Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Ther Innov Regul Sci ; 57(2): 386-395, 2023 03.
Article in English | MEDLINE | ID: mdl-36401076

ABSTRACT

The U.S. Food and Drug Administration and European Commission have developed successful orphan drug legislation to promote the research, development, and marketing approval of drugs to treat rare diseases. Central to these regulations are the concepts of structural similarity and clinical superiority/significant benefit to achieve orphan drug exclusivity. However, differences in health authority expectations remain regarding the qualification for an orphan drug designation, defining structural similarity, and demonstrating clinical superiority/significant benefit. These differences can create sponsor company uncertainty regarding the approvability of products (e.g., blocking risk by an existing orphan product) and divergent orphan drug decisions among health authorities. A comprehensive assessment of current regulations, case studies in exclusivities, and recommendations for improvement are presented.


Subject(s)
Drug Approval , Orphan Drug Production , United States , Humans , European Union , Rare Diseases/drug therapy , Marketing
4.
Oncotarget ; 8(27): 44605-44624, 2017 Jul 04.
Article in English | MEDLINE | ID: mdl-28574834

ABSTRACT

A key task in developing the field of personalized cancer therapy is the identification of novel molecular targets that enable treatment of cancers not susceptible to other means of specific therapy. The collagen receptor uPARAP/Endo180 is overexpressed by malignant cells in several non-epithelial cancers, notably including sarcomas, glioblastomas and subsets of acute myeloid leukemia. In contrast, in healthy adult individuals, expression is restricted to minor subsets of mesenchymal cells. Functionally, uPARAP/Endo180 is a rapidly recycling endocytic receptor that delivers its cargo directly into the endosomal-lysosomal system, thus opening a potential route of entry into receptor-positive cells. This combination of specific expression and endocytic function appears well suited for targeting of uPARAP/Endo180-positive cancers by antibody-drug conjugate (ADC) mediated drug delivery. Therefore, we utilized a specific monoclonal antibody against uPARAP/Endo180, raised through immunization of a uPARAP/Endo180 knock-out mouse, which reacts with both the human and the murine receptor, to construct a uPARAP-directed ADC. This antibody was coupled to the highly toxic dolastatin derivative, monomethyl auristatin E, via a cathepsin-labile valine-citrulline linker. With this ADC, we show strong and receptor-dependent cytotoxicity in vitro in uPARAP/Endo180-positive cancer cell lines of sarcoma, glioblastoma and leukemic origin. Furthermore, we demonstrate the potency of the ADC in vivo in a xenograft mouse model with human uPARAP/Endo180-positive leukemic cells, obtaining a complete cure of all tested mice following intravenous ADC treatment with no sign of adverse effects. Our study identifies uPARAP/Endo180 as a promising target for novel therapy against several highly malignant cancer types.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Immunoconjugates/pharmacology , Mannose-Binding Lectins/antagonists & inhibitors , Membrane Glycoproteins/antagonists & inhibitors , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Mitogen/antagonists & inhibitors , Animals , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Survival , Disease Models, Animal , Endocytosis , Gene Expression , Humans , Leukemia/drug therapy , Leukemia/metabolism , Leukemia/mortality , Leukemia/pathology , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Molecular Targeted Therapy , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Mitogen/genetics , Receptors, Mitogen/metabolism , Sarcoma/drug therapy , Sarcoma/metabolism , Sarcoma/mortality , Sarcoma/pathology , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
5.
Sci Rep ; 6: 25833, 2016 05 12.
Article in English | MEDLINE | ID: mdl-27169360

ABSTRACT

C4.4A is a modular glycolipid-anchored Ly6/uPAR/α-neurotoxin multidomain protein that exhibits a prominent membrane-associated expression in stratified squamous epithelia. C4.4A is also expressed in various solid cancer lesions, where high expression levels often are correlated to poor prognosis. Circumstantial evidence suggests a role for C4.4A in cell adhesion, migration, and invasion, but a well-defined biological function is currently unknown. In the present study, we have generated and characterized the first C4.4A-deficient mouse line to gain insight into the functional significance of C4.4A in normal physiology and cancer progression. The unchallenged C4.4A-deficient mice were viable, fertile, born in a normal Mendelian distribution and, surprisingly, displayed normal development of squamous epithelia. The C4.4A-deficient mice were, nonetheless, significantly lighter than littermate controls predominantly due to differences in fat mass. Congenital C4.4A deficiency delayed migration of keratinocytes enclosing incisional skin wounds in male mice. In chemically induced bladder carcinomas, C4.4A deficiency attenuated the incidence of invasive lesions despite having no effect on total tumour burden. This new C4.4A-deficient mouse line provides a useful platform for future studies on functional aspects of C4.4A in tumour cell invasion in vivo.


