1
|
Kutuk T, Tolakanahalli R, McAllister NC, Hall MD, Tom MC, Rubens M, Appel H, Gutierrez AN, Odia Y, Mohler A, Ahluwalia MS, Mehta MP, Kotecha R. Pulsed-Reduced Dose Rate (PRDR) Radiotherapy for Recurrent Primary Central Nervous System Malignancies: Dosimetric and Clinical Results. Cancers (Basel) 2022; 14:2946. [PMID: 35740612 PMCID: PMC9221236 DOI: 10.3390/cancers14122946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/07/2022] [Accepted: 06/14/2022] [Indexed: 02/05/2023] Open
Abstract
PURPOSE The objective was to describe PRDR outcomes and report EQD2 OAR toxicity thresholds. METHODS Eighteen patients with recurrent primary CNS tumors treated with PRDR at a single institution between April 2017 and September 2021 were evaluated. The radiotherapy details, cumulative OAR doses, progression-free survival (PFS), overall survival (OS), and toxicities were collected. RESULTS The median PRDR dose was 45 Gy (range: 36-59.4 Gy); the median cumulative EQD2 prescription dose was 102.7 Gy (range: 93.8-120.4 Gy). The median cumulative EQD2 D0.03cc for the brain was 111.4 Gy (range: 82.4-175.2 Gy). Symptomatic radiation necrosis occurred in three patients, for which the median EQD2 brain D0.03cc was 115.9 Gy (110.4-156.7 Gy). The median PFS and OS after PRDR were 6.3 months (95%CI: 0.9-11.6 months) and 8.6 months (95%CI: 4.9-12.3 months), respectively. The systematic review identified five peer-reviewed studies with a median cumulative EQD2 prescription dose of 110.3 Gy. At a median follow-up of 8.7 months, the median PFS and OS were 5.7 months (95%CI: 2.1-15.4 months) and 6.7 months (95%CI: 3.2-14.2 months), respectively. CONCLUSION PRDR re-irradiation is a relatively safe and feasible treatment for recurrent primary CNS tumors. Despite high cumulative dose to OARs, the risk of high-grade, treatment-related toxicity within the first year of follow-up remains acceptable.
Collapse
Affiliation(s)
- Tugce Kutuk
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA; (T.K.); (R.T.); (N.C.M.); (M.D.H.); (M.C.T.); (H.A.); (A.N.G.); (M.P.M.)
| | - Ranjini Tolakanahalli
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA; (T.K.); (R.T.); (N.C.M.); (M.D.H.); (M.C.T.); (H.A.); (A.N.G.); (M.P.M.)
- Department of Radiation Oncology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Nicole C. McAllister
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA; (T.K.); (R.T.); (N.C.M.); (M.D.H.); (M.C.T.); (H.A.); (A.N.G.); (M.P.M.)
| | - Matthew D. Hall
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA; (T.K.); (R.T.); (N.C.M.); (M.D.H.); (M.C.T.); (H.A.); (A.N.G.); (M.P.M.)
- Department of Radiation Oncology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Martin C. Tom
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA; (T.K.); (R.T.); (N.C.M.); (M.D.H.); (M.C.T.); (H.A.); (A.N.G.); (M.P.M.)
- Department of Radiation Oncology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Muni Rubens
- Department of Clinical Informatics, Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA;
| | - Haley Appel
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA; (T.K.); (R.T.); (N.C.M.); (M.D.H.); (M.C.T.); (H.A.); (A.N.G.); (M.P.M.)
| | - Alonso N. Gutierrez
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA; (T.K.); (R.T.); (N.C.M.); (M.D.H.); (M.C.T.); (H.A.); (A.N.G.); (M.P.M.)
- Department of Radiation Oncology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Yazmin Odia
- Department of Neuro-Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA; (Y.O.); (A.M.)
| | - Alexander Mohler
- Department of Neuro-Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA; (Y.O.); (A.M.)
| | - Manmeet S. Ahluwalia
- Department of Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA;
| | - Minesh P. Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA; (T.K.); (R.T.); (N.C.M.); (M.D.H.); (M.C.T.); (H.A.); (A.N.G.); (M.P.M.)
- Department of Radiation Oncology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Rupesh Kotecha
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA; (T.K.); (R.T.); (N.C.M.); (M.D.H.); (M.C.T.); (H.A.); (A.N.G.); (M.P.M.)
