Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
1.
Photochem Photobiol Sci ; 17(5): 539-551, 2018 May 16.
Article in English | MEDLINE | ID: mdl-29565434

ABSTRACT

Triple-negative breast cancer (TNBC) and malignant melanoma are highly aggressive cancers that widely express the cell surface chondroitin sulfate proteoglycan 4 (CSPG4/NG2). CSPG4 plays an important role in tumor cell growth and survival and promotes chemo- and radiotherapy resistance, suggesting that CSPG4 is an attractive target in cancer therapy. In the present work, we applied the drug delivery technology photochemical internalization (PCI) in combination with the novel CSPG4-targeting immunotoxin 225.28-saporin as an efficient and specific strategy to kill aggressive TNBC and amelanotic melanoma cells. Light-activation of the clinically relevant photosensitizer TPCS2a (fimaporfin) and 225.28-saporin was found to act in a synergistic manner, and was superior to both PCI of saporin and PCI-no-drug (TPCS2a + light only) in three TNBC cell lines (MDA-MB-231, MDA-MB-435 and SUM149) and two BRAFV600E mutated malignant melanoma cell lines (Melmet 1 and Melmet 5). The cytotoxic effect was highly dependent on the light dose and expression of CSPG4 since no enhanced cytotoxicity of PCI of 225.28-saporin compared to PCI of saporin was observed in the CSPG4-negative MCF-7 cells. The PCI of a smaller, and clinically relevant CSPG4-targeting toxin (scFvMEL-rGel) validated the CSPG4-targeting concept in vitro and induced a strong inhibition of tumor growth in the amelanotic melanoma xenograft A-375 model. In conclusion, the combination of the drug delivery technology PCI and CSPG4-targeting immunotoxins is an efficient, specific and light-controlled strategy for the elimination of aggressive cells of TNBC and malignant melanoma origin. This study lays the foundation for further preclinical evaluation of PCI in combination with CSPG4-targeting.


Subject(s)
Antineoplastic Agents/pharmacology , Chondroitin Sulfate Proteoglycans/antagonists & inhibitors , Immunotoxins/pharmacology , Melanoma/drug therapy , Membrane Proteins/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Chondroitin Sulfate Proteoglycans/metabolism , Dose-Response Relationship, Drug , Drug Delivery Systems , Drug Screening Assays, Antitumor , Humans , Immunotoxins/chemistry , Light , Melanoma/metabolism , Melanoma/pathology , Membrane Proteins/metabolism , Mice , Photochemical Processes , Structure-Activity Relationship , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured
2.
Oncogene ; 35(17): 2145-55, 2016 04 28.
Article in English | MEDLINE | ID: mdl-26300004

ABSTRACT

UNLABELLED: Fibroblast growth factor-inducible 14 (Fn14; TNFRSF12A) is the cell surface receptor for the tumor necrosis factor (TNF) family member TNF-like weak inducer of apoptosis (TWEAK). The Fn14 gene is normally expressed at low levels in healthy tissues but expression is significantly increased after tissue injury and in many solid tumor types, including glioblastoma (GB; formerly referred to as 'GB multiforme'). GB is the most common and aggressive primary malignant brain tumor and the current standard-of-care therapeutic regimen has a relatively small impact on patient survival, primarily because glioma cells have an inherent propensity to invade into normal brain parenchyma, which invariably leads to tumor recurrence and patient death. Despite major, concerted efforts to find new treatments, a new GB therapeutic that improves survival has not been introduced since 2005. In this review article, we summarize studies indicating that (i) Fn14 gene expression is low in normal brain tissue but is upregulated in advanced brain cancers and, in particular, in GB tumors exhibiting the mesenchymal molecular subtype; (ii) Fn14 expression can be detected in glioma cells residing in both the tumor core and invasive rim regions, with the maximal levels found in the invading glioma cells located within normal brain tissue; and (iii) TWEAK: Fn14 engagement as well as Fn14 overexpression can stimulate glioma cell migration, invasion and resistance to chemotherapeutic agents in vitro. We also discuss two new therapeutic platforms that are currently in development that leverage Fn14 overexpression in GB tumors as a way to deliver cytotoxic agents to the glioma cells remaining after surgical resection while sparing normal healthy brain cells.


