respiratory Diseases, Peking Union Medical College Hospital, Beijing, China
| | - Shafei Wu
- Department of Pathology, Peking Union Medical College Hospital, Beijing, China
| | - Hanping Wang
- Department of Respiratory Diseases, Peking Union Medical College Hospital, Beijing, China
| | - Xiaotong Zhang
- Department of Respiratory Diseases, Peking Union Medical College Hospital, Beijing, China
| | - Mengzhao Wang
- Department of Respiratory Diseases, Peking Union Medical College Hospital, Beijing, China
| | - Xuan Zeng
- Department of Pathology, Peking Union Medical College Hospital, Beijing, China
| | - Li Zhang
- Department of Respiratory Diseases, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
31
|
Schultheis B, Reuter D, Ebert MP, Siveke J, Kerkhoff A, Berdel WE, Hofheinz R, Behringer DM, Schmidt WE, Goker E, De Dosso S, Kneba M, Yalcin S, Overkamp F, Schlegel F, Dommach M, Rohrberg R, Steinmetz T, Bulitta M, Strumberg D. Gemcitabine combined with the monoclonal antibody nimotuzumab is an active first-line regimen in KRAS wildtype patients with locally advanced or metastatic pancreatic cancer: a multicenter, randomized phase IIb study. Ann Oncol 2018; 28:2429-2435. [PMID: 28961832 DOI: 10.1093/annonc/mdx343] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background This randomized study was designed to investigate the superiority of gemcitabine (gem) plus nimotuzumab (nimo), an anti-epidermal growth factor receptor monoclonal antibody, compared with gem plus placebo as first-line therapy in patients with advanced pancreatic cancer. Patients and methods Patients with previously untreated, unresectable, locally advanced or metastatic pancreatic cancer were randomly assigned to receive gem: 1000 mg/m2, 30-min i.v. once weekly (d1, 8, 15; q29) and nimo: fixed dose of 400 mg once weekly as a 30-min infusion, or gem plus placebo, until progression or unacceptable toxicity. The primary end point was overall survival (OS), secondary end points included time to progression, overall response rate, safety and quality of life. Results A total of 192 patients were randomized, with 186 of them being assessable for efficacy and safety (average age 63.6 years). One-year OS/progression-free survival (PFS) was 34%/22% for gem plus nimo compared with 19%/10% for gem plus placebo (HR = 0.69; P = 0.03/HR = 0.68; P = 0.02). Median OS/PFS was 8.6/5.1 months for gem plus nimo versus 6.0/3.4 mo in the gem plus placebo group (HR = 0.69; P = 0.0341/HR = 0.68; P = 0.0163), with very few grade 3/4 toxicities. KRAS wildtype patients experienced a significantly better OS than those with KRAS mutations (11.6 versus 5.6 months, P = 0.03). Conclusion This randomized study showed that nimo in combination with gem is safe and well tolerated. The 1-year OS and PFS rates for the entire population were significantly improved. Especially, those patients with KRAS wildtype seem to benefit. The study was registered as protocol ID OSAG101-PCS07, NCT00561990 and EudraCT 2007-000338-38.
Collapse
Affiliation(s)
- B Schultheis
- Department of Hematology/Oncology, University Bochum, Marien Hospital Herne, Herne;.
| | - D Reuter
- Oncoscience AG, Wedel (recently Schenefeld)
| | - M P Ebert
- Klinikum Rechts der Isar TU München, München
| | - J Siveke
- Klinikum Rechts der Isar TU München, München
| | | | | | - R Hofheinz
- Department of Hematology and Medical Oncology, University Medical Center Mannheim, Mannheim
| | | | - W E Schmidt
- St. Josef Hospital, Med. Klinik I, Bochum, Germany
| | - E Goker
- Ege University Medical School, Izmir, Turkey
| | - S De Dosso
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - M Kneba
- Department of Medicine, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - S Yalcin
- Hacettepe University Hospital, Ankara, Turkey
| | - F Overkamp
- Medical Practice for Oncology and Hematology, Recklinghausen
| | | | - M Dommach
- Sana-Kliniken, Medizinisches Versorgungszentrum Onkologie, Düsseldorf
| | - R Rohrberg
- Gemeinschaftspraxis und Tagesklinik fuer Haematologie, Onkologie und Gastroenterologie, Halle
| | - T Steinmetz
- Group Practice Hematology/Oncology Cologne, Cologne
| | - M Bulitta
- CRM Biometrics GmbH, Rheinbach, Germany
| | - D Strumberg
- Department of Hematology/Oncology, University Bochum, Marien Hospital Herne, Herne
| |
Collapse
|
32
|
Kato K, Ura T, Koizumi W, Iwasa S, Katada C, Azuma M, Ishikura S, Nakao Y, Onuma H, Muro K. Nimotuzumab combined with concurrent chemoradiotherapy in Japanese patients with esophageal cancer: A phase I study. Cancer Sci 2018; 109:785-793. [PMID: 29285832 PMCID: PMC5834813 DOI: 10.1111/cas.13481] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 12/19/2017] [Accepted: 12/24/2017] [Indexed: 12/14/2022] Open
Abstract