- Department of Radiation Oncology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
- Department of Translational Oncology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
2
|
Ruiz de Garibay G, García de Jalón E, Stigen E, Lund KB, Popa M, Davidson B, Safont MM, Rygh CB, Espedal H, Barrett TM, Haug BE, McCormack E. Repurposing 18F-FMISO as a PET tracer for translational imaging of nitroreductase-based gene directed enzyme prodrug therapy. Am J Cancer Res 2021; 11:6044-6057. [PMID: 33897898 PMCID: PMC8058731 DOI: 10.7150/thno.55092] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/25/2021] [Indexed: 12/25/2022] Open
Abstract
Nitroreductases (NTR) are a family of bacterial enzymes used in gene directed enzyme prodrug therapy (GDEPT) that selectively activate prodrugs containing aromatic nitro groups to exert cytotoxic effects following gene transduction in tumours. The clinical development of NTR-based GDEPT has, in part, been hampered by the lack of translational imaging modalities to assess gene transduction and drug cytotoxicity, non-invasively. This study presents translational preclinical PET imaging to validate and report NTR activity using the clinically approved radiotracer, 18F-FMISO, as substrate for the NTR enzyme. Methods: The efficacy with which 18F-FMISO could be used to report NfsB NTR activity in vivo was investigated using the MDA-MB-231 mammary carcinoma xenograft model. For validation, subcutaneous xenografts of cells constitutively expressing NTR were imaged using 18F-FMISO PET/CT and fluorescence imaging with CytoCy5S, a validated fluorescent NTR substrate. Further, examination of the non-invasive functionality of 18F-FMISO PET/CT in reporting NfsB NTR activity in vivo was assessed in metastatic orthotopic NfsB NTR expressing xenografts and metastasis confirmed by bioluminescence imaging. 18F-FMISO biodistribution was acquired ex vivo by an automatic gamma counter measuring radiotracer retention to confirm in vivo results. To assess the functional imaging of NTR-based GDEPT with 18F-FMISO, PET/CT was performed to assess both gene transduction and cytotoxicity effects of prodrug therapy (CB1954) in subcutaneous models. Results:18F-FMISO retention was detected in NTR+ subcutaneous xenografts, displaying significantly higher PET contrast than NTR- xenografts (p < 0.0001). Substantial 18F-FMISO retention was evident in metastases of orthotopic xenografts (p < 0.05). Accordingly, higher 18F-FMISO biodistribution was prevalent ex vivo in NTR+ xenografts. 18F-FMISO NfsB NTR PET/CT imaging proved useful for monitoring in vivo NTR transduction and the cytotoxic effect of prodrug therapy. Conclusions:18F-FMISO NfsB NTR PET/CT imaging offered significant contrast between NTR+ and NTR- tumours and effective resolution of metastatic progression. Furthermore, 18F-FMISO NfsB NTR PET/CT imaging proved efficient in monitoring the two steps of GDEPT, in vivo NfsB NTR transduction and response to CB1954 prodrug therapy. These results support the repurposing of 18F-FMISO as a readily implementable PET imaging probe to be employed as companion diagnostic test for NTR-based GDEPT systems.
Collapse
|
3
|
Harper K, Yatsyna A, Charbonneau M, Brochu-Gaudreau K, Perreault A, Jeldres C, McDonald PP, Dubois CM. The Chicken Chorioallantoic Membrane Tumor Assay as a Relevant In Vivo Model to Study the Impact of Hypoxia on Tumor Progression and Metastasis. Cancers (Basel) 2021; 13:cancers13051093. [PMID: 33806378 PMCID: PMC7961795 DOI: 10.3390/cancers13051093] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Hypoxia is a negative prognostic factor known to be closely associated with tumor progression and metastasis. However, existing animal models with the ability to recreate the tumor hypoxic microenvironment have disadvantages that limit our ability to understand and target this pathological condition. The chicken ChorioAllantoic Membrane (CAM) assay is increasingly used as a rapid cost-effective drug-testing model that recapitulates many aspects of human cancers. Whether this model recreates the hypoxic environment of tumors remains understudied. Here, we demonstrate that the CAM model effectively supports the development of hypoxic zones in a variety of tumor types. Treatment of tumors with angiogenesis inhibitors or inducers significantly modulated the formation of hypoxic zones as well as tumor progression and metastasis. Our findings suggest that the CAM-based tumor model is a relevant in vivo platform to further understand the pathological responses to hypoxia and test therapeutic interventions aimed at targeting hypoxic cancers. Abstract Hypoxia in the tumor microenvironment is a negative prognostic factor associated with tumor progression and metastasis, and therefore represents an attractive therapeutic target for anti-tumor therapy. To test the effectiveness of novel hypoxia-targeting drugs, appropriate preclinical models that recreate tumor hypoxia are essential. The chicken ChorioAllantoic Membrane (CAM) assay is increasingly used as a rapid cost-effective in vivo drug-testing platform that recapitulates many aspects of human cancers. However, it remains to be determined whether this model recreates the hypoxic microenvironment of solid tumors. To detect hypoxia in the CAM model, the hypoxic marker pimonidazole was injected into the vasculature of tumor-bearing CAM, and hypoxia-dependent gene expression was analyzed. We observed that the CAM model effectively supports the development of hypoxic zones in a variety of human tumor cell line-derived and patient’s tumor fragment-derived xenografts. The treatment of both patient and cell line-derived CAM xenografts with modulators of angiogenesis significantly altered the formation of hypoxic zones within the xenografts. Furthermore, the changes in hypoxia translated into modulated levels of chick liver metastasis as measured by Alu-based assay. These findings demonstrate that the CAM xenograft model is a valuable in vivo platform for studying hypoxia that could facilitate the identification and testing of drugs targeting this tumor microenvironment.
