Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Q J Nucl Med Mol Imaging ; 58(3): 290-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25265250

ABSTRACT

Nuclear medicine contributes important tools to support anti-inflammatory drug discovery and development. The support provided is manifold: new molecular entities (NME, either small molecules or biologics) labeled with radioisotopes can be applied in animal models and humans to measure biodistribution, target engagement, and pharmacokinetics. In addition, nuclear imaging techniques can be used to select or enrich the patient populations in clinical trials, to assess disease activity, target status and distribution and to quantify response to therapeutic interventions. In the first part of this review we will outline how nuclear imaging techniques can be applied to support informed decision making in drug development. In the second part, we will briefly high-light the use of nuclear imaging of inflammation in drug development in selected diseases, specifically rheumatoid arthritis (RA), inflammatory bowel diseases (IBD), atherosclerosis and - as an emerging topic - cancer.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Drug Design , Inflammation/diagnostic imaging , Inflammation/drug therapy , Outcome Assessment, Health Care/methods , Radiopharmaceuticals , Tomography, Emission-Computed/methods , Animals , Humans , Isotope Labeling/methods , Treatment Outcome
2.
Article in English | MEDLINE | ID: mdl-24829127

ABSTRACT

Nuclear medicine contributes important tools to support antiinflammatory drug discovery and development in many ways. The support provided is manifold: new molecular entities (NME, either small molecules or biologics) labeled with radioisotopes can be applied in animal models and humans to measure biodistribution, target engagement, and pharmacokinetics. In addition, nuclear imaging techniques can be used to select or enrich the patient populations in clinical trials, to assess disease activity, target status and distribution and to quantify response to therapeutic interventions. In the first part of this review we will outline how nuclear imaging techniques can be applied to support informed decision making in drug development. In the second part, we will briefly highlight the use of nuclear imaging of inflammation in drug development in selected diseases, specifically rheumatoid arthritis (RA), inflammatory bowel diseases (IBD), atherosclerosis (ATS) and as an emerging topic cancer.

3.
Curr Med Chem ; 19(28): 4786-93, 2012.
Article in English | MEDLINE | ID: mdl-22873664

ABSTRACT

Noninvasive fluorescence imaging (NFI) is a powerful tool to study physiology and pathophysiology in animal disease models. NFI has been successfully applied in a number of animal disease models including cancer, arthritis, and stroke. Furthermore, several applications in humans have been described. NFI is widely available in research laboratories because it has a number of advantages: It uses non-ionizing radiation and requires comparably simple, inexpensive instrumentation, and easy to handle. Fluorochromes can be detected with high sensitivity, and image acquisition time is relatively short. Furthermore, a plethora of fluorescent imaging agents is available including unspecific, target-specific, and activatable imaging probes. With these probes, biological processes such as inflammation, cell death or enzyme activity, and many others can be visualized in living animals. This review offers an overview of current approaches in NFI of stroke pathophysiology in animal models of cerebral ischemia. First, the instrumentation and the different types of imaging agents for NFI are described. Second, a short introduction to animal models of stroke is provided. Third, examples for NFI in animal models of stroke are given. Finally, the use of NFI in human stroke is critically discussed.


Subject(s)
Stroke/diagnosis , Animals , Blood-Brain Barrier/metabolism , Disease Models, Animal , Fluorescent Dyes/chemistry , Humans , Luminescent Measurements , Matrix Metalloproteinases/metabolism , Quantum Dots , Tomography, Optical
4.
Neuroscience ; 158(3): 1161-73, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-18983900

ABSTRACT

Inflammation is crucially involved in many diseases of the CNS. Immune cells may attack the CNS, as in multiple sclerosis, and therefore be responsible for primary damage. Immune cells may also be activated by injury to the CNS, as for example in stroke or brain trauma, secondarily enhancing lesion growth. In general, CNS inflammation involves a complex interplay of pro- and anti-inflammatory cells and molecules. The blood-brain barrier loses its integrity, plasma proteins leak into the CNS parenchyma, followed by invasion of blood-borne immune cells, and activation of resident microglial cells and astrocytes. However, inflammation not only exacerbates CNS disease, it is also indispensable in containment and resolution of tissue damage, as well as repair and regeneration. The time course and the contribution of inflammatory processes to the pathophysiology of the disease depend on several factors including the type of injury and the time point after injury, and can exhibit a high individual variability. Imaging technologies that enable specific visualization of these inflammatory processes non-invasively are therefore highly desirable. They provide powerful tools to further evaluate the contribution of specific processes to the pathophysiology of CNS disease. Moreover, these technologies may be valuable in detecting and assessing disease progression, in stratifying patients for therapy, and in monitoring therapy. Among the existing non-invasive imaging methods to visualize neuroinflammation in the CNS, we here review the current status of nuclear and optical imaging techniques, with particular emphasis on the sensitivity, specificity, as well as the limitations of these approaches.