Subject(s)
Glioblastoma/drug therapy , Glioblastoma/genetics , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factors/genetics , Apoptosis/genetics , Cell Movement/genetics , Cytokine TWEAK , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Glioblastoma/surgery , Humans , Neoplasm Invasiveness/genetics , Receptors, Tumor Necrosis Factor/biosynthesis , TWEAK Receptor , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factors/biosynthesis
3.
Oncogene ; 34(44): 5582-92, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-25684137

ABSTRACT

The number of epidermal growth factor receptor (EGFR)-targeting drugs in the development for cancer treatment is continuously increasing. Currently used EGFR-targeted monoclonal antibodies and tyrosine kinase inhibitors have specific limitations related to toxicity and development of resistance, and there is a need for alternative treatment strategies to maximize the clinical potential of EGFR as a molecular target. This study describes the design and production of a novel EGFR-targeted fusion protein, rGel/EGF, composed of the recombinant plant toxin gelonin and EGF. rGel/EGF was custom-made for administration by photochemical internalization (PCI), a clinically tested modality for cytosolic release of macromolecular therapeutics. rGel/EGF lacks efficient mechanisms for endosomal escape and is therefore minimally toxic as monotherapy. However, PCI induces selective and efficient cytosolic release of rGel/EGF in EGFR-expressing target cells by light-directed activation of photosensitizers accumulated selectively in tumor tissue. PCI of rGel/EGF was shown to be highly effective against EGFR-expressing cell lines, including head and neck squamous cell carcinoma (HNSCC) cell lines resistant to cetuximab (Erbitux). Apoptosis, necrosis and autophagy were identified as mechanisms of action following PCI of rGel/EGF in vitro. PCI of rGel/EGF was further shown as a highly tumor-specific and potent modality in vivo, with growth inhibitory effects demonstrated on A-431 squamous cell carcinoma (SCC) xenografts and reduction of tumor perfusion and necrosis induction in SCC-026 HNSCC tumors. Considering the small amount of rGel/EGF injected per animal (0.1 mg/kg), the presented in vivo results are highly promising and warrant optimization and production of rGel/EGF for further preclinical evaluation with PCI.


Subject(s)
ErbB Receptors/metabolism , Toxins, Biological/pharmacology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Apoptosis/drug effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cetuximab/pharmacology , Drug Delivery Systems/methods , Epidermal Growth Factor/metabolism , Female , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Humans , Mice , Mice, Nude , Photochemistry/methods , Recombinant Fusion Proteins/metabolism , Ribosome Inactivating Proteins, Type 1/metabolism , Squamous Cell Carcinoma of Head and Neck
4.
Oncogene ; 33(4): 429-39, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-23376850

ABSTRACT

Targeted therapeutics are potential therapeutic agents because of their selectivity and efficacy against tumors resistant to conventional therapy. The goal of this study was to determine the comparative activity of monovalent, engineered anti-Her2/neu immunotoxins fused to recombinant gelonin (rGel) to the activity of bivalent IgG-containing immunoconjugates. Utilizing Herceptin and its derived humanized single-chain antibody (single-chain fragment variable, designated 4D5), we generated bivalent chemical Herceptin/rGel conjugate, and the corresponding monovalent recombinant immunotoxins in two orientations, 4D5/rGel and rGel/4D5. All the constructs showed similar affinity to Her2/neu-overexpressing cancer cells, but significantly different antitumor activities. The rGel/4D5 orientation construct and Herceptin/rGel conjugate were superior to 4D5/rGel construct in in vitro and in vivo efficacy. The enhanced activity was attributed to improved intracellular toxin uptake into target cells and efficient downregulation of Her2/neu-related signaling pathways. The Her2/neu-targeted immunotoxins effectively targeted cells with Her2/neu expression level >1.5 × 10(5) sites per cell. Cells resistant to Herceptin or chemotherapeutic agents were not cross-resistant to rGel-based immunotoxins. Against SK-OV-3 tumor xenografts, the rGel/4D5 construct with excellent tumor penetration showed impressive tumor inhibition. Although Herceptin/rGel conjugate demonstrated comparatively longer serum half-life, the in vivo efficacy of the conjugate was similar to the rGel/4D5 fusion. These comparative studies demonstrate that the monovalent, engineered rGel/4D5 construct displayed comparable in vitro and in vivo antitumor efficacy as bivalent Herceptin/rGel conjugate. Immunotoxin orientation can significantly impact the overall functionality and performance of these agents. The recombinant rGel/4D5 construct with excellent tumor penetration and rapid blood clearance may reduce the unwanted toxicity when administrating to patients, and warrants consideration for further clinical evaluation.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Immunoconjugates/pharmacology , Immunotoxins/pharmacology , Molecular Targeted Therapy/methods , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Blotting, Western , Cell Line, Tumor , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Immunoconjugates/chemistry , Immunohistochemistry , Immunotoxins/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology , Ribosome Inactivating Proteins, Type 1/pharmacology , Trastuzumab , Xenograft Model Antitumor Assays
5.
Leukemia ; 26(8): 1786-96, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22373785

