Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
JNCI Cancer Spectr ; 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730548

ABSTRACT

BACKGROUND: Traditional constraints specify that 700 cc of liver should be spared a hepatotoxic dose when delivering liver-directed radiotherapy to reduce the risk of inducing liver failure. We investigated the role of single photon emission computed tomography (SPECT) to identify and preferentially avoid functional liver during liver-directed radiation treatment planning in patients with preserved liver function but limited functional liver volume after receiving prior hepatotoxic chemotherapy or surgical resection. METHODS: This phase I trial with a 3 + 3 design evaluated the safety of liver-directed radiotherapy using escalating functional liver radiation dose constraints in patients with liver metastases. Dose limiting toxicities (DLTs) were assessed 6-8 weeks and 6 months after completing radiotherapy. RESULTS: All twelve patients had colorectal liver metastases and received prior hepatotoxic chemotherapy. Eight patients underwent prior liver resection. Median computed tomography (CT) anatomical non-tumor liver volume was 1,584 cc (range 764-2,699 cc). Median SPECT functional liver volume was 1,117 cc (range 570-1,928cc). Median non-target CT and SPECT liver volumes below the volumetric dose constraint were 997 cc (range 544-1,576 cc) and 684 cc (range 429-1,244 cc), respectively. The prescription dose was 67.5-75 Gy in 15 fractions or 75-100 Gy in 25 fractions. No DLTs were observed during follow-up. One-year in-field control was 57%. One-year overall survival was 73%. CONCLUSION: Liver-directed radiotherapy can be safely delivered to high doses when incorporating functional SPECT into the radiation treatment planning process which may enable sparing of lower volumes of liver than traditionally accepted in patients with preserved liver function. TRIAL REGISTRATION: NCT02626312.

2.
Lancet Oncol ; 24(12): 1387-1398, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38039992

ABSTRACT

BACKGROUND: Stereotactic body radiotherapy (SBRT) has the potential to ablate localised pancreatic ductal adenocarcinoma. Selective dismutase mimetics sensitise tumours while reducing normal tissue toxicity. This trial was designed to establish the efficacy and toxicity afforded by the selective dismutase mimetic avasopasem manganese when combined with ablative SBRT for localised pancreatic ductal adenocarcinoma. METHODS: In this adaptive, randomised, double-blind, placebo-controlled, phase 1b/2 trial, patients aged 18 years or older with borderline resectable or locally advanced pancreatic cancer who had received at least 3 months of chemotherapy and had an Eastern Cooperative Oncology Group performance status of 0-2 were enrolled at six academic sites in the USA. Eligible patients were randomly assigned (1:1), with block randomisation (block sizes of 6-12) with a maximum of 24 patients per group, to receive daily avasopasem (90 mg) or placebo intravenously directly before (ie, within 180 min) SBRT (50, 55, or 60 Gy in five fractions, adaptively assigned in real time by Bayesian estimates of 90-day safety and efficacy). Patients and physicians were masked to treatment group allocation, but not to SBRT dose. The primary objective was to find the optimal dose of SBRT with avasopasem or placebo as determined by the late onset EffTox method. All analyses were done on an intention-to-treat basis. This study is registered with ClinicalTrials.gov, NCT03340974, and is complete. FINDINGS: Between Jan 25, 2018, and April 29, 2020, 47 patients were screened, of whom 42 were enrolled (median age was 71 years [IQR 63-75], 23 [55%] were male, 19 [45%] were female, 37 [88%] were White, three [7%] were Black, and one [2%] each were unknown or other races) and randomly assigned to avasopasem (n=24) or placebo (n=18); the placebo group was terminated early after failing to meet prespecified efficacy parameters. At data cutoff (June 28, 2021), the avasopasem group satisfied boundaries for both efficacy and toxicity. Late onset EffTox efficacy response was observed in 16 (89%) of 18 patients at 50 Gy and six (100%) of six patients at 55 Gy in the avasopasem group, and was observed in three (50%) of six patients at 50 Gy and nine (75%) of 12 patients at 55 Gy in the placebo group, and the Bayesian model recommended 50 Gy or 55 Gy in five fractions with avasopasem for further study. Serious adverse events of any cause were reported in three (17%) of 18 patients in the placebo group and six (25%) of 24 in the avasopasem group. In the placebo group, grade 3 adverse events within 90 days of SBRT were abdominal pain, acute cholangitis, pyrexia, increased blood lactic acid, and increased lipase (one [6%] each); no grade 4 events occurred. In the avasopasem group, grade 3-4 adverse events within 90 days of SBRT were acute kidney injury, increased blood alkaline phosphatase, haematoma, colitis, gastric obstruction, lung infection, abdominal abscess, post-surgical atrial fibrillation, and pneumonia leading to respiratory failure (one [4%] each).There were no treatment-related deaths but one late death in the avasopasem group due to sepsis in the setting of duodenal obstruction after off-study treatment was reported as potentially related to SBRT. INTERPRETATION: SBRT that uses 50 or 55 Gy in five fractions can be considered for patients with localised pancreatic ductal adenocarcinoma. The addition of avasopasem might further enhance disease outcomes. A larger phase 2 trial (GRECO-2, NCT04698915) is underway to validate these results. FUNDING: Galera Therapeutics.


