Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Curr Med Res Opin ; 38(6): 999-1009, 2022 06.
Article in English | MEDLINE | ID: mdl-35392751

ABSTRACT

OBJECTIVE: The totality-of-evidence approach requires that similarity between a proposed biosimilar and a reference biologic is demonstrated across a range of analytical, preclinical, and clinical parameters to establish biosimilarity. We describe the totality of evidence for Sandoz biosimilar pegfilgrastim (LA-EP2006 [marketed as Ziextenzo]) that supported its regulatory approval in Europe and the United States. METHODS: Analytical similarity to the reference biologic [marketed by Amgen as Neulasta] was first investigated with regard to physiochemical quality attributes such as primary structure, pegylation, higher-order structures, variants and impurities, molecular size variants, and formulation (protein content, pH, excipients, etc.). In vitro biological activity studies were performed to examine the primary mechanism of action of pegfilgrastim. Bioequivalence (clinical pharmacokinetics [PK] and pharmacodynamics [PD]) of Sandoz biosimilar pegfilgrastim to the reference biologic was studied in healthy volunteers; efficacy, safety, and immunogenicity were assessed during confirmatory clinical efficacy studies in patients undergoing treatment for breast cancer. RESULTS: No meaningful or relevant differences were identified between Sandoz biosimilar pegfilgrastim and the reference biologic during analytical testing. Similar receptor binding and induction of cellular proliferation in vitro confirmed no functional differences between the biologics. Clinical studies in healthy adult participants demonstrated PK/PD biosimilarity and a similar safety profile between biosimilar and reference pegfilgrastim. Clinical studies in a sensitive patient population also demonstrated similar efficacy, safety, and immunogenicity between Sandoz biosimilar pegfilgrastim and the reference biologic. CONCLUSIONS: The totality of evidence confirms that Sandoz biosimilar pegfilgrastim matches the reference biologic and will therefore provide equivalent efficacy and safety in all eligible indications.


Subject(s)
Biosimilar Pharmaceuticals , Adult , Biosimilar Pharmaceuticals/adverse effects , Filgrastim/therapeutic use , Humans , Polyethylene Glycols/therapeutic use , Therapeutic Equivalency , United States
2.
AAPS J ; 22(1): 7, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31792633

ABSTRACT

For biosimilar drug development programs, it is essential to demonstrate that there are no clinically significant differences between the proposed biosimilar therapeutic (biosimilar) and its reference product (originator). Based on a stepwise comprehensive comparability exercise, the biosimilar must demonstrate similarity to the originator in physicochemical characteristics, biological activity, pharmacokinetics, efficacy, and safety, including immunogenicity. The goal of the immunogenicity assessment is to evaluate potential differences between the proposed biosimilar product and the originator product in the incidence and severity of human immune responses. Establishing that there are no clinically meaningful differences in the immune response between the products is a key element in the demonstration of biosimilarity. An issue of practical, regulatory, and financial importance is to establish whether a two-assay (based on the biosimilar and originator respectively) or a one-assay approach (based on the biosimilar) is optimal for the comparative immunogenicity assessment. This paper recommends the use of a single, biosimilar-based assay for assessing immunogenic similarity in support of biosimilar drug development. The development and validation of an ADA assay used for a biosimilar program should include all the assessments recommended for an innovator program (10-16, 29). In addition, specific parameters also need to be evaluated, to gain confidence that the assay can detect antibodies against both the biosimilar and the originator. Specifically, the biosimilar and the originator should be compared in antigenic equivalence, to assess the ability of the biosimilar and the originator to bind in a similar manner to the positive control(s), as well as in the confirmatory assay and drug tolerance experiments. Practical guidance for the development and validation of anti-drug antibody (ADA) assays to assess immunogenicity of a biosimilar in comparison to the originator, using the one-assay approach, are described herein.