ABSTRACT

Acute lymphoblastic leukemia (ALL) is the most common malignancy affecting children and a major cause of mortality from hematopoietic malignancies in adults. A substantial number of patients become drug resistant during chemotherapy, necessitating the development of alternative modes of treatment. rGel (recombinant Gelonin)/BlyS (B-lymphocyte stimulator) is a toxin-cytokine fusion protein used for selective killing of malignant B-cells expressing receptors for B-cell-activating factor (BAFF/BLyS) by receptor-targeted delivery of the toxin, Gelonin. Here, we demonstrate that rGel/BLyS binds to ALL cells expressing BAFF receptor (BAFF-R) and upon internalization, it induces apoptosis of these cells and causes downregulation of survival genes even in the presence of stromal protection. Using an immunodeficient transplant model for human ALL, we show that rGel/BLyS prolongs survival of both Philadelphia chromosome-positive and negative ALL-bearing mice. Furthermore, we used AMD3100, a CXCR4 antagonist, to mobilize the leukemic cells protected in the bone marrow (BM) microenvironment and the combination with rGel/BLyS resulted in a significant reduction of the tumor load in the BM and complete eradication of ALL cells from the circulation. Thus, a combination treatment with the B-cell-specific fusion toxin rGel/BLyS and the mobilizing agent AMD3100 could be an effective alternative approach to chemotherapy for the treatment of primary and relapsed ALL.


Subject(s)
Antineoplastic Agents/therapeutic use , B-Cell Activating Factor/therapeutic use , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Recombinant Fusion Proteins/therapeutic use , Ribosome Inactivating Proteins, Type 1/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/metabolism , Apoptosis/drug effects , B-Cell Activating Factor/administration & dosage , B-Cell Activating Factor/genetics , B-Cell Activating Factor/metabolism , Benzylamines , Bone Marrow/pathology , Cell Line, Tumor , Cyclams , Drug Synergism , Gene Expression Regulation, Leukemic/drug effects , Heterocyclic Compounds/pharmacology , Humans , Mice , Mice, Nude , Mice, SCID , NF-kappa B/antagonists & inhibitors , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/metabolism , Ribosome Inactivating Proteins, Type 1/administration & dosage , Ribosome Inactivating Proteins, Type 1/genetics , Ribosome Inactivating Proteins, Type 1/metabolism , Toxins, Biological/administration & dosage , Toxins, Biological/metabolism , Toxins, Biological/therapeutic use , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
Curr Pharm Des ; 15(23): 2676-92, 2009.
Article in English | MEDLINE | ID: mdl-19689338

ABSTRACT

Recombinant fusion proteins are an expanding, important class of novel therapeutic agents. The designs of these constructs typically involve a cell-targeting motif genetically fused to a highly toxic class of enzymes capable of ruthlessly attacking critical cellular machinery once delivered successfully to the cytoplasm of the target cell. Initial development of this class of constructs typically contained recombinant growth factors or single-chain antibodies as the cell-targeting motif fused to highly cytotoxic plant or bacterial toxins. This review describes second-generation molecules composed of cell-targeting molecules fused to highly cytotoxic human enzymes capable of generating intense apoptotic response once delivered to the cytoplasm. The human serine protease granzyme B has been shown to be extremely effective as a cytotoxic molecule when incorporated into numerous cell-targeting constructs. The biological activity of GrB-containing constructs rivals that of plant or bacterial toxins and appears to represent a new generation and class of completely human proteins with unique biological activities.


Subject(s)
Drug Discovery/methods , Granzymes/pharmacology , Recombinant Fusion Proteins/pharmacology , Apoptosis/drug effects , Drug Delivery Systems/methods , Granzymes/immunology , Humans , Immunotoxins/pharmacology , Models, Biological , Recombinant Fusion Proteins/biosynthesis
7.
Phys Rev E Stat Nonlin Soft Matter Phys ; 64(4 Pt 2): 045202, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11690077

ABSTRACT

We propose a method for experimental detection of directionality of weak coupling between two self-sustained oscillators from bivariate data. The technique is applicable to both noisy and chaotic systems that can be nonidentical or even structurally different. We introduce an index that quantifies the asymmetry in coupling.