Collapse
Affiliation(s)
- Kelly Harper
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC J1H 5H3, Canada; (K.H.); (A.Y.); (M.C.); (K.B.-G.); (A.P.)
| | - Anna Yatsyna
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC J1H 5H3, Canada; (K.H.); (A.Y.); (M.C.); (K.B.-G.); (A.P.)
| | - Martine Charbonneau
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC J1H 5H3, Canada; (K.H.); (A.Y.); (M.C.); (K.B.-G.); (A.P.)
| | - Karine Brochu-Gaudreau
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC J1H 5H3, Canada; (K.H.); (A.Y.); (M.C.); (K.B.-G.); (A.P.)
| | - Alexis Perreault
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC J1H 5H3, Canada; (K.H.); (A.Y.); (M.C.); (K.B.-G.); (A.P.)
| | - Claudio Jeldres
- Department of Surgery, Division of Urology, Université de Sherbrooke, Sherbrooke, QC J1H 5H3, Canada;
| | - Patrick P. McDonald
- Department of Medicine, Pulmonary Division, Université de Sherbrooke, Sherbrooke, QC J1H 5H3, Canada;
| | - Claire M. Dubois
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC J1H 5H3, Canada; (K.H.); (A.Y.); (M.C.); (K.B.-G.); (A.P.)
- Correspondence:
| |
Collapse
|
4
|
Wilson GD, Wilson TG, Hanna A, Dabjan M, Buelow K, Torma J, Marples B, Galoforo S. Dacomitinib and gedatolisib in combination with fractionated radiation in head and neck cancer. Clin Transl Radiat Oncol 2020; 26:15-23. [PMID: 33251343 PMCID: PMC7677653 DOI: 10.1016/j.ctro.2020.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 01/05/2023] Open
Abstract
We evaluated radiation with dual EGFR and PI3K targeting in head and neck cancer. Dacomitinib, showed an inverse correlation between growth inhibition and EGFR expression. Gedatolisib was effective in each cell line. Neither drug caused radiosensitization in vitro. Gedatolisib was relatively ineffective in vivo in combination with dacomitinib and/or radiation. Dacomitinib was highly effective alone and in combination with radiation and/or gedatolisib. Immunoblotting studies in vivo mirrored the effects seen with growth delay.
Background and purpose There has been little success targeting individual genes in combination with radiation in head and neck cancer. In this study we investigated whether targeting two key pathways simultaneously might be more effective. Materials and methods We studied the effect of combining dacomitinib (pan-HER, irreversible inhibitor) and gedatolisib (dual PI3K/MTOR inhibitor) with radiation in well characterized, low passage xenograft models of HNSCC in vitro and in vivo. Results Dacomitinib showed differential growth inhibition in vitro that correlated to EGFR expression whilst gedatolisib was effective in both cell lines. Neither agent radiosensitized the cell lines in vitro. In vivo studies demonstrated that dacomitinib was an effective agent alone and in combination with radiation whilst the addition of gedatolisib did not enhance the effect of these two modalities despite inhibiting phosphorylation of key genes in the PI3K/MTOR pathway. Conclusions Our results showed that combining two drugs with radiation provided no added benefit compared to the single most active drug. Dacomitinib deserves more investigation as a radiation sensitizing agent in HNSCC.