Nimotuzumab is a humanized anti‐epidermal growth factor receptor IgG1 monoclonal antibody. This phase I study assessed the tolerability, safety, efficacy, and pharmacokinetics of nimotuzumab in combination with chemoradiotherapy in Japanese patients with esophageal cancer. Patients with stage II, III, and IV esophageal cancer were enrolled. Patients were planned to receive nimotuzumab (level 1: 200 mg/wk for 25 weeks; or level 2: 400 mg/wk in the chemoradiation period, 400 mg biweekly in an additional chemotherapy period [8 weeks after the chemoradiation period] and a maintenance therapy period [after chemotherapy to 25 weeks]) combined with cisplatin (75 mg/m2 on day 1) and fluorouracil (1000 mg/m2 on days 1‐4) in the chemoradiation and additional chemotherapy periods. Radiotherapy was given concurrently at 50.4 Gy. A total of 10 patients were enrolled in level 1. Dose‐limiting toxicities were observed in 2 patients (grade 3 infection and renal disorder). Maximum‐tolerated dose was estimated to be at least 200 mg/wk and the dose was not escalated to level 2. The most common grade ≥3 toxicities were lymphopenia (90%), leukopenia (60%), neutropenia (50%), and febrile neutropenia, decreased appetite, hyponatremia, and radiation esophagitis (30% each). Neither treatment‐related death nor grade ≥3 skin toxicity was observed in any patient. Complete response rate was 50%. Progression‐free survival was 13.9 months. One‐ and 3‐year survival rates were 75% and 37.5%, respectively. Immunogenicity was not reported in any patient. Nimotuzumab in combination with concurrent chemoradiotherapy was tolerable and effective for Japanese patients with esophageal cancer.
Collapse
Affiliation(s)
- Ken Kato
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Takashi Ura
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Wasaburo Koizumi
- Department of Gastroenterology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Satoru Iwasa
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Chikatoshi Katada
- Department of Gastroenterology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Mizutomo Azuma
- Department of Gastroenterology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Satoshi Ishikura
- Department of Radiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yoshinori Nakao
- Pharmacovigilance Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Hiroshi Onuma
- Oncology Clinical Development Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| |
Collapse
|
33
|
Toffoli G, De Mattia E, Cecchin E, Biason P, Masier S, Corona G. Pharmacology of Epidermal Growth Factor Inhibitors. Int J Biol Markers 2018; 22:24-39. [DOI: 10.1177/17246008070221s404] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Research into the molecular bases of malignant diseases has yielded the development of many novel agents with potential antitumor activity. Evidence for a causative role for the epidermal growth factor receptor (EGFR), which is now regarded as an excellent target for cancer chemotherapy in human cancer, leads to the development of EGFR inhibitors. Two classes of anti-EGFR agents are currently in clinical use: monoclonal antibodies directed at the extracellular domain of the receptor, and the low-molecular-weight receptor tyrosine kinase inhibitors acting intracellularly by competing with adenosine triphosphate for binding to the tyrosine kinase portion of the EGFR. The effect on the receptor interferes with key biological functions including cell cycle arrest, potentiation of apoptosis, inhibition of angiogenesis and cell invasion and metastasis. Cetuximab, a monoclonal antibody, and the receptor tyrosine kinase inhibitors gefitinib and erlotinib are currently approved for the treatment of patients with cancer. New agents with clinical activity are entering the clinic, and new combinatorial approaches are being explored with the aim of improving the potency and pharmacokinetics of EGFR inhibition, to increase the synergistic activity in combination with chemotherapy and overcome resistance to the EGFR inhibitors.
Collapse
Affiliation(s)
- G. Toffoli
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, Aviano, Pordenone - Italy
| | - E. De Mattia
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, Aviano, Pordenone - Italy
| | - E. Cecchin
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, Aviano, Pordenone - Italy
| | - P. Biason
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, Aviano, Pordenone - Italy
| | - S. Masier
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, Aviano, Pordenone - Italy
| | - G. Corona
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, Aviano, Pordenone - Italy
| |
Collapse
|
34
|
de Castro Junior G, Segalla JG, de Azevedo SJ, Andrade CJ, Grabarz D, de Araújo Lima França B, Del Giglio A, Lazaretti NS, Álvares MN, Pedrini JL, Kussumoto C, de Matos Neto JN, Forones NM, Fernandes Júnior HJ, Borges G, Girotto G, da Silva IDCG, Maluf-Filho F, Skare NG. A randomised phase II study of chemoradiotherapy with or without nimotuzumab in locally advanced oesophageal cancer: NICE trial. Eur J Cancer 2018; 88:21-30. [DOI: 10.1016/j.ejca.2017.10.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 09/25/2017] [Accepted: 10/02/2017] [Indexed: 01/25/2023]
|
35
|
Mazorra Z, Chao L, Lavastida A, Sanchez B, Ramos M, Iznaga N, Crombet T. Nimotuzumab: beyond the EGFR signaling cascade inhibition. Semin Oncol 2018; 45:18-26. [DOI: 10.1053/j.seminoncol.2018.04.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 04/20/2018] [Indexed: 12/11/2022]
|
36
|
