1
|
Mousavikia SN, Darvish L, Firouzjaei AA, Toossi MTB, Azimian H. PI3K/AKT/mTOR Targeting in Colorectal Cancer Radiotherapy: A Systematic Review. J Gastrointest Cancer 2025; 56:52. [PMID: 39849185 DOI: 10.1007/s12029-024-01160-1] [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] [Accepted: 12/11/2024] [Indexed: 01/25/2025]
Abstract
BACKGROUND Radioresistance is a major challenge in the treatment of patients with colorectal cancer (CRC) and impairs the efficacy of radiotherapy. The PI3K/AKT/mTOR signaling pathway plays a critical role in CRC and contributes to the development of radioresistance. Accordingly, targeting this signaling pathway may be a promising strategy to improve oncotherapy. METHODS We performed a systematic search of Scopus, PubMed, Web of Science, Embase, and Medline databases. We included articles that investigated the effects of PI3K/AKT/mTOR pathway inhibitors on improving the efficacy of radiotherapy. RESULT Of the 32 articles included in our review, 27 were preclinical studies and 5 were clinical trials. We examined the effects of various signaling pathway inhibitors in combination with radiotherapy. While the efficacy of these therapies when used alone is limited, their combination is associated with reduced survival, induction of apoptosis, and cell cycle arrest, which may increase radiosensitivity. Despite the limited number of studies, this combination therapy has shown favorable treatment outcomes in patients with CRC. CONCLUSION PI3K/AKT/mTOR is a critical signaling pathway for cancer cell survival. By inhibiting this pathway, we can increase the efficacy of radiotherapy. These results provide valuable insights for the further development of research and clinical practice in the treatment of colorectal cancer.
Collapse
Affiliation(s)
- S N Mousavikia
- Medical Physics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - L Darvish
- Department of Radiology, Faculty of Paramedicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Mother and Child Welfare Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - A A Firouzjaei
- Bioinformatics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - M T Bahreyni Toossi
- Medical Physics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - H Azimian
- Medical Physics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Peng D, Liang M, Li L, Yang H, Fang D, Chen L, Guan B. Circ_BBS9 as an early diagnostic biomarker for lung adenocarcinoma: direct interaction with IFIT3 in the modulation of tumor immune microenvironment. Front Immunol 2024; 15:1344954. [PMID: 39139574 PMCID: PMC11320841 DOI: 10.3389/fimmu.2024.1344954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 07/02/2024] [Indexed: 08/15/2024] Open
Abstract
Background Introduction: Circular RNAs (circRNAs) have been identified as significant contributors to the development and advancement of cancer. The objective of this study was to examine the expression and clinical implications of circRNA circ_BBS9 in lung adenocarcinoma (LUAD), as well as its potential modes of action. Methods The expression of Circ_BBS9 was examined in tissues and cell lines of LUAD through the utilization of microarray profiling, quantitative real-time polymerase chain reaction (qRT-PCR), and western blot analysis. In this study, we assessed the impact of circ_BBS9 on the proliferation of LUAD cells, as well as its influence on ferroptosis and tumor formation. To analyze these effects, we employed CCK-8 assays and ferroptosis assays. The identification of proteins that interact with Circ_BBS9 was achieved through the utilization of RNA pull-down and mass spectrometry techniques. A putative regulatory network comprising circ_BBS9, miR-7150, and IFIT3 was established using bioinformatics study. The investigation also encompassed the examination of the correlation between the expression of IFIT3 and the invasion of immune cells. Results Circ_BBS9 was significantly downregulated in LUAD tissues and cell lines. Low circ_BBS9 expression correlated with poor prognosis. Functional experiments showed that circ_BBS9 overexpression inhibited LUAD cell proliferation and promoted ferroptosis in vitro and suppressed tumor growth in vivo. Mechanistically, circ_BBS9 was found to directly interact with IFIT3 and regulate its expression by acting as a sponge for miR-7150. Additionally, IFIT3 expression correlated positively with immune infiltration in LUAD. Conclusion Circ_BBS9 has been identified as a tumor suppressor in lung adenocarcinoma (LUAD) and holds promise as a diagnostic biomarker. The potential mechanism of action involves the modulation of ferroptosis and the immunological microenvironment through direct interaction with IFIT3 and competitive binding to miR-7150. The aforementioned findings offer new perspectives on the pathophysiology of LUAD and highlight circ_BBS9 as a potentially valuable target for therapeutic interventions.
Collapse
Affiliation(s)
- Daijun Peng
- Department of Pathology, Jinshan Branch of Shanghai Sixth People’s Hospital, Shanghai, China
| | - Mingyu Liang
- Department of Automation, Shanghai Jiao Tong University, Shanghai, China
| | - Lingyu Li
- Department of Pathology, Jinshan Branch of Shanghai Sixth People’s Hospital, Shanghai, China
| | - Haisheng Yang
- Department of Pathology, Jinshan Branch of Shanghai Sixth People’s Hospital, Shanghai, China
| | - Di Fang
- Department of Pathology, Jinshan Branch of Shanghai Sixth People’s Hospital, Shanghai, China
| | - Lingling Chen
- Department of Pathology, Jinshan Branch of Shanghai Sixth People’s Hospital, Shanghai, China
| | - Bing Guan
- Department of Pathology, Jinshan Branch of Shanghai Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
3
|
Rachamala HK, Madamsetty VS, Angom RS, Nakka NM, Dutta SK, Wang E, Mukhopadhyay D, Pal K. Targeting mTOR and survivin concurrently potentiates radiation therapy in renal cell carcinoma by suppressing DNA damage repair and amplifying mitotic catastrophe. J Exp Clin Cancer Res 2024; 43:159. [PMID: 38840237 PMCID: PMC11155143 DOI: 10.1186/s13046-024-03079-8] [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: 12/18/2023] [Accepted: 05/24/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) was historically considered to be less responsive to radiation therapy (RT) compared to other cancer indications. However, advancements in precision high-dose radiation delivery through single-fraction and multi-fraction stereotactic ablative radiotherapy (SABR) have led to better outcomes and reduced treatment-related toxicities, sparking renewed interest in using RT to treat RCC. Moreover, numerous studies have revealed that certain therapeutic agents including chemotherapies can increase the sensitivity of tumors to RT, leading to a growing interest in combining these treatments. Here, we developed a rational combination of two radiosensitizers in a tumor-targeted liposomal formulation for augmenting RT in RCC. The objective of this study is to assess the efficacy of a tumor-targeted liposomal formulation combining the mTOR inhibitor everolimus (E) with the survivin inhibitor YM155 (Y) in enhancing the sensitivity of RCC tumors to radiation. EXPERIMENTAL DESIGN We slightly modified our previously published tumor-targeted liposomal formulation to develop a rational combination of E and Y in a single liposomal formulation (EY-L) and assessed its efficacy in RCC cell lines in vitro and in RCC tumors in vivo. We further investigated how well EY-L sensitizes RCC cell lines and tumors toward radiation and explored the underlying mechanism of radiosensitization. RESULTS EY-L outperformed the corresponding single drug-loaded formulations E-L and Y-L in terms of containing primary tumor growth and improving survival in an immunocompetent syngeneic mouse model of RCC. EY-L also exhibited significantly higher sensitization of RCC cells towards radiation in vitro than E-L and Y-L. Additionally, EY-L sensitized RCC tumors towards radiation therapy in xenograft and murine RCC models. EY-L mediated induction of mitotic catastrophe via downregulation of multiple cell cycle checkpoints and DNA damage repair pathways could be responsible for the augmentation of radiation therapy. CONCLUSION Taken together, our study demonstrated the efficacy of a strategic combination therapy in sensitizing RCC to radiation therapy via inhibition of DNA damage repair and a substantial increase in mitotic catastrophe. This combination therapy may find its use in the augmentation of radiation therapy during the treatment of RCC patients.
Collapse
Affiliation(s)
- Hari K Rachamala
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA
| | - Vijay S Madamsetty
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA
- PolyARNA Therapeutics, One Kendal Square, Cambridge, MA, 01329, USA
| | - Ramcharan S Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA
| | - Naga M Nakka
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA
| | - Shamit Kumar Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA.
| | - Krishnendu Pal
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA.
| |
Collapse
|
4
|
Rachamala HK, Madamsetty VS, Angom RS, Nakka NM, Kumar Dutta S, Wang E, Mukhopadhyay D, Pal K. Targeting mTOR and Survivin Concurrently Potentiates Radiation Therapy in Renal Cell Carcinoma by Suppressing DNA Damage Repair and Amplifying Mitotic Catastrophe. RESEARCH SQUARE 2023:rs.3.rs-3770403. [PMID: 38196607 PMCID: PMC10775360 DOI: 10.21203/rs.3.rs-3770403/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Background Renal cell carcinoma (RCC) was historically considered to be less responsive to radiation therapy (RT) compared to other cancer indications. However, advancements in precision high-dose radiation delivery through single-fraction and multi-fraction stereotactic ablative radiotherapy (SABR) have led to better outcomes and reduced treatment-related toxicities, sparking renewed interest in using RT to treat RCC. Moreover, numerous studies have revealed that certain therapeutic agents including chemotherapies can increase the sensitivity of tumors to RT, leading to a growing interest in combining these treatments. Here, we developed a rational combination of two radiosensitizers in a tumor-targeted liposomal formulation for augmenting RT in RCC. The objective of this study is to assess the efficacy of a tumor-targeted liposomal formulation combining the mTOR inhibitor everolimus (E) with the survivin inhibitor YM155 (Y) in enhancing the sensitivity of RCC tumors to radiation. Experimental Design We slightly modified our previously published tumor-targeted liposomal formulation to develop a rational combination of E and Y in a single liposomal formulation (EY-L) and assessed its efficacy in RCC cell lines in vitro and in RCC tumors in vivo. We further investigated how well EY-L sensitizes RCC cell lines and tumors toward radiation and explored the underlying mechanism of radiosensitization. Results EY-L outperformed the corresponding single drug-loaded formulations E-L and Y-L in terms of containing primary tumor growth and improving survival in an immunocompetent syngeneic mouse model of RCC. EY-L also exhibited significantly higher sensitization of RCC cells towards radiation in vitro than E-L and Y-L. Additionally, EY-L sensitized RCC tumors towards radiation therapy in xenograft and murine RCC models. EY-L mediated induction of mitotic catastrophe via downregulation of multiple cell cycle checkpoints and DNA damage repair pathways could be responsible for the augmentation of radiation therapy. Conclusion Taken together, our study demonstrated the efficacy of a strategic combination therapy in sensitizing RCC to radiation therapy via inhibition of DNA damage repair and a substantial increase in mitotic catastrophe. This combination therapy may find its use in the augmentation of radiation therapy during the treatment of RCC patients.
