1
|
Mohammad-Jafari K, Naghib SM, Mozafari MR. Cisplatin-based Liposomal Nanocarriers for Drug Delivery in Lung Cancer Therapy: Recent Progress and Future Outlooks. Curr Pharm Des 2024; 30:2850-2881. [PMID: 39051580 DOI: 10.2174/0113816128304923240704113319] [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: 02/13/2024] [Accepted: 05/23/2024] [Indexed: 07/27/2024]
Abstract
In order to improve the treatment of lung cancer, this paper looks at the development of cisplatinbased liposomal nanocarriers. It focuses on addressing the drawbacks of conventional cisplatin therapy, including systemic toxicity, inadequate tumor targeting, and drug resistance. Liposomes, or spherical lipid vesicles, offer a potentially effective way to encapsulate cisplatin, enhancing its transport and minimizing harmful effects on healthy tissues. The article discusses many liposomal cisplatin formulations, including pH-sensitive liposomes, sterically stabilized liposomes, and liposomes coupled with specific ligands like EGFR antibodies. These novel formulations show promise in reducing cisplatin resistance, optimizing pharmacokinetics, and boosting therapeutic results in the two in vitro and in vivo models. They also take advantage of the Enhanced Permeability and Retention (EPR) effect in the direction of improved tumor accumulation. The study highlights the need for more investigation to move these liposomal formulations from experimental to clinical settings, highlighting their potential to offer less harmful and more effective cancer therapy alternatives.
Collapse
Affiliation(s)
- Kave Mohammad-Jafari
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - M R Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
2
|
Zhang Y, Gu W, Shao Y. The therapeutic targets of N6-methyladenosine (m6A) modifications on tumor radioresistance. Discov Oncol 2023; 14:141. [PMID: 37522921 PMCID: PMC10390431 DOI: 10.1007/s12672-023-00759-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 07/24/2023] [Indexed: 08/01/2023] Open
Abstract
Radiation therapy is an important tool for malignant tumors, and its tolerance needs to be addressed. In recent years, several studies have shown that regulators of aberrant m6A methylation play an important role in the formation, development and invasion and metastasis of tumors. A large number of studies have confirmed aberrant m6A methylation as a new target for tumour therapy, but research on whether it can play a role in tumor sensitivity to radiotherapy has not been extensive and thorough enough. Recent studies have shown that all three major enzymes of m6A methylation have significant roles in radioresistance, and that the enzymes that play a role differ in different tumor types and by different mechanisms, including regulating tumor cell stemness, affecting DNA damage and repair, and controlling the cell cycle. Therefore, elucidating the mechanisms of m6A methylation in the radiotherapy of malignant tumors is essential to counteract radioresistance, improve the efficacy of radiotherapy, and even propose targeted treatment plans for specific tumors. The latest research progress on m6A methylation and radioresistance is reviewed in this article.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Changzhou, 213003, China
| | - Wendong Gu
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Changzhou, 213003, China.
| | - Yingjie Shao
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Changzhou, 213003, China.
| |
Collapse
|
3
|
Koch JP, Roth SM, Quintin A, Gavini J, Orlando E, Riedo R, Pozzato C, Hayrapetyan L, Aebersold R, Stroka DM, Aebersold DM, Medo M, Zimmer Y, Medová M. A DNA-PK phosphorylation site on MET regulates its signaling interface with the DNA damage response. Oncogene 2023; 42:2113-2125. [PMID: 37188738 PMCID: PMC10289896 DOI: 10.1038/s41388-023-02714-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/21/2023] [Accepted: 05/02/2023] [Indexed: 05/17/2023]
Abstract
The DNA damage response (DDR) is intertwined with signaling pathways downstream of oncogenic receptor tyrosine kinases (RTKs). To drive research into the application of targeted therapies as radiosensitizers, a better understanding of this molecular crosstalk is necessary. We present here the characterization of a previously unreported MET RTK phosphosite, Serine 1016 (S1016) that represents a potential DDR-MET interface. MET S1016 phosphorylation increases in response to irradiation and is mainly targeted by DNA-dependent protein kinase (DNA-PK). Phosphoproteomics unveils an impact of the S1016A substitution on the overall long-term cell cycle regulation following DNA damage. Accordingly, the abrogation of this phosphosite strongly perturbs the phosphorylation of proteins involved in the cell cycle and formation of the mitotic spindle, enabling cells to bypass a G2 arrest upon irradiation and leading to the entry into mitosis despite compromised genome integrity. This results in the formation of abnormal mitotic spindles and a lower proliferation rate. Altogether, the current data uncover a novel signaling mechanism through which the DDR uses a growth factor receptor system for regulating and maintaining genome stability.
Collapse
Affiliation(s)
- Jonas P Koch
- Department for BioMedical Research, Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, 3008, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3010, Bern, Switzerland
| | - Selina M Roth
- Department for BioMedical Research, Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, 3008, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3010, Bern, Switzerland
| | - Aurélie Quintin
- Department for BioMedical Research, Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, 3008, Bern, Switzerland
| | - Jacopo Gavini
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3010, Bern, Switzerland
- Department for BioMedical Research, Visceral Surgery, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Eleonora Orlando
- Department for BioMedical Research, Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, 3008, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3010, Bern, Switzerland
| | - Rahel Riedo
- Department for BioMedical Research, Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, 3008, Bern, Switzerland
| | - Chiara Pozzato
- Department for BioMedical Research, Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, 3008, Bern, Switzerland
| | - Liana Hayrapetyan
- Department for BioMedical Research, Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, 3008, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3010, Bern, Switzerland
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, 8093, Zürich, Switzerland
- Faculty of Science, University of Zürich, 8057, Zürich, Switzerland
| | - Deborah M Stroka
- Department for BioMedical Research, Visceral Surgery, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Daniel M Aebersold
- Department for BioMedical Research, Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, 3008, Bern, Switzerland
| | - Matúš Medo
- Department for BioMedical Research, Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, 3008, Bern, Switzerland
| | - Yitzhak Zimmer
- Department for BioMedical Research, Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, 3008, Bern, Switzerland
| | - Michaela Medová
- Department for BioMedical Research, Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, 3008, Bern, Switzerland.
| |
Collapse
|
4
|
Predicting tumour radiosensitivity to deliver precision radiotherapy. Nat Rev Clin Oncol 2023; 20:83-98. [PMID: 36477705 DOI: 10.1038/s41571-022-00709-y] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2022] [Indexed: 12/13/2022]
Abstract
Owing to advances in radiotherapy, the physical properties of radiation can be optimized to enable individualized treatment; however, optimization is rarely based on biological properties and, therefore, treatments are generally planned with the assumption that all tumours respond similarly to radiation. Radiation affects multiple cellular pathways, including DNA damage, hypoxia, proliferation, stem cell phenotype and immune response. In this Review, we summarize the effect of these pathways on tumour responses to radiotherapy and the current state of research on genomic classifiers designed to exploit these variations to inform treatment decisions. We also discuss whether advances in genomics have generated evidence that could be practice changing and whether advances in genomics are now ready to be used to guide the delivery of radiotherapy alone or in combination.
Collapse
|
5
|
Ghorbanpour M, Soltani B, Mota A, Jahanbin Sardroodi J, Mehdizadeh Aghdam E, Shayanfar A, Molavi O, Mohammad-Rezaei R, Ebadi-Nahari M, Ziegler CJ. Copper (II) complexes with N, S donor pyrazole-based ligands as anticancer agents. Biometals 2022; 35:1095-1111. [PMID: 36001216 DOI: 10.1007/s10534-022-00426-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 07/19/2022] [Indexed: 11/25/2022]
Abstract
A group of bidentate nitrogen and sulfur donor pyrazole derivative ligands abbreviated as Na[RNCS(Pz)], Na[RNCS(PzMe2)], Na[RNCS(PzMe3)], Na[RNCS(PzPhMe)], Na[RNCS(PzPh2)], where (R = Et, Ph), and their Cu (II) complexes were synthesized and characterized by spectroscopic and physicochemical methods. The crystal structure of [Cu(PhNCSPzMe3)2] was determined by X-ray crystallography analysis and the results described a distorted square planar coordination geometry for this complex. Also, the cyclic voltammetry investigations indicated that the synthesized copper complex is an electrochemically active species. Moreover, the cytotoxic activity of all of the twenty synthesized compounds was evaluated using MTT assay against the MCF-7 (human breast carcinoma) cell lines, in vitro. Cu (II) complexes indicate significant cytotoxicity against the MCF-7 cell lines as compared with the free ligands. The docking studies showed that the copper complexes have better interactions with EGFR and CDK2 proteins, compared to the free ligands, and most of the studied compounds have a higher value of binding energy relative to the studied controls. The results of QSAR analysis suggest that dipole moment is in direct correlation with the obtained IC50 values, and it strongly impact the anticancer effects generated by the compounds. Our findings suggest that the developed copper complexes can be good candidates for further evaluations as chemotherapeutic agents in the treatment of cancer.
Collapse
Affiliation(s)
- Monireh Ghorbanpour
- Department of Chemistry, Faculty of Basic Science, Azarbaijan Shahid Madani University, P. O. Box 53714-161, Tabriz, Iran
| | - Behzad Soltani
- Department of Chemistry, Faculty of Basic Science, Azarbaijan Shahid Madani University, P. O. Box 53714-161, Tabriz, Iran.
| | - Ali Mota
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaber Jahanbin Sardroodi
- Department of Chemistry, Faculty of Basic Science, Azarbaijan Shahid Madani University, P. O. Box 53714-161, Tabriz, Iran
| | - Elnaz Mehdizadeh Aghdam
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shayanfar
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rahim Mohammad-Rezaei
- Department of Chemistry, Faculty of Basic Science, Azarbaijan Shahid Madani University, P. O. Box 53714-161, Tabriz, Iran
| | - Mostafa Ebadi-Nahari
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | | |
Collapse
|
6
|
Papini F, Sundaresan J, Leonetti A, Tiseo M, Rolfo C, Peters GJ, Giovannetti E. Hype or hope - Can combination therapies with third-generation EGFR-TKIs help overcome acquired resistance and improve outcomes in EGFR-mutant advanced/metastatic NSCLC? Crit Rev Oncol Hematol 2021; 166:103454. [PMID: 34455092 DOI: 10.1016/j.critrevonc.2021.103454] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/07/2021] [Accepted: 08/10/2021] [Indexed: 02/08/2023] Open
Abstract
Three generations of epidermal growth factor receptor - tyrosine kinase inhibitors (EGFR-TKIs) have been developed for treating advanced/metastatic non-small cell lung cancer (NSCLC) patients harboring EGFR-activating mutations, while a fourth generation is undergoing preclinical assessment. Although initially effective, acquired resistance to EGFR-TKIs usually arises within a year due to the emergence of clones harboring multiple resistance mechanisms. Therefore, the combination of EGFR-TKIs with other therapeutic agents has emerged as a potential strategy to overcome resistance and improve clinical outcomes. However, results obtained so far are ambiguous and ideal therapies for patients who experience disease progression during treatment with EGFR-TKIs remain elusive. This review provides an updated landscape of EGFR-TKIs, along with a description of the mechanisms causing resistance to these drugs. Moreover, it discusses the current knowledge, limitations, and future perspective regarding the use of EGFR-TKIs in combination with other anticancer agents, supporting the need for bench-to-bedside approaches in selected populations.
Collapse
Affiliation(s)
- Filippo Papini
- Department of Medical Oncology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, the Netherlands; Fondazione Pisana per la Scienza, Pisa, Italy
| | - Janani Sundaresan
- Department of Medical Oncology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Alessandro Leonetti
- Department of Medical Oncology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Medicine and Surgery, University of Parma, Parma, Italy; Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Marcello Tiseo
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Christian Rolfo
- The Center of Thoracic Oncology at the Tisch Cancer Institute, Mount Sinai, NYC, United States
| | - Godefridus J Peters
- Department of Medical Oncology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Biochemistry, Medical University of Gdansk, Poland
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, the Netherlands; Fondazione Pisana per la Scienza, Pisa, Italy.
| |
Collapse
|
7
|
Kummer B, Löck S, Gurtner K, Hermann N, Yaromina A, Eicheler W, Baumann M, Krause M, Jentsch C. Value of functional in-vivo endpoints in preclinical radiation research. Radiother Oncol 2021; 158:155-161. [PMID: 33639191 DOI: 10.1016/j.radonc.2021.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 02/11/2021] [Accepted: 02/18/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND Cancer research faces the problem of high rates of clinical failure of new treatment approaches after positive preclinical data. We hypothesize that a major confounding factor to this problem in radiooncology is the choice of the preclinical endpoint. METHODS We present a comprehensive re-evaluation of large-scale preclinical in-vivo data on fractionated irradiation alone or simultaneously with Epidermal Growth Factor Receptor inhibition. Taking the permanent local tumour control assay as a gold standard, we evaluated different tumour volume dependent endpoints that are widely used for preclinical experiments. RESULTS The analysis showed the highest correlations between volume related and local tumour control endpoints after irradiation alone. For combined treatments, wide inter-tumoural variations were observed with reduced correlation between the endpoints. Evaluation of growth delay per Gray (GD/Gy) based on two or more dose levels showed closest correlation with local tumour control dose 50% (TCD50). CONCLUSIONS GD/Gy with at least two dose groups correlates with TCD50, but cannot replace the latter as the goldstandard.