Subject(s)
Adenocarcinoma , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Radiosurgery , Humans , Male , Female , Aged , Adenocarcinoma/radiotherapy , Adenocarcinoma/drug therapy , Pancreatic Neoplasms/radiotherapy , Pancreatic Neoplasms/drug therapy , Radiosurgery/adverse effects , Bayes Theorem , Carcinoma, Pancreatic Ductal/radiotherapy , Carcinoma, Pancreatic Ductal/drug therapy , Double-Blind Method , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
3.
Mol Imaging Biol ; 25(6): 991-1019, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37845582

ABSTRACT

Critical advances in radionuclide therapy have led to encouraging new options for cancer treatment through the pairing of clinically useful radiation-emitting radionuclides and innovative pharmaceutical discovery. Of the various subatomic particles used in therapeutic radiopharmaceuticals, alpha (α) particles show great promise owing to their relatively large size, delivered energy, finite pathlength, and resulting ionization density. This review discusses the therapeutic benefits of α-emitting radiopharmaceuticals and their pairing with appropriate diagnostics, resulting in innovative "theranostic" platforms. Herein, the current landscape of α particle-emitting radionuclides is described with an emphasis on their use in theranostic development for cancer treatment. Commonly studied radionuclides are introduced and recent efforts towards their production for research and clinical use are described. The growing popularity of these radionuclides is explained through summarizing the biological effects of α radiation on cancer cells, which include DNA damage, activation of discrete cell death programs, and downstream immune responses. Examples of efficient α-theranostic design are described with an emphasis on strategies that lead to cellular internalization and the targeting of proteins involved in therapeutic resistance. Historical barriers to the clinical deployment of α-theranostic radiopharmaceuticals are also discussed. Recent progress towards addressing these challenges is presented along with examples of incorporating α-particle therapy in pharmaceutical platforms that can be easily converted into diagnostic counterparts.


Subject(s)
Neoplasms , Radiopharmaceuticals , Radiopharmaceuticals/therapeutic use , Alpha Particles/therapeutic use , Radioisotopes/therapeutic use , Pharmaceutical Preparations , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Neoplasms/radiotherapy
4.
Radiat Res ; 198(4): 336-346, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35939823

ABSTRACT

Small molecule inhibitors are currently in preclinical and clinical development for the treatment of selected cancers, particularly those with existing genetic alterations in DNA repair and DNA damage response (DDR) pathways. Keen interest has also been expressed in combining such agents with other targeted antitumor strategies such as radiotherapy. Radiotherapy exerts its cytotoxic effects primarily through DNA damage-induced cell death; therefore, inhibiting DNA repair and the DDR should lead to additive and/or synergistic radiosensitizing effects. In this study we screened the response to X-ray or proton radiation in cell lines treated with DDR inhibitors (DDRis) targeting ATM, ATR, DNA-PKcs, Rad51, and PARP, with survival metrics established using clonogenic assays. We observed that DDRis generate significant radiosensitization in cancer and primary cells derived from normal tissue. Existing genetic defects in cancer cells appear to be an important consideration when determining the optimal inhibitor to use for synergistic combination with radiation. We also show that while greater radiosensitization can be achieved with protons (9.9 keV/µm) combined with DDRis, the relative biological effectiveness is unchanged or in some cases reduced. Our results indicate that while targeting the DDR can significantly radiosensitize cancer cells to such combinations, normal cells may also be equally or more severely affected, depending on the DDRi used. These data highlight the importance of identifying genetic defects as predictive biomarkers of response for combination treatment.


Subject(s)
Neoplasms , Radiation-Sensitizing Agents , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA , DNA Damage , DNA Repair , Humans , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Protons , Rad51 Recombinase/metabolism , Radiation-Sensitizing Agents/pharmacology , X-Rays
5.
Med Phys ; 49(9): 6221-6236, 2022 09.
Article in English | MEDLINE | ID: mdl-35831779

ABSTRACT

BACKGROUND: Proton relative biological effectiveness (RBE) is known to depend on physical factors of the proton beam, such as its linear energy transfer (LET), as well as on cell-line specific biological factors, such as their ability to repair DNA damage. However, in a clinical setting, proton RBE is still considered to have a fixed value of 1.1 despite the existence of several empirical models that can predict proton RBE based on how a cell's survival curve (linear-quadratic model [LQM]) parameters α and ß vary with the LET of the proton beam. Part of the hesitation to incorporate variable RBE models in the clinic is due to the great noise in the biological datasets on which these models are trained, often making it unclear which model, if any, provides sufficiently accurate RBE predictions to warrant a departure from RBE = 1.1. PURPOSE: Here, we introduce a novel model of proton RBE based on how a cell's intrinsic radiosensitivity varies with LET, rather than its LQM parameters. METHODS AND MATERIALS: We performed clonogenic cell survival assays for eight cell lines exposed to 6 MV x-rays and 1.2, 2.6, or 9.9 keV/µm protons, and combined our measurements with published survival data (n = 397 total cell line/LET combinations). We characterized how radiosensitivity metrics of the form DSF% , (the dose required to achieve survival fraction [SF], e.g., D10% ) varied with proton LET, and calculated the Bayesian information criteria associated with different LET-dependent functions to determine which functions best described the underlying trends. This allowed us to construct a six-parameter model that predicts cells' proton survival curves based on the LET dependence of their radiosensitivity, rather than the LET dependence of the LQM parameters themselves. We compared the accuracy of our model to previously established empirical proton RBE models, and implemented our model within a clinical treatment plan evaluation workflow to demonstrate its feasibility in a clinical setting. RESULTS: Our analyses of the trends in the data show that DSF% is linearly correlated between x-rays and protons, regardless of the choice of the survival level (e.g., D10% , D37% , or D50% are similarly correlated), and that the slope and intercept of these correlations vary with proton LET. The model we constructed based on these trends predicts proton RBE within 15%-30% at the 68.3% confidence level and offers a more accurate general description of the experimental data than previously published empirical models. In the context of a clinical treatment plan, our model generally predicted higher RBE-weighted doses than the other empirical models, with RBE-weighted doses in the distal portion of the field being up to 50.7% higher than the planned RBE-weighted doses (RBE = 1.1) to the tumor. CONCLUSIONS: We established a new empirical proton RBE model that is more accurate than previous empirical models, and that predicts much higher RBE values in the distal edge of clinical proton beams.