Collapse
|
5
|
Non-Small Cell Lung Cancer Treatment with Molecularly Targeted Therapy and Concurrent Radiotherapy—A Review. Int J Mol Sci 2023; 24:ijms24065858. [PMID: 36982933 PMCID: PMC10052930 DOI: 10.3390/ijms24065858] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/13/2023] [Accepted: 03/13/2023] [Indexed: 03/22/2023] Open
Abstract
Lung cancer is the leading cause of death worldwide for both men and women. Surgery can be offered as a radical treatment at stages I and II and selected cases of stage III (III A). Whereas at more advanced stages, combined modalities of treatment are applied: radiochemotherapy (IIIB) and molecularly targeted treatment (small molecule tyrosine kinase inhibitors, VEGF receptor inhibitors, monoclonal antibodies, and immunological treatment with monoclonal antibodies). Combination treatment, composed of radiotherapy and molecular therapy, is increasingly employed in locally advanced and metastatic lung cancer management. Recent studies have indicated a synergistic effect of such treatment and modification of immune response. The combination of immunotherapy and radiotherapy may result in the enhancement of the abscopal effect. Anti-angiogenic therapy, in combination with RT, is associated with high toxicity and should be not recommended. In this paper, the authors discuss the role of molecular treatment and the possibility of its concurrent use with radiotherapy in non-small cell lung cancer (NSCLC).
Collapse
|
6
|
The PIK3CA-E545K-SIRT4 signaling axis reduces radiosensitivity by promoting glutamine metabolism in cervical cancer. Cancer Lett 2023; 556:216064. [PMID: 36646410 DOI: 10.1016/j.canlet.2023.216064] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/04/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
The mutation of glutamic acid 545 to lysine (E545K) in PIK3CA, as the most common missense mutation of this gene in various cancer types, is frequently observed in cervical cancer and has been shown to reduce cervical cancer radiosensitivity. However, the underlying mechanisms remain unclear. Here, we implicate the alterations of glutamine metabolism in PIK3CA-E545K-mediated radioresistance of cervical cancer. Specifically, PIK3CA mutation negatively regulated the expression of SIRT4 via the epigenetic regulator EP300 independently of the canonical mTORC1 pathway. PIK3CA-E545K-induced SIRT4 downregulation promoted cell proliferation, migration, and radiation-induced DNA repair and apoptosis, while SIRT4 overexpression reversed the radioresistance phenotype mediated by PIK3CA mutation. Mechanistically, SIRT4 modulated glutamine metabolism and thus cellular apoptosis by negatively regulating a glutamate pyruvate transaminase GPT1. Moreover, the PI3K inhibitor BYL719, but not mTOR inhibitors, exerted remarkable synergistic effects with radiotherapy by inhibiting glutamine metabolism in vitro and in vivo. Collectively, this study reveals the role of PIK3CA-E545K-SIRT4 axis in regulating glutamine metabolism and the radioresistance in cervical cancer, which provides a necessary preliminary basis for clinical research of PI3K inhibitors as radiosensitizing agents.
Collapse
|
7
|
Pierrard J, Deheneffe S, Longton E, Henry S, Van Houtte P, Remouchamps V. Radiation therapy-induced left vocal cord paralysis following lung stereotactic body radiation therapy: A case report and review of the literature. Cancer Radiother 2023; 27:69-74. [PMID: 35872055 DOI: 10.1016/j.canrad.2022.05.004] [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: 03/02/2022] [Revised: 05/02/2022] [Accepted: 05/09/2022] [Indexed: 02/06/2023]
Abstract
We report the case of a 50-year old women with an oncological history of metastatic breast carcinoma who underwent lung stereotactic body radiation therapy (SBRT) of 60Gy in 8 fractions for a left upper lobe metastatic lesion. Seven months later, she complains about hoarseness and weakness of voice. Tumoral relapse and other frequent etiologies were excluded. The diagnosis of radiation induced left recurrent laryngeal nerve paralysis causing left vocal cord paralysis (VCP) was made. The symptomatology did not improve till the disease progression and death of the patient 29 months after SBRT. VCP after lung SBRT is a rare adverse event that has not yet been well described in the medical literature.
Collapse
Affiliation(s)
- J Pierrard
- Department of Radiotherapy, CHU UCL Namur, Site Ste Elisabeth, Namur, Belgium.
| | - S Deheneffe
- Department of Radiotherapy, CHU UCL Namur, Site Ste Elisabeth, Namur, Belgium
| | - E Longton
- Department of Radiotherapy, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - S Henry
- Department of Medical Oncology, CHU UCL Namur, Site Ste Elisabeth, Namur, Belgium
| | - P Van Houtte
- Department of Radiotherapy, Institut Jules-Bordet, Université Libre Bruxelles, Brussels, Belgium
| | - V Remouchamps
- Department of Radiotherapy, CHU UCL Namur, Site Ste Elisabeth, Namur, Belgium
| |
Collapse
|
8
|
Yuan G, Hu B, Ma J, Zhang C, Xie H, Wei T, Yang Y, Ni B. Histone lysine methyltransferase
SETDB2
suppresses
NRF2
to restrict tumor progression and modulates chemotherapy sensitivity in lung adenocarcinoma. Cancer Med 2022; 12:7258-7272. [PMID: 36504353 PMCID: PMC10067124 DOI: 10.1002/cam4.5451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Aberrant epigenetic remodeling represents a molecular hallmark in lung adenocarcinoma (LUAD). We aim to investigate the biological roles of SETDB2 and its underlying associations with oxidative stress, providing therapeutic targets for individualized treatment of LUAD. METHODS Differential analysis was conducted via Limma package, and Kaplan-Meier analysis was performed with survival package. CCK-8, cell proliferation assay, transwell assay, and in vivo assays were conducted to assess the function of SETDB2. Western blot assay, RT-qPCR, and immunohistochemistry (IHC) were conducted to assess the expression levels of SETDB2/NRF2. Chromatin immunoprecipitation (ChIP) assay and ChIP-qPCR were conducted to assess the epigenetic roles of SETDB2. RESULTS We found that SETDB2 expression is decreased in tumor samples versus normal tissues in TCGA-LUAD cohort, LUAD-EAS cohort, GSE72094 dataset, and independent Soochow-LUAD dataset. Patients with low SETDB2 levels had a worse prognosis relative to those with high SETDB2. SETDB2 inhibition could significantly promote cell growth, migration ability, and stemness maintenance. Gene set enrichment analysis (GSEA) suggested that SETDB2 correlated with oxidative stress crosstalk and regulated NRF2 mRNA levels. ChIP assay suggested that SETDB2 mainly recruited the H3K9me3 enrichment at the NRF2 promoter region to suppress the mRNA levels of NRF2. Downregulated SETDB2 could activate NRF2 transcription and expression, thereby promoting its downstream targets, like NQO1, FTH1, and ME1. Functional experiments demonstrated that low SETDB2 allowed NRF2 to drive malignant processes of LUAD. SETDB2 overexpression attenuated the ability of NRF2 signaling to neutralize cellular reactive oxygen species (ROS) levels, leading to enhanced cell apoptosis. Overexpressed SETDB2 could inhibit tumor progression in vivo and further render LUAD cells sensitive to chemotherapy. CONCLUSIONS In conclusion, these findings uncovered the suppressive role of SETDB2 in LUAD. SETDB2 negatively regulates NRF2 signaling to modulate tumor progression, which creates a therapeutic vulnerability in LUAD.
Collapse
Affiliation(s)
- Guangda Yuan
- Department of Thoracic Surgery The First Affiliated Hospital of Soochow University Suzhou China
| | - Bowen Hu
- Department of Thoracic Surgery The First Affiliated Hospital of Soochow University Suzhou China
- Department of Thoracic Surgery The Affiliated Suzhou Hospital of Nanjing Medical University Suzhou China
| | - Jun Ma
- Department of Thoracic Surgery The Affiliated Suzhou Hospital of Nanjing Medical University Suzhou China
| | - Chuanyu Zhang
- Department of Thoracic Surgery The Affiliated Suzhou Hospital of Nanjing Medical University Suzhou China
| | - Hongya Xie
- Department of Thoracic Surgery The Affiliated Suzhou Hospital of Nanjing Medical University Suzhou China
| | - Tengteng Wei
- Department of Thoracic Surgery The Affiliated Suzhou Hospital of Nanjing Medical University Suzhou China
| | - Yong Yang
- Department of Thoracic Surgery The Affiliated Suzhou Hospital of Nanjing Medical University Suzhou China
| | - Bin Ni
- Department of Thoracic Surgery The First Affiliated Hospital of Soochow University Suzhou China
| |
Collapse
|
9
|
Tseng SC, Lee HY, Nishino M. Imaging of Drug-Related Pneumonitis in Oncology. Semin Respir Crit Care Med 2022; 43:887-898. [PMID: 36307109 DOI: 10.1055/s-0042-1755569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Clinical applications of novel anticancer agents in the past few decades brought marked advances in cancer treatment, enabling remarkable efficacy and effectiveness; however, these novel agents are also associated with toxicities. Among various toxicities, drug-related pneumonitis is one of the major clinical challenges in the management of cancer patients. Imaging plays a key role in detection, diagnosis, and monitoring of drug-related pneumonitis during cancer treatment. In the current era of precision oncology, pneumonitis from molecular targeted therapy and immune-checkpoint inhibitors (ICI) has been recognized as an event of clinical significance. Additionally, further advances of therapeutic approaches in cancer have brought several emerging issues in diagnosis and monitoring of pneumonitis. This article will describe the computed tomography (CT) pattern-based approach for drug-related pneumonitis that has been utilized to describe the imaging manifestations of pneumonitis from novel cancer therapies. Then, we will discuss pneumonitis from representative agents of precision cancer therapy, including mammalian target of rapamycin inhibitors, epidermal growth factor receptor inhibitors, and ICI, focusing on the incidence, risk factors, and the spectrum of CT patterns. Finally, the article will address emerging challenges in the diagnosis and monitoring of pneumonitis, including pneumonitis from combination ICI and radiation therapy and from antibody conjugate therapy, as well as the overlapping imaging features of drug-related pneumonitis and coronavirus disease 2019 pneumonia. The review is designed to provide a practical overview of drug-related pneumonitis from cutting-edge cancer therapy with emphasis on the role of imaging.
Collapse
Affiliation(s)
- Shu-Chi Tseng
- Department of Radiology, Brigham and Women's Hospital and Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ho Yun Lee
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-Gu, Seoul, Korea
| | - Mizuki Nishino
- Department of Radiology, Brigham and Women's Hospital and Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
10
|
Qiao L, Chen Y, Liang N, Xie J, Deng G, Chen F, Wang X, Liu F, Li Y, Zhang J. Targeting Epithelial-to-Mesenchymal Transition in Radioresistance: Crosslinked Mechanisms and Strategies. Front Oncol 2022; 12:775238. [PMID: 35251963 PMCID: PMC8888452 DOI: 10.3389/fonc.2022.775238] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Radiotherapy exerts a crucial role in curing cancer, however, its treatment efficiency is mostly limited due to the presence of radioresistance. Epithelial-to-mesenchymal transition (EMT) is a biological process that endows the cancer cells with invasive and metastatic properties, as well as radioresistance. Many potential mechanisms of EMT-related radioresistance being reported have broaden our cognition, and hint us the importance of an overall understanding of the relationship between EMT and radioresistance. This review focuses on the recent progresses involved in EMT-related mechanisms in regulating radioresistance, irradiation-mediated EMT program, and the intervention strategies to increase tumor radiosensitivity, in order to improve radiotherapy efficiency and clinical outcomes of cancer patients.