Subject(s)
Biosimilar Pharmaceuticals , Immunologic Techniques , Validation Studies as Topic
3.
Br J Clin Pharmacol ; 84(12): 2790-2801, 2018 12.
Article in English | MEDLINE | ID: mdl-30079636

ABSTRACT

AIMS: This study aimed to demonstrate that the pharmacokinetic (PK) and pharmacodynamic (PD) profile of Sandoz proposed biosimilar pegfilgrastim (LA-EP2006) matches reference pegfilgrastim (Neulasta® ) in healthy subjects. Safety and immunogenicity were also assessed. METHODS: The phase I, randomized, double-blind, two-period crossover study consisted of two treatment periods separated by an 8-week washout period. Healthy subjects aged 18-45 were randomized to either proposed biosimilar/reference pegfilgrastim or reference pegfilgrastim/proposed biosimilar. Proposed biosimilar and reference pegfilgrastim were administered on Day 1 of each treatment period (single 6 mg subcutaneous injection). Blood samples for PK/PD analysis were taken predose and ≤336 h postdose. PK/PD similarity was claimed if 90% (PK) and 95% (PD) confidence intervals (CI) for geometric mean ratios of the area under the serum concentration-time curve (AUC) from time of dosing and extrapolated to infinity (AUC0-inf ), or to the last measurable concentration (AUC0-last ), maximum observed serum concentration (Cmax ), absolute neutrophil count (ANC) area under the effect curve from the time of dosing to the last measurable concentration (AUEC0-last ) and ANC maximum effect attributable to the therapy under investigation (Emax ) were completely contained within the predefined margin (0.8 to 1.25). RESULTS: Overall, 169 subjects completed the study. PK/PD similarity was demonstrated; 90% CIs of geometric mean ratio of proposed biosimilar/reference for PK: AUC0-inf (1.0559-1.2244), AUC0-last (1.0607-1.2328), Cmax (1.0312-1.1909) and 95% CIs for PD (ANC): AUEC0-last (0.9948-1.0366), Emax (0.9737-1.0169) were completely contained within predefined margin of 0.8 to 1.25. Both biologics had similar safety profiles, were well tolerated and had low incidence of anti-drug antibodies. No neutralizing or clinically relevant antibodies were detected. CONCLUSIONS: PK/PD similarity of Sandoz proposed biosimilar pegfilgrastim and reference pegfilgrastim was confirmed. No clinically meaningful differences in safety, tolerability and immunogenicity were observed in healthy subjects.


Subject(s)
Biosimilar Pharmaceuticals/pharmacokinetics , Filgrastim/pharmacokinetics , Polyethylene Glycols/pharmacokinetics , Adult , Cross-Over Studies , Double-Blind Method , Female , Filgrastim/adverse effects , Filgrastim/immunology , Filgrastim/pharmacology , Healthy Volunteers , Humans , Male , Polyethylene Glycols/adverse effects , Polyethylene Glycols/pharmacology
4.
Am J Nephrol ; 46(5): 364-370, 2017.
Article in English | MEDLINE | ID: mdl-29084409

ABSTRACT

BACKGROUND: HX575 (biosimilar epoetin alfa) was approved in Europe in 2007 for the treatment of chronic kidney disease (CKD)-related anemia. This study assessed the clinical equivalence of HX575 with the US-licensed reference epoetin alfa (Epogen®/Procrit®, Amgen/Janssen) following subcutaneous (SC) administration in dialysis patients with CKD-related anemia. METHODS: This randomized, double-blind, parallel-group, multicenter study (NCT01693029) was conducted at 49 US clinical sites. Eligible patients were aged ≥18 years, had end-stage renal disease, were on hemodialysis or peritoneal dialysis for ≥6 months (or ≥12 months in the case of a failed kidney transplant), and were receiving treatment with stable SC doses of epoetin alfa. Eligible patients also had mean hemoglobin (Hb) concentration between 9.0 and 11.5 g/dL during the screening period. The primary endpoint was the mean absolute change in Hb concentration between the screening/baseline period (week-4 to week-1) and the evaluation period (weeks 21 to 28). RESULTS: Hb values at the end of the evaluation period and the Hb change from baseline to evaluation period were similar between treatment groups. The estimated difference between groups in mean absolute change in Hb concentration was -0.093 g/dL, with 90% CI (-0.23 to 0.04) entirely within the pre-specified equivalence limits (-0.5 to 0.5 g/dL). The safety profile of each medicine was similar and as expected in dialysis patients, and neither method of treatment led to the development of neutralizing, clinically relevant antibodies. CONCLUSIONS: SC HX575 in dialysis patients with renal anemia was therapeutically equivalent to the reference medicine in terms of maintaining stable Hb levels and safety.