Subject(s)
Proton Therapy , Protons , Bayes Theorem , Proton Therapy/methods , Radiation Tolerance , Relative Biological Effectiveness , X-Rays
6.
Med Phys ; 49(9): 6098-6109, 2022 09.
Article in English | MEDLINE | ID: mdl-35754208

ABSTRACT

PURPOSE: We assessed whether adding sodium borocaptate (BSH) or 4-borono-l-phenylalanine (BPA) to cells irradiated with proton beams influenced the biological effectiveness of those beams against prostate cancer cells to investigate if the alpha particles generated through proton-boron nuclear reactions would be sufficient to enhance the biological effectiveness of the proton beams. METHODS: We measured clonogenic survival in DU145 cells treated with 80.4-ppm BSH or 86.9-ppm BPA, or their respective vehicles, after irradiation with 6-MV X-rays, 1.2-keV/µm (low linear energy transfer [LET]) protons, or 9.9-keV/µm (high-LET) protons. We also measured γH2AX and 53BP1 foci in treated cells at 1 and 24 h after irradiation with the same conditions. RESULTS: We found that BSH radiosensitized DU145 cells across all radiation types. However, no difference was found in relative radiosensitization, characterized by the sensitization enhancement ratio or the relative biological effectiveness, for vehicle- versus BSH-treated cells. No differences were found in numbers of γH2AX or 53BP1 foci or γH2AX/53BP1 colocalized foci for vehicle- versus BSH-treated cells across radiation types. BPA did not radiosensitize DU145 cells nor induced any significant differences when comparing vehicle- versus BPA-treated cells for clonogenic cell survival or γH2AX and 53BP1 foci or γH2AX/53BP1 colocalized foci. CONCLUSIONS: Treatment with 11 B, at concentrations of 80.4 ppm from BSH or 86.9 ppm from BPA, had no effect on the biological effectiveness of proton beams in DU145 prostate cancer cells. Our results agree with published theoretical calculations indicating that the contribution of alpha particles from such reactions to the total absorbed dose and biological effectiveness is negligible. We also found that BSH radiosensitized DU145 cells to X-rays, low-LET protons, and high-LET protons but that the radiosensitization was not related to DNA damage.


Subject(s)
Boron Neutron Capture Therapy , Prostatic Neoplasms , Proton Therapy , Boron Compounds/pharmacology , Boron Compounds/therapeutic use , Humans , Male , Phenylalanine/pharmacology , Phenylalanine/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , Protons , Relative Biological Effectiveness
7.
Int J Radiat Oncol Biol Phys ; 112(2): 426-436, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34610390

ABSTRACT

PURPOSE: BRCA1/2 pathogenic variant (PV) mutations confer radiation sensitivity preclinically, but there are limited data regarding breast cancer outcomes after radiation therapy (RT) among patients with documented BRCA1/2 PV mutations versus no PV mutations. METHODS AND MATERIALS: This retrospective cohort study included women with clinical stage I-III breast cancer who received definitive surgery and RT and underwent BRCA1/2 genetic evaluation at the The University of Texas MD Anderson Cancer Center. Rates of locoregional recurrence (LRR), disease-specific death (DSD), toxicities, and second cancers were compared by BRCA1/2 PV status. RESULTS: Of the 2213 women who underwent BRCA1/2 testing, 63% self-reported their race as White, 13.6% as Black/African American, 17.6% as Hispanic, and 5.8% as Asian/American Indian/Alaska Native; 124 had BRCA1 and 100 had BRCA2 mutations; and 1394 (63%) received regional nodal RT. The median follow-up time for all patients was 7.4 years (95% confidence interval [CI], 7.1-7.7 years). No differences were found between the groups with and without BRCA1/2 PV mutations in 10-year cumulative incidences of LRR (with mutations: 11.6% [95% CI, 7.0%-17.6%]; without mutations: 6.6% [95% CI, 5.3%-8.0%]; P = .466) and DSD (with mutations: 12.3% [95% CI, 8.0%-17.7%]; without mutations: 13.8% [95% CI, 12.0%-15.8%]; P = .716). On multivariable analysis, BRCA1/2 status was not associated with LRR or DSD, but Black/African American patients (P = .036) and Asians/American Indians/Alaska Native patients (P = .002) were at higher risk of LRR compared with White patients, and Black/African American patients were at higher risk of DSD versus White patients (P = .004). No in-field, nonbreast second cancers were observed in the BRCA1/2 PV group. Rates of acute and late grade ≥3 radiation-related toxicity in the BCRA1/2 PV group were 5.4% (n = 12) and 0.4% (n = 1), respectively. CONCLUSIONS: Oncologic outcomes in a diverse cohort of patients with breast cancer who had a germline BRCA1/2 PV mutation and were treated with RT were similar to those of patients with no mutation, supporting the use of RT according to standard indications in patients with a germline BRCA1/2 PV mutation.