Collapse
Affiliation(s)
- Lili Qiao
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Province Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China.,Department of Oncology, Shandong First Medical University, Jinan, China
| | - Yanfei Chen
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Province Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China.,Department of Oncology, Shandong First Medical University, Jinan, China
| | - Ning Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Province Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China.,Department of Oncology, Shandong First Medical University, Jinan, China
| | - Jian Xie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Province Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China.,Department of Oncology, Shandong First Medical University, Jinan, China
| | - Guodong Deng
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Province Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China.,Department of Oncology, Shandong First Medical University, Jinan, China
| | - Fangjie Chen
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Province Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China.,Department of Oncology, Shandong First Medical University, Jinan, China
| | - Xiaojuan Wang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Province Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China.,Department of Oncology, Shandong First Medical University, Jinan, China
| | - Fengjun Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Province Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China.,Department of Oncology, Shandong First Medical University, Jinan, China
| | - Yupeng Li
- Department of Oncology, Shandong First Medical University, Jinan, China.,Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Jiandong Zhang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Province Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China.,Department of Oncology, Shandong First Medical University, Jinan, China
| |
Collapse
|
11
|
Mardanshahi A, Gharibkandi NA, Vaseghi S, Abedi SM, Molavipordanjani S. The PI3K/AKT/mTOR signaling pathway inhibitors enhance radiosensitivity in cancer cell lines. Mol Biol Rep 2021; 48:1-14. [PMID: 34357550 DOI: 10.1007/s11033-021-06607-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 07/29/2021] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Radiotherapy is one of the most common types of cancer treatment modalities. Radiation can affect both cancer and normal tissues, which limits the whole delivered dose. It is well documented that radiation activates phosphatidylinositol 3-kinase (PI3K) and AKT signaling pathway; hence, the inhibition of this pathway enhances the radiosensitivity of tumor cells. The mammalian target of rapamycin (mTOR) is a regulator that is involved in autophagy, cell growth, proliferation, and survival. CONCLUSION The inhibition of mTOR as a downstream mediator of the PI3K/AKT signaling pathway represents a vital option for more effective cancer treatments. The combination of PI3K/AKT/mTOR inhibitors with radiation can increase the radiosensitivity of malignant cells to radiation by autophagy activation. Therefore, this review aims to discuss the impact of such inhibitors on the cell response to radiation.
Collapse
Affiliation(s)
- Alireza Mardanshahi
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nasrin Abbasi Gharibkandi
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Samaneh Vaseghi
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Seyed Mohammad Abedi
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sajjad Molavipordanjani
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
12
|
Fehrenbach U, Rodríguez-Laval V, Jann H, Fernández CMP, Pavel M, Denecke T. Everolimus-induced pneumonitis in neuroendocrine neoplasms: correlation of CT findings and clinical signs. Acta Radiol 2021; 62:1006-1015. [PMID: 32819165 DOI: 10.1177/0284185120950100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Everolimus, a mammalian target of rapamycin (mTOR)-inhibitor, is approved for the treatment of advanced neuroendocrine neoplasms (NEN). A rare major adverse event is the occurrence of drug-induced pneumonitis. PURPOSE To evaluate the correlation between clinical signs and computed tomography (CT) findings in everolimus-induced pneumonitis in patients with NEN. MATERIAL AND METHODS Ninety patients with NEN treated with everolimus were retrospectively enrolled (approved by our Institutional Review Board). All patients received chest CTs before the initiation of everolimus and during the treatment along with physical examinations. Clinical signs of pneumonitis were scored (symptomatic score) according to CTCAE v5.0. Pulmonary function tests (PFT) were evaluated if available. CT images were analyzed based on the severity of interstitial lung disease (ILD), the overall pneumonitis extent (PnE), and regarding presence of typical lung opacification patterns. Follow-up examinations of patients with pneumonitis were analyzed. RESULTS Pneumonitis was diagnosed in 18 (20%) patients. There was no significant correlation between symptomatic score or PFT and ILD score or PnE. In case of a cryptogenic organizing pneumonia pattern (n = 14), symptomatic scores were significantly lower (P = 0.035) than in case of other opacification patterns (n = 4). In the follow-up analysis, we could identify four different clinical courses. CONCLUSION CT detects everolimus-induced pneumonitis at a subclinical stage. In this setting, CT findings, clinical severity, and PFT do not clearly correlate. Opacification pattern analysis seems to be of importance when assessing the severity of CT findings. Asymptomatic patients with positive CT findings should be closely monitored to timely initiate specific treatment.
Collapse
Affiliation(s)
- Uli Fehrenbach
- Department of Radiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | | | - Henning Jann
- Department of Internal Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | | | - Marianne Pavel
- Department of Internal Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Internal Medicine, University Hospital Erlangen, Erlangen, Germany
| | - Timm Denecke
- Department of Radiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Radiology, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
13
|
Danesh Pazhooh R, Rahnamay Farnood P, Asemi Z, Mirsafaei L, Yousefi B, Mirzaei H. mTOR pathway and DNA damage response: A therapeutic strategy in cancer therapy. DNA Repair (Amst) 2021; 104:103142. [PMID: 34102579 DOI: 10.1016/j.dnarep.2021.103142] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/26/2021] [Accepted: 05/31/2021] [Indexed: 10/21/2022]
Abstract
The mammalian target of rapamycin (mTOR) is a conserved serine/threonine-protein kinase, comprising two subunit protein complexes: mTORC1 and mTORC2. In response to insult and cancer, the mTOR pathway plays a crucial role in regulating growth, metabolism, cell survival, and protein synthesis. Key subunits of mTORC1/2 catalyze the phosphorylation of various molecules, including eukaryotic translation initiation factor 4E binding protein 1 (4E-BP1), ribosomal protein S6 kinase β-1 (S6K1). The DNA damage response (DDR) maintains genomic stability and provides an opportunity for treating tumors with defects caused by DNA damaging agents. Many mTOR inhibitors are utilized for the treatment of cancers. However, several clinical trials are still assessing the efficacy of mTOR inhibitors. This paper discusses the role of the mTOR signaling pathway and its regulators in developing cancer. In the following, we will review the interaction between DDR and mTOR signaling and the innovative therapies applied in preclinical and clinical trials for treating cancers.
Collapse
Affiliation(s)
| | | | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Liaosadat Mirsafaei
- Department of Cardiology, Ramsar Campus, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
14
|
High in vitro and in vivo synergistic activity between mTORC1 and PLK1 inhibition in adenocarcinoma NSCLC. Oncotarget 2021; 12:859-872. [PMID: 33889306 PMCID: PMC8057272 DOI: 10.18632/oncotarget.27930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 03/15/2021] [Indexed: 01/22/2023] Open
Abstract
Significant rational is available for specific targeting of PI3K/AKT/mTOR pathway in the treatment of non-small cell lung cancer (NSCLC). However, almost all clinical trials that have evaluated Pi3K pathway-based monotherapies/combinations did not observe an improvement of patient’s outcome. The aim of our study was therefore to define combination of treatment based on the determination of predictive markers of resistance to the mTORC1 inhibitor RAD001/Everolimus. An in vivo study showed high efficacy of RAD001 in NSCLC Patient-Derived Xenografts (PDXs). When looking at biomarkers of resistance by RT-PCR study, three genes were found to be highly expressed in resistant tumors, i.e., PLK1, CXCR4, and AXL. We have then focused our study on the combination of RAD001 + Volasertib, a PLK1 inhibitor, and observed a high antitumor activity of the combination in comparison to each monotherapy; similarly, a clear synergistic effect between the two compounds was found in an in vitro study. Pharmacodynamics study demonstrated that this synergy was due to (1) tumor vascularization decrease, increase of the HIF1 protein expression and decrease of the intracellular pH, and (2) decrease of the Carbonic Anhydrase 9 (CAIX) protein that could not correct intracellular acidosis. In conclusion, all these preclinical data strongly suggest that the inhibition of mTORC1 and PLK1 proteins may be a promising therapeutic approach for NSCLC patients.
Collapse
|
15
|
Haslett K, Koh P, Hudson A, Ryder W, Falk S, Mullan D, Taylor B, Califano R, Blackhall F, Faivre-Finn C. Phase I trial of the MEK inhibitor selumetinib in combination with thoracic radiotherapy in non-small cell lung cancer. Clin Transl Radiat Oncol 2021; 28:24-31. [PMID: 33748440 PMCID: PMC7970011 DOI: 10.1016/j.ctro.2021.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/19/2021] [Accepted: 02/20/2021] [Indexed: 12/25/2022] Open
Abstract
Background The RAS/RAF/MEK/ERK signalling pathway has a pivotal role in cancer proliferation and modulating treatment response. Selumetinib inhibits MEK and enhances effects of radiotherapy in preclinical studies. Patients and methods Single-arm, single-centre, open-label phase I trial. Patients with stage III NSCLC unsuitable for concurrent chemo-radiotherapy, or stage IV with dominant thoracic symptoms, were recruited to a dose-finding stage (Fibonacci 3 + 3 design; maximum number = 18) then an expanded cohort (n = 15). Oral selumetinib was administered twice daily (starting dose 50 mg) commencing 7 days prior to thoracic radiotherapy, then with radiotherapy (6-6.5 weeks; 60-66 Gy/30-33 fractions). The primary objective was to determine the recommended phase II dose (RP2D) of selumetinib in combination with thoracic radiotherapy. Results 21 patients were enrolled (06/2010-02/2015). Median age: 62y (range 50-73). M:F ratio 12(57%):9(43%). ECOG PS 0:1, 7(33%):14(67%). Stage III 16(76%); IV 5(24%). Median GTV 64 cm3 (range 1-224 cm3). 15 patients comprised the expanded cohort at starting dose. All 21 patients completed thoracic radiotherapy as planned and received induction chemotherapy. 13 (62%) patients received the full dose of selumetinib.In the starting cohort no enhanced radiotherapy-related toxicity was seen. Two patients had dose-limiting toxicity (1x grade 3 diarrhoea/fatigue and 1x pulmonary embolism). Commonest grade 3-4 adverse events: lymphopaenia (19/21 patients) and hypertension (7/21 patients). One patient developed grade 3 oesophagitis. No patients developed grade ≥3 radiation pneumonitis. Two patients were alive at the time of analysis (24 and 26 months follow-up, respectively). Main cause of first disease progression: distant metastases ± locoregional progression (12/21 [57.1%] patients). Six patients had confirmed/suspected pneumocystis jiroveci pneumonia. Conclusion We report poor outcome and severe lymphopenia in most patients treated with thoracic radiotherapy and selumetinib at RP2D in combination, contributing to confirmed/clinically suspected pneumocystis jiroveci pneumonia. These results suggest that this combination should not be pursued in a phase II trial.ClinicalTrials.gov reference: NCT01146756.