Subject(s)
Diagnostic Imaging/methods , Encephalitis/diagnostic imaging , Optics and Photonics/methods , Positron-Emission Tomography/methods , Tomography, Emission-Computed, Single-Photon/methods , Animals , Brain Edema/diagnostic imaging , Brain Edema/physiopathology , Chemotaxis, Leukocyte/physiology , Diagnostic Imaging/trends , Encephalitis/pathology , Encephalitis/physiopathology , Fluorescent Dyes , Glucose/metabolism , Humans , Luminescent Proteins , Optics and Photonics/trends , Positron-Emission Tomography/trends , Predictive Value of Tests , Tomography, Emission-Computed, Single-Photon/trends
5.
Neurodegener Dis ; 5(5): 296-303, 2008.
Article in English | MEDLINE | ID: mdl-18520164

ABSTRACT

Fluorescence molecular brain imaging is a new modality allowing the detection of specific contrast agents down to very low concentration ranges (picomolar) in disease models. Here we demonstrate a first noninvasive application of fluorescence imaging in the human brain, where concentrations down to about 100 nM of a nonspecific dye were detected. We argue that due to its high sensitivity, optical molecular imaging of the brain is feasible, which - together with its bedside applicability - makes it a promising technique for use in patients.


Subject(s)
Brain/pathology , Fluorescence , Spectroscopy, Near-Infrared/methods , Animals , Brain/physiopathology , Humans , Mice , Molecular Biology/methods , Sensitivity and Specificity
6.
Rheumatology (Oxford) ; 44(11): 1341-9, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15972356

ABSTRACT

Molecular imaging is a rapidly emerging field in biomedical research, aiming at the visualization, characterization and quantification of molecular and cellular processes non-invasively within intact living organisms. To sense biological processes such as gene expression, angiogenesis, apoptosis or cell trafficking in vivo, imaging reporter agents that interact specifically with molecular targets and appropriate imaging systems are currently under development. In rheumatoid arthritis, these novel tools will be used to evaluate physiological and pathophysiological processes, to facilitate diagnosis and monitor therapeutic regimens, to enable reliable prognosis and to support the development of new therapies. In this review, we summarize the basic principles of molecular imaging, such as the development of molecular imaging agents, the actual capabilities of different imaging modalities and the most recent advances in molecular imaging, demonstrating the potential of this technology. With regard to their applicability in rheumatic diseases, we discuss potential molecular targets, current experimental approaches and the future prospects for molecular imaging in rheumatoid arthritis.


Subject(s)
Arthritis, Rheumatoid/diagnosis , Animals , Biomarkers/analysis , Contrast Media , Humans , Magnetic Resonance Imaging/methods , Mice , Molecular Probes , Positron-Emission Tomography/methods
7.
Rofo ; 177(3): 326-37, 2005 Mar.
Article in German | MEDLINE | ID: mdl-15719293

ABSTRACT

The recent years have seen significant advances in both molecular biology, allowing the identification of genes and pathways related to disease, and imaging technologies that allow for improved spatial and temporal resolution, enhanced sensitivity, better depth penetration, improved image processing, and beneficial combinations of different imaging modalities. These advances have led to a paradigm shift in the scope of diagnostic imaging. The traditional role of radiological diagnostic imaging is to define gross anatomy and structure in order to detect pathological abnormalities. Available contrast agents are mostly non-specific and can be used to image physiological processes such as changes in blood volume, flow, and perfusion but not to demonstrate pathological alterations at molecular levels. However, alterations at the anatomical-morphological level are relatively late manifestations of underlying molecular changes. Using molecular probes or markers that bind specifically to molecular targets allows for the non-invasive visualization and quantitation of biological processes such as gene expression, apoptosis, or angiogenesis at the molecular level within intact living organisms. This rapidly evolving, multidisciplinary approach, referred to as molecular imaging, promises to enable early diagnosis, can provide improved classification of stage and severity of disease, an objective assessment of treatment efficacy, and a reliable prognosis. Furthermore, molecular imaging is an important tool for the evaluation of physiological and pathophysiological processes, and for the development of new therapies. This article comprises a review of current technologies of molecular imaging, describes the development of contrast agents and various imaging modalities, new applications in specific disease models, and potential future developments.