Collapse
Affiliation(s)
- Berit Kummer
- Department of Radiation Oncology and OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Steffen Löck
- Department of Radiation Oncology and OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center, Heidelberg, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany; German Cancer Research Center (DKFZ) Heidelberg, Germany
| | - Kristin Gurtner
- Department of Radiation Oncology and OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany; German Cancer Research Center (DKFZ) Heidelberg, Germany
| | - Nadine Hermann
- Department of Radiation Oncology and OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Ala Yaromina
- Department of Radiation Oncology and OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany; The D-Lab and The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Wolfgang Eicheler
- Department of Radiation Oncology and OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Michael Baumann
- Department of Radiation Oncology and OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany; German Cancer Consortium (DKTK), Core Center Heidelberg, Germany; German Cancer Research Center (DKFZ) Heidelberg, Germany
| | - Mechthild Krause
- Department of Radiation Oncology and OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center, Heidelberg, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany; German Cancer Research Center (DKFZ) Heidelberg, Germany.
| | - Christina Jentsch
- Department of Radiation Oncology and OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany; German Cancer Research Center (DKFZ) Heidelberg, Germany
| |
Collapse
|
8
|
Zhang P, Song E, Jiang M, Song Y. Celecoxib and Afatinib synergistic enhance radiotherapy sensitivity on human non-small cell lung cancer A549 cells. Int J Radiat Biol 2020; 97:170-178. [PMID: 33164600 DOI: 10.1080/09553002.2021.1846817] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE Radioresistance is highly correlated with radiotherapy failure in clinical cancer treatment. In the current study, we sought to examine the efficacy of Celecoxib and Afatinib co-treatment as radiosensitizers in the management of non-small cell lung cancer (NSCLC) A549 cells. MATERIALS AND METHODS Generally, A549 cells were cultured with the treatment of Celecoxib and/or Afatinib for 24 h. Then, the cells were exposed to irradiation at 2 Gy/min for 1 min. After the end of treatment, cell viability, clonogenic survival, apoptosis and Prostaglandin E2 (PGE2) Elisa assays were performed. Transcriptional levels of Cyclooxygenase-2 (COX-2) affected by Celecoxib and/or Afatinib were measured by RT-qPCR. Posttranscriptional level of epidermal growth factor receptor (EGFR)-related gene was measured by Western blotting analysis. RESULTS Here, we, for the first time, reported that the co-treatment of Celecoxib and Afatinib regulates the resistance of NSCLC A549 cells to radiation. The co-treatment of Celecoxib and Afatinib sensitized radiotherapy through the radiation-induced loss of cell viability and colony formation, as well as apoptosis. Mechanistically, Celecoxib and Afatinib-treated cells showed the inhibition of COX-2 and EGFR expression, which may be responsible for the A549 cells' increased resistance to radiation. CONCLUSION Our results suggested that Celecoxib and Afatinib regulate cell sensitivity to apoptosis, and thus modulate the resistance of NSCLC to radiation.
Collapse
Affiliation(s)
- Pan Zhang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China
| | - Erqun Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China
| | - Mingdong Jiang
- Department of Radiation Oncology, The Ninth People's Hospital of Chongqing, Chongqing, People's Republic of China
| | - Yang Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China
| |
Collapse
|
9
|
Jiang L, Meng X, Zhao X, Xing L, Yu J. Perspective on treatment for unresectable locally advanced non-small cell lung cancer with oncogene-driven mutation: a narrative review. Transl Lung Cancer Res 2020; 9:2137-2144. [PMID: 33209632 PMCID: PMC7653115 DOI: 10.21037/tlcr-20-722] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The standard treatment of unresectable locally advanced non-small cell lung cancer (LA NSCLC) is concurrent chemoradiotherapy. With the addition of immunotherapy, patients with LA NSCLC received a significantly prolonged outcome, while patients with harboring epidermal growth factor receptor (EGFR) mutation benefited less. Tyrosine kinase inhibitors (TKIs) have revolutionized the treatment of stage IV with harboring EGFR mutation and anaplastic lymphoma kinase rearrangement, but there are few recommendations indicating whether TKI treatment is effective in unresectable NSCLC. Preclinical studies have shown that TKIs could have a radiosensitizing effect, which provided a rationale to consider the application TKI with radiotherapy. In this review, we summarize the clinical studies that have used TKIs in LA-NSCLC as well as ongoing trials, and discuss recent progress in research related to the efficacy of TKI for unresectable LA NSCLC patients. Recent results of small studies evaluating TKI therapy for LA NSCLC patients in combination with radiation or chemoradiation demonstrated promising efficacy, improved outcomes with a tolerable toxicity profile. However, there is a lack of strong evidence for TKI treatment in unresectable LA NSCLC, because of unpowered statistics, lack of molecular selection, or lack of large randomized arms. We prospect the combination of TKI and radiation or chemoradiation therapy might eventually replace the current standard treatment for patients with LA NSCLC harboring oncogene-driven mutation.
Collapse
Affiliation(s)
- Liyang Jiang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xianguang Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Ligang Xing
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
10
|
Yu Y, Guan H, Jiang L, Li X, Xing L, Sun X. Nimotuzumab, an EGFR‑targeted antibody, promotes radiosensitivity of recurrent esophageal squamous cell carcinoma. Int J Oncol 2020; 56:945-956. [PMID: 32319582 DOI: 10.3892/ijo.2020.4981] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 12/12/2019] [Indexed: 11/06/2022] Open
Abstract
Local tumor recurrence is one of the main causes for the failure of esophageal cancer treatment following radiotherapy. Previous studies have demonstrated that epidermal growth factor receptor (EGFR)‑targeted therapy combined with radiotherapy is expected to become an effective means to control tumor recurrence. The aim of the present study was to investigate the effect and mechanism of nimotuzumab (an EGFR‑targeted antibody) in the treatment of recurrent esophageal carcinoma. The radiation responses of two esophageal squamous carcinoma cell lines, EC109 and TE‑1, with or without nimotuzumab, were first evaluated by CCK‑8 assay. Colony formation and apoptosis were used to measure anti‑proliferation effects. It was demonstrated that nimotuzumab arrested the cell cycle at the G2 phase in vitro. Western blotting and immunofluorescence analysis were used to determine signaling pathway changes. It was observed that nimotuzumab inhibited phosphorylation of EGFR in EC109 cells. Furthermore, recurrent tumor models were established and it was identified that the degree of tumor hypoxia was positively associated with EGFR overexpression. In EC109 cell xenografts, nimotuzumab combined with radiation led to a significant delay in recurrent tumor growth compared with that of radiation alone (P<0.001 for 0 Gy pre‑irradiation, P=0.005 for 20 Gy pre‑irradiation, P=0.005 for 10 Gy pre‑irradiation). These results suggest that nimotuzumab combined with radiation may be an effective means to control recurrent esophageal squamous cell carcinoma with EGFR overexpression.
Collapse
Affiliation(s)
- Yang Yu
- School of Medicine and Life Sciences, Shandong Academy of Medical Sciences, University of Jinan, Jinan, Shandong 250031, P.R. China
| | - Hui Guan
- Department of Radiation Oncology, The Fourth People's Hospital of Jinan, Jinan, Shandong 250031, P.R. China
| | - Liyang Jiang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Xiaolin Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Ligang Xing
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Xiaorong Sun
- Department of Nuclear Medicine, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
11
|
Spagnuolo A, Muto M, Monaco F, Colantuoni G, Gridelli C. The optional approach of oncogene-addicted non-small cell lung cancer with brain metastases in the new generation targeted therapies era. Transl Lung Cancer Res 2019; 8:1134-1151. [PMID: 32010591 PMCID: PMC6976372 DOI: 10.21037/tlcr.2019.12.24] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 11/13/2019] [Indexed: 12/11/2022]
Abstract
In recent years, the study of the molecular characteristics of non-small cell lung cancer (NSCLC) has highlighted a specific role of some genes that represent important therapeutic targets, including epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), c-ros oncogene 1 (ROS-1) and v-Raf murine sarcoma viral oncogene homolog B1 (BRAF). Patients with oncogene-addicted cancer benefit more from therapy with tyrosine kinase inhibitors (TKIs) than from chemotherapy. The brain is a preferred site for tumor spread in these patients. In addition, given greater control of extracranial disease and prolonged survival, the brain is often the first site of progression. Therefore, there is great interest in therapeutic approaches that optimize the control of intracranial disease associated with systemic drugs that, by penetrating the blood-brain barrier (BBB), may improve local control. On the latter, radiotherapy provides excellent efficacy but following the results of clinical trials with new brain penetrant drugs, the question of how and especially when to perform brain radiotherapy in patients with oncogene-addicted NSCLC remains open. Prospective studies may indicate which patients are most likely to benefit from combined use or in what sequence they will undergo systemic and radiotherapy treatment. Due to the heterogeneity of patients and the introduction of new generation TKIs, a multidisciplinary assessment for the best management of therapies in NSCLC patients with molecular driver alterations and brain metastases (BM) is required.
Collapse
Affiliation(s)
- Alessia Spagnuolo
- Division of Medical Oncology, ‘S. G. Moscati’ Hospital, Avellino, Italy
| | - Matteo Muto
- Division of Radiotherapy, ‘S. G. Moscati’ Hospital, Avellino, Italy
| | - Fabio Monaco
- Division of Radiation Protection, ‘S. G. Moscati’ Hospital, Avellino, Italy
| | | | - Cesare Gridelli
- Division of Medical Oncology, ‘S. G. Moscati’ Hospital, Avellino, Italy
| |
Collapse
|
12
|
Combination of chemotherapy and radiotherapy: A thirty years evolution. Cancer Radiother 2019; 23:662-665. [PMID: 31473087 DOI: 10.1016/j.canrad.2019.07.157] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 07/06/2019] [Indexed: 02/07/2023]
Abstract
Chemoradiotherapy is now considered the standard of care for many locally advanced diseases. Cytotoxic drugs have been largely evaluated in this setting, with cisplatin and 5FU the most often used drugs. A large amount of pre-clinical studies has demonstrated the synergy between both modalities. Concomitant administration seems the more beneficial in many diseases. Emergence of new approaches, combining targeted therapies and radiotherapy (RT) is now a reality. The main example is the association of cetuximab and RT in head and neck carcinomas, even if, 14 years after the initial publication, the best way to use it is still unknown. New compounds as inhibitors of DNA-repair or immune checkpoints are under investigation and showed early promising results.
Collapse
|
13
|
Gurtner K, Kryzmien Z, Koi L, Wang M, Benes CH, Hering S, Willers H, Baumann M, Krause M. Radioresistance of KRAS/TP53-mutated lung cancer can be overcome by radiation dose escalation or EGFR tyrosine kinase inhibition in vivo. Int J Cancer 2019; 147:472-477. [PMID: 31359406 DOI: 10.1002/ijc.32598] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 07/02/2019] [Indexed: 12/25/2022]
Abstract
Recent clinical data have linked KRAS/TP53 comutation (mut) to resistance to radiotherapy (RT), but supporting laboratory in vivo evidence is lacking. In addition, the ability of different radiation doses, with/without epidermal growth factor receptor (EGFR)-directed treatment, to achieve local tumor control as a function of KRAS status is unknown. Here, we assessed clonogenic radiation survival of a panel of annotated lung cancer cell lines. KRASmut/TP53mut was associated with the highest radioresistance in nonisogenic and isogenic comparisons. To validate these findings, isogenic TP53mut NCI-H1703 models, KRASmut or wild-type (wt), were grown as heterotopic xenografts in nude mice. A clinical RT schedule of 30 fractions over 6 weeks was employed. The dose that controlled 50% of tumors (TCD50 ) was calculated. The TCD50 for KRASwt/TP53mut xenografts was 43.1 Gy whereas KRASmut/TP53mut tumors required a 1.9-fold higher TCD50 of 81.4 Gy. The EGFR inhibitor erlotinib radiosensitized KRASmut but not KRASwt cells and xenografts. The TCD50 associated with adding erlotinib to RT was 58.8 Gy for KRASmut, that is, a ~1.4-fold dose enhancement. However, the EGFR antibody cetuximab did not have a radiosensitizing effect. In conclusion, we demonstrate for the first time that KRASmut in a TP53mut background confers radioresistance when studying a clinical RT schedule and local control rather than tumor growth delay. Despite the known unresponsiveness of KRASmut tumors to EGFR inhibitors, erlotinib radiosensitized KRASmut tumors. Our data highlight KRAS/TP53 comutation as a candidate biomarker of radioresistance that can be at least partially reversed by dose escalation or the addition of a targeted agent.