Collapse
Affiliation(s)
- George D Wilson
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States
| | - Thomas G Wilson
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States
| | - Alaa Hanna
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States
| | - Mohamad Dabjan
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States
| | - Katie Buelow
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States
| | - John Torma
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States
| | - Brian Marples
- Department of Radiation Oncology, University of Rochester, Rochester, NY, United States
| | - Sandra Galoforo
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States
| |
Collapse
|
5
|
Nicolay NH, Wiedenmann N, Mix M, Weber WA, Werner M, Grosu AL, Kayser G. Correlative analyses between tissue-based hypoxia biomarkers and hypoxia PET imaging in head and neck cancer patients during radiochemotherapy-results from a prospective trial. Eur J Nucl Med Mol Imaging 2019; 47:1046-1055. [PMID: 31811344 DOI: 10.1007/s00259-019-04598-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/04/2019] [Indexed: 12/24/2022]
Abstract
PURPOSE Tumor hypoxia impairs the response of head-and-neck cancer (HNSCC) patients to radiotherapy and can be detected both by tissue biomarkers and PET imaging. However, the value of hypoxia biomarkers and imaging for predicting HNSCC patient outcomes are incompletely understood, and potential correlations between tissue and PET data remain to be elucidated. Here, we performed exploratory analyses of potential correlations between tissue-based hypoxia biomarkers and longitudinal hypoxia imaging in a prospective trial of HNSCC patients. METHODS Forty-nine patients undergoing chemoradiation for locally advanced HNSCCs were enrolled in this prospective trial. They underwent baseline biopsies and [18F]FDG PET imaging and [18F]FMISO PET at weeks 0, 2, and 5 during treatment. Immunohistochemical analyses for p16, Ki67, CD34, HIF1α, CAIX, Ku80, and CD44 were performed, and HPV status was assessed. Biomarker expression was correlated with biological imaging information and patient outcome data. RESULTS High HIF1α tumor levels significantly correlated with increased tumor hypoxia at week 2 as assessed by the difference in the [18F]FMISO tumor-to-background ratios, and high HIF1α and CAIX expressions were both associated with a deferred decrease in hypoxia between weeks 2 and 5. Loco-regional recurrence rates after radiotherapy were significantly higher in patients with high CAIX expression and also increased for high levels of the DNA repair factor Ku80. HPV status did not correlate with any of the tested hypoxia biomarkers, and HPV-positive patients showed higher loco-regional control rates and progression-free survival independent of their hypoxia dynamics. CONCLUSION In this exploratory trial, high expression of the tissue-based hypoxia biomarkers HIF1α and CAIX correlated with adverse hypoxia dynamics in HNSCCs during chemoradiation as assessed by PET imaging, and high CAIX levels were associated with increased loco-regional recurrence rates. Hence, hypoxia biomarkers warrant further investigations as potential predictors of hypoxia dynamics and hypoxia-associated radiation resistance.
Collapse
Affiliation(s)
- Nils H Nicolay
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany. .,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Nicole Wiedenmann
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Mix
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wolfgang A Weber
- Department of Nuclear Medicine, Technical University of Munich, Munich, Germany
| | - Martin Werner
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Surgical Pathology, Department of Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anca L Grosu
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gian Kayser
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Surgical Pathology, Department of Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
6
|
Li J, Zhao Z, Du G, Dai T, Zhen X, Cai H, Liao D, Xiang M, Wen Y, Geng L, Yang X, Feng G, Zhang Y, Bai J, Liu L, Du X. Safety and efficacy of pulsed low-dose rate radiotherapy for local recurrent esophageal squamous cell carcinoma after radiotherapy: Study protocol for a prospective multi-center phase II trial. Medicine (Baltimore) 2019; 98:e16176. [PMID: 31261551 PMCID: PMC6620741 DOI: 10.1097/md.0000000000016176] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION Re-irradiation after radiotherapy is a common treatment for locally recurrent esophageal cancer. However, the side effects of re-irradiation are serious. The most serious adverse reactions of re-irradiation include esophageal perforation and hemorrhage caused by esophageal perforation. Studies have shown that pulsed low-dose rate radiotherapy (PLDR) induces a hypersensitivity effect on tumor tissue and a hyper-repair effect on normal tissue, which can simultaneously reduce damage on the normal tissue and increase the therapeutic effect on the tumor. The objective of this study is to explore whether PLDR can reduce rate of esophageal perforation and improve efficacy in patients with recurrent esophageal squamous cell carcinoma (ESCC) after radiotherapy. METHODS AND ANALYSIS This study is a prospective, multi-center, open, single-arm clinical trial designed to enroll 27 patients with locally recurrent ESCC after radiotherapy with or without chemotherapy. Re-irradiation will be performed using intensity modulated radiation therapy in 50 Gy/25 fractions. The strategy of PLDR includes dividing 2 Gy into 10 fractions, and administering each irradiating dose of 20 cGy at an interval of 3 minutes before the next low-dose irradiation. The actual dose rate of administration each time will be 16.67 cGy /minute. The primary endpoint in this study is the rate of esophageal perforation. The secondary endpoints are the objective remission rate, the palliative effect on quality of life and pain, and the time of disease progression. The observation time is 2 years after the end of the study. TRIAL REGISTRATION Clinical trial number: ChiCTR1900020609.