8.
Phys Rev E Stat Nonlin Soft Matter Phys ; 63(5 Pt 2): 058201, 2001 May.
Article in English | MEDLINE | ID: mdl-11415052

ABSTRACT

Chen et al. [Phys. Rev. E 61, 2559 (2000)] recently proposed an extension of the concept of phase for discrete chaotic systems. Using the newly introduced definition of phase they studied the dynamics of coupled map lattices and compared these dynamics with phase synchronization of coupled continuous-time chaotic systems. In this paper we illustrate by two simple counterexamples that the angle variable introduced by Chen et al. fails to satisfy the basic requirements to the proper phase. Furthermore, we argue that an extension of the notion of phase synchronization to generic discrete maps is doubtful.

9.
Int J Cancer ; 88(2): 267-73, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11004679

ABSTRACT

Over-expression of the proto-oncogene HER2/neu in breast cancer and certain other tumors appears to be a central mechanism that may be partly responsible for cellular progression of the neoplastic phenotype. Transfection of mammalian cells and over-expression of HER2/neu appears to result in reduced sensitivity to the cytotoxic effects of tumor necrosis factor (TNF) and reduced sensitivity to immune effector killing. The single-chain recombinant antibody sFv23 recognizes the cell-surface domain of HER2/neu. The cDNA for this antibody was fused to the cDNA encoding human TNF, and this sFv23/TNF fusion construct was cloned into a plasmid for expression in Escherichia coli. The fusion protein was expressed and purified by ion-exchange chromatography. SDS-PAGE demonstrated a single band at the expected m.w. (43 kDa). Western analysis confirmed the presence of both the antibody component and the TNF component in the final fusion product. The fusion construct was tested for TNF activity against L-929 cells and found to have biological activity similar to that of authentic TNF (SA 420 nM). The scFv23/TNF construct bound to SKBR-3 (HER2-positive) but not to A-375 human melanoma (HER2-negative) cells. Cytotoxicity studies against log-phase human breast carcinoma cells (SKBR-3-HP) over-expressing HER2/neu demonstrate that the sFv23/TNF fusion construct was 1, 000-fold more active than free TNF. Tumor cells expressing higher levels of HER2/neu (SKBR-3-LP) were relatively resistant to both the fusion construct and native TNF. These studies suggest that fusion constructs targeting the HER2/neu surface domain and containing TNF are more effective cytotoxic agents in vitro than native TNF and may be effective against tumor cells expressing intermediate, but not high, levels of HER2/neu.


Subject(s)
Cell Survival/drug effects , Genes, erbB-2 , Immunotoxins/toxicity , Receptor, ErbB-2/genetics , Tumor Necrosis Factor-alpha/toxicity , Animals , Breast Neoplasms , Cloning, Molecular , Escherichia coli , Female , Humans , Immunotoxins/genetics , Immunotoxins/pharmacokinetics , Kinetics , L Cells , Melanoma , Mice , Proto-Oncogene Mas , Receptor, ErbB-2/analysis , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/toxicity , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/pharmacokinetics
10.
Cancer Chemother Pharmacol ; 44(4): 343-8, 1999.
Article in English | MEDLINE | ID: mdl-10447583