Garrido G, Rabasa A, Garrido C, Chao L, Garrido F, García-Lora ÁM, Sánchez-Ramírez B. Upregulation of HLA Class I Expression on Tumor Cells by the Anti-EGFR Antibody Nimotuzumab. Front Pharmacol 2017; 8:595. [PMID: 29056908 PMCID: PMC5635422 DOI: 10.3389/fphar.2017.00595] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/17/2017] [Indexed: 12/21/2022] Open
Abstract
Defining how epidermal growth factor receptor (EGFR)-targeting therapies influence the immune response is essential to increase their clinical efficacy. A growing emphasis is being placed on immune regulator genes that govern tumor – T cell interactions. Previous studies showed an increase in HLA class I cell surface expression in tumor cell lines treated with anti-EGFR agents. In particular, earlier studies of the anti-EGFR blocking antibody cetuximab, have suggested that increased tumor expression of HLA class I is associated with positive clinical response. We investigated the effect of another commercially available anti-EGFR antibody nimotuzumab on HLA class I expression in tumor cell lines. We observed, for the first time, that nimotuzumab increases HLA class I expression and its effect is associated with a coordinated increase in mRNA levels of the principal antigen processing and presentation components. Moreover, using 7A7 (a specific surrogate antibody against murine EGFR), we obtained results suggesting the importance of the increased MHC-I expression induced by EGFR-targeted therapies display higher in antitumor immune response. 7A7 therapy induced upregulation of tumor MHC-I expression in vivo and tumors treated with this antibody display higher susceptibility to CD8+ T cells-mediated lysis. Our results represent the first evidence suggesting the importance of the adaptive immunity in nimotuzumab-mediated antitumor activity. More experiments should be conducted in order to elucidate the relevance of this mechanism in cancer patients. This novel immune-related antitumor mechanism mediated by nimotuzumab opens new perspectives for its combination with various immunotherapeutic agents and cancer vaccines.
Collapse
Affiliation(s)
- Greta Garrido
- Tumor Immunology Direction, Molecular Immunology Institute, Center of Molecular Immunology, Havana, Cuba
| | - Ailem Rabasa
- Tumor Immunology Direction, Molecular Immunology Institute, Center of Molecular Immunology, Havana, Cuba
| | - Cristina Garrido
- Servicio de Análisis Clínicos e Inmunología, Hospital Universitario Virgen de las Nieves, Granada, Spain.,Departamento de Bioquímica, Biología Molecular e Inmunología III, Universidad de Granada, Granada, Spain
| | - Lisset Chao
- Tumor Immunology Direction, Molecular Immunology Institute, Center of Molecular Immunology, Havana, Cuba
| | - Federico Garrido
- Servicio de Análisis Clínicos e Inmunología, Hospital Universitario Virgen de las Nieves, Granada, Spain.,Departamento de Bioquímica, Biología Molecular e Inmunología III, Universidad de Granada, Granada, Spain
| | - Ángel M García-Lora
- Servicio de Análisis Clínicos e Inmunología, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Belinda Sánchez-Ramírez
- Tumor Immunology Direction, Molecular Immunology Institute, Center of Molecular Immunology, Havana, Cuba
| |
Collapse
|
37
|
A randomized, phase II study of gefitinib alone versus nimotuzumab plus gefitinib after platinum-based chemotherapy in advanced non-small cell lung cancer (KCSG LU12-01). Oncotarget 2017; 8:15943-15951. [PMID: 27823977 PMCID: PMC5362536 DOI: 10.18632/oncotarget.13056] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 10/26/2016] [Indexed: 12/11/2022] Open
Abstract
We aimed to evaluate the efficacy of dual inhibition of epidermal growth factor receptor (EGFR) with nimotuzumab (EGFR monoclonal antibody) plus gefitinib (EGFR-tyrosine kinase inhibitor) in advanced non-small cell lung cancer (NSCLC) after platinum-based chemotherapy. An open label, randomized, phase II trial was conducted at 6 centers; 160 patients were randomized (1:1) to either gefitinib alone or nimotuzumab (200 mg, i.v. weekly) plus gefitinib (250 mg p.o. daily) until disease progression or intolerable toxicity. The primary endpoint was progression-free survival (PFS) at 3 months. Of the total 160 enrolled patients, 155 (77: gefitinib, 78: nimotuzumab plus gefitinib) received at least one dose and could be evaluated for efficacy and toxicity. The majority had adenocarcinoma (65.2%) and ECOG performance status of 0 to 1 (83.5%). The median follow-up was 22.1 months, and the PFS rate at 3 months was 48.1% in gefitinib and 37.2% in nimotuzumab plus gefitinib (P = not significant, NS). The median PFS and OS were 2.8 and 13.2 months in gefitinib and 2.0 and 14.0 months in nimotuzumab plus gefitinib. Combined treatment was not associated with superior PFS to gefitinib alone in patients with EGFR mutation (13.5 vs. 10.2 months in gefitinib alone, P=NS) or those with wild-type EGFR (0.9 vs. 2.0 months in gefitinib alone, P=NS). Combined treatment did not increase EGFR inhibition-related adverse events with manageable toxicities. The dual inhibition of EGFR with nimotuzumab plus gefitinib was not associated with better outcomes than gefitinib alone as a second-line treatment of advanced NSCLC (NCT01498562).