Subject(s)
Breast Neoplasms , Neoplasm Recurrence, Local , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Breast Neoplasms/radiotherapy , Breast Neoplasms/surgery , Cohort Studies , Female , Germ Cells/pathology , Germ-Line Mutation , Humans , Mutation , Neoplasm Recurrence, Local/genetics , Retrospective Studies
8.
Int J Radiat Oncol Biol Phys ; 112(2): 437-444, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34582940

ABSTRACT

PURPOSE: Multigene panel testing has increased the detection of germline mutations in patients with breast cancer. The implications of using radiation therapy (RT) to treat patients with pathogenic variant (PV) mutations are not well understood and have been studied mostly in women with only BRCA1 or BRCA2 PVs. We analyzed oncologic outcomes and toxicity after adjuvant RT in a contemporary, diverse cohort of patients with breast cancer who underwent genetic panel testing. METHODS AND MATERIALS: We retrospectively reviewed the records of 286 women with clinical stage I-III breast cancer diagnosed from 1995 to 2017 who underwent surgery, breast or chest wall RT with or without regional nodal irradiation, multigene panel testing, and evaluation at a large cancer center's genetic screening program. We evaluated rates of overall survival, locoregional recurrence, disease-specific death, and radiation-related toxicities in 3 groups: BRCA1/2 PV carriers, non-BRCA1/2 PV carriers, and patients without PV mutations. RESULTS: PVs were detected in 25.2% of the cohort (12.6% BRCA1/2 and 12.6% non-BRCA1/2). The most commonly detected non-BRCA1/2 mutated genes were ATM, CHEK2, PALB2, CDH1, TP53, and PTEN. The median follow-up time for the entire cohort was 4.4 years (95% confidence interval, 3.8-4.9 years). No differences were found in overall survival, locoregional recurrence, or disease-specific death between groups (P > .1 for all). Acute and late toxicities were comparable across groups. CONCLUSION: Oncologic and toxicity outcomes after RT in women with PV germline mutations detected by multigene pane testing are similar to those in patients without detectable mutations, supporting the use of adjuvant RT as a standard of care when indicated.


Subject(s)
Breast Neoplasms , Germ-Line Mutation , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Breast Neoplasms/radiotherapy , Breast Neoplasms/surgery , Female , Genes, BRCA2 , Genetic Predisposition to Disease , Germ-Line Mutation/genetics , Humans , Neoplasm Recurrence, Local/genetics , Retrospective Studies , Treatment Outcome
9.
Int J Radiat Oncol Biol Phys ; 111(5): 1298-1309, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34400267

ABSTRACT

PURPOSE: To estimate the effects of interfractional anatomic changes on dose to organs at risk (OARs) and tumors, as measured with cone beam computed tomography (CBCT) image guidance for pancreatic stereotactic body radiation therapy. METHODS AND MATERIALS: We evaluated 11 patients with pancreatic cancer whom were treated with stereotactic body radiation therapy (33-40 Gy in 5 fractions) using daily CT-on-rails (CTOR) image guidance immediately before treatment with breath-hold motion management. CBCT alignment was simulated in the treatment planning software by aligning the original planning CT to each fractional CTOR image set via fiducial markers. CTOR data sets were used to calculate fractional doses after alignment by applying the rigid shift of the planning CT and CTOR image sets to the planning treatment isocenter and recalculating the fractional dose. Accumulated dose to the gross tumor volume (GTV), tumor vessel interface, duodenum, small bowel, and stomach were calculated by summing the 5 fractional absolute dose-volume histograms into a single dose-volume histogram for comparison with the original planned dose. RESULTS: Four patients had a GTV D100% of at least 1.5 Gy less than the fractional planned value in several fractions; 4 patients had fractional underestimation of duodenum dose by 1.0 Gy per fraction. The D1.0 cm3 <35 Gy constraint was violated for at least 1 OAR in 3 patients, with either the duodenum (n = 2) or small bowel (n = 1) D1.0 cm3 being higher on the accumulated dose distribution (P = .01). D100% was significantly lower according to accumulated dose GTV (P = .01) and tumor vessel interface (P = .02), with 4 and 2 patients having accumulated D100%  ≥4 Gy lower than the planned value for the GTV and tumor vessel interface, respectively. CONCLUSIONS: For some patients, CBCT image guidance based on fiducial alignment may cause large dosimetric uncertainties for OARs and target structures, according to accumulated dose.


Subject(s)
Radiosurgery , Radiotherapy Planning, Computer-Assisted , Cone-Beam Computed Tomography , Humans , Pancreas , Radiometry , Radiotherapy Dosage
10.
Med Phys ; 48(6): 3243-3261, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33837540