ABSTRACT

Immunotoxins are a class of targeted therapeutic agents under development by various research groups. The murine monoclonal antibody designated ZME-018 recognizes a high molecular weight glycoprotein present on most human melanoma cells and biopsy specimens and has been utilized for clinical imaging studies in patients with melanoma. The plant toxin gelonin is a ribosome-inactivating protein (RIP) with n-glycosidase activity similar to that of ricin A chain. In previous studies by our group, the gelonin toxin was sequenced, cloned and expressed in E. coli. The purified recombinant gelonin (RG) was found to have identical protein synthesis inhibitory activity to that of natural gelonin (NG). For comparative purposes, chemical conjugates of antibody ZME and either RG or NG were produced using the heterobifunctional crosslinking reagents SPDP and SMPT. The ZME-NG and ZME-RG immunotoxins were found to be 10(4)- to 10(5)-fold more cytotoxic to antigen-positive human melanoma cells than free toxin. NG toxin alone was cytotoxic to intact cells (IC(50) = 100 nM) while RG was nontoxic to cells at doses up to 1 microM. Both ZME-NG and ZME-RG immunoconjugates were nontoxic to antigen-negative (Me-180) cells. ZME-RG immunotoxins constructed with the more stable SMPT reagent were slightly more effective in culture than conjugates made with SPDP. Tissue distribution studies in tumor-bearing nude mice demonstrated that tumor uptake of the ZME-RG immunotoxin was similar to that of the intact ZME antibody with reduced distribution to normal organs compared to an immunoconjugate produced with NG. Pharmacokinetic studies showed that the terminal-phase plasma half-life of ZME-RG was similar to that of ZME itself (42 h vs 50 h) and almost threefold higher than that of ZME-NG (11.5 h). The area under the concentration curve (Cxt) for ZME-RG was 50% lower than that for ZME due to an increased apparent volume of distribution (Vd(a)) but was almost tenfold higher than the Cxt for ZME-NG. These studies suggest that immunoconjugates comprising RG demonstrate identical in vitro cytotoxic effects to immunoconjugates produced with NG and immunotoxins with RG display improved in vivo pharmacodynamics and tissue distribution compared to immunotoxins containing NG.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/toxicity , Immunotoxins/pharmacokinetics , Immunotoxins/toxicity , Melanoma/drug therapy , Plant Proteins/pharmacokinetics , Plant Proteins/toxicity , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/toxicity , Cross-Linking Reagents/chemistry , Immunotoxins/chemistry , Melanoma/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Synthesis Inhibitors/pharmacokinetics , Protein Synthesis Inhibitors/toxicity , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/toxicity , Ribosome Inactivating Proteins, Type 1 , Succinimides/chemistry , Tumor Cells, Cultured
11.
Clin Cancer Res ; 5(5): 953-61, 1999 May.
Article in English | MEDLINE | ID: mdl-10353726

ABSTRACT

The tumor-associated glycoprotein 72 (TAG-72) antigen is present on a high percentage of tumor types including ovarian carcinomas. Antibody B72.3 is a murine monoclonal recognizing the surface domain of the TAG-72 antigen and has been widely used in human clinical trials. After our initial encouraging studies (M. G. Rosenblum et al., J. Natl. Cancer Inst., 83: 1629-1636, 1991) of tissue disposition, metabolism, and pharmacokinetics in 9 patients with ovarian cancer, we designed an escalating dose, multi-arm Phase I study of 90Y-labeled B72.3 i.p. administration. In the first arm of the study, patients (3 pts/dose level) received an i.p. infusion of either 2 or 10 mg of B72.3 labeled with either 1, 10, 15, or 25 mCi of 90Y. Pharmacokinetic studies demonstrated that concentrations of 90Y-labeled B72.3 persist in peritoneal fluid with half-lives >24 h after i.p. administration. In addition, 90Y-labeled B72.3 was absorbed rapidly into the plasma with peak levels achieved within 48 h, and levels declined slowly thereafter. Cumulative urinary excretion of the 90Y label was 10-20% of the administered dose which suggests significant whole-body retention of the radiolabel. Biopsy specimens of bone and marrow obtained at 72 h after administration demonstrated significant content of the label in bone (0.015% of the dose/g) with relatively little in marrow (0.005% of the dose/g). The maximal tolerated dose was determined to be 10 mCi because of hematological toxicity and platelet suppression. This typically occurred on the 29th day after administration and was thought to be a consequence of the irradiation of the marrow from the bony deposition of the radiolabel. In an effort to suppress the bone uptake of 90Y, patients were treated with a continuous i.v. infusion of EDTA (25 mg/kg/12 h x 6) infused immediately before i.p. administration of the radiolabeled antibody. Patients (3 pts/dose level) were treated with doses of 10, 15, 20, 25, 30, 35, 40, or 45 mCi of 90Y-labeled B72.3 for a total of 38 patients. EDTA administration resulted in significant myeloprotection, which allowed escalation to the maximal tolerated dose of 40 mCi. Dose-limiting toxicity was thrombocytopenia and neutropenia. Studies of plasma and peritoneal fluid pharmacokinetics demonstrate no changes compared with patients without EDTA pretreatment. Cumulative urinary excretion of the radiolabel was not increased in patients pretreated with EDTA compared with the untreated group. However, analysis of biopsy specimens of bone and marrow demonstrated that bone and marrow content of the 90Y label was 15-fold lower (<0.001% injected dose/g) than a companion group without EDTA. Four responses were noted in patients who received 15-30 mCi of 90Y-labeled B72.3 with response durations of 1-12 months. These results demonstrate the myeloprotective ability of EDTA, which allows safe i.p. administration of higher doses of 90Y-labeled B72.3 and, therefore, clearly warrant an expanded Phase II trial in patients with minimal residual disease after standard chemotherapy or for the palliation of refractory ascites.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Antigens, Neoplasm/immunology , Bone Marrow Diseases/prevention & control , Carcinoma/radiotherapy , Chelating Agents/therapeutic use , Edetic Acid/therapeutic use , Glycoproteins/immunology , Ovarian Neoplasms/radiotherapy , Radiation Injuries/prevention & control , Radioimmunotherapy , Radioisotopes/therapeutic use , Ytterbium/therapeutic use , Adult , Aged , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Neoplasm/administration & dosage , Antibodies, Neoplasm/immunology , Ascites/radiotherapy , Ascitic Fluid/chemistry , Bone Marrow/chemistry , Bone Marrow/radiation effects , Bone Marrow Diseases/chemically induced , Bone and Bones/chemistry , Carcinoma/pathology , Carcinoma/secondary , Carcinoma/therapy , Chelation Therapy , Dose-Response Relationship, Immunologic , Dose-Response Relationship, Radiation , Edetic Acid/administration & dosage , Edetic Acid/pharmacology , Fallopian Tube Neoplasms/pathology , Fallopian Tube Neoplasms/radiotherapy , Fallopian Tube Neoplasms/therapy , Female , Half-Life , Humans , Injections, Intraperitoneal , Mice , Middle Aged , Neoplasm, Residual , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Peritoneal Neoplasms/radiotherapy , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/therapy , Radiation Injuries/chemically induced , Radioimmunotherapy/adverse effects , Radioisotopes/administration & dosage , Radioisotopes/adverse effects , Radioisotopes/pharmacokinetics , Radiotherapy Dosage , Tissue Distribution , Treatment Outcome , Ytterbium/administration & dosage , Ytterbium/adverse effects , Ytterbium/pharmacokinetics
12.
Nature ; 399(6735): 461-5, 1999 Jun 03.
Article in English | MEDLINE | ID: mdl-10365957

