Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Cancer Chemother Pharmacol ; 92(2): 151-155, 2023 08.
Article in English | MEDLINE | ID: mdl-37369852

ABSTRACT

BACKGROUND: Rectal carcinomas are tumors that arise from the last 12 cm of the large intestine closest to the anus. They generally have a modest prognosis exacerbated by a high local recurrence rate if radiosensitizing chemotherapy is not given during radiotherapy. This case report discusses the clinical trial treatment of a patient with rectal adenocarcinoma by a new ropidoxuridine-capecitabine-radiotherapy combination. This case report is novel due to the patient's participation in an accelerated titration phase I clinical trial and the resultant rare adverse event of treatment-related sigmoid typhlitis. CASE PRESENTATION: The patient was an 82-year-old female who noticed hematochezia and change in stool caliber over a period of 3 months. A rectal mass was identified by biopsy as a microsatellite stable adenocarcinoma. A planned total neoadjuvant treatment involved eight cycles of leucovorin calcium (folinic acid)-fluorouracil-oxaliplatin (mFOLFOX6) chemotherapy, followed by a clinical trial combination of ropidoxuridine-capecitabine-radiotherapy, prior to definitive surgery. The patient began daily intensity modulated pelvic radiotherapy with concurrent twice-daily oral ropidoxuridine and twice-daily oral capecitabine to be given over 6 weeks. After 14 days of ropidoxuridine-capecitabine-radiotherapy, the patient developed sigmoid typhlitis requiring a 10-day hospitalization and 14-day disruption of treatment. The patient died 27 days after the start of ropidoxuridine-capecitabine-radiotherapy. This adverse event was listed as a definite attribution to the ropidoxuridine-capecitabine treatment; pharmacokinetic and pharmacodynamic data showed low ropidoxuridine metabolite DNA incorporation and high capecitabine metabolite concentration. The accelerated titration phase I clinical trial has been subsequently closed to accrual (NCT04406857). CONCLUSIONS: We believe this case report demonstrates the decision-making process for terminating a phase I accelerated titration designed clinical trial. The report also presents the rare complication of sigmoid typhlitis as a treatment-attributed adverse event. In this case, a ropidoxuridine-capecitabine combination was used as an investigational radiosensitizing treatment now with a narrower future clinical development pathway.


Subject(s)
Adenocarcinoma , Rectal Neoplasms , Typhlitis , Female , Humans , Aged, 80 and over , Capecitabine , Fluorouracil , Typhlitis/drug therapy , Typhlitis/etiology , Typhlitis/pathology , Rectal Neoplasms/drug therapy , Rectal Neoplasms/radiotherapy , Neoadjuvant Therapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Leucovorin , Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Neoplasm Staging
2.
Front Oncol ; 13: 1179316, 2023.
Article in English | MEDLINE | ID: mdl-37025589
3.
Front Oncol ; 10: 586232, 2020.
Article in English | MEDLINE | ID: mdl-33335855

ABSTRACT

PURPOSE: To reduce patient and procedure identification errors by human interactions in radiotherapy delivery and surgery, a Biometric Automated Patient and Procedure Identification System (BAPPIS) was developed. BAPPIS is a patient identification and treatment procedure verification system using fingerprints. METHODS: The system was developed using C++, the Microsoft Foundation Class Library, the Oracle database system, and a fingerprint scanner. To register a patient, the BAPPIS system requires three steps: capturing a photograph using a web camera for photo identification, taking at least two fingerprints, and recording other specific patient information including name, date of birth, allergies, etc. To identify a patient, the BAPPIS reads a fingerprint, identifies the patient, verifies with a second fingerprint to confirm when multiple patients have same fingerprint features, and connects to the patient's record in electronic medical record (EMR) systems. To validate the system, 143 and 21 patients ranging from 36 to 98 years of ages were recruited from radiotherapy and breast surgery, respectively. The registration process for surgery patients includes an additional module, which has a 3D patient model. A surgeon could mark 'O' on the model and save a snap shot of patient in the preparation room. In the surgery room, a webcam displayed the patient's real-time image next to the 3D model. This may prevent a possible surgical mistake. RESULTS: 1,271 (96.9%) of 1,311 fingerprints were verified by BAPPIS using patients' 2nd fingerprints from 143 patients as the system designed. A false positive recognition was not reported. The 96.9% completion ratio is because the operator did not verify with another fingerprint after identifying the first fingerprint. The reason may be due to lack of training at the beginning of the study. CONCLUSION: We successfully demonstrated the use of BAPPIS to correctly identify and recall patient's record in EMR. BAPPIS may significantly reduce errors by limiting the number of non-automated steps.