Subject(s)
Anemia/drug therapy , Biosimilar Pharmaceuticals/therapeutic use , Epoetin Alfa/therapeutic use , Hematinics/therapeutic use , Kidney Failure, Chronic/drug therapy , Adult , Aged , Aged, 80 and over , Anemia/blood , Double-Blind Method , Female , Hemoglobins/analysis , Humans , Kidney Failure, Chronic/blood , Male , Middle Aged , Renal Dialysis , Therapeutic Equivalency , Treatment Outcome , United States , Young Adult
5.
Clin Nephrol ; 88(10): 190-197, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28766493

ABSTRACT

AIM: To assess the safety and immunogenicity of subcutaneous (SC) HX575 (epoetin-α) in dialysis- and nondialysis-dependent adult patients with chronic kidney disease (CKD). METHODS: Open-label, single-arm, multicenter study in patients (n = 416) from Germany, Italy, Poland, Romania, Russia, Turkey, and Ukraine. RESULTS: Mean (standard deviation (SD)) age was 52.3 (15.8) years, all patients were Caucasian, and similar proportions were male/female. 250 patients (60.1%) were erythropoiesis-stimulating agent (ESA)-naïve, and 166 (39.9%) were receiving ESA maintenance therapy at study start; mean (SD) on-study treatment duration with HX575 was 43.4 (15.8) weeks and 45.3 (13.7) weeks, respectively. Binding antierythropoietin (EPO) antibodies were detected by radioimmunoprecipitation (RIP) assay in 7 patients (1.7%; incidence 0.019); 5 of these were ESA-naïve at study entry. No patient developed neutralizing antibodies as determined in a cell-based epoetin neutralizing assay. Of the 7 patients with a positive binding anti-EPO RIP assay, 4 tested negative at later time points while continuing HX575 treatment. Three patients had low titers of anti-EPO antibodies at the last study assessment. There were no clinical signs of immunogenicity or hypersensitivity. CONCLUSIONS: SC HX575 was effective for correcting and maintaining correction of anemia, and the mean weekly dose remained stable over time.
.


Subject(s)
Anemia/drug therapy , Epoetin Alfa/adverse effects , Hematinics/adverse effects , Recombinant Proteins/adverse effects , Renal Dialysis/adverse effects , Renal Insufficiency, Chronic/complications , Adult , Aged , Anemia/etiology , Epoetin Alfa/therapeutic use , Erythropoietin , Europe , Female , Hematinics/therapeutic use , Humans , Male , Middle Aged , Recombinant Proteins/therapeutic use , Renal Insufficiency, Chronic/therapy
6.
Oncologist ; 21(7): 789-94, 2016 07.
Article in English | MEDLINE | ID: mdl-27091420

