Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Xenobiotica ; 40(11): 730-42, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20836725

ABSTRACT

PF-592379 is a potent, selective agonist of the dopamine 3 receptor, for the treatment of male erectile dysfunction and female sexual dysfunction. In vivo, PF-592379 has low-moderate clearance relative to liver blood flow of 6.3 and 8.5 ml/min/kg in dog and 44.8 and 58.2 ml/min/kg in rat. It has high permeability in Caco-2 cells and was completely absorbed in rat and dog pharmacokinetic studies with an oral bioavailability of 28% in both rats and 61 and 87% in the dogs. These data are consistent with the physicochemical properties of PF-592379, which indicate complete absorption by the transcellular route. Elimination of PF-592379 was predominantly metabolic in nature. In vitro routes of metabolism studies indicate that metabolism in the rat is a combination of P450 mechanisms and N-glucuronidation, whereas in dog and human, N-glucuronidation is the major route. NMR analysis indicates that N-glucuronidation is non-quaternary in nature and occurs on both the pyridyl amine and ring nitrogen. Rates of clearance via N-glucuronidation were predicted to be low in humans compared with acyl or phenolic glucuronidation. PF-592379 was predicted to have complete absorption from the gastrointestinal tract and an oral bioavailability of >60% in the clinic. Clinical data verified that PF-592379 is a low clearance compound in human, with a mean oral clearance of 6.5 ml/min/kg following a 200 mg oral dose. PF-592379 has ideal pharmacokinetic properties for an oral D3 agonist, intended for on demand dosing.


Subject(s)
Aminopyridines/pharmacokinetics , Dopamine Agonists/pharmacokinetics , Glucuronosyltransferase/metabolism , Morpholines/pharmacokinetics , Receptors, Dopamine D3/agonists , Administration, Oral , Aminopyridines/administration & dosage , Aminopyridines/metabolism , Animals , Biometry , Blood Proteins/metabolism , Caco-2 Cells , Dogs , Dopamine Agonists/administration & dosage , Dopamine Agonists/metabolism , Drug Evaluation, Preclinical , Erectile Dysfunction/drug therapy , Female , Humans , Infusions, Intravenous , Male , Metabolic Clearance Rate , Microsomes, Liver/metabolism , Morpholines/administration & dosage , Morpholines/metabolism , Rats , Rats, Sprague-Dawley
2.
Clin Pharmacol Ther ; 103(2): 174-176, 2018 02.
Article in English | MEDLINE | ID: mdl-29274086

ABSTRACT

If we are to improve our low success rate and rising costs in the pharmaceutical industry, we need to use every tool available. Reverse translation can particularly inform discovery and early clinical development via appropriate quantitative integration of relevant data. This commentary reports on a crowd-sourced survey (2017) that sought to evaluate the integration of reverse translation in pharma. The results indicate that these methods are being applied, to varying degrees, across most respondents.


Subject(s)
Data Mining/methods , Drug Development/methods , Drug Discovery/methods , Drug Industry/methods , Evidence-Based Medicine/methods , Translational Research, Biomedical/methods , Animals , Databases, Factual , Humans , Learning , Models, Animal , Models, Theoretical , Patient Safety , Risk Assessment , Stakeholder Participation , Surveys and Questionnaires
3.
Stat Methods Med Res ; 27(9): 2694-2721, 2018 09.
Article in English | MEDLINE | ID: mdl-28067121

ABSTRACT

Characterizing clinical dose-response is a critical step in drug development. Uncertainty in the dose-response model when planning a dose-ranging study can often undermine efficiency in both the design and analysis of the trial. Results of a previous meta-analysis on a portfolio of small molecule compounds from a large pharmaceutical company demonstrated a consistent dose-response relationship that was well described by the maximal effect model. Biologics are different from small molecules due to their large molecular sizes and their potential to induce immunogenicity. A model-based meta-analysis was conducted on the clinical efficacy of 71 distinct biologics evaluated in 91 placebo-controlled dose-response studies published between 1995 and 2014. The maximal effect model, arising from receptor occupancy theory, described the clinical dose-response data for the majority of the biologics (81.7%, n = 58). Five biologics (7%) with data showing non-monotonic trend assuming the maximal effect model were identified and discussed. A Bayesian model-based hierarchical approach using different joint specifications of prior densities for the maximal effect model parameters was used to meta-analyze the whole set of biologics excluding these five biologics ( n = 66). Posterior predictive distributions of the maximal effect model parameters were reported and they could be used to aid the design of future dose-ranging studies. Compared to the meta-analysis of small molecules, the combination of fewer doses, narrower dosing ranges, and small sample sizes further limited the information available to estimate clinical dose-response among biologics.


