Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Clin Pharmacol Ther ; 102(2): 219-227, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28474798

ABSTRACT

Severely-debilitating or life-threatening (SDLT) diseases include conditions in which life expectancy is short or quality of life is greatly diminished despite available therapies. As such, the medical context for SDLT diseases is comparable to advanced cancer and the benefit vs. risk assessment and development of SDLT disease therapeutics should be similar to that of advanced cancer therapeutics. A streamlined development approach would allow patients with SDLT conditions earlier access to therapeutics and increase the speed of progression through development. In addition, this will likely increase the SDLT disease therapeutic pipeline, directly benefiting patients and reducing the economic and societal burden of SDLT conditions. Using advanced-stage heart failure (HF) as an example that illustrates the concepts applicable to other SDLT indications, this article proposes a streamlined development paradigm for SDLT disease therapeutics and recommends development of aligned global regulatory guidance.


Subject(s)
Disease Progression , Drug Discovery/methods , Heart Failure/diagnosis , Heart Failure/drug therapy , Severity of Illness Index , Animals , Communicable Diseases/diagnosis , Communicable Diseases/drug therapy , Communicable Diseases/epidemiology , Drug Discovery/trends , Drug Evaluation/methods , Drug Evaluation/trends , Heart Failure/epidemiology , Humans , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/epidemiology
3.
Vet Pathol ; 47(4): 614-29, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20516294

ABSTRACT

Chronic 6- and 12-month rat toxicology studies were evaluated for their ability to predict tumor outcome in 2-year rat carcinogenicity studies for 80 pharmaceuticals from commercial and Merck databases. The data consisted of 62 (6-month) and 54 (12-month) studies, which included 30 rat carcinogens and 50 noncarcinogens in 2-year studies. The histopathology findings considered as evidence of potential preneoplasia in the chronic studies were hyperplasia, cellular hypertrophy, and atypical cellular foci. The authors hypothesized that a whole animal-based approach should be taken, wherein (1) evidence of potential preneoplasia in any tissue may serve as a sensitive predictor of tumor outcome in any tissue in the whole animal and not necessarily the same tissue and (2) the absence of evidence for potential preneoplasia in all tissues may serve as a strong negative predictor of tumor outcome in any tissue. Based on this whole animal approach, 25 of 30 rat carcinogens showed histopathologic signals in chronic toxicology studies, yielding a test sensitivity of 83%. The negative predictivity of the absence of histopathology findings in chronic toxicology studies of 50 nontumorigenic compounds was 88%. The value of the extra 6-month treatment was not apparent. The 5 false negatives (negative chronic studies but positive 2-year studies) are for marketed drugs approved for non-life-threatening conditions and associated with rat-specific mechanisms. The absence of preneoplasia in the whole animal is a reliable predictor of negative tumor outcome in 2-year rat studies, and approximately 50% rat carcinogenicity studies could be eliminated for the 80 pharmaceuticals examined, with no risk to humans and with a substantial reduction in animal usage and drug development time.


Subject(s)
Carcinogenicity Tests/methods , Neoplasms/chemically induced , Pharmaceutical Preparations/metabolism , Animals , Carcinogenicity Tests/standards , Humans , Pharmaceutical Preparations/administration & dosage , Predictive Value of Tests , Rats , Retrospective Studies
4.
Clin Pharmacol Ther ; 82(2): 210-4, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17507920

ABSTRACT

Toxicology studies in animals are required by regulatory authorities worldwide to provide assurances that clinical testing of pharmaceutical candidates can be conducted safely. Safety concerns from animal studies account for over 20% of attritions from drug development. As discordance between humans and animals is expected, two goals of safe and efficient drug development must be (1) to improve the human relevance of animal testing with new models and technologies, and (2) to advance quickly to clinical testing armed with improved safety biomarker tools.