Collapse
Affiliation(s)
- Kristin Gurtner
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany.,Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,National Center for Tumour Diseases (NCT), Dresden, Germany
| | - Zofia Kryzmien
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany.,Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Lydia Koi
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany.,Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Meng Wang
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Cyril H Benes
- Center for Cancer Research, Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
| | - Sandra Hering
- Institute of Legal Medicine, Medical Faculty Carl Gustav Carus, University of Technology, Dresden, Germany
| | - Henning Willers
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Michael Baumann
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany.,Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,National Center for Tumour Diseases (NCT), Dresden, Germany.,Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany.,Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,National Center for Tumour Diseases (NCT), Dresden, Germany.,Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany.,German Cancer Consortium (DKTK), Dresden, Germany
| |
Collapse
|
14
|
Liu R, Wei S, Zhang Q, Zhang X, Luo H, Tian J, Li Y, Ge L, Wang X. Epidermal growth factor receptor tyrosine kinase inhibitors combined with thoracic radiotherapy or chemoradiotherapy for advanced or metastatic non-small cell lung cancer: A systematic review and meta-analysis of single-arm trials. Medicine (Baltimore) 2019; 98:e16427. [PMID: 31335695 PMCID: PMC6708798 DOI: 10.1097/md.0000000000016427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 05/24/2019] [Accepted: 06/17/2019] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Preclinical in vitro experiments demonstrated that epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) might have synergistic effect in combination with radiotherapy on Non-small cell lung cancer (NSCLC), but the clinical trials showed inconsistence results in NSCLC patients with EGFR status unknow or mutations. This study aimed to determine if added TKIs to Thoracic radiotherapy (TRT) improve primary disease response rate (RR) and survival outcomes in advanced or metastatic NSCLC. METHODS We searched MEDLINE, EMBASE, and Cochrane Library from January 2000 to December 2017 for eligible studies where patients received concurrent EGFR TKIs and TRT or CRT. Concerned outcomes were primary tumor RR, overall survival (OS), and adverse events (AEs). The meta-analysis was performed using Stata software (version 12.0). Random effects models were used to pool outcomes across studies. Sensitivity analysis was performed to determine if the results would be different. RESULTS We found 16 prospective clinical trials with mature results for meta-analyses. Twelve studies including 446 patients reported the RR and survival outcomes of TRT combined TKIs. The CR, PR, SD, and PD, respectively, were 0.06 (95% CI 0.03-0.09, I = 0%), 0.44 (95% CI 0.38-0.49, I = 64.9%), 0.29 (95% CI 0.24-0.34, I = 78.4%), and 0.15 (95% CI 0.11-0.19, I = 84.2%). One- and 2-year OS, respectively, were 0.52 (95% CI 0.44-0.60, I = 38.8%) and 0.26 (95% CI 0.18-0.33, I = 0%). Four studies including 182 patients reported the RR and survival outcomes of CRT combined TKIs. The pooled CR, PR, SD, and PD, respectively, were 0.12 (95% CI 0.02-0.22, I = 69.1%), 0.41 (95% CI 0.27-0.55, I71.6%), 0.31 (95% CI 0.16-0.46, I = 79%), and 0.14 (95% CI -0.01-0.30, I = 87.8%). Only 1 study reported the survival event rate, 1- and 2-year OS, respectively, were 0.83 (95% CI 0.71-0.94) and 0.67 (95% CI 0.54-0.81). There were not severe adverse events (SAEs) reported either TRT combined TKIs or CRT combined TKIs. CONCLUSION There is evidence, albeit of low quality, that added the TKIs to TRT or CRT may improve RR and survival outcomes in patients with EGFR mutant status unknown advanced or metastatic NSCLC relative to other studies of TKIs alone, TRT alone or CRT.
Collapse
Affiliation(s)
- Ruifeng Liu
- The First Clinical Medical College of Lanzhou University
- Radiotherapy Oncology Department, Gansu Provincial Cancer Hospital
| | - Shihong Wei
- Radiotherapy Oncology Department, Gansu Provincial Cancer Hospital
| | - Qiuning Zhang
- The First Clinical Medical College of Lanzhou University
- Radiotherapy Oncology Department, Gansu Provincial Cancer Hospital
| | - Xueliang Zhang
- Radiotherapy Oncology Department, Gansu Provincial Cancer Hospital
| | - Hongtao Luo
- The First Clinical Medical College of Lanzhou University
- Radiotherapy Oncology Department, Gansu Provincial Cancer Hospital
| | - Jinhui Tian
- Center of Evidence Based Medicine, Lanzhou University, Lanzhou
| | - Yi Li
- The First Clinical Medical College of Lanzhou University
- Hebei provincial chest Hospital, Shijiazhuang, PR China
| | - Long Ge
- The First Clinical Medical College of Lanzhou University
- Center of Evidence Based Medicine, Lanzhou University, Lanzhou
| | - Xiaohu Wang
- The First Clinical Medical College of Lanzhou University
- Radiotherapy Oncology Department, Gansu Provincial Cancer Hospital
| |
Collapse
|
15
|
Rieckmann T, Kriegs M. The failure of cetuximab-based de-intensified regimes for HPV-positive OPSCC: A radiobiologists perspective. Clin Transl Radiat Oncol 2019; 17:47-50. [PMID: 31206086 PMCID: PMC6558227 DOI: 10.1016/j.ctro.2019.05.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 05/20/2019] [Indexed: 01/17/2023] Open
Abstract
Recent trial results show inferiority of cetuximab- to cisplatin-radiotherapy in HPV+ OPSCC. Previous data also question the benefit of cetuximab in HNSCC in the curative setting. The data provide guidance for research on radiosensitization through molecular targeting.
Human Papillomavirus-positive oropharyngeal cancer is a rising tumor entity with unique characteristics and favorable prognosis. Because current multimodal therapies are associated with severe toxicity, different strategies for treatment de-intensification are being tested in clinical trials. In this context two phase 3 studies, which examined the potential of the monoclonal anti-EGFR antibody cetuximab to replace concomitant cisplatin-based chemotherapy, have concordantly reported inferiority of this de-intensification approach. In this opinion article we discuss these recent negative results in the light of previous clinical and preclinical research on the combination of EGFR-inhibition and irradiation. Collectively these data question the effectiveness of EGFR-inhibition in the curative treatment of both HPV-positive and HPV-negative head and neck cancer but provide guidance for future translational research.
Collapse
Affiliation(s)
- Thorsten Rieckmann
- Laboratory of Radiobiology & Experimental Radiation Oncology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
- Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Hamburg, Germany
- Corresponding author at: Laboratory of Radiobiology & Department of Otolaryngology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | - Malte Kriegs
- Laboratory of Radiobiology & Experimental Radiation Oncology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| |
Collapse
|
16
|
Li J, Wang L, Qiu Z, Su Y. Time profile of nimotuzumab for enhancing radiosensitivity of the Eca109 cell line. Oncol Lett 2019; 17:2763-2769. [PMID: 30854050 PMCID: PMC6365957 DOI: 10.3892/ol.2019.9897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 08/16/2018] [Indexed: 12/29/2022] Open
Abstract
The aim of the present study was to investigate the ability of Nimotuzumab to increase radiosensitivity at different delivery times in the mixed cancer cell line Eca109, to determine the optimal delivery time. Cultured Eca109 cells were classified into five groups: Control with no treatment (O group); irradiation without Nimotuzumab treatment (R group); treatment with Nimotuzumab 24 h prior to or after irradiation (24NR or 24RN group, respectively); and Nimotuzumab combined with irradiation simultaneously (NR group). Following cells reaching the logarithmic-growth phase, cell survival after exposure to Nimotuzumab was evaluated using an MTT assay; thereafter, the 50% inhibitory concentration (IC50) of the cell line was calculated. Cell-survival curves were generated using a colony-forming assay. Flow cytometry analysis was used to detect apoptosis rates and cell-cycle distribution. The expression level of epidermal growth factor receptor was measured in Eca109 cells with western blotting. Growth inhibition was only observed 72 h after exposure to Nimotuzumab. The IC50 was 768 µg/ml. At a dose of 0.2 IC50 or 0.3 IC50, the sensitization enhancement ratio of radiosensitivity was highest in the 24NR group. Nimotuzumab enhanced radiation-induced apoptosis in Eca109 cells, with the optimal delivery time at 24 h prior to irradiation (P=0.035). The concentration of Nimotuzumab administered was directly proportional to the increase in radiosensitivity of the cells.
Collapse
Affiliation(s)
- Jiancheng Li
- Department of Radiation Oncology, Fujian Provincial Tumor Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou, Fujian 350014, P.R. China
| | - Linghua Wang
- Department of Radiation Oncology, Fujian Provincial Tumor Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou, Fujian 350014, P.R. China
| | - Zidan Qiu
- Department of Radiation Oncology, Fujian Provincial Tumor Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou, Fujian 350014, P.R. China
| | - Ying Su
- Department of Radiation Oncology, Fujian Provincial Tumor Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou, Fujian 350014, P.R. China
| |
Collapse
|
17
|
Arcangeli S, Jereczek-Fossa BA, Alongi F, Aristei C, Becherini C, Belgioia L, Buglione M, Caravatta L, D'Angelillo RM, Filippi AR, Fiore M, Genovesi D, Greco C, Livi L, Magrini SM, Marvaso G, Mazzola R, Meattini I, Merlotti A, Palumbo I, Pergolizzi S, Ramella S, Ricardi U, Russi E, Trovò M, Sindoni A, Valentini V, Corvò R. Combination of novel systemic agents and radiotherapy for solid tumors - part I: An AIRO (Italian association of radiotherapy and clinical oncology) overview focused on treatment efficacy. Crit Rev Oncol Hematol 2019; 134:87-103. [PMID: 30658886 DOI: 10.1016/j.critrevonc.2018.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 11/13/2018] [Accepted: 11/20/2018] [Indexed: 02/07/2023] Open
Abstract
Over the past century, technologic advances have promoted the evolution of radiation therapy into a precise treatment modality allowing for the maximal administration of dose to tumors while sparing normal tissues. In parallel with this technological maturation, the rapid expansion in understanding the basic biology and heterogeneity of cancer has led to the development of several compounds that target specific pathways. Many of them are in advanced steps of clinical development for combination treatments with radiotherapy, and can be incorporated into radiation oncology practice for a personalized approach to maximize the therapeutic gain. This review describes the rationale for combining novel agents with radiation, and provides an overview of the current landscape focused on treatment efficacy.
Collapse
Affiliation(s)
- Stefano Arcangeli
- Department of Radiation Oncology, Policlinico S. Gerardo and University of Milan "Bicocca", Milan, Italy.
| | | | - Filippo Alongi
- Department of Radiation Oncology, Sacro Cuore Don Calabria Cancer Care Center, Negrar-Verona, and University of Brescia, Brescia, Italy
| | - Cynthia Aristei
- Radiation Oncology Section, Department of Surgical and Biomedical Science, University of Perugia, Perugia General Hospital, Perugia, Italy
| | - Carlotta Becherini
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Firenze, Italy
| | - Liliana Belgioia
- Department of Radiation Oncology, Ospedale Policlinico San Martino and University of Genoa, Genoa, Italy
| | - Michela Buglione
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Luciana Caravatta
- Department of Radiation Oncology, SS. Annunziata Hospital, G. D'Annunzio University of Chieti, Chieti, Italy
| | | | | | - Michele Fiore
- Radiotherapy Unit, Campus Bio-Medico University, Rome, Italy
| | - Domenico Genovesi
- Department of Radiation Oncology, SS. Annunziata Hospital, G. D'Annunzio University of Chieti, Chieti, Italy
| | - Carlo Greco
- Radiotherapy Unit, Campus Bio-Medico University, Rome, Italy
| | - Lorenzo Livi
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Firenze, Italy
| | - Stefano Maria Magrini
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Giulia Marvaso
- Deparment of Radiation Oncology of IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Rosario Mazzola
- Department of Radiation Oncology, Sacro Cuore Don Calabria Cancer Care Center, Negrar-Verona, and University of Brescia, Brescia, Italy
| | - Icro Meattini
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Firenze, Italy
| | - Anna Merlotti
- Department of Radiation Oncology, S. Croce and Carle Teaching Hospital, Cuneo, Italy
| | - Isabella Palumbo
- Radiation Oncology Section, Department of Surgical and Biomedical Science, University of Perugia, Perugia General Hospital, Perugia, Italy
| | - Stefano Pergolizzi
- Department of Biomedical Sciences and Morphological and Functional Images, University of Messina, Italy
| | - Sara Ramella
- Radiotherapy Unit, Campus Bio-Medico University, Rome, Italy
| | | | - Elvio Russi
- Department of Radiation Oncology, S. Croce and Carle Teaching Hospital, Cuneo, Italy
| | - Marco Trovò
- Department of Radiation Oncology, Azienda Sanitaria Universitaria Integrata of Udine, Udine, Italy
| | - Alessandro Sindoni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Vincenzo Valentini
- Gemelli Advanced Radiation Therapy Center, Fondazione Policlinico Universitario "A. Gemelli", Catholic University of Sacred Heart, Rome, Italy
| | - Renzo Corvò
- Department of Radiation Oncology, Ospedale Policlinico San Martino and University of Genoa, Genoa, Italy
| |
Collapse
|
18
|
Fahim Golestaneh A, Lecker LSM, Schlegel J, Nowrouzi A, Schwager C, Meister S, Weichert W, Debus J, Abdollahi A. Large scale in vivo micro-RNA loss of function screen identified miR-29a, miR-100 and miR-155 as modulators of radioresistance and tumor-stroma communication. Int J Cancer 2019; 144:2774-2781. [PMID: 30478850 DOI: 10.1002/ijc.32019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 11/06/2018] [Accepted: 11/13/2018] [Indexed: 12/25/2022]
Abstract
Micro RNAs (miR) are master regulators of cellular transcriptome. We aimed to investigate the role of miR regulation on tumor radiosensitivity and development of local tumor recurrence by a novel large-scale in vivo loss of function screen. For stable miR silencing, human A431 tumor cells were transduced with lentiviral constructs against 170 validated human miR (miRzip library). Fractionated radiotherapy (5x6Gy) was applied to A431 miRzip library growing s.c. in NCr nude mice. Enrichment of miRZip and miR expression was assessed using multiplexed qRT-PCR. The modulatory effect of miR on tumor and tumor microenvironment response to ionizing radiation was further evaluated by clonogenic survival, apoptosis (Caspase 3/7), DNA double-strand breaks (DSB, nuclear γH2AX foci), tumor microvessel density (MVD), transcriptome and protein analysis. Fractionated irradiation of the A431 miRzip library led to regression of tumors. However, after a latency period, tumors ultimately progressed and formed local recurrences indicating the survival of a subpopulation of miRzip expressing tumor clones. Among the selected miR for subsequent validation studies, loss of miR-29a, miR-100 and miR-155 was found to enhance clonogenic survival, reduce apoptosis and residual γH2AX foci of irradiated tumor cells. Moreover, knockdown of miR increased tumor angiogenesis correlating with elevated VEGF and TGFα expression levels. This phenomenon was most evident after tumor irradiation in vivo suggesting a critical role for tumor-stroma communication in development of the radioresistant phenotype. Engineering radioresistant tumors in vivo by modulating miR expression may lead to identification of critical targets for conquering local therapy failure.