ABSTRACT

BACKGROUND: Pegfilgrastim is widely used for the prevention of chemotherapy-induced neutropenia. In highly regulated markets, there are currently no approved biosimilars of pegfilgrastim. Pegfilgrastim Randomized Oncology (Supportive Care) Trial to Evaluate Comparative Treatment (PROTECT-2) was a confirmatory efficacy and safety study designed to compare proposed biosimilar LA-EP2006 with reference pegfilgrastim (Neulasta, Amgen) in early-stage breast cancer patients receiving adjuvant or neoadjuvant myelosuppressive chemotherapy. METHODS: A total of 308 patients were randomized to LA-EP2006 or reference pegfilgrastim. Each patient received TAC (intravenous docetaxel 75 mg/m(2), doxorubicin 50 mg/m(2), and cyclophosphamide 500 mg/m(2)) on day 1 of each cycle, for six or more cycles. Pegfilgrastim (LA-EP2006 or reference) was given subcutaneously (6 mg in 0.6 mL) on day 2 of each cycle. The primary endpoint was duration of severe neutropenia (DSN) during cycle 1 (number of consecutive days with an absolute neutrophil count <0.5 × 10(9)/L), with equivalence confirmed if 90% and 95% confidence intervals (CIs) were within a 1-day margin. RESULTS: Baseline characteristics were well balanced. DSN was equivalent between groups at mean ± SD 1.36 ± 1.13 (LA-EP2006, n = 155) and 1.19 ± 0.98 (reference, n = 153) in cycle 1. With a treatment difference (reference minus LA-EP2006) of -0.16 days (90% CI -0.36 to 0.04; 95% CI -0.40 to 0.08), LA-EP2006 was equivalent to reference pegfilgrastim. Secondary efficacy parameters were similar between groups during cycle 1 and across cycles. Safety profiles were also similar between groups. No neutralizing antibodies against pegfilgrastim, filgrastim, or polyethylene glycol were detected. CONCLUSION: LA-EP2006 and reference pegfilgrastim were therapeutically equivalent and comparable regarding efficacy and safety in the prevention of neutropenia in patients with early-stage breast cancer receiving TAC. IMPLICATIONS FOR PRACTICE: The granulocyte colony-stimulating factor pegfilgrastim is widely used for the prevention of chemotherapy-induced neutropenia. Biosimilars are biologics with similar quality, safety, and efficacy to a reference product that may increase the affordability of treatment compared with their reference compounds. There are currently no approved biosimilars of pegfilgrastim in highly regulated markets. No previous phase III studies have been performed with LA-EP2006. PROTECT-2 was conducted to confirm the similarity of the proposed biosimilar LA-EP2006 to pegfilgrastim. Biosimilar pegfilgrastim (LA-EP2006) may benefit oncology patients by offering increased access to biological treatments that may improve clinical outcomes. This means that patients could potentially be treated prophylactically with biologics rather than only after complications have occurred.


Subject(s)
Biosimilar Pharmaceuticals/therapeutic use , Breast Neoplasms/drug therapy , Granulocyte Colony-Stimulating Factor/therapeutic use , Neoadjuvant Therapy , Neutropenia/prevention & control , Adult , Bone Marrow/drug effects , Breast Neoplasms/pathology , Double-Blind Method , Female , Filgrastim , Granulocyte Colony-Stimulating Factor/adverse effects , Humans , Middle Aged , Neoplasm Staging , Polyethylene Glycols , Prospective Studies , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use
7.
J Neurochem ; 107(2): 418-31, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18702663

ABSTRACT

Repulsive guidance molecule A (RGM A) was recently described as a potent inhibitor of neuroregeneration in a rat spinal cord injury model. The receptor mediating RGM A's repulsive activity was shown to be Neogenin, a member of the Deleted in Colorectal Cancer (DCC) family of netrin receptors. Binding of RGM A to Neogenin induces activation of the small GTPase RhoA and of its effector Rho-kinase by an unknown mechanism. Here we show, that the cytoplasmic tail of Neogenin interacts directly with the transcriptional coactivator LIM domain only 4 (LMO4) in human SH-SY5Y cells, human Ntera neurons, and in embryonic rat cortical neurons. RGM A binding to Neogenin but not binding of Netrin-1, induces release of LMO4 from Neogenin. Down-regulation of LMO4 neutralizes the repulsive activity of RGM A in neuronal cell lines and embryonic rat cortical neurons and prevents RhoA activation. These results show for the first time that an interaction of Neogenin with LMO4 is involved in the RGM A - Neogenin signal transduction pathway for RhoA activation.