Collapse
Affiliation(s)
- Jie Li
- Department of Head and Neck Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan
- Department of Oncology, Mian Yang Central Hospital, Mianyang
| | - Zhenhua Zhao
- Department of Oncology, Mian Yang Central Hospital, Mianyang
| | - Guobo Du
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nan Chong
| | - Tangzhi Dai
- Department of Oncology, Mian Yang Central Hospital, Mianyang
| | - Xuhai Zhen
- Department of Oncology, Mian Yang Central Hospital, Mianyang
| | - Hongwei Cai
- Department of Oncology, Lang Zhong People‘s Hospital, Lang Zhong
| | - Dongbiao Liao
- Department of Oncology, Mian Yang Central Hospital, Mianyang
| | - Miao Xiang
- Department of Oncology, Mian Yang Central Hospital, Mianyang
| | - Yixue Wen
- Department of Oncology, Mian Yang Central Hospital, Mianyang
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nan Chong
| | - Lidan Geng
- Department of Oncology, Mian Yang Central Hospital, Mianyang
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nan Chong
| | - Xiyue Yang
- Department of Oncology, Mian Yang Central Hospital, Mianyang
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nan Chong
| | - Gang Feng
- Department of Oncology, Mian Yang Central Hospital, Mianyang
| | - Yu Zhang
- Department of Oncology, Mian Yang Central Hospital, Mianyang
| | - Jie Bai
- Department of Rehabilitation Medicine, Mian Yang Central Hospital, Mianyang, China
| | - Lei Liu
- Department of Head and Neck Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan
| | - Xiaobo Du
- Department of Oncology, Mian Yang Central Hospital, Mianyang
| |
Collapse
|
7
|
How to Modulate Tumor Hypoxia for Preclinical In Vivo Imaging Research. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:4608186. [PMID: 30420794 PMCID: PMC6211155 DOI: 10.1155/2018/4608186] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/24/2018] [Accepted: 08/13/2018] [Indexed: 01/20/2023]
Abstract
Tumor hypoxia is related with tumor aggressiveness, chemo- and radiotherapy resistance, and thus a poor clinical outcome. Therefore, over the past decades, every effort has been made to develop strategies to battle the negative prognostic influence of tumor hypoxia. For appropriate patient selection and follow-up, noninvasive imaging biomarkers such as positron emission tomography (PET) radiolabeled ligands are unprecedentedly needed. Importantly, before being able to implement these new therapies and potential biomarkers into the clinical setting, preclinical in vivo validation in adequate animal models is indispensable. In this review, we provide an overview of the different attempts that have been made to create differential hypoxic in vivo cancer models with a particular focus on their applicability in PET imaging studies.
Collapse
|
8
|
Hou Z, Yang Y, Li S, Yan J, Ren W, Liu J, Wang K, Liu B, Wan S. Radiomic analysis using contrast-enhanced CT: predict treatment response to pulsed low dose rate radiotherapy in gastric carcinoma with abdominal cavity metastasis. Quant Imaging Med Surg 2018; 8:410-420. [PMID: 29928606 DOI: 10.21037/qims.2018.05.01] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background To determine the feasibility of radiomic analysis for predicting the therapeutic response of gastric carcinoma (GC) with abdominal cavity metastasis (GCACM) to pulsed low dose rate radiotherapy (PLDRT) using contrast-enhanced computed tomography (CECT) images. Methods Pretreatment CECT images of 43 GCACM patients were analyzed. Patients with complete response (CR) and partial response (PR) were considered responders, while stable disease (SD) and progressive disease (PD) as non-responders. A total of 1,117 image features were quantified from tumor region that segmented from arterial phase CT images. Intra-class correlation coefficient (ICC) and absolute correlation coefficient (ACC) were calculated for selecting influential feature subset. The capability of each influential feature on treatment response classification was assessed using Kruskal-Wallis test and receiver operating characteristic (ROC) analysis. Moreover, artificial neural network (ANN) and k-nearest neighbor (KNN) predictive models were constructed based on the training set (18 responders, 14 non-responders) and the testing set (6 responders, 5 non-responders) validated the reliability of the models. Comparison between the performances of the models was performed by using McNemar's test. Results The analyses showed that 6 features (1 first order-based, 1 texture-based, 1 LoG-based, and 3 wavelet-based) were significantly different between responders and non-responders (AUCs range from 0.686 to 0.728). Both two prediction models based on features extracted from CECT showed potential in predicting the treatment response with higher accuracies (ANN: 0.714, KNN: 0.749 for the training set; ANN: 0.816, KNN: 0.816 for the testing set). No statistical difference was observed between the performance of ANN and KNN (P=0.999). Conclusions Pretreatment radiomic analysis using CECT can potentially provide important information regarding the therapeutic response to PLDRT for GCACM, improving risk stratification.