Subject(s)
Clinical Trials as Topic/methods , Drug Evaluation, Preclinical/methods , Drugs, Investigational/pharmacology , Animals , Disease Models, Animal , Drug Approval/legislation & jurisprudence , Drug Approval/methods , Drug Evaluation, Preclinical/standards , Drug-Related Side Effects and Adverse Reactions/chemically induced , Drug-Related Side Effects and Adverse Reactions/prevention & control , Drugs, Investigational/adverse effects , Drugs, Investigational/therapeutic use , Humans
5.
Toxicol Sci ; 49(2): 143-55, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10416259

ABSTRACT

At the present time, there are no uniform standards for the duration of non-rodent chronic toxicity studies. The European Union (EU) requires a 6-month non-rodent study. In Japan, a 6-month study is sufficient for most, but not all, compounds. The U.S. Food and Drug Administration (FDA) maintains its standard duration of 12 months for non-rodents, with 6-month studies accepted for some clinical indications on a case-by-case basis. To achieve harmonization on the duration of non-rodent toxicity studies, each member regulatory region (EU, U.S., and Japan) of the International Conference on Harmonization (ICH) collected non-rodent studies with significant new toxicological findings that had occurred after 6 months. An ICH expert working group was organized that included representatives from the regulatory authorities of each ICH region, to jointly review all available case studies for the purpose of arriving at a consensus on the best duration time for non-rodent toxicity studies. Eighteen case studies were identified and evaluated (16 original cases plus 2 additional FDA cases); most of the toxicities identified fell into the following categories: (1) toxicities identified at 6 months; (2) toxicities observed at 12 months, which were absent or considered isolated and not noteworthy findings at 6 months; (3) drug-related deaths or morbidity that occurred between 6 and 12 months, with a pattern of toxicity that permitted the interpolation of findings to an intermediate interval between 6 and 12 months; and (4) a shift in the dose response for toxicity with increasing duration of drug exposure. Of the 18 cases evaluated, 11 supported a study-duration of 9-12 months, 4 supported a duration of 12 months, and the 3 remaining cases indicated that a 6-month study would be adequate. The working group concluded that there was sufficient evidence to support a harmonized 9-month duration for non-rodent toxicity studies, which would be applicable for most categories of pharmaceuticals.


Subject(s)
International Agencies , Toxicity Tests , Animals , Drug Evaluation , Drug-Related Side Effects and Adverse Reactions , Europe , International Cooperation , Japan , Time Factors , United States
6.
Clin Cancer Res ; 5(5): 1161-7, 1999 May.
Article in English | MEDLINE | ID: mdl-10353752

ABSTRACT

Rodent and nonrodent toxicology studies are currently expected to support Phase I trials of antineoplastic drugs in the United States. To determine the predictive value of these studies, we initiated a project to compare preclinical and clinical toxicity data within various drug classes. The first class analyzed was the platinum anticancer drugs. Twelve platinum analogues that had both preclinical (mice, rats and/or dogs) and clinical data from matching drug administration schedules were identified. The rodent LD10 (the dose that causes lethality in 10% of treated animals) or dog toxic dose high (a dose that when doubled causes lethality in dogs) correlated well with the human maximally tolerated dose on a mg/m2 basis. For every platinum analogue investigated, one-third the rodent LD10 or one-third the dog toxic dose high in mg/m2 gave a starting dose and a first escalation dose that did not exceed the clinical maximally tolerated dose. The dose-limiting toxicities in patients were previously observed in 7 of 7, 7 of 8, and 9 of 11 mouse, rat, and dog studies, respectively. Our data indicate that mice, rats, and dogs all had value in predicting a safe starting dose and the qualitative toxicities in humans for platinum anticancer compounds. The efficiency of Phase 1 trials could have been improved without sacrificing patient safety by allowing higher starting doses for this drug class than conventionally permitted.


Subject(s)
Antineoplastic Agents/toxicity , Organoplatinum Compounds/toxicity , Toxicity Tests , Animals , Antineoplastic Agents/administration & dosage , Clinical Trials, Phase I as Topic , Dogs , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Evaluation Studies as Topic , Humans , Mice , Organoplatinum Compounds/administration & dosage , Rats , Single-Blind Method , Species Specificity , Toxicity Tests/standards
7.
Environ Health Perspect ; 106 Suppl 1: 71-80, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9539006