ABSTRACT

There is evidence that physiological signals under healthy conditions may have a fractal temporal structure. Here we investigate the possibility that time series generated by certain physiological control systems may be members of a special class of complex processes, termed multifractal, which require a large number of exponents to characterize their scaling properties. We report on evidence for multifractality in a biological dynamical system, the healthy human heartbeat, and show that the multifractal character and nonlinear properties of the healthy heart rate are encoded in the Fourier phases. We uncover a loss of multifractality for a life-threatening condition, congestive heart failure.


Subject(s)
Fractals , Heart/physiology , Myocardial Contraction/physiology , Adult , Aged , Databases, Factual , Female , Heart/physiopathology , Heart Failure/physiopathology , Hemodynamics , Humans , Male , Middle Aged , Models, Cardiovascular
13.
Clin Cancer Res ; 5(4): 865-74, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10213223

ABSTRACT

TAB-250 and BACH-250 are murine and human chimeric antibodies directed at the extracellular domain of the gp185c-erb-2 (HER2/neu) growth factor receptor overexpressed in a variety of tumor types, including ovarian and breast carcinoma. The ribosome-inhibiting plant toxin gelonin (rGel) was chemically coupled to both antibodies, and the resulting immunotoxins were purified and tested in vitro against human tumor cells expressing various levels of HER-2/neu and in vivo against human tumor xenograft models. The binding of both BACH-250 and BACH-250/rGel conjugate to target cells was essentially equivalent. Against SKOV-3 cells, the IC50 of BACH-250/rGel was 97 pM (17 ng/ml), whereas BACH-250 and rGel alone showed no cytotoxic effects. There was a clear correlation between expression levels of HER-2/neu and cytoimmunotoxin. Tissue distribution studies showed that the antibody and immunotoxin both concentrate 2-10-fold higher in tumors than in normal tissues, with optimal tumor uptake occurring 48-96 h after administration. Plasma clearance curves for BACH-250 and BACH-250/rGel showed terminal-phase half-lives of 26 and 72 h, respectively. In athymic mice bearing s.c. or i.p. SKOV-3 tumors, immunotoxin treatment slowed tumor growth by 99 and 94 % at days 35 and 49 after implantation, respectively, and lengthened the median survival by 40% (from 30 to 50 days) in mice bearing lethal i.p. tumors. We conclude that clinical development of BACH-250/rGel may be warranted in patients with HER2/neu-expressing malignancies.