Subject(s)
Cell Adhesion Molecules/genetics , Epidermis/embryology , Epidermis/metabolism , GPI-Linked Proteins/genetics , Gene Deletion , Animals , Body Weight , Carcinoma, Lewis Lung , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/metabolism , Energy Metabolism/genetics , Epidermis/ultrastructure , Epithelium/ultrastructure , Female , GPI-Linked Proteins/deficiency , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Developmental , Gene Targeting , Magnetic Resonance Imaging , Male , Mice, Inbred C57BL , Neoplasm Transplantation , Phenotype , Subcellular Fractions/metabolism , Thinness/metabolism , Tomography, X-Ray Computed , Urinary Bladder/pathology , Water Loss, Insensible , Wound Healing
6.
Clin Cancer Res ; 22(10): 2427-34, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26673797

ABSTRACT

PURPOSE: To investigate the prognostic and predictive biomarker value of type IV collagen in colorectal cancer. EXPERIMENTAL DESIGN: Retrospective evaluation of two independent cohorts of patients with colorectal cancer included prospectively in 2004-2005 (training set) and 2006-2008 (validation set). Plasma samples were available from 297 (training set) and 482 (validation set) patients. Type IV collagen determinations were performed using an ELISA. From the training set, 222 tumors were available for IHC. Clinical and follow-up data were retrieved from patient files and national registries. RESULTS: High levels of type IV collagen showed independent prognostic significance in both cohorts with hazard ratios (HRs; for a one-unit change on the log base 2 scale) of 2.25 [95% confidence intervals (CIs), 1.78-2.84; P < 0.0001] and 2.24 (95% CI, 1.75-2.86; P < 0.0001) for the training and validation set, respectively. The prognostic impact was present both in patients with metastatic and nonmetastatic disease. The predictive value of the marker was investigated in stage II and III patients. In the training set, type IV collagen was prognostic both in the subsets of patients receiving and not receiving adjuvant antineoplastic therapy. However, in the validation set, the prognostic effect of the marker vanished when looking at patients who received adjuvant antineoplatic therapy (HR 0.90; 95% CI, 0.42-1.93) but was still present in the group not receiving adjuvant chemotherapy (HR 2.88; 95% CI, 1.98-4.21). CONCLUSIONS: The results indicate clinical validity of type IV collagen as a prognostic biomarker in colorectal cancer, although the suggested predictive role of the marker should be validated. Clin Cancer Res; 22(10); 2427-34. ©2015 AACR.


Subject(s)
Collagen Type IV/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Aged , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Chemotherapy, Adjuvant/methods , Colorectal Neoplasms/drug therapy , Female , Humans , Male , Neoplasm Staging/methods , Prognosis , Proportional Hazards Models , Retrospective Studies
7.
Mol Carcinog ; 55(2): 193-208, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25594187

ABSTRACT

Metastatic growth by colorectal cancer cells in the liver requires the ability of the cancer cells to interact with the new microenvironment. This interaction results in three histological growth patterns of liver metastases: desmoplastic, pushing, and replacement. In primary colorectal cancer several proteases, involved in the degradation of extracellular matrix components, are up-regulated. In liver metastases, their expression is growth pattern dependent. Tissue inhibitor of matrix metalloproteinase-1 (TIMP-1) is a strong prognostic marker in plasma from colorectal cancer patients, with significant higher levels in patients with metastatic disease. We therefore wanted to determine the expression pattern of TIMP-1 in primary colorectal cancers and their matching liver metastases. TIMP-1 mRNA was primarily seen in α-smooth-muscle actin (α-SMA)-positive cells. In all primary tumors and liver metastases with desmoplastic growth pattern, TIMP-1 mRNA was primarily found in α-SMA-positive myofibroblasts located at the invasive front. Some α-SMA-positive cells with TIMP-1 mRNA were located adjacent to CD34-positive endothelial cells, identifying them as pericytes. This indicates that TIMP-1 in primary tumors and liver metastases with desmoplastic growth pattern has dual functions; being an MMP-inhibitor at the cancer periphery and involved in tumor-induced angiogenesis in the pericytes. In the liver metastases with pushing or replacement growth patterns, TIMP-1 was primarily expressed by activated hepatic stellate cells at the metastasis/liver parenchyma interface. These cells were located adjacent to CD34-positive endothelial cells, suggesting a function in tumor-induced angiogenesis. We therefore conclude that TIMP-1 expression is growth pattern dependent in colorectal cancer liver metastases.