ABSTRACT

There is general agreement in the scientific community on the need to improve carcinogenicity testing and the assessment of human carcinogenic risk and to incorporate more information on mechanisms and modes of action into the risk assessment process. Advances in molecular biology have identified a growing number of genes such as protooncogenes and tumor-suppressor genes that are highly conserved across species and are associated with a wide variety of human and animal cancers. In vivo transgenic rodent models incorporating such mechanisms are used to identify mechanisms involved in tumor formation and as selective tests for carcinogens. Transgenic methods can be considered an extension of genetic manipulation by selective breeding, which long has been employed in science and agriculture. The use of two rodent species in carcinogenicity testing is especially important for identifying transspecies carcinogens. The capacity of a substance to induce neoplasia across species suggests that the mechanism(s) involved in the induction of the neoplasia are conserved and therefore may have significance for humans. Based on available information there is sufficient experience with some in vivo transgenic rodent carcinogenicity models to support their application as complementary second species studies in conjunction with a single 2-year rodent carcinogenicity study. The optional substitution of a second 2-year rodent carcinogenicity study with an alternative study such as an in vivo transgenic carcinogenicity study is part of the International Conference on Harmonization guidance S1B: Testing for Carcinogenicity of Pharmaceuticals. This guidance is intended to be flexible enough to accommodate a wide range of possible carcinogenicity assessment models currently under consideration or models that may be developed in the future. The use of an in vivo transgenic mouse model in place of a second 2-year mouse study will improve the assessment of carcinogenic risk by contributing insights into the mechanisms of tumorigenesis and potential human relevance not available from a standard 2-year bioassay. It is envisioned that this will stimulate the further development of more efficient and relevant methods for identifying and assessing potential human carcinogenic risk, which will benefit public health.


Subject(s)
Carcinogenicity Tests , Mice, Transgenic , Animals , Biological Assay , Genes, p53/physiology , Humans , Mice
8.
Cancer Chemother Pharmacol ; 41(3): 173-85, 1998.
Article in English | MEDLINE | ID: mdl-9443633

ABSTRACT

The entry of new anticancer treatments into phase I clinical trials is ordinarily based on relatively modest preclinical data. This report defines the battery of preclinical tests important for assessing safety under an Investigational New Drug application (IND) and outlines a basis for extrapolating starting doses of investigational anticancer drugs in phase I clinical trials from animal toxicity studies. Types of preclinical studies for the support of marketing of a new anticancer drug are also discussed. This report addresses differences and similarities in the preclinical development of cytotoxic drugs (including photosensitizers and targeted delivery products), drugs used chronically (chemopreventive drugs, hormonal drugs, immunomodulators), and drugs intended to enhance the efficacy (MDR-reversing agents and radiation/chemotherapy sensitizers) or diminish the toxicity of currently used anticancer therapies. Factors to consider in the design of preclinical studies of combination therapies, alternative therapies, and adjuvant therapies in the treatment of cancer, and to support changes in clinical formulations or route of administration, are also discussed.


Subject(s)
Antineoplastic Agents , Drug Evaluation, Preclinical/methods , Drugs, Investigational , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Clinical Trials, Phase I as Topic , Drug Approval , Drug Evaluation, Preclinical/standards , Drugs, Investigational/pharmacokinetics , Drugs, Investigational/therapeutic use , Drugs, Investigational/toxicity , Humans , United States , United States Food and Drug Administration
9.
Regul Toxicol Pharmacol ; 25(2): 130-45, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9185889

ABSTRACT

The results of rat and mouse carcinogenicity studies for 282 human pharmaceuticals in the FDA database were analyzed and compared as part of an International Conference on Harmonization (ICH) evaluation of rodent carcinogenicity studies and their utility for carcinogenicity testing. A majority of the carcinogenicity studies in the FDA database were carried out in Sprague-Dawley-derived rats and Swiss-Webster-derived CD-1 mice in contrast to Fisher 344 rats and B6C3F1 mice employed in National Toxicology Program (NTP) studies. Despite the differences in rodent strains, the relative proportion of compounds with positive findings (44.3%) and the degree of overall concordance between rats and mice (74.1%) in the FDA database were similar to the NTP rodent carcinogenicity database. Carcinogenicity studies in two rodent species are necessary primarily to identify trans-species tumorigens, which are considered to pose a relatively greater potential risk to humans than single species positive compounds. Two-year carcinogenicity studies in both rats and mice may not be the only means of identifying trans-species tumorigens. Sufficient experience is now available for some alternative in vivo carcinogenicity models to support their application as complementary studies in combination with a single 2-year carcinogenicity study to identify trans-species tumorigens. Our analysis of the rodent carcinogenicity studies supports such an approach for assessing carcinogenic potential without compromising the public health.