4.
Clin Cancer Res ; 25(20): 6035-6043, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31337643

ABSTRACT

PURPOSE: Iododeoxyuridine (IUdR) is a potent radiosensitizer; however, its clinical utility is limited by dose-limiting systemic toxicities and the need for prolonged continuous infusion. 5-Iodo-2-pyrimidinone-2'-deoxyribose (IPdR) is an oral prodrug of IUdR that, compared with IUdR, is easier to administer and less toxic, with a more favorable therapeutic index in preclinical studies. Here, we report the clinical and pharmacologic results of a first-in-human phase I dose escalation study of IPdR + concurrent radiation therapy (RT) in patients with advanced metastatic gastrointestinal (GI) cancers. PATIENTS AND METHODS: Adult patients with metastatic GI cancers referred for palliative RT to the chest, abdomen, or pelvis were eligible for study. Patients received IPdR orally once every day × 28 days beginning 7 days before the initiation of RT (37.5 Gy in 2.5 Gy × 15 fractions). A 2-part dose escalation scheme was used, pharmacokinetic studies were performed at multiple time points, and all patients were assessed for toxicity and response to Day 56. RESULTS: Nineteen patients were entered on study. Dose-limiting toxicity was encountered at 1,800 mg every day, and the recommended phase II dose is 1,200 mg every day. Pharmacokinetic analyses demonstrated achievable and sustainable levels of plasma IUdR ≥1 µmol/L (levels previously shown to mediate radiosensitization). Two complete, 3 partial, and 9 stable responses were achieved in target lesions. CONCLUSIONS: Administration of IPdR orally every day × 28 days with RT is feasible and tolerable at doses that produce plasma IUdR levels ≥1 µmol/L. These results support the investigation of IPdR + RT in phase II studies.


Subject(s)
Chemoradiotherapy/methods , Gastrointestinal Neoplasms/therapy , Idoxuridine/pharmacokinetics , Pyrimidine Nucleosides/administration & dosage , Radiation-Sensitizing Agents/administration & dosage , Administration, Oral , Adult , Aged , Aged, 80 and over , Dose Fractionation, Radiation , Feasibility Studies , Female , Gastrointestinal Neoplasms/pathology , Humans , Idoxuridine/administration & dosage , Idoxuridine/toxicity , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Prodrugs/toxicity , Pyrimidine Nucleosides/pharmacokinetics , Pyrimidine Nucleosides/toxicity , Radiation-Sensitizing Agents/pharmacokinetics , Radiation-Sensitizing Agents/toxicity , Treatment Outcome
5.
Cancer ; 125(16): 2732-2746, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31017664

ABSTRACT

Colorectal cancer (CRC) represents a major public health problem as the second leading cause of cancer-related mortality in the United States. Of an estimated 140,000 newly diagnosed CRC cases in 2018, roughly one-third of these patients will have a primary tumor located in the distal large bowel or rectum. The current standard-of-care approach includes curative-intent surgery, often after preoperative (neoadjuvant) radiotherapy (RT), to increase rates of tumor down-staging, clinical and pathologic response, as well as improving surgical resection quality. However, despite advancements in surgical techniques, as well as sharpened precision of dosimetry offered by contemporary RT delivery platforms, the oncology community continues to face challenges related to disease relapse. Ongoing investigations are aimed at testing novel radiosensitizing agents and treatments that might exploit the systemic antitumor effects of RT using immunotherapies. If successful, these treatments may usher in a new curative paradigm for rectal cancers, such that surgical interventions may be avoided. Importantly, this disease offers an opportunity to correlate matched paired biopsies, radiographic response, and molecular mechanisms of treatment sensitivity and resistance with clinical outcomes. Herein, the authors highlight the available evidence from preclinical models and early-phase studies, with an emphasis on promising developmental therapeutics undergoing prospective validation in larger scale clinical trials. This review by the National Cancer Institute's Radiation Research Program Colorectal Cancer Working Group provides an updated, comprehensive examination of the continuously evolving state of the science regarding radiosensitizer drug development in the curative treatment of CRC.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/radiotherapy , Radiation-Sensitizing Agents/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Biological Products , HSP90 Heat-Shock Proteins/metabolism , Herpesvirus 1, Human , Humans , Immunotherapy/methods , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Molecular Targeted Therapy , National Cancer Institute (U.S.) , Protein Kinase C/antagonists & inhibitors , Pyrimidine Nucleosides/pharmacology , Radiation-Sensitizing Agents/pharmacology , United States
6.
Adv Radiat Oncol ; 3(3): 245-251, 2018.
Article in English | MEDLINE | ID: mdl-30202794