Subject(s)
Biological Factors/administration & dosage , Dose-Response Relationship, Drug , Drug Dosage Calculations , Algorithms , Bayes Theorem , Humans , Models, Statistical
4.
J Pharm Biomed Anal ; 43(2): 666-76, 2007 Jan 17.
Article in English | MEDLINE | ID: mdl-16971087

ABSTRACT

An in vitro cell-based bioassay was developed and validated to assess the pharmacokinetic profiles of two novel therapeutic recombinant proteins (EP1 and EP2) with erythropoiesis stimulating properties in Sprague-Dawley rats. While immunoassays are the standard choice for evaluating the pharmacokinetic parameters of drugs, no immunoassay was available for EP2, necessitating the need for a quantitative bioassay capable of measuring both EP1 and EP2 separately so that appropriate comparisons could be made. The bioassay described here utilizes a sub clone of the murine 32D cell line transfected with the gene encoding for the human erythopoietin (HuEPO) receptor. Erythropoietin (EPO), EP1 and EP2 exert their proliferative effect on the cell line by signaling through the HuEPO receptor. The proliferation induced by the erythropoietic proteins was measured by [methyl-(3)H]thymidine incorporation into the cellular DNA. The assay was conducted in 96-well microtiter plates and had relatively high throughput. The Guidelines of the International Conference on Harmonization (ICH) were followed for the validation of the different assay parameters including robustness, linearity, accuracy, precision, limit of quantitation (LOQ) and specificity. The robustness of the bioassay is demonstrated by the lack of an effect of age of the 32D cell culture on the performance of the EP2 bioassay. The bioassay demonstrated good linearity, yielding a coefficient of determination of 0.99 or higher for both EP1 and EP2. The assay showed reproducible dose-response curves for EP1 in the range of 0.039-2.5 ng/mL and for EP2 in the range of 0.125-8 ng/mL. The accuracy estimates ranged between 98% and 108% for EP1 and between 90% and 110% for EP2 in the reproducible range mentioned above. Intermediate precision (within-plate R.S.D.) in the same range was within 26% and 17% for the EP1 and EP2 bioassays, respectively. The validated bioassays for EP1 and EP2 were utilized to quantitatively analyze serum samples from a pharmacokinetic study conducted to compare the profiles of the two compounds in Sprague-Dawley rats.


Subject(s)
Biological Assay/methods , Cell Proliferation/drug effects , Erythropoiesis/drug effects , Receptors, Erythropoietin/drug effects , Recombinant Proteins/pharmacokinetics , Animals , Cell Line , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Guidelines as Topic , Linear Models , Male , Mice , Random Allocation , Rats , Rats, Sprague-Dawley , Recombinant Proteins/blood , Recombinant Proteins/pharmacology , Reproducibility of Results , Sensitivity and Specificity , Transfection
5.
Clin Pharmacokinet ; 45(2): 199-211, 2006.
Article in English | MEDLINE | ID: mdl-16485917

ABSTRACT

BACKGROUND AND OBJECTIVE: The pharmacokinetics of darbepoetin alfa after intravenous (IV) administration in the oncology setting have not been previously reported. The objective of this study was to evaluate the pharmacokinetics of IV or subcutaneous (SC) darbepoetin alfa in patients with non-myeloid malignancies undergoing multicycle chemotherapy. METHODS: Fifty-six patients (haemoglobin

Subject(s)
Erythropoietin/analogs & derivatives , Neoplasms/metabolism , Absorption , Adult , Aged , Area Under Curve , Darbepoetin alfa , Erythropoietin/administration & dosage , Erythropoietin/pharmacokinetics , Erythropoietin/therapeutic use , Female , Half-Life , Humans , Injections, Intravenous , Injections, Subcutaneous , Male , Metabolic Clearance Rate , Middle Aged , Neoplasms/drug therapy , Tissue Distribution
6.
MAbs ; 8(5): 983-90, 2016 07.
Article in English | MEDLINE | ID: mdl-27049478