Collapse
Affiliation(s)
- Zhen Hou
- State Key Laboratory of Bioelectronics, Laboratory for Medical Electronics, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yang Yang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing 210000, China
| | - Shuangshuang Li
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing 210000, China
| | - Jing Yan
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing 210000, China
| | - Wei Ren
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing 210000, China
| | - Juan Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing 210000, China
| | - Kangxin Wang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing 210000, China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing 210000, China
| | - Suiren Wan
- State Key Laboratory of Bioelectronics, Laboratory for Medical Electronics, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
9
|
Blas K, Wilson TG, Tonlaar N, Galoforo S, Hana A, Marples B, Wilson GD. Dual blockade of PI3K and MEK in combination with radiation in head and neck cancer. Clin Transl Radiat Oncol 2018; 11:1-10. [PMID: 30014041 PMCID: PMC6019866 DOI: 10.1016/j.ctro.2018.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/16/2018] [Accepted: 04/20/2018] [Indexed: 02/07/2023] Open
Abstract
Background and purpose In this study we have combined fractionated radiation treatment (RT) with two molecular targeted agents active against key deregulated signaling pathways in head and neck cancer. Materials and methods We used two molecularly characterized, low passage HNSCC cell lines of differing biological characteristics to study the effects of binimetinib and buparlisib in combination with radiation in vitro and in vivo. Results Buparlisib was active against both cell lines in vitro whereas binimetinib was more toxic to UT-SCC-14. Neither agent modified radiation sensitivity in vitro. Buparlisib significantly inhibited growth of UT-SSC-15 alone or in combination with RT but was ineffective in UT-SCC-14. Binimetinib did cause a significant delay with RT in UT-SCC-14 and it significantly reduced growth of the UT-SCC-15 tumors both alone and with RT. The tri-modality treatment was not as effective as RT with a single effective agent. Conclusions No significant benefit was gained by the combined use of the two agents with RT even though each was efficacious when used alone.
Collapse
Affiliation(s)
- Kevin Blas
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States
| | - Thomas G Wilson
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States
| | - Nathan Tonlaar
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States
| | - Sandra Galoforo
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States
| | - Alaa Hana
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States
| | - Brian Marples
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - George D Wilson
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, MI, United States.,Beaumont BioBank, William Beaumont Hospital, Royal Oak, MI, United States
| |
Collapse
|
10
|
Yan J, Yang J, Yang Y, Ren W, Liu J, Gao S, Li S, Kong W, Zhu L, Yang M, Qian X, Liu B. Use of Pulsed Low-Dose Rate Radiotherapy in Refractory Malignancies. Transl Oncol 2018; 11:175-181. [PMID: 29306203 PMCID: PMC5756059 DOI: 10.1016/j.tranon.2017.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/07/2017] [Accepted: 12/07/2017] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Most tumor cell lines exhibited low-dose hyperradiosensitivity (LDHRS) to radiation doses lower than 0.3 Gy. Pulsed low-dose rate radiotherapy (PLDR) took advantage of LDHRS and maximized the tumor control process. In this study, we retrospectively analyzed patients receiving PLDR for refractory malignancies. PATIENTS AND METHODS In total, 22 patients were included in our study: 9 females and 13 males. The median age was 61 years old. All the patients previously received multiline treatments and failed with an estimated survival less than 6 months. Thus, palliative PLDR was given. The PLDR was delivered using 10 fractions of 2 Gy/day, with an interval of 3 minutes, for 5 days per week. The dose rate was 6.67 cGy/min. The median follow-up was 1 year (range 8-30 months). Nine patients underwent PLDR for reirradiation due to locally recurrent diseases. The time interval from last irradiation was 11 to 168 months. Ten patients received PLDR due to poor performance status. Three patients were given PLDR for bulky tumor. The irradiated sites included primary disease (seven patients), locally recurrent disease (nine patients), and retroperitoneal adenopathy (six patients). RESULTS Five patients developed grade 3 or 4 toxicities. No grade 5 toxicities occurred. All the toxicities recovered after treatments. In general, the 1-year local-regional control rate was approximately 40%, and almost all the patients developed progression at the second year after PLDR. The 6-month survival rate was 76%, and the 1-year survival rate was 69%. For the three patients given PLDR for bulky tumor, all of them achieved partial remission 1 month after the PLDR, and one patient achieved complete response at the fourth month. CONCLUSION PLDR is an effective and safe option not only for reirradiation but also for patients with poor performance status or bulky tumors. A prospective clinical trial (NCT03061162) is ongoing to validate our results.