Collapse
Affiliation(s)
- Azadeh Fahim Golestaneh
- Division of Molecular & Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital (UKHD), Germany.,German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
| | - Laura S M Lecker
- Division of Molecular & Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital (UKHD), Germany.,German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
| | - Julian Schlegel
- Division of Molecular & Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital (UKHD), Germany.,German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
| | - Ali Nowrouzi
- Division of Molecular & Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital (UKHD), Germany.,German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
| | - Christian Schwager
- Division of Molecular & Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital (UKHD), Germany.,German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
| | - Sarah Meister
- Division of Molecular & Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital (UKHD), Germany.,German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
| | - Wilko Weichert
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Jürgen Debus
- Division of Molecular & Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital (UKHD), Germany.,German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
| | - Amir Abdollahi
- Division of Molecular & Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital (UKHD), Germany.,German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
| |
Collapse
|
19
|
Wrona A, Dziadziuszko R, Jassem J. Management of brain metastases in non-small cell lung cancer in the era of tyrosine kinase inhibitors. Cancer Treat Rev 2018; 71:59-67. [DOI: 10.1016/j.ctrv.2018.10.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 10/11/2018] [Accepted: 10/19/2018] [Indexed: 01/16/2023]
|
20
|
Pogue BW, Wilson BC. Optical and x-ray technology synergies enabling diagnostic and therapeutic applications in medicine. JOURNAL OF BIOMEDICAL OPTICS 2018; 23:1-17. [PMID: 30350489 PMCID: PMC6197862 DOI: 10.1117/1.jbo.23.12.121610] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/24/2018] [Indexed: 05/10/2023]
Abstract
X-ray and optical technologies are the two central pillars for human imaging and therapy. The strengths of x-rays are deep tissue penetration, effective cytotoxicity, and the ability to image with robust projection and computed-tomography methods. The major limitations of x-ray use are the lack of molecular specificity and the carcinogenic risk. In comparison, optical interactions with tissue are strongly scatter dominated, leading to limited tissue penetration, making imaging and therapy largely restricted to superficial or endoscopically directed tissues. However, optical photon energies are comparable with molecular energy levels, thereby providing the strength of intrinsic molecular specificity. Additionally, optical technologies are highly advanced and diversified, being ubiquitously used throughout medicine as the single largest technology sector. Both have dominant spatial localization value, achieved with optical surface scanning or x-ray internal visualization, where one often is used with the other. Therapeutic delivery can also be enhanced by their synergy, where radio-optical and optical-radio interactions can inform about dose or amplify the clinical therapeutic value. An emerging trend is the integration of nanoparticles to serve as molecular intermediates or energy transducers for imaging and therapy, requiring careful design for the interaction either by scintillation or Cherenkov light, and the nanoscale design is impacted by the choices of optical interaction mechanism. The enhancement of optical molecular sensing or sensitization of tissue using x-rays as the energy source is an important emerging field combining x-ray tissue penetration in radiation oncology with the molecular specificity and packaging of optical probes or molecular localization. The ways in which x-rays can enable optical procedures, or optics can enable x-ray procedures, provide a range of new opportunities in both diagnostic and therapeutic medicine. Taken together, these two technologies form the basis for the vast majority of diagnostics and therapeutics in use in clinical medicine.
Collapse
Affiliation(s)
- Brian W. Pogue
- Dartmouth College, Thayer School of Engineering, Geisel School of Medicine, Hanover, New Hampshire, United States
| | - Brian C. Wilson
- University of Toronto, Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
21
|
Liu B, Han D, Zhang T, Cheng G, Lu Y, Wang J, Zhao H, Zhao Z. Hypoxia-induced autophagy promotes EGFR loss in specific cell contexts, which leads to cell death and enhanced radiosensitivity. Int J Biochem Cell Biol 2018; 111:12-18. [PMID: 30278227 DOI: 10.1016/j.biocel.2018.09.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 09/19/2018] [Accepted: 09/20/2018] [Indexed: 10/28/2022]
Abstract
Treatment failure through radioresistance of tumors is associated with activation of the epidermal growth factor receptor (EGFR). Tumor cell proliferation, DNA-repair, hypoxia and metastases-formation are four mechanisms in which EGFR signaling has an important role. However, the effect of hypoxia on EGFR expression is still controversial. In this study, we demonstrated that hypoxia enhanced EGFR expression and sustained cell survival in SiHa, CAL 27 and A549 cells at both low and high cell desnities, while in MCF-7, MDA-MB-231 and HeLa cells, EGFR and cell survival were regulated by hypoxic treatment in a cell-density dependent manner: upregulated at low cell density and downregulated at high cell density. In MCF-7 and HeLa xenografts in nude mice, EGFR expression varied inversely with the pimonidazole level that was used as an indicator of hypoxia, accordant with the effect of hypoxia at high cell density in vitro. Hypoxia induced more remarkable cell autophagy at high cell density than at low cell density. Autophagy inhibitor 3MA, rather than proteasome inhibitor MG132 inhibited hypoxia-mediated EGFR loss and shifted cell death to cell survival in HeLa cells. The MCF7 cells' sensitivity to ionizing radiation (IR) under hypoxia was also conditional on the cell densities when the hypoxia treatment was introduced, inversely associated with the expression levels of EGFR. Altogether, hypoxia can decrease EGFR expression in some cell lines by enhancing autophagy at high cell density, leading to cell death and hypersensitivity to radiotherapy. This study may help to understand how hypoxia influences EGFR expression and radiosensitivity.
Collapse
Affiliation(s)
- Baocai Liu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Dongmei Han
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Tingting Zhang
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Guanghui Cheng
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China.
| | - Yinliang Lu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Jinbao Wang
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Hongfu Zhao
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Zhipeng Zhao
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| |
Collapse
|
22
|
Bhattacharya P, Shetake NG, Pandey BN, Kumar A. Receptor tyrosine kinase signaling in cancer radiotherapy and its targeting for tumor radiosensitization. Int J Radiat Biol 2018; 94:628-644. [DOI: 10.1080/09553002.2018.1478160] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Poushali Bhattacharya
- Radiation Signaling and Cancer Biology Section, Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Neena G. Shetake
- Radiation Signaling and Cancer Biology Section, Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Badri N. Pandey
- Radiation Signaling and Cancer Biology Section, Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Amit Kumar
- Radiation Signaling and Cancer Biology Section, Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| |
Collapse
|
23
|
Yomo S, Oda K. Impacts of EGFR-mutation status and EGFR-TKI on the efficacy of stereotactic radiosurgery for brain metastases from non-small cell lung adenocarcinoma: A retrospective analysis of 133 consecutive patients. Lung Cancer 2018; 119:120-126. [DOI: 10.1016/j.lungcan.2018.03.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/12/2018] [Accepted: 03/13/2018] [Indexed: 12/15/2022]
|
24
|
Pinto C, Di Bisceglie M, Di Fabio F, Bochicchio A, Latiano T, Cordio S, Rosati G, Aschele C, Marino A, Bergamo F, Bustreo S, Frassineti L, Ciardiello F, Damato A, Giaquinta S, Baldari D, Boni L. Phase II Study of Preoperative Treatment with External Radiotherapy Plus Panitumumab in Low-Risk, Locally Advanced Rectal Cancer (RaP Study/STAR-03). Oncologist 2018. [PMID: 29523646 DOI: 10.1634/theoncologist.2017-0484] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Treatment with fluoropyrimidines and concomitant long-course external radiotherapy (RTE) is the standard of care in locally advanced rectal cancer (LARC) preoperative chemoradiation. A randomized phase II study (RaP/STAR-03) was conducted that aimed to evaluate the activity and safety of the monoclonal antibody anti-epidermal growth factor receptor panitumumab as a single agent in combination with radiotherapy in low-risk LARC preoperative treatment. MATERIALS AND METHODS Patients had adenocarcinoma of the mid-low rectum, cT3N- or cT2-T3N+, KRAS wild-type status, and negative circumferential radial margin. Panitumumab was administered concomitant to RTE. Rectal surgery was performed 6-8 weeks after the end of preoperative treatment. The adjuvant chemotherapy regimen was FOLFOX. The primary endpoint was the pathologic complete response (pCR) rate. The sample size was calculated using Simon's two-stage design. A pCR of 16% was considered to qualify the experimental treatment for further testing. RESULTS Ninety-eight patients were enrolled in 13 Italian centers from October 2012 to October 2015. Three panitumumab infusions were administered in 92 (93.4%) patients. The RTE compliance was median dose 50.4 Gy; ≥28 fractions in 82 (83.7%) patients. Surgical treatment was performed in 92 (93.9%) patients, and no severe intraoperative complications were observed. A pCR was observed in 10 (10.9%) patients (95% confidence interval, 4.72%-17.07%). Pathological downstaging occurred in 45 (45.9%) patients. Grade 3 toxicities were observed in 22 (22.3%) patients, and the common adverse events were skin rash in 16 (16.3%) patients. No grade 4 toxicities were reported. CONCLUSION The pCR rate (our primary endpoint), at only 10.9%, did not reach the specified level considered suitable for further testing. However, the analysis showed a good toxicity profile and compliance to concomitant administration of panitumumab and RTE in preoperative treatment of LARC. The pCR evaluation in all wild-type RAS is ongoing. IMPLICATIONS FOR PRACTICE The aim of the RaP/STAR-03 study was to evaluate the activity and safety of monoclonal antibody anti-epidermal growth factor receptor (EGFR) panitumumab as a single agent without chemotherapy in low-risk, locally advanced rectal cancer (LARC) preoperative treatment. Nevertheless, the use of panitumumab in combination with radiotherapy in preoperative treatment in patients with KRAS wild type and low-risk LARC did not reach the pathologic complete response primary endpoint. This study showed a good toxicity profile and compliance to combination treatment. Further analysis of NRAS and BRAF on tissue and circulating levels of the EGFR ligands and vascular factors (soluble vascular endothelial growth factor, E-selectin) may provide insight on the potential molecular pathways involved in the anti-EGFR response.
Collapse
Affiliation(s)
- Carmine Pinto
- Medical Oncology Unit, Clinical Cancer Centre, IRCCS-Arcispedale S. Maria Nuova, Reggio Emilia, Italy
| | | | | | | | - Tiziana Latiano
- Medical Oncology Unit, IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Stefano Cordio
- Medical Oncology Unit, Ospedale Garibaldi, Catania, Italy
| | - Gerardo Rosati
- Medical Oncology Unit, Ospedale San Carlo, Potenza, Italy
| | - Carlo Aschele
- Medical Oncology Unit, Ospedale Civile, La Spezia, Italy
| | | | - Francesca Bergamo
- Medical Oncology Unit 1, Istituto Oncologico Veneto IOV - IRCCS, Padova, Italy
| | - Sara Bustreo
- Medical Oncology Unit, IRST-IRCCS, Meldola, Italy
| | - Luca Frassineti
- Medical Oncology Unit, AOU Città della Salute e della Scienza, Turin, Italy
| | | | - Angela Damato
- Medical Oncology Unit, Clinical Cancer Centre, IRCCS-Arcispedale S. Maria Nuova, Reggio Emilia, Italy
| | | | | | - Luca Boni
- Clinical Trial Center, Policlinico Careggi, Florence, Italy
| |
Collapse
|
25
|
Moncharmont C, Guy JB, Wozny AS, Gilormini M, Battiston-Montagne P, Ardail D, Beuve M, Alphonse G, Simoëns X, Rancoule C, Rodriguez-Lafrasse C, Magné N. Carbon ion irradiation withstands cancer stem cells' migration/invasion process in Head and Neck Squamous Cell Carcinoma (HNSCC). Oncotarget 2018; 7:47738-47749. [PMID: 27374096 PMCID: PMC5216975 DOI: 10.18632/oncotarget.10281] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 05/28/2016] [Indexed: 12/12/2022] Open
Abstract
Cancer Stem Cells (CSCs) in Head and Neck Squamous Cell Carcinoma (HNSCC) have extremely aggressive profile (high migratory and invasive potential). These characteristics can explain their resistance to conventional treatment. Efficacy of photon and carbon ion irradiation with addition of cetuximab (5 nM) is studied on clonogenic death, migration and invasion of two HNSCC populations: SQ20B and SQ20B/CSCs. SQ20B express E-cadherin and overexpress EGFR while SQ20B/CSCs express N-cadherin and low EGFR. Cetuximab strongly inhibits SQ20B proliferation but has no effect on SQ20B/CSCs. 2 Gy photon irradiation enhances migration and invasiveness in both populations (p < 0.05), while cetuximab only stops SQ20B migration (p < 0.005). Carbon irradiation significantly inhibits invasion in both populations (p < 0.05), and the association with cetuximab significantly inhibits invasion in both populations (p < 0.005). These results highlight CSCs characteristics: EGFRLow, cetuximab-resistant, and highly migratory. Carbon ion irradiation appears to be a very promising therapeutic modality counteracting migration/invasion process in both parental cells and CSCs in contrast to photon irradiation.