ABSTRACT

PURPOSE: Single-fraction radiation surgery for spine metastases is highly effective. However, a high rate (20-39%) of vertebral body fracture (VBF) has been associated with large, single-fraction doses. We report our experience using multifraction stereotactic body radiation therapy (SBRT). METHODS AND MATERIALS: All patients who were treated with multifraction SBRT for spine metastases at our institution between 2009 and 2017 were retrospectively analyzed. SBRT was delivered in 2 to 5 fractions using the Cyberknife System (Accuray, Sunnyvale, CA). Patients were followed clinically and with magnetic resonance imaging every 3 to 6 months. Local control, complications (including VBF), and overall survival were evaluated. Patient, disease, and treatment variables were analyzed for a statistical association with outcomes. RESULTS: A total of 83 patients were treated to 98 spine lesions with a median follow-up of 7.6 months. Histologies included non-small cell lung cancer (NSCLC; 24%), renal cell carcinoma (RCC; 18%), and breast cancer (12%). Surgery or vertebroplasty were performed before SBRT in 21% of cases. Patients received a median SBRT dose of 24 Gy in a median of 3 fractions. Local control was 93% at 6 months and 84% at 1 year. Higher prescribed dose, higher biologic effective dose, higher minimum dose to 90% of the planning target volume, tumor histology, and smaller tumor volume predicted improved local control. The cumulative dose was 23 Gy versus 26 Gy for patients with and without failure (P = .02), higher biologic effective dose 39 Gy versus 46 Gy, (P = .01), and higher minimum dose to 90% of the planning target volume 23 Gy versus 26 Gy (P = .03). VBF occurred in 4.2% of all cases and 5.3% of those without surgery or vertebroplasty prior to SBRT. Only preexisting VBF predicted risk of post-SBRT VBF (P < .01). CONCLUSIONS: Multifraction SBRT results in a high local control rate for metastatic spinal disease with a low VBF rate, which suggests a favorable therapeutic ratio compared with single-fraction SBRT.

7.
World Neurosurg ; 104: 589-593, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28450235

ABSTRACT

PURPOSE/OBJECTIVES: The outcomes of repeat stereotactic radiosurgery (SRS) after failure of previous SRS are not well established. We report our overall experience using SRS for the retreatment of locally recurrent brain metastases. METHODS: Patients with brain metastases diagnosed between 2003 and 2015 who underwent repeat SRS for local tumor progression following prior SRS were identified. Rates of local control, radiation necrosis, and overall survival were analyzed. Factors affecting local failure and radiation necrosis were assessed by chi-square test. RESULTS: Twenty-four lesions in 22 patients underwent repeat SRS in a single fraction. Median age was 59 years. The median SRS-1 dose was 18 Gy, and the median SRS-2 dose was 15.5 Gy. The median SRS-1 target volume was 2.25 cm3, and the median SRS-2 target volume was 3.30 cm3. The median follow-up from SRS-2 was 8.8 months. The actuarial local controls for SRS-2 were 94.1% and 61.1% at 6 and 12 months, respectively. The incidences of actuarial radiation necrosis were 9.2% and 9.2% at 6 and 12 months, respectively. Volume of tumor >4 cm3 correlated with increased risk of local failure (P = 0.006) with no local failures recorded with volumes ≤4 cm3. SRS-2 dose, cumulative SRS dose, receipt of whole brain radiotherapy, and use of SRS-2 as boost after surgery did not correlate with local failure or radiation necrosis. Median overall survival after SRS-2 was 8.78 months. CONCLUSION: Repeat SRS is feasible for select patients, particularly for those with tumor volume ≤4 cm3. Further evaluation is needed to establish the most appropriate treatment doses and volumes for this approach.