ABSTRACT

PURPOSE: To show that intrinsic radiosensitivity varies greatly for protons and carbon (C) ions in addition to photons, and that DNA repair capacity remains important in governing this variability. METHODS: We measured or obtained from the literature clonogenic survival data for a number of human cancer cell lines exposed to photons, protons (9.9 keV/µm), and C-ions (13.3-77.1 keV/µm). We characterized their intrinsic radiosensitivity by the dose for 10% or 50% survival (D10% or D50% ), and quantified the variability at each radiation quality by the coefficient of variation (COV) in D10% and D50% . We also treated cells with DNA repair inhibitors prior to irradiation to assess how DNA repair capacity affects their variability. RESULTS: We found no statistically significant differences in the COVs of D10% or D50% between any of the radiation qualities investigated. The same was true regardless of whether the cells were treated with DNA repair inhibitors, or whether they were stratified into histologic subsets. Even within histologic subsets, we found remarkable differences in radiosensitivity for high LET C-ions that were often greater than the variations in RBE, with brain cancer cells varying in D10% (D50% ) up to 100% (131%) for 77.1 keV/µm C-ions, and non-small cell lung cancer and pancreatic cancer cell lines varying up to 55% (76%) and 51% (78%), respectively, for 60.5 keV/µm C-ions. The cell lines with modulated DNA repair capacity had greater variability in intrinsic radiosensitivity across all radiation qualities. CONCLUSIONS: Even for cell lines of the same histologic type, there are remarkable variations in intrinsic radiosensitivity, and these variations do not differ significantly between photon, proton or C-ion radiation. The importance of DNA repair capacity in governing the variability in intrinsic radiosensitivity is not significantly diminished for higher LET radiation.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carbon , Cell Line , Cell Survival , Humans , Protons , Radiation Tolerance , Relative Biological Effectiveness
11.
Phys Med Biol ; 66(5)2021 02 26.
Article in English | MEDLINE | ID: mdl-33227715

ABSTRACT

The treatment of cancer with proton radiation therapy was first suggested in 1946 followed by the first treatments in the 1950s. As of 2020, almost 200 000 patients have been treated with proton beams worldwide and the number of operating proton therapy (PT) facilities will soon reach one hundred. PT has long moved from research institutions into hospital-based facilities that are increasingly being utilized with workflows similar to conventional radiation therapy. While PT has become mainstream and has established itself as a treatment option for many cancers, it is still an area of active research for various reasons: the advanced dose shaping capabilities of PT cause susceptibility to uncertainties, the high degrees of freedom in dose delivery offer room for further improvements, the limited experience and understanding of optimizing pencil beam scanning, and the biological effect difference compared to photon radiation. In addition to these challenges and opportunities currently being investigated, there is an economic aspect because PT treatments are, on average, still more expensive compared to conventional photon based treatment options. This roadmap highlights the current state and future direction in PT categorized into four different themes, 'improving efficiency', 'improving planning and delivery', 'improving imaging', and 'improving patient selection'.


Subject(s)
Neoplasms , Proton Therapy , Biology , Humans , Neoplasms/radiotherapy , Photons , Physics , Proton Therapy/methods , Radiotherapy Dosage , Radiotherapy Planning, Computer-Assisted/methods
12.
Med Phys ; 47(1): 272-281, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31677156

ABSTRACT

PURPOSE: High energetic carbon (C-) ion beams undergo nuclear interactions with tissue, producing secondary nuclear fragments. Thus, at depth, C-ion beams are composed of a mixture of different particles with different linear energy transfer (LET) values. We developed a technique to enable isolation of DNA damage response (DDR) in mixed radiation fields using beam line microscopy coupled with fluorescence nuclear track detectors (FNTDs). METHODS: We imaged live cells on a coverslip made of FNTDs right after C-ion, proton or photon irradiation using an in-house built confocal microscope placed in the beam path. We used the FNTD to link track traversals with DNA damage and separated DNA damage induced by primary particles from fragments. RESULTS: We were able to spatially link physical parameters of radiation tracks to DDR in live cells to investigate spatiotemporal DDR in multi-ion radiation fields in real time, which was previously not possible. We demonstrated that the response of lesions produced by the high-LET primary particles associates most strongly with cell death in a multi-LET radiation field, and that this association is not seen when analyzing radiation induced foci in aggregate without primary/fragment classification. CONCLUSIONS: We report a new method that uses confocal microscopy in combination with FNTDs to provide submicrometer spatial-resolution measurements of radiation tracks in live cells. Our method facilitates expansion of the radiation-induced DDR research because it can be used in any particle beam line including particle therapy beam lines. CATEGORY: Biological Physics and Response Prediction.


Subject(s)
Carbon , DNA Damage , Fluorescent Dyes/metabolism , Linear Energy Transfer , Cell Line, Tumor , Cell Survival , Humans , Molecular Imaging , Time Factors
13.
Int J Radiat Oncol Biol Phys ; 105(5): 1119-1125, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31425731

ABSTRACT

PURPOSE: This study seeks to identify biological factors that may yield a therapeutic advantage of proton therapy versus photon therapy. Specifically, we address the role of nonhomologous end-joining (NHEJ) and homologous recombination (HR) in the survival of cells in response to clinical photon and proton beams. METHODS AND MATERIALS: We irradiated HT1080, M059K (DNA-PKcs+/+), and HCC1937 human cancer cell lines and their isogenic counterparts HT1080-shDNA-PKcs, HT1080-shRAD51IND, M059J (DNA-PKcs-/-), and HCC1937-BRCA1 (BRCA1 complemented) to assess cell clonogenic survival and γ-H2AX radiation-induced foci. Cells were irradiated with either clinically relevant photons or 1 of 3 proton linear energy transfer (LET) values. RESULTS: Our results indicate that NHEJ deficiency is more important in dictating cell survival than proton LET. Cells with disrupted HR through BRCA1 mutation showed increased radiosensitivity only for high-LET protons whereas RAD51 depletion showed increased radiosensitivity for both photons and protons. DNA double strand breaks, assessed by γ-H2AX radiation-induced foci, showed greater numbers after 24 hours in cells exposed to higher LET protons. We also observed that NHEJ-deficient cells were unable to repair the vast majority of double strand breaks after 24 hours. CONCLUSIONS: BRCA1 mutation significantly sensitizes cells to protons, but not photons. Loss of NHEJ renders cells hypersensitive to radiation, whereas the relative importance of HR increases with LET across several cell lines. This may be attributable to the more clustered damage induced by higher LET protons, which are harder to repair through NHEJ. This highlights the importance of tumor biology in dictating treatment modality and suggests BRCA1 as a potential biomarker for proton therapy response. Our data also support the use of pharmacologic inhibitors of DNA repair to enhance the sensitivity to different radiation types, although this raises issues for normal tissue toxicity.