Collapse
|
38
|
Mazorra Z, Lavastida A, Concha-Benavente F, Valdés A, Srivastava RM, García-Bates TM, Hechavarría E, González Z, González A, Lugiollo M, Cuevas I, Frómeta C, Mestre BF, Barroso MC, Crombet T, Ferris RL. Nimotuzumab Induces NK Cell Activation, Cytotoxicity, Dendritic Cell Maturation and Expansion of EGFR-Specific T Cells in Head and Neck Cancer Patients. Front Pharmacol 2017; 8:382. [PMID: 28674498 PMCID: PMC5474456 DOI: 10.3389/fphar.2017.00382] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/31/2017] [Indexed: 12/19/2022] Open
Abstract
Survival benefit and long-term duration of clinical response have been seen using the epidermal growth factor receptor (EGFR)-targeted monoclonal antibody (mAb) nimotuzumab. Blocking EGFR signaling may not be the only mechanism of action underlying its efficacy. As an IgG1 isotype mAb, nimotuzumab's capacity of killing tumor cells by antibody dependent cellular cytotoxicity (ADCC) and to induce an immune response in cancer patients have not been studied. ADCC-induced by nimotuzumab was determined using a 51Cr release assay. The in vitro effect of nimotuzumab on natural killer (NK) cell activation and dendritic cell (DC) maturation and the in vivo frequency of circulating regulatory T cells (Tregs) and NK cells were assessed by flow cytometry. Cytokine levels in supernatants were determined by ELISA. ELISpot was carried out to quantify EGFR-specific T cells in nimotuzumab-treated head and neck cancer (HNSCC) patients. Nimotuzumab was able to kill EGFR+ tumor cells by NK cell-mediated ADCC. Nimotuzumab-activated NK cells promoted DC maturation and EGFR-specific CD8+ T cell priming. Interestingly, nimotuzumab led to upregulation of some immune checkpoint molecules on NK cells (TIM-3) and DC (PD-L1), to a lower extent than another EGFR mAb, cetuximab. Furthermore, circulating EGFR-specific T cells were identified in nimotuzumab-treated HNSCC patients. Notably, nimotuzumab combined with cisplatin-based chemotherapy and radiation increased the frequency of peripheral CD4+CD39+FOXP3+Tregs which otherwise were decreased to baseline values when nimotuzumab was used as monotherapy. The frequency of circulating NK cells remained constant during treatment. Nimotuzumab-induced, NK cell-mediated DC priming led to induction of anti-EGFR specific T cells in HNSCC patients. The association between EGFR-specific T cells and patient clinical benefit with nimotuzumab treatment should be investigated.
Collapse
Affiliation(s)
- Zaima Mazorra
- Department of Clinical Immunology, Clinical Direction, Center of Molecular ImmunologyHavana, Cuba
| | - Anabel Lavastida
- Department of Clinical Immunology, Clinical Direction, Center of Molecular ImmunologyHavana, Cuba
| | | | - Anet Valdés
- Department of Clinical Immunology, Clinical Direction, Center of Molecular ImmunologyHavana, Cuba
| | | | - Tatiana M García-Bates
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, PittsburghPA, United States
| | - Esperanza Hechavarría
- Department of Clinical Immunology, Clinical Direction, Center of Molecular ImmunologyHavana, Cuba
| | - Zuyen González
- Department of Clinical Immunology, Clinical Direction, Center of Molecular ImmunologyHavana, Cuba
| | - Amnely González
- Department of Clinical Immunology, Clinical Direction, Center of Molecular ImmunologyHavana, Cuba
| | | | - Iván Cuevas
- National Institute of Oncology and RadiobiologyHavana, Cuba
| | - Carlos Frómeta
- National Institute of Oncology and RadiobiologyHavana, Cuba
| | | | - Maria C Barroso
- Clinical Direction, Center of Molecular ImmunologyHavana, Cuba
| | - Tania Crombet
- Clinical Direction, Center of Molecular ImmunologyHavana, Cuba
| | - Robert L Ferris
- Department of Immunology, University of Pittsburgh, PittsburghPA, United States.,Department of Otolaryngology, University of Pittsburgh, PittsburghPA, United States.,Cancer Immunology Program, University of Pittsburgh Cancer Institute, PittsburghPA, United States
| |
Collapse
|
39
|
Schmitt LC, Rau A, Seifert O, Honer J, Hutt M, Schmid S, Zantow J, Hust M, Dübel S, Olayioye MA, Kontermann RE. Inhibition of HER3 activation and tumor growth with a human antibody binding to a conserved epitope formed by domain III and IV. MAbs 2017; 9:831-843. [PMID: 28421882 DOI: 10.1080/19420862.2017.1319023] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human epidermal growth factor receptor 3 (HER3, also known as ErbB3) has emerged as relevant target for antibody-mediated tumor therapy. Here, we describe a novel human antibody, IgG 3-43, recognizing a unique epitope formed by domain III and parts of domain IV of the extracellular region of HER3, conserved between HER3 and mouse ErbB3. An affinity of 11 nM was determined for the monovalent interaction. In the IgG format, the antibody bound recombinant bivalent HER3 with subnanomolar affinity (KD = 220 pM) and HER3-expressing tumor cells with EC50 values in the low picomolar range (27 - 83 pM). The antibody competed with binding of heregulin to HER3-expressing cells, efficiently inhibited phosphorylation of HER3 as well as downstream signaling, and induced receptor internalization and degradation. Furthermore, IgG 3-43 inhibited heregulin-dependent proliferation of several HER3-positive cancer cell lines and heregulin-independent colony formation of HER2-overexpressing tumor cell lines. Importantly, inhibition of tumor growth and prolonged survival was demonstrated in a FaDu xenograft tumor model in SCID mice. These findings demonstrate that by binding to the membrane-proximal domains III and IV involved in ligand binding and receptor dimerization, IgG 3-43 efficiently inhibits activation of HER3, thereby blocking tumor cell growth both in vitro and in vivo.