Subject(s)
Homeodomain Proteins/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/physiology , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Amides/pharmacology , Analysis of Variance , Animals , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , GPI-Linked Proteins , Gene Expression/drug effects , Green Fluorescent Proteins/biosynthesis , Homeodomain Proteins/biosynthesis , Humans , LIM Domain Proteins , Neurites/drug effects , Neurites/physiology , Neurons/cytology , Neurons/drug effects , Protein Structure, Tertiary , Pyridines/pharmacology , RNA, Small Interfering/pharmacology , Rats , Transcription Factors/biosynthesis , Transfection/methods , Tubulin/pharmacology , Two-Hybrid System Techniques , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
9.
Mol Cell ; 23(6): 887-97, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16973440

ABSTRACT

Aberrant folding and fibrillar aggregation by polyglutamine (polyQ) expansion proteins are associated with cytotoxicity in Huntington's disease and other neurodegenerative disorders. Hsp70 chaperones have an inhibitory effect on fibril formation and can alleviate polyQ cytotoxicity. Here we show that the cytosolic chaperonin, TRiC, functions synergistically with Hsp70 in this process and is limiting in suppressing polyQ toxicity in a yeast model. In vitro reconstitution experiments revealed that TRiC, in cooperation with the Hsp70 system, promotes the assembly of polyQ-expanded fragments of huntingtin (Htt) into soluble oligomers of approximately 500 kDa. Similar oligomers were observed in yeast cells upon TRiC overexpression and were found to be benign, in contrast to conformationally distinct Htt oligomers of approximately 200 kDa, which accumulated at normal TRiC levels and correlated with inhibition of cell growth. We suggest that TRiC cooperates with the Hsp70 system as a key component in the cellular defense against amyloid-like protein misfolding.


Subject(s)
Chaperonins/physiology , Peptides/chemistry , Chaperonins/metabolism , DNA Repeat Expansion , Green Fluorescent Proteins/analysis , HSP40 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Peptides/metabolism , Protein Folding , Recombinant Fusion Proteins/analysis , Repetitive Sequences, Amino Acid , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
10.
Philos Trans R Soc Lond B Biol Sci ; 361(1473): 1513-29, 2006 Sep 29.
Article in English | MEDLINE | ID: mdl-16939972

ABSTRACT

During the development of the nervous system, outgrowing axons often have to travel long distances to reach their target neurons. In this process, outgrowing neurites tipped with motile growth cones rely on guidance cues present in their local environment. These cues are detected by specific receptors expressed on growth cones and neurites and influence the trajectory of the growing fibres. Neurite growth, guidance, target innervation and synapse formation and maturation are the processes that occur predominantly but not exclusively during embryonic or early post-natal development in vertebrates. As a result, a functional neural network is established, which is usually remarkably stable. However, the stability of the neural network in higher vertebrates comes at an expensive price, i.e. the loss of any significant ability to regenerate injured or damaged neuronal connections in their central nervous system (CNS). Most importantly, neurite growth inhibitors prevent any regenerative growth of injured nerve fibres. Some of these inhibitors are associated with CNS myelin, others are found at the lesion site and in the scar tissue. Traumatic injuries in brain and spinal cord of mammals induce upregulation of embryonic inhibitory or repulsive guidance cues and their receptors on the neurites. An example for embryonic repulsive directional cues re-expressed at lesion sites in both the rat and human CNS is provided with repulsive guidance molecules, a new family of directional guidance cues.


Subject(s)
Central Nervous System/embryology , Central Nervous System/growth & development , Ephrin-A5/metabolism , Neurons/cytology , Animals , Central Nervous System/metabolism , Ephrin-A5/chemistry , Neurons/metabolism , Signal Transduction
11.
Biochem Biophys Res Commun ; 333(4): 1202-10, 2005 Aug 12.
Article in English | MEDLINE | ID: mdl-15978545

ABSTRACT

Protein aggregation is the key event in a number of human diseases such as Alzheimer's and Parkinson's disease. We present a general method to quantify and characterize protein aggregates by dual-colour scanning for intensely fluorescent targets (SIFT). In addition to high sensitivity, this approach offers a unique opportunity to study co-aggregation processes. As the ratio of two fluorescently labelled components can be analysed for each aggregate separately in a homogeneous assay, the molecular composition of aggregates can be studied even in samples containing a mixture of different types of aggregates. Using this method, we could show that wild-type alpha-synuclein forms co-aggregates with a mutant variant found in familial Parkinson's disease. Moreover, we found a striking increase in aggregate formation at non-equimolar mixing ratios, which may have important therapeutic implications, as lowering the relative amount of aberrant protein may cause an increase of protein aggregation leading to adverse effects.