Collapse
Affiliation(s)
- K. Haslett
- The Christie NHS Foundation Trust, United Kingdom
| | - P. Koh
- University of Manchester, United Kingdom
- New Cross Hospital, United Kingdom
| | - A. Hudson
- The Christie NHS Foundation Trust, United Kingdom
| | - W.D. Ryder
- University of Manchester, United Kingdom
| | - S. Falk
- The Christie NHS Foundation Trust, United Kingdom
| | - D. Mullan
- The Christie NHS Foundation Trust, United Kingdom
| | - B. Taylor
- The Christie NHS Foundation Trust, United Kingdom
| | - R. Califano
- The Christie NHS Foundation Trust, United Kingdom
- University of Manchester, United Kingdom
| | - F. Blackhall
- The Christie NHS Foundation Trust, United Kingdom
- University of Manchester, United Kingdom
| | - C. Faivre-Finn
- The Christie NHS Foundation Trust, United Kingdom
- University of Manchester, United Kingdom
- Corresponding author at: The Christie NHS Foundation Trust, United Kingdom.
| |
Collapse
|
16
|
Baumann BC, Mitra N, Harton JG, Xiao Y, Wojcieszynski AP, Gabriel PE, Zhong H, Geng H, Doucette A, Wei J, O'Dwyer PJ, Bekelman JE, Metz JM. Comparative Effectiveness of Proton vs Photon Therapy as Part of Concurrent Chemoradiotherapy for Locally Advanced Cancer. JAMA Oncol 2020; 6:237-246. [PMID: 31876914 DOI: 10.1001/jamaoncol.2019.4889] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Importance Concurrent chemoradiotherapy is the standard-of-care curative treatment for many cancers but is associated with substantial morbidity. Concurrent chemoradiotherapy administered with proton therapy might reduce toxicity and achieve comparable cancer control outcomes compared with conventional photon radiotherapy by reducing the radiation dose to normal tissues. Objective To assess whether proton therapy in the setting of concurrent chemoradiotherapy is associated with fewer 90-day unplanned hospitalizations (Common Terminology Criteria for Adverse Events, version 4 [CTCAEv4], grade ≥3) or other adverse events and similar disease-free and overall survival compared with concurrent photon therapy and chemoradiotherapy. Design, Setting, and Participants This retrospective, nonrandomized comparative effectiveness study included 1483 adult patients with nonmetastatic, locally advanced cancer treated with concurrent chemoradiotherapy with curative intent from January 1, 2011, through December 31, 2016, at a large academic health system. Three hundred ninety-one patients received proton therapy and 1092, photon therapy. Data were analyzed from October 15, 2018, through February 1, 2019. Interventions Proton vs photon chemoradiotherapy. Main Outcomes and Measures The primary end point was 90-day adverse events associated with unplanned hospitalizations (CTCAEv4 grade ≥3). Secondary end points included Eastern Cooperative Oncology Group (ECOG) performance status decline during treatment, 90-day adverse events of at least CTCAEv4 grade 2 that limit instrumental activities of daily living, and disease-free and overall survival. Data on adverse events and survival were gathered prospectively. Modified Poisson regression models with inverse propensity score weighting were used to model adverse event outcomes, and Cox proportional hazards regression models with weighting were used for survival outcomes. Propensity scores were estimated using an ensemble machine-learning approach. Results Among the 1483 patients included in the analysis (935 men [63.0%]; median age, 62 [range, 18-93] years), those receiving proton therapy were significantly older (median age, 66 [range, 18-93] vs 61 [range, 19-91] years; P < .01), had less favorable Charlson-Deyo comorbidity scores (median, 3.0 vs 2.0; P < .01), and had lower integral radiation dose to tissues outside the target (mean [SD] volume, 14.1 [6.4] vs 19.1 [10.6] cGy/cc × 107; P < .01). Baseline grade ≥2 toxicity (22% vs 24%; P = .37) and ECOG performance status (mean [SD], 0.62 [0.74] vs 0.68 [0.80]; P = .16) were similar between the 2 cohorts. In propensity score weighted-analyses, proton chemoradiotherapy was associated with a significantly lower relative risk of 90-day adverse events of at least grade 3 (0.31; 95% CI, 0.15-0.66; P = .002), 90-day adverse events of at least grade 2 (0.78; 95% CI, 0.65-0.93; P = .006), and decline in performance status during treatment (0.51; 95% CI, 0.37-0.71; P < .001). There was no difference in disease-free or overall survival. Conclusions and Relevance In this analysis, proton chemoradiotherapy was associated with significantly reduced acute adverse events that caused unplanned hospitalizations, with similar disease-free and overall survival. Prospective trials are warranted to validate these results.
Collapse
Affiliation(s)
- Brian C Baumann
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia.,Department of Radiation Oncology, Washington University in St Louis, St Louis, Missouri.,Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia
| | - Nandita Mitra
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia.,Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia
| | - Joanna G Harton
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia
| | - Ying Xiao
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia
| | | | - Peter E Gabriel
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia
| | - Haoyu Zhong
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia
| | - Huaizhi Geng
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia
| | - Abigail Doucette
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia
| | - Jenny Wei
- currently a medical student at Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Peter J O'Dwyer
- Division of Medical Oncology, University of Pennsylvania, Philadelphia.,Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Justin E Bekelman
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia.,Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia.,Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - James M Metz
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia.,Abramson Cancer Center, University of Pennsylvania, Philadelphia
| |
Collapse
|
17
|
Fife K, Bang A. Combined Radiotherapy and New Systemic Therapies - Have We Moved Beyond Palliation? Clin Oncol (R Coll Radiol) 2020; 32:758-765. [PMID: 32863071 DOI: 10.1016/j.clon.2020.07.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/23/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022]
Abstract
The new systemic therapies for cancer are having major impacts on the prognosis of patients with advanced cancers, some achieving long-term survival with targeted therapy or immune checkpoint inhibitors. Interactions of radiotherapy with the new systemic therapies are reviewed. Many agents increase radiosensitivity and particular caution is required combining BRAF inhibitors and radiotherapy because of significant toxicity. Most new systemic therapies can be used safely with palliative doses of radiotherapy, but it is important to be aware of overlapping toxicities depending on the site treated. DNA damage response modulators increase radiosensitivity and may potentially increase radiation toxicity but are at an earlier stage of development. Stereotactic ablative radiotherapy may produce further survival gains in patients responding to targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- K Fife
- Oncology Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - A Bang
- Division of Radiation Oncology, University of British Columbia/BC Cancer, Victoria, British Columbia, Canada
| |
Collapse
|
18
|
Is Host Metabolism the Missing Link to Improving Cancer Outcomes? Cancers (Basel) 2020; 12:cancers12092338. [PMID: 32825010 PMCID: PMC7564800 DOI: 10.3390/cancers12092338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 12/11/2022] Open
Abstract
For the past 100 years, oncologists have relentlessly pursued the destruction of tumor cells by surgical, chemotherapeutic or radiation oncological means. Consistent with this focus, treatment plans are typically based on key characteristics of the tumor itself such as disease site, histology and staging based on local, regional and systemic dissemination. Precision medicine is similarly built on the premise that detailed knowledge of molecular alterations of tumor cells themselves enables better and more effective tumor cell destruction. Recently, host factors within the tumor microenvironment including the vasculature and immune systems have been recognized as modifiers of disease progression and are being targeted for therapeutic gain. In this review, we argue that—to optimize the impact of old and new treatment options—we need to take account of an epidemic that occurs independently of—but has major impact on—the development and treatment of malignant diseases. This is the rapidly increasing number of patients with excess weight and its’ attendant metabolic consequences, commonly described as metabolic syndrome. It is well established that patients with altered metabolism manifesting as obesity, metabolic syndrome and chronic inflammation have an increased incidence of cancer. Here, we focus on evidence that these patients also respond differently to cancer therapy including radiation and provide a perspective how exercise, diet or pharmacological agents may be harnessed to improve therapeutic responses in this patient population.
Collapse
|
19
|
Wang Y, Xie S, He B. Mannose shows antitumour properties against lung cancer via inhibiting proliferation, promoting cisplatin‑mediated apoptosis and reducing metastasis. Mol Med Rep 2020; 22:2957-2965. [PMID: 32700756 PMCID: PMC7453596 DOI: 10.3892/mmr.2020.11354] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 07/09/2019] [Indexed: 01/10/2023] Open
Abstract
It has been reported that mannose exerts antitumour effects against certain types of cancer. The present study was designed to evaluate whether mannose exerted potential anticancer effects on A549 and H1299 non-small cell lung cancer (NSCLC) cells in vitro, which has not been reported previously. A Cell Counting Kit-8 cell viability assay was used to assess the antiproliferative effects of mannose on NSCLC cells. Flow cytometry-based methods were used to evaluate the effects of mannose on the cell cycle distribution and cisplatin-mediated apoptosis of NSCLC cells. Transwell migration and invasion assays were conducted to examine whether mannose could inhibit the invasive abilities of NSCLC cells. The effects of mannose on the PI3K/AKT and ERK signalling pathways were explored through western blot analysis assessing the expression of phosphorylated (p)-AKT and p-ERK1/2. It was found that mannose showed potential anticancer effects against NSCLC cells in vitro by inhibiting proliferation, inducing G0/G1 cell cycle arrest, promoting cisplatin-induced apoptosis and decreasing the invasive abilities. These data indicate the potential anticancer properties of mannose and suggest the application of mannose-based therapies to treat NSCLC.
Collapse
Affiliation(s)
- Youyu Wang
- Department of Thoracic Surgery, Sichuan Academy Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610000, P.R. China
| | - Shenglong Xie
- Department of Thoracic Surgery, Sichuan Academy Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610000, P.R. China
| | - Bin He
- Department of Thoracic Surgery, Sichuan Academy Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610000, P.R. China
| |
Collapse
|
20
|
Reda M, Bagley AF, Zaidan HY, Yantasee W. Augmenting the therapeutic window of radiotherapy: A perspective on molecularly targeted therapies and nanomaterials. Radiother Oncol 2020; 150:225-235. [PMID: 32598976 DOI: 10.1016/j.radonc.2020.06.041] [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] [Received: 11/27/2019] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/25/2022]
Abstract
Radiation therapy is a cornerstone of modern cancer therapy alongside surgery, chemotherapy, and immunotherapy, with over half of all cancer patients receiving radiation therapy as part of their treatment regimen. Development of novel radiation sensitizers that can improve the therapeutic window of radiation therapy are sought after, particularly for tumors at an elevated risk of local and regional recurrence such as locally-advanced lung, head and neck, and gastrointestinal tumors. This review discusses clinical strategies to enhance radiotherapy efficacy and decrease toxicity, hence, increasing the overall therapeutic window. A focus is given to the molecular targets that have been identified and their associated mechanisms of action in enhancing radiotherapy. Examples include cell survival and proliferation signaling such as the EGFR and PI3K/AKT/mTOR pathways, DNA repair genes including PARP and ATM/ATR, angiogenic growth factors, epigenetic regulators, and immune checkpoint proteins. By manipulating various mechanisms of tumor resistance to ionizing radiation (IR), targeted therapies hold significant value to increase the therapeutic window of radiotherapy. Further, the use of novel nanoparticles to enhance radiotherapy is also reviewed, including nanoparticle delivery of chemotherapies, metallic (high-Z) nanoparticles, and nanoparticle delivery of targeted therapies - all of which may improve the therapeutic window of radiotherapy by enhancing the tumor response to IR or reducing normal tissue toxicity.