Collapse
Affiliation(s)
- Jing Yan
- The Comprehensive Cancer Centre of Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China
| | - Ju Yang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China
| | - Yang Yang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China
| | - Wei Ren
- The Comprehensive Cancer Centre of Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China
| | - Juan Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China
| | - Shanbao Gao
- The Comprehensive Cancer Centre of Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China
| | - Shuangshuang Li
- The Comprehensive Cancer Centre of Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China
| | - Weiwei Kong
- The Comprehensive Cancer Centre of Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China
| | - Lijing Zhu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China
| | - Mi Yang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China
| | - Xiaoping Qian
- The Comprehensive Cancer Centre of Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China.
| |
Collapse
|
11
|
Antitumor activity of the dual PI3K/MTOR inhibitor, PF-04691502, in combination with radiation in head and neck cancer. Radiother Oncol 2017; 124:504-512. [PMID: 28823407 DOI: 10.1016/j.radonc.2017.08.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/27/2017] [Accepted: 08/02/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND PURPOSE Head and neck squamous cell carcinoma (HNSCC) remains a clinical challenge where new treatments are required to supplement the current-standard-of care of concurrent chemoradiation. The PI3K/AKT/MTOR pathway has been identified from several next generation DNA sequencing studies to be commonly altered and activated in HNSCC. MATERIAL AND METHODS In this study we investigated the activity of PF-04691502, an orally active ATP-competitive, dual inhibitor of PI3K and mTOR, in combination with a clinically relevant fractionated radiation treatment in two contrasting, well characterized, low passage HNSCC models. RESULTS We found that PF-04691502 combined synergistically with radiation in the UT-SCC-14 model derived from a primary cancer but was ineffective in the UT-SCC-15 model which was derived from a nodal recurrence. Further examination of the status of key signaling pathways combined with next generation DNA sequencing of a panel of 160 cancer-associated genes revealed crucial differences between the two models that could account for the differential effect. The UT-SCC-15 cell line was characterized by a higher mutational burden, an excess of variants in the PI3K/AKT/MTOR pathway, increased constitutive activity of PI3K, AKT1 and 2 and MTOR and an inability to inhibit key phosphorylation events in response to the treatments. CONCLUSION This study clearly highlights the promise of agents such as PF-04691502 in selected HNSCCs but also emphasizes the need for molecular characterization and alternative treatment strategies in non-responsive HNSCCs.
Collapse
|
12
|
Meyer K, Krueger SA, Kane JL, Wilson TG, Hanna A, Dabjan M, Hege KM, Wilson GD, Grills I, Marples B. Pulsed Radiation Therapy With Concurrent Cisplatin Results in Superior Tumor Growth Delay in a Head and Neck Squamous Cell Carcinoma Murine Model. Int J Radiat Oncol Biol Phys 2016; 96:161-9. [PMID: 27511853 DOI: 10.1016/j.ijrobp.2016.04.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/13/2016] [Accepted: 04/30/2016] [Indexed: 01/09/2023]
Abstract
PURPOSE To assess the efficacy of 3-week schedules of low-dose pulsed radiation treatment (PRT) and standard radiation therapy (SRT), with concurrent cisplatin (CDDP) in a head and neck squamous cell carcinoma xenograft model. METHODS AND MATERIALS Subcutaneous UT-SCC-14 tumors were established in athymic NIH III HO female mice. A total of 30 Gy was administered as 2 Gy/d, 5 d/wk for 3 weeks, either by PRT (10 × 0.2 Gy/d, with a 3-minute break between each 0.2-Gy dose) or SRT (2 Gy/d, uninterrupted delivery) in combination with concurrent 2 mg/kg CDDP 3 times per week in the final 2 weeks of radiation therapy. Treatment-induced growth delays were defined from twice-weekly tumor volume measurements. Tumor hypoxia was assessed by (18)F-fluoromisonidazole positron emission tomography imaging, and calculated maximum standardized uptake values compared with tumor histology. Tumor vessel density and hypoxia were measured by quantitative immunohistochemistry. Normal tissues effects were evaluated in gut and skin. RESULTS Untreated tumors grew to 1000 mm(3) in 25.4 days (±1.2), compared with delays of 62.3 days (±3.5) for SRT + CDDP and 80.2 days (±5.0) for PRT + CDDP. Time to reach 2× pretreatment volume ranged from 8.2 days (±1.8) for untreated tumors to 67.1 days (±4.7) after PRT + CDDP. Significant differences in tumor growth delay were observed for SRT versus SRT + CDDP (P=.04), PRT versus PRT + CDDP (P=.035), and SRT + CDDP versus PRT + CDDP (P=.033), and for survival between PRT versus PRT + CDDP (P=.017) and SRT + CDDP versus PRT + CDDP (P=.008). Differences in tumor hypoxia were evident by (18)F-fluoromisonidazole positron emission tomography imaging between SRT and PRT (P=.025), although not with concurrent CDDP. Tumor vessel density differed between SRT + CDDP and PRT + CDDP (P=.011). No differences in normal tissue parameters were seen. CONCLUSIONS Concurrent CDDP was more effective in combination PRT than SRT at restricting tumor growth. Significant differences in tumor vascular density were evident between PRT and SRT, suggesting a preservation of vascular network with PRT.