Collapse
Affiliation(s)
- Coralie Moncharmont
- Université Lyon 1, Faculté de Médecine-Lyon-Sud, Oullins, 69921, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, Institut de Physique Nucléaire de Lyon, IPNL, Villeurbanne, 69622, France.,Département de Radiothérapie, Institut de Cancérologie de la Loire - Lucien Neuwirth, St Priest en Jarez, 42270, France
| | - Jean-Baptiste Guy
- Université Lyon 1, Faculté de Médecine-Lyon-Sud, Oullins, 69921, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, Institut de Physique Nucléaire de Lyon, IPNL, Villeurbanne, 69622, France.,Département de Radiothérapie, Institut de Cancérologie de la Loire - Lucien Neuwirth, St Priest en Jarez, 42270, France
| | - Anne-Sophie Wozny
- Université Lyon 1, Faculté de Médecine-Lyon-Sud, Oullins, 69921, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, Institut de Physique Nucléaire de Lyon, IPNL, Villeurbanne, 69622, France.,Hospices Civils de Lyon, Lyon, 69229, France
| | - Marion Gilormini
- Université Lyon 1, Faculté de Médecine-Lyon-Sud, Oullins, 69921, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, Institut de Physique Nucléaire de Lyon, IPNL, Villeurbanne, 69622, France
| | - Priscilla Battiston-Montagne
- Université Lyon 1, Faculté de Médecine-Lyon-Sud, Oullins, 69921, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, Institut de Physique Nucléaire de Lyon, IPNL, Villeurbanne, 69622, France
| | - Dominique Ardail
- Université Lyon 1, Faculté de Médecine-Lyon-Sud, Oullins, 69921, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, Institut de Physique Nucléaire de Lyon, IPNL, Villeurbanne, 69622, France.,Hospices Civils de Lyon, Lyon, 69229, France
| | - Michael Beuve
- Institut de Physique Nucléaire de Lyon, IPNL, Villeurbanne, 69622, France
| | - Gersende Alphonse
- Laboratoire de Radiobiologie Cellulaire et Moléculaire, Institut de Physique Nucléaire de Lyon, IPNL, Villeurbanne, 69622, France.,Hospices Civils de Lyon, Lyon, 69229, France
| | - Xavier Simoëns
- Département de Pharmacologie Clinique et d'Innovation, Institut de Cancérologie de la Loire - Lucien Neuwirth, St Priest en Jarez, 42270, France
| | - Chloé Rancoule
- Département de Radiothérapie, Institut de Cancérologie de la Loire - Lucien Neuwirth, St Priest en Jarez, 42270, France
| | - Claire Rodriguez-Lafrasse
- Université Lyon 1, Faculté de Médecine-Lyon-Sud, Oullins, 69921, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, Institut de Physique Nucléaire de Lyon, IPNL, Villeurbanne, 69622, France.,Hospices Civils de Lyon, Lyon, 69229, France
| | - Nicolas Magné
- Université Lyon 1, Faculté de Médecine-Lyon-Sud, Oullins, 69921, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, Institut de Physique Nucléaire de Lyon, IPNL, Villeurbanne, 69622, France.,Département de Radiothérapie, Institut de Cancérologie de la Loire - Lucien Neuwirth, St Priest en Jarez, 42270, France
| |
Collapse
|
26
|
Carbonic Anhydrase IX (CAIX), Cancer, and Radiation Responsiveness. Metabolites 2018; 8:metabo8010013. [PMID: 29439394 PMCID: PMC5874614 DOI: 10.3390/metabo8010013] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/06/2018] [Accepted: 02/07/2018] [Indexed: 12/23/2022] Open
Abstract
Carbonic anhydrase IX has been under intensive investigation as a therapeutic target in cancer. Studies demonstrate that this enzyme has a key role in pH regulation in cancer cells, allowing these cells to adapt to the adverse conditions of the tumour microenviroment. Novel CAIX inhibitors have shown efficacy in both in vitro and in vivo pre-clinical cancer models, adversely affecting cell viability, tumour formation, migration, invasion, and metastatic growth when used alone. In co-treatments, CAIX inhibitors may enhance the effects of anti-angiogenic drugs or chemotherapy agents. Research suggests that these inhibitors may also increase the response of tumours to radiotherapy. Although many of the anti-tumour effects of CAIX inhibition may be dependent on its role in pH regulation, recent work has shown that CAIX interacts with several of the signalling pathways involved in the cellular response to radiation, suggesting that pH-independent mechanisms may also be an important basis of its role in tumour progression. Here, we discuss these pH-independent interactions in the context of the ability of CAIX to modulate the responsiveness of cancer to radiation.
Collapse
|
27
|
Yang Z, Tam KY. Combination Strategies Using EGFR-TKi in NSCLC Therapy: Learning from the Gap between Pre-Clinical Results and Clinical Outcomes. Int J Biol Sci 2018; 14:204-216. [PMID: 29483838 PMCID: PMC5821041 DOI: 10.7150/ijbs.22955] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 12/09/2017] [Indexed: 01/04/2023] Open
Abstract
Although epidermal growth factor receptor (EGFR) inhibitors have been used to treat non-small cell lung cancer (NSCLC) for decades with great success in patients with EGFR mutations, acquired resistance inevitably occurs after long-term exposure. More recently, combination therapy has emerged as a promising strategy to overcome this issue. Several experiments have been carried out to evaluate the synergism of combination therapy both in vitro and in vivo. Additionally, many clinical studies have been carried out to investigate the feasibility of treatment with EGFR-tyrosine kinase inhibitors (TKi) combined with other NSCLC treatments, including radiotherapy, cytotoxic chemotherapies, targeted therapies, and emerging immunotherapies. However, a significant gap still exists when applying pre-clinical results to clinical scenarios, which hinders the development and use of these strategies. This article is a literature review analysing the rationalities and controversies in the transition from pre-clinical investigation to clinical practice associated with various combination strategies. It also highlights clues and challenges regarding future combination therapeutic options in NSCLC treatment.
Collapse
Affiliation(s)
| | - Kin Yip Tam
- Faculty of Health Science, University of Macau, Taipa, Macau, China
| |
Collapse
|
28
|
Kriegs M, Kasten-Pisula U, Riepen B, Hoffer K, Struve N, Myllynen L, Braig F, Binder M, Rieckmann T, Grénman R, Petersen C, Dikomey E, Rothkamm K. Radiosensitization of HNSCC cells by EGFR inhibition depends on the induction of cell cycle arrests. Oncotarget 2018; 7:45122-45133. [PMID: 27281611 PMCID: PMC5216710 DOI: 10.18632/oncotarget.9161] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 04/18/2016] [Indexed: 12/28/2022] Open
Abstract
The increase in cellular radiosensitivity by EGF receptor (EGFR) inhibition has been shown to be attributable to the induction of a G1-arrest in p53-proficient cells. Because EGFR targeting in combination with radiotherapy is used to treat head and neck squamous cell carcinomas (HNSCC) which are predominantly p53 mutated, we tested the effects of EGFR targeting on cellular radiosensitivity, proliferation, apoptosis, DNA repair and cell cycle control using a large panel of HNSCC cell lines. In these experiments EGFR targeting inhibited signal transduction, blocked proliferation and induced radiosensitization but only in some cell lines and only under normal (pre-plating) conditions. This sensitization was not associated with impaired DNA repair (53BP1 foci) or induction of apoptosis. However, it was associated with the induction of a lasting G2-arrest. Both, the radiosensitization and the G2-arrest were abrogated if the cells were re-stimulated (delayed plating) with actually no radiosensitization being detectable in any of the 14 tested cell lines. Therefore we conclude that EGFR targeting can induce a reversible G2 arrest in p53 deficient HNSCC cells, which does not consequently result in a robust cellular radiosensitization. Together with recent animal and clinical studies our data indicate that EGFR inhibition is no effective strategy to increase the radiosensitivity of HNSCC cells.
Collapse
Affiliation(s)
- Malte Kriegs
- Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg - Eppendorf, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, 20246 Hamburg, Germany
| | - Ulla Kasten-Pisula
- Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg - Eppendorf, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, 20246 Hamburg, Germany
| | - Britta Riepen
- Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg - Eppendorf, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, 20246 Hamburg, Germany
| | - Konstantin Hoffer
- Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg - Eppendorf, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, 20246 Hamburg, Germany
| | - Nina Struve
- Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg - Eppendorf, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, 20246 Hamburg, Germany
| | - Laura Myllynen
- Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg - Eppendorf, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, 20246 Hamburg, Germany
| | - Friederike Braig
- Department of Oncology and Hematology, BMT with section Pneumology, University Medical Center Hamburg - Eppendorf, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, 20246 Hamburg, Germany
| | - Mascha Binder
- Department of Oncology and Hematology, BMT with section Pneumology, University Medical Center Hamburg - Eppendorf, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, 20246 Hamburg, Germany
| | - Thorsten Rieckmann
- Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg - Eppendorf, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, 20246 Hamburg, Germany.,Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Hamburg - Eppendorf, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, 20246 Hamburg, Germany
| | - Reidar Grénman
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Turku and Turku University Hospital, 20521 Turku, Finland
| | - Cordula Petersen
- Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg - Eppendorf, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, 20246 Hamburg, Germany
| | - Ekkehard Dikomey
- Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg - Eppendorf, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, 20246 Hamburg, Germany
| | - Kai Rothkamm
- Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg - Eppendorf, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, 20246 Hamburg, Germany
| |
Collapse
|
29
|
Koi L, Löck S, Linge A, Thurow C, Hering S, Baumann M, Krause M, Gurtner K. EGFR-amplification plus gene expression profiling predicts response to combined radiotherapy with EGFR-inhibition: A preclinical trial in 10 HNSCC-tumour-xenograft models. Radiother Oncol 2017; 124:496-503. [PMID: 28807520 DOI: 10.1016/j.radonc.2017.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/03/2017] [Accepted: 07/06/2017] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND PURPOSE Improvement of the results of radiotherapy by EGFR inhibitors is modest, suggesting significant intertumoural heterogeneity of response. To identify potential biomarkers, a preclinical trial was performed on ten different human squamous cell carcinoma xenografts of the head and neck (HNSCC) studying in vivo and ex vivo the effect of fractionated irradiation and EGFR inhibition. Local tumour control and tumour growth delay were correlated with potential biomarkers, e.g. EGFR gene amplification and radioresponse-associated gene expression profiles. MATERIAL AND METHODS Local tumour control 120days after end of irradiation was determined for fractionated radiotherapy alone (30f, 6weeks) or after simultaneous EGFR-inhibition with cetuximab. The EGFR gene amplification status was determined using FISH. Gene expression analyses were performed using an in-house gene panel. RESULTS Six out of 10 investigated tumour models showed a significant increase in local tumour control for the combined treatment of cetuximab and fractionated radiotherapy compared to irradiation alone. For 3 of the 6 responding tumour models, an amplification of the EGFR gene could be demonstrated. Gene expression profiling of untreated tumours revealed significant differences between amplified and non-amplified tumours as well as between responder and non-responder tumours to combined radiotherapy and cetuximab. CONCLUSION The EGFR amplification status, in combination with gene expression profiling, may serve as a predictive biomarker for personalized interventional strategies regarding combined treatment of cetuximab and fractionated radiotherapy and should, as a next step, be clinically validated.
Collapse
Affiliation(s)
- Lydia Koi
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Germany
| | - Steffen Löck
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Annett Linge
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), partner site Dresden, Germany
| | - Cedric Thurow
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany
| | - Sandra Hering
- Institute for Legal Medicine, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Michael Baumann
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), partner site Dresden, Germany
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Germany; German Cancer Consortium (DKTK), partner site Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), partner site Dresden, Germany
| | - Kristin Gurtner
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), partner site Dresden, Germany.
| |
Collapse
|
30
|
Higgins GS, Krause M, McKenna WG, Baumann M. Personalized Radiation Oncology: Epidermal Growth Factor Receptor and Other Receptor Tyrosine Kinase Inhibitors. Recent Results Cancer Res 2017; 198:107-22. [PMID: 27318683 DOI: 10.1007/978-3-662-49651-0_5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Molecular biomarkers are currently evaluated in preclinical and clinical studies in order to establish predictors for treatment decisions in radiation oncology. The receptor tyrosine kinases (RTK) are described in the following text. Among them, the most data are available for the epidermal growth factor receptor (EGFR) that plays a major role for prognosis of patients after radiotherapy, but seems also to be involved in mechanisms of radioresistance, specifically in repopulation of tumour cells between radiotherapy fractions. Monoclonal antibodies against the EGFR improve locoregional tumour control and survival when applied during radiotherapy, however, the effects are heterogeneous and biomarkers for patient selection are warranted. Also other RTK´s such as c-Met and IGF-1R seem to play important roles in tumour radioresistance. Beside the potential to select patients for molecular targeting approaches combined with radiotherapy, studies are also needed to evluate radiotherapy adaptation approaches for selected patients, i.e. adaptation of radiation dose, or, more sophisticated, of target volumes.
Collapse
Affiliation(s)
- Geoff S Higgins
- Gray Laboratories, Department of Oncology, Cancer Research UK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology (NCRO), Carl Gustav Carus Faculty of Medicine, University Hospital, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.
- German Cancer Consortium (DKTK) Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Helmholtz-Zentrum Dresden-Rossendorf, Insititute of Radiooncology, Dresden, Germany.
- Department of Radiation Oncology, Carl Gustav Carus Faculty of Medicine, University Hospital, Technische Universität Dresden, Dresden, Germany.
| | - W Gillies McKenna
- Gray Laboratories, Department of Oncology, Cancer Research UK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Michael Baumann
- OncoRay - National Center for Radiation Research in Oncology (NCRO), Carl Gustav Carus Faculty of Medicine, University Hospital, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK) Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Insititute of Radiooncology, Dresden, Germany
- Department of Radiation Oncology, Carl Gustav Carus Faculty of Medicine, University Hospital, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
31
|
Samson P, Lockhart AC. Biologic therapy in esophageal and gastric malignancies: current therapies and future directions. J Gastrointest Oncol 2017; 8:418-429. [PMID: 28736629 PMCID: PMC5506284 DOI: 10.21037/jgo.2016.11.13] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/27/2016] [Indexed: 12/14/2022] Open
Abstract
Biologic agents, including targeted antibodies as well as immunomodulators, are demonstrating unparalleled development and study across the entire spectrum of human malignancy. This review summarizes the current state of biologic therapies for esophageal, esophagogastric, and gastric malignancies, including those that target human epidermal growth factor receptor 2 (HER2), epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), c-Met, mechanistic target of rapamycin (mTOR) and immunomodulators. We focus primarily on agents that have been included in phase II and III clinical trials in locally advanced, progressive, or metastatic esophageal and gastric malignancies. At this time, only two biologic therapies are recommended by the National Comprehensive Cancer Network (NCCN): trastuzumab for patients with esophageal/esophagogastric or gastric adenocarcinomas with HER2 overexpression and ramucirumab, a VEGFR-2 inhibitor, as a second-line therapy for metastatic disease. However, recent reports of increases in overall and progression-free survival for agents including pertuzumab, apatinib, and pembrolizumab will likely increase the use of targeted biologic therapy in clinical practice for esophageal and gastric malignancies.