Subject(s)
Diagnostic Imaging , Magnetic Resonance Imaging , Molecular Biology , Nuclear Medicine , Tomography, X-Ray Computed , Ultrasonography , Animals , Biomedical Research , Contrast Media , Forecasting , Humans , Image Processing, Computer-Assisted , Mice , Molecular Probes , Prognosis
8.
Rheumatology (Oxford) ; 43(9): 1097-105, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15199219

ABSTRACT

OBJECTIVE: To evaluate the anti-arthritic effects of the new inflammation-targeted drug MTX-HSA and to investigate whether peripheral blood mononuclear cells (PBMC) are potential target cells for albumin-mediated drug delivery. METHODS: The murine model of collagen-induced arthritis (CIA) was used to measure the anti-arthritic effect of MTX, MTX-HSA or a combination of both (n = 30 to 35 per group). In addition, the uptake of fluorescence-labelled albumin (AFLc-HSA) in PBMC of 14 patients with RA was measured by fluorescence-activated cell sorting (FACS). RESULTS: In equivalent doses of 7.5 mg/kg intravenously (IV) twice a week, MTX-HSA is significantly (P<0.02) superior to MTX in inhibiting the development of CIA and reducing the joint count as well as the number of affected paws. When given in lower doses as combination therapy, both drugs act synergistically (P<0.03). A mean of 96, 72 and 64% of the CD14-, CD16- and CD20-positive cells from peripheral blood of rheumatoid arthritis (RA) patients showed an uptake of albumin after incubation with AFLc-HSA in vitro. This finding was not significantly different in comparison to healthy controls. In contrast, the number of CD3-positive cells taking up albumin is increased significantly in RA patients in comparison to controls (26.3 +/- 12.9% s.d. vs 11.6 +/- 7.3% s.d.; P = 0.005). CONCLUSION: The data show that the effectiveness of MTX-HSA in CIA is superior to MTX and that both drugs act synergistically. In addition, albumin appears to be taken up by peripheral blood cells, suggesting that they might be one of the potential target cells of this novel anti-arthritic treatment approach.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Methotrexate/administration & dosage , Serum Albumin/administration & dosage , Animals , Antibodies/blood , Antigens, CD/analysis , Collagen , Collagen Type II/immunology , Disease Models, Animal , Drug Administration Schedule , Drug Carriers , Drug Combinations , Drug Synergism , Flow Cytometry/methods , Humans , Injections, Intravenous , Leukocytes, Mononuclear/immunology , Male , Mice , Mice, Inbred DBA , Serum Albumin/pharmacokinetics , Treatment Outcome
9.
Ann Rheum Dis ; 63(7): 884-6, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15194591

ABSTRACT

OBJECTIVE: To investigate the effect of methotrexate (MTX) and albumin coupled with methotrexate (MTX-HSA) on cartilage invasion and induction of perichondrocytic degradation by rheumatoid arthritis synovial fibroblasts (RA SF) in vivo. METHODS: Human cartilage and RA SF were co-transplanted in three groups of severe combined immunodeficient mice (SCID), which received 1 mg/kg free MTX (n = 9), 1 mg/kg MTX-HSA (n = 6), or 0.9% NaCl (n = 5), respectively, intraperitoneally twice a week. After 4 weeks' treatment, the mice were killed and the implants analysed histologically. RESULTS: The control group had a mean (SEM) score for cartilage invasion of RA SF of 2.0 (0.26) and for perichondrocytic cartilage degradation of 1.5 (0.34). In contrast, mice which received MTX showed a significantly reduced invasion (0.78 (0.14), p<0.01) and a reduction in perichondrocytic cartilage degradation scores (0.69 (0.2), p<0.05) in comparison with the control group. Mice treated with MTX-HSA also had significantly reduced scores for RA SF invasion into the cartilage (0.92 (0.41), p<0.05) and for cartilage degradation (0.83 (0.44), p<0.05) compared with controls. The effects of MTX and MTX-HSA were not significantly different between these two groups. CONCLUSION: Treatment with MTX or MTX-HSA significantly ameliorates cartilage destruction in the SCID mouse model for human RA.