Collapse
Affiliation(s)
- Lisa C Schmitt
- a Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart , Germany
| | - Alexander Rau
- a Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart , Germany
| | - Oliver Seifert
- a Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart , Germany
| | - Jonas Honer
- a Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart , Germany
| | - Meike Hutt
- a Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart , Germany
| | - Simone Schmid
- a Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart , Germany
| | - Jonas Zantow
- b Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig , Braunschweig , Germany
| | - Michael Hust
- b Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig , Braunschweig , Germany
| | - Stefan Dübel
- b Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig , Braunschweig , Germany
| | - Monilola A Olayioye
- a Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart , Germany.,c Stuttgart Research Center Systems Biology, University of Stuttgart , Stuttgart , Germany
| | - Roland E Kontermann
- a Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart , Germany.,c Stuttgart Research Center Systems Biology, University of Stuttgart , Stuttgart , Germany
| |
Collapse
|
40
|
Stengl A, Hörl D, Leonhardt H, Helma J. A Simple and Sensitive High-Content Assay for the Characterization of Antiproliferative Therapeutic Antibodies. SLAS DISCOVERY 2016; 22:309-315. [PMID: 27909235 PMCID: PMC5322830 DOI: 10.1177/1087057116677821] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Monoclonal antibodies (mAbs) have become a central class of therapeutic agents in particular as antiproliferative compounds. Their often complex modes of action require sensitive assays during early, functional characterization. Current cell-based proliferation assays often detect metabolites that are indicative of metabolic activity but do not directly account for cell proliferation. Measuring DNA replication by incorporation of base analogues such as 5-bromo-2'-deoxyuridine (BrdU) fills this analytical gap but was previously restricted to bulk effect characterization in enzyme-linked immunosorbent assay formats. Here, we describe a cell-based assay format for the characterization of antiproliferative mAbs regarding potency and mode of action in a single experiment. The assay makes use of single cell-based high-content-analysis (HCA) for the reliable quantification of replicating cells and DNA content via 5-ethynyl-2'-deoxyuridine (EdU) and 4',6-diamidino-2-phenylindole (DAPI), respectively, as sensitive measures of antiproliferative mAb activity. We used trastuzumab, an antiproliferative therapeutic antibody interfering with HER2 cell surface receptor-mediated growth signal transduction, and HER2-overexpressing cell lines BT474 and SKBR3 to demonstrate up to 10-fold signal-to-background (S/B) ratios for treated versus untreated cells and a shift in cell cycle profiles indicating antibody-induced cell cycle arrest. The assay is simple, cost-effective, and sensitive, providing a cell-based format for preclinical characterization of therapeutic mAbs.
Collapse
Affiliation(s)
- Andreas Stengl
- 1 Department of Biology II, LMU Munich, Planegg-Martinsried, Germany
| | - David Hörl
- 1 Department of Biology II, LMU Munich, Planegg-Martinsried, Germany
| | | | - Jonas Helma
- 1 Department of Biology II, LMU Munich, Planegg-Martinsried, Germany
| |
Collapse
|
41
|
Lazăr DC, Tăban S, Cornianu M, Faur A, Goldiş A. New advances in targeted gastric cancer treatment. World J Gastroenterol 2016; 22:6776-6799. [PMID: 27570417 PMCID: PMC4974579 DOI: 10.3748/wjg.v22.i30.6776] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/13/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023] Open
Abstract
Despite a decrease in incidence over past decades, gastric cancer remains a major global health problem. In the more recent period, survival has shown only minor improvement, despite significant advances in diagnostic techniques, surgical and chemotherapeutic approaches, the development of novel therapeutic agents and treatment by multidisciplinary teams. Because multiple genetic mutations, epigenetic alterations, and aberrant molecular signalling pathways are involved in the development of gastric cancers, recent research has attempted to determine the molecular heterogeneity responsible for the processes of carcinogenesis, spread and metastasis. Currently, some novel agents targeting a part of these dysfunctional molecular signalling pathways have already been integrated into the standard treatment of gastric cancer, whereas others remain in phases of investigation within clinical trials. It is essential to identify the unique molecular patterns of tumours and specific biomarkers to develop treatments targeted to the individual tumour behaviour. This review analyses the global impact of gastric cancer, as well as the role of Helicobacter pylori infection and the efficacy of bacterial eradication in preventing gastric cancer development. Furthermore, the paper discusses the currently available targeted treatments and future directions of research using promising novel classes of molecular agents for advanced tumours.