Collapse
Affiliation(s)
- Pamela Samson
- Division of Cardiothoracic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - A. Craig Lockhart
- Division of Oncology, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
32
|
Xu H, Zong H, Ma C, Ming X, Shang M, Li K, He X, Du H, Cao L. Epidermal growth factor receptor in glioblastoma. Oncol Lett 2017; 14:512-516. [PMID: 28693199 PMCID: PMC5494611 DOI: 10.3892/ol.2017.6221] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 03/21/2017] [Indexed: 12/11/2022] Open
Abstract
Mutations in the epidermal growth factor receptor (EGFR) are commonly occurring in glioblastoma. Enhanced activation of EGFR can occur through a variety of different mechanisms, both ligand-dependent and ligand-independent. Numerous evidence has suggested that EGFR is overexpressed in most of primary glioblastomas and some of the secondary glioblastomas and is characteristic of more aggressive glioblastoma phenotypes. Additionally, recent studies have revealed that wild-type EGFR, and to a greater extent hyper-activating EGFR mutants induced a substantial upregulation of Fyn expression. Furthermore, it was determined that Fyn expression is upregulated across a panel of patient-derived glioblastoma stem cells (GSCs) relative to normal progenitor controls. Moreover, researchers are continuously involved in elucidation of novel mechanism linking EGFR EGFR-expressing glioblastoma. The present review highlights current aspects of EGFR receptor in glioblastoma and concludes that the concept of EGFR signaling and related receptors and associated factors is evolving, however, it needs detailed evaluation for future clinical applications in cancer patients.
Collapse
Affiliation(s)
- Hongsheng Xu
- Department of Neurosurgery, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| | - Hailiang Zong
- Department of Neurosurgery, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| | - Chong Ma
- Department of Neurosurgery, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| | - Xing Ming
- Department of Neurosurgery, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| | - Ming Shang
- Department of Neurosurgery, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| | - Kai Li
- Department of Neurosurgery, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| | - Xiaoguang He
- Department of Neurosurgery, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| | - Hai Du
- Department of Neurosurgery, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| | - Lei Cao
- Department of Neurosurgery, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| |
Collapse
|
33
|
Couñago F, Rodríguez A, Calvo P, Luna J, Monroy JL, Taboada B, Díaz V, Rodríguez de Dios N. Targeted therapy combined with radiotherapy in non-small-cell lung cancer: a review of the Oncologic Group for the Study of Lung Cancer (Spanish Radiation Oncology Society). Clin Transl Oncol 2017; 19:31-43. [PMID: 27106020 DOI: 10.1007/s12094-016-1512-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 04/09/2016] [Indexed: 02/06/2023]
Abstract
In recent years, major advances in our understanding of the molecular biology of lung cancer, together with significant improvements in radiotherapy technologies, have revolutionized the treatment of non-small cell lung cancer (NSCLC). This has led to the development of new therapies that target molecular mutations specific to each tumor type, acting on the cell surface antigens or intracellular signaling pathways, or directly affecting cell survival. At the same time, ablative dose radiotherapy can be delivered safely in the context of metastatic disease. In this article, the GOECP/SEOR (Oncological Group for Study of Lung Cancer/Spanish Society of Radiation Oncology) reviews the role of new targeted therapies used in combination with radiotherapy in patients with locally advanced (stage III) NSCLC and in patients with advanced, metastatic (stage IV) NSCLC.
Collapse
Affiliation(s)
- F Couñago
- Department of Radiation Oncology, Hospital Universitario Quirón, Calle Diego de Velázquez, 1, 28223, Pozuelo de Alarcón, Madrid, Spain
| | - A Rodríguez
- Department of Radiation Oncology, Hospital Ruber Internacional, Calle de la Masó, 38, 28034, Madrid, Spain
| | - P Calvo
- Department of Radiation Oncology, Hospital Universitario Santiago de Compostela, Travesía de Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - J Luna
- Department of Radiation Oncology, Hospital Universitario Fundación Jiménez Díaz, Avda. Reyes Católicos, 2, 28040, Madrid, Spain
| | - J L Monroy
- Department of Radiation Oncology, Hospital Universitario La Ribera, Carretera de Corbera, km 1, 46600, Alzira, Valencia, Spain
| | - B Taboada
- Department of Radiation Oncology, Hospital Universitario Santiago de Compostela, Travesía de Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - V Díaz
- Department of Radiation Oncology, Hospital Universitario Puerta del Mar, Av. Ana de Viya, 21, 11009, Cádiz, Spain
| | - N Rodríguez de Dios
- Department of Radiation Oncology, Hospital de la Esperanza, Parc de Salut Mar, Barcelona, Spain.
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.
- Universitat Pompeu Fabra, San josé de la Montaña 12, 08024, Barcelona, Spain.
| |
Collapse
|
34
|
Alongi F, Arcangeli S, Ramella S, Giaj-Levra N, Borghetti P, D'angelillo R, Ricchetti F, Maddalo M, Mazzola R, Trovò M, Russi E, Magrini SM. From chemotherapy to target therapies associated with radiation in the treatment of NSCLC: a durable marriage? Expert Rev Anticancer Ther 2016; 17:157-165. [PMID: 27984919 DOI: 10.1080/14737140.2017.1273111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION The integration between radiotherapy and drugs, from chemotherapy to recently available target therapies, continues to have a relevant role in the treatment of locally advanced and metastatic Non-small cell lung cancer (NSCLC). Aim of the present review is to evaluate the promising and emerging application of the best interaction between new drugs and new modalities of radiotherapy. Areas covered: We searched Medline, Google Scholar, PubMed, ProQuest Dissertation, and Theses databases for reports published in English. A study was included when it reported on cancer-related radiotherapy and included patients with NSCLC treated with chemo and/or target therapies. Review articles were excluded from the analysis. Expert commentary: Chemo-radiotherapy still represents the standard of choice in locally advanced NSCLC, while to date the addition of target therapies to chemo-radiotherapy did not demonstrate any robust advantage in this stage of disease. Considering the absence of randomized controlled trials, the role of target therapies in early stage adjuvant NSCLC is not yet recommended in clinical practice. On the contrary, in the setting of oligometastatic and oligoprogressive disease, new molecules demonstrated to be safe and effective, opening to a promising and emerging application of the best interaction between new drugs and new modalities of radiotherapy.
Collapse
Affiliation(s)
- Filippo Alongi
- a Radiation Oncology , Sacro Cuore Don Calabria Cancer Care Center , Negrar, Verona , Italy
| | - Stefano Arcangeli
- b Radiation Oncology , San Camillo and Forlanini Hospital , Rome , Italy
| | - Sara Ramella
- c Department of Radiotherapy , Campus Bio-Medico University , Rome , Italy
| | - Niccolò Giaj-Levra
- a Radiation Oncology , Sacro Cuore Don Calabria Cancer Care Center , Negrar, Verona , Italy
| | - Paolo Borghetti
- d Department of Radiation Oncology , Spedali Civili of Brescia , Brescia , Italy
| | | | - Francesco Ricchetti
- a Radiation Oncology , Sacro Cuore Don Calabria Cancer Care Center , Negrar, Verona , Italy
| | - Marta Maddalo
- d Department of Radiation Oncology , Spedali Civili of Brescia , Brescia , Italy
| | - Rosario Mazzola
- a Radiation Oncology , Sacro Cuore Don Calabria Cancer Care Center , Negrar, Verona , Italy
| | - Marco Trovò
- e Radiation Oncology Department , CRO Aviano , Aviano , Italy
| | - Elvio Russi
- f Department of Radiation Oncology , S. Croce University Hospital , Cuneo , Italy
| | | | | |
Collapse
|
35
|
Gargioni E, Schulz F, Raabe A, Burdak-Rothkamm S, Rieckmann T, Rothkamm K. Targeted nanoparticles for tumour radiotherapy enhancement-the long dawn of a golden era? ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:523. [PMID: 28151534 DOI: 10.21037/atm.2016.12.46] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Despite considerable progress in (I) our understanding of the aetiopathology of head and neck cancer and (II) the precise delivery of radiotherapy, long-term survival rates for many patients with head and neck cancer remain disappointingly low. Over the past years, gold nanoparticles (NP) have emerged as promising radiation dose enhancers. In a recent study published in Nanoscale, Popovtzer et al. have used gold NP coated with an antibody against the epidermal growth factor receptor (EGFR) in an attempt to enhance radiation-induced tumour cell killing in a head and neck cancer xenograft model. They report a significant impact of the combined treatment with radiation and gold NP on tumour growth and suggest an involvement of apoptosis, inhibition of angiogenesis and diminished tissue repair. In this perspective, we illustrate the underlying radiobiophysical concepts and discuss some of the challenges associated with this and related nanoparticle-radiotherapy studies from a physics, chemistry, biology and therapy angle. We conclude that strong interdisciplinary collaborations spanning all these areas are crucially important to proceed towards effective cancer treatment with gold NP "from bench to bedside".
Collapse
Affiliation(s)
- Elisabetta Gargioni
- Department of Radiotherapy and Radio-Oncology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Florian Schulz
- Institute for Physical Chemistry, University of Hamburg, Hamburg, Germany
| | - Annette Raabe
- Department of Radiotherapy and Radio-Oncology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | | | - Thorsten Rieckmann
- Department of Radiotherapy and Radio-Oncology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Kai Rothkamm
- Department of Radiotherapy and Radio-Oncology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| |
Collapse
|
36
|
Economopoulou P, Mountzios G. Non-small cell lung cancer (NSCLC) and central nervous system (CNS) metastases: role of tyrosine kinase inhibitors (TKIs) and evidence in favor or against their use with concurrent cranial radiotherapy. Transl Lung Cancer Res 2016; 5:588-598. [PMID: 28149754 DOI: 10.21037/tlcr.2016.12.06] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Central nervous system (CNS) metastases, including brain metastases (BM) and leptomeningeal metastases (LM) represent a frequent complication of non-small cell lung cancer (NSCLC). Patients with BM comprise a heterogeneous group, with a median survival that ranges from 3 to 14 months. However, in the majority of patients, the occurrence of CNS metastases is usually accompanied by severe morbidity and substantial deterioration in quality of life. Local therapies, such as whole brain radiotherapy (WBRT), stereotactic radiosurgery (SRS) or surgical resection, either alone or as part of a multimodality treatment are available treatment strategies for BM and the choice of therapy varies depending on patient group and prognosis. Meanwhile, introduction of tyrosine kinase inhibitors (TKIs) in clinical practice has led to individualization of therapy based upon the presence of the exact abnormality, resulting in a major therapeutic improvement in patients with NSCLC who harbor epidermal growth factor receptor (EGFR) activating mutations or anaplastic lymphoma kinase (ALK) gene rearrangements, respectively. Based on their clinical activity in systemic disease, such molecular agents could offer the promise of improved BM control without substantial toxicity; however, their role in combination with radiotherapy is controversial. In this review, we discuss the controversy regarding the use of TKIs in combination with radiotherapy and illustrate future perspectives in the treatment of BM in NSCLC.
Collapse
Affiliation(s)
| | - Giannis Mountzios
- Department of Medical Oncology, 251 Airforce General Hospital, Athens, Greece
| |
Collapse
|
37
|
Dal Pra A, Locke JA, Borst G, Supiot S, Bristow RG. Mechanistic Insights into Molecular Targeting and Combined Modality Therapy for Aggressive, Localized Prostate Cancer. Front Oncol 2016; 6:24. [PMID: 26909338 PMCID: PMC4754414 DOI: 10.3389/fonc.2016.00024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 01/22/2016] [Indexed: 12/12/2022] Open
Abstract
Radiation therapy (RT) is one of the mainstay treatments for prostate cancer (PCa). The potentially curative approaches can provide satisfactory results for many patients with non-metastatic PCa; however, a considerable number of individuals may present disease recurrence and die from the disease. Exploiting the rich molecular biology of PCa will provide insights into how the most resistant tumor cells can be eradicated to improve treatment outcomes. Important for this biology-driven individualized treatment is a robust selection procedure. The development of predictive biomarkers for RT efficacy is therefore of utmost importance for a clinically exploitable strategy to achieve tumor-specific radiosensitization. This review highlights the current status and possible opportunities in the modulation of four key processes to enhance radiation response in PCa by targeting the: (1) androgen signaling pathway; (2) hypoxic tumor cells and regions; (3) DNA damage response (DDR) pathway; and (4) abnormal extra-/intracell signaling pathways. In addition, we discuss how and which patients should be selected for biomarker-based clinical trials exploiting and validating these targeted treatment strategies with precision RT to improve cure rates in non-indolent, localized PCa.