Subject(s)
Colorectal Neoplasms/blood supply , Liver Neoplasms/secondary , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Actins/metabolism , Adult , Aged , Cell Line, Tumor , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Middle Aged , Pericytes/metabolism
8.
Clin Exp Metastasis ; 32(6): 543-54, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26040548

ABSTRACT

Urokinase-type plasminogen activator (uPA) is an extracellular protease that plays a pivotal role in tumor progression. uPA activity is spatially restricted by its anchorage to high-affinity uPA receptors (uPAR) at the cell surface. High tumor tissue expression of uPA and uPAR is associated with poor prognosis in lung, breast, and colon cancer patients in clinical studies. Genetic deficiency of uPA leads to a significant reduction in metastases in the murine transgenic MMTV-PyMT breast cancer model, demonstrating a causal role for uPA in cancer dissemination. To investigate the role of uPAR in cancer progression, we analyze the effect of uPAR deficiency in the same cancer model. uPAR is predominantly expressed in stromal cells in the mouse primary tumors, similar to human breast cancer. In a cohort of MMTV-PyMT mice [uPAR-deficient (n = 31) or wild type controls (n = 33)], tumorigenesis, tumor growth, and tumor histopathology were not significantly affected by uPAR deficiency. Lung and lymph node metastases were also not significantly affected by uPAR deficiency, in contrast to the significant reduction seen in uPA-deficient mice. Taken together, our data show that the genetic absence of uPAR does not influence the outcome of the MMTV-PyMT cancer model.


Subject(s)
Breast Neoplasms/pathology , Lung Neoplasms/secondary , Lymph Nodes/pathology , Receptors, Urokinase Plasminogen Activator/physiology , Stromal Cells/pathology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , Humans , Immunoenzyme Techniques , In Situ Hybridization , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lymph Nodes/metabolism , Lymphatic Metastasis , Mice , Mice, Transgenic , Receptors, Urokinase Plasminogen Activator/antagonists & inhibitors , Stromal Cells/metabolism
9.
Neoplasia ; 17(5): 421-33, 2015 May.
Article in English | MEDLINE | ID: mdl-26025665

ABSTRACT

The stroma of breast cancer can promote the disease's progression, but whether its composition and functions are shared among different subtypes is poorly explored. We compared stromal components of a luminal [mouse mammary tumor virus (MMTV)-Neu] and a triple-negative/basal-like [C3(1)-Simian virus 40 large T antigen (Tag)] genetically engineered breast cancer mouse model. The types of cytokines and their expression levels were very different in the two models, as was the extent of innate immune cell infiltration; however, both models showed infiltration of innate immune cells that expressed matrix metalloproteinase 9 (MMP9), an extracellular protease linked to the progression of many types of cancer. By intercrossing with Mmp9 null mice, we found that the absence of MMP9 delayed tumor onset in the C3(1)-Tag model but had no effect on tumor onset in the MMTV-Neu model. We discovered that protein levels of insulin-like growth factor binding protein-1 (IGFBP-1), an MMP9 substrate, were increased in C3(1)-Tag;Mmp9(-/-) compared to C3(1)-Tag;Mmp9(+/+) tumors. In contrast, IGFBP-1 protein expression was low in MMTV-Neu tumors regardless of Mmp9 status. IGFBP-1 binds and antagonizes IGFs, preventing them from activating their receptors to promote cell proliferation and survival. Tumors from C3(1)-Tag;Mmp9(-/-) mice had reduced IGF-1 receptor phosphorylation, consistent with slower tumor onset. Finally, gene expression analysis of human breast tumors showed that high expression of IGFBP mRNA was strongly correlated with good prognosis but not when MMP9 mRNA was also highly expressed. In conclusion, MMP9 has different effects on breast cancer progression depending on whether IGFBPs are expressed.