ABSTRACT

IMA-638 and IMA-026 are humanized IgG1 monoclonal antibodies (mAbs) that target non-overlapping epitopes of IL-13. Separate first-in-human single ascending dose studies were conducted for each mAb. These studies had similar study designs, but mild to moderate asthmatics were recruited for the IMA-638 study and healthy subjects were recruited for the IMA-026 study. IMA-638 and IMA-026 showed similar pharmacokinetic (PK) profiles, but very different total IL-13 (free and drug bound IL-13) profiles; free IL13 was not measured. IMA-026 treatment induced a dose-dependent accumulation of total IL-13, while IMA-638 treatment led to a much smaller accumulation without any clear dose-response. To understand the differences between the two total IL-13 profiles and to predict the free IL-13 profiles for each mAb treatment, a mechanistic PK/pharmacodynamic model was developed. PK-related parameters were first fit to the mean PK profiles of each mAb separately; thereafter, the target-related parameters were fit to both total IL-13 profiles simultaneously. The IL-13 degradation rate was assumed to be the same for asthma patients and healthy subjects. The model suggests that an approximately 100× faster elimination of IL-13-IMA-638 complex than IL-13-IMA-026 complex could be responsible for the differences observed in total IL-13 profiles for the two mAbs. Furthermore, the model predicts that IMA-638 administration results in greater and more prolonged free IL-13 inhibition than equivalent dosing of IMA-026 despite similar binding KD and PK profile. In conclusion, joint modeling of two similar molecules provided mechanistic insight that the elimination rate of mAb-target complex can regulate the degree of free target inhibition.


Subject(s)
Anti-Asthmatic Agents/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal/pharmacokinetics , Asthma/drug therapy , Interleukin-13/antagonists & inhibitors , Models, Theoretical , Adult , Female , Humans , Immunoglobulin G/pharmacology , Male
8.
Clin Pharmacol Ther ; 72(5): 546-55, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12426518

ABSTRACT

OBJECTIVE: The aim of this multicenter, randomized, open-label study was to compare the pharmacokinetic and pharmacodynamic profiles of darbepoetin alfa, a new erythropoiesis-stimulating protein, and recombinant human erythropoietin (epoetin) after repeated intravenous dosing in patients with chronic kidney disease receiving hemodialysis. METHODS: Forty-seven patients were randomized to receive darbepoetin alfa administered once weekly (n = 17) or 3 times weekly (n = 15) or epoetin administered 3 times weekly (n = 15) for up to 52 weeks. Pharmacokinetic profiles were measured during weeks 1 and 12 and at hemoglobin steady state (defined as a hemoglobin concentration within the target range for 4 consecutive weeks after week 12 with no change in study drug dose) or between weeks 36 and 40, whichever occurred first. RESULTS: At each of the 3 time points evaluated, the terminal half-life of darbepoetin alfa was 2 to 3 times longer and the clearance approximately 4 times slower than those of epoetin. At week 12, the terminal half-life was 23.4 hours with darbepoetin alfa once weekly, 18.3 hours with darbepoetin alfa 3 times weekly, and 8.0 hours with epoetin 3 times weekly. The pharmacokinetics of darbepoetin alfa was not dependent on dose or time. Mean hemoglobin values at steady state were all approximately 11 g/dL, within the target range of 9.0 to 13.0 g/dL. Safety analyses revealed no differences between darbepoetin alfa and epoetin. CONCLUSIONS: The pharmacokinetic and pharmacodynamic profiles and safety data for darbepoetin alfa demonstrate that it can be administered less frequently than epoetin in patients with chronic kidney disease receiving hemodialysis, thus simplifying anemia management.