Subject(s)
Amyloid/chemistry , Amyloid/ultrastructure , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/ultrastructure , Spectrometry, Fluorescence/methods , Amyloid/analysis , Binding Sites , Dimerization , Multiprotein Complexes/analysis , Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , Nerve Tissue Proteins/analysis , Protein Binding , Synucleins , alpha-Synuclein
12.
Mol Cell ; 15(1): 95-105, 2004 Jul 02.
Article in English | MEDLINE | ID: mdl-15225551

ABSTRACT

The expression of polyglutamine-expanded mutant proteins in Huntington's disease and other neurodegenerative disorders is associated with the formation of intraneural inclusions. These aggregates could potentially cause cellular toxicity by sequestering essential proteins possessing normal polyQ repeats, including the transcription factors TBP and CBP. We show, in vitro and in cells, that monomers or small soluble oligomers of huntingtin exon1 accumulate in the nucleus and inhibit the function of TBP in a polyQ-dependent manner. FRET experiments indicate that these toxic forms are generated through a conformational rearrangement in huntingtin. Interaction of toxic huntingtin with the benign polyQ repeat of TBP structurally destabilizes the transcription factor, independent of the formation of insoluble coaggregates. Hsp70/Hsp40 chaperones interfere with the conformational change in mutant huntingtin and inhibit the deactivation of TBP. These results outline a molecular mechanism of cellular toxicity in polyQ disease and can explain the beneficial effects of molecular chaperones.


Subject(s)
Nerve Tissue Proteins/toxicity , Nuclear Proteins/toxicity , Peptides/genetics , Transcription Factors/antagonists & inhibitors , Trinucleotide Repeat Expansion/genetics , Animals , CREB-Binding Protein , Cell Line, Tumor , Cell Nucleus/metabolism , Exons , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Huntingtin Protein , Macromolecular Substances , Mice , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Mutation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peptides/metabolism , Protein Conformation , Protein Folding , Saccharomyces cerevisiae , TATA-Box Binding Protein/antagonists & inhibitors , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/metabolism , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Semin Cell Dev Biol ; 15(1): 17-29, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15036203

ABSTRACT

Human misfolding diseases result from the failure of proteins to reach their active state or from the accumulation of aberrantly folded proteins. The mechanisms by which molecular chaperones influence the development of these diseases is beginning to be understood. Mutations that compromise the activity of chaperones lead to several rare syndromes. In contrast, the more frequent amyloid-related neurodegenerative diseases are caused by a gain of toxic function of misfolded proteins. Toxicity in these disorders may result from an imbalance between normal chaperone capacity and production of dangerous protein species. Increased chaperone expression can suppress the neurotoxicity of these molecules, suggesting possible therapeutic strategies.


Subject(s)
Disease/etiology , Molecular Chaperones/physiology , Protein Folding , Proteins/chemistry , Amyloid/chemistry , Amyloid/physiology , Chaperonin 60/genetics , Chaperonin 60/physiology , Chaperonins/physiology , Cysteine Endopeptidases/physiology , Cytosol/physiology , Endoplasmic Reticulum/physiology , Eye Proteins/genetics , Eye Proteins/physiology , GTP-Binding Proteins , Group II Chaperonins , HSP70 Heat-Shock Proteins/physiology , HSP90 Heat-Shock Proteins/physiology , Heat-Shock Proteins/genetics , Heat-Shock Proteins/physiology , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Models, Biological , Molecular Chaperones/genetics , Multienzyme Complexes/physiology , Mutation , Proteasome Endopeptidase Complex , Proteins/physiology , Ubiquitins/physiology , alpha-Crystallins/genetics , alpha-Crystallins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...