Subject(s)
Cell Death/genetics , DNA End-Joining Repair/physiology , Genes, BRCA1 , Homologous Recombination/physiology , Linear Energy Transfer , Photons , Protons , Calcium-Binding Proteins/genetics , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/radiation effects , DNA Breaks, Double-Stranded , Gene Silencing , Histones/analysis , Humans , Mutation , Rad51 Recombinase/genetics , Radiation Tolerance/genetics , Radiation Tolerance/radiation effects , Time Factors
14.
Int J Radiat Oncol Biol Phys ; 105(4): 698-712, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31381960

ABSTRACT

The advent of affordable and rapid next-generation DNA sequencing technology, along with the US Supreme Court ruling invalidating gene patents, has led to a deluge of germline and tumor genetic variant tests that are being rapidly incorporated into clinical cancer decision-making. A major concern for clinicians is whether the presence of germline mutations may increase the risk of radiation toxicity or secondary malignancies. Because scarce clinical data exist to inform decisions at this time, the American Society for Radiation Oncology convened a group of radiation science experts and clinicians to summarize potential issues, review relevant data, and provide guidance for adult patients and their care teams regarding the impact, if any, that genetic testing should have on radiation therapy recommendations. During the American Society for Radiation Oncology workshop, several main points emerged, which are discussed in this manuscript: (1) variants of uncertain significance should be considered nondeleterious until functional genomic data emerge to demonstrate otherwise; (2) possession of germline alterations in a single copy of a gene critical for radiation damage responses does not necessarily equate to increased risk of radiation-induced toxicity; (3) deleterious ataxia-telangiesctasia gene mutations may modestly increase second cancer risk after radiation therapy, and thus follow-up for these patients after indicated radiation therapy should include second cancer screening; (4) conveying to patients the difference between relative and absolute risk is critical to decision-making; and (5) more work is needed to assess the impact of tumor somatic alterations on the probability of response to radiation therapy and the potential for individualization of radiation doses. Data on radiosensitivity related to specific genetic mutations is also briefly discussed.


Subject(s)
Genetic Testing , Mutation , Neoplasms/genetics , Neoplasms/radiotherapy , Radiation Oncologists , Radiation Tolerance/genetics , Adult , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Clinical Decision-Making , Consensus , DNA Repair/genetics , Genes, BRCA1 , Genes, BRCA2 , Genetic Variation , Germ-Line Mutation , Health Care Surveys , Heterozygote , Humans , Neoplasms, Radiation-Induced/genetics , Neoplasms, Second Primary/genetics , Syndrome , Terminology as Topic
15.
Cancer Res ; 79(9): 2327-2338, 2019 05 01.
Article in English | MEDLINE | ID: mdl-31043430

ABSTRACT

When pancreatic cancer cannot be removed surgically, patients frequently experience morbidity and death from progression of their primary tumor. Radiation therapy (RT) cannot yet substitute for an operation because radiation causes fatal bleeding and ulceration of the nearby stomach and intestines before achieving tumor control. There are no FDA-approved medications that prevent or reduce radiation-induced gastrointestinal injury. Here, we overcome this fundamental problem of anatomy and biology with the use of the oral EGLN inhibitor FG-4592, which selectively protects the intestinal tract from radiation toxicity without protecting tumors. A total of 70 KPC mice with autochthonous pancreatic tumors received oral FG-4592 or vehicle control ± ablative RT to a cumulative 75 Gy administered in 15 daily fractions to a limited tumor field. Although ablative RT reduced complications from local tumor progression, fatal gastrointestinal bleeding was observed in 56% of mice that received high-dose RT with vehicle control. However, radiation-induced bleeding was completely ameliorated in mice that received high-dose RT with FG-4592 (0% bleeding, P < 0.0001 compared with vehicle). Furthermore, FG-4592 reduced epithelial apoptosis by half (P = 0.002) and increased intestinal microvessel density by 80% compared with vehicle controls. EGLN inhibition did not stimulate cancer growth, as treatment with FG-4592 alone, or overexpression of HIF2 within KPC tumors independently improved survival. Thus, we provide a proof of concept for the selective protection of the intestinal tract by the EGLN inhibition to enable ablative doses of cytotoxic therapy in unresectable pancreatic cancer by reducing untoward morbidity and death from radiation-induced gastrointestinal bleeding. SIGNIFICANCE: Selective protection of the intestinal tract by EGLN inhibition enables potentially definitive doses of radiation therapy. This might allow radiation to be a surgical surrogate for unresectable pancreatic cancer.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/9/2327/F1.large.jpg.