Collapse
Affiliation(s)
- Moataz Reda
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, United States; PDX Pharmaceuticals, Portland, OR 97239, United States
| | - Alexander F Bagley
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | | | - Wassana Yantasee
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, United States; PDX Pharmaceuticals, Portland, OR 97239, United States.
| |
Collapse
|
21
|
Fu Q, Yu Z. Phosphoglycerate kinase 1 (PGK1) in cancer: A promising target for diagnosis and therapy. Life Sci 2020; 256:117863. [PMID: 32479953 DOI: 10.1016/j.lfs.2020.117863] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 12/22/2022]
Abstract
Phosphoglycerate kinase 1 (PGK1) is the first critical enzyme to produce ATP in the glycolytic pathway. PGK1 is not only a metabolic enzyme but also a protein kinase, which mediates the tumor growth, migration and invasion through phosphorylation some important substrates. Moreover, PGK1 is associated with poor treatment and prognosis of cancers. This manuscript reviews the structure, functions, post-translational modifications (PTMs) of PGK1 and its relationship with tumors, which demonstrates that PGK1 has indispensable value in the tumor progression. The current review highlights the important role of PGK1 in anticancer treatments.
Collapse
Affiliation(s)
- Qi Fu
- Department of Reproductive Medicine, Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, PR China.; College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province, PR China
| | - Zhenhai Yu
- Department of Reproductive Medicine, Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, PR China..
| |
Collapse
|
22
|
Chen K, Shang Z, Dai AL, Dai PL. Novel PI3K/Akt/mTOR pathway inhibitors plus radiotherapy: Strategy for non-small cell lung cancer with mutant RAS gene. Life Sci 2020; 255:117816. [PMID: 32454155 DOI: 10.1016/j.lfs.2020.117816] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/07/2020] [Accepted: 05/16/2020] [Indexed: 02/07/2023]
Abstract
Non-small cell lung cancer (NSCLC) with RAS -mutant gene has been the most difficult obstacle to overcome. Over 25% of muted lung adenocarcinomas have RAS mutation. The prognosis of NSCLC patients with RAS-mutant genes is always poor because there is no effective drug to suppress RAS-mutant genes. NSCLC patients with RAS-mutant usually develop resistance to radiotherapy and chemotherapy, which in some cases leads to a 5-10% survival rate for non-small cell lung cancer (NSCLC). As little clinical symptom of NSCLC was presented at its early stages, thus it always brings in disappointing treatment outcome. Currently, NSCLC presents the highest morbidity and mortality all over the world. The combination of PI3K/AKT/mTOR pathway inhibitors with radiotherapy is a novel strategy to improve radiosensitivity and therapeutic outcome of NSCLC with a RAS-mutant gene. There have been many preclinical studies and clinical trials on the effect of PI3K/AKT/mTOR pathway inhibitors combined with radiotherapy in NSCLC with a RAS-mutant gene have been reported in the past years. This review provides current knowledge of the combination of PI3K/Akt/mTOR pathway inhibitors with radiotherapy, which prove to be a significant improvement for the treatment of NSCLC patients with RAS mutations and will benefit NSCLC patients with RAS mutations.
Collapse
Affiliation(s)
- Kai Chen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhongjun Shang
- Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming 650118, China
| | - Ai-Lin Dai
- Kunming Medical University Haiyuan School, Kunming 650100, China; Maternal and Child Health and Family Planning Service Center of Wenshan state, 663000, China
| | - Pei-Ling Dai
- Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming 650118, China; Kunming Medical University, Kunming 650100, China.
| |
Collapse
|
23
|
Wanigasooriya K, Tyler R, Barros-Silva JD, Sinha Y, Ismail T, Beggs AD. Radiosensitising Cancer Using Phosphatidylinositol-3-Kinase (PI3K), Protein Kinase B (AKT) or Mammalian Target of Rapamycin (mTOR) Inhibitors. Cancers (Basel) 2020; 12:E1278. [PMID: 32443649 PMCID: PMC7281073 DOI: 10.3390/cancers12051278] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/08/2020] [Accepted: 05/12/2020] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy is routinely used as a neoadjuvant, adjuvant or palliative treatment in various cancers. There is significant variation in clinical response to radiotherapy with or without traditional chemotherapy. Patients with a good response to radiotherapy demonstrate better clinical outcomes universally across different cancers. The PI3K/AKT/mTOR pathway upregulation has been linked to radiotherapy resistance. We reviewed the current literature exploring the role of inhibiting targets along this pathway, in enhancing radiotherapy response. We identified several studies using in vitro cancer cell lines, in vivo tumour xenografts and a few Phase I/II clinical trials. Most of the current evidence in this area comes from glioblastoma multiforme, non-small cell lung cancer, head and neck cancer, colorectal cancer, and prostate cancer. The biological basis for radiosensitivity following pathway inhibition was through inhibited DNA double strand break repair, inhibited cell proliferation, enhanced apoptosis and autophagy as well as tumour microenvironment changes. Dual PI3K/mTOR inhibition consistently demonstrated radiosensitisation of all types of cancer cells. Single pathway component inhibitors and other inhibitor combinations yielded variable outcomes especially within early clinical trials. There is ample evidence from preclinical studies to suggest that direct pharmacological inhibition of the PI3K/AKT/mTOR pathway components can radiosensitise different types of cancer cells. We recommend that future in vitro and in vivo research in this field should focus on dual PI3K/mTOR inhibitors. Early clinical trials are needed to assess the feasibility and efficacy of these dual inhibitors in combination with radiotherapy in brain, lung, head and neck, breast, prostate and rectal cancer patients.
Collapse
Affiliation(s)
- Kasun Wanigasooriya
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Science, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (J.D.B.-S.); (Y.S.); (A.D.B.)
- The New Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Edgbaston, Birmingham B15 2TH, UK; (R.T.); (T.I.)
| | - Robert Tyler
- The New Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Edgbaston, Birmingham B15 2TH, UK; (R.T.); (T.I.)
| | - Joao D. Barros-Silva
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Science, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (J.D.B.-S.); (Y.S.); (A.D.B.)
| | - Yashashwi Sinha
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Science, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (J.D.B.-S.); (Y.S.); (A.D.B.)
- The New Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Edgbaston, Birmingham B15 2TH, UK; (R.T.); (T.I.)
| | - Tariq Ismail
- The New Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Edgbaston, Birmingham B15 2TH, UK; (R.T.); (T.I.)
| | - Andrew D. Beggs
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Science, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (J.D.B.-S.); (Y.S.); (A.D.B.)
- The New Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Edgbaston, Birmingham B15 2TH, UK; (R.T.); (T.I.)
| |
Collapse
|
24
|
Chargari C, Levy A, Paoletti X, Soria JC, Massard C, Weichselbaum RR, Deutsch E. Methodological Development of Combination Drug and Radiotherapy in Basic and Clinical Research. Clin Cancer Res 2020; 26:4723-4736. [PMID: 32409306 DOI: 10.1158/1078-0432.ccr-19-4155] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/14/2020] [Accepted: 05/12/2020] [Indexed: 01/03/2023]
Abstract
Newer technical improvements in radiation oncology have been rapidly implemented in recent decades, allowing an improved therapeutic ratio. The development of strategies using local and systemic treatments concurrently, mainly targeted therapies, has however plateaued. Targeted molecular compounds and immunotherapy are increasingly being incorporated as the new standard of care for a wide array of cancers. A better understanding of possible prior methodology issues is therefore required and should be integrated into upcoming early clinical trials including individualized radiotherapy-drug combinations. The outcome of clinical trials is influenced by the validity of the preclinical proofs of concept, the impact on normal tissue, the robustness of biomarkers and the quality of the delivery of radiation. Herein, key methodological aspects are discussed with the aim of optimizing the design and implementation of future precision drug-radiotherapy trials.
Collapse
Affiliation(s)
- Cyrus Chargari
- Department of Radiation Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Université Paris-Sud, Orsay, France
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Institut de Recherche Biomédicale des Armées, Brétigny sur Orge, France
| | - Antonin Levy
- Department of Radiation Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France.
- Université Paris-Sud, Orsay, France
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Xavier Paoletti
- University of Versailles St. Quentin, France
- Institut Curie INSERM U900, Biostatistics for Personalized Medicine Team, St. Cloud, France
| | | | - Christophe Massard
- Université Paris-Sud, Orsay, France
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France.
- Université Paris-Sud, Orsay, France
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
25
|
Reda M, Ngamcherdtrakul W, Gu S, Bejan DS, Siriwon N, Gray JW, Yantasee W. PLK1 and EGFR targeted nanoparticle as a radiation sensitizer for non-small cell lung cancer. Cancer Lett 2019; 467:9-18. [PMID: 31563561 PMCID: PMC6927399 DOI: 10.1016/j.canlet.2019.09.014] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/31/2019] [Accepted: 09/24/2019] [Indexed: 02/07/2023]
Abstract
Radiation sensitizers that can selectively act on cancer cells hold great promise to patients who receive radiation therapy. We developed a novel targeted therapy and radiation sensitizer for non-small cell lung cancer (NSCLC) based on cetuximab conjugated nanoparticle that targets epidermal growth factor receptor (EGFR) and delivers small interfering RNA (siRNA) against polo-like kinase 1 (PLK1). EGFR is overexpressed in 50% of lung cancer patients and a mediator of DNA repair, while PLK1 is a key mitotic regulator whose inhibition enhances radiation sensitivity. The nanoparticle construct (C-siPLK1-NP) effectively targets EGFR + NSCLC cells and reduces PLK1 expression, leading to G2/M arrest and cell death. Furthermore, we show a synergistic combination between C-siPLK1-NP and radiation, which was confirmed in vivo in A549 flank tumors. We also demonstrate the translational potential of C-siPLK1-NP as a systemic therapeutic in an orthotopic lung tumor model, where administration of C-siPLK1-NP reduced tumor growth and led to prolonged survival. Our findings demonstrate that C-siPLK1-NP is effective as a targeted therapy and as a potent radiation sensitizer for NSCLC. Potential application to other EGFR + cancer types such as colorectal and breast cancer is also demonstrated.