Subject(s)
Brain Neoplasms/secondary , Brain Neoplasms/surgery , Neoplasm Recurrence, Local/surgery , Radiosurgery/methods , Adult , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/mortality , Retrospective Studies , Survival Analysis , Treatment Failure , Tumor Burden
8.
World Neurosurg ; 98: 266-272, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27838432

ABSTRACT

BACKGROUND: Stereotactic radiosurgery (SRS) alone is an attractive option for treatment of brain metastases. SRS avoids whole-brain radiotherapy (WBRT)-associated morbidity, but is limited by regional central nervous system (CNS) failures and short survival in some patients. We evaluated a subgroup of patients with controlled systemic disease that could represent a favorable patient population for SRS alone. METHODS: All patients with brain metastases treated with SRS without WBRT at our institution between 2004 and 2014 were grouped into two cohorts: those with controlled systemic disease (CSD) for 1 year or longer before prior to presentation with brain metastases and those without (i.e., uncontrolled systemic disease [USD]). Rates of local and regional CNS failure, and overall survival were assessed with χ2 and Student t tests. Cox regression analysis was performed to evaluate independent predictors of regional control and overall survival. RESULTS: Two hundred ninety-four patients underwent SRS to 697 lesions, of which 65 patients had CSD. Median follow-up was 9.7 months. There was no difference in local control between the two cohorts (P = 0.795). Regional CNS control was significantly better for patients with CSD (68% vs. 48%; P = 0.001). Overall survival at 1 and 5 years for CSD were 65% and 13% with USD yielding 41% and 7%, respectively (P < 0.001). Multivariate analysis demonstrated that USD (relative CSD) independently predicts regional failure (hazard ratio [HR], 1.75; P = 0.008) and shorter overall survival (HR, 1.55; P = 0.007). CONCLUSIONS: Patients with brain metastases after 1 year or longer of primary and systemic disease control represent a particularly favorable cohort, with lower regional CNS failure and prolonged survival, for an approach of SRS alone.


Subject(s)
Brain Neoplasms/diagnostic imaging , Brain Neoplasms/radiotherapy , Radiosurgery/methods , Adult , Aged , Aged, 80 and over , Brain Neoplasms/secondary , Cohort Studies , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis , Radiosurgery/trends , Retrospective Studies , Time Factors , Treatment Outcome
9.
Front Oncol ; 6: 203, 2016.
Article in English | MEDLINE | ID: mdl-27672621

ABSTRACT

INTRODUCTION: Sacral chordomas represent one half of all chordomas, a rare neoplasm of notochordal remnants. Current NCCN guidelines recommend surgical resection with or without adjuvant radiotherapy or definitive radiation for unresectable cases. Recent advances in radiation for chordomas include conformal photon and proton beam radiation. We investigated incidence, treatment, and survival outcomes to observe any trends in response to improvements in surgical and radiation techniques over a near 40-year time period. MATERIALS AND METHODS: Three hundred forty-five microscopically confirmed cases of sacral chordoma were identified between 1974 and 2011 from the surveillance, epidemiology, and end results program of the National Cancer Institute. Cases were divided into three cohorts by calendar year, 1974-1989, 1990-1999, and 2000-2011, as well as into two groups by age ≤65 versus >65 to investigate trends over time and age via Chi-square analysis. Kaplan-Meier analyses were performed to determine effects of treatment on survival. Multivariate Cox regression analysis was performed to determine predictors of overall survival (OS). RESULTS: Five-year OS for the entire cohort was 60.0%. OS correlated significantly with treatment modality, with 44% surviving at 5 years with no treatment, 52% with radiation alone, 82% surgery alone, and 78% surgery and radiation (p < 0.001). Age >65 was significantly associated with non-surgical management with radiation alone or no treatment (p < 0.001). Relatively, fewer patients received radiation between 2000 and 2011 compared to prior time periods (p = 0.03) versus surgery, for which rates which did not vary significantly over time (p = 0.55). However, 5-year OS was not significantly different by time period. Age group and treatment modality were predictive for OS on multivariate analysis (p < 0.001). CONCLUSION: Surgery remains an important component in the treatment of sacral chordomas in current practice. Fewer patients were treated with radiation more recently despite advances in photon and proton beam radiation. OS remains unchanged. Additional analyses of margin status, radiation modality, and local control in current practice are warranted.