Collapse
|
42
|
Frequent co-expression of EGFR and NeuGcGM3 ganglioside in cancer: it's potential therapeutic implications. Clin Exp Metastasis 2016; 33:717-25. [PMID: 27449755 PMCID: PMC5035325 DOI: 10.1007/s10585-016-9811-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 07/11/2016] [Indexed: 12/25/2022]
Abstract
Interaction between epidermal growth factor receptor (EGFR) signaling with GM3 ganglioside expression has been previously described. However, little is known about EGFR and NeuGcGM3 co-expression in cancer patients and their therapeutic implications. In this paper, we evaluate the co-expression of EGFR and NeuGcGM3 ganglioside in tumors from 92 patients and in two spontaneous lung metastasis models of mice (Lewis lung carcinoma (3LL-D122) in C57BL/6 and mammary carcinoma (4T1) in BALB/c). As results, co-expression of EGFR and NeuGcGM3 ganglioside was frequently observed in 63 of 92 patients (68 %), independently of histological subtype. Moreover, EGFR is co-expressed with NeuGcGM3 ganglioside in the metastasis of 3LL-D122 and 4T1 murine models. Such dual expression appears to be therapeutically relevant, since combined therapy with mAbs against these two molecules synergistically increase the survival of mice treated. Overall, our results suggest that NeuGcGM3 and EGFR may coordinately contribute to the tumor cell biology and that therapeutic combinations against these two targets might be a valid strategy to explore.
Collapse
|
43
|
Xu S, Ramos-Suzarte M, Bai X, Xu B. Treatment outcome of nimotuzumab plus chemotherapy in advanced cancer patients: a single institute experience. Oncotarget 2016; 7:33391-407. [PMID: 27050148 PMCID: PMC5078104 DOI: 10.18632/oncotarget.8516] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/02/2016] [Indexed: 12/18/2022] Open
Abstract
Nimotuzumab is a humanized anti-EGFR IgG1 monoclonal antibody and demonstrates a better safety profile than other anti-EGFR antibodies due to its intermediate affinity. Since it was approved in China for the treatment of nasopharyngeal cancer (NPC), it has been widely used in NPC and in many clinical trials for other cancer types. However, the optimal dose and administration frequency of nimotuzumab that should be used and which kind of cancer patients will be more benefited from nimotuzumab is still unknown. In this retrospective study, 205 advanced cancer patients with colorectal cancer, esophageal cancer, head and neck cancer, gastric cancer, non-small cell lung cancer, or other cancers from mainland China, treated with nimotuzumab in combination with chemotherapy, were enrolled. Over 60% of these patients received nimotuzumab > 6 doses and ≥ 400 mg/week as maintenance therapy. It was well tolerated in real-life patients. This report demonstrates that age, sex and previous treatment might be potential predictive factors for survival, and patients received nimotuzumab > 6 doses and > 200 mg/week might benefit more from nimotuzumab therapy. Using these factors for stratification analysis may form a predictive differential clinical strategy for nimotuzumab to maximize the benefit in patients with different epithelial tumors.
Collapse
Affiliation(s)
- Shuping Xu
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
- Department of Medical Affairs, Biotech Pharmaceuticals Co., Ltd., P. R. China
| | - Mayra Ramos-Suzarte
- Department of Clinical Research, Center of Molecular Immunology, Havana, Cuba
| | - Xianhong Bai
- Department of Medical Affairs, Biotech Pharmaceuticals Co., Ltd., P. R. China
| | - Binghe Xu
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| |
Collapse
|
44
|
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
|
45
|
Zhao J, Zhuo M, Wang Z, Duan J, Wang Y, Wang S, An T, Wu M, Wang J. A phase I study of nimotuzumab plus docetaxel in chemotherapy-refractory/resistant patients with advanced non-small-cell lung cancer. Chin J Cancer Res 2016; 28:12-18. [PMID: 27041923 PMCID: PMC4779759 DOI: 10.3978/j.issn.1000-9604.2016.02.07] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 02/02/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND To determine the safety and therapeutic efficacy of nimotuzumab (h-R3) combined with docetaxel in advanced non-small-cell lung cancer (NSCLC) patients who have failed to respond to prior first-line chemotherapy. METHODS In this single-center, open-label, dose-escalating phase I trial, patients with epidermal growth factor receptor (EGFR)-expressing stage IV NSCLC were treated with nimotuzumab plus docetaxel according to a dose escalation schedule. The safety and efficacy of the combination treatment were observed and analyzed. RESULTS There were 12 patients with EGFR-expressing stage IV NSCLC enrolled. The dose of nimotuzumab was escalated from 200 to 600 mg/week. The longest administration of study drug was 40 weeks at the 600 mg/week dose level. Grade III-IV toxicities included neutropenia and fatigue, and other toxicities included rash. Dose-limiting toxicity occurred with Grade 3 fatigue at the 200 mg dose level of nimotuzumab and Grade 4 neutropenia with pneumonia at the 600 mg dose level of nimotuzumab. No objective responses were observed, and stable disease was observed in eight patients (66.7%). The median progression-free survival (PFS) was 4.4 months in all patients, 1.3 months in patients with the EGFR mutation, and 4.4 months in those with wild type EGFR (EGFR WT). The median survival time (MST) was 21.1 months in all patients, 21.1 months in patients with EGFR mutation, and 26.4 months in patients with EGFR WT. CONCLUSIONS Nimotuzumab and docetaxel combination therapy was found to be well tolerated and efficacious. Further study of nimotuzumab is warranted in advanced NSCLC patients.