Collapse
Affiliation(s)
- Moataz Reda
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, 97239, USA
| | | | - Shenda Gu
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, 97239, USA
| | | | - Natnaree Siriwon
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Joe W Gray
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Wassana Yantasee
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, 97239, USA; PDX Pharmaceuticals, LLC, Portland, OR, 97239, USA.
| |
Collapse
|
26
|
Cuomo F, Altucci L, Cobellis G. Autophagy Function and Dysfunction: Potential Drugs as Anti-Cancer Therapy. Cancers (Basel) 2019; 11:cancers11101465. [PMID: 31569540 PMCID: PMC6826381 DOI: 10.3390/cancers11101465] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 12/24/2022] Open
Abstract
Autophagy is a highly conserved catabolic and energy-generating process that facilitates the degradation of damaged organelles or intracellular components, providing cells with components for the synthesis of new ones. Autophagy acts as a quality control system, and has a pro-survival role. The imbalance of this process is associated with apoptosis, which is a “positive” and desired biological choice in some circumstances. Autophagy dysfunction is associated with several diseases, including neurodegenerative disorders, cardiomyopathy, diabetes, liver disease, autoimmune diseases, and cancer. Here, we provide an overview of the regulatory mechanisms underlying autophagy, with a particular focus on cancer and the autophagy-targeting drugs currently approved for use in the treatment of solid and non-solid malignancies.
Collapse
Affiliation(s)
- Francesca Cuomo
- Department of Precision Medicine, University of Campania "L. Vanvitelli", via L. De Crecchio, 7, 80138 Naples, Italy.
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "L. Vanvitelli", via L. De Crecchio, 7, 80138 Naples, Italy.
| | - Gilda Cobellis
- Department of Precision Medicine, University of Campania "L. Vanvitelli", via L. De Crecchio, 7, 80138 Naples, Italy.
| |
Collapse
|
27
|
Visy A, Bachelot T, Racadot S. Radiation recall syndrome in a patient with breast cancer, after introduction of everolimus. Cancer Radiother 2019; 23:423-425. [DOI: 10.1016/j.canrad.2019.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 01/05/2019] [Accepted: 01/24/2019] [Indexed: 10/26/2022]
|
28
|
Gkountakos A, Sartori G, Falcone I, Piro G, Ciuffreda L, Carbone C, Tortora G, Scarpa A, Bria E, Milella M, Rosell R, Corbo V, Pilotto S. PTEN in Lung Cancer: Dealing with the Problem, Building on New Knowledge and Turning the Game Around. Cancers (Basel) 2019; 11:cancers11081141. [PMID: 31404976 PMCID: PMC6721522 DOI: 10.3390/cancers11081141] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/27/2019] [Accepted: 07/29/2019] [Indexed: 12/15/2022] Open
Abstract
Lung cancer is the most common malignancy and cause of cancer deaths worldwide, owing to the dismal prognosis for most affected patients. Phosphatase and tensin homolog deleted in chromosome 10 (PTEN) acts as a powerful tumor suppressor gene and even partial reduction of its levels increases cancer susceptibility. While the most validated anti-oncogenic duty of PTEN is the negative regulation of the PI3K/mTOR/Akt oncogenic signaling pathway, further tumor suppressor functions, such as chromosomal integrity and DNA repair have been reported. PTEN protein loss is a frequent event in lung cancer, but genetic alterations are not equally detected. It has been demonstrated that its expression is regulated at multiple genetic and epigenetic levels and deeper delineation of these mechanisms might provide fertile ground for upgrading lung cancer therapeutics. Today, PTEN expression is usually determined by immunohistochemistry and low protein levels have been associated with decreased survival in lung cancer. Moreover, available data involve PTEN mutations and loss of activity with resistance to targeted treatments and immunotherapy. This review discusses the current knowledge about PTEN status in lung cancer, highlighting the prevalence of its alterations in the disease, the regulatory mechanisms and the implications of PTEN on available treatment options.
Collapse
Affiliation(s)
- Anastasios Gkountakos
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy
| | - Giulia Sartori
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, 37134 Verona, Italy
| | - Italia Falcone
- Medical Oncology 1, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Geny Piro
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Medical Oncology, Università Cattolica Del Sacro Cuore, 00168 Rome, Italy
| | - Ludovica Ciuffreda
- SAFU Laboratory, Department of Research, Advanced Diagnostics, and Technological Innovation, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Carmine Carbone
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Medical Oncology, Università Cattolica Del Sacro Cuore, 00168 Rome, Italy
| | - Giampaolo Tortora
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Medical Oncology, Università Cattolica Del Sacro Cuore, 00168 Rome, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy
- Center for Applied Research on Cancer (ARC-NET), University of Verona, 37134 Verona, Italy
| | - Emilio Bria
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Medical Oncology, Università Cattolica Del Sacro Cuore, 00168 Rome, Italy
| | - Michele Milella
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, 37134 Verona, Italy
| | - Rafael Rosell
- Germans Trias i Pujol, Health Sciences Institute and Hospital, Campus Can Ruti, 08916 Badalona, Spain
| | - Vincenzo Corbo
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy.
- Center for Applied Research on Cancer (ARC-NET), University of Verona, 37134 Verona, Italy.
| | - Sara Pilotto
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, 37134 Verona, Italy.
| |
Collapse
|
29
|
mTORC1 inhibitor RAD001 (everolimus) enhances non-small cell lung cancer cell radiosensitivity in vitro via suppressing epithelial-mesenchymal transition. Acta Pharmacol Sin 2019; 40:1085-1094. [PMID: 30796356 DOI: 10.1038/s41401-019-0215-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 01/16/2019] [Indexed: 12/19/2022]
Abstract
Resistance to radiotherapy causes non-small cell lung cancer (NSCLC) treatment failure associated with local recurrence and metastasis. Thus, understanding the radiosensitization of NSCLC cells is crucial for developing new treatments and improving prognostics. mTORC1 has been shown to regulate tumor cell radiosensitivity, but the underlying mechanisms are unclear. Moreover, mTORC1 also regulates epithelial-mesenchymal transition (EMT) that is important to metastasis and recurrence. In this study we explored whether mTORC1 regulated NSCLC cell radiosensitivity by altering EMT. We performed immunohistichemical analysis using tumor, adjacent and normal tissues from 50 NSCLC patients, which confirmed significantly elevated mTOR protein expression in NSCLC tissue. Then we used NCI-H460 and NCI-H661 cell lines to examine the effects of the mTORC1 inhibitor RAD001 (everolimus) on in vitro radiosensitivity, protein expression and dose-survival curves. RAD001 (10 nmol/L) significantly inhibited the mTORC1 pathway in both the cell lines. Pretreatment with RAD001 (0.1 nmol/L) enhanced the radiosensitivity in NCI-H661 cells with wild-type PIK3CA and KRAS but not in NCI-H460 cells with mutant PIK3CA and KRAS; the sensitivity enhancement ratios in the two NSCLC cell lines were 1.40 and 1.03, respectively. Furthermore, pretreatment with RAD001 (0.1 nmol/L) significantly decreased the migration and invasion with altered expression of several EMT-associated proteins (significantly increased E-cadherin and decreased vimentin expression) in irradiated NCI-H661 cells. Publicly available expression data confirmed that irradiation affected mTOR and EMT-associated genes at the transcript level in NSCLC cells. These results suggest that mTORC1 inhibition enhances the in vitro radiosensitivity of NSCLC cells with wild-type PIK3CA and KRAS by affecting EMT. Our preclinical data may provide a potential new strategy for NSCLC treatment.
Collapse
|
30
|
Phase IB Study of Induction Chemotherapy With XELOX, Followed by Radiation Therapy, Carboplatin, and Everolimus in Patients With Locally Advanced Esophageal Cancer. Am J Clin Oncol 2019; 42:331-336. [PMID: 30789414 DOI: 10.1097/coc.0000000000000524] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE Preclinical studies have shown synergy between everolimus, an mTOR inhibitor, radiation, and platinum agents. We conducted a phase IB trial to determine the recommended phase II dose of everolimus with carboplatin and radiation. MATERIALS AND METHODS Patients with stage II/III esophageal cancer were enrolled. Following 2 cycles of Capecitabine/Oxaliplatin (XELOX), patients with no disease progression, received 50.4 Gy in 28 fractions and concurrent weekly carboplatin (area under the curve=2), with escalating doses of everolimus. A standard 3+3 dose escalation design was used. RESULTS Nineteen patients were enrolled. Two patients were screen failures and 4 were removed due to poor tolerance to XELOX (n=2) or disease progression (n=2). All treated patients had adenocarcinoma. Median age was 58 (44 to 71 y) and 85% were male patients. One patient at dose level 1 was replaced due to ongoing anxiety. One of 6 patients had a dose-limiting toxicity of bowel ischemia that was fatal. At dose level 2, two of 6 patients had a dose-limiting toxicity (fever with neutropenia and nausea). The recommended phase II dose of everolimus was 2.5 mg QOD. Grade ≥3 toxicities included lymphopenia (11%), nausea (10%), low white blood cell (8.0%) vomiting (5.5%), decreased neutrophils (4.0%). All patients achieved an R0 resection with a pathologic response rate of 40% and a pathologic complete response (ypCR) rate of 23%. The 2-year progression-free survival and overall survival were 50% and 49.6%, respectively. CONCLUSIONS The recommended phase II dose of everolimus with concurrent weekly carboplatin and radiation is 2.5 mg QOD.
Collapse
|
31
|
Tian T, Li X, Zhang J. mTOR Signaling in Cancer and mTOR Inhibitors in Solid Tumor Targeting Therapy. Int J Mol Sci 2019; 20:ijms20030755. [PMID: 30754640 PMCID: PMC6387042 DOI: 10.3390/ijms20030755] [Citation(s) in RCA: 403] [Impact Index Per Article: 67.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 01/28/2019] [Accepted: 02/01/2019] [Indexed: 12/12/2022] Open
Abstract
The mammalian or mechanistic target of rapamycin (mTOR) pathway plays a crucial role in regulation of cell survival, metabolism, growth and protein synthesis in response to upstream signals in both normal physiological and pathological conditions, especially in cancer. Aberrant mTOR signaling resulting from genetic alterations from different levels of the signal cascade is commonly observed in various types of cancers. Upon hyperactivation, mTOR signaling promotes cell proliferation and metabolism that contribute to tumor initiation and progression. In addition, mTOR also negatively regulates autophagy via different ways. We discuss mTOR signaling and its key upstream and downstream factors, the specific genetic changes in the mTOR pathway and the inhibitors of mTOR applied as therapeutic strategies in eight solid tumors. Although monotherapy and combination therapy with mTOR inhibitors have been extensively applied in preclinical and clinical trials in various cancer types, innovative therapies with better efficacy and less drug resistance are still in great need, and new biomarkers and deep sequencing technologies will facilitate these mTOR targeting drugs benefit the cancer patients in personalized therapy.
Collapse
Affiliation(s)
- Tian Tian
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China.
| | - Xiaoyi Li
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China.
| | - Jinhua Zhang
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China.
| |
Collapse
|
32
|
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 II: An AIRO (Italian association of radiotherapy and clinical oncology) overview focused on treatment toxicity. Crit Rev Oncol Hematol 2019; 134:104-119. [PMID: 30658887 DOI: 10.1016/j.critrevonc.2018.11.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 11/13/2018] [Accepted: 11/20/2018] [Indexed: 12/31/2022] Open
Abstract
Clinical development and use of novel systemic agents in combination with radiotherapy (RT) is at nowadays most advanced in the field of treatment of solid tumors. Although for many of these substances preclinical studies provide sufficient evidences on their principal capability to enhance radiation effects, the majority of them have not been investigated in even phase I clinical trials for safety in the context of RT. In clinical practice, unexpected acute and late side effects may emerge especially in combination with RT. As a matter of fact, despite combined modality treatment holds potential for enhancing the therapeutic ratio, some concerns are raised from the lack of high-quality clinical data to guide the care of patients who are treated with novel compounds in conjunction with RT. The aim of this review is to provide, from a radio-oncological point of view, an overview of the most advanced combined treatment concepts for solid tumors focusing on treatment toxicity.