10.
Adv Radiat Oncol ; 1(4): 294-299, 2016.
Article in English | MEDLINE | ID: mdl-28740900

ABSTRACT

PURPOSE: Many patients treated with stereotactic radiosurgery (SRS) alone as initial treatment require 1 or more subsequent salvage therapies. This study aimed to determine if commonly used salvage strategies are associated with differing risks of radiation necrosis (RN). METHODS AND MATERIALS: All patients treated with upfront SRS alone for brain metastases at our institution were retrospectively analyzed. Salvage treatment details were obtained for brain failures. Patients who underwent repeat SRS to the same lesion were excluded. RN was determined based on pathological confirmation or advanced brain imaging consistent with RN in a symptomatic patient. Patients were grouped according to salvage treatment and rates of RN were compared via Fisher's exact tests. RESULTS: Of 284 patients treated with upfront SRS alone, 132 received salvage therapy and 44 received multiple salvage treatments. This included 31 repeat SRS alone, 58 whole brain radiation therapy (WBRT) alone, 28 SRS and WBRT, 7 surgery alone, and 8 surgery with adjuvant radiation. With a median follow-up of 10 months, the rate of RN among all patients was 3.17% (9/284), salvaged patients 4.55% (6/132), and never salvaged patients 1.97% (3/152). Receiving salvage therapy did not significantly increase RN risk (P = .31). Of the patients requiring salvage treatments, the highest RN rate was among patients that had both salvage SRS and WBRT (delivered as separate salvage therapies) (6/28, 21.42%). RN rate in this group was significantly higher than in those treated with repeat SRS alone (0/31), WBRT alone (0/58), surgery alone (0/7), and surgery with adjuvant radiation (0/8). Comparing salvage WBRT doses <30 Gy versus ≥30 Gy revealed no effect of dose on RN rate. Additionally, among patients who received multiple SRS treatments, number of treated lesions was not predictive of RN incidence. CONCLUSION: Our results suggest that initial management approach for recurrent brain metastasis after upfront SRS does not affect the rate of RN. However, the risk of RN significantly increases when patients are treated with both repeat SRS and salvage WBRT. Methods to improve prediction of toxicity and optimize patient selection for salvage treatments are needed.

11.
R I Med J (2013) ; 98(11): 17-9, 2015 Nov 02.
Article in English | MEDLINE | ID: mdl-26517249

ABSTRACT

Central nervous system (CNS) tumors are a heterogeneous group of neoplasms divided into two broad categories, glial and non-glial. Non-glial tumors are derived from such diverse structures as the pineal gland, meninges, germ cells, and hematopoietic cells, as well as metastases. Primary glial neoplasms, or those which originate from astrocytes, oligodendrocytes, or ependymal cells, include astrocytomas, oligodendrogliomas, ependymomas, and mixed gliomas. Each entity has a unique morphology and pattern of biologic behavior which portends a distinct prognosis and outcome. Individual outcomes show some variability based on tumor location and age of symptom onset; however, the underlying aggressiveness of the tumor often dictates the time course of the disease. With the advent and widespread use of fluorescent in-situ hybridization and polymerase chain reaction (PCR) techniques, molecular phenotyping of brain tumors has become mainstream and is now an integral part of patient care. The molecular genetics of CNS tumors is a rapidly growing field, and the volume of discoveries is growing at an ever increasing rate, compelling the need for updates in this exciting area of science.