Subject(s)
Carcinogens/chemistry , Pharmaceutical Services , Animals , Humans , Information Systems , Mice , Rats , Social Control, Formal , United States , United States Food and Drug Administration
10.
Regul Toxicol Pharmacol ; 25(2): 189-93, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9185894

ABSTRACT

The standard approaches for the preclinical development of chronically administered drugs also apply to most respiratory drugs. Modifications from the standard preclinical development plan, however, may be necessary if the drug is administered intranasally or by inhalation. Administration by these routes may result in airway toxicity and the intended patient population is often particularly susceptible. Current and former representatives of the Division of Pulmonary Drug Products (CDER, U.S. FDA) present this article to describe general principles of preclinical development for respiratory drug indications. The article addresses drugs intended for administration by the intranasal or inhalation routes. The article describes the types of studies recommended, considers the initial human dose, and discusses dose-escalation strategies in clinical trials. Other areas of special concern with intranasal or inhalation administration include immunotoxicity, reproductive toxicity, types of dosing apparatus, excipients and extractables, and formulation changes. The approaches described in this article are intended as general information and should be adapted to the scientific considerations and circumstances of a particular drug under development.


Subject(s)
Research , Respiratory System Agents/toxicity , Humans , Research Design
12.
Toxicol Pathol ; 23(2): 220-5, 1995.
Article in English | MEDLINE | ID: mdl-7569678

ABSTRACT

The importance of drug kinetics for interpretation of toxicity findings and for cross-species toxicity assessment has been long recognized. Recently, an international effort was initiated to standardize guidance on the kinetic data to be collected in conjunction with toxicity studies. The guidance addresses the kinetic data to be included in studies on carcinogenicity, reproduction toxicity, genotoxicity, and single- and repeat-dose toxicity. In various stages of development or implementation, the guidance is intentionally nondetailed regarding the specific kinetic assessments to be performed. This is to allow flexibility in study design and ensures that scientific judgment is used to determine the appropriate kinetic endpoints to achieve study- and drug-specific goals. Some examples of how kinetics have been used at the Food and Drug Administration in review of toxicity studies submitted in drug applications are presented. The examples discussed demonstrate successful and unsuccessful integration of kinetics into study design and interpretation and highlight the impact on the drug development program from a regulatory perspective.


Subject(s)
Chemistry, Pharmaceutical/methods , Pharmacokinetics , Toxicology/legislation & jurisprudence , Toxicology/methods , Animals , Humans , United States , United States Food and Drug Administration
13.
Article in English | MEDLINE | ID: mdl-7894318

ABSTRACT

The broad concept of chemoprevention applies to the prevention of clinical cancer by the administration of chemical agents. Current approaches to the development and marketing approval of drugs to prevent cancer are described by a Working Group from the National Cancer Institute and the Food and Drug Administration. A strategy is presented that identifies candidate drugs, with examples that illustrate how drugs are characterized for efficacy through in vitro transformation modulation and mechanistic assays, and in vivo tumor modulation models of carcinogenesis. Requirements and recommendations for safety evaluation in toxicology testing are given, and the evaluation of pharmacokinetic and pharmacodynamic drug effects and potential surrogate end point biomarkers in Phase I trials are discussed. Appropriate subject populations are identified. Phase II trials should emphasize the evaluation of surrogate end point biomarkers that are highly correlated with cancer incidence and may serve as an estimate of cancer incidence reduction. In Phase III trials the interim analysis of a validated surrogate end point of cancer incidence may facilitate timely and cost-effective marketing of efficacious drugs.