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, 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, 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
|
33
|
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
|
34
|
De Mello RA, Aguiar PN, Tadokoro H, Farias-Vieira TM, Castelo-Branco P, de Lima Lopes G, Pozza DH. MetaLanc9 as a novel biomarker for non-small cell lung cancer: promising treatments via a PGK1-activated AKT/mTOR pathway. J Thorac Dis 2018; 10:S2076-S2078. [PMID: 30023123 PMCID: PMC6036006 DOI: 10.21037/jtd.2018.04.122] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/16/2018] [Indexed: 01/08/2023]
Affiliation(s)
- Ramon Andrade De Mello
- Algarve Biomedical Center, Department of Biomedical Sciences and Medicine, Division of Oncology, University of Algarve, 8005-139 Faro, Portugal
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Translational Research Centre, Division of Medical Oncology, Oncology & Hematology Nucleus of Ceará, Hospital São Mateus, Fortaleza, CE, Brazil
| | | | - Hakaru Tadokoro
- Division of Medical Oncology, São Paulo University Hospital, Federal University of São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| | | | - Pedro Castelo-Branco
- Algarve Biomedical Center, Department of Biomedical Sciences and Medicine, Division of Oncology, University of Algarve, 8005-139 Faro, Portugal
| | | | - Daniel Humberto Pozza
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Faculty of Nutrition and Food Sciences, University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
35
|
Levy A, Doyen J. Metformin for non-small cell lung cancer patients: Opportunities and pitfalls. Crit Rev Oncol Hematol 2018; 125:41-47. [PMID: 29650275 DOI: 10.1016/j.critrevonc.2018.03.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/17/2018] [Accepted: 03/01/2018] [Indexed: 12/11/2022] Open
Abstract
Despite exciting advances of the anticancer armamentarium in the recent years, mortality of non-small cell lung cancer (NSCLC) remains high and novel treatments are requisite. Therapy intensification is explored with promising, but expensive and potentially toxic new compounds. Repositioning already existing drugs for cancer treatment could save money and improve patient outcomes in specific contexts. Observational data suggest that use of the standard antidiabetic agent metformin decreases lung cancer incidence and mortality. Several basic researches have shown various anticancer effects of metformin, acting both on the glycolytic metabolism and on the tumoral immune microenvironment. Synergistic actions of metformin with antitumoral agents in preclinical NSCLC models have then been highlighted. Recent retrospective studies advocated improved outcomes in NSCLC diabetic patients receiving metformin with chemoradiotherapy or systemic compounds (including conventional platinum-based chemotherapy and EGFR tyrosine kinase inhibitors). Several prospective randomized trials are therefore currently assessing the addition of metformin to standard therapy in non-diabetic lung cancer patients. This article reviews promises and possible limitations of concurrent metformin used as an anticancer agent in NSCLC patients.
Collapse
Affiliation(s)
- Antonin Levy
- Department of Radiation Oncology, Institut d'Oncologie Thoracique (IOT), Gustave Roussy, Université Paris-Saclay, F-94805, Villejuif, France; Univ Paris Sud, Université Paris-Saclay, F-94270, Le Kremlin-Bicêtre, France; INSERM U1030, Molecular Radiotherapy, Gustave Roussy, Université Paris-Saclay, F-94805, Villejuif, France.
| | - Jérôme Doyen
- Department of Radiation Oncology, Centre Antoine Lacassagne, 33 Avenue de Valombrose, 06189, Nice Cedex 2, France; University of Côte d'Azur, Nice, France; Institut for Research on Cancer and Aging (IRCAN), CNRS 7284 "Normal and Pathological Angiogenesis", Nice, France
| |
Collapse
|
36
|
Prognostic value of tumor mutations in radically treated locally advanced non-small cell lung cancer patients. Oncotarget 2018; 8:25189-25199. [PMID: 28445990 PMCID: PMC5421921 DOI: 10.18632/oncotarget.15966] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 02/15/2017] [Indexed: 12/25/2022] Open
Abstract
Introduction Chemo-radiation is standard treatment in locally advanced non-small cell lung cancers (NSCLC). The prognostic value of mutations has been poorly explored in this population. RESULTS Clinical data were collected from 190 patients and mutational profiles were obtained in 78 of them; 58 (74%) were males, 31 (40%) current smokers, 47/31 stage IIIA/IIIB and 40 (51%) adenocarcinoma. The following mutations were identified: EGFR 12% (9/78), KRAS 15% (12/78), BRAF 5% (3/65), PI3KCA 2% (1/57), NRAS 3% (1/32), and ALK+ (FISH) 4% (2/51). HER2 was not detected. Median follow-up was 3.1 years. Overall survival was evaluated by group; no significant differences were identified in median overall survival (p = 0.21), with 29.4 months for the EGFR/ALK group (n = 11), 12.8 months for other mutations (n = 17), and 23.4 months for wild-type (n = 50). The EGFR/ALK and other mutations groups had poorer median progression-free survival (9.6 and 6.0 months) compared to the wild-type group (12.0 months; multivariate hazard ratio 2.0 [95% CI, 0.9–4.2] and 2.8 [95% CI, 1.5–5.2] respectively, p = 0.003). Materials and Methods We retrospectively reviewed all patients receiving radical treatment for locally advanced NSCLC in a single institution between January 2002 and June 2013. Next generation sequencing was performed on DNA from paraffin-embedded tissue. ALK rearrangements were detected by immunohistochemistry and/or FISH. Mutational prognostic value for Kaplan-Meier survival parameters was determined by log-rank tests and Cox proportional hazards models. Conclusions Selected gene alterations may be associated with poorer progression-free survival in locally advanced radically treated NSCLC and their prognostic and/or predictive value merits further evaluation in a larger population.
Collapse
|
37
|
Vestergaard HH, Christensen MR, Lassen UN. A systematic review of targeted agents for non-small cell lung cancer. Acta Oncol 2018; 57:176-186. [PMID: 29172833 DOI: 10.1080/0284186x.2017.1404634] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND advanced-stage non-small cell lung cancer (NSCLC) is characterized by having limited treatment options and thus a poor prognosis. However, new treatment options, in the form of targeted agents (TA), have emerged during recent years. This systematic review aims to provide an overview of the accessible literature in PubMed evaluating TA used on NSCLC patients, and the resulting survival outcomes. METHOD this systematic literature review was conducted by reviewing all relevant literature in PubMed. Six separate searches were performed: Three searches where controlled entry terms were used and three free text searches. Furthermore, other relevant publications were included manually. A total of seventy-two studies met the search criteria and were thus further analyzed and evaluated. RESULTS In the included studies, various TAs and their effect on different molecular targets have been evaluated. Clinical responses vary considerably among the different genetic aberrations. The majority of studies evaluated TA for epidermal growth factor receptor (EGFR) mutations and TA for echinoderm microtubule-associated protein-like 4-anaplastic lymphoma kinase (EML4-ALK) rearrangements. Studies regarding the use of TA for Rat sarcoma (RAS), rapidly accelerated fibrosarcoma (RAF), ROS proto-oncogene 1 (ROS1) rearrangement, Receptor tyrosine-protein kinase erbB-2 (ERBB2), Phosphatidylinositol 3-kinase (PIK3CA)/v-akt murine thymoma viral oncogene homolog; protein kinase B(AKT)/Phosphatase and tensin homolog deleted on chromosome 10(PTEN), The mammalian target of rapamycin (mTOR), and Mesenchymal-epithelial transition factor (MET) were included as well. In general, studies comparing treatment outcomes in EGFR-mutated patients and EML4-ALK (ALK) rearranged patients after use of either TA or standard chemotherapy, present significant better results after TA. CONCLUSIONS This systematic review provides an overview of available literature in PubMed regarding NSCLC and TA. Included studies point toward that TA appears to be a promising therapeutic tool in treating NSCLC patients and use of TA is expected to result in improved treatment outcomes.
Collapse
|
38
|
Levy A, Bardet E, Lacas B, Pignon JP, Adam J, Lacroix L, Artignan X, Verrelle P, Le Péchoux C. A phase II open-label multicenter study of gefitinib in combination with irradiation followed by chemotherapy in patients with inoperable stage III non-small cell lung cancer. Oncotarget 2017; 8:15924-15933. [PMID: 27764781 PMCID: PMC5362534 DOI: 10.18632/oncotarget.12741] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 10/12/2016] [Indexed: 12/25/2022] Open
Abstract
Background Gefitinib is an oral EGFR tyrosine kinase inhibitors which may act as a radiosensitizer. Patients and Methods This phase II study evaluated the efficacy of gefitinib 250 mg once daily in combination with thoracic radiotherapy (66 Gy in 6.5 weeks, 2 Gy/day, 5 fractions/week) followed by consolidation chemotherapy (IV cisplatin and vinorelbine) as first line treatment in a population of unselected stage IIIB NSCLC patients according to EGFR mutation status. Results Due to a low accrual rate in this study, the sample size (n = 50) was not reached. Sixteen patients were included in four centers, 50% had adenocarcinoma and 75% were male. Genomic alterations (7 patients studied) retrieved TP53 mutation in 2 patients and no EGFR mutation. Four weeks after radiotherapy, 3 patients (19%) had a partial response, 6 (38%) had a stable disease, and 7 had a progression (44%). Median overall survival was 11 months and median progression-free survival was 5 months. At the time of the last contact, 5 patients (31%) were still alive. Main toxicities were gastrointestinal (81%), cutaneous (81%), general (56%), and respiratory (50%). There were 12>G3 adverse events in 7 (47%) patients, and there was one toxic-death during the concomitant period due to an interstitial pneumonitis. There were two possible adverse events-related deaths during the chemotherapy period (pulmonary embolism (n = 1) and sudden death after the administration of the 3rd course of chemotherapy (n = 1)). Conclusion The benefit of Gefitinib-RT could not be confirmed due to premature trial discontinuation. Further evaluation is required, especially in patients with EGFR mutated NSCLC.