Subject(s)
Erythropoietin/analogs & derivatives , Erythropoietin/pharmacology , Erythropoietin/pharmacokinetics , Renal Dialysis , Adult , Aged , Darbepoetin alfa , Epoetin Alfa , Erythropoietin/adverse effects , Female , Half-Life , Hemoglobins/analysis , Humans , Male , Middle Aged , Models, Biological , Recombinant Proteins
9.
Eur J Pharmacol ; 505(1-3): 93-101, 2004 Nov 28.
Article in English | MEDLINE | ID: mdl-15556141

ABSTRACT

Studies have suggested that erythropoietin (EPO) may be used to treat stroke in both animals and humans. It is thought to exert its effects directly on the brain and studies with therapeutic doses have shown that it can cross the blood-brain barrier. Here, we compared in a blinded fashion the ability of three erythropoietic agents (murine erythropoietin, human erythropoietin, and darbepoetin alfa, an analog of human erythropoietin in clinical use) to cross the blood-brain barrier of the mouse. High-performance liquid chromatography (HPLC) results showed that all three erythropoietic agents were enzymatically resistant in brain and blood. The unidirectional blood-to-brain influx rates (Ki) as measured by multiple-time regression analysis showed that all the erythropoietic agents crossed the blood-brain barrier at about the same rate as albumin, suggesting that they cross the blood-brain barrier by way of the extracellular pathways. No saturable component to influx was found, but indirect evidence suggested a brain-to-blood efflux system. The percent of the intravenously injected dose taken up per gram of brain (%Inj/g) ranged from 0.05 to 0.1 %Inj/g among the three erythropoietic agents and peaked about 3 h after IV injection. For other substances, this range of %Inj/g is known to produce direct effects on brain function. We conclude that erythropoietic agents cross the blood-brain barrier by way of the extracellular pathways in amounts that are likely sufficient to explain their neuroprotective effects.


Subject(s)
Blood-Brain Barrier/metabolism , Erythropoietin/analogs & derivatives , Erythropoietin/pharmacokinetics , Algorithms , Animals , Brain/metabolism , Capillaries/metabolism , Chromatography, High Pressure Liquid , Darbepoetin alfa , Erythropoietin/metabolism , Humans , Iodine Radioisotopes , Male , Metabolic Clearance Rate , Mice
10.
Oncology (Williston Park) ; 16(10 Suppl 11): 37-44, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12435172

ABSTRACT

Our objective was to assess, using clinical trial simulation, the feasibility of a fixed 200-microg dose of darbepoetin alfa (Aranesp) administered every 2 weeks in chemotherapy-induced anemia. A pharmacokinetic/pharmacodynamic model was developed using clinical data from 547 cancer patients who received darbepoetin alfa at various doses and schedules. Monte Carlo simulations were performed for weight-based (3 microg/kg every 2 weeks) and fixed-dose (200 microg every 2 weeks) regimens and were compared with observed clinical data. Mean hemoglobin changes from baseline to end of treatment were +1.61 g/dL, +1.83 g/dL, and +1.79 g/dL for observed data, the weight-based simulation, and the fixed-dose simulation, respectively. The rates of required transfusions (hemoglobin < or = 8 g/dL) were also similar between groups. For patients between 45 and 95 kg (over 90% of the population), the impact of a fixed dose on mean hemoglobin change was negligible. There was a slight weight effect at body weight extremes (< 45 kg and > 95 kg). Clinical outcomes from simulations of weight-based andfixed dosing of darbepoetin alfa were similar to those of observed weight-based data. Given the weight distribution of a typical cancer population, the majority would be expected to benefit equally from weight-based and fixed-dose darbepoetin alfa in the amelioration of chemotherapy-induced anemia.


Subject(s)
Anemia/chemically induced , Anemia/drug therapy , Clinical Trials as Topic/methods , Computer Simulation , Erythropoietin/analogs & derivatives , Erythropoietin/administration & dosage , Adult , Aged , Aged, 80 and over , Anemia/blood , Body Weight/drug effects , Breast Neoplasms/blood , Breast Neoplasms/drug therapy , Darbepoetin alfa , Drug Administration Schedule , Erythropoietin/pharmacokinetics , Erythropoietin/pharmacology , Female , Gastrointestinal Neoplasms/blood , Gastrointestinal Neoplasms/drug therapy , Genital Neoplasms, Female/blood , Genital Neoplasms, Female/drug therapy , Genital Neoplasms, Male/blood , Genital Neoplasms, Male/drug therapy , Hemoglobins/metabolism , Humans , Lung Neoplasms/blood , Lung Neoplasms/drug therapy , Male , Middle Aged , Monte Carlo Method
11.
AAPS J ; 8(3): E552-63, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-17025273