Subject(s)
Breast Neoplasms/pathology , Insulin-Like Growth Factor Binding Protein 1/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Animals , Antigens, Viral, Tumor , Breast Neoplasms/metabolism , Disease Models, Animal , Disease Progression , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse , Mice , Mice, Knockout , Polyomavirus Infections , Protein Array Analysis , Retroviridae Infections , Simian virus 40 , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Virus Infections
10.
Adv Clin Chem ; 44: 65-102, 2007.
Article in English | MEDLINE | ID: mdl-17682340

ABSTRACT

The cellular receptor for urokinase, urokinase-type plasminogen activator receptor (uPAR) plays a central role in localizing its ligand, urokinase-type plasminogen activator (uPA) and thereby the plasminogen activation to the cell surface. uPA converts the proenzyme plasminogen to plasmin, which is involved in degradation of the extracellular matrix. In addition, uPA also cleaves uPAR, liberating the ligand-binding domain I, uPAR(I) and leaving the cleaved form, uPAR(II-III) on the cell surface. This cleavage inactivates the binding potential of uPAR toward uPA and vitronectin. uPAR can be shed from the cell surface and both intact and cleaved uPAR variants have been identified in tissue and body fluids. Identification and characterization of cleaved uPAR variants are dependent on monoclonal antibodies with known epitope specificity. Some of these have also been useful for the immunohistochemical localization of uPAR in tumor tissue. A number of immunoassays have been designed to measure uPAR and the collective amounts of all uPAR forms measured by enzyme-linked immunosorbent assay (ELISA) in tumor lysates or blood correlate to prognosis in several forms of cancer. However, the amounts of uPAR(I) and uPAR(II-III) may be directly related to the uPA activity and therefore be even stronger prognostic markers. Immunoassays measuring the individual uPAR forms have recently been designed and can be used to investigate this. This chapter is focused on the mechanism of uPAR cleavage and characterization and identification of the different uPAR forms in biological tissues and body fluids using immunologic methods. Monoclonal antibodies against uPAR are reviewed as well as different immunoassays used to investigate the prognostic potential of uPAR. Finally, an overview of the localization and prognostic significance of uPAR in different cancers and other malignancies is included.


Subject(s)
Antibodies, Monoclonal/immunology , Neoplasms/diagnosis , Neoplasms/metabolism , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Body Fluids/enzymology , Body Fluids/immunology , Body Fluids/metabolism , Humans , Immunoassay , Neoplasms/blood , Neoplasms/immunology , Receptors, Cell Surface/chemistry , Receptors, Urokinase Plasminogen Activator , Urokinase-Type Plasminogen Activator/immunology
11.
J Biol Chem ; 279(23): 24226-35, 2004 Jun 04.
Article in English | MEDLINE | ID: mdl-15024017

ABSTRACT

Protein-tyrosine phosphatases (PTPs) are considered important therapeutic targets because of their pivotal role as regulators of signal transduction and thus their implication in several human diseases such as diabetes, cancer, and autoimmunity. In particular, PTP1B has been the focus of many academic and industrial laboratories because it was found to be an important negative regulator of insulin and leptin signaling, and hence a potential therapeutic target in diabetes and obesity. As a result, significant progress has been achieved in the design of highly selective and potent PTP1B inhibitors. In contrast, little attention has been given to other potential drug targets within the PTP family. Guided by x-ray crystallography, molecular modeling, and enzyme kinetic analyses with wild type and mutant PTPs, we describe the development of a general, low molecular weight, non-peptide, non-phosphorus PTP inhibitor into an inhibitor that displays more than 100-fold selectivity for PTPbeta over PTP1B. Of note, our structure-based design principles, which are based on extensive bioinformatics analyses of the PTP family, are general in nature. Therefore, we anticipate that this strategy, here applied to PTPbeta, in principle can be used in the design and development of selective inhibitors of many, if not most PTPs.


Subject(s)
Enzyme Inhibitors/pharmacology , Protein Tyrosine Phosphatases/antagonists & inhibitors , Cloning, Molecular , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/chemistry , Hydrogen Bonding , Insulin/metabolism , Kinetics , Leptin/metabolism , Ligands , Models, Chemical , Models, Molecular , Mutation , Phthalimides/chemistry , Protein Conformation , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/chemistry , Signal Transduction , Structure-Activity Relationship , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...