Subject(s)
Immunotoxins/pharmacokinetics , Immunotoxins/therapeutic use , Ovarian Neoplasms/drug therapy , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use , Animals , Antibodies, Monoclonal , Binding, Competitive/drug effects , Cell Line , Dose-Response Relationship, Drug , Female , Humans , Immunoconjugates , Immunotoxins/toxicity , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Recombinant Fusion Proteins/toxicity , Survival Rate , Tissue Distribution , Tumor Stem Cell Assay
14.
Article in English | MEDLINE | ID: mdl-11969830

ABSTRACT

We investigate synchronization between cardiovascular and respiratory systems in healthy humans under free-running conditions. For this aim we analyze nonstationary irregular bivariate data, namely, electrocardiograms and measurements of respiratory flow. We briefly discuss a statistical approach to synchronization in noisy and chaotic systems and illustrate it with numerical examples; effects of phase and frequency locking are considered. Next, we present and discuss methods suitable for the detection of hidden synchronous epochs from such data. The analysis of the experimental records reveals synchronous regimes of different orders n:m and transitions between them; the physiological significance of this finding is discussed.


Subject(s)
Cardiovascular System , Respiratory System , Humans , Models, Statistical , Oscillometry , Time Factors
16.
Clin Cancer Res ; 4(8): 1971-6, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9717827

ABSTRACT

The recently characterized immunotoxin HuM195-gelonin consists of a humanized anti-CD33 monoclonal antibody conjugated to the single-chain plant toxin gelonin. Binding of the immunotoxin to hematopoietic cells that express the CD33 differentiation antigen has been demonstrated and results in cytotoxicity due to ribosomal inactivation by gelonin. Blast cells from most patients with acute myelogenous leukemia express CD33, whereas normal stem cells necessary for maintenance of hematopoiesis do not. We asked whether an immunoconjugate using recombinant gelonin rather than plant gelonin is toxic to acute myelogenous leukemia (AML) cell lines and primary AML blasts obtained from patients and exposed to the immunotoxin in vitro. The CD33pos cell lines HL60, OCI/AML2, and OCI/ AML5 showed decreased proliferation when exposed to immunotoxin for 24-72 h. The CD33neg cell line OCI/AML3 was relatively resistant to HuM195, and all cell lines were resistant to equimolar concentrations of unconjugated antibody and gelonin. Primary blast cultures from seven patients with AML had CD33 detectable on 75.7-99.8% of cells by flow cytometry, and all showed dose-dependent decreases in clonogenic cell survival during 24-h incubation with the immunotoxin. Cells selected for low CD33 expression by cell sorting or by prolonged incubation with immunotoxin could reexpress CD33 at baseline levels and remained sensitive to immunotoxin. We conclude that humanized M195 conjugated to recombinant gelonin has antileukemic activity and should be considered for clinical testing in Phase I trials.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Antineoplastic Agents, Phytogenic/pharmacology , Immunotoxins/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Plant Proteins/pharmacology , Antibodies, Monoclonal/immunology , Bone Marrow Transplantation , Cryopreservation , Humans , Leukemia, Myeloid, Acute/pathology , Recombinant Proteins/pharmacology , Ribosome Inactivating Proteins, Type 1 , Sensitivity and Specificity , Sialic Acid Binding Ig-like Lectin 3 , Tumor Cells, Cultured
18.
Physica A ; 249: 587-93, 1998.
Article in English | MEDLINE | ID: mdl-11541514

ABSTRACT

We find that a universal homogeneous scaling form describes the distribution of cardiac variations for a group of healthy subjects, which is stable over a wide range of time scales. However, a similar scaling function does not exist for a group with a common cardiopulmonary instability associated with sleep apnea. Subtle differences in the distributions for the day- and night-phase dynamics for healthy subjects are detected.