ABSTRACT

Anemia is frequently observed in patients undergoing chemotherapy. Administration of darbepoetin alfa, a recombinant erythropoiesis-stimulating agent that has longer residence time than endogenous erythropoietin, to patients with chemotherapy-induced anemia (CIA) increases mean hemoglobin concentration, reduces risk of red blood cell transfusions, and improves patient-reported outcomes. A pharmacokinetic/pharmacodynamic (PkPd) model was developed using data from patients with nonmyeloid malignancies and CIA who were receiving darbepoetin alfa. A 2-compartment Pk model with linear elimination described the Pk data obtained in 140 CIA patients after intravenous and subcutaneous (s.c.) doses of 2.25 microg/kg every week and s.c. doses of 6.75 microg/kg every 3 weeks. The population typical values of key Pk parameters were clearance, 2010 mL/day; steady-state volume of distribution, 3390 mL; and bioavailability, 44.3%. A modified indirect response model, wherein serum concentrations stimulated the production of hemoglobin through an Emax-type equation, described the hemoglobin levels after s.c. doses of 0.5 microg/kg every week to 15 microg/kg every 3 weeks in 573 CIA patients. The estimated incremental maximum stimulation of hemoglobin production was 43.7% and darbepoetin alfa serum concentration at half-maximal stimulation was 3.68 ng/mL. The impact of covariates (body weight and platinum-containing chemotherapy) on the PkPd response was evaluated based on point and interval estimates of parameters, rather than through stepwise hypothesis testing. The final PkPd model adequately predicted hemoglobin response in a test data set, thereby confirming the predictive capability of the model. Based on simulations, it was not possible to categorize the influence of any covariate as clinically important.


Subject(s)
Erythropoietin/analogs & derivatives , Neoplasms/metabolism , Anemia/chemically induced , Darbepoetin alfa , Drug Therapy , Erythropoietin/adverse effects , Erythropoietin/pharmacokinetics , Evaluation Studies as Topic , Female , Humans , Male , Middle Aged , Models, Biological , Neoplasms/drug therapy , Population
12.
Pharm Res ; 23(9): 2060-6, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16951999

ABSTRACT

PURPOSE: To determine the contribution of the lymphatics to the systemic availability of darbepoetin alfa (DA) using an established sheep model. MATERIALS AND METHODS: DA was administered either by intravenous (IV) injection (0.2, 0.5 or 2 microg/kg) or by subcutaneous (SC) administration (2 microg/kg) into the interdigital space of the hind leg. A SC control group was used to determine the absolute bioavailability (F (sys)). Cannulation of the peripheral lymphatics in a parallel SC group allowed the continuous collection of lymph draining the injection site and determination of the cumulative amount of DA absorbed via the lymphatics. Serum and lymph concentrations of DA were determined by ELISA. The fraction of the dose absorbed into the lymphatics (F (lymph)) relative to the fraction absorbed directly into the blood (F (blood)) was determined using a compartmental approach. RESULTS: Dose-linear pharmacokinetics was observed within the dose range investigated. The bioavailability was virtually complete following SC injection into the interdigital space (88.4 +/- 15.7%). A high proportion of the administered dose was recovered in peripheral lymph (90.2 +/- 4.4%) resulting in a substantial reduction in the systemic availability in lymph cannulated animals (3.7%). CONCLUSION: The high recovery of DA in the peripheral lymph demonstrated near complete absorption of this recombinant protein via the lymphatics in a lymph cannulated sheep model.