Collapse
Affiliation(s)
- Alan Dal Pra
- Radiation Medicine Program, Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Jennifer A Locke
- Radiation Medicine Program, Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Gerben Borst
- Radiation Medicine Program, Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Stephane Supiot
- Integrated Center of Oncology (ICO) René Gauducheau , Nantes , France
| | - Robert G Bristow
- Radiation Medicine Program, Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
38
|
Huang S, Peter Rodemann H, Harari PM. Molecular Targeting of Growth Factor Receptor Signaling in Radiation Oncology. Recent Results Cancer Res 2016; 198:45-87. [PMID: 27318681 DOI: 10.1007/978-3-662-49651-0_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Ionizing radiation has been shown to activate and interact with multiple growth factor receptor pathways that can influence tumor response to therapy. Among these receptor interactions, the epidermal growth factor receptor (EGFR) has been the most extensively studied with mature clinical applications during the last decade. The combination of radiation and EGFR-targeting agents using either monoclonal antibody (mAb) or small-molecule tyrosine kinase inhibitor (TKI) offers a promising approach to improve tumor control compared to radiation alone. Several underlying mechanisms have been identified that contribute to improved anti-tumor capacity after combined treatment. These include effects on cell cycle distribution, apoptosis, tumor cell repopulation, DNA damage/repair, and impact on tumor vasculature. However, as with virtually all cancer drugs, patients who initially respond to EGFR-targeted agents may eventually develop resistance and manifest cancer progression. Several potential mechanisms of resistance have been identified including mutations in EGFR and downstream signaling molecules, and activation of alternative member-bound tyrosine kinase receptors that bypass the inhibition of EGFR signaling. Several strategies to overcome the resistance are currently being explored in preclinical and clinical models, including agents that target the EGFR T790 M resistance mutation or target multiple EGFR family members, as well as agents that target other receptor tyrosine kinase and downstream signaling sites. In this chapter, we focus primarily on the interaction of radiation with anti-EGFR therapies to summarize this promising approach and highlight newly developing opportunities.
Collapse
Affiliation(s)
- Shyhmin Huang
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue K4/336 CSC, Madison, WI, 53792, USA
- Department of Human Oncology, University of Wisconsin Comprehensive Cancer Center, WIMR 3136, 1111 Highland Ave Madison, Madison, WI, 53705, USA
| | - H Peter Rodemann
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tübingen, Röntgenweg, 72076, Tübingen, Germany
| | - Paul M Harari
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue K4/336 CSC, Madison, WI, 53792, USA.
| |
Collapse
|
39
|
Swaminath A, Wright JR, Tsakiridis TK, Ung YC, Pond GR, Sur R, Corbett TB, Okawara G, Levine MN. A Phase II Trial of Erlotinib and Concurrent Palliative Thoracic Radiation for Patients With Non-Small-Cell Lung Cancer. Clin Lung Cancer 2015; 17:142-9. [PMID: 26521036 DOI: 10.1016/j.cllc.2015.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 09/25/2015] [Accepted: 09/29/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND The downstream signaling pathways of the epidermal growth factor receptor might influence radiation resistance. Data from preclinical work support the hypothesis that erlotinib concurrent with radiation therapy (RT) might increase cancer cell killing. The present trial was designed to examine the efficacy and toxicity of combined erlotinib and palliative chest thoracic RT in non-small-cell lung cancer (NSCLC). MATERIALS AND METHODS Patients with newly diagnosed stage III-IV (American Joint Committee on Cancer, version 6) or recurrent NSCLC received 3 weeks of erlotinib at a dose of 150 mg daily, starting 1 week before palliative thoracic RT to 30 Gy in 10 fractions within 2 weeks. The primary outcome was a change in the quality of life, as measured by the Lung Cancer Symptom Scale (LCSS) question on the "symptoms of lung cancer" from baseline to 4 weeks after treatment. RESULTS A total of 40 patients were recruited from 2 institutions. Of the 40 patients, 22 (55%) were men, with an average age of 71 years, and 60% had stage IV disease. A total of 26 patients (65%) completed the full course of erlotinib, and 35 (88%) completed the planned RT. Twenty-five patients (62.5%) reported LCSS scores at 4 weeks after treatment, with an average change (improvement) of -12.5 U (95% confidence interval, -23.0 to -1.9; 2P = .023). This was less than the a priori hypothesis of a change of -17.5 U. The median overall and progression-free survival was 5.2 and 3.2 months, respectively. CONCLUSION The present single-arm, phase II trial did not demonstrate additional symptomatic benefit from concurrent erlotinib therapy with standard palliative thoracic RT for patients with locally advanced or metastatic NSCLC.
Collapse
Affiliation(s)
- Anand Swaminath
- Department of Oncology, McMaster University, Hamilton, Ontario, Canada; Juravinski Cancer Centre, Hamilton Health Sciences, Hamilton, Ontario, Canada.
| | - James R Wright
- Department of Oncology, McMaster University, Hamilton, Ontario, Canada; Juravinski Cancer Centre, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Theodoros K Tsakiridis
- Department of Oncology, McMaster University, Hamilton, Ontario, Canada; Juravinski Cancer Centre, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Yee C Ung
- Odette Cancer Centre and University of Toronto, Toronto, Ontario, Canada
| | - Gregory R Pond
- Department of Oncology, McMaster University, Hamilton, Ontario, Canada; Ontario Clinical Oncology Group, Juravinski Hospital, Hamilton, Ontario, Canada
| | - Ranjan Sur
- Department of Oncology, McMaster University, Hamilton, Ontario, Canada; Juravinski Cancer Centre, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Thomas B Corbett
- Department of Oncology, McMaster University, Hamilton, Ontario, Canada; Juravinski Cancer Centre, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Gordon Okawara
- Department of Oncology, McMaster University, Hamilton, Ontario, Canada; Juravinski Cancer Centre, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Mark N Levine
- Department of Oncology, McMaster University, Hamilton, Ontario, Canada; Juravinski Cancer Centre, Hamilton Health Sciences, Hamilton, Ontario, Canada; Ontario Clinical Oncology Group, Juravinski Hospital, Hamilton, Ontario, Canada
| |
Collapse
|
40
|
Myllynen L, Kwiatkowski M, Gleißner L, Riepen B, Hoffer K, Wurlitzer M, Petersen C, Dikomey E, Rothkamm K, Schlüter H, Kriegs M. Quantitative proteomics unveiled: Regulation of DNA double strand break repair by EGFR involves PARP1. Radiother Oncol 2015; 116:423-30. [DOI: 10.1016/j.radonc.2015.09.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 08/19/2015] [Accepted: 09/14/2015] [Indexed: 11/29/2022]
|
41
|
Adding Erlotinib to Chemoradiation Improves Overall Survival but Not Progression-Free Survival in Stage III Non-Small Cell Lung Cancer. Int J Radiat Oncol Biol Phys 2015; 92:317-24. [PMID: 25968826 PMCID: PMC4432249 DOI: 10.1016/j.ijrobp.2015.02.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 01/27/2015] [Accepted: 02/02/2015] [Indexed: 01/10/2023]
Abstract
Background Concurrent chemoradiotherapy is the standard of care for inoperable stage III non-small cell lung cancer (NSCLC) for patients who can tolerate it. We explored if adding erlotinib would increase the effectiveness of chemoradiotherapy without increasing toxicity in a single-arm prospective phase II trial. Methods Forty-eight patients with previously untreated NSCLC received intensity-modulated radiation therapy (63 Gy/35 fractions) on Monday–Friday, with chemotherapy (paclitaxel 45 mg/m2, carboplatin AUC=2) on Mondays, for 7 weeks. All patients also received the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor erlotinib (150 mg orally 1/day) on Tuesday–Sunday for 7 weeks followed by consolidation paclitaxel–carboplatin. The primary endpoint was time to progression; secondary endpoints were overall survival (OS), toxicity, response, and disease control and whether any endpoint differed by EGFR mutation status. Results Of 46 patients evaluable for response, 40 were former or never-smokers and 41 were evaluable for EGFR mutations (37 wild-type [wt] and 4 [mutated; all adenocarcinoma]). Median time to progression was 14.0 months and did not differ by EGFR status. Toxicity was acceptable (no grade 5, one grade 4, eleven grade 3). Twelve patients (26%) had complete responses (10 wt, 2 mutated), 27 (59%) partial (21 wt, 2 mutated, 4 unknown), and 7 (15%) none (6 wt, 2 mutated, 1 unknown) (P=0.610). At 37.0 months’ follow-up (range 3.6–76.5 months) for all patients, median OS time was 36.5 months and 1-, 2-, and 5-year OS rates were 82.6%, 67.4%, and 35.9%; none differed by mutation status. Twelve patients had no progression and 34 had local and/or distant failure. Eleven of 27 distant failures were in the brain (7 wt, 3 mutated, 1 unknown). Conclusions Toxicity and OS were promising, but time to progression did not meet expectations. The prevalence of distant failures underscores the need for effective systemic therapy.
Collapse
|
42
|
Schaue D, McBride WH. Opportunities and challenges of radiotherapy for treating cancer. Nat Rev Clin Oncol 2015; 12:527-40. [PMID: 26122185 DOI: 10.1038/nrclinonc.2015.120] [Citation(s) in RCA: 503] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The past 20 years have seen dramatic changes in the delivery of radiation therapy, but the impact of radiobiology on the clinic has been far less substantial. A major consideration in the use of radiotherapy has been on how best to exploit differences between the tumour and host tissue characteristics, which in the past has been achieved empirically by radiation-dose fractionation. New advances are uncovering some of the mechanistic processes that underlie this success story. In this Review, we focus on how these processes might be targeted to improve the outcome of radiotherapy at the individual patient level. This approach would seem a more productive avenue of treatment than simply trying to increase the radiation dose delivered to the tumour.
Collapse
Affiliation(s)
- Dörthe Schaue
- Department of Radiation Oncology, Room B3-109, Center for Health Sciences, Westwood, University of California, Los Angeles (UCLA), Los Angeles, CA 90095-1714, USA
| | - William H McBride
- Department of Radiation Oncology, Room B3-109, Center for Health Sciences, Westwood, University of California, Los Angeles (UCLA), Los Angeles, CA 90095-1714, USA
| |
Collapse
|
43
|
Teng K, Zhang Y, Hu X, Ding Y, Gong R, Liu L. Nimotuzumab enhances radiation sensitivity of NSCLC H292 cells in vitro by blocking epidermal growth factor receptor nuclear translocation and inhibiting radiation-induced DNA damage repair. Onco Targets Ther 2015; 8:809-18. [PMID: 25926742 PMCID: PMC4403694 DOI: 10.2147/ott.s77283] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background The epidermal growth factor receptor (EGFR) signaling pathway plays a significant role in radiation resistance. There is evidence that EGFR nuclear translocation is associated with DNA double-strand breaks (DSB) repair. Nimotuzumab has shown the effect of radiosensitization in various cancer cells, but little is known about the relationship between nimotuzumab and EGFR nuclear translocation in non-small cell lung cancer (NSCLC) cell lines. In this study, we selected two NSCLC cell lines, namely, H292 (with high EGFR expression) and H1975 (with low EGFR expression) and explored the mechanisms underlying radiation sensitivity. Methods MTT assay, clonogenic survival assay, and flow cytometry were performed separately to test cell viability, radiation sensitivity, cell cycle distribution, and apoptosis. Protein γ-H2AX, DNA-PK/p-DNA-PK, and EGFR/p-EGFR expression were further compared both in the cytoplasm and the nucleus with the western blot. Results Nimotuzumab reduced the viability of H292 cells and sensitized H292 cells to ionizing radiation. The radiation sensitivity enhancement ratio (SER) was 1.304 and 1.092 for H292 and H1975 cells, respectively. H292 cells after nimotuzumab administration were arrested at the G0/G1 phase in response to radiation. Apoptosis was without statistical significance in both cell lines. γ-H2AX formation in the combination group (nimotuzumab and radiation) increased both in the cytoplasm and the nucleus along with the decreased expression of nuclear EGFR/p-EGFR and p-DNA-PK in H292 cells (P<0.05) that was more significant than that in H1975 cells. Conclusion Our research revealed a possible mechanism to explain the radiosensitivity in H292 cells. Nimotuzumab decreased the radiation-induced activation of DNA-PK by blocking EGFR nuclear translocation and impairing DNA DSB repair, thus enhancing radiosensitivity in H292 cells. Because these results represent early research, the matters of how γ-H2AX and DNA-PK dynamically change simultaneously with nuclear EGFR and the best time to administer nimotuzumab will require further exploration.
Collapse
Affiliation(s)
- Kai Teng
- Department of Thoracic Oncology, Cancer Center of Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China ; Department of Radiation Oncology, Hainan Cancer Hospital, Haikou, Hainan, People's Republic of China
| | - Yong Zhang
- Department of Thoracic Oncology, Cancer Center of Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Xiaoyan Hu
- Department of Thoracic Oncology, Cancer Center of Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yihui Ding
- Department of Thoracic Oncology, Cancer Center of Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Rui Gong
- Department of Thoracic Oncology, Cancer Center of Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Li Liu
- Department of Thoracic Oncology, Cancer Center of Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
44
|
Tsai YC, Ho PY, Tzen KY, Tuan TF, Liu WL, Cheng AL, Pu YS, Cheng JCH. Synergistic Blockade of EGFR and HER2 by New-Generation EGFR Tyrosine Kinase Inhibitor Enhances Radiation Effect in Bladder Cancer Cells. Mol Cancer Ther 2015; 14:810-20. [PMID: 25589492 DOI: 10.1158/1535-7163.mct-13-0951] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 12/27/2014] [Indexed: 11/16/2022]
Abstract
Blockade of EGFR has been proved useful in enhancing the effect of radiotherapy, but the advantages of new-generation EGFR tyrosine kinase inhibitors (TKI) in radiosensitization are not well known. We used two human bladder cancer cells with wild-type EGFR to study the synergism between irradiation and afatinib (an EGFR/HER2 dual kinase inhibitor) or erlotinib (an EGFR kinase inhibitor). Here, we showed that afatinib has better radiosensitizing effect than erlotinib in increasing cancer cell killing, the percentage of apoptotic cells, and DNA damage. Afatinib is also superior to erlotinib in combining radiation to decrease tumor size, inhibit glucose metabolism, and enhance apoptotic proteins in vivo. Finally, erlotinib suppressed cell growth and induced more DNA damage in bladder cancer cells transfected with HER2 shRNA, but not in control vector-treated cells. In conclusion, concomitant blockade of radiation-activated EGFR and HER2 signaling by a new-generation EGFR TKI better inhibits the growth of bladder cancer cells both in vitro and in vivo. The absence of radiosensitization by EGFR inhibition alone and the greater radiosensitizing effect of EGFR inhibitor in HER2 knocked down cells suggest the synergism between HER2 and EGFR in determining radiosensitivity. The regained radiosensitizing activity of erlotinib implies that with proper HER2 inhibition, EGFR tyrosine kinase is still a potential target to enhance radiotherapy effect in these seemingly unresponsive bladder cancer cells.