Subject(s)
Biomarkers, Tumor , Central Nervous System Neoplasms/pathology , Glioma/pathology , Adult , Central Nervous System Neoplasms/genetics , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Glioma/genetics , Humans , Isocitrate Dehydrogenase/genetics , Prognosis , Tumor Suppressor Proteins/genetics
12.
Front Oncol ; 5: 110, 2015.
Article in English | MEDLINE | ID: mdl-26029666

ABSTRACT

PURPOSE: To identify predictors for prolonged survival free from salvage whole brain radiation therapy (WBRT) in patients with brain metastases treated with stereotactic radiosurgery (SRS) as their initial radiotherapy approach. MATERIALS AND METHODS: Patients with brain metastases treated with SRS from 2001 to 2013 at our institution were identified. SRS without WBRT was typically offered to patients with 1-4 brain metastases, Karnofsky performance status ≥70, and life expectancy ≥3 months. Three hundred and eight patients met inclusion criteria for analysis. Medical records were reviewed for patient, disease, and treatment information. Two comparison groups were identified: those with ≥1-year WBRT-free survival (N = 104), and those who died or required salvage WBRT within 3 months of SRS (N = 56). Differences between these groups were assessed by univariate and multivariate analyses. RESULTS: Median survival for all patients was 11 months. Among patients with ≥1-year WBRT-free survival, median survival was 33 months (12-107 months) with only 21% requiring salvage WBRT. Factors significantly associated with prolonged WBRT-free survival on univariate analysis (p < 0.05) included younger age, asymptomatic presentation, RTOG RPA class I, fewer brain metastases, surgical resection, breast primary, new or controlled primary, absence of extracranial metastatic disease, and oligometastatic disease burden (≤5 metastatic lesions). After controlling for covariates, asymptomatic presentation, breast primary, single brain metastasis, absence of extracranial metastases, and oligometastatic disease burden remained independent predictors for favorable WBRT-free survival. CONCLUSION: A subset of patients with brain metastases can achieve long-term survival after upfront SRS without the need for salvage WBRT. Predictors identified in this study can help select patients that might benefit most from a treatment strategy of SRS alone.

13.
Am J Clin Oncol ; 37(5): 444-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-23388562

ABSTRACT

OBJECTIVES: Paclitaxel poliglumex (PPX), a drug conjugate that links paclitaxel to poly-L-glutamic acid, is a potent radiation sensitizer. Prior studies in esophageal cancer have demonstrated that PPX (50 mg/m/wk) can be administered with concurrent radiation with acceptable toxicity. The primary objective of this study was to determine the safety of the combination of PPX with temozolomide and concurrent radiation for high-grade gliomas. METHODS: Eligible patients were required to have WHO grade 3 or 4 gliomas. Patients received weekly PPX (50 mg/m/wk) combined with standard daily temozolomide (75 mg/m) for 6 weeks with concomitant radiation (2.0 Gy, 5 d/wk for a total dose of 60 Gy). RESULTS: Twenty-five patients were enrolled, 17 with glioblastoma and 8 with grade 3 gliomas. Seven of 25 patients had grade 4 myelosuppression. Hematologic toxicity lasted up to 5 months suggesting a drug interaction between PPX and temozolomide. For patients with glioblastoma, the median progression-free survival was 11.5 months and the median overall survival was 18 months. CONCLUSIONS: PPX could not be safely combined with temozolomide due to grade 4 hematologic toxicity. However, the favorable progression-free and overall survival suggest that PPX may enhance radiation for glioblastoma. A randomized study of single agent PPX/radiation versus temozolomide/radiation for glioblastoma without MGMT methylation is underway.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brachytherapy , Brain Neoplasms/therapy , Chemoradiotherapy/methods , Glioma/therapy , Adult , Aged , Brain Neoplasms/mortality , Combined Modality Therapy , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Disease-Free Survival , Female , Glioma/mortality , Humans , Male , Middle Aged , Paclitaxel/administration & dosage , Paclitaxel/analogs & derivatives , Polyglutamic Acid/administration & dosage , Polyglutamic Acid/analogs & derivatives , Survival Analysis , Temozolomide
14.
Front Oncol ; 3: 288, 2013.
Article in English | MEDLINE | ID: mdl-24324931