Subject(s)
Data Interpretation, Statistical , Fractals , Heart Rate , Sleep Apnea Syndromes/physiopathology , Adult , Circadian Rhythm , Female , Humans , Male , Middle Aged , Time Factors
19.
Clin Cancer Res ; 3(9): 1547-55, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9815842

ABSTRACT

Preclinical studies have demonstrated that recombinant IFN-alpha (rIFN-alpha) can enhance the tumor associated glycoprotein 72 (TAG-72) on tumors. To determine whether rIFN-alpha could enhance TAG-72 expression in vivo in patients, 15 women with breast cancer were randomized to receive daily injections of rIFN-alpha (3 x 10(6) units/m2 for 14 days) beginning on day 1 (group 1 = 7 patients) or on day 6 (group 2 = 8 patients). On day 3, all patients received a 10-20-mCi tracer dose of 131I-CC49, a high-affinity murine monoclonal antibody reactive against TAG-72, followed by a therapy dose of 60-75 mCi/m2 of 131I-CC49 on day 6. Whole body and single-photon emission computed tomography scans along with whole blood pharmacokinetics were performed following tracer and treatment phases. Hematological toxicity was considerable; reversible grade 3-4 neutropenia and thrombocytopenia was observed in 12 of 15 patients. Twelve of 14 patients tested developed human antimouse antibodies 3-6 weeks after treatment. For group 1 patients, whole blood residence time increased significantly between that predicted from the tracer doses and therapy doses (42.6 +/- 4.7 versus 51.5 +/- 4.8 h, respectively; P < 0.01). The calculated radiation absorbed dose to red marrow from therapy compared to tracer activity was also significantly higher for this group (1.25 +/- 0.35 versus 1. 07 +/- 0.26 cGy/mCi; P < 0.05). Treatment with rIFN-alpha was found to enhance TAG-72 expression in tumors from patients receiving rIFN-alpha (group 1) by 46 +/- 19% (P < 0.05) compared to only 1.3 +/- 0.95% in patients not initially receiving IFN (group 2). The uptake of CC49 in tumors was also significantly increased in rIFN-alpha-treated patients. One partial and two minor tumor responses were seen. In summary, rIFN-alpha treatment altered the pharmacokinetics and tumor uptake of 131I-CC49 in patients at the expense of increased toxicity.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antigens, Neoplasm/immunology , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Glycoproteins/immunology , Immunoconjugates/pharmacokinetics , Interferon-alpha/pharmacology , Iodine Radioisotopes/pharmacokinetics , Radioimmunotherapy , Adult , Animals , Antibodies, Anti-Idiotypic/biosynthesis , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/therapeutic use , Antibody Specificity , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Bone Marrow/radiation effects , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/immunology , Breast Neoplasms/radiotherapy , Combined Modality Therapy , Drug Administration Schedule , Female , Gene Expression Regulation, Neoplastic/drug effects , Glycoproteins/biosynthesis , Glycoproteins/genetics , Humans , Immunoconjugates/adverse effects , Immunoconjugates/therapeutic use , Interferon alpha-2 , Interferon-alpha/administration & dosage , Interferon-alpha/therapeutic use , Iodine Radioisotopes/adverse effects , Iodine Radioisotopes/therapeutic use , Lymphatic Metastasis/radiotherapy , Mice , Middle Aged , Neoplasm Metastasis , Neutropenia/chemically induced , Recombinant Proteins , Thrombocytopenia/chemically induced , Tissue Distribution , Tomography, Emission-Computed, Single-Photon , Treatment Outcome
20.
Nature ; 383(6598): 323-7, 1996 Sep 26.
Article in English | MEDLINE | ID: mdl-8848043

ABSTRACT

Biological time-series analysis is used to identify hidden dynamical patterns which could yield important insights into underlying physiological mechanisms. Such analysis is complicated by the fact that biological signals are typically both highly irregular and non-stationary, that is, their statistical character changes slowly or intermittently as a result of variations in background influences. Previous statistical analyses of heartbeat dynamics have identified long-range correlations and power-law scaling in the normal heartbeat, but not the phase interactions between the different frequency components of the signal. Here we introduce a new approach, based on the wavelet transform and an analytic signal approach, which can characterize non-stationary behaviour and elucidate such phase interactions. We find that, when suitably rescaled, the distributions of the variations in the beat-to-beat intervals for all healthy subjects are described by a single function stable over a wide range of timescales. However, a similar scaling function does not exist for a group with cardiopulmonary instability caused by sleep apnoea. We attribute the functional form of the scaling observed in the healthy subjects to underlying nonlinear dynamics, which seem to be essential to normal heart function. The approach introduced here should be useful in the analysis of other nonstationary biological signals.


Subject(s)
Electrocardiography , Heart Rate/physiology , Models, Cardiovascular , Models, Statistical , Adult , Algorithms , Female , Fourier Analysis , Humans , Male , Middle Aged , Reference Values , Sleep Apnea Syndromes/physiopathology , Time
SELECTION OF CITATIONS
SEARCH DETAIL
...