Subject(s)
Erythropoietin/analogs & derivatives , Lymphatic System/metabolism , Absorption , Algorithms , Animals , Area Under Curve , Biological Availability , Darbepoetin alfa , Enzyme-Linked Immunosorbent Assay , Erythropoietin/administration & dosage , Erythropoietin/blood , Erythropoietin/pharmacokinetics , Half-Life , Injections, Intravenous , Injections, Subcutaneous , Male , Nonlinear Dynamics , Sheep
13.
J Pharmacol Exp Ther ; 313(1): 345-51, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15579493

ABSTRACT

The contribution of the lymphatics to the absorption and systemic availability of recombinant human epoetin alfa (rHuEPO) following s.c. injection was examined using a cannulated sheep model. Parallel studies were conducted in sheep where a single bolus dose was administered either by i.v. (10, 100, or 1000 IU/kg) or s.c. (400 IU/kg) injection. The first s.c. group served as a control for the calculation of absolute bioavailability. In the second group, the efferent popliteal lymphatic duct was cannulated and peripheral lymph draining the injection site was continuously collected. In the third group, the thoracic duct was cannulated to allow collection of central lymph just prior to entry into the systemic circulation. Blood was periodically sampled from all animals, and concentrations in serum and lymph were determined by enzyme-linked immunosorbent assay. The cumulative amount of rHuEPO recovered in peripheral and central lymph was 83.9 +/- 6.6% and 75.3 +/- 3.9% of the administered dose, respectively, indicating almost complete absorption from the s.c. injection site and minimal clearance during transit through the lymphatic system. After i.v. administration, the systemic clearance of rHuEPO decreased with increasing dose, reflecting capacity-limited elimination kinetics. A pharmacokinetic model was developed to simultaneously fit experimental data for all treatment groups and estimate bioavailability. The direct measurement of >75% of the dose in peripheral and central lymph independently verifies the calculated bioavailability of 87% and demonstrates the major role of the lymphatic route in the overall s.c. bioavailability of rHuEPO after s.c. administration with this animal model.


Subject(s)
Erythropoietin/pharmacokinetics , Lymphatic System/metabolism , Absorption , Algorithms , Animals , Area Under Curve , Biological Availability , Epoetin Alfa , Erythropoietin/administration & dosage , Half-Life , Injections, Intravenous , Injections, Subcutaneous , Male , Recombinant Proteins , Sheep
14.
Br J Haematol ; 122(4): 623-36, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12899718

ABSTRACT

Despite its frequency and impact on clinical outcomes, anaemia in cancer patients remains poorly understood and suboptimally treated. The definition of optimum treatment schedules with erythropoietic agents requires a suitable model of chemotherapy-induced progressive anaemia. This study investigated novel strategies such as once-per-chemotherapy-cycle dosing, synchronization between erythroid supportive care and chemotherapy, and definition of the optimum timing of erythroid support. A murine model of carboplatin chemotherapy/radiotherapy (CRT)-induced anaemia was used, which caused progressive anaemia across multiple cycles. Weekly administration of recombinant human erythropoietin (rHuEPO) was effective, but the longer-acting darbepoetin alpha resulted in superior responses. In all animals, anaemia became progressive and more refractory across cycles because of accumulated bone marrow damage. Exploiting a specific enzyme-linked immunosorbent assay, which could distinguish between darbepoetin alpha and endogenous erythropoietin, the effect of CRT upon the pharmacokinetics of darbepoetin alpha showed that clearance of darbepoetin alpha, and presumably erythropoietin, was at least partially dependent on a chemotherapy-sensitive pathway. Scheduling data suggested that administration of erythropoietic agents prior to chemotherapy was more effective than administration after chemotherapy. There was no evidence that erythropoietic agents exacerbated anaemia, even when administered immediately prior to CRT in an attempt to "prime" erythroid cells for the effects of CRT.


Subject(s)
Anemia/prevention & control , Antineoplastic Agents/adverse effects , Erythropoietin/analogs & derivatives , Erythropoietin/therapeutic use , Hematopoiesis/drug effects , Radiation Injuries/prevention & control , Anemia/etiology , Anemia/physiopathology , Animals , Carboplatin/adverse effects , Combined Modality Therapy , Darbepoetin alfa , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Erythropoietin/blood , Female , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/radiation effects , Hemoglobins/metabolism , Mice , Mice, Inbred Strains , Radiation Injuries/etiology , Radiation Injuries/physiopathology , Radiotherapy/adverse effects , Recombinant Proteins
SELECTION OF CITATIONS
SEARCH DETAIL