Subject(s)
Antineoplastic Agents , Drug Approval , Neoplasms/prevention & control , Animals , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Drug Screening Assays, Antitumor , Humans , National Institutes of Health (U.S.) , United States , United States Food and Drug Administration
14.
Clin Exp Metastasis ; 12(3): 213-25, 1994 May.
Article in English | MEDLINE | ID: mdl-8194196

ABSTRACT

This study investigates the incorporation of three intravenously administered radiolabeled fatty acids, [9,10-3H]palmitate (3H-PAM), [1-14C]arachidonate (14C-ACH) and [1-14C]docosahexaenoate (14C-DHA), into lipids of intracerebrally implanted tumor and contralateral brain cortex in awake rats. A suspension of Walker 256 carcinosarcoma cells (1 x 10(6) cells) was implanted into the right cerebral hemisphere of an 8- to 9-week-old Fischer-344 rat. Seven days later, the awake rat was infused intravenously for 5 min with 3H-PAM (6.4 mCi/kg), 14C-ACH (170 microCi/kg) or 14C-DHA (100 microCi/kg). Twenty min after the start of infusion, the rat was killed and intracranial tumor mass and brain cortex were removed for lipids analysis. Each radiolabel was incorporated more into tumor than into brain cortex. Ratios of net incorporation rate coefficients (k*) into tumor as compared with brain were 4.5, 3.4 and 1.7 for 3H-PAM, 14C-ACH and 14C-DHA, respectively. Lipid radioactivity comprised more than 80% of total tumor or brain radioactivity for each probe. Phospholipids contained 58%, 89% and 68% of tumor lipid radioactivity, and 58%, 82% and 74% of brain lipid radioactivity, for 3H-PAM, 14C-ACH and 14C-DHA, respectively. Incorporation coefficients (k*i) for a phospholipid class (i)--choline phosphoglycerides (PC), inositol monophosphoglycerides (PI), ethanolamine phosphoglycerides (PE), serine phosphoglycerides (PS), and sphingomyelin (SM)--were greater in tumor than in brain for each fatty acid probe, except that values for k*PE and k*PS using 14C-DHA were equivalent. Differences in k*i between tumor and brain were largest for SM and PC and the change in k*PC accounted for 65-90% of the increase in the net phospholipid incorporation rate for each probe. Differences in k*PI, k*PE and k*PS were smaller than those in were smaller than those in k*PC and k*SM, and varied with the probe. Differences in k*i were related to differences in tumor and brain phospholipid composition and metabolism. The results indicate that suitably radiolabeled fatty acids may be used to image and characterize metabolism of lipid compartments of a brain tumor in vivo using positron emission tomography.


Subject(s)
Brain Neoplasms/metabolism , Brain/metabolism , Fatty Acids/metabolism , Phospholipids/metabolism , Animals , Carcinoma 256, Walker/metabolism , Male , Rats , Rats, Inbred F344 , Wakefulness
16.
Antisense Res Dev ; 4(4): 299-301, 1994.
Article in English | MEDLINE | ID: mdl-7734946

ABSTRACT

This article describes pharmacology and toxicity studies for oligonucleotide drugs that are recommended for inclusion in the initial Investigational New Drug Application (IND), a first request to use an investigational drug in clinical trials. Recent observations of non-sequence-dependent cardiovascular toxicity and deaths in monkeys following intravenous infusions of phosphorothioates have raised a potential safety concern for oligonucleotide drugs. This concern should be considered by drug sponsors in designing pre-IND nonclinical development programs and Phase I clinical protocols. Pre-IND conduct of pharmacodynamic cardiovascular screening is highly recommended for defining safe clinical dosing regimens for phosphorothioate (and, possibly, other charged-backbone) oligomers. Additionally, drug sponsors are encouraged to (1) conduct research into-the mechanisms responsible for this dose-limiting toxicity, (2) institute liberal publication policies for research conducted under industrial sponsorship, and (3) communicate with reviewing divisions at FDA for updated guidance in this field when planning pre-IND safety studies. Recommendations for nonclinical studies during development of oligonucleotides will be modified as new information regarding the biological properties of oligonucleotides becomes available.