Subject(s)
Glycine/analogs & derivatives , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Isoquinolines/pharmacology , Pancreatic Neoplasms/mortality , Radiation Injuries/prevention & control , Radiation-Protective Agents/pharmacology , Radiotherapy/mortality , Animals , Apoptosis , Female , Glycine/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/radiotherapy , Proto-Oncogene Proteins p21(ras)/physiology , Radiation Injuries/etiology , Radiation Injuries/mortality , Radiotherapy/adverse effects , Transcription Factors/physiology , Tumor Suppressor Protein p53/physiology
16.
Radiat Prot Dosimetry ; 183(1-2): 264-269, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30726978

ABSTRACT

Human exposure to α-particles from radon and other radionuclides is associated with carcinogenesis, but if well controlled and targeted to cancer cells, α-particles may be used in radiotherapy. Thus, it is important to understand the biological effects of α-particles to predict cancer risk and optimise radiotherapy. To enable studies of α-particles in cells, we developed and characterised an α-particle automated irradiation rig that allows exposures at a shallow angle (70° to the normal) of cell monolayers in a 30 mm diameter dish to complement standard perpendicular irradiations. The measured incident energy of the α-particles was 3.3 ± 0.5 MeV (LET in water = 120 keV µm-1), with a maximum incident dose rate of 1.28 ± 0.02 Gy min-1, which for a 5 µm cell monolayer corresponds to a mean dose rate of 1.57 ± 0.02 Gy min-1 and a mean LET in water of 154 keV µm-1. The feasibility of resolving radiation-induced DNA double-strand breaks (DSB) foci along the track of α-particles was demonstrated using immunofluorescent labelling with γH2AX and 53BP1 in normal MRC-5 human lung cells.


Subject(s)
Alpha Particles , Cells, Cultured/radiation effects , Lung/cytology , Radiobiology/instrumentation , DNA Breaks, Double-Stranded , Equipment Design , Humans , Linear Energy Transfer
17.
Adv Radiat Oncol ; 3(4): 693-700, 2018.
Article in English | MEDLINE | ID: mdl-30370371

ABSTRACT

PURPOSE: There are limited treatment options for locally advanced, unresectable pancreatic cancer (LAPC) and no likelihood of cure without surgery. Radiation offers an option for local control, but radiation dose has previously been limited by nearby bowel toxicity. Advances in on-board imaging and treatment planning may allow for dose escalation not previously feasible and improve local control. In preparation for development of clinical trials of dose escalation in LAPC, we undertook a dosimetric study to determine the maximum possible dose escalation while maintaining known normal tissue constraints. METHODS AND MATERIALS: Twenty patients treated at our institution with either SBRT or dose-escalated hypofractionated IMRT (DE-IMRT) were re-planned using dose escalated SBRT to 70 Gy in 5 fractions to the GTV and 40 Gy in 5 fractions to the PTV. Standard accepted organ at risk (OAR) constraints were used for planning. Descriptive statistics were generated for homogeneity, conformality, OAR's and GTV/PTV. RESULTS: Mean iGTV coverage by 50 Gy was 91% (±0.07%), by 60 Gy was 61.3% (±0.08%) and by 70 Gy was 24.4% (±0.05%). Maximum PTV coverage by 70 Gy was 33%. Maximum PTV coverage by 60 Gy was 77.5%. The following organ at risk (OAR) constraints were achieved for 90% of generated plans: Duodenum V20 < 30 cc, V30 < 3 cc, V35 < 1 cc; Small Bowel V20 < 15 cc, V30 < 1 cc, V35 < 0.1 cc; Stomach V20 < 20 cc, V30 < 2 cc, V35 < 1 cc. V40 < 0.5 cc was achieved for all OAR. CONCLUSIONS: Dose escalation to 60 Gy is dosimetrically feasible with adequate GTV coverage. The identified constraints for OAR's will be used in ongoing clinical trials.

18.
Med Phys ; 45(2): 884-897, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29178457

ABSTRACT

PURPOSE: The purpose of this study was to acquire beam data for an MR-linac, with and without a 1.5 T magnetic field, by using a variety of commercially available detectors to assess their relative response in the magnetic field. The impact of the magnetic field on the measured dose distribution was also assessed. METHODS: An MR-safe 3D scanning water phantom was used to measure output factors, depth dose curves, and off-axis profiles for various depths and for field sizes between 2 × 2 cm2 and 22 × 22 cm2 for an Elekta MR-linac beam with the orthogonal 1.5 T magnetic field on or off. An on-board MV portal imaging system was used to ensure that the reproducibility of the detector position, both with and without the magnetic field, was within 0.1 mm. The detectors used included ionization chambers with large, medium, and small sensitive volumes; a diamond detector; a shielded diode; and an unshielded diode. RESULTS: The offset of the effective point of measurement of the ionization chambers was found to be reduced by at least half for each chamber in the direction parallel with the beam. A lateral shift of similar magnitude was also introduced to the chambers' effective point of measurement toward the average direction of the Lorentz force. A similar lateral shift (but in the opposite direction) was also observed for the diamond and diode detectors. The measured lateral shift in the dose distribution was independent of depth and field size for each detector for fields between 2 × 2 cm2 and 10 × 10 cm2 . The shielded diode significantly misrepresented the dose distribution in the lateral direction perpendicular to the magnetic field, making it seem more symmetric. The percentage depth dose was generally found to be lower with the magnetic field than without, but this difference was reduced as field size increased. The depth of maximum dose showed little dependence on field size in the presence of the magnetic field, with values from 1.2 cm to 1.3 cm between the 2 × 2 cm2 and 22 × 22 cm2 fields. Output factors measured in the magnetic field at the center of the beam profile produced a larger spread of values between detectors for fields smaller than 10 × 10 cm2 (with a spread of 2% at 3 × 3 cm2 ). The spread of values was more consistent when the output factors were measured at the point of peak intensity of the lateral dose distribution instead (except for the shielded diode which differed by up to 2% depending on field size). CONCLUSIONS: The magnetic field of the MR-linac alters the effective point of measurement of ionization chambers, shifting it both downstream and laterally. Shielded diodes produce incorrect and misleading dose profiles. The output factor measured at the point of peak intensity in the lateral dose distribution is more robust than the conventional output factor (measured at central axis). Diodes are not recommended for output factor measurements in the magnetic field.