ABSTRACT

PURPOSE: Pre-operative chemoradiation (CRT) is currently the standard of care for patients with clinical stage II and III rectal cancer but only about 45% of patients achieve tumor downstaging and <20% of patients achieve a pathologic complete response. Better methods to stratify patients according to potential neoadjuvant treatment response are needed. We used microarray analysis to identify a genetic signature that correlates with a pathological complete response (pCR) to neoadjuvant CRT. We performed a gene network analysis to identify potential signaling pathways involved in determining response to neoadjuvant treatment. PATIENTS AND METHODS: We identified 31 T3-4 N0-1 rectal cancer patients who were treated with neoadjuvant fluorouracil-based CRT. Eight patients were identified to have achieved a pCR to treatment while 23 patients did not. mRNA expression was analyzed using cDNA microarrays. The correlation between mRNA expression and pCR from pre-treatment tumor biopsies was determined. Gene network analysis was performed for the genes represented by the predictive signature. RESULTS: A genetic signature represented by expression levels of the three genes EHBP1, STAT1, and GAPDH was found to correlate with a pCR to neoadjuvant treatment. The difference in expression levels between patients who achieved a pCR and those who did not was greatest for EHBP1. Gene network analysis showed that the three genes can be connected by the gene ubiquitin C (UBC). CONCLUSION: This study identifies a 3-gene signature expressed in pre-treatment tumor biopsies that correlates with a pCR to neoadjuvant CRT in patients with clinical stage II and III rectal cancer. These three genes can be connected by the gene UBC, suggesting that ubiquitination is a molecular mechanism involved in determining response to treatment. Validating this genetic signature in a larger number of patients is proposed.

15.
IET Syst Biol ; 7(4): 114-24, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23919954

ABSTRACT

DNA mismatch repair (MMR) is involved in processing DNA damage following treatment with ionising radiation (IR) and various classes of chemotherapy drugs including iododeoxyuridine (IUdR), a known radiosensitiser. In this study, the authors have developed asynchronous probabilistic cell cycle models to assess the isolated effects of IUdR and IR and the combined effects of IUdR + IR treatments on MMR damage processing. The authors used both synchronous and asynchronous MMR-proficient/MMR-deficient cell populations and followed treated cells for up to two cell cycle times. They have observed and quantified differential cell cycle responses to MMR damage processing following IR and IUdR + IR treatments, principally in the duration of both G1 and G2/M cell cycle phases. The models presented in this work form the foundation for the development of an approach to maximise the therapeutic index for IR and IUdR + IR treatments in MMR-deficient (damage tolerant) cancers.


Subject(s)
Colorectal Neoplasms/therapy , DNA Mismatch Repair , Idoxuridine/pharmacology , Cell Cycle/drug effects , Cell Cycle/radiation effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , HCT116 Cells , Humans , Models, Biological , Radiation, Ionizing , Radiation-Sensitizing Agents/pharmacology , Systems Biology
16.
IET Syst Biol ; 7(2): 27-37, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23847811

ABSTRACT

Base excision repair (BER) is a major DNA repair pathway involved in the processing of exogenous non-bulky base damages from certain classes of cancer chemotherapy drugs as well as ionising radiation (IR). Methoxyamine (MX) is a small molecule chemical inhibitor of BER that is shown to enhance chemotherapy and/or IR cytotoxicity in human cancers. In this study, the authors have analysed the inhibitory effect of MX on the BER pathway kinetics using a computational model of the repair pathway. The inhibitory effect of MX depends on the BER efficiency. The authors have generated variable efficiency groups using different sets of protein concentrations generated by Latin hypercube sampling, and they have clustered simulation results into high, medium and low efficiency repair groups. From analysis of the inhibitory effect of MX on each of the three groups, it is found that the inhibition is most effective for high efficiency BER, and least effective for low efficiency repair.