Subject(s)
Drugs, Investigational/pharmacology , Oligonucleotides/pharmacology , Animals , Drugs, Investigational/adverse effects , Drugs, Investigational/metabolism , Humans , Injections, Intravenous , Investigational New Drug Application , Oligonucleotides/metabolism , Protein Binding
17.
Anesthesiology ; 80(1): 151-8, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8291704

ABSTRACT

BACKGROUND: Barbiturates are reported to reduce brain oxidative metabolism and brain free fatty acid release during ischemia by mechanisms that are as yet unclear. To elucidate their action on brain lipid metabolism, an in vivo method was used to quantify the effect of pentobarbital on the incorporation of radiolabeled palmitic acid from blood into lipids of the rat brain. METHODS: [9,10(-3)H]-Palmitate was infused intravenously in an awake rat or in a rat lightly anesthetized or made comatose by pentobarbital. Twenty minutes after infusion was begun, the rat was killed and non-3H2O radioactivity in individual brain lipid compartments was determined. Incorporation coefficients (k*) were calculated by dividing the lipid compartment radioactivities by the integrated plasma radioactivity to 20 min. RESULTS: Net brain k* for [9,10(-3)H]-palmitate was reduced by 36-40% in pentobarbital-anesthetized rats. This reduction was unrelated to depth of anesthesia or to the presence of hypercapnia and acidosis, because breathing 7.5% CO2 had no effect on k* in awake rats. Anesthesia reduced radiolabel incorporation into phospholipids by 46-53% and into neutral lipids by 20-26% but did not change the distribution of radiolabel among phospholipid or neutral lipid classes. CONCLUSIONS: Pentobarbital has a profound effect on brain lipid metabolism. It reduces incorporation of plasma palmitate into brain, more so into phospholipids than into neutral lipids, independently of changes in cerebral blood flow. Reduced incorporation likely reflects reduced turnover of palmitate within brain lipids (mainly phosphatidylcholine), consistent with evidence that barbiturates also reduce release of free fatty acids during brain ischemia.


Subject(s)
Brain/metabolism , Palmitates/pharmacokinetics , Pentobarbital/pharmacology , Animals , Brain/drug effects , Male , Palmitates/blood , Rats , Rats, Inbred F344
18.
Clin Exp Metastasis ; 11(2): 141-9, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8444007

ABSTRACT

This paper investigates the incorporation of intravenously (i.v.) administered radiolabelled fatty acids--[9,10(3)-H]palmitate (3H-PA), [1-14C]arachidonate (14C-AA) and [1-14C]docosahexaenoate (14C-DHA)--into intracerebrally implanted tumours in awake Fischer-344 rats. A suspension of Walker 256 carcinosarcoma tumour cells (1 x 10(6) cells) was implanted into the right cerebral hemisphere of 8- to 9-week-old rats. Seven days after implantation, the awake rat was infused i.v. for 5 min with 3H-PA (6.4 mCi/kg), 14C-AA (170 microCi/kg) or 14C-DHA (100 microCi/kg). Twenty minutes after the start of infusion, the rat was killed and coronal brain sections were obtained for quantitative autoradiography and histology. Each fatty acid showed well-demarcated incorporation into tumour tissue. Areas of necrosis or haemorrhage showed no or small levels of incorporation. The ratios of incorporation into the tumour to incorporation into contralateral brain regions were 2.8-5.5 for 3H-PA, 2.1-3.3 for 14C-AA and 1.5-2.2 for 14C-DHA. The mean ratios differed significantly between the fatty acids (P < 0.01). 3H-PA was not incorporated into necrotic tumours despite the presence of an open blood-tumour barrier, indicated by extravasated horseradish peroxidase. The incorporation rate constant of 3H-PA was similar for small intracerebral and large extracerebral tumours. The results show that 3H-PA, 14C-AA and 14C-DHA are incorporated more readily into tumour tissue than into brain, and that the increase is primarily due to increased utilization of fatty acids by tumour cells and not due to a high blood-tumour permeability. The relative increases in rates of incorporation for the different fatty acids may be related to lipid composition of the tumour and to the requirement of and specific role of these fatty acids in tumour cell growth and division.