Subject(s)
Albumins/therapeutic use , Arthritis, Rheumatoid/drug therapy , Cartilage/pathology , Immunosuppressive Agents/therapeutic use , Methotrexate/therapeutic use , Synovial Membrane/pathology , Animals , Arthritis, Rheumatoid/pathology , Cell Movement/drug effects , Drug Therapy, Combination , Female , Fibroblasts/pathology , Humans , Mice , Mice, SCID , Models, Animal , Serum Albumin/therapeutic use
10.
Z Rheumatol ; 62(Suppl 2): II33-6, 2003.
Article in German | MEDLINE | ID: mdl-14648088

ABSTRACT

Advances in molecular biology and technical developments in the field of imaging are increasingly allowing for non-invasive visualization and quantitation of biological processes at the molecular level. Such technologies, defined as molecular imaging, promise early diagnosis and improved classification of the stage and severity of disease, objective assessment of treatment efficacy, and reliable prognosis based on so-called molecular markers. Furthermore, molecular imaging is an important tool for the evaluation of physiological and pathophysiological processes and for drug development. Various different imaging modalities are available, such as conventional radiography (CR), computed tomography (CT), nuclear imaging, magnetic resonance imaging (MRI), ultrasound (US), as well as other methods including fluorescence-based optical imaging. These methods differ with respect to resolution and their potential to gather information at the anatomical, physiological, cellular and molecular level. Therefore, the choice of the imaging modality for molecular imaging depends on the questions that need to be answered. This review discusses the potential of imaging modalities for molecular imaging in rheumatoid arthritis (RA).


Subject(s)
Arthritis, Rheumatoid/diagnosis , Biomarkers/analysis , Diagnostic Imaging/methods , Image Enhancement/methods , Image Processing, Computer-Assisted/methods , Animals , Arthritis, Experimental/classification , Arthritis, Rheumatoid/classification , Humans , Mice , Sensitivity and Specificity
11.
Neurol Res ; 22(5): 481-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10935221

ABSTRACT

To delineate the tumor margins of malignant gliomas laser-induced fluorescence detection technique was applied using 5-aminofluorescein-albumin as the fluorescent dye. The 5-aminofluorescein was linked to serum albumin (= AFlc-SA) as a cumulative protein label using residualizing markers. In a C6-glioma model the biodistribution and pharmacokinetics of the injected dye were investigated by labeling the protein conjugate with 111In-DTPA. Twenty-four hours after intravenous injection of the dye, fluorescence was activated by an argon laser and inspected in the C6-gliomas. Histological examinations were performed to compare the microscopic margins of the fluorescence-stained tumors with hematoxylin/eosin. The tumor uptake 24 h after dye injection was 23-fold higher than in the surrounding brain. Fluorescence inspection under laser activation demonstrated clearly stained and sharply demarcated tumors. The microscopic borders of the tumors corresponded exactly with the fluorescence, also demonstrating intracellular tumor uptake of the dye. In a preliminary study, three patients with malignant gliomas were operated using laser-induced fluorescence detection technique after injection of AFlc-SA. In all patients, the borders of the malignant gliomas were clearly stained by AFlc-SA during surgery. Laser-induced fluorescence imaging using the albumin conjugate AFlc-SA may be a promising method for delineating tumor margins which are hard to detect under the operating microscope alone.