Subject(s)
DNA Repair/drug effects , DNA Repair/genetics , DNA, Neoplasm/genetics , Hydroxylamines/administration & dosage , Models, Biological , Neoplasms/genetics , Neoplasms/therapy , Animals , Antineoplastic Agents/administration & dosage , Computer Simulation , DNA Repair/radiation effects , Drug Design , Drug Therapy, Computer-Assisted/methods , Humans , Molecular Targeted Therapy/methods
17.
Front Oncol ; 1: 7, 2011.
Article in English | MEDLINE | ID: mdl-22645711
18.
Front Oncol ; 1: 14, 2011.
Article in English | MEDLINE | ID: mdl-22649754

ABSTRACT

Photodynamic therapy (PDT) for cutaneous malignancies has been found to be an effective treatment with a range of photosensitizers. The phthalocyanine Pc 4 was developed initially for PDT of primary or metastatic cancers in the skin. A Phase I trial was initiated to evaluate the safety and pharmacokinetic profiles of systemically administered Pc 4 followed by red light (Pc 4-PDT) in cutaneous malignancies. A dose-escalation study of Pc 4 (starting dose 0.135 mg/m(2)) at a fixed light fluence (135 J/cm(2) of 675-nm light) was initiated in patients with primary or metastatic cutaneous malignancies with the aim of establishing the maximum tolerated dose (MTD). Blood samples were taken at intervals over the first 60 h post-PDT for pharmacokinetic analysis, and patients were evaluated for toxicity and tumor response. A total of three patients (two females with breast cancer and one male with cutaneous T-cell lymphoma) were enrolled and treated over the dose range of 0.135 mg/m(2) (first dose level) to 0.54 mg/m(2) (third dose level). Grade 3 erythema within the photoirradiated area was induced in patient 2, and transient tumor regression in patient 3, in spite of the low photosensitizer doses. Pharmacokinetic observations fit a three-compartment exponential elimination model with an initial rapid distribution phase (∼0.2 h) and relatively long terminal elimination phase (∼28 h), Because of restrictive exclusion criteria and resultant poor accrual, the trial was closed before MTD could be reached. While the limited accrual to this initial Phase I study did not establish the MTD nor establish a complete pharmacokinetic and safety profile of intravenous Pc 4-PDT, these preliminary data support further Phase I testing of this new photosensitizer.

19.
Front Oncol ; 1: 20, 2011.
Article in English | MEDLINE | ID: mdl-22649757

ABSTRACT

Over the last 7 years, we have focused our experimental and computational research efforts on improving our understanding of the biochemical, molecular, and cellular processing of iododeoxyuridine (IUdR) and ionizing radiation (IR) induced DNA base damage by DNA mismatch repair (MMR). These coordinated research efforts, sponsored by the National Cancer Institute Integrative Cancer Biology Program (ICBP), brought together system scientists with expertise in engineering, mathematics, and complex systems theory and translational cancer researchers with expertise in radiation biology. Our overall goal was to begin to develop computational models of IUdR- and/or IR-induced base damage processing by MMR that may provide new clinical strategies to optimize IUdR-mediated radiosensitization in MMR deficient (MMR(-)) "damage tolerant" human cancers. Using multiple scales of experimental testing, ranging from purified protein systems to in vitro (cellular) and to in vivo (human tumor xenografts in athymic mice) models, we have begun to integrate and interpolate these experimental data with hybrid stochastic biochemical models of MMR damage processing and probabilistic cell cycle regulation models through a systems biology approach. In this article, we highlight the results and current status of our integration of radiation biology approaches and computational modeling to enhance IUdR-mediated radiosensitization in MMR(-) damage tolerant cancers.

20.
Front Oncol ; 1: 8, 2011.
Article in English | MEDLINE | ID: mdl-22655228

ABSTRACT

A more flexible and higher-yielding in vitro DNA mismatch repair (MMR) substrate construction method, which was developed initially by Wang and Hays, is described for the construction of a nucleotide-based chemical mismatch (G/IU) and a G/T mismatch. Our modifications use the combination of two endonuclease enzymes (NheI and BciVI) and two new redesigned plasmids (pWDAH1A and pWDSH1B). In our modified methodology, plasmids are initially digested with the nicking endonucleases, followed by the streptavidin treatment. The mismatch-containing oligo is then annealed to the gap DNA and finally ligated to produce a mismatch-containing DNA substrate. We report a high efficiency (up to 90%) of these mismatch substrates and confirm recognition using a functional assay. These modifications, coupled with the use of the redesigned plasmids, can be applied for the construction of other types of chemically induced mismatches as well as insertion-deletion loops for future in vitro studies of MMR processing by our group and others.

SELECTION OF CITATIONS
SEARCH DETAIL
...