Collapse
Affiliation(s)
- Kurt Meyer
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, Michigan
| | - Sarah A Krueger
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, Michigan
| | - Jonathan L Kane
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, Michigan
| | - Thomas G Wilson
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, Michigan
| | - Alaa Hanna
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, Michigan
| | - Mohamad Dabjan
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, Michigan
| | - Katie M Hege
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, Michigan
| | - George D Wilson
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, Michigan
| | - Inga Grills
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, Michigan
| | - Brian Marples
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, Michigan.
| |
Collapse
|
13
|
Kane JL, Krueger SA, Hanna A, Raffel TR, Wilson GD, Madlambayan GJ, Marples B. Effect of Irradiation on Tumor Microenvironment and Bone Marrow Cell Migration in a Preclinical Tumor Model. Int J Radiat Oncol Biol Phys 2016; 96:170-8. [PMID: 27511854 DOI: 10.1016/j.ijrobp.2016.04.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 04/06/2016] [Accepted: 04/28/2016] [Indexed: 01/29/2023]
Abstract
PURPOSE To characterize the tumor microenvironment after standard radiation therapy (SRT) and pulsed radiation therapy (PRT) in Lewis lung carcinoma (LLC) allografts. METHODS AND MATERIALS Subcutaneous LLC tumors were established in C57BL/6 mice. Standard RT or PRT was given at 2 Gy/d for a total dose of 20 Gy using a 5 days on, 2 days off schedule to mimic clinical delivery. Radiation-induced tumor microenvironment changes were examined after treatment using flow cytometry and antibody-specific histopathology. Normal tissue effects were measured using noninvasive (18)F-fluorodeoxyglucose positron emission tomography/computed tomography after naïve animals were given whole-lung irradiation to 40 Gy in 4 weeks using the same 2-Gy/d regimens. RESULTS Over the 2 weeks of therapy, PRT was more effective than SRT at reducing tumor growth rate (0.31 ± 0.02 mm(3)/d and 0.55 ± 0.04 mm(3)/d, respectively; P<.007). Histopathology showed a significant comparative reduction in the levels of Ki-67 (14.5% ± 3%), hypoxia (10% ± 3.5%), vascular endothelial growth factor (2.3% ± 1%), and stromal-derived factor-1α (2.5% ± 1.4%), as well as a concomitant decrease in CD45(+) bone marrow-derived cell (BMDC) migration (7.8% ± 2.2%) after PRT. The addition of AMD3100 also decreased CD45(+) BMDC migration in treated tumors (0.6% ± 0.1%). Higher vessel density was observed in treated tumors. No differences were observed in normal lung tissue after PRT or SRT. CONCLUSIONS Pulsed RT-treated tumors exhibited slower growth and reduced hypoxia. Pulsed RT eliminated initiation of supportive mechanisms utilized by tumors in low oxygen microenvironments, including angiogenesis and recruitment of BMDCs.
Collapse
Affiliation(s)
- Jonathan L Kane
- Department of Biological Sciences, Oakland University, Rochester, Michigan; Department of Radiation Oncology, William Beaumont Health System, Royal Oak, Michigan
| | - Sarah A Krueger
- Department of Radiation Oncology, William Beaumont Health System, Royal Oak, Michigan
| | - Alaa Hanna
- Department of Radiation Oncology, William Beaumont Health System, Royal Oak, Michigan
| | - Thomas R Raffel
- Department of Biological Sciences, Oakland University, Rochester, Michigan
| | - George D Wilson
- Department of Radiation Oncology, William Beaumont Health System, Royal Oak, Michigan
| | | | - Brian Marples
- Department of Radiation Oncology, William Beaumont Health System, Royal Oak, Michigan.
| |
Collapse
|