Subject(s)
Brain Neoplasms/diagnosis , Fluorescein , Glioma/diagnosis , Lasers , Serum Albumin , Animals , Brain Neoplasms/pathology , Contrast Media , Female , Fluorescence , Glioma/pathology , Humans , Indium Radioisotopes , Magnetic Resonance Imaging , Neoplasm Transplantation , Pentetic Acid , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured
14.
Anticancer Drugs ; 10(8): 785-90, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10573211

ABSTRACT

Albumin-based drug carrier systems have been developed in the field of chemotherapy to improve the passive tumor targeting properties of anti-cancer drugs. Usually, serum albumins of different species are used as carrier proteins, mostly of bovine (BSA), human (HSA) or rat (RSA) origin. The resulting albumin conjugates are often tested for anticancer activity in heterologous tumor models. No data is available whether the choice of the albumin species might influence the pharmacokinetics or the tumor uptake rates of the conjugates in vivo. Residualizingly ([111In]DTPA) radiolabeled RSA, BSA or HSA were administered to Walker-256 carcinoma-bearing rats. No significant difference was found in the absolute or the weight-adjusted tumor uptake rates of the three albumin tracers. The tumors were the major catabolic sites accumulating 14-18% of the injected dose (ID). Low hepatic uptake rates were determined for all albumins (below 100% ID). Minor differences were found for hepatic uptake in favor of the autologous RSA (5.8% ID) versus HSA (6.9%) and BSA (8.0%). These differences might have occurred during the commercial preparation or the radiolabeling of the different batches. In addition, there are structural differences between the three albumins, which might have contributed, despite high sequence homologies above 70% for RSA, HSA and BSA. These minor differences in the distribution patterns of RSA, HSA or BSA might not decisively influence the results of drug targeting experiments in rats. For further studies with albumin conjugates, HSA was chosen as drug carrier in rodent animal models when considering later human use. In rats or nude mice multiple injections of various HSA-drug conjugates were well tolerated without signs of allergy or anaphylaxis.


Subject(s)
Carcinoma 256, Walker/metabolism , Drug Carriers/pharmacokinetics , Serum Albumin/pharmacokinetics , Animals , Antineoplastic Agents/pharmacokinetics , Drug Carriers/chemistry , Drug Delivery Systems/methods , Female , Humans , Liver/metabolism , Mice , Neoplasm Transplantation , Rats , Serum Albumin/chemistry , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/pharmacokinetics , Species Specificity
15.
Anticancer Drugs ; 10(4): 405-11, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10378676

ABSTRACT

Based on the rationale of a preferred albumin uptake by tumors, conjugates comprising of rat serum albumin (RSA) as a drug carrier and of methotrexate (MTX) as chemotherapeutic drug were prepared. For a comparative study of MTX-RSA and MTX we chose a slow growing Dunning R3327 HI prostate cancer model. In a radiopharmacologic study blood kinetics and the tumor and organ distribution pattern of residualizingly labeled MTX-RSA were determined, and were found to be similar to that of residualizingly labeled RSA. The MTD was established for Copenhagen rats at a total four injections of 2 mg/kg MTX or MTX-RSA administered at days 0, 4, 8 and 12. Tumor volume measurements and tumor removal showed a small non-significant growth delay in the MTX treatment group, suggesting MTX resistance for the Dunning R3327 HI prostate carcinoma. In contrast, treatment with MTX-RSA resulted in a significant (50%) growth inhibition of the Dunning R3327 HI tumor. The cellular mechanisms responsible for MTX resistance in Dunning HI tumor cells is not known. The improved therapeutic effects seen during MTX-RSA treatment in this slow growing adenocarcinoma might be a result of prolonged tumor exposure time and an altered cellular uptake by a lysosomal route. MTX-albumin conjugates have shown antitumor activity exceeding that of MTX in several tumor xenografts in nude mice, including human prostate cancer. The recently initiated clinical development of MTX-human serum albumin will be continued and cancer of the prostate will be included as a potential target tumor during further clinical phase II testing.


Subject(s)
Methotrexate/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Serum Albumin/therapeutic use , Animals , Body Weight , Cell Division/drug effects , Cell Line , Drug Carriers , Drug Resistance, Neoplasm , Humans , Lysosomes/metabolism , Male , Methotrexate/blood , Methotrexate/pharmacokinetics , Mice , Mice, Nude , Rats , Serum Albumin/pharmacokinetics , Tissue Distribution , Transplantation, Heterologous
16.
Clin Cancer Res ; 5(4): 753-9, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10213209