Collapse
Affiliation(s)
- Jun Zhao
- Departments of Thoracic Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Minglei Zhuo
- Departments of Thoracic Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Zhijie Wang
- Departments of Thoracic Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Jianchun Duan
- Departments of Thoracic Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Yuyan Wang
- Departments of Thoracic Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Shuhang Wang
- Departments of Thoracic Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Tongtong An
- Departments of Thoracic Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Meina Wu
- Departments of Thoracic Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Jie Wang
- Departments of Thoracic Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing 100142, China
| |
Collapse
|
46
|
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
|
47
|
Li K, Li J. Current Molecular Targeted Therapy in Advanced Gastric Cancer: A Comprehensive Review of Therapeutic Mechanism, Clinical Trials, and Practical Application. Gastroenterol Res Pract 2016; 2016:4105615. [PMID: 26880889 PMCID: PMC4736909 DOI: 10.1155/2016/4105615] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 11/10/2015] [Indexed: 02/06/2023] Open
Abstract
Despite the great progress in the treatment of gastric cancer, it is still the third leading cause of cancer death worldwide. Patients often miss the opportunity for a surgical cure, because the cancer has already developed into advanced cancer when identified. Compared to best supportive care, chemotherapy can improve quality of life and prolong survival time, but the overall survival is often short. Due to the molecular study of gastric cancer, new molecular targeted drugs have entered the clinical use. Trastuzumab, an antibody targeting human epidermal growth factor receptor 2 (HER2), can significantly improve survival in advanced gastric cancer patients with HER2 overexpression. Second-line treatment of advanced gastric cancer with ramucirumab, an antibody targeting VEGFR-2, alone or in combination with paclitaxel, has been proved to provide a beneficial effect. The VEGFR-2 tyrosine kinase inhibitor, apatinib, can improve the survival of advanced gastric cancer patients after second-line chemotherapy failure. Unfortunately, none of the EGFR targeting antibodies (cetuximab or panitumumab), VEGF targeting monoclonal antibodies (bevacizumab), mTOR inhibitor (everolimus), or HGF/MET pathway targeting drugs has a significant survival benefit. Many other clinical trials based on molecular markers are underway. This review will summarize targeted therapies for advanced gastric cancer.
Collapse
Affiliation(s)
- Kaichun Li
- Tianyou Hospital, Tongji University, Shanghai 200331, China
| | - Jin Li
- Tianyou Hospital, Tongji University, Shanghai 200331, China
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Department of Medical Oncology, Shanghai 200032, China
| |
Collapse
|
48
|
Nimotuzumab suppresses epithelial-mesenchymal transition and enhances apoptosis in low-dose UV-C treated salivary adenoid cystic carcinoma cell lines in vitro. Anticancer Drugs 2015; 25:1052-60. [PMID: 25035960 DOI: 10.1097/cad.0000000000000139] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Salivary adenoid cystic carcinoma (SACC), which is one of the most common malignant tumors of the salivary glands, is associated with a poor long-term outcome. There are currently few therapeutic options for patients with SACC. Recent studies have shown the potential of the application of ultraviolet-C (UV-C) irradiation for the treatment of human cancer. In the present study, we investigated the effects of UV-C in the SACC cell lines SACC-83 and SACC-LM. High-dose UV-C (200 J/m) induced apoptosis and inhibited colony formation significantly. However, low-dose UV-C (10 J/m), which had little effect on apoptosis and colony formation, increased the ability of migration in SACC cells accompanied by a decrease in E-cadherin and an increase in vimentin, suggesting the occurrence of epithelial-mesenchymal transition (EMT). Low-dose UV-C (10 J/m) also resulted in upregulation of the phosphorylated forms of epidermal growth factor receptor (EGFR) and Akt (p-EGFR and p-Akt, respectively). Pretreatment with Nimotuzumab, an anti-EGFR monoclonal antibody, reversed the EMT as well as upregulation of p-EGFR/p-Akt induced by UV-C. Moreover, Nimotuzumab enhanced UV-C induced apoptosis and inhibition of colony formation. Our results indicate that EMT exerts a protective effect against apoptosis induced by low-dose UV-C. Thus, the combined application of Nimotuzumab and low-dose UV-C in vitro has an advantageous antitumor effect in SACC compared with the application of UV-C alone.