Subject(s)
Diamond , Electrical Equipment and Supplies , Magnetic Resonance Imaging , Particle Accelerators , Radiation Dosage , Radiometry/instrumentation , Imaging, Three-Dimensional , Phantoms, Imaging , Water
19.
Med Phys ; 44(7): 3830-3838, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28432792

ABSTRACT

PURPOSE: The aim of this study was to examine the effect of submillimeter air gaps that may exist between an ionization chamber and solid phantoms when measurements are performed in a magnetic field. METHODS: Geant4 Monte Carlo simulations were performed using a model of a PTW 30013 Farmer chamber in a water phantom. Symmetrical and asymmetrical air gaps of various thicknesses were modeled surrounding the chamber, and the dose to the air cavity of the chamber was scored in each case. Magnetic fields were modeled parallel to the long axis of the chamber with strengths of 0, 0.35 T, 1.0 T, and 1.5 T. To examine the phenomenon in more detail, the gyroradii of the electrons responsible for the energy deposited in the chamber were scored as they entered the chamber and the total energy deposited was split into three components: energy originating from inside the chamber, in the immediate vacinity of the chamber, or outside the chamber. RESULTS: Differences in the chamber dose of 1.6% were observed for asymmetric air gaps just 0.2 mm thick. No effect greater than 0.5% was observed for the symmetrical air gaps investigated in this work (1.4 mm thick or less) for this chamber/magnetic field configuration. The mean gyroradius of contributing electrons as they first enter the chamber was 4 mm. The presence of the air gap reduced the energy contributions from electrons released in the immediate vicinity of the chamber, and this loss was not completely compensated for when a magnetic field was present. CONCLUSIONS: The gyroradius of most electrons was too large to be responsible for the air gap effect via the electron return effect; instead, the effect is attributed to the loss of energy contributions from electrons originating inside the air gap volume, which is not completely compensated for by more distant electrons owing to their reduced range in the magnetic field. When the chamber is parallel with the magnetic field, symmetric air gaps have a smaller effect (< 0.5%) compared to asymmetric air-gaps (up to 1.6%) on the chamber response.


Subject(s)
Monte Carlo Method , Phantoms, Imaging , Radiometry , Air , Electrons , Magnetic Fields
20.
Int J Radiat Oncol Biol Phys ; 97(2): 323-332, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28068240

ABSTRACT

PURPOSE: To determine whether severity of lymphopenia is dependent on radiation dose and fractional volume of spleen irradiated unintentionally during definitive chemoradiation (CRT) in patients with locally advanced pancreatic cancer (LAPC). METHODS: 177 patients with LAPC received induction chemotherapy (mainly gemcitabine-based regimens) followed by CRT (median 50.4 Gy with concurrent capecitabine) from January 2006 to December 2012. Absolute lymphocyte count (ALC) was recorded at baseline, before CRT, and 2 to 10 weeks after CRT. Splenic dose-volume histogram (DVH) parameters were reported as mean splenic dose (MSD) and percentage of splenic volume receiving at least 5- (V5), 10- (V10), 15- (V15), and 20-Gy (V20) dose. Overall survival (OS) was analyzed with use of the Cox model, and development of post-CRT severe lymphopenia (ALC <0.5 K/UL) was assessed by multivariate logistic regression with use of baseline and treatment factors. RESULTS: The median post-CRT ALC (0.68 K/UL; range, 0.13-2.72) was significantly lower than both baseline ALC (1.42 K/UL; range, 0.34-3.97; P<.0001) and pre-CRT ALC (1.32 K/UL, range 0.36-4.82; P<.0001). Post-CRT ALC <0.5 K/UL was associated with inferior OS on univariate analysis (median, 11.1 vs 15.3 months; P=.01) and multivariate analysis (hazard ratio = 1.66, P=.01). MSD (9.8 vs 6 Gy, P=.03), median V10 (32.6 vs 16%, P=.04), V15 (23.2 vs 9.5%, P=.03), and V20 (15.4 vs 4.6%, P=.02) were significantly higher in patients with severe lymphopenia than in those without. On multivariate analysis, postinduction lymphopenia (P<.001; odds ratio [OR] = 5.25) and MSD (P=.002; OR= 3.42) were independent predictors for the development of severe post-CRT lymphopenia. CONCLUSION: Severe post-CRT lymphopenia is an independent predictor of poor OS in LAPC patients receiving CRT. Higher splenic doses increase the risk for the development of severe post-CRT lymphopenia. When clinically indicated, assessment of splenic DVHs before the acceptance of treatment plans may minimize the risk of severe post-CRT lymphopenia.


Subject(s)
Chemoradiotherapy/adverse effects , Lymphopenia/etiology , Organs at Risk/radiation effects , Pancreatic Neoplasms/therapy , Spleen/radiation effects , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Female , Humans , Induction Chemotherapy , Logistic Models , Lymphocyte Count , Lymphopenia/mortality , Male , Middle Aged , Odds Ratio , Proportional Hazards Models , Radiotherapy Dosage , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...