Subject(s)
Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Carbon Radioisotopes , Fatty Acids/pharmacokinetics , Phospholipids/metabolism , Tritium , Animals , Arachidonic Acid/pharmacokinetics , Autoradiography , Blood-Brain Barrier/physiology , Docosahexaenoic Acids/pharmacokinetics , Fatty Acids/blood , Injections, Intravenous , Male , Palmitic Acid , Palmitic Acids/pharmacokinetics , Radionuclide Imaging , Rats , Rats, Inbred F344
19.
Antisense Res Dev ; 3(4): 399-404, 1993.
Article in English | MEDLINE | ID: mdl-8155981

ABSTRACT

This article focuses on pharmacology and toxicology data that should be included in an Investigational New Drug Application (IND), a request to use an investigational drug in clinical trials. In general, pharmacology and toxicology testing programs for antisense compounds are held to the same regulatory standards applied to other new therapeutic classes. Biological properties of oligonucleotide therapeutics are mentioned where they may pertain to clinical safety issues. Nonclinical data submitted to the IND should characterize the pharmacology, disposition, and toxicology of a new drug; these data form the basis for clinical risk assessment. Concomitant evaluation of pharmacokinetics allows for better interpretation of in vivo studies and increased accuracy of dose extrapolation to humans. Recommendations for nonclinical drug development will be modified as new information regarding the biological properties of oligonucleotides becomes available.


Subject(s)
Drugs, Investigational/pharmacology , Investigational New Drug Application , Oligonucleotides, Antisense/pharmacology , Animals , Bone Marrow Purging , Clinical Trials as Topic , Drugs, Investigational/adverse effects , Drugs, Investigational/pharmacokinetics , Humans , Oligonucleotides, Antisense/adverse effects , Oligonucleotides, Antisense/pharmacokinetics , United States , United States Food and Drug Administration
20.
Brain Res Brain Res Rev ; 17(3): 187-214, 1992.
Article in English | MEDLINE | ID: mdl-1467810

ABSTRACT

An experimental method and its associated mathematical model are described to quantitate in vivo incorporation rates into and turnovers of fatty acids (FAs) within stable brain metabolic compartments, particularly phospholipids. A radiolabeled FA is injected i.v. in a rat, and arterial plasma unacylated FA radioactivities and unlabeled concentrations are sampled until the animal is killed after 15 min, when the brain is analyzed biochemically or with quantitative autoradiography. Unbound unacylated label in blood easily crosses the blood-brain barrier; rapidly equilibrates in the unacylated FA, acyl-CoA and phosphatidate-diacylglycerol brain pools; then is incorporated into phospholipids and other stable metabolic compartments. Uptake and incorporation of labeled FAs are independent of cerebral blood flow at constant brain blood volume. Different labeled FAs enter specific sn positions of different brain phospholipids, suggesting that a combination of probes can be used to investigate metabolism of these phospholipids. Thus, [9,10-3-H]palmitate preferentially labels the sn1 position of phosphatidylcholine; [1-14C]arachidonate the sn2 positions of phosphatidylinositol and phosphatidylcholine; and [1-14C]docosahexaenoate the sn2 positions of phosphatidylethanolamine and phosphatidylcholine. The FA model provides an operational equation for rates of incorporation of FAs into brain phospholipids, taking into account intracerebral recycling and de novo synthesis of the FA, as well as entry into brain of FA from acylated blood sources. The equation is essentially independent of specific details of the proposed model, and can be used to calculate turnovers and half-lives of FAs within different phospholipid classes. For the model to be most applicable, experiments should satisfy conditions for pulse-labeling of the phospholipids, with brain sampling times short enough to minimize exchange of label between stable metabolic compartments. A 15-20 min sampling time satisfies these criteria. The FA method has been used to elucidate the dynamics of brain phospholipids metabolism in relation to brain development, brain tumor, chronically reduced auditory input, transient ischemic insult, axotomy with and without nerve regeneration, and cholinergic stimulation in animals with or without a chronic unilateral lesion of the nucleus basalis magnocellularis.


Subject(s)
Brain/metabolism , Cerebrovascular Circulation/physiology , Fatty Acids/metabolism , Phospholipids/metabolism , Animals , Autoradiography , Fatty Acids/blood , Half-Life , Humans , Models, Neurological
SELECTION OF CITATIONS
SEARCH DETAIL
...