Collapse
Affiliation(s)
- Antonin Levy
- Department of Radiation Oncology, Gustave Roussy, Université Paris-Saclay, Institut Thoracique d'Oncologie (IOT), Villejuif, France.,INSERM U1030, Molecular Radiotherapy, Gustave Roussy, Université Paris-Saclay, Villejuif, France.,Univ Paris Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Etienne Bardet
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest, Nantes, France
| | - Benjamin Lacas
- Gustave Roussy, Université Paris-Saclay, Department of Biostatistics and Epidemiology, Villejuif, France.,INSERM U1018, CESP, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Jean-Pierre Pignon
- Gustave Roussy, Université Paris-Saclay, Department of Biostatistics and Epidemiology, Villejuif, France.,INSERM U1018, CESP, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Julien Adam
- Department of Medical Biology and Pathology, Translational Research Laboratory and Biobank (UMS3655 CNRS / US23 INSERM), INSERM Unit U981, Villejuif, France
| | - Ludovic Lacroix
- Department of Medical Biology and Pathology, Translational Research Laboratory and Biobank (UMS3655 CNRS / US23 INSERM), INSERM Unit U981, Villejuif, France
| | - Xavier Artignan
- Department of Radiation Oncology, University Hospital Grenoble, Grenoble, France.,Department of Radiation Oncology, St Grégoire Hospital, St Grégoire, France
| | - Pierre Verrelle
- Department of Radiation Oncology, Centre Jean Perrin, Clermont-Ferrand, France
| | - Cécile Le Péchoux
- Department of Radiation Oncology, Gustave Roussy, Université Paris-Saclay, Institut Thoracique d'Oncologie (IOT), Villejuif, France
| |
Collapse
|
39
|
Kunz-Schughart LA, Dubrovska A, Peitzsch C, Ewe A, Aigner A, Schellenburg S, Muders MH, Hampel S, Cirillo G, Iemma F, Tietze R, Alexiou C, Stephan H, Zarschler K, Vittorio O, Kavallaris M, Parak WJ, Mädler L, Pokhrel S. Nanoparticles for radiooncology: Mission, vision, challenges. Biomaterials 2016; 120:155-184. [PMID: 28063356 DOI: 10.1016/j.biomaterials.2016.12.010] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/08/2016] [Accepted: 12/09/2016] [Indexed: 12/29/2022]
Abstract
Cancer is one of the leading non-communicable diseases with highest mortality rates worldwide. About half of all cancer patients receive radiation treatment in the course of their disease. However, treatment outcome and curative potential of radiotherapy is often impeded by genetically and/or environmentally driven mechanisms of tumor radioresistance and normal tissue radiotoxicity. While nanomedicine-based tools for imaging, dosimetry and treatment are potential keys to the improvement of therapeutic efficacy and reducing side effects, radiotherapy is an established technique to eradicate the tumor cells. In order to progress the introduction of nanoparticles in radiooncology, due to the highly interdisciplinary nature, expertise in chemistry, radiobiology and translational research is needed. In this report recent insights and promising policies to design nanotechnology-based therapeutics for tumor radiosensitization will be discussed. An attempt is made to cover the entire field from preclinical development to clinical studies. Hence, this report illustrates (1) the radio- and tumor-biological rationales for combining nanostructures with radiotherapy, (2) tumor-site targeting strategies and mechanisms of cellular uptake, (3) biological response hypotheses for new nanomaterials of interest, and (4) challenges to translate the research findings into clinical trials.
Collapse
Affiliation(s)
- Leoni A Kunz-Schughart
- 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
| | - Anna Dubrovska
- 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
| | - Claudia Peitzsch
- 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
| | - Alexander Ewe
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Germany
| | - Achim Aigner
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Germany
| | - Samuel Schellenburg
- Institute of Pathology, University Hospital, Carl Gustav Carus, TU Dresden, Germany
| | - Michael H Muders
- Institute of Pathology, University Hospital, Carl Gustav Carus, TU Dresden, Germany
| | - Silke Hampel
- Leibniz Institute of Solid State and Material Research Dresden, 01171 Dresden, Germany
| | - Giuseppe Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy
| | - Francesca Iemma
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy
| | - Rainer Tietze
- ENT-Department, Section for Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius Professorship, University Hospital Erlangen, Erlangen, Germany
| | - Christoph Alexiou
- ENT-Department, Section for Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius Professorship, University Hospital Erlangen, Erlangen, Germany
| | - Holger Stephan
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01314 Dresden, Germany
| | - Kristof Zarschler
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01314 Dresden, Germany
| | - Orazio Vittorio
- Children's Cancer Institute Australia, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and Australian Centre for NanoMedicine, Sydney, UNSW, Australia
| | - Maria Kavallaris
- Children's Cancer Institute Australia, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and Australian Centre for NanoMedicine, Sydney, UNSW, Australia
| | - Wolfgang J Parak
- Fachbereich Physik, Philipps Universität Marburg, 35037 Marburg, Germany; CIC Biomagune, 20009 San Sebastian, Spain
| | - Lutz Mädler
- Foundation Institute of Materials Science (IWT), Department of Production Engineering, University of Bremen, 28359 Bremen, Germany
| | - Suman Pokhrel
- Foundation Institute of Materials Science (IWT), Department of Production Engineering, University of Bremen, 28359 Bremen, Germany.
| |
Collapse
|
40
|
|
41
|
Daste A, de Mones E, Dupin C, François L, Ravaud A, Digue L. m-TOR inhibitor as potential radiosensitizer for head and neck squamous cell carcinoma: A case report of an organ transplant patient and review of the literature. Oral Oncol 2016; 62:e1-e2. [PMID: 27589913 DOI: 10.1016/j.oraloncology.2016.08.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 08/19/2016] [Accepted: 08/22/2016] [Indexed: 01/02/2023]
Affiliation(s)
- Amaury Daste
- Department of Medical Oncology, Saint-André Hospital, University Hospital, CHU Bordeaux, France; Bordeaux University, Bordeaux, France.
| | - Erwan de Mones
- Department of Otolaryngology - Head and Neck Surgery, Pellegrin Hospital, University Hospital, CHU Bordeaux, France
| | - Charles Dupin
- Department of Radiation Therapy, Haut Leveque Hospital, University Hospital, CHU Bordeaux, France
| | - Louis François
- Department of Medical Oncology, Saint-André Hospital, University Hospital, CHU Bordeaux, France; Bordeaux University, Bordeaux, France
| | - Alain Ravaud
- Department of Medical Oncology, Saint-André Hospital, University Hospital, CHU Bordeaux, France; Bordeaux University, Bordeaux, France
| | - Laurence Digue
- Department of Medical Oncology, Saint-André Hospital, University Hospital, CHU Bordeaux, France
| |
Collapse
|
42
|
de Melo AC, Grazziotin-Reisner R, Erlich F, Fontes Dias MS, Moralez G, Carneiro M, Ingles Garces ÁH, Guerra Alves FV, Novaes Neto B, Fuchshuber-Moraes M, Morando J, Suarez-Kurtz G, Ferreira CG. A phase I study of mTOR inhibitor everolimus in association with cisplatin and radiotherapy for the treatment of locally advanced cervix cancer: PHOENIX I. Cancer Chemother Pharmacol 2016; 78:101-9. [PMID: 27206639 DOI: 10.1007/s00280-016-3064-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 05/11/2016] [Indexed: 01/29/2023]
Abstract
BACKGROUND Cervix cancer (CC) represents the fourth most common cancer in women. Treatment involving cisplatin and radiotherapy has been the standard for locally advanced disease. Everolimus inhibits the aberrant activity of mTOR that is part of carcinogenesis in CC. Further everolimus inactivates the HPV E7 oncoprotein and inhibits its proliferation. Preclinical models have suggested that everolimus sensitizes tumoral cells and vasculature to cisplatin and radiotherapy. METHODS In a 3 + 3 design, the trial aimed to treat three dose levels of at least three patients with daily doses of everolimus (2.5, 5 and 10 mg/day), cisplatin and radiotherapy delivered in a 9-week interval in CC patients, stage IIB, IIIA or IIIB. Patients received everolimus from day -7 up to the last day of brachytherapy. Primary objective was to evaluate safety, toxicity and the maximum-tolerated dose (MTD) of everolimus in association with cisplatin and radiotherapy. Pharmacokinetic (PK) parameters and response rates were analyzed as secondary objectives. RESULTS Thirteen patients were enrolled, 6 at 2.5 mg, 3 at 5 mg and 4 at 10 mg. Four patients did not complete the planned schedule, 1 at 2.5 mg presented grade 4 acute renal failure interpreted as dose-limiting toxicity (DLT) and 3 at 10 mg: 1 with disease progression, and 2 with DLTs-1 grade 3 rash and 1 grade 4 neutropenia. PK results were characterized by dose-dependent increases in AUC and C max. CONCLUSIONS The MTD of everolimus in combination with cisplatin and radiotherapy has been defined as 5 mg/day. The data regarding safety and response rates support further studies.
Collapse
Affiliation(s)
| | | | - Felipe Erlich
- Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | | | | | | - Carlos Gil Ferreira
- Brazilian Clinical Cancer Research Network (RNPCC) - INCA/Decit/MS, D'or Institute of Research and Education (IDOR), Rio de Janeiro, Brazil
| |
Collapse
|
43
|
Mirghani H, Amen F, Tao Y, Deutsch E, Levy A. Increased radiosensitivity of HPV-positive head and neck cancers: Molecular basis and therapeutic perspectives. Cancer Treat Rev 2015; 41:844-52. [PMID: 26476574 DOI: 10.1016/j.ctrv.2015.10.001] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/22/2015] [Accepted: 10/04/2015] [Indexed: 12/27/2022]
Abstract
Human papillomavirus driven head and neck squamous cell carcinoma (HNSCC), particularly oropharyngeal squamous cell carcinoma (OPSCC), are characterized by a significant survival advantage over their HPV-negative counterparts. Although the reasons behind this are still not fully elucidated, it is widely accepted that these tumors have a higher response to ionizing radiation that might explain their favorable outcomes. Potential underlying intrinsic mechanisms include impaired DNA repair abilities, differences in activated repopulation-signaling pathways and cell cycle control mechanisms. The role of the microenvironment is increasingly highlighted, particularly tumor oxygenation and the immune response. Recent studies have shown a distinct pattern of intratumoral immune cell infiltrates, according to HPV status, and have suggested that an increased cytotoxic T-cell based antitumor immune response is involved in improved prognosis of patients with HPV-positive OPSCC. These significant milestones, in the understanding of HPV-induced HNSCC, pave the way to new therapeutic opportunities. This article reviews the current evidence on the biological basis of increased radiosensitivity in HPV-positive HNSCC and discusses potential therapeutic implications.
Collapse
Affiliation(s)
- Haïtham Mirghani
- Department of Otolaryngology - Head and Neck Surgery, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, Villejuif, France.
| | - Furrat Amen
- Department of Otolaryngology, Peterborough City Hospital and Addenbrooke's Hospital, Cambridge, UK
| | - Yungan Tao
- Department of Radiation Oncology, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, Villejuif, France
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, Villejuif, France; Université Paris Sud, Faculté de Médecine, Kremlin Bicêtre 94270, France; INSERM U1030 Molecular Radiotherapy, Cancer Research Institute, Villejuif, France
| | - Antonin Levy
- Department of Radiation Oncology, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, Villejuif, France; Université Paris Sud, Faculté de Médecine, Kremlin Bicêtre 94270, France; INSERM U1030 Molecular Radiotherapy, Cancer Research Institute, Villejuif, France
| |
Collapse
|