ABSTRACT

Methotrexate-albumin conjugate (MTX-HSA) is a novel human albumin-based prodrug conjugate of methotrexate (MTX). A low MTX loading rate provided optimal tumor targeting and therapeutic efficacy during preclinical testing. The objectives of this first Phase I study of MTX-HSA were to determine dose-limiting toxicity (DLT) and maximum tolerated dose (MTD) in a weekly regimen. Seventeen cancer patients who were no longer amenable to standard treatment were enrolled and were evaluable for DLT. Up to eight injections were performed in weekly intervals. Dose escalation was as follows: 20, 40, 50, and then 60 mg/m2 MTX-HSA (based on the amount of MTX bound to albumin). Additional MTX-HSA courses were feasible in case of tumor response. DLT (mainly stomatitis, Common Toxicity Criteria grade 3) occurred, beginning at the 50 mg/m2 dose level after repeated administrations; in one case, thrombocytopenia was dose-limiting. Two events of DLT occurred at the 60 mg/m2 dose level within the first two administrations. Mild anemia, transaminitis, and one case of skin toxicity were found. No significant leukopenia, nausea, renal toxicity, or other toxicities were observed. MTX-HSA was well tolerated. Drug accumulation occurred on the weekly schedule. The half-life of the drug was estimated to be up to 3 weeks. Tumor responses were seen in three patients: (a) a partial response was seen in one patient with renal cell carcinoma (response duration, 30 months, ongoing); (b) a minor response was seen in one patient with pleural mesothelioma (response duration, 31 months, ongoing); and (c) a minor response was seen in one patient with renal cell carcinoma (response duration, 14 months until progression). Poststudy treatment was administered at 2-4-week intervals. No signs of toxicity or drug accumulation were seen. Altered pharmacological properties of MTX-HSA such as plasma half-life, tumor targeting, or intracellular metabolism might have contributed to these responses. The MTD for weekly administration was 4 x 50 mg/m2 MTX-HSA during short-term treatment. A regimen with MTX-HSA injections of 50 mg/m2 every 2 weeks was recommended for a further clinical Phase I study.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Methotrexate/administration & dosage , Methotrexate/therapeutic use , Neoplasms/drug therapy , Serum Albumin/administration & dosage , Serum Albumin/therapeutic use , Adolescent , Adult , Aged , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Carcinoma, Renal Cell/drug therapy , Dose-Response Relationship, Drug , Female , Humans , Kidney Neoplasms/drug therapy , Male , Mesothelioma/drug therapy , Methotrexate/pharmacokinetics , Methotrexate/toxicity , Middle Aged , Pleural Neoplasms/drug therapy , Remission Induction , Serum Albumin/pharmacokinetics , Serum Albumin/toxicity , Spinal Cord Neoplasms/drug therapy , Spinal Cord Neoplasms/secondary
20.
Int J Cancer ; 76(6): 884-90, 1998 Jun 10.
Article in English | MEDLINE | ID: mdl-9626357

ABSTRACT

We have recently reported that albumin accumulates in solid tumors and serves there as a source of nitrogen and energy. Methotrexate-albumin conjugates [MTX(I)-RSA] derivatized at a molar ratio of 1:1 differ favorably from original MTX in terms of plasma presence and tumor uptake. The purpose of this study was to evaluate the therapeutic efficacy of these novel conjugates in a comparative study with low m.w. MTX is Sprague-Dawley rats bearing a Walker-256 carcinoma. The maximum tolerated dose (MTD) for MTX and MTX(I)-RSA was determined (2 mg/kg based on MTX injected on days 1, 3 and 8). The tumor-bearing rats received injections of either the MTD or MTD/2 of MTX, MTX-albumin or mixtures containing the MTD/2 or MTD/4 of both MTX and MTX-albumin. No toxic side effects were observed. Cure rate and tumor growth retardation were slightly better for the conjugate compared with MTX alone in the MTD group (16 complete remissions vs. 14 of 20 rats). The best results were achieved for the combination treatment with MTX and MTX-albumin, with complete remission in all 20 rats. In conclusion, MTX-albumin conjugates show therapeutic activity in vivo without toxic side effects. Additive effects were observed for a combination of MTX-albumin and MTX. These effects might be caused by the much longer tumor exposition time of the conjugate in conjunction with a different route of uptake (pinocytosis for MTX-albumin vs. folate receptors for MTX).


Subject(s)
Albumins/administration & dosage , Antimetabolites, Antineoplastic/administration & dosage , Carcinoma 256, Walker/drug therapy , Methotrexate/administration & dosage , Animals , Female , Methotrexate/toxicity , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...