Collapse
Affiliation(s)
- Yu-Chieh Tsai
- Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Pei-Yin Ho
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Kai-Yuan Tzen
- Department of Nuclear Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan. Molecular Imaging Center, National Taiwan University, Taipei, Taiwan
| | - Tsung-Fan Tuan
- Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wei-Lin Liu
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ann-Lii Cheng
- Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan. Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan. Cancer Research Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yeong-Shiau Pu
- Department of Urology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan. Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Jason Chia-Hsien Cheng
- Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan. Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan. Cancer Research Center, National Taiwan University College of Medicine, Taipei, Taiwan. Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
45
|
JUNG JOOHEE, JEONG SEONGYUN, PARK SEOKSOON, SHIN SEOLHWA, JU EUNJIN, CHOI JINHYANG, PARK JAESOOK, LEE JAEHEE, KIM INKI, SUH YOUNGAH, HWANG JUNGJIN, KURODA SHUN, LEE JUNGSHIN, SONG SIYEOL, CHOI EUNKYUNG. A cisplatin-incorporated liposome that targets the epidermal growth factor receptor enhances radiotherapeutic efficacy without nephrotoxicity. Int J Oncol 2014; 46:1268-74. [DOI: 10.3892/ijo.2014.2806] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 12/09/2014] [Indexed: 11/06/2022] Open
|
46
|
Gurtner K, Ebert N, Pfitzmann D, Eicheler W, Zips D, Baumann M, Krause M. Effect of combined irradiation and EGFR/Erb-B inhibition with BIBW 2992 on proliferation and tumour cure in cell lines and xenografts. Radiat Oncol 2014; 9:261. [PMID: 25444177 PMCID: PMC4271482 DOI: 10.1186/s13014-014-0261-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 11/12/2014] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND AND PURPOSE In previous experiments an enhanced anti-proliterative effect of the EGFR/ErbB tyrosine kinase inhibitor (TKI) BIBW 2992 with single dose irradiation was observed in FaDu tumour xenografts. Aim of the present experiment was to determine if this effect can also be seen in combination with a fractionated radiotherapy. Secondly we investigate the efficacy of BIBW 2992 on local tumour control for UT-SCC-15. MATERIAL AND METHODS Tumour pieces of FaDu, UT-SCC-14, A431, UT-SCC-15 (squamous cell carcinomas) and A7 (glioma) tumour models were transplanted onto the right hind leg of NMRI (nu/nu) nude mice. For evaluation of tumour growth mice were either treated daily orally with BIBW 2992 (30 mg/kg body weight), or carrier up to a final tumour size of 15 mm or with a fractionated radiotherapy (15f/15d, 30 Gy) with simultaneous application of BIBW 2992 or carrier. For local tumour control UT-SCC-15 tumours were treated with a fractionated radiotherapy (30f/6weeks) or received 30f/6 weeks in combination with daily orally BIBW 2992 (22.5 mg/kg b.w.) during RT. RESULTS A significant effect on tumour growth time was observed in all tumour models for BIBW 2992 application alone. However, substantial intertumoural heterogeneity could be seen. In the UT-SCC-14, UT-SCC-15 and A431 tumour models a total regression of the tumours and no recurrence during treatment time (73 days) were determined where as for the A7 tumour only a slight effect was noticeable. For the combined treatment of fractionated radiotherapy (15f/15d) and BIBW 2992 administration a significant effect on tumour growth time was seen compared to irradiation alone for A7, UT-SCC-15 and A431 (ER 1.2 - 3.7), this advantage could not be demonstrated for FaDu and UT-SCC-14. However, the local tumour control was not altered for the UT-SCC-15 tumour model when adding BIBW 2992 to fractionated irradiation (30f/6weeks). CONCLUSION A heterogeneous effect on tumour growth time of BIBW 2992 alone as well as in combination with fractionated irradiation could be demonstrated for all tumour models. However, the significant effect on tumour growth time did not translate into an improvement of local tumour control for the UT-SCC-15 tumour model.
Collapse
Affiliation(s)
- Kristin Gurtner
- Department of Radiation Oncology, UniversityHospital C.G. Carus, Fetscherstr. 74, 01307, Dresden, Germany. .,OncoRay - National Centerfor Radiation Research in Oncology, Medical Faculty and University Hospital Carl Gustav Carus, TechnischeUniversität and Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany.
| | - Nadja Ebert
- Department of Radiation Oncology, UniversityHospital C.G. Carus, Fetscherstr. 74, 01307, Dresden, Germany. .,OncoRay - National Centerfor Radiation Research in Oncology, Medical Faculty and University Hospital Carl Gustav Carus, TechnischeUniversität and Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany.
| | - Dorothee Pfitzmann
- Department of Radiation Oncology, UniversityHospital C.G. Carus, Fetscherstr. 74, 01307, Dresden, Germany. .,OncoRay - National Centerfor Radiation Research in Oncology, Medical Faculty and University Hospital Carl Gustav Carus, TechnischeUniversität and Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany.
| | - Wolfgang Eicheler
- Department of Radiation Oncology, UniversityHospital C.G. Carus, Fetscherstr. 74, 01307, Dresden, Germany. .,OncoRay - National Centerfor Radiation Research in Oncology, Medical Faculty and University Hospital Carl Gustav Carus, TechnischeUniversität and Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany.
| | - Daniel Zips
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany.
| | - Michael Baumann
- Department of Radiation Oncology, UniversityHospital C.G. Carus, Fetscherstr. 74, 01307, Dresden, Germany. .,OncoRay - National Centerfor Radiation Research in Oncology, Medical Faculty and University Hospital Carl Gustav Carus, TechnischeUniversität and Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany. .,German Cancer consortium (DKTK) Dresden and German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany. .,Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany.
| | - Mechthild Krause
- Department of Radiation Oncology, UniversityHospital C.G. Carus, Fetscherstr. 74, 01307, Dresden, Germany. .,OncoRay - National Centerfor Radiation Research in Oncology, Medical Faculty and University Hospital Carl Gustav Carus, TechnischeUniversität and Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany. .,German Cancer consortium (DKTK) Dresden and German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany. .,Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany.
| |
Collapse
|
47
|
Preoperative chemoradiation therapy in combination with panitumumab for patients with resectable esophageal cancer: the PACT study. Int J Radiat Oncol Biol Phys 2014; 90:190-6. [PMID: 25195993 DOI: 10.1016/j.ijrobp.2014.05.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 05/10/2014] [Accepted: 05/19/2014] [Indexed: 11/22/2022]
Abstract
PURPOSE Preoperative chemoradiation therapy (CRT) has become the standard treatment strategy for patients with resectable esophageal cancer. This multicenter phase 2 study investigated the efficacy of the addition of the epidermal growth factor receptor (EGFR) inhibitor panitumumab to a preoperative CRT regimen with carboplatin, paclitaxel, and radiation therapy in patients with resectable esophageal cancer. METHODS AND MATERIALS Patients with resectable cT1N1M0 or cT2-3N0 to -2M0 tumors received preoperative CRT consisting of panitumumab (6 mg/kg) on days 1, 15, and 29, weekly administrations of carboplatin (area under the curve [AUC] = 2), and paclitaxel (50 mg/m(2)) for 5 weeks and concurrent radiation therapy (41.4 Gy in 23 fractions, 5 days per week), followed by surgery. Primary endpoint was pathologic complete response (pCR) rate. We aimed at a pCR rate of more than 40%. Furthermore, we explored the predictive value of biomarkers (EGFR, HER 2, and P53) for pCR. RESULTS From January 2010 until December 2011, 90 patients were enrolled. Patients were diagnosed predominantly with adenocarcinoma (AC) (80%), T3 disease (89%), and were node positive (81%). Three patients were not resected due to progressive disease. The primary aim was unmet, with a pCR rate of 22%. Patients with AC and squamous cell carcinoma reached a pCR of 14% and 47%, respectively. R0 resection was achieved in 95% of the patients. Main grade 3 toxicities were rash (12%), fatigue (11%), and nonfebrile neutropenia (11%). None of the biomarkers was predictive for response. CONCLUSIONS The addition of panitumumab to CRT with carboplatin and paclitaxel was safe and well tolerated but could not improve pCR rate to the preset criterion of 40%.
Collapse
|
48
|
Zarschler K, Prapainop K, Mahon E, Rocks L, Bramini M, Kelly PM, Stephan H, Dawson KA. Diagnostic nanoparticle targeting of the EGF-receptor in complex biological conditions using single-domain antibodies. NANOSCALE 2014; 6:6046-6056. [PMID: 24777583 DOI: 10.1039/c4nr00595c] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
For effective localization of functionalized nanoparticles at diseased tissues such as solid tumours or metastases through biorecognition, appropriate targeting vectors directed against selected tumour biomarkers are a key prerequisite. The diversity of such vector molecules ranges from proteins, including antibodies and fragments thereof, through aptamers and glycans to short peptides and small molecules. Here, we analyse the specific nanoparticle targeting capabilities of two previously suggested peptides (D4 and GE11) and a small camelid single-domain antibody (sdAb), representing potential recognition agents for the epidermal growth factor receptor (EGFR). We investigate specificity by way of receptor RNA silencing techniques and look at increasing complexity in vitro by introducing increasing concentrations of human or bovine serum. Peptides D4 and GE11 proved problematic to employ and conjugation resulted in non-receptor specific uptake into cells. Our results show that sdAb-functionalized particles can effectively target the EGFR, even in more complex bovine and human serum conditions where targeting specificity is largely conserved for increasing serum concentration. In human serum however, an inhibition of overall nanoparticle uptake is observed with increasing protein concentration. For highly affine targeting ligands such as sdAbs, targeting a receptor such as EGFR with low serum competitor abundance, receptor recognition function can still be partially realised in complex conditions. Here, we stress the value of evaluating the targeting efficiency of nanoparticle constructs in realistic biological milieu, prior to more extensive in vivo studies.
Collapse
Affiliation(s)
- K Zarschler
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, D-01328 Dresden, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Addeo R, Zappavigna S, Parlato C, Caraglia M. Erlotinib: early clinical development in brain cancer. Expert Opin Investig Drugs 2014; 23:1027-37. [PMID: 24836441 DOI: 10.1517/13543784.2014.918950] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Glioblastoma (GBM) is the most common brain cancer in adults. It is also, unfortunately, the most aggressive type and the least responsive to therapy. Overexpression of EGFR and/or EGFRvIII is frequently found in GBM and is frequently associated with the more malignant phenotype of the disease and a poor clinical outcome. EGFR-targeted therapy represents a promising anti-GBM therapy. Two EGFR kinase inhibitors, gefitinib and erlotinib have been tested in clinical trials for malignant gliomas. However, the clinical efficacy of EGFR-targeted therapy has been only modest in GBM patients. AREAS COVERED The authors provide an evaluation of erlotinib as a potential therapy for GBM. The authors highlight experiences drawn from clinical trials and discuss the challenges, which include the insufficient penetration through the blood-brain barrier (BBB) and chemoresistance. EXPERT OPINION Malignant brain tumours have a very complex signalling network that is not only driven by EGFR. This complexity dictates tumour sensitivity to EGFR-targeted therapies. Alternative kinase signalling pathways may be involved in parallel with the inhibited target, so that a single target's inactivation is not sufficient to block downstream oncogenic signalling. The use of nanocarriers offers many opportunities, such as the release of the drug to specific cells or tissues, together with the ability to overcome different biological barriers, like the BBB.
Collapse
|
50
|
Searle EJ, Illidge TM, Stratford IJ. Emerging opportunities for the combination of molecularly targeted drugs with radiotherapy. Clin Oncol (R Coll Radiol) 2014; 26:266-76. [PMID: 24602563 DOI: 10.1016/j.clon.2014.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 01/29/2014] [Accepted: 02/11/2014] [Indexed: 02/08/2023]
Abstract
Recent drug discovery developments in the field of small molecule targeted agents have led to much interest in combining these with radiotherapy. There are good preclinical data to suggest this approach worthy of investigation and in this review we discuss how this has translated into recent clinical trials. The outcome of clinical trials investigating radiotherapy/targeted drug combinations published in the last 5 years is discussed, as are trials in progress. The perceived future opportunities and challenges in the development of this exciting area are considered.
Collapse
Affiliation(s)
- E J Searle
- Manchester Pharmacy School, University of Manchester, Manchester, UK; Targeted Therapy Group, Institute of Cancer Sciences, University of Manchester, Manchester, UK.
| | - T M Illidge
- Targeted Therapy Group, Institute of Cancer Sciences, University of Manchester, Manchester, UK
| | - I J Stratford
- Manchester Pharmacy School, University of Manchester, Manchester, UK
| |
Collapse
|