Collapse
|
49
|
Jin T, Zhu Y, Luo JL, Zhou N, Li DC, Ju HX, Fan YT, Liu Y, Zhu YP, Feng HY, Liu LY. Prospective phase II trial of nimotuzumab in combination with radiotherapy and concurrent capecitabine in locally advanced rectal cancer. Int J Colorectal Dis 2015; 30:337-45. [PMID: 25564344 DOI: 10.1007/s00384-014-2097-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/11/2014] [Indexed: 02/04/2023]
Abstract
PURPOSE The aim of the study was to evaluate the safety and efficacy of adding concurrent nimotuzumab to preoperative radiotherapy with concurrent capecitabine in locally advanced rectal cancer. METHODS AND MATERIALS Patients with rectal cancer (clinical stage T3/4 or N+) were scheduled to receive weekly nimotuzumab (400 mg; days -6, 1, 8, 15, 22, and 29). Capecitabine (825 mg/m(2)) was delivered orally twice daily for the duration of radiotherapy. Radiotherapy was administered at 50.4 Gy (45 + 5.4 Gy). The main endpoint was the pathologic complete response (pCR) rate. RESULTS Twenty-one patients with T3 or T4 disease were enrolled; 66.7 % were nodal-positive; the median distance from the anal verge was 5.5 cm. A pCR was achieved in four patients (19.0 %); 71.4 % patients obtained moderate or good tumor regression (Grade 2 and 3). Downstaging occurred in 15/21 (71.4 %) patients by T stage and 11/14 (78.6 %) by N stage. The actual dose intensities (median/mean, %) were nimotuzumab (100, 100) and capecitabine (100, 99.5). The most frequent Grade 1/2 toxicities were radiation dermatitis (57.1 %), nausea/vomiting (52.4 %), leukocytopenia (47.6 %), diarrhea (47.6 %), and proctitis (38.1 %). Grade 3 diarrhea was observed in 9.5 % of patients and Grade 3 leukocytopenia in 4.8 %. CONCLUSION These preliminary results indicate that nimotuzumab can be safely combined with radiotherapy plus concurrent capecitabine. The efficacy of this regimen (pCR = 19.0 %) was significantly higher than that observed in previous phase II trials of preoperative radiotherapy with concurrent capecitabine and cetuximab in rectal cancer. Further investigation of concurrent nimotuzumab with radiotherapy plus capecitabine is warranted.
Collapse
Affiliation(s)
- Ting Jin
- Department of Radiation Oncology, Zhejiang Cancer Hospital, 38 Guang Ji Road, Hangzhou, 310022, Zhejiang, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Liu H, Yang W, Gao H, Jiang T, Gu B, Dong Q, Xu W, Wu S, Sun X. Nimotuzumab abrogates acquired radioresistance of KYSE-150R esophageal cancer cells by inhibiting EGFR signaling and cellular DNA repair. Onco Targets Ther 2015; 8:509-18. [PMID: 25750543 PMCID: PMC4348136 DOI: 10.2147/ott.s76958] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Background Acquired radioresistance of cancer is common after repeated irradiation and often leads to treatment failure. This study aimed to examine the effects of nimotuzumab on acquired radioresistance in human esophageal carcinoma cells and to investigate its underlying mechanisms. Methods The radioresistant human esophageal carcinoma cell line KYSE-150R was generated by using fractionated irradiation. KYSE-150R cells were pretreated with or without nimotuzumab before ionizing radiation. Cell growth and colony formation were measured to quantitate the effects of radiation. The γ-H2AX foci assay was employed to determine cellular DNA-repairing capacity. The phosphorylation of key molecules involved in the epidermal growth factor receptor (EGFR) signaling pathway and cellular DNA repair was measured by Western blot analysis. Results Nimotuzumab enhanced radiation-induced inhibition on cell growth and clonogenic survival in KYSE-150R cells. The average number of γ-H2AX foci increased in the irradiated cells treated with nimotuzumab. Nimotuzumab inhibited phosphorylation of the EGFR and its downstream molecules AKT and ERK. Phosphorylation of the DNA repair-related proteins DNA-PKcs, ATM, and RAD51 was also inhibited by nimotuzumab. Conclusions These results indicate that nimotuzumab can inhibit key cancer survival mechanisms, the EGFR signaling pathway, and DNA repair and thereby reverse acquired radioresistance in KYSE-150R cell line.
Collapse
Affiliation(s)
- Hai Liu
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, Sir Run Run Shaw Institute of Clinical Medicine of Zhejiang University, Hangzhou, People's Republic of China
| | - Weifang Yang
- Department of Radiation Oncology, Laboratory of Cellular and Molecular Radiation Oncology, Taizhou Hospital, Wenzhou Medical College, Taizhou, People's Republic of China
| | - Huaping Gao
- Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Tingting Jiang
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, Sir Run Run Shaw Institute of Clinical Medicine of Zhejiang University, Hangzhou, People's Republic of China
| | - Bengxin Gu
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, Sir Run Run Shaw Institute of Clinical Medicine of Zhejiang University, Hangzhou, People's Republic of China
| | - Qinghua Dong
- Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Wenhong Xu
- Department of Radiation Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, People's Republic of China
| | - Shixiu Wu
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Wushan District, Hangzhou First People's Hospital, Hangzhou, People's Republic of China
| | - Xiaonan Sun
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, Sir Run Run Shaw Institute of Clinical Medicine of Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|